CN116472047A - Arylamine derivative, preparation method and medical application thereof - Google Patents

Arylamine derivative, preparation method and medical application thereof Download PDF

Info

Publication number
CN116472047A
CN116472047A CN202280007466.7A CN202280007466A CN116472047A CN 116472047 A CN116472047 A CN 116472047A CN 202280007466 A CN202280007466 A CN 202280007466A CN 116472047 A CN116472047 A CN 116472047A
Authority
CN
China
Prior art keywords
compound
alkyl
aryl
formula
halogen
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202280007466.7A
Other languages
Chinese (zh)
Inventor
闫旭
田卫学
陈士柱
殷惠军
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
National Institutes of Pharmaceutical R&D Co Ltd
Original Assignee
National Institutes of Pharmaceutical R&D Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by National Institutes of Pharmaceutical R&D Co Ltd filed Critical National Institutes of Pharmaceutical R&D Co Ltd
Publication of CN116472047A publication Critical patent/CN116472047A/en
Pending legal-status Critical Current

Links

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

Abstract

The present disclosure relates to arylamine derivatives, methods of preparation and medical uses thereof. In particular, it relates to compounds of formula (I), methods for their preparation, pharmaceutical compositions containing them and their use as Toll-like receptor (TLR) agonists for the treatment of diseases associated with TLR8 activity. The definition of each group in the general formula (I) is the same as that in the specification.

Description

Arylamine derivative, preparation method and medical application thereof Technical Field
The invention relates to arylamine derivatives, a preparation method and medical application thereof. In particular, the invention relates to compounds of formula (I), methods for their preparation, pharmaceutical compositions containing them and their use as Toll-like receptor (TLR) agonists for the treatment of diseases associated with TLR8 activity.
Background
Toll-like receptors (TLRs) are a class of pattern recognition receptors that recognize and respond to microorganisms. TLR family members play an important role in the immune system, both as important elements involved in innate immunity and as a bridge linking innate immunity to specific immunity. The receptor can specifically recognize microorganisms and initiate an immune response.
The TLRs are type I transmembrane glycoproteins, consisting of an extracellular domain of a repeat sequence (LRR) rich in 16-28 leucine, a transmembrane domain, and a cytoplasmic Toll/IL-1 receptor (TIR) domain. X-ray crystallography analysis determined that the TLR LRR domains were horseshoe-like structures. To date 11 members have been found in humans, with TLR1, 2, 4, 5, 6, 10 and 11 located on the cell surface and TLR3, 7, 8, 9 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 adjacent X chromosomes (Xp 22). Upon binding of the LRR of a TLR to a ligand, the TIR domain conformation changes, which in turn triggers activation of the TLR signaling pathway. The TIR domain of a TLR can recruit a variety of signaling molecules, including tumor necrosis factor receptor-related factor 6 and myeloid differentiation factor 88 (MyD 88), among others. Wherein TLR8 depends on MyD88 signal path, induces activation of proteinase-1 (AP-1) and nuclear factor κB (NF- κB) to transfer into nucleus, induces expression of related genes in nucleus, secretes chemotactic factors and inflammatory factors, and plays a role in transcriptional regulation. In addition, TLR8 can activate mitogen-activated protein kinase (MAPK) signaling pathways, including p38, ERK, JNK, etc., mainly involved in the regulation of cell proliferation, cell differentiation, cell transformation, apoptosis, etc., and is closely related to various diseases such as inflammation and tumor (journal of immunology, 2017, 33, 813).
Hepatitis B Virus (HBV) is a particulate double stranded DNA virus. Activating TLR8 can effectively inhibit the replication of hepatitis B virus in vivo and in vitro, so that the TLR8 becomes a target for developing and treating chronic hepatitis B virus. The study shows that the TLR8 agonist ssRNA40 can selectively activate the innate immune cells around the liver to generate a large amount of IFN-gamma so as to inhibit the replication of hepatitis B virus, thereby having potential application as treatment of hepatitis virus infection. Stimulation of PBMCs with TLR8 agonists was found to induce high levels of IFN- γ and TNF- α production, thereby inhibiting HBV replication (Current Opinion in Virology,2018, 30,9).
The TLR is expressed not only on immune cells, but also in various tumor cells, and participates in tumor immune monitoring, and plays different roles in tumor growth. Wherein TLR8 enhances natural killer cell (NK cell) activity upon activation and enhances antibody-dependent cell-mediated cytotoxicity (ADCC) and induces Th1 polarization. TLR8 agonists are a potential adjuvant in cancer therapy, with the aim of inducing specific immune responses against tumor cells, improving the clinical efficacy of approved monoclonal antibody therapies, especially in individuals with reduced ADCC.
Given the important potential of TLR-8 agonists for the treatment of a variety of diseases, there is an urgent clinical need for novel TLR-8 agonists with potent activity and high selectivity.
Disclosure of Invention
Through intensive research, the inventor designs and synthesizes a series of aromatic amine compounds which show excellent TLR8 agonistic activity and can be developed into medicaments for treating diseases related to TLR 8.
It is therefore an object of the present invention to provide a compound of the general formula (I) or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer or mixture thereof, or a pharmaceutically acceptable salt thereof,
wherein,
X 1 is CR (CR) 1 Or N;
X 2 is CR (CR) 2 Or N;
X 3 is CR (CR) 3 Or N;
X 4 is CR (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 1 Selected from the group consisting of hydrogen, halogen, amino, nitro, cyano, hydroxy, mercapto, carboxyl, alkyl, alkoxy, haloalkyl, haloalkoxy;
R 2 selected from hydrogen, halogen, cyano, oxo, alkyl, alkenyl, alkynyl, -OR a 、-SR a 、-NR a R b Cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl is optionally further substituted with one or more Q 1 Substituted with a group;
R 3 selected from hydrogen, halogen, cyano, oxo, alkyl, alkenyl, alkynyl, -OR a 、-SR a 、-NR a R b Cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl is optionally further substituted with one or more Q 2 Substituted with a group;
R 4 selected from the group consisting of hydrogen, halogen, amino, nitro, cyano, hydroxy, mercapto, carboxyl, alkyl, alkoxy, haloalkyl, haloalkoxy;
R 5 and R is 6 Each independently selected from hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl; which is a kind ofOptionally further selected from the group consisting of deuterated, 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)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 Is substituted with one or more groups;
Q 1 and Q 2 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 the group consisting of halogen, amino, nitro, cyano, carboxyl, ester, oxo, alkyl, alkoxy, 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)NR c R d 、-NR c C(O)R d 、-S(O) n R a 、-S(O) n NR c R d and-NR c S(O) n R d Is substituted with one or more groups;
R a and R is b Each independently selected from the group consisting of hydrogen, halogen, hydroxy, nitro, cyano, oxo, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein the alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl is optionally further selected from the group consisting of 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)NR c R d 、-NR c C(O)R d 、-S(O) n R a 、-S(O) n NR c R d and-NR c S(O) n R d Is substituted with one or more groups;
or R is a And R is b Together with the nitrogen atom to which they are attached, form a nitrogen-containing heterocyclic group optionally further containing one OR more heteroatoms selected from N, O, S in addition to N, said nitrogen-containing heterocyclic group optionally being further substituted with a moiety selected from halogen, nitro, cyano, oxo, carboxyl, ester, alkyl, haloalkyl, 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)NR c R d 、-NR c C(O)R d 、-S(O) n R a 、-S(O) n NR c R d and-NR c S(O) n R d Is substituted with one or more groups;
R c and R is d Each independently selected from the group consisting of hydrogen, halogen, hydroxy, nitro, cyano, oxo, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein the alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl is optionally further substituted with one or more groups selected from the group consisting of halogen, amino, nitro, cyano, hydroxy, mercapto, carboxyl, ester, oxo, alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl, and heteroaryl;
Or R is c And R is d Together with the nitrogen atom to which they are attached, form a nitrogen-containing heterocyclic group optionally further containing one or more heteroatoms selected from N, O, S in addition to N, the nitrogen-containing heterocyclic group optionally being further substituted with a moiety selected from halogen, nitro, cyano, oxo, hydroxy, mercapto, carboxyl, ester, alkyl, alkoxy, cycloalkyl, heterocyclic, aryl and heteroaryl;
n is 1 or 2;
v is an integer from 1 to 6;
t is 0 to 6.
In a specific embodiment, the compounds of formula (I) according to the invention or stereoisomers, tautomers, meso, racemates, enantiomers, diastereomers, or mixtures thereof, or pharmaceutically acceptable salts thereof, are compounds of formula (II) or stereoisomers, tautomers, meso, racemates, enantiomers, diastereomers, or mixtures thereof, or pharmaceutically acceptable salts thereof,
wherein X is 1 、X 2 、X 3 、L、R 4 、R 5 、R 6 As defined by formula (I).
In another specific embodiment, the compound of formula (I) according to the present invention or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, is a compound of formula (III) or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof,
Therein, L, R 1 、R 3 、R 4 、R 5 、R 6 As defined by formula (I).
In another specific embodiment, the compound of formula (I) according to the present invention or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, is a compound of formula (IV) or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof,
therein, L, R 1 、R 2 、R 4 、R 5 、R 6 As defined by formula (I).
In another specific embodiment, the compound of formula (I) according to the present invention or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, is a compound of formula (V) or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof,
therein, L, R 2 、R 4 、R 5 、R 6 As defined by formula (I).
In another specific embodiment, the compounds according to the invention of formulae (I) to (V) or stereoisomers, tautomers, meso, racemates, enantiomers, diastereomers, or mixtures thereof, or pharmaceutically acceptable salts thereof, wherein:
L is selected from a bond or-C (O) -; preferably a key.
In another specific embodiment, the compounds according to the invention of formulae (I) to (V) or stereoisomers, tautomers, meso, racemates, enantiomers, diastereomers, or mixtures thereof, or pharmaceutically acceptable salts thereof, wherein:
l is selected from- (CH) 2 ) v -; v is 1 or 2, preferably 1.
In another specific embodiment, the compounds of formulae (I) to (V) according to the invention or stereoisomers, tautomers, meso, racemates, enantiomers, diastereomers, or mixtures thereof, or pharmaceutically acceptable salts thereof, are compounds of formulae (I-1), (II-1), (III-1), (IV-1), (V-1) or stereoisomers, tautomers, meso, racemates, enantiomers, diastereomers, or mixtures thereof, or pharmaceutically acceptable salts thereof,
wherein X is 1 、X 2 、X 3 、X 4 、R 1 、R 2 、R 3 、R 4 、R 5 、R 6 As defined by formula (I).
In another specific embodiment, the compounds of formulae (I) to (V) or formulae (I-1), (II-1), (III-1), (IV-1), (V-1) according to the invention or stereoisomers, tautomers, meso, racemates, enantiomers, diastereomers, or mixtures thereof, or pharmaceutically acceptable salts thereof, wherein,
R 3 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 optionally further substituted with one or more Q 2 Substituted with a group;
Q 2 selected from halogen, C 1 -C 6 Alkyl, 4-6 membered heterocyclyl, 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 selected from halogen, C 1 -C 6 Alkyl, - (CH) 2 ) v -NR c R d Is substituted with one or more groups;
R a and R is b Each independently selected from hydrogen, C 1 -C 6 An alkyl group;
or R is a And R is b Together with the nitrogen atom to which they are attached, form a 4-6 membered nitrogen containing heterocyclic group, said 4-6 membered nitrogen containing heterocyclic group optionally further containing one or more heteroatoms selected from N, O, S in addition to N, said 4-6 membered nitrogen containing heterocyclic group optionally being 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 Is substituted with one or more groups;
R c and R is d Each independently selected from hydrogen, C 1 -C 6 An alkyl group;
or R is c And R is d Together with the nitrogen atom to which they are attached, form a 4-6 membered nitrogen containing heterocyclic group, said 4-6 membered nitrogen containing heterocyclic group optionally further containing one or more heteroatoms selected from N, O, S in addition to N, said 4-6 membered nitrogen containing heterocyclic group optionally being further selected from halogen, C 1 -C 6 One or more groups of the alkyl group are substituted;
v is an integer from 1 to 6.
In another particular embodiment, the compounds of the formulae (I) to (V) or of the formulae (I-1), (II-1), (III-1), (IV-1), (V-1) according to the invention or stereoisomers, tautomers, meso, racemates, enantiomers, diastereomers, or mixtures thereof, or pharmaceutically acceptable salts thereof, wherein R 3 Selected from hydrogen, halogen, C 1 -C 6 Alkyl groups, preferably hydrogen.
In another specific embodiment, the compounds of formulae (I) to (V) or formulae (I-1), (II-1), (III-1), (IV-1), (V-1) according to the invention or stereoisomers, tautomers, meso, racemates, enantiomers, diastereomers, or mixtures thereof, or pharmaceutically acceptable salts thereof, wherein,
R 3 selected from C 6 -C 10 Aryl and 5 to 10 membered heteroaryl, preferably phenyl or 5-6 membered heteroaryl; wherein said C 6 -C 10 Aryl and 5 to 10 membered heteroaryl optionally further substituted with one or more Q 2 Substituted with a group;
Q 2 selected from halogen, C 1 -C 6 Alkyl, 4-6 membered heterocyclyl, 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 selected from halogen, C 1 -C 6 One or more groups of the alkyl group are substituted;
R a and R is b Each independently selected from hydrogen, C 1 -C 6 An alkyl group;
or R is a And R is b Together with the nitrogen atom to which they are attached, form a 4-6 membered nitrogen containing heterocyclic group, said 4-6 membered nitrogen containing heterocyclic group optionally further containing one or more heteroatoms selected from N, O, S in addition to N, said 4-6 membered nitrogen containing heterocyclic group optionally being 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 Is substituted with one or more groups;
R c and R is d Each independently selected from hydrogen, C 1 -C 6 An alkyl group;
or R is c And R is d Together with the nitrogen atom to which they are attached, form a 4-6 membered nitrogen containing heterocyclic group, said 4-6 membered nitrogen containing heterocyclic group optionally further containing one or more heteroatoms selected from N, O, S in addition to N, said 4-6 membered nitrogen containing heterocyclic group optionally being further substitutedSelected from halogen, C 1 -C 6 One or more groups of the alkyl group are substituted v is an integer from 1 to 6, preferably 1 or 2.
In another specific embodiment, the compounds of formulae (I) to (V) or formulae (I-1), (II-1), (III-1), (IV-1), (V-1) according to the invention or stereoisomers, tautomers, meso, racemates, enantiomers, diastereomers, or mixtures thereof, or pharmaceutically acceptable salts thereof, wherein,
R 3 Selected from 5 to 10 membered heteroaryl, preferably 5 or 6 membered heteroaryl, more preferably pyridyl, pyrimidinyl, imidazolyl, pyrazolyl, pyrrolyl, pyridoimidazolyl, pyridopyrrolyl, pyridopyrazolyl, benzimidazolyl, benzopyrazolyl, benzopyrrolyl; the heteroaryl is optionally further substituted with one or more Q 2 Substituted with a group;
Q 2 selected from halogen, C 1 -C 6 Alkyl, C 6 -C 10 Aryl is preferably phenyl, 5 to 10 membered heteroaryl is preferably 5 or 6 membered heteroaryl, wherein the aryl and heteroaryl are optionally further selected from halogen, C 1 -C 6 One or more groups of the alkyl group are substituted.
In another specific embodiment, the compounds of formulae (I) to (V) or formulae (I-1), (II-1), (III-1), (IV-1), (V-1) according to the invention or stereoisomers, tautomers, meso, racemates, enantiomers, diastereomers, or mixtures thereof, or pharmaceutically acceptable salts thereof, wherein,
R 3 selected from 5 to 10 membered heteroaryl, preferably 5 or 6 membered heteroaryl, more preferably pyridyl, pyrimidinyl, imidazolyl, pyrazolyl, pyrrolyl, pyridoimidazolyl, pyridopyrrolyl, pyridopyrazolyl, benzimidazolyl, benzopyrazolyl, benzopyrrolyl; the heteroaryl is optionally further substituted with one or more Q 2 Substituted with a group;
Q 2 selected from halogen, C 1 -C 6 Alkyl, 4-6 membered heterocyclyl or-NR a R b preferably-NR a R b
R a And R is b Each independently selected from hydrogen, C 1 -C 6 An alkyl group;
or R is a And R is b Together with the nitrogen atom to which they are attached, form a 4-6 membered nitrogen containing heterocyclic group, said 4-6 membered nitrogen containing heterocyclic group optionally further containing one or more heteroatoms selected from N, O, S in addition to N, said 4-6 membered nitrogen containing heterocyclic group optionally being 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 Is substituted with one or more groups;
R c and R is d Each independently selected from hydrogen, C 1 -C 6 An alkyl group;
or R is c And R is d Together with the nitrogen atom to which they are attached, form a 4-6 membered nitrogen containing heterocyclic group, said 4-6 membered nitrogen containing heterocyclic group optionally further containing one or more heteroatoms selected from N, O, S in addition to N, said 4-6 membered nitrogen containing heterocyclic group optionally being further selected from halogen, C 1 -C 6 One or more groups of the alkyl group are substituted v is an integer from 1 to 6, preferably 1 or 2.
In another specific embodiment, the compounds of formulae (I) to (V) or formulae (I-1), (II-1), (III-1), (IV-1), (V-1) according to the invention or stereoisomers, tautomers, meso, racemates, enantiomers, diastereomers, or mixtures thereof, or pharmaceutically acceptable salts thereof, wherein,
R 3 Selected from C 6 -C 10 Aryl, preferably phenyl; the aryl group is optionally further Q 2 Substituted with a group;
Q 2 selected from 5-or 6-membered heteroaryl, wherein the 5-or 6-membered heteroaryl is optionally further selected from halogen, C 1 -C 6 One or more groups of the alkyl group are substituted.
In another specific embodiment, the compounds of formulae (I) to (V) or formulae (I-1), (II-1), (III-1), (IV-1), (V-1) according to the invention or stereoisomers, tautomers, meso, racemates, enantiomers, diastereomers, or mixtures thereof, or pharmaceutically acceptable salts thereof, wherein,
R 3 selected from C 1 -C 6 Alkyl, preferably methyl; the C is 1 -C 6 Alkyl is optionally further substituted with Q 2 Substituted with a group;
Q 2 selected from C 6 -C 10 Aryl, preferably phenyl, wherein said C 6 -C 10 Aryl is optionally further substituted by- (CH) 2 ) v -NR c R d Substitution;
R c and R is d Together with the nitrogen atom to which they are attached, form a 4-6 membered nitrogen containing heterocyclic group, said 4-6 membered nitrogen containing heterocyclic group optionally further containing one or more heteroatoms selected from N, O, S in addition to N, said 4-6 membered nitrogen containing heterocyclic group optionally being further selected from halogen, C 1 -C 6 One or more groups of the alkyl group are substituted;
v is an integer from 1 to 6, preferably 1 or 2.
In another specific embodiment, the compounds of formulae (I) to (V) or formulae (I-1), (II-1), (III-1), (IV-1), (V-1) according to the invention or stereoisomers, tautomers, meso, racemates, enantiomers, diastereomers, or mixtures thereof, or pharmaceutically acceptable salts thereof, wherein,
R 2 Selected from hydrogen, halogen, C 1 -C 6 Alkyl, -NR a R b The C is 1 -C 6 Alkyl is optionally further substituted with Q 1 Substitution;
Q 1 selected from C 6 -C 10 Aryl, 5 to 10 membered heteroaryl, wherein the C is 6 -C 10 Aryl and 5 to 10 membered heteroaryl are optionally further selected from halogen, C 1 -C 6 Alkyl, - (CH) 2 ) v -NR c R d Is substituted with one or more groups;
R a 、R b each independently selected from hydrogen, C 1 -C 6 An alkyl group;
R c and R is d Each independently selected from hydrogen, C 1 -C 6 An alkyl group;
or R is c And R is d Together with the nitrogen atom to which they are attached, form a 4-6 membered nitrogen containing heterocyclic group, said 4-6 membered nitrogen containing heterocyclic group optionally further containing one or more heteroatoms selected from N, O, S in addition to N, said 4-6 membered nitrogen containing heterocyclic group optionally being further selected from halogen, C 1 -C 6 One or more groups of the alkyl group are substituted;
v is an integer from 1 to 6, preferably 1 or 2.
In another particular embodiment, the compounds of the formulae (I) to (V) or of the formulae (I-1), (II-1), (III-1), (IV-1), (V-1) according to the invention or stereoisomers, tautomers, meso, racemates, enantiomers, diastereomers, or mixtures thereof, or pharmaceutically acceptable salts thereof, wherein R 2 Selected from hydrogen, halogen, C 1 -C 6 Alkyl, -NR a R b
R a 、R b Each independently selected from hydrogen, C 1 -C 6 An alkyl group.
In another specific embodiment, the compounds of formulae (I) to (V) or formulae (I-1), (II-1), (III-1), (IV-1), (V-1) according to the invention or stereoisomers, tautomers, meso, racemates, enantiomers, diastereomers, or mixtures thereof, or pharmaceutically acceptable salts thereof, wherein,
R 2 selected from C 1 -C 6 Alkyl, preferably methyl; the C is 1 -C 6 Alkyl is optionally further substituted with Q 2 Substituted with a group;
Q 2 selected from C 6 -C 10 Aryl, preferably phenyl, wherein said C 6 -C 10 Aryl is optionally further substituted by- (CH) 2 ) v -NR c R d Substitution;
R c and R is d Together with the nitrogen atom to which they are attached, form a 4-6 membered nitrogen containing heterocyclic group, said 4-6 membered nitrogen containing heterocyclic group optionally further containing one or more heteroatoms selected from N, O, S in addition to N, said 4-6 membered nitrogen containing heterocyclic group optionally being further selected from halogen, C 1 -C 6 One or more groups of the alkyl group are substituted;
v is an integer from 1 to 6, preferably 1 or 2.
In another particular embodiment, the compounds of the formulae (I) to (V) or of the formulae (I-1), (II-1), (III-1), (IV-1), (V-1) according to the invention or stereoisomers, tautomers, meso, racemates, enantiomers, diastereomers, or mixtures thereof, or pharmaceutically acceptable salts thereof, wherein R 1 Selected from hydrogen, halogen, C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy, C 1 -C 6 Haloalkyl, C 1 -C 6 Haloalkoxy groups; preferably, R 1 Is hydrogen or halogen.
In another particular embodiment, the general formulae (I) to (V) or according to the inventionA compound represented by the general formula (I-1), (II-1), (III-1), (IV-1), (V-1) or a stereoisomer, a tautomer, a meso form, a racemate, an enantiomer, a diastereomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof, wherein R 4 Selected from hydrogen, halogen, C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy, C 1 -C 6 Haloalkyl, C 1 -C 6 Haloalkoxy groups; preferably, R 4 Is hydrogen or halogen.
In another specific embodiment, the compounds of formulae (I) to (V) or formulae (I-1), (II-1), (III-1), (IV-1), (V-1) according to the invention or stereoisomers, tautomers, meso, racemates, enantiomers, diastereomers, or mixtures thereof, or pharmaceutically acceptable salts thereof, wherein,
R 5 and R is 6 Each independently selected from hydrogen and C 1 -C 12 Alkyl, said C 1 -C 12 The alkyl group is optionally further selected from deuterated, -OR a 、-SR a 、-NR a R b Is substituted with one or more groups;
R a selected from hydrogen, C 1 -C 6 An alkyl group;
R b selected from hydrogen, C 1 -C 6 Alkyl, C 3 -C 6 Cycloalkyl and 5 to 7 membered heterocyclyl;
preferably, R 5 Is hydrogen, R 6 Is C 1 -C 12 Alkyl, said C 1 -C 12 The alkyl group is optionally further substituted with one or more groups selected from deuterated, -OH.
Typical compounds of the present invention include, but are not limited to:
or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof.
The present invention further provides a process for preparing a compound of formula (III-1) according to the present invention or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, comprising the steps of:
the compound of formula A3 is reacted with boric acid or a pinacol borate compound via a metal-catalyzed cross-coupling reaction (e.g., suzuki coupling) to give a compound of formula A4; then deprotecting the residue with a suitable acid (e.g., trifluoroacetic acid) to give a compound represented by the general formula (III-1); catalysts such as Pd (PPh) 3 ) 4 、K 2 CO 3 、Cs 2 CO 3
Or,
the compound of formula A7 is subjected to metal-catalyzed cross-coupling reaction (such as Suzuki coupling) with boric acid or a pinacol borate compound to obtain a compound shown as a general formula (III-1); catalysts such as Pd (PPh) 3 ) 4 、K 2 CO 3 、Cs 2 CO 3
Wherein: r is R 1 、R 3 、R 4 、R 5 、R 6 As defined by the general formula (III-1).
The present invention further provides a process for preparing a compound of formula (IV-1) according to the present invention or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, comprising the steps of:
the compound of formula B3 is reacted with boric acid or a pinacol borate compound via a metal-catalyzed cross-coupling reaction (e.g., a Suzuki coupling) to give a compound of formula B4; then deprotecting the residue with a suitable acid (e.g., trifluoroacetic acid) to give a compound represented by the general formula (IV-1); catalysts such as Pd (PPh) 3 ) 4 、K 2 CO 3 、Cs 2 CO 3
Or,
cross-coupling reaction of a Compound of formula B7 with boric acid or a pinacol ester Compound by Metal catalysisShould (e.g., suzuki coupling) give compounds of formula (IV-1); catalysts such as Pd (PPh) 3 ) 4 、K 2 CO 3 、Cs 2 CO 3
Wherein: r is R 1 、R 2 、R 4 、R 5 、R 6 As defined by the general formula (IV-1).
The present invention further provides a process for preparing a compound of formula (V-1) according to the present invention or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, comprising the steps of:
The compound of formula C3 is reacted with boric acid or a pinacol borate compound via a metal-catalyzed cross-coupling reaction (e.g., suzuki coupling) to give a compound of formula C4; then deprotecting the residue with a suitable acid (e.g., trifluoroacetic acid) to give a compound represented by the general formula (V-1); catalysts such as Pd (PPh) 3 ) 4 、K 2 CO 3 、Cs 2 CO 3
Alternatively, the compound represented by the general formula (V-1) can be produced by the following scheme 6:
the compound of formula C7 is reacted with boric acid or a pinacol ester compound of boric acid or R 2 The Y compound is subjected to metal-catalyzed cross-coupling reaction (such as Suzuki coupling) to obtain a compound shown as a general formula (V-1); catalysts such as Pd (PPh) 3 ) 4 、K 2 CO 3 、Cs 2 CO 3
Wherein: r is R 2 、R 4 、R 5 、R 6 As defined by the general formula (V-1).
The present invention further provides a pharmaceutical composition comprising a compound of the general formula or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, according to the invention, and a pharmaceutically acceptable carrier or excipient.
The invention further relates to the use of a compound of the general formula according to the invention or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same, for the preparation of a TLR8 agonist.
The invention further relates to the use of a compound of the general formula according to the invention or a stereoisomer, tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof or a pharmaceutical composition comprising the same, for the manufacture of a medicament for the prevention or treatment of a TLR 8-related disease, which may be a viral infectious disease or a malignancy, such as viral hepatitis b, HIV virus infection, 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, ovarian cancer, bladder cancer, liver cancer, fallopian tube tumor, ovarian tumor, peritoneal tumor, melanoma, solid tumor, glioma, glioblastoma, hepatocellular carcinoma, mastoid kidney tumor, head and neck tumor, leukemia, lymphoma, myeloma and non-small cell lung cancer.
The invention further relates to a compound of the general formula according to the invention or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same, for use as a TLR8 agonist.
The invention further relates to a compound of the general formula according to the invention or a stereoisomer, tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutical composition comprising the same, for use in the prevention or treatment of a TLR 8-related disease, which may be a viral infectious disease, such as viral hepatitis b, HIV virus infection, or a malignancy, 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, ovarian cancer, bladder cancer, liver cancer, fallopian tube tumor, ovarian tumor, peritoneal tumor, melanoma, solid tumor, glioma, glioblastoma, hepatocellular carcinoma, mastoid kidney tumor, head and neck tumor, leukemia, lymphoma, myeloma and non-small cell lung cancer.
The invention further relates to a method of agonizing TLR8, comprising administering to a patient in need thereof an effective amount of a compound of the general formula according to the invention or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same.
The invention further relates to a method for preventing or treating TLR 8-related diseases comprising administering to a patient in need thereof an effective amount of a compound of formula (la) according to the invention or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same; the disease may be a viral infectious disease, such as viral hepatitis b, HIV viral infection, or a malignancy, 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, ovarian cancer, bladder cancer, liver cancer, fallopian tube tumors, ovarian tumors, peritoneal tumors, melanoma, solid tumors, glioma, neuroglioblastoma, hepatocellular carcinoma, mastoid kidney tumor, head and neck tumors, leukemia, lymphoma, myeloma, and non-small cell lung cancer.
The compounds of the present invention may form pharmaceutically acceptable base addition salts or acid addition salts with bases or acids according to methods conventional in the art to which the present invention pertains. The base includes inorganic bases and organic bases, acceptable organic bases include diethanolamine, ethanolamine, N-methylglucamine, triethanolamine, tromethamine, and the like, and acceptable inorganic bases include aluminum hydroxide, calcium hydroxide, potassium hydroxide, sodium carbonate, sodium hydroxide, and the like. The acids include inorganic acids and organic acids, and acceptable inorganic acids include hydrochloric acid, sulfuric acid, nitric acid, phosphoric acid, hydrobromic acid, and the like. Acceptable organic acids include acetic acid, trifluoroacetic acid, formic acid, anti-cyclic acid, and the like.
Pharmaceutical compositions containing the active ingredient may be in a form suitable for oral administration, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs. Oral compositions may be prepared according to any method known in the art for preparing pharmaceutical compositions, and such compositions may contain one or more ingredients selected from the group consisting of: sweeteners, flavoring agents, coloring agents and preservatives to provide a pleasing and palatable pharmaceutical preparation. Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets. These excipients may be inert excipients, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example 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 they may be coated by known techniques to mask the taste of the drug or delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, water-soluble taste masking substances such as hydroxypropyl methylcellulose or hydroxypropyl cellulose, or extended time substances such as ethylcellulose, cellulose acetate butyrate may be used.
Oral formulations may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with a water-soluble carrier, for example polyethylene glycol or an oil vehicle, for example peanut oil, liquid paraffin or olive oil.
Aqueous suspensions contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions. Such excipients are suspending agents, for example sodium carboxymethyl cellulose, methyl cellulose, hydroxypropyl methyl cellulose, sodium alginate, polyvinylpyrrolidone and acacia; the dispersing or wetting agent may be a naturally occurring phospholipid such as lecithin, or a condensation product of an alkylene oxide with a fatty acid such as polyoxyethylene stearate, or a condensation product of ethylene oxide with a long chain fatty alcohol such as heptadecaethyleneoxy cetyl alcohol, or a condensation product of ethylene oxide with a partial ester derived from a fatty acid and a hexitol such as polyethylene oxide sorbitol monooleate, or a condensation product of ethylene oxide with a partial ester derived from a fatty acid and a hexitol anhydride such as polyethylene oxide sorbitan monooleate. The aqueous suspension may also contain one or more preservatives such as ethyl or Jin Zhengbing esters of nipagin, one or more coloring agents, one or more flavoring agents and one or more sweetening agents, such as sucrose, saccharin or aspartame.
Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin. The oil suspension may contain a thickening agent, such as beeswax, hard paraffin or cetyl alcohol. The above-described sweeteners and flavoring agents 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 use in the preparation of an aqueous suspension by the addition of water provide the active ingredient in combination with a dispersing or wetting agent, suspending agent or one or more preservatives. Suitable dispersing or wetting agents and suspending agents are as described above. Other excipients, for example sweetening, flavoring and coloring agents, may also be added. These compositions are preserved by the addition of an antioxidant such as ascorbic acid.
The pharmaceutical compositions of the present invention may also be in the form of an oil-in-water emulsion. The oily phase may be a vegetable oil, for example olive oil or arachis oil, or a mineral oil, for example liquid paraffin or mixtures thereof. Suitable emulsifiers may be naturally occurring phospholipids, such as soy lecithin, and esters or partial esters derived from fatty acids and hexitol anhydrides, such as sorbitan monooleate, and condensation products of the partial esters and ethylene oxide, such as polyethylene oxide sorbitol 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, a colorant and an antioxidant.
The pharmaceutical compositions of the present invention may be in the form of sterile injectable aqueous solutions. Acceptable vehicles and solvents that may be used 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 an oil phase. For example, the active ingredient is dissolved in a mixture of soybean oil and lecithin. The oil solution is then treated to form a microemulsion by adding it to a mixture of water and glycerol. The injection or microemulsion may be injected into the patient's blood stream by local bolus injection. Alternatively, it may be desirable to administer the solutions and microemulsions in a manner that maintains a constant circulating concentration of the compounds of the present invention. To maintain this constant concentration, a continuous intravenous delivery device may be used.
The pharmaceutical compositions of the present invention may be in the form of sterile injectable aqueous or oleaginous suspensions for intramuscular and subcutaneous administration. The suspensions may be formulated according to known techniques using those suitable dispersing or wetting agents and suspending agents as described above. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example as a solution in 1, 3-butanediol. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any blend stock oil may be used, including synthetic mono-or diglycerides. In addition, fatty acids such as oleic acid may be used in the preparation of injectables.
The compounds of the present 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. Such materials include cocoa butter, glycerogelatin, hydrogenated vegetable oils, polyethylene glycols of various molecular weights and mixtures of fatty acid esters of polyethylene glycols.
It is well known to those skilled in the art that the amount of drug administered depends on a variety of factors, including but not limited to the following: the activity of the particular compound used, the age of the patient, the weight of the patient, the health of the patient, the patient's integument, the patient's diet, the time of administration, the mode of administration, the rate of excretion, the combination of the drugs, etc. In addition, the optimal mode of treatment, such as the mode of treatment, the daily amount of the compound of formula (I) or the type of pharmaceutically acceptable salt, can be verified according to conventional treatment protocols.
The invention can contain the compound of the general formula and pharmaceutically acceptable salt, hydrate or solvate thereof as active ingredients, and is mixed with pharmaceutically acceptable carriers or excipients to prepare a composition and a clinically acceptable dosage form. The derivatives of the present invention may be used in combination with other active ingredients as long as they do not exert other adverse effects such as allergic reactions and the like. The compounds of the present invention may be used as the sole active ingredient, or in combination with other drugs for the treatment of diseases associated with tyrosine kinase activity. Combination therapy is achieved by simultaneous, separate or sequential administration of the individual therapeutic components.
Description of the terms
Unless stated to the contrary, the terms used in the specification and claims have the following meanings.
The term "alkyl" refers to a saturated aliphatic hydrocarbon group which is a straight or branched chain group containing from 1 to 20 carbon atoms, preferably an alkyl group containing from 1 to 12 carbon atoms, more preferably an alkyl group containing from 1 to 6 carbon atoms. Non-limiting examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1-dimethylpropyl, 1, 2-dimethylpropyl, 2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2-methylpropyl, 1, 2-trimethylpropyl, 1-dimethylbutyl, 1, 2-dimethylbutyl, 2-dimethylbutyl, 1, 3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2, 3-dimethylbutyl, n-heptyl, 2-methylhexyl, 3-methylhexyl, 4-methylhexyl 5-methylhexyl, 2, 3-dimethylpentyl, 2, 4-dimethylpentyl, 2-dimethylpentyl, 3-dimethylpentyl, 2-ethylpentyl, 3-ethylpentyl, n-octyl, 2, 3-dimethylhexyl, 2, 4-dimethylhexyl, 2, 5-dimethylhexyl, 2-dimethylhexyl, 3-dimethylhexyl 4, 4-dimethylhexyl, 2-ethylhexyl, 3-ethylhexyl, 4-ethylhexyl, 2-methyl-2-ethylpentyl, 2-methyl-3-ethylpentyl, n-nonyl, 2-methyl-2-ethylhexyl, 2-methyl-3-ethylhexyl, 2-diethylpentyl, n-decyl, 3-diethylhexyl, 2-diethylhexyl, and various branched isomers thereof. More preferred are lower alkyl groups containing 1 to 6 carbon atoms, and non-limiting examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1-dimethylpropyl, 1, 2-dimethylpropyl, 2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2-methylpropyl, 1, 2-trimethylpropyl, 1-dimethylbutyl, 1, 2-dimethylbutyl, 2-dimethylbutyl, 1, 3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2, 3-dimethylbutyl, and the like. The alkyl group may be substituted or unsubstituted, and when substituted, the substituent may be substituted at any available point of attachment, preferably one or more groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, oxo, carboxyl or carboxylate.
The term "alkenyl" refers to an alkyl group as defined above consisting of at least two carbon atoms and at least one carbon-carbon double bond, such as vinyl, 1-propenyl, 2-propenyl, 1-, 2-, or 3-butenyl, and the like. Alkenyl groups may be substituted or unsubstituted, and when substituted, the substituents are preferably one or more groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio.
The term "alkynyl" refers to an alkyl group as defined above consisting of at least two carbon atoms and at least one carbon-carbon triple bond, such as ethynyl, propynyl, butynyl, and the like. Alkynyl groups may be substituted or unsubstituted, and when substituted, the substituents are preferably one or more groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio.
The term "cycloalkyl" refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent, the cycloalkyl ring containing from 3 to 20 carbon atoms, preferably from 3 to 12 carbon atoms, more preferably from 3 to 10 carbon atoms, even more preferably from 3 to 8 carbon atoms, and most preferably from 3 to 6 carbon atoms. Non-limiting examples of monocyclic cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cycloheptatrienyl, cyclooctyl, and the like; polycyclic cycloalkyl groups include spiro, fused and bridged cycloalkyl groups.
The term "spirocycloalkyl" refers to a polycyclic group sharing one carbon atom (referred to as a spiro atom) between 5-to 20-membered monocyclic rings, which may contain one or more double bonds, but no ring has a fully conjugated pi-electron system. Preferably 6 to 14 membered, more preferably 7 to 10 membered. The spirocycloalkyl group is classified into a single spirocycloalkyl group, a double spirocycloalkyl group or a multiple spirocycloalkyl group according to the number of common spiro atoms between rings, and preferably a single spirocycloalkyl group and a double spirocycloalkyl group. More preferably 4-membered/4-membered, 4-membered/5-membered, 4-membered/6-membered, 5-membered/5-membered or 5-membered/6-membered monocyclocycloalkyl. Non-limiting examples of spirocycloalkyl groups include:
The term "fused ring alkyl" refers to a 5 to 20 membered, all carbon polycyclic group wherein each ring in the system shares an adjacent pair of carbon atoms with the other rings in the system, wherein one or more of the rings may contain one or more double bonds, but none of the rings has a fully conjugated pi electron system. Preferably 6 to 14 membered, more preferably 7 to 10 membered. The number of constituent rings may be classified as a bicyclic, tricyclic, tetracyclic or polycyclic fused ring alkyl group, preferably a bicyclic or tricyclic, more preferably a 5-membered/5-membered or 5-membered/6-membered bicycloalkyl group. Non-limiting examples of fused ring alkyl groups include:
the term "bridged cycloalkyl" refers to an all-carbon polycyclic group of 5 to 20 members, any two rings sharing two carbon atoms not directly attached, which may contain one or more double bonds, but no ring has a fully conjugated pi-electron system. Preferably 6 to 14 membered, more preferably 7 to 10 membered. Cycloalkyl groups which may be classified as bicyclic, tricyclic, tetracyclic or polycyclic bridged according to the number of constituent rings are preferably bicyclic, tricyclic or tetracyclic, more preferably bicyclic or tricyclic. Non-limiting examples of bridged cycloalkyl groups include:
the cycloalkyl ring may be fused to an aryl, heteroaryl, or heterocycloalkyl ring, where the ring attached to the parent structure is cycloalkyl, non-limiting examples include indanyl, tetrahydronaphthyl, benzocycloheptyl, and the like. Cycloalkyl groups may be optionally substituted or unsubstituted, and when substituted, the substituents are preferably one or more groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, oxo, carboxyl, or carboxylate groups.
The term "heterocyclyl" refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent containing from 3 to 20 ring atoms in which one or more ring atoms are selected from nitrogen, oxygen or S (O) m (wherein m is an integer from 0 to 2), but does not include a ring moiety of-O-O-, -O-S-, or-S-S-, and the remaining ring atoms are carbon. Preferably containing 3 to 12 ring atoms, of which 1 to 4 are heteroatoms; most preferably from 3 to 8 ring atoms, of which 1 to 3 are heteroatoms; most preferably from 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, tetrahydrothienyl, dihydroimidazolyl, dihydrofuranyl, dihydropyrazolyl, dihydropyrrolyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, homopiperazinyl, pyranyl, and the like, preferably 1, 2, 5-oxadiazolyl, pyranyl, or morpholinyl. Polycyclic heterocyclyl groups include spiro, fused and bridged heterocyclic groups.
The term "spiroheterocyclyl" refers to a polycyclic heterocyclic group having a single ring of 5 to 20 members sharing one atom (referred to as the spiro atom) between them, wherein one or more of the ring atoms is selected from nitrogen, oxygen or S (O) m (wherein m is an integer from 0 to 2) and the remaining ring atoms are carbon. Which may contain one or more double bonds, but none of the rings has a fully conjugated pi-electron system. Preferably 6 to 14 membered, more preferably 7 to 10 membered. The spiroheterocyclyl groups are classified into a single spiroheterocyclyl group, a double spiroheterocyclyl group or a multiple spiroheterocyclyl group according to the number of common spiro atoms between rings, and preferably a single spiroheterocyclyl group and a double spiroheterocyclyl group. More preferably 4/4, 4/5, 4/6, 5/5 or 5/6 units A spiroheterocyclyl group. Non-limiting examples of spiroheterocyclyl groups include:
the term "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 the other rings in the system, one or more of which may contain one or more double bonds, but none of which has a fully conjugated pi electron system in which one or more ring atoms are selected from nitrogen, oxygen or S (O) m (wherein m is an integer from 0 to 2) and the remaining ring atoms are carbon. Preferably 6 to 14 membered, more preferably 7 to 10 membered. The number of constituent rings may be classified as a bicyclic, tricyclic, tetracyclic or polycyclic fused heterocyclic group, preferably a bicyclic or tricyclic, more preferably a 5-membered/5-membered or 5-membered/6-membered bicyclic fused heterocyclic group. Non-limiting examples of fused heterocyclyl groups include:
the term "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 fully conjugated pi electron system in which one or more ring atoms are selected from nitrogen, oxygen, or S (O) m (wherein m is an integer from 0 to 2) and the remaining ring atoms are carbon. Preferably 6 to 14 membered, more preferably 7 to 10 membered. Heterocyclic groups which may be classified as bicyclic, tricyclic, tetracyclic or polycyclic bridged according to the number of constituent rings are preferably bicyclic, tricyclic or tetracyclic, more preferably bicyclic or tricyclic. Non-limiting examples of bridged heterocyclyl groups include:
The heterocyclyl ring may be fused to an aryl, heteroaryl or cycloalkyl ring, wherein the ring attached to the parent structure is heterocyclyl, non-limiting examples of which include:
etc.
The heterocyclic group may be optionally substituted or unsubstituted, and when substituted, the substituent is preferably one or more groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, oxo, carboxyl, or carboxylate groups.
The term "aryl" refers to a 6 to 14 membered all-carbon monocyclic or fused polycyclic (i.e., rings sharing adjacent pairs of carbon atoms) group having a conjugated pi-electron system, preferably 6 to 10 membered, such as phenyl and naphthyl. More preferably phenyl. The aryl ring may be fused to a heteroaryl, heterocyclyl or cycloalkyl ring, wherein the ring attached 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 groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, carboxyl, or carboxylate groups.
The term "heteroaryl" refers to a heteroaromatic system containing from 1 to 4 heteroatoms, from 5 to 14 ring atoms, wherein the heteroatoms are selected from oxygen, sulfur and nitrogen. Heteroaryl groups are preferably 5 to 10 membered, containing 1 to 3 heteroatoms; more preferably 5 or 6 membered, containing 1 to 2 heteroatoms; preferably, for example, imidazolyl, furyl, thienyl, thiazolyl, pyrazolyl, oxazolyl, pyrrolyl, tetrazolyl, pyridyl, pyrimidinyl, thiadiazole, pyrazinyl, and the like, preferably imidazolyl, thiazolyl, pyrazolyl or pyrimidinyl, thiazolyl; more preferably pyrazolyl or thiazolyl. The heteroaryl ring may be fused to an aryl, heterocyclyl, or cycloalkyl ring, wherein the ring attached to the parent structure is a heteroaryl ring, non-limiting examples of which include:
heteroaryl groups may be optionally substituted or unsubstituted, and when substituted, the substituents are preferably one or more groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, carboxyl, or carboxylate groups.
The term "alkoxy" refers to-O- (alkyl) and-O- (unsubstituted cycloalkyl), wherein alkyl is as defined above. Non-limiting examples of alkoxy groups include: methoxy, ethoxy, propoxy, butoxy, cyclopropoxy, cyclobutoxy, cyclopentoxy, cyclohexyloxy. The alkoxy groups may be optionally substituted or unsubstituted, and when substituted, the substituents are preferably one or more groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, carboxyl, or carboxylate groups.
The term "haloalkyl" refers to an alkyl group substituted with one or more halogens, wherein alkyl is as defined above.
The term "haloalkoxy" refers to an alkoxy group substituted with one or more halogens, wherein the alkoxy group is as defined above.
The term "hydroxyalkyl" refers to an alkyl group substituted with one or more hydroxyl groups, wherein alkyl is as defined above.
The term "hydroxy" refers to an-OH group.
The term "halogen" refers to fluorine, chlorine, bromine or iodine.
The term "amino" refers to-NH 2
The term "cyano" refers to-CN.
The term "nitro" refers to-NO 2
The term "oxo" refers to = O.
The term "carboxy" refers to-C (O) OH.
The term "mercapto" refers to-SH.
The term "ester group" refers to a-C (O) O (alkyl) or-C (O) O (cycloalkyl), wherein alkyl and cycloalkyl are as defined above.
The term "acyl" refers to compounds containing a-C (O) R group, wherein R is alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl.
"optional" or "optionally" means that the subsequently described event or circumstance may but need not occur, and that the description includes instances where the event or circumstance occurs or does not. For example, "a heterocyclic group optionally substituted with an alkyl group" means that an alkyl group may be, but is not necessarily, present, and the description includes cases where the heterocyclic group is substituted with an alkyl group and cases where the heterocyclic group is not substituted with an alkyl group.
"substituted" means that one or more hydrogen atoms, preferably up to 5, more preferably 1 to 3 hydrogen atoms in the group are independently substituted with a corresponding number of substituents. It goes without saying that substituents are only in their possible chemical positions, and that the person skilled in the art is able to determine (by experiment or theory) possible or impossible substitutions without undue effort. For example, amino or hydroxyl groups having free hydrogen may be unstable when bound to carbon atoms having unsaturated (e.g., olefinic) bonds.
"pharmaceutical composition" means a mixture comprising one or more of the compounds described herein or a physiologically/pharmaceutically acceptable salt or prodrug thereof, and other chemical components, such as physiologically/pharmaceutically acceptable carriers and excipients. The purpose of the pharmaceutical composition is to promote the administration to organisms, facilitate the absorption of active ingredients and thus exert biological activity.
By "pharmaceutically acceptable salts" is meant salts of the compounds of the present invention which are safe and effective when used in a mammal, and which possess the desired biological activity.
Synthesis method of compound of the invention
To accomplish the objects of the present invention, the present invention employs the following synthetic schemes for preparing the compounds of the general formula (I) of the present invention.
When the compound represented by the general formula (I) is a compound represented by the general formula (III-1), the compound represented by the general formula (III-1) is prepared by the following scheme 1:
scheme 1
The compound of formula A1 is reacted with a nucleophilic amine in the presence of a suitable base (e.g., DIEA) at room temperature to give a compound of formula A2; then, under the heating condition, reacting the compound of the formula A2 with 2, 4-dimethoxy benzylamine to obtain a compound of the formula A3; the compounds of formula A3 may be prepared by metal catalysis (e.g. Pd (PPh 3 ) 4 、K 2 CO 3 、Cs 2 CO 3 ) Is reacted with boric acid or a pinacol ester compound to give a compound of formula A4; deprotection with a suitable acid (e.g., trifluoroacetic acid) to provide a compound of formula (III-1);
wherein: r is R 1 、R 3 、R 4 、R 5 、R 6 As defined by the general formula (III-1).
Alternatively, the compound represented by the general formula (III-1) can be prepared by the following scheme 2:
scheme 2
Reacting a compound of formula A6 with a nucleophilic amine in the presence of a condensing agent (e.g., BOP) to give a coupling product, a compound of formula A7; the compounds of formula A7 may be prepared by metal catalysis (e.g. Pd (PPh 3 ) 4 、K 2 CO 3 、Cs 2 CO 3 ) The cross-coupling reaction (e.g., suzuki coupling) with boric acid or a pinacol ester compound of boric acid to give a compound represented by the general formula (III-1);
wherein: r is R 1 、R 3 、R 4 、R 5 、R 6 As defined by the general formula (III-1).
When the compound represented by the general formula (I) is a compound represented by the general formula (IV-1), the compound represented by the general formula (IV-1) is prepared by the following scheme 3:
scheme 3
The compound of formula B1 is reacted with a nucleophilic amine in the presence of a suitable base (e.g., DIEA) at room temperature to give the compound of formula B2; then, under the heating condition, reacting the compound of the formula B2 with 2, 4-dimethoxy benzylamine to obtain a compound of the formula B3; the compounds of formula B3 may be prepared by metal catalysis (e.g. Pd (PPh 3 ) 4 、K 2 CO 3 、Cs 2 CO 3 ) Is reacted with boric acid or a pinacol ester compound to give a compound of formula B4; deprotection with a suitable acid (e.g., trifluoroacetic acid) to provide a compound of formula (IV-1);
wherein: r is R 1 、R 2 、R 4 、R 5 、R 6 As defined by the general formula (IV-1).
Alternatively, the compound represented by the general formula (IV-1) can be prepared by the following scheme 4:
scheme 4
Reacting a compound of formula B6 with a nucleophilic amine in the presence of a condensing agent (e.g., BOP) to give a coupling product, a compound of formula B7; the compounds of formula B7 may be prepared by metal catalysis (e.g. Pd (PPh 3 ) 4 、K 2 CO 3 、Cs 2 CO 3 ) The cross-coupling reaction (e.g., suzuki coupling) with boric acid or a pinacol ester compound of boric acid to give a compound represented by the general formula (IV-1);
wherein: r is R 1 、R 2 、R 4 、R 5 、R 6 As defined by the general formula (IV-1).
When the compound represented by the general formula (I) is a compound represented by the general formula (V-1), the compound represented by the general formula (V-1) is prepared by the following scheme 5:
scheme 5
The compound of formula C1 is reacted with a nucleophilic amine in the presence of a suitable base (e.g., DIEA) at room temperature to give a compound of formula C2; then, under the heating condition, reacting the compound of the formula C2 with 2, 4-dimethoxy benzylamine to obtain a compound of the formula C3; the compounds of formula C3 may be prepared by metal catalysis (e.g. Pd (PPh 3 ) 4 、K 2 CO 3 、Cs 2 CO 3 ) Is reacted with boric acid or a pinacol ester compound to give a compound of formula C4; deprotection with a suitable acid (e.g., trifluoroacetic acid) to provide a compound of formula (V-1);
wherein: r is R 2 、R 4 、R 5 、R 6 As defined by the general formula (V-1).
Alternatively, the compound represented by the general formula (V-1) can be produced by the following scheme 6:
scheme 6
Reacting a compound of formula C6 with a nucleophilic amine in the presence of a condensing agent (e.g., BOP) to give a coupling product compound of formula C7; the compounds of formula C7 may be prepared by metal catalysis (e.g. Pd (PPh 3 ) 4 、K 2 CO 3 、Cs 2 CO 3 ) The cross-coupling reaction (e.g., suzuki coupling) with boric acid or a pinacol ester compound of boric acid to give a compound represented by the general formula (V-1);
wherein: r is R 2 、R 4 、R 5 、R 6 As defined by the general formula (V-1).
Drawings
FIG. 1 shows parameters of the compound of example 52 in cynomolgus monkey serum.
Detailed Description
The compounds of the present invention and their preparation are further understood by the examples which illustrate some methods of making or using the compounds. However, it is to be understood that these examples do not limit the present invention. Variations of the invention now known or further developed are considered to fall within the scope of the invention described and claimed herein.
The compounds of the present invention are prepared using convenient starting materials and general preparation procedures. Typical or preferential reaction conditions are given in the present invention, such as reaction temperature, time, solvent, pressure, molar ratio of reactants. But other reaction conditions can be adopted unless specifically stated. The optimization conditions may vary with the particular reactants or solvents used, but in general, both the reaction optimization steps and conditions can be determined.
In addition, some protecting groups may be used in the present invention to protect certain functional groups from unwanted reactions. Protecting groups suitable for various functional groups and their protecting or deprotecting conditions are well known to those skilled in the art. For example, T.W.Greene and G.M.Wuts in organic preparation of protecting groups (3 rd edition, wiley, new York,1999 and literature citations) describe in detail the protection or deprotection of a large number of protecting groups.
The separation and purification of the compounds and intermediates may be carried out by any suitable method or procedure depending on the particular needs, such as filtration, extraction, distillation, crystallization, column chromatography, thin layer chromatography, high performance liquid chromatography or a combination thereof. The specific methods of use thereof may be found in the examples described herein. Of course, other similar isolation and purification means may be employed. It can be characterized using conventional methods, including physical constants and spectral data.
The structure of the compounds is determined by Nuclear Magnetic Resonance (NMR) or/and Mass Spectrometry (MS). NMR shift at 10 -6 Units of (ppm) are given. NMR was performed using Bruker dps 300 nuclear magnetic resonance apparatus with deuterated dimethyl sulfoxide (DMSO-d) 6 ) Deuterated chloroform (CDCl) 3 ) Deuterated methanol (CD) 3 OD), internal standard Tetramethylsilane (TMS).
The MS was determined by LC (Waters 2695)/MS (Quattro Premier x E) mass spectrometer (manufacturer: watt) (Photodiode Array Detector).
The preparation liquid chromatography used an lc6000 high performance liquid chromatograph (manufacturer: innovative). The column was Daisogel C18 μm 100A (30 mm. Times.250 mm), mobile phase: acetonitrile/water.
The Thin Layer Chromatography (TLC) uses Qingdao ocean chemical GF254 silica gel plate, the specification of the silica gel plate used for reaction monitoring is 0.20 mm-0.25 mm, and the specification of the silica gel plate used for preparing the thin layer chromatography is 0.5mm.
The silica gel column chromatography uses Qingdao ocean silica gel 100-200 mesh, 200-300 mesh and 300-400 mesh silica gel as carrier.
The known starting materials of the present invention may be synthesized using or according to methods known in the art or may be purchased from commercial establishments, beijing couplings, sigma, carbofuran, yi Shiming, shanghai Shuya, shanghai Enoki, an Naiji chemistry, shanghai Pico, and the like.
The examples are not particularly described, and the reactions can all be carried out under nitrogen atmosphere.
An argon or nitrogen atmosphere means that the reactor flask is connected to a balloon of argon or nitrogen of about 1L volume.
The reaction solvent, the organic solvent or the inert solvent are each expressed as a solvent which does not participate in the reaction under the reaction conditions described, and include, for example, benzene, toluene, acetonitrile, tetrahydrofuran (THF), dimethylformamide (DMF), chloroform, methylene chloride, diethyl ether, methanol, nitrogen-methylpyrrolidone (NMP), pyridine, etc. The examples are not specifically described, and the solution refers to an aqueous solution.
The chemical reactions described in the present invention are generally carried out at atmospheric pressure. The reaction temperature is between-78 ℃ and 200 ℃. The reaction time and conditions are, for example, between-78 ℃ and 200 ℃ at one atmosphere, completed in about 1 to 24 hours. If the reaction is overnight, the reaction time is typically 16 hours. The reaction temperature is room temperature and is 20-30 deg.c without specific explanation in the examples.
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art. In addition, any methods and materials similar or equivalent to those described herein can be used in the methods of the present invention.
Example 1: preparation of (R) -2- ((2-aminopyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (1)
Step 1: preparation of 1- ((tert-butyldimethylsilyl) oxy) propan-2-one (1 b)
Hydroxyacetone 1a (100 g,1.35 mol) was dissolved in Dichloromethane (DCM) (1L) at room temperature. The solution was cooled to 0deg.C and imidazole (175 g,2.57 mol) and tert-butyldimethylchlorosilane (TBDMSCl) (245 g,1.63 mol) were added sequentially. The reaction solution was stirred at 0℃for 1 hour, then slowly warmed to room temperature, and stirring was continued for 12 hours. After completion of the reaction, the reaction mixture was washed with water (3X 1L), and the organic phase was concentrated under reduced pressure to give Compound 1b (200 g, 78.7%) as a pale yellow liquid.
1 H-NMR(CDCl 3 )δ:4.15(s,2H),2.17(s,3H),0.93(s,9H),0.09(s,6H)。
Step 2: preparation of (S, E) -N- (1- ((tert-butyldimethylsilyl) oxy) propan-2-ylidene) -2-methylpropane-2-sulfonamide (1 c)
Compound 1b (200 g,1.06 mol) and S-tert-butylsulfinamide (129 g,1.06 mol) were dissolved in tetrahydrofuran (3.6L) at room temperature. The reaction was added dropwise to tetraisopropyl orthotitanate (800 mL,2.66 mol). The reaction solution was stirred at 70℃for 12 hours. After the reaction was completed, it was cooled to room temperature. The reaction solution was concentrated under reduced pressure, the resulting brown liquid was added to ice water (1L), the resulting solid was removed by filtration, ethyl acetate (3X 500 mL) was added for extraction, the combined organic phases were washed with saturated brine (500 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude brown oil was purified by silica gel column chromatography (mobile phase: ethyl acetate/petroleum ether=3% -20%) to give compound 1c (50 g, 16.1%) as a tan liquid.
1 H NMR (300 MHz, chloroform-d) delta 4.23 (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] +
Step 3: preparation of (S) -N- ((R) -1- ((tert-butyldimethylsilyl) oxy) -2-methylhex-2-yl) -2-methylpropane-2-sulfonamide (1 d)
Compound 1c (50 g,0.171 mol) was dissolved in toluene (500 mL) at room temperature under nitrogen. The reaction mixture was added dropwise to a heptanes solution of trimethylaluminum (207 mL,1mol/L,0.21 mol) at-78℃and, after completion of the addition, stirring was continued for 0.5 hours. Then, n-hexane solution (102 mL,2.5mol/L,0.26 mol) of n-butylaluminum was added dropwise thereto at-78℃and, after completion of the addition, the mixture was stirred at-78℃for 4 hours. After completion of the reaction, water (500 mL) was added thereto, followed by quenching, filtration, extraction with ethyl acetate (3X 200 mL) was performed, and the combined organic phases were washed with saturated brine (300 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The resulting yellow crude product was purified by silica gel column chromatography (mobile phase: ethyl acetate/petroleum ether=7% -10%) to give compound 1d (26 g, 43.4%) as a yellow liquid.
1 H NMR (400 MHz, chloroform-d) delta 3.66 (s, 1H), 3.51 (d, j=9.4 hz, 1H), 3.32 (d, j=9.4 hz, 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).
LC-MS:m/z 350.3[M+H] +
Step 4: preparation of (R) -1- ((tert-butyldimethylsilyl) oxy) -2-methylhex-2-amine (1 e)
Compound 1d (26 g,0.074 mol) was dissolved in tetrahydrofuran (250 mL) and water (50 mL) at room temperature, and elemental iodine (3.78 g,0.015 mol) was added to the reaction mixture, which was stirred at 50℃overnight. After the completion of the reaction, water (200 mL) was added to dilute the reaction solution, tetrahydrofuran was removed under reduced pressure, ethyl acetate (4X 200 mL) was added to extract, and the combined organic phases were washed successively with a sodium thiosulfate solution (400 mL) and a saturated saline solution (400 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give compound 1e (15 g, 82.1%) as a brown oily liquid, which was used in the next step without purification.
1 H NMR (400 MHz, chloroform-d) delta 3.30 (q, j=9.4 hz, 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] +
Step 5: preparation of 2, 4-dichloropyrido [4,3-d ] pyrimidine (1 g)
Pyrido [4,3-d ] pyrimidine-2, 4 (1H, 3H) -dione 1f (50 mg,0.31 mmol) was dissolved in phosphorus oxychloride (1.5 ml) at room temperature, then N, N-Diisopropylethylamine (DIEA) (356 mg,2.75 mmol) was slowly added, the reaction was warmed to 100℃and stirred for 4 hours. After completion of the reaction, the reaction mixture was cooled to room temperature, diluted with ice water (30 mL), extracted rapidly with ethyl acetate (3X 8 mL), and the organic phases were combined, washed with saturated brine (30 mL), dried over anhydrous sodium sulfate, and the resulting ethyl acetate solution of 1g of the compound was directly used for the next reaction.
LC-MS:m/z 200.0[M+H] +
Step 6: preparation of (R) -N- ((1- ((tert-butyldimethylsilyl) oxy) -2-methylhex-2-yl) -2-chloropyrido [4,3-d ] pyrimidin-4-amine (1 h)
To the above 1g of ethyl acetate solution were added (R) -1- ((tert-butyldimethylsilyl) oxy) -2-methylhex-2-amine 1e (90.4 mg,0.37 mmol) and N, N-diisopropylethylamine (198mg, 1.53 mmol) at room temperature. The reaction solution was stirred at room temperature for 4 hours. After completion of the reaction, the reaction mixture was diluted with water (30 mL), extracted with ethyl acetate (3X 30 mL), and the organic phases were combined, washed with saturated brine (30 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product obtained was purified by column chromatography on silica gel (petroleum ether: ethyl acetate=1:1). The obtained product was concentrated under reduced pressure to obtain a yellow oily compound (16 mg, 12.7%) for 1 h.
LC-MS:m/z 409.2[M+H] +
Step 7: (R) -N 4 - (1- ((tert-butyldimethylsilyl) oxy) -2-methylhex-2-yl) -N 2 - (2, 4-dimethoxybenzyl) pyrido [4,3-d ]]Preparation of pyrimidine-2, 4-diamine (1 i)
Compound 1h (16 mg,0.039 mmol) was reacted with 2, 4-dimethoxybenzylamine (DMB-NH) at room temperature 2 ) (45.7 mg,0.274 mmol) in dioxane (1 mL) and then N, N-diisopropylethylamine (15 mg,0.117 mmol) was added to the system. The reaction solution was stirred at 100℃for 4 hours. After the completion of the reaction, the reaction was cooled to room temperature, diluted with water (30 mL), extracted with ethyl acetate (3X 30 mL), The organic phases were combined, washed with saturated brine (30 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product obtained was purified by column chromatography on silica gel (petroleum ether: ethyl acetate=1:1). The obtained product was concentrated under reduced pressure to obtain compound 1i (14 mg, 66.7%) as a yellow oil.
LC-MS:m/z 540.3[M+H] +
Step 8: preparation of (R) -2- ((2-aminopyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (1)
Compound 1i (14 mg,0.027 mmol) was dissolved in trifluoroacetic acid (TFA) (1 mL) at room temperature and stirred overnight at 40 ℃. After completion of the reaction, the reaction mixture was concentrated under reduced pressure, diluted with saturated sodium hydrogencarbonate solution (20 mL), extracted with ethyl acetate (3X 30 mL), and the organic phases were combined, washed with saturated brine (20 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product was purified by preparative chromatography (column: XBridge Shield RP OBD column, 5um,19 x 150mm; mobile phase A: water (10 mmol/L ammonium bicarbonate), mobile phase B: acetonitrile; flow rate: 25mL/min; gradient: 18% -35% acetonitrile in 8 min; detection wavelength: 220 nm) to give compound 1 (3.4 mg, 44%) as a white solid.
1 H NMR (400 MHz, methanol-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] +
Example 2: preparation of (R) -2- ((2-amino-7- (2- (4-methylpiperazin-1-yl) pyrimidin-5-yl) pyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (2)
Step 1: preparation of 4-amino-6-chloronicotinamide (2 b)
4-amino-6-chloronicotinic acid 2a (1.00 g,5.795 mmol) was dissolved in 1, 4-dioxane (15 mL) at room temperature, and thionyl chloride (8 mL) was slowly added dropwise thereto, and the reaction mixture was stirred at 90℃for 2 hours. The reaction was concentrated under reduced pressure, and the residue was diluted with Tetrahydrofuran (THF) (5 mL). Ammonia water (10 mL) was slowly added dropwise to the reaction solution at room temperature, and stirring was continued for 1 hour. The resulting mixture was concentrated under reduced pressure to give compound 2b (961 mg, 96.65%) as a yellow solid.
LC-MS:m/z 172.0[M+H] +
Step 2: preparation of 7-chloropyrido [4,3-d ] pyrimidine-2, 4 (1H, 3H) -dione (2 c)
Compound 2b (961 mg,5.60 mmol) was dissolved in N, N-Dimethylformamide (DMF) (10 mL), N-Carbonyldiimidazole (CDI) (3.63 g,22.39 mmol) and 1, 8-diazabicyclo undec-7-ene (DBU) (2.13 g,13.99 mmol) were sequentially added, and the reaction was stirred at 80℃for 1 hour. After the completion of the reaction, the reaction mixture was quenched with water (30 mL), the pH of the reaction mixture was adjusted to 4 with dilute hydrochloric acid, ethyl acetate (3X 40 mL) was added, the combined organic phases were washed with saturated brine (100 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give compound 2c (910 mg, 82.23%) as a dark yellow solid.
LC-MS:m/z 198.0[M+H] +
Step 3: preparation of 2,4, 7-trichloropyrido [4,3-d ] pyrimidine (2 d)
Compound 2c (457 mg,2.31 mmol) was mixed with phosphorus oxychloride (5 mL) at 0deg.C, and N, N-diisopropylethylamine (1.49 g,11.53 mmol) was slowly added dropwise. The reaction mixture was stirred at 100℃for 2 hours. After the completion of the reaction, the reaction solution was cooled to room temperature and concentrated under reduced pressure to give crude compound 2d (540 mg, crude) as a black oil, which was used in the next step without purification.
LC-MS:m/z 233.9[M+H] +
Step 4: preparation of (R) -N- ((1- ((tert-butyldimethylsilyl) oxy) -2-methylhex-2-yl) -2, 7-dichloropyrido [4,3-d ] pyrimidin-4-amine (2 e)
Compound 2d (540 mg,2.30 mmol) was dissolved in 1, 4-dioxane (5 mL) and (R) -1- ((tert-butyldimethylsilyl) oxy) -2-methylhex-2-amine 1e (1.13 g,4.61 mmol) and N, N-diisopropylethylamine (2.98 g,23.03 mmol) were added sequentially and reacted at room temperature for 1.5 hours. After completion of the reaction, the reaction mixture was diluted with water (30 mL), extracted with ethyl acetate (3×30 mL), and the combined organic phases were washed with saturated brine (100 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the obtained crude product was purified by silica gel column chromatography (mobile phase: ethyl acetate/petroleum ether=0-5%) to give compound 2e (367 mg, 35.93%) as a yellow oil.
LC-MS:m/z 443.2[M+H] +
Step 5: (R) -N 4 - (1- ((tert-butyldimethylsilyl) oxy) -2-methylhex-2-yl) -7-chloro-N 2 - (2, 4-dimethoxybenzyl) pyrido [4,3-d ]]Preparation of pyrimidine-2, 4-diamine (2 f)
Compound 2e (367 mg,0.83 mmol) was dissolved in 1, 4-dioxane (5 mL) and 2, 4-dimethoxybenzylamine (969 mg,5.80 mmol) and N, N-diisopropylethylamine (321 mg,2.48 mmol) were added sequentially at room temperature. The reaction mixture was stirred at 100℃overnight. After the reaction was completed, it was cooled to room temperature. The reaction solution was diluted with water (20 mL), extracted with dichloromethane (3×20 mL), the organic phases were combined, washed with saturated ammonium chloride solution (2×30 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the obtained crude product was separated and purified by silica gel column chromatography (mobile phase: ethyl acetate/petroleum ether=0-8%) to give compound 2f (264 mg, 78.70%) as a pale yellow solid.
LC-MS:m/z 574.3[M+H] +
Step 6: (R) -N 4 - (1- ((tert-butyldimethylsilyl) oxy) -2-methylhex-2-yl) -N 2 - (2, 4-dimethoxybenzyl) -7- (2- (4-methylpiperazin-1-yl) pyrimidin-5-yl) pyrido [4,3-d]Preparation of pyrimidine-2, 4-diamine (2 g)
Compound 2f (204 mg,0.36 mmol) was dissolved in 1, 4-dioxane (2 mL) and water (0.4 mL) at room temperature, and 2- (4-methylpiperazin-1-yl) pyrimidine-5-boronic acid pinacol ester (CAS: 942922-07-8) (2 h) (264 mg,1.05 mmol), potassium carbonate (148 mg,1.07 mmol) and [1,1' -bis (diphenylphosphino) ferrocene were added sequentially ]Palladium dichloride (Pd (dppf) Cl) 2 ) (26 mg,0.036 mmol). The reaction was stirred overnight at 95℃under nitrogen. After completion of the reaction, cooled to room temperature, 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 concentrated under reduced pressure, and the resulting crude product was isolated and purified by preparative thin layer chromatography (mobile phase: ethyl acetate/petroleum ether=1:2) to give 2g (33 mg, 12.97%) of a yellow oily compound.
LC-MS:m/z 716.4[M+H] +
Step 7: preparation of (R) -2- ((2-amino-7- (2- (4-methylpiperazin-1-yl) pyrimidin-5-yl) pyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (2)
2g (33 mg,0.046 mmol) of the compound was dissolved in trifluoroacetic acid (1 mL) at room temperature. The reaction was stirred at 40℃overnight. After completion of the reaction, the reaction mixture was concentrated under reduced pressure, and the obtained crude product was separated and purified by preparative thin layer chromatography (mobile phase: dichloromethane/methanol/triethylamine=100:20:1) to give compound 2 (6.4 mg, 28.66%) as an off-white solid.
1 H NMR (300 MHz, methanol-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] +
Example 3: preparation of (R) -2- ((2-amino-7- (6- (4-methylpiperazin-1-yl) pyridin-3-yl) pyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (3)
The same procedure as in example 2 was followed, except that 2- (4-methylpiperazin-1-yl) pyrimidine-5-boronic acid pinacol ester (2 h) was replaced with 2- (4-methylpiperazin-1-yl) pyridine-5-boronic acid pinacol ester (CAS: 918524-63-7) (3B), and the crude product was purified by preparative chromatography (column: sunFire Prep C18 OBD column, 5um,19 x 150mm; mobile phase A: water (0.1% formic acid), mobile phase B: acetonitrile; flow rate: 25mL/min; gradient: 6% -17% acetonitrile in 8 min; detection wavelength: 220 nm) to give formate salt of compound 3.
1 H NMR (300 MHz, methanol-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] +
Example 4: preparation of (R) -2- ((2-amino-7- (5-methyl-6- (4-methylpiperazin-1-yl) pyridin-3-yl) pyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (4)
Step 1: preparation of (5-methyl-6- (4-methylpiperazin-1-yl) pyridin-3-yl) boronic acid (4 b)
The compound 1- (5-bromo-3-methylpyridin-2-yl) -4-methylpiperazine 4a (500 mg,1.85 mmol) was dissolved in 1, 4-dioxane (10.0 mL) at room temperature, followed by the pinacol diboronate (B) 2 (Pin) 2 ) (704 mg,2.78 mmol), potassium acetate (545 mg,5.55 mmol) and [1,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride dichloroMethane (151 mg,0.19 mmol). The reaction solution was stirred at 80℃for 3 hours under nitrogen atmosphere. After completion of the reaction, the reaction mixture was diluted with water (20 mL), extracted with ethyl acetate (2X 20 mL), and the resulting aqueous phase was concentrated under reduced pressure, and the crude product was purified by reverse phase column chromatography (column: ai Jieer 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: 220/254 nm) to give off-white solid compound 4B (370 mg, 85.04%).
LC-MS:m/z 236.1[M+H] +
The remaining procedure was the same as in the preparation of example 2, except that the compound 2- (4-methylpiperazin-1-yl) pyrimidine-5-boronic acid pinacol ester (2 h) was replaced with (5-methyl-6- (4-methylpiperazin-1-yl) pyridin-3-yl) boronic acid (4B), and the resulting crude product was purified by preparative chromatography (column: XBRID Prep C18 OBD column, 5um,19 x 150mm; mobile phase A: water (10 mmol/L ammonium bicarbonate), mobile phase B: acetonitrile; flow rate: 25mL/min; gradient: 22% -46% acetonitrile in 8 min; detection wavelength: 254/220 nm) to give compound 4.
1 H NMR (400 MHz, methanol-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] +
Example 5: preparation of (R) -2- ((2-amino-7- (6- (pyrrolidin-1-yl) pyridin-3-yl) pyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (5)
The procedure was followed in the same manner as in example 2, except that the compound 2- (4-methylpiperazin-1-yl) pyrimidine-5-boronic acid pinacol ester (2 h) was replaced with 6- (pyrrolidin-1-yl) pyridine-3-boronic acid (CAS: 1150114-75-2) (5B), and the crude product obtained was purified by separation using a preparative chromatography column (column: sunFire Prep C18 OBD column, 5um,19 x 150mm; mobile phase A: water (0.1% formic acid), mobile phase B: acetonitrile; flow rate: 25mL/min; gradient: 7% -21% acetonitrile; detection wavelength: 220 nm) to give formate salt of compound 5.
1 H NMR (300 MHz, methanol-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] +
Example 6: preparation of (R) -2- ((2-amino-7- (6- (piperidin-1-yl) pyridin-3-yl) pyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (6)
The same procedure as in example 2 was followed, except that 2- (4-methylpiperazin-1-yl) pyrimidine-5-boronic acid pinacol ester (2 h) was replaced with 2- (piperidin-1-yl) pyridine-5-boronic acid pinacol ester (CAS: 852228-08-1) (6B), and the crude product obtained was purified by preparative chromatography (column: sunFire Prep C18 OBD column, 5um,19 x 150mm; mobile phase A: water (0.1% formic acid), mobile phase B: acetonitrile; flow rate: 25mL/min; gradient: 13% -29% acetonitrile in 8 min; detection wavelength: 220 nm) to give formate salt of compound 6.
1 H NMR (400 MHz, methanol-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] +
Example 7: preparation of (R) -2- ((2-amino-7- (6- (4-hydroxypiperidin-1-yl) pyridin-3-yl) pyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol formate salt (7)
In the same manner as in example 2, except that 2- (4-methylpiperazin-1-yl) pyrimidine-5-boronic acid pinacol ester (2 h) was replaced with 6- (4-hydroxypiperidin-1-yl) pyridine-3-boronic acid pinacol ester (CAS: 1251948-86-3) (7B), the crude product was purified by preparative chromatography (column: XBridge Shield RP OBD column, 5um,19 x 150mm; mobile phase A: water (0.1% formic acid), mobile phase B: acetonitrile; flow rate: 25mL/min; gradient: 10% -32% acetonitrile; detection wavelength: 254 nm) to obtain formate salt of compound 7.
1 H NMR (300 MHz, methanol-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] +
Example 8: preparation of (R) -2- ((2-amino-7- (imidazo [1,2-a ] pyridin-6-yl) pyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (8)
The same procedure as in example 2 was followed, except that 2- (4-methylpiperazin-1-yl) pyrimidine-5-boronic acid pinacol ester (2 h) was replaced with imidazo [1,2-a ] pyridine-6-boronic acid pinacol ester (CAS: 1204742-76-6) (8B), and the crude product was purified by preparative chromatography (column: sunFire Prep C18 OBD column, 5um,19 x 150mm; mobile phase A: water (0.1% formic acid), mobile phase B: acetonitrile; flow rate: 25mL/min; gradient: 7% -15% acetonitrile in 8 min; detection wavelength: 254 nm) to give formate salt of compound 8.
1 H NMR (300 MHz, methanol-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] +
Example 9: preparation of (R) -2- ((2-amino-7- (6- (4-cyclopropylpiperazin-1-yl) pyridin-3-yl) pyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (9)
Step 1: preparation of 2-amino-7-chloropyrido [4,3-d ] pyrimidin-4 (3H) -one (9 b)
4, 6-Dichloronicotinic acid 9a (5.0 g,26.04 mmol) and guanidine hydrochloride (2.74 g,28.646 mmol) were dissolved in N, N-dimethylformamide (100 mL) at room temperature, and cesium carbonate (16.97 g,52.084 mmol) and cuprous iodide (0.99 g,5.208 mmol) were added sequentially to the reaction solution. The reaction was stirred overnight at 110℃under nitrogen. After the reaction, the solid in the reaction mixture was removed by filtration, and the filtrate was concentrated under reduced pressure. The resulting residue was added to a saturated sodium carbonate solution and extracted with n-butanol (5X 50 mL). The combined organic phases were washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give compound 9b (4.2 g, 82.04%) as an off-white to pale green solid.
LC-MS:m/z 197.0[M+H] +
Step 2: preparation of (R) -2- ((2-amino-7-chloropyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (9 c)
Compound 9b (500.00 mg,2.543 mmol), (R) -2-amino-2-methylhex-1-ol (500.61 mg,3.815 mmol), a catter condensing agent (BOP) (1462.34 mg,3.306 mmol), 1, 8-diazabicyclo undec-7-ene (DBU) (1161.58 mg,7.629 mmol) was dissolved in N, N-dimethylformamide (8 mL) at room temperature. The reaction was stirred at room temperature under nitrogen overnight. After the completion of the reaction, the reaction mixture was quenched with water (50 mL), ethyl acetate (3×40 mL) was added to the reaction mixture, the combined organic phases were washed with saturated brine (50 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the crude product was purified by silica gel column chromatography (mobile phase: ethyl acetate/petroleum ether=1:1) to give yellow semisolid compound 9c (200 mg, 22.84%).
LC-MS:m/z 310.1[M+H] +
Step 3: preparation of 2- (4-cyclopropylpiperazin-1-yl) pyridine-5-boronic acid pinacol ester (9 d)
6-chloropyridine-3-boronic acid pinacol ester (50 mg,0.209 mmol) was dissolved in dimethyl sulfoxide (1 mL) at room temperature, and 1-cyclopropylpiperazine (52.69 mg,0.418 mmol) was added to the reaction solution, followed by stirring at 150℃for 3 hours under nitrogen atmosphere. After the completion of the reaction, the reaction was quenched with water (20 mL), ethyl acetate (3X 20 mL) was added to the system, 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 to give compound 9d (72 mg, crude) as a brown solid, which was used in the next step without purification.
LC-MS:m/z 330.2[M+H] +
Step 4: preparation of (R) -2- ((2-amino-7- (6- (4-cyclopropylpiperazin-1-yl) pyridin-3-yl) pyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (9)
Compound 9c (32.00 mg,0.103 mmol) and compound 9d (68.02 mg,0.206 mmol) were dissolved in 1, 4-dioxane (1 mL) and water (0.1 mL) at room temperature, and potassium carbonate (42.83 mg,0.309 mmol) and [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride (16.83 mg,0.021 mmol) were added sequentially. The reaction was stirred at 95℃overnight under nitrogen. After completion of the reaction, cooled to room temperature, diluted with water (10 mL), extracted with ethyl acetate (3×20 mL), the combined organic phases were washed with saturated brine (50 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate concentrated under reduced pressure, and the resulting crude product was isolated and purified by preparative thin layer chromatography (mobile phase: ethyl acetate/triethylamine=20:1) to give a crude product as a yellow oil. The product was further purified by preparative chromatography (column: XSelect CSH Prep C OBD column, 5um,19 x 150mm; mobile phase A: water (0.05% ammonia), mobile phase B: acetonitrile; flow rate: 25mL/min; gradient: 32% -52% acetonitrile in 8 min; detection wavelength: 254/220 nm) to give off-white solid compound 9 (17.5 mg, 35.55%).
1 H NMR (300 MHz, methanol-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] +
Example 10: preparation of (R) -4- (5- (2-amino-4- ((1-hydroxy-2-methylhex-2-yl) amino) pyrido [4,3-d ] pyrimidin-7-yl) pyridin-2-yl) -1-methylpiperazin-2-one (10)
Compound 10 was prepared in the same manner as in example 9, except that 1-cyclopropylpiperazine was replaced with 1-methylpiperazin-2-one.
1 H NMR (300 MHz, methanol-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] +
Example 11: preparation of (R) -2- ((2-amino-7- (6- (4-ethylpiperazin-1-yl) pyridin-3-yl) pyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (11)
In the same manner as in example 9, except that the compound 1-cyclopropylpiperazine was replaced with 1-ethylpiperazine, compound 11 was produced.
1 H NMR (400 MHz, methanol-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] +
Example 12: preparation of (R) -2- ((2-amino-7- (6- (4-isopropylpiperazin-1-yl) pyridin-3-yl) pyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (12)
Compound 12 was produced in the same manner as in the production method of example 9, except that the compound 1-cyclopropylpiperazine was replaced with 1-isopropylpiperazine.
1 H NMR (300 MHz, methanol-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] +
Example 13: preparation of (R) -2- ((2-amino-7- (6- (4- (2-methoxyethyl) piperazin-1-yl) pyridin-3-yl) pyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (13)
In the same manner as in example 9, except that the compound 1-cyclopropylpiperazine was replaced with 1- (2-methoxyethyl) piperazine, compound 13 was produced.
1 H NMR (400 MHz, methanol-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] +
Example 14: preparation of (2R) -2- ((2-amino-7- (6- (3, 4-dimethylpiperazin-1-yl) pyridin-3-yl) pyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (14)
In the same manner as in example 9, except that 1-cyclopropylpiperazine was replaced with 1, 2-dimethylpiperazine, compound 14 was produced.
1 H NMR (400 MHz, methanol-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] +
Example 15: preparation of (2R) -2- ((2-amino-7- (4-methyl-6- (4-methylpiperazin-1-yl) pyridin-3-yl) pyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (15)
In the same manner as in example 9, except that the compound 6-chloropyridine-3-boronic acid pinacol ester was replaced with 4-methyl-6-chloropyridine-3-boronic acid pinacol ester and 1-cyclopropylpiperazine was replaced with 1-methylpiperazine, compound 15 was produced.
1 H NMR (400 MHz, methanol-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] +
Example 16: preparation of (R) -2- ((2-amino-7- (2-methyl-6- (4-methylpiperazin-1-yl) pyridin-3-yl) pyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (16)
In the same manner as in example 9, except that the compound 6-chloropyridine-3-boronic acid pinacol ester was replaced with 2-methyl-6-chloropyridine-3-boronic acid pinacol ester and 1-cyclopropylpiperazine was replaced with 1-methylpiperazine, compound 16 was produced.
1 H NMR (400 MHz, methanol-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] +
Example 17: preparation of (R) -2- ((2-amino-7- (4-fluoro-6- (4-methylpiperazin-1-yl) pyridin-3-yl) pyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (17)
Step 1: preparation of 1- (4-fluoropyridin-2-yl) -4-methylpiperazine (17 a)
2-bromo-4-fluoropyridine (400 mg,2.273 mmol) and N-methylpiperazine were dissolved in toluene (10.0 mL) at room temperature, followed by sequential addition of Rac-BINAP-Pd-G3 (225.56 mg,0.227 mmol) and cesium carbonate (2962.20 mg,9.092 mmol). The reaction was stirred overnight at 95℃under nitrogen. After completion of the reaction, the reaction mixture was diluted with water (30 mL), extracted with ethyl acetate (3X 20 mL), and the organic phases were combined, washed with saturated brine (30 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product was purified by column chromatography (mobile phase: petroleum ether/ethyl acetate=2:1) to give compound 17a (119 mg, 26.82%) as a dark yellow oil.
LC-MS:m/z 196.1[M+H] +
Step 2: preparation of 1- (5-bromo-4-fluoropyridin-2-yl) -4-methylpiperazine (17 b)
The compound 1- (4-fluoropyridin-2-yl) -4-methylpiperazine 17a (119 mg,0.610 mmol) was dissolved in Acetonitrile (ACN) (3 mL) at room temperature, N-bromosuccinimide (NBS) (130.18 mg,0.732 mmol) was added and reacted overnight at room temperature under nitrogen at a dark place. After completion of the reaction, the mixture was concentrated under reduced pressure, the residue was diluted with water (10 mL), extracted with ethyl acetate (3X 10 mL), and the organic phases were combined, washed with saturated brine (20 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product was purified by column chromatography (mobile phase: petroleum ether/ethyl acetate=2:1) to give 17b (166 mg, 99.35%) as an orange semisolid compound.
LC-MS:m/z 274.0[M+H] +
Step 3: preparation of 2- (4-methylpiperazin-1-yl) -4-fluoropyridine-5-boronic acid (17 c)
The compound 1- (5-bromo-4-fluoropyridin-2-yl) -4-methylpiperazine 17b (166 mg,0.606 mmol) was dissolved in 1, 4-dioxane (3.0 mL) at room temperature, followed by the sequential addition of pinacol biborate (230.65 mg,0.909 mmol), potassium acetate (178.29 mg,1.818 mmol) and [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride dichloromethane (98.66 mg,0.121 mmol), and the reaction was stirred overnight at 80℃under nitrogen. After completion of the reaction, the reaction solution was diluted with water (10 mL), extracted with ethyl acetate (2×10 mL), and the resulting aqueous phase was concentrated under reduced pressure to give compound 17c (150 mg, crude product) as a brown solid, which was used in the next step without purification.
LC-MS:m/z 240.1[M+H] +
The remaining procedure was as in example 9, except that 2- (4-cyclopropylpiperazin-1-yl) pyridine-5-boronic acid pinacol ester (9 d) was replaced with 2- (4-methylpiperazin-1-yl) -4-fluoropyridine-5-boronic acid (17 c) to give compound 17.
1 H NMR (400 MHz, methanol-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] +
Example 18: preparation of (R) -2- ((2-amino-7- (6- (4- (dimethylamino) piperidin-1-yl) pyridin-3-yl) pyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (18)
Compound 18 was prepared in the same manner as in example 9, except that 1-cyclopropylpiperazine was replaced with 4-dimethylaminopiperidine.
1 H NMR (400 MHz, methanol-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] +
Example 19: preparation of (R) -2- ((2-amino-7- (6- (4- (2-hydroxyethyl) piperazin-1-yl) pyridin-3-yl) pyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (19)
Compound 19 was produced in the same manner as in example 9, except that 1-cyclopropylpiperazine was replaced with N-hydroxyethylpiperazine.
1 H NMR (400 MHz, methanol-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] +
Example 20: preparation of (R) -2- ((2-amino-7- (6- (4-propylpiperazin-1-yl) pyridin-3-yl) pyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (20)
Step 1: preparation of 2- (4-propylpiperazin-1-yl) pyridine-5-boronic acid pinacol ester (20 a)
1-n-propylpiperazine dibromoate (484.39 mg, 1.640 mmol) and potassium carbonate (464.99 mg,3.340 mmol) were dissolved in dimethyl sulfoxide (5 mL) at room temperature, 2-chloropyridine-5-boronic acid pinacol ester (200 mg,0.835 mmol) was added, and stirred under nitrogen atmosphere at 150℃for 2 hours. After the completion of the reaction, the reaction was quenched with water (30 mL), ethyl acetate (3X 15 mL) was added to the system, 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 to give a brown yellow liquid compound 20a (549 mg, crude product), which was used in the next step without purification.
LC-MS:m/z 332.2[M+H] +
The remaining procedure was as in example 9, except that 2- (4-cyclopropylpiperazin-1-yl) pyridine-5-boronic acid pinacol ester (9 d) was replaced with 2- (4-propylpiperazin-1-yl) pyridine-5-boronic acid pinacol ester (20 a), to give compound 20.
1 H NMR (400 MHz, methanol-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] +
Example 21: preparation of (R) -2- ((2-amino-7- (6- (4- (2, 2-difluoroethyl) piperazin-1-yl) pyridin-3-yl) pyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (21)
In the same manner as in example 20, except that 1-n-propylpiperazine dibromoate was replaced with 1- (2, 2-difluoroethyl) piperazine hydrochloride, compound 21 was produced.
1 H NMR (300 MHz, methanol-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] +
Example 22: preparation of (R) -2- ((2-amino-7- (6- (4- (2-fluoroethyl) piperazin-1-yl) pyridin-3-yl) pyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (22)
Step 1: preparation of 2- (4- (2-fluoroethyl) piperazin-1-yl) pyridine-5-boronic acid pinacol ester (22 a)
2-chloropyridine-5-boronic acid pinacol ester (150 mg, 0.616 mmol) and 1- (2-fluoroethyl) piperazine dihydrochloride (256.90 mg,1.252 mmol) were dissolved in dimethyl sulfoxide (2 mL) at room temperature, N-diisopropylethylamine (323.77 mg,2.504 mmol) was added, and stirred under nitrogen at 150℃for 2 hours. After completion of the reaction, the reaction was quenched with water (20 mL), ethyl acetate (3×20 mL) was added to the system for extraction, 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 to give brown semisolid compound 22a (198 mg, crude), which was used in the next step without purification.
LC-MS:m/z 336.2[M+H] +
The remaining procedure was as in example 9, except that 2- (4-cyclopropylpiperazin-1-yl) pyridine-5-boronic acid pinacol ester (9 d) was replaced with 2- (4- (2-fluoroethyl) piperazin-1-yl) pyridine-5-boronic acid pinacol ester (22 a), to give compound 22.
1 H NMR (400 MHz, methanol-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] +
Example 23: preparation of (R) -2- ((2-amino-7- (6- (4- (2, 2-trifluoroethyl) piperazin-1-yl) pyridin-3-yl) pyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (23)
In the same manner as in example 22, except that 1- (2-fluoroethyl) piperazine dihydrochloride was replaced with 1- (2, 2-trifluoroethyl) piperazine dihydrochloride, compound 23 was produced.
1 H NMR (400 MHz, methanol-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] +
Example 24: preparation of (R) -2- ((2-amino-7- (6- (4-propylpiperidin-1-yl) pyridin-3-yl) pyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (24)
In the same manner as in example 22 except that 1- (2-fluoroethyl) piperazine dihydrochloride was replaced with 4-n-propylpiperidine hydrochloride, compound 24 was produced.
1 H NMR (300 MHz, methanol-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] +
Example 25: preparation of (R) -2- ((2-amino-7- (6- (3-ethoxyazetidin-1-yl) pyridin-3-yl) pyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (25)
In the same manner as in example 22, except that 1- (2-fluoroethyl) piperazine dihydrochloride was replaced with 3-ethoxyazetidine hydrochloride, compound 25 was prepared.
1 H NMR (400 MHz, methanol-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] +
Example 26: preparation of (R) -2- ((2-amino-7- (6- (4-ethoxypiperidin-1-yl) pyridin-3-yl) pyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (26)
Compound 26 was prepared in the same manner as in example 22, except that 1- (2-fluoroethyl) piperazine dihydrochloride was replaced with 4-ethoxypiperidine.
1 H NMR (300 MHz, methanol-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] +
Example 27: preparation of (R) -2- ((2-amino-7- (6- (3- (dipropylamino) azetidin-1-yl) pyridin-3-yl) pyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (27)
In the same manner as in example 22, except that 1- (2-fluoroethyl) piperazine dihydrochloride was replaced with N, N-dipropylazetidin-3-amine, compound 27 was prepared.
1 H NMR (300 MHz, methanol-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] +
Example 28: preparation of (R) -2- ((2-amino-7- (6- (4-diethylaminopiperidin-1-yl) pyridin-3-yl) pyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (28)
Compound 28 was prepared in the same manner as in example 9, except that 1-cyclopropylpiperazine was replaced with 4-diethylaminopiperidine.
1 H NMR (300 MHz, methanol-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] +
Example 29: preparation of (R) -2- ((2-amino-7- (6- (3- (diethylamino) azetidin-1-yl) pyridin-3-yl) pyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (29)
In the same manner as in example 22, except that 1- (2-fluoroethyl) piperazine dihydrochloride was replaced with 3- (diethylamino) azetidine dihydrochloride, compound 29 was prepared.
1 H NMR (300 MHz, methanol-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] +
Example 30: preparation of (R) -2- ((2-amino-7- (2- (4- (2-fluoroethyl) piperazin-1-yl) pyrimidin-5-yl) pyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (30)
Step 1: preparation of 2- (4- (2-fluoroethyl) piperazin-1-yl) pyrimidine-5-boronic acid (30 a)
2-chloropyrimidine-5-boronic acid (148 mg,0.935 mmol) and 1- (2-fluoroethyl) piperazine dihydrochloride (209.92 mg,1.029 mmol) were dissolved in ethanol (2 mL) at room temperature, triethylamine (331.02 mg, 3.271mmol) was added to the reaction solution, and the mixture was stirred under nitrogen at 75℃for 1 hour. After the completion of the reaction, the reaction solution was cooled to room temperature and concentrated under reduced pressure to give compound 30a (260 mg, crude product) as a yellow solid, which was used in the next step without purification.
LC-MS:m/z 255.1[M+H] +
The remaining procedure was the same as in example 9 except that compound 2- (4-cyclopropylpiperazin-1-yl) pyridine-5-boronic acid pinacol ester (9 d) was replaced with (30 a), to obtain compound 30.
1 H NMR (300 MHz, methanol-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] +
Example 31: preparation of (R) -2- ((2-amino-7- (2- (4-propylpiperazin-1-yl) pyrimidin-5-yl) pyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (31)
In the same manner as in example 30, except that 1- (2-fluoroethyl) piperazine dihydrochloride was replaced with 1-n-propylpiperazine dibromoate, compound 31 was obtained.
1 H NMR (400 MHz, methanol-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] +
Example 32: preparation of (R) -2- ((2-amino-7- (2- (4-isopropylpiperazin-1-yl) pyrimidin-5-yl) pyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (32)
Compound 32 was prepared in the same manner as in example 30, except that 1- (2-fluoroethyl) piperazine dihydrochloride was replaced with 1-isopropylpiperazine.
1 H NMR (300 MHz, methanol-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] +
Example 33: preparation of (R) -2- ((2-amino-7- (2- (4- (dimethylamino) piperidin-1-yl) pyrimidin-5-yl) pyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (33)
In the same manner as in example 30 except that 1- (2-fluoroethyl) piperazine dihydrochloride was replaced with 4-dimethylaminopiperidine, compound 33 was produced.
1 H NMR (400 MHz, methanol-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] +
Example 34: preparation of (R) -2- ((2-amino-7- (4- (4-methylpiperazin-1-yl) phenyl) pyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (34)
In the same manner as in example 9 except that 2- (4-cyclopropylpiperazin-1-yl) pyridine-5-boronic acid pinacol ester (9 d) was replaced with 4- (4-methylpiperazin-1-yl) phenylboronic acid pinacol ester (CAS: 747413-21-4) (34 a), compound 34 was produced.
1 H NMR (300 MHz, methanol-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] +
Example 35: preparation of (R) -2- ((2-amino-7- (1- (1-methylpiperidin-4-yl) -1H-pyrazol-4-yl) pyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (35)
In the same manner as in example 9 except that 2- (4-cyclopropylpiperazin-1-yl) pyridine-5-boronic acid pinacol ester (9 d) was replaced with (1-methylpiperidin-4-yl) -1H-pyrazole-4-boronic acid pinacol ester (CAS: 1323919-64-7) (35 a), compound 35 was produced.
1 H NMR (300 MHz, methanol-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] +
Example 36: preparation of (R) -2- ((2-amino-7- (1- (tetrahydropyran-4-yl) -1H-pyrazol-4-yl) pyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (36)
In the same manner as in example 9 except that 2- (4-cyclopropylpiperazin-1-yl) pyridine-5-boronic acid pinacol ester (9 d) was replaced with 1- (tetrahydropyran-4-yl) -1H-pyrazole-4-boronic acid pinacol ester (CAS: 1040377-03-4) (36 a), compound 36 was prepared.
1 H NMR (400 MHz, methanol-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] +
Example 37: preparation of (R) -2- ((2-amino-7- (4- (3-pyridinyl) phenyl) pyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (37)
In the same manner as in example 9 except that 2- (4-cyclopropylpiperazin-1-yl) pyridine-5-boronic acid pinacol ester (9 d) was replaced with 4- (3-pyridyl) phenylboronic acid pinacol ester (CAS: 929203-04-3) (37 a), compound 37 was produced.
1 H NMR (400 MHz, methanol-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] +
Example 38: preparation of (R) -2- ((2-amino-7- (6- (piperazin-1-yl) pyridin-3-yl) pyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (38)
Step 1: preparation of (R) -4- (5- (2-amino-4- ((1-hydroxy-2-methylhex-2-yl) amino) pyrido [4,3-d ] pyrimidin-7-yl) pyridin-2-yl) piperazine-1-carboxylic acid tert-butyl ester (38 b)
In the same manner as in step 4 of example 9 except that 2- (4-cyclopropylpiperazin-1-yl) pyridine-5-boronic acid pinacol ester (9 d) was replaced with 4- [4- (N-Boc) piperazin-1-yl ] phenylboronic acid pinacol ester (CAS: 496786-98-2) (38 a), compound 38b was produced.
Step 2: preparation of (R) -2- ((2-amino-7- (6- (piperazin-1-yl) pyridin-3-yl) pyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (38)
Compound 38b (50 mg,0.093 mmol) was dissolved in methylene chloride (1 mL) at room temperature, trifluoroacetic acid (1 mL) was added to the reaction mixture, and the mixture was stirred at room temperature under a nitrogen atmosphere for 2 hours. After the reaction, the reaction solution was concentrated under reduced pressure and purified by preparative thin layer chromatography (mobile phase: dichloromethane/methanol=10:1) to obtain a crude product. The crude product was further purified by preparative chromatography (column: gemini-NX C18 AXAI packet column, 5um,21.2 x 150mm; mobile phase A: water (0.05% ammonia), mobile phase B: acetonitrile; flow rate: 25mL/min; gradient: 23% -41% acetonitrile in 8 min; detection wavelength: 254/220 nm) to give off-white solid compound 38 (2.1 mg, 4.76%).
1 H NMR (400 MHz, methanol-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] +
Example 39: preparation of (R) -2- ((7- ([ 2,3' -bipyridyl ] -5-yl) -2-aminopyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (39)
Step 1: preparation of 5-bromo-2, 3' -bipyridine (39 a)
3-Pyridineboronic acid (500.09 mg,4.069 mmol) and 5-bromo-2-iodopyridine (1050 mg,3.699 mmol) were dissolved in 1, 4-dioxane (10.0 mL) and potassium carbonate (1533.49 mg,11.097 mmol) and tetrakis (triphenylphosphine) palladium (427.39 mg,0.37 mmol) were added in sequence, and the reaction stirred overnight at 80℃under nitrogen. After completion of the reaction, the reaction mixture was diluted with water (30 mL), extracted with ethyl acetate (3X 30 mL), and the organic phases were combined, washed with saturated brine (30 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product was purified by column chromatography (mobile phase: petroleum ether/ethyl acetate=4:1) to give compound 39a (148 mg, 17.02%) as a yellow solid.
LC-MS:m/z 235.0[M+H] +
Step 2: preparation of 2,3' -bipyridine-5-boronic acid (39 b)
Compound 39a (148 mg,0.63 mmol) was dissolved in 1, 4-dioxane (3.0 mL) at room temperature, and pinacol biborate (239.81 mg,0.944 mmol), potassium acetate (185.36 mg,1.889 mmol) and [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride dichloromethane complex (51.29 mg,0.063 mmol) were sequentially added to the reaction solution, and the reaction solution was stirred at 80℃for 3 hours under nitrogen atmosphere. After completion of the reaction, the reaction mixture was diluted with water (10 mL), extracted with ethyl acetate (3×10 mL), and the organic phases were combined, washed with saturated brine (20 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give compound 39b (314 mg, crude) as a brown solid, which was used in the next step without purification.
LC-MS:m/z 201.1[M+H] +
The remaining procedure was as in example 9, except that compound 2- (4-cyclopropylpiperazin-1-yl) pyridine-5-boronic acid pinacol ester (9 d) was replaced with 2,3' -bipyridine-5-boronic acid (39 b), producing compound 39.
1 H NMR (400 MHz, methanol-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] +
Example 40: preparation of (R) -2- ((7- ([ 2,4' -bipyridyl ] -5-yl) -2-aminopyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (40)
Compound 40 was produced in the same manner as in example 39, except that 3-pyridineboronic acid was replaced with 4-pyridineboronic acid.
1 H NMR (400 MHz, methanol-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] +
Example 41: preparation of (R) -2- ((2-amino-7- (6 '-methyl- [2,3' -bipyridin ] -5-yl) pyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (41)
In the same manner as in example 39, except that the compound 3-pyridineboronic acid was replaced with 6-methylpyridine-3-boronic acid, compound 41 was prepared.
1 H NMR (400 MHz, methanol-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] +
Example 42: preparation of (R) -2- ((7- ([ 2,2' -bipyridyl ] -5-yl) -2-aminopyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (42)
In the same manner as in example 39 except that the compound 5-bromo-2, 3 '-bipyridine (39 a) was replaced with 5-bromo-2, 2' -bipyridine (42 a), compound 42 was produced.
1 H 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] +
Example 43: preparation of (R) -2- ((2-amino-7- (4- (pyrrolidin-1-ylmethyl) benzyl) pyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (43)
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 ]]Preparation of pyrimidine-2, 4-diamine (43 a)
Compound 2f (150 mg,0.26 mmol) was dissolved in 1, 4-dioxane (2 mL) and water (0.5 mL) at room temperature, followed by the addition of 4-chlorobenzyl boronic acid pinacol ester (CAS: 475250-49-8) (43 b) (264 mg,1.05 mmol), potassium carbonate (255 mg,1.83 mmol) and tetrakis (triphenylphosphine) palladium (60 mg,0.052 mmol). The reaction solution was stirred under nitrogen at 95℃for 3 hours. After completion of the reaction, the reaction mixture was diluted with water (15 mL), extracted with ethyl acetate (3×10 mL), the organic phases were combined, washed with saturated brine (30 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the obtained crude product was isolated and purified by preparative thin layer chromatography (mobile phase: ethyl acetate/petroleum ether=1:2) to give compound 43a (92 mg, 53.01%) as an orange oil.
LC-MS:m/z 664.3[M+H] +
Step 2: (R) -N 4 - (1- ((tert-butyldimethylsilyl) oxy) -2-methylhex-2-yl) -N 2 - (2, 4-dimethoxybenzyl) -7- (4- (pyrrolidin-1-ylmethyl) benzyl) pyrido [4,3-d]Preparation of pyrimidine-2, 4-diamine (43 c)
Compound 43a (92 mg,0.14 mmol) was dissolved in 1, 4-dioxane (2 mL) and water (0.5 mL) at room temperature, followed by the addition of potassium (1-pyrrolidinylmethyl) trifluoroborate (37 mg,0.19 mmol), potassium carbonate (57.8 mg,0.41 mmol) and XPhos Pd G3 (11.7 mg,0.014 mmol). The reaction was stirred overnight at 100℃under nitrogen. After completion of the reaction, the reaction mixture was diluted with water (15 mL), extracted with ethyl acetate (3×10 mL), and the organic phases were combined, washed with saturated brine (30 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the obtained crude product was isolated and purified by preparative thin layer chromatography (mobile phase: ethyl acetate/petroleum ether=1:2) to give compound 43b (33 mg, 33.4%) as an orange oil.
LC-MS:m/z 713.4[M+H] +
Step 3: preparation of (R) -2- ((2-amino-7- (4- (pyrrolidin-1-ylmethyl) benzyl) pyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (43)
Compound 43b (33 mg,0.046 mmol) was dissolved in dichloromethane (1 mL) at room temperature, and trifluoroacetic acid (1 mL) was slowly added dropwise. The reaction was stirred at 40℃overnight. After the reaction was completed, the reaction mixture was concentrated under reduced pressure, and the crude product obtained was purified by preparative chromatography (column type: XBridge Shield RP OBD column, 5um,19 x 150mm; mobile phase A: water (10 mmol/L ammonium bicarbonate), mobile phase B: acetonitrile; flow rate: 25mL/min; gradient: 11% -47% acetonitrile in 8 minutes; detection wavelength: 254/220 nm) to give compound 43 (1.6 mg, 7.41%) as a white solid.
1 H NMR (400 MHz, methanol-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] +
Example 44: n (N) 4 -propylpyrido [3,4-d ]]Preparation of pyrimidine-2, 4-diamine (44)
Step 1: preparation of 2-aminopyrido [3,4-d ] pyrimidin-4 (3H) -one (44 b)
3-Bromoiisonicotinic acid 44a (5.0 g,24.75 mmol) was dissolved in N, N-dimethylformamide (120 mL) at room temperature, cesium carbonate (16.1 g,49.50 mmol), cuprous iodide (0.94 g,4.95 mmol), guanidine hydrochloride (2.60 g,27.23 mmol) were added in this order, and the reaction was stirred overnight at 110℃under nitrogen. After the completion of the reaction, the solid in the system was filtered, and the residue was washed with methanol (3X 100 mL). The collected filtrate was concentrated under reduced pressure, and the crude product was purified by silica gel column chromatography (mobile phase: methanol/dichloromethane=20% -30%) to give compound 44b (3.2 g, 79.4%) as a pale yellow solid.
LC-MS:m/z 163.1[M+H] +
Step 2: n (N) 4 -propylpyrido [3,4-d ]]Preparation of pyrimidine-2, 4-diamine (44)
Compound 44b (40 mg,0.247 mmol) was dissolved in N, N-dimethylformamide (3 mL) at room temperature, and a catter condensing agent (BOP) (218 mg,0.494 mmol), 1, 8-diazabicyclo undec-7-ene (DBU) (75.1 mg,0.494 mmol), N-propylamine (29.2 mg,0.494 mmol) and the reaction mixture was reacted at room temperature under a nitrogen atmosphere for 4 hours. After completion of the reaction, the mixture was quenched with water (10 mL) and extracted with ethyl acetate (3X 20 mL). The organic phases were combined and washed with saturated brine (2X 20 mL). The organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure to give crude product. The crude product was purified by preparative reverse phase chromatography (column: XBridge Shield RP OBD column, 5um,19 x 150mm; mobile phase A: water (10 mmol/L ammonium bicarbonate), mobile phase B: acetonitrile; flow rate: 25mL/min; gradient: 47% -53% acetonitrile in 8 min; detection wavelength: 254/220 nm) to give compound 44 (10.2 mg, 20.8%) as a white solid.
1 H NMR (400 MHz, methanol-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] +
Example 45: n (N) 4 -n-butylpyrido [3,4-d ]]Preparation of pyrimidine-2, 4-diamine (45)
Compound 45 was produced in the same manner as in example 44, except that n-propylamine was replaced with n-butylamine.
1 H NMR (400 MHz, methanol-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] +
Example 46: n (N) 4 -n-pentylpyrido [3,4-d ]]Preparation of pyrimidine-2, 4-diamine (46)
Compound 46 was prepared in the same manner as in example 44, except that n-propylamine was replaced with n-pentylamine.
1 H NMR (400 MHz, methanol-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] +
Example 47: preparation of 2- ((2-aminopyridine [3,4-d ] pyrimidin-4-yl) amino) -1-pentanol (47)
Compound 47 was prepared in the same manner as in example 44, except that n-propylamine was replaced with DL-2-amino-1-pentanol. 1 H NMR (400 MHz, methanol-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] +
Example 48: preparation of 2- ((2-aminopyridine [3,4-d ] pyrimidin-4-yl) amino) -1-hexanol (48)
Compound 47 was prepared in the same manner as in example 44, except that n-propylamine was replaced with DL-2-amino-1-hexanol. 1 H NMR (400 MHz, methanol-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] +
Example 49: preparation of (R) -2- ((2-aminopyrido [3,4-d ] pyrimidin-4-yl) amino) -2-methylbutan-1-ol (49)
Compound 49 was prepared in the same manner as in example 44, except that n-propylamine was substituted for (R) -2-amino-2-methylbutan-1-ol (49 a).
1 H NMR (400 MHz, methanol-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] +
Example 50: preparation of (R) -2- ((2-aminopyrido [3,4-d ] pyrimidin-4-yl) amino) -2-methylpentan-1-ol (50)
Compound 50 was prepared in the same manner as in example 44, except that n-propylamine was replaced with (R) -2-amino-2-methylpentan-1-ol (50 a).
1 H NMR (400 MHz, methanol-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] +
Example 51: preparation of (R) -2- ((2-aminopyrido [3,4-d ] pyrimidin-4-yl) amino) -2-methylhept-1-ol (51)
In the same manner as in example 44, except that n-propylamine was replaced with (R) -2-amino-2-methylhept-1-ol (51 a), compound 51 was obtained. 1 H NMR (400 MHz, methanol-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] +
Examples 52 and 53: preparation of (R) -2- ((2-aminopyrido [3,4-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (52) and (S) -2- ((2-aminopyrido [3,4-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (53)
Step 1: preparation of (R) -N- ((1- ((tert-butyldimethylsilyl) oxy) -2-methylhex-2-yl) -2-chloropyrido [3,4-d ] pyrimidin-4-amine (52 b)
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. To the reaction solution was added successively (R) -1- ((tert-butyldimethylsilyl) oxy) -2-methylhex-2-amine (1 e) (123 mg,0.50 mmol) and N, N-diisopropylethylamine (0.26. Mu.L, 1.49 mmol), and the mixture was stirred at room temperature for 2 hours. After the reaction was completed, it was cooled to room temperature. The reaction mixture was diluted with water (20 mL), extracted with ethyl acetate (3X 20 mL), and the organic phases were combined, washed with saturated brine (20 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product was purified by silica gel column chromatography (mobile phase: petroleum ether/ethyl acetate=4/1) to give compound 52b (110 mg, 53.79%) as a yellow oil.
LC-MS:m/z 409.1[M+H] +
Step 2: (R) -N 4 - (1- ((tert-butyldimethylsilyl) oxy) -2-methylhex-2-yl) -N 2 - (2, 4-dimethoxybenzyl) pyrido [3,4-d ]]Preparation of pyrimidine-2, 4-diamine (52 c)
Compound 52b (110 mg,0.27 mmol) was dissolved in 2, 4-dimethoxybenzylamine (1.0 mL) at room temperature. N, N-diisopropylethylamine (140. Mu.l, 0.80 mmol) was added to the reaction mixture, and the mixture was stirred at 80℃for 2 hours. After the reaction was completed, it was cooled to room temperature. The reaction mixture was diluted with water (20 mL), extracted with ethyl acetate (3X 20 mL), and the organic phases were combined, washed with saturated brine (20 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product was purified by column chromatography on silica gel (mobile phase: petroleum ether/ethyl acetate=3:2) to give compound 52c (114 mg, 78.53%) as a yellow oil.
LC-MS:m/z 508.3[M+H] +
Step 3: preparation of (R) -2- ((2-aminopyrido [3,4-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (52 d)
Compound 52c (114 mg,0.21 mmol) was dissolved in dichloromethane (0.5 mL) at room temperature. Trifluoroacetic acid (0.5 mL) was added to the reaction mixture, and the mixture was stirred at room temperature for 12 hours. After the reaction was completed, the reaction mixture was concentrated under reduced pressure, and the crude product obtained was purified by preparative chromatography (column type: XBridge Shield RP OBD column, 5um,19 x 150mm; mobile phase A: water (10 mmol/L ammonium bicarbonate), mobile phase B: acetonitrile; flow rate: 25mL/min; gradient: 20% -38% acetonitrile in 8 minutes; detection wavelength: 220 nm) to give a white solid compound 52d (12 mg, 20.18%).
1 H NMR (300 MHz, methanol-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] +
Step 4: preparation of (R) -2- ((2-aminopyrido [3,4-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (52) and (S) -2- ((2-aminopyrido [3,4-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (53)
The compound 52d was isolated by chiral separation (column: optiChiral A6-5 column, 3X 25cm,5um; mobile phase A: carbon dioxide; mobile phase B: methanol (0.1% 2M methanolic ammonia solution), flow rate: 50mL/min; gradient: 50% B etc., detection wavelength: 220 nm) to give the compound 52 as a white solid product, t R =2.052 min, ee value (enantiomeric excess): 100%; and white solid product compound 53, t R =2.383min,
ee value (enantiomeric excess): 92.8%.
Compound 52:
1 h NMR (300 MHz, methanol-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] +
Compound 53:
1 h NMR (400 MHz, methanol-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] +
Example 54: preparation of 2- ((2-aminopyrido [3,4-d ] pyrimidin-4-yl) amino) -1-heptanol (54)
Compound 54 was prepared in the same manner as in steps 1 to 3 of example 52 except that (R) -1- ((tert-butyldimethylsilyl) oxy) -2-methylhex-2-amine (1 e) was replaced with 2-amino-1-heptanol.
1 H NMR (300 MHz, methanol-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] +
Example 55: preparation of (R) -2- ((2-amino-8-fluoropyrido [3,4-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (55)
Step 1: preparation of 2-amino-8-fluoropyrido [3,4-d ] pyrimidin-4 (3H) -one (55 b)
3-chloro-2-fluoroisonicotinic acid (55 a) (1.0 g,5.70 mmol) was dissolved in N, N-dimethylformamide (15 mL) at room temperature, cesium carbonate (5.57 g,17.1 mmol), guanidine carbonate (1.03 g,5.72 mmol), cuprous iodide (217 mg,1.14 mmol) were added in this order, and the reaction solution was stirred under nitrogen at 110℃for 4 hours. After the completion of the reaction, the reaction mixture was quenched with water (100 mL), chloroform (3×50 mL) was added to the reaction mixture, the combined organic phases were washed with saturated brine (100 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the crude product was purified by silica gel column chromatography (mobile phase: methanol/dichloromethane=0 to 10%) to give 55b (83.9 mg, 8.2%) as a pale yellow solid.
LC-MS:m/z 181.0[M+H] +
Step 2: (R) -N 4 - (1- ((tert-butyldimethylsilyl) oxy) -2-methylhex-2-yl) -8-fluoropyrido [3,4-d ]]Preparation of pyrimidine-2, 4-diamine (55 c)
Compound 55b (30 mg,0.167 mmol) was dissolved in N, N-dimethylformamide (5 mL), and (R) -1- ((tert-butyldimethylsilyl) oxy) -2-methylhex-2-amine (1 e) (122 mg,0.50 mmol), 1, 8-diazabicyclo undec-7-ene (76.1 mg,0.50 mmol), and a Kate condensing agent (110 mg,0.25 mmol) were added sequentially, and the reaction was stirred overnight at room temperature under nitrogen. After the completion of the reaction, the reaction mixture was quenched with water (50 mL), ethyl acetate (3×20 mL) was added to the reaction mixture, the combined organic phases were washed with saturated brine (50 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the crude product was purified by silica gel column chromatography (mobile phase: ethyl acetate/petroleum ether=15-30%) to give compound 55c (10 mg, 14.7%) as a white solid.
LC-MS:m/z 408.3[M+H] +
Step 3: preparation of (R) -2- ((2-amino-8-fluoropyrido [3,4-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (55)
Compound 55c (10 mg,0.025 mmol) was dissolved in a mixture of trifluoroacetic acid (2 mL) and dichloromethane (2 mL) at room temperature. The reaction solution was stirred at 45℃for 2 hours. After the reaction, the reaction mixture was concentrated under reduced pressure, the crude product was diluted with water (30 mL), methylene chloride (3X 15 mL) was added to the system, the combined organic phases were washed with saturated brine (30 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give the crude product, which was purified by preparative reverse phase chromatography (column: XBridge Shield RP OBD column, 5um, 19X 150mm; mobile phase A: water (10 mmol/L ammonium bicarbonate), mobile phase B: acetonitrile; flow rate: 25mL/min; gradient: 25% -42% acetonitrile in 8 minutes; detection wavelength: 254/220 nm), to give the product as a white solid 55 (1.4 mg, 19.4%).
1 H NMR (400 MHz, methanol-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] +
Example 56: preparation of (R) -2- ((2-amino-5-fluoropyrido [3,4-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (56)
Step 1: preparation of 3-amino-5-fluoroisonicotinamide (56 b)
Ammonia (30 mL) was added to methyl 3-amino-5-fluoroisonicotinate (56 a) (900 mg,5.29 mmol) at room temperature. The reaction was stirred at 60℃for 3 hours. After the completion of the reaction, the reaction mixture was cooled to room temperature, diluted with water, extracted with ethyl acetate (3×50 mL), and the combined organic phases were washed with saturated brine (50 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the obtained crude product was purified by silica gel column chromatography (mobile phase: ethyl acetate/petroleum ether=1:1) to give a yellow solid compound 56b (700 mg, 84.7%).
LC-MS:m/z 156.0[M+H] +
Step 2: preparation of 5-fluoropyrido [3,4-d ] pyrimidine-2, 4 (1H, 3H) -dione (56 c)
Compound 56b (700 mg,4.49 mmol) was dissolved in 1, 4-dioxane (15 mL) at room temperature, and diphosgene (3.09 g,15.7 mmol) was added. The reaction solution was stirred at 120℃for 16 hours. After the completion of the reaction, the reaction mixture was cooled to room temperature, quenched with water, and then extracted with ethyl acetate (3×40 mL), the combined organic phases were washed with saturated brine (50 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the crude product was purified by silica gel column chromatography (mobile phase: ethyl acetate/petroleum ether=1:1) to give compound 56c (350 mg, 43.1%) as a yellow solid.
LC-MS:m/z 182.0[M+H] +
Step 3: preparation of 2, 4-dichloro-5-fluoropyrido [3,4-d ] pyrimidine (56 d)
Compound 56c (350 mg,1.93 mmol) was mixed with phosphorus oxychloride (5.0 mL) at 0deg.C and N, N-diisopropylethylamine (1.24 g,9.65 mmol) was added. The reaction solution was stirred at 100℃for 0.5 hours. After the completion of the reaction, the reaction mixture was cooled to room temperature and concentrated under reduced pressure to give a brown crude compound 56d (70.0 mg, 16.7%).
LC-MS:m/z 218.0[M+H] +
Step 4: preparation of (R) -N- ((1- ((tert-butyldimethylsilyl) oxy) -2-methylhex-2-yl) -2-chloro-5-fluoropyrido [3,4-d ] pyrimidin-4-amine (56 e)
Compound 56d (70.0 mg,0.32 mmol) was dissolved in 1, 4-dioxane (5.0 mL) at room temperature. (R) -1- ((tert-Butyldimethylsilyl) oxy) -2-methylhex-2-amine (1 e) (94.4 mg,0.39 mmol) and N, N-diisopropylethylamine (144 mg,1.12 mmol) were added sequentially to the reaction solution, and the mixture was reacted at room temperature for 12 hours. After completion of the reaction, the reaction mixture was diluted with water (15 mL), extracted with ethyl acetate (3×10 mL), the organic phases were combined, washed with saturated brine (15 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the obtained crude product was isolated and purified by preparative thin layer chromatography (mobile phase: ethyl acetate/petroleum ether=1:8) to give compound 56e (55.0 mg, 40.2%) as a brown oil.
LC-MS:m/z 427.2[M+H] +
Step 5: (R) -N 4 - (1- ((tert-butyldimethylsilyl) oxy) -2-methylhex-2-yl) -N 2 - (2, 4-dimethoxybenzyl) -5-fluoropyrido [3,4-d ]]Preparation of pyrimidine-2, 4-diamine (56 f)
Compound 56e (55 mg,0.13 mmol) was dissolved in 1, 4-dioxane (5 mL) at room temperature. To the reaction mixture was added 2, 4-dimethoxybenzylamine (21 mg,0.15 mmol) and N, N-diisopropylethylamine (58 mg,0.45 mmol) in this order, and the mixture was stirred at 100℃for 16 hours. After completion of the reaction, the reaction mixture was diluted with water (15 mL), extracted with ethyl acetate (3×10 mL), and the organic phases were combined, washed with saturated brine (15 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the obtained crude product was isolated and purified by preparative thin layer chromatography (mobile phase: ethyl acetate/petroleum ether=1:10) to give compound 56f (30.0 mg, 44.3%) as a brown oil.
LC-MS:m/z 558.3[M+H] +
Step 6: preparation of (R) -2- ((2-amino-5-fluoropyrido [3,4-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (56)
Compound 56f (30 mg,0.057 mmol) was dissolved in dichloromethane (3 mL) at room temperature. Trifluoroacetic acid (1 mL) was added to the reaction mixture, and the mixture was stirred at room temperature for 3 hours. After completion of the reaction, the reaction mixture was concentrated under reduced pressure, diluted with saturated sodium hydrogencarbonate solution (10 mL), extracted with ethyl acetate (3X 10 mL), and the organic phases were combined, washed with saturated brine (20 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product was purified by preparative chromatography (column: XBridge Shield RP OBD column, 5um,19 x 150mm; mobile phase A: water (10 mmol/L ammonium bicarbonate), mobile phase B: acetonitrile, flow rate: 25mL/min; gradient: 25% -45% acetonitrile in 8 min; detection wavelength: 254/220 nm) to give compound 56 (7.30 mg, 43.7%) as a white solid.
1 H NMR (400 MHz, methanol-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] +
Example 57: preparation of (R) -2- ((2-amino-6-fluoropyrido [3,4-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (57)
Step 1: preparation of 5-amino-2-fluoroisonicotinamide (57 b)
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℃for 1 hour. After the completion of the reaction, the reaction mixture was concentrated under reduced pressure, the thus-obtained brown oily liquid compound was dissolved in tetrahydrofuran (5 mL), and the mixture was slowly added dropwise to aqueous ammonia (15 mL) at 0℃and the reaction mixture was stirred at 0℃for 1 hour. After the completion of the reaction, water was added to dilute the reaction, ethyl acetate (3×50 mL) was added to the system to extract, and the combined organic phases were washed with saturated brine (100 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the obtained crude product was separated and purified by silica gel column chromatography (mobile phase: ethyl acetate/petroleum ether=1:1) to give crude compound 57b (960 mg, 64.4%) as a yellow solid.
LC-MS:m/z 156.0[M+H] +
The remaining procedure was as in example 56, except that 3-amino-5-fluoroisonicotinamide (56 b) was replaced with 5-amino-2-fluoroisonicotinamide (57 b), to obtain compound 57.
1 H NMR (300 MHz, methanol-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] +
Example 58: preparation of (R) -2- ((2-amino-7-fluoropyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (58)
Step 1: preparation of 2-fluoro-5-iodo-4-aminopyridine (58 b) and 2-fluoro-3-iodo-4-aminopyridine (58 c)
The compound 2-fluoro-4-aminopyridine 58a (3.0 g,26.8 mmol) was dissolved in ethanol (100 mL) at room temperature, iodine (6.79 g,26.8 mmol) and silver sulfate (8.34 g,26.8 mmol) were added in this order, and the mixture was stirred at room temperature for 16 hours. After the reaction is finished, filtering, and directly decompressing and concentrating filtrate to obtain a crude product. The crude product was purified by column chromatography on silica gel (mobile phase: ethyl acetate/petroleum ether=20% -40%) to give compound 58b (0.96 g, 15.0%) and compound 58c (3.10 g, 48.7%).
LC-MS:m/z 58b 238.9[M+H] + ;LC-MS:m/z 58c 238.9[M+H] +
Step 2: preparation of 4-amino-6-fluoro-nicotinic acid methyl ester (58 d)
Compound 58b (2.33 g,9.79 mmol) was dissolved in methanol (40 mL) at room temperature, and triethylamine (0.99 g,9.79 mmol) and [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride (0.72 g,0.98 mmol) were sequentially added and reacted at 100℃under 20 atm of carbon monoxide for 6 hours. After the reaction is finished, filtering, and directly decompressing and concentrating filtrate to obtain a crude product. The crude product was purified by column chromatography on silica gel (mobile phase: ethyl acetate/petroleum ether=20% -40%) to give compound 58d (543 mg, 32.6%) as a pale yellow solid.
LC-MS:m/z 171.0[M+H] +
The remaining procedure is as in example 56, except that 3-amino-5-fluoroisonicotinamide (56 b) is replaced with methyl 4-amino-6-fluoro-nicotinate (58 d), producing compound 58.
LC-MS:m/z 294.2[M+H] +
1 H NMR (300 MHz, methanol-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)。
Example 59: preparation of (R) -2- ((2-amino-5-fluoropyrido [4,3-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (59)
Step 1: preparation of 4-amino-6-fluoro-nicotinic acid methyl ester (59 a)
2-fluoro-3-iodo-4-aminopyridine 58c (3.10 g,13.0 mmol) was dissolved in methanol (60 mL) at room temperature, and triethylamine (1.32 g,13.0 mmol) and [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride (1.06 g,1.30 mmol) were sequentially added to react at 100℃for 6 hours under 20 atm of carbon monoxide. After the reaction is finished, filtering, and directly decompressing and concentrating filtrate to obtain a crude product. The crude product was purified by column chromatography on silica gel (mobile phase: ethyl acetate/petroleum ether=20% -40%) to give compound 59a (1.6 g, 72.2%) as a pale yellow solid.
LC-MS:m/z 171.0[M+H] +
The remaining procedure was as in example 56, except that 3-amino-5-fluoroisonicotinamide (56 b) was replaced with 4-amino-6-fluoro-nicotinic acid methyl ester (59 a), to give compound 59.
LC-MS:m/z 294.2[M+H] +
1 H NMR (300 MHz, methanol-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)。
Example 60: preparation of 2- ((2-amino-6- (4- (pyrrolidin-1-ylmethyl) benzyl) pyridin [3,4-d ] pyrimidin-4-yl) amino) hex-1-ol (60)
Step 1: preparation of methyl 5-amino-2-chloroisonicotinate (60 b)
5-amino-2-chloropyridine-4-carboxylic acid 60a (25 g,145 mmol) was dissolved in methanol (300 mL) at 0deg.C, and thionyl chloride (100 mL) was added dropwise to the above solution (keeping the internal temperature of the reaction system at 20-25deg.C), and after the completion of the dropwise addition, the temperature was raised to 70deg.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, filtered and the cake dried to give compound 60b (16 g, 59.1%) as a yellow solid.
LC-MS:m/z 187.0[M+H] +
Step 2: preparation of 2-amino-6-chloropyrido [3,4-d ] pyrimidin-4 (3H) -one (60 c)
In a 100mL glass bottle, 60b (3.0 g,16.1 mmol), chlorformamidine hydrochloride (3.7 g,32.2 mmol) and dimethyl sulfone (10 g) were successively added at room temperature, and the reaction mixture was heated to 150℃and stirred for 4 hours. After the reaction was completed, the reaction mixture was diluted with water (100 mL), filtered, and the obtained cake was rinsed with water for 2 times, and the obtained cake was dried to obtain a yellow solid compound 60c (3.7 g, crude product).
LC-MS:m/z 197.0[M+H] +
Step 3: preparation of 2- ((2-amino-6-chloropyridin [3,4-d ] pyrimidin-4-yl) amino) hex-1-ol (60 d)
Compound 60c (130 mg,0.66 mmol) and DL-2-amino-1-hexanol (155 mg,1.32 mmol) were dissolved in N, N-dimethylformamide (2 mL) at room temperature, after which a Kate condensing agent (351 mg,0.79 mmol) and 1, 8-diazabicyclo undec-7-ene (302 mg,1.98 mmol) were added sequentially to the system. The reaction solution was stirred at room temperature for 4 hours. After completion of the reaction, the reaction mixture was diluted with water (30 mL), extracted with ethyl acetate (3X 30 mL), and the organic phases were combined, washed with saturated brine (30 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The resulting crude product was purified by silica gel column chromatography (mobile phase: petroleum ether: ethyl acetate=1:1) to give compound 60d (120 mg, 61%) as a yellow oil.
LC-MS:m/z 296.1[M+H] +
Step 4: preparation of 2- ((2-amino-6- (4-chlorobenzyl) pyridin [3,4-d ] pyrimidin-4-yl) amino) hex-1-ol (60 e)
Compound 60d (120 mg,0.41 mmol) and 4-chlorobenzyl boronic acid pinacol ester (43 b) (512 mg,2.02 mmol) were dissolved in dioxane (2 mL) and water (0.4 mL) at room temperature, and potassium carbonate (168 mg,1.21 mmol) and tetrakis (triphenylphosphine) palladium (93 mg,0.081 mmol) were then added sequentially to the system. The reaction solution was stirred under nitrogen at 95℃for 4 hours. After completion of the reaction, the reaction mixture was cooled to room temperature, diluted with water (30 mL), extracted with ethyl acetate (3×30 mL), and the organic phases were combined, washed with saturated brine (30 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The resulting crude product was purified by silica gel column chromatography (mobile phase: petroleum ether: ethyl acetate=1:1) to give compound 60e (60 mg, 38.3%) as a yellow oil.
LC-MS:m/z 386.2[M+H] +
Step 5: preparation of 2- ((2-amino-6- (4- (pyrrolidin-1-ylmethyl) benzyl) pyridin [3,4-d ] pyrimidin-4-yl) amino) hex-1-ol (60)
Compound 60e (60 mg,0.155 mmol) and potassium (1-pyrrolidinylmethyl) trifluoroborate (35 mg,0.18 mmol) were dissolved in dioxane (1 mL) and water (0.2 mL) at room temperature, and Xphos Pd G3 (9.9 mg,0.012 mmol) and potassium carbonate (64 mg,0.46 mmol) were then added sequentially to the system. The reaction solution was stirred under nitrogen at 100℃for 4 hours. After the completion of the reaction, the reaction mixture was cooled to room temperature, diluted with water (30 mL), extracted with ethyl acetate (3X 30 mL), and the organic phases were combined, washed with saturated brine (30 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure, and the crude product obtained was purified by separation using preparative chromatography (column: XBridge Shield RP OBD column, 5um, 19X 150mm; mobile phase A: water (0.1% formic acid), mobile phase B: acetonitrile; flow rate: 25mL/min; gradient: 6% -18% acetonitrile within 8 minutes; detection wavelength: 220 nm) to give off-white solid compound 60 (19.5 mg, 26.4%).
1 H NMR (400 MHz, methanol-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] +
Example 61: preparation of (R) -2- ((2-amino-6- (4- (pyrrolidin-1-ylmethyl) benzyl) pyridin [3,4-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (61)
Step 1: (R) -N 4 - (1- ((tert-butyldimethylsilyl) oxy) -2-methylhex-2-yl) -6-chloropyrido [3,4-d]Preparation of pyrimidine-2, 4-diamine (61 a)
Compound 60c (300 mg,1.52 mmol) was dissolved in N, N-dimethylformamide (2 mL) with (R) -1- ((tert-butyldimethylsilyl) oxy) -2-methylhex-2-amine (1 e) (749 mg,3.05 mmol) and then the catter condensing agent (720 mg,1.83 mmol) and 1, 8-diazabicycloundec-7-ene (697 mg,4.57 mmol) were added sequentially to the system at room temperature. The reaction was stirred at room temperature for 4 hours. After completion of the reaction, the reaction mixture was diluted with water (30 mL), extracted with ethyl acetate (3X 30 mL), and the organic phases were combined, washed with saturated brine (30 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The resulting crude product was purified by silica gel column chromatography (mobile phase: petroleum ether: ethyl acetate=1:1) to give compound 61a (102 mg, 76%) as a yellow oil.
LC-MS:m/z 424.2[M+H] +
Step 2: (R) -N 4 - (1- ((tert-butyldimethylsilyl) oxy) -2-methylhex-2-yl) -6- (4-chlorobenzyl) pyrido [3,4-d ]Preparation of pyrimidine-2, 4-diamine (61 b)
Compound 61a (100 mg,0.24 mmol) and 4-chlorobenzyl boronic acid pinacol ester (43 b) (294 mg,1.17 mmol) were dissolved in dioxane (2 mL) and water (0.4 mL) at room temperature, and tetrakis (triphenylphosphine) palladium (54 mg,0.04 mmol) and potassium carbonate (97 mg,0.7 mmol) were then added sequentially to the system. The reaction solution was stirred under nitrogen at 95℃for 4 hours. After completion of the reaction, the reaction mixture was cooled to room temperature, diluted with water (30 mL), extracted with ethyl acetate (3×30 mL), and the organic phases were combined, washed with saturated brine (30 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The resulting crude product was purified by silica gel column chromatography (mobile phase: petroleum ether: ethyl acetate=1:1) to give compound 61b (62 mg, 51%) as a yellow oil.
LC-MS:m/z 514.3[M+H] +
Step 3: (R) -N 4 - (1- ((tert-butyldimethylsilyl) oxy) -2-methylhex-2-yl) -6- (4- (pyrrolidin-1-ylmethyl) benzyl) pyrido [3,4-d]Preparation of pyrimidine-2, 4-diamine (61 c)
Compound 61b (60 mg,0.12 mmol) and potassium (1-pyrrolidinylmethyl) trifluoroborate (26 mg,0.14 mmol) were dissolved in dioxane (1 mL) and water (0.2 mL) at room temperature, then potassium carbonate (48 mg, 0.35 mmol) and Xphos Pd G3 (7.4 mg,0.009 mmol) were added sequentially to the system. The reaction solution was stirred under nitrogen at 100℃for 4 hours. After completion of the reaction, the reaction mixture was cooled to room temperature, diluted with water (30 mL), extracted with ethyl acetate (3×30 mL), and the organic phases were combined, washed with saturated brine (30 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The resulting crude product was purified by silica gel column chromatography (mobile phase: petroleum ether: ethyl acetate=2:1) to give compound 61c (8 mg, 12%) as a yellow oil.
LC-MS:m/z 563.4[M+H] +
Step 4: preparation of (R) -2- ((2-amino-6- (4- (pyrrolidin-1-ylmethyl) benzyl) pyridin [3,4-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (61)
Compound 61c (8 mg,0.014 mmol) was dissolved in dichloromethane (1.0 mL) at room temperature. Trifluoroacetic acid (0.5 mL) was added to the reaction mixture, and the mixture was stirred at room temperature for 1 hour. After completion of the reaction, the reaction mixture was concentrated under reduced pressure, diluted with saturated sodium hydrogencarbonate solution (10 mL), extracted with ethyl acetate (3X 10 mL), and the organic phases were combined, washed with saturated brine (20 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product was purified by preparative chromatography (column: XBridge Shield RP OBD column, 5um,19 x 150mm; mobile phase A: water (0.05% ammonia), mobile phase B: acetonitrile; flow rate: 25mL/min; gradient: 30% -65% acetonitrile in 8 min; detection wavelength: 220 nm) to give compound 61 (1.2 mg, 8.3%) as a white solid.
1 H NMR (400 MHz, methanol-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] +
Example 62: preparation of (R) -2- ((2-amino-6- (4- ((dimethylamino) methyl) benzyl) pyridin [3,4-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (62)
Step 1: preparation of (R) -2- ((2-amino-6-chloropyridin [3,4-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (62 a)
Compound 60c (300 mg,1.56 mmol), (R) -2-amino-2-methylhex-1-ol (300 mg,2.27 mmol), a carbomer (81mg, 1.83 mmol), 1, 8-diazabicycloundec-7-ene (697 mg,4.59 mmol) was dissolved in N, N-dimethylformamide (5 mL) at room temperature. The reaction solution was stirred at room temperature for 12 hours. After the completion of the reaction, the reaction mixture was diluted with water (20 mL), ethyl acetate (3×30 mL) was added to the reaction mixture, 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, and the obtained yellow crude product was separated and purified by silica gel column chromatography (mobile phase: methanol/dichloromethane/triethylamine=1/12/0.1) to give a yellow solid compound 62a (300 mg, 62.03%).
LC-MS:m/z 310.1[M+H] +
Step 2: preparation of (R) -2- ((2-amino-6- (4-chlorobenzyl) pyridin [3,4-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (62 b)
Compound 62a (150 mg,0.48 mmol), 4-chlorobenzyl boronic acid pinacol ester (43 b) (248 mg,0.97 mmol), potassium carbonate (201 mg,1.45 mmol), tetrakis (triphenylphosphine) palladium were dissolved in 1, 4-dioxane (2 mL) and water (0.5 mL) at room temperature. The reaction solution was stirred under nitrogen at 95℃for 12 hours. After the completion of the reaction, the reaction mixture was diluted with water (20 mL), ethyl acetate (3×30 mL) was added to the reaction mixture, 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, and the obtained yellow crude product was separated and purified by silica gel column chromatography (mobile phase: methanol/dichloromethane/triethylamine=1/13/0.1) to give a yellow solid compound 62b (30 mg, 15.53%).
LC-MS:m/z 400.2[M+H] +
Step 3: preparation of (R) -2- ((2-amino-6- (4- ((dimethylamino) methyl) benzyl) pyridin [3,4-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (62)
Compound 62b (20 mg,0.05 mmol), (dimethylaminomethyl) potassium trifluoroborate (165 mg,0.08 mmol), cesium carbonate (49 mg,0.15 mmol), 2-dicyclohexylphosphorus-2, 4, 6-triisopropylbiphenyl (5 mg,0.01 mmol), palladium acetate (1 mg,0.005 mmol) was dissolved in 1, 4-dioxane (1 mL) and water (0.2 mL) at room temperature. The reaction solution was replaced with nitrogen three times and stirred at 100℃for 12 hours. After the reaction, water (20 mL) was added to dilute the reaction, ethyl acetate (3X 30 mL) was added to the system to extract, 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, and the crude product obtained was purified by preparative chromatography (column type: XBridge Shield RP OBD column, 10um, 19X 250mm; mobile phase A: water (0.05% ammonia water), mobile phase B: acetonitrile; flow rate: 25mL/min; gradient: 30% -55% acetonitrile in 8 minutes; detection wavelength: 254/220 nm). The product was collected and lyophilized to give compound 62 (1.1 mg, 5.21%) as a white oil.
1 H NMR (400 MHz, methanol-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] +
Example 63: preparation of (R) -2- ((2-amino-6-methylpyridin [3,4-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (63)
Compound 62a (50.0 mg,0.161 mmol), trimethylcyclotriboroxane (50 wt% in THF, 60.78mg,0.242 mmol), cesium carbonate (157.76 mg, 0.284 mmol), tetrakis (triphenylphosphine) palladium (37.30 mg,0.032 mmol) and water (0.2 mL) were dissolved in 1, 4-dioxane at room temperature. The reaction solution was stirred at 100℃for 12 hours under nitrogen atmosphere. After the reaction, the reaction mixture was concentrated under reduced pressure, and the residue was purified by preparative thin layer chromatography (mobile phase: ethyl acetate/methanol=30:1), and the crude product obtained was further purified by preparative chromatography (column: XSelect CSH Prep C OBD column, 5um,19 x 150mm; mobile phase a: water (0.1% formic acid), mobile phase B: acetonitrile; flow rate: 25mL/min; gradient: 10% -22% acetonitrile in 8 minutes; detection wavelength: 254/220 nm). The product was collected and lyophilized to give formate salt of compound 63 as a yellow-green solid (13.8 mg, 25.49%).
1 H NMR (400 MHz, methanol-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] +
Example 64: preparation of (R) -2- ((2-amino-6-ethylpyridin [3,4-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (64)
Compound 64 was produced in the same manner as in example 63, except that the compound trimethylboroxine was replaced with ethylboric acid.
1 H NMR (300 MHz, methanol-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] +
Example 65: preparation of (R) -2- ((2-amino-6- (dimethylamino) pyridin [3,4-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (65)
Compound 62a (40 mg,0.129 mmol), dimethylamine hydrochloride (15.79 mg,0.194 mmol), cesium carbonate (168.27 mg,0.516 mmol), pd-PEPSI-IPent were reacted at room temperature Cl O-methylpyridine (CAS 1612891-29-8, 10.85mg,0.013 mmol) was dissolved in 1, 4-dioxane (1.5 mL). The reaction solution was stirred under nitrogen at 90℃for 12 hours. After completion of the reaction, the reaction mixture was diluted with water (20 mL), extracted with ethyl acetate (3X 20 mL), and the organic phases were combined, washed with saturated brine (40 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product obtained is separated and purified by a preparative chromatographic column (column type: XB ridge Prep C18 OBD column, 5um,19 x 150mm; mobile phase A: water (10 mmol/L ammonium bicarbonate), mobile phase B: acetonitrile; flow rate: 25mL/min; gradient: 25% -40% acetonitrile in 8 min; detection wavelength: 254/220 nm). The product was collected and lyophilized to give compound 65 (2.3 mg, 5.43%) as a yellow-green solid.
1 H NMR (400 MHz, methanol-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] +
Examples 66 and 67: preparation of (R) -2- ((2-aminopyrimido [5,4-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (66) and (R) -2- ((6-aminopyrimido [5,4-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (67)
Step 1: preparation of (R) -N- ((1- ((tert-butyldimethylsilyl) oxy) -2-methylhex-2-yl) -2,6, 8-trichloropyrimido [5,4-d ] pyrimidin-4-amine (66 b)
The compound 2,4,6, 8-tetrachloropyrimido [5,4-d ] pyrimidine 66a (700 mg,2.59 mmol) was dissolved in anhydrous tetrahydrofuran (5 mL) at room temperature, and after cooling to-78 ℃, N-diisopropylethylamine (402 mg,3.11 mmol), (R) -1- ((tert-butyldimethylsilyl) oxy) -2-methylhex-2-amine (1 e) (637 mg,2.59 mmol) was added in sequence and the reaction stirred under nitrogen at-78℃for 1 hour. After the completion of the reaction, the reaction mixture was quenched with water (20 mL), ethyl acetate (3X 30 mL) was added to the reaction mixture, the combined organic phases were washed with saturated brine (50 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give crude 66b as a pale yellow solid (510 mg, 41.1%).
LC-MS:m/z 478.1[M+H] +
Step 2: preparation of (R) -N- ((1- ((tert-butyldimethylsilyl) oxy) -2-methylhex-2-yl) -2, 6-dichloropyrimido [5,4-d ] pyrimidin-4-amine (66 c)
Compound 66b (480 mg,1.00 mmol) was dissolved in methanol (5 mL) at room temperature, followed by the addition of 10% palladium on carbon (48 mg, water content 10%). The reaction system was stirred at room temperature for 1 hour under a hydrogen atmosphere. After the completion of the reaction, the reaction solution was concentrated by filtration to give compound 66c (300 mg, 67.3%) as an off-white solid.
LC-MS:m/z 444.2[M+H] +
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-diamines (66 d) and (R) -N 8 - (1- ((tert-butyl)Phenyl dimethyl silyl) oxy) -2-methyl hex-2-yl) -6-chloro-N 2 - (2, 4-dimethoxybenzyl) pyrimido [5,4-d]Preparation of pyrimidine-2, 8-diamine (67 a)
Compound 66c (300 mg,0.675 mmol) was dissolved in 1, 4-dioxane (3 mL) at room temperature, N-diisopropylethylamine (174 mg,1.35 mmol) and 2, 4-dimethoxybenzylamine (113 mg,0.675 mmol) were added sequentially. The reaction system was stirred at 100℃for 3 hours. After the completion of the reaction, the reaction mixture was quenched with water (20 mL), extracted with ethyl acetate (3X 20 mL), and the combined organic phases were dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product was purified by column chromatography on silica gel (mobile phase: ethyl acetate/petroleum ether=20% -40%) to give a mixture of 66d and 67a as a yellow oily liquid (80 mg, 21.8%).
LC-MS:m/z 575.3[M+H] +
Step 4: (R) -N 4 - (1- ((tert-butyldimethylsilyl) oxy) -2-methylhex-2-yl) -N 2 - (2, 4-dimethoxybenzyl) pyrimido [5,4-d]Pyrimidine-2, 4-diamines (66 e) and (R) -N 8 - (1- ((tert-butyldimethylsilyl) oxy) -2-methylhex-2-yl) -N 2 - (2, 4-dimethoxybenzyl) pyrimido [5,4-d]Preparation of pyrimidine-2, 8-diamine (67 b)
A mixture of compounds 66d and 67a (80.0 mg,0.148 mmol) was dissolved in methanol (3 mL) at room temperature. 10% Palladium on carbon (8 mg, moisture content 10%), ammonium formate (28.0 mg,0.444 mmol) was added. The reaction solution was stirred at room temperature for 1 hour. After completion of the reaction, the reaction was quenched with water (10 mL), ethyl acetate (3X 10 mL) was added to the system, the combined organic phases were dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give a mixture of crude products 66e and 67b as a yellow semisolid (40.0 mg, 53.3%).
LC-MS:m/z 541.3[M+H] +
Step 5: preparation of (R) -2- ((2-aminopyrimido [5,4-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (66) and (R) -2- ((6-aminopyrimido [5,4-d ] pyrimidin-4-yl) amino) -2-methylhex-1-ol (67)
A mixture of compounds 66e and 67b (40 mg,0.0787 mmol) was dissolved in trifluoroacetic acid (1 mL) at room temperature. The reaction solution was stirred at 40℃for 4 hours. After the reaction was completed, the crude product was obtained by concentrating under reduced pressure, and purified by preparative chromatography (column: XSelect CSH Prep C OBD column, 5um,19 x 150mm; mobile phase A: water (0.1% formic acid), mobile phase B: acetonitrile; flow rate: 25mL/min; gradient: 10% -30% acetonitrile in 8 minutes; detection wavelength: 254/220 nm), to obtain white solid 66 (1.0 mg, 4.9%) and white solid 67 (5.6 mg, 27.4%).
Compound 66:
1 h NMR (400 MHz, methanol-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] +
Compound 67:
1 h NMR (400 MHz, methanol-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] +
Example 68: preparation of (R) -2- ((2-aminopyrido [3,4-d ] pyrimidin-4-yl) amino) -2-methylhex-1, 1-dideutero-1-ol (68)
Step 1: preparation of (R) -3-methyl-5-phenyl-5, 6-dihydro-2H-1, 4-oxazin-2-one (68 a)
Compound (R) -2-amino-2-phenylethanol (5 g,36.44 mmol) and ethyl pyruvate (4.23 g,36.44 mmol) were dissolved in trifluoroethanol (110 mL) at room temperature and the temperature was raised to 75℃for 16 hours. After the reaction was completed, the mixture was cooled to room temperature, filtered, and the cake was washed with ethyl acetate (3X 30 mL). The filtrate was concentrated under reduced pressure, and the resulting yellow crude product was purified by silica gel column chromatography (mobile phase: ethyl acetate/petroleum ether=0-10%) to give 68a (2.4 g, 34.8%) as a white solid.
LC-MS:m/z 190.1[M+H] +
Step 2: preparation of (3R, 5R) -3-butyl-3-methyl-5-phenylmorpholin-2-one (68 b)
Compound 68a (2.4 g,12.68 mmol) was dissolved in dry tetrahydrofuran (80 mL) at room temperature, the reaction solution was cooled to-78deg.C, and boron trifluoride etherate (3.3 mL) was slowly added dropwise under nitrogen atmosphere and the reaction was maintained at-78deg.C for 1.5 hours. Then, a tetrahydrofuran solution (13.5 mL,2 mol/L) of n-butylmagnesium chloride was slowly added dropwise thereto, and the reaction was continued at-78℃for 2 hours. After the completion of the reaction, the reaction was slowly warmed to room temperature, quenched with an aqueous ammonium chloride solution, extracted with ethyl acetate (3×50 mL), and the combined organic phases were washed with saturated brine (120 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure, and the resulting crude brown oil was separated and purified by silica gel column chromatography (mobile phase: ethyl acetate/petroleum ether=0 to 5%) to give 68b (1.35 g, 43.0%) as a white solid.
LC-MS:m/z 248.2[M+H] +
Step 3: preparation of (R) -2- (((R) -2-hydroxy-1-phenylethyl) amino) -2-methylhex-1, 1-dideugenol-1-ol (68 c)
Compound 68b (500 mg,2.02 mmol) was dissolved in dry tetrahydrofuran (5 mL) at room temperature, the reaction was placed at 0deg.C and lithium deuterated aluminum tetrahydride (169.7 mg,4.04 mmol) was added. The reaction solution was stirred at room temperature for 2 hours. After the completion of the reaction, the reaction was quenched with water in an ice bath, ethyl acetate (3×6 mL) was added to the system to extract, and the combined organic phases were washed with saturated brine (15 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a pale yellow solid 68c (360 mg, 70.3%).
LC-MS:m/z 254.3[M+H] +
Step 4: preparation of (R) -2-amino-2-methylhex-1, 1-dideutero-1-ol hydrochloride (68 d)
Compound 68c (360 mg,1.42 mmol) was dissolved in absolute ethanol (5 mL) at room temperature, and palladium on carbon hydroxide (144 mg,0.42 mmol) and hydrogen chloride dioxane solution (0.5 mL,4 mol/L) were added. The reaction solution was reacted under a hydrogen atmosphere (5 atm) at 70℃for 16 hours. After the reaction was completed, the mixture was cooled to room temperature, filtered, and the cake was washed with ethanol (3X 3 mL). Concentrating the filtrate under reduced pressure to obtain yellow crude product. Water (10 mL) was added and ethyl acetate (3X 8 mL) was added to the system for extraction, and the aqueous phase was concentrated under reduced pressure to give product 68d (185 mg, 76.7%) as a yellow oil
LC-MS:m/z 134.0[M+H] +
Step 5: preparation of (R) -2- ((2-aminopyrido [3,4-d ] pyrimidin-4-yl) amino) -2-methylhex-1, 1-dideutero-1-ol (68)
Compound 68d (90 mg,0.67 mmol) and 2-aminopyridine [3,4-d ] pyrimidin-4 (3H) -one (44 b) (100 mg,0.62 mmol) were dissolved in N, N-dimethylformamide (2 mL) at room temperature, and a catter condensing agent (327 mg,0.74 mmol) and 1, 8-diazabicyclo undec-7-ene (282 mg,1.85 mmol) were added and reacted at room temperature for 16 hours. After completion of the reaction, the reaction mixture was diluted with water (10 mL), extracted with ethyl acetate (3X 8 mL), and the organic phases were combined, washed with saturated brine (3X 15 mL), and dried over anhydrous sodium sulfate. The organic phase was concentrated under reduced pressure and the crude product obtained was purified by preparative chromatography (Column: XBridge Shield RP OBD Column,5um,19 x 150mm; mobile phase A: water (0.05% ammonia), mobile phase B: acetonitrile; flow rate: 25mL/min; gradient: 25% -40% acetonitrile in 8 min; detection wavelength: 254/220 nm) to give a white solid product 68 (13.6 mg, 7.8%).
1 H NMR (300 MHz, methanol-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] +
Example 69: preparation of (R) -2- ((2-aminopyrido [3,4-d ] pyrimidin-4-yl) amino) -2-ethylhexyl-1-ol (69)
Step 1: preparation of (R) -3-ethyl-5-phenyl-5, 6-dihydro-2H-1, 4-oxazin-2-one (69 a)
Compound (R) -2-amino-2-phenylethanol (4.11 g,29.96 mmol) and methyl 2-oxobutyrate (3.48 g,29.96 mmol) were dissolved in trifluoroethanol (90 mL) at room temperature, and the temperature was raised to 75℃for 16 hours. After the reaction was completed, the mixture was cooled to room temperature, filtered, and the cake was washed with ethyl acetate (3X 30 mL). The filtrate was concentrated under reduced pressure, and the resulting yellow crude product was purified by silica gel column chromatography (mobile phase: ethyl acetate/petroleum ether=0 to 10%) to give 69a (2.3 g, 37.8%) as a white solid.
LC-MS:m/z 204.2[M+H] +
Step 2: preparation of (3R, 5R) -3-butyl-3-ethyl-5-phenylmorpholin-2-one (69 b)
Compound 69a (2.3 g,11.32 mmol) was dissolved in dry tetrahydrofuran (75 mL) at room temperature, the reaction solution was cooled to-78deg.C, and boron trifluoride etherate (3.0 mL) was slowly added dropwise under nitrogen atmosphere and the reaction was maintained at-78deg.C for 1.5 hours. Then, a tetrahydrofuran solution (11.9 mL,2 mol/L) of n-butylmagnesium chloride was slowly added dropwise thereto, and the reaction was continued at-78℃for 2 hours. After the completion of the reaction, the reaction was slowly warmed to room temperature, quenched with an aqueous ammonium chloride solution, extracted with ethyl acetate (3×50 mL), and the combined organic phases were washed with saturated brine (120 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure, and the obtained crude brown oil was separated and purified by silica gel column chromatography (mobile phase: ethyl acetate/petroleum ether=0 to 5%) to give 69b (1.23 g, 41.6%) as a white solid.
LC-MS:m/z 262.3[M+H] +
Step 3: preparation of (R) -2-ethyl-2- (((R) -2-hydroxy-1-phenylethyl) amino) hex-1-ol (69 c)
Compound 69b (1.23 g,4.71 mmol) was dissolved in dry tetrahydrofuran (10 mL) at room temperature, the reaction was placed at 0deg.C, and a solution of lithium borohydride in tetrahydrofuran (4.7 mL,2 mol/L) was added. The reaction solution was reacted at room temperature for 2 hours. After completion of the reaction, the reaction was quenched with water in an ice bath, ethyl acetate (3×12 mL) was added to the system to extract, and 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 to give 69c (1.1 g, 88.1%) as a pale yellow solid.
LC-MS:m/z 266.2[M+H] +
Step 4: preparation of (R) -2-amino-2-ethylhexyl-1-hydrochloride (69 d)
Compound 69c (1.1 g,4.15 mmol) was dissolved in absolute ethanol (20 mL) at room temperature, and palladium on carbon hydroxide (426 mg,1.23 mmol) and hydrogen chloride in dioxane (1.5 mL,4 mol/L) were added. The reaction solution was reacted under a hydrogen atmosphere (5 atm) at 70℃for 16 hours. After the reaction was completed, the mixture was cooled to room temperature, filtered, and the cake was washed with ethanol (3X 8 mL). Concentrating the filtrate under reduced pressure to obtain yellow crude product. Water (30 mL) was added, and ethyl acetate (3X 25 mL) was added to the system for extraction, and the aqueous phase was concentrated under reduced pressure to give 69d (750 mg, 99.6%) as a yellow oil
LC-MS:m/z 146.0[M+H] +
Step 5: preparation of (R) -2- ((2-aminopyrido [3,4-d ] pyrimidin-4-yl) amino) -2-ethylhexyl-1-ol (69)
Compound 69d (202 mg,1.39 mmol) and 2-aminopyridine [3,4-d ] pyrimidin-4 (3H) -one (44 b) (150 mg,0.93 mmol) were dissolved in N, N-dimethylformamide (3 mL) at room temperature, and a catter condensing agent (491 mg,1.11 mmol) and 1, 8-diazabicycloundec-7-ene (428 mg,2.78 mmol) were added and reacted at room temperature for 16 hours. After completion of the reaction, the reaction mixture was diluted with water (10 mL), extracted with ethyl acetate (3X 8 mL), and the organic phases were combined, washed with saturated brine (3X 15 mL), and dried over anhydrous sodium sulfate. The organic phase was concentrated under reduced pressure and the crude product obtained was purified by preparative chromatography (Column: XBridge Shield RP OBD Column,5um,19 x 150mm; mobile phase A: water (0.05% ammonia), mobile phase B: acetonitrile; flow rate: 25mL/min; gradient: 25% -50% acetonitrile in 8 min; detection wavelength: 254/220 nm) to give white solid product 69 (16.3 mg, 6.0%).
1 H NMR (300 MHz, methanol-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] +
Biological assay
Test example 1: agonistic activity of the compounds of the invention on hTLR8 and hTLR7
In vitro analysis of receptor binding Activity of the Compounds of the invention on hTLR8 and hTLR7 HEK-Blue purchased from Invivogen Co TM hTLR8 cells and HEK-Blue TM hTLR7 cells. The cell is a reporter gene for a secreted alkaline phosphatase (SEAP) co-transfected with the hTLR8 or hTLR7 gene in HEK293 cells. Wherein the gene for SEAP is placed downstream of the IFN- β minimal promoter, which consists of 5 NF-. Kappa.B and AP-1 binding sites. The effect of compounds was evaluated by detecting SEAP levels by activating NF- κb and AP-1 promoters with a stimulator of hTLR8 or hTLR7 to produce SEAP.
Test reagent:
HEK-Blue hTLR8 cells and HEK-Blue hTLR7 cells (from Invivogen Co.)
HEK-BlueTM assay reagent (from Invivogen Co.)
DMEM medium (from Gibco company)
Fetal bovine serum (from Gibco company)
Normocin TM Zeocin and blasticidin (Blasticidine) (from Invivogen Corp.)
The test process comprises the following steps:
1. collecting cells in the cell culture flask, and adjusting cell density to 2.2X10 5 Per mL, cells were resuspended with HEK-BlueTM assay reagent and 45. Mu.L of cell suspension was seeded into 384 well plates 10000 cells per well.
2. Compound plate preparation: test compounds were diluted 3-fold with DMSO from 2mM for 10 gradients. mu.L of diluted compound was added to 38. Mu.L of HEK-blue detection reagent for 20-fold intermediate dilution. Cells were plated in 0.5% dmso wells as negative control wells for low reading. Cells were added to 1. Mu.M GS-9688 (see WO2016141092A1 synthetic route) wells as positive control wells for high read values.
3. 5. Mu.L of the intermediate diluted compound was added to 384-well plates, which had been inoculated with 45. Mu.L of cells, and the drug was 10-fold diluted to a final DMSO concentration of 0.5%.
4. Placing 384-well plate containing cells and compound into 37deg.C, 5% CO 2 Is cultured for 16 hours.
5. After 16 hours, the plates were removed and SEAP light absorption at 620nm was measured using the instrument VICTOR Nivo.
6. Analysis of data using GraphPad Prism 8 software gave EC for each compound 50
The average of the data for each concentration and positive and negative controls was determined. The percent activity was calculated from the formula:
activity% = (compound reading-negative well reading)/(positive well reading-negative well reading) ×100.
Calculation of IC for each compound by fitting data to a nonlinear regression equation 50
Y=minimum + (maximum-minimum)/(1+10 ((LogEC) 50 -X) X hill slope);
wherein X is the logarithm of the compound concentration and Y is the percentage of activity.
The agonist activity of the compounds of the invention on TLR8/TLR7 is shown in table 1 below.
TABLE 1 agonism of compounds of the invention for TLR8/TLR7 EC 50 Value of
Examples 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
Conclusion: the compounds of the invention are capable of selectively activating TLR8.
Test example 2: study of the absorption mechanism of the Compound of example 52
This experiment examined the possible permeation and absorption of the test compounds by measuring their permeability coefficient in the Caco-2 cell model, using LC/MS/MS to measure the concentration of the compounds in the test sample, and calculating the apparent permeability coefficient (Papp) of the test compounds in the Caco-2 cell membrane. Caco-2 cells were purchased from the American type culture Collection,no. HTB-37, after about 14 days of culture, was completely pooled and differentiation was completed. The test is bi-directional administration, i.e., the simultaneous detection of 1) the rate of compound transport from the apical to the basal end, 2) the rate of compound transport from the basal end to the apical is determined.
The main reagent information for the parts involved in the experiments are shown in the following table:
1. caco-2 cell preparation
Caco-2 cells (American type culture Collection, HTB-37) were diluted to 6.86X 10 with medium 5 Each cell/mL, and 50. Mu.L of the cell suspension was added to the filter wells of a 96-well Transwell plate (Cat. No. 3391). Cell culture plates were incubated at 37℃with 5% CO 2 Culturing in a cell culture incubator with 95% relative humidity for 14-18 days. Cell culture medium was changed every other day.
2. Cell monolayer stability assessment
Old media was removed prior to evaluation and replaced with pre-warmed fresh media. Transepithelial resistance (TEER) across monolayers was measured using a Millicell Epithelial Volt-Ohm measurement system (Millipore, USA). After the measurement is completed, the plate is returned to the incubator.
TEER values were calculated according to the following formula:
TEER measurement (ohms) x membrane area (cm) 2 ) TEER value (ohm cm) 2 )
Wherein the TEER value is greater than 230ohm cm 2 The Caco-2 monolayer film is qualified and can be used for subsequent tests.
3. Preparation of compound solutions
The compound of example 52 was accurately weighed, dissolved in DMSO to prepare a stock solution at a concentration of 2mM, and diluted with HBSS (Gibico, 10mM HEPES,pH 7.4) to obtain a 10. Mu.M working solution. Metoprolol and digoxin were used as control compounds.
4. Drug delivery test
The monolayers were washed twice with pre-heated HBSS (10mM HEPES,pH 7.4) prior to testing. The plates were then incubated at 37℃for 30 minutes.
(1) Determining the rate of drug transport from the apical to the basal lateral direction: 125 μ working solution was added to the Transwell (apical compartment) and immediately 50 μl samples were transferred from the apical compartment to 200 μl new 96-well plates containing IS (100 nM alprazolam, 200nM caffeine and 100nM tolbutamide) as initial donor samples (AB). Vortex at 1000rpm for 10 minutes. The wells in the receiver plate (basolateral compartment) were filled with 235 μl of buffer.
(2) Determining the rate of drug transport from the basolateral to the root tip: 285 μl of working solution was added to the receiver plate wells (basolateral compartments) and 50 μl of sample was transferred.
The plates were incubated at 37℃for 2 hours. At the end of incubation, 50 μl samples from the donor side (ap→apical compartment of bl→basolateral compartment of bl→ap) and the receiver side (ap→basolateral compartment of bl→apical compartment of bl→ap) were transferred to wells of a new 96-well plate, followed by 200 μl acetonitrile containing IS (100 nM alprazolam, 200nM caffeine and 100nM tolbutamide). Samples were vortexed for 10 minutes, 50 μl samples were transferred to wells of a new 96-well plate, and then 50 μl HEPRS and 200 μl IS were added. All samples were vortexed for 10 minutes and then centrifuged at 3,220g for 40 minutes. Before analysis, 150. Mu.L of the supernatant was mixed with an appropriate amount of ultrapure water and subjected to test analysis by LC-MS/MS.
Apparent permeability (P) was determined using the following equation app ):
Wherein P is app Is apparent permeability (cm/s.times.10) -6 ) dQ/dt is the rate of drug transport (pmol/sec), A is the surface area of the membrane (cm 2 ) D0 is the initial donor concentration (nM; pmol/cm 3 )
The outflow ratio is determined using the following equation:
wherein P is app(B-A) Represents apparent permeability coefficient, P, from the outside of the substrate to the root tip app(A-B) The apparent permeability coefficient in the direction from the root tip to the outer side of the substrate is shown.
The test results are shown in Table 2.
TABLE 2 Caco-2 test of example 52 compound and example 4 compound of WO2018045144A1
Test parameters
Parameters (parameters) Example 52 WO2018045144A1 (example 4)
P app(A-B) 19.2 2.72
P app(B-A) 31.1 30.0
Flow ratio 1.62 11.0
Recovery rate (%) < AP-BL- 107 97.2
Recovery rate (%) < BL-AP- 93.5 102
Conclusion: the compound of example 52 has a better permeability coefficient and a lower risk of efflux transporter substrates. The compound of example 4 of WO2018045144A1 has a lower permeability and a higher efflux ratio, likely to be an efflux transporter substrate. In comparison, the in vitro Caco-2 test results of example 52 for the compound of the present invention showed better permeability than the compound of example 4 of WO2018045144A 1.
Test example 3: rat pharmacokinetic study of the Compound of example 52
This experiment was designed to evaluate the pharmacokinetic behavior of the compound of example 52 following intravenous drip or intragastric administration in rats. Intravenous drip administration: the tested compound is prepared into a clear solution with the concentration of 0.5 mg/ml, and the solvent is 2% ethanol/40% polyethylene glycol 300/58%0.01 mol hydrochloric acid; gastric lavage administration: the test compound was formulated as a clear solution of 0.5 mg/ml with a vehicle of 2% ethanol/40% polyethylene glycol 300/58%0.01 mole hydrochloric acid.
The concentration of the test compound in the plasma was determined by high performance liquid chromatography-tandem mass spectrometry (LC-MS/MS). By WinNonlin TM Non-compartmental model of Version 8.3 (Pharsight, mountain View, CA) pharmacokinetic software plasma and tissue concentrations were processed and pharmacokinetic parameters were calculated using the linear log trapezium method.
The rat pharmacokinetic parameters for the compound of example 52 are shown in table 3 below.
TABLE 3 parameters related to the pharmacokinetics of intravenous drip and gastric lavage in rats of the compound of example 52
The parameters relevant to the study of the tissue distribution of the compound of example 52 in rats at a dose of 5mg/kg administered by gavage are shown in table 4 below.
TABLE 4 study of parameters related to the distribution of the gastric lavage tissue of the rats of the compound of example 52
The intestinal bioavailability and liver first pass study parameters of the compound of example 52 at 5mg/kg administered by gavage are shown in table 5 below.
Table 5 gastrointestinal bioavailability and liver first pass study parameters of the compound of example 52 in rats
The relevant parameters for the rat excretion study at a dose of 1mg/kg administered for 0.5 hour by intravenous infusion of the compound of example 52 are shown in Table 6 below.
Table 6 parameters relevant to the study of the intravenous drip excretion of the rat of the compound of example 52
Conclusion: the compound of example 52 has lower systemic oral bioavailability but higher intestinal bioavailability, and can specifically enrich liver tissue for excretion mainly through the intestinal tract.
Test example 4: cynomolgus monkey pharmacodynamics study of the Compounds of example 52
This test was intended to evaluate the pharmacodynamics of the compound of example 52 following oral administration in male cynomolgus monkeys.
Oral administration: the test compound is prepared into 10mg/mL, and the solvent is 10% ethanol, 40% polyethylene glycol 300 and 50% deionized water. An appropriate amount of the test compound is weighed into an appropriate amount of ethanol and the mixture is completely dissolved under stirring and/or ultrasound. An appropriate volume of polyethylene glycol 300 is then added with stirring. Finally, a suitable volume of deionized water is added with stirring to obtain the final concentration of the formulation. The formulation will be stirred at room temperature for at least 10 minutes prior to and during administration.
Each group was observed 2 times per day by 1 cynomolgus monkey (purchased from beijing xielxin biological resource) per sex. Before administration, 30min, 1hr, 2hr, 4hr, 8hr and 24hr after administration, and 0.3mL serum was collected for each animal sample. The biochemical indicators IL12p40, IFN-alpha and TNF-alpha concentrations in serum were determined by a hypersensitive multifactorial electrochemiluminescence analyzer.
The reagents and instrumentation involved in the test are shown in the following table:
vendor MSD full name: meso Scale Discovery.
The serum cytokine concentration test process is:
1) U-PLEX 96 microwells were pre-incubated with coating solution and incubated for 1 hour at room temperature.
2) The plate was washed 3 times with 300 μl 1x wash buffer.
3) 25 μl of Diluent (Diluent 43) was added to each well to be tested, and the sides of the plate were gently tapped to evenly distribute Diluent43 at the bottom of the MSD microplate.
4) mu.L of TNF-alpha, IL-12 or INF-alpha standard or 25. Mu.L of serum sample was added to each well and incubated with shaking at room temperature for 1 hour using a plate membrane seal.
5) Plates were washed 3 times with 300ul 1 Xwash buffer (PBS-0.05% Tween-20).
6) 50 μl of detection antibody solution was added to each well, membrane-sealed with a sealing plate, and incubated with shaking at room temperature for 1 hour.
7) The plate was washed 3 times with 300 μl 1x wash buffer.
8) Mu. L MSD GOLD Read Buffer B was added per well and the wells were read with MSD reader (model: MESO SECTOR S600) read U-PLEX 96 microwell plates.
9) Data were analyzed using GraphPad Prism 8 software to obtain the area under the 'cytokine concentration-time' curve (AUC).
FIG. 1 shows parameters of the compound of example 52 in cynomolgus monkey serum.
As can be seen from FIG. 1, the compound of example 52 had a significant activation of IL-12p40 in cynomolgus monkey serum but had little effect on TNF- α and INF- α following oral administration of 10 mg/kg.

Claims (20)

  1. A compound of formula (I) or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer or mixture thereof, or a pharmaceutically acceptable salt thereof,
    Wherein,
    X 1 is CR (CR) 1 Or N;
    X 2 is CR (CR) 2 Or N;
    X 3 is CR (CR) 3 Or N;
    X 4 is CR (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 1 Selected from the group consisting of hydrogen, halogen, amino, nitro, cyano, hydroxy, mercapto, carboxyl, alkyl, alkoxy, haloalkyl, haloalkoxy;
    R 2 selected from hydrogen, halogen, cyano, oxo, alkyl, alkenyl, alkynyl, -OR a 、-SR a 、-NR a R b Cycloalkyl, heterocyclyl, aryl and heteroarylWherein the alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl is optionally further substituted with one or more Q 1 Substituted with a group;
    R 3 selected from hydrogen, halogen, cyano, oxo, alkyl, alkenyl, alkynyl, -OR a 、-SR a 、-NR a R b Cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl is optionally further substituted with one or more Q 2 Substituted with a group;
    R 4 selected from the group consisting of hydrogen, halogen, amino, nitro, cyano, hydroxy, mercapto, carboxyl, alkyl, alkoxy, haloalkyl, haloalkoxy;
    R 5 and R is 6 Each independently selected from hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl; wherein the alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl is optionally further selected from deuterated, 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)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 Is substituted with one or more groups;
    Q 1 and Q 2 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 the group consisting of halogen, amino, nitro, cyano, carboxyl, ester, oxo, alkyl, alkoxy, 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)NR c R d 、-NR c C(O)R d 、-S(O) n R a 、-S(O) n NR c R d and-NR c S(O) n R d Is substituted with one or more groups;
    R a and R is b Each independently selected from the group consisting of hydrogen, halogen, hydroxy, nitro, cyano, oxo, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein the alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl is optionally further selected from the group consisting of 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)NR c R d 、-NR c C(O)R d 、-S(O) n R a 、-S(O) n NR c R d and-NR c S(O) n R d Is substituted with one or more groups;
    or R is a And R is b Together with the nitrogen atom to which they are attached, form a nitrogen-containing heterocyclic group optionally further containing one OR more heteroatoms selected from N, O, S in addition to N, said nitrogen-containing heterocyclic group optionally being further substituted with a moiety selected from halogen, nitro, cyano, oxo, carboxyl, ester, alkyl, haloalkyl, 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)NR c R d 、-NR c C(O)R d 、-S(O) n R a 、-S(O) n NR c R d and-NR c S(O) n R d Is substituted with one or more groups;
    R c and R is d Each independently selected from the group consisting of hydrogen, halogen, hydroxy, nitro, cyano, oxo, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein the alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl is optionally further substituted with one or more groups selected from the group consisting of halogen, amino, nitro, cyano, hydroxy, mercapto, carboxyl, ester, oxo, alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl, and heteroaryl;
    or R is c And R is d Together with the nitrogen atom to which they are attached, form a nitrogen-containing heterocyclic group optionally further containing one or more heteroatoms selected from N, O, S in addition to N, the nitrogen-containing heterocyclic group optionally being further substituted with a moiety selected from halogen, nitro, cyano, oxo, hydroxy, mercapto, carboxyl, ester, alkyl, alkoxy, cycloalkyl, heterocyclic, aryl and heteroaryl;
    n is 1 or 2;
    v is an integer from 1 to 6;
    t is 0 to 6.
  2. The compound of the formula (I) according to claim 1, which is a compound of the formula (II) or a stereoisomer, a tautomer, a meso, a racemate, an enantiomer, a diastereomer or a mixture thereof, or a pharmaceutically acceptable salt thereof,
    Wherein X is 1 、X 2 、X 3 、L、R 4 、R 5 、R 6 As defined in claim 1.
  3. The compound of the formula (I) according to claim 1, which is a compound of the formula (III) or a stereoisomer, a tautomer, a meso, a racemate, an enantiomer, a diastereomer or a mixture thereof, or a pharmaceutically acceptable salt thereof,
    therein, L, R 1 、R 3 、R 4 、R 5 、R 6 As defined in claim 1.
  4. The compound of the formula (I) according to claim 1, which is a compound of the formula (IV) or a stereoisomer, a tautomer, a meso, a racemate, an enantiomer, a diastereomer or a mixture thereof, or a pharmaceutically acceptable salt thereof,
    therein, L, R 1 、R 2 、R 4 、R 5 、R 6 As defined in claim 1.
  5. The compound of the formula (I) according to claim 1, which is a compound of the formula (V) or a stereoisomer, a tautomer, a meso, a racemate, an enantiomer, a diastereomer or a mixture thereof, or a pharmaceutically acceptable salt thereof,
    Therein, L, R 2 、R 4 、R 5 、R 6 As defined in claim 1.
  6. A compound of formula (I) according to any one of claims 1 to 5, or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein: l is selected from a bond or-C (O) -; preferably a key.
  7. A compound of the formula (I) according to claim 1 to 6, or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer or mixture thereof, or a pharmaceutically acceptable salt thereof, which is a compound of the formula (I-1), (II-1), (III-1), (IV-1), (V-1) or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer or mixture thereof, or a pharmaceutically acceptable salt thereof,
    wherein X is 1 、X 2 、X 3 、X 4 、R 1 、R 2 、R 3 、R 4 、R 5 、R 6 As defined in claim 1.
  8. A compound of the general formula (I) according to claim 1 to 3, 6 to 7, or a stereoisomer, a tautomer, a meso form, a racemate, an enantiomer, a diastereomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof,
    Wherein,
    R 3 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 optionally further substituted with one or more Q 2 Substituted with a group;
    Q 2 selected from halogen, C 1 -C 6 Alkyl, 4-6 membered heterocyclyl, 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 selected from halogen, C 1 -C 6 Alkyl, - (CH) 2 ) v -NR c R d Is substituted with one or more groups;
    R a and R is b Each independently selected from hydrogen, C 1 -C 6 An alkyl group;
    or R is a And R is b Together with the nitrogen atom to which they are attached, form a 4-6 membered nitrogen containing heterocyclic group, said 4-6 membered nitrogen containing heterocyclic group optionally further containing one or more heteroatoms selected from N, O, S in addition to N, said 4-6 membered nitrogen containing heterocyclic group optionally being 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 Is substituted with one or more groups;
    R c and R is d Each independently of the otherIs selected from hydrogen, C 1 -C 6 An alkyl group;
    or R is c And R is d Together with the nitrogen atom to which they are attached, form a 4-6 membered nitrogen containing heterocyclic group, said 4-6 membered nitrogen containing heterocyclic group optionally further containing one or more heteroatoms selected from N, O, S in addition to N, said 4-6 membered nitrogen containing heterocyclic group optionally being further selected from halogen, C 1 -C 6 One or more groups of the alkyl group are substituted;
    v is an integer from 1 to 6.
  9. The compound represented by the general formula (I) according to claim 8, or a stereoisomer, a tautomer, a meso, a racemate, an enantiomer, a diastereomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof,
    R 3 selected from C 6 -C 10 Aryl and 5 to 10 membered heteroaryl, preferably phenyl or 5-6 membered heteroaryl; wherein said C 6 -C 10 Aryl and 5 to 10 membered heteroaryl optionally further substituted with one or more Q 2 Substituted with a group;
    Q 2 selected from halogen, C 1 -C 6 Alkyl, 4-6 membered heterocyclyl, 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 selected from halogen, C 1 -C 6 One or more groups of the alkyl group are substituted;
    R a and R is b Each independently selected from hydrogen, C 1 -C 6 An alkyl group;
    or R is a And R is b Together with the nitrogen atom to which they are attached, form a 4-6 membered nitrogen containing heterocyclic group, said 4-6 membered nitrogen containing heterocyclic group optionally further containing one or more heteroatoms selected from N, O, S in addition to NA child, 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 Is substituted with one or more groups;
    R c and R is d Each independently selected from hydrogen, C 1 -C 6 An alkyl group;
    or R is c And R is d Together with the nitrogen atom to which they are attached, form a 4-6 membered nitrogen containing heterocyclic group, said 4-6 membered nitrogen containing heterocyclic group optionally further containing one or more heteroatoms selected from N, O, S in addition to N, said 4-6 membered nitrogen containing heterocyclic group optionally being further selected from halogen, C 1 -C 6 One or more groups of the alkyl group are substituted v is an integer from 1 to 6, preferably 1 or 2.
  10. A compound of the general formula (I) according to claim 1 to 2, 4 to 7, or a stereoisomer, a tautomer, a meso form, a racemate, an enantiomer, a diastereomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof,
    wherein R is 2 Selected from hydrogen, halogen, C 1 -C 6 Alkyl, -NR a R b The C is 1 -C 6 Alkyl is optionally further substituted with Q 1 Substitution;
    Q 1 selected from C 6 -C 10 Aryl, 5 to 10 membered heteroaryl, wherein the C is 6 -C 10 Aryl and 5 to 10 membered heteroaryl are optionally further selected from halogen, C 1 -C 6 Alkyl, - (CH) 2 ) v -NR c R d Is substituted with one or more groups;
    R a 、R b each independently selected from hydrogen, C 1 -C 6 An alkyl group;
    R c and R is d Each independently selected from hydrogen, C 1 -C 6 An alkyl group;
    or R is c And R is d Together with the nitrogen atom to which they are attached, form a 4-6 membered nitrogen containing heterocyclic group, said 4-6 membered nitrogen containing heterocyclic group optionally further containing one or more heteroatoms selected from N, O, S in addition to N, said 4-6 membered nitrogen containing heterocyclic group optionally being further selected from halogen, C 1 -C 6 One or more groups of the alkyl group are substituted;
    v is an integer from 1 to 6, preferably 1 or 2.
  11. A compound of the general formula (I) according to claim 1 to 10, or a stereoisomer, a tautomer, a meso form, a racemate, an enantiomer, a diastereomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof,
    wherein R is 1 Selected from hydrogen, halogen, C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy, C 1 -C 6 Haloalkyl, C 1 -C 6 Haloalkoxy groups; preferably, R 1 Is hydrogen or halogen.
  12. A compound of the general formula (I) according to claim 1 to 11, or a stereoisomer, a tautomer, a meso form, a racemate, an enantiomer, a diastereomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof,
    wherein R is 4 Selected from hydrogen, halogen, C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy, C 1 -C 6 Haloalkyl, C 1 -C 6 Haloalkoxy groups; preferably, R 4 Is hydrogen or halogen.
  13. A compound of the general formula (I) according to claim 1 to 12, or a stereoisomer, a tautomer, a meso form, a racemate, an enantiomer, a diastereomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof,
    wherein:
    R 5 and R is 6 Each independently selected from hydrogen and C 1 -C 12 Alkyl, said C 1 -C 12 The alkyl group is optionally further selected from deuterated, -OR a 、-SR a 、-NR a R b Is substituted with one or more groups;
    R a selected from hydrogen, C 1 -C 6 An alkyl group;
    R b selected from hydrogen, C 1 -C 6 Alkyl, C 3 -C 6 Cycloalkyl and 5 to 7 membered heterocyclyl;
    preferably, R 5 Is hydrogen, R 6 Is C 1 -C 12 Alkyl, said C 1 -C 12 The alkyl group is optionally further substituted with one or more groups selected from deuterated, -OH.
  14. A compound of formula (I) according to any one of claims 1 to 13, or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, selected from:
  15. a process for preparing a compound of formula (III-1) or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, comprising the steps of:
    the compound of formula A3 is coupled with boric acid or a pinacol borate compound by a metal-catalyzed cross-coupling reaction such as Suzuki coupling to give a compound of formula A4; then deprotecting the residue with a suitable acid such as trifluoroacetic acid to give a compound represented by the general formula (III-1);
    Or,
    the compound of the formula A7 and boric acid or a pinacol borate compound are coupled through a metal-catalyzed cross coupling reaction such as Suzuki coupling to obtain a compound shown in a general formula (III-1);
    R 1 、R 3 、R 4 、R 5 、R 6 as defined in claim 7.
  16. A process for preparing a compound of formula (IV-1) or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, comprising the steps of:
    the compound of formula B3 is coupled with boric acid or a pinacol borate compound by a metal-catalyzed cross-coupling reaction such as Suzuki coupling to give a compound of formula B4; then deprotecting the residue with a suitable acid such as trifluoroacetic acid to give a compound represented by the general formula (IV-1);
    or,
    the compound of the formula B7 and boric acid or a pinacol borate compound are coupled through a metal-catalyzed cross coupling reaction such as Suzuki coupling to obtain a compound shown in a general formula (IV-1);
    wherein: r is R 1 、R 2 、R 4 、R 5 、R 6 As defined in claim 7.
  17. A process for preparing a compound of formula (V-1) or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, comprising the steps of:
    The compound of formula C3 is coupled with boric acid or a pinacol borate compound by a metal-catalyzed cross-coupling reaction such as Suzuki coupling to give a compound of formula C4; then deprotecting the residue with a suitable acid such as trifluoroacetic acid to give a compound represented by the general formula (V-1);
    alternatively, the compound represented by the general formula (V-1) can be produced by the following scheme 6:
    the compound of formula C7 is coupled with boric acid or a pinacol borate compound through a metal-catalyzed cross coupling reaction such as Suzuki coupling to obtain a compound shown in a general formula (V-1);
    wherein: r is R 2 、R 4 、R 5 、R 6 As defined in claim 7.
  18. A pharmaceutical composition comprising a compound of general formula (I) according to any one of claims 1 to 14 or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier or excipient.
  19. Use of a compound of general formula (I) according to any one of claims 1 to 14 or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 18, for the preparation of a TLR8 agonist.
  20. Use of a compound of general formula (I) according to any one of claims 1 to 14, or a stereoisomer, tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 18, for the manufacture of a medicament for the prevention or treatment of TLR 8-related diseases, preferably viral infectious diseases, such as viral hepatitis b, HIV viral infection, 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, ovarian cancer, bladder cancer, liver cancer, fallopian tube tumors, ovarian tumors, peritoneal tumors, melanoma, solid tumors, glioma, neuroglioblastoma, hepatoma, renal carcinoma, head and neck tumors, leukemia, lymphoma, myeloma and non-small cell lung cancer.
CN202280007466.7A 2021-11-05 2022-11-01 Arylamine derivative, preparation method and medical application thereof Pending CN116472047A (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
CN2021113042021 2021-11-05
CN202111304202 2021-11-05
PCT/CN2022/128963 WO2023078241A1 (en) 2021-11-05 2022-11-01 Aromatic amine derivatives and preparation method and medical use thereof

Publications (1)

Publication Number Publication Date
CN116472047A true CN116472047A (en) 2023-07-21

Family

ID=86240671

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202280007466.7A Pending CN116472047A (en) 2021-11-05 2022-11-01 Arylamine derivative, preparation method and medical application thereof

Country Status (3)

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

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9670205B2 (en) * 2015-03-04 2017-06-06 Gilead Sciences, Inc. Toll like receptor modulator compounds
WO2018045144A1 (en) * 2016-09-02 2018-03-08 Gilead Sciences, Inc. Toll like receptor modulator compounds
CN108069963B (en) * 2017-11-17 2020-01-14 清华大学 Pyridopyrimidine derivative or salt thereof, and preparation method, pharmaceutical composition and application thereof
MX2020013785A (en) * 2018-07-03 2021-03-25 Jiangsu Hengrui Medicine Co Pyridopyrimidine derivative, preparation method therefor and medical use thereof.
JP2022501350A (en) * 2018-09-19 2022-01-06 チア タイ ティエンチン ファーマシューティカル グループ カンパニー リミテッドChia Tai Tianqing Pharmaceutical Group Co., Ltd. TLR8 agonist
AU2019361697A1 (en) * 2018-10-19 2021-05-20 Chia Tai Tianqing Pharmaceutical Group Co., Ltd. TLR8 agonist

Also Published As

Publication number Publication date
TW202327601A (en) 2023-07-16
WO2023078241A1 (en) 2023-05-11

Similar Documents

Publication Publication Date Title
JP6919922B2 (en) FGFR4 inhibitor, its manufacturing method and pharmaceutical application
US7723347B2 (en) Substituted phenylamino-pyrimidines
CN113637005A (en) KRAS inhibitors for cancer treatment
US11179391B2 (en) Compound with kinase inhibitory activity and preparation method and use thereof
TW200829589A (en) Compound for inhibiting mitotic progression
CN110167941B (en) Substituted fused heteroaryl compounds as kinase inhibitors and uses thereof
CN113135910A (en) Pyrimidine-4 (3H) -ketone heterocyclic compound, preparation method and pharmaceutical application thereof
WO2013118817A1 (en) Quinolyl pyrrolopyrimidine compound or salt thereof
TWI781607B (en) A kind of immunosuppressant, its preparation method and application
CN113754682B (en) Compound having macrocyclic structure and use thereof
CN115710266A (en) Pyridopyrimidine derivative, preparation method thereof and application thereof in medicines
CN111566100A (en) Pyrimidine compound, preparation method and medical application thereof
WO2023169226A1 (en) Substituted tricyclic compounds as parp inhibitors and the use thereof
US20230114765A1 (en) Tricyclic compounds as inhibitors of kras
CN115362155A (en) Arylamine derivatives, and preparation method and medical application thereof
CN110312717B (en) Substituted fused heteroaromatic tricyclic compounds as kinase inhibitors and uses thereof
JP2022517723A (en) Macrocycle compound as a CDK inhibitor, its production method and its application in pharmaceutical products
WO2020221209A1 (en) Cd73 inhibitor, preparation method therefor and application thereof
CN116472047A (en) Arylamine derivative, preparation method and medical application thereof
CN114072412B (en) Tricyclic compound, pharmaceutical composition and application thereof
CN114394965B (en) Triazolopyridine compound and preparation method and application thereof
US20090258888A1 (en) Pyrimidine Derivatives
CN114591351A (en) Polycyclic compound and preparation method and application thereof
CN116529251A (en) Substituted fused bicyclic compounds as kinase inhibitors and their use
CN111163775B (en) Novel [1,6] naphthyridine compounds and derivatives as CDK8/CDK19 inhibitors

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 40087550

Country of ref document: HK