WO2023078241A1 - 芳胺类衍生物及其制备方法和医药用途 - Google Patents

芳胺类衍生物及其制备方法和医药用途 Download PDF

Info

Publication number
WO2023078241A1
WO2023078241A1 PCT/CN2022/128963 CN2022128963W WO2023078241A1 WO 2023078241 A1 WO2023078241 A1 WO 2023078241A1 CN 2022128963 W CN2022128963 W CN 2022128963W WO 2023078241 A1 WO2023078241 A1 WO 2023078241A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
compound
general formula
aryl
halogen
Prior art date
Application number
PCT/CN2022/128963
Other languages
English (en)
French (fr)
Inventor
闫旭
田卫学
陈士柱
殷惠军
Original Assignee
中国医药研究开发中心有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 中国医药研究开发中心有限公司 filed Critical 中国医药研究开发中心有限公司
Priority to CN202280007466.7A priority Critical patent/CN116472047A/zh
Publication of WO2023078241A1 publication Critical patent/WO2023078241A1/zh

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems

Definitions

  • the present invention relates to aromatic amine derivatives and their preparation method and medical application. Specifically, the present invention relates to a compound represented by general formula (I), a preparation method thereof, a pharmaceutical composition containing it, and its use as a Toll-like receptor (TLR) agonist for treating diseases related to TLR8 activity .
  • TLR Toll-like receptor
  • TLRs Toll-like receptors
  • TLRs are all type I transmembrane glycoproteins, consisting of 16-28 leucine-rich repeat (LRR) extracellular domain, transmembrane domain and cytoplasmic Toll/IL-1 receptor (TIR) structure domain composition. X-ray crystallography analysis confirmed that the TLR and LRR domains were horseshoe-shaped. So far, 11 members have been found in humans, among which TLR1, 2, 4, 5, 6, 10 and 11 are located on the cell surface, and TLR3, 7, 8, and 9 are located on the endosomal membrane. TLR8 and TLR7 are phylogenetically close to each other and have a high degree of sequence homology, and are located on the adjacent X chromosome (Xp22).
  • LRR leucine-rich repeat
  • TIR cytoplasmic Toll/IL-1 receptor
  • TLR8 relies on the MyD88 signaling pathway, induces and activates protease-1 (AP-1) and nuclear factor ⁇ B (NF- ⁇ B) to transfer to the nucleus, induces the expression of related genes in the nucleus, secretes chemokines and inflammatory factors, etc., and plays a role in transcription. role of regulation.
  • MyD88 myeloid differentiation factor 88
  • TLR8 can also activate the mitogen-activated protein kinase (MAPK) signaling pathway, including p38, ERK, and JNK, etc., which are mainly involved in the regulation of cell proliferation, cell differentiation, cell transformation, and apoptosis, and are closely related to inflammation, tumors, etc. A variety of diseases are closely related (Journal of Immunology, 2017, 33, 813).
  • MAPK mitogen-activated protein kinase
  • Hepatitis B virus is a particulate double-stranded DNA virus. Activation of TLR8 can effectively inhibit HBV replication in vivo and in vitro, thus becoming a target for the development of treatments for chronic HBV.
  • TLR8 agonist ssRNA40 can selectively activate perihepatic innate immune cells to produce a large amount of IFN- ⁇ , thereby inhibiting the replication of hepatitis B virus, which has the potential to be used as a treatment for hepatitis virus infection.
  • Stimulation of PBMC with TLR8 agonists was found to induce high levels of IFN- ⁇ and TNF- ⁇ production, thereby inhibiting HBV replication (Current Opinion in Virology, 2018, 30, 9).
  • TLR is not only expressed on immune cells, but also expressed in various tumor cells, participates in tumor immune surveillance, and plays different roles in tumor growth. Among them, the activation of TLR8 enhances the activity of natural killer cells (NK cells), improves antibody-dependent cell-regulated cytotoxicity (ADCC) and induces Th1 polarization. TLR8 agonists serve as potential adjuvants in cancer therapy, aiming to induce specific immune responses against tumor cells and improve the clinical efficacy of approved monoclonal antibody therapies, especially in individuals with reduced ADCC.
  • NK cells natural killer cells
  • ADCC antibody-dependent cell-regulated cytotoxicity
  • TLR-8 agonists In view of the important potential of TLR-8 agonists in the treatment of various diseases, there is an urgent clinical need for new TLR-8 agonists with strong activity and high selectivity.
  • the inventors designed and synthesized a series of arylamine compounds, which exhibit excellent TLR8 agonistic activity, and can be developed as drugs for treating TLR8-related diseases.
  • the object of the present invention is to provide a compound represented by general formula (I) or its stereoisomer, tautomer, mesoform, racemate, enantiomer, diastereomer Enantiomers, or mixtures thereof, or pharmaceutically acceptable salts thereof,
  • X 1 is CR 1 or N
  • X 2 is CR 2 or N
  • X 3 is CR 3 or N
  • X 4 is CR 4 or N
  • L is selected from a bond, -(CH 2 ) v -, -C(O)(CH 2 ) t - or -(CH 2 ) t C(O)-;
  • R is selected from hydrogen, halogen, amino, nitro, cyano, hydroxyl, mercapto, carboxyl, alkyl, alkoxy, haloalkyl, haloalkoxy;
  • R is selected from hydrogen, halogen, cyano, oxo, alkyl, alkenyl, alkynyl, -OR a , -SR a , -NR a R b , cycloalkyl, heterocyclyl, aryl and hetero Aryl, wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl are optionally further substituted by one or more Q groups;
  • R is selected from hydrogen, halogen, cyano, oxo, alkyl, alkenyl, alkynyl, -OR a , -SR a , -NR a R b , cycloalkyl, heterocyclyl, aryl and hetero Aryl, wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl are optionally further substituted by one or more Q groups;
  • R is selected from hydrogen, halogen, amino, nitro, cyano, hydroxyl, mercapto, carboxyl, alkyl, alkoxy, haloalkyl, haloalkoxy;
  • R and R are each independently selected from hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl; wherein the alkyl, alkenyl , alkynyl, cycloalkane Base, heterocyclyl, aryl, heteroaryl is optionally further selected from deuterium, halogen, nitro, cyano, oxo, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, Aryl, Heteroaryl, -OR a , -SR a , -NR a R b , -C(O)R a , -O(O)CR a , -C(O)OR a , -C(O) One or more of NR a R b , -NR a C(O)R b , -S(O) n R a , -S(O
  • Q 1 and Q 2 are each independently selected from halogen, nitro, cyano, oxo, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -OR a , -SR a , -(CH 2 ) v -NR a R b , -NR a R b , -C(O)R a , -O(O)CR a , -C(O)OR a , -C(O )NR a R b , -NR a C(O)R b , -S(O) n R a , -S(O) n NR a R b and -NR a S(O) n R b , wherein the Alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl are optionally further selected from
  • R and R are each independently selected from hydrogen, halogen, hydroxyl, nitro, cyano, oxo, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein The alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl are optionally further selected from halogen, amino, nitro, cyano, carboxyl, ester, oxo, Alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -OR c , -SR c , -(CH 2 ) v -OR c , -(CH 2 ) v -NR c R d , -NR c R d , -C(O)R c , -O(O)CR c , -C(O)OR c , -C(O
  • R a and R b form a nitrogen-containing heterocyclic group together with their connected nitrogen atoms, and the nitrogen-containing heterocyclic group optionally further contains one or more heteroatoms selected from N, O, and S in addition to N , the nitrogen-containing heterocyclic group is optionally further selected from halogen, nitro, cyano, oxo, carboxyl, ester, alkyl, haloalkyl, cycloalkyl, heterocyclic, aryl, heteroaryl group, -OR c , -SR c , -(CH 2 ) v -OR c , -(CH 2 ) v -NR c R d , -NR c R d , -C(O)R c , -O(O )CR c , -C(O)OR c , -C(O)NR c R d , -NR c C(O)R d , -S(O)
  • R and R are each independently selected from hydrogen, halogen, hydroxyl, nitro, cyano, oxo, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein The alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl are optionally further selected from halogen, amino, nitro, cyano, hydroxyl, mercapto, carboxyl, ester, One or more groups in oxo, alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl and heteroaryl are substituted;
  • R c and R d form a nitrogen-containing heterocyclic group together with the nitrogen atom they are connected to, and the nitrogen-containing heterocyclic group optionally further contains one or more heteroatoms selected from N, O, and S in addition to N , the nitrogen-containing heterocyclic group is optionally further selected from halogen, nitro, cyano, oxo, hydroxyl, mercapto, carboxyl, ester, alkyl, alkoxy, cycloalkyl, heterocyclic, Aryl and heteroaryl;
  • n 1 or 2;
  • v is an integer from 1 to 6;
  • t 0 to 6.
  • the compound represented by general formula (I) according to the present invention or its stereoisomer, tautomer, mesoform, racemate, enantiomer isomers, diastereoisomers, or mixtures thereof, or pharmaceutically acceptable salts thereof which are compounds represented by general formula (II) or their stereoisomers, tautomers, mesomers, Racemates, enantiomers, diastereomers, or mixtures thereof, or pharmaceutically acceptable salts thereof,
  • X 1 , X 2 , X 3 , L, R 4 , R 5 , and R 6 are as defined in general formula (I).
  • the compound represented by general formula (I) according to the present invention or its stereoisomer, tautomer, mesoform, racemate, enantiomer isomers, diastereomers, or mixtures thereof, or pharmaceutically acceptable salts thereof which are compounds represented by general formula (III) or stereoisomers, tautomers, and mesomers thereof , racemate, enantiomer, diastereomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof,
  • L, R 1 , R 3 , R 4 , R 5 , and R 6 are as defined in general formula (I).
  • the compound represented by general formula (I) according to the present invention or its stereoisomer, tautomer, mesoform, racemate, enantiomer isomers, diastereoisomers, or mixtures thereof, or pharmaceutically acceptable salts thereof which are compounds represented by general formula (IV) or stereoisomers, tautomers, and mesomers thereof , racemate, enantiomer, diastereomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof,
  • L, R 1 , R 2 , R 4 , R 5 , and R 6 are as defined in general formula (I).
  • the compound represented by general formula (I) according to the present invention or its stereoisomer, tautomer, mesoform, racemate, enantiomer isomers, diastereomers, or mixtures thereof, or pharmaceutically acceptable salts thereof which are compounds represented by general formula (V) or stereoisomers, tautomers, and mesomers thereof , racemate, enantiomer, diastereomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof,
  • L, R 2 , R 4 , R 5 , and R 6 are as defined in general formula (I).
  • L is selected from a bond or -C(O)-; preferably a bond.
  • L is selected from -(CH 2 ) v -; v is 1 or 2, preferably 1.
  • X 1 , X 2 , X 3 , X 4 , R 1 , R 2 , R 3 , R 4 , R 5 , and R 6 are as defined in general formula (I).
  • R 3 is selected from hydrogen, halogen, C 1 -C 6 alkyl, C 6 -C 10 aryl and 5 to 10 membered heteroaryl, wherein the C 1 -C 6 alkyl, C 6 -C 10 aryl and 5 to 10 membered heteroaryl are optionally further substituted by one or more Q groups;
  • Q 2 is selected from halogen, C 1 -C 6 alkyl, 4-6 membered heterocyclic group, C 6 -C 10 aryl, 5 to 10 membered heteroaryl, -NR a R b , wherein said C 6 - C 10 aryl and 5 to 10 membered heteroaryl are optionally further substituted by one or more groups selected from halogen, C 1 -C 6 alkyl, -(CH 2 ) v -NR c R d ;
  • R a and R b are each independently selected from hydrogen, C 1 -C 6 alkyl
  • R a and R b form a 4-6 membered nitrogen-containing heterocyclic group together with their connected nitrogen atoms.
  • the 4-6 membered nitrogen-containing heterocyclic group optionally further contains a group selected from N, O, One or more heteroatoms of S, the 4-6 membered nitrogen-containing heterocyclic group is optionally further selected from oxo, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, C 3 -C 6 Cycloalkyl, -OR c , -SR c , -(CH 2 ) v -OR c , -NR c R d are substituted by one or more groups;
  • R c and R d are each independently selected from hydrogen, C 1 -C 6 alkyl
  • R c and R d form a 4-6 membered nitrogen-containing heterocyclic group together with their connected nitrogen atoms, and the 4-6 membered nitrogen-containing heterocyclic group may optionally further contain a group selected from N, O, One or more heteroatoms of S, the 4-6 membered nitrogen-containing heterocyclic group is optionally further substituted by one or more groups selected from halogen, C 1 -C 6 alkyl;
  • v is an integer of 1 to 6.
  • R 3 is selected from C 6 -C 10 aryl and 5 to 10 membered heteroaryl, preferably phenyl or 5-6 membered heteroaryl; wherein the C 6 -C 10 aryl and 5 to 10 membered heteroaryl Optionally further substituted by one or more Q2 groups;
  • Q 2 is selected from halogen, C 1 -C 6 alkyl, 4-6 membered heterocyclic group, C 6 -C 10 aryl, 5 to 10 membered heteroaryl, -NR a R b , wherein said C 6 - C 10 aryl and 5 to 10 membered heteroaryl are optionally further substituted by one or more groups selected from halogen, C 1 -C 6 alkyl;
  • R a and R b are each independently selected from hydrogen, C 1 -C 6 alkyl
  • R a and R b form a 4-6 membered nitrogen-containing heterocyclic group together with their connected nitrogen atoms.
  • the 4-6 membered nitrogen-containing heterocyclic group optionally further contains a group selected from N, O, One or more heteroatoms of S, the 4-6 membered nitrogen-containing heterocyclic group is optionally further selected from oxo, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, C 3 -C 6 Cycloalkyl, -OR c , -SR c , -(CH 2 ) v -OR c , -NR c R d are substituted by one or more groups;
  • R c and R d are each independently selected from hydrogen, C 1 -C 6 alkyl
  • R c and R d form a 4-6 membered nitrogen-containing heterocyclic group together with their connected nitrogen atoms
  • the 4-6 membered nitrogen-containing heterocyclic group may optionally further contain a group selected from N, O, One or more heteroatoms of S
  • the 4-6 membered nitrogen-containing heterocyclic group is optionally further substituted by one or more groups selected from halogen, C 1 -C 6 alkyl, and v is an integer from 1 to 6 , preferably 1 or 2.
  • R is selected from 5 to 10 membered heteroaryl, preferably 5 or 6 membered heteroaryl, more preferably pyridyl, pyrimidinyl, imidazolyl, pyrazolyl, pyrrolyl, pyridoimidazolyl, pyridopyrrolyl, pyridine Pyrazolyl, benzimidazolyl, benzopyrazolyl, benzopyrrolyl; the heteroaryl is optionally further substituted by one or more Q2 groups;
  • Q 2 is selected from halogen, C 1 -C 6 alkyl, C 6 -C 10 aryl, preferably phenyl, 5 to 10 membered heteroaryl, preferably 5 or 6 membered heteroaryl, wherein the aryl and heteroaryl Optionally further substituted by one or more groups selected from halogen, C 1 -C 6 alkyl.
  • R is selected from 5 to 10 membered heteroaryl, preferably 5 or 6 membered heteroaryl, more preferably pyridyl, pyrimidinyl, imidazolyl, pyrazolyl, pyrrolyl, pyridoimidazolyl, pyridopyrrolyl, pyridine Pyrazolyl, benzimidazolyl, benzopyrazolyl, benzopyrrolyl; the heteroaryl is optionally further substituted by one or more Q2 groups;
  • Q 2 is selected from halogen, C 1 -C 6 alkyl, 4-6 membered heterocyclic group or -NR a R b , preferably -NR a R b ;
  • R a and R b are each independently selected from hydrogen, C 1 -C 6 alkyl
  • R a and R b form a 4-6 membered nitrogen-containing heterocyclic group together with their connected nitrogen atoms.
  • the 4-6 membered nitrogen-containing heterocyclic group optionally further contains a group selected from N, O, One or more heteroatoms of S, the 4-6 membered nitrogen-containing heterocyclic group is optionally further selected from oxo, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, C 3 -C 6 Cycloalkyl, -OR c , -SR c , -(CH 2 ) v -OR c , -NR c R d are substituted by one or more groups;
  • R c and R d are each independently selected from hydrogen, C 1 -C 6 alkyl
  • R c and R d form a 4-6 membered nitrogen-containing heterocyclic group together with their connected nitrogen atoms
  • the 4-6 membered nitrogen-containing heterocyclic group may optionally further contain a group selected from N, O, One or more heteroatoms of S
  • the 4-6 membered nitrogen-containing heterocyclic group is optionally further substituted by one or more groups selected from halogen, C 1 -C 6 alkyl, and v is an integer from 1 to 6 , preferably 1 or 2.
  • R 3 is selected from C 6 -C 10 aryl, preferably phenyl; the aryl is optionally further substituted by Q 2 groups;
  • Q 2 is selected from 5- or 6-membered heteroaryl, wherein the 5- or 6-membered heteroaryl is optionally further substituted by one or more groups selected from halogen, C 1 -C 6 alkyl.
  • R 3 is selected from C 1 -C 6 alkyl, preferably methyl; said C 1 -C 6 alkyl is optionally further substituted by a Q 2 group;
  • Q 2 is selected from C 6 -C 10 aryl, preferably phenyl, wherein the C 6 -C 10 aryl is optionally further substituted by -(CH 2 ) v -NR c R d ;
  • R c and R d form a 4-6 membered nitrogen-containing heterocyclic group together with the nitrogen atom they are connected to.
  • the 4-6 membered nitrogen-containing heterocyclic group may further contain, in addition to N, a group selected from N, O, S One or more heteroatoms, the 4-6 membered nitrogen-containing heterocyclic group is optionally further substituted by one or more groups selected from halogen, C 1 -C 6 alkyl;
  • v is an integer of 1 to 6, preferably 1 or 2.
  • R 2 is selected from hydrogen, halogen, C 1 -C 6 alkyl, -NR a R b , and the C 1 -C 6 alkyl is optionally further substituted by Q 1 ;
  • Q 1 is selected from C 6 -C 10 aryl, 5 to 10 membered heteroaryl, wherein the C 6 -C 10 aryl and 5 to 10 membered heteroaryl are optionally further selected from halogen, C 1 -C One or more groups of 6 alkyl, -(CH 2 ) v -NR c R d are substituted;
  • R a and R b are each independently selected from hydrogen, C 1 -C 6 alkyl
  • R c and R d are each independently selected from hydrogen, C 1 -C 6 alkyl
  • R c and R d form a 4-6 membered nitrogen-containing heterocyclic group together with their connected nitrogen atoms, and the 4-6 membered nitrogen-containing heterocyclic group may optionally further contain a group selected from N, O, One or more heteroatoms of S, the 4-6 membered nitrogen-containing heterocyclic group is optionally further substituted by one or more groups selected from halogen, C 1 -C 6 alkyl;
  • v is an integer of 1 to 6, preferably 1 or 2.
  • R a and R b are each independently selected from hydrogen, C 1 -C 6 alkyl.
  • R 2 is selected from C 1 -C 6 alkyl, preferably methyl; said C 1 -C 6 alkyl is optionally further substituted by a Q 2 group;
  • Q 2 is selected from C 6 -C 10 aryl, preferably phenyl, wherein the C 6 -C 10 aryl is optionally further substituted by -(CH 2 ) v -NR c R d ;
  • R c and R d form a 4-6 membered nitrogen-containing heterocyclic group together with the nitrogen atom they are connected to.
  • the 4-6 membered nitrogen-containing heterocyclic group may further contain, in addition to N, a group selected from N, O, S One or more heteroatoms, the 4-6 membered nitrogen-containing heterocyclic group is optionally further substituted by one or more groups selected from halogen, C 1 -C 6 alkyl;
  • v is an integer of 1 to 6, preferably 1 or 2.
  • R 5 and R 6 are each independently selected from hydrogen and C 1 -C 12 alkyl, and the C 1 -C 12 alkyl is optionally further selected from deuterated, -OR a , -SR a , -NR a R One or more groups of b are substituted;
  • R a is selected from hydrogen, C 1 -C 6 alkyl
  • R b is selected from hydrogen, C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl and 5 to 7 membered heterocyclyl;
  • R 5 is hydrogen
  • R 6 is C 1 -C 12 alkyl
  • the C 1 -C 12 alkyl is optionally further substituted by one or more groups selected from deuterated, -OH.
  • Typical compounds of the invention include, but are not limited to:
  • the present invention further provides a method for preparing the compound represented by the general formula (III-1) according to the present invention or its stereoisomer, tautomer, mesoform, racemate, enantiomer isomers, diastereoisomers, or a mixture thereof, or a pharmaceutically acceptable salt thereof, comprising the following steps:
  • R 1 , R 3 , R 4 , R 5 , R 6 are as defined in general formula (III-1).
  • the present invention further provides a method for preparing the compound represented by the general formula (IV-1) according to the present invention or its stereoisomer, tautomer, mesoform, racemate, enantiomer isomers, diastereoisomers, or a mixture thereof, or a pharmaceutically acceptable salt thereof, comprising the following steps:
  • R 1 , R 2 , R 4 , R 5 , R 6 are as defined in general formula (IV-1).
  • the present invention further provides a method for preparing the compound represented by the general formula (V-1) according to the present invention or its stereoisomer, tautomer, mesoform, racemate, enantiomer isomers, diastereoisomers, or a mixture thereof, or a pharmaceutically acceptable salt thereof, comprising the following steps:
  • Metal-catalyzed cross-coupling e.g. Suzuki coupling
  • boronic acid or boric acid pinacol ester compounds affords compounds of formula C4; deprotection with a suitable acid (e.g. trifluoroacetic acid) then affords Compounds represented by general formula (V-1); catalysts such as Pd(PPh 3 ) 4 , K 2 CO 3 , Cs 2 CO 3 ;
  • the compound of formula C7 and boric acid or boric acid pinacol ester compound or R 2 Y compound pass metal-catalyzed cross-coupling reaction (such as Suzuki coupling), obtain the compound shown in general formula (V-1); Catalyst such as Pd ( PPh 3 ) 4 , K 2 CO 3 , Cs 2 CO 3 ;
  • R 2 , R 4 , R 5 , R 6 are as defined in general formula (V-1).
  • the present invention further provides a pharmaceutical composition, which comprises the compound of the general formula according to the present invention or its stereoisomers, tautomers, mesoforms, racemates, enantiomers, Diastereomers, or mixtures thereof, or pharmaceutically acceptable salts thereof, and pharmaceutically acceptable carriers or excipients.
  • the present invention further relates to compounds of the general formula according to the present invention or their stereoisomers, tautomers, mesoforms, racemates, enantiomers, diastereoisomers, or Use of its mixture form, or its pharmaceutically acceptable salt, or a pharmaceutical composition comprising it in the preparation of a TLR8 agonist.
  • the present invention further relates to compounds of the general formula according to the present invention or their stereoisomers, tautomers, mesoforms, racemates, enantiomers, diastereoisomers, or Use of its mixture form, or its pharmaceutically acceptable salt, or a pharmaceutical composition comprising it in the preparation of a medicament for preventing or treating a TLR8-related disease
  • the disease may be a viral infectious disease or a malignant tumor, and the viral infectious disease
  • the viral infectious disease such as viral hepatitis B, HIV virus infection, the malignant tumors such as breast cancer, cervical cancer, colon cancer, lung cancer, stomach cancer, rectal cancer, pancreatic cancer, brain cancer, skin cancer, oral cancer, prostate cancer, bone cancer, Renal cancer, ovarian cancer, bladder cancer, liver cancer, fallopian tube tumor, ovarian tumor, peritoneal tumor, melanoma, solid tumor, glioma, glioblastoma, hepatocellular carcinoma, papillary renal tumor, head and neck
  • the present invention further relates to a compound of the general formula according to the present invention or its stereoisomers, tautomers, mesoforms, racemates, enantiomers, diastereoisomers , or a mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same, for use as a TLR8 agonist.
  • the present invention further relates to a compound of the general formula according to the present invention or its stereoisomers, tautomers, mesoforms, racemates, enantiomers, diastereoisomers , or a mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising it, which is used for preventing or treating a TLR8-related disease
  • the disease may be a viral infectious disease or a malignant tumor, and the viral infectious disease such as Viral hepatitis B, HIV virus infection, the malignant tumors such as breast cancer, cervical cancer, colon cancer, lung cancer, stomach cancer, rectal cancer, pancreatic cancer, brain cancer, skin cancer, oral cancer, prostate cancer, bone cancer, kidney cancer Carcinoma, ovarian cancer, bladder cancer, liver cancer, fallopian tube tumor, ovarian tumor, peritoneal tumor, melanoma, solid tumor, glioma, glioblastoma, hepatocellular carcinoma, papillary renal tumor, head and neck tumor
  • the present invention further relates to a method for activating TLR8, which comprises administering an effective amount of the compound of the general formula according to the present invention or its stereoisomers, tautomers, mesoforms, exo Racemates, enantiomers, diastereomers, or mixtures thereof, or pharmaceutically acceptable salts thereof or pharmaceutical compositions comprising the same.
  • the present invention further relates to a method for preventing or treating TLR8-related diseases, which comprises administering an effective amount of the compound of the general formula according to the present invention or its stereoisomers, tautomers, mesoform, racemate, enantiomer, diastereoisomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same;
  • the disease may be viral infectivity Disease or malignant tumor, the viral infectious disease such as hepatitis B, HIV virus infection, the malignant tumor such as breast cancer, cervical cancer, colon cancer, lung cancer, gastric cancer, rectal cancer, pancreatic cancer, brain cancer, skin cancer Carcinoma, Oral Cancer, Prostate Cancer, Bone Cancer, Kidney Cancer, Ovarian Cancer, Bladder Cancer, Liver Cancer, Fallopian Tube Tumor, Ovarian Tumor, Peritoneal Tumor, Melanoma, Solid Tumor, Glioma, Glioblastoma, Hepatocyte Carcino
  • the compounds of the present invention can form pharmaceutically acceptable base addition salts or acid addition salts with bases or acids.
  • the base includes inorganic bases and organic bases.
  • Acceptable organic bases include diethanolamine, ethanolamine, N-methylglucamine, triethanolamine, tromethamine, etc.
  • Acceptable inorganic bases include aluminum hydroxide, hydroxide Calcium, Potassium Hydroxide, Sodium Carbonate and Sodium Hydroxide etc.
  • the acid includes inorganic acid and organic acid, acceptable inorganic acid includes hydrochloric acid, sulfuric acid, nitric acid, phosphoric acid, hydrobromic acid and the like.
  • Acceptable organic acids include acetic acid, trifluoroacetic acid, formic acid, ascorbic acid, and the like.
  • the pharmaceutical composition containing the active ingredient may be in a form suitable for oral administration, such as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixir.
  • Oral compositions can be prepared according to any method known in the art for the preparation of pharmaceutical compositions, and such compositions can contain one or more ingredients selected from the group consisting of sweeteners, flavoring agents, coloring agents and preservatives, To provide pleasing and palatable medicinal preparations. Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients suitable for the manufacture of tablets.
  • excipients can be inert excipients such as calcium carbonate, sodium carbonate, lactose, calcium or sodium phosphate; granulating and disintegrating agents such as microcrystalline cellulose, croscarmellose sodium, corn starch or alginic acid; binders such as starch, gelatin, polyvinylpyrrolidone or acacia; and lubricants such as magnesium stearate, stearic acid or talc.
  • These tablets may be uncoated or coated by known techniques to mask the taste of the drug or to delay disintegration and absorption in the gastrointestinal tract, thus providing sustained release over an extended period of time.
  • water-soluble taste-masking materials such as hydroxypropylmethylcellulose or hydroxypropylcellulose, or time-extending materials such as ethylcellulose, cellulose acetate butyrate may be used.
  • Hard gelatin capsules in which the active ingredient is admixed with an inert solid diluent such as calcium carbonate, calcium phosphate, or kaolin, or in which the active ingredient is admixed with a water-soluble carrier such as polyethylene glycol or an oil vehicle such as peanut oil, liquid paraffin, or olive oil may also be used.
  • Soft gelatin capsules provide an oral formulation.
  • Aqueous suspensions contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions.
  • excipients are suspending agents, such as sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethylcellulose, sodium alginate, polyvinylpyrrolidone, and acacia; dispersing or wetting agents, which may be natural
  • the resulting phospholipids such as lecithin, or condensation products of alkylene oxides with fatty acids, such as polyoxyethylene stearate, or condensation products of ethylene oxide with long-chain fatty alcohols, such as heptadecanylethyleneoxycetate Wax alcohols (heptadeca ethyl ene oxy cetanol), or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitols, such as polyethylene oxide sorbitan monooleate, or ethylene oxide with Condensation products of partial esters derived from fatty acids and hexi
  • Aqueous suspensions may also contain one or more preservatives, such as ethyl or n-propylparaben, one or more coloring agents, one or more flavoring agents and one or more sweeteners.
  • preservatives such as ethyl or n-propylparaben
  • coloring agents such as ethyl or n-propylparaben
  • flavoring agents such as sucrose, saccharin, or aspartame.
  • Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil, such as arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin.
  • Oily suspensions may contain a thickening agent, such as beeswax, hard paraffin or cetyl alcohol. Sweetening and flavoring agents as mentioned above may be added to provide a palatable preparation. These compositions can be preserved by the addition of antioxidants such as butylated hydroxyanisole or alpha-tocopherol.
  • Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient with a dispersing or wetting agent, suspending agent or one or more preservatives for admixture. Suitable dispersing or wetting agents and suspending agents are mentioned above. Other excipients, for example sweetening, flavoring and coloring agents, may also be added. These compositions are preserved by the addition of antioxidants such as ascorbic acid.
  • the pharmaceutical compositions of the invention may also be in the form of oil-in-water emulsions.
  • the oily phase may be a vegetable oil such as olive oil or arachis oil, or a mineral oil such as liquid paraffin or mixtures thereof.
  • Suitable emulsifiers may be naturally occurring phospholipids, such as soybean lecithin, and esters or partial esters derived from fatty acids and hexitol anhydrides, such as sorbitan monooleate, and the condensation of said partial esters with ethylene oxide Products such as polyethylene oxide sorbitan monooleate.
  • the emulsions may also contain sweetening, flavoring, preservative and antioxidant agents.
  • Syrups and elixirs may be formulated with sweetening agents, for example glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative, coloring agents and antioxidants.
  • sweetening agents for example glycerol, propylene glycol, sorbitol or sucrose.
  • Such formulations may also contain a demulcent, a preservative, coloring agents and antioxidants.
  • the pharmaceutical compositions of the present invention may be in the form of sterile injectable aqueous solutions.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • the sterile injectable preparation may be a sterile injectable oil-in-water microemulsion in which the active ingredient is dissolved in the oily phase.
  • the active ingredient is dissolved in a mixture of soybean oil and lecithin.
  • the oil solution is then treated in a mixture of water and glycerol to form a microemulsion.
  • the injectable solution or microemulsion can be injected into the patient's bloodstream by local bolus injection.
  • solutions and microemulsions are preferably administered in a manner that maintains a constant circulating concentration of the compounds of the invention. To maintain this constant concentration, a continuous intravenous delivery device can be used.
  • the pharmaceutical composition of the present invention may be in the form of sterile injectable aqueous or oily suspension for intramuscular and subcutaneous administration.
  • This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above.
  • the sterile injectable preparation can also be a sterile injectable solution or suspension prepared in a non-toxic parenterally acceptable diluent or solvent, for example a solution in 1,3-butanediol.
  • sterile fixed oils are conveniently employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid are prepared as injectables.
  • the compounds of this invention may be administered in the form of suppositories for rectal administration.
  • These pharmaceutical compositions can be prepared by mixing the drug with a suitable non-irritating excipient which is solid at ordinary temperatures but liquid in the rectum and will therefore melt in the rectum to release the drug.
  • suitable non-irritating excipient include cocoa butter, glycerinated gelatin, hydrogenated vegetable oils, polyethylene glycols of various molecular weights and mixtures of fatty acid esters of polyethylene glycols.
  • the dosage of the drug depends on many factors, including but not limited to the following factors: the activity of the specific compound used, the patient's age, the patient's body weight, the patient's health status, the patient's behavior, the patient's Diet, administration time, administration method, excretion rate, drug combination, etc.
  • the optimal treatment method such as the treatment mode, the daily dosage of the compound of the general formula or the type of pharmaceutically acceptable salt can be verified according to the traditional treatment plan.
  • the present invention may contain the compound of the general formula, and its pharmaceutically acceptable salt, hydrate or solvate as the active ingredient, mixed with a pharmaceutically acceptable carrier or excipient to prepare a composition, and prepared into a clinically acceptable dosage form.
  • the derivatives of the present invention can be used in combination with other active ingredients as long as they do not produce other adverse effects such as allergic reactions and the like.
  • the compound of the present invention can be used as the only active ingredient, and can also be used in combination with other drugs for treating diseases related to tyrosine kinase activity. Combination therapy is achieved by the simultaneous, separate or sequential administration of the individual therapeutic components.
  • alkyl refers to a saturated aliphatic hydrocarbon group, which is a straight or branched chain group containing 1 to 20 carbon atoms, preferably an alkyl group containing 1 to 12 carbon atoms, more preferably 1 to 6 carbon atoms atom of the alkyl group.
  • Non-limiting examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1,1-dimethylpropyl, 1 ,2-Dimethylpropyl, 2,2-Dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2- Methylpropyl, 1,1,2-trimethylpropyl, 1,1-dimethylbutyl, 1,2-dimethylbutyl, 2,2-dimethylbutyl, 1,3 -Dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2,3-dimethylbutyl, n-heptyl, 2 -Methylhexyl, 3-methylhexyl, 4-methylhe
  • lower alkyl groups containing 1 to 6 carbon atoms include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl Base, n-pentyl, 1,1-dimethylpropyl, 1,2-dimethylpropyl, 2,2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2-methylpropyl, 1,1,2-trimethylpropyl, 1,1-dimethylbutyl, 1,2-dimethyl Dimethylbutyl, 2,2-dimethylbutyl, 1,3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl group, 2,3-dimethylbutyl group, etc.
  • Alkyl groups may be substituted or unsubstituted, and when substituted, substituents may be substituted at any available point of attachment, said substituents being preferably one or more of the following groups independently selected from alkyl radical, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxyl, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkane Oxy group, heterocycloalkoxy group, cycloalkylthio group, heterocycloalkylthio group, oxo group, carboxyl group or carboxylate group.
  • alkenyl means an alkyl group as defined above consisting of at least two carbon atoms and at least one carbon-carbon double bond, for example vinyl, 1-propenyl, 2-propenyl, 1-, 2- or 3- -butenyl etc.
  • Alkenyl groups may be substituted or unsubstituted, and when substituted, the substituents are preferably one or more of the following groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkylthio, Alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycle Alkylthio.
  • alkynyl refers to an alkyl group as defined above consisting of at least two carbon atoms and at least one carbon-carbon triple bond, eg ethynyl, propynyl, butynyl and the like.
  • Alkynyl groups may be substituted or unsubstituted, and when substituted, the substituents are preferably one or more of the following groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkylthio, Alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycle Alkylthio.
  • cycloalkyl refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent, the cycloalkyl ring containing 3 to 20 carbon atoms, preferably containing 3 to 12 carbon atoms, more preferably containing 3 to 10 carbon atoms, more preferably contain 3 to 8 carbon atoms, most preferably contain 3 to 6 carbon atoms.
  • Non-limiting examples of monocyclic cycloalkyls include cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cycloheptatriene Base, cyclooctyl, etc.; polycyclic cycloalkyl includes spiro ring, fused ring and bridged ring cycloalkyl.
  • spirocycloalkyl refers to a polycyclic group of 5 to 20 membered monocyclic rings sharing one carbon atom (called a spiro atom), which may contain one or more double bonds, but none of the rings has complete conjugation The ⁇ -electron system. Preferably it is 6 to 14 yuan, more preferably 7 to 10 yuan. According to the number of spiro atoms shared between the rings, the spirocycloalkyl group can be divided into single spirocycloalkyl, double spirocycloalkyl or polyspirocycloalkyl, preferably single spirocycloalkyl and double spirocycloalkyl.
  • spirocycloalkyl groups include:
  • fused cycloalkyl refers to a 5 to 20 membered all-carbon polycyclic group in which each ring of the system shares an adjacent pair of carbon atoms with other rings in the system, wherein one or more rings may contain one or Multiple double bonds, but none of the rings have a fully conjugated ⁇ -electron system.
  • it is 6 to 14 yuan, more preferably 7 to 10 yuan.
  • it can be divided into bicyclic, tricyclic, tetracyclic or polycyclic condensed cycloalkyl groups, preferably bicyclic or tricyclic, more preferably 5-membered/5-membered or 5-membered/6-membered bicycloalkyl groups.
  • fused cycloalkyl groups include:
  • bridged cycloalkyl refers to a 5 to 20 membered, all-carbon polycyclic group having any two rings sharing two carbon atoms not directly attached, which may contain one or more double bonds, but none of the rings has a complete Conjugated ⁇ -electron systems. Preferably it is 6 to 14 yuan, more preferably 7 to 10 yuan. According to the number of constituent rings, it can be divided into bicyclic, tricyclic, tetracyclic or polycyclic bridged cycloalkyl groups, preferably bicyclic, tricyclic or tetracyclic, more preferably bicyclic or tricyclic.
  • bridged cycloalkyl groups include:
  • the cycloalkyl ring may be fused to an aryl, heteroaryl or heterocycloalkyl ring where the ring bonded to the parent structure is a cycloalkyl, non-limiting examples include indanyl, tetrahydronaphthalene base, benzocycloheptyl, etc.
  • Cycloalkyl groups may be optionally substituted or unsubstituted, and when substituted, the substituents are preferably one or more of the following groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkane Thio, alkylamino, halogen, mercapto, hydroxyl, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio , heterocycloalkylthio, oxo, carboxyl or carboxylate.
  • heterocyclyl refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent comprising 3 to 20 ring atoms, one or more of which is selected from nitrogen, oxygen or S(O) m (where m is an integer from 0 to 2), but excluding ring portions of -OO-, -OS- or -SS-, the remaining ring atoms being carbon.
  • ring atoms Preferably contain 3 to 12 ring atoms, of which 1 to 4 are heteroatoms; most preferably contain 3 to 8 ring atoms, of which 1 to 3 are heteroatoms; most preferably contain 5 to 7 ring atoms, of which 1 to 2 or 1 to 3 are heteroatoms.
  • Non-limiting examples of monocyclic heterocyclyl groups include pyrrolidinyl, imidazolidinyl, tetrahydrofuranyl, tetrahydrothiophenyl, dihydroimidazolyl, dihydrofuranyl, dihydropyrazolyl, dihydropyrrolyl, piperidine group, piperazinyl, morpholinyl, thiomorpholinyl, homopiperazinyl, pyranyl, etc., preferably 1, 2, 5-oxadiazolyl, pyranyl or morpholinyl.
  • Polycyclic heterocyclyls include spiro, fused and bridged heterocyclyls.
  • spiroheterocyclyl refers to a polycyclic heterocyclic group that shares one atom (called a spiro atom) between 5 to 20-membered monocyclic rings, wherein one or more ring atoms are selected from nitrogen, oxygen or S(O ) m (wherein m is an integer from 0 to 2), the remaining ring atoms are carbon. It may contain one or more double bonds, but none of the rings has a fully conjugated pi-electron system. Preferably it is 6 to 14 yuan, more preferably 7 to 10 yuan.
  • the spiroheterocyclyl can be divided into single spiroheterocyclyl, double spiroheterocyclyl or polyspiroheterocyclyl, preferably single spiroheterocyclyl and double spiroheterocyclyl. More preferably, it is a 4-membered/4-membered, 4-membered/5-membered, 4-membered/6-membered, 5-membered/5-membered or 5-membered/6-membered monospiro heterocyclic group.
  • spiroheterocyclyls include:
  • fused heterocyclyl refers to a 5 to 20 membered polycyclic heterocyclic group in which each ring in the system shares an adjacent pair of atoms with other rings in the system, and one or more rings may contain one or more double bond, but none of the rings has a fully conjugated ⁇ -electron system, where one or more ring atoms are heteroatoms selected from nitrogen, oxygen, or S(O) m (where m is an integer from 0 to 2), and the remaining ring
  • the atom is carbon.
  • it is 6 to 14 yuan, more preferably 7 to 10 yuan.
  • bicyclic, tricyclic, tetracyclic or polycyclic fused heterocyclic groups preferably bicyclic or tricyclic, more preferably 5-membered/5-membered or 5-membered/6-membered bicyclic fused heterocyclic groups.
  • fused heterocyclic groups include:
  • bridged heterocyclyl refers to a 5 to 14 membered polycyclic heterocyclic group in which any two rings share two atoms not directly attached, which may contain one or more double bonds, but none of the rings has a complete shared bond.
  • it is 6 to 14 yuan, more preferably 7 to 10 yuan.
  • bridged heterocyclyl groups include:
  • the heterocyclyl ring may be fused to an aryl, heteroaryl, or cycloalkyl ring where the ring bonded to the parent structure is a heterocyclyl, non-limiting examples of which include:
  • Heterocyclic groups may be optionally substituted or unsubstituted, and when substituted, the substituents are preferably one or more of the following groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alk Thio, alkylamino, halogen, mercapto, hydroxyl, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio , heterocycloalkylthio, oxo, carboxyl or carboxylate.
  • aryl refers to a 6 to 14 membered all-carbon monocyclic or fused polycyclic (that is, rings sharing adjacent pairs of carbon atoms) group, preferably 6 to 10 membered, having a conjugated pi-electron system, such as benzene base and naphthyl. Phenyl is more preferred.
  • the aryl ring may be fused to a heteroaryl, heterocyclyl or cycloalkyl ring, where the ring bonded to the parent structure is an aryl ring, non-limiting examples of which include:
  • Aryl groups may be substituted or unsubstituted, and when substituted, the substituents are preferably one or more of the following groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, Alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycle Alkylthio, carboxyl or carboxylate.
  • heteroaryl refers to a heteroaromatic system comprising 1 to 4 heteroatoms, 5 to 14 ring atoms, wherein the heteroatoms are selected from oxygen, sulfur and nitrogen.
  • Heteroaryl is preferably 5 to 10 membered, containing 1 to 3 heteroatoms; more preferably 5 or 6 membered, containing 1 to 2 heteroatoms; preferred examples are imidazolyl, furyl, thienyl, thiazolyl, pyryl Azolyl, oxazolyl, pyrrolyl, tetrazolyl, pyridyl, pyrimidinyl, thiadiazole, pyrazinyl, etc., preferably imidazolyl, thiazolyl, pyrazolyl or pyrimidinyl, thiazolyl; more preferably pyrazolyl or thiazolyl.
  • the heteroaryl ring may be fused to an aryl, heterocyclyl or cycloalkyl
  • Heteroaryl groups may be optionally substituted or unsubstituted, and when substituted, the substituents are preferably one or more of the following groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkane Thio, alkylamino, halogen, mercapto, hydroxyl, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio , heterocycloalkylthio, carboxyl or carboxylate.
  • alkoxy refers to -O-(alkyl) and -O-(unsubstituted cycloalkyl), wherein alkyl is as defined above.
  • alkoxy include: methoxy, ethoxy, propoxy, butoxy, cyclopropoxy, cyclobutoxy, cyclopentyloxy, cyclohexyloxy.
  • Alkoxy may be optionally substituted or unsubstituted, and when substituted, the substituent is preferably one or more of the following groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkoxy Thio, alkylamino, halogen, mercapto, hydroxyl, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio , heterocycloalkylthio, carboxyl or carboxylate.
  • the substituent is preferably one or more of the following groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkoxy Thio, alkylamino, halogen, mercapto, hydroxyl, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl,
  • haloalkyl refers to an alkyl group substituted with one or more halo, wherein alkyl is as defined above.
  • haloalkoxy refers to an alkoxy group substituted with one or more halogens, wherein alkoxy group is as defined above.
  • hydroxyalkyl refers to an alkyl group substituted with one or more hydroxy groups, wherein alkyl is as defined above.
  • hydroxyl refers to a -OH group.
  • halogen refers to fluorine, chlorine, bromine or iodine.
  • amino refers to -NH2 .
  • cyano refers to -CN.
  • nitro refers to -NO2 .
  • mercapto refers to -SH.
  • ester group refers to -C(O)O(alkyl) or -C(O)O(cycloalkyl), wherein alkyl and cycloalkyl are as defined above.
  • acyl refers to compounds containing the group -C(O)R, where R is alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl.
  • Optional or “optionally” means that the subsequently described event or circumstance can but need not occur, and that the description includes instances where the event or circumstance occurs or does not occur.
  • a heterocyclic group optionally substituted with an alkyl group means that an alkyl group may but need not be present, and the description includes cases where the heterocycle group is substituted with an alkyl group and cases where the heterocycle group is not substituted with an alkyl group .
  • Substituted means that one or more hydrogen atoms in a group, preferably up to 5, more preferably 1 to 3 hydrogen atoms are independently substituted by the corresponding number of substituents. It goes without saying that substituents are only in their possible chemical positions and that a person skilled in the art can determine (by experiment or theory) possible or impossible substitutions without undue effort. For example, an amino or hydroxyl group with free hydrogen may be unstable when bonded to a carbon atom with an unsaturated (eg, ethylenic) bond.
  • “Pharmaceutical composition” means a mixture containing one or more compounds described herein, or a physiologically/pharmaceutically acceptable salt or prodrug thereof, and other chemical components, and other components such as a physiologically/pharmaceutically acceptable carrier and excipients.
  • the purpose of the pharmaceutical composition is to promote the administration to the organism, facilitate the absorption of the active ingredient and thus exert biological activity.
  • “Pharmaceutically acceptable salt” refers to the salt of the compound of the present invention, which is safe and effective when used in mammals, and has proper biological activity.
  • the present invention adopts following synthetic scheme to prepare general formula (I) compound of the present invention.
  • the compound of formula A1 is reacted with a nucleophilic amine to obtain a compound of formula A2; then, under heating conditions, the compound of formula A2 is combined with 2,4-dimethyl Oxybenzylamine reaction to obtain the compound of formula A3; the compound of formula A3 can be cross-coupling reaction (such as Suzuki coupling) by metal catalysis (such as Pd(PPh 3 ) 4 , K 2 CO 3 , Cs 2 CO 3 ) Reaction with boric acid or boric acid pinacol ester compound to obtain a compound of formula A4; deprotection with a suitable acid (such as trifluoroacetic acid) to obtain a compound shown in general formula (III-1);
  • a suitable base such as DIEA
  • R 1 , R 3 , R 4 , R 5 , R 6 are as defined in general formula (III-1).
  • the compound of formula A6 reacts with a nucleophilic amine in the presence of a condensing agent (such as BOP) to obtain a compound of the coupled product formula A7;
  • a condensing agent such as BOP
  • the compound of formula A7 can be catalyzed by a metal (such as Pd(PPh 3 ) 4 , K 2 CO 3 , Cs 2 CO 3 ) cross-coupling reaction (such as Suzuki coupling) reacts with boric acid or boric acid pinacol ester compound to obtain the compound shown in general formula (III-1);
  • R 1 , R 3 , R 4 , R 5 , R 6 are as defined in general formula (III-1).
  • the compound of formula B1 is reacted with a nucleophilic amine to obtain a compound of formula B2; then, under heating conditions, the compound of formula B2 is combined with 2,4-dimethyl Oxybenzylamine reaction to obtain the compound of formula B3; the compound of formula B3 can be cross-coupling reaction (such as Suzuki coupling) by metal catalysis (such as Pd(PPh 3 ) 4 , K 2 CO 3 , Cs 2 CO 3 ) Reaction with boric acid or boric acid pinacol ester compound to obtain a compound of formula B4; deprotection with a suitable acid (such as trifluoroacetic acid) to obtain a compound shown in general formula (IV-1);
  • a suitable base such as DIEA
  • R 1 , R 2 , R 4 , R 5 , R 6 are as defined in general formula (IV-1).
  • the compound of formula B6 reacts with a nucleophilic amine in the presence of a condensing agent (such as BOP) to obtain a compound of the coupled product formula B7;
  • a condensing agent such as BOP
  • the compound of formula B7 can be catalyzed by a metal (such as Pd(PPh 3 ) 4 , K 2 CO 3 , Cs 2 CO 3 ) cross-coupling reaction (such as Suzuki coupling) reacts with boric acid or boric acid pinacol ester compound to obtain the compound shown in general formula (IV-1);
  • R 1 , R 2 , R 4 , R 5 , R 6 are as defined in general formula (IV-1).
  • the compound of formula C1 reacts with a nucleophilic amine to obtain a compound of formula C2; then, under heating conditions, the compound of formula C2 reacts with 2,4-dimethyl Oxybenzylamine reaction to give the compound of formula C3; the compound of formula C3 can be cross-coupling reaction (such as Suzuki coupling) by metal catalysis (such as Pd(PPh 3 ) 4 , K 2 CO 3 , Cs 2 CO 3 ) Reaction with boric acid or boric acid pinacol ester compound to obtain a compound of formula C4; deprotection with a suitable acid (such as trifluoroacetic acid) to obtain a compound shown in general formula (V-1);
  • a suitable base such as DIEA
  • R 2 , R 4 , R 5 , R 6 are as defined in general formula (V-1).
  • the compound of formula C6 reacts with a nucleophilic amine in the presence of a condensing agent (such as BOP) to obtain a compound of the coupled product formula C7;
  • a condensing agent such as BOP
  • the compound of formula C7 can be catalyzed by a metal (such as Pd(PPh 3 ) 4 , K 2 CO 3 , Cs 2 CO 3 ) cross-coupling reaction (such as Suzuki coupling) reacts with boric acid or boric acid pinacol ester compound to obtain the compound shown in general formula (V-1);
  • R 2 , R 4 , R 5 , R 6 are as defined in general formula (V-1).
  • Figure 1 shows the parameters of the compound of Example 52 in the serum of cynomolgus monkeys.
  • the compounds of the present invention are prepared utilizing convenient starting materials and general preparative procedures.
  • the present invention gives typical or preferred reaction conditions, such as reaction temperature, time, solvent, pressure, molar ratio of reactants. But unless otherwise specified, other reaction conditions can also be adopted. Optimum conditions may vary with specific reactants or solvents used, but in general, reaction optimization steps and conditions can be identified.
  • protecting groups may be used in the present invention to protect certain functional groups from unnecessary reactions.
  • Suitable protecting groups for various functional groups and their protection or deprotection conditions are widely known to those skilled in the art.
  • Protecting Groups in Organic Preparations by T.W. Greene and G.M. Wuts (3rd Edition, Wiley, New York, 1999 and citations in the book) describes in detail the protection or deprotection of a large number of protecting groups.
  • the separation and purification of compounds and intermediates takes appropriate methods and steps according to specific needs, such as filtration, extraction, distillation, crystallization, column chromatography, preparative thin-layer plate chromatography, preparative high-performance liquid chromatography or a combination of the above methods.
  • For its specific usage method please refer to the examples described in the present invention.
  • other similar separation and purification means can also be used. They can be characterized using conventional methods, including physical constants and spectral data.
  • NMR nuclear magnetic resonance
  • MS mass spectroscopy
  • MS was determined by LC (Waters 2695)/MS (Quattro Premier x E) mass spectrometer (manufacturer: Waters) (Photodiode Array Detector).
  • the lc6000 high performance liquid chromatograph (manufacturer: Innovation Tongheng) was used for the preparative liquid chromatography.
  • Chromatographic column is Daisogel C18 10 ⁇ m 100A (30mm ⁇ 250mm), mobile phase: acetonitrile/water.
  • TLC Thin-layer chromatography
  • Silica gel column chromatography uses Qingdao ocean silica gel 100-200 mesh, 200-300 mesh and 300-400 mesh silica gel as the carrier.
  • the known starting materials of the present invention can be adopted or synthesized according to methods known in the art, or can be purchased from Wanghua Mall, Beijing Coupling, Sigma, Bailingwei, Yi Shiming, Shanghai Shuya, Shanghai Yinuokai, Anaiji Chemical, Shanghai Biide and other companies.
  • the reactions can all be carried out under a nitrogen atmosphere.
  • the argon atmosphere or nitrogen atmosphere means that the reaction bottle is connected to an argon or nitrogen balloon with a volume of about 1 L.
  • Reaction solvents organic solvents or inert solvents are each expressed as the solvent used does not participate in the reaction under the described reaction conditions, including, such as benzene, toluene, acetonitrile, tetrahydrofuran (THF), dimethylformamide (DMF), chloroform , dichloromethane, ether, methanol, nitrogen-methylpyrrolidinone (NMP), pyridine, etc.
  • the solution refers to an aqueous solution.
  • the chemical reactions described in the present invention are generally carried out under normal pressure.
  • the reaction temperature is between -78°C and 200°C.
  • the reaction time and conditions are, for example, between -78°C and 200°C under one atmospheric pressure, and the reaction is completed within about 1 to 24 hours. If the reaction is overnight, the reaction time is generally 16 hours. Unless otherwise specified in the examples, the reaction temperature is room temperature, which is 20°C to 30°C.
  • the reaction solution was concentrated under reduced pressure, the resulting brown liquid was added to ice water (1 L), the resulting solid was removed by filtration, extracted by adding ethyl acetate (3 ⁇ 500 mL), the combined organic phases were washed with saturated brine (500 mL), and anhydrous Dry over sodium sulfate, filter, and concentrate the filtrate under reduced pressure.
  • reaction solution was cooled to room temperature, diluted with ice water (30 mL), extracted quickly with ethyl acetate (3 ⁇ 8 mL), the organic phases were combined, washed with saturated brine (30 mL), and dried over anhydrous sodium sulfate to obtain The ethyl acetate solution of compound 1g was directly used in the next reaction.
  • Step 7 (R)-N 4 -(1-((tert-butyldimethylsilyl)oxy)-2-methylhex-2-yl)-N 2 -(2,4-dimethoxy Preparation of benzyl)pyrido[4,3-d]pyrimidine-2,4-diamine (1i)
  • Step 8 Preparation of (R)-2-((2-aminopyrido[4,3-d]pyrimidin-4-yl)amino)-2-methylhexan-1-ol (1)
  • the obtained crude product was separated and purified by preparative chromatographic column (column type: XBridge Shield RP18 OBD column, 5um, 19*150mm; mobile phase A: water (10mmol/L ammonium bicarbonate), mobile phase B: acetonitrile; flow rate: 25mL/min ; Gradient: 18%-35% acetonitrile in 8 minutes; detection wavelength: 220 nm) to obtain compound 1 (3.4 mg, 44%) as a white solid.
  • preparative chromatographic column column (column type: XBridge Shield RP18 OBD column, 5um, 19*150mm; mobile phase A: water (10mmol/L ammonium bicarbonate), mobile phase B: acetonitrile; flow rate: 25mL/min ; Gradient: 18%-35% acetonitrile in 8 minutes; detection wavelength: 220 nm) to obtain compound 1 (3.4 mg, 44%) as a white solid.
  • compound 2b (961mg, 5.60mmol) was dissolved in N,N-dimethylformamide (DMF) (10mL), and N,N-carbonyldiimidazole (CDI) (3.63g, 22.39mmol) was added successively and 1,8-diazabicycloundec-7-ene (DBU) (2.13 g, 13.99 mmol), and the reaction solution was stirred at 80° C. for 1 hour.
  • DMF N,N-dimethylformamide
  • DBU 1,8-diazabicycloundec-7-ene
  • compound 2d (540 mg, 2.30 mmol) was dissolved in 1,4-dioxane (5 mL), and (R)-1-((tert-butyldimethylsilyl)oxy)-2 was added successively -Methylhexan-2-amine 1e (1.13g, 4.61mmol) and N,N-diisopropylethylamine (2.98g, 23.03mmol), react at room temperature for 1.5 hours.
  • Step 5 (R)-N 4 -(1-((tert-butyldimethylsilyl)oxy)-2-methylhex-2-yl)-7-chloro-N 2 -(2,4 Preparation of -dimethoxybenzyl)pyrido[4,3-d]pyrimidine-2,4-diamine (2f)
  • Step 6 (R)-N 4 -(1-((tert-butyldimethylsilyl)oxy)-2-methylhex-2-yl)-N 2 -(2,4-dimethoxy Preparation of benzyl)-7-(2-(4-methylpiperazin-1-yl)pyrimidin-5-yl)pyrido[4,3-d]pyrimidine-2,4-diamine (2g)
  • compound 2f (204 mg, 0.36 mmol) was dissolved in 1,4-dioxane (2 mL) and water (0.4 mL), and 2-(4-methylpiperazin-1-yl) pyrimidine- 5-Boronic acid pinacol ester (CAS: 942922-07-8) (2h) (264mg, 1.05mmol), potassium carbonate (148mg, 1.07mmol) and [1,1'-bis(diphenylphosphoryl) di Ferrocene]palladium dichloride (Pd(dppf) Cl2 ) (26mg, 0.036mmol). The reaction was stirred overnight at 95°C under a nitrogen atmosphere.
  • Step 7 (R)-2-((2-Amino-7-(2-(4-methylpiperazin-1-yl)pyrimidin-5-yl)pyrido[4,3-d]pyrimidine-4 Preparation of -yl)amino)-2-methylhexan-1-alcohol (2)
  • the preparation method is the same as in Example 2, except that the compound 2-(4-methylpiperazin-1-yl)pyrimidine-5-boronic acid pinacol ester (2h) is replaced by 2-(4-methylpiperazine- 1-yl) pyridine-5-boronic acid pinacol ester (CAS: 918524-63-7) (3b), the obtained crude product was separated and purified by preparative chromatographic column (column type: SunFire Prep C18 OBD column, 5um, 19*150mm ; Mobile phase A: water (0.1% formic acid), mobile phase B: acetonitrile; flow rate: 25mL/min; gradient: 6%-17% acetonitrile in 8 minutes; detection wavelength: 220nm), obtain the formic acid of compound 3 Salt.
  • reaction solution was diluted with water (20mL), and then extracted with ethyl acetate (2 ⁇ 20mL).
  • the resulting aqueous phase was concentrated under reduced pressure, and the crude product was separated and purified by reverse-phase column chromatography (column type: Agel C18 column; mobile phase A: water (0.1% formic acid), mobile phase B: acetonitrile; flow rate: 60mL/min; gradient: 5%-20% acetonitrile in 10 minutes; detection wavelength: 220/254nm), to obtain off-white Solid compound 4b (370mg, 85.04%).
  • the preparation method is the same as in Example 2, except that the compound 2-(4-methylpiperazin-1-yl)pyrimidine-5-boronic acid pinacol ester (2h) is replaced by 6-(pyrrolidin-1-yl) Pyridine-3-boronic acid (CAS: 1150114-75-2) (5b), the obtained crude product was separated and purified by preparative chromatographic column (column type: SunFire Prep C18 OBD column, 5um, 19*150mm; mobile phase A: water (0.1 % formic acid), mobile phase B: acetonitrile; flow rate: 25mL/min; gradient: 7%-21% acetonitrile in 8 minutes; detection wavelength: 220nm), the formate salt of compound 5 was prepared.
  • preparative chromatographic column column (column type: SunFire Prep C18 OBD column, 5um, 19*150mm; mobile phase A: water (0.1 % formic acid), mobile phase B: acetonitrile; flow rate: 25mL/min; gradient:
  • the preparation method is the same as in Example 2, except that the compound 2-(4-methylpiperazin-1-yl)pyrimidine-5-boronic acid pinacol ester (2h) is replaced by 2-(piperidin-1-yl) Pyridine-5-boronic acid pinacol ester (CAS: 852228-08-1) (6b), the obtained crude product was separated and purified by preparative chromatographic column (column type: SunFire Prep C18 OBD column, 5um, 19*150mm; mobile phase A : water (0.1% formic acid), mobile phase B: acetonitrile; flow rate: 25mL/min; gradient: 13%-29% acetonitrile in 8 minutes; detection wavelength: 220nm), the formate salt of compound 6 was obtained.
  • preparative chromatographic column column (column type: SunFire Prep C18 OBD column, 5um, 19*150mm; mobile phase A : water (0.1% formic acid), mobile phase B: acetonitrile; flow rate: 25mL
  • the preparation method is the same as in Example 2, except that the compound 2-(4-methylpiperazin-1-yl)pyrimidine-5-boronic acid pinacol ester (2h) is replaced by 6-(4-hydroxypiperidine-1 -yl) pyridine-3-boronic acid pinacol ester (CAS: 1251948-86-3) (7b), the obtained crude product was separated and purified by preparative chromatographic column (column type: XBridge Shield RP18 OBD column, 5um, 19*150mm; Mobile phase A: water (0.1% formic acid), mobile phase B: acetonitrile; flow rate: 25mL/min; gradient: 10%-32% acetonitrile in 8 minutes; detection wavelength: 254nm), obtained the formic acid of compound 7 Salt.
  • the preparation method is the same as in Example 2, except that the compound 2-(4-methylpiperazin-1-yl)pyrimidine-5-boronic acid pinacol ester (2h) is replaced by imidazo[1,2-a]pyridine -6-boronic acid pinacol ester (CAS: 1204742-76-6) (8b), the obtained crude product was separated and purified by preparative chromatographic column (column type: SunFire Prep C18 OBD column, 5um, 19*150mm; mobile phase A: Water (0.1% formic acid), mobile phase B: acetonitrile; flow rate: 25mL/min; gradient: 7%-15% acetonitrile in 8 minutes; detection wavelength: 254nm), the formate salt of compound 8 was obtained.
  • preparative chromatographic column column (column type: SunFire Prep C18 OBD column, 5um, 19*150mm; mobile phase A: Water (0.1% formic acid), mobile phase B: acetonitrile; flow rate: 25mL/min; gradient
  • 6-chloropyridine-3-boronic acid pinacol ester 50 mg, 0.209 mmol was dissolved in dimethyl sulfoxide (1 mL), and 1-cyclopropylpiperazine (52.69 mg, 0.418 mmol), stirred at 150° C. for 3 hours under nitrogen atmosphere. After the reaction was complete, add water (20 mL) to quench the reaction, add ethyl acetate (3 ⁇ 20 mL) to the system for extraction, wash the combined organic phase with saturated brine (40 mL), dry over anhydrous sodium sulfate, filter, and concentrate the filtrate under reduced pressure , to obtain brown solid compound 9d (72 mg, crude product), which was directly used in the next step without purification.
  • Step 4 (R)-2-((2-Amino-7-(6-(4-cyclopropylpiperazin-1-yl)pyridin-3-yl)pyrido[4,3-d]pyrimidine- Preparation of 4-yl)amino)-2-methylhexan-1-ol (9)
  • the preparation method was the same as in Example 9, except that 1-cyclopropylpiperazine was replaced by 1-methylpiperazin-2-one, to prepare compound 10.
  • the preparation method was the same as that of Example 9, except that the compound 1-cyclopropylpiperazine was replaced by 1-ethylpiperazine to prepare compound 11.
  • the preparation method was the same as that of Example 9, except that compound 1-cyclopropylpiperazine was replaced by 1-isopropylpiperazine to prepare compound 12.
  • the preparation method was the same as that of Example 9, except that the compound 1-cyclopropylpiperazine was replaced by 1-(2-methoxyethyl)piperazine to prepare compound 13.
  • the preparation method was the same as in Example 9, except that the compound 1-cyclopropylpiperazine was replaced by 1,2-dimethylpiperazine to prepare compound 14.
  • the preparation method is the same as in Example 9, except that the compound 6-chloropyridine-3-boronic acid pinacol ester is replaced by 4-methyl-6-chloropyridine-3-boronic acid pinacol ester, and 1-cyclopropane Substituting 1-methylpiperazine for 1-methylpiperazine yielded compound 15.
  • the preparation method is the same as in Example 9, except that the compound 6-chloropyridine-3-boronic acid pinacol ester is replaced by 2-methyl-6-chloropyridine-3-boronic acid pinacol ester, and 1-cyclopropane Substituting 1-methylpiperazine for 1-methylpiperazine yielded compound 16.
  • reaction solution was diluted with water (10 mL), and then extracted with ethyl acetate (2 ⁇ 10 mL), and the obtained aqueous phase was concentrated under reduced pressure to obtain brown solid compound 17c (150 mg, crude product), which was directly used in the following step.
  • the preparation method was the same as in Example 9, except that 1-cyclopropylpiperazine was replaced by 4-dimethylaminopiperidine, to obtain compound 18.
  • the preparation method was the same as in Example 9, except that 1-cyclopropylpiperazine was replaced by N-hydroxyethylpiperazine to prepare compound 19.
  • the preparation method was the same as in Example 20, except that 1-n-propylpiperazine dibromide was replaced by 1-(2,2-difluoroethyl)piperazine hydrochloride to prepare compound 21.
  • the preparation method is the same as in Example 22, except that 1-(2-fluoroethyl)piperazine dihydrochloride is replaced by 1-(2,2,2-trifluoroethyl)piperazine dihydrochloride to obtain Compound 23.
  • Compound 24 was prepared in the same manner as in Example 22, except that 1-(2-fluoroethyl)piperazine dihydrochloride was replaced by 4-n-propylpiperidine hydrochloride.
  • Compound 25 was prepared in the same manner as in Example 22, except that 1-(2-fluoroethyl)piperazine dihydrochloride was replaced by 3-ethoxyazetidine hydrochloride.
  • Compound 26 was prepared in the same manner as in Example 22, except that 1-(2-fluoroethyl)piperazine dihydrochloride was replaced by 4-ethoxypiperidine.
  • the preparation method was the same as in Example 22, except that 1-(2-fluoroethyl)piperazine dihydrochloride was replaced by N,N-dipropylazetidin-3-amine to prepare compound 27.
  • the preparation method was the same as in Example 9, except that 1-cyclopropylpiperazine was replaced by 4-diethylaminopiperidine, to obtain compound 28.
  • Compound 29 was prepared in the same manner as in Example 22, except that 1-(2-fluoroethyl)piperazine dihydrochloride was replaced by 3-(diethylamino)azetidine dihydrochloride.
  • Compound 31 was prepared in the same manner as in Example 30, except that 1-(2-fluoroethyl)piperazine dihydrochloride was replaced by 1-n-propylpiperazine dibromide.
  • Compound 32 was prepared in the same manner as in Example 30, except that 1-(2-fluoroethyl)piperazine dihydrochloride was replaced by 1-isopropylpiperazine.
  • Compound 33 was prepared in the same manner as in Example 30, except that 1-(2-fluoroethyl)piperazine dihydrochloride was replaced by 4-dimethylaminopiperidine.
  • the preparation method is the same as that of Example 9, except that 2-(4-cyclopropylpiperazin-1-yl)pyridine-5-boronic acid pinacol ester (9d) is replaced by (1-methylpiperidine-4 -yl)-1H-pyrazole-4-boronic acid pinacol ester (CAS: 1323919-64-7) (35a), to obtain compound 35.
  • the preparation method is the same as in Example 9, except that 2-(4-cyclopropylpiperazin-1-yl)pyridine-5-boronic acid pinacol ester (9d) is replaced by 1-(tetrahydropyran-4- base)-1H-pyrazole-4-boronic acid pinacol ester (CAS: 1040377-03-4) (36a), to obtain compound 36.
  • Step 1 (R)-4-(5-(2-amino-4-((1-hydroxy-2-methylhex-2-yl)amino)pyrido[4,3-d]pyrimidine-7- yl)pyridin-2-yl)piperazine-1-carboxylate tert-butyl ester (38b)
  • Step 2 (R)-2-((2-Amino-7-(6-(piperazin-1-yl)pyridin-3-yl)pyrido[4,3-d]pyrimidin-4-yl)amino ) Preparation of -2-methylhexan-1-ol (38)
  • 3-pyridineboronic acid 500.09 mg, 4.069 mmol
  • 5-bromo-2-iodopyridine 1050 mg, 3.699 mmol
  • potassium carbonate 1533.49mg, 11.097mmol
  • tetrakis(triphenylphosphine)palladium 427.39mg, 0.37mmol
  • reaction solution was diluted with water (30 mL), then extracted with ethyl acetate (3 ⁇ 30 mL), the combined organic phases were washed with saturated brine (30 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure.
  • the preparation method was the same as in Example 39, except that 3-pyridineboronic acid was replaced by 4-pyridineboronic acid to prepare compound 40.
  • the preparation method was the same as that of Example 39, except that compound 3-pyridineboronic acid was replaced by 6-methylpyridine-3-boronic acid to prepare compound 41.
  • the preparation method was the same as that of Example 39, except that compound 5-bromo-2,3'-bipyridine (39a) was replaced by 5-bromo-2,2'-bipyridine (42a), to prepare compound 42.
  • Step 1 (R)-N 4 -(1-((tert-butyldimethylsilyl)oxy)-2-methylhex-2-yl)-7-(4-chlorobenzyl)-N
  • 2- (2,4-dimethoxybenzyl)pyrido[4,3-d]pyrimidine-2,4-diamine 43a
  • compound 2f 150 mg, 0.26 mmol was dissolved in 1,4-dioxane (2 mL) and water (0.5 mL), and 4-chlorobenzylboronic acid pinacol ester (CAS: 475250-49 -8) (43b) (264mg, 1.05mmol), potassium carbonate (255mg, 1.83mmol) and tetrakis(triphenylphosphine)palladium (60mg, 0.052mmol). The reaction solution was stirred at 95° C. for 3 hours under nitrogen atmosphere.
  • Step 2 (R)-N 4 -(1-((tert-butyldimethylsilyl)oxy)-2-methylhex-2-yl)-N 2 -(2,4-dimethoxy Preparation of benzyl)-7-(4-(pyrrolidin-1-ylmethyl)benzyl)pyrido[4,3-d]pyrimidine-2,4-diamine (43c)
  • compound 43a (92 mg, 0.14 mmol) was dissolved in 1,4-dioxane (2 mL) and water (0.5 mL), and (1-pyrrolidinylmethyl) potassium trifluoroborate (37 mg, 0.19mmol), potassium carbonate (57.8mg, 0.41mmol) and XPhos Pd G3 (11.7mg, 0.014mmol). The reaction solution was stirred overnight at 100° C. under a nitrogen atmosphere.
  • Step 3 (R)-2-((2-Amino-7-(4-(pyrrolidin-1-ylmethyl)benzyl)pyrido[4,3-d]pyrimidin-4-yl)amino) - Preparation of 2-methylhexan-1-ol (43)
  • Step 2 Preparation of N 4 -propylpyrido[3,4-d]pyrimidine-2,4-diamine (44)
  • compound 44b (40mg, 0.247mmol) was dissolved in N,N-dimethylformamide (3mL), and Carter’s condensing agent (BOP) (218mg, 0.494mmol), 1,8-diazepine Dicycloundec-7-ene (DBU) (75.1 mg, 0.494 mmol), n-propylamine (29.2 mg, 0.494 mmol), and the reaction liquid were reacted at room temperature for 4 hours under nitrogen atmosphere. After the reaction was completed, it was quenched by adding water (10 mL), and extracted with ethyl acetate (3 ⁇ 20 mL). The organic phases were combined and washed with saturated brine (2 ⁇ 20 mL).
  • BOP condensing agent
  • DBU 1,8-diazepine Dicycloundec-7-ene
  • n-propylamine 29.2 mg, 0.494 mmol
  • the organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure to obtain a crude product.
  • the crude product was purified with a preparative reverse-phase chromatographic column (column type: XBridge Shield RP18 OBD column, 5um, 19*150mm; mobile phase A: water (10mmol/L ammonium bicarbonate), mobile phase B: acetonitrile; flow rate: 25mL/min ; Gradient: 47%-53% acetonitrile in 8 minutes; detection wavelength: 254/220nm) to give compound 44 (10.2 mg, 20.8%) as a white solid.
  • the preparation method was the same as that of Example 44, except that n-propylamine was replaced by n-butylamine, and compound 45 was prepared.
  • Compound 46 was prepared in the same manner as in Example 44, except that n-propylamine was replaced by n-pentylamine.
  • Compound 49 was prepared in the same manner as in Example 44, except that n-propylamine was replaced by (R)-2-amino-2-methylbutan-1-ol (49a).
  • Compound 50 was prepared in the same manner as in Example 44, except that n-propylamine was replaced by (R)-2-amino-2-methylpentan-1-ol (50a).
  • Compound 51 was prepared in the same manner as in Example 44, except that n-propylamine was replaced by (R)-2-amino-2-methylheptan-1-ol (51a).
  • Example 52 and Example 53 (R)-2-((2-aminopyrido[3,4-d]pyrimidin-4-yl)amino)-2-methylhexan-1-ol (52) and Preparation of (S)-2-((2-aminopyrido[3,4-d]pyrimidin-4-yl)amino)-2-methylhexan-1-ol (53)
  • Step 1 (R)-N-((1-((tert-butyldimethylsilyl)oxy)-2-methylhex-2-yl)-2-chloropyrido[3,4-d ] Preparation of pyrimidine-4-amine (52b)
  • 2,4-Dichloropyrido[3,4-d]pyrimidine 52a (100 mg, 0.50 mmol) was dissolved in 1,4-dioxane (2.0 mL) at room temperature.
  • (R)-1-((tert-butyldimethylsilyl)oxy)-2-methylhexan-2-amine (1e) (123mg, 0.50mmol) and N,N-di Isopropylethylamine (0.26 ⁇ L, 1.49 mmol) stirred at room temperature for 2 hours. After the reaction is complete, cool to room temperature. The reaction solution was diluted with water (20 mL), and extracted with ethyl acetate (3 ⁇ 20 mL).
  • Step 2 (R)-N 4 -(1-((tert-butyldimethylsilyl)oxy)-2-methylhex-2-yl)-N 2 -(2,4-dimethoxy Preparation of benzyl)pyrido[3,4-d]pyrimidine-2,4-diamine (52c)
  • Step 3 Preparation of (R)-2-((2-aminopyrido[3,4-d]pyrimidin-4-yl)amino)-2-methylhexan-1-ol (52d)
  • Step 4 (R)-2-((2-Aminopyrido[3,4-d]pyrimidin-4-yl)amino)-2-methylhexan-1-ol (52) and (S)-2 Preparation of -((2-aminopyrido[3,4-d]pyrimidin-4-yl)amino)-2-methylhexan-1-ol (53)
  • 3-chloro-2-fluoroisonicotinic acid (55a) (1.0g, 5.70mmol) was dissolved in N,N-dimethylformamide (15mL), and cesium carbonate (5.57g, 17.1mmol) was added successively ), guanidine carbonate (1.03g, 5.72mmol), cuprous iodide (217mg, 1.14mmol), and the reaction solution was stirred at 110°C for 4 hours under a nitrogen atmosphere.
  • Step 2 (R)-N 4 -(1-((tert-butyldimethylsilyl)oxy)-2-methylhex-2-yl)-8-fluoropyrido[3,4-d ] Preparation of pyrimidine-2,4-diamine (55c)
  • compound 55b (30 mg, 0.167 mmol) was dissolved in N,N-dimethylformamide (5 mL), and (R)-1-((tert-butyldimethylsilyl)oxy) was added successively -2-Methylhex-2-amine (1e) (122mg, 0.50mmol), 1,8-diazabicycloundec-7-ene (76.1mg, 0.50mmol), Carter condensing agent (110mg, 0.25 mmol), and the reaction solution was stirred overnight at room temperature under a nitrogen atmosphere.
  • Step 3 Preparation of (R)-2-((2-amino-8-fluoropyrido[3,4-d]pyrimidin-4-yl)amino)-2-methylhexan-1-ol (55)
  • Aqueous ammonia (30 mL) was added to methyl 3-amino-5-fluoroisonicotinate (56a) (900 mg, 5.29 mmol) at room temperature.
  • the reaction was stirred at 60°C for 3 hours.
  • Step 4 (R)-N-((1-((tert-Butyldimethylsilyl)oxy)-2-methylhex-2-yl)-2-chloro-5-fluoropyrido[3 ,4-d] Preparation of pyrimidin-4-amine (56e)
  • reaction solution was diluted with water (15 mL), extracted with ethyl acetate (3 ⁇ 10 mL), and the combined organic phases were washed with saturated brine (15 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure.
  • Step 5 (R)-N 4 -(1-((tert-butyldimethylsilyl)oxy)-2-methylhex-2-yl)-N 2 -(2,4-dimethoxy Preparation of benzyl)-5-fluoropyrido[3,4-d]pyrimidine-2,4-diamine (56f)
  • Step 6 Preparation of (R)-2-((2-amino-5-fluoropyrido[3,4-d]pyrimidin-4-yl)amino)-2-methylhexan-1-ol (56)
  • the obtained crude product was purified with a preparative chromatographic column (column type: XBridge Shield RP18 OBD column, 5um, 19*150mm; mobile phase A: water (10mmol/L ammonium bicarbonate), mobile phase B: acetonitrile; flow rate: 25mL/min; Gradient: 25%-45% acetonitrile in 8 minutes; detection wavelength: 254/220nm) to give compound 56 (7.30 mg, 43.7%) as a white solid.
  • a preparative chromatographic column column (column type: XBridge Shield RP18 OBD column, 5um, 19*150mm; mobile phase A: water (10mmol/L ammonium bicarbonate), mobile phase B: acetonitrile; flow rate: 25mL/min; Gradient: 25%-45% acetonitrile in 8 minutes; detection wavelength: 254/220nm) to give compound 56 (7.30 mg, 43.7%) as a white solid.
  • 5-Amino-2-fluoro-4-pyridinecarboxylic acid 57a (1.50 g, 9.62 mmol) was dissolved in thionyl chloride (20 mL) at room temperature. The reaction was refluxed at 80°C for 1 hour. After the reaction was complete, it was directly concentrated under reduced pressure, and the resulting brown oily liquid compound was dissolved in tetrahydrofuran (5 mL), and slowly added dropwise into ammonia water (15 mL) at 0°C, and the reaction solution was stirred at 0°C for 1 hour.
  • Step 1 Preparation of 2-fluoro-5-iodo-4-aminopyridine (58b) and 2-fluoro-3-iodo-4-aminopyridine (58c)
  • Step 1 Preparation of 4-amino-6-fluoro-nicotinic acid methyl ester (59a)
  • Step 1 Preparation of 5-amino-2-chloroisonicotinic acid methyl ester (60b)
  • 5-amino-2-chloropyridine-4-carboxylic acid 60a (25 g, 145 mmol) was dissolved in methanol (300 mL), and then thionyl chloride (100 mL) was added dropwise to the above solution (keep the reaction system The inner temperature is 20-25°C), after the dropwise addition, the temperature was raised to 70°C and stirred overnight. The reaction system was cooled to room temperature. The mixture was concentrated under reduced pressure, and the resulting residue was slurried twice with aqueous sodium bicarbonate solution, filtered, and the filter cake was dried to obtain compound 60b (16 g, 59.1%) as a yellow solid.
  • compound 60b (3.0g, 16.1mmol), chloroformamidine hydrochloride (3.7g, 32.2mmol), and dimethyl sulfone (10g) were sequentially added to a 100mL glass bottle, and the reaction solution was heated to 150°C and stirred for 4 Hour. After the reaction was completed, the reaction liquid was diluted with water (100 mL), filtered, and the obtained filter cake was rinsed with water twice, and the obtained filter cake was dried to obtain yellow solid compound 60c (3.7 g, crude product).
  • Step 3 Preparation of 2-((2-amino-6-chloropyridin[3,4-d]pyrimidin-4-yl)amino)hexan-1-ol (60d)
  • Step 4 Preparation of 2-((2-amino-6-(4-chlorobenzyl)pyridin[3,4-d]pyrimidin-4-yl)amino)hexan-1-ol (60e)
  • Step 1 (R)-N 4 -(1-((tert-butyldimethylsilyl)oxy)-2-methylhex-2-yl)-6-chloropyrido[3,4-d ] Preparation of pyrimidine-2,4-diamine (61a)
  • compound 60c 300 mg, 1.52 mmol was mixed with (R)-1-((tert-butyldimethylsilyl)oxy)-2-methylhexan-2-amine (1e) (749 mg, 3.05 mmol) was dissolved in N,N-dimethylformamide (2mL), then Carter condensing agent (810mg, 1.83mmol) and 1,8-diazabicycloundec-7-ene (697mg, 4.57mmol ) into the system in turn. The reaction was stirred at room temperature for 4 hours. After the reaction was completed, the reaction solution was diluted with water (30 mL), and then extracted with ethyl acetate (3 ⁇ 30 mL).
  • Step 2 (R)-N 4 -(1-((tert-butyldimethylsilyl)oxy)-2-methylhex-2-yl)-6-(4-chlorobenzyl)pyrido Preparation of [3,4-d]pyrimidine-2,4-diamine (61b)
  • Step 3 (R)-N 4 -(1-((tert-butyldimethylsilyl)oxy)-2-methylhex-2-yl)-6-(4-(pyrrolidine-1- Preparation of methyl)benzyl)pyrido[3,4-d]pyrimidine-2,4-diamine (61c)
  • the obtained crude product was separated and purified by preparative chromatographic column (column type: XBridge Shield RP18 OBD column, 5um, 19*150mm; mobile phase A: water (0.05% ammonia water), mobile phase B: acetonitrile; flow rate: 25mL/min; gradient: 30%-65% acetonitrile within 8 minutes; detection wavelength: 220 nm), to obtain compound 61 (1.2 mg, 8.3%) as a white solid.
  • Step 1 Preparation of (R)-2-((2-amino-6-chloropyridin[3,4-d]pyrimidin-4-yl)amino)-2-methylhexan-1-ol (62a)
  • compound 60c (300mg, 1.56mmol), (R)-2-amino-2-methylhexan-1-ol (300mg, 2.27mmol), Carter condensing agent (810mg, 1.83mmol), 1,8 - Diazabicycloundec-7-ene (697 mg, 4.59 mmol) was dissolved in N,N-dimethylformamide (5 mL). The reaction was stirred at room temperature for 12 hours.
  • Step 3 (R)-2-((2-amino-6-(4-((dimethylamino)methyl)benzyl)pyridin[3,4-d]pyrimidin-4-yl)amino)-2 - Preparation of methylhexan-1-ol (62)
  • compound 62b (20 mg, 0.05 mmol), potassium (dimethylaminomethyl) trifluoroborate (165 mg, 0.08 mmol), cesium carbonate (49 mg, 0.15 mmol), 2-dicyclohexylphosphine-2,4 , 6-triisopropylbiphenyl (5 mg, 0.01 mmol), palladium acetate (1 mg, 0.005 mmol) were dissolved in 1,4-dioxane (1 mL) and water (0.2 mL). The reaction solution was replaced with nitrogen three times, and stirred at 100° C. for 12 hours.
  • the preparation method was the same as that of Example 63, except that the compound trimethylboroxine was replaced by ethyl boroxane to obtain compound 64.
  • compound 62a 40 mg, 0.129 mmol
  • dimethylamine hydrochloride 15.79 mg, 0.194 mmol
  • cesium carbonate 168.27 mg, 0.516 mmol
  • Pd-PEPPSI-IPent Cl -o-picoline CAS1612891-29-8, 10.85 mg, 0.013 mmol
  • 1,4-dioxane 1.5 mL
  • reaction solution was diluted with water (20 mL), extracted with ethyl acetate (3 ⁇ 20 mL), the combined organic phases were washed with saturated brine (40 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure.
  • the obtained crude product was separated and purified by preparative chromatographic column (column type: XBridge Prep C18 OBD column, 5um, 19*150mm; mobile phase A: water (10mmol/L ammonium bicarbonate), mobile phase B: acetonitrile; flow rate: 25mL/min ; gradient: 25%-40% acetonitrile in 8 minutes; detection wavelength: 254/220nm).
  • the product was collected and lyophilized to obtain compound 65 (2.3 mg, 5.43%) as a yellow-green solid.
  • Step 1 (R)-N-((1-((tert-butyldimethylsilyl)oxy)-2-methylhex-2-yl)-2,6,8-trichloropyrimido[ Preparation of 5,4-d]pyrimidin-4-amine (66b)
  • Step 3 (R)-N 4 -(1-((tert-butyldimethylsilyl)oxy)-2-methylhex-2-yl)-6-chloro-N 2 -(2,4 -dimethoxybenzyl)pyrimido[5,4-d]pyrimidine-2,4-diamine (66d) and (R)-N 8 -(1-((tert-butyldimethylsilyl) Oxy)-2-methylhex-2-yl)-6-chloro-N 2 -(2,4-dimethoxybenzyl)pyrimido[5,4-d]pyrimidine-2,8-di Preparation of amine (67a)
  • compound 66c (300mg, 0.675mmol) was dissolved in 1,4-dioxane (3mL), and N,N-diisopropylethylamine (174mg, 1.35mmol) and 2,4- Dimethoxybenzylamine (113 mg, 0.675 mmol).
  • the reaction system was stirred at 100°C for 3 hours.
  • water (20 mL) was added to quench the reaction
  • ethyl acetate (3 ⁇ 20 mL) was added to the system for extraction, the combined organic phases were dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure.
  • Step 4 (R)-N 4 -(1-((tert-butyldimethylsilyl)oxy)-2-methylhex-2-yl)-N 2 -(2,4-dimethoxy benzyl)pyrimido[5,4-d]pyrimidine-2,4-diamine (66e) and (R)-N 8 -(1-((tert-butyldimethylsilyl)oxy)- Preparation of 2-methylhex-2-yl)-N 2 -(2,4-dimethoxybenzyl)pyrimido[5,4-d]pyrimidine-2,8-diamine (67b)
  • Step 5 (R)-2-((2-aminopyrimido[5,4-d]pyrimidin-4-yl)amino)-2-methylhexan-1-ol (66) and (R)-2 Preparation of -((6-aminopyrimido[5,4-d]pyrimidin-4-yl)amino)-2-methylhexan-1-ol (67)
  • compound 68a (2.4 g, 12.68 mmol) was dissolved in dry tetrahydrofuran (80 mL), the reaction solution was cooled to -78 ° C, and boron trifluoride ether solution (3.3 mL) was slowly added dropwise under nitrogen atmosphere, Keep at -78°C for 1.5 hours. Then a solution of n-butylmagnesium chloride in tetrahydrofuran (13.5 mL, 2 mol/L) was slowly added dropwise, and the reaction was maintained at -78°C for 2 hours.
  • Step 3 Preparation of (R)-2-(((R)-2-hydroxy-1-phenylethyl)amino)-2-methylhexa-1,1-dideutero-1-ol (68c)
  • Step 5 (R)-2-((2-Aminopyrido[3,4-d]pyrimidin-4-yl)amino)-2-methylhexan-1,1-dideutero-1-ol (68 ) preparation
  • compound 68d (90mg, 0.67mmol) and 2-aminopyridin[3,4-d]pyrimidin-4(3H)-one (44b) (100mg, 0.62mmol) were dissolved in N,N-dimethyl Add formamide (2 mL), add Carter's condensing agent (327 mg, 0.74 mmol) and 1,8-diazabicycloundec-7-ene (282 mg, 1.85 mmol), and react at room temperature for 16 hours. After the reaction was complete, the reaction solution was diluted with water (10 mL), and then extracted with ethyl acetate (3 ⁇ 8 mL).
  • Step 1 Preparation of (R)-3-ethyl-5-phenyl-5,6-dihydro-2H-1,4-oxazin-2-one (69a)
  • compound 69a (2.3 g, 11.32 mmol) was dissolved in dry tetrahydrofuran (75 mL), the reaction solution was cooled to -78 ° C, and boron trifluoride ether solution (3.0 mL) was slowly added dropwise under nitrogen atmosphere, Keep at -78°C for 1.5 hours. Then a solution of n-butylmagnesium chloride in tetrahydrofuran (11.9 mL, 2 mol/L) was slowly added dropwise, and the reaction was maintained at -78°C for 2 hours.
  • Step 3 Preparation of (R)-2-ethyl-2-(((R)-2-hydroxy-1-phenylethyl)amino)hexan-1-ol (69c)
  • Step 5 Preparation of (R)-2-((2-aminopyrido[3,4-d]pyrimidin-4-yl)amino)-2-ethylhexan-1-ol (69)
  • compound 69d (202mg, 1.39mmol) and 2-aminopyridin[3,4-d]pyrimidin-4(3H)-one (44b) (150mg, 0.93mmol) were dissolved in N,N-dimethyl Add formamide (3 mL), add Carter's condensing agent (491 mg, 1.11 mmol) and 1,8-diazabicycloundec-7-ene (422 mg, 2.78 mmol), and react at room temperature for 16 hours.
  • reaction solution was diluted with water (10 mL), extracted with ethyl acetate (3 ⁇ 8 mL), the organic phases were combined, washed with saturated brine (3 ⁇ 15 mL), and dried over anhydrous sodium sulfate.
  • the organic phase was concentrated under reduced pressure, and the obtained crude product was separated and purified by preparative chromatographic column (column type: XBridge Shield RP18OBD Column, 5um, 19*150mm; mobile phase A: water (0.05% ammonia water), mobile phase B: acetonitrile; flow rate: 25 mL/min; gradient: 25%-50% acetonitrile in 8 minutes; detection wavelength: 254/220 nm), the product 69 was obtained as a white solid (16.3 mg, 6.0%).
  • Test Example 1 The agonistic activity of the compound of the present invention on hTLR8 and hTLR7
  • HEK-Blue TM hTLR8 cells and HEK-Blue TM hTLR7 cells purchased from Invivogen were used.
  • the cells were co-transfected with hTLR8 or hTLR7 gene and a reporter gene of secreted alkaline phosphatase (SEAP) in HEK293 cells.
  • SEAP secreted alkaline phosphatase
  • the SEAP gene is placed downstream of the IFN- ⁇ minimal promoter, and the IFN- ⁇ minimal promoter is composed of five NF- ⁇ B and AP-1 binding sites.
  • Stimulators of hTLR8 or hTLR7 will activate NF- ⁇ B and AP-1 promoters to produce SEAP, and evaluate the effect of compounds by detecting SEAP levels.
  • Compound plate preparation starting from 2 mM, the compound to be tested was diluted 3 times with DMSO and diluted 10 gradients. Take 2 ⁇ L of diluted compound and add 38 ⁇ L HEK-BlueTM detection reagent for 20-fold intermediate dilution. Wells with cells plus 0.5% DMSO served as negative control wells for low readings. Cells were added with 1 ⁇ M GS-9688 (refer to WO2016141092A1 synthesis route) wells as positive control wells with high readings.
  • Activity % (compound reading - negative well reading) / (positive well reading - negative well reading) x 100.
  • TLR8/TLR7 The agonistic activity of the compounds of the present invention on TLR8/TLR7 is shown in Table 1 below.
  • the compound of the present invention can selectively activate TLR8.
  • Test Example 2 Research on the absorption mechanism of the compound of Example 52
  • Dilute Caco-2 cells (American Type Culture Collection, HTB-37) with culture medium to 6.86 ⁇ 105 cells/mL, and add 50 ⁇ L of cell suspension to a 96-well Transwell plate (Cat.No.3391) in the filter hole.
  • the cell culture plates were incubated for 14-18 days in a cell culture incubator at 37°C, 5% CO 2 , 95% relative humidity. Change the cell culture medium every other day.
  • Transepithelial electrical resistance (TEER) across the monolayer was measured using the Millicell Epithelial Volt-Ohm measurement system (Millipore, USA). Return the plate to the incubator after the measurement is complete.
  • the TEER value is calculated according to the following formula:
  • TEER measurement value (ohms) x membrane area (cm 2 ) TEER value (ohm ⁇ cm 2 )
  • the TEER value is greater than 230ohm ⁇ cm 2 , indicating that the Caco-2 monolayer film is qualified and can be used for subsequent tests.
  • Example 52 The compound of Example 52 was accurately weighed, dissolved in DMSO to prepare a stock solution with a concentration of 2 mM, and diluted with HBSS (Gibico, 10 mM HEPES, pH 7.4) to obtain a 10 ⁇ M working solution. Metoprolol and digoxin were used as control compounds.
  • the monolayer was washed twice with pre-warmed HBSS (10 mM HEPES, pH 7.4) before the experiment. The plates were then incubated at 37°C for 30 minutes.
  • the plates were incubated at 37°C for 2 hours. At the end of the incubation, pipette 50 ⁇ L of the solution from the donor side (Ap ⁇ Bl's apical compartment, Bl ⁇ Ap's basolateral compartment) and the recipient side (Ap ⁇ Bl's basolateral compartment and Bl ⁇ Ap's apical compartment). The samples were transferred to the wells of a new 96-well plate, and then 200 ⁇ L of acetonitrile containing IS (100 nM alprazolam, 200 nM caffeine, and 100 nM tolbutamide) was added.
  • acetonitrile containing IS 100 nM alprazolam, 200 nM caffeine, and 100 nM tolbutamide
  • P app is the apparent permeability (cm/s ⁇ 10 -6 )
  • dQ/dt is the rate of drug transport (pmol/s)
  • A is the surface area of the membrane (cm 2 )
  • D0 is the initial donor concentration (nM ; pmol/cm 3 )
  • P app(BA) represents the apparent permeability coefficient from the basolateral to the apex direction
  • P app(AB) represents the apparent permeability coefficient from the apex to the basolateral direction
  • the compound of Example 52 has a good permeability coefficient, and the risk of efflux transporter substrate is low.
  • the compound in Example 4 of WO2018045144A1 has low permeability and high efflux ratio, and is likely to be an efflux transporter substrate.
  • the in vitro Caco-2 test results of the compound Example 52 of the present invention show better permeability than the compound of Example 4 of WO2018045144A1.
  • This experiment aims to evaluate the pharmacokinetic behavior of the compound of Example 52 after intravenous infusion or gavage administration in rats.
  • Intravenous infusion the test compound is prepared as a clear solution of 0.5 mg/ml, and the solvent is 2% ethanol/40% polyethylene glycol 300/58% 0.01 molar hydrochloric acid;
  • intragastric administration the test compound is prepared as 0.5 mg/ml clear solution in 2% ethanol/40% polyethylene glycol 300/58% 0.01 molar hydrochloric acid.
  • Plasma concentrations of test compounds were determined by high performance liquid chromatography-tandem mass spectrometry (LC-MS/MS). Plasma and tissue concentrations were processed using the non-compartmental model of the pharmacokinetic software WinNonlin TM Version 8.3 (Pharsight, Mountain View, CA), and pharmacokinetic parameters were calculated using the linear log trapezoidal method.
  • Table 5 below shows the intestinal bioavailability of the compound of Example 52 at a dose of 5 mg/kg in rats and the relevant parameters of the liver first-pass study.
  • the compound of Example 52 has low systemic oral bioavailability, but high intestinal bioavailability, can be specifically enriched in liver tissue, and is mainly excreted through the intestinal tract.
  • the purpose of this test is to evaluate the pharmacodynamics of the compound of Example 52 after oral administration in male cynomolgus monkeys.
  • test compound was formulated at 10 mg/mL, and the vehicle was 10% ethanol, 40% polyethylene glycol 300, and 50% deionized water. Weigh an appropriate amount of the test compound in an appropriate amount of ethanol, and dissolve the mixture completely under stirring and/or ultrasonication. An appropriate volume of polyethylene glycol 300 is then added with stirring. Finally, the appropriate volume of deionized water is added with stirring to obtain the final concentration of the formulation. Formulations will be stirred at room temperature for at least 10 minutes prior to and during dosing.
  • MSD GOLD Read Buffer B add 150 ⁇ L MSD GOLD Read Buffer B to each well, and use MSD reader (model: MESO SECTOR S600) to read the U-PLEX 96 microwell plate.
  • Figure 1 shows the parameters of the compound of Example 52 in the serum of cynomolgus monkeys.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Virology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Molecular Biology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Epidemiology (AREA)
  • AIDS & HIV (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Hydrogenated Pyridines (AREA)

Abstract

本公开涉及芳胺类衍生物及其制备方法和医药用途。具体地,涉及通式(I)所示的化合物,其制备方法,含有其的药物组合物,以及其作为Toll样受体(TLR)激动剂用于治疗与TLR8活性相关的疾病的用途。通式(I)中的各基团的定义与说明书中的定义相同。

Description

芳胺类衍生物及其制备方法和医药用途 技术领域
本发明涉及芳胺类衍生物及其制备方法和医药用途。具体地,本发明涉及通式(I)所示的化合物,其制备方法,含有其的药物组合物,以及其作为Toll样受体(TLR)激动剂用于治疗与TLR8活性相关的疾病的用途。
背景技术
Toll样受体(TLR)是一类模式识别受体,可以识别微生物并对其产生应答。TLR家族成员在免疫***中起着重要作用,既是参与先天免疫的重要元件,也是连接先天免疫和特异性免疫的桥梁。该受体可以特异性地识别微生物,并启动免疫应答。
TLR均为I型跨膜糖蛋白,由富含16-28个亮氨酸的重复序列(LRR)的胞外结构域、跨膜结构域和胞质Toll/IL-1受体(TIR)结构域组成。X射线晶体衍射分析确定TLR LRR结构域均为马蹄状结构。至今在人类已发现11个成员,其中TLR1、2、4、5、6、10和11位于细胞表面,TLR3、7、8、9位于胞内体膜上。TLR8与TLR7在种系发生上彼此接近并且具有高度的序列同源性,并位于相邻的X染色体上(Xp22)。当TLR的LRR与配体结合后,TIR结构域构象发生改变,随后触发TLR信号通路的激活。TLR的TIR结构域可以募集多种信号分子,包括肿瘤坏死因子受体相关因子6和髓样分化因子88(MyD88)等。其中TLR8依赖MyD88信号通路,诱导激活蛋白酶-1(AP-1)和核因子κB(NF-κB)转移到核内,诱导核内相关基因的表达,分泌趋化因子和炎症因子等,发挥转录调控的作用。此外,TLR8还可以激活丝裂原活化的蛋白激酶(MAPK)信号通路,包括p38、ERK和JNK等,主要参与细胞增殖、细胞分化、细胞转化和细胞凋亡等的调节,与炎症和肿瘤等多种疾病密切相关(免疫学杂志,2017,33,813)。
乙型肝炎病毒(HBV)是一种颗粒状的双链DNA病毒。激活TLR8可以有效地抑制体内和体外乙肝病毒复制,因此成为开发治疗慢性乙型肝炎病毒的靶点。研究发现TLR8激动剂ssRNA40可以选择性地激活肝周的先天免疫细胞产生大量的IFN-γ从而抑制乙肝病毒复制,从而有潜力用作肝炎病毒感染的治疗。利用TLR8激动剂刺激PBMC后发现可以诱导高水平的IFN-γ和TNF-α的产生,从而抑制HBV复制(Current Opinion in Virology,2018,30,9)。
TLR不但表达于免疫细胞上,还可在各种肿瘤细胞中表达,参与肿瘤免疫监视,对肿瘤的生长发挥不同的作用。其中TLR8激活后增强自然杀伤细胞(NK cells)的活性,并提高抗体依赖的细胞调节的细胞毒性(ADCC)以及诱导Th1极化。TLR8激动剂在癌症治疗中作为一种有潜力的佐剂,目的在于诱导针对肿瘤细胞的特异性免疫应答,提高已获批准的单克隆抗体疗法临床疗效,尤其是在ADCC降 低的个体。
鉴于TLR-8激动剂具有治疗多种疾病的重要潜力,临床上对于活性强、选择性高的新型TLR-8激动剂具有迫切需求。
发明内容
本发明人经过潜心研究,设计合成了一系列芳胺类化合物,其显示出优异的TLR8激动活性,可以开发为治疗与TLR8相关的疾病的药物。
因此,本发明的目的是提供一种通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,
Figure PCTCN2022128963-appb-000001
其中,
X 1为CR 1或N;
X 2为CR 2或N;
X 3为CR 3或N;
X 4为CR 4或N;
L选自一个键、-(CH 2) v-、-C(O)(CH 2) t-或-(CH 2) tC(O)-;
R 1选自氢、卤素、氨基、硝基、氰基、羟基、巯基、羧基、烷基、烷氧基、卤代烷基、卤代烷氧基;
R 2选自氢、卤素、氰基、氧代基、烷基、烯基、炔基、-OR a、-SR a、-NR aR b、环烷基、杂环基、芳基和杂芳基,其中所述烷基、烯基、炔基、环烷基、杂环基、芳基、杂芳基任选进一步被一个或多个Q 1基团所取代;
R 3选自氢、卤素、氰基、氧代基、烷基、烯基、炔基、-OR a、-SR a、-NR aR b、环烷基、杂环基、芳基和杂芳基,其中所述烷基、烯基、炔基、环烷基、杂环基、芳基、杂芳基任选进一步被一个或多个Q 2基团所取代;
R 4选自氢、卤素、氨基、硝基、氰基、羟基、巯基、羧基、烷基、烷氧基、卤代烷基、卤代烷氧基;
R 5和R 6各自独立地选自氢、烷基、烯基、炔基、环烷基、杂环基、芳基、杂芳基;其中所述烷基、烯基、炔基、环烷基、杂环基、芳基、杂芳基任选进一步被选自氘代、卤素、硝基、氰基、氧代基、烷基、烯基、炔基、环烷基、杂环基、芳基、杂芳基、-OR a、-SR a、-NR aR b、-C(O)R a、-O(O)CR a、-C(O)OR a、-C(O)NR aR b、-NR aC(O)R b、-S(O) nR a、-S(O) nNR aR b和-NR aS(O) nR b的一个或多个基团取代;
Q 1和Q 2各自独立地选自卤素、硝基、氰基、氧代基、烷基、烯基、炔基、环烷基、杂环基、芳基、杂芳基、-OR a、-SR a、-(CH 2) v-NR aR b、-NR aR b、-C(O)R a、-O(O)CR a、-C(O)OR a、-C(O)NR aR b、-NR aC(O)R b、-S(O) nR a、-S(O) nNR aR b和-NR aS(O) nR b,其中所述烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基任选进一步被选自卤素、氨基、硝基、氰基、羧基、酯基、氧代基、烷基、烷氧基、环烷基、杂环基、芳基、杂芳基、-OR c、-SR c、-(CH 2) v-OR c、-(CH 2) v-NR cR d、-NR cR d、-C(O)R c、-O(O)CR c、-C(O)OR c、-C(O)NR cR d、-NR cC(O)R d、-S(O) nR a、-S(O) nNR cR d和-NR cS(O) nR d的一个或多个基团取代;
R a和R b各自独立地选自氢、卤素、羟基、硝基、氰基、氧代、烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基,其中所述烷基、烯基、炔基、环烷基、杂环基、芳基、杂芳基任选进一步被选自卤素、氨基、硝基、氰基、羧基、酯基、氧代基、烷基、环烷基、杂环基、芳基、杂芳基、-OR c、-SR c、-(CH 2) v-OR c、-(CH 2) v-NR cR d、-NR cR d、-C(O)R c、-O(O)CR c、-C(O)OR c、-C(O)NR cR d、-NR cC(O)R d、-S(O) nR a、-S(O) nNR cR d和-NR cS(O) nR d中的一个或多个基团取代;
或者R a和R b与他们连接的氮原子一起形成含氮杂环基,所述含氮杂环基除了N之外,任选进一步含有选自N、O、S的一个或多个杂原子,所述含氮杂环基任选进一步被选自卤素、硝基、氰基、氧代基、羧基、酯基、烷基、卤代烷基、环烷基、杂环基、芳基、杂芳基、-OR c、-SR c、-(CH 2) v-OR c、-(CH 2) v-NR cR d、-NR cR d、-C(O)R c、-O(O)CR c、-C(O)OR c、-C(O)NR cR d、-NR cC(O)R d、-S(O) nR a、-S(O) nNR cR d和-NR cS(O) nR d的一个或多个基团取代;
R c和R d各自独立地选自氢、卤素、羟基、硝基、氰基、氧代、烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基,其中所述烷基、烯基、炔基、环烷基、杂环基、芳基、杂芳基任选进一步被选自卤素、氨基、硝基、氰基、羟基、巯基、羧基、酯基、氧代基、烷基、烷氧基、环烷基、杂环基、芳基和杂芳基中的一个或多个基团取代;
或者R c和R d与他们连接的氮原子一起形成含氮杂环基,所述含氮杂环基除了N之外,任选进一步含有选自N、O、S的一个或多个杂原子,所述含氮杂环基任选进一步被选自卤素、硝基、氰基、氧代基、羟基、巯基、羧基、酯基、烷基、烷氧基、环烷基、杂环基、芳基和杂芳基;
n为1或2;
v为1至6的整数;
t为0至6。
在一个具体的实施方案中,根据本发明所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,其为通式(II)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、 或其可药用盐,
Figure PCTCN2022128963-appb-000002
其中,X 1、X 2、X 3、L、R 4、R 5、R 6如通式(I)所定义。
在另一个具体的实施方案中,根据本发明所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,其为通式(III)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,
Figure PCTCN2022128963-appb-000003
其中,L、R 1、R 3、R 4、R 5、R 6如通式(I)所定义。
在另一个具体的实施方案中,根据本发明所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,其为通式(IV)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,
Figure PCTCN2022128963-appb-000004
其中,L、R 1、R 2、R 4、R 5、R 6如通式(I)所定义。
在另一个具体的实施方案中,根据本发明所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,其为通式(V)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,
Figure PCTCN2022128963-appb-000005
其中,L、R 2、R 4、R 5、R 6如通式(I)所定义。
在另一个具体的实施方案中,根据本发明所述的通式(I)~(V)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,其中:
L选自一个键或-C(O)-;优选一个键。
在另一个具体的实施方案中,根据本发明所述的通式(I)~(V)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,其中:
L选自-(CH 2) v-;v为1或2,优选1。
在另一个具体的实施方案中,根据本发明所述的通式(I)~(V)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,其为通式(I-1)、(II-1)、(III-1)、(IV-1)、(V-1)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,
Figure PCTCN2022128963-appb-000006
其中,X 1、X 2、X 3、X 4、R 1、R 2、R 3、R 4、R 5、R 6如通式(I)所定义。
在另一个具体的实施方案中,根据本发明所述的通式(I)~(V)或通式(I-1)、(II-1)、(III-1)、(IV-1)、(V-1)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,其中,
R 3选自氢、卤素、C 1-C 6烷基、C 6-C 10芳基和5至10元杂芳基,其中所述C 1-C 6烷基、C 6-C 10芳基和5至10元杂芳基任选进一步被一个或多个Q 2基团所取代;
Q 2选自卤素、C 1-C 6烷基、4-6元杂环基、C 6-C 10芳基、5至10元杂芳基、-NR aR b,其中所述C 6-C 10芳基和5至10元杂芳基任选进一步被选自卤素、C 1-C 6烷基、-(CH 2) v-NR cR d的一个或多个基团取代;
R a和R b各自独立地选自氢、C 1-C 6烷基;
或者R a和R b与他们连接的氮原子一起形成4-6元含氮杂环基,所述4-6元含氮杂环基除了N之外,任选进一步含有选自N、O、S的一个或多个杂原子,所述4-6元含氮杂环基任选进一步被选自氧代基、C 1-C 6烷基、C 1-C 6卤代烷基、C 3-C 6环烷基、-OR c、-SR c、-(CH 2) v-OR c、-NR cR d的一个或多个基团取代;
R c和R d各自独立地选自氢、C 1-C 6烷基;
或者R c和R d与他们连接的氮原子一起形成4-6元含氮杂环基,所述4-6元含氮杂环基除了N之外,任选进一步含有选自N、O、S的一个或多个杂原子,所述4-6元含氮杂环基任选进一步被选自卤素、C 1-C 6烷基的一个或多个基团取代;
v为1至6的整数。
在另一个具体的实施方案中,根据本发明所述的通式(I)~(V)或通式(I-1)、(II-1)、(III-1)、(IV-1)、(V-1)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,其中,R 3选自氢、卤素、C 1-C 6烷基,优选氢。
在另一个具体的实施方案中,根据本发明所述的通式(I)~(V)或通式(I-1)、(II-1)、(III-1)、(IV-1)、(V-1)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,其中,
R 3选自C 6-C 10芳基和5至10元杂芳基,优选苯基或5-6元杂芳基;其中所述C 6-C 10芳基和5至10元杂芳基任选进一步被一个或多个Q 2基团所取代;
Q 2选自卤素、C 1-C 6烷基、4-6元杂环基、C 6-C 10芳基、5至10元杂芳基、-NR aR b,其中所述C 6-C 10芳基和5至10元杂芳基任选进一步被选自卤素、C 1-C 6烷基的一个或多个基团取代;
R a和R b各自独立地选自氢、C 1-C 6烷基;
或者R a和R b与他们连接的氮原子一起形成4-6元含氮杂环基,所述4-6元含氮杂环基除了N之外,任选进一步含有选自N、O、S的一个或多个杂原子,所述4-6元含氮杂环基任选进一步被选自氧代基、C 1-C 6烷基、C 1-C 6卤代烷基、C 3-C 6环烷基、-OR c、-SR c、-(CH 2) v-OR c、-NR cR d的一个或多个基团取代;
R c和R d各自独立地选自氢、C 1-C 6烷基;
或者R c和R d与他们连接的氮原子一起形成4-6元含氮杂环基,所述4-6元含氮杂环基除了N之外,任选进一步含有选自N、O、S的一个或多个杂原子,所述4-6元含氮杂环基任选进一步被选自卤素、C 1-C 6烷基的一个或多个基团取代v为1至6的整数,优选1或2。
在另一个具体的实施方案中,根据本发明所述的通式(I)~(V)或通式(I-1)、(II-1)、(III-1)、(IV-1)、(V-1)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,其中,
R 3选自5至10元杂芳基,优选5或6元杂芳基,更优选吡啶基、嘧啶基、咪唑基、吡唑基、吡咯基、吡啶并咪唑基、吡啶并吡咯基、吡啶并吡唑基、苯并咪唑基、苯并吡唑基、苯并吡咯基;所述杂芳基任选进一步被一个或多个Q 2基团所取代;
Q 2选自卤素、C 1-C 6烷基、C 6-C 10芳基优选苯基、5至10元杂芳基优选5或6元杂芳基,其中所述芳基和杂芳基任选进一步被选自卤素、C 1-C 6烷基的一个或多个基团取代。
在另一个具体的实施方案中,根据本发明所述的通式(I)~(V)或通式(I-1)、(II-1)、(III-1)、(IV-1)、(V-1)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,其中,
R 3选自5至10元杂芳基,优选5或6元杂芳基,更优选吡啶基、嘧啶基、咪唑基、吡唑基、吡咯基、吡啶并咪唑基、吡啶并吡咯基、吡啶并吡唑基、苯并咪唑基、苯并吡唑基、苯并吡咯基;所述杂芳基任选进一步被一个或多个Q 2基团所取代;
Q 2选自卤素、C 1-C 6烷基、4-6元杂环基或-NR aR b,优选-NR aR b
R a和R b各自独立地选自氢、C 1-C 6烷基;
或者R a和R b与他们连接的氮原子一起形成4-6元含氮杂环基,所述4-6元含氮杂环基除了N之外,任选进一步含有选自N、O、S的一个或多个杂原子,所述4-6元含氮杂环基任选进一步被选自氧代基、C 1-C 6烷基、C 1-C 6卤代烷基、C 3-C 6环烷基、-OR c、-SR c、-(CH 2) v-OR c、-NR cR d的一个或多个基团取代;
R c和R d各自独立地选自氢、C 1-C 6烷基;
或者R c和R d与他们连接的氮原子一起形成4-6元含氮杂环基,所述4-6元含氮杂环基除了N之外,任选进一步含有选自N、O、S的一个或多个杂原子,所述4-6元含氮杂环基任选进一步被选自卤素、C 1-C 6烷基的一个或多个基团取代v为1至6的整数,优选1或2。
在另一个具体的实施方案中,根据本发明所述的通式(I)~(V)或通式(I-1)、(II-1)、(III-1)、(IV-1)、(V-1)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,其中,
R 3选自C 6-C 10芳基,优选苯基;所述芳基任选进一步被Q 2基团所取代;
Q 2选自5或6元杂芳基,其中所述5或6元杂芳基任选进一步被选自卤素、C 1-C 6烷基的一个或多个基团取代。
在另一个具体的实施方案中,根据本发明所述的通式(I)~(V)或通式(I-1)、 (II-1)、(III-1)、(IV-1)、(V-1)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,其中,
R 3选自C 1-C 6烷基,优选甲基;所述C 1-C 6烷基任选进一步被Q 2基团所取代;
Q 2选自C 6-C 10芳基,优选苯基,其中所述C 6-C 10芳基任选进一步被-(CH 2) v-NR cR d取代;
R c和R d与他们连接的氮原子一起形成4-6元含氮杂环基,所述4-6元含氮杂环基除了N之外,任选进一步含有选自N、O、S的一个或多个杂原子,所述4-6元含氮杂环基任选进一步被选自卤素、C 1-C 6烷基的一个或多个基团取代;
v为1至6的整数,优选1或2。
在另一个具体的实施方案中,根据本发明所述的通式(I)~(V)或通式(I-1)、(II-1)、(III-1)、(IV-1)、(V-1)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,其中,
R 2选自氢、卤素、C 1-C 6烷基、-NR aR b,所述C 1-C 6烷基任选进一步被Q 1取代;
Q 1选自C 6-C 10芳基、5至10元杂芳基,其中所述C 6-C 10芳基和5至10元杂芳基任选进一步被选自卤素、C 1-C 6烷基、-(CH 2) v-NR cR d的一个或多个基团取代;
R a、R b各自独立地选自氢、C 1-C 6烷基;
R c和R d各自独立地选自氢、C 1-C 6烷基;
或者R c和R d与他们连接的氮原子一起形成4-6元含氮杂环基,所述4-6元含氮杂环基除了N之外,任选进一步含有选自N、O、S的一个或多个杂原子,所述4-6元含氮杂环基任选进一步被选自卤素、C 1-C 6烷基的一个或多个基团取代;
v为1至6的整数,优选1或2。
在另一个具体的实施方案中,根据本发明所述的通式(I)~(V)或通式(I-1)、(II-1)、(III-1)、(IV-1)、(V-1)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,其中,R 2选自氢、卤素、C 1-C 6烷基、-NR aR b
R a、R b各自独立地选自氢、C 1-C 6烷基。
在另一个具体的实施方案中,根据本发明所述的通式(I)~(V)或通式(I-1)、(II-1)、(III-1)、(IV-1)、(V-1)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,其中,
R 2选自C 1-C 6烷基,优选甲基;所述C 1-C 6烷基任选进一步被Q 2基团所取代;
Q 2选自C 6-C 10芳基,优选苯基,其中所述C 6-C 10芳基任选进一步被-(CH 2) v-NR cR d取代;
R c和R d与他们连接的氮原子一起形成4-6元含氮杂环基,所述4-6元含氮杂环基 除了N之外,任选进一步含有选自N、O、S的一个或多个杂原子,所述4-6元含氮杂环基任选进一步被选自卤素、C 1-C 6烷基的一个或多个基团取代;
v为1至6的整数,优选1或2。
在另一个具体的实施方案中,根据本发明所述的通式(I)~(V)或通式(I-1)、(II-1)、(III-1)、(IV-1)、(V-1)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,其中,R 1选自氢、卤素、C 1-C 6烷基、C 1-C 6烷氧基、C 1-C 6卤代烷基、C 1-C 6卤代烷氧基;优选,R 1为氢或卤素。
在另一个具体的实施方案中,根据本发明所述的通式(I)~(V)或通式(I-1)、(II-1)、(III-1)、(IV-1)、(V-1)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,其中,R 4选自氢、卤素、C 1-C 6烷基、C 1-C 6烷氧基、C 1-C 6卤代烷基、C 1-C 6卤代烷氧基;优选,R 4为氢或卤素。
在另一个具体的实施方案中,根据本发明所述的通式(I)~(V)或通式(I-1)、(II-1)、(III-1)、(IV-1)、(V-1)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,其中,
R 5和R 6各自独立地选自氢和C 1-C 12烷基,所述C 1-C 12烷基任选进一步被选自氘代、-OR a、-SR a、-NR aR b的一个或多个基团取代;
R a选自氢、C 1-C 6烷基;
R b选自氢、C 1-C 6烷基、C 3-C 6环烷基和5至7元杂环基;
优选地,R 5为氢,R 6为C 1-C 12烷基,所述C 1-C 12烷基任选进一步被选自氘代、-OH的一个或多个基团取代。
本发明的典型化合物,包括但不限于:
Figure PCTCN2022128963-appb-000007
Figure PCTCN2022128963-appb-000008
Figure PCTCN2022128963-appb-000009
Figure PCTCN2022128963-appb-000010
Figure PCTCN2022128963-appb-000011
Figure PCTCN2022128963-appb-000012
Figure PCTCN2022128963-appb-000013
Figure PCTCN2022128963-appb-000014
Figure PCTCN2022128963-appb-000015
Figure PCTCN2022128963-appb-000016
Figure PCTCN2022128963-appb-000017
Figure PCTCN2022128963-appb-000018
Figure PCTCN2022128963-appb-000019
或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐。
本发明进一步提供一种制备根据本发明所述的通式(III-1)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐的方法,其包括以下步骤:
Figure PCTCN2022128963-appb-000020
式A3的化合物与硼酸或硼酸频哪醇酯化合物通过金属催化的交叉偶联反应 (例如Suzuki偶联)得到式A4的化合物;然后,用合适的酸(例如三氟乙酸)脱保护基,得到通式(III-1)所示的化合物;催化剂如Pd(PPh 3) 4、K 2CO 3、Cs 2CO 3
或者,
Figure PCTCN2022128963-appb-000021
式A7的化合物与硼酸或硼酸频哪醇酯化合物通过金属催化的交叉偶联反应(例如Suzuki偶联),得到通式(III-1)所示的化合物;催化剂如Pd(PPh 3) 4、K 2CO 3、Cs 2CO 3
其中:R 1、R 3、R 4、R 5、R 6如通式(III-1)所定义。
本发明进一步提供一种制备根据本发明所述的通式(IV-1)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐的方法,其包括以下步骤:
Figure PCTCN2022128963-appb-000022
式B3的化合物与硼酸或硼酸频哪醇酯化合物通过金属催化的交叉偶联反应(例如Suzuki偶联)得到式B4的化合物;然后,用合适的酸(例如三氟乙酸)脱保护基,得到通式(IV-1)所示的化合物;催化剂如Pd(PPh 3) 4、K 2CO 3、Cs 2CO 3
或者,
Figure PCTCN2022128963-appb-000023
式B7的化合物与硼酸或硼酸频哪醇酯化合物通过金属催化的交叉偶联反应(例如Suzuki偶联),得到通式(IV-1)所示的化合物;催化剂如Pd(PPh 3) 4、K 2CO 3、Cs 2CO 3
其中:R 1、R 2、R 4、R 5、R 6如通式(IV-1)所定义。
本发明进一步提供一种制备根据本发明所述的通式(V-1)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐的方法,其包括以下步骤:
Figure PCTCN2022128963-appb-000024
式C3的化合物与硼酸或硼酸频哪醇酯化合物通过金属催化的交叉偶联反应(例如Suzuki偶联)得到式C4的化合物;然后,用合适的酸(例如三氟乙酸)脱保护基,得到通式(V-1)所示的化合物;催化剂如Pd(PPh 3) 4、K 2CO 3、Cs 2CO 3
或者,可以通过以下方案6制备通式(V-1)所示的化合物:
Figure PCTCN2022128963-appb-000025
式C7的化合物与硼酸或硼酸频哪醇酯化合物或R 2Y化合物通过金属催化的交叉偶联反应(例如Suzuki偶联),得到通式(V-1)所示的化合物;催化剂如Pd(PPh 3) 4、K 2CO 3、Cs 2CO 3
其中:R 2、R 4、R 5、R 6如通式(V-1)所定义。
本发明进一步提供一种药物组合物,其包含根据本发明所述的通式化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,以及药学上可接受的载体或赋形剂。
本发明进一步涉及根据本发明所述的通式化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐或包含其的药物组合物在制备TLR8激动剂中的用途。
本发明进一步涉及根据本发明所述的通式化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐或包含其的药物组合物在制备预防或治疗TLR8相关疾病的药物中的用途,所述疾病可以为病毒感染性疾病或恶性肿瘤,所述病毒感染性疾病例如乙型病毒性肝炎、HIV病毒感染,所述恶性肿瘤例如乳腺癌、***、结肠癌、肺癌、胃癌、直肠癌、胰腺癌、脑癌、皮肤癌、口腔癌、***癌、骨癌、肾癌、卵巢癌、膀胱癌、肝癌、输卵管肿瘤、卵巢瘤、腹膜肿瘤、黑色素瘤、实体瘤、神经胶质瘤、神经胶母细胞瘤、肝细胞癌、乳突肾性瘤、头颈部肿瘤、白血病、淋巴瘤、骨髓瘤和非小细胞肺癌。
本发明进一步涉及一种根据本发明所述的通式化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐或包含其的药物组合物,其用作TLR8激动剂。
本发明进一步涉及一种根据本发明所述的通式化合物或其立体异构体、互变 异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐或包含其的药物组合物,其用于预防或治疗TLR8相关疾病,所述疾病可以为病毒感染性疾病或恶性肿瘤,所述病毒感染性疾病例如乙型病毒性肝炎、HIV病毒感染,所述恶性肿瘤例如乳腺癌、***、结肠癌、肺癌、胃癌、直肠癌、胰腺癌、脑癌、皮肤癌、口腔癌、***癌、骨癌、肾癌、卵巢癌、膀胱癌、肝癌、输卵管肿瘤、卵巢瘤、腹膜肿瘤、黑色素瘤、实体瘤、神经胶质瘤、神经胶母细胞瘤、肝细胞癌、乳突肾性瘤、头颈部肿瘤、白血病、淋巴瘤、骨髓瘤和非小细胞肺癌。
本发明进一步涉及一种激动TLR8的方法,其包括向有需要的患者施用有效量的根据本发明所述的通式化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐或包含其的药物组合物。
本发明进一步涉及一种用于预防或治疗TLR8相关疾病的方法,其包括向有需要的患者施用有效量的根据本发明所述的通式化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐或包含其的药物组合物;所述疾病可以为病毒感染性疾病或恶性肿瘤,所述病毒感染性疾病例如乙型病毒性肝炎、HIV病毒感染,所述恶性肿瘤例如乳腺癌、***、结肠癌、肺癌、胃癌、直肠癌、胰腺癌、脑癌、皮肤癌、口腔癌、***癌、骨癌、肾癌、卵巢癌、膀胱癌、肝癌、输卵管肿瘤、卵巢瘤、腹膜肿瘤、黑色素瘤、实体瘤、神经胶质瘤、神经胶母细胞瘤、肝细胞癌、乳突肾性瘤、头颈部肿瘤、白血病、淋巴瘤、骨髓瘤和非小细胞肺癌。
按照本发明所属领域的常规方法,本发明化合物可以与碱或者酸生成药学上可接受的碱式加成盐或酸式加成盐。所述碱包括无机碱和有机碱,可接受的有机碱包括二乙醇胺、乙醇胺、N-甲基葡糖胺、三乙醇胺、氨丁三醇等,可接受的无机碱包括氢氧化铝、氢氧化钙、氢氧化钾、碳酸钠和氢氧化钠等。所述酸包括无机酸和有机酸,可接受的无机酸包括盐酸、硫酸、硝酸、磷酸、氢溴酸等。可接受的有机酸包括乙酸、三氟乙酸、甲酸、抗环血酸等。
含活性成分的药物组合物可以是适用于口服的形式,例如片剂、糖锭剂、锭剂、水或油混悬液、可分散粉末或颗粒、乳液、硬或软胶囊,或糖浆剂或酏剂。可按照本领域任何已知制备药用组合物的方法制备口服组合物,此类组合物可含有一种或多种选自以下的成分:甜味剂、矫味剂、着色剂和防腐剂,以提供悦目和可口的药用制剂。片剂含有活性成分和用于混合的适宜制备片剂的无毒的可药用的赋形剂。这些赋形剂可以是惰性赋形剂,如碳酸钙、碳酸钠、乳糖、磷酸钙或磷酸钠;造粒剂和崩解剂,例如微晶纤维素、交联羧甲基纤维素钠、玉米淀粉或藻酸;粘合剂,例如淀粉、明胶、聚乙烯吡咯烷酮或***胶;和润滑剂,例如硬脂酸镁、硬脂酸或滑石粉。这些片剂可以不包衣或可通过掩盖药物的味道或 在胃肠道中延迟崩解和吸收,因而在较长时间内提供缓释作用的已知技术将其包衣。例如,可使用水溶性味道掩蔽物质,例如羟丙基甲基纤维素或羟丙基纤维素,或延长时间物质例如乙基纤维素、醋酸丁酸纤维素。
也可用其中活性成分与惰性固体稀释剂例如碳酸钙、磷酸钙或高岭土混合的硬明胶胶囊,或其中活性成分与水溶性载体例如聚乙二醇或油溶媒例如花生油、液体石蜡或橄榄油混合的软明胶胶囊提供口服制剂。
水混悬液含有活性物质和用于混合的适宜制备水混悬液的赋形剂。此类赋形剂是悬浮剂,例如羧基甲基纤维素钠、甲基纤维素、羟丙基甲基纤维素、藻酸钠、聚乙烯吡咯烷酮和***胶;分散剂或湿润剂,可以是天然产生的磷脂例如卵磷脂,或烯化氧与脂肪酸的缩合产物,例如聚氧乙烯硬脂酸酯,或环氧乙烷与长链脂肪醇的缩合产物,例如十七碳亚乙基氧基鲸蜡醇(heptadeca乙基ene氧基cetanol),或环氧乙烷与由脂肪酸和己糖醇衍生的部分酯的缩合产物,例如聚环氧乙烷山梨醇单油酸酯,或环氧乙烷与由脂肪酸和己糖醇酐衍生的偏酯的缩合产物,例如聚环氧乙烷脱水山梨醇单油酸酯。水混悬液也可以含有一种或多种防腐剂例如尼泊金乙酯或尼泊金正丙酯、一种或多种着色剂、一种或多种矫味剂和一种或多种甜味剂,例如蔗糖、糖精或阿司帕坦。
油混悬液可通过使活性成分悬浮于植物油如花生油、橄榄油、芝麻油或椰子油,或矿物油例如液体石蜡中配制而成。油混悬液可含有增稠剂,例如蜂蜡、硬石蜡或鲸蜡醇。可加入上述的甜味剂和矫味剂,以提供可口的制剂。可通过加入抗氧化剂例如丁羟茴醚或α-生育酚保存这些组合物。
通过加入水,适用于制备水混悬液的可分散粉末和颗粒可以提供活性成分和用于混合的分散剂或湿润剂、悬浮剂或一种或多种防腐剂。适宜的分散剂或湿润剂和悬浮剂如上所述。也可加入其他赋形剂例如甜味剂、矫味剂和着色剂。通过加入抗氧化剂例如抗坏血酸保存这些组合物。
本发明的药物组合物也可以是水包油乳剂的形式。油相可以是植物油例如橄榄油或花生油,或矿物油例如液体石蜡或其混合物。适宜的乳化剂可以是天然产生的磷脂,例如大豆卵磷脂,和由脂肪酸和己糖醇酐衍生的酯或偏酯,例如山梨坦单油酸酯,和所述偏酯和环氧乙烷的缩合产物,例如聚环氧乙烷山梨醇单油酸酯。乳剂也可以含有甜味剂、矫味剂、防腐剂和抗氧剂。可用甜味剂例如甘油、丙二醇、山梨醇或蔗糖配制的糖浆和酏剂。此类制剂也可含有缓和剂、防腐剂、着色剂和抗氧剂。
本发明的药物组合物可以是无菌注射水溶液形式。可以使用的可接受的溶媒和溶剂有水、林格氏液和等渗氯化钠溶液。无菌注射制剂可以是其中活性成分溶于油相的无菌注射水包油微乳。例如将活性成分溶于大豆油和卵磷脂的混合物中。然后将油溶液加入水和甘油的混合物中处理形成微乳。可通过局部大量注射,将注射液或微乳注入患者的血流中。或者,最好按可保持本发明化合物恒定循环浓 度的方式给予溶液和微乳。为保持这种恒定浓度,可使用连续静脉内递药装置。
本发明的药物组合物可以是用于肌内和皮下给药的无菌注射水或油混悬液的形式。可按已知技术,用上述那些适宜的分散剂或湿润剂和悬浮剂配制该混悬液。无菌注射制剂也可以是在无毒肠胃外可接受的稀释剂或溶剂中制备的无菌注射溶液或混悬液,例如在1,3-丁二醇中制备的溶液。此外,可方便地用无菌固定油作为溶剂或悬浮介质。为此目的,可使用包括合成甘油单或二酯在内的任何调和固定油。此外,脂肪酸例如油酸也可以制备注射剂。
可按用于直肠给药的栓剂形式给予本发明化合物。可通过将药物与在普通温度下为固体但在直肠中为液体,因而在直肠中会溶化而释放药物的适宜的无刺激性赋形剂混合来制备这些药物组合物。此类物质包括可可脂、甘油明胶、氢化植物油、各种分子量的聚乙二醇和聚乙二醇的脂肪酸酯的混合物。
本领域技术人员熟知,药物的给药剂量依赖于多种因素,包括但并非限定于以下因素:所用特定化合物的活性、病人的年龄、病人的体重、病人的健康状况、病人的行被、病人的饮食、给药时间、给药方式、***的速率、药物的组合等。另外,最佳的治疗方式如治疗的模式、通式化合物的日用量或可药用的盐的种类可以根据传统的治疗方案来验证。
本发明可以含有通式化合物,及其药学上可接受的盐、水合物或溶剂化物作为活性成分,与药学上可接受的载体或赋型剂混合制备成组合物,并制备成临床上可接受的剂型。本发明的衍生物可以与其他活性成分组合使用,只要它们不产生其他不利的作用,例如过敏反应等。本发明化合物可作为唯一的活性成分,也可以与其它治疗与酪氨酸激酶活性相关的疾病的药物联合使用。联合治疗通过将各个治疗组分同时、分开或相继给药来实现。
术语说明
除非有相反陈述,在说明书和权利要求书中使用的术语具有下述含义。
术语“烷基”指饱和脂肪族烃基团,其为包含1至20个碳原子的直链或支链基团,优选含有1至12个碳原子的烷基,更优选含有1至6个碳原子的烷基。非限制性实例包括甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、仲丁基、正戊基、1,1-二甲基丙基、1,2-二甲基丙基、2,2-二甲基丙基、1-乙基丙基、2-甲基丁基、3-甲基丁基、正己基、1-乙基-2-甲基丙基、1,1,2-三甲基丙基、1,1-二甲基丁基、1,2-二甲基丁基、2,2-二甲基丁基、1,3-二甲基丁基、2-乙基丁基、2-甲基戊基、3-甲基戊基、4-甲基戊基、2,3-二甲基丁基、正庚基、2-甲基己基、3-甲基己基、4-甲基己基、5-甲基己基、2,3-二甲基戊基、2,4-二甲基戊基、2,2-二甲基戊基、3,3-二甲基戊基、2-乙基戊基、3-乙基戊基、正辛基、2,3-二甲基己基、2,4-二甲基己基、2,5-二甲基己基、2,2-二甲基己基、3,3-二甲基己基、4,4-二甲基己基、2-乙基己基、3-乙基己基、4-乙基己基、2-甲基-2-乙基戊基、2-甲基-3-乙基戊基、正壬基、2-甲基-2-乙基己基、2-甲基-3-乙基己基、2,2-二乙基戊基、正癸基、3,3-二乙 基己基、2,2-二乙基己基,及其各种支链异构体等。更优选的是含有1至6个碳原子的低级烷基,非限制性实施例包括甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、仲丁基、正戊基、1,1-二甲基丙基、1,2-二甲基丙基、2,2-二甲基丙基、1-乙基丙基、2-甲基丁基、3-甲基丁基、正己基、1-乙基-2-甲基丙基、1,1,2-三甲基丙基、1,1-二甲基丁基、1,2-二甲基丁基、2,2-二甲基丁基、1,3-二甲基丁基、2-乙基丁基、2-甲基戊基、3-甲基戊基、4-甲基戊基、2,3-二甲基丁基等。烷基可以是取代的或非取代的,当被取代时,取代基可以在任何可使用的连接点上被取代,所述取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、氧代基、羧基或羧酸酯基。
术语“烯基”指由至少由两个碳原子和至少一个碳-碳双键组成的如上定义的烷基,例如乙烯基、1-丙烯基、2-丙烯基、1-、2-或3-丁烯基等。烯基可以是取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基。
术语“炔基”指由至少由两个碳原子和至少一个碳-碳三键组成的如上定义的烷基,例如乙炔基、丙炔基、丁炔基等。炔基可以是取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基。
术语“环烷基”指饱和或部分不饱和单环或多环环状烃取代基,环烷基环包含3至20个碳原子,优选包含3至12个碳原子,更优选包含3至10个碳原子,进一步优选包含3至8个碳原子,最优选包含3至6个碳原子。单环环烷基的非限制性实例包括环丙基、环丁基、环戊基、环戊烯基、环己基、环己烯基、环己二烯基、环庚基、环庚三烯基、环辛基等;多环环烷基包括螺环、稠环和桥环的环烷基。
术语“螺环烷基”指5至20元的单环之间共用一个碳原子(称螺原子)的多环基团,其可以含有一个或多个双键,但没有一个环具有完全共轭的π电子***。优选为6至14元,更优选为7至10元。根据环与环之间共用螺原子的数目将螺环烷基分为单螺环烷基、双螺环烷基或多螺环烷基,优选为单螺环烷基和双螺环烷基。更优选为4元/4元、4元/5元、4元/6元、5元/5元或5元/6元单螺环烷基。螺环烷基的非限制性实例包括:
Figure PCTCN2022128963-appb-000026
术语“稠环烷基”指5至20元,***中的每个环与体系中的其他环共享毗邻的一对碳原子的全碳多环基团,其中一个或多个环可以含有一个或多个双键,但没有一个环具有完全共轭的π电子***。优选为6至14元,更优选为7至10元。根据组成环的数目可以分为双环、三环、四环或多环稠环烷基,优选为双环或三环,更优选为5元/5元或5元/6元双环烷基。稠环烷基的非限制性实例包括:
Figure PCTCN2022128963-appb-000027
术语“桥环烷基”指5至20元,任意两个环共用两个不直接连接的碳原子的全碳多环基团,其可以含有一个或多个双键,但没有一个环具有完全共轭的π电子***。优选为6至14元,更优选为7至10元。根据组成环的数目可以分为双环、三环、四环或多环桥环烷基,优选为双环、三环或四环,更有选为双环或三环。桥环烷基的非限制性实例包括:
Figure PCTCN2022128963-appb-000028
所述环烷基环可以稠合于芳基、杂芳基或杂环烷基环上,其中与母体结构连接在一起的环为环烷基,非限制性实例包括茚满基、四氢萘基、苯并环庚烷基等。环烷基可以是任选取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、氧代基、羧基或羧酸酯基。
术语“杂环基”指饱和或部分不饱和单环或多环环状烃取代基,其包含3至20个环原子,其中一个或多个环原子为选自氮、氧或S(O) m(其中m是整数0至2)的杂原子,但不包括-O-O-、-O-S-或-S-S-的环部分,其余环原子为碳。优选包含3至12个环原子,其中1~4个是杂原子;最优选包含3至8个环原子,其中1~3个是杂原子;最优选包含5至7个环原子,其中1~2或1~3个是杂原子。单环杂环基的非限制性实例包括吡咯烷基、咪唑烷基、四氢呋喃基、四氢噻吩基、二氢咪唑基、二氢呋喃基、二氢吡唑基、二氢吡咯基、哌啶基、哌嗪基、吗啉基、硫代吗啉基、高哌嗪基、吡喃基等,优选1、2、5-噁二唑基、吡喃基或吗啉基。多环杂环基包括螺环、稠环和桥环的杂环基。
术语“螺杂环基”指5至20元的单环之间共用一个原子(称螺原子)的多环 杂环基团,其中一个或多个环原子为选自氮、氧或S(O) m(其中m是整数0至2)的杂原子,其余环原子为碳。其可以含有一个或多个双键,但没有一个环具有完全共轭的π电子***。优选为6至14元,更优选为7至10元。根据环与环之间共用螺原子的数目将螺杂环基分为单螺杂环基、双螺杂环基或多螺杂环基,优选为单螺杂环基和双螺杂环基。更优选为4元/4元、4元/5元、4元/6元、5元/5元或5元/6元单螺杂环基。螺杂环基的非限制性实例包括:
Figure PCTCN2022128963-appb-000029
术语“稠杂环基”指5至20元,***中的每个环与体系中的其他环共享毗邻的一对原子的多环杂环基团,一个或多个环可以含有一个或多个双键,但没有一个环具有完全共轭的π电子***,其中一个或多个环原子为选自氮、氧或S(O) m(其中m是整数0至2)的杂原子,其余环原子为碳。优选为6至14元,更优选为7至10元。根据组成环的数目可以分为双环、三环、四环或多环稠杂环基,优选为双环或三环,更优选为5元/5元或5元/6元双环稠杂环基。稠杂环基的非限制性实例包括:
Figure PCTCN2022128963-appb-000030
术语“桥杂环基”指5至14元,任意两个环共用两个不直接连接的原子的多环杂环基团,其可以含有一个或多个双键,但没有一个环具有完全共轭的π电子***,其中一个或多个环原子为选自氮、氧或S(O) m(其中m是整数0至2)的杂原子,其余环原子为碳。优选为6至14元,更优选为7至10元。根据组成环的数目可以分为双环、三环、四环或多环桥杂环基,优选为双环、三环或四环,更有选为双环或三环。桥杂环基的非限制性实例包括:
Figure PCTCN2022128963-appb-000031
所述杂环基环可以稠合于芳基、杂芳基或环烷基环上,其中与母体结构连接在一起的环为杂环基,其非限制性实例包括:
Figure PCTCN2022128963-appb-000032
等。
杂环基可以是任选取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、氧代基、羧基或羧酸酯基。
术语“芳基”指具有共轭的π电子体系的6至14元全碳单环或稠合多环(也就是共享毗邻碳原子对的环)基团,优选为6至10元,例如苯基和萘基。更优选苯基。所述芳基环可以稠合于杂芳基、杂环基或环烷基环上,其中与母体结构连接在一起的环为芳基环,其非限制性实例包括:
Figure PCTCN2022128963-appb-000033
芳基可以是取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、羧基或羧酸酯基。
术语“杂芳基”指包含1至4个杂原子、5至14个环原子的杂芳族体系,其中杂原子选自氧、硫和氮。杂芳基优选为5至10元,含1至3个杂原子;更优选为5元或6元,含1至2个杂原子;优选例如咪唑基、呋喃基、噻吩基、噻唑基、吡唑基、噁唑基、吡咯基、四唑基、吡啶基、嘧啶基、噻二唑、吡嗪基等,优选为咪唑基、噻唑基、吡唑基或嘧啶基、噻唑基;更有选吡唑基或噻唑基。所述杂芳基环可以稠合于芳基、杂环基或环烷基环上,其中与母体结构连接在一起的环为杂芳基环,其非限制性实例包括:
Figure PCTCN2022128963-appb-000034
杂芳基可以是任选取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、羧基或羧酸酯基。
术语“烷氧基”指-O-(烷基)和-O-(非取代的环烷基),其中烷基的定义如上所述。烷氧基的非限制性实例包括:甲氧基、乙氧基、丙氧基、丁氧基、环丙氧基、环丁氧基、环戊氧基、环己氧基。烷氧基可以是任选取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷 氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、羧基或羧酸酯基。
术语“卤代烷基”指被一个或多个卤素取代的烷基,其中烷基如上所定义。
术语“卤代烷氧基”指被一个或多个卤素取代的烷氧基,其中烷氧基如上所定义。
术语“羟烷基”指被一个或多个羟基取代的烷基,其中烷基如上所定义。
术语“羟基”指-OH基团。
术语“卤素”指氟、氯、溴或碘。
术语“氨基”指-NH 2
术语“氰基”指-CN。
术语“硝基”指-NO 2
术语“氧代基”指=O。
术语“羧基”指-C(O)OH。
术语“巯基”指-SH。
术语“酯基”指-C(O)O(烷基)或-C(O)O(环烷基),其中烷基和环烷基如上所定义。
术语“酰基”指含有-C(O)R基团的化合物,其中R为烷基、环烷基、杂环基、芳基、杂芳基。
“任选”或“任选地”意味着随后所描述的事件或环境可以但不必发生,该说明包括该事件或环境发生或不发生的场合。例如,“任选被烷基取代的杂环基团”意味着烷基可以但不必须存在,该说明包括杂环基团被烷基取代的情形和杂环基团不被烷基取代的情形。
“取代的”指基团中的一个或多个氢原子,优选为最多5个,更优选为1~3个氢原子彼此独立地被相应数目的取代基取代。不言而喻,取代基仅处在它们的可能的化学位置,本领域技术人员能够在不付出过多努力的情况下确定(通过实验或理论)可能或不可能的取代。例如,具有游离氢的氨基或羟基与具有不饱和(如烯属)键的碳原子结合时可能是不稳定的。
“药物组合物”表示含有一种或多种本文所述化合物或其生理学上/可药用的盐或前体药物与其他化学组分的混合物,以及其他组分例如生理学/可药用的载体和赋形剂。药物组合物的目的是促进对生物体的给药,利于活性成分的吸收进而发挥生物活性。
“可药用盐”是指本发明化合物的盐,这类盐用于哺乳动物体内时具有安全性和有效性,且具有应有的生物活性。
本发明化合物的合成方法
为了完成本发明的目的,本发明采用如下合成方案制备本发明的通式(I)化 合物。
当通式(I)所示的化合物为通式(III-1)所示的化合物时,通过以下方案1制备通式(III-1)所示的化合物:
Figure PCTCN2022128963-appb-000035
方案1
在室温,合适的碱(例如DIEA)存在下,式A1的化合物通过与亲核性胺的反应,得到式A2的化合物;然后,在加热条件下,式A2的化合物与2,4-二甲氧基苄胺反应,得到式A3的化合物;式A3的化合物可以通过金属催化(例如Pd(PPh 3) 4、K 2CO 3、Cs 2CO 3)的交叉偶联反应(例如Suzuki偶联)与硼酸或硼酸频哪醇酯化合物反应,得到式A4的化合物;用合适的酸(例如三氟乙酸)脱保护基,得到通式(III-1)所示的化合物;
其中:R 1、R 3、R 4、R 5、R 6如通式(III-1)所定义。
或者,可以通过以下方案2制备通式(III-1)所示的化合物:
Figure PCTCN2022128963-appb-000036
方案2
式A6的化合物与亲核性胺在缩合剂(例如BOP)存在下反应,得到偶联产物式A7的化合物;式A7的化合物可以通过金属催化(例如Pd(PPh 3) 4、K 2CO 3、Cs 2CO 3)的交叉偶联反应(例如Suzuki偶联)与硼酸或硼酸频哪醇酯化合物反应,得到通式(III-1)所示的化合物;
其中:R 1、R 3、R 4、R 5、R 6如通式(III-1)所定义。
当通式(I)所示的化合物为通式(IV-1)所示的化合物时,通过以下方案3制备通式(IV-1)所示的化合物:
Figure PCTCN2022128963-appb-000037
方案3
在室温,合适的碱(例如DIEA)存在下,式B1的化合物通过与亲核性胺的反应,得到式B2的化合物;然后,在加热条件下,式B2的化合物与2,4-二甲氧基苄胺反应,得到式B3的化合物;式B3的化合物可以通过金属催化(例如Pd(PPh 3) 4、K 2CO 3、Cs 2CO 3)的交叉偶联反应(例如Suzuki偶联)与硼酸或硼酸频哪醇酯化合物反应,得到式B4的化合物;用合适的酸(例如三氟乙酸)脱保护基,得到通式(IV-1)所示的化合物;
其中:R 1、R 2、R 4、R 5、R 6如通式(IV-1)所定义。
或者,可以通过以下方案4制备通式(IV-1)所示的化合物:
Figure PCTCN2022128963-appb-000038
方案4
式B6的化合物与亲核性胺在缩合剂(例如BOP)存在下反应,得到偶联产物式B7的化合物;式B7的化合物可以通过金属催化(例如Pd(PPh 3) 4、K 2CO 3、Cs 2CO 3)的交叉偶联反应(例如Suzuki偶联)与硼酸或硼酸频哪醇酯化合物反应,得到通式(IV-1)所示的化合物;
其中:R 1、R 2、R 4、R 5、R 6如通式(IV-1)所定义。
当通式(I)所示的化合物为通式(V-1)所示的化合物时,通过以下方案5制备通式(V-1)所示的化合物:
Figure PCTCN2022128963-appb-000039
方案5
在室温,合适的碱(例如DIEA)存在下,式C1的化合物通过与亲核性胺的反应,得到式C2的化合物;然后,在加热条件下,式C2的化合物与2,4-二甲氧基苄胺反应,得到式C3的化合物;式C3的化合物可以通过金属催化(例如Pd(PPh 3) 4、K 2CO 3、Cs 2CO 3)的交叉偶联反应(例如Suzuki偶联)与硼酸或硼酸频哪醇酯化合物反应,得到式C4的化合物;用合适的酸(例如三氟乙酸)脱保护基,得到通式(V-1)所示的化合物;
其中:R 2、R 4、R 5、R 6如通式(V-1)所定义。
或者,可以通过以下方案6制备通式(V-1)所示的化合物:
Figure PCTCN2022128963-appb-000040
方案6
式C6的化合物与亲核性胺在缩合剂(例如BOP)存在下反应,得到偶联产物式C7的化合物;式C7的化合物可以通过金属催化(例如Pd(PPh 3) 4、K 2CO 3、Cs 2CO 3)的交叉偶联反应(例如Suzuki偶联)与硼酸或硼酸频哪醇酯化合物反应,得到通式(V-1)所示的化合物;
其中:R 2、R 4、R 5、R 6如通式(V-1)所定义。
附图说明
图1为实施例52化合物在食蟹猴血清中的参数。
具体实施方式
进一步通过实施例来理解本发明的化合物及其制备,这些实施例说明了一些 制备或使用所述化合物的方法。然而,要理解的是,这些实施例不限制本发明。现在已知的或进一步开发的本发明的变化被认为落入本文中描述的和要求保护的本发明范围之内。
本发明化合物是利用便利的起始原料和通用的制备步骤来完成制备的。本发明给出了典型的或倾向性的反应条件,诸如反应温度、时间、溶剂、压力、反应物的摩尔比。但是除非特殊说明,其他反应条件也能采纳。优化条件可能随着具体的反应物或溶剂的使用而改变,但在通常情况下,反应优化步骤和条件都能得到确定。
另外,本发明中可能用到了一些保护基团来保护某些官能团避免不必要的反应。适宜于各种官能团的保护基以及它们的保护或脱保护条件已经为本领域技术人员广泛熟知。例如T.W.Greene和G.M.Wuts的《有机制备中的保护基团》(第3版,Wiley,New York,1999和书中的引用文献)详细描述了大量的保护基团的保护或脱保护。
化合物和中间体的分离和纯化依据具体的需求采取适当的方法和步骤,例如过滤、萃取、蒸馏、结晶、柱层析、制备薄层板色谱、制备高效液相色谱或上述方法的混合使用。其具体使用方法可参阅本发明描述的实例。当然,其他类似的分离和纯化手段也是可以采用的。可以使用常规方法(包括物理常数和波谱数据)对其进行表征。
化合物的结构是通过核磁共振(NMR)或/和质谱(MS)来确定的。NMR位移以10 -6(ppm)的单位给出。NMR的测定是用Brukerdps 300型核磁仪,测定溶剂为氘代二甲基亚砜(DMSO-d 6)、氘代氯仿(CDCl 3)、氘代甲醇(CD 3OD)、内标为四甲基硅烷(TMS)。
MS的测定用LC(Waters 2695)/MS(Quattro Premier x E)质谱仪(生产商:沃特世)(Photodiode Array Detector)。
制备液相色谱法使用lc6000高效液相色谱仪(生产商:创新通恒)。色谱柱为Daisogel C18 10μm 100A(30mm×250mm),流动相:乙腈/水。
薄层色谱法(TLC)使用青岛海洋化工GF254硅胶板,反应监测用薄层色谱法使用的硅胶板采用的规格是0.20mm~0.25mm,制备薄层色谱法使用的硅胶板采用的规格是0.5mm。
硅胶柱层析色谱法使用青岛海洋硅胶100~200目、200~300目和300~400目硅胶为载体。
本发明的已知的起始原料可以采用或按照本领域已知的方法来合成,或可购买自网化商城、北京偶合、Sigma、百灵威、易世明、上海书亚、上海伊诺凯、安耐吉化学、上海毕得等公司。
实施例中无特殊说明,反应能够均在氮气氛下进行。
氩气氛或氮气氛是指反应瓶连接一个约1L容积的氩气或氮气气球。
反应溶剂,有机溶剂或惰性溶剂各自表述为使用的该溶剂在所描述的反应条件下不参与反应,包括,如苯、甲苯、乙腈、四氢呋喃(THF)、二甲基甲酰胺(DMF)、氯仿、二氯甲烷、***、甲醇、氮-甲基吡咯碄酮(NMP)、吡啶等。实施例中无特殊说明,溶液是指水溶液。
本发明中所描述的化学反应一般在常压下进行。反应温度在-78℃至200℃之间。反应时间和条件为,例如,一个大气压下,-78℃至200℃之间,大约1至24小时内完成。如果反应过夜,则反应时间一般为16小时。实施例中无特殊说明,反应的温度为室温,为20℃~30℃。
除非另行定义,文中所使用的所有专业与科学用语与本领域熟练人员所熟悉的意义相同。此外,任何与所记载内容相似或均等的方法及材料皆可应用于本发明方法中。
实施例1:(R)-2-((2-氨基吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(1)的制备
Figure PCTCN2022128963-appb-000041
步骤1:1-((叔丁基二甲基硅烷基)氧基)丙-2-酮(1b)的制备
于室温,将羟基丙酮1a(100g,1.35mol)溶于二氯甲烷(DCM)(1L)中。将溶液降温至0℃,依次加入咪唑(175g,2.57mol)和叔丁基二甲基氯硅烷 (TBDMSCl)(245g,1.63mol)。反应液在0℃搅拌1小时,然后缓慢升至室温,继续搅拌12小时。反应完全后,将反应液用水洗涤(3×1L),有机相减压浓缩,得淡黄色液体化合物1b(200g,78.7%)。
1H-NMR(CDCl 3)δ:4.15(s,2H),2.17(s,3H),0.93(s,9H),0.09(s,6H)。
步骤2:(S,E)-N-(1-((叔丁基二甲基硅烷基)氧基)丙-2-亚基)-2-甲基丙-2-磺酰胺(1c)的制备
于室温,将化合物1b(200g,1.06mol)和S-叔丁基亚磺酰胺(129g,1.06mol)溶于四氢呋喃(3.6L)中。反应液滴加入原钛酸四异丙酯(800mL,2.66mol)。反应液在70℃搅拌12小时。反应完成后,冷却至室温。将反应液减压浓缩,将所得棕色液体加入冰水(1L),过滤除去产生的固体,加入乙酸乙酯(3×500mL)萃取,合并的有机相用饱和食盐水(500mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。所得棕色油状粗品用硅胶柱层析色谱法分离纯化(流动相:乙酸乙酯/石油醚=3%-20%),得棕黄色液体化合物1c(50g,16.1%)。
1H NMR(300MHz,氯仿-d)δ4.23(s,2H),2.32(s,3H),1.23(s,9H),0.90(s,9H),0.07(s,6H)。
LC-MS:m/z 292.2[M+H] +
步骤3:(S)-N-((R)-1-((叔丁基二甲基硅烷基)氧基)-2-甲基己-2-基)-2-甲基丙-2-磺酰胺(1d)的制备
于室温,在氮气氛下,将化合物1c(50g,0.171mol)溶于甲苯(500mL)。反应液在-78℃滴加入三甲基铝的庚烷溶液(207mL,1mol/L,0.21mol),滴加完毕后,继续搅拌0.5小时。然后在-78℃再滴加正丁基铝的正己烷溶液(102mL,2.5mol/L,0.26mol),滴加完毕后,在-78℃搅拌4小时。反应完毕后,加水(500mL)淬灭,过滤,加入乙酸乙酯(3×200mL)萃取,合并的有机相用饱和食盐水(300mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。所得黄色粗品用硅胶柱层析色谱法分离纯化(流动相:乙酸乙酯/石油醚=7%-10%),得到黄色液体化合物1d(26g,43.4%)。
1H NMR(400MHz,氯仿-d)δ3.66(s,1H),3.51(d,J=9.4Hz,1H),3.32(d,J=9.4Hz,1H),1.72–1.62(m,2H),1.35–1.24(m,4H),1.18(s,9H),1.14(s,3H),0.92–0.87(m,12H),0.05(s,3H),0.05(s,3H)。
LC-MS:m/z 350.3[M+H] +
步骤4:(R)-1-((叔丁基二甲基硅烷基)氧基)-2-甲基己-2-胺(1e)的制备
于室温,将化合物1d(26g,0.074mol)溶于四氢呋喃(250mL)和水(50mL)中,反应液加入碘单质(3.78g,0.015mol),在50℃搅拌过夜。反应完毕后,加水(200mL)稀释,反应液减压除去四氢呋喃,加入乙酸乙酯(4×200mL)萃取,合并的有机相依次用硫代硫酸钠溶液(400mL)和饱和食盐水(400mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得到棕色油状液体化合物1e(15g,82.1%), 其未经纯化直接用于下一步。
1H NMR(400MHz,氯仿-d)δ3.30(q,J=9.4Hz,2H),1.54(s,2H),1.40–1.20(m,6H),0.98(s,3H),0.93–0.87(m,12H),0.04(s,6H)。
LC-MS:m/z 246.2[M+H] +
步骤5:2,4-二氯吡啶并[4,3-d]嘧啶(1g)的制备
在室温下,将吡啶并[4,3-d]嘧啶-2,4(1H,3H)-二酮1f(50mg,0.31mmol)溶于三氯氧磷(1.5ml)中,然后缓慢加入N,N-二异丙基乙胺(DIEA)(356mg,2.75mmol),反应液升温至100℃并搅拌4小时。反应完毕,将反应液冷却至室温,用冰水(30mL)稀释,快速用乙酸乙酯(3×8mL)萃取,有机相合并后用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,所得化合物1g的乙酸乙酯溶液直接用于下步反应。
LC-MS:m/z 200.0[M+H] +
步骤6:(R)-N-((1-((叔丁基二甲基硅烷基)氧基)-2-甲基己-2-基)-2-氯吡啶并[4,3-d]嘧啶-4-胺(1h)的制备
于室温,在上述1g的乙酸乙酯溶液中加入(R)-1-((叔丁基二甲基硅烷基)氧基)-2-甲基己-2-胺1e(90.4mg,0.37mmol)和N,N-二异丙基乙胺(198mg,1.53mmol)。反应液在室温搅拌4小时。反应完毕,反应液用水(30mL)稀释,再用乙酸乙酯(3×30mL)萃取,有机相合并后用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。将所得粗品用硅胶柱层析色谱法纯化(石油醚:乙酸乙酯=1:1)。所得产品经减压浓缩,得黄色油状化合物1h(16mg,12.7%)。
LC-MS:m/z 409.2[M+H] +
步骤7:(R)-N 4-(1-((叔丁基二甲基硅烷基)氧基)-2-甲基己-2-基)-N 2-(2,4-二甲氧基苄基)吡啶并[4,3-d]嘧啶-2,4-二胺(1i)的制备
于室温,将化合物1h(16mg,0.039mmol)与2,4-二甲氧基苄胺(DMB-NH 2)(45.7mg,0.274mmol)溶于二氧六环(1mL),然后将N,N-二异丙基乙胺(15mg,0.117mmol)加入体系。反应液在100℃搅拌4小时。反应完毕,将反应冷却至室温,用水(30mL)稀释,再用乙酸乙酯(3×30mL)萃取,有机相合并后用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。将所得粗品用硅胶柱层析色谱法纯化(石油醚:乙酸乙酯=1:1)。所得产品经减压浓缩,得黄色油状化合物1i(14mg,66.7%)。
LC-MS:m/z 540.3[M+H] +
步骤8:(R)-2-((2-氨基吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(1)的制备
于室温,将化合物1i(14mg,0.027mmol)溶于三氟乙酸(TFA)(1mL),于40℃搅拌过夜。反应完全后,反应液减压浓缩,加入饱和碳酸氢钠溶液(20mL)稀释,再用乙酸乙酯(3×30mL)萃取,有机相合并后用饱和食盐水(20mL)洗 涤,无水硫酸钠干燥,过滤,滤液减压浓缩。所得粗品用制备型色谱柱分离纯化(柱型:XBridge Shield RP18 OBD柱,5um,19*150mm;流动相A:水(10mmol/L碳酸氢铵),流动相B:乙腈;流速:25mL/min;梯度:在8分钟内18%-35%乙腈;检测波长:220nm),得到白色固体化合物1(3.4mg,44%)。
1H NMR(400MHz,甲醇-d 4)δ9.04(s,1H),8.24(d,J=6.0Hz,1H),7.03(d,J=5.9Hz,1H),3.97(d,J=11.2Hz,1H),3.62(d,J=11.2Hz,1H),2.24–2.07(m,1H),1.74–1.58(m,1H),1.37(s,3H),1.32–1.16(m,4H),0.80(t,J=7.0Hz,3H)。
LC-MS:m/z 276.2[M+H] +
实施例2:(R)-2-((2-氨基-7-(2-(4-甲基哌嗪-1-基)嘧啶-5-基)吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(2)的制备
Figure PCTCN2022128963-appb-000042
步骤1:4-氨基-6-氯烟酰胺(2b)的制备
于室温,将4-氨基-6-氯烟酸2a(1.00g,5.795mmol)溶于1,4-二氧六环(15mL)中,缓慢滴加氯化亚砜(8mL),反应液于90℃搅拌反应2小时。反应液减 压浓缩,残留物用四氢呋喃(THF)(5mL)稀释。于室温,向上述反应液中缓慢滴加氨水(10mL),并继续搅拌1小时。所得混合物经减压浓缩得到黄色固体化合物2b(961mg,96.65%)。
LC-MS:m/z 172.0[M+H] +
步骤2:7-氯吡啶并[4,3-d]嘧啶-2,4(1H,3H)-二酮(2c)的制备
于室温,将化合物2b(961mg,5.60mmol)溶于N,N-二甲基甲酰胺(DMF)(10mL)中,依次加入N,N-羰基二咪唑(CDI)(3.63g,22.39mmol)和1,8-二氮杂二环十一碳-7-烯(DBU)(2.13g,13.99mmol),反应液于80℃搅拌反应1小时。反应完毕后,加水(30mL)淬灭,反应液用稀盐酸调节pH至4,加入乙酸乙酯(3×40mL)萃取,合并的有机相用饱和食盐水(100mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得到暗黄色固体化合物2c(910mg,82.23%)。
LC-MS:m/z 198.0[M+H] +
步骤3:2,4,7-三氯吡啶并[4,3-d]嘧啶(2d)的制备
于0℃,将化合物2c(457mg,2.31mmol)与三氯氧磷(5mL)混合,缓慢滴加N,N-二异丙基乙胺(1.49g,11.53mmol)。反应液在100℃搅拌反应2小时。反应完毕后,将反应液冷却至室温,减压浓缩,得黑色油状粗品化合物2d(540mg,粗品),其未经纯化直接用于下一步。
LC-MS:m/z 233.9[M+H] +
步骤4:(R)-N-((1-((叔丁基二甲基硅烷基)氧基)-2-甲基己-2-基)-2,7-二氯吡啶并[4,3-d]嘧啶-4-胺(2e)的制备
于室温,将化合物2d(540mg,2.30mmol)溶于1,4-二氧六环(5mL),依次加入(R)-1-((叔丁基二甲基硅烷基)氧基)-2-甲基己-2-胺1e(1.13g,4.61mmol)和N,N-二异丙基乙胺(2.98g,23.03mmol),室温反应1.5小时。反应完全后,反应液加水(30mL)稀释,用乙酸乙酯(3×30mL)萃取,合并的有机相用饱和食盐水(100mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,所得粗品用硅胶柱层析色谱法分离纯化(流动相:乙酸乙酯/石油醚=0-5%),得到黄色油状化合物2e(367mg,35.93%)。
LC-MS:m/z 443.2[M+H] +
步骤5:(R)-N 4-(1-((叔丁基二甲基硅烷基)氧基)-2-甲基己-2-基)-7-氯-N 2-(2,4-二甲氧基苄基)吡啶并[4,3-d]嘧啶-2,4-二胺(2f)的制备
于室温,将化合物2e(367mg,0.83mmol)溶于1,4-二氧六环(5mL),依次加入2,4-二甲氧基苄胺(969mg,5.80mmol)和N,N-二异丙基乙胺(321mg,2.48mmol)。反应液于100℃搅拌反应过夜。反应完全后,冷却至室温。反应液加水(20mL)稀释,再用二氯甲烷(3×20mL)萃取,有机相合并后用饱和氯化铵溶液(2×30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,所得粗品用硅胶柱层析色谱法分离纯化(流动相:乙酸乙酯/石油醚=0-8%),得到淡黄色固体化合物 2f(374mg,78.70%)。
LC-MS:m/z 574.3[M+H] +
步骤6:(R)-N 4-(1-((叔丁基二甲基硅烷基)氧基)-2-甲基己-2-基)-N 2-(2,4-二甲氧基苄基)-7-(2-(4-甲基哌嗪-1-基)嘧啶-5-基)吡啶并[4,3-d]嘧啶-2,4-二胺(2g)的制备
于室温,将化合物2f(204mg,0.36mmol)溶于1,4-二氧六环(2mL)和水(0.4mL),依次加入2-(4-甲基哌嗪-1-基)嘧啶-5-硼酸频那醇酯(CAS:942922-07-8)(2h)(264mg,1.05mmol)、碳酸钾(148mg,1.07mmol)和[1,1’-双(二苯基磷基)二茂铁]二氯化钯(Pd(dppf)Cl 2)(26mg,0.036mmol)。反应液在氮气氛下于95℃搅拌过夜。反应完全后,冷却至室温,加水(20mL)稀释,用乙酸乙酯(3×20mL)萃取,合并的有机相用饱和食盐水(40mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,所得粗品用制备薄层色谱法分离纯化(流动相:乙酸乙酯/石油醚=1:2),得黄色油状化合物2g(33mg,12.97%)。
LC-MS:m/z 716.4[M+H] +
步骤7:(R)-2-((2-氨基-7-(2-(4-甲基哌嗪-1-基)嘧啶-5-基)吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(2)的制备
于室温,将化合物2g(33mg,0.046mmol)溶于三氟乙酸(1mL)。反应液于40℃搅拌过夜。反应完全后,反应液减压浓缩,所得粗品用制备薄层色谱法分离纯化(流动相:二氯甲烷/甲醇/三乙胺=100:20:1),得到类白色固体化合物2(6.4mg,28.66%)。
1H NMR(300MHz,甲醇-d 4)δ9.19(d,J=0.7Hz,1H),8.96(s,2H),7.40(d,J=0.7Hz,1H),4.09(d,J=11.2Hz,1H),3.96(t,J=5.2Hz,4H),3.75(d,J=11.3Hz,1H),2.55(t,J=5.2Hz,4H),2.37(s,3H),2.31–2.20(m,1H),1.86–1.72(m,1H),1.50(s,3H),1.42–1.32(m,4H),0.96–0.89(m,3H)。
LC-MS:m/z 452.2[M+H] +
实施例3:(R)-2-((2-氨基-7-(6-(4-甲基哌嗪-1-基)吡啶-3-基)吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(3)的制备
Figure PCTCN2022128963-appb-000043
Figure PCTCN2022128963-appb-000044
与实施例2的制备方法相同,除了将化合物2-(4-甲基哌嗪-1-基)嘧啶-5-硼酸频那醇酯(2h)替换为2-(4-甲基哌嗪-1-基)吡啶-5-硼酸频那醇酯(CAS:918524-63-7)(3b),所得粗品用制备型色谱柱分离纯化(柱型:SunFire Prep C18 OBD柱,5um,19*150mm;流动相A:水(0.1%的甲酸),流动相B:乙腈;流速:25mL/min;梯度:在8分钟内6%-17%乙腈;检测波长:220nm),得到化合物3的甲酸盐。
1H NMR(300MHz,甲醇-d 4)δ9.41(s,1H),8.95(d,J=2.0Hz,1H),8.35(d,J=8.9Hz,1H),7.68(s,1H),7.07(d,J=8.8Hz,1H),4.21(d,J=11.4Hz,1H),4.13–3.88(m,4H),3.74(d,J=11.3Hz,1H),3.45–3.35(m,4H),2.96(s,3H),2.38–2.18(m,1H),1.87–1.72(m,1H),1.56(s,3H),1.49–1.29(m,4H),1.02–0.87(m,3H)。
LC-MS:m/z 451.3[M+H] +
实施例4:(R)-2-((2-氨基-7-(5-甲基-6-(4-甲基哌嗪-1-基)吡啶-3-基)吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(4)的制备
Figure PCTCN2022128963-appb-000045
Figure PCTCN2022128963-appb-000046
步骤1:(5-甲基-6-(4-甲基哌嗪-1-基)吡啶-3-基)硼酸(4b)的制备
于室温,将化合物1-(5-溴-3-甲基吡啶-2-基)-4-甲基哌嗪4a(500mg,1.85mmol)溶于1,4-二氧六环(10.0mL)中,依次加入联硼酸频那醇酯(B 2(Pin) 2)(705mg,2.78mmol)、醋酸钾(545mg,5.55mmol)和[1,1′-双(二苯基膦基)二茂铁]二氯化钯二氯甲烷(151mg,0.19mmol)。反应液在氮气氛下,在80℃搅拌3小时。反应完全后,反应液用水(20mL)稀释,再用乙酸乙酯(2×20mL)萃取,所得水相减压浓缩,粗品用反相柱层析色谱法分离纯化(柱型:艾杰尔C18柱;流动相A:水(0.1%的甲酸),流动相B:乙腈;流速:60mL/min;梯度:在10分钟内5%-20%乙腈;检测波长:220/254nm),得到灰白色的固体化合物4b(370mg,85.04%)。
LC-MS:m/z 236.1[M+H] +
其余步骤与实施例2的制备方法相同,除了将化合物2-(4-甲基哌嗪-1-基)嘧啶-5-硼酸频那醇酯(2h)替换为(5-甲基-6-(4-甲基哌嗪-1-基)吡啶-3-基)硼酸(4b),所得粗品用制备型色谱柱分离纯化(柱型:XBridge Prep C18 OBD柱,5um,19*150mm;流动相A:水(10mmol/L碳酸氢铵),流动相B:乙腈;流速:25mL/min;梯度:在8分钟内22%-46%乙腈;检测波长:254/220nm),制得化合物4。
1H NMR(400MHz,甲醇-d 4)δ9.19(d,J=0.7Hz,1H),8.69(d,J=2.4Hz,1H),8.13–8.08(m,1H),7.44(d,J=0.7Hz,1H),4.07(d,J=11.2Hz,1H),3.74(d,J=11.2Hz,1H),3.34–3.23(m,4H),2.75–2.61(m,4H),2.39(s,3H),2.39(s,3H),2.30–2.18(m,1H),1.83–1.72(m,1H),1.48(s,3H),1.42–1.30(m,4H),0.94–0.86(m,3H)。
LC-MS:m/z 465.2[M+H] +
实施例5:(R)-2-((2-氨基-7-(6-(吡咯烷-1-基)吡啶-3-基)吡啶并[4,3-d]嘧啶-4-基) 氨基)-2-甲基己-1-醇(5)的制备
Figure PCTCN2022128963-appb-000047
与实施例2的制备方法相同,除了将化合物2-(4-甲基哌嗪-1-基)嘧啶-5-硼酸频那醇酯(2h)替换为6-(吡咯烷-1-基)吡啶-3-硼酸(CAS:1150114-75-2)(5b),所得粗品用制备型色谱柱分离纯化(柱型:SunFire Prep C18 OBD柱,5um,19*150mm;流动相A:水(0.1%的甲酸),流动相B:乙腈;流速:25mL/min;梯度:在8分钟内7%-21%乙腈;检测波长:220nm),制得化合物5的甲酸盐。
1H NMR(300MHz,甲醇-d 4)δ9.36(s,1H),8.80(d,J=1.9Hz,1H),8.33–8.15(m,1H),7.54(s,1H),6.67(d,J=8.9Hz,1H),4.21(d,J=11.3Hz,1H),3.75(d,J=11.4Hz,1H),3.64–3.48(m,4H),2.38–2.20(m,1H),2.18–2.01(m,4H),1.88–1.70(m,1H),1.56(s,3H),1.50–1.25(m,4H),1.03–0.88(m,3H)。
LC-MS:m/z 422.2[M+H] +
实施例6:(R)-2-((2-氨基-7-(6-(哌啶-1-基)吡啶-3-基)吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(6)的制备
Figure PCTCN2022128963-appb-000048
Figure PCTCN2022128963-appb-000049
与实施例2的制备方法相同,除了将化合物2-(4-甲基哌嗪-1-基)嘧啶-5-硼酸频那醇酯(2h)替换为2-(哌啶-1-基)吡啶-5-硼酸频那醇酯(CAS:852228-08-1)(6b),所得粗品用制备型色谱柱分离纯化(柱型:SunFire Prep C18 OBD柱,5um,19*150mm;流动相A:水(0.1%的甲酸),流动相B:乙腈;流速:25mL/min;梯度:在8分钟内13%-29%乙腈;检测波长:220nm),制得化合物6的甲酸盐。
1H NMR(400MHz,甲醇-d 4)δ9.35(s,1H),8.82(d,J=2.3Hz,1H),8.20(dd,J=9.1,2.5Hz,1H),7.54(s,1H),6.90(d,J=9.1Hz,1H),4.18(d,J=11.3Hz,1H),3.72(d,J=11.3Hz,1H),3.70–3.64(m,4H),2.34–2.19(m,1H),1.84–1.70(m,3H),1.70–1.61(m,4H),1.53(s,3H),1.45–1.29(m,4H),0.98–0.87(m,3H)。
LC-MS:m/z 436.4.1[M+H] +
实施例7:(R)-2-((2-氨基-7-(6-(4-羟基哌啶-1-基)吡啶-3-基)吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇甲酸盐(7)的制备
Figure PCTCN2022128963-appb-000050
与实施例2的制备方法相同,除了将化合物2-(4-甲基哌嗪-1-基)嘧啶-5-硼酸频那醇酯(2h)替换为6-(4-羟基哌啶-1-基)吡啶-3-硼酸频那醇酯(CAS:1251948-86-3)(7b),所得粗品用制备型色谱柱分离纯化(柱型:XBridge Shield RP18 OBD柱,5um,19*150mm;流动相A:水(0.1%的甲酸),流动相B:乙腈;流速:25mL/min;梯度:在8分钟内10%-32%乙腈;检测波长:254nm),制得化合物7的甲酸盐。
1H NMR(300MHz,甲醇-d 4)δ9.35(d,J=1.6Hz,1H),8.85(d,J=2.3Hz,1H),8.22(d,J=8.9Hz,1H),7.56(s,1H),6.95(d,J=9.2Hz,1H),4.30–4.12(m,3H),4.00–3.84(m,1H),3.74(d,J=11.3Hz,1H),3.32–3.23(m,2H),2.37–2.20(m,1H),2.04–1.91(m,2H),1.88–1.71(m,1H),1.65–1.48(m,5H),1.46–1.23(m,4H),1.02–0.88(m,3H)。
LC-MS:m/z 452.2[M+H] +
实施例8:(R)-2-((2-氨基-7-(咪唑并[1,2-a]吡啶-6-基)吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(8)的制备
Figure PCTCN2022128963-appb-000051
与实施例2的制备方法相同,除了将化合物2-(4-甲基哌嗪-1-基)嘧啶-5-硼酸频那醇酯(2h)替换为咪唑并[1,2-a]吡啶-6-硼酸频那醇酯(CAS:1204742-76-6)(8b),所得粗品用制备型色谱柱分离纯化(柱型:SunFire Prep C18 OBD柱,5um,19*150mm;流动相A:水(0.1%的甲酸),流动相B:乙腈;流速:25mL/min;梯度:在8分钟内7%-15%乙腈;检测波长:254nm),制得化合物8的甲酸盐。
1H NMR(300MHz,甲醇-d 4)δ9.46(s,1H),9.41(s,1H),8.20–8.12(m,1H),8.07(s,1H),7.87–7.68(m,3H),4.23(d,J=11.4Hz,1H),3.76(d,J=11.4Hz,1H),2.38–2.21(m,1H),1.90–1.74(m,1H),1.58(s,3H),1.50–1.27(m,4H),1.03–0.88(m,3H)。
LC-MS:m/z 392.3[M+H] +
实施例9:(R)-2-((2-氨基-7-(6-(4-环丙基哌嗪-1-基)吡啶-3-基)吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(9)的制备
Figure PCTCN2022128963-appb-000052
步骤1:2-氨基-7-氯吡啶并[4,3-d]嘧啶-4(3H)-酮(9b)的制备
于室温,将4,6-二氯烟酸9a(5.0g,26.04mmol)和盐酸胍(2.74g,28.646mmol)溶于N,N-二甲基甲酰胺(100mL)中,反应液中依次加入碳酸铯(16.97g,52.084mmol)和碘化亚铜(0.99g,5.208mmol)。反应液在氮气氛下,110℃搅拌过夜。反应完毕后,将反应液中固体过滤除去,滤液减压浓缩。所得残留物加入饱和碳酸钠溶液,用正丁醇(5×50mL)萃取。合并的有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得到类白色至淡绿色固体化合物9b(4.2g,82.04%)。
LC-MS:m/z 197.0[M+H] +
步骤2:(R)-2-((2-氨基-7-氯吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(9c)的制备
于室温,将化合物9b(500.00mg,2.543mmol)、(R)-2-氨基-2-甲基己-1-醇(500.61mg,3.815mmol)、卡特缩合剂(BOP)(1462.34mg,3.306mmol)、1,8-二氮杂二环十一碳-7-烯(DBU)(1161.58mg,7.629mmol)溶于N,N-二甲基甲酰 胺(8mL)中。反应液在氮气氛下于室温搅拌过夜。反应完毕后,加水(50mL)淬灭反应,体系中加入乙酸乙酯(3×40mL)萃取,合并的有机相用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,所得粗品用硅胶柱层析色谱法分离纯化(流动相:乙酸乙酯/石油醚=1:1),得黄色半固体化合物9c(200mg,22.84%)。
LC-MS:m/z 310.1[M+H] +
步骤3:2-(4-环丙基哌嗪-1-基)吡啶-5-硼酸频那醇酯(9d)的制备
于室温,将6-氯吡啶-3-硼酸频那醇酯(50mg,0.209mmol)溶于二甲基亚砜(1mL)中,反应液中加入1-环丙基哌嗪(52.69mg,0.418mmol),在氮气氛下,150℃搅拌3小时。反应完毕后,加水(20mL)淬灭反应,体系中加入乙酸乙酯(3×20mL)萃取,合并的有机相用饱和食盐水(40mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得棕色固体化合物9d(72mg,粗品),其未经纯化直接用于下一步。
LC-MS:m/z 330.2[M+H] +
步骤4:(R)-2-((2-氨基-7-(6-(4-环丙基哌嗪-1-基)吡啶-3-基)吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(9)的制备
于室温,将化合物9c(32.00mg,0.103mmol)和化合物9d(68.02mg,0.206mmol)溶于1,4-二氧六环(1mL)和水(0.1mL),依次加入碳酸钾(42.83mg,0.309mmol)和[1,1’-双(二苯基磷基)二茂铁]二氯化钯(16.83mg,0.021mmol)。在氮气氛下,将反应液于95℃搅拌过夜。反应完全后,冷却至室温,加水(10mL)稀释,用乙酸乙酯(3×20mL)萃取,合并的有机相用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,所得粗品用制备薄层色谱法分离纯化(流动相:乙酸乙酯/三乙胺=20:1),得黄色油状粗品。产品进一步经制备型色谱柱分离纯化(柱型:XSelect CSH Prep C18 OBD柱,5um,19*150mm;流动相A:水(0.05%氨水),流动相B:乙腈;流速:25mL/min;梯度:在8分钟内32%-52%乙腈;检测波长:254/220nm),得到类白色固体化合物9(17.5mg,35.55%)。
1H NMR(300MHz,甲醇-d 4)δ9.18(d,J=0.7Hz,1H),8.77(d,J=2.4Hz,1H),8.17(dd,J=9.0,2.5Hz,1H),7.41(d,J=0.8Hz,1H),6.95(d,J=9.1Hz,1H),4.08(d,J=11.3Hz,1H),3.75(d,J=11.3Hz,1H),3.70–3.60(m,4H),2.84–2.75(m,4H),2.36–2.19(m,1H),1.85–1.69(m,2H),1.50(s,3H),1.43–1.32(m,4H),0.98–0.88(m,3H),0.60–0.47(m,4H)。
LC-MS:m/z 477.4[M+H] +
实施例10:(R)-4-(5-(2-氨基-4-((1-羟基-2-甲基己-2-基)氨基)吡啶并[4,3-d]嘧啶-7-基)吡啶-2-基)-1-甲基哌嗪-2-酮(10)的制备
Figure PCTCN2022128963-appb-000053
与实施例9的制备方法相同,除了将1-环丙基哌嗪替换为1-甲基哌嗪-2-酮,制得化合物10。
1H NMR(300MHz,甲醇-d 4)δ9.20(s,1H),8.82(d,J=2.4Hz,1H),8.22(dd,J=8.9,2.5Hz,1H),7.43(s,1H),6.95(d,J=9.0Hz,1H),4.26(s,2H),4.09(d,J=11.3Hz,1H),4.02–3.94(m,2H),3.75(d,J=11.3Hz,1H),3.61–3.53(m,2H),3.06(s,3H),2.34–2.19(m,1H),1.87–1.71(m,1H),1.50(s,3H),1.45–1.31(m,4H),0.99–0.87(m,3H)。
LC-MS:m/z 465.1[M+H] +
实施例11:(R)-2-((2-氨基-7-(6-(4-乙基哌嗪-1-基)吡啶-3-基)吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(11)的制备
Figure PCTCN2022128963-appb-000054
Figure PCTCN2022128963-appb-000055
与实施例9的制备方法相同,除了将化合物1-环丙基哌嗪替换为1-乙基哌嗪,制得化合物11。
1H NMR(400MHz,甲醇-d 4)δ9.16(s,1H),8.76(d,J=2.4Hz,1H),8.15(dd,J=9.0,2.5Hz,1H),7.39(s,1H),6.94(d,J=9.0Hz,1H),4.06(d,J=11.3Hz,1H),3.73(d,J=11.2Hz,1H),3.71–3.64(m,4H),2.63(t,J=5.0Hz,4H),2.53(q,J=7.2Hz,2H),2.32–2.18(m,1H),1.84–1.71(m,1H),1.48(s,3H),1.44–1.30(m,4H),1.17(t,J=7.3Hz,3H),0.91(t,J=6.9Hz,3H)。
LC-MS:m/z 465.2[M+H] +
实施例12:(R)-2-((2-氨基-7-(6-(4-异丙基哌嗪-1-基)吡啶-3-基)吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(12)的制备
Figure PCTCN2022128963-appb-000056
与实施例9的制备方法相同,除了将化合物1-环丙基哌嗪替换为1-异丙基哌嗪,制得化合物12。
1H NMR(300MHz,甲醇-d 4)δ9.18(d,J=0.7Hz,1H),8.78(d,J=2.4Hz,1H),8.17(dd,J=9.0,2.5Hz,1H),7.41(d,J=0.7Hz,1H),6.96(d,J=9.1Hz,1H),4.08(d,J=11.3Hz,1H),3.75(d,J=11.3Hz,1H),3.72–3.65(m,4H),2.83–2.77(m,1H),2.74(t,J=5.2Hz,4H),2.33–2.19(m,1H),1.84–1.70(m,1H),1.50(s,3H),1.45–1.31(m,4H),1.16(d,J=6.5Hz,6H),0.99–0.89(m,3H)。
LC-MS:m/z 479.2[M+H] +
实施例13:(R)-2-((2-氨基-7-(6-(4-(2-甲氧基乙基)哌嗪-1-基)吡啶-3-基)吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(13)的制备
Figure PCTCN2022128963-appb-000057
与实施例9的制备方法相同,除了将化合物1-环丙基哌嗪替换为1-(2-甲氧基乙基)哌嗪,制得化合物13。
1H NMR(400MHz,甲醇-d 4)δ9.16(s,1H),8.75(d,J=2.5Hz,1H),8.15(dd,J=9.0,2.5Hz,1H),7.39(s,1H),6.93(d,J=9.0Hz,1H),4.07(d,J=11.3Hz,1H),3.73(d,J=11.3Hz,1H),3.67(t,J=5.2Hz,4H),3.60(t,J=5.5Hz,2H),3.37(s,3H),2.71–2.62(m,6H),2.32–2.17(m,1H),1.83–1.71(m,1H),1.48(s,3H),1.42–1.30(m,4H),0.91(t,J=6.9Hz,3H)。
LC-MS:m/z 495.3[M+H] +
实施例14:(2R)-2-((2-氨基-7-(6-(3,4-二甲基哌嗪-1-基)吡啶-3-基)吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(14)的制备
Figure PCTCN2022128963-appb-000058
与实施例9的制备方法相同,除了将化合物1-环丙基哌嗪替换为1,2-二甲基哌嗪,制得化合物14。
1H NMR(400MHz,甲醇-d 4)δ9.19–9.13(m,1H),8.75(d,J=2.5Hz,1H),8.19–8.10(m,1H),7.42–7.34(m,1H),6.94(d,J=9.1Hz,1H),4.31–4.17(m,2H),4.06(d,J=11.3Hz,1H),3.73(d,J=11.3Hz,1H),3.18–3.05(m,1H),3.01–2.89(m,1H),2.80–2.66(m,1H),2.44–2.32(m,4H),2.32–2.18(m,2H),1.83–1.70(m,1H),1.48(s,3H),1.43–1.26(m,4H),1.19(d,J=6.3Hz,3H),0.91(d,J=6.9Hz,3H)。
LC-MS:m/z 465.5[M+H] +
实施例15:(2R)-2-((2-氨基-7-(4-甲基-6-(4-甲基哌嗪-1-基)吡啶-3-基)吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(15)的制备
Figure PCTCN2022128963-appb-000059
Figure PCTCN2022128963-appb-000060
与实施例9的制备方法相同,除了将化合物6-氯吡啶-3-硼酸频那醇酯替换为4-甲基-6-氯吡啶-3-硼酸频那醇酯,并将1-环丙基哌嗪替换为1-甲基哌嗪,制得化合物15。
1H NMR(400MHz,甲醇-d 4)δ9.18(s,1H),8.12(s,1H),7.13(s,1H),6.78(s,1H),4.08(d,J=11.3Hz,1H),3.73(d,J=11.3Hz,1H),3.65–3.58(m,4H),2.59(t,J=5.1Hz,4H),2.37(s,3H),2.34(s,3H),2.30–2.20(m,1H),1.82–1.70(m,1H),1.48(s,3H),1.40–1.30(m,4H),0.97–0.87(m,3H)。
LC-MS:m/z 465.4[M+H] +
实施例16:(R)-2-((2-氨基-7-(2-甲基-6-(4-甲基哌嗪-1-基)吡啶-3-基)吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(16)的制备
Figure PCTCN2022128963-appb-000061
与实施例9的制备方法相同,除了将化合物6-氯吡啶-3-硼酸频那醇酯替换为2-甲基-6-氯吡啶-3-硼酸频那醇酯,并将1-环丙基哌嗪替换为1-甲基哌嗪,制得化合物16。
1H NMR(400MHz,甲醇-d 4)δ9.16(s,1H),7.60(d,J=8.7Hz,1H),7.12(s,1H), 6.72(d,J=8.7Hz,1H),4.08(d,J=11.2Hz,1H),3.73(d,J=11.2Hz,1H),3.68–3.57(m,4H),2.57(t,J=5.1Hz,4H),2.43(s,3H),2.35(s,3H),2.31–2.20(m,1H),1.82–1.70(m,1H),1.48(s,3H),1.41–1.27(m,4H),0.91(t,J=6.9Hz,3H)。
LC-MS:m/z 465.4[M+H] +
实施例17:(R)-2-((2-氨基-7-(4-氟-6-(4-甲基哌嗪-1-基)吡啶-3-基)吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(17)的制备
Figure PCTCN2022128963-appb-000062
步骤1:1-(4-氟吡啶-2-基)-4-甲基哌嗪(17a)的制备
于室温,将2-溴-4-氟吡啶(400mg,2.273mmol)和N-甲基哌嗪溶于甲苯(10.0mL)中,然后依次加入Rac-BINAP-Pd-G3(225.56mg,0.227mmol)和碳酸铯(2962.20mg,9.092mmol)。反应液在氮气氛下在95℃搅拌过夜。反应完全后,反应液用水(30mL)稀释,再用乙酸乙酯(3×20mL)萃取,有机相合并后用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。粗品用柱层析色谱法分离纯化(流动相:石油醚/乙酸乙酯=2:1),得到深黄色油状化合物17a(119mg,26.82%)。
LC-MS:m/z 196.1[M+H] +
步骤2:1-(5-溴-4-氟吡啶-2-基)-4-甲基哌嗪(17b)的制备
于室温,将化合物1-(4-氟吡啶-2-基)-4-甲基哌嗪17a(119mg,0.610mmol)溶于乙腈(ACN)(3mL)中,加入N-溴代丁二酰亚胺(NBS)(130.18mg,0.732mmol),在氮气氛下于室温避光反应过夜。反应完全后减压浓缩,残留物中加入水(10mL)稀释,再用乙酸乙酯(3×10mL)萃取,有机相合并后用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。粗品用柱层析色谱法分离纯化(流动相:石油醚/乙酸乙酯=2:1),得到橘黄色半固体化合物17b(166mg, 99.35%)。
LC-MS:m/z 274.0[M+H] +
步骤3:2-(4-甲基哌嗪-1-基)-4-氟吡啶-5-硼酸(17c)的制备
于室温,将化合物1-(5-溴-4-氟吡啶-2-基)-4-甲基哌嗪17b(166mg,0.606mmol)溶于1,4-二氧六环(3.0mL)中,然后依次加入联硼酸频那醇酯(230.65mg,0.909mmol)、醋酸钾(178.29mg,1.818mmol)和[1,1′-双(二苯基膦基)二茂铁]二氯化钯二氯甲烷(98.66mg,0.121mmol),反应液在氮气氛下于80℃搅拌过夜。反应完全后,反应液用水(10mL)稀释,再用乙酸乙酯(2×10mL)萃取,所得水相减压浓缩,得到棕色固体化合物17c(150mg,粗品),其未经纯化直接用于下一步。
LC-MS:m/z 240.1[M+H] +
其余步骤与实施例9相同,除了将2-(4-环丙基哌嗪-1-基)吡啶-5-硼酸频那醇酯(9d)替换为2-(4-甲基哌嗪-1-基)-4-氟吡啶-5-硼酸(17c),制得化合物17。
1H NMR(400MHz,甲醇-d 4)δ9.08(s,1H),8.58(d,J=11.4Hz,1H),7.30(d,J=1.0Hz,1H),6.57(d,J=14.8Hz,1H),3.97(d,J=11.3Hz,1H),3.63(d,J=11.3Hz,1H),3.57(t,J=5.2Hz,4H),2.46(t,J=5.1Hz,4H),2.26(s,3H),2.21–2.08(m,1H),1.72–1.60(m,1H),1.38(s,3H),1.32–1.18(m,4H),0.81(t,J=6.9Hz,3H)。
LC-MS:m/z 469.3[M+H] +
实施例18:(R)-2-((2-氨基-7-(6-(4-(二甲氨基)哌啶-1-基)吡啶-3-基)吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(18)的制备
Figure PCTCN2022128963-appb-000063
与实施例9的制备方法相同,除了将1-环丙基哌嗪替换为4-二甲氨基哌啶, 制得化合物18。
1H NMR(400MHz,甲醇-d 4)δ9.15(s,1H),8.73(s,1H),8.12(dd,J=8.9,2.2Hz,1H),7.37(s,1H),6.94(d,J=9.1Hz,1H),4.50(d,J=13.5Hz,2H),4.06(d,J=11.3Hz,1H),3.73(d,J=11.3Hz,1H),2.93(t,J=12.9Hz,2H),2.53(s,1H),2.41–2.30(m,6H),2.30–2.18(m,1H),2.06–1.94(m,2H),1.83–1.71(m,1H),1.61–1.44(m,5H),1.42–1.26(m,4H),0.91(t,J=6.9Hz,3H)。
LC-MS:m/z 479.3[M+H] +
实施例19:(R)-2-((2-氨基-7-(6-(4-(2-羟乙基)哌嗪-1-基)吡啶-3-基)吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(19)的制备
Figure PCTCN2022128963-appb-000064
与实施例9的制备方法相同,除了将1-环丙基哌嗪替换为N-羟乙基哌嗪,制得化合物19。
1H NMR(400MHz,甲醇-d 4)δ9.31(s,1H),8.86(s,1H),8.25(d,J=8.9Hz,1H),7.57–7.50(m,1H),6.99(d,J=9.0Hz,1H),4.14(d,J=11.3Hz,1H),3.91–3.76(m,6H),3.72(d,J=11.3Hz,1H),3.08–2.97(m,4H),2.97–2.87(m,2H),2.32–2.20(m,1H),1.82–1.72(m,1H),1.52(s,3H),1.43–1.28(m,4H),0.92(t,J=6.9Hz,3H)。
LC-MS:m/z 481.4[M+H] +
实施例20:(R)-2-((2-氨基-7-(6-(4-丙基哌嗪-1-基)吡啶-3-基)吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(20)的制备
Figure PCTCN2022128963-appb-000065
步骤1:2-(4-丙基哌嗪-1-基)吡啶-5-硼酸频那醇酯(20a)的制备
于室温,将1-正丙基哌嗪二溴酸盐(484.39mg,1.670mmol)、碳酸钾(464.99mg,3.340mmol)溶于二甲基亚砜(5mL)中,加入2-氯吡啶-5-硼酸频那醇酯(200mg,0.835mmol),在氮气氛下,150℃搅拌2小时。反应完毕后,加水(30mL)淬灭反应,体系中加入乙酸乙酯(3×15mL)萃取,合并的有机相用饱和食盐水(15mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得棕黄色液体化合物20a(549mg,粗品),其未经纯化直接用于下一步。
LC-MS:m/z 332.2[M+H] +
其余步骤与实施例9相同,除了2-(4-环丙基哌嗪-1-基)吡啶-5-硼酸频那醇酯(9d)替换为2-(4-丙基哌嗪-1-基)吡啶-5-硼酸频那醇酯(20a),制得化合物20。
1H NMR(400MHz,甲醇-d 4)δ9.16(s,1H),8.75(d,J=2.4Hz,1H),8.15(dd,J=9.0,2.5Hz,1H),7.39(s,1H),6.93(d,J=9.0Hz,1H),4.06(d,J=11.3Hz,1H),3.73(d,J=11.3Hz,1H),3.67(t,J=5.1Hz,4H),2.63(t,J=5.1Hz,4H),2.48–2.36(m,2H),2.31–2.18(m,1H),1.83–1.70(m,1H),1.69–1.53(m,2H),1.48(s,3H),1.42–1.28(m,4H),0.96(t,J=7.4Hz,3H),0.91(t,J=6.9Hz,3H)。
LC-MS:m/z 479.3[M+H] +
实施例21:(R)-2-((2-氨基-7-(6-(4-(2,2-二氟乙基)哌嗪-1-基)吡啶-3-基)吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(21)的制备
Figure PCTCN2022128963-appb-000066
Figure PCTCN2022128963-appb-000067
与实施例20的制备方法相同,除了将1-正丙基哌嗪二溴酸盐替换为1-(2,2-二氟乙基)哌嗪盐酸盐,制得化合物21。
1H NMR(300MHz,甲醇-d 4)δ9.28(s,1H),8.82(s,1H),8.21(d,J=8.8Hz,1H),7.50(s,1H),6.94(d,J=8.9Hz,1H),6.05(tt,J=55.9,4.3Hz,1H),4.15(d,J=11.3Hz,1H),3.75(d,J=11.3Hz,1H),3.70(t,J=5.1Hz,4H),2.84(td,J=15.2,4.3Hz,2H),2.75(t,J=5.0Hz,4H),2.36–2.17(m,1H),1.88–1.70(m,1H),1.53(s,3H),1.47–1.26(m,4H),1.00–0.87(m,3H)。
LC-MS:m/z 501.4[M+H] +
实施例22:(R)-2-((2-氨基-7-(6-(4-(2-氟乙基)哌嗪-1-基)吡啶-3-基)吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(22)的制备
Figure PCTCN2022128963-appb-000068
步骤1:2-(4-(2-氟乙基)哌嗪-1-基)吡啶-5-硼酸频那醇酯(22a)的制备
于室温,将2-氯吡啶-5-硼酸频那醇酯(150mg,0.626mmol)和1-(2-氟乙基)哌嗪二盐酸盐(256.90mg,1.252mmol)溶于二甲基亚砜(2mL)中,加入N,N-二异丙基乙胺(323.77mg,2.504mmol),在氮气氛下,150℃搅拌2小时。反应完毕后,加水(20mL)淬灭反应,体系中加入乙酸乙酯(3×20mL)萃取,合并的 有机相用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得棕色半固体化合物22a(198mg,粗品),其未经纯化直接用于下一步。
LC-MS:m/z 336.2[M+H] +
其余步骤与实施例9相同,除了将2-(4-环丙基哌嗪-1-基)吡啶-5-硼酸频那醇酯(9d)替换为2-(4-(2-氟乙基)哌嗪-1-基)吡啶-5-硼酸频那醇酯(22a),制得化合物22。
1H NMR(400MHz,甲醇-d 4)δ9.19(s,1H),8.72(d,J=2.4Hz,1H),8.11(dd,J=9.0,2.5Hz,1H),7.41(s,1H),6.83(d,J=9.1Hz,1H),4.66–4.56(m,1H),4.54–4.44(m,1H),4.05(d,J=11.4Hz,1H),3.68–3.56(m,5H),2.79–2.72(m,1H),2.71–2.66(m,1H),2.62(t,J=5.1Hz,4H),2.25–2.09(m,1H),1.75–1.61(m,1H),1.42(s,3H),1.35–1.16(m,4H),0.82(t,J=6.9Hz,3H)。
LC-MS:m/z 483.4[M+H] +
实施例23:(R)-2-((2-氨基-7-(6-(4-(2,2,2-三氟乙基)哌嗪-1-基)吡啶-3-基)吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(23)的制备
Figure PCTCN2022128963-appb-000069
与实施例22的制备方法相同,除了1-(2-氟乙基)哌嗪二盐酸盐替换为1-(2,2,2-三氟乙基)哌嗪二盐酸盐,制得化合物23。
1H NMR(400MHz,甲醇-d 4)δ9.30(s,1H),8.82(d,J=2.4Hz,1H),8.25–8.16(m,1H),7.52(s,1H),6.93(d,J=9.0Hz,1H),4.15(d,J=11.3Hz,1H),3.76–3.65(m,5H),3.14(q,J=9.8Hz,2H),2.80(t,J=5.1Hz,4H),2.33–2.19(m,1H),1.83–1.71(m,1H),1.52(s,3H),1.41–1.32(m,4H),0.92(t,J=6.9Hz,3H).。
LC-MS:m/z 519.5[M+H] +
实施例24:(R)-2-((2-氨基-7-(6-(4-丙基哌啶-1-基)吡啶-3-基)吡啶并[4,3-d]嘧啶 -4-基)氨基)-2-甲基己-1-醇(24)的制备
Figure PCTCN2022128963-appb-000070
与实施例22的制备方法相同,除了将1-(2-氟乙基)哌嗪二盐酸盐替换为4-正丙基哌啶盐酸盐,制得化合物24。
1H NMR(300MHz,甲醇-d 4)δ9.16(s,1H),8.73(d,J=2.4Hz,1H),8.12(dd,J=9.0,2.6Hz,1H),7.39(s,1H),6.92(d,J=9.2Hz,1H),4.47–4.35(m,2H),4.08(d,J=11.2Hz,1H),3.75(d,J=11.3Hz,1H),3.00–2.87(m,2H),2.32–2.20(m,1H),1.89–1.76(m,3H),1.67–1.54(m,1H),1.50(s,3H),1.48–1.19(m,10H),1.00–0.89(m,6H)。
LC-MS:m/z 478.5[M+H] +
实施例25:(R)-2-((2-氨基-7-(6-(3-乙氧基氮杂环丁烷-1-基)吡啶-3-基)吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(25)的制备
Figure PCTCN2022128963-appb-000071
Figure PCTCN2022128963-appb-000072
与实施例22的制备方法相同,除了将1-(2-氟乙基)哌嗪二盐酸盐替换为3-乙氧基氮杂环丁烷盐酸盐,制得化合物25。
1H NMR(400MHz,甲醇-d 4)δ9.15(s,1H),8.68(d,J=2.4Hz,1H),8.14(dd,J=8.8,2.4Hz,1H),7.36(s,1H),6.53(d,J=8.8Hz,1H),4.53–4.45(m,1H),4.35–4.27(m,2H),4.05(d,J=11.2Hz,1H),3.96–3.88(m,2H),3.73(d,J=11.3Hz,1H),3.54(q,J=7.0Hz,2H),2.30–2.18(m,1H),1.83–1.70(m,1H),1.47(s,3H),1.42–1.30(m,4H),1.23(t,J=7.0Hz,3H),0.91(t,J=6.9Hz,3H)。
LC-MS:m/z 452.5[M+H] +
实施例26:(R)-2-((2-氨基-7-(6-(4-乙氧基哌啶-1-基)吡啶-3-基)吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(26)的制备
Figure PCTCN2022128963-appb-000073
与实施例22的制备方法相同,除了将1-(2-氟乙基)哌嗪二盐酸盐替换为4-乙氧基哌啶,制得化合物26。
1H NMR(300MHz,甲醇-d 4)δ9.17(s,1H),8.75(d,J=2.4Hz,1H),8.14(dd,J=9.0,2.5Hz,1H),7.39(s,1H),6.95(d,J=9.2Hz,1H),4.63–4.59(m,1H),4.19–4.11(m,1H),4.07(d,J=11.5Hz,1H),3.75(d,J=11.2Hz,1H),3.68–3.54(m,3H), 2.32–2.14(m,1H),2.09–1.94(m,2H),1.86–1.74(m,1H),1.68–1.54(m,2H),1.49(s,3H),1.44–1.27(m,6H),1.22(t,J=7.0Hz,3H),0.97–0.88(m,3H)。
LC-MS:m/z 480.1[M+H] +
实施例27:(R)-2-((2-氨基-7-(6-(3-(二丙氨基)氮杂环丁烷-1-基)吡啶-3-基)吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(27)的制备
Figure PCTCN2022128963-appb-000074
与实施例22的制备方法相同,除了将1-(2-氟乙基)哌嗪二盐酸盐替换为N,N-二丙基氮杂环丁烷-3-胺,制得化合物27。
1H NMR(300MHz,甲醇-d 4)δ9.18(s,1H),8.74–8.68(m,1H),8.17(dd,J=8.8,2.4Hz,1H),7.39(s,1H),6.57(d,J=8.8Hz,1H),4.21(t,J=7.8Hz,2H),4.08(d,J=11.3Hz,1H),4.01–3.91(m,2H),3.87–3.78(m,1H),3.75(d,J=11.3Hz,1H),2.60–2.46(m,4H),2.34–2.16(m,1H),1.87–1.71(m,1H),1.62–1.51(m,4H),1.50(s,3H),1.44–1.32(m,4H),1.01–0.87(m,9H)。
LC-MS:m/z 507.4[M+H] +
实施例28:(R)-2-((2-氨基-7-(6-(4-二乙氨基哌啶-1-基)吡啶-3-基)吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(28)的制备
Figure PCTCN2022128963-appb-000075
与实施例9的制备方法相同,除了将1-环丙基哌嗪替换为4-二乙氨基哌啶,制得化合物28。
1H NMR(300MHz,甲醇-d 4)δ9.37(s,1H),8.87(d,J=2.4Hz,1H),8.26(dd,J=9.1,2.5Hz,1H),7.62(s,1H),7.03(d,J=9.1Hz,1H),4.74–4.54(m,overlapped with solvent,2H),4.17(d,J=11.4Hz,1H),3.78–3.65(m,2H),3.31–3.24(m,overlapped with solvent,4H),3.07(t,J=12.7Hz,2H),2.31–2.15(m,3H),1.91–1.71(m,3H),1.55(s,3H),1.49–1.27(m,10H),1.02–0.86(m,3H)。
LC-MS:m/z 507.5[M+H] +
实施例29:(R)-2-((2-氨基-7-(6-(3-(二乙氨基)氮杂环丁烷-1-基)吡啶-3-基)吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(29)的制备
Figure PCTCN2022128963-appb-000076
Figure PCTCN2022128963-appb-000077
与实施例22的制备方法相同,除了将1-(2-氟乙基)哌嗪二盐酸盐替换为3-(二乙氨基)氮杂环丁烷二盐酸盐,制得化合物29。
1H NMR(300MHz,甲醇-d 4)δ9.17(d,J=0.7Hz,1H),8.71(d,J=2.4Hz,1H),8.17(dd,J=8.8,2.4Hz,1H),7.39(d,J=0.7Hz,1H),6.58(d,J=8.8Hz,1H),4.23(t,J=7.8Hz,2H),4.08(d,J=11.3Hz,1H),4.03–3.92(m,2H),3.87–3.78(m,1H),3.75(d,J=11.2Hz,1H),2.67(q,J=7.2Hz,4H),2.35–2.18(m,1H),1.88–1.71(m,1H),1.50(s,3H),1.43–1.28(m,4H),1.10(t,J=7.2Hz,6H),0.93(t,J=6.9Hz,3H)。
LC-MS:m/z 479.4[M+H] +
实施例30:(R)-2-((2-氨基-7-(2-(4-(2-氟乙基)哌嗪-1-基)嘧啶-5-基)吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(30)的制备
Figure PCTCN2022128963-appb-000078
步骤1:2-(4-(2-氟乙基)哌嗪-1-基)嘧啶-5-硼酸(30a)的制备
于室温,将2-氯嘧啶-5-硼酸(148mg,0.935mmol)和1-(2-氟乙基)哌嗪二盐酸盐(209.92mg,1.029mmol)溶于乙醇(2mL)中,反应液中加入三乙胺(331.02mg,3.271mmol),在氮气氛下,75℃搅拌1小时。反应完毕后,反应液降至室温 后减压浓缩,得黄色固体化合物30a(260mg,粗品),其未经纯化直接用于下一步。
LC-MS:m/z 255.1[M+H] +
其余步骤与实施例9的相同,除了将化合物2-(4-环丙基哌嗪-1-基)吡啶-5-硼酸频那醇酯(9d)替换为(30a),制得化合物30。
1H NMR(300MHz,甲醇-d 4)δ9.22(s,1H),8.98(s,2H),7.44(s,1H),4.76–4.71(m,1H),4.59–4.55(m,1H),4.11(d,J=11.2Hz,1H),3.97(t,J=5.1Hz,4H),3.75(d,J=11.3Hz,1H),2.87–2.82(m,1H),2.78–2.73(m,1H),2.72–2.64(m,4H),2.34–2.19(m,1H),1.86–1.71(m,1H),1.51(s,3H),1.46–1.27(m,4H),0.93(t,J=6.9Hz,3H)。
LC-MS:m/z 484.3[M+H] +
实施例31:(R)-2-((2-氨基-7-(2-(4-丙基哌嗪-1-基)嘧啶-5-基)吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(31)的制备
Figure PCTCN2022128963-appb-000079
与实施例30的制备方法相同,除了将1-(2-氟乙基)哌嗪二盐酸盐替换为1-正丙基哌嗪二溴酸盐,制得化合物31。
1H NMR(400MHz,甲醇-d 4)δ9.18(d,J=0.7Hz,1H),8.95(s,2H),7.39(d,J=0.8Hz,1H),4.07(d,J=11.3Hz,1H),3.99–3.89(m,4H),3.73(d,J=11.3Hz,1H),2.60(t,J=5.0Hz,4H),2.46–2.38(m,2H),2.30–2.19(m,1H),1.83–1.71(m,1H),1.66–1.54(m,2H),1.48(s,3H),1.41–1.26(m,4H),0.96(t,J=7.4Hz,3H),0.91(t,J=7.0Hz,3H)。
LC-MS:m/z 480.4[M+H] +
实施例32:(R)-2-((2-氨基-7-(2-(4-异丙基哌嗪-1-基)嘧啶-5-基)吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(32)的制备
Figure PCTCN2022128963-appb-000080
与实施例30的制备方法相同,除了将1-(2-氟乙基)哌嗪二盐酸盐替换为1-异丙基哌嗪,制得化合物32。
1H NMR(300MHz,甲醇-d 4)δ9.19(s,1H),8.96(s,2H),7.41(s,1H),4.09(d,J=11.3Hz,1H),3.99–3.92(m,4H),3.75(d,J=11.3Hz,1H),2.83–2.74(m,1H),2.68(t,J=5.2Hz,4H),2.32–2.20(m,1H),1.85–1.71(m,1H),1.50(s,3H),1.44–1.26(m,4H),1.16(s,3H),1.14(s,3H),0.93(t,J=6.9Hz,3H)。
LC-MS:m/z 480.3[M+H] +
实施例33:(R)-2-((2-氨基-7-(2-(4-(二甲氨基)哌啶-1-基)嘧啶-5-基)吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(33)的制备
Figure PCTCN2022128963-appb-000081
Figure PCTCN2022128963-appb-000082
与实施例30的制备方法相同,除了将1-(2-氟乙基)哌嗪二盐酸盐替换为4-二甲氨基哌啶,制得化合物33。
1H NMR(400MHz,甲醇-d 4)δ9.17(s,1H),8.92(s,2H),7.38(s,1H),4.67–4.54(m,2H),4.07(d,J=11.3Hz,1H),3.73(d,J=11.3Hz,1H),2.96(t,J=12.9Hz,2H),2.71–2.55(m,1H),2.37(s,6H),2.30–2.18(m,1H),2.07–1.96(m,2H),1.82–1.70(m,1H),1.47(s,3H),1.44–1.24(m,6H),0.91(t,J=6.7Hz,3H)。
LC-MS:m/z 480.4[M+H] +
实施例34:(R)-2-((2-氨基-7-(4-(4-甲基哌嗪-1-基)苯基)吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(34)的制备
Figure PCTCN2022128963-appb-000083
与实施例9的的制备方法相同,除了将2-(4-环丙基哌嗪-1-基)吡啶-5-硼酸频那醇酯(9d)替换为4-(4-甲基哌嗪-1-基)苯硼酸频那醇酯(CAS:747413-21-4)(34a),制得化合物34。
1H NMR(300MHz,甲醇-d 4)δ9.15(s,1H),7.95–7.84(m,2H),7.42(s,1H),7.14–7.03(m,2H),4.07(d,J=11.3Hz,1H),3.75(d,J=11.3Hz,1H),3.36–3.33(m,4H),2.65(t,J=5.1Hz,4H),2.38(s,3H),2.33–2.19(m,1H),1.87–1.71(m,1H),1.49(s,3H),1.45–1.28(m,4H),0.93(t,J=6.8Hz,3H)。
LC-MS:m/z 450.2[M+H] +
实施例35:(R)-2-((2-氨基-7-(1-(1-甲基哌啶-4-基)-1H-吡唑-4-基)吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(35)的制备
Figure PCTCN2022128963-appb-000084
与实施例9的的制备方法相同,除了将2-(4-环丙基哌嗪-1-基)吡啶-5-硼酸频那醇酯(9d)替换为(1-甲基哌啶-4-基)-1H-吡唑-4-硼酸频那醇酯(CAS:1323919-64-7)(35a),制得化合物35。
1H NMR(300MHz,甲醇-d 4)δ9.11(s,1H),8.31(s,1H),8.08(s,1H),7.31(s,1H),4.36–4.18(m,1H),4.08(d,J=11.3Hz,1H),3.74(d,J=11.3Hz,1H),3.13–2.97(m,2H),2.37(s,3H),2.34–2.06(m,7H),1.86–1.69(m,1H),1.49(s,3H),1.45–1.27(m,4H),0.93(t,J=6.9Hz,3H)。
LC-MS:m/z 439.2[M+H] +
实施例36:(R)-2-((2-氨基-7-(1-(四氢吡喃-4-基)-1H-吡唑-4-基)吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(36)的制备
Figure PCTCN2022128963-appb-000085
与实施例9的制备方法相同,除了将2-(4-环丙基哌嗪-1-基)吡啶-5-硼酸频那醇酯(9d)替换为1-(四氢吡喃-4-基)-1H-吡唑-4-硼酸频那醇酯(CAS:1040377-03-4)(36a),制得化合物36。
1H NMR(400MHz,甲醇-d 4)δ9.14(s,1H),8.32(s,1H),8.08(s,1H),7.32(s,1H),4.54–4.42(m,1H),4.14–4.03(m,3H),3.72(d,J=11.3Hz,1H),3.65–3.54(m,2H),2.32–2.19(m,1H),2.17–2.06(m,4H),1.83–1.69(m,1H),1.48(s,3H),1.44– 1.25(m,4H),0.91(t,J=7.0Hz,3H)。
LC-MS:m/z 426.2[M+H] +
实施例37:(R)-2-((2-氨基-7-(4-(3-吡啶基)苯基)吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(37)的制备
Figure PCTCN2022128963-appb-000086
与实施例9的制备方法相同,除了将2-(4-环丙基哌嗪-1-基)吡啶-5-硼酸频那醇酯(9d)替换为4-(3-吡啶基)苯硼酸频那醇酯(CAS:929203-04-3)(37a),制得化合物37。
1H NMR(400MHz,甲醇-d 4)δ9.14(s,1H),8.80(d,J=2.4Hz,1H),8.45(dd,J=4.9,1.6Hz,1H),8.13–8.06(m,1H),8.06–7.99(m,2H),7.79–7.69(m,2H),7.52–7.42(m,2H),3.99(d,J=11.3Hz,1H),3.65(d,J=11.3Hz,1H),2.22–2.09(m,1H),1.74–1.62(m,1H),1.40(s,3H),1.34–1.21(m,4H),0.82(t,J=6.9Hz,3H)。
LC-MS:m/z 429.3[M+H] +
实施例38:(R)-2-((2-氨基-7-(6-(哌嗪-1-基)吡啶-3-基)吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(38)的制备
Figure PCTCN2022128963-appb-000087
Figure PCTCN2022128963-appb-000088
步骤1:(R)-4-(5-(2-氨基-4-((1-羟基-2-甲基己-2-基)氨基)吡啶并[4,3-d]嘧啶-7-基)吡啶-2-基)哌嗪-1-羧酸叔丁酯(38b)的制备
与实施例9的步骤4相同,除了将2-(4-环丙基哌嗪-1-基)吡啶-5-硼酸频那醇酯(9d)替换为4-[4-(N-Boc)哌嗪-1-基]苯硼酸频那醇酯(CAS:496786-98-2)(38a),制得化合物38b。
步骤2:(R)-2-((2-氨基-7-(6-(哌嗪-1-基)吡啶-3-基)吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(38)的制备
于室温,将化合物38b(50mg,0.093mmol)溶于二氯甲烷(1mL)中,反应液加入中加入三氟乙酸(1mL),在氮气氛下,室温搅拌2小时。反应完毕后,反应液减压浓缩后经制备薄层色谱法分离纯化(流动相:二氯甲烷/甲醇=10:1)得粗品。粗品进一步经制备型色谱柱分离纯化(柱型:Gemini-NX C18 AXAI Packed柱,5um,21.2*150mm;流动相A:水(0.05%氨水),流动相B:乙腈;流速:25mL/min;梯度:在8分钟内23%-41%乙腈;检测波长:254/220nm),得到类白色固体化合物38(2.1mg,4.76%)。
1H NMR(400MHz,甲醇-d 4)δ9.15(d,J=0.7Hz,1H),8.75(dd,J=2.6,0.7Hz,1H),8.14(dd,J=9.0,2.5Hz,1H),7.38(d,J=0.7Hz,1H),6.92(d,J=9.0Hz,1H),4.06(d,J=11.3Hz,1H),3.73(d,J=11.3Hz,1H),3.64–3.58(m,4H),2.98–2.90(m,4H),2.30–2.22(m,1H),1.83–1.71(m,1H),1.48(s,3H),1.40–1.32(m,4H),0.94–0.90(m,3H)。
LC-MS:m/z 437.3[M+H] +
实施例39:(R)-2-((7-([2,3’-联吡啶]-5-基)-2-氨基吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(39)的制备
Figure PCTCN2022128963-appb-000089
步骤1:5-溴-2,3’-联吡啶(39a)的制备
于室温,将3-吡啶硼酸(500.09mg,4.069mmol)和5-溴2-碘吡啶(1050mg,3.699mmol)溶于1,4-二氧六环(10.0mL)中,依次加入碳酸钾(1533.49mg,11.097mmol)和四(三苯基膦)钯(427.39mg,0.37mmol),反应液在氮气氛下,在80℃搅拌过夜。反应完全后,反应液用水(30mL)稀释,再用乙酸乙酯(3×30mL)萃取,有机相合并后用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。粗品用柱层析色谱法分离纯化(流动相:石油醚/乙酸乙酯=4:1),得到黄色固体化合物39a(148mg,17.02%)。
LC-MS:m/z 235.0[M+H] +
步骤2:2,3’-联吡啶-5-硼酸(39b)的制备
于室温,将化合物39a(148mg,0.63mmol)溶于1,4-二氧六环(3.0mL)中,反应液中依次加入联硼酸频那醇酯(239.81mg,0.944mmol)、醋酸钾(185.36mg,1.889mmol)和[1,1′-双(二苯基膦基)二茂铁]二氯化钯二氯甲烷络合物(51.29mg,0.063mmol),反应液在氮气氛下,在80℃搅拌3小时。反应完全后,反应液用水(10mL)稀释,再用乙酸乙酯(3×10mL)萃取,有机相合并后用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得到棕色固体化合物39b(314mg,粗品),其未经纯化直接用于下一步。
LC-MS:m/z 201.1[M+H] +
其余步骤与实施例9相同,除了将化合物2-(4-环丙基哌嗪-1-基)吡啶-5-硼酸频那醇酯(9d)替换为2,3’-联吡啶-5-硼酸(39b),制得化合物39。
1H NMR(400MHz,甲醇-d 4)δ9.33–9.30(m,1H),9.29–9.24(m,2H),8.62(dd,J=4.9,1.6Hz,1H),8.56–8.48(m,2H),8.10(d,J=8.4Hz,1H),7.63–7.57(m,2H), 4.09(d,J=11.3Hz,1H),3.74(d,J=11.3Hz,1H),2.31–2.22(m,1H),1.83–1.73(m,1H),1.50(s,3H),1.43–1.28(m,4H),0.92(t,J=7.0Hz,3H)。
LC-MS:m/z 430.3[M+H] +
实施例40:(R)-2-((7-([2,4’-联吡啶]-5-基)-2-氨基吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(40)的制备
Figure PCTCN2022128963-appb-000090
与实施例39的制备方法相同相同,除了将3-吡啶硼酸替换为4-吡啶硼酸,制得化合物40。
1H NMR(400MHz,甲醇-d 4)δ9.35(d,J=2.2Hz,1H),9.29(s,1H),8.72–8.66(m,2H),8.55(dd,J=8.3,2.3Hz,1H),8.19(d,J=8.2Hz,1H),8.17–8.13(m,2H),7.63(s,1H),4.10(d,J=11.4Hz,1H),3.74(d,J=11.3Hz,1H),2.33–2.21(m,1H),1.83–1.71(m,1H),1.50(s,3H),1.43–1.29(m,4H),0.92(t,J=6.9Hz,3H)。
LC-MS:m/z 430.4[M+H] +
实施例41:(R)-2-((2-氨基-7-(6’-甲基-[2,3’-联吡啶]-5-基)吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(41)的制备
Figure PCTCN2022128963-appb-000091
Figure PCTCN2022128963-appb-000092
与实施例39的制备方法相同,除了将化合物3-吡啶硼酸替换为6-甲基吡啶-3-硼酸,制得化合物41。
1H NMR(400MHz,甲醇-d 4)δ9.38(s,1H),9.25(s,1H),9.02(s,1H),8.46(d,J=7.9Hz,1H),8.34(d,J=8.0Hz,1H),7.96(d,J=7.9Hz,1H),7.66(s,1H),7.40(d,J=8.0Hz,1H),4.18(d,J=11.4Hz,1H),3.76(d,J=11.4Hz,1H),2.59(s,3H),2.33–2.19(m,1H),1.85–1.71(m,1H),1.54(s,3H),1.47–1.24(m,4H),0.93(t,J=6.9Hz,3H)。
LC-MS:m/z 444.3[M+H] +
实施例42:(R)-2-((7-([2,2’-联吡啶]-5-基)-2-氨基吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(42)的制备
Figure PCTCN2022128963-appb-000093
与实施例39的制备方法相同,除了将化合物5-溴-2,3’-联吡啶(39a)替换为5-溴-2,2’-联吡啶(42a),制得化合物42。
1H NMR(300MHz,Methanol-d 4)δ9.35–9.25(m,2H),8.75–8.65(m,1H),8.60–8.51(m,1H),8.51–8.39(m,2H),8.00(t,J=7.5Hz,1H),7.65(s,1H),7.56–7.43(m,1H),4.12(d,J=11.3Hz,1H),3.77(d,J=11.3Hz,1H),2.37–2.17(m,1H),1.88 –1.73(m,1H),1.52(s,3H),1.47–1.30(m,4H),0.94(t,J=6.8Hz,3H)。
LC-MS:m/z 430.1[M+H] +
实施例43:(R)-2-((2-氨基-7-(4-(吡咯烷-1-基甲基)苄基)吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(43)的制备
Figure PCTCN2022128963-appb-000094
步骤1:(R)-N 4-(1-((叔丁基二甲基硅烷基)氧基)-2-甲基己-2-基)-7-(4-氯苄基)-N 2-(2,4-二甲氧基苄基)吡啶并[4,3-d]嘧啶-2,4-二胺(43a)的制备
于室温,将化合物2f(150mg,0.26mmol)溶于1,4-二氧六环(2mL)和水(0.5mL),依次加入4-氯苄基硼酸频那醇酯(CAS:475250-49-8)(43b)(264mg,1.05mmol)、碳酸钾(255mg,1.83mmol)和四(三苯基膦)钯(60mg,0.052mmol)。反应液在氮气氛下于95℃搅拌3小时。反应完成后,反应液加水(15mL)稀释,用乙酸乙酯(3×10mL)萃取,有机相合并后用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,所得粗品用制备薄层色谱法分离纯化(流动相:乙酸乙酯/石油醚=1:2)得到橙色油状化合物43a(92mg,53.01%)。
LC-MS:m/z 664.3[M+H] +
步骤2:(R)-N 4-(1-((叔丁基二甲基硅烷基)氧基)-2-甲基己-2-基)-N 2-(2,4-二甲氧基苄基)-7-(4-(吡咯烷-1-基甲基)苄基)吡啶并[4,3-d]嘧啶-2,4-二胺(43c)的制备
于室温,将化合物43a(92mg,0.14mmol)溶于1,4-二氧六环(2mL)和水 (0.5mL),依次加入(1-吡咯烷基甲基)三氟硼酸钾(37mg,0.19mmol)、碳酸钾(57.8mg,0.41mmol)和XPhos Pd G3(11.7mg,0.014mmol)。反应液在氮气氛下于100℃搅拌过夜。反应完全后,反应液加水(15mL)稀释,用乙酸乙酯(3×10mL)萃取,有机相合并后用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,所得粗品用制备薄层色谱法分离纯化(流动相:乙酸乙酯/石油醚=1:2)得到橙色油状化合物43b(33mg,33.4%)。
LC-MS:m/z 713.4[M+H] +
步骤3:(R)-2-((2-氨基-7-(4-(吡咯烷-1-基甲基)苄基)吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(43)的制备
于室温,将化合物43b(33mg,0.046mmol)溶于二氯甲烷(1mL),缓慢滴加三氟乙酸(1mL)。反应液于40℃搅拌反应过夜。反应完成后,反应液减压浓缩,所得粗品用制备型色谱柱分离纯化(柱型:XBridge Shield RP18 OBD柱,5um,19*150mm;流动相A:水(10mmol/L碳酸氢铵),流动相B:乙腈;流速:25mL/min;梯度:在8分钟内11%-47%乙腈;检测波长:254/220nm),得到白色固体化合物43(1.6mg,7.41%)。
1H NMR(400MHz,甲醇-d 4)δ8.97(s,1H),7.26–7.16(m,4H),6.79(s,1H),4.50(s,2H),4.02(s,2H),3.95(d,J=11.2Hz,1H),3.60(d,J=11.3Hz,1H),3.42–3.34(m,2H),3.08–2.99(m,2H),2.19–2.08(m,1H),1.82–1.72(m,4H),1.68–1.59(m,1H),1.35(s,3H),1.31–1.19(m,4H),0.80(t,J=6.9Hz,3H)。
LC-MS:m/z 449.3[M+H] +
实施例44:N 4-丙基吡啶并[3,4-d]嘧啶-2,4-二胺(44)的制备
Figure PCTCN2022128963-appb-000095
步骤1:2-氨基吡啶并[3,4-d]嘧啶-4(3H)-酮(44b)的制备
于室温,将3-溴异烟酸44a(5.0g,24.75mmol)溶于N,N-二甲基甲酰胺(120mL)中,依次加入碳酸铯(16.1g,49.50mmol)、碘化亚铜(0.94g,4.95mmol)、盐酸胍(2.60g,27.23mmol),反应液在氮气氛下110℃搅拌过夜。反应完毕后,将体系中固体过滤,滤渣用甲醇洗涤(3×100mL)。将收集到的滤液减压浓缩,粗品用硅胶柱层析色谱法纯化(流动相:甲醇/二氯甲烷=20%-30%),得到淡黄色固体化合物44b(3.2g,79.4%)。
LC-MS:m/z 163.1[M+H] +
步骤2:N 4-丙基吡啶并[3,4-d]嘧啶-2,4-二胺(44)的制备
于室温,将化合物44b(40mg,0.247mmol)溶于N,N-二甲基甲酰胺(3mL)中,依次加入卡特缩合剂(BOP)(218mg,0.494mmol)、1,8-二氮杂二环十一碳-7-烯(DBU)(75.1mg,0.494mmol)、正丙胺(29.2mg,0.494mmol),反应液在氮气氛下,于室温反应4小时。反应完毕后,加入水(10mL)淬灭,用乙酸乙酯(3×20mL)萃取。有机相合并后用饱和食盐水(2×20mL)洗涤。有机相用无水硫酸钠干燥后,减压浓缩得粗品。粗品用制备型反相色谱柱纯化(柱型:XBridge Shield RP18 OBD柱,5um,19*150mm;流动相A:水(10mmol/L碳酸氢铵),流动相B:乙腈;流速:25mL/min;梯度:在8分钟内47%-53%乙腈;检测波长:254/220nm),得白色固体化合物44(10.2mg,20.8%)。
1H NMR(400MHz,甲醇-d 4)δ8.50(s,1H),8.06(d,J=5.6Hz,1H),7.69(d,J=5.7Hz,1H),3.49–3.41(m,2H),1.70–1.57(m,2H),0.90(t,J=7.4Hz,3H)。
LC-MS:m/z 204.2[M+H] +
实施例45:N 4-正丁基吡啶并[3,4-d]嘧啶-2,4-二胺(45)的制备
Figure PCTCN2022128963-appb-000096
与实施例44的制备方法相同,除了将正丙胺替换为正丁胺,制得化合物45。
1H NMR(400MHz,甲醇-d 4)δ8.50(d,J=0.9Hz,1H),8.06(d,J=5.6Hz,1H),7.69(dd,J=5.6,0.9Hz,1H),3.49(t,J=7.3Hz,2H),1.67–1.55(m,2H),1.41–1.29(m,2H),0.89(t,J=7.4Hz,3H)。
LC-MS:m/z 218.2[M+H] +
实施例46:N 4-正戊基吡啶并[3,4-d]嘧啶-2,4-二胺(46)的制备
Figure PCTCN2022128963-appb-000097
与实施例44的制备方法相同,除了将正丙胺替换为正戊胺,制得化合物46。
1H NMR(400MHz,甲醇-d 4)δ8.50(d,J=0.9Hz,1H),8.06(dd,J=5.6,0.8Hz,1H),7.69(dd,J=5.6,0.9Hz,1H),3.51–3.44(m,2H),1.69–1.56(m,2H),1.37–1.25(m,4H),0.91–0.78(m,3H)。
LC-MS:m/z 232.2[M+H] +
实施例47:2-((2-氨基吡啶[3,4-d]嘧啶-4-基)氨基)-1-戊醇(47)的制备
Figure PCTCN2022128963-appb-000098
与实施例44的制备方法相同,除了将正丙胺替换为DL-2-氨基-1-戊醇,制得化合物47。 1H NMR(400MHz,甲醇-d 4)δ8.50(s,1H),8.07(d,J=5.7Hz,1H),7.80(dd,J=5.6,0.9Hz,1H),4.47–4.39(m,1H),3.58(dd,J=5.4,1.4Hz,2H),1.68–1.51(m,2H),1.43–1.28(m,2H),0.88(t,J=7.3Hz,3H)。
LC-MS:m/z 248.2[M+H] +
实施例48:2-((2-氨基吡啶[3,4-d]嘧啶-4-基)氨基)-1-己醇(48)的制备
Figure PCTCN2022128963-appb-000099
与实施例44的制备方法相同,除了将正丙胺替换为DL-2-氨基-1-己醇,制得化合物47。 1H NMR(400MHz,甲醇-d 4)δ8.51(d,J=0.9Hz,1H),8.07(d,J=5.6Hz,1H),7.80(dd,J=5.6,0.9Hz,1H),4.45–4.37(m,1H),3.58(dd,J=5.4,1.1Hz,2H),1.73–1.62(m,1H),1.62–1.51(m,1H),1.51–1.40(m,1H),1.36–1.22(m,3H),0.85– 0.78(m,3H)。
LC-MS:m/z 262.2[M+H] +
实施例49:(R)-2-((2-氨基吡啶并[3,4-d]嘧啶-4-基)氨基)-2-甲基丁-1-醇(49)的制备
Figure PCTCN2022128963-appb-000100
与实施例44的制备方法相同,除了将正丙胺替换为(R)-2-氨基-2-甲基丁-1-醇(49a),制得化合物49。
1H NMR(400MHz,甲醇-d 4)δ8.79(s,1H),8.53(d,J=5.6Hz,1H),8.19(d,J=5.6Hz,1H),4.16(d,J=11.4Hz,1H),3.73(d,J=11.4Hz,1H),2.37–2.22(m,1H),1.89–1.73(m,1H),1.52(s,3H),0.93(t,J=7.5Hz,3H)。
LC-MS:m/z 248.3[M+H] +
实施例50:(R)-2-((2-氨基吡啶并[3,4-d]嘧啶-4-基)氨基)-2-甲基戊-1-醇(50)的制备
Figure PCTCN2022128963-appb-000101
与实施例44的制备方法相同,除了将正丙胺替换为(R)-2-氨基-2-甲基戊-1-醇(50a),制得化合物50。
1H NMR(400MHz,甲醇-d 4)δ8.59(d,J=0.9Hz,1H),8.16(d,J=5.6Hz,1H),7.90(dd,J=5.6,0.9Hz,1H),4.05(d,J=11.4Hz,1H),3.73(d,J=11.3Hz,1H),2.23–2.13(m,1H),1.81–1.69(m,1H),1.47(s,3H),1.44–1.28(m,2H),0.94(t,J=7.3Hz,3H)。
LC-MS:m/z 262.2[M+H] +
实施例51:(R)-2-((2-氨基吡啶并[3,4-d]嘧啶-4-基)氨基)-2-甲基庚-1-醇(51)的制备
Figure PCTCN2022128963-appb-000102
与实施例44的制备方法相同,除了将正丙胺替换为(R)-2-氨基-2-甲基庚-1-醇(51a),制得化合物51。 1H NMR(400MHz,甲醇-d 4)δ8.59(s,1H),8.16(d,J=5.6Hz,1H),7.90(d,J=5.3Hz,1H),4.06(d,J=11.3Hz,1H),3.72(d,J=11.3Hz,1H),2.28–2.19(m,1H),1.80–1.72(m,1H),1.47(s,3H),1.34–1.25(m,6H),0.90–0.83(m,3H)。
LC-MS:m/z 290.2[M+H] +
实施例52和实施例53:(R)-2-((2-氨基吡啶并[3,4-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(52)和(S)-2-((2-氨基吡啶并[3,4-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(53)的制备
Figure PCTCN2022128963-appb-000103
步骤1:(R)-N-((1-((叔丁基二甲基硅烷基)氧基)-2-甲基己-2-基)-2-氯吡啶并[3,4-d]嘧啶-4-胺(52b)的制备
于室温,将2,4-二氯吡啶并[3,4-d]嘧啶52a(100mg,0.50mmol)溶于1,4-二氧六环(2.0mL)中。反应液中依次加入(R)-1-((叔丁基二甲基硅烷基)氧基)-2-甲基己-2-胺(1e)(123mg,0.50mmol)和N,N-二异丙基乙胺(0.26μL,1.49mmol),于室温搅拌2小时。反应完全后,冷却至室温。反应液用水(20mL)稀释,再用乙酸乙酯(3×20mL)萃取,有机相合并后用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。所得粗品用硅胶柱层析色谱法分离纯化(流动相:石油醚/乙酸乙酯=4/1),得到黄色的油状化合物52b(110mg,53.79%)。
LC-MS:m/z 409.1[M+H] +
步骤2:(R)-N 4-(1-((叔丁基二甲基硅烷基)氧基)-2-甲基己-2-基)-N 2-(2,4-二甲氧基苄基)吡啶并[3,4-d]嘧啶-2,4-二胺(52c)的制备
于室温,将化合物52b(110mg,0.27mmol)溶于2,4-二甲氧基苄胺(1.0mL)中。反应液中加入N,N-二异丙基乙胺(140μl,0.80mmol),于80℃搅拌2小时。反应完全后,冷却至室温。反应液用水(20mL)稀释,再用乙酸乙酯(3×20mL)萃取,有机相合并后用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。所得粗品用硅胶柱层析色谱法分离纯化(流动相:石油醚/乙酸乙酯=3:2),得到黄色的油状化合物52c(114mg,78.53%)。
LC-MS:m/z 508.3[M+H] +
步骤3:(R)-2-((2-氨基吡啶并[3,4-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(52d)的制备
于室温,将化合物52c(114mg,0.21mmol)溶于二氯甲烷(0.5mL)中。反应液中加入三氟乙酸(0.5mL),于室温搅拌12小时。反应完全后,反应液减压浓缩,所得粗品用制备型色谱柱分离纯化(柱型:XBridge Shield RP18 OBD柱,5um,19*150mm;流动相A:水(10mmol/L碳酸氢铵),流动相B:乙腈;流速:25mL/min;梯度:在8分钟内20%-38%乙腈;检测波长:220nm),得到白色的固体化合物52d(12mg,20.18%)。
1H NMR(300MHz,甲醇-d 4)δ8.61(d,J=0.9Hz,1H),8.18(d,J=5.6Hz,1H),7.92(dd,J=5.6,0.9Hz,1H),4.09(d,J=11.3Hz,1H),3.74(d,J=11.2Hz,1H),2.32–2.18(m,1H),1.86–1.71(m,1H),1.49(s,3H),1.41–1.28(m,4H),0.92(t,J=6.9Hz,3H)。
LC-MS:m/z 275.9[M+H] +
步骤4:(R)-2-((2-氨基吡啶并[3,4-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(52)和(S)-2-((2-氨基吡啶并[3,4-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(53)的制备
化合物52d通过手性分离(柱型:OptiChiral A6-5柱,3*25cm,5um;流动相A:二氧化碳,流动相B:甲醇(0.1%的2M氨甲醇溶液);流速:50mL/min;梯度:50%B等梯度;检测波长:220nm),得到白色的固体产品化合物52,t R=2.052min,ee值(对映体过量):100%;和白色的固体产品化合物53,t R=2.383min,
ee值(对映体过量):92.8%。
化合物52:
1H NMR(300MHz,甲醇-d 4)δ8.61(s,1H),8.17(d,J=5.6Hz,1H),7.91(d,J=5.6Hz,1H),4.08(d,J=11.3Hz,1H),3.74(d,J=11.3Hz,1H),2.33–2.15(m,1H),1.89–1.71(m,1H),1.49(s,3H),1.44–1.21(m,4H),0.98–0.82(m,3H)。
LC-MS:m/z 276.3[M+H] +
化合物53:
1H NMR(400MHz,甲醇-d 4)δ8.64(s,1H),8.26(d,J=5.6Hz,1H),7.97(d,J=5.6Hz,1H),4.12–4.07(m,1H),3.72(d,J=11.2Hz,1H),2.29–2.17(m,1H),1.82–1.71(m,1H),1.49(s,3H),1.38–1.29(m,4H),0.89(t,J=6.9Hz,3H)。
LC-MS:m/z 276.2[M+H] +
实施例54:2-((2-氨基吡啶并[3,4-d]嘧啶-4-基)氨基)-1-庚醇(54)的制备
Figure PCTCN2022128963-appb-000104
与实施例52的步骤1至步骤3相同,除了将(R)-1-((叔丁基二甲基硅烷基)氧基)-2-甲基己-2-胺(1e)替换为2-氨基-1-庚醇,制得化合物54。
1H NMR(300MHz,甲醇-d 4)δ8.62(d,J=0.9Hz,1H),8.18(d,J=5.6Hz,1H),7.91(dd,J=5.6,0.9Hz,1H),4.59–4.46(m,1H),3.74–3.68(m,2H),1.87–1.59(m,2H),1.50–1.27(m,6H),0.97–0.84(m,3H)。
LC-MS:m/z 275.9[M+H] +
实施例55:(R)-2-((2-氨基-8-氟吡啶并[3,4-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(55)的制备
Figure PCTCN2022128963-appb-000105
步骤1:2-氨基-8-氟吡啶并[3,4-d]嘧啶-4(3H)-酮(55b)的制备
于室温,将3-氯-2-氟异烟酸(55a)(1.0g,5.70mmol)溶于N,N-二甲基甲酰胺(15mL)中,依次加入碳酸铯(5.57g,17.1mmol)、碳酸胍(1.03g,5.72mmol)、碘化亚铜(217mg,1.14mmol),反应液在氮气氛下110℃搅拌4小时。反应完毕后,加水(100mL)淬灭反应,体系中加入氯仿(3×50mL)萃取,合并的有机相用饱和食盐水(100mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,粗品用硅胶柱层析色谱法分离纯化(流动相:甲醇/二氯甲烷=0-10%),得淡黄色固体55b(83.9mg,8.2%)。
LC-MS:m/z 181.0[M+H] +
步骤2:(R)-N 4-(1-((叔丁基二甲基硅烷基)氧基)-2-甲基己-2-基)-8-氟吡啶并[3,4-d]嘧啶-2,4-二胺(55c)的制备
于室温,将化合物55b(30mg,0.167mmol)溶于N,N-二甲基甲酰胺(5mL)中,依次加入(R)-1-((叔丁基二甲基硅烷基)氧基)-2-甲基己-2-胺(1e)(122mg,0.50mmol)、1,8-二氮杂二环十一碳-7-烯(76.1mg,0.50mmol)、卡特缩合剂(110mg,0.25mmol),反应液在氮气氛下于室温搅拌过夜。反应完毕后,加水(50mL)淬灭反应,体系中加入乙酸乙酯(3×20mL)萃取,合并的有机相用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,粗品用硅胶柱层析色谱法分离纯化(流动相:乙酸乙酯/石油醚=15-30%),得白色固体化合物55c(10mg,14.7%)。
LC-MS:m/z 408.3[M+H] +
步骤3:(R)-2-((2-氨基-8-氟吡啶并[3,4-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(55)的制备
于室温,将化合物55c(10mg,0.025mmol)溶于三氟乙酸(2mL)和二氯甲烷(2mL)的混合液中。反应液在45℃搅拌2小时。反应完毕后,反应液减压浓缩,粗品加水(30mL)稀释,体系中加入二氯甲烷(3×15mL)萃取,合并的有机相用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩后得粗品,粗品用制备型反相色谱柱纯化(柱型:XBridge Shield RP18 OBD柱,5um,19*150mm;流动相A:水(10mmol/L碳酸氢铵),流动相B:乙腈;流速:25mL/min;梯度:在8分钟内25%-42%乙腈;检测波长:254/220nm),得到白色固体产物55(1.4mg,19.4%)。
1H NMR(400MHz,甲醇-d 4)δ7.69(d,J=5.7Hz,1H),7.59(dd,J=5.7,1.3Hz,1H),3.98(d,J=11.2Hz,1H),3.61(d,J=11.2Hz,1H),2.20–2.06(m,1H),1.72–1.60(m,1H),1.37(s,3H),1.32–1.14(m,4H),0.80(t,J=7.0Hz,3H)。
LC-MS:m/z 293.9[M+H] +
实施例56:(R)-2-((2-氨基-5-氟吡啶并[3,4-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(56)的制备
Figure PCTCN2022128963-appb-000106
步骤1:3-氨基-5-氟异烟酰胺(56b)的制备
于室温,将氨水(30mL)加入3-氨基-5-氟异烟酸甲酯(56a)(900mg,5.29mmol)中。反应于60℃搅拌3小时。反应完毕后,将反应液冷却至室温,加入水稀释反应,向体系中加入乙酸乙酯(3×50mL)萃取,合并的有机相使用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,所得粗品用硅胶柱层析色谱 法分离纯化(流动相:乙酸乙酯/石油醚=1:1),得黄色固体化合物56b(700mg,84.7%)。
LC-MS:m/z 156.0[M+H] +
步骤2:5-氟吡啶并[3,4-d]嘧啶-2,4(1H,3H)-二酮(56c)的制备
于室温,将化合物56b(700mg,4.49mmol)溶于1,4-二氧六环(15mL)中,加入双光气(3.09g,15.7mmol)。反应液在120℃搅拌16小时。反应完毕,将反应液冷却至室温,加入水淬灭反应后,向体系中加入乙酸乙酯(3×40mL)萃取,合并的有机相使用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,所得粗品用硅胶柱层析色谱法分离纯化(流动相:乙酸乙酯/石油醚=1:1),得黄色固体化合物56c(350mg,43.1%)。
LC-MS:m/z 182.0[M+H] +
步骤3:2,4-二氯-5-氟吡啶并[3,4-d]嘧啶(56d)的制备
于0℃,将化合物56c(350mg,1.93mmol)与三氯氧磷(5.0mL)混合,加入N,N-二异丙基乙胺(1.24g,9.65mmol)。反应液在100℃搅拌0.5小时。反应完毕,将反应液冷却至室温,减压浓缩,得褐色粗品化合物56d(70.0mg,16.7%)。
LC-MS:m/z 218.0[M+H] +
步骤4:(R)-N-((1-((叔丁基二甲基硅烷基)氧基)-2-甲基己-2-基)-2-氯-5-氟吡啶并[3,4-d]嘧啶-4-胺(56e)的制备
于室温,将化合物56d(70.0mg,0.32mmol)溶于1,4-二氧六环(5.0mL)中。反应液中依次加入(R)-1-((叔丁基二甲基硅烷基)氧基)-2-甲基己-2-胺(1e)(94.4mg,0.39mmol)和N,N-二异丙基乙胺(144mg,1.12mmol),于室温反应12小时。反应完全后,反应液用水(15mL)稀释,再用乙酸乙酯(3×10mL)萃取,有机相合并后用饱和食盐水(15mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,所得粗品用制备薄层色谱法分离纯化(流动相:乙酸乙酯/石油醚=1:8)得到棕色油状化合物56e(55.0mg,40.2%)。
LC-MS:m/z 427.2[M+H] +
步骤5:(R)-N 4-(1-((叔丁基二甲基硅烷基)氧基)-2-甲基己-2-基)-N 2-(2,4-二甲氧基苄基)-5-氟吡啶并[3,4-d]嘧啶-2,4-二胺(56f)的制备
于室温,将化合物56e(55mg,0.13mmol)溶于1,4-二氧六环(5mL)中。反应液中依次加入2,4-二甲氧基苄胺(21mg,0.15mmol)和N,N-二异丙基乙胺(58mg,0.45mmol),于100℃搅拌16小时。反应完全后,反应液用水(15mL)稀释,再用乙酸乙酯(3×10mL)萃取,有机相合并后用饱和食盐水(15mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,所得粗品用制备薄层色谱法分离纯化(流动相:乙酸乙酯/石油醚=1:10)得到棕色油状化合物56f(30.0mg,44.3%)。
LC-MS:m/z 558.3[M+H] +
步骤6:(R)-2-((2-氨基-5-氟吡啶并[3,4-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(56) 的制备
于室温,将化合物56f(30mg,0.057mmol)溶于二氯甲烷(3mL)中。反应液中加入三氟乙酸(1mL),于室温搅拌3小时。反应完全后,反应液减压浓缩,加入饱和碳酸氢钠溶液(10mL)稀释,再用乙酸乙酯(3×10mL)萃取,有机相合并后用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。所得粗品用制备型色谱柱纯化(柱型:XBridge Shield RP18 OBD柱,5um,19*150mm;流动相A:水(10mmol/L碳酸氢铵),流动相B:乙腈;流速:25mL/min;梯度:在8分钟内25%-45%乙腈;检测波长:254/220nm),得到白色固体化合物56(7.30mg,43.7%)。
1H NMR(400MHz,甲醇-d 4)δ8.33(d,J=1.2Hz,1H),7.92(d,J=2.9Hz,1H),3.81(d,J=11.1Hz,1H),3.60(d,J=11.0Hz,1H),2.02–1.88(m,1H),1.87–1.73(m,1H),1.38(s,3H),1.31–1.19(m,4H),0.87–0.77(m,3H)。
LC-MS:m/z 293.9[M+H] +
实施例57:(R)-2-((2-氨基-6-氟吡啶并[3,4-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(57)的制备
Figure PCTCN2022128963-appb-000107
步骤1:5-氨基-2-氟异烟酰胺(57b)的制备
于室温,将5-氨基-2-氟-4-吡啶羧酸57a(1.50g,9.62mmol)溶于氯化亚砜(20mL)中。反应于80℃回流1小时。反应完毕后,直接减压浓缩,将所得的褐色油状液体化合物溶于四氢呋喃(5mL),于0℃缓慢滴加入下氨水(15mL)中,反应液于0℃搅拌1小时。反应完毕后,加入水稀释反应,向体系中加入乙酸乙酯(3 ×50mL)萃取,合并的有机相使用饱和食盐水(100mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,所得粗品用硅胶柱层析色谱法分离纯化(流动相:乙酸乙酯/石油醚=1:1),得黄色固体粗品化合物57b(960mg,64.4%)。
LC-MS:m/z 156.0[M+H] +
其余步骤与实施例56相同,除了将3-氨基-5-氟异烟酰胺(56b)替换为5-氨基-2-氟异烟酰胺(57b),制得化合物57。
1H NMR(300MHz,甲醇-d 4)δ8.29(s,1H),7.74(s,1H),4.11(d,J=11.3Hz,1H),3.73(d,J=11.3Hz,1H),2.12–2.01(m,1H),1.68–1.56(m,1H),1.40(s,3H),1.36–1.31(m,4H),0.96–0.86(m,3H)。
LC-MS:m/z 293.9[M+H] +
实施例58:(R)-2-((2-氨基-7-氟吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(58)的制备
Figure PCTCN2022128963-appb-000108
步骤1:2-氟-5-碘-4-氨基吡啶(58b)和2-氟-3-碘-4-氨基吡啶(58c)的制备
于室温,将化合物2-氟-4-氨基吡啶58a(3.0g,26.8mmol)溶于乙醇(100mL) 中,依次加入碘(6.79g,26.8mmol)、硫酸银(8.34g,26.8mmol),于室温搅拌16小时。反应完毕后,过滤,滤液直接减压浓缩得粗品。粗品用硅胶柱层析色谱法分离纯化(流动相:乙酸乙酯/石油醚=20%-40%)得化合物58b(0.96g,15.0%)和化合物58c(3.10g,48.7%)。
LC-MS:m/z  58b 238.9[M+H] +;LC-MS:m/z  58c 238.9[M+H] +
步骤2:4-氨基-6-氟-烟酸甲酯(58d)的制备
于室温,将化合物58b(2.33g,9.79mmol)溶于甲醇(40mL)中,依次加入三乙胺(0.99g,9.79mmol)、[1,1'-双(二苯基膦基)二茂铁]二氯化钯(0.72g,0.98mmol),于一氧化碳20个大气压、100℃反应6小时。反应完毕后,过滤,滤液直接减压浓缩得粗品。粗品用硅胶柱层析纯化(流动相:乙酸乙酯/石油醚=20%-40%),得浅黄色固体化合物58d(543mg,32.6%)。
LC-MS:m/z 171.0[M+H] +
其余步骤与实施例56相同,除了将3-氨基-5-氟异烟酰胺(56b)替换为4-氨基-6-氟-烟酸甲酯(58d),制得化合物58。
LC-MS:m/z 294.2[M+H] +
1H NMR(300MHz,甲醇-d 4)δ7.95(d,J=5.7Hz,1H),7.01(d,J=6.0Hz,1H),3.93(d,J=11.2Hz,1H),3.71(d,J=11.0Hz,1H),2.15–1.83(m,2H),1.50(s,3H),1.44–1.31(m,4H),0.94(t,J=6.5Hz,3H)。
实施例59:(R)-2-((2-氨基-5-氟吡啶并[4,3-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(59)的制备
Figure PCTCN2022128963-appb-000109
Figure PCTCN2022128963-appb-000110
步骤1:4-氨基-6-氟-烟酸甲酯(59a)的制备
于室温,将2-氟-3-碘-4-氨基吡啶58c(3.10g,13.0mmol)溶于甲醇(60mL)中,依次加入三乙胺(1.32g,13.0mmol)、[1,1'-双(二苯基膦基)二茂铁]二氯化钯(1.06g,1.30mmol),于一氧化碳20个大气压、100℃反应6小时。反应完毕后,过滤,滤液直接减压浓缩得粗品。粗品用硅胶柱层析纯化(流动相:乙酸乙酯/石油醚=20%-40%),得浅黄色固体化合物59a(1.6g,72.2%)。
LC-MS:m/z 171.0[M+H] +
其余步骤与实施例56相同,除了将3-氨基-5-氟异烟酰胺(56b)替换为4-氨基-6-氟-烟酸甲酯(59a),制得化合物59。
LC-MS:m/z 294.2[M+H] +
1H NMR(300MHz,甲醇-d 4)δ7.94(dd,J=6.0,0.8Hz,1H),7.01(dd,J=6.0,1.9Hz,1H),3.93(d,J=11.1Hz,1H),3.71(d,J=11.0Hz,1H),2.14–1.99(m,1H),1.98–1.83(m,1H),1.50(s,3H),1.45–1.25(m,4H),1.02–0.87(m,3H)。
实施例60:2-((2-氨基-6-(4-(吡咯烷-1-基甲基)苄基)吡啶[3,4-d]嘧啶-4-基)氨基)己-1-醇(60)的制备
Figure PCTCN2022128963-appb-000111
Figure PCTCN2022128963-appb-000112
步骤1:5-氨基-2-氯异烟酸甲酯(60b)的制备
在0℃,将5-氨基-2-氯吡啶-4-羧酸60a(25g,145mmol)溶于甲醇(300mL)中,然后将二氯亚砜(100mL)滴加入上述溶液中(保持反应体系内温为20-25℃),滴加完成后,升温至70℃并搅拌过夜。将反应体系冷却至室温。混合液减压浓缩,所得残余物用碳酸氢钠水溶液打浆两遍,过滤后滤饼烘干得到黄色固体化合物60b(16g,59.1%)。
LC-MS:m/z 187.0[M+H] +
步骤2:2-氨基-6-氯吡啶并[3,4-d]嘧啶-4(3H)-酮(60c)的制备
于室温,在100mL玻璃瓶中依次加入化合物60b(3.0g,16.1mmol)、氯甲脒盐酸盐(3.7g,32.2mmol)、二甲基砜(10g),反应液升温至150℃搅拌4小时。反应完毕后,反应液加水(100mL)稀释,过滤,将所得滤饼用水淋洗2遍,所得滤饼经烘干得黄色固体化合物60c(3.7g,粗品)。
LC-MS:m/z 197.0[M+H] +
步骤3:2-((2-氨基-6-氯吡啶[3,4-d]嘧啶-4-基)氨基)己-1-醇(60d)的制备
于室温,将化合物60c(130mg,0.66mmol)与DL-2-氨基-1-己醇(155mg,1.32mmol)溶于N,N-二甲基甲酰胺(2mL),后将卡特缩合剂(351mg,0.79mmol)和1,8-二氮杂二环十一碳-7-烯(302mg,1.98mmol)依次加入体系。反应液在于室温搅拌4小时。反应完毕,反应液用水(30mL)稀释,再用乙酸乙酯(3×30mL)萃取,有机相合并后用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。将所得粗品用硅胶柱层析色谱法纯化(流动相:石油醚:乙酸乙酯=1:1),得黄色油状化合物60d(120mg,61%)。
LC-MS:m/z 296.1[M+H] +
步骤4:2-((2-氨基-6-(4-氯苄基)吡啶[3,4-d]嘧啶-4-基)氨基)己-1-醇(60e)的制备
于室温,将化合物60d(120mg,0.41mmol)与4-氯苄基硼酸频那醇酯(43b) (512mg,2.02mmol)溶于二氧六环(2mL)和水(0.4mL),然后将碳酸钾(168mg,1.21mmol)和四(三苯基膦)钯(93mg,0.081mmol)依次加入体系。反应液在氮气氛下95℃搅拌4小时。反应完毕,将反应液冷却至室温,用水(30mL)稀释,再用乙酸乙酯(3×30mL)萃取,有机相合并后用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。将所得粗品用硅胶柱层析色谱法纯化(流动相:石油醚:乙酸乙酯=1:1),得黄色油状化合物60e(60mg,38.3%)。
LC-MS:m/z 386.2[M+H] +
步骤5:2-((2-氨基-6-(4-(吡咯烷-1-基甲基)苄基)吡啶[3,4-d]嘧啶-4-基)氨基)己-1-醇(60)的制备
于室温,将化合物60e(60mg,0.155mmol)与(1-吡咯烷基甲基)三氟硼酸钾(35mg,0.18mmol)溶于二氧六环(1mL)和水(0.2mL),然后将Xphos Pd G3(9.9mg,0.012mmol)和碳酸钾(64mg,0.46mmol)依次加入体系。反应液在氮气氛下100℃搅拌4小时。反应完毕,将反应液冷却至室温,用水(30mL)稀释,再用乙酸乙酯(3×30mL)萃取,有机相合并后用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,所得粗品用制备型色谱柱分离纯化(柱型:XBridge Shield RP18 OBD柱,5um,19*150mm;流动相A:水(0.1%的甲酸),流动相B:乙腈;流速:25mL/min;梯度:在8分钟内6%-18%乙腈;检测波长:220nm),得到类白色固体化合物60(19.5mg,26.4%)。
1H NMR(400MHz,甲醇-d 4)δ8.73(s,1H),8.14(s,1H),7.50–7.36(m,4H),4.68–4.55(m,1H),4.32(s,2H),4.26(s,2H),3.79–3.65(m,2H),3.30–3.27(m,2H),2.15–1.99(m,4H),1.83–1.62(m,2H),1.46–1.32(m,4H),0.96–0.88(m,3H)。
LC-MS:m/z 435.2[M+H] +
实施例61:(R)-2-((2-氨基-6-(4-(吡咯烷-1-基甲基)苄基)吡啶[3,4-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(61)的制备
Figure PCTCN2022128963-appb-000113
Figure PCTCN2022128963-appb-000114
步骤1:(R)-N 4-(1-((叔丁基二甲基硅烷基)氧基)-2-甲基己-2-基)-6-氯吡啶并[3,4-d]嘧啶-2,4-二胺(61a)的制备
于室温,将化合物60c(300mg,1.52mmol)与(R)-1-((叔丁基二甲基硅烷基)氧基)-2-甲基己-2-胺(1e)(749mg,3.05mmol)溶于N,N-二甲基甲酰胺(2mL),然后将卡特缩合剂(810mg,1.83mmol)和1,8-二氮杂二环十一碳-7-烯(697mg,4.57mmol)依次加入体系。反应液于室温搅拌4小时。反应完毕,反应液用水(30mL)稀释,再用乙酸乙酯(3×30mL)萃取,有机相合并后用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。将所得粗品用硅胶柱层析色谱法纯化(流动相:石油醚:乙酸乙酯=1:1),得黄色油状化合物61a(102mg,76%)。
LC-MS:m/z 424.2[M+H] +
步骤2:(R)-N 4-(1-((叔丁基二甲基硅烷基)氧基)-2-甲基己-2-基)-6-(4-氯苄基)吡啶并[3,4-d]嘧啶-2,4-二胺(61b)的制备
于室温,将化合物61a(100mg,0.24mmol)与4-氯苄基硼酸频那醇酯(43b)(297mg,1.17mmol)溶于二氧六环(2mL)和水(0.4mL),然后将四(三苯基膦)钯(54mg,0.04mmol)和碳酸钾(97mg,0.7mmol)依次加入体系。反应液在氮气氛下95℃搅拌4小时。反应完毕,将反应液冷却至室温,用水(30mL)稀释,再用乙酸乙酯(3×30mL)萃取,有机相合并后用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。将所得粗品用硅胶柱层析色谱法纯化(流动相:石油醚:乙酸乙酯=1:1),得黄色油状化合物61b(62mg,51%)。
LC-MS:m/z 514.3[M+H] +
步骤3:(R)-N 4-(1-((叔丁基二甲基硅烷基)氧基)-2-甲基己-2-基)-6-(4-(吡咯烷-1-基甲基)苄基)吡啶并[3,4-d]嘧啶-2,4-二胺(61c)的制备
于室温,将化合物61b(60mg,0.12mmol)与(1-吡咯烷基甲基)三氟硼酸钾(26mg,0.14mmol)溶于二氧六环(1mL)和水(0.2mL),然后将碳酸钾(48mg, 0.35mmol)和Xphos Pd G3(7.4mg,0.009mmol)依次加入体系。反应液在氮气氛下100℃搅拌4小时。反应完毕,将反应液冷却至室温,用水(30mL)稀释,再用乙酸乙酯(3×30mL)萃取,有机相合并后用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。将所得粗品用硅胶柱层析色谱法纯化(流动相:石油醚:乙酸乙酯=2:1),得黄色油状化合物61c(8mg,12%)。
LC-MS:m/z 563.4[M+H] +
步骤4:(R)-2-((2-氨基-6-(4-(吡咯烷-1-基甲基)苄基)吡啶[3,4-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(61)的制备
于室温,将化合物61c(8mg,0.014mmol)溶于二氯甲烷(1.0mL)中。反应液中加入三氟乙酸(0.5mL),于室温搅拌1小时。反应完全后,反应液减压浓缩,加入饱和碳酸氢钠溶液(10mL)稀释,再用乙酸乙酯(3×10mL)萃取,有机相合并后用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。所得粗品用制备型色谱柱分离纯化(柱型:XBridge Shield RP18 OBD柱,5um,19*150mm;流动相A:水(0.05%氨水),流动相B:乙腈;流速:25mL/min;梯度:在8分钟内30%-65%乙腈;检测波长:220nm),得到白色固体化合物61(1.2mg,8.3%)。
1H NMR(400MHz,甲醇-d 4)δ8.73(s,1H),8.24(s,1H),7.42(q,J=8.1Hz,4H),4.32(s,2H),4.26(s,2H),4.17(d,J=11.4Hz,1H),3.70(d,J=11.4Hz,1H),3.30–
3.27(m,4H),2.29–2.19(m,1H),2.13–2.01(m,4H),1.83–1.72(m,1H),1.52(s,
3H),1.40–1.29(m,4H),0.90(t,J=6.9Hz,3H)。
LC-MS:m/z 449.2[M+H] +
实施例62:(R)-2-((2-氨基-6-(4-((二甲氨基)甲基)苄基)吡啶[3,4-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(62)的制备
Figure PCTCN2022128963-appb-000115
步骤1:(R)-2-((2-氨基-6-氯吡啶[3,4-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(62a)的制备
于室温,将化合物60c(300mg,1.56mmol)、(R)-2-氨基-2-甲基己-1-醇(300mg,2.27mmol)、卡特缩合剂(810mg,1.83mmol)、1,8-二氮杂二环十一碳-7-烯(697mg,4.59mmol)溶于N,N-二甲基甲酰胺(5mL)中。反应液于室温搅拌12小时。反应完毕后,加水(20mL)稀释反应,体系中加入乙酸乙酯(3×30mL)萃取,合并的有机相用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,所得黄色粗品用硅胶柱层析色谱法分离纯化(流动相:甲醇/二氯甲烷/三乙胺=1/12/0.1),得黄色固体化合物62a(300mg,62.03%)。
LC-MS:m/z 310.1[M+H] +
步骤2:(R)-2-((2-氨基-6-(4-氯苄基)吡啶[3,4-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(62b)的制备
于室温,将化合物62a(150mg,0.48mmol)、4-氯苄基硼酸频那醇酯(43b)(245mg,0.97mmol)、碳酸钾(201mg,1.45mmol)、四(三苯基膦)钯溶于1,4-二氧六环(2mL)和水(0.5mL)中。反应液在氮气氛下于95℃搅拌12小时。反应完毕后,加水(20mL)稀释反应,体系中加入乙酸乙酯(3×30mL)萃取,合并的有机相用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,所得黄色粗品用硅胶柱层析色谱法分离纯化(流动相:甲醇/二氯甲烷/三乙胺=1/13/0.1),得黄色固体化合物62b(30mg,15.53%)。
LC-MS:m/z 400.2[M+H] +
步骤3:(R)-2-((2-氨基-6-(4-((二甲氨基)甲基)苄基)吡啶[3,4-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(62)的制备
于室温,将化合物62b(20mg,0.05mmol)、(二甲氨基甲基)三氟硼酸钾(165mg,0.08mmol)、碳酸铯(49mg,0.15mmol)、2-二环己基磷-2,4,6-三异丙基联苯(5mg,0.01mmol)、醋酸钯(1mg,0.005mmol)溶于1,4-二氧六环(1mL)和水(0.2mL)。反应液用氮气置换三次,于100℃搅拌12小时。反应完毕后,加水(20mL)稀释反应,体系中加入乙酸乙酯(3×30mL)萃取,合并的有机相用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,所得粗品用制备型色谱柱分离纯化(柱型:XBridge Shield RP18 OBD柱,10um,19*250mm;流动相A:水(0.05%氨水),流动相B:乙腈;流速:25mL/min;梯度:在8分钟内30%-55%乙腈;检测波长:254/220nm)。收集产品,冷冻干燥,得到白色油状化合物62(1.1mg,5.21%)。
1H NMR(400MHz,甲醇-d 4)δ8.44(d,J=0.8Hz,1H),7.80(d,J=0.8Hz,1H),7.15(s,4H),4.07(s,2H),3.95(d,J=11.3Hz,1H),3.61(d,J=11.2Hz,1H),3.56–3.53(m,1H),3.40(s,2H),2.16(s,6H),2.14–2.07(m,1H),1.72–1.59(m,1H),1.36(s,3H),1.28–1.15(m,4H),0.79(t,J=7.0Hz,3H)。
LC-MS:m/z 423.3[M+H] +
实施例63:(R)-2-((2-氨基-6-甲基吡啶[3,4-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(63)的制备
Figure PCTCN2022128963-appb-000116
于室温,将化合物62a(50.0mg,0.161mmol)、三甲基环三硼氧烷(50wt%在THF中,60.78mg,0.242mmol)、碳酸铯(157.76mg,0.484mmol)、四(三苯基膦)钯(37.30mg,0.032mmol)溶于1,4-二氧六环(1.8mL)和水(0.2mL)。反应液在氮气氛下于100℃搅拌12小时。反应完毕后,反应液减压浓缩,残余物用制备薄层色谱法分离纯化(流动相:乙酸乙酯/甲醇=30:1),所得粗品进一步经制备型色谱柱分离纯化(柱型:XSelect CSH Prep C18 OBD柱,5um,19*150mm;流动相A:水(0.1%的甲酸),流动相B:乙腈;流速:25mL/min;梯度:在8分钟内10%-22%乙腈;检测波长:254/220nm)。收集产品,冷冻干燥,得到黄绿色固体化合物63的甲酸盐(13.8mg,25.49%)。
1H NMR(400MHz,甲醇-d 4)δ8.88(br s,1H),8.23(s,1H),4.17(d,J=11.3Hz,1H),3.70(d,J=11.3Hz,1H),2.64(s,3H),2.32–2.18(m,1H),1.83–1.71(m,1H),1.53(s,3H),1.42–1.28(m,4H),0.91(t,J=6.9Hz,3H)。
LC-MS:m/z 290.3[M+H] +
实施例64:(R)-2-((2-氨基-6-乙基吡啶[3,4-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(64)的制备
Figure PCTCN2022128963-appb-000117
Figure PCTCN2022128963-appb-000118
与实施例63的制备方法相同,除了将化合物三甲基环三硼氧烷替换为乙基硼酸,制得化合物64。
1H NMR(300MHz,甲醇-d 4)δ8.65(s,1H),8.03(s,1H),4.17(d,J=11.3Hz,1H),3.73(d,J=11.3Hz,1H),2.92(q,J=7.6Hz,2H),2.36–2.19(m,1H),1.86–1.71(m,1H),1.53(s,3H),1.41–1.35(m,4H),0.93(t,J=7.1Hz,3H)。
LC-MS:m/z 303.9[M+H] +
实施例65:(R)-2-((2-氨基-6-(二甲氨基)吡啶[3,4-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(65)的制备
Figure PCTCN2022128963-appb-000119
于室温,将化合物62a(40mg,0.129mmol)、二甲胺盐酸盐(15.79mg,0.194mmol)、碳酸铯(168.27mg,0.516mmol)、Pd-PEPPSI-IPent Cl-o-甲基吡啶(CAS1612891-29-8,10.85mg,0.013mmol)溶于1,4-二氧六环(1.5mL)。反应液在氮气氛下于90℃搅拌12小时。反应完毕,反应液用水(20mL)稀释,再用乙酸乙酯(3×20mL)萃取,有机相合并后用饱和食盐水(40mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。所得粗品经制备型色谱柱分离纯化(柱型:XBridge Prep C18 OBD柱,5um,19*150mm;流动相A:水(10mmol/L碳酸氢铵),流动相B:乙腈;流速:25mL/min;梯度:在8分钟内25%-40%乙腈;检测波长:254/220nm)。收集产品,冷冻干燥,得到黄绿色固体化合物65(2.3mg,5.43%)。
1H NMR(400MHz,甲醇-d 4)δ8.34(d,J=0.9Hz,1H),6.98(d,J=0.9Hz,1H),4.04(d,J=11.3Hz,1H),3.72(d,J=11.3Hz,1H),3.10(s,6H),2.31–2.18(m,1H),1.83–1.72(m,1H),1.47(s,3H),1.43–1.26(m,4H),0.90(t,J=7.1Hz,3H)。
LC-MS:m/z 319.2[M+H] +
实施例66和67:(R)-2-((2-氨基嘧啶并[5,4-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(66) 和(R)-2-((6-氨基嘧啶并[5,4-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(67)的制备
Figure PCTCN2022128963-appb-000120
步骤1:(R)-N-((1-((叔丁基二甲基硅烷基)氧基)-2-甲基己-2-基)-2,6,8-三氯嘧啶并[5,4-d]嘧啶-4-胺(66b)的制备
于室温,将化合物2,4,6,8-四氯嘧啶并[5,4-d]嘧啶66a(700mg,2.59mmol)溶于无水四氢呋喃(5mL)中,冷却至-78℃后,依次加入N,N-二异丙基乙胺(402mg,3.11mmol)、(R)-1-((叔丁基二甲基硅烷基)氧基)-2-甲基己-2-胺(1e)(637mg,2.59mmol),反应液在氮气氛下-78℃搅拌1小时。反应完毕后,加水(20mL)淬灭反应,体系中加入乙酸乙酯(3×30mL)萃取,合并的有机相用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得粗品66b,为淡黄色固体(510mg,41.1%)。
LC-MS:m/z 478.1[M+H] +
步骤2:(R)-N-((1-((叔丁基二甲基硅烷基)氧基)-2-甲基己-2-基)-2,6-二氯嘧啶并[5,4-d]嘧啶-4-胺(66c)的制备
于室温,将化合物66b(480mg,1.00mmol)溶于甲醇(5mL)中,依次加入10%钯碳(48mg,含水量10%)。反应体系在氢气氛下室温搅拌1小时。反应完毕后,反应液过滤浓缩得类白色固体化合物66c(300mg,67.3%)。
LC-MS:m/z 444.2[M+H] +
步骤3:(R)-N 4-(1-((叔丁基二甲基硅烷基)氧基)-2-甲基己-2-基)-6-氯-N 2-(2,4-二甲氧基苄基)嘧啶并[5,4-d]嘧啶-2,4-二胺(66d)和(R)-N 8-(1-((叔丁基二甲基硅烷基)氧基)-2-甲基己-2-基)-6-氯-N 2-(2,4-二甲氧基苄基)嘧啶并[5,4-d]嘧啶-2,8-二胺 (67a)的制备
于室温,将化合物66c(300mg,0.675mmol)溶于1,4-二氧六环(3mL)中,依次加入N,N-二异丙基乙胺(174mg,1.35mmol)和2,4-二甲氧基苄胺(113mg,0.675mmol)。反应体系于100℃搅拌3小时。反应完毕后,加水(20mL)淬灭反应,体系中加入乙酸乙酯(3×20mL)萃取,合并的有机相用无水硫酸钠干燥,过滤,滤液减压浓缩。粗品用硅胶柱层析色谱法纯化(流动相:乙酸乙酯/石油醚=20%-40%)得66d和67a的混合物,为黄色油状液体(80mg,21.8%)。
LC-MS:m/z 575.3[M+H] +
步骤4:(R)-N 4-(1-((叔丁基二甲基硅烷基)氧基)-2-甲基己-2-基)-N 2-(2,4-二甲氧基苄基)嘧啶并[5,4-d]嘧啶-2,4-二胺(66e)和(R)-N 8-(1-((叔丁基二甲基硅烷基)氧基)-2-甲基己-2-基)-N 2-(2,4-二甲氧基苄基)嘧啶并[5,4-d]嘧啶-2,8-二胺(67b)的制备
于室温,将化合物66d和67a的混合物(80.0mg,0.148mmol)溶于甲醇(3mL)中。加入10%钯碳(8mg,含水量10%)、甲酸铵(28.0mg,0.444mmol)。反应液于室温搅拌1小时。反应完毕后,加水(10mL)淬灭反应,体系中加入乙酸乙酯(3×10mL)萃取,合并的有机相用无水硫酸钠干燥,过滤,滤液减压浓缩后得粗品66e和67b的混合物,为黄色半固体(40.0mg,53.3%)。
LC-MS:m/z 541.3[M+H] +
步骤5:(R)-2-((2-氨基嘧啶并[5,4-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(66)和(R)-2-((6-氨基嘧啶并[5,4-d]嘧啶-4-基)氨基)-2-甲基己-1-醇(67)的制备
于室温,将化合物66e和67b的混合物(40mg,0.0787mmol)溶于三氟乙酸(1mL)中。反应液在40℃搅拌4小时。反应完毕后,减压浓缩后得粗品,粗品用制备型色谱柱纯化(柱型:XSelect CSH Prep C18 OBD柱,5um,19*150mm;流动相A:水(0.1%的甲酸),流动相B:乙腈;流速:25mL/min;梯度:在8分钟内10%-30%乙腈;检测波长:254/220nm),得白色固体66(1.0mg,4.9%)和白色固体67(5.6mg,27.4%)。
化合物66:
1H NMR(400MHz,甲醇-d 4)δ9.06(s,1H),8.93(s,1H),3.87(d,J=11.2Hz 1H),3.62(d,J=11.2Hz 1H),2.05-1.97(m,1H),1.87-1.81(m,1H),1.45(s,3H),1.29-1.18(m,4H),0.85(t,J=6.8Hz,3H)。
LC-MS:m/z 277.2[M+H] +
化合物67:
1H NMR(400MHz,甲醇-d 4)δ8.74(s,1H),8.13(s,1H),3.83(d,J=11.2Hz 1H),3.61(d,J=11.2Hz 1H),2.02-1.94(m,1H),1.80-1.71(m,1H),1.39(s,3H),1.25-1.15(m,4H),0.82(t,J=6.8Hz,3H)。
LC-MS:m/z 277.2[M+H] +
实施例68:(R)-2-((2-氨基吡啶并[3,4-d]嘧啶-4-基)氨基)-2-甲基己-1,1-二氘-1-醇(68)的制备
Figure PCTCN2022128963-appb-000121
步骤1:(R)-3-甲基-5-苯基-5,6-二氢-2H-1,4-噁嗪-2-酮(68a)的制备
于室温,将化合物(R)-2-氨基2-苯基乙醇(5g,36.44mmol)、丙酮酸乙酯(4.23g,36.44mmol)溶于三氟乙醇(110mL)中,升温至75℃反应16小时。反应完毕后,冷却至室温,过滤,滤饼用乙酸乙酯(3×30mL)洗涤。将滤液减压浓缩,所得黄色粗品用硅胶柱层析色谱法分离纯化(流动相:乙酸乙酯/石油醚=0-10%),得白色固体68a(2.4g,34.8%)。
LC-MS:m/z 190.1[M+H] +
步骤2:(3R,5R)-3-丁基-3-甲基-5-苯基吗啉-2-酮(68b)的制备
于室温,将化合物68a(2.4g,12.68mmol)溶于干燥的四氢呋喃(80mL)中,将反应液冷却至-78℃,在氮气氛下缓慢滴加三氟化硼***溶液(3.3mL),保持-78℃反应1.5小时。然后缓慢滴加正丁基氯化镁的四氢呋喃溶液(13.5mL,2mol/L),保持-78℃反应2小时。反应完毕后,缓慢升至室温,加氯化铵水溶液淬灭反应,向体系中加入乙酸乙酯(3×50mL)萃取,合并的有机相使用饱和食盐水(120mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,所得棕色油状粗品用硅胶柱层析分离纯化(流动相:乙酸乙酯/石油醚=0-5%),得白色固体68b(1.35g,43.0%)。
LC-MS:m/z 248.2[M+H] +
步骤3:(R)-2-(((R)-2-羟基-1-苯基乙基)氨基)-2-甲基己-1,1-二氘-1-醇(68c)的制备
于室温,将化合物68b(500mg,2.02mmol)溶于干燥的四氢呋喃(5mL) 中,将反应液置于0℃,加入氘代四氢铝锂(169.7mg,4.04mmol)。反应液于室温搅拌2小时。反应完毕,冰浴下加水淬灭反应,向体系中加入乙酸乙酯(3×6mL)萃取,合并的有机相使用饱和食盐水(15mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得淡黄色固体68c(360mg,70.3%)。
LC-MS:m/z 254.3[M+H] +
步骤4:(R)-2-氨基-2-甲基己-1,1-二氘-1-醇盐酸盐(68d)的制备
于室温,将化合物68c(360mg,1.42mmol)溶于无水乙醇(5mL)中,加入氢氧化钯碳(144mg,0.42mmol)和氯化氢的二氧六环溶液(0.5mL,4mol/L)。反应液在70℃氢气氛下(5atm)反应16小时。反应完毕后,冷却至室温,过滤,滤饼用乙醇(3×3mL)洗涤。将滤液减压浓缩,得黄色粗品。加入水(10mL),并向体系中加入乙酸乙酯(3×8mL)萃取,水相减压浓缩,得黄色油状产品68d(185mg,76.7%)
LC-MS:m/z 134.0[M+H] +
步骤5:(R)-2-((2-氨基吡啶并[3,4-d]嘧啶-4-基)氨基)-2-甲基己-1,1-二氘-1-醇(68)的制备
于室温,将化合物68d(90mg,0.67mmol)和2-氨基吡啶[3,4-d]嘧啶-4(3H)-酮(44b)(100mg,0.62mmol)溶于N,N-二甲基甲酰胺(2mL),加入卡特缩合剂(327mg,0.74mmol)和1,8-二氮杂二环十一碳-7-烯(282mg,1.85mmol),于室温反应16小时。反应完全后,反应液用水(10mL)稀释,再用乙酸乙酯(3×8mL)萃取,有机相合并后饱和食盐水(3×15mL)洗涤,无水硫酸钠干燥。有机相减压浓缩,所得粗品用制备型色谱柱分离纯化(柱型:XBridge Shield RP18 OBD Column,5um,19*150mm;流动相A:水(0.05%的氨水),流动相B:乙腈;流速:25mL/min;梯度:在8分钟内25%-40%乙腈;检测波长:254/220nm),得到白色的固体产品68(13.6mg,7.8%)。
1H NMR(300MHz,甲醇-d 4)δ8.61(d,J=0.8Hz,1H),8.18(d,J=5.6Hz,1H),7.92(dd,J=5.7,0.9Hz,1H),2.32–2.16(m,1H),1.85–1.70(m,1H),1.48(s,3H),1.43–1.29(m,4H),0.97–0.86(m,3H)。
LC-MS:m/z 278.4[M+H] +
实施例69:(R)-2-((2-氨基吡啶并[3,4-d]嘧啶-4-基)氨基)-2-乙基己-1-醇(69)的制备
Figure PCTCN2022128963-appb-000122
Figure PCTCN2022128963-appb-000123
步骤1:(R)-3-乙基-5-苯基-5,6-二氢-2H-1,4-噁嗪-2-酮(69a)的制备
于室温,将化合物(R)-2-氨基2-苯基乙醇(4.11g,29.96mmol)、2-氧代丁酸甲酯(3.48g,29.96mmol)溶于三氟乙醇(90mL)中,升温至75℃反应16小时。反应完毕后,冷却至室温,过滤,滤饼用乙酸乙酯(3×30mL)洗涤。将滤液减压浓缩,所得黄色粗品用硅胶柱层析分离纯化(流动相:乙酸乙酯/石油醚=0-10%),得白色固体69a(2.3g,37.8%)。
LC-MS:m/z 204.2[M+H] +
步骤2:(3R,5R)-3-丁基-3-乙基-5-苯基吗啉-2-酮(69b)的制备
于室温,将化合物69a(2.3g,11.32mmol)溶于干燥的四氢呋喃(75mL)中,将反应液冷却至-78℃,在氮气氛下缓慢滴加三氟化硼***溶液(3.0mL),保持-78℃反应1.5小时。然后缓慢滴加正丁基氯化镁的四氢呋喃溶液(11.9mL,2mol/L),保持-78℃反应2小时。反应完毕后,缓慢升至室温,加氯化铵水溶液淬灭反应,向体系中加入乙酸乙酯(3×50mL)萃取,合并的有机相使用饱和食盐水(120mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,所得棕色油状粗品用硅胶柱层析分离纯化(流动相:乙酸乙酯/石油醚=0-5%),得白色固体69b(1.23g,41.6%)。
LC-MS:m/z 262.3[M+H] +
步骤3:(R)-2-乙基-2-(((R)-2-羟基-1-苯基乙基)氨基)己-1-醇(69c)的制备
于室温,将化合物69b(1.23g,4.71mmol)溶于干燥的四氢呋喃(10mL)中,将反应液置于0℃,加入硼氢化锂的四氢呋喃溶液(4.7mL,2mol/L)。反应液于室温反应2小时。反应完毕,冰浴下加水淬灭反应,向体系中加入乙酸乙酯(3×12mL)萃取,合并的有机相使用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得淡黄色固体69c(1.1g,88.1%)。
LC-MS:m/z 266.2[M+H] +
步骤4:(R)-2-氨基-2-乙基己-1-醇盐酸盐(69d)的制备
于室温,将化合物69c(1.1g,4.15mmol)溶于无水乙醇(20mL)中,加入氢氧化钯碳(421mg,1.23mmol)和氯化氢的二氧六环溶液(1.5mL,4mol/L)。 反应液在70℃氢气氛下(5atm)反应16小时。反应完毕后,冷却至室温,过滤,滤饼用乙醇(3×8mL)洗涤。将滤液减压浓缩,得黄色粗品。加入水(30mL),并向体系中加入乙酸乙酯(3×25mL)萃取,水相减压浓缩,得黄色油状产品69d(750mg,99.6%)
LC-MS:m/z 146.0[M+H] +
步骤5:(R)-2-((2-氨基吡啶并[3,4-d]嘧啶-4-基)氨基)-2-乙基己-1-醇(69)的制备
于室温,将化合物69d(202mg,1.39mmol)和2-氨基吡啶[3,4-d]嘧啶-4(3H)-酮(44b)(150mg,0.93mmol)溶于N,N-二甲基甲酰胺(3mL),加入卡特缩合剂(491mg,1.11mmol)和1,8-二氮杂二环十一碳-7-烯(422mg,2.78mmol),于室温反应16小时。反应完全后,反应液用水(10mL)稀释,再用乙酸乙酯(3×8mL)萃取,有机相合并后饱和食盐水(3×15mL)洗涤,无水硫酸钠干燥。有机相减压浓缩,所得粗品用制备型色谱柱分离纯化(柱型:XBridge Shield RP18OBD Column,5um,19*150mm;流动相A:水(0.05%的氨水),流动相B:乙腈;流速:25mL/min;梯度:在8分钟内25%-50%乙腈;检测波长:254/220nm),得到白色的固体产品69(16.3mg,6.0%)。
1H NMR(300MHz,甲醇-d 4)δ8.61(d,J=0.8Hz,1H),8.18(d,J=5.6Hz,1H),7.92(dd,J=5.6,0.9Hz,1H),3.94(d,J=1.4Hz,2H),2.15–1.83(m,4H),1.46–1.22(m,4H),0.99–0.85(m,6H)。
LC-MS:m/z 290.2[M+H] +
生物学试验
试验例1:本发明化合物对hTLR8和hTLR7的激动活性
本发明化合物对hTLR8和hTLR7的受体结合活性的体外分析使用从Invivogen公司购买的HEK-Blue TMhTLR8细胞和HEK-Blue TMhTLR7细胞。该细胞是在HEK293细胞中共转染hTLR8或hTLR7基因和一个分泌型的碱性磷酸酶(SEAP)的报告基因。其中SEAP的基因是放在IFN-β最小启动子下游,IFN-β最小启动子是由5个NF-κB和AP-1结合位点组成。用hTLR8或hTLR7的刺激剂会激活NF-κB和AP-1启动子而产生SEAP,通过检测SEAP水平来评价化合物的作用。
试验试剂:
HEK-Blue hTLR8细胞和HEK-Blue hTLR7细胞(来源于Invivogen公司)
HEK-BlueTM检测试剂(来源于Invivogen公司)
DMEM培养基(来源于Gibco公司)
胎牛血清(来源于Gibco公司)
Normocin TM、Zeocin和杀稻瘟菌素(Blasticidine)(来源于Invivogen公司)
试验过程:
1、收取细胞培养瓶中的细胞,调整细胞密度为2.2×10 5/mL,用HEK-BlueTM检测试剂重悬细胞,向384孔板中接种45μL细胞悬液,每孔10000个细胞。
2、化合物板准备:待测化合物从2mM起用DMSO稀释3倍稀释10个梯度。取2μL稀释的化合物加入38μL HEK-BlueTM检测试剂进行20倍中间稀释。细胞加0.5%DMSO孔,作为低读值的阴性对照孔。细胞加1μM GS-9688(参考WO2016141092A1合成路线)孔,作为高读值的阳性对照孔。
3、取5μL中间稀释的化合物加入到已接种45μL细胞的384孔板中,对药物进行10倍稀释,DMSO的终浓度为0.5%。
4、将含有细胞和化合物的384孔板放入37℃、5%CO 2的培养箱培养16小时。
5、16小时后,取出培养板,使用仪器VICTOR Nivo检测SEAP在620nm的光吸收。
6、利用GraphPad Prism 8软件分析数据,得到各化合物的EC 50
求取各浓度以及阳性和阴性对照的数据的平均值。由下式计算活性百分比:
活性%=(化合物读值-阴性孔读值)/(阳性孔读值-阴性孔读值)×100。
通过使数据与非线性回归方程式拟合来计算各化合物的IC 50
Y=最低值+(最高值-最低值)/(1+10^((LogEC 50-X)×希尔斜率));
其中,X为化合物浓度的对数,Y为活性百分比。
本发明化合物对TLR8/TLR7的激动活性见下表1。
表1本发明化合物对TLR8/TLR7的激动EC 50
实施例 hTLR8EC 50(μM) hTLR7EC 50(μM)
1 >10 >90
2 0.694 >90
3 0.2718 >90
4 0.5162 >10
5 0.9821 >90
6 1.095 >90
7 2.129 >90
8 4.968 >90
9 0.8597 >90
10 1.57 >90
11 0.1356 >90
12 0.06071 >10
13 0.1443 >90
14 0.07661 >10
15 2.744 >90
16 0.404 >90
17 1.122 >90
18 0.03949 >10
19 0.2503 >90
20 0.02429 >10
21 0.1472 >10
22 0.1271 >90
23 0.1525 >10
24 1.118 >10
25 1.621 >90
26 1.15 >90
27 0.06586 10
28 0.04618 >90
29 0.06834 >90
30 1.18 >90
31 0.1319 >90
32 0.1007 >10
33 0.2339 >10
34 0.2012 >10
35 0.7754 >90
36 1.93 >90
37 2.864 >90
38 1.229 >90
39 0.4759 >90
40 0.6079 >90
41 0.281 >10
42 2.952 >90
43 1.755 >90
44 >10 >90
45 >10 1.772
46 >10 1.176
47 1.461 5.196
48 1.723 6.801
49 >10 >90
50 1.121 >90
51 6.211 >90
52 0.021 3.07
53 1.168 >90
54 3.402 6.388
55 1.238 60.484
56 0.5489 >90
57 >10 >90
58 >10 >90
59 >10 >90
60 0.1372 0.106
61 0.08516 >10
62 0.2409 >90
63 7.693 >90
64 6.859 >90
65 2.777 >90
66 2.295 >90
67 >10 >90
68 0.1296 3.559
69 0.3225 40.765
结论:本发明化合物能够选择性地激活TLR8。
试验例2:实施例52化合物的吸收机制研究
本实验通过检测受试物在Caco-2细胞模型的渗透系数,考察其可能的渗透和吸收情况,使用LC/MS/MS检测化合物在测试样品中的浓度,并计算受试化合物在Caco-2细胞膜的表观渗透系数(Papp)。Caco-2细胞购自美国典型菌种保藏中心,
Figure PCTCN2022128963-appb-000124
编号为HTB-37,经过14天左右的培养后,完全汇合并完成分化。测试为双向给药,即同时检测1)化合物由顶端到基底端的转运速率,2)测定化合物由基底端到顶端的转运速率。
实验涉及到的部分主要试剂信息见下表:
Figure PCTCN2022128963-appb-000125
1、Caco-2细胞准备
用培养基将Caco-2细胞(美国典型菌种保藏中心,HTB-37)稀释至6.86×10 5 个细胞/mL,并将50μL细胞悬液加入到96孔Transwell板(Cat.No.3391)的过滤孔中。将细胞培养板在37℃、5%CO 2、95%相对湿度的细胞培养箱中培养14-18天。每隔一天更换一次细胞培养基。
2、细胞单层稳定性评估
评估前移出旧培养基并更换为预热的新鲜培养基。使用Millicell Epithelial Volt-Ohm测量***(Millipore,USA)测量跨单层的跨上皮电阻(TEER)。测量完成后将板放回培养箱。
TEER值根据以下公式计算:
TEER测量值(ohms)×膜面积(cm 2)=TEER值(ohm·cm 2)
其中TEER值大于230ohm·cm 2,表明Caco-2单层膜合格,可以用于后续测试。
3、制备化合物溶液
准确称取实施例52化合物,利用DMSO进行溶解制备得到浓度为2mM的储备溶液,并用HBSS(Gibico,10mM HEPES,pH 7.4)稀释,得到10μM工作溶液。美托洛尔和地高辛用作对照化合物。
4、进行药物运输试验
试验前利用预热的HBSS(10mM HEPES,pH 7.4)清洗单层两次。然后将培养板置于37℃孵育30分钟。
①确定药物从顶端到基外侧方向的转运速率:125μ工作溶液添加到Transwell(顶端隔室)中,并立即将50μL样品从顶端隔室转移到200μL含有IS(100nM阿普***、200nM咖啡因和100nM甲苯磺丁脲)的新的96孔板作为初始供体样品(AB)。以1000rpm的速度涡旋10分钟。用235μL的缓冲液填充接收器板(基底外侧隔室)中的孔。
②确定药物从基底外侧到根尖方向的转运速率:将285μL的工作溶液添加到接收器板孔(基底外侧隔室)中,并转移50μL样品。
将培养板在37℃孵育2小时。孵育结束时,将来自供体侧(Ap→Bl的顶端隔室,Bl→Ap的基底外侧隔室)和接收侧(Ap→Bl的基底外侧隔室和Bl→Ap的顶端隔室)的50μL样品转移到新的96孔板的孔中,然后加入200μL含有IS(100nM阿普***、200nM咖啡因和100nM甲苯磺丁脲)的乙腈。将样品涡旋10分钟,转移50μL样品到新的96孔板的孔中,然后加入50μL HEPRS和200μL IS。将所有样品涡旋混合10分钟,然后以3,220g离心40分钟。分析之前,将150μL的上清液与适量的超纯水混合后并利用LC-MS/MS进行测试分析。
使用以下等式确定表观渗透率(P app):
Figure PCTCN2022128963-appb-000126
其中P app是表观渗透率(cm/s×10 -6),dQ/dt是药物转运的速率(pmol/秒),A 是膜的表面积(cm 2),D0是初始供体浓度(nM;pmol/cm 3)
使用以下等式确定流出比:
Figure PCTCN2022128963-appb-000127
其中P app(B-A)表示基底外侧至根尖方向的表观渗透系数,P app(A-B)表示根尖至基底外侧方向的表观渗透系数。
试验结果如表2所示。
表2实施例52化合物以及WO2018045144A1的实施例4化合物的Caco-2试
验测试参数
参数 实施例52 WO2018045144A1(实施例4)
P app(A-B) 19.2 2.72
P app(B-A) 31.1 30.0
流出比 1.62 11.0
回收率(%) AP-BL 107 97.2
回收率(%) BL-AP 93.5 102
结论:实施例52化合物具有较好的渗透系数,外排转运体底物风险较低。WO2018045144A1的实施例4化合物渗透性较低,而且有较高的外排比率,很有可能是外排转运体底物。相较而言,本发明化合物实施例52的体外Caco-2测试结果显示比WO2018045144A1的实施例4化合物有更好的渗透性。
试验例3:实施例52化合物的大鼠药物代谢动力学研究
本实验旨在评价实施例52化合物在大鼠体内静脉滴注或灌胃给药后的药代动力学行为。静脉滴注给药:受试化合物配制成0.5毫克/毫升的澄清溶液,溶媒为2%乙醇/40%聚乙二醇300/58%0.01摩尔的盐酸;灌胃给药:受试化合物配制成0.5毫克/毫升的澄清溶液,溶媒为2%乙醇/40%聚乙二醇300/58%0.01摩尔的盐酸。
受试化合物在血浆中的浓度由高效液相色谱-串联质谱(LC-MS/MS)进行测定。采用WinNonlin TM Version 8.3(Pharsight,Mountain View,CA)药动学软件的非房室模型处理血浆和组织浓度,使用线性对数梯形法计算药动学参数。
实施例52化合物的大鼠药代动力学相关参数如下表3所示。
表3实施例52化合物的大鼠静脉滴注和灌胃药代动力学相关参数
Figure PCTCN2022128963-appb-000128
Figure PCTCN2022128963-appb-000129
实施例52化合物在灌胃给药5mg/kg剂量下的大鼠组织分布研究相关参数如下表4所示。
表4实施例52化合物的大鼠灌胃组织分布研究相关参数
Figure PCTCN2022128963-appb-000130
实施例52化合物在灌胃给药5mg/kg剂量下的大鼠肠生物利用度和肝脏首过研究相关参数如下表5所示。
表5实施例52化合物的大鼠灌胃肠生物利用度和肝脏首过研究相关参数
Figure PCTCN2022128963-appb-000131
实施例52化合物静脉滴注0.5小时给药剂量1mg/kg下的大鼠***研究的相关参数如下表6所示。
表6实施例52化合物的大鼠静脉滴注***研究相关参数
Figure PCTCN2022128963-appb-000132
Figure PCTCN2022128963-appb-000133
结论:实施例52化合物具有较低的全身性口服生物利用度,但具有较高的肠生物利用度,可以特异性的富集肝脏组织,主要通过肠道进行***。
试验例4:实施例52化合物的食蟹猴药效学研究
本试验旨在评价实施例52化合物在雄性食蟹猴体内口服给药后的药效学。
口服给药:受试化合物配制为10mg/mL,溶媒为10%乙醇、40%聚乙二醇300、50%去离子水。将适量的受试化合物称量在适量的乙醇中,在搅拌和/或超声作用下使混合物完全溶化。然后在搅拌下加入适当体积的聚乙二醇300。最后,在搅拌下加入适当体积的去离子水,以获得最终浓度的配方。配方在投药前和投药期间将在室温下搅拌至少10分钟。
每组每性别各1只食蟹猴(购自北京协尔鑫生物资源),每天笼旁观察2次。于给药前,给药后30min、1hr、2hr、4hr、8hr和24hr,每只动物每个样品采集0.3mL血清。血清中的生化指标IL12p40、IFN-α和TNF-α浓度由超敏多因子电化学发光分析仪进行测定。
试验涉及到的试剂和仪器见下表:
Figure PCTCN2022128963-appb-000134
供应商MSD全名:Meso Scale Discovery。
血清细胞因子浓度测试过程为:
1)用包被溶液对U-PLEX 96微孔板进行预孵育,室温孵育1小时。
2)用300μL 1x清洗缓冲液洗板3次。
3)每个待测孔加入25μL稀释液(Diluent43),轻轻拍打板子的侧面使Diluent43均匀的分布在MSD微孔板底部。
4)每孔加入25μL TNF-α、IL-12或INF-α标准品或25μL血清样品,用封板膜封板室温震荡孵育1小时。
5)用300ul 1x清洗缓冲液(PBS-0.05%吐温-20)洗板3次。
6)每孔加50μL检测抗体溶液,用封板膜封板,室温下震荡孵育1小时。
7)用300μL 1x清洗缓冲液洗板3次。
8)每孔加150μL MSD GOLD Read Buffer B,用MSD reader(型号:MESO SECTOR S600)读U-PLEX 96微孔板。
9)利用GraphPad Prism 8软件分析数据,得到‘细胞因子浓度-时间’曲线下 面积(AUC)。
图1为实施例52化合物在食蟹猴血清中的参数。
由图1可知,实施例52化合物经10mg/kg口服给药后,化合物对于食蟹猴血清中IL-12p40有明显激活作用,但对TNF-α和INF-α影响不大。

Claims (20)

  1. 一种通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,
    Figure PCTCN2022128963-appb-100001
    其中,
    X 1为CR 1或N;
    X 2为CR 2或N;
    X 3为CR 3或N;
    X 4为CR 4或N;
    L选自一个键、-(CH 2) v-、-C(O)(CH 2) t-或-(CH 2) tC(O)-;
    R 1选自氢、卤素、氨基、硝基、氰基、羟基、巯基、羧基、烷基、烷氧基、卤代烷基、卤代烷氧基;
    R 2选自氢、卤素、氰基、氧代基、烷基、烯基、炔基、-OR a、-SR a、-NR aR b、环烷基、杂环基、芳基和杂芳基,其中所述烷基、烯基、炔基、环烷基、杂环基、芳基、杂芳基任选进一步被一个或多个Q 1基团所取代;
    R 3选自氢、卤素、氰基、氧代基、烷基、烯基、炔基、-OR a、-SR a、-NR aR b、环烷基、杂环基、芳基和杂芳基,其中所述烷基、烯基、炔基、环烷基、杂环基、芳基、杂芳基任选进一步被一个或多个Q 2基团所取代;
    R 4选自氢、卤素、氨基、硝基、氰基、羟基、巯基、羧基、烷基、烷氧基、卤代烷基、卤代烷氧基;
    R 5和R 6各自独立地选自氢、烷基、烯基、炔基、环烷基、杂环基、芳基、杂芳基;其中所述烷基、烯基、炔基、环烷基、杂环基、芳基、杂芳基任选进一步被选自氘代、卤素、硝基、氰基、氧代基、烷基、烯基、炔基、环烷基、杂环基、芳基、杂芳基、-OR a、-SR a、-NR aR b、-C(O)R a、-O(O)CR a、-C(O)OR a、-C(O)NR aR b、-NR aC(O)R b、-S(O) nR a、-S(O) nNR aR b和-NR aS(O) nR b的一个或多个基团取代;
    Q 1和Q 2各自独立地选自卤素、硝基、氰基、氧代基、烷基、烯基、炔基、环烷基、杂环基、芳基、杂芳基、-OR a、-SR a、-(CH 2) v-NR aR b、-NR aR b、-C(O)R a、-O(O)CR a、-C(O)OR a、-C(O)NR aR b、-NR aC(O)R b、-S(O) nR a、-S(O) nNR aR b和-NR aS(O) nR b,其中所述烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基任选进一步被选自卤素、氨基、硝基、氰基、羧基、酯基、氧代基、烷基、烷氧基、环烷基、杂环基、芳基、杂芳基、-OR c、-SR c、-(CH 2) v-OR c、-(CH 2) v-NR cR d、-NR cR d、 -C(O)R c、-O(O)CR c、-C(O)OR c、-C(O)NR cR d、-NR cC(O)R d、-S(O) nR a、-S(O) nNR cR d和-NR cS(O) nR d的一个或多个基团取代;
    R a和R b各自独立地选自氢、卤素、羟基、硝基、氰基、氧代、烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基,其中所述烷基、烯基、炔基、环烷基、杂环基、芳基、杂芳基任选进一步被选自卤素、氨基、硝基、氰基、羧基、酯基、氧代基、烷基、环烷基、杂环基、芳基、杂芳基、-OR c、-SR c、-(CH 2) v-OR c、-(CH 2) v-NR cR d、-NR cR d、-C(O)R c、-O(O)CR c、-C(O)OR c、-C(O)NR cR d、-NR cC(O)R d、-S(O) nR a、-S(O) nNR cR d和-NR cS(O) nR d中的一个或多个基团取代;
    或者R a和R b与他们连接的氮原子一起形成含氮杂环基,所述含氮杂环基除了N之外,任选进一步含有选自N、O、S的一个或多个杂原子,所述含氮杂环基任选进一步被选自卤素、硝基、氰基、氧代基、羧基、酯基、烷基、卤代烷基、环烷基、杂环基、芳基、杂芳基、-OR c、-SR c、-(CH 2) v-OR c、-(CH 2) v-NR cR d、-NR cR d、-C(O)R c、-O(O)CR c、-C(O)OR c、-C(O)NR cR d、-NR cC(O)R d、-S(O) nR a、-S(O) nNR cR d和-NR cS(O) nR d的一个或多个基团取代;
    R c和R d各自独立地选自氢、卤素、羟基、硝基、氰基、氧代、烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基,其中所述烷基、烯基、炔基、环烷基、杂环基、芳基、杂芳基任选进一步被选自卤素、氨基、硝基、氰基、羟基、巯基、羧基、酯基、氧代基、烷基、烷氧基、环烷基、杂环基、芳基和杂芳基中的一个或多个基团取代;
    或者R c和R d与他们连接的氮原子一起形成含氮杂环基,所述含氮杂环基除了N之外,任选进一步含有选自N、O、S的一个或多个杂原子,所述含氮杂环基任选进一步被选自卤素、硝基、氰基、氧代基、羟基、巯基、羧基、酯基、烷基、烷氧基、环烷基、杂环基、芳基和杂芳基;
    n为1或2;
    v为1至6的整数;
    t为0至6。
  2. 根据权利要求1所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,其为通式(II)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,
    Figure PCTCN2022128963-appb-100002
    Figure PCTCN2022128963-appb-100003
    其中,X 1、X 2、X 3、L、R 4、R 5、R 6如权利要求1所定义。
  3. 根据权利要求1所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,其为通式(III)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,
    Figure PCTCN2022128963-appb-100004
    其中,L、R 1、R 3、R 4、R 5、R 6如权利要求1所定义。
  4. 根据权利要求1所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,其为通式(IV)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,
    Figure PCTCN2022128963-appb-100005
    其中,L、R 1、R 2、R 4、R 5、R 6如权利要求1所定义。
  5. 根据权利要求1所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,其为通式(V)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,
    Figure PCTCN2022128963-appb-100006
    其中,L、R 2、R 4、R 5、R 6如权利要求1所定义。
  6. 根据权利要求1至5任一项所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,其中:L选自一个键或-C(O)-;优选一个键。
  7. 根据权利要求1至6中任一项所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,其为通式(I-1)、(II-1)、(III-1)、(IV-1)、(V-1)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,
    Figure PCTCN2022128963-appb-100007
    其中,X 1、X 2、X 3、X 4、R 1、R 2、R 3、R 4、R 5、R 6如权利要求1所定义。
  8. 根据权利要求1至3、6至7中任一项所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,
    其中,
    R 3选自氢、卤素、C 1-C 6烷基、C 6-C 10芳基和5至10元杂芳基,其中所述C 1-C 6烷基、C 6-C 10芳基和5至10元杂芳基任选进一步被一个或多个Q 2基团所取代;
    Q 2选自卤素、C 1-C 6烷基、4-6元杂环基、C 6-C 10芳基、5至10元杂芳基、-NR aR b,其中所述C 6-C 10芳基和5至10元杂芳基任选进一步被选自卤素、C 1-C 6烷基、-(CH 2) v-NR cR d的一个或多个基团取代;
    R a和R b各自独立地选自氢、C 1-C 6烷基;
    或者R a和R b与他们连接的氮原子一起形成4-6元含氮杂环基,所述4-6元含氮杂环基除了N之外,任选进一步含有选自N、O、S的一个或多个杂原子,所述4-6元含氮杂环基任选进一步被选自氧代基、C 1-C 6烷基、C 1-C 6卤代烷基、C 3-C 6环烷基、-OR c、-SR c、-(CH 2) v-OR c、-NR cR d的一个或多个基团取代;
    R c和R d各自独立地选自氢、C 1-C 6烷基;
    或者R c和R d与他们连接的氮原子一起形成4-6元含氮杂环基,所述4-6元含氮杂环基除了N之外,任选进一步含有选自N、O、S的一个或多个杂原子,所述4-6元含氮杂环基任选进一步被选自卤素、C 1-C 6烷基的一个或多个基团取代;
    v为1至6的整数。
  9. 根据权利要求8所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,其中,
    R 3选自C 6-C 10芳基和5至10元杂芳基,优选苯基或5-6元杂芳基;其中所述C 6-C 10芳基和5至10元杂芳基任选进一步被一个或多个Q 2基团所取代;
    Q 2选自卤素、C 1-C 6烷基、4-6元杂环基、C 6-C 10芳基、5至10元杂芳基、-NR aR b,其中所述C 6-C 10芳基和5至10元杂芳基任选进一步被选自卤素、C 1-C 6烷基的一个或多个基团取代;
    R a和R b各自独立地选自氢、C 1-C 6烷基;
    或者R a和R b与他们连接的氮原子一起形成4-6元含氮杂环基,所述4-6元含氮杂环基除了N之外,任选进一步含有选自N、O、S的一个或多个杂原子,所述4-6元含氮杂环基任选进一步被选自氧代基、C 1-C 6烷基、C 1-C 6卤代烷基、C 3-C 6环烷基、-OR c、-SR c、-(CH 2) v-OR c、-NR cR d的一个或多个基团取代;
    R c和R d各自独立地选自氢、C 1-C 6烷基;
    或者R c和R d与他们连接的氮原子一起形成4-6元含氮杂环基,所述4-6元含氮杂环基除了N之外,任选进一步含有选自N、O、S的一个或多个杂原子,所述4-6元含氮杂环基任选进一步被选自卤素、C 1-C 6烷基的一个或多个基团取代v为1至6的整数,优选1或2。
  10. 根据权利要求1至2、4至7中任一项所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,
    其中,R 2选自氢、卤素、C 1-C 6烷基、-NR aR b,所述C 1-C 6烷基任选进一步被Q 1取代;
    Q 1选自C 6-C 10芳基、5至10元杂芳基,其中所述C 6-C 10芳基和5至10元杂芳基任选进一步被选自卤素、C 1-C 6烷基、-(CH 2) v-NR cR d的一个或多个基团取代;
    R a、R b各自独立地选自氢、C 1-C 6烷基;
    R c和R d各自独立地选自氢、C 1-C 6烷基;
    或者R c和R d与他们连接的氮原子一起形成4-6元含氮杂环基,所述4-6元含氮杂环基除了N之外,任选进一步含有选自N、O、S的一个或多个杂原子,所述4-6元含氮杂环基任选进一步被选自卤素、C 1-C 6烷基的一个或多个基团取代;
    v为1至6的整数,优选1或2。
  11. 根据权利要求1至10中任一项所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,
    其中,R 1选自氢、卤素、C 1-C 6烷基、C 1-C 6烷氧基、C 1-C 6卤代烷基、C 1-C 6卤代烷氧基;优选,R 1为氢或卤素。
  12. 根据权利要求1至11中任一项所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,
    其中,R 4选自氢、卤素、C 1-C 6烷基、C 1-C 6烷氧基、C 1-C 6卤代烷基、C 1-C 6卤代烷氧基;优选,R 4为氢或卤素。
  13. 根据权利要求1至12中任一项所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,
    其中:
    R 5和R 6各自独立地选自氢和C 1-C 12烷基,所述C 1-C 12烷基任选进一步被选自氘代、-OR a、-SR a、-NR aR b的一个或多个基团取代;
    R a选自氢、C 1-C 6烷基;
    R b选自氢、C 1-C 6烷基、C 3-C 6环烷基和5至7元杂环基;
    优选地,R 5为氢,R 6为C 1-C 12烷基,所述C 1-C 12烷基任选进一步被选自氘代、-OH的一个或多个基团取代。
  14. 根据权利要求1至13中任一项所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,其选自:
    Figure PCTCN2022128963-appb-100008
    Figure PCTCN2022128963-appb-100009
    Figure PCTCN2022128963-appb-100010
    Figure PCTCN2022128963-appb-100011
  15. 一种制备通式(III-1)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐的方法,其包括以下步骤:
    Figure PCTCN2022128963-appb-100012
    式A3的化合物与硼酸或硼酸频哪醇酯化合物通过金属催化的交叉偶联反应如Suzuki偶联,得到式A4的化合物;然后,用合适的酸如三氟乙酸脱保护基,得到通式(III-1)所示的化合物;
    或者,
    Figure PCTCN2022128963-appb-100013
    式A7的化合物与硼酸或硼酸频哪醇酯化合物通过金属催化的交叉偶联反应如Suzuki偶联,得到通式(III-1)所示的化合物;
    R 1、R 3、R 4、R 5、R 6如权利要求7所定义。
  16. 一种制备通式(IV-1)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐的方法,其包括以下步骤:
    Figure PCTCN2022128963-appb-100014
    式B3的化合物与硼酸或硼酸频哪醇酯化合物通过金属催化的交叉偶联反应如Suzuki偶联,得到式B4的化合物;然后,用合适的酸如三氟乙酸脱保护基,得到通式(IV-1)所示的化合物;
    或者,
    Figure PCTCN2022128963-appb-100015
    式B7的化合物与硼酸或硼酸频哪醇酯化合物通过金属催化的交叉偶联反应如Suzuki偶联,得到通式(IV-1)所示的化合物;
    其中:R 1、R 2、R 4、R 5、R 6如权利要求7所定义。
  17. 一种制备通式(V-1)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐的方法,其包括以下步骤:
    Figure PCTCN2022128963-appb-100016
    式C3的化合物与硼酸或硼酸频哪醇酯化合物通过金属催化的交叉偶联反应如Suzuki偶联,得到式C4的化合物;然后,用合适的酸如三氟乙酸脱保护基,得到通式(V-1)所示的化合物;
    或者,可以通过以下方案6制备通式(V-1)所示的化合物:
    Figure PCTCN2022128963-appb-100017
    式C7的化合物与硼酸或硼酸频哪醇酯化合物通过金属催化的交叉偶联反应如Suzuki偶联,得到通式(V-1)所示的化合物;
    其中:R 2、R 4、R 5、R 6如权利要求7所定义。
  18. 一种药物组合物,其包含根据权利要求1至14中任一项所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,以及药学上可接受的载体或赋形剂。
  19. 根据权利要求1至14中任一项所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐或根据权利要求18所述的药物组合物在制备TLR8激动剂中的用途。
  20. 根据权利要求1至14中任一项所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐或根据权利要求18所述的药物组合物在制备预防或治疗TLR8相关疾病的药物中的用途,所述疾病优选病毒感染性疾病和恶性肿瘤,所述病毒感染性疾病例如乙型病毒性肝炎、HIV病毒感染,所述恶性肿瘤例如乳腺癌、***、结肠癌、肺癌、胃癌、直肠癌、胰腺癌、脑癌、皮肤癌、口腔癌、***癌、骨癌、肾癌、卵巢癌、膀胱癌、肝癌、输卵管肿瘤、卵巢瘤、腹膜肿瘤、黑色素瘤、实体瘤、神经胶质瘤、神经胶母细胞瘤、肝细胞癌、乳突肾性瘤、头颈部肿瘤、白血病、淋巴瘤、骨髓瘤和非小细胞肺癌。
PCT/CN2022/128963 2021-11-05 2022-11-01 芳胺类衍生物及其制备方法和医药用途 WO2023078241A1 (zh)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202280007466.7A CN116472047A (zh) 2021-11-05 2022-11-01 芳胺类衍生物及其制备方法和医药用途

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202111304202 2021-11-05
CN202111304202.1 2021-11-05

Publications (1)

Publication Number Publication Date
WO2023078241A1 true WO2023078241A1 (zh) 2023-05-11

Family

ID=86240671

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/128963 WO2023078241A1 (zh) 2021-11-05 2022-11-01 芳胺类衍生物及其制备方法和医药用途

Country Status (3)

Country Link
CN (1) CN116472047A (zh)
TW (1) TW202327601A (zh)
WO (1) WO2023078241A1 (zh)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107108615A (zh) * 2015-03-04 2017-08-29 吉利德科学公司 Toll样受体调节性4,6‑二氨基‑吡啶并[3,2‑D]嘧啶化合物
CN108069963A (zh) * 2017-11-17 2018-05-25 清华大学 吡啶并嘧啶衍生物或其盐及其制法、药物组合物和用途
CN109923106A (zh) * 2016-09-02 2019-06-21 吉利德科学公司 toll样受体调节剂化合物
WO2020007275A1 (zh) * 2018-07-03 2020-01-09 江苏恒瑞医药股份有限公司 吡啶并嘧啶类衍生物、其制备方法及其在医药上的应用
WO2020057604A1 (zh) * 2018-09-19 2020-03-26 正大天晴药业集团股份有限公司 Tlr8激动剂
WO2020078455A1 (zh) * 2018-10-19 2020-04-23 南京明德新药研发有限公司 Tlr8激动剂

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107108615A (zh) * 2015-03-04 2017-08-29 吉利德科学公司 Toll样受体调节性4,6‑二氨基‑吡啶并[3,2‑D]嘧啶化合物
CN109923106A (zh) * 2016-09-02 2019-06-21 吉利德科学公司 toll样受体调节剂化合物
CN108069963A (zh) * 2017-11-17 2018-05-25 清华大学 吡啶并嘧啶衍生物或其盐及其制法、药物组合物和用途
WO2020007275A1 (zh) * 2018-07-03 2020-01-09 江苏恒瑞医药股份有限公司 吡啶并嘧啶类衍生物、其制备方法及其在医药上的应用
WO2020057604A1 (zh) * 2018-09-19 2020-03-26 正大天晴药业集团股份有限公司 Tlr8激动剂
WO2020078455A1 (zh) * 2018-10-19 2020-04-23 南京明德新药研发有限公司 Tlr8激动剂

Also Published As

Publication number Publication date
CN116472047A (zh) 2023-07-21
TW202327601A (zh) 2023-07-16

Similar Documents

Publication Publication Date Title
AU2018226315B2 (en) FGFR inhibitor and application thereof
WO2021218110A1 (zh) 一类苯并噻唑基联芳基类化合物、制备方法和用途
CN105085483B (zh) 激酶抑制剂及其应用
EA025466B1 (ru) Определенные триазолопиридины и триазолопиразины, их композиции и способы их применения
WO2021143701A1 (zh) 嘧啶-4(3h)-酮类杂环化合物、其制备方法及其在医药学上的应用
TW200829589A (en) Compound for inhibiting mitotic progression
CN110167941B (zh) 取代的稠合杂芳基化合物作为激酶抑制剂及其应用
CN107312009B (zh) 喹啉类化合物、其制备方法、中间体、药物组合物和应用
TWI781607B (zh) 一種免疫抑制劑、其製備方法和應用
WO2020200069A1 (zh) 吡咯并杂环类衍生物、其制备方法及其在医药上的应用
CN115710266A (zh) 吡啶并嘧啶类衍生物、其制备方法及其在医药上的应用
TW201910331A (zh) 成纖維細胞生長因子受體抑制劑、含有其的藥物製劑及其用途
WO2022247816A1 (zh) 含氮杂环类化合物、其制备方法及其在医药上的应用
WO2022228543A1 (zh) 桥环类化合物、其制备方法及其在医药上的应用
WO2022213980A1 (zh) Tyk2抑制剂及其用途
WO2022017408A1 (zh) 芳胺类衍生物及其制备方法和医药用途
CN110312717B (zh) 取代的稠合杂芳三环化合物作为激酶抑制剂及其应用
CN115197208A (zh) 杂芳基类化合物、其制备方法及其在医药上的应用
WO2021249319A1 (zh) 三环化合物、药物组合物及其应用
WO2020221209A1 (zh) 一种cd73抑制剂,其制备方法和应用
WO2023078241A1 (zh) 芳胺类衍生物及其制备方法和医药用途
WO2018171611A1 (zh) 6-吡唑-[1,2,4]***并[4,3-a]吡啶-3-酰胺类衍生物、其制备方法及其在医药上的应用
WO2023040771A1 (zh) 含氮稠环类化合物及其制备方法和医药用途
WO2024046471A9 (zh) Usp1抑制剂
WO2023088425A1 (zh) 15-pgdh抑制剂及用途

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 202280007466.7

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 22889265

Country of ref document: EP

Kind code of ref document: A1