CN113072521B - ROR gamma t inhibitor and application thereof in medicines - Google Patents

ROR gamma t inhibitor and application thereof in medicines Download PDF

Info

Publication number
CN113072521B
CN113072521B CN202011615479.1A CN202011615479A CN113072521B CN 113072521 B CN113072521 B CN 113072521B CN 202011615479 A CN202011615479 A CN 202011615479A CN 113072521 B CN113072521 B CN 113072521B
Authority
CN
China
Prior art keywords
mmol
methyl
trifluoromethyl
dcm
compound
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN202011615479.1A
Other languages
Chinese (zh)
Other versions
CN113072521A (en
Inventor
刘兵
潘伟
张英俊
何为
王峰
李旭珂
许娟
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.)
Guangdong HEC Pharmaceutical
Original Assignee
Guangdong HEC Pharmaceutical
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 Guangdong HEC Pharmaceutical filed Critical Guangdong HEC Pharmaceutical
Publication of CN113072521A publication Critical patent/CN113072521A/en
Application granted granted Critical
Publication of CN113072521B publication Critical patent/CN113072521B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D295/00Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms
    • C07D295/04Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms
    • C07D295/12Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by singly or doubly bound nitrogen atoms
    • C07D295/135Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by singly or doubly bound nitrogen atoms with the ring nitrogen atoms and the substituent nitrogen atoms separated by carbocyclic rings or by carbon chains interrupted by carbocyclic rings
    • 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
    • A61P11/06Antiasthmatics
    • 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
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D211/00Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings
    • C07D211/04Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D211/06Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D211/00Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings
    • C07D211/04Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D211/06Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D211/36Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D211/40Oxygen atoms
    • C07D211/42Oxygen atoms attached in position 3 or 5
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/06Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having one double bond between ring members or between a ring member and a non-ring member
    • C07D239/08Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having one double bond between ring members or between a ring member and a non-ring member with hetero atoms directly attached in position 2
    • C07D239/10Oxygen or sulfur atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/10Spiro-condensed systems
    • C07D491/107Spiro-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Pulmonology (AREA)
  • Rheumatology (AREA)
  • Pain & Pain Management (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention relates to a RORγt inhibitor compound which is a compound shown as a formula (I). The compounds or pharmaceutical compositions comprising the compounds are useful for modulating retinoic acid related lone-core receptors γt (Retinoid-related orphan receptor gamma t, rorγt). The invention also relates to a method for preparing the compound and/or the pharmaceutical composition, and the application of the compound and/or the pharmaceutical composition in preparing medicines for treating or preventing inflammatory or autoimmune diseases mediated by RORγt of mammals, especially human beings.

Description

ROR gamma t inhibitor and application thereof in medicines
Technical Field
The invention belongs to the technical field of medicines, and particularly relates to a small molecular compound, a composition, a preparation method and application thereof, wherein the compound or the composition can be used as an inhibitor of retinoic acid related lone-core receptor gamma t (Retinoid-related orphan receptor gamma t, ROR gamma t) and used for preventing or treating immune related diseases.
Background
Retinoic acid related lone nuclear receptors are subfamilies of transcription factors in the steroid hormone nuclear receptor superfamily. The tretinoin related lone core receptor family includes rorα, rorβ and rorγ, encoded by different genes (RORA, RORB and RORC), respectively. Retinoic acid related lone nuclear receptors contain four major domains: an N-terminal A/B domain, a DNA binding domain, a hinge domain, and a ligand binding domain.
Retinoic acid related lone-core receptor γt (Retinoid-related orphan receptor gamma t, rorγt) is one of two isoforms of retinoic acid related lone-core receptor γ (Retinoid-related orphan receptor gamma, rorγ), also known as rorγ2. There were studies showing that roryt is expressed only in lymphoid lineage and embryonic lymphoid tissue inducer cells (Sun et al Science 288:2369-2372,2000;Eberl et al, nat immunol.5:64-73,2004). RORγt, which is a transcription factor characteristic of helper T cells (Th 17), plays an important role in Th17 cell differentiation, and is a key regulator of Th17 cell differentiation (Ivanov, II, mcKenzie BS, zhou L, tadokoro CE, lepelley A, lafaille JJ, et al cell 2006;126 (6): 1121-33).
Th17 is capable of secreting interleukin 17 (IL-17) and other pro-inflammatory cytokines, and has important implications in autoimmune diseases and body defense responses. IL-17 is a pro-inflammatory cytokine for the development of inflammation and various autoimmune diseases, and is closely related to various autoimmune diseases and inflammatory diseases, such as rheumatoid arthritis, psoriasis, psoriatic arthritis, spondyloarthritis, asthma, inflammatory bowel disease, systemic lupus erythematosus, multiple sclerosis, and the like (Jetten et al, nucl. Receiver. Signal,2009,7:e003;Manel et al, nat. Immunol.,2008,9,641-649).
The role of rorγt in the pathogenesis of autoimmune diseases or inflammation has been widely studied and fully elucidated (Jetten et al, adv. Dev. Biol,2006,16:313-355;Meier et al.Immunity,2007,26:643-654; aloisi et al, nat. Rev. Immunol.,2006,6:205-217; jager et al, j. Immunol.,2009,183:7169-7177;Barnes et al, nat. Rev. Immunol.,2008, 8:183-192). Thus, inhibition of roryt will effectively inhibit Th17 cell differentiation, regulate IL-17 and other pro-inflammatory cytokine production and secretion levels, and thus regulate the immune system of the body, treating immune and inflammatory diseases associated with roryt regulation.
Summary of The Invention
The following is merely a general description of some aspects of the invention and is not limited in this regard. These aspects and others are described more fully below. All references in this specification are incorporated herein by reference in their entirety. When the disclosure of the present specification is different from that of the cited document, the disclosure of the present specification controls.
The invention provides a compound with retinoic acid related lone nucleus receptor gamma t (Retinoid-related orphan receptor gamma t, ROR gamma t) inhibitory activity, which is used for preparing medicines for preventing or treating inflammation or autoimmune diseases mediated by ROR gamma t, such as psoriasis, rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis, inflammatory bowel disease, colitis, ulcerative colitis, rheumatic arthritis, autoimmune ocular disease, ankylosing spondylitis, asthma, chronic obstructive pulmonary disease, osteoarthritis, allergic rhinitis, allergic dermatitis, crohn's disease or Kawasaki disease and the like; the compound can well inhibit RORγt and has excellent physicochemical property and pharmacokinetic property.
The invention also provides methods of preparing these compounds and pharmaceutical compositions comprising these compounds and methods of using these compounds or compositions in the treatment of the above-described diseases in mammals, especially humans.
Specifically:
in one aspect, the present invention relates to a compound of formula (I) or a stereoisomer, a geometric isomer, a tautomer, a nitroxide, a hydrate, a solvate, a metabolite, an ester, a pharmaceutically acceptable salt or a prodrug thereof,
wherein:
r is C 1-4 Alkyl, C 1-4 Alkoxy, C 1-4 Alkylamino, C 3-6 Cycloalkylamino, -C 1-4 alkylene-C 3-6 Cycloalkyl, C 3-6 Cycloalkyl, C 1-4 Haloalkyl or C 1-4 Haloalkoxy groups;
Z 1 、Z 2 、Z 3 、Z 4 、Z 5 、Z 6 、Z 7 and Z 8 Each independently of the otherGround is CR 1 Or N;
each R is 1 Independently hydrogen, deuterium, cyano, fluorine, chlorine, bromine, iodine, C 1-6 Alkyl, C 1-6 Haloalkyl, hydroxy-substituted C 1-6 Alkyl, -C 1-6 alkylene-C 1-6 Alkoxy, C 1-6 Alkoxy, C 3-8 Cycloalkyl or 3-8 membered heterocyclyl;
ring A is a 6 membered heterocyclic ring, said 6 membered heterocyclic ring optionally being substituted with 1, 2, 3 or 4R 2 Substitution;
each R is 2 Independently is fluoro, chloro, bromo, iodo, hydroxy, oxo (=o), amino, nitro, cyano, C 1-6 Alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, C 6-10 Aryl, C 3-8 Cycloalkyl, -C 0-6 alkylene-OR a 、-C 0-4 Alkylene- (C (=O)) m -R b 、-C 0-4 Alkylene- (O) n -(C(=O)) m -NR c R d 5-to 10-membered heteroaryl, -C 0-4 Alkylene- (3-7 membered heterocyclyl), -C 0-4 Alkylene- (5-12 membered spiroheterocyclyl) or-C 0-4 Alkylene- (4-12 membered fused heterocyclyl); the R is 2 Optionally by 1, 2 or 3R f Substitution;
alternatively, two R's, optionally attached to the same carbon atom 2 And the carbon atoms to which they are attached together form C 3-6 Cycloalkyl or 4-6 membered heterocycle;
each R is a Independently hydrogen, C 1-6 Alkyl, C 1-6 Haloalkyl, C 6-10 Aryl, C 3-8 Cycloalkyl, 5-10 membered heteroaryl, 3-7 membered heterocyclyl, 5-12 membered spiroheterocyclyl or 4-12 membered fused heterocyclyl; each R is b 、R c And R is d Independently hydrogen, hydroxy, C 1-6 Alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, C 6-10 Aryl, C 3-8 Cycloalkyl, 4-12 membered fused ring group, 5-12 membered spiro ring group, 5-10 membered heteroaryl group, 3-7 membered heterocyclic group, 5-12 membered spiro heterocyclic group or 4-12 membered fused heterocyclic group; or R is c 、R d And the N atoms to which they are attached together form 4-7 memberedA heterocycle; each R is as follows a 、R b 、R c And R is d Independently optionally substituted with 1, 2, 3, 4, 5 or 6R g Substitution;
each R is f And R is g Independently is fluorine, chlorine, bromine, iodine, oxo, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, hydroxy substituted C 1-6 Alkyl, C 1-6 Haloalkyl, C 6-10 Aryl, C 3-8 Cycloalkyl, 4-12 membered fused carbocyclyl, 5-12 membered spirocarbocyclyl, 5-10 membered heteroaryl, 3-12 membered heterocyclyl, 5-12 membered spiroheterocyclyl, 4-12 membered fused heterocyclyl, C 1-6 Alkoxy, C 1-6 Alkylamino or C 1-6 Haloalkoxy groups;
ring B is C 6-10 Aryl, 4-12 membered fused heterocyclyl, 4-12 membered fused carbocyclyl, 5-12 membered spirocarbocyclyl or C 3-8 Cycloalkyl, wherein the B ring is optionally substituted with 1, 2, 3 or 4R' s e Substitution;
each R is e Independently deuterium, fluorine, chlorine, bromine, iodine, cyano, nitro, hydroxy, amino, C 1-4 Alkyl, C 1-4 Alkoxy, C 3-6 Cycloalkyl, 3-8 membered heterocyclyl, hydroxy substituted C 1-4 Alkyl, hydroxy substituted C 1-4 Haloalkyl, C 1-4 Haloalkyl or C 1-4 Haloalkoxy groups;
alternatively, any two adjacent R e And together with the atoms to which they are attached form C 3-8 Cycloalkyl or 3-8 membered heterocyclyl;
wherein, C is as follows 1-4 Alkoxy optionally substituted by 1 3-to 6-membered heterocyclyl or C 3-8 Cycloalkyl substitution;
L 1 is-S (=O) 2 -NH-、-NH-S(=O) 2 -S (=o) -NH-, -NH-S (=o) -, -C (=o) NH-, or-NHC (=o) -;
L 2 is-CR 3 R 4
L 3 Is carbonyl, -S-, -O-, -NR 5 -or-CR 6 R 7 -;
R 3 C substituted by hydrogen or hydroxy 1-6 Substituted by alkyl or cyano groupsC 1-6 An alkyl group;
R 4 c substituted by hydrogen or hydroxy 1-6 C substituted by alkyl or cyano 1-6 An alkyl group;
R 5 is hydrogen, C 1-6 Alkyl, C 1-6 Haloalkyl, C 3-8 Cycloalkyl or 3-6 membered heterocyclyl;
each R is 6 And R is 7 Independently hydrogen, C 1-6 Alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 3-8 Cycloalkyl or 3-6 membered heterocyclyl;
each m and n is independently 0 or 1.
In some embodiments, R is methyl, ethyl, n-propyl, isopropyl, monofluoromethyl, difluoromethyl, trifluoromethyl, 1, 2-difluoroethyl, methoxy, ethoxy, n-propoxy, isopropoxy, methylamino, ethylamino, dimethylamino, cyclopropylamino, cyclobutylamino, cyclopentylamino, cyclohexylamino, methylenecyclopropyl, methylenecyclobutyl, methylenecyclopentyl, methylenecyclohexyl, ethylenecyclopropyl, ethylenecyclobutyl, ethylenecyclopentyl, ethylenecyclohexyl, cyclopropyl, cyclopentyl, or cyclohexyl.
In some embodiments, each R 1 Independently is hydrogen, deuterium, cyano, fluorine, chlorine, bromine, iodine, methyl, ethyl, n-propyl, isopropyl, difluoromethyl, trifluoromethyl, 1, 2-difluoroethyl, 1-difluoroethyl, methoxy, ethoxy, n-propoxy, isopropoxy, methoxymethylene, ethoxymethylene, n-propoxymethyl, isopropoxymethyl, cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl.
In some embodiments, R 3 Is hydrogen, hydroxymethyl, hydroxyethyl, hydroxy-n-propyl, cyanomethyl, cyanoethyl or cyano-n-propyl; r is R 4 Is hydrogen, hydroxymethyl, hydroxyethyl, hydroxy-n-propyl, cyanomethyl, cyanoethyl or cyano-n-propyl.
In some embodiments, the B ring is phenyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, The B ring is optionally substituted with 1,2, 3 or 4R' s e And (3) substitution.
In some embodiments, each R e Independently deuterium, fluorine, chlorine, bromine, iodine, cyano, nitro, hydroxy, amino, methyl, ethyl, n-propyl, isopropyl, hydroxymethyl, 2-hydroxyethyl, 3-hydroxy-n-propyl, 2-hydroxy-1, 3-hexafluoroisopropyl, monofluoromethyl, difluoromethyl, trifluoromethyl, 1, 2-difluoroethyl, methoxy, ethoxy, n-propoxy, t-butoxy, monofluoromethoxy, difluoromethoxy, trifluoromethoxy or 1, 2-difluoroethoxy; wherein said methoxy, ethoxy, n-propoxy, t-butoxy is optionally substituted with 1 substituent selected from the group consisting of oxiranyl, azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, pyrazolidinyl, imidazolidinyl, tetrahydrofuranyl, tetrahydrothienyl, 1, 3-dioxacyclopentyl, dithiocyclopentyl, tetrahydropyranyl, 4H-pyranyl, tetrahydrothiopyranyl, piperidinyl, morpholinyl, thiomorpholinyl, and piperazinyl.
In some embodiments, R 5 Is hydrogen, methyl, ethyl, isopropyl, n-propyl, isobutyl, tert-butyl, n-butyl, monofluoromethyl, difluoromethyl, trifluoromethyl, 1, 2-difluoroethyl, 2-difluoroethyl, methoxy, ethoxy, n-propoxy, isopropoxy, cyclopropyl, cyclobutyl or azetidinyl;
each R is 6 And R is 7 Independently hydrogen, methyl, ethyl, isopropyl, n-propyl, isobutyl, tert-butyl, n-butyl, monofluoromethyl, difluoromethyl, trifluoromethyl, 1, 2-difluoroethyl, 1-difluoroethyl, methoxy, ethoxy, n-propoxy, isopropoxy, monofluoromethoxy, difluoromethoxy, trifluoromethoxy, 1, 2-difluoroethoxy, 2-difluoroethoxy, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, azetidine, oxetanyl, tetrahydrofuranA group, tetrahydropyranyl, pyrrolidinyl, piperidinyl or piperazinyl.
In some embodiments, the A ring is Wherein the A ring is optionally substituted with 1,2, 3 or 4R' s 2 And (3) substitution.
In some embodiments, each R 2 Independently is fluorine, chlorine, bromine, iodine, hydroxyl, oxo, amino, nitro, cyano, C 1-4 Alkyl, C 1-4 Haloalkyl, C 1-4 Alkoxy, phenyl, C 3-6 Cycloalkyl, -C 0-4 alkylene-OR a 、-C 0-4 Alkylene- (C (=O)) m -R b 、-C 0-4 Alkylene- (O) n -(C(=O)) m -NR c R d 5-6 membered heteroaryl, -C 0-4 Alkylene- (3-7 membered heterocyclyl), -C 0-4 Alkylene- (5-12 membered spiroheterocyclyl) or-C 0-4 Alkylene- (4-12 membered fused heterocyclyl); wherein the R is 2 Optionally by 1,2 or 3R f Substitution; alternatively, two R's, optionally attached to the same carbon atom 2 And the carbon atoms to which they are attached together form C 3-6 Cycloalkyl or 4-6 membered heterocycle.
In some embodiments, each R a Independently hydrogen, C 1-4 Alkyl, C 1-4 Haloalkyl, C 6-10 Aryl, C 3-6 Cycloalkyl, 5-7 membered heteroaryl, or 3-7 membered heterocyclyl; each R is b 、R c And R is d Independently hydrogen, hydroxy, C 1-4 Alkyl, C 1-4 Haloalkyl, C 1-4 Alkoxy, C 6-10 Aryl, C 3-6 Cycloalkyl, 3-7 membered heterocyclyl or 5-12 membered spiroheterocyclyl; or R is c 、R d And the N atoms to which they are attached together form a 4-7 membered heterocyclic ring;
wherein each R is a 、R b 、R c And R is d Independent and independentOptionally by 1,2, 3, 4, 5 or 6R g And (3) substitution.
In some embodiments, each R 2 Independently is fluoro, chloro, bromo, iodo, hydroxy, oxo, amino, nitro, cyano, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, t-butyl, monofluoromethyl, difluoromethyl, trifluoromethyl, monofluoroethyl, 1-difluoroethyl, 1, 2-difluoroethyl, methoxy, ethoxy, n-propoxy, isopropoxy, phenyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, -OR a 、-CH 2 -OR a 、-CH 2 CH 2 -OR a 、-CH 2 (CH 2 ) 2 -OR a 、-CH 2 CH(CH 3 )-OR a 、-C(CH 3 ) 2 CH 2 -OR a 、-CH 2 (CH 2 ) 3 -OR a 、-CH(CH 3 )CH(CH 3 )-OR a 、-CH 2 C(CH 3 ) 2 -OR a 、-(C=O) m -R b 、-CH 2 -(C(=O)) m -R b 、-CH 2 CH 2 -(C(=O)) m -R b 、-CH 2 (CH 2 ) 2 -(C(=O)) m -R b 、-CH 2 CH(CH 3 )-(C(=O)) m -R b 、-C(CH 3 ) 2 CH 2 -(C(=O)) m -R b 、-CH 2 (CH 2 ) 3 -(C(=O)) m -R b 、-CH(CH 3 )CH(CH 3 )-(C(=O)) m -R b 、-CH 2 C(CH 3 ) 2 -(C(=O)) m -R b 、-(O) n -(C(=O)) m -NR c R d 、-CH 2 -(O) n -(C(=O)) m -NR c R d 、-CH 2 CH 2 -(O) n -(C(=O)) m -NR c R d 、-CH 2 (CH 2 ) 2 -(O) n -(C(=O)) m -NR c R d 、-CH 2 CH(CH 3 )-(O) n -(C(=O)) m -NR c R d 、-C(CH 3 ) 2 CH 2 -(O) n -(C(=O)) m -NR c R d 、-CH 2 (CH 2 ) 3 -(O) n -(C(=O)) m -NR c R d 、-CH(CH 3 )CH(CH 3 )-(O) n -(C(=O)) m -NR c R d 、-CH 2 C(CH 3 ) 2 -(O) n -(C(=O)) m -NR c R d Oxazolyl, thiazolyl, thienyl, imidazolyl, pyridazinyl, pyridyl, Wherein the R is 2 Optionally by 1,2 or 3R f Substitution;
alternatively, two R's, optionally attached to the same carbon atom 2 And the carbon atoms to which they are attached together form cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, pyrazolidinyl, imidazolidinyl, tetrahydrofuranyl, tetrahydrothienyl, 1, 3-dioxacyclopentyl, dithiocyclopentyl, tetrahydropyranyl, 4H-pyranyl, tetrahydrothiopyranyl, piperidinyl, morpholinyl, thiomorpholinyl or piperazinyl.
In some embodiments, each R a Independently is hydrogen, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, monofluoromethyl, difluoromethyl, trifluoromethyl, monofluoroethyl, 2-difluoroethyl, 1, 2-difluoroethyl, 2-trifluoroethyl, Phenyl group,Oxetanyl, tetrahydrofuranyl, tetrahydropyranyl, oxetanyl, cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl; each R is b 、R c And R is d Is independently hydrogen, hydroxy, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, monofluoromethyl, difluoromethyl, trifluoromethyl, monofluoroethyl, 2-difluoroethyl, 1, 2-difluoroethyl, 2-trifluoroethyl, methoxy, ethoxy, n-propoxy, isopropoxy, phenyl, oxetanyl, tetrahydrofuranyl, tetrahydropyranyl, oxetanyl, cycloheptyl, Cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl; or R is c And R is d Together with the N atoms to which they are attached, form a 4-7 membered heterocyclic ring;
wherein each R is a 、R b 、R c And R is d Independently optionally substituted with 1,2, 3, 4, 5 or 6R g And (3) substitution.
In some embodiments, each R f And R is g Independently is fluorine, chlorine, bromine, iodine, oxo, hydroxy, amino, nitro, cyano, methyl, ethyl, n-propyl, isopropyl, n-butyl, -CH 2 OH, monofluoromethyl, difluoromethyl, trifluoromethyl, monofluoroethyl, 1-difluoroethyl, 1, 2-difluoroethyl, phenyl, pyridyl, pyrimidinyl, pyrazolyl, oxiranyl, oxetanyl, tetrahydrofuranyl, tetrahydropyranyl, Cyclopropyl, cyclobutyl, cyclohexyl, methoxy, ethoxy, n-propoxyAn isopropoxy group, a monofluoromethoxy group, a difluoromethoxy group or a trifluoromethoxy group.
In another aspect, the present invention relates to a pharmaceutical composition comprising a compound of formula (I) or a stereoisomer, a geometric isomer, a tautomer, a nitroxide, a hydrate, a solvate, a metabolite, a pharmaceutically acceptable salt or a prodrug thereof, according to the present invention, and a pharmaceutically acceptable excipient, carrier, adjuvant or combination thereof.
In some embodiments, the pharmaceutical compositions of the present invention comprise other agents that prevent or treat inflammatory syndromes or autoimmune diseases or any combination thereof.
In another aspect, the invention relates to the use of a compound of formula (I) or a pharmaceutical composition thereof for the manufacture of a medicament for the prevention or treatment of an inflammatory or autoimmune disease mediated by roryt in a mammal.
In some embodiments, the present invention relates to the use of a compound of formula (I) or a pharmaceutical composition thereof for the preparation of a medicament for the prevention or treatment of psoriasis, rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis, inflammatory bowel disease, colitis, ulcerative colitis, rheumatoid arthritis, autoimmune ocular disease, ankylosing spondylitis, asthma, chronic obstructive pulmonary disease, osteoarthritis, allergic rhinitis, allergic dermatitis, crohn's disease, or kawasaki disease.
In another aspect, the present invention relates to methods for the preparation, isolation and purification of compounds of formula (I).
Biological test results show that the compound provided by the invention has better inhibition activity on RORγt and good pharmacokinetic characteristics.
Any of the embodiments of any of the aspects of the invention may be combined with other embodiments, provided that they do not contradict. Furthermore, in any of the embodiments of any of the aspects of the present invention, any technical feature may be applied to the technical feature in other embodiments as long as they do not contradict.
The foregoing merely outlines certain aspects of the invention and is not limited in this regard. These and other aspects are described more fully below.
Detailed description of the invention
Definitions and general terms
Reference will now be made in detail to certain embodiments of the invention, examples of which are illustrated in the accompanying structural and chemical formulas. The invention is intended to cover all alternatives, modifications and equivalents, which may be included within the scope of the invention as defined by the appended claims. Those skilled in the art will recognize that many methods and materials similar or equivalent to those described herein can be used in the practice of the present invention. The present invention is in no way limited to the methods and materials described herein. In the event of one or more of the incorporated references, patents and similar materials differing from or contradictory to the present application (including but not limited to defined terms, term application, described techniques, etc.), the present application controls.
It should further be appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the invention, which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable sub-combination.
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. All patents and publications referred to herein are incorporated by reference in their entirety.
The following definitions as used herein should be applied unless otherwise indicated. For the purposes of the present invention, chemical elements are in accordance with CAS version of the periodic Table of the elements, and handbook of chemistry and physics, 75 th edition, 1994. In addition, general principles of organic chemistry may be referenced to the descriptions in "Organic Chemistry", thomas Sorrell, university Science Books, sausalato:1999, and "March's Advanced Organic Chemistry" by Michael b.smith and Jerry March, john Wiley & Sons, new york:2007, the entire contents of which are incorporated herein by reference.
The articles "a," "an," and "the" are intended to include "at least one" or "one or more" unless the context clearly dictates otherwise or otherwise. Thus, as used herein, these articles refer to one or to more than one (i.e., to at least one) object. For example, "a component" refers to one or more components, i.e., more than one component is contemplated as being employed or used in embodiments of the described embodiments.
The term "subject" as used herein refers to an animal. Typically the animal is a mammal. The subject, for example, also refers to a primate (e.g., human, male or female), cow, sheep, goat, horse, dog, cat, rabbit, rat, mouse, fish, bird, and the like. In certain embodiments, the subject is a primate. In other embodiments, the subject is a human.
The term "patient" as used herein refers to a human (including adults and children) or other animals. In some embodiments, "patient" refers to a human.
The term "comprising" is an open-ended expression, i.e., including what is indicated by the invention, but not excluding other aspects.
"stereoisomers" refer to compounds having the same chemical structure but different arrangements of atoms or groups in space. Stereoisomers include enantiomers, diastereomers, conformational isomers (rotamers), geometric isomers (cis/trans), atropisomers, and the like.
"chiral" is a molecule that has properties that do not overlap with its mirror image; and "achiral" refers to a molecule that may overlap with its mirror image.
"enantiomer" refers to two isomers of a compound that do not overlap but are in mirror image relationship to each other.
"diastereoisomers" refers to stereoisomers which have two or more chiral centers and whose molecules are not mirror images of each other. Diastereomers have different physical properties, such as melting point, boiling point, spectral properties, and reactivity. The diastereomeric mixture may be separated by high resolution analytical procedures such as electrophoresis and chromatography, e.g., HPLC.
The stereochemical definitions and rules used in the present invention generally follow S.P. Parker, ed., mcGraw-Hill Dictionary of Chemical Terms (1984) McGraw-Hill Book Company, new York; and Eliel, e.and Wilen, s., "Stereochemistry of Organic Compounds", john Wiley & Sons, inc., new York,1994.
Many organic compounds exist in optically active form, i.e., they have the ability to rotate the plane of plane polarized light. In describing optically active compounds, the prefixes D and L or R and S are used to represent the absolute configuration of the molecule with respect to one or more of its chiral centers. The prefixes d and l or (+) and (-) are symbols for specifying the rotation of plane polarized light by a compound, where (-) or l indicates that the compound is left-handed. The compound prefixed with (+) or d is dextrorotatory. One particular stereoisomer is an enantiomer, and a mixture of such isomers is referred to as an enantiomeric mixture. A50:50 mixture of enantiomers is referred to as a racemic mixture or racemate, which can occur when there is no stereoselectivity or stereospecificity in a chemical reaction or process.
Any asymmetric atom (e.g., carbon, etc.) of the disclosed compounds may exist in racemic or enantiomerically enriched form, such as in the (R) -, (S) -or (R, S) -configuration. In certain embodiments, each asymmetric atom has at least 50% enantiomeric excess, at least 60% enantiomeric excess, at least 70% enantiomeric excess, at least 80% enantiomeric excess, at least 90% enantiomeric excess, at least 95% enantiomeric excess, or at least 99% enantiomeric excess in the (R) -or (S) -configuration.
Depending on the choice of starting materials and methods, the compounds of the invention may be present in the form of one of the possible isomers or mixtures thereof, for example racemates and diastereomeric mixtures, depending on the number of asymmetric carbon atoms. Optically active (R) -or (S) -isomers can be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques. If the compound contains a double bond, the substituent may be in the E or Z configuration; if the compound contains a disubstituted cycloalkyl group, the substituents of the cycloalkyl group may have cis or trans configuration.
The resulting mixture of any stereoisomers may be separated into pure or substantially pure geometric isomers, enantiomers, diastereomers, e.g., by chromatography and/or fractional crystallization, depending on the differences in the physicochemical properties of the components.
Any of the resulting racemates of the end products or intermediates can be resolved into the optical enantiomers by methods familiar to those skilled in the art, e.g., by separation of the diastereoisomeric salts thereof obtained, using known methods. The racemic product can also be separated by chiral chromatography, e.g., high performance liquid chromatography using chiral adsorbents (HPLC: column: chiralpak AD-H (4.6 mm x 250mm,5 μm), mobile phase: n-hexane: ethanol=40:60, isocratic elution, flow rate: 1 mL/min). In particular, enantiomers may be prepared by asymmetric synthesis, for example, reference may be made to Jacques, et al, encomers, racemates and Resolutions (Wiley Interscience, new York, 1981); principles of Asymmetric Synthesis (2) nd Ed.Robert E.Gawley,Jeffrey Aubé,Elsevier,Oxford,UK,2012);Eliel,E.L.Stereochemistry of Carbon Compounds(McGraw-Hill,NY,1962);Wilen,S.H.Tables of Resolving Agents and Optical Resolutions p.268(E.L.Eliel,Ed.,Univ.of Notre Dame Press,Notre Dame,IN 1972);Chiral Separation Techniques:A Practical Approach(Subramanian,G.Ed.,Wiley-VCH Verlag GmbH&Co.KGaA,Weinheim,Germany,2007)。
The compounds of the invention may be optionally substituted with one or more substituents, as described in the present invention, such as the compounds of the general formula above, or as specific examples within the examples, subclasses, and classes of compounds encompassed by the invention.
In general, the term "substituted" means that one or more hydrogen atoms in a given structure are replaced with a specific substituent. Unless otherwise indicated, a substituted group may have a substituent substituted at each substitutable position of the group. When more than one position in a given formula can be substituted with one or more substituents selected from a particular group, then the substituents may be the same or different at each position.
The term "unsubstituted" means that the specified group does not carry a substituent.
The term "optionally substituted with … …" may be used interchangeably with the term "unsubstituted or substituted with … …," i.e., the structure is unsubstituted or substituted with one or more substituents described herein. Substituents described herein include, but are not limited to, D, oxo (=o), F, cl, br, I, N 3 、CN、NO 2 、OH、SH、NH 2 Carboxyl, aldehyde, alkyl, haloalkyl, alkenyl, alkynyl, alkoxy, haloalkoxy, alkylamino, carbocyclyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -alkylene-cycloalkyl, cycloalkylamino, -alkylene-alkoxy, and the like.
In addition, unless explicitly indicated otherwise, the descriptions used in this disclosure of the manner in which each … is independently "and" … is independently "and" … is independently "are to be construed broadly as meaning that particular items expressed between the same symbols in different groups do not affect each other, or that particular items expressed between the same symbols in the same groups do not affect each other.
In the various parts of the present specification, substituents of the presently disclosed compounds are disclosed in terms of the type or scope of groups. It is specifically noted that the present invention includes each individual subcombination of the individual members of these group classes and ranges. For example, the term "C 1-6 Alkyl "means in particular methyl, ethyl, C independently disclosed 3 Alkyl, C 4 Alkyl, C 5 Alkyl and C 6 An alkyl group.
In the various parts of the invention, linking substituents are described. When the structure clearly requires a linking group, the markush variables recited for that group are understood to be linking groups. For example, if the structure requires a linking group and the markush group definition for that variable enumerates an "alkyl" or "aryl" group, it will be understood that the "alkyl" or "aryl" represents a linked alkylene group or arylene group, respectively.
The term "alkyl" or "alkyl group" as used herein refers to a saturated, straight or branched chain monovalent hydrocarbon group, wherein the alkyl group may be optionally substituted with one or more substituents as described herein. Unless otherwise specified, alkyl groups contain 1 to 20 carbon atoms. In one embodiment, the alkyl group contains 1 to 12 carbon atoms; in another embodiment, the alkyl group contains 3 to 12 carbon atoms; in another embodiment, the alkyl group contains 1 to 6 carbon atoms; in yet another embodiment, the alkyl group contains 1 to 4 carbon atoms. In yet another embodiment, the alkyl group contains 1 to 3 carbon atoms.
Examples of alkyl groups include, but are not limited to, methyl (Me, -CH 3 ) Ethyl (Et, -CH) 2 CH 3 ) N-propyl (n-Pr, -CH) 2 CH 2 CH 3 ) Isopropyl (i-Pr, -CH (CH) 3 ) 2 ) N-butyl (n-Bu, -CH) 2 CH 2 CH 2 CH 3 ) Isobutyl (i-Bu, -CH) 2 CH(CH 3 ) 2 ) Sec-butyl (s-Bu, -CH (CH) 3 )CH 2 CH 3 ) Tert-butyl (t-Bu, -C (CH) 3 ) 3 ) N-pentyl (-CH) 2 CH 2 CH 2 CH 2 CH 3 ) 2-pentyl (-CH (CH) 3 )CH 2 CH 2 CH 3 ) 3-pentyl (-CH (CH) 2 CH 3 ) 2 ) 2-methyl-2-butyl (-C (CH) 3 ) 2 CH 2 CH 3 ) 3-methyl-2-butyl (-CH (CH) 3 )CH(CH 3 ) 2 ) 3-methyl-1-butyl (-CH) 2 CH 2 CH(CH 3 ) 2 ) 2-methyl-1-butyl (-CH) 2 CH(CH 3 )CH 2 CH 3 ) N-hexyl (-CH) 2 CH 2 CH 2 CH 2 CH 2 CH 3 ) 2-hexyl (-CH (CH) 3 )CH 2 CH 2 CH 2 CH 3 ) 3-hexyl (-CH (CH) 2 CH 3 )(CH 2 CH 2 CH 3 ) 2-methyl-2-pentyl (-C (CH) 3 ) 2 CH 2 CH 2 CH 3 ) 3-methyl-2-pentyl (-CH (CH) 3 )CH(CH 3 )CH 2 CH 3 ) 4-methyl-2-pentyl (-CH (CH) 3 )CH 2 CH(CH 3 ) 2 ) 3-methyl-3-pentyl (-C (CH) 3 )(CH 2 CH 3 ) 2 ) 2-methyl-3-pentyl (-CH (CH) 2 CH 3 )CH(CH 3 ) 2 ) 2, 3-dimethyl-2-butyl (-C (CH) 3 ) 2 CH(CH 3 ) 2 ) 3, 3-dimethyl-2-butyl (-CH (CH) 3 )C(CH 3 ) 3 ) N-heptyl, n-octyl, and the like.
The term "alkylene" means a saturated divalent hydrocarbon group resulting from the removal of two hydrogen atoms from a saturated straight or branched hydrocarbon group. Unless otherwise specified, alkylene groups contain 1 to 12 carbon atoms. In one embodiment, the alkylene group contains 1 to 6 carbon atoms; in another embodiment, the alkylene group contains 1 to 4 carbon atoms; in yet another embodiment, the alkylene group contains 1 to 3 carbon atoms; in yet another embodiment, the alkylene group contains 1 to 2 carbon atoms. Examples of this include methylene (-CH) 2 (-), ethylene (-CH) 2 CH 2 (-), isopropylidene (-CH (CH) 3 )CH 2 (-), etc.
The term "alkoxy" means that the alkyl group is attached to the remainder of the molecule through an oxygen atom, wherein the alkyl group has the meaning as described herein. Unless otherwise specified, the alkoxy groups contain 1 to 12 carbon atoms. In one embodiment, the alkoxy group contains 1 to 6 carbon atoms; in another embodiment, the alkoxy group contains 1 to 4 carbon atoms; in yet another embodiment, the alkoxy group contains 1 to 3 carbon atoms. The alkoxy group may be optionally substituted with one or more substituents described herein.
Examples of alkoxy groups include, but are not limited to, methoxy (MeO, -OCH) 3 ) Ethoxy (EtO, -OCH) 2 CH 3 ) 1-propoxy (n-PrO, n-propoxy, -OCH) 2 CH 2 CH 3 ) 2-propoxy (i-PrO, i-propoxy, -OCH (CH) 3 ) 2 ) 1-butoxy (n-BuO, n-butoxy, -OCH) 2 CH 2 CH 2 CH 3 ) 2-methyl-l-propoxy (i-BuO, i-butoxy, -OCH) 2 CH(CH 3 ) 2 ) 2-butoxy (s-BuO, s-butoxy, -OCH (CH) 3 )CH 2 CH 3 ) 2-methyl-2-propoxy (t-BuO, t-butoxy, -OC (CH) 3 ) 3 ) 1-pentoxy (n-pentoxy, -OCH) 2 CH 2 CH 2 CH 2 CH 3 ) 2-pentoxy (-OCH (CH) 3 )CH 2 CH 2 CH 3 ) 3-pentoxy (-OCH (CH) 2 CH 3 ) 2 ) 2-methyl-2-butoxy (-OC (CH) 3 ) 2 CH 2 CH 3 ) 3-methyl-2-butoxy (-OCH (CH) 3 )CH(CH 3 ) 2 ) 3-methyl-l-butoxy (-OCH) 2 CH 2 CH(CH 3 ) 2 ) 2-methyl-l-butoxy (-OCH) 2 CH(CH 3 )CH 2 CH 3 ) And so on.
The term "alkylamino" or "alkylamino" includes "N-alkylamino" and "N, N-dialkylamino" in which the amino groups are each independently substituted with one or two alkyl groups; the alkyl groups have the meaning described in the present invention. Some of these are, for example, alkylamino groups of one or two C 1-6 Alkyl groups are attached to lower alkylamino groups formed on the nitrogen atom. Still other embodiments are where the alkylamino group is one or two C 1-4 Is linked to an alkylamino group formed on the nitrogen atom. Other embodiments are where the alkylamino group is one or two C 1-3 Is linked to an alkylamino group formed on the nitrogen atom. Suitable alkylamino groups may be mono-or dialkylamino, such examples include, but are not limited to, N-methylamino (methylamino), N-ethylamino (ethylamino), N-dimethylamino (dimethylamino), N-diethylamino (diethylamino), and the like.
The term "haloalkyl", "haloalkoxy" or "haloalkylamino" means an alkyl, alkoxy or alkylamino group substituted with one or more halogen atoms, wherein alkyl, alkoxy or alkylamino groups have the meaning as described herein, such examples include, but are not limited to, difluoromethyl, trifluoromethyl, 2-trifluoroethyl, 2, 3-tetrafluoropropyl, difluoromethoxy, trifluoromethoxy, trifluoromethylamino and the like.
The term "hydroxy-substituted haloalkyl" means that the haloalkyl is substituted with one or more hydroxy groups, wherein haloalkyl has the meaning as described herein, such examples include, but are not limited to, 2-hydroxy-1, 3-hexafluoroisopropyl, and the like.
The term "hydroxy-substituted alkyl" means that the alkyl group is substituted with one or more hydroxy groups, wherein the alkyl group has the meaning as described herein, examples of which include, but are not limited to, hydroxymethyl, 1-hydroxyethyl, 2-hydroxyethyl, and the like. E.g. "hydroxy-substituted C 1-4 Alkyl "means an alkyl group having 1 to 4 carbon atoms substituted with one or more hydroxyl groups.
The term "alkoxyalkyl" or "alkoxy-alkylene" means an alkoxy group attached to the remainder of the molecule through an alkyl group, wherein the alkoxy, alkyl and alkylene groups have the meanings as described herein. Examples include, but are not limited to, methoxymethyl, ethoxymethyl, isopropoxymethyl, 1-methoxyethyl, 2-methoxyethyl, and the like.
The term "cycloalkyl" denotes a monovalent or polyvalent saturated monocyclic, bicyclic or tricyclic hydrocarbon group containing 3 to 12 carbon atoms. In one embodiment, cycloalkyl groups contain 7 to 12 carbon atoms; in yet another embodiment, cycloalkyl groups contain 3 to 8 carbon atoms; in yet another embodiment, cycloalkyl groups contain 3 to 6 carbon atoms. The cycloalkyl groups may independently be unsubstituted or substituted with one or more substituents described herein.
The term "cycloalkylamino" includes "N-cycloalkylamino" and "N, N-dicycloalkylamino" in which the amino groupEach independently substituted with one or two cycloalkyl groups; the cycloalkyl has the meaning described in the present invention. Some of these embodiments are those wherein the cycloalkylamino group is one or two C 3-8 Cycloalkyl groups are attached to nitrogen atoms to form cycloalkylamino groups. Other embodiments are where the cycloalkylamino group is one or two C 3-6 A cycloalkylamino group formed by linking a cycloalkyl group of (a) to a nitrogen atom. Suitable cycloalkylamino groups may be monocycloalkylamino or bicycloalkylamino, such examples include, but are not limited to, N-cyclopropylamino, N-cyclobutylamino, N-cyclohexylamino, N-dicyclohexylamino, and the like.
The terms "cycloalkylalkyl" and "cycloalkyl-alkylene" are used interchangeably and each denote a cycloalkyl group attached to the remainder of the molecule through an alkyl group, where cycloalkyl and alkyl groups have the meanings as described herein. Examples include, but are not limited to, cyclopropylmethylene, cyclobutylmethylene, cyclopentylmethylene, cyclohexylmethylene, cyclopropylethylene, cyclobutylethylene, cyclopentylethylene, cyclohexylethylene, and the like.
The terms "heterocyclyl" and "heterocycle" are used interchangeably herein to refer to a saturated or partially unsaturated, non-aromatic, monovalent or multivalent monocyclic ring comprising 3 to 12 ring atoms, at least one of which is selected from the group consisting of nitrogen, sulfur and oxygen atoms. Wherein in some embodiments 3 to 12 ring atoms of the heterocyclyl contain 2 to 9 carbon atoms; in still other embodiments, 3 to 12 ring atoms of the heterocyclyl contain 2 to 8 carbon atoms; in still other embodiments, 3 to 12 ring atoms of the heterocyclyl contain 2 to 6 carbon atoms; in still other embodiments, 3 to 12 ring atoms of the heterocyclyl contain 2 to 5 carbon atoms. Unless otherwise indicated, a heterocyclic group may be attached to other groups in the molecule through a carbon atom, may be attached to other groups in the molecule through a nitrogen atom, and-CH 2 The group may optionally be replaced by-C (=o) -. The sulfur atom of the ring may optionally be oxidized to an S-oxide. The nitrogen atom of the ring may optionally be oxidized to an N-oxide. Examples of heterocyclyl groups include, but are not limited to: oxirane and azaCyclobutyl, oxetanyl, thietanyl, pyrrolidinyl, 2-pyrrolinyl, 3-pyrrolinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, dihydrothienyl, 1, 3-dioxacyclopentyl, dithiocyclopentyl, tetrahydropyranyl, dihydropyranyl, 2H-pyranyl, 4H-pyranyl, tetrahydrothiopyranyl, piperidinyl, morpholinyl, thiomorpholinyl, piperazinyl, dioxanyl, dithianyl, thiazalkyl, homopiperazinyl, homopiperidinyl, diazepanyl, oxepinyl, thietanyl, oxazazinylRadical, diaza->Radical, thiazal->A base. In heterocyclic groups-CH 2 Examples of the substitution of the-group by-C (=o) -include, but are not limited to, 2-oxo-pyrrolidinyl, oxo-1, 3-thiazolidinyl, 2-piperidonyl, 3, 5-dioxopiperidyl, pyrimidinedionyl. Examples of sulfur atoms in the heterocyclic group that are oxidized include, but are not limited to, sulfolane, 1-dioxothiomorpholino. The heterocyclyl group may be optionally substituted with one or more substituents described herein.
The term "spirocarbocyclyl" refers to a saturated or partially unsaturated, non-aromatic, mono-or polyvalent bi-or tricyclic ring comprising 5 to 12 ring atoms, wherein the rings share a carbon atom with the rings, and wherein the ring atoms are all carbon atoms. Unless otherwise indicated, spiro groups may be attached to other groups in the molecule through carbon atoms, and-CH 2 The group may optionally be replaced by-C (=o) -. Examples of spirocycles include, but are not limited to:
the terms "spiroheterocyclyl" and "spiroheterocycle" are used interchangeably herein to refer to a saturated or partially unsaturated, non-aromatic, monovalent or polyvalent bicyclic or tricyclic ring comprising 5 to 12 ring atoms, wherein at least one ring atom is selected from the group consisting of nitrogen, sulfur and oxygen atoms, and the ring shares a carbon atom with the ring. Wherein in some embodiments, from 4 to 9 carbon atoms are contained in 5 to 12 ring atoms of the spiroheterocyclyl; in still other embodiments, from 4 to 8 carbon atoms are contained in 5 to 12 ring atoms of the spiroheterocyclyl; in still other embodiments, the spiroheterocyclyl contains 4 to 6 carbon atoms in 5 to 12 ring atoms. Unless otherwise indicated, a spiroheterocyclyl group may be attached to other groups in the molecule through a carbon atom, may be attached to other groups in the molecule through a nitrogen atom, and-CH 2 The group may optionally be replaced by-C (=o) -. The sulfur atom of the ring may optionally be oxidized to an S-oxide. The nitrogen atom of the ring may optionally be oxidized to an N-oxide. Examples of spiroheterocycles include, but are not limited to:
the term "fused carbocyclyl" refers to a saturated or partially unsaturated, non-aromatic, mono-or polyvalent bi-or tricyclic ring containing 4 to 12 ring atoms, wherein the rings share a ring edge with the rings, and wherein the ring atoms are all carbon atoms. Unless otherwise indicated, fused ring groups may be attached to other groups in the molecule through carbon atoms, and-CH 2 The group may optionally be replaced by-C (=o) -. Examples of fused rings include, but are not limited to:
the terms "fused heterocyclyl" and "fused heterocycle" are used interchangeably herein to refer to a saturated or partially unsaturated, non-aromatic, monovalent or polyvalent bicyclic or tricyclic ring comprising 4 to 12 ring atoms, wherein at least one ring atom is selected from the group consisting of nitrogen, sulfur and oxygen atoms, and the ring and ring share a ring edge. Wherein, in some embodimentsThe 4-12 ring atoms of the condensed heterocyclic group contain 3-9 carbon atoms; in still other embodiments, from 3 to 8 carbon atoms in 4 to 12 ring atoms of the fused heterocyclic group; in still other embodiments, the fused heterocyclic group contains 3 to 6 carbon atoms in 4 to 12 ring atoms. Unless otherwise indicated, a fused heterocyclic group may be attached to other groups in the molecule through a carbon atom, may be attached to other groups in the molecule through a nitrogen atom, and-CH 2 The group may optionally be replaced by-C (=o) -. The sulfur atom of the ring may optionally be oxidized to an S-oxide. The nitrogen atom of the ring may optionally be oxidized to an N-oxide. Examples of fused heterocycles include, but are not limited to:
the term "unsaturated" as used in the present invention means that the group contains one or more unsaturations.
The term "heteroatom" refers to O, S, N, P and Si, including N, S and any oxidation state forms of P; primary, secondary, tertiary and quaternary ammonium salt forms; or a form in which the hydrogen on the nitrogen atom of the heterocycle is substituted, for example, N (like N in 3, 4-dihydro-2H-pyrrolyl), NH (like NH in pyrrolidinyl) or NR (like NR 'in N-substituted pyrrolidinyl), R' is a substituent according to the invention.
The term "halogen" or "halogen atom" refers to a fluorine atom (F), a chlorine atom (Cl), a bromine atom (Br) or an iodine atom (I).
The term "cyano" or "CN" means a cyano structure, which group may be attached to other groups.
The term "nitro" or "NO 2 "means a nitro structure, which may be attached to other groups.
The term "amino" or "NH 2 "means an amino structure, which may be attached to other groups.
The term "aryl" means a monocyclic, bicyclic or tricyclic, fully carbocyclic ring system containing 6 to 14 ring atoms, or 6 to 12 ring atoms, or 6 to 10 ring atoms, wherein at least one ring is aromatic and has one or more attachment points attached to the remainder of the molecule. The term "aryl" may be used interchangeably with the term "aromatic ring". In one embodiment, aryl groups are carbocyclic ring systems consisting of 6 to 10 ring atoms and containing at least one aromatic ring therein. Examples of aryl groups may include phenyl, naphthyl and anthracenyl. The aryl groups may independently be optionally substituted with one or more substituents described herein.
The term "heteroaryl" means a single, double, or triple ring containing 5-12 ring atoms, wherein at least one ring is aromatic and at least one ring contains one or more heteroatoms and has one or more attachment points to the remainder of the molecule. The term "heteroaryl" may be used interchangeably with the term "heteroaromatic ring" or "heteroaromatic compound". Wherein in some embodiments, from 1 to 9 carbon atoms are contained in 5 to 12 ring atoms of the heteroaryl group; in still other embodiments, from 1 to 7 carbon atoms in 5 to 12 ring atoms of the heteroaryl group; in still other embodiments, from 1 to 5 carbon atoms are contained in 5 to 12 ring atoms of the heteroaryl group; the heteroaryl group is optionally substituted with one or more substituents described herein. In one embodiment, heteroaryl is a heteroaryl group consisting of 5-12 ring atoms comprising 1, 2, 3, or 4 heteroatoms independently selected from O, S and N; in one embodiment, heteroaryl is a heteroaryl group consisting of 5 to 10 ring atoms comprising 1, 2, 3, or 4 heteroatoms independently selected from O, S and N; in another embodiment, heteroaryl is a heteroaryl group consisting of 5-6 ring atoms containing 1, 2, 3, or 4 heteroatoms independently selected from O, S and N.
Examples of heteroaryl groups include, but are not limited to, furyl (e.g., 2-furyl, 3-furyl), imidazolyl (e.g., N-imidazolyl, 2-imidazolyl, 4-imidazolyl, 5-imidazolyl), isoxazolyl (e.g., 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl), oxazolyl (e.g., 2-oxazolyl, 4-oxazolyl, 5-oxazolyl), oxadiazolyl (e.g., 1,2, 3-oxadiazolyl, 1,2, 5-oxadiazolyl, 1,2, 4-oxadiazolyl), oxazolyl (e.g., 1,2,3, 4-oxazolyl), thiazolyl (e.g., 2-thiazolyl, 4-thiazolyl, 5-thiazolyl), isothiazolyl, 2-thiadiazolyl (e.g., 1,3, 4-thiadiazolyl) 1,2, 3-thiadiazolyl, 1,2, 5-thiadiazolyl), thiatriazolyl (e.g., 1,2,3, 4-thiatriazolyl), tetrazolyl (e.g., 2H-1,2,3, 4-tetrazolyl), 1H-1,2,3, 4-tetrazolyl), triazolyl (e.g., 2H-1,2, 3-triazolyl, 1H-1,2, 4-triazolyl, 4H-1,2, 4-triazolyl), thienyl (e.g., 2-thienyl, 3-thienyl), 1H-pyrazolyl (e.g., 1H-pyrazol-3-yl, 1H-pyrazol-4-yl, 1H-pyrazol-5-yl), 1,2, 3-thiodiazolyl, 1,3, 4-thiodiazolyl, 1,2, 5-thiodiazolyl, pyrrolyl (e.g., N-pyrrolyl), 2-pyrrolyl, 3-pyrrolyl), pyridinyl (e.g., 2-pyridinyl, 3-pyridinyl, 4-pyridinyl), pyrimidinyl (e.g., 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl), pyridazinyl (e.g., 3-pyridazinyl, 4-pyridazinyl), 2-pyrazinyl, triazinyl (e.g., 1,3, 5-triazine), tetrazinyl (e.g., 1,2,4, 5-tetrazine, 1,2,3, 5-tetrazine); the following bicyclic rings are also included, but are in no way limited to: benzimidazolyl, benzofuranyl, benzothienyl, indolyl (e.g., 2-indolyl), purinyl, quinolinyl (e.g., 2-quinolinyl, 3-quinolinyl, 4-quinolinyl), isoquinolinyl (e.g., 1-isoquinolinyl, 3-isoquinolinyl, or 4-isoquinolinyl), imidazo [1,2-a ] pyridinyl, pyrazolo [1,5-a ] pyrimidinyl, imidazo [1,2-b ] pyridazinyl, [1,2,4] triazolo [4,3-b ] pyridazinyl, [1,2,4] triazolo [1,5-a ] pyrimidinyl, [1,2,4] triazolo [1,5-a ] pyridinyl, and the like.
The term "carboxy", whether used alone or in combination with other terms, such as "carboxy", means-CO 2 H is formed; the term "carbonyl", whether used alone or in combination with other terms, such as "aminocarbonyl" or "acyloxy", means- (C=O) -.
The term "j-k membered" means that the cyclic group consists of j-k ring atoms including carbon atoms and/or O, N, S, P and like heteroatoms; each of j and k is independently any non-zero natural number, and k > j; the term "j-k" includes j, k and any natural number therebetween. For example, "3-6 membered", "3-7 membered", "5-10 membered", "5-12 membered", "4-12 membered" or "5-6 membered" means that the cyclic group is composed of 3-6 (i.e., 3, 4, 5 or 6), 3-7 (i.e., 3, 4, 5, 6 or 7), 5-10 (i.e., 5, 6, 7, 8, 9 or 10), 5-12 (i.e., 5, 6, 7, 8, 9, 10, 11 or 12), 4-12 (i.e., 4, 5, 6, 7, 8, 9, 10, 11 or 12) or 5-6 (i.e., 5 or 6) ring atoms, which ring atoms include heteroatoms such as carbon atoms and/or O, N, S, P.
As described herein, the attachment of a linkage to a ring system (as shown in formula c) represents that the ring may be attached to the remainder of the molecule at any available position on the ring system via the linkage. Formula c represents that the ring may be attached to the remainder of the molecule at any possible attachment position on the ring, including, but not limited to, those shown in formulas c 1-c 6.
The term "protecting group" or "PG" refers to a substituent that is commonly used to block or protect a particular functionality when reacted with other functional groups. For example, by "protecting group for an amino group" is meant a substituent attached to the amino group to block or protect the functionality of the amino group in the compound, suitable amino protecting groups include acetyl, trifluoroacetyl, t-butoxycarbonyl (BOC ), benzyloxycarbonyl (CBZ ) and 9-fluorenylmethoxycarbonyl (Fmoc). Similarly, "hydroxy protecting group" refers to the functionality that a substituent of a hydroxy group serves to block or protect the hydroxy group, and suitable protecting groups include acetyl and silyl. "carboxyl protecting group" refers to the functionality of a substituent of a carboxyl group to block or protect the carboxyl group, and typically the carboxyl protecting group includes-CH 2 CH 2 SO 2 Ph, cyanoethyl, 2- (trimethylsilyl) ethyl, 2- (trimethylsilyl) ethoxymethyl, 2- (p-toluenesulfonyl) ethyl, 2- (p-nitrobenzenesulfonyl) ethyl, 2- (diphenylphosphino) ethyl, nitroethyl, and the like. General description of protecting groups can be found in the literature: t W.Greene, protective Groups in Organic Synthesis, john Wiley&Sons,New York,1991;and P.J.Kocienski,Protecting Groups,Thieme,Stuttgart,2005.
The term "prodrug" as used herein, means a compound that is converted in vivo Is a compound shown in a formula (I). Such conversion is effected by hydrolysis of the prodrug in the blood or enzymatic conversion to the parent structure in the blood or tissue. The prodrug of the invention can be ester, and in the prior invention, the ester can be phenyl ester, aliphatic (C 1 -C 24 ) Esters, acyloxymethyl esters, carbonates, carbamates and amino acid esters. For example, one compound of the invention may contain a hydroxyl group, i.e., it may be acylated to provide the compound in a prodrug form. Other prodrug forms include phosphates, such as those obtained by phosphorylation of a hydroxyl group on the parent. For a complete discussion of prodrugs, reference may be made to the following documents: higuchi and V.stilla, pro-drugs as Novel Delivery Systems, vol.14 of the A.C.S. symposium Series, edward B.Roche, ed., bioreversible Carriers in Drug Design, american Pharmaceutical Association and Pergamon Press,1987,J.Rautio et al, prodrug: design and Clinical Applications, nature Review Drug Discovery,2008,7,255-270,and S.J.Hecker et al, prodrugs of Phosphates and Phosphonates, journal of Medicinal Chemistry,2008,51,2328-2345.
"metabolite" refers to a product obtained by metabolizing a specific compound or salt thereof in vivo. The metabolites of a compound may be identified by techniques well known in the art and their activity may be characterized by employing the assay methods as described herein. Such products may be obtained by oxidation, reduction, hydrolysis, amidization, deamination, esterification, degreasing, enzymatic cleavage, etc. of the administered compound. Accordingly, the present invention includes metabolites of compounds, including metabolites produced by contacting a compound of the present invention with a mammal for a period of time sufficient.
As used herein, "pharmaceutically acceptable salts" refers to organic and inorganic salts of the compounds of the present invention. Pharmaceutically acceptable salts are well known in the art, as in the literature: S.M. Berge et al describe pharmaceutically acceptable salts in detail in J.pharmaceutical Sciences,1977,66:1-19The above. Pharmaceutically acceptable non-toxic acid forming salts include, but are not limited to, inorganic acid salts such as hydrochloride, hydrobromide, phosphate, sulfate, perchlorate, and organic acid salts such as acetate, oxalate, maleate, tartrate, citrate, succinate, malonate, or by other methods described in the book literature such as ion exchange. Other pharmaceutically acceptable salts include adipic acid salts, alginates, ascorbates, aspartic acid salts, benzenesulfonates, benzoic acid salts, bisulfate salts, borates, butyric acid salts, camphoric acid salts, cyclopentylpropionates, digluconate, dodecylsulfate, ethanesulfonate, formate, fumaric acid salts, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, caproate, hydroiodic acid salts, 2-hydroxy-ethanesulfonate, lactobionic acid salts, lactate, laurate, lauryl sulfate, malate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, palmitate, pamoate, pectate, persulfate, 3-phenylpropionate, picrate, pivalate, propionate, stearate, thiocyanate, p-toluenesulfonate, undecanoate, valerate, and the like. Salts obtained by suitable bases include alkali metals, alkaline earth metals, ammonium and N + (C 1-4 Alkyl group 4 Is a salt of (a). The present invention also contemplates quaternary ammonium salts formed from any compound containing a group of N. The water-soluble or oil-soluble or dispersible product may be obtained by quaternization. Alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. The pharmaceutically acceptable salts further include suitable, non-toxic ammonium, quaternary ammonium salts and counter-ion forming amine cations, such as halides, hydroxides, carboxylates, sulphates, phosphates, nitrates, C 1-8 Sulfonate and aromatic sulfonate.
"solvate" according to the present invention refers to an association of one or more solvent molecules with a compound according to the present invention. Solvents that form solvates include, but are not limited to, water, isopropanol, ethanol, methanol, dimethylsulfoxide, ethyl acetate, acetic acid, and aminoethanol. The term "hydrate" refers to an association of solvent molecules that are water.
When the solvent is water, the term "hydrate" may be used. In some embodiments, a molecule of a compound of the invention may be associated with a water molecule, such as a monohydrate; in other embodiments, one of the present compound molecules may be associated with more than one water molecule, such as a dihydrate, and in still other embodiments, one of the present compound molecules may be associated with less than one water molecule, such as a hemihydrate. It should be noted that the hydrates described in the present invention retain the biological effectiveness of the compounds in a non-hydrated form.
The term "treating" any disease or disorder as used herein refers to all slowing, interrupting, arresting, controlling or stopping the progression of the disease or disorder, but does not necessarily mean that the symptoms of all diseases or disorders are all absent, and includes prophylactic treatment of such symptoms, particularly in patients susceptible to such diseases or disorders. In some embodiments, ameliorating a disease or disorder (i.e., slowing or preventing or alleviating the progression of the disease or at least one clinical symptom thereof). In other embodiments, "treating" refers to moderating or improving at least one physical parameter, including physical parameters that may not be perceived by the patient. In other embodiments, "treating" refers to modulating a disease or disorder physically (e.g., stabilizing a perceived symptom) or physiologically (e.g., stabilizing a parameter of the body) or both. In other embodiments, "treating" refers to preventing or delaying the onset, or exacerbation of a disease or disorder.
The term "therapeutically effective amount" or "therapeutically effective dose" as used herein refers to an amount of a compound of the invention that is capable of eliciting a biological or medical response in an individual (e.g., reducing or inhibiting enzyme or protein activity, or ameliorating symptoms, alleviating a condition, slowing or delaying the progression of a disease, or preventing a disease, etc.). In one non-limiting embodiment, the term "therapeutically effective amount" refers to an amount that, when administered to an individual, is effective for: (1) At least partially alleviating, inhibiting, preventing and/or ameliorating a disorder or disease that is (i) mediated by roryt, or (ii) associated with roryt activity, or (iii) characterized by aberrant activity of roryt; or (2) reduce or inhibit roryt activity; or (3) reduce or inhibit expression of roryt. In another embodiment, the term "therapeutically effective amount" means an amount that, when administered to a cell, or organ, or non-cellular biological substance, or medium, at least partially reduces or inhibits roryt activity; or an amount of a compound of the invention that is effective to at least partially reduce or inhibit roryt expression.
The terms "administration" and "administering" of a compound as used herein should be understood to mean providing a compound of the invention or a prodrug of a compound of the invention to an individual in need thereof. It will be appreciated that one skilled in the art treats a patient presently suffering from such a disorder or prophylactically treats a patient suffering from such a disorder by administering an effective amount of a compound of the present invention.
The term "composition" as used herein refers to a product comprising the specified ingredients in the specified amounts, as well as any product that results, directly or indirectly, from combination of the specified ingredients in the specified amounts. The meaning of such terms in relation to pharmaceutical compositions includes products comprising the active ingredient(s) and the inert ingredient(s) that make up the carrier, as well as any products that result directly or indirectly from mixing, compounding or aggregation of any two or more ingredients, or from decomposition of one or more ingredients, or from other types of reactions or interactions of one or more ingredients. Accordingly, the pharmaceutical compositions of the present invention include any composition prepared by admixing a compound of the present invention and a pharmaceutically acceptable carrier.
Description of the Compounds of the invention
The invention discloses a sulfonyl ester derivative, pharmaceutically acceptable salt, a pharmaceutical preparation and a composition thereof, which can be used as ROR gamma t inhibitor and has potential application to the treatment of inflammation or autoimmune diseases mediated by ROR gamma t, such as psoriasis, rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis, inflammatory bowel disease, colitis, ulcerative colitis, rheumatic arthritis, autoimmune ophthalmopathy, ankylosing spondylitis, asthma, chronic obstructive pulmonary disease, osteoarthritis, allergic rhinitis, allergic dermatitis, crohn's disease or Kawasaki disease.
In one aspect, the present invention relates to a compound of formula (I) or a stereoisomer, nitroxide, solvate, metabolite, pharmaceutically acceptable salt or prodrug thereof of a compound of formula (I),
wherein, ring A, ring B, L 1 、L 2 、L 3 、R、Z 1 、Z 2 、Z 3 、Z 4 、Z 5 、Z 6 、Z 7 And Z 8 Having the meaning as described in the present invention; * Represents the direction of the linking of the A ring with the right (hetero) aryl group.
In some embodiments, R is C 1-4 Alkyl, C 1-4 Alkoxy, C 1-4 Alkylamino, C 3-6 Cycloalkylamino, -C 1-4 alkylene-C 3-6 Cycloalkyl, C 3-6 Cycloalkyl, C 3-6 Cycloalkyl, C 1-4 Haloalkyl or C 1-4 Haloalkoxy groups.
In other embodiments, R is methyl, ethyl, n-propyl, isopropyl, monofluoromethyl, difluoromethyl, trifluoromethyl, 1, 2-difluoroethyl, methoxy, ethoxy, n-propoxy, isopropoxy, methylamino, ethylamino, dimethylamino, cyclopropylamino, cyclobutylamino, cyclopentylamino, cyclohexylamino, methylenecyclopropyl, methylenecyclobutyl, methylenecyclopentyl, methylenecyclohexyl, ethylenecyclopropyl, ethylenecyclobutyl, ethylenecyclopentyl, ethylenecyclohexyl, cyclopropyl, cyclopentyl, or cyclohexyl.
In some embodiments, Z 1 Is CR (CR) 1 Or N, wherein R 1 Has the meaning as described in the present invention.
In some embodiments, Z 2 Is CR 1 Or N, wherein R 1 Has the meaning as described in the present invention.
In some embodiments, Z 3 Is CR (CR) 1 Or N, wherein R 1 Has the meaning as described in the present invention.
In some embodiments, Z 4 Is CR (CR) 1 Or N, wherein R 1 Has the meaning as described in the present invention.
In some embodiments, Z 5 Is CR (CR) 1 Or N, wherein R 1 Has the meaning as described in the present invention.
In some embodiments, Z 6 Is CR (CR) 1 Or N, wherein R 1 Has the meaning as described in the present invention.
In some embodiments, Z 7 Is CR (CR) 1 Or N, wherein R 1 Has the meaning as described in the present invention.
In some embodiments, Z 8 Is CR (CR) 1 Or N, wherein R 1 Has the meaning as described in the present invention.
In some embodiments, each R 1 Independently hydrogen, deuterium, cyano, fluorine, chlorine, bromine, iodine, C 1-6 Alkyl, C 1-6 Haloalkyl, hydroxy-substituted C 1-6 Alkyl, -C 1-6 alkylene-C 1-6 Alkoxy, C 1-6 Alkoxy, C 3-8 Cycloalkyl or 3-8 membered heterocyclyl.
In other embodiments, each R 1 Independently is hydrogen, cyano, fluoro, chloro, bromo, iodo, methyl, ethyl, n-propyl, isopropyl, difluoromethyl, trifluoromethyl, 1, 2-difluoroethyl, 1-difluoroethyl, methoxy, ethoxy, n-propoxy, isopropoxy, methoxymethylene, ethoxymethylene, n-propoxymethylene, isopropoxy methylene, cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl.
In some embodiments, ring B is C 6-10 Aryl, 5-12 membered heteroaryl, 3-7 membered heterocyclyl, 4-12 membered fused heterocyclyl, 5-12 membered spiroheterocyclyl, 4-12 membered fused carbocyclyl, 5-12 membered spirocarbocyclyl or C 3-8 Cycloalkyl, wherein the B ring is optionally substituted with 1,2, 3 or 4R' s e Substitution; r is R e Has the meaning as described in the present invention.
In other embodiments, the B ring is phenyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, The B ring is optionally substituted with 1,2, 3 or 4R' s e Substitution; r is R e Has the meaning as described in the present invention.
In some embodiments, each R e Independently deuterium, fluorine, chlorine, bromine, iodine, cyano, nitro, hydroxy, amino, C 1-4 Alkyl, C 1-4 Alkoxy, C 3-6 Cycloalkyl, 3-8 membered heterocyclyl, hydroxy substituted C 1-4 Alkyl, hydroxy substituted C 1-4 Haloalkyl, C 1-4 Haloalkyl or C 1-4 Haloalkoxy groups; alternatively, any two adjacent R e And together with the atoms to which they are attached form C 3-8 Cycloalkyl or 3-8 membered heterocyclyl; wherein, C is as follows 1-4 Alkoxy optionally substituted by 1 3-to 6-membered heterocyclyl or C 3-8 Cycloalkyl substitution.
In other embodiments, each R e Independently deuterium, fluorine, chlorine, bromine, iodine, cyano, nitro, hydroxy, amino, methyl, ethyl, n-propyl, isopropyl, hydroxymethyl, 2-hydroxyethyl, 3-hydroxy-n-propyl, 2-hydroxy-1, 3-hexafluoroisopropyl, monofluoromethyl, difluoromethyl, trifluoromethyl, 1, 2-difluoroethyl, methoxy, ethoxy, n-propoxy, t-butoxy, monofluoromethoxy, difluoromethoxy, trifluoromethoxy or 1, 2-difluoroethoxy; wherein said methoxy, ethoxy, n-propoxy, t-butoxy is optionally substituted with 1 member selected from the group consisting of oxiranyl, azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, pyrazolidinyl, imidazolidinyl, tetrahydrofuranyl, tetrahydrothienyl, 1, 3-dioxacyclopentyl, dithiocyclopentyl Substituted with tetrahydropyranyl, 4H-pyranyl, tetrahydrothiopyranyl, piperidinyl, morpholinyl, thiomorpholinyl and piperazinyl.
In some embodiments, L 1 is-S (=O) 2 -NH-、-NH-S(=O) 2 -S (=o) -NH-, -NH-S (=o) -, -C (=o) NH-, or-NHC (=o) -.
In some embodiments, L 2 Is CR (CR) 3 R 4 The method comprises the steps of carrying out a first treatment on the surface of the Wherein R is 3 And R is 4 Having the meaning described in the present invention.
In some embodiments, R 3 C substituted by hydrogen or hydroxy 1-6 C substituted by alkyl or cyano 1-6 An alkyl group; r is R 4 C substituted by hydrogen or hydroxy 1-6 C substituted by alkyl or cyano 1-6 An alkyl group.
In other embodiments, R 3 Is hydrogen, hydroxymethyl, hydroxyethyl, hydroxy-n-propyl, cyanomethyl, cyanoethyl or cyano-n-propyl; r is R 4 Is hydrogen, hydroxymethyl, hydroxyethyl, hydroxy-n-propyl, cyanomethyl, cyanoethyl or cyano-n-propyl.
In some embodiments, L 3 Is carbonyl, -S-, -O-, -NR 5 -or-CR 6 R 7 -; wherein R is 5 、R 6 And R is 7 Having the meaning described in the present invention.
In some embodiments, R 5 Is hydrogen, C 1-6 Alkyl, C 1-6 Haloalkyl, C 3-8 Cycloalkyl or 3-6 membered heterocyclyl; each R is 6 And R is 7 Independently hydrogen, C 1-6 Alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy or 3-6 membered heterocyclyl.
In other embodiments, R 5 Is hydrogen, methyl, ethyl, isopropyl, n-propyl, isobutyl, tert-butyl, n-butyl, monofluoromethyl, difluoromethyl, trifluoromethyl, 1, 2-difluoroethyl, 1-difluoroethyl, methoxy, ethoxy, n-propoxy, isopropoxy, cyclopropyl, cyclobutyl or azetidinyl;
each R is 6 And R is 7 Independently is hydrogen, methyl, ethyl, isopropyl, n-propyl, isobutyl, tert-butyl, n-butyl, monofluoromethyl, difluoromethyl, trifluoromethyl, 1, 2-difluoroethyl, 2-difluoroethyl, methoxy, ethoxy, n-propoxy, isopropoxy, monofluoromethoxy, difluoromethoxy, trifluoromethoxy, 1, 2-difluoroethoxy, 2-difluoroethoxy, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, azetidinyl, oxetanyl, tetrahydrofuranyl, tetrahydropyranyl, pyrrolidinyl, piperidinyl or piperazinyl.
In some embodiments, the a ring is a 6 membered heterocycle, further said 6 membered heterocycle optionally being substituted with 1,2, 3 or 4R 2 Substitution; wherein R is 2 Having the meaning described in the present invention.
In other embodiments, the A ring is Further said A ring is optionally substituted with 1,2, 3 or 4R' s 2 Substitution; wherein R is 2 Having the meaning described in the present invention.
In some embodiments, each R 2 Independently is fluoro, chloro, bromo, iodo, hydroxy, oxo (=o), amino, nitro, cyano, C 1-6 Alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, C 6-10 Aryl, C 3-8 Cycloalkyl, -C 0-6 alkylene-OR a 、-C 0-4 Alkylene- (C (=O)) m -R b 、-C 0-4 Alkylene- (O) n -(C(=O)) m -NR c R d 5-to 10-membered heteroaryl, -C 0-4 Alkylene- (3-7 membered heterocyclyl), -C 0-4 Alkylene- (5-12 membered spiroheterocyclyl) or-C 0-4 Alkylene- (4-12 membered fused heterocyclyl); the R is 2 Optionally by 1, 2 or 3R f Substitution; alternatively, two R's, optionally attached to the same carbon atom 2 And the carbon atoms to which they are attached together form C 3-6 Cycloalkyl radicalsOr a 4-6 membered heterocycle; wherein, m, n and R a 、R b 、R c 、R d And R is f Having the meaning described in the present invention.
In some embodiments, each R a Independently hydrogen, C 1-4 Alkyl, C 1-4 Haloalkyl, C 6-10 Aryl, C 3-6 Cycloalkyl, 5-7 membered heteroaryl, or 3-7 membered heterocyclyl; each R is b 、R c And R is d Independently hydrogen, hydroxy, C 1-4 Alkyl, C 1-4 Haloalkyl, C 1-4 Alkoxy, C 6-10 Aryl, C 3-6 Cycloalkyl, 3-7 membered heterocyclyl or 5-12 membered spiroheterocyclyl; or R is c 、R d And the N atoms to which they are attached together form a 4-7 membered heterocyclic ring;
wherein each R is a 、R b 、R c And R is d Independently optionally substituted with 1,2, 3, 4, 5 or 6R g Substitution; wherein the R is g Having the meaning described in the present invention.
In some embodiments, each R 2 Independently is fluoro, chloro, bromo, iodo, hydroxy, oxo (=o), amino, nitro, cyano, C 1-4 Alkyl, C 1-4 Haloalkyl, C 1-4 Alkoxy, phenyl, C 3-6 Cycloalkyl, -C 0-4 alkylene-OR a 、-C 0-4 Alkylene- (C (=O)) m -R b 、-C 0-4 Alkylene- (O) n -(C(=O)) m -NR c R d 5-6 membered heteroaryl, -C 0-4 Alkylene- (3-7 membered heterocyclyl), -C 0-4 Alkylene- (5-12 membered spiroheterocyclyl) or-C 0-4 Alkylene- (4-12 membered fused heterocyclyl); wherein the R is 2 Optionally by 1,2 or 3R f Substitution;
alternatively, two R's, optionally attached to the same carbon atom 2 And the carbon atoms to which they are attached together form C 3-6 Cycloalkyl or 4-6 membered heterocycle;
wherein, m, n and R a 、R b 、R c 、R d And R is f With the invention described inMeaning.
In other embodiments, each R 2 Independently is fluoro, chloro, bromo, iodo, hydroxy, oxo, amino, nitro, cyano, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, t-butyl, monofluoromethyl, difluoromethyl, trifluoromethyl, monofluoroethyl, 1-difluoroethyl, 1, 2-difluoroethyl, methoxy, ethoxy, n-propoxy, isopropoxy, phenyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, -OR a 、-CH 2 -OR a 、-CH 2 CH 2 -OR a 、-CH 2 (CH 2 ) 2 -OR a 、-CH 2 CH(CH 3 )-OR a 、-C(CH 3 ) 2 CH 2 -OR a 、-CH 2 (CH 2 ) 3 -OR a 、-CH(CH 3 )CH(CH 3 )-OR a 、-CH 2 C(CH 3 ) 2 -OR a 、-(C=O) m -R b 、-CH 2 -(C(=O)) m -R b 、-CH 2 CH 2 -(C(=O)) m -R b 、-CH 2 (CH 2 ) 2 -(C(=O)) m -R b 、-CH 2 CH(CH 3 )-(C(=O)) m -R b 、-C(CH 3 ) 2 CH 2 -(C(=O)) m -R b 、-CH 2 (CH 2 ) 3 -(C(=O)) m -R b 、-CH(CH 3 )CH(CH 3 )-(C(=O)) m -R b 、-CH 2 C(CH 3 ) 2 -(C=O) m -R b 、-(O) n -(C(=O)) m -NR c R d 、-CH 2 -(O) n -(C(=O)) m -NR c R d 、-CH 2 CH 2 -(O) n -(C(=O)) m -NR c R d 、-CH 2 (CH 2 ) 2 -(O) n -(C(=O)) m -NR c R d 、-CH 2 CH(CH 3 )-(O) n -(C(=O)) m -NR c R d 、-C(CH 3 ) 2 CH 2 -(O) n -(C(=O)) m -NR c R d 、-CH 2 (CH 2 ) 3 -(O) n -(C(=O)) m -NR c R d 、-CH(CH 3 )CH(CH 3 )-(O) n -(C(=O)) m -NR c R d 、-CH 2 C(CH 3 ) 2 -(O) n -(C(=O)) m -NR c R d Oxazolyl, thiazolyl, thienyl, imidazolyl, pyridazinyl, pyridyl, Wherein the R is 2 Optionally by 1, 2 or 3R f Substitution;
alternatively, two R's, optionally attached to the same carbon atom 2 And the carbon atoms to which they are attached together form cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, pyrazolidinyl, imidazolidinyl, tetrahydrofuranyl, tetrahydrothienyl, 1, 3-dioxacyclopentyl, dithiocyclopentyl, tetrahydropyranyl, 4H-pyranyl, tetrahydrothiopyranyl, piperidinyl, morpholinyl, thiomorpholinyl, or piperazinyl;
wherein, m, n and R a 、R b 、R c 、R d And R is f Having the meaning described in the present invention.
In some embodiments, each R a Independently hydrogen, C 1-6 Alkyl, C 1-6 Haloalkyl, C 6-10 Aryl, C 3-8 Cycloalkyl, 5-10 membered heteroaryl, 3-12 membered heterocyclyl, 5-12 membered spiroheterocyclyl or 4-12 membered fused heterocyclyl; each R is b 、R c And R is d Independently hydrogen, hydroxy, C 1-6 Alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, C 6-10 Aryl, C 3-8 Cycloalkyl, 4-12 membered fused carbocyclyl, 5-12 membered spirocarbocyclyl, 5-10 membered heteroaryl, 3-7 membered heterocyclyl, 5-1A 2-membered spiroheterocyclyl or a 4-12 membered fused heterocyclyl; or R is c 、R d And the N atoms to which they are attached together form a 4-7 membered heterocyclic ring;
each R is as follows a 、R b 、R c And R is d Independently optionally substituted with 1,2, 3, 4, 5 or 6R g Substitution; wherein the R is g Having the meaning described in the present invention.
In other embodiments, each R a Independently is hydrogen, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, monofluoromethyl, difluoromethyl, trifluoromethyl, monofluoroethyl, 2-difluoroethyl, 1, 2-difluoroethyl, 2-trifluoroethyl, Phenyl, oxetanyl, tetrahydrofuranyl, tetrahydropyranyl, oxetanyl, cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl; each R is b 、R c And R is d Is independently hydrogen, hydroxy, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, monofluoromethyl, difluoromethyl, trifluoromethyl, monofluoroethyl, 2-difluoroethyl, 1, 2-difluoroethyl, 2-trifluoroethyl, methoxy, ethoxy, n-propoxy, isopropoxy, phenyl, oxetanyl, tetrahydrofuranyl, tetrahydropyranyl, oxetanyl, cycloheptyl, Cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl; or R is c 、R d And the N atoms to which they are attached together form a 4-7 membered heterocyclic ring;
Wherein each R is a 、R b 、R c And R is d Independently optionally substituted with 1,2, 3, 4, 5 or 6R g Substitution; the R is g Having the meaning described in the present invention.
In some embodiments, each R f And R is g Independently is fluoro, chloro, bromo, iodo, oxo (=o), hydroxy, amino, nitro, cyano, C 1-6 Alkyl, hydroxy substituted C 1-6 Alkyl, C 1-6 Haloalkyl, C 6-10 Aryl, C 3-8 Cycloalkyl, 4-12 membered fused carbocyclyl, 5-12 membered spirocarbocyclyl, 5-10 membered heteroaryl, 3-12 membered heterocyclyl, 5-12 membered spiroheterocyclyl, 4-12 membered fused heterocyclyl, C 1-6 Alkoxy, C 1-6 Alkylamino or C 1-6 Haloalkoxy groups.
In other embodiments, each R f And R is g Independently is fluorine, chlorine, bromine, iodine, oxo, hydroxy, amino, nitro, cyano, methyl, ethyl, n-propyl, isopropyl, n-butyl, -CH 2 OH, monofluoromethyl, difluoromethyl, trifluoromethyl, monofluoroethyl, 1-difluoroethyl, 1, 2-difluoroethyl, phenyl, pyridyl, pyrimidinyl, pyrazolyl, oxiranyl, oxetanyl, tetrahydrofuranyl, tetrahydropyranyl, Cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, methoxy, ethoxy, n-propoxy, isopropoxy, monofluoromethoxy, difluoromethoxy or trifluoromethoxy.
In some embodiments, each m and n is independently 0 or 1.
In some embodiments, the present invention relates to compounds of one of the following, or stereoisomers, nitrogen oxides, solvates, metabolites, pharmaceutically acceptable salts or prodrugs thereof, but in no way limited to these compounds:
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
stereoisomers, solvates, metabolites, pharmaceutically acceptable salts and prodrugs of the compounds of formula (I) are included within the scope of the invention unless otherwise specified.
In another aspect, the present invention relates to a pharmaceutical composition comprising a compound of formula (I) or a stereoisomer, a geometric isomer, a tautomer, a nitroxide, a hydrate, a solvate, a metabolite, a pharmaceutically acceptable salt or a prodrug thereof, according to the present invention, and a pharmaceutically acceptable excipient, carrier, adjuvant or combination thereof.
In some embodiments, the pharmaceutical composition comprises other agents that prevent or treat inflammatory syndromes or autoimmune diseases or any combination thereof.
In one embodiment, the pharmaceutical composition may be in the form of a liquid, solid, semi-solid, gel or spray.
In another aspect, the invention relates to the use of a compound of formula (I) or a pharmaceutical composition thereof for the manufacture of a medicament for the prevention or treatment of an inflammatory or autoimmune disease mediated by roryt in a mammal.
In some embodiments, the present invention relates to the use of a compound of formula (I) or a pharmaceutical composition thereof for the preparation of a medicament for the prevention or treatment of psoriasis, rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis, inflammatory bowel disease, colitis, ulcerative colitis, rheumatoid arthritis, autoimmune ocular disease, ankylosing spondylitis, asthma, chronic obstructive pulmonary disease, osteoarthritis, allergic rhinitis, allergic dermatitis, crohn's disease, or kawasaki disease.
In another aspect, the present invention relates to methods for the preparation, isolation and purification of compounds of formula (I).
In another aspect, the present invention relates to intermediates for preparing compounds of formula (I).
The disclosed compounds may contain asymmetric or chiral centers and thus may exist in different stereoisomeric forms. The present invention is intended to encompass all stereoisomeric forms of the compounds of formula (I), including but not limited to diastereomers, enantiomers, atropisomers and geometric (or conformational) isomers, as well as mixtures thereof, such as racemic mixtures, as part of the present invention.
In the structures disclosed herein, when the stereochemistry of any particular chiral atom is not indicated, then all stereoisomers of that structure are contemplated as being within the present invention and are included as presently disclosed compounds. When stereochemistry is indicated by the solid wedge (solid wedge) or dashed line representing a particular configuration, then the stereoisomers of that structure are so defined and defined.
The compounds of formula (I) may exist in different tautomeric forms and all such tautomers are included within the scope of the invention.
The compounds of formula (I) may be present in the form of salts. In one embodiment, the salt refers to a pharmaceutically acceptable salt. The term "pharmaceutically acceptable" means that the substance or composition must be chemically and/or toxicologically compatible with the other ingredients comprising the formulation and/or the mammal being treated therewith. In another embodiment, the salt is not necessarily a pharmaceutically acceptable salt, and may be an intermediate for preparing and/or purifying the compound of formula (I) and/or for separating enantiomers of the compound of formula (I).
Pharmaceutically acceptable acid addition salts may be formed from the reaction of a compound of the present disclosure with an inorganic or organic acid, such as acetate, aspartate, benzoate, benzenesulfonate, bromide/hydrobromide, bicarbonate/carbonate, bisulfate/sulfate, camphorsulfonate, chloride/hydrochloride, chlorotheophylline salt, citrate, ethanedisulfonate, fumarate, glucoheptonate, gluconate, glucuronate, hippurate, hydroiodite/iodide, isethionate, lactate, lactobionic aldehyde, lauryl sulfate, malate, maleate, malonate, mandelate, methanesulfonate, methylsulfate, naphthoate, naphthalenesulfonate, nicotinate, nitrate, stearate, oleate, oxalate, palmitate, pamoate, phosphate/hydrogen phosphate/dihydrogen phosphate, polygalactoate, propionate, stearate, succinate, sulfosalicylate, tartrate, tosylate and trifluoroacetate.
Pharmaceutically acceptable base addition salts may be formed from the compounds of the present disclosure by reaction with inorganic or organic bases.
Inorganic bases from which salts may be derived include, for example, ammonium salts and metals of groups I to XII of the periodic Table. In certain embodiments, the salt is derived from sodium, potassium, ammonium, calcium, magnesium, iron, silver, zinc, and copper; particularly suitable salts include ammonium, potassium, sodium, calcium and magnesium salts.
Organic bases from which salts may be derived include primary, secondary and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, basic ion exchange resins, and the like. Some organic amines include, for example, isopropylamine, benzathine (benzathine), choline salts (choline), diethanolamine, diethylamine, lysine, meglumine (meglumine), piperazine and tromethamine.
Pharmaceutically acceptable salts of the invention can be synthesized from the parent compound, basic or acidic moiety using conventional chemical methods. In general, such salts can be prepared by reacting the free acid forms of these compounds with a stoichiometric amount of a suitable base (e.g., na, ca, mg or K hydroxide, carbonate, bicarbonate, etc.), or by reacting the free base forms of these compounds with a stoichiometric amount of a suitable acid. Such reactions are generally carried out in water or an organic solvent or a mixture of both. Generally, it is desirable to use a non-aqueous medium such as diethyl ether, ethyl acetate, ethanol, isopropanol or acetonitrile where appropriate. In, for example, "Remington's Pharmaceutical Sciences", 20 th edition, mack Publishing Company, easton, pa., (1985); and "manual of pharmaceutically acceptable salts: a list of further suitable salts can be found in Properties, selection and application (Handbook of Pharmaceutical Salts: properties, selection, and Use) ", stahl and Wermuth (Wiley-VCH, weinheim, germany, 2002).
In addition, the compounds disclosed herein, including their salts, may also be obtained in the form of their hydrates or in the form of solvents (e.g., ethanol, DMSO, etc.) containing them, for their crystallization. The disclosed compounds may form solvates inherently or by design with pharmaceutically acceptable solvents (including water); accordingly, the present invention is intended to include both solvated and unsolvated forms of the presently disclosed compounds.
Any formulae given herein are also intended to represent non-isotopically enriched forms as well as isotopically enriched forms of such compounds. Isotopically enriched compounds have structures depicted by the formulae given herein except that one or more atoms are replaced by an atom having a selected atomic or mass number. Exemplary isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, and chlorine, such as 2 H、 3 H、 11 C、 13 C、 14 C、 15 N、 17 O、 18 O、 18 F、 31 P、 32 P、 35 S、 36 Cl and Cl 125 I。
In another aspect, the compounds of the invention include isotopically enriched compounds defined in the invention, e.g., wherein a radioisotope, such as 3 H、 14 C and C 18 F, or in which non-radioactive isotopes are present, e.g 2 H and 13 Those of C. Such isotopically enriched compounds are useful in metabolic studies (using 14 C) Reaction kinetics studies (using, for example 2 H or 3 H) Detection or imaging techniques, such as Positron Emission Tomography (PET) or Single Photon Emission Computed Tomography (SPECT) including drug or substrate tissue distribution assays, or may be used in radiation therapy of a patient. 18 F-enriched compounds are particularly desirable for PET or SPECT studies. Isotopically enriched compounds of formula (I) can be described by conventional techniques familiar to those skilled in the art or by the examples and procedures of preparation of the invention using a suitable isotopic labelThe marking reagent is prepared in place of the originally used unlabeled reagent.
In addition, heavier isotopes are in particular deuterium (i.e., 2 substitution of H or D) may provide certain therapeutic advantages, which are brought about by a higher metabolic stability. For example, increased in vivo half-life or reduced dosage requirements or improved therapeutic index. It is to be understood that deuterium in the present invention is considered as a substituent of the compound of formula (I). The concentration of such heavier isotopes, particularly deuterium, can be defined by an isotopic enrichment factor. The term "isotopically enriched factor" as used herein refers to the ratio between the isotopic abundance and the natural abundance of a specified isotope. If a substituent of a compound of the invention is designated as deuterium, the compound has an isotopic enrichment factor for each designated deuterium atom of at least 3500 (52.5% deuterium incorporation at each designated deuterium atom), at least 4000 (60% deuterium incorporation), at least 4500 (67.5% deuterium incorporation), at least 5000 (75% deuterium incorporation), at least 5500 (82.5% deuterium incorporation), at least 6000 (90% deuterium incorporation), at least 6333.3 (95% deuterium incorporation), at least 6466.7 (97% deuterium incorporation), at least 6600 (99% deuterium incorporation), or at least 6633.3 (99.5% deuterium incorporation). Pharmaceutically acceptable solvates of the invention include those wherein the crystallization solvent may be isotopically substituted, e.g. D 2 O, acetone-d 6 、DMSO-d 6 Those solvates of (a).
Pharmaceutical compositions, formulations and administration of the compounds of the invention
The present invention provides a pharmaceutical composition comprising a compound of the present disclosure, such as the compounds listed in the examples; and a pharmaceutically acceptable excipient, carrier, adjuvant, or combination thereof.
The present invention provides methods of treating, preventing or ameliorating a disease or disorder comprising administering a safe and effective amount of a combination comprising a compound of the present disclosure and one or more therapeutically active agents. Wherein the combination comprises one or more other agents for preventing or treating inflammatory syndromes, disorders or diseases, and the active therapeutic agents include, but are not limited to:
1) TNF-alpha inhibitors; 2) A non-selective COX-1/COX-2 inhibitor; 3) COX-2 inhibitors; 4) Other therapeutic agents useful in the treatment of inflammatory syndromes and autoimmune diseases, including glucocorticoids, methotrexate, leflunomide (leflunomide), sulfasalazine, azathioprine, cyclosporine, tacrolimus (tacrolimus), penicillamine, buspiramine, aclitalopril, mizoribine, clozapride, ciclesonide, hydroxychloroquine, gold thiomalate (aurothiomalate), auranofin, cyclophosphamide, BAFF/APRIL inhibitors, CTLA-4-immunoglobulins, or the like; 5) Leukotriene biosynthesis inhibitors, 5-lipoxygenase inhibitors or 5-lipoxygenase activating protein (FLAP) antagonists; 6) LTD4 receptor antagonists; 7) PDE4 inhibitors; 8) Antihistaminic HI receptor antagonists; alpha 1-and alpha 2-adrenoreceptor agonists; 9) Anticholinergic agents; 10 P-adrenergic receptor agonists; 11 Insulin-like growth factor type I analogues; 12 Kinase inhibitors selected from the group consisting of Janus kinase inhibitors (JAK 1 and/or JAK2 and/or JAK3 and/or TYK 2), p38 MAPK and IKK2;13 B cell targeted biologic agents such as rituximab; 14 Selective co-stimulatory modulators such as abafpu; 15 Interleukin inhibitor selected from the group consisting of IL-1 inhibitors such as anakinra, IL-6 inhibitors such as tolizumab and IL-12/IL-23 inhibitors such as Utilizumab.
The amount of the compound in the pharmaceutical composition disclosed by the invention means that the inhibition of retinoic acid related lone-core receptor gamma t in a biological sample or a patient can be effectively detected. The dosage of the active ingredient in the compositions of the present invention may vary, however, the amount of active ingredient must be such that an appropriate dosage form is obtained. The active ingredient may be administered to patients (animals and humans) in need of such treatment in a dosage that provides optimal pharmaceutical efficacy. The selected dosage depends on the desired therapeutic effect, on the route of administration and the duration of the treatment. The dosage will vary from patient to patient depending on the nature and severity of the disease, the weight of the patient, the particular diet of the patient, the concurrent medication, and other factors that will be recognized by those skilled in the art. The dosage range is typically about 0.5mg to 1.0g per patient per day, and may be administered in single or multiple doses. In one embodiment, the dosage ranges from about 0.5mg to 500mg per patient per day; in another embodiment about 0.5mg to 200mg per patient per day; in yet another embodiment about 5mg to 50mg per patient per day.
It will also be appreciated that certain compounds of the invention may exist in free form or, if appropriate, in the form of pharmaceutically acceptable derivatives thereof. Pharmaceutically acceptable derivatives include pharmaceutically acceptable prodrugs, salts, esters, salts of such esters, or any additional adducts or derivatives that provide, directly or indirectly, the compounds of the invention or metabolites or residues thereof when administered to a patient in need thereof.
The disclosed pharmaceutical or pharmaceutical compositions may be prepared and packaged in bulk (bulk) form, wherein a safe and effective amount of the compound of formula (I) may be extracted and then administered to a patient in powder or syrup form. Typically, the patient is administered at a dosage level of between 0.0001 and 10mg/kg body weight per day to obtain an effective inhibition of retinoic acid related lone core receptors γt. Alternatively, the pharmaceutical compositions disclosed herein may be prepared and packaged in unit dosage form, wherein each physically discrete unit contains a safe and effective amount of a compound of formula (I). When prepared in unit dosage form, the presently disclosed pharmaceutical compositions may generally contain, for example, from 0.5mg to 1g, or from 1mg to 700mg, or from 5mg to 100mg of a presently disclosed compound.
When the pharmaceutical compositions of the present invention contain one or more other active ingredients in addition to the compound of the present invention, the compound weight ratio of the compound of the present invention to the second active ingredient may vary and will depend on the effective dose of each ingredient. Typically, an effective dose of each is used. Thus, for example, when a compound of the present invention is mixed with another agent, the weight ratio of the compound of the present invention to the other agent typically ranges from about 1000:1 to about 1:1000, e.g., about 200:1 to about 1:200. mixtures of the compounds according to the invention with other active ingredients are generally also within the abovementioned ranges, but in each case an effective dose of each active ingredient should be used.
As used herein, "pharmaceutically acceptable excipient" means a pharmaceutically acceptable material, mixture or vehicle associated with consistency of administration dosage form or pharmaceutical composition. Each excipient must be compatible with the other ingredients of the pharmaceutical composition when mixed to avoid interactions that would greatly reduce the efficacy of the disclosed compounds and interactions that would result in a pharmaceutical composition that is not pharmaceutically acceptable when administered to a patient. Furthermore, each excipient must be pharmaceutically acceptable, e.g., of sufficiently high purity.
Suitable pharmaceutically acceptable excipients will vary depending upon the particular dosage form selected. Furthermore, pharmaceutically acceptable excipients may be selected according to their particular function in the composition. For example, certain pharmaceutically acceptable excipients may be selected that can aid in the production of a uniform dosage form. Certain pharmaceutically acceptable excipients that can aid in the production of stable dosage forms can be selected. Certain pharmaceutically acceptable excipients that facilitate carrying or transporting the disclosed compounds from one organ or portion of the body to another organ or portion of the body when administered to a patient may be selected. Certain pharmaceutically acceptable excipients that enhance patient compliance may be selected.
Suitable pharmaceutically acceptable excipients include the following types of excipients: diluents, fillers, binders, disintegrants, lubricants, glidants, granulating agents, coating agents, wetting agents, solvents, co-solvents, suspending agents, emulsifiers, sweeteners, flavoring agents, taste masking agents, colorants, anti-caking agents, humectants, chelating agents, plasticizers, tackifiers, antioxidants, preservatives, stabilizers, surfactants, and buffers. The skilled artisan will recognize that certain pharmaceutically acceptable excipients may provide more than one function, and alternative functions, depending on how much of the excipient is present in the formulation and which other excipients are present in the formulation.
The skilled artisan will know and be familiar with the art to which they will be able to select the appropriate amount of suitable pharmaceutically acceptable excipients for use in the present invention. Furthermore, there are a number of resources available to the skilled person, who describe pharmaceutically acceptable excipients and are used to select the appropriate pharmaceutically acceptable excipient. Examples include Remington's Pharmaceutical Sciences (Mack Publishing Company), the Handbook of Pharmaceutical Additives (Gower Publishing Limited), and The Handbook of Pharmaceutical Excipients (the American Pharmaceutical Association and the Pharmaceutical Press).
Various carriers for formulating pharmaceutically acceptable compositions, and well known techniques for their preparation, are disclosed in Remington, the Science and Practice of Pharmacy,21st edition,2005,ed.D.B.Troy,Lippincott Williams&Wilkins,Philadelphia,and Encyclopedia of Pharmaceutical Technology,eds.J.Swarbrick and J.C.Boylan,1988-1999,Marcel Dekker,New York, the contents of each of which are incorporated herein by reference. It is within the scope of the present invention to contemplate its use in addition to any common carrier that is incompatible with the disclosed compounds of the present invention, such as by producing any undesirable biological effect, or by interacting in a deleterious manner with any other component of the pharmaceutically acceptable composition.
The pharmaceutical compositions disclosed herein are prepared using techniques and methods known to those skilled in the art. Some methods commonly used in the art are described in Remington's Pharmaceutical Sciences (Mack Publishing Company).
Thus, in another aspect, the present invention relates to a process for preparing a pharmaceutical composition comprising a compound of the present disclosure and a pharmaceutically acceptable excipient, carrier, adjuvant or combination thereof, which process comprises mixing the various ingredients. Pharmaceutical compositions comprising the compounds of the present disclosure may be prepared by mixing, for example, at ambient temperature and atmospheric pressure.
The compounds disclosed herein are generally formulated in a dosage form suitable for administration to a patient by the desired route. For example, dosage forms include those suitable for the following routes of administration: (1) Oral administration, such as tablets, capsules, caplets, pills, troches, powders, syrups, elixirs, suspensions, solutions, emulsions, sachets and cachets; (2) Parenteral administration, such as sterile solutions, suspensions, and reconstituted powders; (3) transdermal administration, such as transdermal patch tablets; (4) rectal administration, such as suppositories; (5) inhalations, such as aerosols, solutions and dry powders; and (6) topical administration, such as creams, ointments, lotions, solutions, pastes, sprays, foams and gels.
In one embodiment, the presently disclosed compounds may be formulated into oral dosage forms. In another embodiment, the presently disclosed compounds may be formulated into an inhalation dosage form. In another embodiment, the presently disclosed compounds may be formulated for nasal administration. In yet another embodiment, the presently disclosed compounds may be formulated into transdermal dosage forms. In yet another embodiment, the presently disclosed compounds may be formulated into topical dosage forms.
The pharmaceutical compositions provided herein may be provided in compressed tablets, developed tablets, chewable lozenges, instant tablets, reconstituted tablets, or enteric tablets, sugar-coated or film-coated tablets. Enteric-coated tablets are compressed tablets coated with a substance that resists the action of gastric acid but dissolves or disintegrates in the intestine, thereby preventing the active ingredient from contacting the acidic environment of the stomach. Enteric coatings include, but are not limited to, fatty acids, fats, phenyl salicylate, waxes, shellac, aminated shellac, and cellulose acetate phthalate. Dragees are dragee-enclosed compressed tablets that can facilitate masking of unpleasant tastes or odors and prevent oxidation of the tablet. The film coated tablet is a compressed tablet covered with a thin layer or film of a water-soluble substance. Film coatings include, but are not limited to, hydroxyethyl cellulose, sodium carboxymethyl cellulose, polyethylene glycol 4000, and cellulose acetate phthalate. The film coating imparts the same general characteristics as the sugar coating. The composite tablet is a compressed tablet prepared through more than one compression cycle, and comprises a multi-layer tablet and a compression coated or dry coated tablet.
Tablet dosage forms may be prepared from the active ingredient in powder, crystalline or particulate form alone or in combination with one or more carriers or excipients described herein, including binders, disintegrants, controlled release polymers, lubricants, diluents and/or colorants. Flavoring and sweetening agents are particularly useful in forming chewable tablets and lozenges.
The pharmaceutical composition provided by the invention can be provided in a soft capsule or a hard capsule, and can be prepared from gelatin, methylcellulose, starch or calcium alginate. The hard gelatin capsule, also known as a Dry Filled Capsule (DFC), consists of two segments, one segment being filled into the other, thus completely encapsulating the active ingredient. Soft Elastic Capsules (SEC) are soft, spherical shells, such as gelatin shells, which are plasticized by the addition of glycerol, sorbitol or similar polyols. The soft gelatin shell may contain a preservative to prevent microbial growth. Suitable preservatives are those described herein, including methyl and propyl parabens, and sorbic acid. Liquid, semi-solid and solid dosage forms provided herein may be encapsulated in capsules. Suitable liquid and semi-solid dosage forms include solutions and suspensions in propylene carbonate, vegetable oils or triglycerides. Capsules containing such solutions can be prepared as described in U.S. patent nos.4,328,245;4,409,239 and 4,410,545. The capsules may also be coated as known to those skilled in the art to improve or maintain dissolution of the active ingredient.
The pharmaceutical compositions provided herein may be provided in liquid and semi-solid dosage forms, including emulsions, solutions, suspensions, elixirs and syrups. Emulsions are two-phase systems in which one liquid is completely dispersed in the form of pellets in another liquid, which may be oil-in-water or water-in-oil. The emulsion may include pharmaceutically acceptable non-aqueous liquids and solvents, emulsifiers, and preservatives. Suspensions may include pharmaceutically acceptable suspending agents and preservatives. The aqueous alcohol solution may include a pharmaceutically acceptable acetal, such as a di (lower alkyl) acetal of a lower alkyl aldehyde, for example, acetaldehyde diethyl acetal; and water-soluble solvents having one or more hydroxyl groups, such as propylene glycol and ethanol. Elixirs are clear, sweet aqueous alcoholic solutions. Syrups are concentrated aqueous solutions of a sugar, for example sucrose, and may also contain a preservative. For liquid dosage forms, for example, a solution in polyethylene glycol may be diluted with a sufficient amount of a pharmaceutically acceptable liquid carrier, such as water, for accurate and convenient administration.
Other useful liquid and semi-solid dosage forms include, but are not limited to, those comprising the active ingredient provided herein and a secondary mono-or poly-alkylene glycol, including: 1, 2-dimethoxymethane, diglyme, triglyme, tetraglyme, polyethylene glycol-350-dimethyl ether, polyethylene glycol-550-dimethyl ether, polyethylene glycol-750-dimethyl ether, wherein 350, 550, 750 refer to the approximate average molecular weight of polyethylene glycol. These formulations may further include one or more antioxidants such as Butylated Hydroxytoluene (BHT), butylated Hydroxyanisole (BHA), propyl gallate, vitamin E, hydroquinone, hydroxycoumarins, ethanolamine, lecithin, cephalin, ascorbic acid, malic acid, sorbitol, phosphoric acid, bisulphite, sodium metabisulfite, thiodipropionic acid and esters thereof, and dithiocarbamates.
Dosage unit formulations for oral administration may be microencapsulated, as appropriate. It may also be formulated in an extended-release or sustained-release composition, for example, by coating or embedding the particulate material in a polymer, wax or the like.
The oral pharmaceutical compositions provided by the present invention may also be provided in the form of liposomes, micelles, microspheres or nanosystems. Micelle dosage forms may be prepared using the methods described in U.S. Pat. No.6,350,458.
The pharmaceutical compositions provided herein may be provided in non-effervescent or effervescent granules and powders for reconstitution into liquid dosage forms. Pharmaceutically acceptable carriers and excipients used in non-effervescent granules or powders may include diluents, sweeteners and wetting agents. Pharmaceutically acceptable carriers and excipients used in effervescent granules or powders may include organic acids and carbon dioxide sources.
Coloring and flavoring agents may be used in all of the above dosage forms.
The disclosed compounds may also be combined with soluble polymers as targeted drug carriers. Such polymers include polyvinylpyrrolidone, pyran copolymers, polyhydroxypropyl methacrylamide-phenol, polyhydroxyethyl asparaginol or palmitoyl residue substituted polyoxyethylene polylysine. In addition, the disclosed compounds may be combined with a class of biodegradable polymers used in achieving controlled release of drugs, such as polylactic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates, and cross-linked or amphiphilic block copolymers of hydrogels.
The pharmaceutical compositions provided herein may be formulated in immediate or modified release dosage forms, including delayed-, sustained-, pulsed-, controlled-, targeted-, and programmed release forms.
The pharmaceutical compositions provided herein may be co-formulated with other active ingredients that do not impair the intended therapeutic effect, or with substances that supplement the intended effect.
The pharmaceutical compositions provided herein may be administered parenterally, by injection, infusion or implantation, for topical or systemic administration. Parenteral administration as used in the present invention includes intravenous, intra-arterial, intraperitoneal, intrathecal, intraventricular, intraurethral, intrasternal, intracranial, intramuscular, intrasynovial and subcutaneous administration.
The pharmaceutical compositions provided herein may be formulated in any dosage form suitable for parenteral administration, including solutions, suspensions, emulsions, micelles, liposomes, microspheres, nanosystems, and solid forms suitable for making solutions or suspensions in liquids prior to injection. Such dosage forms may be prepared according to conventional methods known to those skilled in The art of pharmaceutical sciences (see Remington: the Science and Practice of Pharmacy, supra).
Pharmaceutical compositions contemplated for parenteral administration may include one or more pharmaceutically acceptable carriers and excipients including, but not limited to, aqueous vehicles, water miscible vehicles, non-aqueous vehicles, antimicrobial or antimicrobial growth preservatives, stabilizers, dissolution enhancers, isotonic agents, buffers, antioxidants, local anesthetics, suspending and dispersing agents, wetting or emulsifying agents, complexing agents, sequestering or chelating agents, freezing point depressants, cryoprotectants, thickening agents, pH adjusting agents, and inert gases.
Suitable aqueous vehicles include, but are not limited to: water, saline, normal saline or Phosphate Buffered Saline (PBS), sodium chloride injection, ringers injection, isotonic dextrose injection, sterile water injection, dextrose and lactated Ringers injection. Non-aqueous vehicles include, but are not limited to, fixed oils of vegetable origin, castor oil, corn oil, cottonseed oil, olive oil, peanut oil, peppermint oil, safflower oil, sesame oil, soybean oil, hydrogenated vegetable oils, medium chain triglycerides of hydrogenated soybean oil and coconut oil, and palm seed oil. Water-miscible vehicles include, but are not limited to, ethanol, 1, 3-butanediol, liquid polyethylene glycols (e.g., polyethylene glycol 300 and polyethylene glycol 400), propylene glycol, glycerol, N-methyl-2-pyrrolidone, N-dimethylacetamide, and dimethylsulfoxide.
Suitable antimicrobial agents or preservatives include, but are not limited to, phenol, cresol, mercuric agents, benzyl alcohol, chlorobutanol, methyl and propyl p-hydroxybenzoates, thimerosal, benzalkonium chloride (e.g., benzethonium chloride), methyl and propyl parabens, and sorbic acid. Suitable isotonic agents include, but are not limited to, sodium chloride, glycerol, and glucose. Suitable buffers include, but are not limited to, phosphates and citrates. Suitable antioxidants are those as described herein, including bisulfites and sodium metabisulfites. Suitable local anesthetics include, but are not limited to procaine hydrochloride. Suitable suspending and dispersing agents are those as described herein, including sodium carboxymethyl cellulose, hydroxypropyl methylcellulose, and polyvinylpyrrolidone. Suitable emulsifiers include those described herein, including polyoxyethylene sorbitan monolaurate, polyoxyethylene sorbitan monooleate 80, and triethanolamine oleate. Suitable sequestering or chelating agents include, but are not limited to, EDTA. Suitable pH adjusting agents include, but are not limited to, sodium hydroxide, hydrochloric acid, citric acid, and lactic acid. Suitable complexing agents include, but are not limited to, cyclodextrins including alpha-cyclodextrin, beta-cyclodextrin, hydroxypropyl-beta-cyclodextrin, sulfobutyl ether-beta-cyclodextrin and sulfobutyl ether-7-beta-cyclodextrin CyDex,Lenexa,KS)。
The pharmaceutical compositions provided by the invention can be formulated for single or multiple dose administration. The single dose formulation is packaged in ampules, vials or syringes. The multi-dose parenteral formulation must contain antimicrobial agents at bacteriostatic or fungistatic concentrations. All parenteral formulations must be sterile, as is known and practiced in the art.
In one embodiment, the pharmaceutical composition is provided as a ready-to-use sterile solution. In another embodiment, the pharmaceutical composition is provided as a sterile dry soluble product, including lyophilized powder and subcutaneous injection tablets, which are reconstituted with a carrier prior to use. In yet another embodiment, the pharmaceutical composition is formulated as a ready-to-use sterile suspension. In yet another embodiment, the pharmaceutical composition is formulated as a sterile dry insoluble product reconstituted with a carrier prior to use. In yet another embodiment, the pharmaceutical composition is formulated as a sterile ready-to-use emulsion.
The pharmaceutical compositions may be formulated as suspensions, solids, semi-solids, or thixotropic liquids for administration as an implanted depot. In one embodiment, the disclosed pharmaceutical compositions are dispersed in a solid inner matrix surrounded by an outer polymeric membrane that is insoluble in body fluids but allows the active ingredient in the pharmaceutical composition to diffuse through.
Suitable internal matrices include polymethyl methacrylate, polymethyl butyl acrylate, plasticized or unplasticized polyvinyl chloride, plasticized nylon, plasticized polyethylene terephthalate, natural rubber, polyisoprene, polyisobutylene, polybutadiene, polyethylene, ethylene vinyl acetate copolymers, silicone rubber, polydimethylsiloxane, silicone carbonate copolymers, hydrogels of hydrophilic polymers such as esters of acrylic and methacrylic acid, collagen, crosslinked polyvinyl alcohol, and partially hydrolyzed polyvinyl acetate for coaches.
Suitable external polymeric films include polyethylene, polypropylene, ethylene/propylene copolymers, ethylene/ethyl acrylate copolymers, ethylene/vinyl acetate copolymers, silicone rubber, polydimethylsiloxane, neoprene, chlorinated polyethylene, polyvinyl chloride, copolymers of chlorinated ethylene and vinyl acetate, vinylidene chloride, ethylene and propylene, ionomer polyethylene terephthalate, butyl rubber chlorohydrin rubber, ethylene/vinyl alcohol copolymers, ethylene/vinyl acetate/vinyl alcohol terpolymers, and ethylene/ethyleneoxy ethanol copolymers.
In another aspect, the disclosed pharmaceutical compositions may be formulated in any dosage form suitable for inhaled administration to a patient, such as dry powder, aerosol, suspension or solution compositions. In one embodiment, the disclosed pharmaceutical compositions can be formulated into dosage forms suitable for administration by inhalation to a patient using dry powders. In yet another embodiment, the disclosed pharmaceutical compositions may be formulated in a dosage form suitable for administration by inhalation to a patient via a nebulizer. Dry powder compositions for delivery to the lungs by inhalation typically comprise a finely powdered compound of the presently disclosed invention and one or more finely powdered pharmaceutically acceptable excipients. Pharmaceutically acceptable excipients particularly suitable for use as dry powders are known to those skilled in the art and include lactose, starch, mannitol, and mono-, di-and polysaccharides. The fine powder can be prepared by, for example, micronization and grinding. In general, the size-reduced (e.g., micronized) compound may be produced by a D of about 1 to 10 microns 50 Values (e.g., measured using laser diffraction methods) are defined.
Aerosols may be formulated by suspending or dissolving the presently disclosed compounds in a liquefied propellant. Suitable propellants include chlorinated hydrocarbons, hydrocarbons and other liquefied gases. Representative propellants include: trichlorofluoromethane (propellant 11), dichlorofluoromethane (propellant 12), dichlorotetrafluoroethane (propellant 114), tetrafluoroethane (HFA-134 a), 1-difluoroethane (HFA-152 a), difluoromethane (HFA-32), pentafluoroethane (HFA-12), heptafluoropropane (HFA-227 a), perfluoropropane, perfluorobutane, perfluoropentane, butane, isobutane and pentane. Aerosols comprising the disclosed compounds are typically administered to patients by means of Metered Dose Inhalers (MDI). Such devices are known to those skilled in the art.
The aerosols may contain additional pharmaceutically acceptable excipients that may be used by MDIs, such as surfactants, lubricants, co-solvents, and other excipients, to improve the physical stability of the formulation, improve valve characteristics, improve solubility, or improve taste.
Pharmaceutical compositions suitable for transdermal administration may be formulated as discrete patches intended to remain in intimate contact with the epidermis of the patient for an extended period of time. For example, the active ingredient may be delivered from a patch by ion permeation, as generally described in Pharmaceutical Research,3 (6), 318 (1986).
Pharmaceutical compositions suitable for topical administration may be formulated as ointments, creams, suspensions, lotions, powders, solutions, pastes, gels, sprays, aerosols or oils. For example, ointments, creams and gels may be formulated with water or oil bases, with appropriate thickening and/or gelling agents and/or solvents. Such a base may include water, and/or oils such as liquid paraffin and vegetable oils (e.g., peanut oil or castor oil), or solvents such as polyethylene glycol. Thickening and gelling agents used according to the nature of the matrix include soft paraffin, aluminum stearate, cetostearyl alcohol, polyethylene glycol, lanolin, beeswax, carbopol and cellulose derivatives, and/or glyceryl monostearate and/or nonionic emulsifiers.
Lotions may be formulated with water or oil and will in general also contain one or more emulsifying agents, stabilizing agents, dispersing agents, suspending agents or thickening agents.
The topical powders may be formed in the presence of any suitable powder base such as talc, lactose or starch. Drops may be formulated with an aqueous or nonaqueous base containing one or more dispersing agents, solubilising agents, suspending agents or preservatives.
Topical formulations may be administered by application to the affected area one or more times per day; a occlusive dressing covering the skin is preferably used. The adhesive reservoir system may allow for continuous or prolonged administration.
Use of the compounds and pharmaceutical compositions of the invention
The compounds or pharmaceutical compositions disclosed herein may be used in the preparation of a medicament for the treatment, prevention, amelioration, control or alleviation of roryt-mediated inflammatory or autoimmune diseases in a mammal, including a human, and may also be used in the preparation of other medicaments for inhibiting roryt.
Specifically, the amount of the compound in the pharmaceutical composition of the present invention can be effective to detectably inhibit rorγt, and the compound of the present invention can be used as a medicament for preventing or treating psoriasis, rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis, inflammatory bowel disease, colitis, ulcerative colitis, rheumatoid arthritis, autoimmune ocular disease, ankylosing spondylitis, asthma, chronic obstructive pulmonary disease, osteoarthritis, allergic rhinitis, allergic dermatitis, crohn's disease or kawasaki disease in humans.
The compounds or pharmaceutical compositions of the invention may be applied to, but are in no way limited to, the use of an effective amount of a compound or pharmaceutical composition of the invention to prevent, treat or ameliorate psoriasis, rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis, inflammatory bowel disease, colitis, ulcerative colitis, rheumatoid arthritis, autoimmune ocular disease, ankylosing spondylitis, asthma, chronic obstructive pulmonary disease, osteoarthritis, allergic rhinitis, allergic dermatitis, crohn's disease or kawasaki disease in a mammal, including a human, to a patient.
The compounds and pharmaceutical compositions of the present invention are useful for veterinary treatment of mammals, in addition to human therapy, in pets, in animals of introduced species and in farm animals. Examples of other animals include horses, dogs, and cats. Herein, the compounds of the present invention include pharmaceutically acceptable derivatives thereof.
General synthetic procedure
For the purpose of illustrating the invention, examples are set forth below. It is to be understood that the invention is not limited to these examples but provides a method of practicing the invention.
In general, the compounds of the invention may be prepared by the methods described herein, wherein the substituents are as defined in formula (I), unless otherwise indicated. The following reaction schemes and examples are provided to further illustrate the present invention.
Those skilled in the art will recognize that: the chemical reactions described herein may be used to suitably prepare many other compounds of the present invention, and other methods for preparing the compounds of the present invention are considered to be within the scope of the present invention. For example, the synthesis of those non-exemplified compounds according to the invention can be successfully accomplished by modification methods, such as appropriate protection of interfering groups, by use of other known reagents in addition to those described herein, or by some conventional modification of the reaction conditions, by those skilled in the art. In addition, the reactions disclosed herein or known reaction conditions are also well-known to be applicable to the preparation of other compounds of the present invention.
The examples described below are given unless otherwise indicated that all temperatures are given in degrees celsius. Reagents were purchased from commercial suppliers such as Aldrich Chemical Company, arco Chemical Company and Alfa Chemical Company and were used without further purification unless otherwise indicated. General reagents were purchased from Shandong Chemicals, guangdong Chemicals, guangzhou Chemicals, tianjin good Chemies, tianjin Fuchen Chemies, wuhan Xinhua Yuan technology development Co., ltd., qingdao Teng Chemies Co., and Qingdao sea chemical Co.
Anhydrous tetrahydrofuran, dioxane, toluene and diethyl ether are obtained by reflux drying of metallic sodium. The anhydrous methylene chloride and chloroform are obtained by reflux drying of calcium hydride. Ethyl acetate, petroleum ether, N-hexane, N-dimethylacetamide and N, N-dimethylformamide were dried over anhydrous sodium sulfate in advance for use.
The following reaction is typically carried out under nitrogen or argon pressure or with a dry tube (unless otherwise indicated) over anhydrous solvent, the reaction flask is capped with a suitable rubber stopper and the substrate is injected through a syringe. The glassware was all dried.
The chromatographic column is a silica gel column. Silica gel (300-400 mesh) was purchased from Qingdao ocean chemical plant.
Nuclear magnetic resonance spectra were recorded using Bruker 400MHz or 600MHz nuclear magnetic resonance spectrometer with CDC1 3 、DMSO-d 6 、CD 3 OD or acetone-d 6 TMS (0 ppm) or chloroform (7.26 ppm) was used as a reference standard for the solvent (in ppm). When multiple peaks occur, the following abbreviations will be used: s (single, singlet), d (doublet ), t (triplet), q (quartet), m (multiplet), br (broadened, broad), dd (doublet of doublets, doublet), dt (doublet of triplets, doublet), ddd (triplet), qd (quadruple). Coupling constant J, expressed in hertz (Hz).
The measurement conditions for low resolution Mass Spectrometry (MS) data are: agilent 6120 four-stage HPLC-M (column type: zorbax SB-C18,2.1x30mm,3.5 μm, 6min, flow rate 0.6mL/min. Mobile phase: 5% -95% (CH containing 0.1% formic acid) 3 CN) in (H containing 0.1% formic acid) 2 O), using electrospray ionization (ESI), at 210nm/254nm, using UV detection.
The purity of the compound was determined by High Performance Liquid Chromatography (HPLC), using Agilent 1260HPLC (column model: agilent zorbax Eclipse Plus C), and detected by a DAD detector, and finally calculated by area normalization.
The following abbreviations are used throughout the present invention:
Ac 2 o acetic anhydride/acetic anhydride; etOH ethanol;
AcOH acetic acid/acetic acid; et (Et) 3 N, TEA triethylamine;
silver AgOTf triflate; EDCI 1-ethyl- (3-dimethylaminopropyl) carbodiimide salt
Boc 2 Di-tert-butyl O dicarbonate; an acid salt;
CDC1 3 deuterated chloroform; HOBT, HOBT 1-hydroxybenzotriazole;
CDI N, N-carbonyldiimidazole; i-PrOH isopropanol;
DCM dichloromethane; i-PrMgBr isopropyl magnesium bromide;
DMFN, N-dimethylformamide; potassium KOAc acetate;
DIPEA N, N-diisopropylethylamine; t-BuOK potassium tert-butoxide;
DMSO dimethyl sulfoxide; CH (CH) 3 CN, ACN acetonitrile;
DMSO-d 6 deuterated dimethyl sulfoxide; meOH methanol;
DMAP 4-dimethylaminopyridine; msCl methylsulfonyl chloride;
EtOAc, EA ethyl acetate; pd (Pd) 2 (dba) 3 Tris (dibenzylideneacetone) dipalladium;
Pd(dppf)Cl 2 [1,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride; g;
Pd(P(o-Tol) 3 ) 2 (OAc) 2 diacetoxybis (tri-o-tolylphosphine) -h hours;
palladium (II); min;
PE petroleum ether; mmol millimoles;
TMSCF 3 (trifluoromethyl) trimethylsilane; temperature of c degrees celsius;
Tf 2 o-trifluoromethanesulfonic anhydride; mL of water;
TBSOTf t-butyldisilyl triflate; rpm revolutions per minute;
ruphos 2-dicyclohexylphosphine-2 ',6' -diisopropyloxybiphenyl; aq. solution;
TMSCF 2 Br (bromodifluoromethyl) trimethylsilane; m moles per liter;
Pd/C palladium/carbon nM nanomole;
TLC thin layer chromatography; mu M micromolar;
THF tetrahydrofuran; DTT dithiothreitol;
TFA trifluoroacetic acid; mu L microliters;
TBAF tetrabutylammonium fluoride; selectFluor selective fluorinating agent;
XantPhos4, 5-bis-diphenylphosphine-9, 9-dimethylxanthene; sodium STAB triacetoxyborohydride;
typical synthetic procedures for preparing the disclosed compounds are shown in the synthetic schemes below. Unless otherwise indicated, ring B, R, R 2 、R 3 、R 4 、Z 1 、Z 2 、Z 3 、Z 4 、Z 5 、Z 6 、Z 7 、Z 8 、R a 、R b 、R c 、R d N and m have the meaning described in the present invention.
In the following synthetic scheme, X represents CH 2 、NR X Or O; wherein R is X Is C 1-6 Alkyl, C 1-6 Haloalkyl, C 3-8 Cycloalkyl, -C 0-6 alkylene-OR a 、-C 0-4 Alkylene- (C (=O)) m -R b 、-C 0-4 Alkylene- (O) n -(C(=O)) m -NR c R d Or a 5-to 10-membered heteroaryl; y represents a halogen atom; PG represents an amino protecting group; PG 2 Represents a hydroxyl protecting group; ms represents a methanesulfonyl leaving group, T 1 And T 2 Is carboxyl or amino, and T 1 And T is 2 And not the same group.
Synthesis scheme 1
Compound (6 a) can be prepared by the following procedure:
the compound (1 a) and the compound (1 a ') or the compound (1 a ') are subjected to substitution reaction to generate a compound (2 a), the compound (2 a) is deprotected to generate a compound (3 a), the compound (3 a) and the compound (3 a ') are subjected to coupling reaction to generate a compound (4 a), and the compound (4 a) is subjected to hydrolysis reaction to generate a compound (5 a). The compound (5 a) is condensed with the compound (5 a') to give the compound (6 a).
Synthesis scheme 2
/>
Compound (9 b) can be prepared by the following procedure:
the compound (1 a) is subjected to substitution reaction to generate a compound (2 b), the compound (2 b) is subjected to deamination under acidic conditions (such as trifluoroacetic acid) to obtain a compound (3 b), the compound (3 b) is subjected to coupling reaction with the compound (3 a ') under the action of a coupling reagent to generate a compound (4 b), the compound (4 b) is subjected to oxygen atom protection group removal to obtain a compound (5 b), the compound (5 b) is reacted with a sulfonyl compound to obtain a compound (6 b), the compound (6 b) is subjected to sulfonyl leaving group removal, the compound (7 b) is subjected to amino substitution reaction to obtain a compound (7 b), the compound (7 b) and the compound (1 a ') are subjected to substitution or coupling reaction to obtain a compound (8 b), and the compound (8 b) and the compound (5 a ') are subjected to condensation reaction to obtain a compound (9 b).
Synthesis scheme 3
The compound (6 c) or (6 c') can be produced by the following process:
the compound (1 c) and the compound (2 c) are subjected to condensation reaction to generate a compound (3 c) or (3 c '), the compound (3 c) or (3 c') and the compound (3 c) are subjected to substitution reaction to generate a compound (4 c) or (4 c '), the compound (4 c) or (4 c') is subjected to deamination protecting group to generate a compound (5 c) or (5 c '), and the compound (5 c) or (5 c') and the compound (5 c ') are reacted to obtain a compound (6 c) or (6 c').
Synthesis scheme 4
Compound (6 d) can be prepared by the following procedure:
the compound (1 d) and the compound (1 d ') are subjected to substitution reaction under alkaline conditions to generate a compound (2 d), the compound (2 d) is deaminated and protected under acidic conditions (such as trifluoroacetic acid) to generate a compound (3 d), the compound (3 d) and the compound (3 a ') are subjected to coupling reaction to generate a compound (4 d), the compound (4 d) is hydrolyzed under alkaline conditions to generate a compound (5 d), and the compound (5 a ') are condensed under the action of a condensation reagent to obtain a compound (6 d).
The compounds, pharmaceutical compositions and uses thereof provided by the present invention are further described below in conjunction with the examples.
Examples
Example 12- (4- (ethylsulfonyl) phenyl) -N- (4- (1- (4- (trifluoromethyl) benzyl) piperidin-3-yl) phenyl) acetamide
Step one: synthesis of tert-butyl 3- (((trifluoromethyl) sulfonyl) oxy) -5, 6-dihydropyridine-1 (2H) -carboxylate
Under nitrogen, 3-oxopiperidine-1-carboxylic acid tert-butyl ester (2.00 g,10.0 mmol) and DMAP (3.80 g,30.8 mmol) were dissolved in DCM (30 mL) and cooled at-10deg.C with stirring to obtain Tf 2 O (2.60 mL,13.0 mmol) was slowly added dropwise to the reaction system, and the mixture was stirred at room temperature for 5h. The reaction was then quenched with water (35 mL) in an ice bath, extracted with DCM (30 mL. Times.3), the organic phases combined and dried over anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by column chromatography on silica gel (eluent: PE/EtOAc (v/v) =10/1) to give a colorless transparent liquid (1.15 g, 35%).
Step two: synthesis of (4- (2- (4- (ethylsulfonyl) phenyl) acetamido) phenyl) boronic acid
(4-aminophenyl) boronic acid (600 mg,3.49 mmol), 2- (4- (ethylsulfonyl) phenyl) acetic acid (800 mg,3.50 mmol), EDCI (1.10 g,5.62 mmol) and HOBt (600 mg,4.30 mmol) were dissolved in DCM (50 mL) and stirred at room temperature for 7h. The reaction was quenched by addition of HCl solution (30 mL,1 mol/L) to precipitate a large amount of a white solid, which was filtered to give a white solid (820 mg, 67%).
MS(ESI,pos.ion)m/z=348.1[M+H] + .
Step three: synthesis of tert-butyl 5- (4- (2- (4- (ethylsulfonyl) phenyl) acetamido) phenyl) -3, 4-dihydropyridine-1 (2H) -carboxylate
(4- (2- (4- (ethylsulfonyl) phenyl) acetamido) phenyl) boronic acid (210 mg,0.60 mmol), 3- (((trifluoromethyl) sulfonyl) oxy) -5, 6-dihydropyridine-1 (2H) -carboxylic acid tert-butyl ester (220 mg,0.66 mmol), pd (dppf) Cl under nitrogen 2 (101 mg,0.13 mmol) and Cs 2 CO 3 (500 mg,1.53 mmol) was dissolved in 1, 4-dioxane (40 mL), and the temperature was raised to 105℃and stirred for 7h. Subsequently cooled to room temperature, filtered, and the filtrate concentrated under reduced pressure, and the crude product was separated by column chromatography on silica gel (eluent: PE/EtOAc (v/v) =3/1) to give a pale yellow solid (103 mg, 35%).
MS(ESI,pos.ion)m/z=485.6[M+H] + .
Step four: synthesis of tert-butyl 3- (4- (2- (4- (ethylsulfonyl) phenyl) acetamido) phenyl) piperidine-1-carboxylate
Pd/C (860 mg, 5%) was added to a solution of tert-butyl 5- (4- (2- (4- (ethylsulfonyl) phenyl) acetamido) phenyl) -3, 4-dihydropyridine-1 (2H) -carboxylate (200 mg,0.41 mmol) in MeOH (50 mL) under hydrogen atmosphere, stirred at room temperature for 4H, followed by filtration over celite, and the filtrate concentrated under reduced pressure to give the crude product which was separated by column chromatography over silica gel (eluent: DCM/MeOH (v/v) =20/1) to give a yellow solid (90 mg, 45%).
MS(ESI,pos.ion)m/z=509.3[M+23] + .
Step five: synthesis of 2- (4- (ethylsulfonyl) phenyl) -N- (4- (piperidin-3-yl) phenyl) acetamide
3- (4- (2- (4- (ethylsulfonyl) phenyl) acetamido) phenyl) piperidine-1-carboxylic acid tert-butyl ester (200 mg,0.41 mmol) was dissolved in DCM (50 mL) and TFA (5 mL) and stirred at room temperature for 0.5h. The reaction solution was concentrated under reduced pressure to give a pale yellow liquid (151 mg, 95%).
MS(ESI,pos.ion)m/z=387.5[M+H] + .
Step six: synthesis of 2- (4- (ethylsulfonyl) phenyl) -N- (4- (1- (4- (trifluoromethyl) benzyl) piperidin-3-yl) phenyl) acetamide
2- (4- (ethylsulfonyl) phenyl) -N- (4- (piperidin-3-yl) phenyl) acetamide (100 mg,0.26 mmol) and 4- (trifluoromethyl) benzaldehyde (0.10 mL,0.70 mmol) were dissolved in MeOH (20 mL) and NaBH was added under ice-bath 3 CN (100 mg,1.29 mmol), the reaction was stirred at room temperature for 5h. The reaction was concentrated under reduced pressure to remove the organic solvent, and the crude product was separated by column chromatography on silica gel (eluent: DCM/MeOH (v/v) =15/1) to give a pale yellow solid (30 mg, 21%).
MS(ESI,pos.ion)m/z=545.6[M+H] + .
1 H NMR(400MHz,CDCl 3 )δ(ppm):7.89(d,J=8.1Hz,2H),7.72(d,J=2.4Hz,1H),7.65(d,J=7.9Hz,2H),7.60–7.54(m,4H),7.40(d,J=8.3Hz,2H),7.08(d,J=8.4Hz,2H),3.92(dd,J=23.6,13.4Hz,2H),3.82(s,2H),3.14(q&m,J=7.3Hz,4H),2.97(d,J=8.8Hz,1H),2.40(t,J=9.7Hz,2H),1.96–1.84(m,3H),1.56–1.46(m,1H),1.30(t,J=7.5Hz,3H).
Example 22- (4- (ethylsulfonyl) phenyl) -N- (4- (4- (4- (trifluoromethyl) benzyl) piperazin-1-yl) phenyl) acetamide
Step one: synthesis of tert-butyl 4- (4-nitrophenyl) piperazine-1-carboxylate
1-fluoro-4-nitrobenzene (4.00 g,27.78 mmol) and piperazine-1-carboxylic acid tert-butyl ester (5.40 g,29.01 mmol) were dissolved in DCM (100 mL) under nitrogen and stirred at room temperature for 63h. The reaction was concentrated under reduced pressure, and the crude product was separated by column chromatography on silica gel (eluent: PE/EtOAc (v/v) =4/1) to give the product as a yellow solid (860 mg, 10%).
Step two: synthesis of tert-butyl 4- (4-aminophenyl) piperazine-1-carboxylate
Pd/C (172 mg, 10%) was added to a solution of tert-butyl 4- (4-nitrophenyl) piperazine-1-carboxylate (860 mg,2.80 mmol) in EtOH (10 mL) under hydrogen atmosphere and stirred at room temperature for 13h. The reaction solution was filtered through celite, and the filtrate was evaporated under reduced pressure, and the crude product was separated by silica gel column chromatography (eluent: PE/EtOAc (v/v) =4/1) to give a pale yellow solid (570 mg, 73%).
Step three: synthesis of tert-butyl 4- (4- (2- (4- (ethylsulfonyl) phenyl) acetamido) phenyl) piperazine-1-carboxylate
Tert-butyl 4- (4-aminophenyl) piperazine-1-carboxylate (570 mg,2.06 mmol), 2- (4- (ethylsulfonyl) phenyl) acetic acid (720 mg,3.15 mmol), EDCI (830 mg,4.33 mmol) and HOBT (560 mg,4.14 mmol) were dissolved in DCM (13 mL) and stirred at room temperature for 12h. The reaction was diluted with DCM (90 mL) and saturated NaHCO 3 The solution (50 mL) was washed, the aqueous phase extracted with DCM (50 mL. Times.3), the organic phases combined and dried over anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by column chromatography on silica gel (eluent: DCM/EtOAc (v/v) =2/1) to give a yellow solid (596 mg, 59%).
MS(ESI,pos.ion)m/z=488.2[M+H] + .
Step four: synthesis of 2- (4-ethylsulfonylphenyl) -N- (4- (piperazin-1-yl) phenyl) acetamide
Tert-butyl 4- (4- (2- (4- (ethylsulfonyl) phenyl) acetamido) phenyl) piperazine-1-carboxylate (412 mg,0.85 mmol) was dissolved in DCM/TFA (10 mL/4 mL) and stirred at room temperature for 5h. The reaction was concentrated under reduced pressure to remove the organic solvent, and the residue was diluted with DCM (130 mL) and taken up in saturated Na 2 CO 3 The solution (50 mL) was washed, the aqueous phase was extracted with DCM (30 ml×3), the organic phases were combined, concentrated under reduced pressure, and the crude product was separated by column chromatography on silica gel (eluent: DCM/MeOH (v/v) =10/1) to give a white solid (300 mg, 92%).
MS(ESI,pos.ion)m/z=388.5[M+H] + .
Step five: synthesis of 2- (4- (ethylsulfonyl) phenyl) -N- (4- (4- (4- (trifluoromethyl) benzyl) piperazin-1-yl) phenyl) acetamide
2- (4- (ethylsulfonyl) phenyl) -N- (4- (piperazin-1-yl) phenyl) acetamide (125 mg,0.32 mmol), 4- (trifluoromethyl) benzaldehyde (170 mg,0.98 mmol) was dissolved in DCM (10 mL) under nitrogen, acOH (19 mg,0.32 mmol) was added, stirred at room temperature for 1h, then NaBH was added 3 CN (45 mg,0.72 mmol), was stirred at room temperature for 19h. Saturated NaHCO is added into the system 3 The reaction was quenched with solution (10 mL), diluted with DCM (90 mL) and successively saturated NaHCO 3 Solution (20 mL) and saturationAnd NaCl solution (30 mL), anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by silica gel column chromatography (eluent: DCM/MeOH (v/v) =20/1) to give a white solid (70 mg, 40%).
MS(ESI,pos.ion)m/z=546.1[M+H] + .
1 H NMR(400MHz,CDCl 3 )δ(ppm):7.86(d,J=7.7Hz,2H),7.58(d,J=7.8Hz,2H),7.53(d,J=7.9Hz,2H),7.47(d,J=7.8Hz,2H),7.34(d,J=8.8Hz,2H),6.85(d,J=8.8Hz,2H),3.75(s,2H),3.60(s,2H),3.15(t,J=4.2Hz,4H),3.11(q,J=7.6Hz,2H),2.60(t,J=4.4Hz,4H),1.28(t,J=7.6Hz,3H).
Example 32- (4- (ethylsulfonyl) phenyl) -N- (4- (4- (((1R, 4R) -4- (trifluoromethyl) cyclohexyl) methyl) piperazin-1-yl) phenyl) acetamide
2- (4- (ethylsulfonyl) phenyl) -N- (4- (piperazin-1-yl) phenyl) acetamide (25 mg,0.07 mmol) and 4- (trifluoromethyl) cyclohexyl formaldehyde (35 mg,0.19 mmol) were dissolved in DCM (5 mL) under nitrogen, acOH (4 mg,0.07 mmol) was added, stirred at room temperature for 1h, then NaBH was added 3 CN (23 mg,0.37 mmol), was stirred at room temperature for 24h. Saturated NaHCO is added into the system 3 The reaction was quenched with solution (6 mL), diluted with DCM (90 mL) and successively saturated NaHCO 3 Saturated solution (20 mL) and saturated NaCl solution (30 mL), the aqueous phase was extracted with DCM (30 mL. Times.3), the organic phases were combined, and the aqueous phase was washed with anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by silica gel column chromatography (eluent: DCM/MeOH (v/v) =30/1) to give a white solid (25 mg, 70%).
MS(ESI,pos.ion)m/z=552.3[M+H] + .
1 H NMR(400MHz,CDCl 3 )δ(ppm):7.88(d,J=8.1Hz,2H),7.54(d,J=8.1Hz,2H),7.33(d,J=8.8Hz,2H),7.20(br,1H),6.85(d,J=8.8Hz,2H),3.77(s,2H),3.18-3.07(m,6H),2.57–2.50(m,4H),2.19(d,J=7.0Hz,2H),2.06-1.88(m,5H),1.78-1.63(m,5H),1.31(t,J=7.3Hz,3H).
Example 42- (4- (ethylsulfonyl) phenyl) -N- (4- (4- ((1R, 4R) -4- (trifluoromethyl) cyclohexylformyl) piperazin-1-yl) phenyl) acetamide
2- (4- (ethylsulfonyl) phenyl) -N- (4- (piperazin-1-yl) phenyl) acetamide (220 mg,0.57 mmol), 4- (trifluoromethyl) cyclohexyl carboxylic acid (67 mg,0.85 mmol), EDCI (219 mg,1.14 mmol) and HOBT (156 mg,1.15 mmol) were dissolved in DCM (10 mL) and stirred at room temperature for 18h. The reaction was diluted with DCM (90 mL) and saturated NaHCO 3 Solution (50 mL) wash, anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by silica gel column chromatography (eluent: DCM/MeOH (v/v) =40/1) to give a white solid (250 mg, 78%).
MS(ESI,pos.ion)m/z=566.1[M+H] + .
1 H NMR(400MHz,CDCl 3 )δ(ppm):7.93(s,1H),7.81(d,J=8.2Hz,2H),7.49(d,J=8.2Hz,2H),7.37(d,J=8.9Hz,2H),6.82(d,J=9.0Hz,2H),3.73(s,4H),3.64(s,2H),3.08(dd,J=14.8,7.4Hz,6H),2.51(ddd,J=11.8,8.7,3.3Hz,1H),2.09–1.99(m,3H),1.87(d,J=14.0Hz,2H),1.66–1.53(m,2H),1.43–1.33(m,2H),1.25(t,J=7.4Hz,3H).
Example 5N- ((R) -1- (5- (ethylsulfonyl) pyridin-2-yl) -2-hydroxyethyl) -4- ((S) -3- (4- (trifluoromethyl) phenoxy) piperidin-1-yl) benzamide
Step one: synthesis of (S) -3- (4- (trifluoromethyl) phenoxy) piperidine-1-carboxylic acid tert-butyl ester
NaH (403 mg,10.1mmol, 60%) was added to a solution of tert-butyl (S) -3-hydroxypiperidine-1-carboxylate (1.0 g,5.0 mmol) and 1-bromo-4- (trifluoromethyl) benzene (2.0 mL,14.0 mmol) in DMF (20 mL) under stirring in an ice bath for 20min, and after stirring the reaction was heated by 80℃in an oil bath for 21h. EtOAc (100 mL) was added to the reaction solution,then using saturated NH 4 Cl (aq.) (30 mL. Times.5) washed, anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by column chromatography on silica gel (eluent: PE/DCM/EtOAc (v/v/v) =10/1/1) to give a yellow solid (629 mg, 37%).
MS(ESI,pos.ion)m/z=290.1[M+H-56] + .
Step two: synthesis of (S) -3- (4- (trifluoromethyl) phenoxy) piperidine
Tert-butyl (S) -3- (4- (trifluoromethyl) phenoxy) piperidine-1-carboxylate (1.35 g,3.91 mmol) was dissolved in HCl in MeOH (20 mL) and stirred at room temperature for 2h. The starting material was essentially converted as tracked by TLC. Saturated Na was added to the reaction solution 2 CO 3 (aq.) (50 mL) quench the reaction, then extract the aqueous phase with DCM (40 mL. Times.3), combine the organic phases and wash with saturated NaCl solution (30 mL), anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by silica gel column chromatography (eluent: DCM/MeOH (v/v) =10/1) to give a yellow oil (639 mg, 67%). MS (ESI, pos.ion) m/z=246.3 [ m+h ]] + .
Step three: synthesis of methyl (S) -4- (3- (4- (trifluoromethyl) phenoxy) piperidin-1-yl) benzoate
(S) -3- (4- (trifluoromethyl) phenoxy) piperidine (200 mg,0.82 mmol), methyl 4-iodobenzoate (430 mg,1.64 mmol), pd under nitrogen 2 (dba) 3 (40 mg,0.044 mmol), xantPhos (35 mg,0.06 mmol) and Cs 2 CO 3 (530 mg,1.62 mmol) was dissolved in 1, 4-dioxane (4 mL) and the reaction was heated in an oil bath at 100deg.C and stirred for 22h. Saturated NaHCO is added into the reaction solution 3 The solution (30 mL) was then extracted with DCM (40 mL. Times.3), the organic phases combined and washed with saturated NaCl solution (20 mL), anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by column chromatography on silica gel (eluent: PE/DCM/EtOAc (v/v/v) =10/1/1) to give a yellow solid (200 mg, 65%). MS (ESI, pos.ion) m/z=380.1 [ m+h ]] + .
Step four: synthesis of (S) -4- (3- (4- (trifluoromethyl) phenoxy) piperidin-1-yl) benzoic acid
LiOH.H 2 O (122 mg,2.91 mmol) was added to (S) -4- (3- (4- (trifluoromethyl) phenoxy) piperazine)Methyl pyridin-1-yl benzoate (200 mg,0.53 mmol) in THF/MeOH (3 mL/3 mL) was stirred at room temperature for 12h. Then heated by an oil bath at 60 ℃ for 1.0h, the raw materials remain in a large quantity. Additional THF/H 2 O (6 mL/3 mL), 60 ℃ oil bath heating for 1.0h, the raw materials are completely converted. The reaction was diluted with DCM (60 mL) and water (20 mL) and then 1.0M HCl solution was added dropwise to adjust the ph=2 to 3, the solution was separated, the aqueous phase was extracted with DCM (20 ml×2), the organic phases were combined and the aqueous Na phase was used 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by column chromatography on silica gel (eluent: DCM/EtOAc (v/v) =5/1) to give a pale yellow solid (190 mg, 98%).
MS(ESI,pos.ion)m/z=366.1[M+H] + .
Step five: synthesis of N- ((R) -1- (5- (ethylsulfonyl) pyridin-2-yl) -2-hydroxyethyl) -4- ((S) -3- (4- (trifluoromethyl) phenoxy) piperidin-1-yl) benzamide
(S) -4- (3- (4- (trifluoromethyl) phenoxy) piperidin-1-yl) benzoic acid (90 mg,0.25 mmol), (R) -2-amino-2- (5- (ethylsulfonyl) pyridin-2-yl) ethanol (90 mg,0.30 mmol), EDCI (98 mg,0.51 mmol) and HOBT (70 mg,0.52 mmol) were dissolved in DCM (6 mL) and TEA (0.18 mL,1.30 mmol) was added and stirred at room temperature for 12h. The reaction was diluted with DCM (60 mL) and saturated NaHCO 3 Solution (20 mL) and NaCl solution (20 mL) washing, anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by column chromatography on silica gel (eluent: DCM/EtOAc (v/v) =1/1) to give a yellow solid (60 mg, 42%).
MS(ESI,pos.ion)m/z=578.0[M+H] + .
1 H NMR(400MHz,DMSO-d 6 )δ(ppm):8.79(s,1H),8.55(d,J=7.7Hz,1H),8.11(dd,J=8.2,1.8Hz,1H),8.03(d,J=8.0Hz,1H),7.77(d,J=8.8Hz,2H),7.64(d,J=8.6Hz,2H),7.15(d,J=8.6Hz,2H),6.96(d,J=8.8Hz,2H),5.16(dd,J=13.8,6.8Hz,1H),5.10(t,J=5.8Hz,1H),4.64(ddd,J=11.6,7.2,4.1Hz,1H),3.83–3.70(m,3H),3.52(m,2H),3.42(m,3H),3.30–3.22(m,1H),3.15(m,1H),2.09–2.15(m,1H),1.89–1.79(m,1H),1.67(s,2H),1.13(t,J=7.4Hz,3H).
Example 6N- ((R) -1- (4- (ethylsulfonyl) phenyl) -2-hydroxyethyl) -4- ((R) -3- (4- (trifluoromethyl) phenoxy) piperidin-1-yl) benzamide
Step one: synthesis of tert-butyl (R) - (3- (4- (trifluoromethyl) phenoxy) piperidin-1-yl) formate
NaH (400 mg,10.00mmol, 60%) was added to a solution of tert-butyl (R) - (3-hydroxypiperidin-1-yl) carboxylate (1.00 g,4.97 mmol) and 1-bromo-4- (trifluoromethyl) benzene (2 mL,14.00 mmol) in DMF (20 mL) under stirring in an ice bath for 20min, and after stirring the reaction was heated in an 80℃oil bath for 21h. The reaction mixture was cooled to room temperature, etOAc (100 mL) was added to the reaction mixture, and the mixture was saturated with NH 4 Cl solution (30 mL. Times.5) washing, anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by column chromatography on silica gel (eluent: PE/DCM/EtOAc (v/v/v) =10/1/1) to give a yellow solid (700 mg, 41%).
MS(ESI,pos.ion)m/z=290.1[M+H-56] + .
Step two: synthesis of (R) -3- (4- (trifluoromethyl) phenoxy) piperidine
Tert-butyl (R) - (3- (4- (trifluoromethyl) phenoxy) piperidin-1-yl) carboxylate (1.35 g,3.91 mmol) was dissolved in HCl in methanol (20 mL) and stirred at room temperature for 2h. Saturated Na was added to the reaction solution 2 CO 3 The reaction was quenched with solution (50 mL), the aqueous phase extracted with DCM (40 mL. Times.3), the organic phases combined and washed with saturated NaCl solution (30 mL), anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by silica gel column chromatography (eluent: DCM/MeOH (v/v) =10/1) to give a yellow oil (639 mg, 67%).
MS(ESI,pos.ion)m/z=246.3[M+H] + .
Step three: synthesis of methyl (R) -4- (3- (4- (trifluoromethyl) phenoxy) piperidin-1-yl) benzoate
(R) -3- (4- (trifluoromethyl) phenoxy) piperidine (200 mg,0.82 mmol), methyl 4-iodobenzoate (430 mg,1.64 mmol), pd under nitrogen 2 (dba) 3 (40 mg,0.04 mmol), xantPhos (35 mg,0.06 mmol) and Cs 2 CO 3 (530mg,1.62 mmol) was dissolved in 1, 4-dioxane (4 mL) and the reaction was heated in an oil bath at 100deg.C and stirred for 22h. The reaction solution was cooled to room temperature and saturated NaHCO was added 3 The solution (30 mL), the aqueous phase was extracted with DCM (40 mL. Times.3), the organic phases combined and washed with saturated NaCl solution (20 mL), anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by column chromatography on silica gel (eluent: PE/DCM/EtOAc (v/v/v) =10/1/1) to give a yellow solid (210 mg, 68%).
MS(ESI,pos.ion)m/z=380.1[M+H] + .
Step four: synthesis of (R) -4- (3- (4- (trifluoromethyl) phenoxy) piperidin-1-yl) benzoic acid
LiOH.H 2 O (122 mg,2.91 mmol) was added to a solution of methyl (R) -4- (3- (4- (trifluoromethyl) phenoxy) piperidin-1-yl) benzoate (200 mg,0.53 mmol) in THF/MeOH (3 mL/3 mL) and the reaction was heated in an oil bath at 60℃under stirring for 8h. The reaction mixture was cooled to room temperature, diluted with DCM (60 mL) and water (20 mL), HCl solution (1 mol/L) was added dropwise to adjust the pH of the system to about 3, the mixture was separated, the aqueous phase was extracted with DCM (20 mL. Times.2), the organic phases were combined and dried over anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by column chromatography on silica gel (eluent: DCM/EtOAc (v/v) =5/1) to give a pale yellow solid (192 mg, 99%).
MS(ESI,pos.ion)m/z=366.1[M+H] + .
Step five: synthesis of N- ((R) -1- (4- (ethylsulfonyl) phenyl) -2-hydroxyethyl) -4- ((S) -3- (4- (trifluoromethyl) phenoxy) piperidin-1-yl) benzamide
(R) -4- (3- (4- (trifluoromethyl) phenoxy) piperidin-1-yl) benzoic acid (192 mg,0.52 mmol), (R) -2-amino-2- (4- (ethylsulfonyl) phenyl) ethanol (145 mg,0.63 mmol), EDCI (200 mg,1.04 mmol) and HOBT (140 mg,1.04 mmol) were dissolved in DCM (10 mL) and TEA (0.22 mL,1.60 mmol) was added and stirred at room temperature for 12h. The reaction was diluted with DCM (60 mL) and successively saturated NaHCO 3 Solution (20 mL) and NaCl solution (20 mL) washing, anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by silica gel column chromatography (eluent: DCM/EtOAc (v/v) =1/1) to give pale yellow powder (210 mg, 70%).
MS(ESI,pos.ion)m/z=577.2[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ(ppm):8.49(d,J=7.8Hz,1H),7.82(d,J=8.3Hz,2H),7.78(d,J=8.7Hz,2H),7.64(d,J=7.3Hz,4H),7.16(d,J=8.6Hz,2H),6.96(d,J=8.9Hz,2H),5.12(dd,J=14.0,7.0Hz,1H),5.01(t,J=5.8Hz,1H),4.68–4.61(m,1H),3.78(dd,J=13.3,2.6Hz,1H),3.70(qd,J=11.4,5.4Hz,2H),3.56-3.48(m,1H),3.25(q,J=7.3Hz,2H),3.29–3.22(m,1H),3.18–3.12(m,1H),2.14–2.06(m,1H),1.89–1.79(m,1H),1.67(t,J=7.9Hz,2H),1.09(t,J=7.3Hz,3H).
Example 7N- ((R) -1- (5- (ethylsulfonyl) pyridin-2-yl) -2-hydroxyethyl) -4- ((R) -3- (4- (trifluoromethyl) phenoxy) piperidin-1-yl) benzamide
(R) -4- (3- (4- (trifluoromethyl) phenoxy) piperidin-1-yl) benzoic acid (90 mg,0.25 mmol), (R) -2-amino-2- (5- (ethylsulfonyl) pyridin-2-yl) ethanol (90 mg,0.30 mmol), EDCI (98 mg,0.51 mmol) and HOBT (70 mg,0.52 mmol) were dissolved in DCM (6 mL) and TEA (0.18 mL,1.30 mmol) was added and stirred at room temperature for 12h. The reaction was diluted with DCM (60 mL) and saturated NaHCO 3 Solution (20 mL) and NaCl solution (20 mL) washing, anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by column chromatography on silica gel (eluent: DCM/EtOAc (v/v) =1/1) to give a yellow solid (70 mg, 49%).
MS(ESI,pos.ion)m/z=578.1[M+H] + .
1 H NMR(400MHz,DMSO-d 6 )δ(ppm):8.79(s,1H),8.55(d,J=7.6Hz,1H),8.11(dd,J=8.1,1.7Hz,1H),8.03(d,J=8.1Hz,1H),7.77(d,J=8.8Hz,2H),7.64(d,J=8.6Hz,2H),7.15(d,J=8.6Hz,2H),6.96(d,J=8.8Hz,2H),5.16(dd,J=13.8,6.7Hz,1H),5.11(t,J=5.7Hz,1H),4.69–4.59(m,1H),3.76(m,3H),3.52(d,J=13.4Hz,1H),3.42(q,J=7.4Hz,2H),3.31–3.21(m,1H),3.19–3.12(m,1H),2.10(d,J=11.2Hz,1H),1.83(d,J=10.2Hz,1H),1.73–1.60(m,2H),1.13(t,J=7.4Hz,3H).
Example 8 (R) -N- (1- (4- (ethylsulfonyl) phenyl) -2-hydroxyethyl) -4- (2-oxo-3- (4- (trifluoromethyl) benzyl) tetrahydropyrimidin-1 (2H) -yl) benzamide
Step one: synthesis of tert-butyl (3- ((4- (trifluoromethyl) benzyl) amino) propyl) carbamate
Tert-butyl (3-aminopropyl) carbamate (1.03 g,5.91 mmol) and 4- (trifluoromethyl) benzaldehyde (1.53 g,8.79 mmol) were dissolved in EtOH (10 mL) in ice bath and stirred at room temperature for 1.0h, naBH was added 3 CN (1.12 g,17.8 mmol), stirring was continued for 6h at RT. Saturated Na was added to the reaction solution 2 CO 3 The reaction was quenched with solution (30 mL), the aqueous phase extracted with DCM (30 Ml. Times.3), the organic phases combined and washed with saturated NaCl solution (20 mL), anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by silica gel column chromatography (eluent: DCM/MeOH (v/v) =10/1) to give a yellow oil (844 mg, 43%).
MS(ESI,pos.ion)m/z=333.3[M+H] + .
Step two: synthesis of 1- (4- (trifluoromethyl) benzyl) tetrahydropyrimidin-2 (1H) -one
t-BuOK (863 mg,7.71 mmol) was added to a solution of tert-butyl (3- ((4- (trifluoromethyl) benzyl) amino) propyl) carbamate (844 mg,2.54 mmol) in THF (10 mL) under nitrogen with stirring in an ice bath, and after the addition, the mixture was stirred at room temperature for 30min, and the reaction was heated in an oil bath at 60℃for 27h with stirring. The reaction was cooled to room temperature, water was added to dissolve the solid, the aqueous phase was extracted with DCM (40 mL. Times.3), the organic phases were combined and washed with saturated NaCl solution (20 mL), anhydrous Na 2 SO 4 Drying and concentration under reduced pressure gave the crude product as a yellow solid (650 mg, 99%).
MS(ESI,pos.ion)m/z=259.3[M+H] + .
Step three: synthesis of methyl 4- (2-oxo-3- (4- (trifluoromethyl) benzyl) tetrahydropyrimidin-1 (2H) -yl) benzoate
Under nitrogen, 1- (4- (trifluoromethyl) benzyl) tetrahydropyrimidin-2 (1H) -one (655 mg,2.54 mmo) l), methyl 4-bromobenzoate (1.03 g,4.79 mmol), pd 2 (dba) 3 (123 mg,0.13 mmol), xantPhos (116 mg,0.20 mmol) and Cs 2 CO 3 (2.51 g,7.65 mmol) was dissolved in 1, 4-dioxane (10 mL) and the reaction was heated in an oil bath at 100deg.C and stirred for 22h. The reaction was cooled to room temperature and concentrated under reduced pressure, and the crude product was separated by column chromatography on silica gel (eluent: PE/DCM/EtOAc (v/v/v) =2/1/1) to give the target as a yellow solid (487 mg, 49%).
MS(ESI,pos.ion)m/z=393.3[M+H] + .
Step four: synthesis of 4- (2-oxo-3- (4- (trifluoromethyl) benzyl) tetrahydropyrimidin-1 (2H) -yl) benzoic acid
LiOH.H 2 O (265 mg,6.32 mmol) methyl 4- (2-oxo-3- (4- (trifluoromethyl) benzyl) tetrahydropyrimidin-1 (2H) -yl) benzoate (487 mg,1.24 mmol) in THF/H 2 In O (10 mL/3 mL) solution, the reaction was stirred for 18h with heating in an oil bath at 50 ℃. Water (20 mL) was added to the reaction solution, HCl solution (1 mol/L) was added dropwise to the system, the pH of the solution was adjusted to about 3, and the aqueous phase was concentrated under reduced pressure to give a crude product as a yellow solid (460 mg, 99%).
Step five: synthesis of (R) -N- (1- (4- (ethylsulfonyl) phenyl) -2-hydroxyethyl) -4- (2-oxo-3- (4- (trifluoromethyl) benzyl) tetrahydropyrimidin-1 (2H) -yl) benzamide
4- (2-oxo-3- (4- (trifluoromethyl) benzyl) tetrahydropyrimidin-1 (2H) -yl) benzoic acid (232 mg,0.6133 mmol), (R) -2-amino-2- (4- (ethylsulfonyl) phenyl) ethanol (173 mg,0.75 mmol), EDCI (235 mg,1.23 mmol) and HOBT (167 mg,1.24 mmol) were dissolved in DCM (12 mL) and TEA (0.26 mL,1.9 mmol) was added and stirred at room temperature for 20H. DCM (60 mL) was added to the reaction, followed by saturated NaCl solution (20 mL) and saturated NaHCO 3 Solution (20 mL) wash, anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by silica gel column chromatography (eluent: DCM/EtOAc (v/v) =1/1) to give a pale yellow solid (225 mg, 62%).
MS(ESI,pos.ion)m/z=590.4[M+H] + .
1 H NMR(400MHz,DMSO-d 6 )δ(ppm):8.74(d,J=7.8Hz,1H),7.88(d,J=8.6Hz,2H),7.85(d,J=8.3Hz,2H),7.73(d,J=8.1Hz,2H),7.67(d,J=8.3Hz,2H),7.53(d,J=8.0Hz,2H),7.43(d,J=8.6Hz,2H),5.15(dd,J=13.7,7.1Hz,1H),5.05(t,J=5.9Hz,1H),4.63(s,2H),3.80–3.66(m,4H),3.39–3.35(m,2H),3.27(q,J=7.3Hz,2H),2.06(dd,J=11.6,6.0Hz,2H),1.10(t,J=7.3Hz,3H).
Example 9N- ((R) -1- (4- (ethylsulfonyl) phenyl) -2-hydroxyethyl) -4- ((S) -6- (methoxymethyl) -2-oxo-3- (4- (trifluoromethyl) benzyl) tetrahydropyrimidin-1 (2H) -yl) benzamide
Step one: synthesis of (S) -2- ((tert-Butoxyformyl) amino) -4- ((4- (trifluoromethyl) benzyl) amino) butanoic acid
(S) -4-amino-2- ((tert-Butoxyformyl) amino) butanoic acid (3.03 g,13.90 mmol) and 4- (trifluoromethyl) benzaldehyde (2.0 mL,15.00 mmol) were dissolved in EtOH (30 mL) in ice bath and stirred at room temperature for 6h, then NaBH was added 3 CN (4.32 g,68.70 mmol) was stirred at room temperature for 10h. The reaction was concentrated under reduced pressure, and the crude product was separated by silica gel column chromatography (eluent: DCM/MeOH (v/v) =10/1) to give a white solid (4.0 g, 77%).
MS(ESI,pos.ion)m/z=377.3[M+H] + .
Step two: synthesis of (S) -4- ((tert-Butoxyformyl) (4- (trifluoromethyl) benzyl) amino) -2- ((tert-Butoxyformyl) amino) butanoic acid
Under ice bath, boc was added to the mixture 2 O (2.40 mL,10.00 mmol) was added (S) -2- ((tert-butoxyformyl) amino) -4- ((4- (trifluoromethyl) benzyl) amino) butanoic acid (4.0 g,11.00 mmol) and NaOH (850 mg,21.25 mmol) in THF/H 2 O (20 mL/20 mL) was added and stirred at room temperature for 8h. HCl solution (1 mol/L) was added to the reaction solution, the pH of the solution was adjusted to about 5, the aqueous phase was extracted with DCM (40 mL. Times.3), the organic phases were combined and the mixture was taken up in anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by column chromatography on silica gel (eluent: PE/EtOAc (v/v) =2/1) to give a pale yellow oil (3.05 g, 60%).
MS(ESI,neg.ion)m/z=475.3[M-H] - .
Step three: synthesis of methyl (S) -2-amino-4- ((4- (trifluoromethyl) benzyl) amino) butyrate
Under ice bath, SOCl 2 (2.8 mL,39 mmol) was added to a solution of (S) -4- ((tert-butoxyformyl) (4- (trifluoromethyl) benzyl) amino) -2- ((tert-butoxyformyl) amino) butanoic acid (3.05 g,6.40 mmol) in MeOH (30 mL), stirred at room temperature for 1h, heated in an oil bath at 80℃for 3h. The reaction solution was cooled to room temperature, concentrated under reduced pressure, and saturated Na was added to the residue 2 CO 3 The reaction was quenched with solution (30 mL), the aqueous phase extracted with DCM (40 mL. Times.3), the organic phases combined and washed with saturated NaCl solution (30 mL), anhydrous Na 2 SO 4 Drying and concentration under reduced pressure gave a pale yellow oily liquid (1.82 g, 98%).
MS(ESI,pos.ion)m/z=291.3[M+H] + .
Step four: synthesis of methyl (S) -2-oxo-1- (4- (trifluoromethyl) benzyl) hexahydropyrimidine-4-carboxylate
CDI (3.00 g,18.50 mmol) was added to a solution of methyl (S) -2-amino-4- ((4- (trifluoromethyl) benzyl) amino) butanoate (1.82 g,6.27 mmol) in THF (60 mL) at-10deg.C, after 30min, stirred at room temperature for 10h. The reaction was concentrated under reduced pressure, and the crude product was separated by column chromatography on silica gel (eluent: DCM/MeOH (v/v) =40/1) to give a yellow transparent liquid (1.20 g, 60%).
MS(ESI,pos.ion)m/z=317.3[M+H] + .
Step five: synthesis of (S) -4- (hydroxymethyl) -1- (4- (trifluoromethyl) benzyl) tetrahydropyrimidin-2 (1H) -one
LiBH is carried out 4 To a solution of methyl (S) -2-oxo-1- (4- (trifluoromethyl) benzyl) hexahydropyrimidine-4-carboxylate (1.20 g,3.79 mmol) in THF (4 mL) was added (6.0 mL,12.00 mmol) and the mixture was stirred at room temperature for 12h. Saturated NaHCO is added into the reaction solution 3 The reaction was quenched with solution (20 mL), the aqueous phase extracted with DCM (30 mL. Times.3), the organic phases combined and washed with saturated NaCl solution (20 mL), anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by silica gel column chromatography (eluent: DCM/MeOH (v/v) =20/1) to give pale yellow solid (274 mg, 25%).
MS(ESI,pos.ion)m/z=289.1[M+H] + .
Step six: synthesis of methyl (S) - (2-oxo-1- (4- (trifluoromethyl) benzyl) hexahydropyrimidin-4-yl) methanesulfonate
MsCl (0.10 mL,1.30 mmol) was added to a solution of (S) -4- (hydroxymethyl) -1- (4- (trifluoromethyl) benzyl) tetrahydropyrimidin-2 (1H) -one (37 mg,0.13 mmol) and TEA (0.60 mL,4.30 mmol) in DCM (4 mL) under ice-bath and stirred at room temperature for 12H. Saturated NaHCO is added into the reaction solution 3 The reaction was quenched with solution (20 mL), the aqueous phase extracted with DCM (30 mL. Times.3), the organic phases combined and dried over anhydrous Na 2 SO 4 Drying and concentration under reduced pressure gave a yellow solid (47 mg, 100%).
MS(ESI,pos.ion)m/z=367.1[M+H] + .
Step seven: synthesis of (S) -4- (methoxymethyl) -1- (4- (trifluoromethyl) benzyl) tetrahydropyrimidin-2 (1H) -one
Will K 2 CO 3 (209 mg,1.51 mmol) was added to a solution of methyl (S) - (2-oxo-1- (4- (trifluoromethyl) benzyl) hexahydropyrimidin-4-yl) methanesulfonate (47 mg,0.13 mmol) in MeOH (10 mL) and the reaction was heated in 66℃oil and stirred for 12h. The reaction was cooled to room temperature, water was added to the reaction until the solid was completely dissolved, the aqueous phase was extracted with DCM (20 mL. Times.3), the organic phases were combined and the aqueous Na was used 2 SO 4 Drying and concentration under reduced pressure gave a yellow solid (38 mg, 98%).
MS(ESI,pos.ion)m/z=303.1[M+H] + .
Step eight: synthesis of methyl (S) -4- (6- (methoxymethyl) -2-oxo-3- (4- (trifluoromethyl) benzyl) tetrahydropyrimidin-1 (2H) -yl) benzoate
(S) -4- (methoxymethyl) -1- (4- (trifluoromethyl) benzyl) tetrahydropyrimidin-2 (1H) -one (38 mg,0.13 mmol), methyl 4-bromobenzoate (54 mg,0.25 mmol), pd under nitrogen 2 (dba) 3 (58 mg,0.06 mmol), xantPhos (54 mg,0.09 mmol) and Cs 2 CO 3 (132 mg,0.40 mmol) was dissolved in 1, 4-dioxane (4 mL) and the reaction was heated in an oil bath at 100deg.C and stirred for 24h. The reaction was cooled to room temperature, concentrated under reduced pressure, and the crude product was separated by column chromatography on silica gel (eluent: PE/DCM/EtOAc (v/v/v) =2/1/1) to give a yellow solid (38 mg, 69%).
MS(ESI,pos.ion)m/z=437.4[M+H] + .
Step nine: synthesis of (S) -4- (6- (methoxymethyl) -2-oxo-3- (4- (trifluoromethyl) benzyl) tetrahydropyrimidin-1 (2H) -yl) benzoic acid
Methyl (S) -4- (6- (methoxymethyl) -2-oxo-3- (4- (trifluoromethyl) benzyl) tetrahydropyrimidin-1 (2H) -yl) benzoate (38 mg,0.09 mmol) and LiOH. H 2 O (80 mg,1.91 mmol) was dissolved in THF/MeOH (1 mL/1 mL) and stirred at room temperature for 5h. Adding water to dissolve solid, adding HCl solution (1 mol/L) dropwise for dissolving, adjusting pH to about 4, extracting the mixed solution with DCM (20 mL×3), mixing the organic phases, and adding anhydrous Na 2 SO 4 Drying and concentration under reduced pressure gave a yellow solid (36 mg, 98%).
MS(ESI,pos.ion)m/z=423.2[M+H] + .
Step ten: synthesis of N- ((R) -1- (4- (ethylsulfonyl) phenyl) -2-hydroxyethyl) -4- ((S) -6- (methoxymethyl) -2-oxo-3- (4- (trifluoromethyl) benzyl) tetrahydropyrimidin-1 (2H) -yl) benzamide
(S) -4- (6- (methoxymethyl) -2-oxo-3- (4- (trifluoromethyl) benzyl) tetrahydropyrimidin-1 (2H) -yl) benzoic acid (36 mg,0.09 mmol), (R) -2-amino-2- (4- (ethylsulfonyl) phenyl) ethanol (30 mg,0.13 mmol), EDCI (50 mg,0.26 mmol) and HOBT (35 mg,0.26 mmol) were dissolved in DCM (4 mL) and TEA (0.10 mL,0.72 mmol) was added and stirred at room temperature for 10H. The reaction was diluted with DCM (60 mL) and saturated NaHCO 3 Solution (20 mL) wash, anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by silica gel column chromatography (eluent: DCM/MeOH (v/v) =20/1) to give a yellow solid (20 mg, 37%).
MS(ESI,pos.ion)m/z=634.2[M+H] + .
1 H NMR(400MHz,DMSO-d 6 )δ(ppm):8.78(d,J=7.8Hz,1H),7.89(d,J=8.5Hz,2H),7.84(d,J=8.3Hz,2H),7.73(d,J=8.0Hz,2H),7.67(d,J=8.3Hz,2H),7.51(d,J=7.9Hz,2H),7.40(d,J=8.5Hz,2H),5.20–5.10(dd,1H),5.05(t,J=5.7Hz,1H),4.61(dd,J=25.2Hz,15.5Hz,2H),4.06-4.14(m,1H),3.80–3.66(m,2H),3.48–3.40(m,2H),3.31(q,J=7.3Hz,2H),3.18(s,3H),2.22–2.13(m,1H),2.10–2.04(m,1H),2.02–1.94(m,2H),1.10(t,J=7.3Hz,3H).
Example 10N- ((R) -1- (4- (ethylsulfonyl) phenyl) -2-hydroxyethyl) -4- ((S) -3- (4- (trifluoromethyl) phenoxy) piperidin-1-yl) benzamide
Step one: synthesis of tert-butyl (S) - (3- (4- (trifluoromethyl) phenoxy) piperidin-1-yl) formate
NaH (403 mg,10.10mmol, 60%) was added to a solution of tert-butyl (S) - (3-hydroxypiperidin-1-yl) carboxylate (1.00 g,4.97 mmol) and 1-bromo-4- (trifluoromethyl) benzene (2 mL,14.00 mmol) in DMF (20 mL) under stirring in an ice bath for 20min, after which the reaction was heated in an 80℃oil bath and stirred for 21h. The reaction mixture was cooled to room temperature, etOAc (100 mL) was added to the reaction mixture, and the mixture was saturated with NH 4 Cl solution (30 mL. Times.5) washing, anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by column chromatography on silica gel (eluent: PE/DCM/EtOAc (v/v/v) =10/1/1) to give a yellow solid (629 mg, 37%).
MS(ESI,pos.ion)m/z=290.1[M+H-56] + .
Step two: synthesis of (S) -3- (4- (trifluoromethyl) phenoxy) piperidine
Tert-butyl (S) - (3- (4- (trifluoromethyl) phenoxy) piperidin-1-yl) carboxylate (1.35 g,3.91 mmol) was dissolved in HCl in MeOH (20 mL) and stirred at room temperature for 2h. Saturated Na was added to the reaction solution 2 CO 3 The reaction was quenched with solution (50 mL), the aqueous phase extracted with DCM (40 mL. Times.3), the organic phases combined and washed with saturated NaCl solution (30 mL), anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by silica gel column chromatography (eluent: DCM/MeOH (v/v) =10/1) to give a yellow oil (639 mg, 67%).
MS(ESI,pos.ion)m/z=246.3[M+H] + .
Step three: synthesis of methyl (S) -4- (3- (4- (trifluoromethyl) phenoxy) piperidin-1-yl) benzoate
(S) -3- (4- (trifluoromethyl) phenoxy) piperidine (200 mg,0.82mm under nitrogen protectionol), methyl 4-iodobenzoate (430 mg,1.64 mmol), pd 2 (dba) 3 (40 mg,0.04 mmol), xantPhos (35 mg,0.06 mmol) and Cs 2 CO 3 (530 mg,1.62 mmol) was dissolved in 1, 4-dioxane (4 mL) and the reaction was heated in an oil bath at 100deg.C and stirred for 22h. The reaction solution was cooled to room temperature and saturated NaHCO was added 3 The solution (30 mL), the aqueous phase was extracted with DCM (40 mL. Times.3), the organic phases combined and washed with saturated NaCl solution (20 mL), anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by column chromatography on silica gel (eluent: PE/DCM/EtOAc (v/v/v) =10/1/1) to give a yellow solid (200 mg, 65%).
MS(ESI,pos.ion)m/z=380.1[M+H] + .
Step four: synthesis of (S) -4- (3- (4- (trifluoromethyl) phenoxy) piperidin-1-yl) benzoic acid
LiOH.H 2 O (122 mg,2.91 mmol) was added to a solution of methyl (S) -4- (3- (4- (trifluoromethyl) phenoxy) piperidin-1-yl) benzoate (200 mg,0.53 mmol) in THF/MeOH (3 mL/3 mL) and the reaction was heated in an oil bath at 60℃under stirring for 8h. The reaction mixture was cooled to room temperature, diluted with DCM (60 mL) and water (20 mL), HCl solution (1 mol/L) was added dropwise to adjust the pH of the system to about 3, the mixture was separated, the aqueous phase was extracted with DCM (20 mL. Times.2), the organic phases were combined and dried over anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by column chromatography on silica gel (eluent: DCM/EtOAc (v/v) =5/1) to give a pale yellow solid (190 mg, 98%).
MS(ESI,pos.ion)m/z=366.1[M+H] + .
Step five: synthesis of N- ((R) -1- (4- (ethylsulfonyl) phenyl) -2-hydroxyethyl) -4- ((S) -3- (4- (trifluoromethyl) phenoxy) piperidin-1-yl) benzamide
(S) -4- (3- (4- (trifluoromethyl) phenoxy) piperidin-1-yl) benzoic acid (190 mg,0.52 mmol), (R) -2-amino-2- (4- (ethylsulfonyl) phenyl) ethanol (145 mg,0.63 mmol), EDCI (200 mg,1.04 mmol) and HOBT (140 mg,1.04 mmol) were dissolved in DCM (10 mL) and TEA (0.22 mL,1.60 mmol) was added and stirred at room temperature for 12h. The reaction was diluted with DCM (60 mL) and successively saturated NaHCO 3 Solution (20 mL) and NaCl solution (20 mL) washing, anhydrous Na 2 SO 4 Drying, concentrating under reduced pressure, and collecting crude product from silica gelColumn chromatography (eluent: DCM/EtOAc (v/v) =1/1) afforded a pale yellow powder (240 mg, 80%).
MS(ESI,pos.ion)m/z=577.1[M+H] + .
1 H NMR(400MHz,DMSO-d 6 )δ(ppm):8.49(d,J=8.0Hz,1H),7.83(d,J=8.2Hz,2H),7.78(d,J=8.7Hz,2H),7.64(d,J=6.2Hz,4H),7.17(d,J=8.8Hz,2H),6.97(d,J=8.9Hz,2H),5.15–5.07(m,1H),5.02(t,J=5.8Hz,1H),4.69–4.61(m,1H),3.83–3.76(m,1H),3.75–3.65(m,2H),3.41-3.61(m,1H),3.47-3.43(m,1H),3.40(q,J=7.4Hz,2H),3.26–3.22(m,1H),2.15–2.05(m,1H),1.89–1.80(m,1H),1.68(dd,J=9.3,7.3Hz,2H),1.10(t,J=7.3Hz,3H).
Example 11N- ((R) -1- (4- (ethylsulfonyl) phenyl) -2-hydroxyethyl) -4- ((2S, 5S) -2- ((difluoromethoxy) methyl) -5- (4- (trifluoromethyl) phenoxy) piperidin-1-yl) benzamide
Step one: synthesis of (2S, 5S) -1-tert-butyl 2-methyl 5- ((tert-butyldimethylsilyl) oxy) piperidine-1, 2-dicarboxylic acid ester
TBSOTf (6.00 mL,26.00 mmol) was slowly added dropwise to (2S, 5S) -1-tert-butyl 2-methyl-5-hydroxypiperidine-1, 2-dicarboxylic acid ester (4.68 g,18.06 mmol) and TEA (5.00 mL,35.97 mmol) in DCM (40 mL) under ice and stirred at room temperature for 12h. Saturated NaHCO is added into the reaction solution 3 The reaction was quenched with solution (50 mL), the aqueous phase extracted with DCM (50 mL. Times.3), the organic phases combined and dried over anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by column chromatography on silica gel (eluent: PE/DCM/EtOAc (v/v/v) =20/1/1) to give a yellow oily liquid (5.07 g, 75%).
MS(ESI,pos.ion)m/z=396.0[M+Na] + .
Step two: synthesis of tert-butyl (2S, 5S) - (5- ((tert-butyldimethylsilyl) oxy) -2- (hydroxymethyl) piperidin-1-yl) carboxylate
LiBH was added at room temperature 4 Is added slowly (2)S, 5S) -1-tert-butyl 2-methyl 5- ((tert-butyldimethylsilyl) oxy) piperidine-1, 2-dicarboxylic acid ester (5.07 g,13.60 mmol) in THF (10 mL) was stirred at room temperature for 5h. Saturated NaHCO is added into the reaction solution 3 The reaction was quenched with solution (40 mL), the aqueous phase extracted with DCM (40 mL. Times.3), the organic phases combined and dried over anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by silica gel column chromatography (eluent: DCM/EtOAc (v/v) =10/1) to give a yellow oil (3.09 g, 66%).
MS(ESI,pos.ion)m/z=368.2[M+Na] + .
Step three: synthesis of (6S, 8S) -6- ((tert-butyldimethylsilyl) oxy) tetrahydro-1H-oxazol [3,4-a ] pyridin-3 (5H) -one
t-BuOK (116 mg,1.03 mmol) was added to a solution of tert-butyl (2S, 5S) -5- ((tert-butyldimethylsilyl) oxy) -2- (hydroxymethyl) piperidine-1-carboxylate (290 mg,0.84 mmol) in THF (10 mL) at-10℃and stirred at room temperature for 12h. Saturated NaHCO is added into the reaction solution 3 The reaction was quenched with solution (30 mL), the aqueous phase extracted with DCM (30 mL. Times.3), the organic phases combined and washed with saturated NaCl solution (20 mL), anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure to give a pale yellow oily liquid (210 mg, 92%).
MS(ESI,pos.ion)m/z=272.1[M+H] + .
Step four: synthesis of (6S, 8S) -6-hydroxytetrahydro-1H-oxazol [3,4-a ] pyridin-3 (5H) -one
A solution of TBAF in THF (14 mL,14.00mmol,1.0 mol/L) was added to (6S, 8S) -6- ((tert-butyldimethylsilyl) oxy) tetrahydro-1H-oxazol [3,4-a ]]In a solution of pyridin-3 (5H) -one (1.82 g,6.71 mmol) in THF (10 mL) was stirred at room temperature for 5H. The reaction was concentrated under reduced pressure, and the crude product was separated by silica gel column chromatography (eluent: DCM/EtOAc (v/v) =1/1) to give a pale yellow solid (697 mg, 66%). MS (ESI, pos.ion) m/z=158.1 [ m+h ]] + .
Step five: synthesis of (6S, 8S) -6- (4- (trifluoromethyl) phenoxy) tetrahydro-1H-oxazol [3,4-a ] pyridin-3 (5H) -one
NaH (860 mg,21.50mmol, 60%) was added to 1-fluoro-4- (trifluoromethyl) benzene (5.20 mL,41.00 mmol) and (6S, 8S) -6-hydroxytetrahydro-1H-oxazole [3 ] under stirring in an ice bath under nitrogen,4-a]A solution of pyridin-3 (5H) -one (2.12 g,13.50 mmol) in DMF (50 mL) was stirred at room temperature for 2H and heated in an oil bath at 60℃for 15H. The reaction was cooled to room temperature, etOAc (120 mL) was added to the reaction, followed by saturated NaHCO 3 Solution (30 mL) and saturated NH 4 Cl solution (20 mL. Times.5) was washed with anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by column chromatography on silica gel (eluent: DCM/EtOAc (v/v) =1/1) to give a yellow solid (2.62 g, 65%).
MS(ESI,pos.ion)m/z=302.2[M+H] + .
Step six: synthesis of ((2S, 5S) -5- (4- (trifluoromethyl) phenoxy) piperidin-2-yl) methanol
Under ice bath, t-BuOK (4.87 g,43.50 mmol) was added to (6S, 8S) -6- (4- (trifluoromethyl) phenoxy) tetrahydro-1H-oxazol [3,4-a ]]i-PrOH/THF/H of pyridin-3 (5H) -one (2.62 g,8.70 mmol) 2 O (20 mL/10mL/1 mL) was added thereto, and the mixture was stirred at room temperature for 16h. Saturated NaHCO is added into the reaction solution 3 The reaction was quenched with solution (50 mL), the aqueous phase extracted with DCM (50 mL. Times.3), the organic phases combined and washed with saturated NaCl solution (30 mL), anhydrous Na 2 SO 4 Drying and concentration under reduced pressure gave a yellow oil (2.30 g, 96%).
MS(ESI,pos.ion)m/z=276.0[M+H] + .
Step seven: synthesis of tert-butyl (2S, 5S) - (2- (hydroxymethyl) -5- (4- (trifluoromethyl) phenoxy) piperidin-1-yl) carboxylate
Boc is to be Boc 2 O (0.27 mL,1.20 mmol) was added ((2S, 5S) -5- (4- (trifluoromethyl) phenoxy) piperidin-2-yl) methanol (217 mg,0.79 mmol) and K 2 CO 3 (330 mg,2.39 mmol) THF/H 2 O (10 mL/3 mL) was stirred at room temperature for 12h. Saturated NaHCO is added into the reaction solution 3 The reaction was quenched with solution (40 mL), the aqueous phase extracted with DCM (40 mL. Times.3), the organic phases combined and dried over anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by silica gel column chromatography (eluent: DCM/EtOAc (v/v) =5/1) to give a white solid (236 mg, 80%). MS (ESI, pos.ion) m/z=398.2 [ m+na] + .
Step eight: synthesis of tert-butyl (2S, 5S) - (2- ((difluoromethoxy) methyl) -5- (4- (trifluoromethyl) phenoxy) piperidin-1-yl) carboxylate
KOAc (250 mg,2.55 mmol) was added to tert-butyl (2S, 5S) - (2- (hydroxymethyl) -5- (4- (trifluoromethyl) phenoxy) piperidin-1-yl) carboxylate (236 mg,0.63 mmol) and TMSCF 2 Br (0.50 mL,3.20 mmol) DCM/H 2 O (2 mL/2 mL) was stirred at room temperature for 12h. Additional TMSCF 2 Br (0.50 mL,3.20 mmol) and stirring was continued for 6h. DCM (80 mL) was added to the reaction with saturated NaHCO 3 Washing with solution (20 mL. Times.2), anhydrous Na was used as the organic phase 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by column chromatography on silica gel (eluent: PE/DCM/EtOAc (v/v/v) =10/1/1) to give a pale yellow solid (197mg, 74%).
MS(ESI,pos.ion)m/z=448.1[M+Na] + .
Step nine: synthesis of (2S, 5S) -2- ((difluoromethoxy) methyl) -5- (4- (trifluoromethyl) phenoxy) piperidine
(2S, 5S) -2- ((difluoromethoxy) methyl) -5- (4- (trifluoromethyl) phenoxy) piperidine-1-carboxylic acid tert-butyl ester (197mg, 0.46 mmol) was dissolved in HCl in MeOH (10 mL, 20%) and stirred at room temperature for 12h. The reaction solution was added with saturated Na 2 CO 3 The aqueous phase was extracted with DCM (30 mL. Times.3) in solution (40 mL), the organic phases combined and taken up in anhydrous Na 2 SO 4 Drying and concentration under reduced pressure gave a pale yellow oil (150 mg, 100%).
MS(ESI,pos.ion)m/z=326.2[M+H] + .
Step ten: synthesis of methyl 4- ((2S, 5S) -2- ((difluoromethoxy) methyl) -5- (4- (trifluoromethyl) phenoxy) piperidin-1-yl) benzoate
(2S, 5S) -2- ((difluoromethoxy) methyl) -5- (4- (trifluoromethyl) phenoxy) piperidine (150 mg,0.46 mmol), methyl 4-iodobenzoate (360 mg,1.37 mmol), pd under nitrogen 2 (dba) 3 (21 mg,0.02 mmol), xantPhos (20 mg,0.04 mmol) and Cs 2 CO 3 (323 mg,0.98 mmol) was dissolved in 1, 4-dioxane (5 mL) and the reaction was heated in an oil bath at 100deg.C and stirred for 22h. The reaction solution was cooled to room temperature, and saturated NaHCO was added to the reaction solution 3 The reaction was quenched with solution (50 mL), the aqueous phase extracted with DCM (40 mL. Times.3), the organic phases combined and dried over anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by column chromatography on silica gel (eluent: PE/DCM/EtOAc (v/v/v) =10/1/1) to give a yellow solid (69 mg, 33%).
MS(ESI,pos.ion)m/z=460.2[M+H] + .
Step eleven: synthesis of 4- ((2S, 5S) -2- ((difluoromethoxy) methyl) -5- (4- (trifluoromethyl) phenoxy) piperidin-1-yl) benzoic acid
LiOH.H 2 O (134 mg,3.19 mmol) was added to a solution of methyl 4- ((2S, 5S) -2- ((difluoromethoxy) methyl) -5- (4- (trifluoromethyl) phenoxy) piperidin-1-yl) benzoate (69 mg,0.15 mmol) in THF/MeOH (1 mL/1 mL) and stirred at room temperature for 12h. Adding LiOH H 2 O (1.20 g,28.60 mmol) and stirring was continued for 6h at room temperature. DCM (80 mL) and water (40 mL) were added to the reaction solution, HCl solution (1 mol/L) was added dropwise to adjust the pH of the system to about 3, the solution was separated, the aqueous phase was extracted with DCM (40 mL. Times.2), the organic phases were combined and the mixture was taken up with anhydrous Na 2 SO 4 Drying and concentration under reduced pressure gave a yellow solid (60 mg, 90%).
MS(ESI,pos.ion)m/z=445.9[M+H] + .
Step twelve: synthesis of N- ((R) -1- (4- (ethylsulfonyl) phenyl) -2-hydroxyethyl) -4- ((2S, 5S) -2- ((difluoromethoxy) methyl) -5- (4- (trifluoromethyl) phenoxy) piperidin-1-yl) benzamide
4- ((2S, 5S) -2- ((difluoromethoxy) methyl) -5- (4- (trifluoromethyl) phenoxy) piperidin-1-yl) benzoic acid (60 mg,0.13 mmol), (R) -2-amino-2- (4- (ethylsulfonyl) phenyl) ethanol (54 mg,0.24 mmol), EDCI (104 mg,0.54 mmol) and HOBT (75 mg,0.56 mmol) were dissolved in DCM (6 mL) and TEA (0.12 mL,0.86 mmol) was added and the reaction stirred at room temperature for 12h. DCM (60 mL) was added to the reaction followed by saturated NaHCO 3 Solution (40 mL) and saturated NaCl solution (20 mL), anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by column chromatography on silica gel (eluent: DCM/EtOAc (v/v) =1/1) to give a yellow solid (50 mg, 57%). MS (ESI, pos.ion) m/z=657.1 [ m+h] + .
1 H NMR(400MHz,DMSO-d 6 )δ(ppm):8.51(d,J=7.8Hz,1H),7.81(t,J=8.9Hz,4H),7.65(t,J=7.7Hz,4H),7.20(d,J=8.5Hz,2H),7.01(d,J=8.9Hz,2H),6.67(t,J=76.0Hz,1H),5.12(dd,J=13.8,6.9Hz,1H),5.00(t,J=5.8Hz,1H),4.68–4.57(m,1H),4.32(s,1H),4.05(dd,J=17.4,9.8Hz,1H),3.95(dd,J=10.2,6.2Hz,2H),3.71(qd,J=11.4,7.0Hz,2H),3.26(q,J=7.4Hz,2H),3.12–3.03(m,1H),2.09(d,J=8.8Hz,1H),1.91(dd,J=10.9,9.3Hz,2H),1.78–1.63(m,1H),1.09(t,J=7.3Hz,3H).
Example 12N- ((R) -1- (4- (ethylsulfonyl) phenyl) -2-hydroxyethyl) -4- ((2S, 5R) -2- ((difluoromethoxy) methyl) -5- (4- (trifluoromethyl) phenoxy) piperidin-1-yl) benzamide
The title compound was prepared as a yellow solid (55 mg, 63%) by the method of step one-step twelve of example 11 starting from (2 s,5 r) -1-tert-butyl 2-methyl-5-hydroxypiperidine-1, 2-dicarboxylic acid ester.
MS(ESI,pos.ion)m/z=657.1[M+H] + .
1 H NMR(400MHz,DMSO-d 6 )δ(ppm):8.44(d,J=7.8Hz,1H),7.82(d,J=8.3Hz,2H),7.74(d,J=8.8Hz,2H),7.63(d,J=8.4Hz,4H),7.12(d,J=8.6Hz,2H),6.89(d,J=9.0Hz,2H),6.68(t,J=75.8Hz,1H),5.11(dd,J=13.8,7.1Hz,1H),4.99(t,J=5.8Hz,1H),4.88-4.80(s,1H),4.43-4.29(m,1H),4.09–4.01(m,1H),3.93(dd,J=16.3,6.3Hz,2H),3.76–3.62(m,2H),3.42–3.35(m,1H),3.25(q,J=7.3Hz,2H),2.02(dd,J=14.7,7.7Hz,2H),1.87-1.80(m,1H),1.75-1.62(m,1H),1.08(d,J=7.3Hz,3H).
Example 13N- ((R) -1- (4- (ethylsulfonyl) phenyl) -2-hydroxyethyl) -4- ((2S, 5S) -2- ((trifluoromethoxy) methyl) -5- (4- (trifluoromethyl) phenoxy) piperidin-1-yl) benzamide
Step one: synthesis of tert-butyl (2S, 5S) - (2- ((trifluoromethoxy) methyl) -5- (4- (trifluoromethyl) phenoxy) piperidin-1-yl) carboxylate
Under nitrogen, 2-fluoropyridine (0.60 mL,7.00 mmol) and TMSCF were reacted 3 (1.20 mL,8.12 mmol) was added successively to a solution of tert-butyl (2S, 5S) - (2- (hydroxymethyl) -5- (4- (trifluoromethyl) phenoxy) piperidin-1-yl) carboxylate (800 mg,2.13 mmol), agOTf (2.21 g,8.60 mmol), selectFluor (1.13 g,3.19 mmol) and KF (503 mg,8.67 mmol) in EtOAc (20 mL) and stirred at room temperature for 72h. The reaction was filtered through celite and the filter cake was washed with DCM (50 mL) and the filtrate was saturated with NaHCO 3 Washing with solution (30 mL. Times.2), anhydrous Na was used as the organic phase 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by column chromatography on silica gel (eluent: PE/DCM/EtOAc (v/v/v) =10/1/1) to give a yellow oil (312 mg, 33%).
MS(ESI,pos.ion)m/z=388.1[M+H-56] + .
Step two: synthesis of (2S, 5S) -2- ((trifluoromethoxy) methyl) -5- (4- (trifluoromethyl) phenoxy) piperidine
Tert-butyl (2S, 5S) - (2- ((trifluoromethoxy) methyl) -5- (4- (trifluoromethyl) phenoxy) piperidin-1-yl) carboxylate (312 mg,0.70 mmol) was dissolved in HCl in methanol (10 mL, 20%) and stirred at room temperature for 4h. Adding saturated Na into the reaction solution 2 CO 3 The aqueous phase was extracted with DCM (30 mL. Times.3) in solution (40 mL), the organic phases combined and taken up in anhydrous Na 2 SO 4 Drying and concentration under reduced pressure gave a pale yellow oil (240 mg, 99%). MS (ESI, pos.ion) m/z=344.1 [ m+h ]] + .
Step three: synthesis of methyl 4- ((2S, 5S) -2- ((trifluoromethoxy) methyl) -5- (4- (trifluoromethyl) phenoxy) piperidin-1-yl) benzoate
(2S, 5S) -2- ((trifluoromethoxy) methyl) -5- (4- (trifluoromethyl) phenoxy) piperidine (240 mg,0.70 mmol), methyl 4-iodobenzoate (550 mg,2.10 mmol), pd under nitrogen 2 (dba) 3 (64 mg,0.07 mmol), xantPhos (60 mg,0.10 mmol) and Cs 2 CO 3 (463 mg,1.41 mmol) was dissolved in 1, 4-dioxane (6 mL) and the reaction was heated in an oil bath at 100deg.C and stirred for 70h. The reaction solution was cooled to room temperature, and saturated NaHCO was added to the reaction solution 3 The reaction was quenched with solution (50 mL), the aqueous phase extracted with DCM (40 mL. Times.3), the organic phases combined and dried over anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by column chromatography on silica gel (eluent: PE/DCM/EtOAc (v/v/v) =5/1/1) to give a yellow oily solid (157 mg, 47%).
MS(ESI,pos.ion)m/z=478.2[M+H] + .
Step four: synthesis of 4- ((2S, 5S) -2- ((trifluoromethoxy) methyl) -5- (4- (trifluoromethyl) phenoxy) piperidin-1-yl) benzoic acid
LiOH.H 2 O (1.51 g,36.00 mmol) methyl 4- ((2S, 5S) -2- ((trifluoromethoxy) methyl) -5- (4- (trifluoromethyl) phenoxy) piperidin-1-yl) benzoate (157 mg,0.33 mmol) THF/MeOH/H 2 In a solution of O (3 mL/3mL/1 mL), the mixture was stirred at room temperature for 72h. DCM (80 mL) and water (40 mL) were added to the reaction solution, HCl solution (2 mol/L) was added dropwise to adjust the pH of the system to about 3, the solution was separated, the aqueous phase was extracted with DCM (40 mL. Times.2), the organic phases were combined and the aqueous phase was extracted with anhydrous Na 2 SO 4 Drying and concentration under reduced pressure gave a yellow solid (124 mg, 81%). MS (ESI, pos.ion) m/z=464.2 [ m+h] + .
Step five: synthesis of N- ((R) -1- (4- (ethylsulfonyl) phenyl) -2-hydroxyethyl) -4- ((2S, 5S) -2- ((trifluoromethoxy) methyl) -5- (4- (trifluoromethyl) phenoxy) piperidin-1-yl) benzamide
4- ((2S, 5S) -2- ((trifluoromethoxy) methyl) -5- (4- (trifluoromethyl) phenoxy) piperidin-1-yl) benzoic acid (124 mg,0.27 mmol), (R) -2-amino-2- (4- (ethylsulfonyl) phenyl) ethanol (75 mg,0.33 mmol), EDCI (106 mg,0.55 mmol) and HOBT (76 mg,0.56 mmol) were dissolved in DCM (6 mL) and TEA (0.16 mL,1.10 mmol) was added and stirred at room temperature for 12h. DCM (80 mL) was added to the reaction followed by saturated NaHCO 3 Solution (30 mL) and saturated NaCl solution (20 mL), anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by column chromatography on silica gel (eluent: DCM/EtOAc (v/v) =2/1) to give a yellow solid (68 mg, 38%).
MS(ESI,pos.ion)m/z=675.1[M+H] + .
1 H NMR(400MHz,DMSO-d 6 )δ(ppm):8.54(d,J=7.9Hz,1H),7.81(t,J=8.6Hz,4H),7.65(t,J=8.3Hz,4H),7.20(d,J=8.5Hz,2H),7.03(d,J=8.8Hz,2H),5.12(dd,J=13.7,6.6Hz,1H),5.02(t,J=5.8Hz,1H),4.63(ddd,J=11.2,8.2,3.8Hz,1H),4.43(dd,J=18.1,7.9Hz,1H),4.36–4.28(m,1H),4.24(dd,J=9.9,6.8Hz,1H),3.97(dd,J=13.1,4.9Hz,1H),3.78–3.63(m,2H),3.26(q,J=7.3Hz,2H),3.16–3.06(m,1H),2.13–2.04(m,1H),2.00-1.88(m,2H),1.78–1.66(m,1H),1.09(t,J=7.3Hz,3H).
Example 14N- ((R) -1- (4- (ethylsulfonyl) phenyl) -2-hydroxyethyl) -4- (1- (4- (trifluoromethyl) benzyl) piperidin-2-yl) benzamide
Step one: synthesis of methyl 4- (5- ((tert-butoxycarbonyl) amino) pentanoyl) benzoate
Methyl 4-iodobenzoate (4.60 g,18.00 mmol) was dissolved in THF (20 mL), cooled to-50deg.C, i-PrMgBr (17 mL,17.00 mmol) was added, stirred for 1h at-50deg.C, tert-butyl 2-oxopiperidine-1-carboxylate (2.90 g,15.00 mmol) was added and the temperature was slowly restored to room temperature. The reaction was carried out for 3h, diluted hydrochloric acid was slowly added to pH acidity, extracted with EtOAc (25 mL. Times.2), the organic phases combined, anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by column chromatography on silica gel (eluent: PE/EtOAc (v/v) =10/1) to give a colorless liquid (2.75 g, 56%). MS (ESI, pos.ion) m/z 358[ M+Na ]] + .
Step two: synthesis of methyl 4- (3, 4,5, 6-tetrahydropyridin-2-yl) benzoate
Methyl 4- (5- ((tert-butoxycarbonyl) amino) pentanoyl) benzoate (2.57 g,7.66 mmol) was dissolved in DCM (10 mL) and TFA (2.85 mL,38.40 mmol) was added and stirred at room temperature for 1h. The reaction was diluted with DCM (30 mL) and washed successively with saturated NaHCO 3 Solution (15 mL) and saturated NaCl solution (15 mL), anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by column chromatography on silica gel (eluent: PE/EtOAc (v/v) =10/1) to give a white solid (1.63 g, 98%).
MS(ESI,pos.ion)m/z:218[M+H] + .
Step three: synthesis of methyl 4- (piperidin-2-yl) benzoate
4- (3, 4,methyl 5, 6-tetrahydropyridin-2-yl benzoate (2.23 g,11.00 mmol) was dissolved in THF (12 mL) and MeOH (5 mL), and NaBH was slowly added at room temperature 4 (0.11 g,2.90 mmol). The reaction was allowed to react for 2h, quenched with water, extracted with EtOAc (15 mL. Times.2), the organic phases combined, anhydrous Na 2 SO 4 Drying and concentration under reduced pressure gave a colorless liquid (2.25 g, 99%).
MS(ESI,pos.ion)m/z:220[M+H] + .
Step four: synthesis of tert-butyl (2- (4- (methoxycarbonyl) phenyl) piperidin-1-yl) carboxylate
Methyl 4- (piperidin-2-yl) benzoate (1.69 g,7.71 mmol) was dissolved in DCM (5 mL) to which TEA (2 mL,15.00 mmol) was added followed by Boc 2 O (1.70 g,7.71 mmol) was stirred at room temperature for 5h. The reaction mixture was diluted with DCM (30 mL) and successively saturated NH 4 Cl solution (15 mL) and saturated NaCl solution (15 mL), anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by column chromatography on silica gel (eluent: PE/EtOAc (v/v) =5/1) to give a colorless liquid (1.98 g, 80.4%).
MS(ESI,pos.ion)m/z:320[M+H] + .
Step five: synthesis of 4- (1- (tert-butoxycarbonyl) piperidin-2-yl) benzoic acid
Tert-butyl (2- (4- (methoxycarbonyl) phenyl) piperidin-1-yl) carboxylate (1.98 g,6.20 mmol) was dissolved in THF (10 mL) and MeOH (10 mL), to which LiOH (1.48 g,61.80 mmol) H was added 2 O (2 mL) solution, stirring at room temperature for 12h, adding HCl solution (2 mol/L) to adjust pH to about 5, extracting with EtOAc (25 mL. Times.2), mixing organic phases, washing with saturated NaCl (20 mL) solution, and anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by silica gel column chromatography (eluent: DCM/MeOH (v/v) =20/1) to give a white solid (1.66 g, 88%).
MS(ESI,pos.ion)m/z:306[M+H] + .
Step six: synthesis of tert-butyl (2- (4- (((R) -1- (4- (ethylsulfonyl) phenyl) -2-hydroxyethyl) amido) phenyl) piperidin-1-yl) carboxylate
4- (1- (tert-Butoxycarbonyl) piperidin-2-yl) benzoic acid (120 mg,0.39 mmol), (R) -2-amino-2- (4-(ethylsulfonyl) phenyl) ethanol (112 mg,0.49 mmol) and HATU (181 mg,0.47 mmol) were dissolved in DCM (12 mL) and stirred at room temperature for 24h after the addition of TEA (0.12 mL,0.92 mmol). The reaction was diluted with DCM (30 mL) and washed successively with saturated NaHCO 3 Solution (15 ml) and saturated NaCl solution (15 ml) washing, anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by column chromatography on silica gel (eluent: DCM/EtOAc (v/v) =1/1) to give a white solid (120 mg, 59%).
MS(ESI,pos.ion)m/z:539[M+H] + .
Step seven: synthesis of N- ((R) -1- (4- (ethylsulfonyl) phenyl) -2-hydroxyethyl) -4- (piperidin-2-yl) benzamide
Tert-butyl (2- (4- (((R) -1- (4- (ethylsulfonyl) phenyl) -2-hydroxyethyl) amido) phenyl) piperidin-1-yl) carboxylate (120 mg,0.23 mmol) was dissolved in DCM (10 mL) after which TFA (0.24 mL,3.20 mmol) was slowly added and stirred at room temperature for 3h. The reaction was diluted with DCM (30 mL) and washed successively with saturated NaHCO 3 Solution (15 ml) and saturated NaCl solution (15 ml) washing, anhydrous Na 2 SO 4 Drying and concentration under reduced pressure gave a colorless liquid (94 mg, 97%).
MS(ESI,pos.ion)m/z:417[M+H] + .
Step eight: synthesis of N- ((R) -1- (4- (ethylsulfonyl) phenyl) -2-hydroxyethyl) -4- (1- (4- (trifluoromethyl) benzyl) piperidin-2-yl) benzamide
N- ((R) -1- (4- (ethylsulfonyl) phenyl) -2-hydroxyethyl) -4- (piperidin-2-yl) benzamide (97 mg,0.23 mmol) was dissolved in MeOH (10 mL) and 4-trifluorobenzaldehyde (48 mg,0.28 mmol) was added before NaBH was added 3 CN (15 mg,0.24 mmol), was stirred at room temperature for 12h. The reaction was diluted with DCM (30 mL) and washed successively with saturated NaHCO 3 Solution (15 ml) and saturated NaCl solution (15 ml) washing, anhydrous Na 2 SO 4 Drying and concentrating under reduced pressure gave a white solid (22 mg, 16%).
MS(ESI,pos.ion)m/z:575[M+H] + .
1 H NMR(600MHz,CDCl 3 )δ(ppm):7.87(dd,J=8.4,2.0Hz,2H),7.82(d,J=8.3Hz,2H),7.61–7.51(m,6H),7.40(d,J=7.6Hz,2H),7.16(d,J=6.7Hz,1H),5.32–5.28(m,1H),4.06(dt,J=11.2,3.6Hz,1H),3.98(dd,J=11.3,4.7Hz,1H),3.72(d,J=14.2Hz,1H),3.24(d,J=9.1Hz,1H),3.10(q,J=7.4Hz,2H),2.95(d,J=13.9Hz,2H),2.01(t,J=11.3Hz,1H),1.82(dd,J=26.3,12.9Hz,3H),1.69–1.57(m,3H),1.29(d,J=7.1Hz,3H).
Example 15N- ((R) -1- (4- (ethylsulfonyl) phenyl) -2-hydroxyethyl) -4- ((2S) -2- ((difluoromethoxy) methyl) -5- ((4- (trifluoromethyl) phenyl) (methyl) amino) piperidin-1-yl) benzamide
Step one: synthesis of (2S) -1-tert-butyl-2-methyl-5- ((4- (trifluoromethyl) phenyl) amino) piperidine-1, 2-dicarboxylic acid ester
4- (trifluoromethyl) aniline (1.03 g,6.39 mmol), (S) -1-tert-butyl 2-methyl 5-oxopiperidine-1, 2-dicarboxylic acid ester (1.83 g,7.11 mmol) and AcOH (0.70 mL,12 mmol) were dissolved in toluene (20 mL), reacted in an oil bath at 80℃with heating and stirring for 5h, and concentrated under reduced pressure to give a concentrated solution. NaBH is carried out 3 CN (4.0 g,64 mmol) was added to a solution of AcOH (0.40 mL,7.0 mmol) and the concentrate above in EtOH (20 mL) and stirred at room temperature for 3h. Then the reaction solution was added with saturated Na 2 CO 3 The aqueous phase was extracted with DCM (50 mL. Times.3) in solution (50 mL), the organic phases combined and washed with saturated NaCl solution (30 mL), anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by column chromatography on silica gel (eluent: PE/DCM/EtOAc (v/v/v) =5/1/1) to give a yellow oil (1.55 g, 62%).
MS(ESI,pos.ion)m/z=403.1[M+H] + .
Step two: (2S, 5R) - (2- (hydroxymethyl) -5- ((4- (trifluoromethyl) phenyl) amino) piperidin-1-yl) carboxylic acid tert-butyl ester
LiBH was added at room temperature 4 To a suspension of (2S) -1-tert-butyl-2-methyl-5- ((4- (trifluoromethyl) phenyl) amino) piperidine-1, 2-dicarboxylic acid ester (1.0 g,2.50 mmol) in THF (4 mL) was added dropwise (7.5 mL,7.50mmol, 1.0M) and stirred for 5h. Follow by TLC, complete conversion of starting material. The reaction solution is added with saturated NaHCO 3 In solution (40 mL) and DCM was then used30ml×3) extraction, combining the organic phases and using anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by silica gel column chromatography (eluent: DCM/EtOAc (v/v) =5/1) to give a yellow solid (254 mg, 32%).
MS(ESI,pos.ion)m/z=319.2[M-56+H] + .
Step three: synthesis of tert-butyl (2S, 5R) - (2- (hydroxymethyl) -5- (methyl- (4- (trifluoromethyl) phenyl) amino) piperidin-1-yl) carboxylate
Tert-butyl (2S, 5R) - (2- (hydroxymethyl) -5- ((4- (trifluoromethyl) phenyl) amino) piperidin-1-yl) carboxylate (222 mg,0.59 mmol), formaldehyde (120 mg,4.00 mmol) and AcOH (0.10 mL,1.70 mmol) were dissolved in toluene (8 mL) and reacted in an oil bath at 80℃with heating and stirring for 6h. Concentrating under reduced pressure to obtain crude product. NaBH is carried out 3 CN (360 mg,5.73 mmol) was added to a solution of the above crude product and AcOH (0.10 mL,1.70 mmol) in EtOH (5 mL) and stirred at room temperature for 9h. Saturated Na was added to the reaction solution 2 CO 3 The reaction was quenched with solution (40 mL) and the aqueous phase was extracted with DCM (40 mL. Times.3), the organic phases combined and taken up in anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by column chromatography on silica gel (eluent: DCM/EtOAc (v/v) =5/1) to give a yellow oil (66 mg, 32%).
MS(ESI,pos.ion)m/z=389.1[M+H] + .
Step four: synthesis of tert-butyl (2S, 5R) -2- ((difluoromethoxy) methyl) -5- (methyl- (4- (trifluoromethyl) phenyl) amino) piperidin-1-yl) carboxylate
KOAc (70 mg,0.71 mmol) was added to tert-butyl (2S, 5R) - (2- (hydroxymethyl) -5- (methyl- (4- (trifluoromethyl) phenyl) amino) piperidin-1-yl) carboxylate (66 mg,0.17 mmol) and TMSCF 2 Br (0.30 mL,1.90 mmol) DCM/H 2 O (1 mL/1 mL) was stirred at room temperature for 22h. DCM (80 mL) was added to the reaction followed by saturated NaHCO 3 The solution (20 mL. Times.2) was washed and the organic phase was washed with anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by column chromatography on silica gel (eluent: PE/DCM/EtOAc (v/v/v) =5/1/1) to give a yellow oily liquid (30 mg, 40%).
MS(ESI,pos.ion)m/z=439.1[M+H] + .
Step five: synthesis of (3R, 6S) -6- ((difluoromethoxy) methyl) -N-methyl-N- (4- (trifluoromethyl) phenyl) piperidin-3-amine
Tert-butyl (2S, 5R) -2- ((difluoromethoxy) methyl) -5- (methyl- (4- (trifluoromethyl) phenyl) amino) piperidin-1-yl) carboxylate (30 mg,0.068 mmol) was dissolved in HCl in MeOH (5 mL, 30%) and stirred at room temperature for 3h. The starting material was essentially converted as tracked by TLC. Saturated Na was added to the reaction solution 2 CO 3 (aq.) (20 mL) then the aqueous phase was extracted with DCM (20 mL. Times.3), the organic phases combined and dried over anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure to give a pale yellow oily solid (23 mg, 99%).
Step six: synthesis of methyl 4- ((2S, 5R) -2- ((difluoromethoxy) methyl) -5- (methyl- (4- (trifluoromethyl) phenyl) amino) piperidin-1-yl) benzoate
(3R, 6S) -6- ((difluoromethoxy) methyl) -N-methyl-N- (4- (trifluoromethyl) phenyl) piperidin-3-amine (23 mg,0.068 mmol), methyl 4-iodobenzoate (60 mg,0.23 mmol), pd under nitrogen 2 (dba) 3 (12 mg,0.013 mmol), xantPhos (10 mg,0.017 mmol) and Cs 2 CO 3 (100 mg,0.30 mmol) was dissolved in 1, 4-dioxane (3 mL) and the reaction was stirred with heating in an oil bath at 100deg.C for 24h. Saturated NaHCO is added into the reaction solution 3 (aq.) (30 mL) quench the reaction, then extract the aqueous phase with DCM (30 mL. Times.3), combine the organic phases and use anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by column chromatography on silica gel (eluent: PE/DCM/EtOAc (v/v/v) =10/1/1) to give the product as a yellow solid (15 mg, 47%).
MS(ESI,pos.ion)m/z=473.2[M+H] + .
Step seven: synthesis of 4- ((2S, 5R) -2- ((difluoromethoxy) methyl) -5- (methyl- (4- (trifluoromethyl) phenyl) amino) piperidin-1-yl) benzoic acid
LiOH.H 2 O (104 mg,2.48 mmol) methyl 4- ((2S, 5R) -2- ((difluoromethoxy) methyl) -5- (methyl- (4- (trifluoromethyl) phenyl) amino) piperidin-1-yl) benzoate (15 mg,0.032 mmol) THF/MeOH/H 2 O (1 mL/1mL/0.2 mL) was stirred at room temperature for 36h. Adding LiOH H 2 O (500 mg,11.92 mmol), stirring was continued at room temperature for 5h with a small amount of raw material remainingThe balance of the preparation method. The reaction was diluted with DCM (60 mL) and water (20 mL), then 1.0M HCl solution was added dropwise to the system to adjust the ph=2-3, the solution was separated, the aqueous phase was extracted with DCM (20 ml×2), the organic phases were combined and the anhydrous Na was used 2 SO 4 Drying and concentration under reduced pressure gave the crude product as a pale yellow solid (14 mg, 96%).
MS(ESI,pos.ion)m/z=459.1[M+H] + .
Step eight: synthesis of N- ((R) -1- (4- (ethylsulfonyl) phenyl) -2-hydroxyethyl) -4- ((2S, 5R) -2- ((difluoromethoxy) methyl) -5- (methyl- (4- (trifluoromethyl) phenyl) amino) piperidin-1-yl) benzamide
4- ((2S, 5R) -2- ((difluoromethoxy) methyl) -5- (methyl- (4- (trifluoromethyl) phenyl) amino) piperidin-1-yl) benzoic acid (14 mg,0.030 mmol), (R) -2-amino-2- (4- (ethylsulfonyl) phenyl) ethanol (23 mg,0.10 mmol), EDCI (34 mg,0.18 mmol) and HOBT (23 mg,0.17 mmol) were dissolved in DCM (4 mL) and TEA (0.10 mL,0.72 mmol) was added and the reaction stirred at room temperature. The reaction was followed by TLC. The reaction was diluted with DCM (60 mL) and then saturated NaHCO 3 Solution (20 mL) and NaCl solution (20 mL) washing, anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was chromatographed on silica gel (eluent: DCM/EtOAc (v/v) =1/1) to give a yellow solid (6 mg, 29%).
MS(ESI,pos.ion)m/z=669.9[M+H] + .
Example 16N- ((R) -1- (4- (ethylsulfonyl) phenyl) -2-hydroxyethyl) -4- ((S) -4- (4- (difluoromethoxy) benzyl) -2- ((difluoromethoxy) methyl) piperazin-1-yl) benzamide
Step one: synthesis of (S) -4-benzyl 2- (hydroxymethyl) piperazine-1, 4-dicarboxylic acid-1-tert-butyl ester
(S) -4-benzyl 1-tert-butyl 2-methylpiperazine-1, 2, 4-tricarboxylic acid ester (5.1 g,13.00 mmol) was dissolved in THF (15 mL), and LiBH was slowly added in portions under ice-bath 4 (700 mg,32.14 mmol) was stirred at room temperature for 24h. Saturated NaHCO is added into the reaction solution 3 The solution (100 mL),the reaction was quenched. Concentrated under reduced pressure, THF was removed, extracted with DCM (120 mL. Times.2), and anhydrous Na was added 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by silica gel column chromatography (eluent: PE/EtOAc (v/v) =1/1) to give a colorless oil (4.6 g, 97%).
MS(ESI,pos.ion)m/z=373.2[M+Na] + .
Step two: synthesis of (S) -4-benzyl 2-tert-butyl ((difluoromethoxy) methyl) piperazine-1, 4-dicarboxylic acid ester
(S) -4-benzyl 2- (hydroxymethyl) piperazine-1, 4-dicarboxylic acid 1-tert-butyl ester (4.6 g,13.00 mmol) was dissolved in DCM/H 2 O (18 mL/18 mL) and KOAc (7.70 g,78.00 mmol) were added, and (bromo (difluoro) methyl) -trimethyl-silane (8.2 mL,53.00 mmol) was slowly added dropwise under ice-bath and stirred at room temperature for 18h. Saturated NaHCO is added into the reaction solution 3 The reaction was quenched with solution (60 mL), extracted with DCM (80 mL. Times.2), and dried over Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by column chromatography over silica gel (eluent: PE/EtOAc (v/v) =6/1) to give a colorless oil (3.59 g, 68%). MS (ESI, pos.ion) m/z 423.3[ M+Na ]] + .
Step three: synthesis of benzyl (S) -3- ((difluoromethoxy) methyl) piperazine-1-carboxylate
(S) -4-benzyl 2-tert-butyl ((difluoromethoxy) methyl) piperazine-1, 4-dicarboxylate (2.10 g,5.20 mmol) was dissolved in EtOAc (5 mL), HCl EtOAc (6 mL,18mmol,3 mol/L) was added to ice bath and stirred at room temperature for 24h. Removing part of solvent by distillation under reduced pressure, adding saturated NaHCO 3 (50 mL), pH of the system was adjusted to about 8, DCM (80 mL. Times.2) was used for extraction, and the organic layer was dried over Na 2 SO 4 Drying and concentration under reduced pressure gave a yellow oil (1.45 g, 92%).
MS(ESI,pos.ion)m/z=301.2[M+H] + .
Step four: synthesis of benzyl (S) -3- ((difluoromethoxy) methyl) -4- (4- (methoxycarbonyl) phenyl) piperazine-1-carboxylate
Benzyl (S) -3- ((difluoromethoxy) methyl) piperazine-1-carboxylate (550 mg,1.83 mmol), methyl 4-iodobenzoate (665 mg,2.54 mmol), pd under nitrogen 2 (dba) 3 (158mg,0.17mmol),Ruphos(197mg,0.42mmol),Cs 2 CO 3 (1.06 g,3.25 mmol) was dissolved in toluene (10 mL) and heated at 110deg.C for 19h. The crude product was isolated by column chromatography over silica gel (eluent: PE/EtOAc (v/v) =3/1) to give a yellow oil (778 mg, 98%).
MS(ESI,pos.ion)m/z=435.1[M+H] + .
Step five: synthesis of methyl (S) -4- (2- ((difluoromethoxy) methyl) piperazin-1-yl) benzoate
Benzyl (S) -3- ((difluoromethoxy) methyl) -4- (4- (methoxycarbonyl) phenyl) piperazine-1-carboxylate (778 mg,1.79 mmol), pd/C (1.06 g,0.99mmol,10 mass%) was dissolved in MeOH/THF (5 mL/5 mL) under hydrogen atmosphere and stirred at room temperature for 12h. The mixture was filtered through celite and concentrated under reduced pressure to give a yellow oil (45 mg, 85%).
MS(ESI,pos.ion)m/z=301.2[M+H] + .
Step six: synthesis of methyl (S) -4- (4- (4- (difluoromethoxy) benzyl) -2- ((difluoromethoxy) methyl) piperazin-1-yl) benzoate
Methyl (S) -4- (2- ((difluoromethoxy) methyl) piperazin-1-yl) benzoate (53 mg,1.78 mmol), 4- (difluoromethoxy) benzaldehyde (0.47 mL,3.60 mmol) were dissolved in EtOH/THF (6 mL/6 mL) and AcOH (0.5 mL,9.00 mmol), STAB (1.9 g,9.00 mmol) was added and reacted at 60℃for 14h. Saturated NaHCO is added into the reaction solution 3 Solution (50 mL), pH of the system was adjusted to about 8, etOAc (80 mL. Times.2) was used for extraction, and anhydrous Na was used as the organic layer 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by column chromatography over silica gel (eluent: PE/EtOAc (v/v) =3/1) to give a yellow oil (612 mg, 76%).
MS(ESI,pos.ion)m/z=457.1[M+H] + .
Step seven: synthesis of (S) -4- (4- (4- (difluoromethoxy) benzyl) -2- ((difluoromethoxy) methyl) piperazin-1-yl) benzoic acid
Methyl (S) -4- (4- (4- (difluoromethoxy) benzyl) -2- ((difluoromethoxy) methyl) piperazin-1-yl) benzoate (612 mg,1.34 mmol), liOH. H 2 O (610 mg,14.54 mmol) was dissolved in THF/MeOH/H2O (6 mL/3mL/3 mL) and reacted at room temperature for 17H. 1.0M HCl solution (8 mL) was added to the reaction mixture, the pH of the system was adjusted to about 4, and EtOAc (20 mL. Times.2) was used for extractionTaking out the anhydrous Na of the organic layer 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by column chromatography on silica gel (eluent: PE/EtOAc (v/v) =3/1) to give a white solid (160 mg, 27%).
MS(ESI,pos.ion)m/z=443.0[M+H] + .
Step eight: synthesis of N- ((R) -1- (4- (ethylsulfonyl) phenyl) -2-hydroxyethyl) -4- ((S) -4- (4- (difluoromethoxy) benzyl) -2- ((difluoromethoxy) methyl) piperazin-1-yl) benzamide
(S) -4- (4- (4- (difluoromethoxy) benzyl) -2- ((difluoromethoxy) methyl) piperazin-1-yl) benzoic acid (160 mg,0.36 mmol), (2R) -2-amino-2- (4- (ethylsulfonyl) phenyl) ethanolic acid (126 mg,0.47 mmol), EDCI (236 mg,1.23 mmol), HOBT (158 mg,1.17 mmol) were dissolved in DCM (5 mL) and DIPEA (0.5 mL,3.00 mmol) was added and stirred at room temperature for 24h. DCM (50 mL) was added to the reaction solution, saturated NaHCO 3 (20 mL. Times.2) washing, saturated NaCl (20 mL), anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by silica gel column chromatography (eluent: DCM/MeOH (v/v) =10/1) to give a white solid (204 mg, 86%).
MS(ESI,pos.ion)m/z=654.1[M+H] + .
1 H NMR(400MHz,DMSO-d 6 )δ(ppm):8.51(d,J=7.2Hz,1H),7.82(t,J=8.7Hz,4H),7.65(d,J=7.7Hz,2H),7.40(d,J=7.4Hz,2H),7.22(s,1H),7.15(d,J=7.8Hz,2H),6.95(d,J=8.1Hz,2H),6.61(t,J=75.8Hz,1H),5.13(d,J=6.3Hz,1H),5.02(t,J=4.8Hz,1H),4.21(dd,J=16.9,7.7Hz,2H),3.75(s,3H),3.59(d,J=12.7Hz,2H),3.48(d,J=13.3Hz,1H),3.26(dd,J=14.0,6.8Hz,2H),3.06(t,J=10.7Hz,1H),2.92(dd,J=19.2,11.3Hz,2H),2.25–2.12(m,2H),1.10(t,J=7.0Hz,3H).
Example 17N- (4- (ethylsulfonyl) benzyl) - (S) -4- (2- ((difluoromethoxy) methyl) -4- (4- (trifluoromethyl) benzyl) piperazin-1-yl) benzamide
Step one: synthesis of methyl (S) -4- (2- ((difluoromethoxy) methyl) -4- (4- (trifluoromethyl) benzyl) piperazin-1-yl) benzoate
Methyl (S) -4- (2- ((difluoromethoxy) methyl) piperazin-1-yl) benzoate (203 mg,0.68 mmol), 4- (trifluoromethyl) benzaldehyde (0.12 mL,0.88 mmol) was dissolved in EtOH/THF (3 mL/3 mL) and AcOH (0.08 mL,1.00 mmol), STAB (299 mg,1.41 mmol) was added and reacted at room temperature for 18h. STAB (2 eq 300 mg) was added and reacted at 60℃for 15h. Saturated NaHCO is added into the reaction solution 3 Solution (30 mL), pH of the system was adjusted to about 8, etOAc (50 mL. Times.2) was used for extraction, and anhydrous Na was used as the organic layer 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by column chromatography over silica gel (eluent: PE/EtOAc (v/v) =3/1) to give a yellow oil (133 mg, 43%).
MS(ESI,pos.ion)m/z=459.3[M+H] + .
Step two: synthesis of (S) -4- (2- ((difluoromethoxy) methyl) -4- (4- (trifluoromethyl) benzyl) piperazin-1-yl) benzoic acid
Methyl (S) -4- (2- ((difluoromethoxy) methyl) -4- (4- (trifluoromethyl) benzyl) piperazin-1-yl) benzoate (133 mg,0.29 mmol), liOH. H 2 O (162 mg,3.86 mmol) was dissolved in THF/MeOH/H2O (2 mL/1mL/0.7 mL) and reacted at room temperature for 22H. 1.0M HCl solution (5 mL) was added to the reaction mixture, the pH of the system was adjusted to about 4, etOAc (20 mL. Times.2) was used for extraction, and anhydrous Na was used as the organic layer 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by column chromatography on silica gel (eluent: PE/EtOAc (v/v) =3/1) to give a white solid (62 mg, 48%).
MS(ESI,pos.ion)m/z=445.2[M+H] + .
Step three: synthesis of N- (4- (ethylsulfonyl) benzyl) - (S) -4- (2- ((difluoromethoxy) methyl) -4- (4- (trifluoromethyl) benzyl) piperazin-1-yl) benzamide
(S) -4- (2- ((difluoromethoxy) methyl) -4- (4- (trifluoromethyl) benzyl) piperazin-1-yl) benzoic acid (106 mg,0.24 mmol), (4- (ethylsulfonyl) phenyl) methylamine (78 mg,0.39 mmol), EDCI (99 mg,0.52 mmol), HOBT (72 mg,0.53 mmol) were dissolved in DCM (5 mL) and DIPEA (0.2 mL,1.00 mmol) was added and stirred at RT for 12h. DCM (50 mL) was added to the reaction solution, saturated NaHCO 3 (20 mL. Times.2) washing, saturated NaCl (20 mL), anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by column chromatography on silica gel (eluent: PE/EtOAc (v/v) =1/3) to give a white solid (52 mg, 35%).
MS(ESI,pos.ion)m/z=626.0[M+H] + .
1 H NMR(400MHz,DMSO-d 6 )δ(ppm):8.89(t,J=5.5Hz,1H),7.82(dd,J=14.2,8.3Hz,4H),7.70(d,J=7.9Hz,2H),7.62–7.53(m,4H),6.96(d,J=8.6Hz,2H),6.62(t,J=75.8Hz,1H),4.56(d,J=5.4Hz,2H),4.30–4.18(m,2H),3.74(dd,J=24.8,9.4Hz,2H),3.65–3.54(m,2H),3.26(dd,J=14.5,7.2Hz,2H),3.09(t,J=10.8Hz,1H),2.93(t,J=9.8Hz,2H),2.23(t,J=13.6Hz,2H),1.09(t,J=7.3Hz,3H).
Example 18N- (1- (4- (ethylsulfonyl) phenyl) -2-hydroxyethyl) - (R) -4- (4- (cyclopentylcarbonyl) piperazin-1-yl) benzamide
Step one: synthesis of tert-butyl 4- (4- (methoxycarbonyl) phenyl) piperazine-1-carboxylate
Tert-butyl piperazine-1-carboxylate (8.20 g,44.00 mmol), methyl 4-iodobenzoate (16.90 g,64.50 mmol), pd under nitrogen 2 (dba) 3 (3.88g,4.24mmol),Ruphos(4.20g,9.00mmol),Cs 2 CO 3 (27.90 g,85.60 mmol) was dissolved in toluene (80 mL) and heated at 110deg.C for 19h. The crude product was isolated by column chromatography on silica gel (eluent: PE/EtOAc (v/v) =3/1) to give a yellow solid (10 g, 71%).
MS(ESI,pos.ion)m/z=321.1[M+H] + .
Step two: synthesis of methyl 4- (piperazin-1-yl) benzoate
Tert-butyl 4- (4- (methoxycarbonyl) phenyl) piperazine-1-carboxylate (3 g,9.36 mmol) was dissolved in DCM (8 mL), and HCl 1, 4-dioxane (16 mL,48.00mmol,3 mol/L) was added in ice bath and stirred at room temperature for 13h. Removing part of the organic solvent by distillation under reduced pressure, precipitating solid, suction-filtering, washing the filter cake with DCM (10 mL. Times.2), adding saturated NaHCO to the filter cake 3 (120 mL), pH of the system was adjusted to about 8, DCM (150 mL. Times.2) was used for extraction, and the organic layer was dried over Na 2 SO 4 Drying and concentration under reduced pressure gave a brown solid (1.9 g, 92%).
MS(ESI,pos.ion)m/z=221.2[M+H] + .
Step three: synthesis of methyl 4- (4- (cyclopentylcarbonyl) piperazin-1-yl) benzoate
Methyl 4- (piperazin-1-yl) benzoate (1.05 g,4.77 mmol), cyclopentanecarboxylic acid (0.68 mL,6.30 mmol), EDCI (1.88 g,9.81 mmol), HOAT (1.35 g,9.92 mmol) were dissolved in DCM (15 mL), and DIPEA (4 mL,24.20 mmol) was added and reacted at room temperature for 12h. DCM (50 mL) was added to the reaction solution, saturated NH 4 Cl solution (30 mL. Times.2) wash, saturated NaHCO 3 Washing with solution (30 mL. Times.2), anhydrous Na was used as the organic layer 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by silica gel column chromatography (eluent: PE/EtOAc (v/v) =2/1) to give a white solid (1.5 g, 99%).
MS(ESI,pos.ion)m/z=317.1[M+H] + .
Step four: synthesis of 4- (4- (cyclopentylcarbonyl) piperazin-1-yl) benzoic acid
Methyl 4- (4- (cyclopentylcarbonyl) piperazin-1-yl) benzoate (1.50 g,4.70 mmol), liOH. H 2 O (2.06 g,49.10 mmol) was dissolved in THF/MeOH/H2O (8 mL/4mL/4 mL) and reacted at room temperature for 20H. Concentrated under reduced pressure, 1.0M HCl solution (25 mL) was added to the reaction mixture, the pH of the system was adjusted to about 4, and a white solid was precipitated, filtered off with suction, and dried to give a white solid (1.4 g, 98%).
MS(ESI,pos.ion)m/z=303.2[M+H] + .
Step five: synthesis of N- (1- (4- (ethylsulfonyl) phenyl) -2-hydroxyethyl) - (R) -4- (4- (cyclopentylcarbonyl) piperazin-1-yl) benzamide
4- (4- (cyclopentylcarbonyl) piperazin-1-yl) benzoic acid (205 mg,0.68 mmol), (2R) -2-amino-2- (4- (ethylsulfonyl) phenyl) ethanol (188 mg,0.82 mmol), EDCI (257 mg,1.34 mmol), HOBT (185 mg,1.37 mmol) were dissolved in DCM (8 mL) and DIPEA (0.8 mL,5.00 mmol) was added and stirred at room temperature for 17h. DCM (50 mL) was added to the reaction solution, saturated NaHCO 3 (20 mL. Times.2) washing, saturated NaCl (20 mL), anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by silica gel column chromatography (eluent: DCM/MeOH (v/v) =15/1) to give a white solid (324 mg, 93%).
MS(ESI,pos.ion)m/z=514.2[M+H] + .
1 H NMR(400MHz,DMSO-d 6 )δ(ppm):8.54(d,J=7.7Hz,1H),7.84(d,J=7.9Hz,4H),7.66(d,J=7.9Hz,2H),7.00(d,J=8.8Hz,2H),5.14(d,J=6.6Hz,1H),5.03(t,J=5.4Hz,1H),3.79–3.69(m,2H),3.64(d,J=15.3Hz,4H),3.26(dd,J=14.5,7.1Hz,6H),3.08–2.98(m,1H),1.78(d,J=7.7Hz,2H),1.65(ddd,J=27.9,13.5,6.7Hz,4H),1.53(dd,J=9.0,5.4Hz,2H),1.10(t,J=7.3Hz,3H).
Example 19N- (4- (ethylsulfonyl) benzyl) -4- (4- (cyclopentylcarbonyl) piperazin-1-yl) benzamide
4- (4- (Cyclopentylcarbonyl) piperazin-1-yl) benzoic acid (204 mg,0.67 mmol), (4- (ethylsulfonyl) phenyl) methylamine (267 mg,1.34 mmol), EDCI (267 mg,1.39 mmol), HOBT (230 mg,1.70 mmol) were dissolved in DCM (8 mL) and DIPEA (0.88 mL,5.30 mmol) was added and stirred at room temperature for 17h. DCM (50 mL) was added to the reaction solution, saturated NaHCO 3 (20 mL. Times.2) washing, saturated NaCl (20 mL), anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by column chromatography on silica gel (eluent: PE/EtOAc (v/v) =1/5) to give a white solid (101 mg, 31%).
MS(ESI,pos.ion)m/z=484.1[M+H] + .
1 H NMR(400MHz,DMSO-d 6 )δ(ppm):8.90(t,J=5.8Hz,1H),7.83(t,J=9.5Hz,4H),7.56(d,J=8.1Hz,2H),7.00(d,J=8.8Hz,2H),4.56(d,J=5.7Hz,2H),3.63(d,J=17.5Hz,4H),3.29(s,2H),3.25(t,J=7.1Hz,4H),3.08–2.99(m,1H),1.78(d,J=7.7Hz,2H),1.73–1.59(m,4H),1.55(dd,J=9.6,6.0Hz,2H),1.09(t,J=7.3Hz,3H).
Example 20 (R) -N- (1- (4- (ethylsulfonyl) phenyl) -2-hydroxyethyl) -4- (4- (4- (trifluoromethyl) benzyl) piperazin-1-yl) benzamide
Step one: synthesis of methyl 4- (4- (4- (trifluoromethyl) benzyl) piperazin-1-yl) benzoate
Methyl 4- (piperazin-1-yl) benzoate (1.04 g,4.72 mmol), 4- (trifluoromethyl) benzaldehyde (0.8 mL,6.00 mmol) was dissolved in THF/EtOH (6 mL/6 mL) and AcOH (1.3 mL,23.00 mmol), STAB (4.80 g,23.00 mmol) was added and reacted at 60℃for 18h. Saturated NaHCO is added into the reaction solution 3 Solution (60 mL), pH of the system was adjusted to about 8, etOAc (100 mL. Times.2) was used for extraction, and anhydrous Na was used as the organic layer 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by column chromatography over silica gel (eluent: PE/EtOAc (v/v) =3/1) to give a brown solid (1.6 g, 90%).
MS(ESI,pos.ion)m/z=379.3[M+H] + .
Step two: synthesis of 4- (4- (4- (trifluoromethyl) benzyl) piperazin-1-yl) benzoic acid
Methyl 4- (4- (4- (trifluoromethyl) benzyl) piperazin-1-yl) benzoate (1.60 g,4.20 mmol), liOH. H 2 O (1.82 g,43.40 mmol) was dissolved in THF/MeOH/H2O (8 mL/4mL/4 mL) and reacted at room temperature for 48H. Concentrated under reduced pressure, 1.0M HCl solution (20 mL) was added to the reaction mixture, the pH of the system was adjusted to about 4, and a brown solid was precipitated, filtered off with suction, and dried to give a brown solid (1.45 g, 94%).
MS(ESI,pos.ion)m/z=365.4[M+H] + .
Step three: synthesis of (R) -N- (1- (4- (ethylsulfonyl) phenyl) -2-hydroxyethyl) -4- (4- (4- (trifluoromethyl) benzyl) piperazin-1-yl) benzamide
4- (4- (4- (trifluoromethyl) benzyl) piperazin-1-yl) benzoic acid (200 mg,0.55 mmol), (2R) -2-amino-2- (4- (ethylsulfonyl) phenyl) ethanol (154 mg,0.67 mmol), EDCI (221 mg,1.15 mmol), HOBT (152 mg,1.12 mmol) were dissolved in DCM (8 mL) and DIPEA (0.7 mL,4.00 mmol) was added and stirred at room temperature for 17h. DCM (50 mL) was added to the reaction solution, saturated NaHCO 3 (20 mL. Times.2) washing, saturated NaCl (20 mL), anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by silica gel column chromatography (eluent: DCM/MeOH (v/v) =15/1) to give a white solid (247 mg, 78%).
MS(ESI,pos.ion)m/z=576.7[M+H] + .
1 H NMR(400MHz,DMSO-d 6 )δ(ppm):8.50(d,J=7.6Hz,1H),7.82(dd,J=12.4,8.5Hz,4H),7.72(d,J=7.9Hz,2H),7.65(d,J=8.2Hz,2H),7.59(d,J=7.9Hz,2H),6.97(d,J=8.8Hz,2H),5.13(dd,J=13.7,6.8Hz,1H),5.02(t,J=5.8Hz,1H),3.78–3.68(m,2H),3.64(s,2H),3.26(dd,J=12.8,5.4Hz,6H),2.53(s,4H),1.10(t,J=7.3Hz,3H).
Example 21N- ((R) -1- (4- (ethylsulfonyl) phenyl) -2-hydroxyethyl) -4- ((R) -2-methyl-4- (4- (trifluoromethyl) benzyl) piperazin-1-yl) benzamide
Step one: synthesis of (R) -4- (4- (methoxycarbonyl) phenyl) -3-methylpiperazine-1-carboxylic acid tert-butyl ester
(R) -3-methylpiperazine-1-carboxylic acid tert-butyl ester (1.10 g,5.50 mmol), methyl 4-iodobenzoate (2.21 g,8.43 mmol), pd under nitrogen 2 (dba) 3 (546mg,0.60mmol),Ruphos(475mg,1.02mmol),Cs 2 CO 3 (3.34 g,10.30 mmol) was dissolved in toluene (20 mL) and heated at 110deg.C for 17h. The crude product was isolated by column chromatography over silica gel (eluent: PE/EtOAc (v/v) =5/1) to give a yellow solid (1.8 g, 98%).
MS(ESI,pos.ion)m/z=335.2[M+H] + .
Step two: synthesis of methyl (R) -4- (2-methylpiperazin-1-yl) benzoate
(R) -4- (4- (methoxycarbonyl) phenyl) -3-methylpiperazine-1-carboxylic acid tert-butyl ester (1.80 g,5.40 mmol) was dissolved in DCM (5 mL), HCl 1, 4-dioxane (12 mL,48.00mmol,4 mol/L) was added under ice-bath and stirred at room temperature for 20h. Concentrating under reduced pressure, removing part of the organic solvent, and adding saturated NaHCO 3 (80 mL), the pH of the system was adjusted to about 8, DCM (100 mL. Times.)2) Extraction, anhydrous Na of organic layer 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by silica gel column chromatography (eluent: DCM/MeOH (v/v) =10/1) to give a brown oil (1.3 g, 100%). MS (ESI, pos.ion) m/z=235.2 [ m+h] + .
Step three: synthesis of methyl (R) -4- (2-methyl-4- (4- (trifluoromethyl) benzyl) piperazin-1-yl) benzoate
Methyl (R) -4- (2-methylpiperazin-1-yl) benzoate (520 mg,2.22 mmol), 4- (trifluoromethyl) benzaldehyde (0.4 mL,3.00 mmol) was dissolved in THF/EtOH (6 mL/6 mL) and AcOH (0.6 mL,10.00 mmol), STAB (2.3 g,11.00 mmol) was added and reacted at 60℃for 19h. Saturated NaHCO is added into the reaction solution 3 Solution (40 mL), pH of the system was adjusted to about 8, etOAc (60 mL. Times.2) was used for extraction, and anhydrous Na was used as the organic layer 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was isolated by column chromatography over silica gel (eluent: PE/EtOAc (v/v) =5/1) to give a yellow solid (730 mg, 84%).
MS(ESI,pos.ion)m/z=393.2[M+H] + .
Step four: synthesis of (R) -4- (2-methyl-4- (4- (trifluoromethyl) benzyl) piperazin-1-yl) benzoic acid
(R) -methyl 4- (2-methyl-4- (4- (trifluoromethyl) benzyl) piperazin-1-yl) benzoate (730 mg,1.87 mmol), liOH. H 2 O (935 mg,22.28 mmol) was dissolved in THF/MeOH/H2O (4 mL/2mL/2 mL) and reacted at room temperature for 48H. Concentrating under reduced pressure, adding 1.0M HCl solution (20 mL) to the reaction solution, adjusting pH to about 4, extracting with EtOAc (50 mL. Times.2), and collecting anhydrous Na as organic layer 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by silica gel column chromatography (eluent: DCM/MeOH (v/v) =10/1) to give a white solid (700 mg, 99%).
MS(ESI,pos.ion)m/z=379.3[M+H] + .
Step five: synthesis of N- ((R) -1- (4- (ethylsulfonyl) phenyl) -2-hydroxyethyl) -4- ((R) -2-methyl-4- (4- (trifluoromethyl) benzyl) piperazin-1-yl) benzamide
(R) -4- (2-methyl-4- (4- (trifluoromethyl) benzyl) piperazin-1-yl) benzoic acid (250 mg,0.66 mmol), (R) -2-amino-2- (4- (ethylsulfonyl) phenyl) ethanol (196 mg,0.85 mmol), EDCI (308 mg,1.61 mmol), HOBT(192 mg,1.42 mmol) was dissolved in DCM (8 mL) and DIPEA (0.55 mL,3.30 mmol) was added and stirred at room temperature for 21h. DCM (50 mL) was added to the reaction solution, saturated NH 4 Cl (20 mL. Times.2), saturated NaCl (20 mL), anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by silica gel column chromatography (eluent: DCM/MeOH (v/v) =15/1) to give a white solid (335 mg, 86%).
MS(ESI,pos.ion)m/z=590.3[M+H] + .
1 H NMR(400MHz,DMSO-d 6 )δ(ppm):8.48(d,J=7.0Hz,1H),7.89–7.77(m,4H),7.72(d,J=7.1Hz,2H),7.65(d,J=7.6Hz,2H),7.60(d,J=6.6Hz,2H),6.91(d,J=7.8Hz,2H),5.13(d,J=6.0Hz,1H),5.02(s,1H),4.18(s,1H),3.78–3.64(m,3H),3.53(dd,J=22.1,13.0Hz,2H),3.26(d,J=7.0Hz,2H),3.05(d,J=10.3Hz,1H),2.90(d,J=8.7Hz,1H),2.70(d,J=9.8Hz,1H),2.31(d,J=8.9Hz,1H),2.19(s,1H),1.10(s,6H).
Example 22N- ((R) -1- (4- (ethylsulfonyl) phenyl) -2-hydroxyethyl) -4- (2, 6-dimethyl-4- (4- (trifluoromethyl) benzyl) piperazin-1-yl) benzamide
Step one: synthesis of tert-butyl 4- (4- (methoxycarbonyl) phenyl) -3, 5-dimethylpiperazine-1-carboxylate
3, 5-Dimethylpiperazine-1-carboxylic acid tert-butyl ester (1.02 g,4.76 mmol), methyl 4-iodobenzoate (1.95 g, 7.44), pd under nitrogen 2 (dba) 3 (433mg,0.47mmol),Ruphos(451mg,0.97mmol),Cs 2 CO 3 (3.10 g,9.50 mmol) was dissolved in toluene (20 mL) and heated at 110deg.C for 22h. The crude product was isolated by column chromatography over silica gel (eluent: PE/EtOAc (v/v) =5/1) to give a yellow oil (400 mg, 24%).
MS(ESI,pos.ion)m/z=349.2[M+H] + .
Step two: synthesis of methyl 4- (2, 6-dimethylpiperazin-1-yl) benzoate
4- (4- (methoxycarbonyl) phenyl)Tert-butyl 3, 5-dimethylpiperazine-1-carboxylate (400 mg,1.15 mmol) was dissolved in DCM (5 mL), HCl 1, 4-dioxane (10 mL,40.00mmol,4 mol/L) was added on ice and stirred at room temperature for 20h. Concentrating under reduced pressure, removing part of the organic solvent, and adding saturated NaHCO 3 (40 mL), pH of the system was adjusted to about 8, DCM (60 mL. Times.2) was used for extraction, and the organic layer was dried over Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by silica gel column chromatography (eluent: DCM/MeOH (v/v) =10/1) to give a brown oil (210 mg, 74%). MS (ESI, pos.ion) m/z=249.1 [ m+h ] ] + .
Step three: synthesis of methyl 4- (2, 6-dimethyl-4- (4- (trifluoromethyl) benzyl) piperazin-1-yl) benzoate
Methyl 4- (2, 6-dimethylpiperazin-1-yl) benzoate (210 mg,0.85 mmol), 4- (trifluoromethyl) benzaldehyde (0.15 mL,1.10 mmol) was dissolved in THF/EtOH (3 mL/3 mL) and AcOH (0.25 mL,4.4 mmol), STAB (1.02 g,4.81 mmol) was added and reacted at 60℃for 18h. Saturated NaHCO is added into the reaction solution 3 Solution (30 mL), pH of the system was adjusted to about 8, etOAc (50 mL. Times.2) was used for extraction, and anhydrous Na was used as the organic layer 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by column chromatography over silica gel (eluent: PE/EtOAc (v/v) =5/1) to give a yellow solid (221 mg, 64%).
MS(ESI,pos.ion)m/z=407.1[M+H] + .
Step four: synthesis of 4- (2, 6-dimethyl-4- (4- (trifluoromethyl) benzyl) piperazin-1-yl) benzoic acid
Methyl 4- (2, 6-dimethyl-4- (4- (trifluoromethyl) benzyl) piperazin-1-yl) benzoate (221 mg,0.54 mmol), liOH. H 2 O (284 mg,6.77 mmol) was dissolved in THF/MeOH/H2O (2 mL/1mL/1 mL) and reacted at room temperature for 21H. Concentrating under reduced pressure, adding 1.0M HCl solution (10 mL) to the reaction solution, adjusting pH to about 4, extracting with EtOAc (30 mL. Times.2), and collecting anhydrous Na as organic layer 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by silica gel column chromatography (eluent: DCM/MeOH (v/v) =10/1) to give a yellow solid (210 mg, 98%).
MS(ESI,pos.ion)m/z=393.2[M+H] + .
Step five: synthesis of N- ((R) -1- (4- (ethylsulfonyl) phenyl) -2-hydroxyethyl) -4- (2, 6-dimethyl-4- (4- (trifluoromethyl) benzyl) piperazin-1-yl) benzamide
4- (2, 6-dimethyl-4- (4- (trifluoromethyl) benzyl) piperazin-1-yl) benzoic acid (210 mg,0.54 mmol), (R) -2-amino-2- (4- (ethylsulfonyl) phenyl) ethanol (150 mg,0.65 mmol), EDCI (287 mg,1.50 mmol), HOBT (148 mg,1.10 mmol) were dissolved in DCM (8 mL) and DIPEA (0.45 mL,2.70 mmol) was added and stirred at room temperature for 48h. DCM (50 mL) was added to the reaction solution, saturated NH 4 Cl (20 mL. Times.2), saturated NaCl (20 mL), anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by column chromatography over silica gel (eluent: DCM/MeOH (v/v) =15/1) to give a yellow solid (101 mg, 31%).
MS(ESI,pos.ion)m/z=604.2[M+H] + .
1 H NMR(400MHz,DMSO-d 6 )δ(ppm):8.48(d,J=7.6Hz,1H),7.82(t,J=8.5Hz,4H),7.73(d,J=7.7Hz,2H),7.64(t,J=6.9Hz,4H),6.88(d,J=8.5Hz,2H),5.13(dd,J=13.1,6.4Hz,1H),5.02(t,J=5.6Hz,1H),3.93(s,2H),3.80–3.68(m,2H),3.64(s,2H),3.26(dd,J=14.5,7.2Hz,2H),2.63(d,J=10.4Hz,2H),2.35(d,J=8.3Hz,2H),1.11(dd,J=17.9,6.6Hz,9H).
Example 23N- ((R) -1- (4- (ethylsulfonyl) phenyl) -2-hydroxyethyl) -4- ((S) -2- ((difluoromethoxy) methyl) -4- (4- (trifluoromethyl) benzyl) piperazin-1-yl) benzamide
(S) -4- (2- ((difluoromethoxy) methyl) -4- (4- (trifluoromethyl) benzyl) piperazin-1-yl) benzoic acid (114 mg,0.26 mmol), (R) -2-amino-2- (4- (ethylsulfonyl) phenyl) ethanol (75 mg,0.33 mmol), EDCI (115 mg,0.60 mmol), HOBT (73 mg,0.54 mmol) were dissolved in DCM (5 mL) and DIPEA (0.21 mL,1.30 mmol) was added and stirred at room temperature for 12h. DCM (50 mL) was added to the reaction solution, saturated NaHCO 3 (20 mL. Times.2) washing, saturated NaCl (20 mL), anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by silica gel column chromatography (eluent: DCM/MeOH (v/v) =15/1) to give a white solid (126 mg, 75%).
MS(ESI,pos.ion)m/z=656.1[M+H] + .
1 H NMR(400MHz,DMSO-d 6 )δ(ppm):8.52(d,J=7.7Hz,1H),7.83(t,J=7.8Hz,4H),7.70(d,J=8.0Hz,2H),7.65(d,J=8.2Hz,2H),7.59(d,J=7.9Hz,2H),6.96(d,J=8.8Hz,2H),6.62(t,J=75.8Hz,1H),5.14(dd,J=13.3,6.7Hz,1H),5.03(t,J=5.6Hz,1H),4.31–4.18(m,2H),3.74(dd,J=21.7,10.1Hz,4H),3.58(d,J=13.5Hz,2H),3.26(q,J=7.4Hz,2H),3.09(t,J=10.9Hz,1H),2.93(t,J=10.2Hz,2H),2.24(dd,J=17.7,10.3Hz,2H),1.10(t,J=7.3Hz,3H).
Example 24 (R) -N- (1- (4- (ethylsulfonyl) phenyl) -2-hydroxyethyl) -4- (8- (4- (trifluoromethyl) benzyl) -2-oxa-5, 8-diazaspiro [3.5] non-5-yl) benzamide
Step one: synthesis of tert-butyl 5- (4- (methoxycarbonyl) phenyl) -2-oxa-5, 8-diazaspiro [3.5] nonane-8-carboxylate
2-oxa-6, 9-diazaspiro [3.5] under nitrogen protection]Nonane-6-carboxylic acid tert-butyl ester (313 mg,2.69 mmol), methyl 4-iodobenzoate (1.1 g,4.20 mmol), pd 2 (dba) 3 (250mg,0.27mmol),Ruphos(249mg,0.53mmol),Cs 2 CO 3 (1.7 g,5.20 mmol) in toluene (8 mL) was heated at 110deg.C for 19h. The crude product was isolated by column chromatography over silica gel (eluent: PE/EtOAc (v/v) =3/1) to give a yellow oil (430 mg, 44%).
MS(ESI,pos.ion)m/z:307.1[M+H-56] + .
Step two: synthesis of methyl 4- (2- (chloromethyl) -2- (hydroxymethyl) piperazin-1-yl) benzoate
5- (4- (methoxycarbonyl) phenyl) -2-oxa-5, 8-diazaspiro [3.5]]Nonane-8-carboxylic acid tert-butyl ester (430 mg,1.19 mmol) was dissolved in DCM (2 mL), HCl 1, 4-dioxane (5 mL,20.00mmol,4 mol/L) was added under ice and stirred at room temperature for 16h. Removing part of the organic solvent by distillation under reduced pressure, and adding saturated NaHCO 3 (20 mL), pH of the system was adjusted to about 8, DCM (30 mL. Times.2) was used for extraction, and the organic layer was dried over Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by silica gel column chromatography (eluent: DCM/MeOH (v/v) =10/1) to give a yellow oil (223 mg, 63%).
MS(ESI,pos.ion)m/z=299.3[M+H] + .
Step three: synthesis of methyl 4- (8- (4- (trifluoromethyl) benzyl) -2-oxa-5, 8-diazaspiro [3.5] nonan-5-yl) benzoate
Methyl 4- (2- (chloromethyl) -2- (hydroxymethyl) piperazin-1-yl) benzoate (223 mg,0.75 mmol), 1- (bromomethyl) -4- (trifluoromethyl) benzene (244 mg,1.02 mmol), K 2 CO 3 (458 mg,3.31 mmol) in CH 3 CN (5 mL) was reacted at 90℃for 6 hours. Concentrating under reduced pressure, and then adding saturated NaHCO to the residue 3 Solution (20 mL), DCM (30 mL. Times.2) extraction, anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by column chromatography on silica gel (eluent: PE/EtOAc (v/v) =4/1) to give a yellow oil (123 mg, 39%).
MS(ESI,pos.ion)m/z=421.9[M+H] + .
Step four: synthesis of 4- (8- (4- (trifluoromethyl) benzyl) -2-oxa-5, 8-diazaspiro [3.5] nonan-5-yl) benzoic acid
4- (8- (4- (trifluoromethyl) benzyl) -2-oxa-5, 8-diazaspiro [ 3.5)]Methyl nonan-5-yl benzoate (123 mg,0.29 mmol), liOH. H 2 O (135 mg,3.22 mmol) was dissolved in THF/MeOH/H2O (1.5 mL/0.8mL/0.8 mL) and reacted at room temperature. Concentrating under reduced pressure, adding 1.0M HCl solution (6 mL) to the reaction solution, adjusting pH to about 4, extracting with EtOAc (30 mL. Times.2), and collecting anhydrous Na as organic layer 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by silica gel column chromatography (eluent: DCM/MeOH (v/v) =10/1) to give a yellow solid (51 mg, 43%).
MS(ESI,pos.ion)m/z=407.8[M+H] + .
Step five: synthesis of (R) -N- (1- (4- (ethylsulfonyl) phenyl) -2-hydroxyethyl) -4- (8- (4- (trifluoromethyl) benzyl) -2-oxa-5, 8-diazaspiro [3.5] non-5-yl) benzamide
4- (8- (4- (trifluoromethyl)) to) Benzyl) -2-oxa-5, 8-diazaspiro [3.5]Nonan-5-yl) benzoic acid (51 mg,0.16 mmol), (R) -2-amino-2- (4- (ethylsulfonyl) phenyl) ethanol (37 mg,0.16 mmol), EDCI (56 mg,0.29 mmol), HOBT (37 mg,0.27 mmol) were dissolved in DCM (5 mL) and DIPEA (0.12 mL,0.73 mmol) was added and stirred at room temperature for 7h. DCM (50 mL) was added to the reaction solution, saturated NH 4 Cl (20 mL. Times.2), saturated NaCl (20 mL), anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the crude product was separated by silica gel column chromatography (eluent: DCM/MeOH (v/v) =12/1) to give a yellow solid (18 mg, 23%).
MS(ESI,pos.ion)m/z=618.2[M+H] + .
1 H NMR(400MHz,DMSO-d 6 )δ(ppm):8.44(d,J=7.6Hz,1H),7.83(d,J=8.2Hz,2H),7.78(d,J=8.7Hz,2H),7.66(dd,J=12.2,8.3Hz,4H),7.56(d,J=7.9Hz,2H),6.86(d,J=8.8Hz,2H),5.12(dd,J=13.3,6.9Hz,1H),5.00(t,J=5.6Hz,1H),3.87(t,J=7.5Hz,3H),3.81–3.66(m,5H),3.61(d,J=15.5Hz,1H),3.29–3.22(m,2H),2.89(d,J=4.5Hz,1H),2.79(s,2H),2.56(dd,J=15.5,10.0Hz,2H),1.10(t,J=7.3Hz,3H).
Biological Activity test
Fluorescence Resonance Energy Transfer (FRET) assay
1. Test method
(1) Preparation of RORγt assay buffer and 10mM DTT
100mL of 1 Xbasic assay buffer (HEPES (pH 7.4), 100mM NaCl,0.01%BSA) was prepared, 154.25mg of DTT was added, and thoroughly mixed.
(2) Preparing gradient concentration of compound
a. Standard compounds were formulated and diluted to 2.5mm in 100% dmso, 3-fold dilution, 11 gradients to a final concentration of 42.34nM;
b. experimental compounds were formulated and reference standard compounds were used.
(3) Preparation of 1x protein solution mixture
a. The required amount of 2x B-RORγt LBD/SA-APC protein mixture is prepared. The concentration of B-RORγtLBD was 40nM and the concentration of SA-APC was 20nM, and the mixture was gently mixed upside down and incubated at room temperature for 15 minutes. 400nM biotin was added and mixed gently upside down and incubated for 10 min at room temperature;
b. the desired amount of 2x Biotin-SRC1/SA-eu protein mixture was formulated. The concentration of Bioin-SRC1 was 40nM and the concentration of SA-eu was 20nM, mixed gently upside down and incubated for 15 min at room temperature. 200nM biotin was added and mixed gently upside down and incubated for 10 min at room temperature;
c.1, 1 uniformly mixing the protein mixture prepared in the step a and the step b, and incubating for 5 minutes at room temperature;
d. adding 25 μl of the mixture in step c to a 384 well plate containing the test compound;
e.1000rpm for one minute;
f. incubate for 1 hour at room temperature.
(4) Data acquisition and calculation
After incubation at room temperature for 1 hour, fluorescence values at 665nm and 615nm were measured with EnVision plate reader, respectively, and inhibition ratios were calculated, and the IC finally obtained 50 The values are shown in Table 1;
inhibition ratio (%) = [ (X-Min)/(Max-Min) ]. Times.100%
X is the number of "665/615" for the test compound; min is the average of "665/615" for the DMSO blank; max is the average of "665/615" for 10. Mu.M SRC.
2. Experimental results
Table 1: evaluation of the inhibitory Activity of the Compounds of the invention on RORγt at the molecular level
Conclusion: the compound has good inhibition activity on RORγt.
Pharmacokinetic evaluation
ICR mice were weighed after 15 hours overnight fast and randomized according to body weight, and test compound formulation vehicle was 5% dmso+5% solutol+90% saline. For intravenous injectionA test group to which the drug was administered, a dose of 1mg/kg was administered to the test animals; for the test group for oral administration, a dose of 5mg/kg was administered to the test animals. Venous blood (about 0.2 mL) was then taken at time points 0, 0.083 (intravenous only), 0.25, 0.5, 1.0, 2.0, 5.0, 7.0 and 24 hours and placed in EDTA-K 2 In an anticoagulation tube, plasma was collected by centrifugation at 11,000rpm for 2 minutes and stored at-20℃or-70℃until LC/MS/MS analysis was performed. The drug concentration in the plasma at each time point was determined and pharmacokinetic parameters were calculated from the drug concentration-time curve.
The pharmacokinetic properties of the compounds of the invention were tested by the above assays. Experimental results show that the compound has good pharmacokinetic characteristics in ICR mice.
Finally, it should be noted that there are other ways to implement the invention. Accordingly, the embodiments of the present invention are to be construed as illustrative, not restrictive of the invention, but may be modified and equivalents added to the scope of the invention as defined by the appended claims. All publications or patents cited herein are incorporated by reference.

Claims (12)

1. A compound represented by the formula (I) or a stereoisomer or a pharmaceutically acceptable salt of the compound represented by the formula (I),
(I),
wherein:
r is C 1-4 An alkyl group;
Z 1 、Z 2 、Z 3 、Z 4 、Z 5 、Z 6 、Z 7 and Z 8 Each independently is CR 1 Or N;
each R is 1 Independently hydrogen, deuterium or C 1-6 An alkyl group;
ring A isOr->The A ring is optionally substituted with 1, 2, 3 or 4R' s 2 Substitution;
each R is 2 Independently is fluorine, chlorine, bromine, iodine, hydroxyl, oxo, amino, nitro, cyano, C 1-4 Alkyl, C 1-4 Haloalkyl, C 1-4 Alkoxy, phenyl, C 3-6 Cycloalkyl, -C 0-4 alkylene-OR a or-C 0-4 Alkylene- (O) n -(C(=O)) m -NR c R d The method comprises the steps of carrying out a first treatment on the surface of the The R is 2 Optionally by 1, 2 or 3R f Substitution;
each R is a Independently hydrogen, C 1-4 Alkyl, C 1-4 Haloalkyl, C 3-6 Cycloalkyl or 3-7 membered heterocyclyl; each R is c And R is d Independently hydrogen, hydroxy, C 1-4 Alkyl, C 1-4 Haloalkyl or C 1-4 An alkoxy group; each R is as follows a 、R c And R is d Independently optionally substituted with 1, 2, 3, 4, 5 or 6R g Substitution;
each R is f And R is g Independently is fluorine, chlorine, bromine, iodine, oxo, hydroxy, amino, nitro, cyano, C 1-6 Alkyl, hydroxy substituted C 1-6 Alkyl, C 1-6 Haloalkyl, C 3-8 Cycloalkyl, 3-12 membered heterocyclyl, C 1-6 Alkoxy, C 1-6 Alkylamino or C 1-6 Haloalkoxy groups;
ring B is C 6-10 Aryl or C 3-8 Cycloalkyl, wherein the B ring is optionally substituted with 1, 2, 3 or 4R' s e Substitution;
each R is e Independently deuterium, fluorine, chlorine, bromine, iodine, cyano, nitro, hydroxy, amino, C 1-4 Alkyl, C 1-4 Alkoxy, C 3-6 Cycloalkyl, 3-8 membered heterocyclyl, hydroxy substituted C 1-4 Alkyl, hydroxy substituted C 1-4 Haloalkyl, C 1-4 Haloalkyl or C 1-4 Haloalkoxy groups;
L 1 is-C (=o) NH-or-NHC (=o) -;
L 2 is-CR 3 R 4 -;
L 3 Is carbonyl, -O-, -NR 5 -or-CR 6 R 7 -;
R 3 Is hydrogen;
R 4 c substituted by hydrogen or hydroxy 1-6 An alkyl group;
R 5 is hydrogen, C 1-6 Alkyl or C 1-6 A haloalkyl group;
each R is 6 And R is 7 Independently hydrogen or C 1-6 An alkyl group;
each m and n is independently 0 or 1.
2. The compound of claim 1, wherein R is methyl, ethyl, n-propyl, or isopropyl;
Each R is 1 Independently hydrogen, deuterium, methyl, ethyl, n-propyl, or isopropyl;
R 3 is hydrogen; r is R 4 Is hydrogen, hydroxymethyl, hydroxyethyl or hydroxy-n-propyl;
R 5 is hydrogen, methyl, ethyl, isopropyl, n-propyl, isobutyl, tert-butyl, n-butyl, monofluoromethyl, difluoromethyl, trifluoromethyl, 1, 2-difluoroethyl or 1, 1-difluoroethyl;
each R is 6 And R is 7 Independently is hydrogen, methyl, ethyl, isopropyl, n-propyl, isobutyl, tert-butyl or n-butyl.
3. A compound according to claim 1 or 2, wherein the B ring is phenyl, cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl; the B ring is optionally substituted with 1,2, 3 or 4R' s e Substitution;
each R is e Is independently deuterium, fluorine, chlorine, bromine, iodine, cyano, nitro, hydroxy, amino, methyl, ethyl, n-propyl, isopropyl, hydroxymethyl, 2-hydroxyethyl, 3-hydroxy-n-propyl, 2-hydroxy-1, 3-hexafluoroisopropyl, monofluoromethyl, difluoromethyl, trifluoromethyl, 1,2-Difluoroethyl, methoxy, ethoxy, n-propoxy, t-butoxy, monofluoromethoxy, difluoromethoxy, trifluoromethoxy or 1, 2-difluoroethoxy.
4. The compound of claim 1 or 2, wherein each R 2 Independently is fluoro, chloro, bromo, iodo, hydroxy, oxo, amino, nitro, cyano, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, t-butyl, monofluoromethyl, difluoromethyl, trifluoromethyl, monofluoroethyl, 1-difluoroethyl, 1, 2-difluoroethyl, methoxy, ethoxy, n-propoxy, isopropoxy, phenyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, -OR a 、-CH 2 -OR a 、-CH 2 CH 2 -OR a 、-CH 2 (CH 2 ) 2 -OR a 、-CH 2 CH(CH 3 )-OR a 、-C(CH 3 ) 2 CH 2 -OR a 、-CH 2 (CH 2 ) 3 -OR a 、-CH(CH 3 )CH(CH 3 )-OR a 、-CH 2 C(CH 3 ) 2 -OR a 、-(O) n -(C(=O)) m -NR c R d 、-CH 2 -(O) n -(C=O) m -NR c R d 、-CH 2 CH 2 -(O) n -(C(=O)) m -NR c R d 、-CH 2 (CH 2 ) 2 -(O) n -(C(=O)) m -NR c R d 、-CH 2 CH(CH 3 )-(O) n -(C(=O)) m -NR c R d 、-C(CH 3 ) 2 CH 2 -(O) n -(C(=O)) m -NR c R d 、-CH 2 (CH 2 ) 3 -(O) n -(C(=O)) m -NR c R d 、-CH(CH 3 )CH(CH 3 )-(O) n -(C(=O)) m -NR c R d or-CH 2 C(CH 3 ) 2 -(O) n -(C(=O)) m -NR c R d The method comprises the steps of carrying out a first treatment on the surface of the Wherein the R is 2 Optionally is covered by1. 2 or 3R f And (3) substitution.
5. A compound according to claim 1 or 2, each R a Independently is hydrogen, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, monofluoromethyl, difluoromethyl, trifluoromethyl, monofluoroethyl, 2-difluoroethyl, 1, 2-difluoroethyl, 2-trifluoroethyl,、/>、/>、/>、/>An oxetanyl, tetrahydrofuranyl, tetrahydropyranyl, oxetanyl, cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl group; each R is c And R is d Independently hydrogen, hydroxy, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, monofluoromethyl, difluoromethyl, trifluoromethyl, monofluoroethyl, 2-difluoroethyl, 1, 2-difluoroethyl, 2-trifluoroethyl, methoxy, ethoxy, n-propoxy or isopropoxy;
Wherein each R is a 、R c And R is d Independently optionally substituted with 1,2, 3, 4, 5 or 6R g And (3) substitution.
6. The compound of claim 1 or 2, wherein each R f And R is g Independently is fluorine, chlorine, bromine, iodine, oxo, hydroxy, amino, nitro, cyano, methyl, ethyl, n-propyl, isopropyl, n-butyl, -CH 2 OH, monofluoromethyl, difluoromethyl, trifluoroMethyl, monoethyl, 1-difluoroethyl, 1, 2-difluoroethyl, oxiranyl, oxetanyl, tetrahydrofuranyl, tetrahydropyranyl,、/>、/>、/>Cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, methoxy, ethoxy, n-propoxy, isopropoxy, monofluoromethoxy, difluoromethoxy or trifluoromethoxy.
7. A compound having the structure of one of:
(6)、/>(20)、(22)、/>(99)、(103)、/>(112)、(113)、/>(114)、(115) Or a pharmaceutically acceptable salt thereof.
8. A pharmaceutical composition comprising a compound of any one of claims 1-7, and a pharmaceutically acceptable excipient, carrier, adjuvant, or combination thereof.
9. Use of a compound according to any one of claims 1-7 or a pharmaceutical composition according to claim 8 for the manufacture of a medicament for the prevention or treatment of a roryt mediated disease, disorder or syndrome in a mammal.
10. The use of claim 9, wherein the disease, disorder or syndrome mediated by roryt is psoriasis, systemic lupus erythematosus, multiple sclerosis, inflammatory bowel disease, rheumatoid arthritis, autoimmune eye disease, ankylosing spondylitis, asthma, chronic obstructive pulmonary disease, osteoarthritis, allergic rhinitis, allergic dermatitis, crohn's disease or kawasaki disease.
11. The use according to claim 10, wherein the inflammatory bowel disease is colitis.
12. The use of claim 11, wherein the colitis is ulcerative colitis.
CN202011615479.1A 2020-01-06 2020-12-31 ROR gamma t inhibitor and application thereof in medicines Active CN113072521B (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN2020100091330 2020-01-06
CN202010009133 2020-01-06

Publications (2)

Publication Number Publication Date
CN113072521A CN113072521A (en) 2021-07-06
CN113072521B true CN113072521B (en) 2024-04-05

Family

ID=76609207

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202011615479.1A Active CN113072521B (en) 2020-01-06 2020-12-31 ROR gamma t inhibitor and application thereof in medicines

Country Status (1)

Country Link
CN (1) CN113072521B (en)

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
CN105980353A (en) * 2013-12-05 2016-09-28 领先制药Cel模型知识产权公司 Ror gamma (rory) modulators
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
WO2018116285A1 (en) * 2016-12-23 2018-06-28 Glenmark Pharmaceuticals S.A. Substituted morpholine derivatives as ror gamma modulators

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10844017B2 (en) * 2017-02-09 2020-11-24 Fudan University Biaryl compound, 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
CN105980353A (en) * 2013-12-05 2016-09-28 领先制药Cel模型知识产权公司 Ror gamma (rory) modulators
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
WO2018116285A1 (en) * 2016-12-23 2018-06-28 Glenmark Pharmaceuticals S.A. Substituted morpholine derivatives as ror gamma modulators

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
From RORγt Agonist to Two Types of RORγt Inverse Agonists;Yonghui Wang et al;《ACS Med. Chem. Lett.》;第9卷(第2期);第120−124页 *

Also Published As

Publication number Publication date
CN113072521A (en) 2021-07-06

Similar Documents

Publication Publication Date Title
AU2018351400B2 (en) Dihydropyrimidine compounds and uses thereof in medicine
CN108689942B (en) Nitrogen-containing bicyclic compound, and preparation method and application thereof
CA2876690C (en) Dihydropyrimidine compounds and their application in pharmaceuticals
CN114502158A (en) IRAK degradation agent and application thereof
CN104918934A (en) 3-substituted pyrazoles and use as DLK inhibitors
CN112513021B (en) ROR gamma antagonist and application thereof in medicines
CN113072542B (en) ROR gamma t inhibitor and preparation method and application thereof
TW201609692A (en) Substituted pyrazoles and uses thereof
WO2015112847A1 (en) Arylpyridinone itk inhibitors for treating inflammation and cancer
CN111333648A (en) 8-substituted aromatic ring vinyl xanthine derivative and application thereof
CN112789277A (en) Guanidine derivatives and use thereof
JP2022531088A (en) Substituted pyrolopyridine as a JAK inhibitor
CN110981876B (en) 8-substituted styrylxanthine derivatives and uses thereof
CN113072476B (en) ROR gamma t inhibitor and preparation method and application thereof
CN113072521B (en) ROR gamma t inhibitor and application thereof in medicines
CN107089955B (en) Sulfonamide derivative and preparation method and application thereof
CN112300165B (en) 8-substituted styrylxanthine derivatives and uses thereof
CN111018856B (en) 8-substituted styrylxanthine derivatives and uses thereof
CN111072676B (en) Nitrogen-containing fused tricyclic derivatives and uses thereof
CN113072538B (en) ROR gamma t inhibitor and application thereof in medicines
CN114075139A (en) Five-membered heteroaromatic ring compound and application thereof in medicines
CN110922408B (en) 3H- [1,2,3] triazolo [4,5-d ] pyrimidin-5-amine derivatives and uses thereof
CN109988169B (en) Octahydropyrrolo [3,4-c ] pyrrole derivatives and uses thereof
CN111072663B (en) 8-substituted styrylxanthine derivatives and uses thereof
CN113072546A (en) Five-membered heteroaromatic derivative and preparation method and application 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
CB02 Change of applicant information
CB02 Change of applicant information

Address after: 523808 No.1, Gongye North Road, Songshanhu Park, Dongguan City, Guangdong Province

Applicant after: Guangdong Dongyangguang Pharmaceutical Co.,Ltd.

Address before: 523808 No.1, Gongye North Road, Songshanhu Park, Dongguan City, Guangdong Province

Applicant before: SUNSHINE LAKE PHARMA Co.,Ltd.

GR01 Patent grant
GR01 Patent grant