CN113666853A - Biaryl compounds useful as ROR gamma modulators - Google Patents

Biaryl compounds useful as ROR gamma modulators Download PDF

Info

Publication number
CN113666853A
CN113666853A CN202110526063.0A CN202110526063A CN113666853A CN 113666853 A CN113666853 A CN 113666853A CN 202110526063 A CN202110526063 A CN 202110526063A CN 113666853 A CN113666853 A CN 113666853A
Authority
CN
China
Prior art keywords
alkyl
halogen
optionally substituted
compound
pharmaceutically acceptable
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
CN202110526063.0A
Other languages
Chinese (zh)
Other versions
CN113666853B (en
Inventor
程耀邦
黄亚飞
周娟
董志强
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.)
Lidi (Hangzhou) Pharmaceutical Technology Co.,Ltd.
Original Assignee
Shanghai Huiqi Biomedical Technology 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 Shanghai Huiqi Biomedical Technology Co ltd filed Critical Shanghai Huiqi Biomedical Technology Co ltd
Publication of CN113666853A publication Critical patent/CN113666853A/en
Application granted granted Critical
Publication of CN113666853B publication Critical patent/CN113666853B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C317/00Sulfones; Sulfoxides
    • C07C317/44Sulfones; Sulfoxides having sulfone or sulfoxide groups and carboxyl groups bound to the same carbon skeleton
    • C07C317/46Sulfones; Sulfoxides having sulfone or sulfoxide groups and carboxyl groups bound to the same carbon skeleton the carbon skeleton being further substituted by singly-bound oxygen atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • A61K31/167Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide having the nitrogen of a carboxamide group directly attached to the aromatic ring, e.g. lidocaine, paracetamol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/397Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having four-membered rings, e.g. azetidine
    • 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
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/02Nasal agents, e.g. decongestants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/08Bronchodilators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • 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
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C317/00Sulfones; Sulfoxides
    • C07C317/14Sulfones; Sulfoxides having sulfone or sulfoxide groups bound to carbon atoms of six-membered aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C317/00Sulfones; Sulfoxides
    • C07C317/44Sulfones; Sulfoxides having sulfone or sulfoxide groups and carboxyl groups bound to the same carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C317/00Sulfones; Sulfoxides
    • C07C317/44Sulfones; Sulfoxides having sulfone or sulfoxide groups and carboxyl groups bound to the same carbon skeleton
    • C07C317/48Sulfones; Sulfoxides having sulfone or sulfoxide groups and carboxyl groups bound to the same carbon skeleton the carbon skeleton being further substituted by singly-bound nitrogen atoms, not being part of nitro or nitroso groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C317/00Sulfones; Sulfoxides
    • C07C317/44Sulfones; Sulfoxides having sulfone or sulfoxide groups and carboxyl groups bound to the same carbon skeleton
    • C07C317/48Sulfones; Sulfoxides having sulfone or sulfoxide groups and carboxyl groups bound to the same carbon skeleton the carbon skeleton being further substituted by singly-bound nitrogen atoms, not being part of nitro or nitroso groups
    • C07C317/50Sulfones; Sulfoxides having sulfone or sulfoxide groups and carboxyl groups bound to the same carbon skeleton the carbon skeleton being further substituted by singly-bound nitrogen atoms, not being part of nitro or nitroso groups at least one of the nitrogen atoms being part of any of the groups, X being a hetero atom, Y being any atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D205/00Heterocyclic compounds containing four-membered rings with one nitrogen atom as the only ring hetero atom
    • C07D205/02Heterocyclic compounds containing four-membered rings with one nitrogen atom as the only ring hetero atom not condensed with other rings
    • C07D205/04Heterocyclic compounds containing four-membered rings with one nitrogen atom as the only ring hetero atom not condensed with other rings having no double bonds between ring members or between ring members and non-ring members
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/62Oxygen or sulfur atoms
    • C07D213/70Sulfur atoms
    • C07D213/71Sulfur atoms to which a second hetero atom is attached
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/07Optical isomers

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Pulmonology (AREA)
  • Immunology (AREA)
  • Diabetes (AREA)
  • Dermatology (AREA)
  • Virology (AREA)
  • Rheumatology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Pain & Pain Management (AREA)
  • Oncology (AREA)
  • Hematology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Communicable Diseases (AREA)
  • Cardiology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Transplantation (AREA)
  • Emergency Medicine (AREA)
  • Endocrinology (AREA)
  • Molecular Biology (AREA)
  • Obesity (AREA)
  • Ophthalmology & Optometry (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Otolaryngology (AREA)
  • Urology & Nephrology (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention relates to compounds of formula (I), stereoisomers, tautomers, stable isotopic variations, pharmaceutically acceptable salts or solvates thereof, pharmaceutical compositions comprising the same, methods of using the same for treating or preventing ROR γ t-related diseases, and uses thereof in the manufacture of medicaments for treating or preventing ROR γ t-related diseases。

Description

Biaryl compounds useful as ROR gamma modulators
Technical Field
The invention belongs to the technical field of chemical medicines, and particularly relates to a biaryl compound with ROR gamma t inhibitory activity, a pharmaceutical composition containing the compound, a method for preparing the compound, and application of the compound in preparing a medicine for preventing or treating ROR gamma t related diseases.
Background
Retinoic acid receptor-Related Orphan Receptors (RORs), also known as NF1R, belong to a subfamily of the nuclear receptor superfamily of ligand-dependent transcription factors. The RORs subfamily mainly includes three subtypes, ROR α, ROR β and ROR γ. ROR γ contains two members: ROR γ 1 (also called ROR γ) and ROR γ 2 (also called ROR γ t), in which ROR γ 1 is distributed in skeletal muscle, thymus, testis, pancreas, prostate, heart, liver, and the like, and ROR γ t is expressed only in some immune cells.
Littman et al first reported that ROR γ T was necessary for the initial differentiation of CD4+ T cells into Th17 cells (Cell,2006126, 1121-1133). During the differentiation process of antigen-stimulated Thp cells to Th17 cells, ROR gamma t is induced and expressed under the action of cytokines such as IL-6, IL-21 and TGF-beta. The ability of Thp cells isolated from ROR γ t-deficient mice to differentiate into the Th17 cell line was significantly reduced. These all indicate that ROR γ t is a key regulator for promoting differentiation of Th17 cells.
Th17 cells are one of the helper T cells, and produce IL-17 and other proinflammatory cytokines. Th17 cells play a key role in many mouse models of autoimmune disease, such as Experimental Allergic Encephalomyelitis (EAE) and collagen-induced arthritis (CIA) animal models. Furthermore, increased IL-17 levels are detectable in some human autoimmune diseases, including Rheumatoid Arthritis (RA), Multiple Sclerosis (MS), Psoriasis (Psoriasis) and Inflammatory Bowel Disease (IBD). The number of Th17 cells found in both tissues and peripheral blood samples from patients with autoimmune disease is increased. Therefore, the Th17 cell or the cytokine IL-17 produced by it are closely related to the pathogenesis of inflammation and autoimmune diseases.
The monoclonal antibody Cosentyx (Secukinumab/AIN457) developed by Nowa, which is used to treat psoriasis by specifically blocking IL-17, was approved by the FDA to be marketed in 1 month 2015, and is the first drug acting on IL-17 in the psoriasis-treating drug market. Subsequently, the monoclonal antibody ixekizumab, targeting the pro-inflammatory cytokine IL-17A, was approved for use in the indications psoriasis, psoriatic arthritis. The clinical success of these monoclonal antibodies demonstrates the importance of the IL-17 signaling pathway in inflammatory and autoimmune diseases and demonstrates the potential for treating inflammatory and autoimmune diseases by affecting the IL-17 signaling pathway through ROR γ t inhibitors.
Therefore, ROR gamma t can be used as a new target of drugs for treating autoimmune diseases, and the search for ROR gamma t small molecule inhibitors and the application of ROR gamma t small molecule inhibitors in treating ROR gamma t mediated diseases such as inflammation and autoimmune diseases have important significance.
To date, a total of 4 small molecules of ROR γ t inhibitors were in clinical stage 2, 7 small molecules of ROR γ t inhibitors were in clinical stage 1, and none of the compounds had entered clinical stage 3. Therefore, there is still a great need to find and develop new ROR γ t inhibitor compounds for the prevention and/or treatment of diseases associated with ROR γ t, such as inflammatory and autoimmune diseases. In addition to satisfactory ROR γ t inhibitory activity, such compounds are expected to have high selectivity for ROR subtypes and good or even improved druggability based on structural optimization, so as to provide more drug options and better therapeutic effects for patients with related diseases.
Brief description of the invention
The present invention relates to compounds useful for the prevention or treatment of diseases associated with ROR γ t. In particular, it has been identified that the compounds of the present invention not only show satisfactory ROR γ t inhibitory activity, have the ability to regulate Th17 cell differentiation, thereby inhibiting IL-17 production, but also show good performance in vivo pharmacokinetic experiments, suggesting improved druggability and improved bioavailability; in addition, the compound also shows good safety and has lower risk of drug interaction. Therefore, the compound of the present invention can not only achieve the purpose for preventing or treating diseases associated with ROR γ t, but also the prepared drug is expected to have improved absorption, enhanced therapeutic effect at the same dose, or provide the same therapeutic effect at a lower dose, longer half-life and/or reduce possible side effects. Thus, the present invention also provides the use of the compounds of the present invention for the preparation of medicaments for the prevention or treatment of diseases associated with ROR γ t, pharmaceutical compositions comprising the compounds and methods for the prevention and/or treatment of diseases associated with ROR γ t by administering the compounds.
Accordingly, in one aspect of the present invention, there is provided a compound of formula (I), a stereoisomer, a tautomer, a stable isotopic variation, a pharmaceutically acceptable salt, or a solvate thereof:
Figure BDA0003065598540000021
wherein:
x is selected from CH or N;
R1selected from hydrogen, halogen, cyano, nitro, C1-C6Alkyl, -O-C1-C6Alkyl, S-C1-C6Alkyl, -NH-C1-C6Alkyl, -N- (C)1-C6Alkyl radical)2、C1-C6alkyl-O-C1-C6Alkyl, -C1-C6alkyl-S-C1-C6Alkyl radical, C1-C6alkyl-NH-C1-C6Alkyl or C1-C6alkyl-N (C)1-C6Alkyl radical)2Wherein said C is1-C6Alkyl radical orOptionally substituted by halogen or cyano;
R2selected from hydrogen, halogen, cyano, nitro or C1-C6Alkyl radical, wherein said C1-C6Alkyl is optionally substituted with substituents independently selected from: -Raor-OH, wherein RaIs C optionally substituted by halogen1-C6An alkyl group;
R3、R4each independently selected from halogen, cyano, nitro, Rb、-ORb、-SRbor-NRbRbWherein R isbIs H or optionally substituted C1-C6Alkyl, each of said substituents being independently selected from hydrogen, halogen, cyano, nitro, -OH, -SH, -NH2、-O-C1-C6Alkyl, -S-C1-C6Alkyl, -NH-C1-C6Alkyl or-N (-C)1-C6Alkyl radical)2
R5Is selected from-ORc、-SRc、-NRcRc、-NHCORc、-NHSO2Rc、-COORc、-CONRcRcor-SO2NRcRcWherein R iscIndependently at each occurrence, is selected from hydrogen, C optionally substituted with halogen1-C6Alkyl or C optionally substituted by halogen3-C7Cycloalkyl, or two R bound to the same N atomcMay form, together with the N atom to which they are attached, a 4-7 membered nitrogen containing heterocycloalkyl group;
R6is selected from C1-C6Alkyl radical, C3-C7Cycloalkyl, 4-7 membered heterocycloalkyl or optionally substituted by C1-C6Alkyl radical, C3-C7Cycloalkyl or 4-7 membered heterocycloalkyl substituted-NH2Wherein said C is1-C6Alkyl radical, C3-C7Cycloalkyl or 4-7 membered heterocycloalkyl is optionally substituted with substituents each independently selected from: hydrogen, halogen, cyano, nitro, -Ra、-ORa、-SRaor-NRaRaOr optionally substituted by halogenSubstituted by elements C3-C7Cycloalkyl radical, RaIndependently at each occurrence, is selected from H or C optionally substituted with halogen1-C6Alkyl, wherein two groups attached to the N atom of the amino group may form a 4-7 membered ring together with the N to which they are attached; and is
n is selected from 0, 1 or 2.
In another aspect of the present invention, there is provided a compound of formula (I), a stereoisomer, a tautomer, a stable isotopic variant, a pharmaceutically acceptable salt or a solvate thereof, having ROR γ t inhibitory activity for use as a medicament, particularly as an inhibitor of ROR γ t.
In another aspect of the invention, there is provided a compound of formula (I), a stereoisomer, a tautomer, a stable isotopic variant, a pharmaceutically acceptable salt, or a solvate thereof, for use in the treatment, particularly in the treatment or prevention of a disease associated with ROR γ t.
In another aspect of the invention, there is provided a pharmaceutical composition comprising a compound of the invention as described above and a pharmaceutically acceptable excipient. In a specific aspect, the pharmaceutical composition of the present invention is provided for the prevention or treatment of diseases associated with roryt. In a particular aspect, the pharmaceutical composition may additionally comprise additional therapeutically active ingredients suitable for use in combination with the compounds of the present invention.
In another aspect of the present invention, there is provided a pharmaceutical combination comprising a compound of the invention as described above and an additional active agent.
In another aspect of the present invention, there is provided a method for the prevention or treatment of diseases associated with roryt in a mammal, particularly a human, which comprises administering an effective amount of a compound of the present invention described herein or a pharmaceutical composition comprising the same.
In another aspect of the present invention, there is provided a use of the above-described compound or pharmaceutical composition of the present invention for preventing or treating a disease associated with roryt.
In another aspect of the present invention, there is provided a use of the above-described compound or pharmaceutical composition of the present invention for the preparation of a medicament for the prevention or treatment of a disease associated with roryt.
In further aspects, methods are provided for the synthesis of compounds of the invention, wherein representative synthetic schemes and routes are described below.
Other objects and advantages of the present invention will become apparent to those skilled in the art from a reading of the following detailed description.
Detailed Description
Definition of
Unless otherwise indicated, each term used in the specification and claims has the following meaning. Where a particular term or phrase is not specifically defined, it should not be considered uncertain or clear, but rather should be understood appropriately according to the context herein or ordinary meaning in the art. Many of the groups defined herein are optionally substituted and the list of substituents given in this definition section are exemplary only and are not intended to limit the substituents defined elsewhere in this specification and claims.
The term "alkyl" as used herein means a straight or branched chain aliphatic hydrocarbon group having the indicated number of carbon atoms. Specifically, the alkyl group can have 1 to 6,1 to 5, 1 to 4, 1 to 3, or 1 to 2 carbon atoms. Suitably C1-6Examples of alkyl groups include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, isopentyl, neopentyl, n-hexyl, and isohexyl. Particular alkyl groups have 1 to 3 carbon atoms.
The term "alkoxy" as used herein means the group-O-alkyl, wherein alkyl has the meaning described herein. In particular, the term refers to the group-O-C1-6An alkyl group. Examples of suitable alkoxy groups include, but are not limited to, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, tert-butoxy, sec-butoxy, n-pentoxy, n-hexoxy, and 1, 2-dimethylbutoxy. Particular alkoxy groups have 1 to 3 carbon atoms.
The term "alkylthio" as used herein means the group-S-alkyl, wherein alkyl has the meaning described herein. In particular, the term refers to the group-S-C1-6An alkyl group. Examples of suitable alkylthio groups include, but are not limited to, methylthio, ethylthio, n-propylthio, isopropylthio, n-butylthio, t-butylthio, sec-butylthio, n-pentylthio, n-hexylthio, and 1, 2-dimethylbutylthio. Particular alkylthio groups have 1 to 3 carbon atoms.
As used herein, the term "halogen-substituted C1-C6Alkyl "means C as defined above1-C6Alkyl, wherein one or more (e.g. 1,2, 3, 4 or 5) hydrogen atoms are replaced by halogen. It will be understood by those skilled in the art that when there is more than one halogen substituent, the halogens may be the same or different and may be located on the same or different C atoms. "halogen-substituted C1-C6Examples of alkyl radicals "are, for example, -CH2F、-CHF2、-CF3、-CCl3、-C2F5、-C2Cl5、-CH2CF3、-CH2Cl、CH2CH2CF3or-CF (CF)3)2And the like.
As used herein, the term "halogen-substituted C1-C6Alkoxy "means C as defined above1-C6Alkoxy, wherein one or more (e.g. 1,2, 3, 4 or 5) hydrogen atoms are replaced by halogen. It will be understood by those skilled in the art that when there is more than one halogen substituent, the halogens may be the same or different and may be located on the same or different C atoms. "halogen-substituted C1-C6Examples of alkoxy "are e.g. -OCH2F、-OCHF2、-OCF3、-OCCl3、-OC2F5、-OC2Cl5、-OCH2CF3、-OCH2Cl or-OCH2CH2CF3And the like.
As used herein, the term "halogen-substituted C1-C6Alkylthio "refers to C as defined above1-C6Alkylthio, wherein one or more (e.g. 1,2, 3, 4 or 5) hydrogen atoms are replaced by halogen. It will be understood by those skilled in the art that when there is more than one halogen substituent, the halogenMay be the same or different and may be located on the same or different C atoms. "halogen-substituted C1-C6Examples of alkylthio "are e.g. -SCH2F、-SCHF2、-SCF3、-SCCl3、-SC2F5、-SC2Cl5、-SCH2CF3、-SCH2Cl or-SCH2CH2CF3And the like.
The term "cycloalkyl" as used herein means a monocyclic, fused polycyclic, bridged polycyclic or spiro non-aromatic saturated hydrocarbon ring structure having the indicated number of ring atoms. The cycloalkyl group can have 3 to 12 carbon atoms, specifically 3 to 10, and more specifically 3 to 7 carbon atoms, i.e., C3-C7A cycloalkyl group. Examples of suitable cycloalkyl groups include, but are not limited to, monocyclic C3-C7Cycloalkyl groups such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl. Particular cycloalkyl groups have 3 to 5 carbon atoms.
As used herein, the term "halogen-substituted C3-C7Cycloalkyl "means C as defined above3-C7Cycloalkyl, wherein one or more (e.g. 1,2, 3, 4 or 5) hydrogen atoms are replaced by halogen. It will be understood by those skilled in the art that when there is more than one halogen substituent, the halogens may be the same or different and may be located on the same or different C atoms. Specific examples include, but are not limited to, 2-fluorocyclopropyl, 2, 3-difluorocyclopropyl, 2,3, 3-tetrafluorocyclopropyl, 2, 3-difluorocyclobutyl, 2, 4-difluorocyclobutyl, and the like.
The term "heterocycloalkyl" as used herein is intended to include one or more heteroatoms independently selected from O, N and S and a monocyclic, fused polycyclic, spiro, or bridged polycyclic non-aromatic saturated ring structure of the indicated number of ring atoms, or an N-oxide thereof, or an S-oxide or S-dioxide thereof. The heterocycloalkyl group can have 4 to 12 ring members, specifically 4 to 10 ring members, and more specifically 4 to 7 ring members. Heterocycloalkyl groups typically contain up to 4 heteroatoms, more typically up to 3 heteroatoms, more typically up to 2, e.g. a single heteroatom, e.g. a 4-7 membered monocyclic heterocycloalkyl having one heteroatom such as N, i.e. a 4-7 membered nitrogen containing heterocycloalkyl. Examples of suitable heterocycloalkyl groups include, but are not limited to, azetidinyl, oxetanyl, thietanyl, pyrrolidinyl (e.g., 1-pyrrolidinyl, 2-pyrrolidinyl, and 3-pyrrolidinyl), tetrahydrofuranyl (e.g., 1-tetrahydrofuranyl, 2-tetrahydrofuranyl, and 3-tetrahydrofuranyl), tetrahydrothienyl (e.g., 1-tetrahydrothienyl, 2-tetrahydrothienyl, and 3-tetrahydrothienyl), piperidinyl (e.g., 1-piperidinyl, 2-piperidinyl, 3-piperidinyl, and 4-piperidinyl), tetrahydropyranyl (e.g., 4-tetrahydropyranyl), tetrahydrothiopyranyl (e.g., 4-tetrahydrothiopyranyl), morpholinyl, thiomorpholinyl, dioxanyl, piperazinyl, or azepanyl.
It will be understood by those of ordinary skill in the art of organic synthesis that stable, chemically feasible heterocycles, whether aromatic or non-aromatic, have a maximum number of heteroatoms or type of heteroatoms included that is determined by the ring size, degree of unsaturation, and the valency of the heteroatom. In general, a heterocycle may have from 1 to 4 heteroatoms, as long as the heterocycle or heteroaromatic ring is chemically feasible and stable.
The term "halo" or "halogen" as used herein means fluorine (F), chlorine (Cl), bromine (Br) and iodine (I). Particular halo groups are fluoro or chloro. The term "halogen-substituted" group as used herein is intended to include monohalogenated or polyhalogenated groups in which one or more of the same or different halogens substitute for one or more hydrogens in the group.
The term "cyano" as used herein means the group-CN.
The term "nitro" as used herein means the group-NO2
The term "amino" as used herein means the group-NH2
The term "optionally substituted with … …" as used herein means that a group may be unsubstituted or substituted with one or more substituents (e.g., 0, 1,2, 3, 4, or 5 or more, or any range derivable therein) as set forth for that group, wherein the substituents may be the same or different. In one embodiment, the optionally substituted group has 1 substituent. In another embodiment, the optionally substituted group has 2 substituents. In another embodiment, the optionally substituted group has 3 substituents. In another embodiment, the optionally substituted group has 4 substituents.
Unless otherwise defined, C as used in the definition of compounds herein1-C6Alkyl radical, C3-C7Cycloalkyl or 4-7 membered heterocycloalkyl optionally carrying one or more substituents which may be selected from H, F, Cl, Br, I, cyano, nitro, C1-C6Alkyl (e.g., methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, pentyl, neopentyl, hexyl, 1, 2-dimethylbutyl, etc.), C3-C7Cycloalkyl (cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl), -OH, -O-C1-C6Alkyl (e.g., methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, tert-butoxy, sec-butoxy, n-pentoxy, n-hexoxy, 1, 2-dimethylbutoxy, etc.), -SH, -S-C1-C6Alkyl (e.g., methylthio, ethylthio, n-propylthio, isopropylthio, n-butylthio, t-butylthio, s-butylthio, n-pentylthio, n-hexylthio, 1, 2-dimethylbutylthio, etc.) or-NH2、-NH-C1-C6Alkyl (e.g., methylamino, ethylamino, propylamino, isopropylamino, N-butylamino, tert-butylamino, sec-butylamino, N-pentylamino, N-hexylamino, and 1, 2-dimethylbutylamino), N (C)1-C6Alkyl radical)2(e.g., dimethylamino, methylethylamino, diethylamino, etc.), wherein C1-C6Alkyl or C3-C7Cycloalkyl is optionally substituted with one or more halogens (preferably F).
The term "compound of the invention" as used herein is intended to encompass a compound of general formula (I) as defined herein or any preferred or specific embodiment thereof, stereoisomers, tautomers, stable isotopic variations, pharmaceutically acceptable salts or solvates thereof, and prodrugs. Similarly, references herein to "intermediates", whether or not they are themselves claimed, are intended to cover the free forms thereof as well as the various derivatives described above, if the context permits.
The term "pharmaceutically acceptable" as used herein means molecular entities and compositions that are approved by or can be approved by the corresponding authorities in the various countries or listed in the generally recognized pharmacopoeias for animals, and more particularly humans, or that do not produce adverse, allergic, or other untoward reactions when administered in moderate amounts to animals, such as humans.
The term "pharmaceutically acceptable salt" as used herein means a salt of a compound of the present invention which is pharmaceutically acceptable and which possesses the desired pharmacological activity of the parent compound. In particular, such salts are non-toxic and may be inorganic acid addition salts or organic acid addition salts and base addition salts. Specifically, such salts include: (1) acid addition salts with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or an acid addition salt formed with an organic acid such as acetic acid, propionic acid, hexanoic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, benzenesulfonic acid, 2-naphthalenesulfonic acid, 4-toluenesulfonic acid, camphorsulfonic acid, glucoheptonic acid, 3-phenylpropionic acid, trimethylacetic acid, tert-butylacetic acid, laurylsulfuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, muconic acid, etc.; or (2) a salt formed when an acidic proton present in the parent compound is replaced with a metal ion such as an alkali metal ion, an alkaline earth metal ion, or an aluminum ion, or coordinated with an organic base such as ethanolamine, diethanolamine, triethanolamine, N-methylglucamine, or the like. The general principles and techniques for preparing pharmaceutically acceptable salts are known to those skilled in the art, for example, those described in Berge et al, Pharm ScL, 66, 1-19 (1977).
The term "prodrug" as used herein means a compound of the present invention having a cleavable group and becoming pharmaceutically active in vivo by solvolysis or under physiological conditions, including derivatives of the compounds of the present invention. Prodrugs include acid derivatives well known in the art, such as esters prepared by reacting the parent acid with a suitable alcohol, or substituted or unsubstituted aminesAmide, or anhydride or mixed anhydride. Simple aliphatic or aromatic esters, amides and anhydrides derived from the pendant acid groups of the compounds of the present invention are particularly useful prodrugs. Particular such prodrugs are C of the compounds of the present invention1-8Alkyl radical, C2-8Alkenyl, optionally substituted C6-10Aryl and (C)6-10Aryl group) - (C1-4Alkyl) esters.
The term "stereoisomer" as used herein denotes an isomer formed as a result of at least one asymmetric center. In compounds having one or more (e.g., 1,2, 3, or 4) asymmetric centers, they can result in racemic mixtures, single enantiomers, diastereomeric mixtures and individual diastereomers. The particular molecule may also exist as geometric isomers (cis/trans). Similarly, the compounds of the invention may exist as a mixture of two or more different structural forms in rapid equilibrium (commonly referred to as tautomers). Representative examples of tautomers include keto-enol tautomers, phenol-keto tautomers, nitroso-oxime tautomers, imine-enamine tautomers, and the like. For example, a nitroso-oxime may exist in solution in equilibrium with the following tautomeric forms:
Figure BDA0003065598540000071
it is to be understood that the scope of this application encompasses all such isomers or mixtures thereof in any ratio (e.g., 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%).
The compounds of the invention may have one or more asymmetric centers and may thus be prepared as (R) -or (S) -stereoisomers, respectively, or as mixtures thereof. Structural formula or structural fragment of compound used in the invention
Figure BDA0003065598540000072
Or
Figure BDA0003065598540000073
Indicating the relative configuration of asymmetric centers, i.e., chiral centers. Accordingly, the relative configuration with respect to the chiral center is denoted by R and S in the nomenclature of the compounds or intermediates provided herein.
The term "solvate" as used herein refers to a form of solvent addition comprising a stoichiometric or non-stoichiometric amount of solvent, including for example solvates with water, such as hydrates, or with organic solvents, such as methanol, ethanol or acetonitrile, i.e. as methanolate, ethanolate or acetonitrilate, respectively; or in the form of any polymorph. It will be understood that such solvates of the compounds of the invention also include solvates of pharmaceutically acceptable salts of the compounds of the invention.
The term "isotopic variation" as used herein refers to a compound in which one or more atoms constituting the compound are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes that can be incorporated into one or more atoms of a compound of the invention include, for example2H、3H、13C、14C、15N、17O、18O、31P、32P、35S、18F and36cl, thereby forming isotopically-varied forms of the compounds of the present invention, whether radioactive or not, are intended to be encompassed within the scope of the present invention. In some embodiments, the incorporated isotope is 2H (deuterium); in other embodiments, the incorporated isotope is 3H (tritium).
The term "disease associated with ROR γ t" as used herein means a disease in which ROR γ t contributes to the onset and progression of the disease, or inhibition of ROR γ t reduces the incidence of disease, reduces or eliminates disease symptoms. For the purposes of the present invention, a "disease associated with ROR γ t" is chosen in particular from inflammatory or autoimmune diseases, cancer and the like, including but not limited to psoriasis, rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, multiple sclerosis, systemic lupus erythematosus, graft-versus-host disease, inflammatory bowel disease, crohn's disease, ulcerative colitis, chronic obstructive pulmonary disease, asthma, glomerulonephritis, lupus nephritis, myocarditis, thyroiditis, dry eye, uveitis, behcet's disease, allergic dermatitis, acne, scleroderma, bronchitis, cutaneous allergic rhinitis, Necrotizing Enterocolitis (NEC), hepatic fibrosis, nonalcoholic steatohepatitis (NASH), New coronaviral pneumonia (New coronavirus pneumanias), insulin-dependent type I diabetes mellitus, triple negative breast cancer, and prostate cancer, among others. Preferred indications of the invention are selected from psoriasis, rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, multiple sclerosis, inflammatory bowel disease, dry eye, allergic dermatitis, Chronic Obstructive Pulmonary Disease (COPD), asthma, necrotizing enterocolitis, liver fibrosis, non-alcoholic steatohepatitis, neocoronavirus pneumonia, triple negative breast cancer and prostate cancer.
The term "subject" or "individual" as used herein includes a human or non-human animal. Exemplary human individuals include human individuals (referred to as patients) having a disease (e.g., a disease described herein) or normal individuals. "non-human animals" in the context of the present invention include all vertebrates, such as non-mammals (e.g., birds, amphibians, reptiles) and mammals, such as non-human primates, livestock and/or domesticated animals (e.g., sheep, dogs, cats, cows, pigs, etc.).
The term "therapeutically effective amount" as used herein means an amount sufficient to alleviate or completely alleviate the symptoms or other deleterious effects of a disorder when administered to a subject to treat a disease; reversing, completely stopping, or slowing the progression of the disorder; or an amount that reduces the risk of worsening the condition, "effective amount" may vary depending on the compound, the disease and its severity, and the age, weight, etc. of the subject to be treated.
The term "prevention" as used herein means the administration of one or more compounds of the invention to a subject, e.g. a mammal, e.g. a human, suspected of suffering from or susceptible to a disease associated with roryt as defined herein, in particular an inflammatory or autoimmune disease, such that the risk of suffering from the defined disease is reduced. The term "prevention" encompasses the use of a compound of the invention prior to diagnosis or determination of any clinical and/or pathological condition.
The term "treating" as used herein refers to administering one or more compounds of the invention described herein to a subject, e.g., a mammal, e.g., a human, suffering from, or having symptoms of, the disease, for the purpose of curing, alleviating, or otherwise affecting the disease or symptoms of the disease. In a particular embodiment of the invention, the disease is a disease associated with ROR γ t as defined herein, in particular an inflammatory or autoimmune disease.
The term "pharmaceutical combination" as used herein means that the compounds of the present invention can be used in combination with other active agents for the purpose of the present invention. The additional active agent may be one or more additional compounds of the invention, or may be a second or additional (e.g., third) compound that is compatible with, i.e., does not adversely affect, each other, or has complementary activity. Such active agents are suitably present in combination in an effective amount to achieve the intended purpose. The additional active agents may be co-administered with the compounds of the present invention in a single pharmaceutical composition, or separately administered in separate discrete units from the compounds of the present invention, either simultaneously or sequentially when administered separately. The sequential administration may be close in time or remote in time.
The term "pharmaceutically acceptable excipient or carrier" as used herein refers to one or more compatible solid or liquid filler or gel substances which are pharmacologically inactive, compatible with the other ingredients of the composition, and which are intended to be acceptable for administration to a warm-blooded animal such as man, and which serve as a carrier or vehicle for the compounds of the present invention in the form in which they are to be administered, examples of which include, but are not limited to, cellulose and its derivatives (e.g., sodium carboxymethylcellulose, cellulose acetate, and the like), gelatin, talc, solid lubricants (e.g., magnesium stearate), calcium sulfate, vegetable oils, polyols (e.g., propylene glycol, glycerol, mannitol, sorbitol, and the like), emulsifiers (e.g., tweens), wetting agents (e.g., sodium lauryl sulfate), colorants, flavors, stabilizers, antioxidants, preservatives, and the like.
Unless otherwise specified, C in the definition of the compounds of the inventionn-n+mOr Cn-Cn+mIncluding n to n + m carbon atoms, e.g. C1-6Comprising C1、C2、C3、C4、C5And C6Also included are any ranges of n to n + m, e.g. C1-6Comprising C1-2、C1-3、C1-4、C2-6、C3-6And the like. Similarly, n-to n + m-membered in the definition of the compounds of the present invention means that the number of ring atoms is n to n + m, for example, 3-to 12-membered rings include 3-membered rings, 4-membered rings, 5-membered rings, 6-membered rings, 12-membered rings and the like, and any range of n to n + m-membered rings is also included, for example, 3-to 12-membered rings include 3-to 6-membered rings, 3-to 9-membered rings, 5-to 6-membered rings, 5-to 7-membered rings, 6-to 8-membered rings, 6-to 10-membered rings and the like.
It is to be understood that the dosages referred to when describing the compounds of the present invention, pharmaceutical compositions, pharmaceutical combinations comprising the same, and related uses and methods, are by weight of the free form, and not by any salt, hydrate, solvate or the like thereof, unless otherwise defined in the specification.
Compounds of the invention
The terms "inventive compound" and the like, as used throughout this application, unless otherwise indicated, encompass compounds of formula (I), as defined in the various embodiments herein and in the specific or preferred embodiments thereof, stereoisomers, tautomers, stable isotopic variations, pharmaceutically acceptable salts or solvates thereof, and prodrugs. The stereoisomers, tautomers, stable isotopic variations, pharmaceutically acceptable salts or solvates, and prodrugs are described in the definitions section above. Preferably, the compounds of the invention are free forms of the compounds of formula (I) or a pharmaceutically acceptable salt or solvate thereof; most preferred is the free form of the compound of formula (I) or a pharmaceutically acceptable salt thereof.
Certain compounds of the present invention may exist in polymorphic or amorphous forms, which also fall within the scope of the present invention. When in a solid crystalline form, the compound of formula (I) may be in the form of a co-crystal with another chemical entity, and the specification includes all such co-crystals.
Where chiral centers are present, the compounds of the present invention may exist as individual enantiomers or as mixtures of enantiomers. According to one embodiment, there is provided a compound of formula (I) or a pharmaceutically acceptable salt thereof, which is a single enantiomer with enantiomeric excess (% ee) > 95, > 98% or > 99%. Preferably, the single enantiomer is present in an enantiomeric excess (% ee) of > 99%.
In particular, in one aspect, the present invention provides compounds of formula (I), stereoisomers, tautomers, stable isotopic variations, pharmaceutically acceptable salts, or solvates thereof:
Figure BDA0003065598540000091
wherein:
x is selected from CH or N;
R1selected from hydrogen, halogen, cyano, nitro, C1-C6Alkyl, -O-C1-C6Alkyl, S-C1-C6Alkyl, -NH-C1-C6Alkyl, -N- (C)1-C6Alkyl radical)2、C1-C6alkyl-O-C1-C6Alkyl, -C1-C6alkyl-S-C1-C6Alkyl radical, C1-C6alkyl-NH-C1-C6Alkyl or C1-C6alkyl-N (C)1-C6Alkyl radical)2Wherein said C is1-C6Alkyl is optionally substituted by halogen or cyano;
R2selected from hydrogen, halogen, cyano, nitro or C1-C6Alkyl radical, wherein said C1-C6Alkyl is optionally substituted with substituents independently selected from: -Raor-OH, wherein RaIs C optionally substituted by halogen1-C6An alkyl group;
R3、R4each independently selected from halogen, cyano, nitro, Rb、-ORb、-SRbor-NRbRbWherein R isbIs H or optionally substituted C1-C6Alkyl, each of said substituents being independently selected from hydrogen, halogen, cyano, nitro, -OH, -SH, -NH2、-O-C1-C6Alkyl, -S-C1-C6Alkyl, -NH-C1-C6Alkyl or-N (-C)1-C6Alkyl radical)2
R5Is selected from-ORc、-SRc、-NRcRc、-NHCORc、-NHSO2Rc、-COORc、-CONRcRcor-SO2NRcRcWherein R iscIndependently at each occurrence, is selected from hydrogen, C optionally substituted with halogen1-C6Alkyl or C optionally substituted by halogen3-C7Cycloalkyl, or two R bound to the same N atomcMay form, together with the N atom to which they are attached, a 4-7 membered nitrogen containing heterocycloalkyl group;
R6is selected from C1-C6Alkyl radical, C3-C7Cycloalkyl, 4-7 membered heterocycloalkyl or optionally substituted by C1-C6Alkyl radical, C3-C7Cycloalkyl or 4-7 membered heterocycloalkyl substituted-NH2Wherein said C is1-C6Alkyl radical, C3-C7Cycloalkyl or 4-7 membered heterocycloalkyl is optionally substituted with substituents each independently selected from: hydrogen, halogen, cyano, nitro, -Ra、-ORa、-SRaor-NRaRaOr C optionally substituted by halogen3-C7Cycloalkyl radical, RaIndependently at each occurrence, is selected from H or C optionally substituted with halogen1-C6Alkyl, wherein two groups attached to the N atom of the amino group may form a 4-7 membered ring together with the N to which they are attached; and is
n is selected from 0, 1 or 2.
In one embodiment of the compounds of formula (I), X is CH.
In one embodiment of the compounds of formula (I), R1Is H.
In one embodiment of the compounds of formula (I), R1Is halogen, such as F, Cl, Br or I; f or Cl is preferred.
In one embodiment of the compounds of formula (I), R1Is C1-C6Alkyl, preferably C1-C3Alkyl, optionally substituted by halogen (preferably F), e.g. methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, -CF3、-CHF2、-CH2CF3、-CF2CF3、-CH2CH2CF3、-CH(CF3)2And the like.
In one embodiment of the compounds of formula (I), R1Is selected from-O-C1-C6Alkyl, -S-C1-C6Alkyl, -NH-C1-C6Alkyl or-N- (C)1-C6Alkyl radical)2Wherein C is1-C6Alkyl is optionally substituted with one or more halogen (preferably F), examples include, but are not limited to-OCH3、-OCF3、-OCHF2、-OCH2CH3、-OCH2CF3、-OCH2CH2CH3、-OCH2CH2CF3、-OCH(CF3)2、-SCH3、-SCF3、-SCH2CH3、-NH2、-NHCH3、-N(CH3)2、-NH-CH2CH3、-N(CH3)(CH2CH3)、-NHCF3、-N(CH3)(CF3) And the like. In a more specific embodiment, R1Is selected from-O-C1-C6Alkyl radical, wherein C1-C6Alkyl radicals substituted by at least three halogens, preferably F, e.g. -OCF3、-OCH2CF3、-OCH2CH2CF3、-CH(CF3)2And the like.
In one embodiment of the compounds of formula (I), R1Is selected from-C1-C6alkyl-O-C1-C6Alkyl, -C1-C6alkyl-S-C1-C6Alkyl, -C1-C6alkyl-NH-C1-C6Alkyl or-C1-C6alkyl-N (C)1-C6Alkyl radical)2Wherein C is1-C6Alkyl is optionally substituted with halogen (preferably F). Specific examples include, but are not limited to, -CH2OCH3、-CH2CH2OCH3、-CH2OCH2CH3、-CH2CH2OCH2CH3、-CH2NHCH3、-CH2N(CH3)2、-CH2OCF3、-CH2CH2OCF3、-CH2OCH2CF3、-CH2NHCF3、-CH2N(CF3)2And the like.
In one embodiment of the compounds of formula (I), R2Is hydrogen.
In one embodiment of the compounds of formula (I), R2Is halogen, such as F, Cl, Br or I; f or Cl is preferred.
In one embodiment of the compounds of formula (I), R2Is C1-C6Alkyl, preferably C1-C3Alkyl, e.g. para to the phenyl radical, wherein said C1-C3C with alkyl substituted by two halogen radicals, preferably F1-C3Alkyl and one-OH substitution, e.g.
Figure BDA0003065598540000101
In a specific embodiment, R1Is H and R2Selected from halogen or
Figure BDA0003065598540000111
In one embodiment of the compounds of formula (I), R3、R4Each independently selected from H, halogen or C optionally substituted by halogen1-C6An alkyl group.
In a compound of formula (I)In the embodiment (1), R3、R4Each independently selected from H or halogen, preferably fluorine or chlorine, most preferably Cl, e.g. R3Is H and R4Is halogen, preferably F or Cl, or vice versa. In a specific embodiment, R3And R4And at the same time halogen, preferably fluorine or chlorine, most preferably at the same time Cl.
In one embodiment of the compounds of formula (I), R3、R4Each independently selected from H or C1-C6Alkyl, optionally substituted with halogen. In a specific embodiment, R3Is H and R4Is C optionally substituted by halogen, preferably F1-C6Alkyl, or vice versa, e.g. R3Is H and R4Is methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, -CF3、-CHF2、-CH2CF3、-CF2CF3、-CH2CH2CF3or-CH (CF)3)2Etc., or vice versa. In a specific embodiment, R3And R4Is selected from C1-C6Alkyl, optionally substituted by halogen (preferably F), e.g. R3And R4While being methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, -CF3、-CHF2、-CH2CF3、-CF2CF3、-CH2CH2CF3or-CH (CF)3)2Etc. or R3And R4Each independently is one of the exemplary groups described above.
In one embodiment of the compounds of formula (I), R3、R4Each independently selected from-ORb、-SRbor-NRbRbWherein R isbIs H or C optionally substituted by halogen1-C6Alkyl radicals, e.g. OH, -SH, -OCH3、-OCF3、-OCHF2、-OCH2CH3、-OCH2CF3、-OCH2CH2CH3、-OCH2CH2CF3、-OCH(CF3)2、-SCH3、-SCF3、-SCH2CH3、-NH2、-NHCH3、-N(CH3)2、-NH-CH2CH3、-N(CH3)(CH2CH3)、-NHCF3、-N(CH3)(CF3) And the like.
In one embodiment of the compounds of formula (I), R5Is selected from-ORcWherein R iscSelected from hydrogen, C optionally substituted by halogen1-C6Alkyl or C optionally substituted by halogen3-C7A cycloalkyl group. In a specific embodiment, R5is-OH. In a specific embodiment, R5is-O-C1-C6Alkyl, optionally substituted by halogen, e.g. -OCH3、-OCH2CH3、-OCH2CH2CH3、-OCH(CH3)、-OCF3、-OCHF2、-OCH2CF3、-OCH2CH2CF3、-OCH(CF3) And the like. In a specific embodiment, R5is-O-C3-C7Cycloalkyl, optionally substituted with halo, such as-O-cyclopropyl, -O-cyclobutyl, -O-cyclopentyl, and the like. In a more specific embodiment, R5is-O-C1-C3Alkyl radicals, e.g. -OCH3、-OCH2CH3、-OCH2CH2CH3、-OCH(CH3) And the like.
In one embodiment of the compounds of formula (I), R5Is selected from-NRcRc、-NHCORcor-NHSO2Rcpreferably-NRcRcor-NHCORcWherein R iscIndependently at each occurrence, is selected from hydrogen, C optionally substituted with halogen1-C6Alkyl or C optionally substituted by halogen3-C7Cycloalkyl, or two R bound to the same N atomcMay form, together with the N atom to which they are attached, a 4-to 7-membered nitrogen-containing heterocycloalkyl group.
In a specific embodiment, R5Is selected from-NRcRcor-NHCORcWherein R iscIndependently at each occurrence, is selected from hydrogen or C optionally substituted with halogen (preferably F)1-C6Alkyl, specific examples include, but are not limited to-NH2、-NHCH3、-N(CH3)2、-NH-CH2CH3、-N(CH3)(CH2CH3)、-N(CH2CH3)(CH2CH3)-、-NH(CH2CH2CH3)、-NHCF3、-N(CH3)(CF3)、-N(CF3)CF3、-N(CH2CF3)CF3or-N (CH)2CF3)(CH2CF3)-、-NHCOCH3、-NHCO-CH2CH3、-NHCO-CH2CF3. In a specific embodiment, R5Is selected from-NRcRcIn which two R are bound to the same N atomcMay form, together with the N atom to which they are attached, a 4-7 membered nitrogen containing heterocycloalkyl group, such as-azetidine, aziridine, piperidinyl and the like. In a more specific embodiment, R5Is selected from-NH2、-NHCH3、-N(CH3)2、-NH-CH2CH3、-NH(CH2CH2CH3)、-N(CH3)(CH2CH3)、-NHCOCH3、-NHCO-CH2CH3-azetidine, aziridine or piperidinyl.
In a specific embodiment, R5Is selected from-NRcRcor-NHCORcWherein R iscIndependently at each occurrence, is selected from hydrogen or C optionally substituted with halogen3-C7Cycloalkyl groups, such as-NH-cyclopropyl, -NH-cyclobutyl, -NH-cyclopentyl, -NHCO-cyclopropyl, -NHCO-cyclobutyl, -NHCO-cyclopentyl, and the like.
In one embodiment of the compounds of formula (I), R5Is selected from-COORc、-CONRcRcor-SO2NRcRcpreferably-COORcor-CONRcRcWherein R iscIndependently at each occurrence, is selected from hydrogen, C optionally substituted with halogen1-C6Alkyl or C optionally substituted by halogen3-C7Cycloalkyl, or two R bound to the same N atomcMay form, together with the N atom to which they are attached, a 4-to 7-membered nitrogen-containing heterocycloalkyl group.
In a specific embodiment, R5Is selected from-COORcor-CONRcRcWherein R iscIndependently at each occurrence, is selected from H or C optionally substituted with halogen (preferably F)1-C6Alkyl radicals, e.g. COOH, -COOCH3、-COOCF3、-COOCH2CH3、-COOCH2CF3、-CONH2、-CONHCH3、-CON(CH3)2、-CON(CH3)(CH2CH3)、-CONHCF3、-CON(CF3)2. In a specific embodiment, R5Is selected from-CONRcRcIn which two R are bound to the same N atomcMay form, together with the N atom to which they are attached, a 4-7 membered nitrogen containing heterocycloalkyl group such as-CO-azetidine, -CO-aziridine, -CO-piperidinyl and the like.
In one embodiment of the compounds of formula (I), R6Is selected from C1-C6Alkyl, preferably C1-C3Alkyl optionally substituted with one or more groups independently selected from: H. halogen, C3-C7Cycloalkyl radical, Ra、-ORa、-SRaor-NRaRaWherein R isaIndependently selected from H or C optionally substituted with one or more halogens1-C6Alkyl, or two R bound to the same N atomaMay form, together with the N atom to which they are attached, a 4-to 7-membered nitrogen-containing heterocycloalkyl group.
In a specific embodiment, R6Is C1-C6Alkyl, preferably C1-C3Alkyl, optionally substituted by F, Cl, Br, I, RaOR-ORaIs substituted in whichRaIs H or C optionally substituted by one or more halogen (preferably F)1-C3An alkyl group. Specific examples include, but are not limited to, methyl, ethyl, propyl or isopropyl, trifluoromethyl, trifluoroethyl, hydroxymethyl, hydroxyethyl, methoxymethyl, methoxyethyl, trifluoromethoxy methyl or trifluoromethoxyethyl, and the like. In more specific embodiments, R6Is C1-C3Alkyl groups such as methyl, ethyl, propyl or isopropyl.
In a specific embodiment, R6Is C1-C6Alkyl, preferably C1-C3Alkyl, C optionally substituted by halogen3-C7Cycloalkyl is substituted. Specific examples include, but are not limited to, methyl, ethyl, propyl or isopropyl, cyclopropylmethyl, cyclopropylethyl, cyclobutylmethyl, cyclobutylethyl, cyclopentylmethyl, cyclopentylethyl and the like.
In specific embodiments, R6Is C1-C6Alkyl, preferably C1-C3Alkyl, optionally substituted by-NRaRaIs substituted in which RaIndependently selected from H or C optionally substituted with one or more halogens1-C3Alkyl, or two R bound to the same N atomaMay form, together with the N atom to which they are attached, a 4-to 7-membered nitrogen-containing heterocycloalkyl group. Specific examples include, but are not limited to, methyl, ethyl, propyl or isopropyl, aminomethyl, aminoethyl, aminopropyl, methylaminomethyl, dimethylaminomethyl, methylethylaminomethyl, azetidinylmethyl, pyrrolidinylmethyl, piperidinylmethyl and the like.
In one embodiment of the compounds of formula (I), R6is-C3-C7Cycloalkyl optionally substituted with one or more groups independently selected from: H. halogen, C optionally substituted by halogen3-C7Cycloalkyl radical, Ra、-ORa、-SRaor-NRaRaWherein R isaIndependently selected from H or C optionally substituted with one or more halogens1-C6Alkyl radicalOr two R's bound to the same N atomaMay form, together with the N atom to which they are attached, a 4-to 7-membered nitrogen-containing heterocycloalkyl group. In specific embodiments, R6is-C3-C5Cycloalkyl groups, such as cyclopropyl, cyclobutyl or cyclopentyl.
In a specific embodiment, R6is-C3-C7Cycloalkyl optionally substituted with one or more groups independently selected from: halogen, Ra、-ORaor-NRaRaWherein R isaIndependently selected from H or C optionally substituted with one or more halogens1-C6An alkyl group. Specific examples include, but are not limited to, 2, 3-difluorocyclopropyl, 2,3, 3-tetrafluorocyclopropyl, 2, 3-difluorocyclobutyl, methylcyclopropyl or cyclobutyl, dimethylcyclopropyl or cyclobutyl, trifluoromethylcyclopropyl or cyclobutyl, hydroxycyclopropyl or cyclobutyl, trifluoromethoxycyclopropyl or cyclobutyl, methylaminocyclopropyl, dimethylaminocyclopropyl, trifluoromethylaminocyclopropyl, and the like.
In one embodiment of the compounds of formula (I), R6Is a 4-7 membered heterocycloalkyl group, optionally substituted with one or more groups independently selected from: H. halogen, Ra、-ORa、-SRaor-NRaRaWherein R isaIndependently selected from H or C optionally substituted with one or more halogens1-C6Alkyl, or two R bound to the same N atomaMay form, together with the N atom to which they are attached, a 4-to 7-membered nitrogen-containing heterocycloalkyl group. In specific embodiments, R6Is a 4-to 7-membered heterocycloalkyl group such as azetidinyl, oxetanyl, thietanyl, pyrrolidinyl (e.g., 1-pyrrolidinyl, 2-pyrrolidinyl and 3-pyrrolidinyl), tetrahydrofuranyl (e.g., 1-tetrahydrofuranyl, 2-tetrahydrofuranyl and 3-tetrahydrofuranyl), tetrahydrothienyl (e.g., 1-tetrahydrothienyl, 2-tetrahydrothienyl and 3-tetrahydrothienyl), piperidinyl (e.g., 1-piperidinyl, 2-piperidinyl, 3-piperidinyl and 4-piperidinyl), tetrahydropyranyl (e.g., 4-tetrahydropyranyl), tetrahydrothiopyranyl (e.g., 4-tetrahydrothiopyranyl), morpholinyl, oxetanyl, thiobutanyl, oxetanyl, and the like,Thiomorpholinyl, dioxanyl, piperazinyl or azepanyl, each of which is optionally substituted by one or more, e.g. 1,2 or 3, independently selected from F, Cl, Br, I, Ra、-ORaor-NRaRaWherein R isaIndependently selected from H or C optionally substituted with one or more halogens1-C3An alkyl group.
In one embodiment of the compounds of formula (I), R6Is optionally substituted by C1-C6Alkyl radical, C3-C7Cycloalkyl or 4-7 membered heterocycloalkyl substituted amino. In a particular embodiment, C substituted on the amino group1-C6Alkyl radical, C3-C7Cycloalkyl or 4-7 membered heterocycloalkyl each have the above embodiment pairs as R6C of (A)1-C6Alkyl radical, C3-C7Each embodiment or specific example as defined for cycloalkyl or 4-7 membered heterocycloalkyl. In a most specific embodiment, R6Is C optionally substituted by halogen1-C6Alkyl-substituted amino, specific examples include, but are not limited to, -NH2、-NHCH3、-NCH3CH3、-N(CH2CH3)CH3、-N(CH2CH3)(CH2CH3)-、-NHCF3、-N(CH3)CF3、-N(CF3)CF3、-N(CH2CF3)CF3or-N (CH)2CF3)(CH2CF3)-、-NH(CH2CH2CH3) And the like.
It is to be understood that the compounds of formula (I) of the present invention encompass each of the above independent embodiments or each specific embodiment, and also encompass embodiments comprised of any combination or subcombination of the above-described embodiments or specific embodiments, as well as embodiments comprised of any combination of any of the above preferences or exemplifications.
Preferably, the present invention provides a compound of formula (I), a stereoisomer, a tautomer, a stable isotopic variation, a pharmaceutically acceptable salt, or a solvate thereof:
Figure BDA0003065598540000141
wherein:
x is selected from CH;
R1selected from hydrogen, -O-C1-C6Alkyl, S-C1-C6Alkyl, -NH-C1-C6Alkyl or-N- (C)1-C6Alkyl radical)2Wherein said C is1-C6Alkyl optionally substituted with halo;
R2selected from hydrogen, halogen or C1-C6Alkyl radical, wherein said C1-C6The alkyl group is substituted with a substituent independently selected from the group consisting of: -Raor-OH, wherein RaIs C substituted by halogen1-C6An alkyl group;
R3、R4each independently selected from H, halogen, cyano or C optionally substituted by halogen1-C6An alkyl group;
R5is selected from-ORc、-SRc、-NRcRc、-NHCORc、-NHSO2Rc、-COORc、-CONRcRcor-SO2NRcRcWherein R iscIndependently at each occurrence, is selected from hydrogen or C optionally substituted with halogen1-C6Alkyl, or two R bound to the same N atomcMay form, together with the N atom to which they are attached, a 4-7 membered nitrogen containing heterocycloalkyl group;
R6is selected from C1-C6Alkyl or-NRaRaWherein said C is1-C6Alkyl is optionally substituted with substituents each independently selected from: halogen, -Raor-NRaRa,RaIndependently at each occurrence, selected from H, C optionally substituted with halogen1-C6Alkyl or C optionally substituted by halogen3-C7Cycloalkyl in which two are bound to the N atom of the amino groupR isaMay form a 4-7 membered ring together with the N to which they are attached; and is
n is selected from 0, 1 or 2.
In one embodiment of one of the preferred compounds of formula (I) above, R1Selected from hydrogen or-O-C1-C6Alkyl radical, wherein said C1-C6Alkyl is optionally substituted with halogen. In a specific embodiment, R1Selected from hydrogen or-O-C1-C6Alkyl radical, wherein said C1-C6Alkyl is substituted with one or more, e.g. one, two or three, halogens (preferably F). In a more specific embodiment, R1is-O-C1-C6Alkyl radical, wherein said C1-C6Alkyl radicals substituted by at least three halogens, preferably F, e.g. -O-CF3、-O-CH2CF3、-O-CH2CH2CF3、-O-CF2CF3And the like.
In one embodiment of the above preferred compounds of formula (I), R2Is hydrogen.
In one embodiment of the above preferred compounds of formula (I), R2Is halogen, such as F, Cl, Br or I; f or Cl is preferred.
In one embodiment of the above preferred compounds of formula (I), R2Is C1-C6Alkyl, preferably C1-C3Alkyl, C substituted by two halogens (preferably F)1-C3Alkyl and one-OH substitution, e.g.
Figure BDA0003065598540000142
In one embodiment of the above preferred compounds of formula (I), R1Is H and R2Selected from halogen or
Figure BDA0003065598540000151
In one embodiment of the above preferred compounds of formula (I), R3、R4Each independently selected from H or halogen, preferably F or Cl, most preferably Cl, e.g. R3Is H and R4Is halogen, preferably F or Cl, or vice versa. In a specific embodiment, R3And R4And at the same time halogen, preferably fluorine or chlorine, most preferably at the same time Cl.
In one embodiment of the above preferred compounds of formula (I), R3、R4Each independently selected from H or C1-C6Alkyl, optionally substituted with halogen. In a specific embodiment, R3Is H and R4Is C optionally substituted by halogen, preferably F1-C6Alkyl, or vice versa, e.g. R3Is H and R4Is methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, -CF3、-CHF2、-CH2CF3、-CF2CF3、-CH2CH2CF3or-CH (CF)3)2Etc., or vice versa. In a specific embodiment, R3And R4Is selected from C1-C6Alkyl radicals, e.g. R3And R4While being methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, -CF3、-CHF2、-CH2CF3、-CF2CF3、-CH2CH2CF3or-CH (CF)3)2Etc. or R3And R4Each independently is one of the exemplary groups described above.
In one embodiment of the above preferred compounds of formula (I), R5Is selected from-ORcWherein R iscSelected from hydrogen or C optionally substituted by halogen1-C6An alkyl group. In a specific embodiment, R5is-OH. In a specific embodiment, R5is-O-C1-C6Alkyl, optionally substituted by halogen, e.g. -OCH3、-OCH2CH3、-OCH2CH2CH3、-OCH(CH3)、-OCF3、-OCH2CF3、-OCH2CH2CF3、-OCH(CF3) And the like. In a more specific embodiment, R5is-O-C1-C3Alkyl radicals, e.g. -OCH3、-OCH2CH3、-OCH2CH2CH3、-OCH(CH3) And the like.
In one embodiment of the above preferred compounds of formula (I), R5Is selected from-NRcRcor-NHCORcWherein R iscIndependently at each occurrence, is selected from hydrogen or C optionally substituted with halogen (preferably F)1-C6Alkyl, specific examples include, but are not limited to-NH2、-NHCH3、-N(CH3)2、-NH-CH2CH3、-N(CH3)(CH2CH3)、-N(CH2CH3)(CH2CH3)-、-NH(CH2CH2CH3)、-NHCF3、-N(CH3)(CF3)、-N(CF3)CF3、-N(CH2CF3)CF3or-N (CH)2CF3)(CH2CF3)-、-NHCOCH3、-NHCO-CH2CH3、-NHCO-CH2CF3. In a specific embodiment, R5Is selected from-NRcRcIn which two R are bound to the same N atomcMay form, together with the N atom to which they are attached, a 4-7 membered nitrogen containing heterocycloalkyl group, such as-azetidine, aziridine, piperidinyl and the like. In a more specific embodiment, R5Is selected from-NH2、-NHCH3、-N(CH3)2、-NH-CH2CH3、-N(CH3)(CH2CH3)、-NH(CH2CH2CH3)、-NHCOCH3、-NHCO-CH2CH3-azetidine, aziridine or piperidinyl.
In one embodiment of the above preferred compounds of formula (I), R5Is selected from-COORcor-CONRcRcWherein R iscIndependently at each occurrence, is selected from H or C optionally substituted with halogen (preferably F)1-C6Alkyl radicals, e.g. COOH, -COOCH3、-COOCH2CH3、-CONH2、-CONHCH3、-CON(CH3)2、-CON(CH3)(CH2CH3). In a specific embodiment, R5Is selected from-CONRcRcIn which two R are bound to the same N atomcMay form, together with the N atom to which they are attached, a 4-7 membered nitrogen containing heterocycloalkyl group such as-CO-azetidine, -CO-aziridine, -CO-piperidinyl and the like.
In one embodiment of the above preferred compounds of formula (I), R6is-C1-C6Alkyl, preferably-C1-C3Alkyl, optionally substituted with halogen. Specific examples include, but are not limited to, methyl, ethyl, propyl or isopropyl, trifluoromethyl, trifluoroethyl, pentafluoroethyl, and the like.
In a more specific embodiment, R6is-C1-C3Alkyl groups such as methyl, ethyl, propyl or isopropyl.
In one embodiment of the above preferred compounds of formula (I), R6is-C1-C6Alkyl, preferably-C1-C3Alkyl, optionally substituted by-NRaRaIs substituted in which RaSelected from H or C optionally substituted by halogen1-C3Alkyl, or two R bound to the same N atomaMay form, together with the N atom to which they are attached, a 4-to 7-membered nitrogen-containing heterocycloalkyl group. Specific examples include, but are not limited to, aminomethyl, aminoethyl, aminopropyl, methylaminomethyl, dimethylaminomethyl, methylethylaminomethyl, azetidinylmethyl, pyrrolidinylmethyl, piperidinylmethyl, and the like.
In one embodiment of the above preferred compounds of formula (I), R6is-C1-C6Alkyl, preferably-C1-C3Alkyl, C optionally substituted by halogen3-C7Cycloalkyl is substituted. Specific examples include, but are not limited to, methyl, ethyl, propyl or isopropyl, cyclopropylmethyl, cyclopropylethyl, cyclobutylmethyl, cyclobutylethyl, cyclopentylmethyl, cyclopentylethylAnd the like.
In one embodiment of the above preferred compounds of formula (I), R6is-NRaRaWherein R isaSelected from H or C optionally substituted by halogen1-C3Alkyl, or two R bound to the same N atomaMay form, together with the N atom to which they are attached, a 4-to 7-membered nitrogen-containing heterocycloalkyl group. Specific examples include, but are not limited to, -NH2、-NHCH3、-NCH3CH3、-N(CH2CH3)CH3、-N(CH2CH3)(CH2CH3)-、-NHCF3、-N(CH3)CF3、-N(CF3)CF3、-N(CH2CF3)CF3or-N (CH)2CF3)(CH2CF3)-、-NH(CH2CH2CH3) Azetidinyl, pyrrolidinyl, piperidinyl and the like.
It is to be understood that the preferred compounds of formula (I) described above encompass each individual embodiment or each specific embodiment, as well as embodiments that consist of any combination or subcombination of the various embodiments or specific embodiments described above.
Preferred embodiments of the compounds of the present invention include the following compounds, stereoisomers, tautomers, stable isotopic variations, pharmaceutically acceptable salts or solvates thereof,
Figure BDA0003065598540000161
Figure BDA0003065598540000171
the invention has the advantages of
The invention provides biaryl compounds with a structure shown in a general formula (I), and researches show that the compounds can effectively inhibit ROR gamma t protein receptors, so that the differentiation of Th17 cells is regulated and controlled, the generation of IL-17 is inhibited, and the biaryl compounds can be used as immunomodulators for treating diseases related to Th17 cell differentiation.
The compound of the invention has the following beneficial effects:
has high inhibitory activity on ROR gamma t receptor;
regulating the differentiation of Th17 cells, and inhibiting the production of IL-17; and/or
Having good pharmacokinetic properties, e.g. having a longer t1/2Thus, for example, the dosing interval can be increased, the half-life is longer, and the patient has better compliance;
the compound has improved AUC0-last data, better druggability and higher bioavailability; and/or
Good safety, excellent properties like membrane permeability, P450 (reduced risk of drug interaction), solubility, etc.
Compounds of the invention for use in therapy or as medicaments
In one aspect, the present invention provides compounds of the invention for use as medicaments, in particular as ROR γ t inhibitors.
In another aspect, the present invention provides a compound of the invention for use in the treatment, especially for use in the treatment and/or prevention of a disease associated with roryt.
In particular embodiments, the invention provides compounds of the invention for use in the treatment and/or prevention of diseases in which ROR γ t contributes to the onset and progression of the disease or in which inhibition of ROR γ t would reduce the incidence of, reduce or eliminate the symptoms of, the disease, e.g., inflammation or autoimmune disease, cancer, etc., including but not limited to psoriasis, rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, multiple sclerosis, systemic lupus erythematosus, graft-versus-host disease, inflammatory bowel disease, crohn's disease, ulcerative colitis, chronic obstructive pulmonary disease, asthma, glomerulonephritis, lupus nephritis, myocarditis, thyroiditis, dry eye, uveitis, behcet disease, allergic dermatitis, acne, scleroderma, bronchitis, dermatomyositis, necrotizing enterocolitis, Hepatic fibrosis, nonalcoholic steatohepatitis (NASH), neocoronavirus pneumonia, insulin-dependent type I diabetes, triple negative breast cancer, prostate cancer, and the like.
Pharmaceutical compositions and their administration
In another aspect, for use of the compounds of the present specification for therapeutic or prophylactic purposes, the compounds of the present invention may be formulated as pharmaceutical compositions in accordance with standard pharmaceutical practice. Meanwhile, based on good pharmacokinetic property, improved AUC0-last and good drug forming property of the compound, the compound can be used for preparing a drug with better pharmacokinetic property and higher bioavailability.
Accordingly, the present invention provides a pharmaceutical composition comprising a compound of the invention as described above and a pharmaceutically acceptable excipient.
In a specific embodiment, said pharmaceutical composition of the invention is provided for use in the prevention or treatment of a disease associated with roryt in a mammalian, e.g. human, subject.
In a particular embodiment, the pharmaceutical composition of the invention may additionally comprise additional therapeutically active ingredients suitable for use in combination with the compounds of the invention. In another specific embodiment, the additional therapeutic agent is as defined herein for the pharmaceutical combination.
The Pharmaceutical compositions of the present invention may be formulated by techniques known to those skilled in the art, such as those disclosed in Remington's Pharmaceutical Sciences, 20 th edition. For example, the pharmaceutical compositions of the present invention described above may be prepared by mixing a compound of the present invention with one or more pharmaceutically acceptable excipients. The preparation may further comprise the step of mixing one or more other active ingredients with the compound of the present invention and one or more pharmaceutically acceptable excipients.
The choice of excipients to include in a particular composition will depend on a variety of factors, such as the mode of administration and the form of the composition provided. Suitable pharmaceutically acceptable excipients are well known to those skilled in the art and are described, for example, in Ansel, Howard C., et al, Ansel's Pharmaceutical Delivery Forms and Drug Delivery systems Philadelphia, Lippincott, Williams & Wilkins,2004, including, for example, adjuvants, diluents (e.g., glucose, lactose or mannitol), carriers, pH adjusting agents, buffers, sweeteners, fillers, stabilizers, surfactants, wetting agents, lubricants, emulsifiers, suspending agents, preservatives, antioxidants, opacifiers, glidants, processing aids, colorants, flavorants, flavoring agents, other known additives.
The pharmaceutical compositions of the present invention may be administered in a standard manner. For example, suitable modes of administration include oral, intravenous, rectal, parenteral, topical, transdermal, ocular, nasal, buccal or pulmonary (inhalation) administration, with parenteral infusion including intramuscular, intravenous, intraarterial, intraperitoneal or subcutaneous administration. For these purposes, the compounds of the present invention may be formulated by methods known in the art into the form of, for example, tablets, capsules, syrups, powders, granules, aqueous or oily solutions or suspensions, (lipid) emulsions, dispersible powders, suppositories, ointments, creams, drops, aerosols, dry powder formulations and sterile injectable aqueous or oily solutions or suspensions.
The size of a prophylactic or therapeutic dose of a compound of the invention will vary depending upon a number of factors, including the severity of the individual, disorder or condition being treated, the rate of administration, the disposition of the compound and the judgment of the prescribing physician. Generally, an effective dose is in the range of about 0.0001 to about 5000mg per kg body weight per day, e.g., about 0.01 to about 1000 mg/kg/day (single or divided administration). For a 70kg human, this may amount to about 0.007 mg/day to about 7000 mg/day, for example about 0.7 mg/day to about 1500 mg/day. Depending on the mode of administration, the amount or amount of a compound of the invention in a pharmaceutical composition may be from about 0.01mg to about 1000mg, suitably 0.1-500mg, preferably 0.5-300mg, more preferably 1-150mg, especially preferably 1-50mg, e.g. 1.5mg, 2mg, 4mg, 10mg, 25mg etc.; accordingly, the pharmaceutical composition of the invention will comprise 0.05 to 99% w/w (weight percentages), such as 0.05 to 80% w/w, such as 0.10 to 70% w/w, such as 0.10 to 50% w/w of the compound of the invention, all weight percentages being based on the total composition. It will be appreciated that it may be necessary in some cases to use dosages outside these limits.
In a specific embodiment, the present invention provides a pharmaceutical composition comprising a compound of the present invention and one or more pharmaceutically acceptable excipients, the composition being formulated for oral administration. The composition may be provided in unit dosage form, for example in the form of a tablet, capsule or oral liquid formulation. Such unit dosage forms may contain, as active ingredient, from 0.1mg to 1g, for example from 5mg to 250mg, of a compound of the invention.
In a specific embodiment, the present invention provides a pharmaceutical composition comprising a compound of the present invention and one or more pharmaceutically acceptable excipients, the composition being formulated for topical administration. Topical administration may be in the form of, for example, a cream, lotion, ointment, or transdermal patch.
In a specific embodiment, the present invention provides a pharmaceutical composition comprising a compound of the present invention and one or more pharmaceutically acceptable excipients, the composition being formulated for administration by inhalation. Inhalation administration can be by oral inhalation, or by intranasal administration. When administered by oral inhalation, the compounds of the invention may be effectively used in the present invention in a daily dose, for example up to 500. mu.g, such as 0.1 to 50. mu.g, 0.1 to 40. mu.g, 0.1 to 30. mu.g, 0.1 to 20. mu.g or 0.1 to 10. mu.g of a compound of the invention. The pharmaceutical compositions of the present invention for oral inhalation may be formulated as a dry powder, a suspension (in a liquid or gas) or a solution (in a liquid), and may be administered in any suitable form and using any suitable inhaler device known in the art, including for example Metered Dose Inhalers (MDIs), Dry Powder Inhalers (DPIs), nebulizers and soft mist inhalers. The multi-compartment device may be used to deliver a compound of the present specification and one or more other active ingredients (when present).
Methods of treatment and uses
Based on the beneficial effects described above with respect to the compounds of the present invention, the compounds of the present invention are useful in methods of treating various conditions in animals, particularly mammals such as humans.
Thus, in another aspect, the invention provides a method of modulating, particularly inhibiting, ROR γ t activity, the method comprising contacting a cell with a compound of the invention as hereinbefore described to modulate, particularly inhibit, ROR γ t activity in the cell.
In another aspect, the present invention provides a method of preventing or treating a disease associated with roryt (e.g. a disease treatable or preventable by roryt inhibition), the method comprising administering to a subject in need thereof an effective amount of a compound of the invention as hereinbefore described or a pharmaceutical composition of the invention comprising the same.
In another aspect, the invention provides the use of a compound of the invention, or a pharmaceutical composition comprising the same, as described hereinbefore, for inhibiting ROR γ t activity, or for the treatment and/or prevention of a disease associated with ROR γ t, for example a disease treatable or preventable by ROR γ t inhibition.
In another aspect, the present invention also provides the use of a compound of the invention or a pharmaceutical composition comprising the same as described above for the manufacture of a medicament, in particular a medicament having ROR γ t receptor inhibitor activity.
In another aspect, the invention provides the use of a compound of the invention, or a pharmaceutical composition comprising the same, as described hereinbefore, optionally in combination with one or more chemotherapeutics or immunotherapeutics, in the manufacture of a medicament for the treatment or prevention of a disease associated with roryt, for example a disease treatable or preventable by roryt inhibition.
Pharmaceutical combination
The compounds of the present invention may be administered as the sole active ingredient or in combination with additional drugs or therapies. The additional drugs or therapies may have or produce the same or different pharmacological effects, provided however that the use in combination with the compounds of the present invention does not result in an undesirable reduction in activity, adverse interaction or side effect.
Thus, in another aspect, the invention provides a pharmaceutical combination comprising or consisting of a compound of the invention as hereinbefore described together with one or more other drugs or therapies acting by the same or different mechanism of action. In a specific embodiment, the pharmaceutical combination is for use in inhibiting ROR γ t activity, or for use in the treatment and/or prevention of a disease associated with ROR γ t.
The compounds of the invention and the other active agents used in combination in the pharmaceutical combination of the invention may be administered simultaneously, separately or sequentially by the same or different routes of administration. The additional active agents may be co-administered in a single pharmaceutical composition with the compounds of the present invention or separately administered in separate discrete units from the compounds of the present invention, e.g., as a combination product, preferably in kit form. When administered separately, may be simultaneous or sequential, which sequential administration may be close in time or remote in time. Furthermore, the compound of the invention and the further medicament may be administered (i) prior to sending the combination product to a physician (e.g. in the case of a kit comprising a compound of the invention and a further medicament); (ii) by the physician himself (or under the direction of the physician) immediately before administration; (iii) the combination therapy is added by the patient himself, for example during sequential administration of the compound of the invention and the further drug, together.
Thus, in a particular embodiment, the invention also provides a kit comprising two or more separate pharmaceutical compositions, at least one of which comprises a compound of the invention, the remainder comprising other active agents used in combination, and means for separately containing said compositions. The kit of the invention is particularly suitable for administration of different dosage forms, such as oral dosage forms and parenteral dosage forms, or for administration of different compositions at different dosage intervals.
In the pharmaceutical combinations of the present invention, the appropriate amounts of the compound of the present invention and the other active agents to be combined will generally be determined by those skilled in the art by, for example, starting from the dosage ranges for the compounds described in the specification and approved or published dosage ranges for the other active compounds, and the dosages of the other drugs co-administered will, of course, vary depending upon such factors as the type of co-drug employed, the particular drug employed, the condition being treated, the general health of the patient, the judgment of the physician or veterinarian, and the like.
For pharmaceutical compositions and combinations of the invention, the additional active agent may be one or more second or additional (e.g., third) compounds that have enhanced, complementary activity that does not adversely affect the compound of the invention, e.g., the active agent may be a compound known to modulate other pathways of biological activity, or may be a compound that modulates different components of the pathway of biological activity to which the compound of the invention relates, or even a compound that overlaps with the biological target of the compound of the invention.
In a particular embodiment, the present invention provides a pharmaceutical combination, for example for use as a medicament for the treatment of one of the diseases listed herein, such as psoriasis, COPD, asthma, psoriatic arthritis or ankylosing spondylitis, comprising a compound of the invention, and at least one active ingredient selected from:
a) a beta-adrenoceptor agonist;
b) a muscarinic receptor antagonist;
c) joint muscarinic receptor antagonists and β -adrenergic receptor agonists; and
d) glucocorticoid receptor agonists (steroidal or non-steroidal);
e) phosphodiesterase-4 (PDE4) inhibitors.
The compounds of the present invention may also be combined with other therapies including, but not limited to, surgery, radiation therapy, transplantation (e.g., stem cell transplantation, bone marrow transplantation), tumor immunotherapy, and the like.
Accordingly, the present invention provides a method for inhibiting ROR γ t activity or for the treatment and/or prevention of a disease associated with ROR γ t, comprising administering a pharmaceutical combination of the invention to a subject in need thereof. The invention also provides the application of the pharmaceutical composition in preparing a medicament for inhibiting the activity of ROR gamma t or treating and/or preventing ROR gamma t related diseases.
For the various aspects described above relating to pharmaceutical compositions, methods of treatment and uses, and pharmaceutical combinations, diseases associated with ROR γ t (e.g., diseases treatable or preventable by ROR γ t inhibition) include inflammatory or autoimmune diseases, cancer, and the like, including, but not limited to, psoriasis, rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, multiple sclerosis, systemic lupus erythematosus, graft-versus-host disease, inflammatory bowel disease, crohn's disease, ulcerative colitis, chronic obstructive pulmonary disease, asthma, glomerulonephritis, lupus nephritis, myocarditis, thyroiditis, dry eye, uveitis, behcet's disease, allergic dermatitis, acne, scleroderma, bronchitis, dermato-muscular allergic rhinitis, necrotizing enterocolitis, hepatic fibrosis, nonalcoholic steatitis (NASH), neoviral pneumonia, neocoronary pneumonia, and the like, Insulin-dependent type I diabetes, triple negative breast cancer, prostate cancer, and the like. Preferred diseases associated with roryt are selected from psoriasis, rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, multiple sclerosis, inflammatory bowel disease, dry eye, allergic dermatitis, Chronic Obstructive Pulmonary Disease (COPD), asthma, necrotizing enterocolitis, liver fibrosis, nonalcoholic steatohepatitis (NASH), neocoronavirus pneumonia, triple negative breast cancer and prostate cancer.
Preferred for the above-described compounds, pharmaceutical compositions, methods, uses, pharmaceutical combinations of the invention are the preferred compounds of formula (I), stereoisomers, tautomers, stable isotopic variations, pharmaceutically acceptable salts or solvates thereof, as described above; more preferred are the specific compounds listed above, e.g., compounds 1-26, or their pharmaceutically acceptable salts or solvates.
When a dosage of a compound or drug for administration is described herein, it is understood that the dosage is based on the weight of the free base, excluding any derivative thereof, unless the specification indicates otherwise.
Synthesis of Compounds of the invention
In another aspect, the invention also provides a process for the preparation of compounds of formula (I), the general synthetic scheme for the synthesis of the compounds of the invention being illustrated below. Suitable reaction conditions for each reaction step are known to those skilled in the art or can be routinely determined. The starting materials and reagents used in the preparation of these compounds are generally commercially available or can be prepared by the methods hereinafter, by methods similar to those given hereinafter, or by methods known in the art, if not otherwise specified. If desired, the starting materials and intermediates in the synthetic reaction schemes can be isolated and purified using conventional techniques, including, but not limited to, filtration, distillation, crystallization, chromatography, and the like. The materials may be characterized using conventional methods including physical constants and spectroscopic data.
In one embodiment, the method comprises the steps of:
synthesis scheme 1:
Figure BDA0003065598540000221
wherein R is1、R2、R3、R4、R6、RcAnd X is as defined above for formula (I), Z is Br or I; and R iscIs not H;
step 1: the compound of formula (I-1) or formula (I-2) and the compound of formula (I-3) are subjected to coupling reaction such as Suzuki coupling reaction under the action of a catalyst such as a palladium catalyst to obtain an intermediate compound of formula (I-4);
wherein the palladium catalyst is a palladium catalyst for Suzuki coupling as is well known in the art, including but not limited to PdCl2(dtbpf)、Pd(dppf)Cl2Etc.; the reaction is preferably carried out in the presence of a suitable base which may be selected from sodium hydroxide, potassium hydroxide, sodium carbonate, potassium carbonate or cesium carbonate, preferably the base is potassium carbonate;
step 2: reacting the compound shown in the formula (I-5) with paraformaldehyde under the action of sodium methoxide to generate a compound shown in the formula (I-6);
and step 3: alkylation of the compound of formula (I-6) to produce a compound of formula (I-7); the alkylation is for example in HBF4With TMSCN or, for example, with an alkyl halide in the presence of a base; the alkali can be selected from butyl lithium, sodium hydroxide, potassium hydroxide, sodium carbonate, potassium carbonate or cesium carbonate;
and 4, step 4: hydrolysis of a compound of formula (I-7) to give a compound of formula (I-8), preferably in the presence of a base selected from lithium hydroxide, sodium hydroxide, potassium hydroxide, sodium carbonate, potassium carbonate or cesium carbonate or an acid selected from hydrochloric acid, sulphuric acid or trifluoroacetic acid; and/or
And 5: the compound of the formula (I-8) and the intermediate compound of the formula (I-4) generate the compound of the formula (I-a) under the action of a condensing agent.
Wherein the condensing agent is one well known in the art for coupling carboxylic acids with amines, including but not limited to EDC, DCC, HATU, N' -tetramethylchloroformamidine hexafluorophosphate, and the like; the reaction is preferably carried out in the presence of a suitable base including, but not limited to, sodium carbonate, potassium carbonate, cesium carbonate, N-diisopropylethylamine, triethylamine, HOBt, N-methylimidazole or pyridine.
In one embodiment, the method comprises the steps of:
synthesis scheme 2:
Figure BDA0003065598540000231
wherein R is1、R2、R3、R4、R6、RcAnd X is as defined above for formula (I).
Step 1: the compound of the formula (II-1) and the compound of the formula (II-2) generate the compound of the formula (II-3) under the action of a condensing agent.
Wherein the condensing agent is one well known in the art for coupling carboxylic acids with amines, including but not limited to EDC, DCC, HATU, N' -tetramethylchloroformamidine hexafluorophosphate, and the like; (ii) a The reaction is preferably carried out in the presence of a suitable base including, but not limited to, sodium carbonate, potassium carbonate, cesium carbonate, N-diisopropylethylamine, triethylamine, HOBt, N-methylimidazole or pyridine;
step 2: reacting the compound of the formula (II-3) with paraformaldehyde under the action of sodium methoxide to generate a compound of a formula (I-b);
step 3 a: alkylating the compound of formula (I-b) to produce a compound of formula (I-a); the alkylation is for example in HBF4With TMSCN or, for example, with an alkyl halide in the presence of a base; the alkali is selected from butyl lithium, sodium hydroxide, potassium hydroxide, and sodium carbonatePotassium carbonate or cesium carbonate;
and step 3 b: reacting the compound shown in the formula (I-b) with methanesulfonyl chloride to generate a compound shown in a formula (II-4); the reaction is preferably carried out in the presence of a suitable base including, but not limited to, sodium carbonate, potassium carbonate, cesium carbonate, N-diisopropylethylamine, triethylamine, HOBt, N-methylimidazole or pyridine; and/or
And 4, step 4: the compound of formula (I-4) and HNRcRcNucleophilic substitution reaction to generate a compound of formula (I-c); the reaction is preferably carried out in the presence of a suitable base including, but not limited to, sodium carbonate, potassium carbonate, cesium carbonate, N-diisopropylethylamine, triethylamine, HOBt, N-methylimidazole or pyridine.
In one embodiment, the method comprises the steps of:
synthesis scheme 3:
Figure BDA0003065598540000241
wherein R is1、R2、R3、R4、R6、RcAnd X is as defined above for formula (I),
step 1a) Compounds of formula (III-1) with RcCOOH or an activated form thereof, such as the corresponding acid chloride or anhydride, to produce a compound of formula (I-d); the reaction is preferably carried out in the presence of a base selected from sodium carbonate, potassium carbonate, cesium carbonate, N-diisopropylethylamine, triethylamine, HOBt or pyridine; the reaction may be carried out in the presence of condensing agents such as, for example, and the like, wherein the condensing agents are well known in the art for the coupling of carboxylic acids to amines, including but not limited to 1-propylphosphoric anhydride (T3P), EDC, DCC, HATU; or
Step 1b) Compounds of formula (III-1) with RcSO2Cl to generate a compound shown as a formula (I-e); the reaction is preferably carried out in the presence of a base selected from sodium carbonate, potassium carbonate, cesium carbonate, N-diisopropylethylamine, triethylamine, HOBt or pyridine;
in one embodiment, the method comprises the steps of:
synthesis scheme 4:
Figure BDA0003065598540000251
wherein R is1、R2、R3、R4、R6、RcAnd X is as defined above for formula (I),
step 1: hydrolysis of the compound of formula (IV-1) to give the compound of formula (IV-2), preferably in the presence of a base selected from lithium hydroxide, sodium hydroxide, potassium hydroxide, sodium carbonate, potassium carbonate or cesium carbonate,
step 2: reacting the compound of formula (IV-2) with the compound of formula (I-4) to obtain the compound of formula (IV-3) under the conditions described in step 1 of FIG. 2;
and step 3: hydrolysis of a compound of formula (IV-3) to give a compound of formula (I-f), preferably in the presence of an acid which may be selected from, for example, hydrochloric acid, sulphuric acid or trifluoroacetic acid;
step 4 a: optionally, a compound of formula (I-f) with RcOH reaction to obtain a compound of formula (I-g); the reaction is preferably carried out in the presence of a base selected from sodium carbonate, potassium carbonate, cesium carbonate, N-diisopropylethylamine, triethylamine, HOBt or pyridine; the reaction may be carried out in the presence of a condensing agent such as, for example, an alcohol, wherein the condensing agent is well known in the art for coupling carboxylic acids to alcohols, including but not limited to 1-propylphosphoric anhydride (T3P), EDC, DCC, or HATU;
and 4 b: optionally, a compound of formula (I-f) with HNRcRcThe reaction gives compounds of formula (I-h) under conditions as described in FIG. 2, step 1;
the reaction in the above step is preferably carried out in an organic solvent which may be selected from alcohol solvents such as (methanol, ethanol or propanol), tetrahydrofuran, ethers (such as diethyl ether, ethylene glycol monomethyl ether, etc.), N-methylpyrrolidone, N-dimethylformamide, N-dimethylacetamide, 1, 4-dioxane, dichloromethane, dimethyl sulfoxide and any combination thereof; mixtures of organic solvents with water can be used for the hydrolysis reaction;
the reaction in the above step is preferably carried out at a suitable temperature, for example, -100 ℃ to 0 ℃, -80 ℃ to-20 ℃, -50 ℃ to 200 ℃, -20 ℃ to 100 ℃, -20 ℃ to 20 ℃, -10 ℃ to 50 ℃, 0-200 ℃, 10-100 ℃, 20-50 ℃ or room temperature (20-25 ℃). The above synthetic schemes are only illustrative of some of the methods of preparation of the compounds of the present invention. The compounds of the present invention or stereoisomers, tautomers, stable isotopic derivatives, pharmaceutically acceptable salts or solvates thereof can be prepared by a variety of methods, including those set forth above, in the examples, or by analogous methods thereto, by one of ordinary skill in the art based on the synthetic schemes described above, in conjunction with routine techniques in the art.
The compounds described in this specification are further illustrated in the following examples. These examples are given by way of illustration only and not by way of limitation.
Detailed Description
The technical solutions of the present invention are further described below with reference to specific examples, but the scope of the present invention is not limited to these examples. All changes, modifications and equivalents that do not depart from the spirit of the invention are intended to be included within the scope thereof.
Experimental procedures without specific conditions noted in the following examples are generally carried out according to conventional conditions for such reactions, or according to conditions recommended by the manufacturer. Unless otherwise indicated, percentages and parts are percentages and parts by weight. Unless otherwise specified, the ratio of liquids is by volume.
The experimental materials and reagents used in the following examples are commercially available, prepared according to methods of the prior art or prepared according to methods similar to those disclosed herein, unless otherwise specified.
Chemical names used herein are IUPAC names and abbreviations used herein have meanings commonly understood in the art unless clearly defined otherwise in the specification. The meanings of the abbreviations used in the specification are listed below:
PdCl2(dtbpf): 1,1' -di-tert-butylphosphino ferrocene palladium dichloride
Pd(dppf)Cl2: [1,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride
xanthophos 4, 5-bis-diphenylphosphine-9, 9-dimethylxanthene
Pd2(dba)3Tris (dibenzylideneacetone) dipalladium
HATU: 2- (7-azobenzotriazol) -N, N, N ', N', -tetramethyluronium hexafluorophosphate
TCFH N, N, N ', N' -tetramethylchloroformamidine hexafluorophosphate
DIEA: n, N-diisopropylethylamine
DMF: n, N-dimethylformamide
DMSO, DMSO: dimethyl sulfoxide
DCM: methylene dichloride
EA ethyl acetate
PE: petroleum ether
rt-room temperature
LC-MS: liquid chromatography-mass spectrometry
ESI electrospray ionization
m/z: mass to charge ratio
TLC thin layer chromatography
Time: retention time
Synthetic examples
In the preparation method of the target compound, the column chromatography adopts silica gel (300-400 meshes) produced by Ningsan Sun drying agent GmbH; thin layer chromatography using GF254(0.25 mm); nuclear magnetic resonance chromatography (NMR) was measured using a Varian-400 nuclear magnetic resonance spectrometer; LC/MS an Agilent technology ESI 6120 LC/MS instrument was used.
In addition, all operations involving easily oxidizable or hydrolyzable raw materials were carried out under nitrogen protection. Unless otherwise indicated, all starting materials used in the present invention are commercially available, can be used without further purification, and are used in the present invention at temperatures in degrees Celsius.
When the structure of the compound of the present invention is inconsistent with the name of the compound, the structural formula is generally subject to the standard unless the name of the compound can be determined to be correct by context.
Example 1: n- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-yl) -2- (4- (methylsulfonyl) phenyl) -3-methoxypropionamide
Figure BDA0003065598540000271
Synthesis of intermediate 2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-amine
Step 1: synthesis of 4-bromo-3, 5-dichloroaniline
Figure BDA0003065598540000272
N-bromosuccinimide (69.2g,389mmol) was added to a solution of 3, 5-dichloroaniline (60.0g,370mmol) in acetonitrile (1.00L) under ice-cooling, and the reaction mixture was stirred at room temperature for 1.5 hours, whereupon the reaction was stopped. The reaction mixture was concentrated under reduced pressure to give a residue, and a mixed solvent of PE/EA (200mL, volume ratio 10:1) was added thereto, and after stirring at room temperature for 10 minutes, the mixture was filtered, and the filter cake was washed with PE (150mL), and dried to obtain the objective compound (70.0g, yield 78.5%, brown solid). LC-MS (ESI) M/z 240.2[ M + H ]]+/242.2[M+2+H]+1H NMR(400MHz,CDCl3)δ8.64(s,2H),6.71(s,2H)。
Step 2: synthesis of 2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-amine
Figure BDA0003065598540000273
To a mixed solution of 4-bromo-3, 5-dichloroaniline (49.5g,206mmol) and (2- (trifluoromethoxy) phenyl) boronic acid (46.5g,226mmol) in water (50.0mL) and 1, 4-dioxane (500mL) was added potassium carbonate (56.8g,411mmol) and PdCl2(dtbpf) (4.03g,6.16 mmol). The reaction mixture was stirred at 60 ℃ for 16 hours under argon protection, and then the reaction was stopped. Cooling the reaction mixture to room temperature, adding water (C)300mL), the mixture was extracted with EA (300mL × 2), the organic phase was separated, washed with saturated brine (200mL × 2), dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the resulting residue was purified by column chromatography (PE: EA 12:1-8:1) to give the title compound (20.5g, yield 31.0%, white solid). LC-MS (ESI) M/z 321.9[ M + H ]]+/323.9[M+2+H]+1H NMR(400MHz,DMSO-d6)δ7.56–7.50(m,1H),7.47–7.41(m,2H),7.35–7.30(m,1H),6.71(s,2H),5.81(s,2H)。
Intermediate: synthesis of 2- (4- (methylsulfonyl) phenyl) -3-methoxypropionic acid
Step 1: synthesis of methyl 2- (4-methylsulfonyl) phenylacetate
Figure BDA0003065598540000281
2- (4-Methylsulfonyl) phenylacetic acid (9.00g,42.0mmol) was dissolved in methanol (100mL) and thionyl chloride (15.0g,126mmol) was added under ice-water bath conditions. The reaction mixture was warmed to room temperature and stirred for 1 hour, and the reaction was stopped. The reaction solution was concentrated under reduced pressure, and the obtained residue was purified by column chromatography (PE: EA ═ 3:1-2:1) to obtain the objective compound (8.60g, yield 89.7%, yellow solid). LC-MS (ESI) M/z 229.0[ M + H ]]+1H NMR(400MHz,CDCl3)δ7.91(d,J=8.4Hz,2H),7.49(d,J=8.5Hz,2H),3.73(s,2H),3.72(s,3H),3.05(s,3H)。
Step 2: synthesis of methyl 2- (4-methylsulfonyl) phenyl-3-hydroxypropionate
Figure BDA0003065598540000282
Methyl 2- (4-methylsulfonyl) phenylacetate (9.40g,41.2mmol) was dissolved in anhydrous DMSO (100.0mL), and paraformaldehyde (1.17g,39.2mmol) and sodium methoxide (89.0mg,1.65mmol) were sequentially added thereto, and the reaction was stopped after the reaction solution was stirred at room temperature for 16 hours. The reaction mixture was added to ice water (300.0mL), the mixture was neutralized with a 1.0M hydrochloric acid solution, extracted with EA (400 mL. times.2), and allowed to stand for separation. The organic phase was washed with saturated brine (60.0 mL. times.3),dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the obtained residue was purified by column chromatography (PE: EA ═ 3:1-1:1) to obtain the objective compound (6.15g, yield 57.8%, light yellow oil).1H NMR(400MHz,DMSO-d6)δ7.87(d,2H),7.57(d,2H),5.08(t,J=4.9Hz,1H),3.95–3.90(m,2H),3.72–3.65(m,1H),3.61(s,3H),3.18(s,3H)。
And step 3: synthesis of methyl 2- (4- (methylsulfonyl) phenyl) -3-methoxypropionate
Figure BDA0003065598540000283
Methyl 2- (4- (methylsulfonyl) phenyl) -3-hydroxypropionate (6.10g,23.6mmol) was dissolved in DCM (140mL), and 50% aqueous fluoroboric acid (4.14g,23.6mmol) and trimethylsilylated diazomethane (11.8mL,23.6mmol,2.0M n-hexane solution) were added to the reaction mixture in an ice-water bath under exclusion of light. After 20 minutes, trimethylsilylated diazomethane (11.8mL,23.6mmol,2.0M n-hexane solution) was added again. After 20 minutes, trimethylsilylated diazomethane (11.8mL,23.6mmol,2.0M n-hexane solution) was added again. After 20 minutes, trimethylsilylated diazomethane (11.8mL,23.6mmol,2.0M n-hexane solution) was added again. The reaction mixture was poured into water (80.0mL), extracted with DCM (200 mL. times.3), and allowed to stand for separation. The organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the obtained residue was purified by column chromatography (PE: EA ═ 5:1-3:1) to obtain the objective compound (4.62g, yield 71.9%, colorless oil). LC-MS (ESI) M/z 273.2[ M + H ]]+1H NMR(400MHz,CDCl3)δ7.90(d,2H),7.53(d,2H),4.00–3.92(m,2H),3.71(s,3H),3.69–3.65(m,1H),3.35(s,3H),3.04(s,3H)。
And 4, step 4: synthesis of 2- (4- (methylsulfonyl) phenyl) -3-methoxypropionic acid
Figure BDA0003065598540000291
Methyl 2- (4- (methylsulfonyl) phenyl) -3-methoxypropionate (3.60g,13.2mmol) was dissolved in 1.4-dioxane (40).0mL) and 6.0M hydrochloric acid (40.0 mL). The reaction mixture was stirred at 80 ℃ for 1 hour, and then the reaction was stopped. The reaction mixture was cooled to room temperature, poured into water (50.0mL), extracted with EA (200 mL. times.2), and allowed to stand for separation. The organic phase was dried over anhydrous sodium sulfate, filtered, the filtrate concentrated under reduced pressure, and the residue was subjected to medium pressure liquid phase preparation (acetonitrile/water containing 0.05% formic acid) to give the title compound (2.00g, yield 58.8%, white solid). MS (ESI) M/z 515.0[2M-H]-1H NMR(400MHz,CDCl3)δ7.91(d,J=8.4Hz,2H),7.56(d,J=8.4Hz,2H),4.02–3.92(m,2H),3.74–3.68(m,1H),3.37(s,3H),3.04(s,3H)。
Synthesis of N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-yl) -2- (4- (methylsulfonyl) phenyl) -3-methoxypropionamide
Figure BDA0003065598540000292
2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl]-4-amine (50.0mg,0.155mmol) and 2- (4- (methylsulfonyl) phenyl) -3-methoxypropionic acid (48.0mg, 0.186mmol) were added to acetonitrile (2.0 mL). To the reaction mixture was added N-methylimidazole (26.8mg, 0.326mmol) and TCFH (52.2mg, 0.186 mmol). The reaction mixture was stirred at room temperature for 1.5 hours. The reaction mixture was directly purified by reverse phase chromatography (C18, methanol: water 0-80%) to obtain the objective compound (40.0mg, yield 45.9%, white solid). LC-MS (ESI) M/z [ M + H ]]+/[M+2+H]+562.0/564.0。1H NMR(500MHz,DMSO-d6)δ10.73(s,1H),7.93(d,J=8.3Hz,2H),7.84(s,2H),7.68(d,J=8.4Hz,2H),7.63–7.59(m,1H),7.54–7.49(m,2H),7.41–7.37(m,1H),4.16–4.12(m,1H),4.04–3.98(m,1H),3.66–3.61(m,1H),3.30(s,3H),3.21(s,3H)。
Examples 2 and 3: rel- (S) -N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-yl) -2- (4- (methylsulfonyl) phenyl) -3-methoxypropionamide or rel- (R) -N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-yl) -2- (4- (methylsulfonyl) phenyl) -3-methoxypropionamide
Figure BDA0003065598540000301
Racemic N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl)]-4-yl) -2- (4- (methylsulfonyl) phenyl) -3-methoxypropionamide (50mg in about 13mL methanol, 1.5mL injection volume) was passed through Waters SFC 150 (room temperature, 100bar,214nm) and 250. mu.25 mm 10Dr
Figure BDA0003065598540000304
OD)) (supercritical carbon dioxide: methanol, 80:20,6.0min,70mL/min) to obtain rel- (S) -N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl)]-4-yl) -2- (4- (methylsulfonyl) phenyl) -3-methoxypropionamide or rel- (R) -N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl]-4-yl) -2- (4- (methylsulfonyl) phenyl) -3-methoxypropionamide (13.8mg, white solid, ret. time. 2.961min, e.e.99%). LC-MS (ESI) M/z 561.9[ M + H ]]+/563.9[M+2+H]+1H NMR(400MHz,CDCl3)δ8.41(s,1H),7.96(d,J=8.4Hz,2H),7.67(s,2H),7.59(d,J=8.4Hz,2H),7.48–7.45(m,1H),7.39–7.35(m,2H),7.26-7.25,7.24-7.23(m,1H),4.06–4.00(m,1H),3.97–3.92(m,1H),3.82–3.78(m,1H),3.51(s,3H),3.06(s,3H)。
rel- (R) -N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl)]-4-yl) -2- (4- (methylsulfonyl) phenyl) -3-methoxypropionamide or rel- (S) -N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl]-4-yl) -2- (4- (methylsulfonyl) phenyl) -3-methoxypropionamide (12.9mg, white solid, ret. time 3.380min, e.e.97%). LC-MS (ESI) M/z 561.8[ M + H ]]+/563.8[M+2+H]+1H NMR(400MHz,CDCl3)δ8.41(s,1H),7.96(d,J=8.4Hz,2H),7.67(s,2H),7.59(d,J=8.4Hz,2H),7.50–7.45(m,1H),7.40–7.35(m,2H),7.26–7.25,7.24-7.23(m,1H),4.06–4.01(m,1H),3.96–3.92(m,1H),3.82–3.78(m,1H),3.51(s,3H),3.06(s,3H)。
Example 4: n- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-yl) -2- (4- (methylsulfonyl) phenyl) -3-hydroxypropionamide
Figure BDA0003065598540000302
Step 1: synthesis of N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-yl) -2- (4- (methylsulfonyl) phenyl) acetamide
Figure BDA0003065598540000303
2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl]-4-amine (150mg, 0.466mmol), 2- (4- (methylsulfonyl) phenyl) acetic acid (120mg, 0.559mmol) and HATU (213mg, 0.559mmol) were added to DCM (10 mL). DIEA (0.231mL, 1.40mmol) was added to the reaction mixture. The reaction mixture was stirred at room temperature for 4 hours. Saturated aqueous sodium bicarbonate (5.0mL) was added. After separating the organic phase, the aqueous phase was extracted with more DCM (10mL), the organic phases were combined and washed with saturated brine, dried over anhydrous sodium sulfate, and filtered. And concentrating the filtrate under reduced pressure to obtain a crude product. Purification by column chromatography (silica gel, EA/PE 0-60%) gave the title compound (240mg, 99.4% yield, white solid). LC-MS (ESI) M/z 518.0[ M + H ]]+
Step 2: synthesis of N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-yl) -2- (4- (methylsulfonyl) phenyl) -3-hydroxypropionamide
Figure BDA0003065598540000311
Mixing N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl)]-4-yl) -2- (4- (methylsulfonyl) phenyl) acetamide (240mg, 0.463mmol) was added to anhydrous DMSO (8 mL). To the reaction mixture was added paraformaldehyde (12.5mg, 0.417mmol) and sodium methoxide (1.03mg, 0.019mmol) in this order. The reaction mixture was stirred at room temperature for 4 hours. EA (15mL) and water (20mL) were added. The organic phase was separated, the aqueous phase was extracted with EA (15mL), and the combined organic phases were washed with saturated brine (15 mL. times.3), dried over anhydrous sodium sulfate, and filtered. And concentrating the filtrate under reduced pressure to obtain a crude product. Warp columnPurification by chromatography (silica gel, EA/PE 0-100%) gave the title compound (149mg, 58.7% yield, white solid). LC-MS (ESI) M/z 548.0[ M + H ]]+1H NMR(400MHz,DMSO-d6)δ10.67(s,1H),7.92(d,J=8.5Hz,2H),7.88–7.83(m,2H),7.66(d,J=8.5Hz,2H),7.63–7.57(m,1H),7.54–7.48(m,2H),7.41–7.37(m,1H),5.14(t,J=5.2Hz,1H),4.13–4.06(m,1H),4.01–3.94(m,1H),3.72–3.63(m,1H),3.20(s,3H)。
Examples 5 and 6: rel- (S) -N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-yl) -2- (4- (methylsulfonyl) phenyl) -3-hydroxypropionamide or rel- (R) -N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-yl) -2- (4- (methylsulfonyl) phenyl) -3-hydroxypropionamide
Figure BDA0003065598540000312
Racemic N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl)]-4-yl) -2- (4- (methylsulfonyl) phenyl) -3-hydroxypropionamide (470mg in about 120mL of methanol, injection volume 3.0mL) was purified by passing Waters SFC 150 (room temperature, 100bar,214nm) and 250 × 25mm 10 μm Dr
Figure BDA0003065598540000313
OD) (supercritical carbon dioxide: methanol, 70:30,3.0min,70mL/min) to obtain rel- (S) -N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl)]-4-yl) -2- (4- (methylsulfonyl) phenyl) -3-hydroxypropionamide or rel- (R) -N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl]-4-yl) -2- (4- (methylsulfonyl) phenyl) -3-hydroxypropionamide (204mg, white solid, ret. time. 4.166min, e.e.99%). LC-MS (ESI) M/z 548.2[ M + H ]]+/550.2[M+2+H]+1H NMR(400MHz,CDCl3)δ8.23(s,1H),7.89(d,J=8.2Hz,2H),7.70(d,J=4.6Hz,2H),7.57(d,J=8.2Hz,2H),7.50–7.44(m,1H),7.40–7.34(m,2H),7.26–7.23(m,1H),4.23–4.15(m,1H),4.04–3.92(m,2H),3.07(s,3H)。
rel- (R) -N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl)]-4-yl) -2- (4- (methylsulfonyl) phenyl) -3-hydroxypropionamide or rel- (S) -N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl]-4-yl) -2- (4- (methylsulfonyl) phenyl) -3-hydroxypropionamide (171mg, white solid, ret. time. 4.453min, e.e.99%). LC-MS (ESI) M/z 548.2[ M + H ]]+/550.2[M+2+H]+1H NMR(400MHz,CDCl3)δ8.04(s,1H),7.99–7.89(m,2H),7.77–7.67(m,2H),7.66–7.56(m,2H),7.53–7.47(m,1H),7.44–7.36(m,2H),7.29–7.24(m,1H),4.32–4.16(m,1H),4.11–3.90(m,2H),3.10(s,3H)。
Example 7: 4- ((2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-yl) amino) -3- (4- (methylsulfonyl) phenyl) -4-oxobutanoic acid
Figure BDA0003065598540000321
Step 1: synthesis of 4-tert-butyl 2- (4- (methylsulfonyl) phenyl) succinate 1-methyl ester
Figure BDA0003065598540000322
Methyl 2- (4- (methylsulfonyl) phenyl) acetate (900mg,3.95mmol) was added to tetrahydrofuran (10mL), cooled to 0 ℃, and NaH (166mg,4.15mmol, 60% in oil) was added and the reaction was continued at 0 ℃ for 30 minutes, then tert-butyl 2-bromoacetate (847mg/0.63mL,4.34mmol) was added to the reaction flask by syringe, the reaction was continued at 0 ℃ for 30 minutes, TLC detected that the starting material was reacted, after the reaction was completed, saturated ammonium chloride (20mL) was added to quench the reaction, EA extraction (20mLx3) was performed, silica gel column separation [ (PE: DCM ═ 2: 1): EA is 100% -65%]The desired product was obtained (920mg, yield 68.1%, colorless oil).1H NMR(400MHz,CDCl3)δ7.93–7.88(m,2H),7.53–7.48(m,2H),4.18–4.10(m,1H),3.69(s,3H),3.18–3.07(m,1H),3.04(s,3H),2.68–2.58(m,1H).
Step 2: synthesis of 4- (tert-butoxy) -2- (4- (methylsulfonyl) phenyl) -4-oxobutanoic acid
Figure BDA0003065598540000323
4-tert-butyl 2- (4- (methylsulfonyl) phenyl) succinate 1-methyl ester (810mg,2.37mmol) and lithium hydroxide (109mg,2.6mmol) were added to a tetrahydrofuran/water (10mL/2mL) solution, reacted at room temperature for 1 hour, 1N hydrochloric acid (3mL) was added, EA (10mLx3) was extracted, washed once with a saturated sodium chloride (20mL) solution, and the solvent was spin-dried under reduced pressure to give the desired product (800mg, colorless oil). LC-MS (ESI) M/z 273.1[ M-56+ H ]]+.
And step 3: synthesis of tert-butyl 4- ((2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-yl) amino) -3- (4- (methylsulfonyl) phenyl) -4-oxobutyrate
Figure BDA0003065598540000331
2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-amine (200mg,0.62mmol), 4- (tert-butoxy) -2- (4- (methylsulfonyl) phenyl) -4-oxobutanoic acid (345mg,1.05mmol), HATU (285mg,0.75mmol) and DIEA (160mg,1.24mmol) were added to DCM (5mL) and reacted overnight at room temperature, TLC checked for completion of the starting material reaction, after which the reaction was completed, washed once with saturated sodium bicarbonate, and silica gel column separated [ (PE: DCM ═ 2: 1): EA 100% -50% ] to obtain the target product (500mg, colorless oil). And 4, step 4: synthesis of 4- ((2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-yl) amino) -3- (4- (methylsulfonyl) phenyl) -4-oxobutanoic acid
Figure BDA0003065598540000332
The preparation method comprises the step of reacting 4- ((2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl)]Tert-butyl (4-yl) amino) -3- (4- (methylsulfonyl) phenyl) -4-oxobutyrate (500mg) was added to DCM (5mL), then trifluoroacetic acid (1mL) was added, the reaction was carried out at room temperature for 2 hours, after completion of the reaction, the solvent was directly spin-dried, then EA (20mL) was added, saturated sodium bicarbonate was washed once, the organic layer was spin-dried to obtain a crude product, and the crude product was subjected to reverse phase column separation (water: methanol is 100%, 0-30%, 70%) The expected product was obtained (270mg, 73.6% yield, white solid). LC-MS (ESI) M/z 576.0[ M + H ]]+1H NMR(400MHz,DMSO-d6)δ12.44(s,1H),10.74(s,1H),7.97–7.89(m,2H),7.84–7.77(m,2H),7.71–7.63(m,2H),7.63–7.56(m,1H),7.54–7.47(m,2H),7.39–7.34(m,1H),4.29–4.18(m,1H),3.25,3.20(s,3H),3.18–3.09(m,1H),2.76–2.67(m,1H).
Example 8: n is a radical of1- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl)]-4-yl) -N4-methyl-2- (4- (methylsulfonyl) phenyl) succinamide
Figure BDA0003065598540000333
The preparation method comprises the step of reacting 4- ((2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl)]-4-yl) amino) -3- (4- (methylsulfonyl) phenyl) -4-oxobutanoic acid (270mg,0.47mmol), methylamine hydrochloride (53mg,0.94mmol), HATU (357mg,0.94mmol) and DIEA (243mg,1.88mmol) were added to DCM (5mL) and reacted overnight at room temperature, TLC checked for completion of the starting material reaction, after which time it was washed with saturated sodium bicarbonate, plate separated (DCM: methanol-40: 1) gave the title product (120mg, 43.3% yield, white solid). LC-MS (ESI) M/z 589.0[ M + H ]]+1H NMR(400MHz,DMSO-d6)δ10.73(s,1H),7.95–7.88(m,3H),7.81(dd,J=6.7,2.0Hz,2H),7.64(d,J=8.5Hz,2H),7.62–7.56(m,1H),7.54–7.47(m,2H),7.37(dd,J=7.9,1.7Hz,1H),4.33–4.26(m,1H),3.20(s,3H),3.05–2.96(m,1H),2.62–2.52(m,4H).
Example 9: n is a radical of1- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl)]-4-yl) -2- (4- (methylsulfonyl) phenyl) succinamide
Figure BDA0003065598540000341
The preparation method comprises the step of reacting 4- ((2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl)]-4-yl) amino) -3- (4- (methylsulfonyl) phenyl) -4-oxobutanoic acid (58mg,0.1mmol), ammonium chloride (10.6mg,0.2mmol), HATU (46mg,0.12mmol) and DIPEA (26mg,0.2mmol) were added to DCM (2mL),the reaction was carried out overnight at room temperature, the starting material was completely reacted by TLC, and after completion of the reaction, it was washed once with saturated sodium bicarbonate and separated on preparative plates (PE: EA: 1) to give the desired product (8mg, yield 13.8%, white solid). LC-MS (ESI) M/z 577.0[ M + H ]]+1H NMR(500MHz,DMSO-d6)δ10.46(s,1H),7.91(d,J=8.4Hz,2H),7.79(dd,J=6.4,2.0Hz,2H),7.70–7.65(m,1H),7.65–7.57(m,3H),7.54–7.48(m,2H),7.42–7.38(m,1H),7.00(s,1H),4.21–4.14(m,1H),3.21(s,3H),3.18–3.11(m,1H),2.82–2.75(m,1H).
Example 10: n- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-yl) -2- (4- (ethylsulfonyl) phenyl) -3-hydroxypropionamide
Figure BDA0003065598540000342
Step 1: synthesis of N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-yl) -2- (4- (ethylsulfonyl) phenyl) acetamide
Figure BDA0003065598540000343
2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl]-4-amine (50.0mg,0.155mmol) and 2- (4- (ethylsulfonyl) phenyl) acetic acid (42.5mg, 0.186mmol) were added to DCM (5 mL). To the reaction mixture was added HATU (70.7mg, 0.186mmol) and DIEA (0.077mL, 0.465mmol) in that order. The reaction mixture was stirred at room temperature overnight. Saturated aqueous sodium bicarbonate (10mL) and DCM (10mL) were added. The aqueous phase was separated and extracted with DCM (10 mL). The combined organic phases were washed with saturated brine, dried over anhydrous sodium sulfate and filtered. And concentrating the filtrate under reduced pressure to obtain a crude product. Purification by column chromatography (silica gel, EA/PE 0-100%) gave the title compound (70.0mg, 84.8% yield, white solid). LC-MS (ESI) M/z [ M + H ]]+532.0。
Step 2: synthesis of N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-yl) -2- (4- (ethylsulfonyl) phenyl) -3-hydroxypropionamide
Figure BDA0003065598540000351
Mixing N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl)]-4-yl) -2- (4- (ethylsulfonyl) phenyl) acetamide (70.0mg, 0.131mmol) was added to anhydrous DMSO (2 mL). To the reaction mixture were added paraformaldehyde (3.54mg, 0.118mmol) and sodium methoxide (0.27mg,0.005mmol) in this order. The reaction mixture was stirred at room temperature overnight. EA (10mL) and water (10mL) were added. The aqueous phase was separated and extracted with EA (10 mL). The combined organic phases were washed with saturated brine, dried over anhydrous sodium sulfate and filtered. And concentrating the filtrate under reduced pressure to obtain a crude product. Purification by column chromatography (silica gel, EA/PE 0-100%) gave the title compound (40.0mg, 54.3% yield, white solid). LC-MS (ESI) M/z 562.0[ M + H ]]+1H NMR(500MHz,DMSO-d6)δ10.68(s,1H),7.90-7.82(m,4H),7.66(d,J=8.3Hz,2H),7.63-7.58(m,1H),7.53-7.47(m,2H),7.40-7.36(m,1H),5.14(t,J=5.2Hz,1H),4.12-4.06(m,1H),4.00-3.95(m,1H),3.71-3.65(m,1H),3.32(s,3H),3.27(q,J=7.3Hz,2H),1.10(t,J=7.3Hz,3H)。
Examples 11 and 12: (R) -N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-yl) -2- (4- (ethylsulfonyl) phenyl) -3-hydroxypropionamide or (S) -N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-yl) -2- (4- (ethylsulfonyl) phenyl) -3-hydroxypropionamide
Figure BDA0003065598540000352
Racemic N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl)]-4-yl) -2- (4- (ethylsulfonyl) phenyl) -3-hydroxypropionamide (50mg in about 12mL methanol, injection volume 4.0mL) was purified by passing Waters SFC 150 (room temperature, 100bar,214nm) and 250 × 25mm 10 μm Dr
Figure BDA0003065598540000353
OD) (supercritical carbon dioxide: methanol, 65:35,3.0min,70mL/min) to obtain (R) -N- (2, 6-dichloro-methane)-2'- (trifluoromethoxy) - [1,1' -biphenyl]-4-yl) -2- (4- (ethylsulfonyl) phenyl) -3-hydroxypropionamide or (S) -N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl]-4-yl) -2- (4- (ethylsulfonyl) phenyl) -3-hydroxypropionamide (19.0mg, white solid, ret. time ═ 1.925min, e.e.98%). LC-MS (ESI) M/z 561.9[ M + H ]]+/563.9[M+2+H]+1H NMR(400MHz,CDCl3)δ7.96(s,1H),7.88(d,J=8.3Hz,2H),7.71–7.67(m,2H),7.57(d,J=8.3Hz,2H),7.51–7.45(m,1H),7.40–7.35(m,2H),7.26–7.25,7.24–7.23(m,1H),4.26–4.16(m,1H),4.06–3.97(m,1H),3.97–3.90(m,1H),3.14(q,J=7.4Hz,2H),2.73(t,J=6.0Hz,1H),1.31(t,J=7.4Hz,3H)。
(S) -N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl)]-4-yl) -2- (4- (ethylsulfonyl) phenyl) -3-hydroxypropionamide or (R) -N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl]-4-yl) -2- (4- (ethylsulfonyl) phenyl) -3-hydroxypropionamide (22.3mg, white solid, ret. time. 2.453min, e.e.99%). LC-MS (ESI) M/z 561.9[ M + H ]]+/563.9[M+2+H]+1H NMR(400MHz,CDCl3)δ7.92(s,1H),7.90(d,J=8.3Hz,2H),7.70–7.66(m,2H),7.58(d,J=8.2Hz,2H),7.50–7.45(m,1H),7.40–7.35(m,2H),7.26–7.25,7.24-7.23(m,1H),4.26–4.16(m,2H),4.06–3.98(m,1H),3.96–3.90(m,1H),3.14(q,J=7.4Hz,2H),2.76–2.68(m,1H),1.31(t,J=7.4Hz,3H)。
Example 13: synthesis of N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-yl) -3-hydroxy-2- (4- (N-methylsulfonylamino) phenyl) propionamide
Figure BDA0003065598540000361
Step 1: synthesis of methyl 2- (4- (chlorosulfonyl) phenyl) acetate
Figure BDA0003065598540000362
Methyl 2- (4- (benzylthio) phenyl) acetate (500mg, 1.84mmol) was addedTo a mixed solvent of acetic acid (5mL) and water (1 mL). N-chlorosuccinimide (737mg, 5.52mmol) was added portionwise to the reaction mixture at 0 ℃. The reaction mixture was warmed to room temperature, and stirred at room temperature for 1 hour. EA (10mL) and water (5mL) were added. The organic phase was separated, washed with saturated brine, dried over anhydrous sodium sulfate, and filtered. And concentrating the filtrate under reduced pressure to obtain a crude product. Purification by column chromatography (silica gel, EA/PE 0-30%) gave the title compound (380mg, yield 83.0%, colorless oil).1H NMR(400MHz,CDCl3)δ8.04–7.97(m,2H),7.55(d,J=8.5Hz,2H),3.76(s,2H),3.74(s,3H)。
Step 2: synthesis of methyl 2- (4- (N-methylsulfonylamino) phenyl) acetate
Figure BDA0003065598540000363
Methyl 2- (4- (chlorosulfonyl) phenyl) acetate (380mg, 1.53mmol) and methylamine hydrochloride (124mg, 1.84mmol) were added to DCM (8 mL). After the reaction mixture was cooled to 0 ℃ under argon, triethylamine (0.638mL, 4.59mmol) was added. The reaction mixture was warmed to room temperature and stirred at room temperature overnight. Water (5mL) was added. The aqueous phase was separated and extracted with DCM (10 mL). The combined organic phases were washed with saturated brine, dried over anhydrous sodium sulfate and filtered. And concentrating the filtrate under reduced pressure to obtain a crude product. Purification by column chromatography (silica gel, EA/(DCM/PE 1:2) ═ 0-50%) gave the title compound (230mg, yield 61.8%, white solid). LC-MS (ESI) M/z 244.1[ M + H ]]+1H NMR(400MHz,DMSO-d6)δ7.76–7.69(m,2H),7.50(d,J=8.3Hz,2H),7.43(q,J=5.0Hz,1H),3.82(s,2H),3.63(s,3H),2.41(d,J=5.0Hz,3H)。
And step 3: synthesis of 2- (4- (N-methylsulfonamido) phenyl) acetic acid
Figure BDA0003065598540000364
Methyl 2- (4- (N-Methanesulfonamido) phenyl) acetate (230mg, 0.945mmol) was added to tetrakisTetrahydrofuran (4 mL). To the mixture was added a solution of lithium hydroxide monohydrate (79.3mg, 1.89mmol) in water (1 mL). After the reaction mixture was stirred at room temperature for 2 hours, dilute hydrochloric acid (1N, 2mL) was added. DCM (10mL) and water (5mL) were added. The organic phase was separated, washed with saturated brine, dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure to give the objective compound (156mg, yield 71.2%, colorless oil). LC-MS (ESI) M/z 230.1[ M + H ]]+
And 4, step 4: synthesis of N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-yl) -2- (4- (N-methylsulfonylamino) phenyl) acetamide
Figure BDA0003065598540000371
2- (4- (N-Methanesulfonamido) phenyl) acetic acid (150mg, 0.654mmol), 2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl]-4-amine (232mg, 0.719mmol), HATU (298mg, 0.785mmol) and DIEA (0.324mL, 1.96mmol) were added sequentially to DCM (8 mL). The reaction mixture was stirred at room temperature overnight. Saturated aqueous sodium bicarbonate (10mL) and DCM (10mL) were added. The organic phase was separated, washed with saturated brine, dried over anhydrous sodium sulfate, and filtered. And concentrating the filtrate under reduced pressure to obtain a crude product. Purification by column chromatography (silica gel, EA/PE 0-50%) gave the title compound (320mg, 91.7% yield, colorless oil). LC-MS (ESI) M/z 533.0[ M + H ]]+
And 5: synthesis of N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-yl) -3-hydroxy-2- (4- (N-methylsulfonylamino) phenyl) propionamide
Figure BDA0003065598540000372
Mixing N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl)]-4-yl) -2- (4- (N-methylsulfonylamino) phenyl) acetamide (320mg, 0.600mmol) was added to anhydrous DMSO (3 mL). To the reaction mixture was added paraformaldehyde (16.2mg, 0.540mmol) and sodium methoxide (1.29mg, 0.024mmol) in that order. The reaction mixture was at room temperatureThe reaction was stirred for 2 hours. EA (15mL) and water (10mL) were added. The aqueous phase was separated and extracted with EA (10 mL). The combined organic phases were washed with saturated brine, dried over anhydrous sodium sulfate and filtered. And concentrating the filtrate under reduced pressure to obtain a crude product. Purification by column chromatography (C18, methanol/water 0-100%) gave the title compound (139mg, yield 41.1%, white solid). LC-MS (ESI) M/z 563.0[ M + H ]]+1H NMR(400MHz,DMSO-d6)δ10.69(s,1H),7.91–7.83(m,2H),7.77(d,J=8.3Hz,2H),7.65–7.57(m,3H),7.55–7.49(m,2H),7.46(q,J=5.0Hz,1H),7.42–7.37(m,1H),5.15(t,J=5.2Hz,1H),4.13–4.04(m,1H),3.99–3.92(m,1H),3.69–3.63(m,1H),2.41(d,J=5.0Hz,3H)。
Example 14: n- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-yl) -3-methoxy-2- (4- (N-methylsulfonylamino) phenyl) propanamide
Figure BDA0003065598540000381
Mixing N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl)]-4-yl) -3-hydroxy-2- (4- (N-methylsulfonylamino) phenyl) propionamide (89mg, 0.158mmol) was added to DCM (3 mL). After the reaction mixture was cooled to 0 ℃, fluoroboric acid (28.9mg, 0.158mmol, 48% aqueous solution) and trimethylsilyl diazomethane (2N, 0.079mL, 0.158mmol) were added sequentially under the exclusion of light. After the reaction mixture was stirred at 0 ℃ for 20 minutes, trimethylsilyldiazomethane (2N, 0.079mL, 0.158mmol) was added again and the reaction mixture was stirred at 0 ℃ for an additional 20 minutes. This step was repeated two more times. Water (10mL) was added to the reaction mixture, and the mixture was stirred at room temperature for 10 minutes. DCM (15mL) was added. The aqueous phase was separated and extracted with DCM (10 mL). The combined organic phases were washed with saturated brine, dried over anhydrous sodium sulfate and filtered. And concentrating the filtrate under reduced pressure to obtain a crude product. Purification by preparative plate separation (silica gel, EA/PE ═ 1:1) afforded the title compound (29mg, yield 31.9%, white solid). LC-MS (ESI) m/z:577.0[ M + H]+1H NMR(400MHz,DMSO-d6)δ10.72(s,1H),7.87–7.81(m,2H),7.77(d,J=8.3Hz,2H),7.63(d,J=8.3Hz,2H),7.61–7.57(m,1H),7.54–7.48(m,2H),7.46(q,J=4.9Hz,1H),7.41–7.36(m,1H),4.14–4.07(m,1H),4.00(t,J=9.1Hz,1H),3.65–3.57(m,1H),3.29(s,3H),2.41(d,J=5.1Hz,3H)。
Example 15: process for preparation of 3-amino-N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-yl) -2- (4- (methylsulfonyl) phenyl) propanamide
Figure BDA0003065598540000382
Step 1: synthesis of methyl 2- (4-methylsulfonyl) phenylacetate
Figure BDA0003065598540000383
2- (4-Methylsulfonyl) phenylacetic acid (5.00g,23.3mmol) was dissolved in methanol (100mL) and thionyl chloride (8.33g,70.0mmol) was added under ice-water bath conditions. The reaction mixture was warmed to room temperature and stirred for 1 hour, and the reaction was stopped. The reaction mixture was concentrated under reduced pressure, and the resulting residue was redissolved in EA (80.0mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The obtained residue was separated and purified by column chromatography (PE: EA ═ 3:1-2:1) to obtain the objective compound (5.00g, yield 94.0%, off-white solid).
Step 2: synthesis of methyl 3- (1, 3-dioxoisoindolin-2-yl) -2- (4- (methylsulfonyl) phenyl) propionate
Figure BDA0003065598540000384
Methyl 2- (4- (methylsulfonyl) phenyl) acetate (2.00g,8.76mmol) was dissolved in ultra-dry tetrahydrofuran (60.0mL), lithium bistrimethylsilylamide (10.5mL,10.5mmol) was slowly added dropwise to the reaction system at-78 ℃ under nitrogen protection, after the reaction mixture was stirred at-78 ℃ for 30 minutes, 2- (bromomethyl) isoindoline-1, 3-dione (2.30g,9.58mmol) (dissolved in ultra-dry tetrahydrofuran) was slowly added dropwise to the reaction system, and after the reaction mixture was stirred at-78 ℃ for 1.5 hours, the reaction was stopped. Saturated ammonium chloride is used for reaction mixture(20.0mL) of the aqueous solution, EA (30.0 mL. times.2) of the extract, washing the organic phase with saturated brine (15.0 mL. times.2), drying over anhydrous sodium sulfate, filtering, and concentrating the filtrate under reduced pressure to obtain the objective compound (3.00g, crude product, gray solid). LC-MS (ESI) M/z 388.0[ M + H ]]+
And step 3: synthesis of 3- (1, 3-dioxoisoindolin-2-yl) -2- (4- (methylsulfonyl) phenyl) propanoic acid
Figure BDA0003065598540000391
To a solution of methyl 3- (1, 3-dioxoisoindolin-2-yl) -2- (4- (methylsulfonyl) phenyl) propionate (3.00g,7.74mmol) in 1, 4-dioxane (20.0mL) was added aqueous hydrochloric acid (6.0N) (20.0 mL). After the reaction mixture was stirred at 80 ℃ for 16 hours, the reaction was stopped. After the reaction mixture was cooled to room temperature, water (20.0mL), EA (20.0 mL. times.2) was added to the mixture to extract, and the organic phase was washed with saturated brine (10.0 mL. times.2), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give the objective compound (3.13g, 95.7% yield in two steps, white solid). LC-MS (ESI) M/z 374.4[ M + H ]]+
Step 4 Synthesis of N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-yl) -3- (1, 3-dioxoisoindolin-2-yl) -2- (4- (methylsulfonyl) phenyl) propionamide
Figure BDA0003065598540000392
At room temperature, 2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl]-4-amine (1.50g,4.66mmol) and 3- (1, 3-dioxoisoindolin-2-yl) -2- (4- (methylsulfonyl) phenyl) propanoic acid (3.13g,8.38mmol) were dissolved in acetonitrile (75.0mL) and then N-methylimidazole (1.91g,23.3mmol) and TCFH (3.92g,14.0mmol) were added in sequence and the reaction was stopped after the reaction solution was stirred for 2 hours at room temperature. Concentrating the reaction solution under reduced pressure, extracting the residue with EA (30.0mL × 2), washing with saturated saline (25.0mL × 2), drying with anhydrous sodium sulfate, filtering, and concentrating under reduced pressure to obtain residue, and separating and purifying by column chromatography(PE: EA ═ 10:1-2:1) to obtain the title compound (4.00g, yield 70.4%, white solid).1H NMR(400MHz,DMSO-d6)δ10.78(s,1H),7.91(d,J=8.3Hz,2H),7.87–7.81(m,4H),7.80–7.78(m,1H),7.76–7.74(m,1H),7.68(d,J=8.3Hz,2H),7.63–7.57(m,1H),7.53–7.47(m,2H),7.39–7.35(m,1H),4.50–4.43(m,1H),4.30–4.23(m,1H),4.14–4.07(m,1H),3.18(s,3H)。
Step 5 Synthesis of 3-amino-N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-yl) -2- (4- (methylsulfonyl) phenyl) propanamide
Figure BDA0003065598540000401
To produce N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl)]To a solution of (4-yl) -3- (1, 3-dioxoisoindolin-2-yl) -2- (4- (methylsulfonyl) phenyl) propanamide (100mg,0.148mmol) in absolute ethanol (2.00mL) was added hydrazine hydrate (98% aqueous solution, 1.00mL), and the reaction mixture was stirred at 80 ℃ for 16 hours, and then the reaction was stopped. After the reaction solution was cooled to room temperature, it was concentrated under reduced pressure, and the obtained residue was purified by preparative (acetonitrile/water containing 0.05% formic acid) to obtain the objective compound (40.8mg, yield 50.5%, white solid). LC-MS (ESI) M/z 547.0[ M + H ]]+/549.0[M+2+H]+1H NMR(400MHz,CD3OD)δ7.97(d,J=8.4Hz,2H),7.83(d,J=2.0Hz,1H),7.76(d,J=2.1Hz,1H),7.68(d,J=8.4Hz,2H),7.56–7.50(m,1H),7.45–7.38(m,2H),7.30–7.27(m,1H),3.97–3.91(m,1H),3.44–3.36(m,1H),3.11(s,3H),3.05–2.98(m,1H)。
Example 16: 3-acetylamino-N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-yl) -2- (4- (methylsulfonyl) phenyl) propanamide
Figure BDA0003065598540000402
3-amino-N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl)]-4-yl) -2- (4- (methylsulfonyl) phenyl) propanamide (100mg,0.183mmol) and triethylamine (46.2mg,0.457mmol) in solutionIn DCM (4.00mL), acetyl chloride (14.4mg,0.183mmol) was slowly added to the mixture under ice bath. The reaction mixture was stirred at room temperature for 1 hour, and then the reaction was stopped. The reaction mixture was poured into water (10.0mL), extracted with DCM (10.0mL × 2), washed with saturated brine (10.0mL × 2), dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the resulting residue was prepared (acetonitrile/water containing 0.05% formic acid) to give the title compound (44.3mg, yield 41.2%, yellow solid). LC-MS (ESI) M/z 588.9[ M + H ]]+/590.9[M+2+H]+1H NMR(400MHz,CD3OD)δ10.37(s,CONH),8.30(t,J=6.4Hz,CH3CONH),7.97(d,J=8.4Hz,2H),7.84–7.68(m,4H),7.56–7.50(m,1H),7.46–7.37(m,2H),7.31–7.26(m,1H),4.42–4.36,4.17–4.07(m,1H),4.02–3.93,3.81–3.71(m,1H),3.68–3.59,3.50–3.46(m,1H),3.11(s,3H),2.14,2.07,1.91(s,3H)。
Example 17: n- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-yl) -3- (methylamino) -2- (4- (methylsulfonyl) phenyl) propionamide formate salt
Figure BDA0003065598540000403
Step 1 Synthesis of 3- ((2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-yl) amino) -2- (4- (methylsulfonyl) phenyl) -3-oxopropyl methanesulfonate
Figure BDA0003065598540000411
Mixing N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl)]-4-yl) -3-hydroxy-2- (4- (methylsulfonyl) phenyl) propionamide (200mg,0.364mmol) and triethylamine (111mg,1.09mmol) were dissolved in DCM (4.00mL) and methanesulfonyl chloride (50.2mg,0.438mmol) was added to the mixture under ice bath. The reaction mixture was stirred at room temperature for 1 hour, and then the reaction was stopped. The reaction mixture was poured into water (10.0mL), extracted with DCM (10.0 mL. times.2), washed with saturated brine (10.0 mL. times.2), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give the title compound (200mg, two steps)Yield 87.6%, white solid). LC-MS (ESI) M/z 625.9[ M + H ]]+/627.9[M+2+H]+
Step 2 Synthesis of N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-yl) -3- (methylamino) -2- (4- (methylsulfonyl) phenyl) propionamide formate salt
Figure BDA0003065598540000412
Methanesulfonic acid 3- ((2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl)]-4-yl) amino) -2- (4- (methylsulfonyl) phenyl) -3-oxopropyl ester (90.0mg,0.144mmol) and triethylamine (29.1mg,0.288mmol) were dissolved in DCM (5.00mL) and methylamine hydrochloride (11.6mg,0.172mmol) was added to the mixture at room temperature. The reaction mixture was stirred at room temperature for 2 hours, and then the reaction was stopped. The reaction mixture was concentrated under reduced pressure, and the resulting residue was purified by preparative chromatography (acetonitrile/water containing 0.05% formic acid) to give the objective compound (20.3mg, yield 23.3%, formate salt, white solid). LC-MS (ESI) M/z 560.9[ M + H ]]+/562.9[M+2+H]+1H NMR(400MHz,CD3OD)δ8.48(s,1H),8.03(d,J=8.3Hz,2H),7.85(d,J=2.0Hz,1H),7.77–7.69(m,3H),7.57–7.50(m,1H),7.46–7.37(m,2H),7.28(dd,J=7.6,1.6Hz,1H),4.31–4.25(m,1H),3.77–3.69(m,1H),3.29–3.27(m,1H),3.13(s,3H),2.72(s,3H)。
Example 18: 3- (azetidin-1-yl) -N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-yl) -2- (4- (methylsulfonyl) phenyl) propanamide
Figure BDA0003065598540000413
Methanesulfonic acid 3- ((2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl)]-4-yl) amino) -2- (4- (methylsulfonyl) phenyl) -3-oxopropyl ester (110mg,0.176mmol) and triethylamine (35.5mg,0.351mmol) were dissolved in DCM (5.00mL) and azetidine (12.0mg,0.210mmol) was added slowly to the mixture while cooling on ice. The reaction mixture was stirred at room temperature for 2 hours, and then the reaction was stopped. Will be reversedThe mixture was concentrated under reduced pressure, and the resulting residue was purified by preparative chromatography (acetonitrile/water containing 0.05% formic acid) to give the title compound (25.9mg, yield 25.1%, white solid). LC-MS (ESI) M/z 587.0[ M + H ]]+/589.0[M+2+H]+1H NMR(400MHz,MeOD)δ7.99(d,J=8.3Hz,2H),7.83(d,J=2.0Hz,1H),7.73(d,J=2.0Hz,1H),7.71(d,J=8.3Hz,2H),7.56–7.51(m,1H),7.46–7.38(m,2H),7.28(dd,J=7.6,1.6Hz,1H),4.03–3.97(m,1H),3.80–3.64(m,5H),3.22–3.16(m,1H),3.12(s,3H),2.33–2.25(m,2H).
Example 19: n- (2-chloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-yl) -3-hydroxy-2- (4- (methylsulfonyl) phenyl) propanamide
Figure BDA0003065598540000421
Step 1 Synthesis of 2-chloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-amine
Figure BDA0003065598540000422
To the microwave tube were added 2- (trifluoromethoxy) phenylboronic acid (1.00g,4.86mmol), 4-bromo-3-chloroaniline (1.10g,5.33mmol), potassium carbonate (1.00g,7.23mmol), Pd (dppf) Cl2(396mg,0.460mmol) and acetonitrile/water (8.00mL/2.00 mL). After the addition, the nitrogen gas was replaced for 3 minutes, and the reaction mixture was stirred at 90 ℃ for 6 hours, and then the reaction was stopped. The reaction mixture was concentrated, extracted with EA (30.0mL × 3), the organic phase was washed with saturated brine (30.0mL × 2), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the resulting crude product was separated and purified by column chromatography (PE: EA ═ 5:1) to give the objective compound (1.20g, yield 85.7%, black solid). LC-MS (ESI) m/z: 287.8[ M + H]+1H NMR(400MHz,DMSO-d6)δ7.51–7.38(m,3H),7.37–7.32(m,1H),6.95(d,J=8.3Hz,1H),6.71(d,J=2.2Hz,1H),6.59–6.53(m,1H),5.53(s,2H)。
Step 2 Synthesis of N- (2-chloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-yl) -2- (4- (methylsulfonyl) phenyl) acetamide
Figure BDA0003065598540000423
To 2-chloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl]N-methylimidazole (285mg,3.48mmol) and TCFH (585mg,2.09mmol) were added to a solution of (E) -4-amine (200mg,0.695mmol) and (4- (methylsulfonyl) phenyl) acetic acid (223mg,1.04mmol) in acetonitrile (5.00mL), and the reaction mixture was stirred at room temperature for 16 hours, whereupon the reaction was stopped. The reaction mixture was concentrated under reduced pressure, extracted with EA (30.0 mL. times.3), and the organic phase was washed with saturated brine (30.0 mL. times.2), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product was isolated and purified by column chromatography (PE: EA ═ 1:1-2:3) to give the title compound (150mg, yield 44.6%, grey solid). LC-MS (ESI) M/z 525.1[ M + ACN + H ]]+
Step 3 Synthesis of N- (2-chloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-yl) -3-hydroxy-2- (4- (methylsulfonyl) phenyl) propanamide
Figure BDA0003065598540000431
To N- (2-chloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl)]A solution of (4-yl) -2- (4- (methylsulfonyl) phenyl) acetamide (100mg,0.207mmol) in anhydrous DMSO (2.00mL) was added paraformaldehyde (4.00mg,0.145mmol), followed by sodium methoxide (0.500mg,0.008mmol), and after addition, the reaction mixture was stirred at room temperature for 2 hours, and LC-MS showed completion of the reaction. The reaction solution was purified by high pressure liquid preparative chromatography (acetonitrile/water containing 0.05% formic acid) to give the objective compound (40.0mg, yield 37.7%, white solid). LC-MS (ESI) M/z 514.2[ M + H ]]+1H NMR(400MHz,DMSO-d6)δ10.57(s,1H),7.99(s,J=2.0Hz,1H),7.92(d,J=8.4Hz,2H),7.66(d,J=8.4Hz,2H),7.59–7.53(m,2H),7.50–7.45(m,2H),7.41–7.38(m,J=7.8,1.7Hz,1H),7.30(d,J=8.4Hz,1H),5.14(s,1H),4.10(t,J=9.3Hz,1H),4.03–3.97(m,J=8.6,5.6Hz,1H),3.70–3.63(m,J=9.8,5.5Hz,1H),3.20(s,3H)。
Example 20: n- (2-trifluoromethyl-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-yl) -3-hydroxy-2- (4- (methylsulfonyl) phenyl) propanamide
Figure BDA0003065598540000432
Step 1 Synthesis of 2-trifluoromethyl-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-amine
Figure BDA0003065598540000433
Into a microwave tube were placed 4-bromo-3-trifluoromethylaniline (500mg,2.08mmol), 2- (trifluoromethoxy) phenylboronic acid (515mg,2.50mmol), potassium carbonate (576mg,4.17mmol), Pd (dppf) Cl in that order2(171mg,0.208mmol) and 1, 4-dioxane/water (8.00mL/2.00 mL). After the nitrogen gas was replaced for 3 minutes, the reaction mixture was stirred at 100 ℃ for 6 hours, and then the reaction was stopped. The reaction mixture was concentrated, extracted with EA (30.0mL × 3), the organic phase was washed with saturated brine (30.0mL × 2), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the resulting crude product was separated and purified by column chromatography (PE: EA ═ 9:1 to 5:1) to give the target compound (600mg, yield 89.6%, gray solid). LC-MS (ESI) M/z 321.9[ M + H ]]+1H NMR(400MHz,CDCl3)δ7.43–7.37(m,1H),7.33–7.27(m,3H),7.11–7.04(m,2H),6.93–6.87(m,1H),4.59(s,2H)。
Step 2 Synthesis of 2- (4- (methylsulfonyl) phenyl) -N- (2'- (trifluoromethoxy) -2- (trifluoromethyl) - [1,1' -biphenyl ] -4-yl) acetamide
Figure BDA0003065598540000434
To 2-trifluoromethyl-2 '- (trifluoromethoxy) - [1,1' -biphenyl]N-methylimidazole (255mg,3.12mmol) and TCFH (524mg,1.87mmol) were added to a solution of (E) -4-amine (200mg,0.623mmol) and (2- (4- (methylsulfonyl) phenyl) acetic acid (200mg,0.934mmol) in acetonitrile (5.00mL), and the reaction mixture was stirred at room temperature for 14 hours, whereupon the reaction was stopped. After the reaction liquid is concentrated,the mixture was extracted with EA (30.0 mL. times.3), and the organic phase was washed with saturated brine (30.0 mL. times.2), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product was isolated and purified by column chromatography (PE: EA ═ 3:2-2:3) to give the title compound (180mg, yield 55.9%, grey solid). LC-MS (ESI) M/z 518.1[ M + H ]]+1H NMR(400MHz,CDCl3)δ7.96–7.86(m,3H),7.80(m,1H),7.68(s,1H),7.56(d,J=8.1Hz,2H),7.46–7.40(m,1H),7.35–7.24(m,4H),3.85,3.83(s,2H),3.07,3.06(s,3H)。
Step 3 Synthesis of N- (2-trifluoromethyl-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-yl) -3-hydroxy-2- (4- (methylsulfonyl) phenyl) propanamide
Figure BDA0003065598540000441
To the reaction solution of 2- (4- (methylsulfonyl) phenyl) -N- (2'- (trifluoromethoxy) -2- (trifluoromethyl) - [1,1' -biphenyl]Polyformaldehyde (4.78mg,0.159mmol) and sodium methoxide (0.430mg,0.008mmol) were added to a DMSO (2.00mL) solution of (100mg,0.199mmol) and the reaction mixture was stirred at room temperature for 2 hours, and the reaction was stopped. The reaction solution was purified by high pressure liquid phase (acetonitrile/water containing 0.05% formic acid) to obtain the objective compound (60.0mg, yield 37.7%, white solid). LC-MS (ESI) M/z 548.1[ M + H ]]+1H NMR(400MHz,DMSO-d6)δ10.71(s,1H),8.29–8.19(m,1H),7.92(d,J=8.3Hz,2H),7.89–7.82(m,1H),7.67(d,J=8.3Hz,2H),7.61–7.52(m,1H),7.49–7.40(m,2H),7.40–7.27(m,2H),5.20–5.10(m,1H),4.24–3.92(m,2H),3.79–3.58(m,1H),3.20(s,3H)。
Example 21: n- (2, 6-dimethyl-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-yl) -3-hydroxy-2- (4- (methylsulfonyl) phenyl) propanamide
Figure BDA0003065598540000442
Step 1: synthesis of N- (2, 6-dimethyl-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-yl) -2- (4- (methylsulfonyl) phenyl) acetamide
Figure BDA0003065598540000443
2, 6-dimethyl-2 '- (trifluoromethoxy) - [1,1' -biphenyl]-4-amine (150mg,0.53mmol), 2- (4- (methylsulfonyl) phenyl) acetic acid (137mg,0.64mmol), HATU (243mg,0.64mmol) and DIEA (137mg,1.06mmol) were added to DCM (3mL) and reacted at room temperature for 2 hours, after which the reaction was completed and silica gel column isolated (PE: DCM ═ 1:1) to give the desired product (250mg, yield 99.2%, white solid).1H NMR(400MHz,CDCl3)δ7.94(d,J=8.3Hz,2H),7.57(d,J=8.3Hz,2H),7.44(s,1H),7.43–7.31(m,3H),7.27–7.25(m,2H),7.16–7.11(m,1H),3.81(s,2H),3.06(s,3H),1.97(s,6H).
Step 2: synthesis of N- (2, 6-dimethyl-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-yl) -3-hydroxy-2- (4- (methylsulfonyl) phenyl) propanamide
Figure BDA0003065598540000451
The preparation method comprises the steps of dissolving N- (2, 6-dimethyl-2 '- (trifluoromethoxy) - [1,1' -biphenyl)]-4-yl) -2- (4- (methylsulfonyl) phenyl) acetamide (100mg,0.21mmol) and paraformaldehyde (5.98mg,0.20mmol) were added to a solution of anhydrous DMSO (2mL), sodium methoxide (0.43mg,0.008mmol) was added with stirring, the reaction was carried out at room temperature for 2 hours, after completion of the reaction, water (20mL) was added, EA (20mLx3) was extracted, the organic layers were combined, washed with water 3 times, the solvent was spin-dried, and the column was reversed-phase separated (water: methanol 100% -75%) to give the desired product (26mg, 24.3% yield, white solid). LC-MS (ESI) M/z 508.0[ M + H ]]+.1H NMR(400MHz,DMSO-d6)δ10.19(s,1H),7.93–7.88(m,2H),7.66(d,J=8.4Hz,2H),7.60–7.44(m,3H),7.43–7.36(m,2H),7.26(dd,J=7.8,1.8Hz,1H),5.14,5.07(t,J=5.2Hz,1H),4.22–4.05(m,1H),4.04–3.95(m,1H),3.70–3.63(m,1H),3.21,3.20(s,2H),1.90(s,6H).
Example 22: n- (2, 6-dichloro-4 '- (1,1,1,3,3, 3-hexafluoro-2-hydroxypropan-2-yl) - [1,1' -biphenyl ] -4-yl) -3-hydroxy-2- (4- (methylsulfonyl) phenyl) propanamide
Figure BDA0003065598540000452
Step 1: synthesis of 2- (4-bromophenyl) -1,1,1,3,3, 3-hexafluoropropane-2-ol
Figure BDA0003065598540000453
Cupric bromide (517mg, 2.32mmol) was dissolved in acetonitrile (5mL), and tert-butyl nitrite (0.347mL, 2.89mmol) was added dropwise to the reaction solution under ice bath and nitrogen atmosphere. After the addition was completed, 2- (4-aminophenyl) -1,1,1,3,3, 3-hexafluoropropane-2-ol (500mg, 1.93mmol) was dissolved in acetonitrile (3mL) and added dropwise to the reaction solution, and after the addition was completed, the reaction mixture was stirred for further 1 hour under ice bath, and the completion of the reaction was monitored by LC-MS. The reaction mixture was poured into ice water (20mL), extracted with methyl tert-butyl ether (15mLx2), and the organic phases were combined, washed with saturated brine (50mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give the title compound (694mg, crude, brown oil). LC-MS (ESI) M/z 323.0[ M-H ]]-
Step 2: synthesis of 1,1,1,3,3, 3-hexafluoro-2- (4- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) phenyl) propan-2-ol
Figure BDA0003065598540000461
2- (4-bromophenyl) -1,1,1,3,3, 3-hexafluoropropan-2-ol (694mg, 2.15mmol), pinacol diboron (600mg, 2.36mmol), potassium acetate (633mg, 6.45mmol) and Pd (dppf) Cl2(157mg, 0.215mmol) was added to 1, 4-dioxane (10 mL). The reaction mixture was stirred at 90 ℃ overnight under nitrogen. The reaction mixture was cooled to room temperature, filtered, and the filtrate was concentrated under reduced pressure to give the title compound (800mg, crude, white solid).
And step 3: synthesis of 2- (4 '-amino-2', 6 '-dichloro- [1,1' -biphenyl ] -4-yl) -1,1,1,3,3, 3-hexafluoropropan-2-ol
Figure BDA0003065598540000462
1,1,1,3,3, 3-hexafluoro-2- (4- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) phenyl) propan-2-ol (800mg, 2.16mmol), 4-bromo-3, 5-dichloroaniline (417mg, 1.73mmol), potassium carbonate (597mg, 4.32mmol) and PdCl2(dtbpf) (141mg, 0.216mmol) was added to a mixed solvent of 1, 4-dioxane (10mL) and water (1 mL). The reaction mixture was stirred at 100 ℃ for 2 hours under nitrogen, and LC-MS monitored the reaction was complete. The reaction mixture was cooled to room temperature, filtered, and the filtrate was concentrated under reduced pressure to give a crude product, which was purified by silica gel column separation (PE: EA ═ 10:1-2:1) to give the target compound (248mg, 31.9% yield in three steps, light brown solid). LC-MS (ESI) M/z 445.0[ M + ACN + H]+
And 4, step 4: synthesis of N- (2, 6-dichloro-4 '- (1,1,1,3,3, 3-hexafluoro-2-hydroxypropan-2-yl) - [1,1' -biphenyl ] -4-yl) -2- (4- (methylsulfonyl) phenyl) acetamide
Figure BDA0003065598540000463
2- (4 '-amino-2', 6 '-dichloro- [1,1' -biphenyl)]-4-yl) -1,1,1,3,3, 3-hexafluoropropan-2-ol (82.0mg, 0.203mmol) and 2- (4- (methylsulfonyl) phenyl) acetic acid (43.5mg, 0.203mmol) were dissolved in anhydrous DCM (3 mL). HATU (116mg, 0.304mmol) and DIEA (0.101mL, 0.609mmol) were added to the reaction mixture at room temperature in that order. After the addition was complete, the reaction mixture was stirred at room temperature overnight. The reaction mixture was poured into water (20mL), extracted with DCM (15mLx2), the organic phases were combined, washed successively with saturated ammonium chloride (20mL) and saturated sodium bicarbonate (20mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give a crude product, which was separated and purified by silica gel column (PE: EA ═ 4:1-2:1) to give the desired compound (110.0mg, yield 90.3%, yellow solid). LC-MS (ESI) M/z 598.0[ M-H ]]-
And 5: synthesis of N- (2, 6-dichloro-4 '- (1,1,1,3,3, 3-hexafluoro-2-hydroxypropan-2-yl) - [1,1' -biphenyl ] -4-yl) -3-hydroxy-2- (4- (methylsulfonyl) phenyl) propionamide
Figure BDA0003065598540000471
Mixing N- (2, 6-dichloro-4 '- (1,1,1,3,3, 3-hexafluoro-2-hydroxypropane-2-yl) - [1,1' -biphenyl ]]-4-yl) -2- (4- (methylsulfonyl) phenyl) acetamide (110mg,0.183mmol) was dissolved in anhydrous DMSO (2 mL). Paraformaldehyde (15.7mg,0.174mmol) and sodium methoxide (0.394mg,0.0073mmol) were sequentially added to the reaction solution at room temperature, and after the addition was completed, the reaction mixture was stirred at room temperature overnight. The reaction solution was poured into ice water (25mL), the mixture was neutralized with 1.0M diluted hydrochloric acid solution, extracted with EA (15mLx3), the organic phases were combined, washed with saturated brine (30mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give a crude product, which was purified by silica gel separation (PE: EA ═ 4:1-0:1) to give the objective compound (16.7mg, yield 14.5%, white solid). LC-MS (ESI) M/z 629.9[ M + H ]]+.
1H NMR(400MHz,CD3OD)δ7.95(d,J=8.4Hz,2H),7.83–7.78(m,4H),7.70(d,J=8.4Hz,2H),7.33(d,J=8.5Hz,2H),4.62–4.57(m,1H),4.27–4.21(m,1H),4.01–3.96(m,1H),3.83–3.76(m,1H),3.11(s,3H).
Example 23: n- (2' -fluoro-4 ' - (1,1,1,3,3, 3-hexafluoro-2-hydroxypropan-2-yl) - [1,1' -biphenyl ] -4-yl) -3-hydroxy-2- (4- (methylsulfonyl) phenyl) propanamide
Figure BDA0003065598540000472
Step 1: synthesis of 2- (4-bromo-3-fluorophenyl) -1,1,1,3,3, 3-hexafluoropropan-2-ol
Figure BDA0003065598540000473
2- (4-amino-3-fluorophenyl) -1,1,1,3,3, 3-hexafluoropropan-2-ol (277mg,1mmol), copper bromide (349.5mg,1.5mmol) and acetonitrile (5mL) are sequentially added into a 25mL single-neck flask, tert-butyl nitrite (155mg,1.5mmol) is slowly dripped into the reaction solution for reaction at room temperature overnight, TLC raw material reaction is completed, water (20mL) and EA (20mLx2) are added for extraction after the reaction is completed, saturated sodium chloride is washed once, and the solvent is dried under reduced pressure to obtain a target product (340mg, yield 100%, orange oily substance).
Step 2: synthesis of 2- (4 '-amino-2-fluoro- [1,1' -biphenyl ] -4-yl) -1,1,1,3,3, 3-hexafluoropropan-2-ol
Figure BDA0003065598540000474
2- (4-bromo-3-fluorophenyl) -1,1,1,3,3, 3-hexafluoropropan-2-ol (341mg,1mmol), 4- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) aniline (263mg,1.2mmol), potassium carbonate (408mg,3mmol) and Pd (dppf) Cl2(37mg, 0.05mmol) was added to 1, 4-dioxane/water (5mL/1mL), the mixture was heated at 110 ℃ for 3 hours under nitrogen protection, TLC detected that the starting material was completely reacted, after completion of the reaction, the mixture was cooled to room temperature, water (20mL) was added, EA (20mLx2) was extracted, and the product was automatically separated by column chromatography on silica gel (PE: EA ═ 1:2) to give the present product (270mg, yield 76.5, white solid). LC-MS (ESI) M/z 354.0[ M + H ]]+
And step 3: synthesis of N- (2' -fluoro-4 ' - (1,1,1,3,3, 3-hexafluoro-2-hydroxypropan-2-yl) - [1,1' -biphenyl ] -4-yl) -2- (4- (methylsulfonyl) phenyl) acetamide
Figure BDA0003065598540000481
2- (4 '-amino-2-fluoro- [1,1' -biphenyl)]-4-yl) -1,1,1,3,3, 3-hexafluoropropan-2-ol (60.0mg, 0.170mmol) and 2- (4- (methylsulfonyl) phenyl) acetic acid (40.0mg, 0.187mmol) were dissolved in anhydrous DCM (3 mL). HATU (96.9mg, 0.255mmol) and DIEA (0.0842mL, 0.510mmol) were added to the reaction mixture at room temperature in that order. After the addition was complete, the reaction mixture was stirred at room temperature overnight. The reaction was poured into water (20mL), extracted with DCM (15mLx2), the organic phases combined, and saturated ammonium chloride (20mL) and saturated carbonic acid sequentiallySodium hydrogen (20mL) was washed, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give a crude product, which was purified by silica gel column separation (PE: EA ═ 4:1-2:1) to give the title compound (59.0mg, yield 63.2%, yellow solid). LC-MS (ESI) M/z 548.1[ M-H ]]-
And 4, step 4: synthesis of N- (2' -fluoro-4 ' - (1,1,1,3,3, 3-hexafluoro-2-hydroxypropan-2-yl) - [1,1' -biphenyl ] -4-yl) -3-hydroxy-2- (4- (methylsulfonyl) phenyl) propionamide
Figure BDA0003065598540000482
Mixing N- (2' -fluoro-4 ' - (1,1,1,3,3, 3-hexafluoro-2-hydroxypropane-2-yl) - [1,1' -biphenyl ]]Preparation of (4-yl) -2- (4- (methylsulfonyl) phenyl) acetamide (59.0mg,0.107mmol) was dissolved in anhydrous DMSO (2 mL). Paraformaldehyde (9.19mg,0.102mmol) and sodium methoxide (0.232mg,0.0043mmol) were sequentially added to the reaction solution at room temperature, and after the addition was completed, the reaction mixture was stirred at room temperature for reaction overnight. The reaction solution was poured into ice water (25mL), the mixture was neutralized with 1.0M diluted hydrochloric acid solution, extracted with EA (15mLx3), the organic phases were combined, washed with saturated brine (30mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give a crude product, which was purified by silica gel separation (PE: EA ═ 4:1-0:1) to give the objective compound (26.2mg, yield 42.1%, white solid). LC-MS (ESI) M/z 580.0[ M + H ]]+.1H NMR(400MHz,DMSO-d6)δ10.39(s,1H),7.88(d,J=8.1Hz,2H),7.71(d,J=8.4Hz,2H),7.68–7.60(m,3H),7.57–7.48(m,4H),5.11–5.05(m,1H),4.10–4.03(m,1H),4.00–3.95(m,1H),3.67–3.60(m,1H),3.16(s,3H).
Example 24: n- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-yl) -2- (4- (ethylsulfonyl) phenyl) -3-methoxypropionamide
Figure BDA0003065598540000491
Step 1: synthesis of methyl 2- (4-ethylsulfonyl) phenylacetate
Figure BDA0003065598540000492
2- (4-Methylsulfonyl) phenylacetic acid (25.0g,110mmol) was dissolved in methanol (150mL) and thionyl chloride (11.9mL,164mmol) was added under ice-water bath conditions. The reaction mixture was warmed to room temperature and stirred for 1 hour, and the reaction was stopped. The reaction solution was concentrated under reduced pressure, and the obtained residue was purified by column chromatography (PE: EA ═ 10:1 to 3:1) to obtain the objective compound (24.2g, yield 91.3%, white solid). MS (ESI) M/z 242.9[ M + H ]]+1H NMR(400MHz,CDCl3)δ7.87(d,J=8.3Hz,2H),7.49(d,J=8.4Hz,2H),3.78–3.69(m,5H),3.11(q,J=7.4Hz,2H),1.29(d,J=7.4Hz,3H).
Step 2: synthesis of methyl 2- (4-ethylsulfonyl) phenyl-3-hydroxypropionate
Figure BDA0003065598540000493
To methyl 2- (4-ethylsulfonyl) phenylacetate (24.2g,100mmol) and paraformaldehyde (2.85g,95.0mmol) in anhydrous DMSO (250mL) was added sodium methoxide (216mg,4.00mmol), and the reaction mixture was stirred at room temperature for 16 hours, whereupon the reaction was stopped. The reaction mixture was poured into ice water (400mL), extracted with EA (600 mL. times.2), and allowed to stand for separation. The organic phase was washed with saturated brine (120mL × 3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the obtained residue was purified by column chromatography (PE: EA ═ 3:1-1:1) to obtain the objective compound (15.9g, yield 58.5%, colorless oil).
And step 3: synthesis of methyl 2- (4- (ethylsulfonyl) phenyl) -3-methoxypropionate
Figure BDA0003065598540000494
Methyl 2- (4- (ethylsulfonyl) phenyl) -3-hydroxypropionate (15.8g,58.0mmol) was dissolved in DCM (140mL), and a 50% aqueous solution of fluoroboric acid (3.72mL,58.0mmol) and trimethylsilylated diazomethane (29.0mL,58.0 mmol) were added to the reaction mixture in an ice-water bath under exclusion of light,2.0M n-hexane solution). After 20 minutes, trimethylsilylated diazomethane (29.0mL,58.0mmol,2.0M n-hexane solution) was added again. After 20 minutes, trimethylsilylated diazomethane (29.0mL,58.0mmol,2.0M n-hexane solution) was added again. After 20 minutes, trimethylsilylated diazomethane (29.0mL,58.0mmol,2.0M n-hexane solution) was added again. The reaction mixture was poured into water (150mL), extracted with DCM (300 mL. times.2), and allowed to stand for separation. The organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the obtained residue was purified by column chromatography (PE: EA ═ 4:1-3:1) to obtain the objective compound (9.60g, yield 57.8%, colorless oil). MS (ESI) M/z 287.1[ M + H]+1H NMR(400MHz,CDCl3)δ7.87(d,J=8.4Hz,2H),7.54(d,J=8.4Hz,2H),4.03–3.93(m,2H),3.72(s,3H),3.69–3.63(m,1H),3.36(s,3H),3.11(q,J=7.4Hz,2H),1.29(t,J=7.4Hz,3H).
And 4, step 4: synthesis of 2- (4- (ethylsulfonyl) phenyl) -3-methoxypropionic acid
Figure BDA0003065598540000501
To a solution of methyl 2- (4- (ethylsulfonyl) phenyl) -3-methoxypropionate (9.60g,33.5mmol) in 1.4-dioxane (100mL) was added 6.0M hydrochloric acid (100 mL). The reaction mixture was stirred at 80 ℃ for 1 hour, and then the reaction was stopped. The reaction mixture was cooled to room temperature, poured into water (80.0mL), extracted with EA (250 mL. times.2), and allowed to stand for separation. The organic phase was dried over anhydrous sodium sulfate, filtered, the filtrate concentrated under reduced pressure, and the residue was subjected to medium pressure liquid phase preparation (acetonitrile/water containing 0.05% formic acid) to give the title compound (6.60g, yield 72.4%, pale yellow oil, which gradually solidified to a white solid after standing for 4 hours).1H NMR(400MHz,CDCl3)δ7.89(d,J=8.4Hz,2H),7.55(d,J=8.3Hz,2H),4.06–3.92(m,2H),3.77–3.62(m,1H),3.39(s,3H),3.11(q,J=7.4Hz,2H),1.29(t,J=7.4Hz,3H).
Step 5 Synthesis of N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-yl) -2- (4- (ethylsulfonyl) phenyl) -3-methoxypropionamide
Figure BDA0003065598540000502
To 2- (4- (ethylsulfonyl) phenyl) -3-methoxypropionic acid (50.7mg,0.186mmol), 2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl]To a solution of (E) -4-amine (50.0mg,0.155mmol) and N-methylimidazole (63.6mg,0.775mmol) in acetonitrile (1.00mL) was added TCFH (130mg,0.465mmol), and the reaction mixture was stirred at room temperature for 1 hour, whereupon the reaction was stopped. The reaction was directly subjected to high pressure liquid phase preparation (acetonitrile/water containing 0.05% formic acid) to give the title compound (34.1mg, yield 38.2%, white solid). LC-MS (ESI) M/z 575.9[ M + H ]]+/577.9[M+2+H]+1H NMR(400MHz,CDCl3)δ8.42(s,1H),7.91(d,J=8.3Hz,2H),7.68(s,2H),7.59(d,J=8.3Hz,2H),7.51–7.44(m,1H),7.41–7.34(m,2H),7.26–7.25,7.24–7.23(m,1H),4.05–4.00(m,1H),3.96–3.91(m,1H),3.82–3.76(m,1H),3.50(s,3H),3.13(q,J=7.4Hz,2H),1.30(t,J=7.4Hz,3H)。
Example 25 and example 26: rel- (S) -N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-yl) -2- (4- (ethylsulfonyl) phenyl) -3-methoxypropionamide or rel- (R) -N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl ] -4-yl) -2- (4- (ethylsulfonyl) phenyl) -3-methoxypropionamide
Figure BDA0003065598540000503
Racemic N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl)]-4-yl) -2- (4- (ethylsulfonyl) phenyl) -3-methoxypropionamide (30mg in ca. 25mL methanol, 2.0mL injection volume) was purified by passing Waters SFC 150 (room temperature, 100bar,214nm) and 250 × 25mm 10 μm Dr
Figure BDA0003065598540000512
OD) (supercritical carbon dioxide: ethanol, 60:40,5.0min,70mL/min) to obtain (S) -N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl)]-4-yl) -2- (4- (ethylsulfonyl) phenyl) -3-methoxypropionamide or (R) -N- (2, 6-dichloro-2' - (trifluoromethoxy) -, [2 ]1,1' -Biphenyl]-4-yl) -2- (4- (ethylsulfonyl) phenyl) -3-methoxypropionamide (11.6mg, white solid, ret. time 1.644min, e.e.100%). LC-MS (ESI) M/z 575.8[ M + H ]]+/577.8[M+2+H]+1H NMR(400MHz,CDCl3)δ8.40(s,1H),7.91(d,J=8.2Hz,2H),7.68(s,2H),7.59(d,J=8.2Hz,2H),7.50–7.45(m,1H),7.40–7.35(m,2H),7.26–7.25),7.24–7.23(m,1H),4.06–4.00(m,1H),3.97–3.91(m,1H),3.82–3.77(m,1H),3.50(s,3H),3.12(q,J=7.4Hz,2H),1.30(t,J=7.4Hz,3H)。
(R) -N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl)]-4-yl) -2- (4- (ethylsulfonyl) phenyl) -3-methoxypropionamide or (S) -N- (2, 6-dichloro-2 '- (trifluoromethoxy) - [1,1' -biphenyl]-4-yl) -2- (4- (ethylsulfonyl) phenyl) -3-methoxypropionamide (9.20mg, white solid, ret. time 2.219min, e.e.98%). LC-MS (ESI) M/z 575.9[ M + H ]]+/577.9[M+2+H]+1H NMR(400MHz,CD3OD)δ7.91(d,J=8.3Hz,2H),7.80–7.78(m,1H),7.77–7.76(m,1H),7.72(d,J=8.4Hz,1H),7.56–7.50(m,1H),7.45–7.38(m,2H),7.31–7.27(m,1H),4.12–4.05(m,1H),3.70–3.62(m,1H),3.39(s,3H),3.20(q,J=7.4Hz,2H),1.21(t,J=7.4Hz,3H)。
Active example 1: in vitro assay of the inhibitory Effect of Compounds on the ROR Gamma t luciferase reporter Gene
The experiment was carried out essentially as described in the literature (Current Chemical Genomics,2010,4, 43-49).
The ROR γ -LBD coding sequence was inserted into the pBIND plasmid (Promega, E1581). The expression vector and reporter vector (pGL4.35 carrying stably integrated GAL4 promoter driven luciferase reporter gene) were co-expressed in HEK293T host cells. When the inhibitor binds to the corresponding chimeric receptor, the chimeric receptor binds to the GAL4 binding site on the reporter vector and inhibits reporter expression. The inhibitory activity of the compound on ROR γ was determined from the intensity of the chemiluminescent signal.
Reagent and consumable
Figure BDA0003065598540000511
The experimental method comprises the following steps:
1 preparation of test Compounds
1.1 all test compounds were diluted in DMSO in 3-fold gradients, 10 dilution gradients, starting at 10 mM.
1.2 Positive control GSK993 was diluted in DMSO in 3-fold gradients, 10 dilution gradients, starting at 10 mM.
1.3 prepare 1000 × Positive control (10mM GSK993) and 1000 × negative control (100% DMSO).
1.4 the compound plate was blocked and shaken for 5 min.
2. Procedure of experiment
2.1 cell suspension preparation and plating
a) All cells were cultured according to ATCC standard procedures, and HEK293T was tested in exponential growth phase.
b) The medium was discarded.
c) Cells were washed 2 times with PBS.
d) The cells were digested by adding pancreatin digest and the digestion was stopped with complete medium.
e) Cells were collected and counted and experiments were only performed if the cell viability rate was greater than 90%.
f) Seed 6 x 106HEK293T cells into 100mm cell culture dishes.
g) The well-seeded cell culture dish was placed at 37 ℃ in 5% CO2The incubator was used for overnight culture.
2.2 transfection of cells
a) Placing the Trans-IT transfection reagent at room temperature for balancing;
b) adding 20 μ l transfection reagent into 600 μ l Opti-MEMTM culture medium, sucking and blowing with pipette tip without touching the tube wall, mixing well, standing at room temperature for 5 min;
c) add 10. mu.g plasmid to transfection reagent (see step 2.2.b), stand for 20 minutes at room temperature;
plasmid: separately, 5. mu.g of pBIND-ROR. gamma. and 5. mu.g of pGL4.35 plasmid were added
d) The transfection reagent mixed with DNA was added to a 100mm cell culture dish (see step 2.1);
e) the Petri dish was placed at 37 ℃ in 5% CO2The incubator is used for 5 h.
2.3 treatment of the Compounds
a) Transfer 25nl of the diluted compound to a cell culture plate with Echo550 (6007680-50, PE);
b) cells (see step 2.2) were seeded into 384 cell culture plates (6007680-50, PE) at 15,000 cells per well in 25. mu.l of 5% charcoal-adsorbed FBS medium;
c) cells were incubated at 37 ℃ with 5% CO2Overnight culture in an incubator
2.4 detection of Compounds:
a) subjecting Steady-GloTMPlacing the detection reagent at room temperature;
b) the 384 cell plate (see step 2.3) was left at room temperature;
c) add 25. mu.L of Steady-Glo to each wellTMDetecting the reagents on the cell culture plate (see step 2.4 b);
d) placing the plate on an oscillator and shaking for 5min in a dark place;
e) chemiluminescence values were measured using Envision 2104.
Calculation of% Inhibition:
RLU fluorescence values of test Compounds
Figure BDA0003065598540000531
Figure BDA0003065598540000532
Mean positive control
Figure BDA0003065598540000533
Average of negative controls
IC of compounds was calculated by fitting% Inhibition and log of compound concentration using Graphad8.050. The determination result shows that the compound of the invention has better inhibitory activity to the ROR gamma t luciferase reporter gene (shown in Table 1).
TABLE 1 determination of ROR gamma t luciferase reporter inhibitory Activity by the example Compounds
Figure BDA0003065598540000534
Figure BDA0003065598540000541
Active example 2: human PBMC Th17 cell differentiation inhibition assay
Test materials:
Figure BDA0003065598540000542
the experimental method comprises the following steps: firstly, PBMC cells are thawed and plated, then the PBMC cells are stimulated to differentiate to Th17 by stimulating factors (anti-hCD 28: 5 mu g/mL; rhTGF-beta 1: 5 ng/mL; rhIL-6: 20 ng/mL; rhIL-23: 10ng/Ml), compounds with different concentrations are added, the maximum concentration is started from 3 mu M, supernatants are collected after 48 hours for IL-17ELISA detection, the inhibition rate of the compounds for inhibiting the IL-17 secretion of Th17 cells is determined by comparing with a solvent group, and IC is fitted by Graphad8.050The value is obtained.
The determination result shows that the compound of the invention has better capability of inhibiting Th17 cells from differentiating and secreting IL-17 (shown in Table 2) for human PBMC.
TABLE 2 Experimental results of IL-17 secretion by inhibiting Th17 cell differentiation by compound
Examples IC50(nM)
1 16.52
2 3.49
4 11.41
5 6.287
9 24.54
12 6.928
16 91.42
19 39.96
20 48.95
22 81.12
23 80.05
25 9.197
Active example 3: human and mouse liver microsome metabolic stability assay
Liver Microsomal metabolic Stability assays for the compounds of the invention are similarly performed according to standard Methods of in vitro metabolic Stability studies conventional in the art, for example as described in (Kerns, Edward H.and Di Li (2008), Drug-like Properties: Concepts, Structure Design and Methods: from ADME to sensitivity Optimization, san Diego: Academic Press, Di, Li et al, Optimization of a high Throughput micro particulate Stability Screening Assay for Profiling Drug Discovery assays, J.biomol. Screen.2003,8(4), 453.).
The incubation system contained 0.5mg protein/mL microsome, cofactor, PBS, preincubated for 3min at 37 deg.C, and substrate (i.e., test compound) was added to initiate the reaction. Samples were taken at 0, 1, 5, 10, 15, 20, 30, 60min from the start of the reaction and appropriate stoppers were added to stop the reaction.
Figure BDA0003065598540000551
Sample treatment (n ═ 3): adding appropriate samples, vortexing, centrifuging at high speed, taking supernatant, and detecting the substrate by HPLC-MS/MS. The peak area at the time point of 0min was taken as 100%. The peak areas at other time points were converted to the residual percentages, the natural logarithm of the residual percentages at each time point was plotted against the incubation time, the slope (-k) was calculated by linear regression, and Clint (uL/min/mg) and the half-life of the compound (T1/2, min) were calculated from the intrinsic clearance (Clint) ((k) volume of incubation)/mass of liver microsomes. The results are shown in Table 3.
TABLE 3 results of the stability of liver microsome metabolism in human and mouse
Figure BDA0003065598540000561
The experimental results show that the compound of the invention has good metabolic stability.
Active example 4: FASSIF solubility assay for Compounds
The solubility Properties of the compounds of the invention were examined as follows, according to standard Methods of solubility determination conventional in the art, which are well known to the person skilled in the art, for example the method described in (Kerns, Edward H.and Di Li (2008), Drug-like Properties: Concepts, Structure Design and Methods: from ADME to sensitivity optimization, san Diego: Academic Press), using FaSSIF (pH6.5), which simulates intestinal fluid in the hunger state before a human meal.
The powder samples of each compound were weighed separately in volumetric flasks, 450. mu.L of a pH6.5 FaSSIF solution were added to obtain a supersaturated suspension, the samples were vortexed for at least 2 minutes, and the volumetric flasks were shaken on a shaker screen at 800 rpm for 24 hours. Then centrifuged at 4000rpm for 20 minutes. The compressed filtrate was added to a high performance liquid chromatography system and the concentration was calculated by a standard curve method, the results are shown in table 4.
The composition of the rapid simulated intestinal fluid FaSSIF is: 0.056% (w/v) lecithin, 0.161% (w/v) sodium taurocholate, 0.39% (w/v) potassium phosphate, 0.77% (w/v) potassium chloride, deionised H2O,pH 6.5。
TABLE 4 solubility of Compounds in FaSSIF
Examples FaSSIF pH=6.5(μg/ml) Examples FaSSIF pH=6.5(μg/ml)
2 82.30 12 33.90
4 137.00 16 296.36
5 23.10 25 73.20
8 32.50 GSK993 5.08
The above experimental results show that the example compounds of the present invention unexpectedly have significantly higher solubility and thus better druggability than the control.
Active example 5: CYP450 enzyme inhibitory Activity assay
The inhibitory effect of the compounds of the invention on Cytochrome P450 enzyme systems is similarly examined as follows, according to standard methods routinely used In the art for the study of Cytochrome P450 enzyme systems, for example as described In (Lin, Tong et al, In Vitro Assessment of Cytochrome P450 Inhibition: Strategies for incorporated LC/MS-Based Assay Throughput Using a One-Point IC50 Method and Multiplexing High-Performance Liquid Chromatography; J.pharm.Sci.2007,96(9), 2485.).
The following substrate stock solutions were prepared with methanol and substrate solutions at the following concentrations were further prepared with PB (phosphate buffer solution 100 mM):
Figure BDA0003065598540000571
stock solutions of the following standard inhibitors were prepared in DMSO at concentrations of 3mM each: α -naphthalenone (α -naphofelone), Sulfaphenazole (sulfophenazole), (+) -N-3-benzylnivanol ((+) -N-3-benzylnivanol), Quinidine (Quinidine) and Ketoconazole (Ketoconazole), and accordingly each solution was further formulated with methanol at a concentration of 300 μ M as a positive control solution so that the test final concentration was 3 μ M.
A0.253 mg/mL human liver microsome solution (Cat No.452117/Lot No.38293, Corning, final concentration: 0.2mg/mL) was prepared with PB (100 mM); and using 33mM MgCl2A10 mM cofactor solution (NADPH, Cat No.00616/Lot No.002162-050403, Chem-impex International, final concentration: 1mM) was prepared.
Stock solutions of 10mM of each test compound were prepared in DMSO and further working solutions at concentrations of 5, 1.50, 0.500, 0.150, 0.050, 0.015, 0.005mM in 1:1DMSO: MeOH, respectively, so that the final test concentrations were 50, 15, 5, 1.5, 0.5, 0.15, 0.05. mu.M.
mu.L of substrate solution was added to each well of the plate and 20. mu.L of PB was added to the blank wells. Add 2. mu.L of test compound and positive control working solution to the reaction wells and 2. mu.L of solvent to the non-inhibitor wells and blank wells. Adding 158 μ L of human liver microsome working solution to each incubation well, preheating the plate in a water bath at 37 ℃ for about 10 minutes; add 20. mu.L of NADPH solution to each incubation well and incubate all CYP in a 37 ℃ water bath for 10 min with mixing. After 60 minutes of reaction, the reaction was stopped by adding 400. mu.L of a cold stop solution (200ng/mL of tolbutamide and labetanol in acetonitrile). The sample was centrifuged at 4000rpm for 20 minutes to precipitate the protein, and 200. mu.L of the supernatant was transferred to 100. mu.L of high performance liquid chromatography water and shaken for 10 minutes for LC/MS/MS analysis, and the amount of the metabolite produced by each probe substrate was detected. The mass spectrum detection condition is electrospray ionization (ESI) source, the selective reaction monitoring (MRM) scanning mode, the chromatographic separation condition is Thermo GLOD, C8 chromatographic column (2.1mm multiplied by 50mm,1.9 mu g), the mobile phase is gradient elution with 0.1% formic acid acetonitrile-0.1% formic acid aqueous solution, the flow rate is 0.5m L min-1, and the semi-Inhibitory Concentration (IC) is calculated by applying Graph pad Prism 5 software50)。
TABLE 5 results of inhibition of CYP450 enzymes by Compounds
Figure BDA0003065598540000581
The above experimental results show that the compounds of the examples of the present invention unexpectedly have significantly lower CYP450 inhibitory activity (especially for CYP2C9, CYP2C19, CYP2D 6) compared to the control, indicating that they have reduced risk of drug interaction, resulting in lower toxicity and higher safety.
Active example 6: compound PK assay in mice
The PK assay for each compound was as follows: 6C 57BL/6 mice (sourced from Shanghai Ling Biotech, Inc.) were divided into two groups of 3 mice each. One group was given Intravenously (IV) at a dose of 1mg/kg, with 5% DMSO/95% (20% Captisol); one group was administered by oral (PO) gavage at a dose of 5mg/kg with vehicle of 0.5% CMC-Na/0.5% Tween 80. Each group was collected via the saphenous vein of the lower leg at 0, 0.083, 0.25, 0.5, 1,2, 4, 6, 8, 24h post-dose. Approximately 40. mu.L of blood was collected in an anticoagulation tube containing EDTA-K2. The tubes were inverted at least 5 times immediately after collection to ensure uniform mixing, and then placed on ice. Blood was collected at each time point and centrifuged at 8000rpm for 5 minutes at 4 ℃ to obtain plasma. Another 1.5mL centrifuge tube was labeled with compound name, animal number, time point and plasma was transferred to the tube. Plasma was stored at-80 ℃ until analysis.
The concentrations of the compounds in plasma were determined by the UPLC-MS/MS method, and pharmacokinetic parameters were calculated on the data obtained with Phoenix WinNolin 6.4 pharmacokinetic software.
The specific experimental results are as follows, and the results show that the compound has better drug absorption and pharmacokinetic advantages.
TABLE 6 in vivo PK results for the example Compounds
Figure BDA0003065598540000591
The structure of the control compound used in the above experiment is as follows:
Figure BDA0003065598540000592
those skilled in the art will appreciate that the foregoing description is exemplary and illustrative in nature and is intended to illustrate the present invention and its preferred embodiments. Obvious modifications and variations will be apparent to those skilled in the art without departing from the spirit of the invention. All such modifications within the scope of the appended claims are intended to be included therein. Accordingly, it is intended that the invention not be limited by the foregoing description, but be defined by the scope of the following claims and their equivalents.
All publications cited in this specification are herein incorporated by reference.

Claims (17)

1. A compound of formula (I), a stereoisomer, a tautomer, a stable isotopic variant, a pharmaceutically acceptable salt, or a solvate thereof,
Figure FDA0003065598530000011
wherein:
x is selected from CH or N;
R1selected from hydrogen, halogen, cyano, nitro, C1-C6Alkyl, -O-C1-C6Alkyl, S-C1-C6Alkyl, -NH-C1-C6Alkyl, -N- (C)1-C6Alkyl radical)2、C1-C6alkyl-O-C1-C6Alkyl, -C1-C6alkyl-S-C1-C6Alkyl radical, C1-C6alkyl-NH-C1-C6Alkyl or C1-C6alkyl-N (C)1-C6Alkyl radical)2Wherein said C is1-C6Alkyl is optionally substituted by halogen or cyano;
R2selected from hydrogen, halogen, cyano, nitro or C1-C6Alkyl radical, wherein said C1-C6Alkyl is optionally substituted with substituents independently selected from: -Raor-OH, wherein RaIs C optionally substituted by halogen1-C6An alkyl group;
R3、R4each independently selected from halogen, cyano, nitro, Rb、-ORb、-SRbor-NRbRbWherein R isbIs H or optionally substituted C1-C6Alkyl, each of said substituents being independently selected from hydrogen, halogen, cyano, nitro, -OH, -SH, -NH2、-O-C1-C6Alkyl, -S-C1-C6Alkyl, -NH-C1-C6Alkyl or-N (-C)1-C6Alkyl radical)2
R5Is selected from-ORc、-SRc、-NRcRc、-NHCORc、-NHSO2Rc、-COORc、-CONRcRcor-SO2NRcRcWherein R iscIndependently at each occurrence, is selected from hydrogen, C optionally substituted with halogen1-C6Alkyl or C optionally substituted by halogen3-C7Cycloalkyl, or two R bound to the same N atomcMay form, together with the N atom to which they are attached, a 4-7 membered nitrogen containing heterocycloalkyl group;
R6is selected from C1-C6Alkyl radical, C3-C7Cycloalkyl, 4-7 membered heterocycloalkyl or optionally substituted by C1-C6Alkyl radical, C3-C7Cycloalkyl or 4-7 membered heterocycloalkyl substituted-NH2Wherein said C is1-C6Alkyl radical, C3-C7Cycloalkyl or 4-7 membered heterocycloalkyl is optionally substituted with substituents each independently selected from: hydrogen, halogen, cyano, nitro, -Ra、-ORa、-SRaor-NRaRaOr C optionally substituted by halogen3-C7Cycloalkyl radical, RaIndependently at each occurrence, is selected from H or C optionally substituted with halogen1-C6Alkyl, wherein two groups attached to the N atom of the amino group may form a 4-7 membered ring together with the N to which they are attached; and is
n is selected from 0, 1 or 2.
2. A compound according to claim 1, a stereoisomer, tautomer, stable isotopic variant, pharmaceutically acceptable salt or solvate thereof, wherein X is CH.
3. A compound according to claim 1 or 2, a stereoisomer, a tautomer, a stable isotopic variation, a pharmaceutically acceptable salt, or a solvate thereof, wherein R is1Is selected from-O-C1-C6Alkyl radical, wherein C1-C6Alkyl substituted by halogen, preferably-OCF3、-OCHF2、-OCH2CF3、-OCH2CH2CF3、-CH(CF3)2
4. A compound according to claim 1 or 2, a stereoisomer, a tautomer, a stable isotopic variation, a pharmaceutically acceptable salt, or a solvate thereof, wherein R is1Is hydrogen, and R2Selected from halogen and
Figure FDA0003065598530000021
5. a compound according to any one of claims 1 to 4, a stereoisomer, a tautomer, a stable isotopic variation, a pharmaceutically acceptable salt, or a solvate thereof, wherein R3、R4Each independently selected from H, halogen or C optionally substituted by halogen1-C6An alkyl group.
6. A compound according to any one of claims 1 to 5, a stereoisomer, a tautomer, a stable isotopic variation, a pharmaceutically acceptable salt, or a solvate thereof, wherein R5is-ORcWherein R iscSelected from hydrogen, C optionally substituted by halogen1-C6An alkyl group.
7. A compound according to any one of claims 1 to 5, a stereoisomer, a tautomer, a stable isotopic variation, a pharmaceutically acceptable salt, or a solvate thereof, wherein R5Is selected from-NRcRcor-NHCORcWherein R iscIndependently at each occurrence, is selected from hydrogen or C optionally substituted with halogen1-C6Alkyl, or two R bound to the same N atomcMay form, together with the N atom to which they are attached, a 4-to 7-membered nitrogen-containing heterocycloalkyl group.
8. A compound according to any one of claims 1 to 5, a stereoisomer, a tautomer, a stable isotopic variation, a pharmaceutically acceptable salt, or a solvate thereof, wherein R5Is selected from-COORcor-CONRcRcWherein R iscIndependently at each occurrence, is selected from H or C optionally substituted with halogen1-C6An alkyl group.
9. A compound according to any one of claims 1 to 8, a stereoisomer, a tautomer, a stable isotopic variation, a pharmaceutically acceptable salt, or a solvate thereof, wherein R6Is selected from C1-C6Alkyl, preferably C1-C3Alkyl, optionally substituted by C3-C7Cycloalkyl is substituted.
10. A compound according to any one of claims 1 to 8, a stereoisomer, a tautomer, a stable isotopic variation, a pharmaceutically acceptable salt, or a solvate thereof, wherein R6Selected from optionally substituted-NH2Said substituent being selected from C optionally substituted by halogen1-C6An alkyl group.
11. A compound selected from
Figure FDA0003065598530000031
Figure FDA0003065598530000041
Stereoisomers, tautomers, stable isotopic variations, pharmaceutically acceptable salts or solvates thereof.
12. A compound according to any one of claims 1 to 11, a stereoisomer, a tautomer, a stable isotopic variant, a pharmaceutically acceptable salt or a solvate thereof, for use as a medicament, particularly as an inhibitor of ROR γ t, or for use in the treatment, particularly in the treatment or prevention of a disease in which ROR γ t is involved.
13. A pharmaceutical composition comprising a compound according to any one of claims 1 to 11, a stereoisomer, a tautomer, a stable isotopic variant, a pharmaceutically acceptable salt, or a solvate thereof, and a pharmaceutically acceptable excipient.
14. A method for the prevention or treatment of diseases related to roryt in mammals, in particular humans, which comprises administering an effective amount of a compound according to any one of claims 1 to 11, a stereoisomer, a tautomer, a stable isotopic variant, a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition according to claim 13.
15. Use of a compound according to any one of claims 1 to 11, a stereoisomer, a tautomer, a stable isotopic variant, a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition according to claim 13 for the prophylaxis or treatment of a disease associated with roryt.
16. Use of a compound according to any one of claims 1 to 11, a stereoisomer, a tautomer, a stable isotopic variant, a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition according to claim 13, for the preparation of a medicament for the prophylaxis or treatment of a disease associated with roryt.
17. A compound according to claim 12, a method according to claim 14 or a use according to claim 15 or 16, wherein the disease associated with roryt is selected from psoriasis, rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, multiple sclerosis, systemic lupus erythematosus, graft-versus-host disease, inflammatory bowel disease, crohn's disease, ulcerative colitis, chronic obstructive pulmonary disease, asthma, glomerulonephritis, lupus nephritis, myocarditis, thyroiditis, dry eye, uveitis, behcet's disease, allergic dermatitis, acne, scleroderma, bronchitis, dermato-allergic rhinitis, necrotizing enterocolitis, liver fibrosis, non-alcoholic steatohepatitis (NASH), neocoronary pneumonia, insulin dependent type I diabetes, triple negative breast cancer and prostate cancer, and the like.
CN202110526063.0A 2020-05-15 2021-05-14 Biaryl compounds useful as ROR gamma modulators Active CN113666853B (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202010411822 2020-05-15
CN2020104118224 2020-05-15

Publications (2)

Publication Number Publication Date
CN113666853A true CN113666853A (en) 2021-11-19
CN113666853B CN113666853B (en) 2023-04-18

Family

ID=78525328

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110526063.0A Active CN113666853B (en) 2020-05-15 2021-05-14 Biaryl compounds useful as ROR gamma modulators

Country Status (3)

Country Link
CN (1) CN113666853B (en)
TW (1) TW202146381A (en)
WO (1) WO2021228215A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023232870A1 (en) 2022-05-31 2023-12-07 Immunic Ag Rorg/rorgt modulators for the treatment of virus infections like covid-19

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013029338A1 (en) * 2011-09-01 2013-03-07 Glaxo Group Limited Novel compounds
WO2013171729A2 (en) * 2013-01-08 2013-11-21 Glenmark Pharmaceuticals S.A. Aryl and heteroaryl amide compounds as rorgamat modulator
CN108430973A (en) * 2015-06-05 2018-08-21 领先制药控股公司 ROR gamma modulators
CN108863850A (en) * 2017-02-09 2018-11-23 复旦大学 Biaryl base class compound and its preparation method and application

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017010399A1 (en) * 2015-07-10 2017-01-19 塩野義製薬株式会社 COMPOUNDS HAVING RORγt INHIBITORY EFFECTS AND PHARMACEUTICAL COMPOSITIONS CONTAINING THE SAME
WO2017157332A1 (en) * 2016-03-18 2017-09-21 江苏恒瑞医药股份有限公司 Aromatic amide derivative, preparation method therefor, and pharmaceutical applications thereof
CA3097519A1 (en) * 2018-05-03 2019-11-07 Jiangsu Hengrui Medicine Co., Ltd. Benzimidazole derivatives as modulators of retinoid-related orphan receptor gamma (ror.gamma.) and pharmaceutical uses thereof
CA3155875A1 (en) * 2019-09-30 2021-04-08 Shanghai Litedd Co., Ltd. Sulfo-substituted biaryl compounds or salts thereof and preparation method and use thereof

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013029338A1 (en) * 2011-09-01 2013-03-07 Glaxo Group Limited Novel compounds
WO2013171729A2 (en) * 2013-01-08 2013-11-21 Glenmark Pharmaceuticals S.A. Aryl and heteroaryl amide compounds as rorgamat modulator
CN108430973A (en) * 2015-06-05 2018-08-21 领先制药控股公司 ROR gamma modulators
CN108863850A (en) * 2017-02-09 2018-11-23 复旦大学 Biaryl base class compound and its preparation method and application

Also Published As

Publication number Publication date
TW202146381A (en) 2021-12-16
CN113666853B (en) 2023-04-18
WO2021228215A1 (en) 2021-11-18

Similar Documents

Publication Publication Date Title
RU2684641C1 (en) Pyrazolopyridine derivatives as modulators of tnf activity
CN108383836B (en) Heterocyclic compounds as MEK inhibitors
RU2685234C1 (en) Condensed bicyclic heteroaromatic derivatives as modulators of tnf activity
JP2021036004A (en) Inhibitors of ret
CN113637005A (en) KRAS inhibitors for cancer treatment
US20170231999A1 (en) Ido inhibitors
CN110678472B (en) 2-oxothiazole derivatives as A2A inhibitors and compounds for the treatment of cancer
AU2017382360A1 (en) Compounds, compositions and methods of use
WO2014172044A1 (en) Substituted benzene compounds
UA74850C2 (en) Amide derivatives as nmda receptor antagonists
JP2021519828A (en) Diaryl macrocycles, pharmaceutical compositions and their uses
JP2024505732A (en) Pyridopyrimidinone derivatives and their production methods and uses
AU2013253541A1 (en) Quinazolinedione derivative
CA3059687A1 (en) Amide derivatives as nav1.7 and nav1.8 blockers
CN113666853B (en) Biaryl compounds useful as ROR gamma modulators
CN113666863A (en) Biaryl compounds useful as ROR gamma modulators
CN113773251A (en) Anilines useful as ROR gamma modulators
JPWO2019235553A1 (en) Azetidine derivatives and their prodrugs
CN107428682B (en) Amide derivatives, preparation method and medical application thereof
JP2024516194A (en) Compounds as PD1/PD-L1 inhibitors and methods thereof
CN114790176A (en) Imidazole compound and preparation method and application thereof
EP4185379A1 (en) Therapeutic agents targeting gpr35
CN114605390A (en) Compound with CDK kinase inhibitory activity, pharmaceutical composition and use thereof
CN114096533B (en) Tri-ring compound, pharmaceutical composition containing same, preparation method and application thereof
CN116535424A (en) Tri-fused ring compounds as MALT1 inhibitors, pharmaceutical compositions 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
REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 40059281

Country of ref document: HK

GR01 Patent grant
GR01 Patent grant
CP03 Change of name, title or address
CP03 Change of name, title or address

Address after: Room 401, Building 4, No. 88 Jiangling Road, Xixing Street, Binjiang District, Hangzhou City, Zhejiang Province, 310051

Patentee after: Lidi (Hangzhou) Pharmaceutical Technology Co.,Ltd.

Address before: Room 1109, 781 Cailun Road, Pudong New Area, Shanghai 201203

Patentee before: Shanghai Huiqi Biomedical Technology Co.,Ltd.