CN112654622B - Ring-fused compound, preparation method and application thereof - Google Patents

Ring-fused compound, preparation method and application thereof Download PDF

Info

Publication number
CN112654622B
CN112654622B CN201980058276.6A CN201980058276A CN112654622B CN 112654622 B CN112654622 B CN 112654622B CN 201980058276 A CN201980058276 A CN 201980058276A CN 112654622 B CN112654622 B CN 112654622B
Authority
CN
China
Prior art keywords
compound
membered
alkyl
pharmaceutically acceptable
aryl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN201980058276.6A
Other languages
Chinese (zh)
Other versions
CN112654622A (en
Inventor
李桂英
何云
游泽金
陈忠辉
田强
宋宏梅
薛彤彤
王利春
王晶翼
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.)
Sichuan Kelun Biotech Biopharmaceutical Co Ltd
Original Assignee
Sichuan Kelun Biotech Biopharmaceutical 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 Sichuan Kelun Biotech Biopharmaceutical Co Ltd filed Critical Sichuan Kelun Biotech Biopharmaceutical Co Ltd
Priority to CN202311141511.0A priority Critical patent/CN117186092A/en
Publication of CN112654622A publication Critical patent/CN112654622A/en
Application granted granted Critical
Publication of CN112654622B publication Critical patent/CN112654622B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • 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 invention belongs to the field of medicines, and relates to a fused ring compound, a preparation method and application thereof. In particular, the present invention relates to compounds of formula I, stereoisomers, tautomers or mixtures thereof, pharmaceutically acceptable salts, co-crystals, polymorphs or solvates of said compounds, or stable isotopic derivatives, metabolites or prodrugs of said compounds.

Description

Ring-fused compound, preparation method and application thereof
Technical Field
The invention belongs to the field of medicines, and relates to a nitrogen-containing parallel-cyclic compound, wherein a stereoisomer, a tautomer or a mixture thereof of the compound, a pharmaceutically acceptable salt, a eutectic, a polymorph or a solvate of the compound, or a stable isotope derivative, a metabolite or a prodrug of the compound. The compounds of the invention are useful as NLRP3 modulators (e.g., agonists) for the treatment of diseases associated with NLRP3 inflammatory small body activity (e.g., neoplastic diseases).
Background
NLRP3 (NLR family pyrin domain containing 3) belongs to the NOD-like receptor family, and is an intracellular model recognition receptor most studied in recent years, mainly expressed in macrophages and neutrophils, and involved in the innate immunity of the organism, and resistant to pathogen infection and stress injury. NLRP3 inflammatory corpuscles play a very clear role in inflammatory and metabolic diseases, and excessive activation thereof can lead to immune diseases such as type 2 diabetes, rheumatoid arthritis and atherosclerosis. However, recent studies have shown that NLRP3 has anti-tumor effects in inhibiting tumor growth and metastasis.
After recognizing the pathogen-associated molecular pattern (PAMP) or the endogenous damage-associated molecular pattern (DAMP), the NOD domain of the NLRP3 protein oligomerizes and recruits proteins such as ASC and pro-caspase-1 to form functional NLRP3 inflammatory bodies. After the pro-caspase-1 is cleaved and activated to caspase-1, the caspase-1 cleaves the pro-IL-1β and pro-IL-18 in large amounts to convert them to active forms IL-1β and IL-18 and release them extracellular, amplifying the inflammatory response. The excited NLRP3 inflammatory corpuscles can obviously improve the level of immune factors IL-1 beta and IL-18 in the tumor microenvironment, and start natural immune killing and subsequent acquired immune response to exert the anti-tumor effect. Specifically, IL-1. Beta. Can induce CD8+ T cells to secrete interferon gamma (IFN-gamma), and also can induce CD4+ cells to secrete IL-17, so that effective anti-tumor immune response is caused; IL-18 can promote NK cell maturation, activate STAT1 downstream signal channels in immune cells, and enhance the killing function of the immune cells. Clinical studies have shown that down-regulation of NLRP3 is significantly inversely related to prognosis in liver cancer patients. Preclinical studies also show that NLRP3 deficient mice have higher colorectal tumor formation rates and worsen colorectal liver metastases. Therefore, NLRP3 plays an important role in tumor microenvironment, and can be used as a key target point of tumor immunotherapy and a tumor prognosis marker.
Despite the potential of NLRP3 agonists for tumor immunotherapy, only two companies currently report compounds with NLRP3 agonistic activity. VentiRx Pharmaceutical, VTX2337, which is a TLR8/NLRP3 inflammatory small body dual agonist, has entered phase II in clinical indications are ovarian and head and neck cancer (plos|one, 2016.02.29). Four patent applications (WO 2017184746, WO2017040670, WO2017184735, WO 2018152396) to Bristol-Myers Squibb company and IFM Therapeutis, INC (purchased by Bristol-Myers Squibb company) cover compounds of the same type of parent nucleus, which have a certain selectivity for TLR7 and TLR8, are currently still in preclinical research. Thus, there is a need to develop new, high-potency, low-toxicity NLRP3 agonists to meet clinical therapeutic needs.
Summary of The Invention
The inventors of the present invention have, through creative efforts, obtained a new class of bicyclic compounds that can act as NLRP3 modulators (e.g., agonists), directly bind or modify NLRP3 at the protein level, and enhance the function of NLRP3 inflammatory bodies by activating, stabilizing, altering NLRP3 distribution or otherwise, thereby providing the invention described below.
In one aspect, the invention relates to a compound of formula I, a stereoisomer, tautomer, or mixture thereof, a pharmaceutically acceptable salt, co-crystal, polymorph, or solvate of the compound, or a stable isotopic derivative, metabolite, or prodrug of the compound:
Wherein:
X 1 is C (R) 7 ) 2 、NR 7 O or S,
X 2 is CR (CR) 7 Or N, or a combination of two,
X 3 c, N, O or S, and satisfies the following conditions:
(1) When X is 3 When C is R 6 Selected from H, C 1-6 Alkyl and C 3-8 Cycloalkyl group, the C 1-6 Alkyl and C 3-8 Cycloalkyl groups are each optionally substituted with one or more of the following groups: halogen, OH, CN, NO 2 、C 1-4 Alkoxy and C 1-4 A hydroxyalkyl group, a hydroxyl group,
(2) When X is 3 In the case of N, O or S, R 6 Is absent and when X 3 When O or S, R 2 There is no time for the existence of the non-woven fabric,
R 1 selected from C 1-8 Alkyl, C 1-8 Alkoxy, C 3-8 Cycloalkyl, 4-10 membered heterocyclyl, C 6-12 Aryl, 5-10 membered heteroaryl and 9-12 membered aryl-heterocyclo, said C 1-8 Alkyl, C 1-8 Alkoxy, C 3-8 Cycloalkyl, 4-10 membered heterocyclyl, C 6-12 Aryl, 5-10 membered heteroaryl, and 9-12 membered aryl-heterocyclo are each optionally substituted with one or more of the following substituents: halogen, CN, NO 2 、C 1-4 Alkyl, C 3-8 Cycloalkyl, C 1-4 Haloalkyl, C 1-4 Alkoxy, C 1-4 Haloalkoxy, C 1-4 Hydroxyalkyl, 4-10 membered heterocyclyl, C 6-12 Aryl, 5-10 membered heteroaryl, 9-12 membered aryl-heterocyclo, CO 2 R 30 、C(O)R 30 、C(O)NR 31 R 32 、NR 33 C(O)R 34 、NR 31 R 32 、OC(O)R 30 、OC(O)NR 31 R 32 、NR 33 C(O)NR 31 R 32 、NR 33 C(O)OR 30 、S(O)NR 31 R 32 、S(O) 2 NR 31 R 32 、S(O)R 35 、S(O) 2 R 35 、OR 37 And SR (Surfural) 37
R 2 Selected from H, C 1-8 Alkyl, C 2-8 Alkenyl, C 2-8 Alkynyl, C 6-12 Aryl, C 3-8 Cycloalkyl, 5-10 membered heteroaryl and 4-10 membered heterocyclyl, said C 1-8 Alkyl, C 2-8 Alkenyl, C 2-8 Alkynyl, C 6-12 Aryl, C 3-8 Cycloalkyl, 5-10 membered heteroaryl, and 4-10 membered heterocyclyl are each optionally substituted with one or more of the following substituents: halogen, C 1-4 Haloalkyl, C 1-4 Hydroxyalkyl, C 1-4 Alkoxy, C 1-4 Haloalkoxy, 4-7 membered heterocyclyl, CN, NO 2 、OR 37 、SR 37 、C(O)R 30 、C(O)NR 31 R 32 、NR 31 R 32 C(O)、C(O)OR 30 、OC(O)R 30 、OC(O)NR 31 R 32 、NR 33 C(O)NR 31 R 32 And NR 31 R 32
R 3 Selected from H, OH, halogen, CN, NO 2 、C 1-8 Alkyl, C 1-8 Haloalkyl, C 1-8 Alkoxy, C 1-8 Haloalkoxy, C 3-8 Cycloalkyl, 4-10 membered heterocyclyl, C 6-12 Aryl, 5-10 membered heteroaryl, 9-12 membered aryl-heterocyclo, 9-12 membered aryl-heteroaryls, 9-12 membered aryl-cycloalkyl, CO 2 R 30 、C(O)NR 31 R 32 、NR 33 S(O) 2 R 34 、NR 33 C(O)R 34 、NR 31 R 32 、SO 2 R 35 、OR 37 、SR 37 、C(O)R 30 、OC(O)R 30 、OC(O)NR 31 R 32 、NR 33 C(O)NR 31 R 32 、NR 33 C(O)OR 30 、C(=NR 38 )NR 31 R 32 、NR 33 C(=NR 38 )NR 31 R 32 、P(R 39 ) 2 、P(OR 39 ) 2 、P(O)R 39 R 40 、P(O)OR 39 OR 30 、S(O)R 35 、S(O) 2 R 35 、S(O)NR 31 R 32 And S (O) 2 NR 31 R 32 The C is 1-8 Alkyl, C 1-8 Haloalkyl, C 1-8 Alkoxy, C 1-8 Haloalkoxy, C 3-8 Cycloalkyl, 4-10 membered heterocyclyl, C 6-12 Aryl, 5-10 membered heteroaryl, 9-12 membered arylalkylheterocyclyl, 9-12 membered arylalkylheteroaryl, and 9-12 membered arylalkylcycloalkyl are each optionally substituted with one or more of the following substituents: halogen, CN, NO 2 、C 1-4 Alkyl, C 3-8 Cycloalkyl, C 1-4 Haloalkyl, C 1-4 Alkoxy, C 1-4 Haloalkoxy, C 1-4 Hydroxyalkyl, 4-10 membered heterocyclyl, C 6-12 Aryl, 5-10 membered heteroaryl, 9-12 membered aryl-heterocyclo, CO 2 R 30 、C(O)R 30 、C(O)NR 31 R 32 、NR 33 C(O)R 34 、NR 31 R 32 、S(O)R 35 、S(O) 2 R 35 、S(O)NR 31 R 32 、S(O) 2 NR 31 R 32 、OR 37 、SR 37 、OC(O)R 30 、OC(O)NR 31 R 32 、NR 33 C(O)NR 31 R 32 、NR 33 C(O)OR 30 、C(=NR 38 )NR 31 R 32 、NR 33 C(=NR 38 )NR 31 R 32 、=NNR 31 R 32 、P(R 39 ) 2 、P(OR 39 ) 2 、P(O)R 39 R 40 And P (O) OR 39 OR 30 The method comprises the steps of carrying out a first treatment on the surface of the Or R is 2 And R is 3 A 5-8 membered ring is formed by ligation;
R 4 and R is 5 Each independently selected from H, C 1-8 Alkyl, C 3-8 Cycloalkyl and C 1-8 Alkoxy group, the C 1-8 Alkyl, C 3-8 Cycloalkyl and C 1-8 The alkoxy groups are each optionally substituted with one or more of the following substituents: halogen, OH, CN, NO 2 、C 1-6 Alkyl, C 1-4 Haloalkyl and C 1-4 A hydroxyalkyl group; or R is 4 、R 5 And the nitrogen atom to which it is attached form a 4-7 membered heterocyclic ring;
R 7 selected from H, C 1-6 Alkyl and C 3-8 Cycloalkyl group, the C 1-6 Alkyl and C 3-8 Cycloalkyl groups are each optionally substituted with one or more of the following groups: halogen, OH, CN, C 1-4 Alkoxy and C 1-4 A hydroxyalkyl group; or two R 7 And the carbon atoms to which they are attached form a 3-5 membered cycloalkyl group; when two R 7 When present at the same time, each R 7 May be the same or different;
v is absent, or is selected from C 1-8 Alkylene, C (R) 36a R 36b )、O、S、NR 33 、SO、SO 2 And CO, said C 1-8 The alkylene group is optionally substituted with one or more of the following substituents: halogen, OH, CN, NO 2 、C 1-6 Alkyl, C 1-4 Haloalkyl, C 1-4 Hydroxyalkyl and C 1-4 An alkoxy group;
v and X 1 Not both a heteroatom N, O or S;
l is- (L) 1 ) n -(L 2 ) p -(L 3 ) q -, wherein L 1 、L 2 And L 3 Identical or different and are each independently selected from C 1-8 Alkylene, C 2-8 Alkenylene, C 2-8 Alkynylene, C 1-8 Halogenated alkylene, C 1-8 Alkyloxy, C 1-8 Haloalkoxy, C 1-8 Hydroxyalkylene, C 3-8 Cycloalkylene, 4-10 membered heterocyclylene, C 6-12 Arylene, 5-10 membered heteroarylene, O, S, NR 33 、SO、SO 2 CO and C (R) 36a R 36b ) The C is 1-8 Alkylene, C 2-8 Alkenylene, C 2-8 Alkynylene, C 1-8 Halogenated alkylene, C 1-8 Alkyloxy, C 1-8 Haloalkoxy, C 1-8 Hydroxyalkylene, C 3-8 Cycloalkylene, 4-10 membered heterocyclylene, C 6-12 Arylene and 5-10 membered heteroarylene are each optionally substituted with one or more of the following substituents: halogen, OH, CN, NO 2 、C 1-6 Alkyl, C 1-4 Haloalkyl, C 1-4 Hydroxyalkyl and C 1-4 An alkoxy group;
n, p and q are each independently 0 or 1;
R 30 、R 37 、R 39 and R is 40 Each independently selected from hydrogen, C 1-8 Alkyl, C 1-8 Hydroxyalkyl, C 1-8 Haloalkyl, C 1-8 Alkoxy, C 1-8 Haloalkoxy, C 3-8 Cycloalkyl, 4-10 membered heterocyclyl, C 6-12 Aryl, 5-10 membered heteroaryl, C 1-8 alkyl-C 6-12 Aryl and C 1-8 Alkyl- (5-10 membered heteroaryl), said C 1-8 Alkyl, C 1-8 Hydroxyalkyl, C 1-8 Haloalkyl, C 1-8 Alkoxy, C 1-8 Haloalkoxy, C 3-8 Cycloalkyl, 4-10 membered heterocyclyl, C 6-12 Aryl and 5-10 membered heteroaryl are each optionally substituted with one or more of the following substituents: OH, CN, NO 2 、C 1-4 Alkyl, C 1-4 Alkoxy, C 1-4 Haloalkyl, halogen, C 1-4 Haloalkoxy, CO 2 (C 1-6 Alkyl group, CONR 31 R 32 、NR 31 R 32 、NR 33 C(O)R 34 、S(O)R 35 、S(O) 2 R 35 、S(O)NR 31 R 32 And S (O) 2 NR 31 R 32
When a plurality of R 30 When present at the same time, each R 30 May be the same or different;
when a plurality of R 37 When present at the same time, each R 37 May be the same or different;
when a plurality of R 39 When present at the same time, each R 39 May be the same or different;
when a plurality of R 40 When present at the same time, each R 40 May be the same or different;
R 31 、R 32 、R 33 And R is 34 Each independently selected from H, C 1-8 Alkyl, C 1-8 Hydroxyalkyl, C 1-8 Alkoxy, 4-10 membered heterocyclyl, C 6-12 Aryl, C 3-8 Cycloalkyl and 5-10 membered heteroaryl, said C 1-8 Alkyl, C 1-8 Hydroxyalkyl, C 1-8 Alkoxy, 4-10 membered heterocyclyl, C 6-12 Aryl, C 3-8 Cycloalkyl and 5-10 membered heteroaryl are each optionally substituted with one or more of the following substituents: OH, CN, halogen, NO 2 、C 1-4 Alkyl, C 1-4 Alkoxy, C 1-4 Hydroxyalkyl, C 1-4 Haloalkyl, C 1-4 Haloalkoxy, 4-10 membered heterocyclyl, C 6-12 Aryl and 5-10 membered heteroaryl; or R is 31 And R is 32 And the N atom to which it is attached form a 3-8 membered heterocyclic group; or R is 33 And R is 34 And the N and C atoms or N and S atoms to which they are attached form a 4-8 membered heterocyclic group;
R 35 selected from C 1-8 Alkyl, C 1-8 Hydroxyalkyl, C 1-8 Haloalkyl, C 1-8 Alkoxy, C 1-8 Haloalkoxy, C 3-8 Cycloalkyl, 4-10 membered heterocyclyl, C 6-12 Aryl, 5-10 membered heteroaryl, C 1-8 alkyl-C 6-12 Aryl and C 1-8 Alkyl- (5-10 membered heteroaryl), said C 1-8 Alkyl, C 1-8 Hydroxyalkyl, C 1-8 Haloalkyl, C 1-8 Alkoxy, C 1-8 Haloalkoxy, C 3-8 Cycloalkyl, 4-10 membered heterocyclyl, C 6-12 Aryl and 5-10 membered heteroaryl are each optionally substituted with one or more of the following substituents: OH, CN, NO 2 、C 1-4 Alkyl, C 1-4 Alkoxy, C 1-4 Haloalkyl, halogen, C 1-4 Haloalkoxy, CO 2 (C 1-6 Alkyl group, CONR 31 R 32 、NR 31 R 32 、NR 33 C(O)R 34 、S(O)Me、S(O) 2 Me、S(O)NR 31 R 32 And S (O) 2 NR 31 R 32 Wherein R is 31 、R 32 、R 33 And R is 34 As defined above;
when a plurality of R 31 When present at the same time, each R 31 May be the same or different;
when a plurality of R 32 When present at the same time, each R 32 May be the same or different;
when a plurality of R 33 When present at the same time, each R 33 May be the same or different;
when a plurality of R 34 When present at the same time, each R 34 May be the same or different;
when a plurality of R 35 When present at the same time, each R 35 May be the same or different;
R 36a and R is 36b Identical or different, each independently selected from H, C 1-6 Alkyl, C 1-6 Alkoxy, C 1-8 Hydroxyalkyl and C 1-8 Haloalkyl, said C 1-6 Alkyl, C 1-6 Alkoxy, C 1-8 Hydroxyalkyl and C 1-8 Haloalkyl groups are each optionally substituted with one or more of the following groups: OH, CN, NH 2 、NHCH 3 And N (CH) 3 ) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or R is 36a 、R 36b And together with the carbon atom to which they are attached form a 3-7 membered cycloalkyl or 3-7 membered heterocyclyl;
when a plurality of R 38 When present at the same time, each R 38 Identical or different, each independently selected from H, OH, CN, NO 2 、S(O)R 35 And S (O) 2 R 35
In a preferred embodiment, R 1 Selected from C 1-4 Alkyl, C 1-4 Alkoxy, C 3-6 Cycloalkyl, 4-7 membered heterocyclyl, C 6-12 Aryl, 5-10 membered heteroaryl and 9-12 membered aryl-heterocyclo, said C 1-4 Alkyl, C 1-4 Alkoxy, C 3-6 Cycloalkyl, 4-7 membered heterocyclyl, C 6-12 Aryl, 5-10 membered heteroaryl and 9-12 membered aryl-heterocyclo are each optionally substituted with one or more of the following substituentsMultiple substitutions: halogen, CN, NO 2 、C 1-4 Alkyl, C 3-8 Cycloalkyl, C 1-4 Haloalkyl, C 1-4 Alkoxy, C 1-4 Haloalkoxy, C 1-4 Hydroxyalkyl, 4-10 membered heterocyclyl, C 6-12 Aryl, 5-10 membered heteroaryl, 9-12 membered aryl-heterocyclo, CO 2 R 30 、C(O)R 30 、C(O)NR 31 R 32 、NR 33 C(O)R 34 、NR 31 R 32 、OC(O)R 30 、OC(O)NR 31 R 32 、NR 33 C(O)NR 31 R 32 、NR 33 C(O)OR 30 、S(O)NR 31 R 32 、S(O) 2 NR 31 R 32 、S(O)R 35 、S(O) 2 R 35 、OR 37 And SR (Surfural) 37
In a preferred embodiment, R 1 Selected from C 1-4 Alkyl, C 6-12 Aryl and 5-10 membered heteroaryl, said C 1-4 Alkyl, C 6-12 Aryl and 5-10 membered heteroaryl are each optionally substituted with one or more of the following substituents: halogen, CN, NO 2 、C 1-4 Alkyl, C 3-8 Cycloalkyl, C 1-4 Haloalkyl, C 1-4 Alkoxy, C 1-4 Haloalkoxy, C 1-4 Hydroxyalkyl, 4-10 membered heterocyclyl, C 6-12 Aryl, 5-10 membered heteroaryl, 9-12 membered aryl-heterocyclo, CO 2 R 30 、C(O)R 30 、C(O)NR 31 R 32 、NR 33 C(O)R 34 、NR 31 R 32 、OC(O)R 30 、OC(O)NR 31 R 32 、NR 33 C(O)NR 31 R 32 、NR 33 C(O)OR 30 、S(O)NR 31 R 32 、S(O) 2 NR 31 R 32 、S(O)R 35 、S(O) 2 R 35 、OR 37 And SR (Surfural) 37
In a preferred embodiment, R 1 Selected from isobutyl group,
In the above formula I, V and X 1 Not simultaneously being hetero atoms N, O or S, meaning when X 1 Is NR (NR) 7 In the case of O or S, V is not O, S, NR 33 SO or SO 2
In a preferred embodiment, V is absent, or selected from CO and C 1-8 Alkylene (e.g., methylene).
In a preferred embodiment, X 1 Selected from O and NR 7 Preferably X 1 Selected from O, NH and N (CH) 3 )。
In a preferred embodiment, X 1 Selected from NR 7 Preferably X 1 Selected from NH and N (CH) 3 )。
In a preferred embodiment, X 1 Is O.
In a preferred embodiment, X 1 S.
In a preferred embodiment, X 2 Is CR (CR) 7 Preferably X 2 CH.
In a preferred embodiment, X 2 Is N.
In a preferred embodiment, X 3 Is N, which is R 2 Substituted, and R 6 Absence of; preferably, R 2 Selected from H, C 1-4 Alkyl, C 3-8 Cycloalkyl and C 6-12 An aryl group; more preferably, R 2 Selected from the group consisting of H, methyl, ethyl and propyl, in particular H and methyl.
In a preferred embodiment, R 3 Selected from OH, C 1-8 Alkyl, 4-10 membered heterocyclyl, C 6-12 Aryl, NR 31 R 32 、NR 33 C(O)R 34 、NR 33 S(O) 2 R 34 、C(O)NR 31 R 32 9-12 membered aryl-heterocyclo, 5-10 membered heteroaryl and NR 33 C(O)NR 31 R 32 The C is 1-8 Alkyl, 4-10 membered heterocyclyl, C 6-12 Aryl groupEach of the 5-to 10-membered heteroaryl and 9-to 12-membered aryl-and heterocyclyl is optionally substituted with one or more of the following substituents: c (C) 1-4 Alkyl, C 1-4 Alkoxy, C 6-12 Aryl, C (O) R 30 、C(O)NR 31 R 32 、NR 33 C(O)R 34 、NR 31 R 32 、S(O) 2 R 35 And OR 37
In a preferred embodiment, R 3 Selected from OH, C 1-8 Alkyl, 4-10 membered heterocyclyl, C 6-12 Aryl, NR 31 R 32 、NR 33 C(O)R 34 、NR 33 S(O) 2 R 34 、C(O)NR 31 R 32 9-12 membered aryl-heterocyclo, 5-10 membered heteroaryl and NR 33 C(O)NR 31 R 32 The C is 1-8 Alkyl, 4-10 membered heterocyclyl, C 6-12 Aryl, 5-10 membered heteroaryl, and 9-12 membered aryl-heterocyclo are each optionally substituted with one or more of the following substituents: methyl, methoxy, phenyl, C (O) CH 3 、C(O)NH 2 、NHC(O)CH 3 、NHC(O)H、NH 2 、NH(CH 3 )、S(O) 2 CH 3 And OH.
In a preferred embodiment, R 3 Selected from OH, -NHC (O) CH 3 、-C(O)NH 2 、-N(CH 2 CH 3 )C(O)N(CH 3 ) 2
In a preferred embodiment, R 4 And R is 5 Each independently selected from H, C 1-3 Alkyl, C 3-6 Cycloalkyl and C 1-3 Alkoxy group, the C 1-3 Alkyl, C 1-3 Alkoxy and C 3-6 Cycloalkyl groups are each optionally substituted with one or more of the following substituents: halogen, OH, CN, NO 2 、C 1-6 Alkyl, C 1-4 Haloalkyl and C 1-4 A hydroxyalkyl group; or R is 4 、R 5 And the nitrogen atom to which it is attached form a 4-7 membered heterocyclic ring; preferably, R 4 And R is 5 All are H.
In a preferred embodiment, L is absent or selected from propylene, methylene, ethylene, O, S, NR 33 、-C 1-3 alkylene-O-, -C 1-3 alkylene-S-and-C 1-3 alkylene-NR 33 -; preferably, R 33 Selected from H and methyl.
Preferably, L is absent or selected from propylene, ethylene, O, S, NR 33 、-C 1-3 alkylene-O-, -C 1-3 alkylene-S-and-C 1-3 alkylene-NR 33 -; preferably, R 33 Selected from H and methyl.
More preferably, L is absent or selected from propylene, ethylene, -C 1-3 alkylene-O- (e.g. -ethylene-O-or-propylene-O-) and-C 1-3 alkylene-NH- (e.g. -ethylene-NH-).
In some embodiments of the invention, the compound has the structure of formula II:
wherein R is 1 、R 2 、R 3 、R 4 、R 5 、X 1 V and L are as defined above for formula I; preferably, wherein R 1 、R 2 、R 3 、R 4 、R 5 And L is as defined in claim 1, V is absent or selected from CO, methylene, ethylene and propylene, X 1 Is NR (NR) 7 ,R 7 Selected from H, C 1-4 Alkyl and C 3-6 Cycloalkyl group, the C 1-4 Alkyl and C 3-6 Cycloalkyl groups are each optionally substituted with one or more of the following groups: halogen, OH, CN, C 1-4 Alkoxy and C 1-4 Hydroxyalkyl radicals, e.g. X 1 Selected from NH and N (CH) 3 )。
In a preferred embodiment of the invention, the compounds haveHaving the structure of formula II, wherein R 1 Selected from C 1-4 Alkyl, C 6-12 Aryl (e.g., phenyl) and 5-10 membered heteroaryl (e.g., pyridinyl or pyrazinyl), said C 1-4 Alkyl, C 6-12 Aryl and 5-10 membered heteroaryl are each optionally substituted with one or more of the following substituents: halogen, CN, NO 2 、C 1-4 Alkyl, C 3-8 Cycloalkyl, C 1-4 Haloalkyl, C 1-4 Alkoxy, C 1-4 Haloalkoxy, C 1-4 Hydroxyalkyl, 4-10 membered heterocyclyl, C 6-12 Aryl, 5-10 membered heteroaryl, 9-12 membered aryl-heterocyclo, CO 2 R 30 、C(O)R 30 、C(O)NR 31 R 32 、NR 33 C(O)R 34 、NR 31 R 32 、OC(O)R 30 、OC(O)NR 31 R 32 、NR 33 C(O)NR 31 R 32 、NR 33 C(O)OR 30 、S(O)NR 31 R 32 、S(O) 2 NR 31 R 32 、S(O)R 35 、S(O) 2 R 35 、OR 37 And SR (Surfural) 37 The method comprises the steps of carrying out a first treatment on the surface of the Preferably, R 1 Selected from isobutyl group,
In a preferred embodiment of the invention, the compounds have the structure of formula II, wherein R 2 Selected from H and methyl.
In a preferred embodiment of the invention, the compounds have the structure of formula II, wherein R 3 Selected from OH, C 1-8 Alkyl, 4-10 membered heterocyclyl, C 6-12 Aryl, NR 31 R 32 、NR 33 C(O)R 34 、NR 33 S(O) 2 R 34 、C(O)NR 31 R 32 9-12 membered aryl-heterocyclo, 5-10 membered heteroaryl and NR 33 C(O)NR 31 R 32 The C is 1-8 Alkyl, 4-10 membered heterocyclyl, C 6-12 Aryl groupEach of the 5-to 10-membered heteroaryl and 9-to 12-membered aryl-and heterocyclyl is optionally substituted with one or more of the following substituents: c (C) 1-4 Alkyl, C 1-4 Alkoxy, C 6-12 Aryl, C (O) R 30 、C(O)NR 31 R 32 、NR 33 C(O)R 34 、NR 31 R 32 、S(O) 2 R 35 And OR 37 The method comprises the steps of carrying out a first treatment on the surface of the Preferably, R 3 Selected from OH, C 1-8 Alkyl, 4-10 membered heterocyclyl, C 6-12 Aryl, NR 31 R 32 、NR 33 C(O)R 34 、NR 33 S(O) 2 R 34 、C(O)NR 31 R 32 9-12 membered aryl-heterocyclo, 5-10 membered heteroaryl and NR 33 C(O)NR 31 R 32 The C is 1-8 Alkyl, 4-10 membered heterocyclyl, C 6-12 Aryl, 5-10 membered heteroaryl, and 9-12 membered aryl-heterocyclo are each optionally substituted with one or more of the following substituents: methyl, methoxy, phenyl, C (O) CH 3 、C(O)NH 2 、NHC(O)CH 3 、NHC(O)H、NH 2 、NH(CH 3 )、S(O) 2 CH 3 And OH.
In a preferred embodiment of the invention, the compounds have the structure of formula II, wherein R 4 And R is 5 Each independently selected from H, C 1-3 Alkyl, C 1-3 Alkoxy and C 3-6 Cycloalkyl group, the C 1-3 Alkyl, C 1-3 Alkoxy and C 3-6 Cycloalkyl groups are each optionally substituted with one or more of the following substituents: halogen, OH, CN, NO 2 、C 1-6 Alkyl, C 1-4 Haloalkyl and C 1-4 A hydroxyalkyl group; or R is 4 、R 5 And the nitrogen atom to which it is attached form a 4-7 membered heterocyclic ring; preferably, R 4 And R is 5 All are H.
In a preferred embodiment of the invention, the compound has the structure of formula II, wherein X 1 Selected from C (R) 7 ) 2 、NR 7 O and S; preferably X 1 Selected from O and NR 7 More preferably, X 1 Selected from O, NH and N (CH) 3 )。
In a preferred embodiment of the invention, the compounds have the structure of formula II, wherein V is absent or selected from C 1-8 Alkylene, C (R) 36a R 36b )、O、S、NR 33 、SO、SO 2 And CO, said C 1-8 The alkylene group is optionally substituted with one or more of the following substituents: halogen, OH, CN, NO 2 、C 1-6 Alkyl, C 1-4 Haloalkyl, C 1-4 Hydroxyalkyl and C 1-4 An alkoxy group; preferably, V is absent, or selected from CO and C 1-8 Alkylene (e.g., methylene).
In a preferred embodiment of the invention, the compounds have the structure of formula II, wherein V and X 1 Not simultaneously being hetero atoms N, O or S, i.e. when X 1 Is NR (NR) 7 In the case of O or S, V is not O, S, NR 33 SO or SO 2
In a preferred embodiment of the invention, the compound has the structure of formula II, wherein L is absent or selected from propylene, methylene, ethylene, O, S, NR 33 、-C 1-3 alkylene-O-, -C 1-3 alkylene-S-and-C 1-3 alkylene-NR 33 -, wherein R is 33 Selected from H and methyl; preferably, L is absent or selected from propylene, ethylene, O, S, NR 33 、-C 1-3 alkylene-O-, -C 1-3 alkylene-S-and-C 1-3 alkylene-NR 33 -, wherein R is 33 Selected from H and methyl; more preferably, L is absent or selected from propylene, ethylene, -C 1-3 alkylene-O- (e.g. -ethylene-O-or-propylene-O-) and-C 1-3 alkylene-NH- (e.g. -ethylene-NH-).
In a preferred embodiment of the invention, the compounds have the structure of formula II, wherein n, p and q are each independently 0.
In a preferred embodiment of the invention, the compounds have the structure of formula II, wherein n, p and q are each independently 1.
In a preferred embodiment of the invention, the compound has the structure of formula II, wherein:
R 1 selected from C 1-4 Alkyl, C 6-12 Aryl (e.g., phenyl) and 5-10 membered heteroaryl (e.g., pyridinyl or pyrazinyl), said C 1-4 Alkyl, C 6-12 Aryl and 5-10 membered heteroaryl are each optionally substituted with one or more of the following substituents: halogen, CN, NO 2 、C 1-4 Alkyl, C 3-8 Cycloalkyl, C 1-4 Haloalkyl, C 1-4 Alkoxy, C 1-4 Haloalkoxy, C 1-4 Hydroxyalkyl, 4-10 membered heterocyclyl, C 6-12 Aryl, 5-10 membered heteroaryl, 9-12 membered aryl-heterocyclo, CO 2 R 30 、C(O)R 30 、C(O)NR 31 R 32 、NR 33 C(O)R 34 、NR 31 R 32 、OC(O)R 30 、OC(O)NR 31 R 32 、NR 33 C(O)NR 31 R 32 、NR 33 C(O)OR 30 、S(O)NR 31 R 32 、S(O) 2 NR 31 R 32 、S(O)R 35 、S(O) 2 R 35 、OR 37 And SR (Surfural) 37
R 2 Selected from H and methyl;
R 3 selected from OH, C 1-8 Alkyl, 4-10 membered heterocyclyl, C 6-12 Aryl, NR 31 R 32 、NR 33 C(O)R 34 、NR 33 S(O) 2 R 34 、C(O)NR 31 R 32 9-12 membered aryl-heterocyclo, 5-10 membered heteroaryl and NR 33 C(O)NR 31 R 32 The C is 1-8 Alkyl, 4-10 membered heterocyclyl, C 6-12 Aryl, 5-10 membered heteroaryl, and 9-12 membered aryl-heterocyclo are each optionally substituted with one or more of the following substituents: c (C) 1-4 Alkyl, C 1-4 Alkoxy, C 6-12 Aryl, C (O) R 30 、C(O)NR 31 R 32 、NR 33 C(O)R 34 、NR 31 R 32 、S(O) 2 R 35 And OR 37 The method comprises the steps of carrying out a first treatment on the surface of the Preferably, R 3 Selected from OH, C 1-8 Alkyl, 4-10 membered heterocyclyl, C 6-12 Aryl, NR 31 R 32 、NR 33 C(O)R 34 、NR 33 S(O) 2 R 34 、C(O)NR 31 R 32 9-12 membered aryl-heterocyclo, 5-10 membered heteroaryl and NR 33 C(O)NR 31 R 32 The C is 1-8 Alkyl, 4-10 membered heterocyclyl, C 6-12 Aryl, 5-10 membered heteroaryl, and 9-12 membered aryl-heterocyclo are each optionally substituted with one or more of the following substituents: methyl, methoxy, phenyl, C (O) CH 3 、C(O)NH 2 、NHC(O)CH 3 、NHC(O)H、NH 2 、NH(CH 3 )、S(O) 2 CH 3 And OH;
R 4 and R is 5 Each independently selected from H, C 1-8 Alkyl, C 3-8 Cycloalkyl and C 1-8 Alkoxy group, the C 1-8 Alkyl, C 1-8 Alkoxy and C 3-8 Cycloalkyl groups are each optionally substituted with one or more of the following substituents: halogen, OH, CN, NO 2 、C 1-6 Alkyl, C 1-4 Haloalkyl and C 1-4 A hydroxyalkyl group; or R is 4 、R 5 And the nitrogen atom to which it is attached form a 4-7 membered heterocyclic ring;
X 1 selected from C (R) 7 ) 2 、NR 7 O and S;
v is absent, or is selected from C 1-8 Alkylene, C (R) 36a R 36b )、O、S、NR 33 、SO、SO 2 And CO, said C 1-8 The alkylene group is optionally substituted with one or more of the following substituents: halogen, OH, CN, NO 2 、C 1-6 Alkyl, C 1-4 Haloalkyl, C 1-4 Hydroxyalkyl and C 1-4 An alkoxy group;
v and X 1 Not both a heteroatom N, O or S;
l is- (L) 1 ) n -(L 2 ) p -(L 3 ) q -, wherein L 1 、L 2 And L 3 Identical or different and are each independently selected from C 1-8 Alkylene (e.g. C 1-3 Alkylene group, C 2-8 Alkenylene, C 2-8 Alkynylene, C 1-8 Halogenated alkylene, C 1-8 Alkyloxy, C 1-8 Haloalkoxy, C 1-8 Hydroxyalkylene, C 3-8 Cycloalkylene, 4-10 membered heterocyclylene, C 6-12 Arylene, 5-10 membered heteroarylene, O, S, NR 33 、SO、SO 2 -CO-and-C (R) 36a R 36b ) -, the C 1-8 Alkylene, C 2-8 Alkenylene, C 2-8 Alkynylene, C 1-8 Halogenated alkylene, C 1-8 Alkyloxy, C 1-8 Haloalkoxy, C 1-8 Hydroxyalkylene, C 3-8 Cycloalkylene, 4-10 membered heterocyclylene, C 6-12 Arylene and 5-10 membered heteroarylene are each optionally substituted with one or more of the following substituents: halogen, OH, CN, NO 2 、C 1-6 Alkyl, C 1-4 Haloalkyl, C 1-4 Hydroxyalkyl and C 1-4 An alkoxy group;
n, p, q are each independently 0 or 1.
In a preferred embodiment of the invention, the compound has the structure of formula II, wherein:
R 1 selected from isobutyl group,
R 2 Selected from H and methyl;
R 3 selected from OH, -NHC (O) CH 3 、-C(O)NH 2 、-N(CH 2 CH 3 )C(O)N(CH 3 ) 2
R 4 And R is 5 All are H;
X 1 selected from O, NH and N (CH) 3 );
V is absent or selected from CO and methylene;
l is absent or selected from propylene, methylene, ethylene, O, S, NR 33 、-C 1-3 alkylene-O-, -C 1-3 alkylene-S-and-C 1-3 alkylene-NR 33 -, wherein R is 33 Selected from H and methyl; preferably, L is absent or selected from propylene, ethylene, O, S, NR 33 、-C 1-3 alkylene-O-, -C 1-3 alkylene-S-and-C 1-3 alkylene-NR 33 -, wherein R is 33 Selected from H and methyl; more preferably, L is absent or selected from propylene, ethylene, -C 1-3 alkylene-O- (e.g. -ethylene-O-or-propylene-O-) and-C 1-3 alkylene-NH- (e.g. -ethylene-NH-).
In some embodiments of the invention, the compound has the structure of formula II-A:
wherein R is 1 、R 2 、R 3 And L is as defined above for formula II.
In a preferred embodiment of the invention, the compound has the structure of formula II-A, wherein:
R 1 selected from isobutyl group,
R 2 Selected from H and methyl;
R 3 selected from OH, -NHC (O) CH 3 、C(O)NH 2 、-N(CH 2 CH 3 )C(O)N(CH 3 ) 2
L is absent or selected from propylene, methylene, ethylene, O, S, NR 33 、-C 1-3 alkylene-O-, -C 1-3 alkylene-S-and-C 1-3 alkylene-NR 33 -, wherein R is 33 Selected from H and methyl; preferably, L is absent or selected from propylene, ethylene, O, S, NR 33 、-C 1-3 alkylene-O-, -C 1-3 alkylene-S-and-C 1-3 alkylene-NR 33 -, wherein R is 33 Selected from H and methyl; more preferably, L is absent or selected from propylene, ethylene, -C 1-3 alkylene-O- (e.g. -ethylene-O-or-propylene-O-) and-C 1-3 alkylene-NH- (e.g. -ethylene-NH-).
In some embodiments of the invention, the compound has the structure of formula III:
wherein R is 1 、R 2 、R 3 、R 4 、R 5 、X 1 V and L are as defined above for formula I.
In a preferred embodiment of the invention, the compound has the structure of formula III, wherein:
R 1 selected from isobutyl, C 6-12 Aryl (e.g., phenyl) and 5-10 membered heteroaryl (e.g., pyridinyl or pyrazinyl), said C 6-12 Aryl and 5-10 membered heteroaryl are each optionally substituted with one or more of the following substituents: halogen, CN, NO 2 、C 1-4 Alkyl, C 3-8 Cycloalkyl, C 1-4 Haloalkyl, C 1-4 Alkoxy, C 1-4 Haloalkoxy, C 1-4 Hydroxyalkyl, 4-10 membered heterocyclyl, C 6-12 Aryl, 5-10 membered heteroaryl, 9-12 membered aryl-heterocyclo, CO 2 R 30 、C(O)R 30 、C(O)NR 31 R 32 、NR 33 C(O)R 34 、NR 31 R 32 、OC(O)R 30 、OC(O)NR 31 R 32 、NR 33 C(O)NR 31 R 32 、NR 33 C(O)OR 30 、S(O)NR 31 R 32 、S(O) 2 NR 31 R 32 、S(O)R 35 、S(O) 2 R 35 、OR 37 And SR (Surfural) 37
R 2 Selected from H and methyl;
R 3 selected from OH, C 1-8 Alkyl, 4-10 membered heterocyclyl, C 6-12 Aryl, NR 31 R 32 、NR 33 C(O)R 34 、NR 33 S(O) 2 R 34 、C(O)NR 31 R 32 9-12 membered aryl-heterocyclo, 5-10 membered heteroaryl and NR 33 C(O)NR 31 R 32 The C is 1-8 Alkyl, 4-10 membered heterocyclyl, C 6-12 Aryl, 5-10 membered heteroaryl, and 9-12 membered aryl-heterocyclo are each optionally substituted with one or more of the following substituents: c (C) 1-4 Alkyl, C 1-4 Alkoxy, C 6-12 Aryl, C (O) R 30 、C(O)NR 31 R 32 、NR 33 C(O)R 34 、NR 31 R 32 、S(O) 2 R 35 And OR 37 The method comprises the steps of carrying out a first treatment on the surface of the Preferably, R 3 Selected from OH, C 1-8 Alkyl, 4-10 membered heterocyclyl, C 6-12 Aryl, NR 31 R 32 、NR 33 C(O)R 34 、NR 33 S(O) 2 R 34 、C(O)NR 31 R 32 9-12 membered aryl-heterocyclo, 5-10 membered heteroaryl and NR 33 C(O)NR 31 R 32 The C is 1-8 Alkyl, 4-10 membered heterocyclyl, C 6-12 Aryl, 5-10 membered heteroaryl, and 9-12 membered aryl-heterocyclo are each optionally substituted with one or more of the following substituents: methyl, methoxy, phenyl, C (O) CH 3 、C(O)NH 2 、NHC(O)CH 3 、NHC(O)H、NH 2 、NH(CH 3 )、S(O) 2 CH 3 And OH;
R 4 and R is 5 Each independently selected fromH、C 1-8 Alkyl, C 3-8 Cycloalkyl and C 1-8 Alkoxy group, the C 1-8 Alkyl, C 3-8 Cycloalkyl and C 1-8 The alkoxy groups are each optionally substituted with one or more of the following substituents: halogen, OH, CN, NO 2 、C 1-6 Alkyl, C 1-4 Haloalkyl and C 1-4 A hydroxyalkyl group; or R is 4 、R 5 And the nitrogen atom to which it is attached form a 4-7 membered heterocyclic ring;
X 1 selected from C (R) 7 ) 2 、NR 7 O and S;
v is absent, or is selected from C 1-8 Alkylene, C (R) 36a R 36b )、O、S、NR 33 、SO、SO 2 And CO, said C 1-8 The alkylene group is optionally substituted with one or more of the following substituents: halogen, OH, CN, NO 2 、C 1-6 Alkyl, C 1-4 Haloalkyl, C 1-4 Hydroxyalkyl and C 1-4 An alkoxy group;
v and X 1 Not both a heteroatom N, O or S;
l is- (L) 1 ) n -(L 2 ) p -(L 3 ) q -, wherein L 1 、L 2 And L 3 Identical or different and are each independently selected from C 1-8 Alkylene (e.g. C 1-3 Alkylene group, C 2-8 Alkenylene, C 2-8 Alkynylene, C 1-8 Halogenated alkylene, C 1-8 Alkyloxy, C 1-8 Haloalkoxy, C 1-8 Hydroxyalkylene, C 3-8 Cycloalkylene, 4-10 membered heterocyclylene, C 6-12 Arylene, 5-10 membered heteroarylene, O, S, NR 33 、SO、SO 2 -CO-and-C (R) 36a R 36b ) -, the C 1-8 Alkylene, C 2-8 Alkenylene, C 2-8 Alkynylene, C 1-8 Halogenated alkylene, C 1-8 Alkyloxy, C 1-8 Haloalkoxy, C 1-8 Hydroxyalkylene, C 3-8 Cycloalkylene, 4-10 membered heterocyclylene, C 6-12 Arylene and 5-10 membered heteroarylene are each optionally substituted withOne or more substitutions: halogen, OH, CN, NO 2 、C 1-6 Alkyl, C 1-4 Haloalkyl, C 1-4 Hydroxyalkyl and C 1-4 An alkoxy group;
n, p, q are each independently 0 or 1.
In a preferred embodiment of the invention, the compound has the structure of formula III, wherein:
R 1 selected from isobutyl group,
R 2 Selected from H and methyl;
R 3 selected from OH, -NHC (O) CH 3 、-C(O)NH 2 、-N(CH 2 CH 3 )C(O)N(CH 3 ) 2/>
R 4 And R is 5 All are H;
X 1 selected from O, NH and N (CH) 3 );
V is absent or selected from CO and methylene;
l is absent or selected from propylene, methylene, ethylene, O, S, NR 33 、-C 1-3 alkylene-O-, -C 1-3 alkylene-S-and-C 1-3 alkylene-NR 33 -, wherein R is 33 Selected from H and methyl; preferably, L is absent or selected from propylene, ethylene, O, S, NR 33 、-C 1-3 alkylene-O-, -C 1-3 Alkylene-S-and-C 1-3 alkylene-NR 33 -, wherein R is 33 Selected from H and methyl; more preferably, L is absent or selected from propylene, ethylene, -C 1-3 alkylene-O- (e.g. -ethylene-O-or-propylene-O-) and-C 1-3 alkylene-NH- (e.g. -ethylene-NH-).
In some embodiments of the invention, the compound has the structure of formula IV:
wherein R is 1 、R 3 、R 4 、R 5 、X 1 V and L are as defined above for formula I.
In a preferred embodiment of the invention, the compound has the structure of formula IV, wherein:
R 1 selected from C 1-4 Alkyl, C 6-12 Aryl (e.g., phenyl) and 5-10 membered heteroaryl (e.g., pyridinyl or pyrazinyl), said C 1-4 Alkyl, C 6-12 Aryl and 5-10 membered heteroaryl are each optionally substituted with one or more of the following substituents: halogen, CN, NO 2 、C 1-4 Alkyl, C 3-8 Cycloalkyl, C 1-4 Haloalkyl, C 1-4 Alkoxy, C 1-4 Haloalkoxy, C 1-4 Hydroxyalkyl, 4-10 membered heterocyclyl, C 6-12 Aryl, 5-10 membered heteroaryl, 9-12 membered aryl-heterocyclo, CO 2 R 30 、C(O)R 30 、C(O)NR 31 R 32 、NR 33 C(O)R 34 、NR 31 R 32 、OC(O)R 30 、OC(O)NR 31 R 32 、NR 33 C(O)NR 31 R 32 、NR 33 C(O)OR 30 、S(O)NR 31 R 32 、S(O) 2 NR 31 R 32 、S(O)R 35 、S(O) 2 R 35 、OR 37 And SR (Surfural) 37
R 3 Selected from OH, C 1-8 Alkyl, 4-10 membered heterocyclyl, C 6-12 Aryl, NR 31 R 32 、NR 33 C(O)R 34 、NR 33 S(O) 2 R 34 、C(O)NR 31 R 32 9-12 membered aryl-heterocyclo, 5-10 membered heteroaryl and NR 33 C(O)NR 31 R 32 The C is 1-8 Alkyl, 4-10 membered heterocyclyl, C 6-12 Aryl, 5-10 membered heteroaryl, and 9-12 membered aryl-heterocyclo are each optionally substituted with one or more of the following substituents: c (C) 1-4 Alkyl, C 1-4 Alkoxy, C 6-12 Aryl, C (O) R 30 、C(O)NR 31 R 32 、NR 33 C(O)R 34 、NR 31 R 32 、S(O) 2 R 35 And OR 37 The method comprises the steps of carrying out a first treatment on the surface of the Preferably, R 3 Selected from OH, C 1-8 Alkyl, 4-10 membered heterocyclyl, C 6-12 Aryl, NR 31 R 32 、NR 33 C(O)R 34 、NR 33 S(O) 2 R 34 、C(O)NR 31 R 32 9-12 membered aryl-heterocyclo, 5-10 membered heteroaryl and NR 33 C(O)NR 31 R 32 The C is 1-8 Alkyl, 4-10 membered heterocyclyl, C 6-12 Aryl, 5-10 membered heteroaryl, and 9-12 membered aryl-heterocyclo are each optionally substituted with one or more of the following substituents: methyl, methoxy, phenyl, C (O) CH 3 、C(O)NH 2 、NHC(O)CH 3 、NHC(O)H、NH 2 、NH(CH 3 )、S(O) 2 CH 3 And OH;
R 4 and R is 5 Each independently selected from H, C 1-8 Alkyl, C 1-8 Alkoxy and C 3-8 Cycloalkyl group, the C 1-8 Alkyl, C 1-8 Alkoxy and C 3-8 Cycloalkyl groups are each optionally substituted with one or more of the following substituents: halogen, OH, CN, NO 2 、C 1-6 Alkyl, C 1-4 Haloalkyl and C 1-4 A hydroxyalkyl group; or R is 4 、R 5 And the nitrogen atom to which it is attached form a 4-7 membered heterocyclic ring;
X 1 selected from C (R) 7 ) 2 、NR 7 O and S;
v is absent, or is selected from C 1-8 Alkylene, C (R) 36a R 36b )、O、S、NR 33 、SO、SO 2 And CO, said C 1-8 The alkylene group is optionally substituted with one or more of the following substituents: halogen, OH, CN, NO 2 、C 1-6 Alkyl, C 1-4 Haloalkyl, C 1-4 Hydroxyalkyl and C 1-4 An alkoxy group;
v and X 1 Not both a heteroatom N, O or S;
l is- (L) 1 ) n -(L 2 ) p -(L 3 ) q -, wherein L 1 、L 2 And L 3 Identical or different and are each independently selected from C 1-8 Alkylene (e.g. C 1-3 Alkylene group, C 2-8 Alkenylene, C 2-8 Alkynylene, C 1-8 Halogenated alkylene, C 1-8 Alkyloxy, C 1-8 Haloalkoxy, C 1-8 Hydroxyalkylene, C 3-8 Cycloalkylene, 4-10 membered heterocyclylene, C 6-12 Arylene, 5-10 membered heteroarylene, O, S, NR 33 、SO、SO 2 -CO-and-C (R) 36a R 36b ) -, the C 1-8 Alkylene, C 2-8 Alkenylene, C 2-8 Alkynylene, C 1-8 Halogenated alkylene, C 1-8 Alkyloxy, C 1-8 Haloalkoxy, C 1-8 Hydroxyalkylene, C 3-8 Cycloalkylene, 4-10 membered heterocyclylene, C 6-12 Arylene and 5-10 membered heteroarylene are each optionally substituted with one or more of the following substituents: halogen, OH, CN, NO 2 、C 1-6 Alkyl, C 1-4 Haloalkyl, C 1-4 Hydroxyalkyl and C 1-4 An alkoxy group;
n, p, q are each independently 0 or 1.
In a preferred embodiment of the invention, the compound has the structure of formula IV, wherein:
R 1 selected from isobutyl group,
R 3 Selected from OH, -NHC (O) CH 3 、-C(O)NH 2 、-N(CH 2 CH 3 )C(O)N(CH 3 ) 2/>
R 4 And R is 5 All are H;
X 1 selected from O, NH and N (CH) 3 );
V is absent or selected from CO and methylene;
l is absent or selected from propylene, ethylene, -C 1-3 alkylene-O- (e.g. -ethylene-O-or-propylene-O-) and-C 1-3 alkylene-NH- (e.g. -ethylene-NH-).
In some embodiments of the invention, the compounds of the invention are selected from, but are not limited to:
/>
in another aspect, the invention provides a pharmaceutical composition comprising a compound as described above, a stereoisomer, tautomer, or mixture thereof, a pharmaceutically acceptable salt, co-crystal, polymorph, or solvate of the compound, or a stable isotopic derivative, metabolite, or prodrug of the compound. Optionally, the pharmaceutical composition further comprises one or more pharmaceutically acceptable carriers.
In some embodiments, the pharmaceutical composition is used to prevent and/or treat a disease associated with NLRP3 inflammatory body activity (e.g., a neoplastic disease).
Optionally, the pharmaceutical composition of the invention further comprises one or more second therapeutic agents. In certain embodiments, the second therapeutic agent comprises other drugs for treating neoplastic diseases and the like.
In another aspect, the invention provides a pharmaceutical formulation comprising a compound as described above, a stereoisomer, tautomer, or mixture thereof, a pharmaceutically acceptable salt, co-crystal, polymorph, or solvate of the compound, or a stable isotopic derivative, metabolite, or prodrug of the compound, or a pharmaceutical composition as described above.
In another aspect, the invention provides the use of a compound as described above, a stereoisomer, tautomer, or mixture thereof, a pharmaceutically acceptable salt, co-crystal, polymorph, or solvate of the compound, or a stable isotopic derivative, metabolite, or prodrug of the compound, or a pharmaceutical composition as described above, for the manufacture of a medicament for the prevention and/or treatment of a disease associated with NLRP3 inflammatory body activity (e.g. a neoplastic disease).
In another aspect, the invention provides the use of a compound as described above, a stereoisomer, tautomer, or mixture thereof, a pharmaceutically acceptable salt, co-crystal, polymorph, or solvate of the compound, or a stable isotopic derivative, metabolite, or prodrug of the compound, or a pharmaceutical composition as described above, for preparing a formulation for modulating (e.g., increasing) the activity of an NLRP3 inflammatory body.
In some embodiments, the formulation is administered to a subject (e.g., mammal; e.g., bovine, equine, ovine, porcine, canine, feline, rodent, primate; e.g., human) in vivo to increase NLRP3 inflammatory body activity in cells in the subject; alternatively, the formulation is administered to cells in vitro (e.g., a cell line or a cell from a subject) to increase the activity of NLRP3 inflammatory bodies in the cells.
In another aspect, the invention provides a method of modulating (e.g., increasing) the activity of an inflammatory small body of NLRP3 in a cell, comprising administering to the cell an effective amount of a compound as described above, a stereoisomer, tautomer, or mixture thereof, a pharmaceutically acceptable salt, co-crystal, polymorph, or solvate of the compound, or a stable isotopic derivative, metabolite, or prodrug of the compound, or a pharmaceutical composition as described above, or a pharmaceutical formulation as described above.
In another aspect, the invention provides a kit for modulating (e.g., increasing) the activity of an NLRP3 inflammatory oligomer, the kit comprising a compound as described above, a stereoisomer, tautomer, or mixture thereof, a pharmaceutically acceptable salt, co-crystal, polymorph, or solvate of the compound, or a stable isotopic derivative, metabolite, or prodrug of the compound, or a pharmaceutical composition as described above, or a formulation as described above.
In another aspect, the invention provides a method of treating a disease associated with NLRP3 inflammatory small body activity (e.g., a neoplastic disease), comprising administering to a subject in need thereof a therapeutically and/or prophylactically effective amount of a compound as described above, a stereoisomer, tautomer, or mixture thereof, a pharmaceutically acceptable salt, co-crystal, polymorph, or solvate of the compound, or a stable isotopic derivative, metabolite, or prodrug of the compound, or a pharmaceutical composition as described above, or a formulation as described above.
Optionally, the method further comprises administering to a subject in need thereof one or more second therapeutic agents. In some embodiments, the second therapeutic agent comprises an additional agent that treats a disease such as a tumor.
In another aspect, the invention provides a compound as described above, a stereoisomer, tautomer, or mixture thereof, a pharmaceutically acceptable salt, co-crystal, polymorph, or solvate of the compound, or a stable isotopic derivative, metabolite, or prodrug of the compound, for use in the prevention and/or treatment of a disease associated with NLRP3 inflammatory body activity (e.g., a neoplastic disease).
In the present invention, the neoplastic diseases include, but are not limited to: brain tumor, lung cancer, squamous cell carcinoma, bladder cancer, stomach cancer, ovarian cancer, peritoneal cancer, pancreatic cancer, breast cancer, head and neck cancer, cervical cancer, endometrial cancer, rectal cancer, liver cancer, kidney cancer, esophageal adenocarcinoma, esophageal squamous cell carcinoma, prostate cancer, female genital tract cancer, carcinoma in situ, lymphoma, neurofibromatosis, thyroid cancer, bone cancer, skin cancer, brain cancer, colon cancer, testicular cancer, gastrointestinal stromal tumor, prostate tumor, mast cell tumor, multiple myeloma, melanoma, glioma, or sarcoma.
In some embodiments, the compounds of the invention are full agonists; in some embodiments, the compounds of the invention are partial agonists (partial agonists).
Detailed Description
Definition of terms
Unless defined otherwise hereinafter, all technical and scientific terms used herein are intended to be identical to what is commonly understood by one of ordinary skill in the art. References to techniques used herein are intended to refer to techniques commonly understood in the art, including variations of those that are obvious to those skilled in the art or alternatives to equivalent techniques. While the following terms are believed to be well understood by those skilled in the art, the following definitions are set forth to better explain the present invention.
The term "agonist" refers to a compound that binds to and activates a receptor to elicit a downstream biological effect or response, including full agonist and moietiesAgonists (partial agonist). Full agonists activate the receptor and produce the greatest effect (maximal effect or E) max ). Partial agonists can bind to and activate receptors, but produce only partial effects (partial effects) relative to full agonists. When a full agonist and a partial agonist coexist, the partial agonist may sometimes become a partial antagonist by competing with the full agonist for a binding site or other mechanism at the receptor. Potency of a partial agonist (potential, available from EC 50 (50% E was produced) max Compound concentration at that time) is measured) is likely to be higher or lower than the efficacy of a full agonist. The NLRP3 agonists of the invention include NLRP3 full agonists and NLRP3 partial agonists.
The term "NLRP3" is generally known as NLR family pyrin domain containing 3 and is an inflammatory body. In the present invention, when referring to "NLRP3", the meaning includes nucleic acids, polynucleotides, oligonucleotides, sense and antisense polynucleotide strands, complementary sequences, short peptides, polypeptides, proteins, homologous or heterologous molecules, subtypes, precursors, mutants, variants, derivatives, various spliceosomes, alleles, different species, and activation fragments of NLRP3, and the like.
The terms "comprising," "including," "having," "containing," or "involving," and other variations thereof herein, are inclusive or open-ended and do not exclude additional unrecited elements or method steps.
The term "halo" refers to substitution by a halogen atom, including F, cl, br or I.
The term "alkyl" is a straight or branched chain saturated aliphatic hydrocarbon group. The term "C 1-8 Alkyl "," C 1-6 Alkyl "and" C 1-4 Alkyl "refers to straight or branched chain alkyl groups having 1 to 8 carbon atoms, 1 to 6 carbon atoms, and 1 to 4 carbon atoms, respectively, such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, or n-hexyl. The alkyl group may be optionally substituted with one or more (such as 1 to 3) identical or different substituents.
The term "alkylene" refers to a saturated divalent hydrocarbon radical obtained by removing two hydrogen atoms from a straight or branched saturated hydrocarbon radical, which contains the indicated number of carbon atoms. For example an alkylene group of 1 to 8 carbon atoms, for example methylene (-CH) 2 (-), ethylene (-CH) 2 CH 2 (-), propylene (-CH) 2 CH 2 CH 2 (-), isopropylidene (-CH (CH) 3 )CH 2 (-), etc. The alkylene group may be optionally substituted with one or more (such as 1 to 3) identical or different substituents.
The term "haloalkyl" refers to an alkyl group substituted with one or more (such as 1 to 3) identical or different halogen atoms, the term "C 1-8 Haloalkyl "," C 1-6 Haloalkyl groups "and" C 1-4 Haloalkyl "refers to haloalkyl groups having 1 to 8 carbon atoms, 1 to 6 carbon atoms and 1 to 4 carbon atoms, respectively, such as-CF 3 、-C 2 F 5 、-CHF 2 、-CH 2 F、-CH 2 CF 3 、-CH 2 Cl、-CH 2 CH 2 CF 3 Etc.
The term "hydroxyalkyl" refers to a group formed by substitution of the hydrogen atom of an alkyl group with one or more hydroxyl groups, e.g., C 1-4 Hydroxyalkyl or C 1-3 Hydroxyalkyl groups, examples of which include, but are not limited to, hydroxymethyl, hydroxyethyl, hydroxypropyl, hydroxybutyl, -CH (OH) CH 3 Etc.
The term "alkenyl" refers to a monovalent straight or branched hydrocarbon radical containing one or more carbon-carbon double bonds, e.g., -ch=ch 2 、-CH 2 CH=CH 2 、-C(CH 3 )=CH 2 、-CH 2 -CH=CH-CH 3 Etc. The alkenyl group may be optionally substituted with one or more (such as 1 to 3) identical or different substituents.
The term "alkenylene" refers to a divalent straight or branched chain aliphatic hydrocarbon radical containing one or more carbon-carbon double bonds, containing the indicated number of carbon atoms, e.g., 2 to 8 carbon atoms, e.g., -ch=ch-, -CH 2 CH=CH-、-C(CH 3 ) =ch-, etc. The alkenylene groups may optionally be the same or different by one or more (such as 1 to 3)Is substituted by a substituent of (a).
The term "alkynyl" refers to monovalent straight or branched hydrocarbon groups having one or more carbon-carbon triple bonds, including but not limited to ethynyl, 2-propynyl, 2-butynyl, 1, 3-butadiynyl, and the like. The alkynyl group may be optionally substituted with one or more (such as 1 to 3) identical or different substituents.
The term "alkynylene" refers to a divalent straight or branched chain hydrocarbon radical having one or more carbon-carbon triple bonds containing a specified number of carbon atoms, e.g., 2 to 8 carbon atoms, including but not limited toEtc. The alkynylene group may be optionally substituted with one or more (such as 1 to 3) identical or different substituents.
The term "alkoxy" means a group, preferably C, having an oxygen atom inserted at any reasonable position of an alkyl group (as defined above) 1-8 Alkoxy, C 1-6 Alkoxy, C 1-4 Alkoxy or C 1-3 An alkoxy group. C (C) 1-6 Representative examples of alkoxy groups include, but are not limited to, methoxy, ethoxy, propoxy, isopropoxy, n-butoxy, isobutoxy, tert-butoxy, pentyloxy, hexyloxy, -CH 2 -OCH 3 Etc. The alkoxy groups may be optionally substituted with one or more (such as 1 to 3) identical or different substituents.
The term "alkyleneoxy" refers to a divalent alkoxy group, e.g., -OCH 2 -、-OCH(CH 3 )CH 2 -、-OCH 2 CH 2 O-、-CH 2 CH 2 O-, and the like. The alkyleneoxy groups may be optionally substituted with one or more (such as 1 to 3) identical or different substituents.
The term "fused ring" or "fused ring" refers to a ring system formed by two or more cyclic structures sharing two adjacent atoms with each other.
The term "spiro" refers to a ring system formed by two or more cyclic structures sharing one ring atom with each other.
The term "bridged ring" refers to a ring system formed by two or more cyclic structures sharing two atoms that are not directly attached to each other.
The term "cycloalkyl" refers to a saturated or unsaturated, non-aromatic, monocyclic or polycyclic (such as bicyclic) hydrocarbon cyclic group including, but not limited to, monocyclic alkyl (such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl) and bicyclic alkyl, including spiro, fused (fused) or bridged ring systems (i.e., spiro alkyl, fused (fused) alkyl and bridged cycloalkyl groups, such as bicyclo [1.1.1] pentyl, bicyclo [2.2.1] heptyl, and the like). In the present invention, cycloalkyl groups may be optionally substituted with one or more (such as 1 to 3) identical or different substituents. The carbon atom on the cycloalkyl group is optionally substituted with an oxo (oxo) group (i.e., forming c=o).
The term "cycloalkylene" refers to a cycloalkyl group, as defined herein, having two monovalent radical centers resulting from the removal of two hydrogen atoms from the same carbon atom or two different carbon atoms of the parent cycloalkyl group. Typical cycloalkylene groups include, but are not limited to, cyclopropylene, cyclobutylene, cyclopentylene, cyclohexylene, cycloheptylene, cyclooctylene, cyclononylene, cyclohexenylene, and the like.
The term "3-7 membered cycloalkyl" refers to cycloalkyl having 3 to 7 ring-forming carbon atoms, which may be a monocyclic alkyl group, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl or cycloheptyl, and may also be a bicyclic alkyl group, such as C 5-7 Spirocycloalkyl, C 5-7 Bridged cycloalkyl or C 4-7 Condensed ring alkyl.
The term "C 3-8 Cycloalkyl "means cycloalkyl having 3 to 8 ring-forming carbon atoms, e.g. C 3-6 Cycloalkyl, which may be a monocycloalkyl, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl or cyclooctyl, or bicycloalkyl, such as C 3-8 Spirocycloalkyl, C 3-8 Bridged cycloalkyl, C 3-8 Condensed ring alkyl, C 3-6 Spirocycloalkyl, C 3-6 Bridged cycloalkyl or C 3-6 Condensed ring alkyl.
The term "aryl" refers to an all-carbon monocyclic or fused-polycyclic aromatic group having a conjugated pi-electron system. As herein described The term "C", as used 6-12 Aryl "means aryl containing 6 to 12 carbon atoms, preferably C 6-10 Aryl is preferably phenyl or naphthyl. Aryl groups optionally substituted with one or more (such as 1 to 3) identical or different substituents (e.g. halogen, OH, CN, NO 2 、C 1-6 Alkyl, etc.) substitution.
The term "arylene" refers to an aryl group as defined herein having two monovalent radical centers resulting from the removal of two hydrogen atoms from the same carbon atom or two different carbon atoms of the parent aryl group. Typical arylene groups include, but are not limited to, phenylene and naphthylene.
The term "aryl-cycloalkyl" refers to a fused ring group formed by aryl and cycloalkyl groups (e.g., monocycloalkyl) sharing two adjacent atoms with each other, wherein the point of attachment to the other groups may be on the aryl or on the cycloalkyl. The term "9-12 membered arylcycloalkyl" means arylcycloalkyl containing a total of 9-12 ring atoms, e.g. benzocyclopentyl, benzocyclohexyl, e.g
The term "heterocyclyl" refers to a mono-or polycyclic (e.g., parallel, spiro, or bridged) group having 2 or more (e.g., 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or 14) carbon atoms, and one or more (e.g., 1, 2, 3, or 4) heteroatoms, including but not limited to oxygen, nitrogen, or sulfur atoms, the carbon and heteroatoms on the heterocyclyl being optionally substituted with oxo groups (e.g., to form c= O, S (=o) or S (=o) 2 )。
The term "heterocyclylene" refers to a heterocyclic group as defined herein having two monovalent radical centers derived from the same carbon atom or two different carbon atoms, from one carbon atom and one heteroatom or from two heteroatoms of the parent heterocyclic group, removed two hydrogen atoms.
The term "3-14 membered heterocyclyl" means heterocyclyl containing 3-14 ring atoms including, but not limited to, 4-10 membered heterocyclyl, 4-8 membered heterocyclyl, 4-7 membered heterocyclyl, 5-6 membered heterocyclyl3-8 membered heterocyclyl, 3-7 membered heterocyclyl, 4-7 membered nitrogen containing heterocyclyl, 4-7 membered oxygen containing heterocyclyl, 4-7 membered sulfur containing heterocyclyl, 5-6 membered nitrogen containing heterocyclyl, 5-6 membered oxygen containing heterocyclyl, 5-6 membered sulfur containing heterocyclyl, and the like, each of which optionally also contains one or more additional heteroatoms selected from oxygen, nitrogen and sulfur. Examples of 3-14 membered heterocyclyl groups include, but are not limited to, oxiranyl, aziridinyl, azetidinyl, oxetanyl, tetrahydrofuranyl, pyrrolidinyl, pyrrolidonyl, imidazolidinyl, pyrazolidinyl, tetrahydropyranyl, piperidinyl, morpholinyl, dithianyl, thiomorpholinyl, piperazinyl, trithianyl, Etc.
In the invention, the heterocyclic group also comprises a parallel ring structure, and the connection point of the parallel ring structure and other groups can be on any ring in the parallel ring structure. Thus, the heterocyclyl of the present invention also includes, but is not limited to, heterocyclyl-heterocyclo-heterocyclyl, heterocyclyl-cycloalkyl, mono-heterocyclyl-mono-heterocyclyl, mono-heterocyclo-monocycloalkyl, e.g., 3-7 membered (mono) heterocyclyl-3-7 membered (mono) heterocyclyl, 3-7 membered (mono) heterocyclyl-mono-cycloalkyl, 3-7 membered (mono) heterocyclyl-C 4-6 (mono) cycloalkyl groups and the like, examples of which include, but are not limited to, pyrrolidinyl-cyclopropyl, cyclopentylazetidinyl, pyrrolidinyl-cyclobutyl, pyrrolidinyl-pyrrolidinyl, pyrrolidinyl-piperidinyl, pyrrolidinyl-piperazinyl, piperidinyl-morpholinyl,Etc.
In the present invention, the heterocyclic group also includes bridged heterocyclic groups and spiro heterocyclic groups.
The term "bridged heterocyclyl" refers to a cyclic structure containing one or more (e.g., 1, 2, 3, or 4) heteroatoms (e.g., oxygen, nitrogen, or sulfur atoms) formed by two saturated rings sharing two ring atoms that are not directly connected, including but not limited to 7-10 membered bridged heterocyclyl, 8-10 membered bridged heterocyclyl,7-10 membered nitrogen-containing bridged heterocyclic group, 7-10 membered oxygen-containing bridged heterocyclic group, 7-10 membered sulfur-containing bridged heterocyclic group, etc., for example Etc. The "nitrogen-containing bridged heterocycle", "oxygen-containing bridged heterocycle", "sulfur-containing bridged heterocycle" each optionally also contains one or more additional heteroatoms selected from oxygen, nitrogen and sulfur.
The term "spiroheterocyclyl" refers to a cyclic structure containing one or more (e.g., 1, 2, 3, or 4) heteroatoms (e.g., oxygen, nitrogen, or sulfur atoms) formed by two or more saturated rings sharing one ring atom, including but not limited to 5-10 membered spiroheterocyclyl, 6-10 membered nitrogen-containing spiroheterocyclyl, 6-10 membered oxygen-containing spiroheterocyclyl, 6-10 membered sulfur-containing spiroheterocyclyl, and the like, e.g. Etc. The "nitrogen-containing spiroheterocycle", "oxygen-containing spiroheterocycle" and "sulfur-containing spiroheterocycle" each optionally also contain one or more additional heteroatoms selected from oxygen, nitrogen and sulfur. The term "6-10 membered nitrogen-containing spiroheterocyclyl" refers to a spiroheterocyclyl containing a total of 6-10 ring atoms, at least one of which is a nitrogen atom. />
The term "aryl-heterocyclyl" refers to a cyclic group formed by aryl and heterocyclyl sharing two adjacent carbon atoms with each other (wherein aryl and heterocyclyl are as defined above) and the point of attachment to the other group may be on the aryl or heterocyclyl. For example, as used herein The term "9-12 membered arylheterocyclo" means a group containing an arylheterocyclo group having 9-12 ring atoms in total, including, but not limited to, 9-10 membered benzoheterocyclyl groups, such as benzo 5-8 membered heterocyclyl groups, such as benzo 5-6 membered mono-heterocyclyl groups, benzo 5-6 membered nitrogen-containing mono-heterocyclyl groups, benzo 5-6 membered oxygen-containing mono-heterocyclyl groups, benzo 5-6 membered sulfur-containing heterocyclyl groups, and the like. The "nitrogen-containing heterocyclyl", "oxygen-containing heterocyclyl" and "sulfur-containing heterocyclyl" each optionally also contain one or more additional heteroatoms selected from oxygen, nitrogen and sulfur. The carbon and heteroatoms on the heterocyclyl are optionally substituted with oxo groups (e.g. to form c= O, S (=o) or S (=o) 2 ). Examples of aryl heterocycles include, but are not limited to: indazolyl group,
The term "heteroaryl" refers to a monocyclic or polycyclic aromatic group containing one or more heteroatoms either the same or different, including monocyclic heteroaryl groups and bicyclic or polycyclic ring systems containing at least one heteroaromatic ring (an aromatic ring system containing at least one heteroatom) which may have 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 ring atoms, for example 5, 6, 7, 8, 9 or 10 ring atoms. The heteroatom may be oxygen, nitrogen or sulfur. The carbon and heteroatoms on the heteroaryl group being optionally substituted with oxo groups (e.g. to form c= O, S (=o) or S (=o) 2 )。
The term "heteroarylene" refers to a heteroaryl group as described above having two hydrogen atoms removed from the same carbon atom or two different carbon atoms of the parent heteroaryl group or two monovalent radical centers resulting from the removal of one hydrogen atom from a carbon atom and one hydrogen atom from a nitrogen atom.
The term "5-10 membered heteroaryl" means heteroaryl containing 5 to 10 ring atoms, including 5-6 membered heteroaryl, 5-6 membered mono-heteroaryl, 5-10 membered nitrogen containing heteroaryl, 5-10 membered oxygen containing heteroaryl, 5-10 membered sulfur containing heteroaryl, 5-6 membered nitrogen containing heteroaryl, 5-6 membered oxygen containing heteroaryl, 5-6 membered sulfur containing heteroaryl, 5-6 membered nitrogen mono-heteroaryl, 5-6 membered oxygen containing mono-heteroaryl, 5-6 membered sulfur containing mono-heteroaryl, and the like. The "nitrogen-containing heteroaryl", "oxygen-containing heteroaryl", "sulfur-containing heteroaryl", "nitrogen-containing mono-heteroaryl", "oxygen-containing mono-heteroaryl" and "sulfur-containing mono-heteroaryl" each optionally also contain one or more other heteroatoms selected from oxygen, nitrogen and sulfur. Examples of 5-10 membered heteroaryl groups include, but are not limited to, thienyl, furyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, triazolyl, tetrazolyl, oxadiazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, and the like, as well as 5-10 membered bicyclic groups containing these groups.
In the present invention, the term "heteroaryl" encompasses fused ring structures, i.e., a fused ring structure in which a heteroaryl ring (e.g., a mono-heteroaryl) may share two adjacent atoms with an aryl group (e.g., a monocyclic aryl group, such as phenyl), a heterocyclyl group (e.g., a mono-heterocyclyl group), a cycloalkyl group (e.g., a mono-cycloalkyl group), or another heteroaryl ring (e.g., another mono-heteroaryl) may form a fused ring structure in which the point of attachment to the other group may be on any heteroaryl ring or on any other ring, including, but not limited to, (mono) heteroaryl-mono-heteroaryl, (mono) aryl, (mono) heteroaryl-mono-heterocyclyl, or (mono) heteroaryl-mono-cycloalkyl, such as 5-6 membered (mono) heteroaryl-phenyl, 5-6 membered (mono) heteroaryl-5-6 membered (mono) heterocyclyl, or 5-6 membered (mono) heteroaryl-C 4-6 (mono) cycloalkyl (e.g., 5-6 membered heteroaryl-cyclobutyl, 5-6 membered heteroaryl-cyclopentyl, or 5-6 membered heteroaryl-cyclohexyl), examples of which include, but are not limited to, indolyl, isoindolyl, indazolyl, benzimidazole, quinolinyl, isoquinolinyl, Etc.
The term "heteroaryl" embraces aryl-containing and ring structures also known as "arylfused heteroaryl" groups, which are fused ring groups formed from an aryl group (e.g., a monocyclic aryl group, such as phenyl) and a heteroaryl group (e.g., a mono-heteroaryl group, such as a 5-6 membered mono-heteroaryl group), which may be attached to an aromatic ring or to a heteroaromatic ring at the point of attachment to other groups. The "aryl-heteroaryl" includes, but is not limited to, monocyclic aryl-mono-heteroaryl. The term "9-12 membered arylalkylheteroaryl" refers to arylalkylheteroaryl groups containing a total of 9-12 ring members, such as benzo 5-6 membered nitrogen containing mono-heteroaryl.
The term "heteroaryl" embraces cycloalkyl-containing fused ring structures also known as "heteroarylfused cycloalkyl" and is heteroaryl (e.g., mono-heteroaryl, e.g., 5-6 membered mono-heteroaryl) and cycloalkyl (e.g., C 4-6 Cycloalkyl) and the point of attachment to other groups may be on the heteroaryl ring or on the cycloalkyl. The "heteroarylcycloalkyl" includes, but is not limited to, mono-heteroaryl monocycloalkyl. The term "9-10 membered heteroarylcycloalkyl" refers to heteroarylcycloalkyl containing a total of 9-10 ring atoms, e.g., 4-6 membered nitrogen containing monoheteroaryl and C 4-6 A monocyclic alkyl group.
The term "substitution" means that one or more (e.g., 1, 2, 3, or 4) hydrogens on the designated atom are replaced with a selection from the indicated group, provided that the designated atom's normal valency is not exceeded, and that the substitution forms a stable compound. Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.
If a substituent is described as "optionally substituted with … …," the substituent may be (1) unsubstituted or (2) substituted. If a carbon of a substituent is described as optionally substituted with one or more of the list of substituents, one or more hydrogens on the carbon (to the extent any hydrogens are present) may each be replaced with an independently selected optional substituent. If the nitrogen of a substituent is described as optionally substituted with one or more of the list of substituents, then one or more hydrogens on the nitrogen (to the extent any hydrogens are present) may each be replaced with an independently selected optional substituent.
If substituents are described as "independently selected from" a group, each substituent is selected independently of the other. Thus, each substituent may be the same as or different from another (other) substituent.
As used herein, the term "one or more" means 1 or more than 1, e.g., 2, 3, 4, 5, 6, 7, 8, 9, or 10, under reasonable conditions.
As used herein, unless indicated, the point of attachment of a substituent may be from any suitable position of the substituent.
The invention also includes all pharmaceutically acceptable isotopic compounds of the present invention, which are identical to those of the present invention except that one or more atoms are replaced by an atom having the same atomic number but an atomic mass or mass number different from the atomic mass or mass number prevailing in nature. Examples of isotopes suitable for inclusion in the compounds of the invention include, but are not limited to, isotopes of hydrogen (e.g 2 H、 3 H) The method comprises the steps of carrying out a first treatment on the surface of the Isotopes of carbon (e.g 11 C、 13 C, C is a metal alloy 14 C) The method comprises the steps of carrying out a first treatment on the surface of the Isotopes of chlorine (e.g 36 Cl); isotopes of fluorine (e.g 18 F) The method comprises the steps of carrying out a first treatment on the surface of the Isotopes of iodine (e.g 123 I, I 125 I) The method comprises the steps of carrying out a first treatment on the surface of the Isotopes of nitrogen (e.g 13 N is N 15 N); isotopes of oxygen (e.g 15 O、 17 O and O 18 O); isotopes of phosphorus (e.g 32 P) is as follows; isotopes of sulfur (e.g 35 S). The term "stable isotope derivative" refers to a stable compound in which one or more atoms in the compound of the present invention are replaced by atoms having the same atomic number but an atomic mass or mass number different from the atomic mass or mass number prevailing in nature.
The term "stereoisomer" refers to an isomer of a compound formed as a result of the compound containing at least one asymmetric center. In compounds having one or more (e.g., 1, 2, 3, or 4) asymmetric centers, they can produce racemic mixtures, single enantiomers, diastereomeric mixtures, and individual diastereomers. Specific individual molecules may also exist as geometric isomers (cis/trans). The compounds of the invention may exist as a mixture of two or more different structural forms (commonly referred to as tautomers) in rapid equilibrium. Representative examples of tautomers include keto-enol tautomers, phenol-keto tautomers, nitroso-oxime tautomers, imine-enamine tautomers, and the like. For example, nitroso-oximes may exist in solution in equilibrium in the following tautomeric forms:
It is to be understood that the scope of the present application encompasses all such isomers in any proportion (e.g., 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%) or mixtures thereof.
Unless otherwise indicated, the compounds of the present application are intended to exist as stereoisomers (which include cis and trans isomers), optical isomers (e.g., R and S enantiomers), diastereomers, geometric isomers, rotamers, conformational isomers, atropisomers, or mixtures thereof. The compounds of the present application may exhibit more than one type of isomerism and consist of mixtures thereof (e.g., racemic mixtures and diastereomeric pairs).
The present application encompasses all possible crystalline forms or polymorphs of the compounds of the present application, which may be single polymorphs or mixtures of any ratio of more than one polymorphs. It will also be appreciated that certain compounds of the application may exist in free form for use in therapy or, where appropriate, in the form of pharmaceutically acceptable derivatives thereof. In the present application, pharmaceutically acceptable derivatives include, but are not limited to: pharmaceutically acceptable salts, solvates, metabolites or prodrugs thereof, which, upon administration to a patient in need thereof, are capable of providing the compounds of the application or metabolites or residues thereof, either directly or indirectly. Thus, when reference is made herein to "a compound of the application" it is also intended to encompass the various derivative forms of the compounds described above.
Pharmaceutically acceptable salts of the compounds of the present invention include acid addition salts and base addition salts thereof. Such as hexafluorophosphate, meglumine salt, and the like. For a review of suitable salts, see Stahl and Wermuth, "Handbook of Pharmaceutical Salts: properties, selection, and Use" (Wiley-VCH, 2002).
By "pharmaceutically acceptable carrier" is meant a diluent, adjuvant, excipient or vehicle with which the therapeutic agent is administered, and which is suitable for contacting the tissues of humans and/or other animals within the scope of sound medical judgment without undue toxicity, irritation, allergic response, or other problem or complication commensurate with a reasonable benefit/risk ratio.
Pharmaceutically acceptable carriers that may be used in the pharmaceutical compositions of the present invention include, but are not limited to, sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. When the pharmaceutical composition is administered intravenously, water is an exemplary carrier. Physiological saline and aqueous solutions of glucose and glycerol can also be used as liquid carriers, in particular for injections. Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, maltose, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene glycol, water, ethanol and the like. The composition may also contain minor amounts of wetting agents, emulsifying agents, or pH buffering agents, as desired. Oral formulations may contain standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, and the like. Examples of suitable pharmaceutically acceptable carriers are described in Remington's Pharmaceutical Sciences (1990).
The pharmaceutical compositions of the present invention may act systematically and/or locally. For this purpose, they may be administered by a suitable route, for example by injection, intravenously, intra-arterially, subcutaneously, intraperitoneally, intramuscularly or transdermally; or by oral, buccal, nasal, transmucosal, topical, in the form of an ophthalmic formulation or by inhalation.
For these routes of administration, the pharmaceutical compositions of the present invention may be administered in suitable dosage forms.
Such dosage forms include, but are not limited to, tablets, capsules, lozenges, hard candies, powders, sprays, creams, ointments, suppositories, gels, pastes, lotions, ointments, aqueous suspensions, injectable solutions, elixirs, syrups.
The term "effective dose" as used herein refers to the amount of a compound that, upon administration, will alleviate to some extent one or more symptoms of the condition being treated.
The dosing regimen may be adjusted to provide the best desired response. For example, a single bolus may be administered, several divided doses may be administered over time, or the doses may be proportionally reduced or increased as indicated by the urgent need for a therapeutic situation. It is noted that the dosage value may vary with the type and severity of the condition to be alleviated, and may include single or multiple doses. It is further understood that for any particular individual, the particular dosage regimen will be adjusted over time according to the individual needs and the professional judgment of the person administering or supervising the administration of the compositions.
The amount of the compound of the invention administered will depend on the severity of the individual, disorder or condition being treated, the rate of administration, the disposition of the compound and the discretion of the prescribing physician. Generally, an effective dose is about 0.0001 to about 50mg, for example about 0.01 to about 10 mg/kg/day per kg body weight per day (single or divided administration). For a 70kg human, this amounts to about 0.007 mg/day to about 3500 mg/day, for example about 0.7 mg/day to about 700 mg/day. In some cases, dosage levels not higher than the lower limit of the aforementioned range may be sufficient, while in other cases larger doses may still be employed without causing any adverse side effects, provided that the larger dose is first divided into several smaller doses for administration throughout the day.
The compounds of the present invention may be present in the pharmaceutical composition in an amount or amount of about 0.01mg to about 1000mg.
As used herein, unless otherwise indicated, the term "treating" means reversing, alleviating, inhibiting the progression of, or preventing, a disorder or condition to which such term applies, or one or more symptoms of such disorder or condition.
As used herein, "individual" includes human or non-human animals. Exemplary human individuals include human individuals (referred to as patients) or normal individuals suffering from a disease (e.g., a disease described herein). "non-human animals" in the context of the present invention include all vertebrates, such as non-mammals (e.g., birds, amphibians, reptiles) and mammals, such as non-human primates, domestic animals and/or domesticated animals (e.g., sheep, dogs, cats, cows, pigs, etc.).
The compounds of the invention may be present in the form of solvates (preferably hydrates) wherein the compounds of the invention comprise a polar solvent as a structural element of the compound lattice, in particular for example water, methanol or ethanol. The polar solvent, in particular water, may be present in stoichiometric or non-stoichiometric amounts.
Also included within the scope of the invention are metabolites of the compounds of the invention, i.e., substances that form in vivo upon administration of the compounds of the invention. Such products may result from, for example, oxidation, reduction, hydrolysis, amidation, deamidation, esterification, degreasing, enzymatic hydrolysis, etc. of the compound being administered. Accordingly, the present invention includes metabolites of the compounds of the present invention, including compounds obtained by a method of contacting a compound of the present invention with a mammal for a time sufficient to produce the metabolites thereof.
The invention further includes within its scope prodrugs of the compounds of the invention, which are certain derivatives of the compounds of the invention which may themselves have little or no pharmacological activity, and which, when administered into or onto the body, are converted to the compounds of the invention having the desired activity by, for example, hydrolytic cleavage. Typically such prodrugs will be functional derivatives of the compounds which are readily convertible in vivo into the desired therapeutically active compound. Additional information regarding the use of prodrugs can be found in "Pro-drugs as Novel Delivery Systems", vol.14, ACS Symposium Series (T. Higuchi and V. Stilla) and "Bioreversible Carriers in Drug Design," Pergamon Press,1987 (E. B. Roche eds., american Pharmaceutical Association). Prodrugs of the invention may be prepared, for example, by replacing the appropriate functional groups present in the compounds of the invention with certain moieties known to those skilled in the art as "pro-moieties" (e.g. "Design of Prodrugs", described in h. Bundegaard (Elsevier, 1985) ".
The invention also encompasses compounds of the invention containing a protecting group. During any process for preparing the compounds of the present invention, it may be necessary and/or desirable to protect sensitive or reactive groups on any of the molecules of interest, thereby forming a chemically protected form of the compounds of the present invention. This can be achieved by conventional protecting groups, for example, in Protective Groups in Organic Chemistry, ed.J.F.W.McOmie, plenum Press,1973; and those described in T.W.Greene & P.G.M.Wuts, protective Groups in Organic Synthesis, john Wiley & Sons,1991, which are incorporated herein by reference. The protecting group may be removed at a suitable subsequent stage using methods known in the art.
As used hereinThe bond in the structural formula is a single bond or a double bond.
As used hereinIndicating that the double bond is not located at a certain position, but still ensures that the ring in which it is located has aromaticity.
As used hereinIndicating the presence or absence of a bond in the structural formula.
As used herein, "room temperature" refers to 15-30deg.C.
Preparation method
Synthesis method 1 of compound II-A
Wherein R is 1 、R 2 、R 3 、R 4 、R 5 And L is as defined above for formula I.
The first step: the compound II-1 is substituted under alkaline conditions to form an intermediate II-2.
The solvent used was DMF, DMSO, THF, CH 3 CN, DCM, etc., and the base used is triethylamine, N-diisopropylethylamine, potassium carbonate, potassium tert-butoxide, sodium hydroxide, etc., at a temperature of-20 to 180 ℃.
And a second step of: the compound II-2 is substituted under alkaline conditions to form intermediate II-3.
The solvent used was DMF, DMSO, THF, CH 3 CN, DCM, etc., and the base used is triethylamine, N-diisopropylethylamine, potassium carbonate, potassium tert-butoxide, sodium hydroxide, etc., at 20-150 ℃.
And a third step of: compound II-3 is reduced to intermediate II-4.
The reducing agent is Zn powder/ammonium formate, zn powder/ammonium chloride, sodium hydrosulfite, fe powder/ammonium formate, etc., the solvent is methanol, ethanol, THF, etc., and the temperature is 20-110 ℃.
Fourth step: the compound II-4 is subjected to condensation reaction to generate an intermediate II-5.
The condensation reagent is HATU, HBTU, TBTU, EDCI, the solvent is THF, DMF, 1, 4-dioxane, etc., and the temperature is 20-100 ℃.
Fifth step: compound II-5 is cyclized under basic conditions to form intermediate II-6.
The base is triethylamine, N-diisopropylethylamine, potassium carbonate, potassium tert-butoxide, sodium hydroxide, etc., the solvent is MeOH, etOH, THF, 1, 4-dioxane, etc., and the temperature is 20-150 ℃.
Sixth step: the compound II-6 is subjected to coupling reaction under alkaline conditions to generate an intermediate II-7.
The coupling reagent is tetrakis (triphenylphosphine) palladium (0), tris (dibenzylideneacetone) dipalladium (0) and the like, and the base is sodium bis (trimethylsilyl) amide or tBuOK, tBuONa, tBuOLi, naH, naOH, KOH, cs 2 CO 3 、K 2 CO 3 、Na 2 CO 3 Etc., the solvent is toluene, xylene, THF, DME, 1, 4-dioxane, DMF, NMP, DMSO, etc., and the temperature is0 ℃ to 160 ℃.
Seventh step: the intermediate II-7 is subjected to deprotection reaction under acidic condition to generate a compound II-A.
The acid is trifluoroacetic acid, trifluoromethanesulfonic acid, acetic acid, hydrochloric acid, sulfuric acid, nitric acid, etc., and the solvent is DCM, 1, 4-dioxane, methanol, EA, etc., or the solvent is not needed, and the solvent is directly used for dissolving, and the temperature is between-20 ℃ and 100 ℃.
Synthesis method 2 of compound II-A
The first step: the compound II-3 is substituted under alkaline conditions to form intermediate II-8.
The solvent used was DMF, DMSO, THF, CH 3 CN, DCM, etc., and the base used is cesium carbonate, triethylamine, N-diisopropylethylamine, potassium carbonate, potassium tert-butoxide, sodium hydroxide, butyllithium, etc., at 20℃to 180 ℃.
And a second step of: compound II-8 is reduced to intermediate II-9.
The reducing agent is Zn powder/ammonium formate, zn powder/ammonium chloride, sodium hydrosulfite, fe powder/ammonium formate, etc., the solvent is methanol, ethanol, THF, etc., and the temperature is 20-110 ℃.
And a third step of: the compound II-9 is subjected to condensation reaction to generate an intermediate II-10.
The condensation reagent is HATU, HBTU, TBTU, EDCI, the solvent is THF, DMF, 1, 4-dioxane, etc., and the temperature is 20-100 ℃.
Fourth step: compound II-10 is cyclized under basic conditions to form intermediate II-11.
The base is triethylamine, N-diisopropylethylamine, potassium carbonate, potassium tert-butoxide, sodium hydroxide, etc., the solvent is MeOH, etOH, THF, 1, 4-dioxane, etc., and the temperature is 20-150 ℃.
Fifth step: the intermediate II-11 is subjected to deprotection reaction under acidic condition to generate a compound II-A.
The acid is trifluoroacetic acid, boron tribromide, boron trichloride, trifluoromethanesulfonic acid, acetic acid, hydrochloric acid, sulfuric acid, nitric acid, etc., and the solvent is 1, 4-dioxane, methanol, EA, etc., or the solvent is not needed, and the temperature is-20 ℃ to 100 ℃ directly.
Advantageous effects
The compound has obvious agonistic activity to NLRP3 and a signal path thereof, has no obvious toxic or side effect, and can be used for treating diseases (such as tumors) related to NLRP3 inflammatory body activity.
Examples
The invention is further described below in connection with examples, which are not intended to limit the scope of the invention.
Abbreviations herein have the following meanings:
the structure of the compound is characterized by nuclear magnetic resonance spectrum 1 H NMR) or Mass Spectrometry (MS).
The reaction was monitored by silica gel Thin Layer Chromatography (TLC) or LC-MS.
The microwave reaction was performed using a biotageinitiator+microwave reactor.
Column chromatography generally uses 200-300 mesh silica gel (Qingdao ocean) as the stationary phase. The system of the eluent comprises: a: dichloromethane and methanol; b: the volume ratio of PE and EA, the solvent is adjusted according to the polarity of the compound.
In the following examples, the reaction temperature was room temperature (15℃to 30 ℃) unless otherwise specified.
The reagents used in the present application are available from Acros Organics, aldrich Chemical Company, tertbe chemistry, etc.
Example 1:3- (4-amino-1-methyl-6- ((3-methylbenzyl) amino) -1H-imidazo [4,5-c ] pyridin-2-yl) propan-1-ol (Compound 1)
The first step: 2, 6-dichloro-N-methyl-3-nitropyridin-4-amine (Compound 1 b)
Compound 1a (5.0 g,24.15 mmol), K 2 CO 3 (6.66 g,48.30 mmol) and methyl iodide (8.29 g,60.375 mmol) were added to acetonitrile (40 mL), heated to 80℃and stirred for reaction for 12h. After the completion of the reaction, the mixture was filtered through celite, and the filtrate was dried by spin-drying to give Compound 1B (4.80 g) by flash column chromatography (eluent system B).
MS(ESI,m/z):222.0[M+H] + .
And a second step of: 6-chloro-N 4 -methyl-3-nitro-N 2 - (2, 4-trimethylpent-2-yl) pyridine-2, 4-diamine (compound 1 c)
Compound 1b (4.80 g,21.71 mmol), 2, 4-trimethylpentan-2-amine (5.61 g,43.42 mmol) and triethylamine (6.58 mg,65.13 mmol) were added to dichloromethane (40 mL), heated to 40℃and reacted under stirring for 12h. After the reaction, the solvent was dried by flash column chromatography (eluent system B) to give compound 1c (5.10 g).
MS(ESI,m/z):315.1[M+H] + .
And a third step of: 6-chloro-N 4 -methyl-N 2 - (2, 4-trimethylpent-2-yl) pyridine-2, 3, 4-triamine (compound 1 d)
Compound 1c (5.0 g,15.87 mmol), zinc powder (5.17 g,79.35 mmol), HCOONH 4 (3.0 g,47.16 mmol) was added to MeOH (30 mL) and the reaction stirred at room temperature for 3h. After the reaction was completed, the mixture was filtered through celite, and the filtrate was dried by spin-drying to obtain Compound 1d (3.60 g) by flash column chromatography (eluent system B).
MS(ESI,m/z):270.2[M+H] + .
Fourth step: 4- (benzyloxy) -N- (6-chloro-4- (methylamino) -2- ((2, 4-trimethylpent-2-yl) amino) pyridin-3-yl) butanamide (Compound 1 e)
Compound 1d (3.60 g, 128.04. Mu. Mol), 4-benzyloxybutyric acid (5.61 g,43.42 mmol), HATU (6.58 mg,65.13 mmol), DIPEA (5.17 g,79.35 mmol) in DMF (15 mL), N 2 The reaction was stirred at room temperature under protection for 12h. After the completion of the reaction, the reaction mixture was poured into ice water and extracted with EA. The organic phase was concentrated and flash column chromatography (eluent system B) gave compound 1e (5.58 g).
MS(ESI,m/z):461.3[M+H] + .
Fifth step: 2- (3- (benzyloxy) propyl) -6-chloro-1-methyl-N- (2, 4-trimethylpent-2-yl) -1H-imidazo [4,5-c ] pyridin-4-amine (Compound 1 f)
Compound 1e (5.50 g,11.96 mmol), naOH (1.44 g,35.88 mmol) were added to ethanol (40 mL), N 2 Heating to 90 ℃ under protection, and stirring to react for 12h. After the completion of the reaction, the mixture was filtered through celite, and the filtrate was dried by spin-drying to give compound 1f (4.20 g) by flash column chromatography (eluent system B).
MS(ESI,m/z):443.2[M+H] + .
Sixth step: 2- (3-Benzyloxypropyl) -1-methyl-N 6 - (3-methylbenzyl) -N 4 - (2, 4-trimethylpent-2-yl) -1H-imidazo [4,5-c]Pyridine-4, 6-diamine (Compound 1 g)
DavePhos (17.74 mg, 45.14. Mu. Mol), pd 2 (dba) 3 (20.65 mg, 22.57. Mu. Mol), tBuOK (151.97 mg,1.35 mmol) was added to a solution of 3-methylbenzylamine (82.06 mg, 677.17. Mu. Mol), compound 1f (200 mg, 451.44. Mu. Mol) in 1, 4-dioxane (3 mL), and then nitrogen was bubbled for two minutes to remove oxygen. The reaction system is placed in a microwave reactor and heated to 100 ℃ for reaction for 1h. Insoluble matter was removed by filtration, and after concentrating the mother liquor, the mixture was purified by preparative plate separation by silica gel thin layer chromatography (eluent system A) to obtain 1g (120 mg) of compound.
Seventh step: 3- (4-amino-1-methyl-6- ((3-methylbenzyl) amino) -1H-imidazo [4,5-c ] pyridin-2-yl) propan-1-ol (Compound 1)
Boron tribromide (27.07 mg, 108.30. Mu. Mol) was added to a solution of compound 1g (30.00 mg, 72.20. Mu. Mol) in DCM (2 mL) and then left to react at room temperature for 5min. The reaction was quenched with water and ph=8 was adjusted with potassium carbonate. The organic phase was dried, concentrated to give crude product, and purified by Prep-HPLC to give compound 1 (10 mg) by EA (10 mL).
Prep-HPLC conditions:
instrument model: agilent 1260; chromatographic column: waters XBridge prep C18 (19 mm. Times.150 mm. Times.5.0 μm); mobile phase: acetonitrile/0.05% NH 4 HCO 3 An aqueous solution; elution gradient: acetonitrile 10% -90% in 0-16 min; flow rate: 24ml/min; detection wavelength: 214nm, 254nm, 280nm; compound collection time: 6.7-7.1min.
MS(ESI,m/z):326.2[M+H] + .
1 H NMR(400MHz,MeOD)δ7.18(dd,J=13.8,6.7Hz,3H),7.04(d,J=6.6Hz,1H),5.69(s,1H),4.35(s,2H),3.65(t,J=6.2Hz,2H),3.55(s,3H),2.91–2.86(m,2H),2.31(s,3H),2.00(dd,J=14.6,6.8Hz,2H).
Example 2:2- (3- (benzyloxy) propyl) -1-methyl-N 6 - (2-methylbenzyl) -1H-imidazo [4,5-c]Pyridine-4, 6-diamine (Compound 2)
The first step: 2- (3- (benzyloxy) propyl) -1-methyl-N 6 - (2-methylbenzyl) -N 4 - (2, 4-trimethylpent-2-yl) -1H-imidazo [4,5-c]Pyridine-4, 6-diamine (Compound 2 a)
Compound 1f (150 mg, 338.58. Mu. Mol), 2-methylbenzylamine (61.54 mg, 507.87. Mu. Mol), pd 2 (dba) 3 (15.49 mg, 16.93. Mu. Mol), davePhos (13.32 mg, 33.86. Mu. Mol), tBuOK (113.98 mg,1.02 mmol) were added to 1, 4-dioxane (5 mL), and nitrogen was bubbled for two minutes to remove oxygen, and the mixture was sealed. The reaction system is placed in a microwave reactor and heated to 100 ℃ for reaction for 1h. Filtration through celite, spin-drying of the filtrate followed by flash column chromatography (eluent system B) afforded compound 2a (110 mg).
MS(ESI,m/z):528.2[M+H] + .
And a second step of: 2- (3- (benzyloxy) propyl) -1-methyl-N 6 - (2-methylbenzyl) -1H-imidazo [4,5-c]Pyridine-4, 6-diamine (Compound 2)
TFA (2 mL) was added to a solution of compound 2a (110 mg, 473.72. Mu. Mol) in DCM (3 mL) and stirred for 4h at 25℃with no apparent starting material remaining as monitored by LC-MS, and compound 2 (9 mg) was isolated by Prep-HPLC and lyophilized.
Prep-HPLC conditions:
instrument model: agilent 1260; chromatographic column: waters SunFire Prep C18 OBD (19 mm. Times.150 mm. Times.5.0 μm); chromatographic column temperature: 40 ℃; flow rate: 28.0mL/min; detection wavelength: 254nm; elution gradient: (0 min:10% A,90% B;16min:90% A,10% B); mobile phase a:100% acetonitrile; mobile phase B:0.05% formic acid in water; compound collection time: 6.1-6.6min.
MS(ESI,m/z):416.2[M+H] + .
1 H NMR(400MHz,MeOD)δ7.35–7.10(m,9H),4.47(s,2H),4.35(s,2H),3.60(t,J=5.9Hz,2H),3.52(s,3H),2.90(t,J=7.5Hz,2H),2.38(s,3H),2.15–2.03(m,2H).
Example 3:3- (4-amino-1-methyl-6- (methyl (3-methylbenzyl) amino) -1H-imidazo [4,5-c ] pyridin-2-yl) propan-1-ol (Compound 3)
The first step: 2- (3- (benzyloxy) propyl) -N 6 1-dimethyl-N 6 - (3-methylbenzyl) -N 4 - (2, 4-trimethylpent-2-yl) -1H-imidazo [4,5-c]Pyridine-4, 6-diamine (Compound 3 a)
Compound 1f (120 mg, 270.87. Mu. Mol), N, 3-dimethylbenzylamine (43.95 mg, 325.04. Mu. Mol), pd 2 (dba) 3 (24.80 mg, 27.09. Mu. Mol), davePhos (21.32 mg, 54.17. Mu. Mol), tBuOK (91.18 mg, 812.60. Mu. Mol) was added to 1, 4-dioxane (3 mL), N 2 Heating to 100 ℃ under protection to react for 3.5h. After the reaction was completed, it was filtered through celite, and the filtrate was dried by spin-drying to give compound 3a (92 mg) by TLC (eluent system B).
MS(ESI,m/z):542.4[M+H] + .
And a second step of: 3- (4-amino-1-methyl-6- (methyl (3-methylbenzyl) amino) -1H-imidazo [4,5-c ] pyridin-2-yl) propan-1-ol (Compound 3)
Compound 3a (92 mg, 169.81. Mu. Mol) was dissolved in methylene chloride (10 mL), and after stirring at room temperature, a mixture of boron tribromide and DCM (30%, 0.2 mL) was added to the system. After the reaction was completed, the reaction was quenched by addition of anhydrous methanol (5 mL), and ph=7 was adjusted with saturated aqueous sodium bicarbonate solution. After concentration, the mixture was diluted with water and extracted three times with EA (20 mL. Times.3). The organic phases were combined and dried over anhydrous sodium sulfate, and compound 3 (18 mg) was isolated by Prep-HPLC.
Prep-HPLC conditions:
instrument model: waters 2489; chromatographic column: waters SunFire Prep C18 OBD (19 mm. Times.150 mm. Times.5.0 μm); chromatographic column temperature: 25 ℃; flow rate: 24.0mL/min; detection wavelength: 214nm, 254nm, 280nm; elution gradient: (0 min:30% A,70% B;4min:30% A,70% B;16.0min:90% A,10% B); mobile phase a:100% acetonitrile; mobile phase B:0.05% ammonium bicarbonate aqueous solution. Compound collection time: 7.6-8.1min.
MS(ESI,m/z):340.2[M+H] + .
1 H NMR(DMSO-d 6 ,400MHz)δ7.15(t,J=7.4Hz,1H),6.99(dd,J=12.5,8.3Hz,3H),5.76(s,1H),5.68(s,2H),4.76(s,2H),4.57(t,J=5.0Hz,1H),3.54–3.46(m,5H),2.86(s,3H),2.80–2.73(m,2H),2.25(s,3H),1.90–1.79(m,2H).
Example 4:3- (4-amino-6- [ (4-bromobenzyl) (methyl) amino ] -1-methyl-1H-imidazo [4,5-c ] pyridin-2-yl) propan-1-ol (Compound 4)
The first step: n (N) 6 - (4-bromobenzyl) -N 4 ,N 6 -dimethyl-3-nitro-N 2 - (2, 4-trimethylpent-2-yl) pyridine-2, 4, 6-triamine (compound 4 a)
Cesium carbonate (309.71 mg, 952.96. Mu. Mol) was added to a solution of N-methyl-4-bromobenzylamine (114.40 mg, 571.77. Mu. Mol), compound 1c (150 mg, 476.48. Mu. Mol) in acetonitrile (5 mL), heated to 80℃and stirred for 16h. The reaction mixture was diluted with EA (10 mL), and water (5 mL. Times.3) was added. After concentrating the organic phase by drying, compound 4a (100 mg) was obtained by separation and purification on a silica gel thin layer chromatography preparation plate.
And a second step of: n (N) 6 - (4-bromobenzyl) -N 4 ,N 6 -dimethyl-N 2 - (2, 4-trimethylpent-2-yl) pyridine-2, 3,4, 6-tetramine (compound 4 b)
Zinc powder (67.93 mg,1.05 mmol), ammonium chloride (55.91 mg,1.05 mmol) were added sequentially to a solution of compound 4a (100 mg, 209.02. Mu. Mol) in methanol (5 mL), and then reacted at room temperature for 5min until the color disappeared. Insoluble matter was removed by filtration, and the mother liquor was concentrated to give compound 4b (60 mg).
And a third step of: 4-benzyloxy-N- [6- [ (4-bromobenzyl) (methyl) amino ] -4- (methylamino) -2- [ ((2, 4-trimethylpent-2-yl) amino) pyridin-3-yl ] butanamide (compound 4 d)
HATU (42.37 mg, 111.50. Mu. Mol), DIPEA (57.64 mg, 445.99. Mu. Mol) were added successively to a solution of 4-benzyloxybutyric acid (25.99 mg, 133.80. Mu. Mol), compound 4b (50 mg, 111.50. Mu. Mol) in DMF (2 mL), and then reacted at 25℃for 4 hours. EA (10 mL) was added followed by a clean water wash (5 mL. Times.3). The EA layer was dried, concentrated, and purified by preparative plate on silica gel thin layer chromatography (eluent system A) to give compound 4d (50 mg).
MS(ESI,m/z):624.2[M+H] + .
Fourth step: 2- (3- (benzyloxy) propyl) -N 6 - (4-bromobenzyl) -N 6 1-dimethyl-N 4 - (2, 4-trimethylpent-2-yl) -1H-imidazo [4,5-c]Pyridine-4, 6-diamine (Compound 4 e)
Sodium hydroxide (12.81 mg, 320.18. Mu. Mol) was added to a solution of compound 4d (50 mg, 80.04. Mu. Mol) in ethanol (2 mL) and then heated to 90℃for 4h. The reaction was concentrated directly to dryness, then water (5 mL) was added. The mixture was extracted with EA (5 mL. Times.3), and the organic phase was dried and concentrated to give Compound 4e (30 mg).
MS(ESI,m/z):606.3[M+H] + .
Fifth step: 2- (3- (benzyloxy) propyl) -N 6 - (4-bromobenzyl) -N 6 1-dimethyl-1H-imidazo [4,5-c ]]Pyridine-4, 6-diamine (Compound 4 f)
TFA (1.00 mL) was added to a solution of compound 4e (30 mg, 49.45. Mu. Mol) in DCM (1.00 mL) and then allowed to react at room temperature for 2h. The solvent was removed by concentration under reduced pressure, then water (5 mL) was added and ph=8 was adjusted with sodium carbonate. The mixture was extracted with EA, and the organic phase was dried and concentrated to give Compound 4f (20 mg).
MS(ESI,m/z):496.1[M+H] + .
Sixth step: 3- [ 4-amino-6- [ (4-bromobenzyl) (methyl) amino ] -1-methyl-1H-imidazo [4,5-c ] pyridin-2-yl ] propan-1-ol (Compound 4)
Boron tribromide (25.28 mg, 101.13. Mu. Mol) was slowly added to a solution of compound 4f (20 mg, 40.45. Mu. Mol) in DCM (4 mL) and the reaction stirred at room temperature for 5min. The reaction was quenched with water and ph=8 was adjusted with sodium carbonate. The organic phase was dried, concentrated, extracted with EA (10 mL) and purified by Prep-HPLC to give compound 4 (3 mg).
Prep-HPLC conditions:
instrument model: agilent 1260; chromatographic column: waters SunFire Prep C18 OBD (19 mm. Times.150 mm. Times.5.0 μm); mobile phase: acetonitrile/0.05% NH 4 HCO 3 An aqueous solution; elution gradient: acetonitrile 30% -49.5% of 0-7.8 min; flow rate: 24ml/min. Detection wavelength: 214nm, 254nm, 280nm; compound collection time: 7.1-7.5min.
MS(ESI,m/z):406.1[M+H] + .
1 H NMR(400MHz,DMSO-d 6 )δ7.48(d,J=8.4Hz,2H),7.18(d,J=8.4Hz,2H),5.78(s,1H),5.73(s,2H),4.79(s,2H),4.60(t,J=5.1Hz,1H),3.55(s,3H),3.51(dd,J=11.4,6.1Hz,2H),2.87(s,3H),2.81–2.76(m,2H),1.90–1.82(m,2H).
Example 5:3- (4-amino-6- ((3-methoxybenzyl) (methyl) amino) -1-methyl-1H-imidazo [4,5-c ] pyridin-2-yl) propan-1-ol (Compound 5)
The first step: 2- (3- (benzyloxy) propyl) -N 6 1-dimethyl-N 4 - (2, 4-trimethylpent-2-yl) -1H-imidazo [4,5-c]Pyridine-4, 6-diamine (Compound 5 a)
Compound 1f (100 mg, 225.73. Mu. Mol), 5b (40.90 mg, 270.88. Mu. Mol), pd 2 (dba) 3 (20.65mg22.57. Mu. Mol), davePhos (17.76 mg, 45.14. Mu. Mol), tBuOK (50.65 mg, 451.46. Mu. Mol) was added to 1, 4-dioxane (5 mL), N 2 Heating to 100 ℃ under protection for reaction for 5h. After the completion of the reaction, the mixture was filtered through celite, and the filtrate was dried by spin-drying to give Compound 5a (70 mg) by flash column chromatography (eluent system B).
MS(ESI,m/z):558.4[M+H] + .
And a second step of: 3- (4-amino-6- ((3-methoxybenzyl) (methyl) amino) -1-methyl-1H-imidazo [4,5-c ] pyridin-2-yl) propan-1-ol (Compound 5)
Compound 5a (70 mg, 12.57. Mu. Mol) was dissolved in TFA (4 mL), N 2 Heating to 80 ℃ under protection, and stirring to react for 12h. After the reaction, the reaction mixture was concentrated to dryness under reduced pressure and redissolved in methanol (2 mL). With 1N NaOH/H 2 The O solution was adjusted to ph=9.0 and Prep-HPLC was performed to give compound 5 (14 mg).
Prep-HPLC conditions:
instrument model: agilent 1260; chromatographic column: waters SunFire Prep C18 OBD (19 mm. Times.150 mm. Times.5.0 μm); chromatographic column temperature: 25 ℃; flow rate: 20.0mL/min; detection wavelength: 214nm; elution gradient: (0 min:10% A,90% B;16.0min:90% A,10% B); mobile phase a:100% acetonitrile; mobile phase B:0.05% ammonium bicarbonate aqueous solution. Compound collection time: 6.25-6.90min.
MS(ESI,m/z):326.3[M+H] + .
1 H NMR(DMSO-d 6 ,400MHz)δ7.27-7.29(m,2H),7.20-7.22(m,3H),5.79(s,1H),5.72(s,2H),4.82(s,2H),4.63(s,1H),3.54(s,3H),3.50-3.52(m,2H),2.88(s,3H),2.78-2.81(m,2H),1.86-1.88(m,2H).
Example 6:3- (4-amino-1-methyl-6- (methyl (pyridin-3-ylmethyl) amino) -1H-imidazo [4,5-c ] pyridin-2-yl) propan-1-ol (Compound 6)
The first step: 2- (3- (benzyloxy) propyl) -N 6 1-dimethyl-N 6 - (pyridin-3-ylmethyl) -N 4 -(24, 4-trimethylpent-2-yl) -1H-imidazo [4,5-c]Pyridine-4, 6-diamine (Compound 6 g)
DavePhos (8.87 mg, 22.57. Mu. Mol), pd 2 (dba) 3 (10.33 mg, 11.29. Mu. Mol), tBuOK (75.98 mg, 677.17. Mu. Mol) was added to a solution of N-methyl-1- (pyridin-3-yl) methylamine (33.09 mg, 270.87. Mu. Mol), compound 1f (100 mg, 225.72. Mu. Mol) in 1, 4-dioxane (3 mL) and then nitrogen was bubbled for two minutes to remove oxygen. The reaction system is placed in a microwave reactor and heated to 100 ℃ for reaction for 1h. Insoluble matter was removed by filtration, and after concentrating the mother liquor, the mixture was purified by preparative plate separation by silica gel thin layer chromatography (eluent system A) to obtain 6g (80 mg) of compound.
And a second step of: 2- (3- (benzyloxy) propyl) -N 6 1-dimethyl-N 6 - (pyridin-3-ylmethyl) -1H-imidazo [4,5-c]Pyridine-4, 6-diamine (Compound 6 h)
TFA (2 mL) was added to a solution of compound 6g (80.00 mg, 151.31. Mu. Mol) in DCM (2 mL) and then allowed to react at room temperature for 4h. The solvent was directly concentrated under reduced pressure to give compound 6h (50 mg).
MS(ESI,m/z):417.2[M+H] + .
And a third step of: 3- (4-amino-1-methyl-6- (methyl (pyridin-3-ylmethyl) amino) -1H-imidazo [4,5-c ] pyridin-2-yl) propan-1-ol (Compound 6)
Boron tribromide (45.02 mg, 180.06. Mu. Mol) was added to a solution of compound 6h (50.00 mg, 120.04. Mu. Mol) in DCM (2 mL) and then left to react at room temperature for 5min. The reaction was quenched with water and ph=8 was adjusted with potassium carbonate. Extraction with EA (10 mL) followed by drying and concentration of the organic phase afforded crude product, which was then subjected to Prep-HPLC to afford compound 6 (25 mg).
Prep-HPLC conditions:
instrument model: agilent 1260; chromatographic column: waters SunFire Prep C18 OBD (19 mm. Times.150 mm. Times.5.0 μm), mobile phase: acetonitrile/0.05% NH 4 HCO 3 An aqueous solution; elution gradient: acetonitrile 10% -60% for 0-16 min; flow rate: 24ml/min; detection wavelength: 214nm, 254nm, 280nm; compound collection time: 6.0-6.5min.
MS(ESI,m/z):327.2[M+H] + .
1 H NMR(400MHz,DMSO-d 6 )δ8.48(d,J=1.7Hz,1H),8.41(dd,J=4.7,1.6Hz,1H),7.61(dt,J=7.8,1.8Hz,1H),7.29(dd,J=7.6,4.9Hz,1H),5.78(s,1H),5.76(s,2H),4.82(s,2H),4.61(s,1H),3.53(s,3H),3.49(t,J=6.1Hz,2H),2.88(s,3H),2.80–2.74(m,2H),1.89–1.81(m,2H).
Example 7:3- (4-amino-6- ((2, 4-difluorobenzyl) amino) -1-methyl-1H-imidazo [4,5-c ] pyridin-2-yl) propan-1-ol (compound 7)
The first step: 2- (3- (benzyloxy) propyl) -N 6 - (2, 4-difluorobenzyl) -1-methyl-N 4 - (2, 4-trimethylpentyl-2-yl) -1H-imidazo [4,5-c]Pyridine-4, 6-diamine (Compound 7 a)
Compound 1f (80 mg, 180.58. Mu. Mol), 2, 4-difluorobenzylamine (31.02 mg, 216.69. Mu. Mol), pd 2 (dba) 3 (16.54 mg, 18.06. Mu. Mol), davePhos (14.21 mg, 36.12. Mu. Mol), tBuOK (60.79 mg, 541.73. Mu. Mol) was added to 1, 4-dioxane (3 mL), N 2 Heating to 100 ℃ under protection to react for 16h. After the reaction was completed, it was filtered through celite, and the filtrate was dried by spin-drying to give compound 7a (70 mg) by TLC (eluent system B).
MS(ESI,m/z):550.3[M+H] + .
And a second step of: 3- (4-amino-6- ((2, 4-difluorobenzyl) amino) -1-methyl-1H-imidazo [4,5-c ] pyridin-2-yl) propan-1-ol (compound 7)
Compound 7a (70 mg, 127.34. Mu. Mol) was dissolved in methylene chloride (10 mL), and after stirring at room temperature, a mixture of boron tribromide and DCM (30% by volume, 0.2 mL) was added to the system. After the reaction was completed, the reaction was quenched by addition of anhydrous methanol (5 mL), and ph=7 was adjusted with saturated aqueous sodium bicarbonate solution. After concentration, the mixture was diluted with water and extracted three times with EA (20 mL. Times.3). The organic phases were combined, dried over anhydrous sodium sulfate and separated by Prep-HPLC to give compound 7 (6 mg).
Prep-HPLC conditions:
instrument model: agilent1260; chromatographic column: XBIdge Prep C18 OBD (19 mm. Times.150 mm. Times.5.0 μm); chromatographic column temperature: 25 ℃; flow rate: 24.0mL/min; detection wavelength: 214nm, 254nm, 280nm; elution gradient: (0 min:10% A,90% B;16.0min:90% A,10% B); mobile phase a:100% acetonitrile; mobile phase B:0.05% ammonium bicarbonate aqueous solution. Compound collection time: 6.6-7.5min.
MS(ESI,m/z):348.2[M+H] + .
1 H NMR(DMSO-d 6 ,400MHz)δ7.46(dd,J=15.6,8.7Hz,1H),7.19(td,J=10.5,2.5Hz,1H),7.01(td,J=8.5,1.9Hz,1H),6.17(s,1H),5.95(s,1H),5.71(s,1H),4.57(s,1H),4.40(d,J=6.1Hz,2H),3.47(d,J=6.8Hz,5H),2.80–2.71(m,2H),1.91–1.76(m,2H).
Example 8:3- (4-amino-1-methyl-6- ((pyrazin-2-ylmethyl) amino) -1H-imidazo [4,5-c ] pyridin-2-yl) propan-1-ol (Compound 8)
The first step: 2- (3- (benzyloxy) propyl) -1-methyl-N 6 - (pyrazin-2-ylmethyl) -N 4 - (2, 4-trimethylpentyl-2-yl) -1H-imidazo [4,5-c]Pyridine-4, 6-diamine (Compound 8 a)
Compound 1f (150 mg, 338.58. Mu. Mol), pyrazin-2-ylmethylamine (48.03 mg, 440.16. Mu. Mol), pd 2 (dba) 3 (31.00 mg, 33.86. Mu. Mol), davePhos (26.65 mg, 67.72. Mu. Mol), tBuOK (113.98 mg,1.02 mmol) was added to 1, 4-dioxane (3 mL), and the mixture was heated to 100℃for 5h. After the reaction was completed, the mixture was filtered through celite, and the filtrate was dried by spin-drying and then separated by Prep-HPLC to give compound 8a (46 mg).
Prep-HPLC conditions:
instrument model: agilent 1260; chromatographic column: XBIdge Prep C18 OBD (19 mm. Times.150 mm. Times.5.0 μm); chromatographic column temperature: 25 ℃; flow rate: 24.0mL/min; detection wavelength: 214nm, 254nm, 280nm; elution gradient: (0 min:20% A,80% B;7.6min:48.5% A,51.5% B); mobile phase a:100% acetonitrile; mobile phase B:0.05% formic acid in water. Compound collection time: 6.8-7.5min.
MS(ESI,m/z):516.3[M+H] + .
And a second step of: 3- (4-amino-1-methyl-6- ((pyrazin-2-ylmethyl) amino) -1H-imidazo [4,5-c ] pyridin-2-yl) propan-1-ol (Compound 8)
Compound 8a (46 mg, 90.39. Mu. Mol) was dissolved in TFA (20 mL) and heated to 75deg.C and stirred overnight. After the reaction was completed, the system was concentrated to dryness, and diluted with methanol (5 mL). The ph=9 was adjusted with saturated aqueous sodium bicarbonate solution and stirred at room temperature for 15min. The system was concentrated again and washed with methanol. Insoluble matter was removed by filtration, and the filtrate was concentrated and separated by Prep-HPLC to give compound 138 (4 mg).
Prep-HPLC conditions:
instrument model: agilent1260; chromatographic column: XBIdge Prep C18 OBD (19 mm. Times.150 mm. Times.5.0 μm); chromatographic column temperature: 25 ℃; flow rate: 24.0mL/min; detection wavelength: 214nm, 254nm, 280nm; elution gradient: (0 min:10% A,90% B;4.6min:18.6% A,81.4% B); mobile phase a:100% acetonitrile; mobile phase B:0.05% ammonium bicarbonate aqueous solution. Compound collection time: 3.6-4.3min.
MS(ESI,m/z):314.2[M+H] + .
1 H NMR(DMSO-d 6 ,400MHz)δ8.63(s,1H),8.57(d,J=2.3Hz,1H),8.48(d,J=2.5Hz,1H),6.24(t,J=6.3Hz,1H),5.67(d,J=5.7Hz,3H),4.63–4.44(m,3H),3.53–3.42(m,5H),2.75(t,J=7.6Hz,2H),1.88–1.77(m,2H).
Drug effect screening method and data
Experimental example 1: agonism of IL-1 beta expression in THP-1 cells after PMA induced differentiation by the compounds of the present invention
The present experiment uses a Homogeneous Time Resolved Fluorescence (HTRF) assay to test the effect of the compounds of the present invention on IL-1β levels of the NLRP3 downstream cytokines to assess the agonism of the compounds on the hNLRP3 inflammatory body or NLRP3 inflammatory body pathway at the cellular level.
Reagent: RPMI 1640 (Hyclone); heat-inactivated FBS (Gibco); PMA (Biyundian)
And (3) cells: THP-1 (Nanjing Ke Bai)
The kit comprises: IL-1. Beta. Assay kit (CISBIO)
The experimental steps are as follows:
1) THP-1 cells in logarithmic growth phase were grown at 5X 10 5 Density of individual/well inoculated in T75 flask, placed at 37℃in 5% CO 2 Culturing for 24h in an incubator of (2) to induce THP-1 suspension cells to become adherent macrophages. Complete growth medium was RPMI 1640, containing 10% heat-inactivated FBS,0.05mM β -mercaptoethanol, 1 μm PMA.
2) After 24h of induction culture, the adherent cells were digested with pancreatin. Centrifuge at 1000rpm for 5min and then remove the supernatant. Cells were resuspended to a density of 2×10 using detection medium RPMI 1640 containing 2% heat-inactivated FBS 6 And each mL. Spreading cells to 96-well plate to 50 μl/well, with cell number per well of 1×10 5 And each.
3) An appropriate amount of 10mM DMSO solution of the test compound was prepared at a 2 Xtest concentration in RPMI 1640 medium containing 2% heat-inactivated FBS. Adding 50 μl/well diluent into 96-well plate cells, mixing thoroughly, placing 96-well plate at 37deg.C, 5% CO 2 Is cultured in an incubator for 6 hours. The supernatant was collected and the level of IL-1. Beta. In the supernatant was measured according to the instructions of the IL-1. Beta. Detection kit.
4) Stimulation of IL-1 beta by test Compounds EC 50 The fit was performed by GraphPad software log (agonist) vs. response-Variable slope four-parameter method.
Agonism of the compounds of the invention on IL-1β expression in THP-1 cells after PMA induced differentiation is shown in Table 1.
Experimental example 2: THP1 cells deleted of NLRP3 after PMA induced differentiation by the compound of the present invention (THP 1- def Agonism of IL-1 beta expression in NLRP3 cells)
The test uses HTRF detection method to test the compound of the invention for THP1- def The effect of IL-1β levels in NLRP3 cells to assess the specificity of the compound for hllrp 3 inflammatory body or NLRP3 inflammatory body pathway agonism.
Reagent: RPMI 1640 (Hyclone); heat-inactivated FBS (Gibco); PMA (Biyundian)
And (3) cells: THP1- def NLRP3(InvivoGen)
The kit comprises: IL-1. Beta. Assay kit (CISBIO)
The experimental steps are as follows:
1) THP1- def NLRP3 cells at 5X 10 5 Density of individual/well inoculated in T75 flask, placed at 37℃in 5% CO 2 Culturing in an incubator for 24h, and inducing THP1- def NLRP3 suspension cells become adherent macrophages. Complete growth medium was RPMI 1640, containing 10% heat-inactivated FBS,0.05mM β -mercaptoethanol, 1 μm PMA.
2) After 24h of induction culture, the adherent cells were digested with pancreatin. Centrifuge at 1000rpm for 5min and then remove the supernatant. Cells were resuspended to a density of 2×10 using detection medium RPMI 1640 containing 2% heat-inactivated FBS 6 And each mL. Spreading cells to 96-well plate to 50 μl/well, with cell number per well of 1×10 5 And each.
3) An appropriate amount of 10mM DMSO solution of the test compound was prepared at a 2 Xtest concentration in RPMI 1640 medium containing 2% heat-inactivated FBS. Adding 50 μl/well diluent into 96-well plate cells, mixing, placing 96-well plate at 37deg.C, and 5% CO 2 Is cultured in an incubator for 6 hours. The supernatant was collected and the level of IL-1. Beta. In the supernatant was measured according to the instructions of the IL-1. Beta. Detection kit.
4) Stimulation of IL-1 beta by test Compounds EC 50 The fit was performed by GraphPad software log (agonist) vs. response-Variable slope four-parameter method.
THP1- def The agonism of IL-1β expression in NLRP3 cells is shown in Table 1.
Experimental example 3: agonism of hTLR7 by the inventive Compounds
This experiment tests the activation of TLR7 signaling pathway by compounds of the invention by detecting luciferase in HEK-hTLR7-NF- κb-luciferase reporter cells to assess the specificity of the compounds for agonism of NLRP3 inflammatory small body pathway.
Reagent: DMEM (High glucose); FBS (Gibco);
and (3) cells: HEK-hTLR7-NF-kB-Luciferase reporter cell (humanized TLR7 NF-kB-Luciferase reporter cell) (Nanjac Bai)
The kit comprises: bright-Glo TM Luciferase detection kit (Promega)
The experimental steps are as follows:
1) HEK-hTLR7-NF-kB-Luciferase reporter gene cells in logarithmic growth phase were centrifuged and resuspended to 2X 10 with medium 6 The cell suspension was added to a 96-well plate at a concentration of 50. Mu.L/well. An appropriate amount of 10mM test compound in DMSO was prepared at 2X test concentration in DMEM (High glucose) medium containing 10% FBS. 50. Mu.L/well of the diluent was added to the cells in the 96-well plate, and the 96-well plate was placed in a cell incubator to incubate for 16 hours. The medium was DMEM (High glucose), containing 10% fbs.
2) After the cell incubation has ended, 100. Mu.l Bright-Glo is added TM Luciferase detection reagent, incubated at room temperature for 5min, and the microplate reader reads the relative Luciferase activity units (Relative Luciferase Unit, RLU).
3) Stimulation of hTLR7 by test Compounds EC 50 The fit was performed by GraphPad software log (agonist) vs. response-Variable slope four-parameter method.
The agonism of hTLR7 by the compounds of the invention is shown in table 1.
Experimental example 4: agonism of hTLR8 by the inventive Compounds
This experiment tests the activation of TLR8 signaling pathway by compounds of the invention by detecting the amount of alkaline phosphatase secretion in HEK-Blue cell lines to assess the specificity of the compounds for agonism of the NLRP3 inflammatory body pathway.
Reagent: DMEM (High glucose); FBS (Gibco); QUANTI-Blue (InvivoGen);
and (3) cells: HEK-BlueTMhTLR8 cells (humanized TLR8 cells) (InvivoGen)
The experimental steps are as follows:
1) HEK-BlueTMhTLR8 cells in logarithmic growth phase were centrifuged and resuspended to 2X 10 with medium 6 The cell suspension was added to a 96-well plate at a concentration of 50. Mu.L/well. Taking a proper amount of 10mM DMSO solution of the compound to be tested, and using a solution containing 10% FBSDMEM (High glucose) medium was formulated at 2 x test concentration. 50. Mu.L/well of the diluent was added to the cells in the 96-well plate, and the 96-well plate was placed in a cell incubator to incubate for 16 hours. The medium was DMEM (High glucose), containing 10% fbs.
2) After the cell incubation, 10. Mu.L of the cell culture supernatant was transferred to a 96-well plate, 90. Mu.L/well of QUANTI-Blue detection solution was added, and incubated at 37℃for 3 hours, with an ELISA reader OD 620 And (5) reading.
3) Stimulation of hTLR8 by test Compounds EC 50 The fit was performed by GraphPad software log (agonist) vs. response-Variable slope four-parameter method.
The agonism of hTLR8 by the compounds of the invention is shown in table 1.
TABLE 1
The results show that the compounds of the invention (e.g., compound 1, compound 2, compound 3, compound 4, compound 5, compound 6, compound 7 and compound 8) have significant agonism for IL-1β expression in THP-1 cells after PMA induced differentiation, but for THP1- def IL-1β expression in NLRP3 cells had no agonism at the highest compound test concentration (27 μM); none of the compounds had significant activation of hTLR7 and hTLR 8. In conclusion, the compounds of the application (e.g., compound 1, compound 2, compound 3, compound 4, compound 5, compound 6, compound 7, and compound 8) have significant agonistic activity on hllrp 3 and its signaling pathway with specificity and selectivity.
Various modifications of the application, in addition to those described herein, will be apparent to those skilled in the art from the foregoing description. Such modifications are also intended to fall within the scope of the appended claims. Each reference cited in this disclosure (including all patents, patent applications, journal articles, books, and any other publications) is hereby incorporated by reference in its entirety.

Claims (20)

1. A compound of formula II, a stereoisomer, tautomer, or mixture thereof, a pharmaceutically acceptable salt of said compound:
wherein:
X 1 is NR (NR) 7
R 1 Selected from C 6-12 Aryl and 5-10 membered heteroaryl, said C 6-12 Aryl and 5-10 membered heteroaryl are each optionally substituted with one or more of the following substituents: halogen, C 1-4 Alkyl and C 1-4 An alkoxy group;
R 2 Selected from H and C 1-6 An alkyl group;
R 3 selected from H, OH and C 1-8 Alkyl, said C 1-8 Alkyl optionally substituted with one or more C 6-12 Aryl substitution;
R 4 and R is 5 All are H;
R 7 selected from H and C 1-6 An alkyl group;
v is selected from C 1-8 An alkylene group;
l is- (L) 1 ) n -(L 2 ) p -(L 3 ) q -, wherein L 1 、L 2 And L 3 Identical or different and are each independently selected from C 1-8 Alkylene and O;
n, p and q are each independently 0 or 1.
2. The compound of claim 1, a stereoisomer, tautomer, or mixture thereof, a pharmaceutically acceptable salt of said compound,
wherein X is 1 Selected from NH and N (CH) 3 )。
3. A compound according to claim 1 or 2, a stereoisomer, tautomer or mixture thereof, a pharmaceutically acceptable salt of said compound,
wherein V is selected from methylene.
4. The compound of claim 1, a stereoisomer, tautomer, or mixture thereof, a pharmaceutically acceptable salt of said compound, said compound having the structure of formula II-a:
wherein R is 1 、R 2 、R 3 And L is as defined in claim 1.
5. The compound of any one of claims 1-2 or 4, a stereoisomer, tautomer, or mixture thereof, a pharmaceutically acceptable salt of said compound, wherein:
R 1 Selected from phenyl, pyridinyl or pyrazinyl, each optionally substituted with one or more of the following substituents: halogen, C 1-4 Alkyl and C 1-4 An alkoxy group.
6. The compound of claim 5, a stereoisomer, tautomer, or mixture thereof, a pharmaceutically acceptable salt of said compound, wherein:
R 1 selected from the group consisting of
7. The compound of any one of claims 1-2, 4, or 6, stereoisomers, tautomers, or mixtures thereof, pharmaceutically acceptable salts of said compound, wherein:
R 2 selected from H, methyl,Ethyl and propyl.
8. The compound of claim 7, a stereoisomer, tautomer, or mixture thereof, a pharmaceutically acceptable salt of said compound, wherein:
R 2 selected from H and methyl.
9. The compound of claim 8, a stereoisomer, tautomer, or mixture thereof, a pharmaceutically acceptable salt of said compound, wherein:
R 2 selected from methyl groups.
10. The compound of any one of claims 1-2, 4, 6, or 8-9, stereoisomers, tautomers, or mixtures thereof, pharmaceutically acceptable salts of the compound, wherein:
R 3 Selected from OH and
11. the compound of any one of claims 1-2, 4, 6, or 8-9, stereoisomers, tautomers, or mixtures thereof, pharmaceutically acceptable salts of the compound, wherein:
l is selected from propylene, ethylene, -ethylene-O-and-propylene-O-.
12. The compound of claim 1, a stereoisomer, tautomer, or mixture thereof, a pharmaceutically acceptable salt of said compound selected from the group consisting of:
13. a pharmaceutical composition comprising a compound according to any one of claims 1-12, a stereoisomer, a tautomer, or a mixture of same, a pharmaceutically acceptable salt of said compound, and one or more pharmaceutically acceptable carriers.
14. The pharmaceutical composition of claim 13 for use in the prevention and/or treatment of diseases associated with NLRP3 inflammatory body activity.
15. The pharmaceutical composition of any one of claims 13-14, further comprising one or more second therapeutic agents.
16. The pharmaceutical composition of claim 15, wherein the one or more second therapeutic agents are one or more additional agents for treating neoplastic disease.
17. A pharmaceutical formulation comprising a compound according to any one of claims 1 to 12, a stereoisomer, a tautomer, or a mixture thereof, a pharmaceutically acceptable salt of said compound, or a pharmaceutical composition according to any one of claims 13 to 16.
18. Use of a compound according to any one of claims 1 to 12, a stereoisomer, a tautomer or a mixture thereof, a pharmaceutically acceptable salt of said compound, or a pharmaceutical composition according to any one of claims 13 to 16, or a pharmaceutical formulation according to claim 17, for the preparation of a medicament for the prevention and/or treatment of a disease associated with NLRP3 inflammatory body activity.
19. The use of claim 18, wherein the medicament is for modulating the activity of NLRP3 inflammatory bodies.
20. The use of claim 19, wherein the medicament is for increasing the activity of NLRP3 inflammatory bodies.
CN201980058276.6A 2018-11-07 2019-10-29 Ring-fused compound, preparation method and application thereof Active CN112654622B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202311141511.0A CN117186092A (en) 2018-11-07 2019-10-29 Ring-fused compound, preparation method and application thereof

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
CN2018113159531 2018-11-07
CN201811315953 2018-11-07
PCT/CN2019/113877 WO2020093905A1 (en) 2018-11-07 2019-10-29 Ring-fused compound, preparation method therefor and application thereof

Related Child Applications (1)

Application Number Title Priority Date Filing Date
CN202311141511.0A Division CN117186092A (en) 2018-11-07 2019-10-29 Ring-fused compound, preparation method and application thereof

Publications (2)

Publication Number Publication Date
CN112654622A CN112654622A (en) 2021-04-13
CN112654622B true CN112654622B (en) 2023-10-03

Family

ID=70610875

Family Applications (2)

Application Number Title Priority Date Filing Date
CN201980058276.6A Active CN112654622B (en) 2018-11-07 2019-10-29 Ring-fused compound, preparation method and application thereof
CN202311141511.0A Pending CN117186092A (en) 2018-11-07 2019-10-29 Ring-fused compound, preparation method and application thereof

Family Applications After (1)

Application Number Title Priority Date Filing Date
CN202311141511.0A Pending CN117186092A (en) 2018-11-07 2019-10-29 Ring-fused compound, preparation method and application thereof

Country Status (2)

Country Link
CN (2) CN112654622B (en)
WO (1) WO2020093905A1 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP4146639A1 (en) 2020-05-06 2023-03-15 Ajax Therapeutics, Inc. 6-heteroaryloxy benzimidazoles and azabenzimidazoles as jak2 inhibitors
MX2023003995A (en) 2020-10-05 2023-06-12 Enliven Inc 5- and 6-azaindole compounds for inhibition of bcr-abl tyrosine kinases.

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007109812A2 (en) * 2006-03-23 2007-09-27 Novartis Ag Immunopotentiating compounds
CN101384593A (en) * 2006-02-17 2009-03-11 辉瑞有限公司 3-deazapurine derivatives as tlr7 modulators
WO2017184735A1 (en) * 2016-04-19 2017-10-26 Ifm Therapeutics, Inc Nlrp3 modulators
WO2017184746A1 (en) * 2016-04-19 2017-10-26 Ifm Therapeutics, Inc Nlrp3 modulators

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101384593A (en) * 2006-02-17 2009-03-11 辉瑞有限公司 3-deazapurine derivatives as tlr7 modulators
WO2007109812A2 (en) * 2006-03-23 2007-09-27 Novartis Ag Immunopotentiating compounds
WO2017184735A1 (en) * 2016-04-19 2017-10-26 Ifm Therapeutics, Inc Nlrp3 modulators
WO2017184746A1 (en) * 2016-04-19 2017-10-26 Ifm Therapeutics, Inc Nlrp3 modulators

Also Published As

Publication number Publication date
CN112654622A (en) 2021-04-13
WO2020093905A1 (en) 2020-05-14
CN117186092A (en) 2023-12-08

Similar Documents

Publication Publication Date Title
CN114127053B (en) Substituted pyrazine compound, preparation method and application thereof
CN111836814B (en) Condensed-cyclic compound, preparation method and application thereof
CN113637005A (en) KRAS inhibitors for cancer treatment
CN111683662B (en) Substituted pyrimidine compounds, pharmaceutical compositions and methods of treatment
AU2009345802B2 (en) Phenoxymethyl heterocyclic compounds
JP7033588B2 (en) Phthalazine derivatives as inhibitors of PARP1, PARP2, and / or tubulin useful in the treatment of cancer
CN113330009B (en) Azacyclic compounds, preparation method and application thereof
US20220056043A1 (en) Nitrogen-containing fused cyclic compound, preparation method therefor and use thereof
CN112654622B (en) Ring-fused compound, preparation method and application thereof
CN116568681A (en) SOS1 inhibitor, pharmaceutical composition containing same and application thereof
CN115073450A (en) KRAS G12C Preparation and application of mutant protein inhibitor
US10807983B2 (en) Imidazo-fused heterocycles and uses thereof
CN114555565B (en) Nitrogen-containing heterocyclic compound, preparation method and application thereof
CN111253390B (en) Ring-fused compound, preparation method and application thereof
JP2024516194A (en) Compounds as PD1/PD-L1 inhibitors and methods thereof
CN115181106A (en) Quinazoline KRAS G12D Preparation and application of mutant protein inhibitor
CN115073451A (en) KRAS G12D Preparation and application of mutant protein inhibitor
CN111825694B (en) Condensed-cyclic compound, preparation method and application thereof
CN117185987A (en) Amidine and guanidine derivatives, preparation method and medical application thereof
CN111808080B (en) Substituted pyridine or pyrimidine compound, preparation method and medical application thereof
CN114269753B (en) Nitrogen-containing bicyclic compound, pharmaceutical composition containing same, preparation method and application thereof
CN117164583A (en) Compound for targeting IRAK4 protein degradation and application thereof
CN117062809A (en) Bicyclic heterocyclic FGFR4 inhibitors, pharmaceutical compositions and formulations comprising the same and uses thereof
CN117355533A (en) Novel compounds as protein kinase inhibitors
TW202334097A (en) Benzo[h]quinazoline-4-amine derivatives for the treatment of cancer

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
GR01 Patent grant
GR01 Patent grant