CN111072667A - Five-membered or six-membered heterocyclic pyrimidine compound and application thereof - Google Patents

Five-membered or six-membered heterocyclic pyrimidine compound and application thereof Download PDF

Info

Publication number
CN111072667A
CN111072667A CN201811228693.4A CN201811228693A CN111072667A CN 111072667 A CN111072667 A CN 111072667A CN 201811228693 A CN201811228693 A CN 201811228693A CN 111072667 A CN111072667 A CN 111072667A
Authority
CN
China
Prior art keywords
alkyl
optionally substituted
substitution
compound
alkenyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN201811228693.4A
Other languages
Chinese (zh)
Inventor
胡允金
刘乐
魏国平
游志先
李小龙
吴国胜
冯加权
董加强
王铁林
阳华
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.)
Luoxin Biotechnology Shanghai Co ltd
Shandong Luoxin Pharmaceutical Group Co Ltd
Original Assignee
Luoxin Biotechnology Shanghai Co ltd
Shandong Luoxin Pharmaceutical Group 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 Luoxin Biotechnology Shanghai Co ltd, Shandong Luoxin Pharmaceutical Group Co Ltd filed Critical Luoxin Biotechnology Shanghai Co ltd
Priority to CN201811228693.4A priority Critical patent/CN111072667A/en
Priority to PCT/CN2019/111625 priority patent/WO2020083089A1/en
Publication of CN111072667A publication Critical patent/CN111072667A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • A61K31/52Purines, e.g. adenine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • A61K31/52Purines, e.g. adenine
    • A61K31/522Purines, e.g. adenine having oxo groups directly attached to the heterocyclic ring, e.g. hypoxanthine, guanine, acyclovir
    • 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
    • A61K31/53Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with three nitrogens as the only ring hetero atoms, e.g. chlorazanil, melamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D473/00Heterocyclic compounds containing purine ring systems
    • C07D473/02Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6
    • C07D473/18Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6 one oxygen and one nitrogen atom, e.g. guanine
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D475/00Heterocyclic compounds containing pteridine ring systems
    • C07D475/06Heterocyclic compounds containing pteridine ring systems with a nitrogen atom directly attached in position 4
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Abstract

The invention relates to a method forTreatment of TLRs7Five-or six-membered heterocyclopyrimidines and pharmaceutical compositions for diseases responsive to receptor activation.

Description

Five-membered or six-membered heterocyclic pyrimidine compound and application thereof
Technical Field
The application relates to as TLRs7Five-or six-membered heterocyclopyrimidines of agonists, pharmaceutical compositions containing such compounds, and uses of such compounds and pharmaceutical compositions.
Background
Toll-like receptors (TLRs) are an important class of protein molecules involved in non-specific immunity (innate immunity) and play a key role in coordinating non-specific immunity and specific immunity against pathogens. TLRs are single transmembrane non-catalytic proteins expressed on a variety of immune cells. TLRs can recognize microorganisms and activate the body to produce an immune cell response when they break through the body's physical barriers, such as skin, mucous membranes, etc. Toll-like receptors recognize highly conserved structural motifs: pathogen-associated microbial patterns (PAMPs) expressed by microbial pathogens or damage-associated molecular patterns (DAMPs) released by necrotic cells. By corresponding pathogen associationThe microbial patterns (PAMP) or damage-associated molecular patterns (DAMP) of (A) stimulate Toll-like receptors to initiate signaling cascades leading to transcription factors such as AP-1, NF-kB and activation of interferon regulatory factors. This results in a variety of cellular responses, including the production of interferons, proinflammatory cytokines, and effector cytokines, to generate an immune response. To date, 13 Toll-like receptors have been discovered in mammals. Toll-like receptors 1,2,4, 5 and 6 are expressed primarily on the cell surface, and Toll-like receptors 3, 7, 8 and 9 are expressed in endosomes. Different Toll-like receptors recognize different pathogen-derived ligands. Toll-like receptor 7 (TLR)7) Induction of secretion of interferon α (IFN- α) primarily by plasmacytoid dendritic cell (pDC) expression and ligand recognition Toll-like receptor 7 (TLR)7) And Toll-like receptor 8 (TLR)8) Highly homologous, therefore TLRs7The ligand is also a TLR in most cases8A ligand. TLRs8The stimulation mainly induces the production of cytokines such as tumor necrosis factor α (TNF- α) and chemokines IFN- α is one of the main drugs for treating chronic hepatitis B or hepatitis C, and TNF- α is a proinflammatory cytokine, and excessive secretion may cause serious side effects7Agonists are clinically important safety implications for the treatment of diseases, including viral infectious diseases. TLRs7The agonists have been reported in clinical development, such as imiquimod, resiquimod, GS-9620. For example, Gilead has converted TLRs7The agonist GS-9620 is advanced to phase II of the clinic for the treatment of patients with chronic hepatitis B (D.Allen, et al, Allen, D.et al, WO 2016/044183). There remains a need in the art to develop new more selective, active and safe TLRs7An agonist.
Disclosure of Invention
In one aspect, the present invention provides five-or six-membered heterocyclopyrimidines that can be used as TLRs7An agonist. In particular, the invention relates to compounds of formula (I):
Figure BDA0001836596920000021
or a pharmaceutically acceptable salt, stereoisomer, solvate, polymorph, tautomer, isotopic compound, metabolite, or prodrug thereof;
wherein:
x is selected from O, S and NRx;RxSelected from H, C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with one or more RASubstitution;
y is selected from O, S, N, or Y is absent;
z is selected from N, O, S, C and CH;
q is selected from O, S and NH;
when Y is N, R1Selected from H, hydroxyl, sulfydryl, amino, halogen, nitro, cyano, C1-C8Alkyl radical, C2-C8Alkenyl radical, C2-C8Alkynyl, C6-C10Aryl and five to ten membered heteroaryl; wherein said alkyl, alkenyl, alkynyl, aryl or heteroaryl is optionally substituted with one or more RBSubstitution;
R2selected from H, hydroxyl, sulfydryl, amino, halogen, nitro, cyano, C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with one or more RCSubstitution;
L1is C1-C8A hydrocarbon chain;
ring a is absent, or ring a is selected from: c3-C10Cycloalkyl radical, C3-C10Heterocyclic group, C6-C10Aryl and five to ten membered heteroaryl; wherein said cycloalkyl, heterocyclyl, aryl, heteroaryl is optionally substituted with one or more RDSubstitution;
L2is absent, or L2Is C1-C8A hydrocarbon chain;
R3is H, C3-C10Cycloalkyl radical, C6-C10Aryl, five-to ten-membered heteroarylRadical or-WR6R7Wherein W is N or CH, wherein
R6And R7Each independently selected from H, C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with one or more RESubstitution; or
R6And R7Together with the N or C atom to which they are attached form a three-to ten-membered heterocyclic ring; said heterocycle being optionally substituted by one or more RESubstitution; and optionally, R6And R7The three-to ten-membered heterocyclic ring formed together with the N or C atom to which it is attached may optionally be fused to the a ring;
R4is selected from C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group; wherein said alkyl, alkenyl or and alkynyl are each optionally substituted with one or more RFSubstitution;
ring B is absent, or ring B is selected from C3-C10Cycloalkyl radical, C3-C10Heterocyclic group, C6-C10Aryl and five to ten membered heteroaryl; wherein said cycloalkyl, heterocyclyl, aryl and heteroaryl are each optionally substituted with one or more RGSubstitution;
R5selected from H, C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with one OR more groups selected from-OR8、-SR8and-NR8R8' is substituted with a substituent;
R8and R8' each is independently selected from H, C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with one or more RHSubstitution; or R8And R8' together with the N atom to which it is attached, form a three-to ten-membered heterocyclic ring, preferably a four-to six-membered heterocyclic ring;
RA、RB、RC、RD、RE、RF、RG、RHeach occurrence is independently selected from halogen, cyano, nitro, -Ra、-ORa、=O、-SRa、-NRaRb、=NRa-C (halogen)3-CR (halogen)2、-CR2(halogen), -OCN, -SCN, -N ═ C ═ O, -NCS, -NO2、-N(Ra)C(=O)Rb、-N(Ra)C(=O)ORb、-N(Ra)C(=O)NRbRc、-C(=O)NRaRb、-C(=O)ORa、-OC(=O)NRaRb、-OC(=O)Ra、-OC(=O)ORa、-C(=O)Ra、-S(=O)2ORa、-S(=O)2Ra、-OS(=O)2Ra、-OS(=O)2ORa、-S(=O)2NRaRb、-S(=O)Ra、-N(Ra)S(=O)2Rb、-N(Ra)S(=O)2NRbRc、-N(Ra)S(=O)2ORb、-OP(=O)(ORa)ORb、-P(=O)(ORa)ORb、-C(=O)Ra、-C(=S)Ra、-C(=O)ORa、-C(=S)ORa、-C(=O)SRa、-C(=S)SRa、-C(=O)NRaRb、-C(=S)NRaRb、-C(=NRa)NRbRc、-NRaC(=NRb)NRcRd;Ra、Rb、RcAnd RdEach occurrence is independently selected from H, C1-C8Alkyl radical, C3-C8Cycloalkyl, a three-to eight-membered heterocyclic group, C6-C10Aryl, five-to ten-membered heteroaryl, C6-C10aryl-C1-C4Alkyl and five-to ten-membered heteroaryl-C1-C4An alkyl group; wherein the alkyl, cycloalkyl, heterocyclyl, aryl, heteroarylRadical, aryl radical-C1-C4Alkyl and heteroaryl-C1-C4Each alkyl group is optionally substituted by one or more halogen, ═ O, -OH, -NH2-SH substitution;
wherein when R isA、RB、RC、RD、RE、RF、RGOr RHEach independently is-NRaRbWhen R isa、RbOptionally together with the N atom to which they are attached, form a three-to ten-membered heterocyclic ring, preferably a four-to six-membered heterocyclic ring.
The present invention also provides compounds of the following formulae (I-1), (I-2), (I-3), (I-4), (I-5), (I-6) and (I-7),
Figure BDA0001836596920000031
Figure BDA0001836596920000041
or a pharmaceutically acceptable salt, stereoisomer, solvate, polymorph, tautomer, isotopic compound, metabolite or prodrug thereof,
wherein R is1、R2、R3、R4、RF、R5、L1、L2X, Y, Z, Q, A and B are as defined for formula (I).
In another aspect, the present invention also provides a pharmaceutical composition comprising a compound of the present invention, or a pharmaceutically acceptable salt, stereoisomer, solvate, polymorph, tautomer, isotopic compound, metabolite, or prodrug thereof, and at least one pharmaceutically acceptable carrier.
In a further aspect, the present invention also provides the use of a compound of the present invention, or a pharmaceutically acceptable salt, stereoisomer, solvate, polymorph, tautomer, isotopic compound, metabolite or prodrug thereof, or a pharmaceutical composition of the present invention, for the manufacture of a medicament for the treatment of a disease, which is a disorder of the present inventionTLR7Diseases responsive to the activation of the receptor.
In another aspect, the invention provides a compound of the invention, or a pharmaceutically acceptable salt, stereoisomer, solvate, polymorph, tautomer, isotopic compound, metabolite or prodrug thereof, or a pharmaceutical composition of the invention, for use in the treatment of a TLR7Diseases responsive to the activation of the receptor.
In a further aspect, the invention provides a method of treating a TLR7A method of a disease responsive to activation of a receptor, the method comprising administering to a subject in need thereof a compound of the invention, or a pharmaceutically acceptable salt, stereoisomer, solvate, polymorph, tautomer, isotopic compound, metabolite or prodrug thereof, or a pharmaceutical composition of the invention.
In one embodiment, the pair of TLRs7The diseases responsive to the activation of the receptor are selected from liver-related diseases, tumors and HIV infection. In a specific embodiment, the tumor type is selected from the group consisting of leukemia, lymphoma, melanoma, or non-small cell lung cancer. In a specific embodiment, the HIV infection is aids.
The present invention also provides a pharmaceutical combination comprising (1) a compound of the invention, or a pharmaceutically acceptable salt, stereoisomer, solvate, polymorph, tautomer, isotopic compound, metabolite or prodrug thereof, or a pharmaceutical composition of the invention, and (2) a PD-1 antibody, a PD-L1 antibody, or a PD-1 inhibitor, a PD-L1 inhibitor, or a PD-1/PD-L1 inhibitor. In one embodiment, the pharmaceutical combination is in the form of a pharmaceutical composition or kit.
Drawings
FIG. 1 shows the mouse plasma and liver IFN- α and TNF- α expression levels induced by Compound 3 and GS-9620, wherein the exposure of Compound 3 in both the mouse plasma and liver is below the lower limit of the LC/MS/MS assay, the position of which is shown by the number 0 only.
Detailed description of the preferred embodiments
General terms and definitions
Unless otherwise defined, terms used herein should be construed to have the same meaning as commonly understood in the art by one of ordinary skill in the art. It will be further understood that terms used herein should be interpreted as having a meaning that is consistent with their meaning in the context of this specification and the relevant art, unless explicitly defined as such herein.
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. It will be understood by those skilled in the art that terms such as "including" and "comprising" encompass the meaning of "consisting of ….
The term "one or more" or similar expressions "at least one" may mean, for example, 1,2, 3,4, 5,6, 7, 8, 9, 10 or more(s).
When the lower and upper limits of a range of values are disclosed, any value falling within the range and any included range is specifically disclosed. In particular, each range of values disclosed herein is to be understood as meaning each and every value and range encompassed within the broader range.
The expression m-n as used herein refers to the range of m to n as well as to the subranges comprised of individual point values therein as well as to individual point values. For example, the expression "C1-C8"covers the range of 1 to 8 carbon atoms and is to be understood to also cover any subrange therein as well as each point value, e.g. C2-C5、C3-C4、C1-C2、C1-C3、C1-C4、C1-C5、C1-C6、C1-C7Etc. and C1、C2、C3、C4、C5、C6、C7、C8And the like. For example, the expression "C3-C10"should also be understood in a similar manner, e.g. to cover any sub-ranges and point values comprised therein, e.g. C3-C9、C6-C9、C6-C8、C6-C7、C7-C10、C7-C9、C7-C8、C8-C9Etc. and C3、C4、C5、C6、C7、C8、C9、C10And the like. Also for example, the expression "three to ten members" should be understood to encompass any subrange therein and each point value, such as 3-5 members, 3-6 members, 3-7 members, 3-8 members, 4-5 members, 4-6 members, 4-7 members, 4-8 members, 5-7 members, 5-8 members, 6-7 members, 7-8 members, 9-10 members, etc., as well as 3,4, 5,6, 7, 8, 9, 10 members, etc. Other similar expressions in this document should be understood in a similar manner.
The terms "optionally" or "optionally" mean that the subsequently described event or circumstance may or may not occur, and that the description includes instances where said event or circumstance occurs and instances where it does not.
The terms "substituted" and "substituted" mean that one or more (e.g., one, two, three, or four) hydrogens on the designated atom is replaced with a selection from the indicated group, provided that the designated atom's normal valency in the current situation is not exceeded and that the substitution results in a stable compound. Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds. When it is stated that a substituent is absent, it is understood that the substituent may be one or more hydrogen atoms, provided that the structure is such that the compound attains a stable state.
If a substituent is described as "optionally … substituted," the substituent may be unsubstituted or may be substituted. If an atom or group is described as optionally substituted with one or more of a list of substituents, one or more hydrogens on the atom or group may be replaced with an independently selected, optional substituent. When the substituent is oxo (i.e., ═ O), it means that two hydrogen atoms are substituted.
Unless indicated, as used herein, the point of attachment of a substituent may be from any suitable position of the substituent. When a bond of a substituent is shown through a bond connecting two atoms in a ring, then such substituent may be bonded to any ring atom in the substitutable ring.
When any variable (e.g., R), and the variable bearing the label (e.g., R)1、R2、R3、R4、R5、R6、R7、R8、R8’、RA、RB、RC、RD、RE、RF、RGEtc.) in the composition or structure of a compound, which at each occurrence is defined independently in each instance. For example, if a group is substituted with 0,1, 2,3, or 4R substituents, the group can optionally be substituted with up to four R substituents, and the options for each R substituent in each case are independent of each other.
The term "halogen" or "halo" is understood to mean a fluorine (F), chlorine (Cl), bromine (Br) or iodine (I) atom, preferably a fluorine, chlorine, bromine or iodine atom.
The term "hydroxy" refers to-OH.
The term "cyano" refers to — CN.
The term "mercapto" refers to-SH.
The term "amino" refers to the group-NH2
The term "alkyl" refers to a straight or branched chain saturated aliphatic hydrocarbon group consisting of carbon and hydrogen atoms, which is attached to the rest of the molecule by a single bond. The "alkyl" group may have 1 to 8 carbon atoms, i.e. "C1-C8Alkyl radicals ", e.g. C1-C4Alkyl radical, C1-C3Alkyl radical, C1-C2Alkyl radical, C3Alkyl radical, C4Alkyl radical, C3-C6An alkyl group. Non-limiting examples of alkyl groups include, but are not limited to, methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl, isobutyl, sec-butyl, tert-butyl, isopentyl, 2-methylbutyl, 1-ethylpropyl, 1, 2-dimethylpropyl, neopentyl, 1-dimethylpropyl, 4-methylpentyl, 3-methylpentyl, 2-methylpentyl, 1-methylpentyl, 2-ethylpentyl, and the likeButyl, 1-ethylbutyl, 3-dimethylbutyl, 2-dimethylbutyl, 1-dimethylbutyl, 2, 3-dimethylbutyl, 1, 3-dimethylbutyl or 1, 2-dimethylbutyl, or isomers thereof.
The term "alkenyl" refers to a straight or branched chain unsaturated aliphatic hydrocarbon group having at least one double bond, consisting of carbon atoms and hydrogen atoms. The alkenyl group may have 2 to 8 carbon atoms, i.e. "C2-C8Alkenyl radicals, e.g. C2-C4Alkenyl radical, C3-C4An alkenyl group. Non-limiting examples of alkenyl groups include, but are not limited to, vinyl, allyl, (E) -2-methylvinyl, (Z) -2-methylvinyl, homoallyl, (E) -but-2-enyl, (Z) -but-2-enyl, (E) -but-1-enyl, (Z) -but-1-enyl, and the like.
The term "alkynyl" refers to a straight or branched chain unsaturated aliphatic hydrocarbon group having at least one triple bond composed of carbon atoms and hydrogen atoms. The alkynyl group may have 2 to 8 carbon atoms, i.e. "C2-C8Alkynyl radicals, e.g. C2-C4Alkynyl, C3-C4Alkynyl. Non-limiting examples of alkynyl groups include, but are not limited to, ethynyl, prop-1-ynyl, prop-2-ynyl, but-1-ynyl, but-2-ynyl, but-3-ynyl, and the like.
The term "cycloalkyl" refers to a saturated or unsaturated, non-aromatic cyclic hydrocarbon group consisting of carbon and hydrogen atoms, preferably containing 1 or 2 rings. The cycloalkyl group may be a monocyclic, fused polycyclic, bridged or spiro ring structure. The cycloalkyl group may have 3 to 10 carbon atoms, i.e. "C3-C10Cycloalkyl radicals ", e.g. C3-C8Cycloalkyl radical, C5Cycloalkyl radical, C6Cycloalkyl radical, C7A cyclic hydrocarbon group. Non-limiting examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, bicyclo [2.2.1 ] n]Heptyl and spiro [3.3 ]]Heptyl, and the like.
The term "heterocyclyl" or "heterocyclic hydrocarbon radical" refers to a monocyclic or bicyclic non-aromatic ring system (three to ten, three to eight, three to six) having, for example, 3 to 10 (suitably 3 to 8, more suitably 3 to 6, especially 4 to 6) ring atoms, wherein at least one ring atom (e.g. 1,2 or 3) is a heteroatom selected from N, O and S, and the remaining ring atoms are C. The ring system may be saturated (also understood as corresponding "heterocycloalkyl") or unsaturated (i.e., having one or more double and/or triple bonds within the ring). The term also covers the case where the C atom may be substituted by oxo (═ O) and/or the S atom on the ring may be substituted by 1 or 2 oxo (═ O).
Heterocyclyl groups may be, for example, 4-membered rings, such as azetidinyl, oxetanyl; or a 5-membered ring such as tetrahydrofuranyl, dioxanyl (dioxalinyl), pyrrolidinyl, imidazolidinyl, pyrazolidinyl, pyrrolinyl, oxopyrrolidinyl, 2-oxoimidazolidin-1-yl, thiazolyl, thiadiazolyl; or a 6-membered ring, such as tetrahydropyranyl, piperidinyl, morpholinyl, dithianyl, thiomorpholinyl, piperazinyl, 1-dioxo-1, 2-thiazinan-2-yl, or trithianyl; or 7-membered rings, e.g. diazepine
Figure BDA0001836596920000071
A base ring. Optionally, the heterocyclic group may be benzo-fused.
Heterocyclyl groups may be bicyclic, without limitation, such as a5, 5-membered ring, for example a hexahydrocyclopenta [ c ] pyrrol-2 (1H) -yl) ring; or a5, 6-membered bicyclic ring, such as a hexahydropyrrolo [1,2-a ] pyrazin-2 (1H) -yl ring.
As mentioned above, the ring containing the nitrogen atom may be partially unsaturated, i.e. it may contain one or more double bonds, without limitation, such as a 2, 5-dihydro-1H-pyrrolyl, 4H- [1,3,4] thiadiazinyl, 4, 5-dihydrooxazolyl or 4H- [1,4] thiazinyl ring, or it may be benzo-fused, without limitation, such as a dihydroisoquinolinyl ring.
The term "aryl" refers to an aromatic cyclic group that is an all-carbon monocyclic or fused polycyclic (e.g., bicyclic) ring having a conjugated pi-electron system. For example, the aryl group may have 6 to 20 carbon atoms, 6 to 14 carbon atoms, suitably 6 to 10, more suitably 6 or 10. Examples of aryl groups include, but are not limited to, phenyl, naphthyl, and anthracenyl and the like.
The term "heteroaryl" is understood as preferably meaning a monovalent monocyclic, bicyclic or tricyclic aromatic ring system having 5,6, 7, 8, 9 or 10 ring atoms ("five-to ten-membered heteroaryl"), in particular 5 or 6 or 9 or 10 ring atoms, and comprising at least one, suitably 1 to 4, more suitably 1 to 3, heteroatom(s), which may be identical or different, of the ring atoms, such as oxygen, nitrogen or sulfur, and furthermore, in each case the heteroaryl group may be benzo-fused. In particular, heteroaryl is selected from thienyl, furyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, thia-4H-pyrazolyl and the like, and their benzo derivatives such as benzofuryl, benzothienyl, benzoxazolyl, benzisoxazolyl, benzimidazolyl, benzotriazolyl, indazolyl, indolyl, isoindolyl and the like; or pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, and the like, and benzo derivatives thereof, such as quinolyl, quinazolinyl, isoquinolyl, and the like; or azocinyl, indolizinyl, purinyl and the like, and benzo derivatives thereof; or cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, naphthyridinyl, carbazolyl, acridinyl and the like.
The term "C" as used herein1-C8The hydrocarbon chain "means a chain-like group composed of carbon atoms and hydrogen atoms, which may be straight-chain or branched, and contains 1 to 8 (particularly 1 to 5, for example 1,2, 3,4 or 5) carbon atoms. The hydrocarbon chain may be saturated (i.e., C)1-C8Alkylene) or may be unsaturated, i.e. may contain one or more (preferably 1) carbon-carbon double or triple bonds. The alkylene group may have 1 to 8 carbon atoms, i.e. "C1-C8Alkylene radicals, e.g. C1-C5Alkylene radical, C1-C4Alkylene radical, C1-C3Alkylene radical, C1-C2Alkylene radical, C3Alkylene, and C1Alkylene, i.e. methylene. Non-limiting examples of alkylene groups include, but are not limited to, methylene (-CH)2-), 1-ethylidene (-CH (CH)3) -), 1, 2-ethylene (-CH)2CH2-), 1-propylene (-CH (CH)2CH3) -), 1, 2-propylene (-CH)2CH(CH3) -), 1, 3-propylene (-CH)2CH2CH2-) 1, 4-butylene (-CH2CH2CH2CH2-) and the like.
By "pharmaceutically acceptable" is meant compatible with the other ingredients of the formulation and not unacceptably toxic to the subject to which it is administered.
In particular, the compounds of the present invention may be prepared by a variety of synthetic methods well known to those skilled in the art. This includes the specific embodiments listed below, embodiments thereof in combination with other chemical synthetic methods and equivalents thereof recognized in the art. Preferred embodiments include, but are not limited to, examples of the present invention.
The chemical reactions of the specific embodiments of the present invention are carried out in a suitable solvent, which is described as being suitable for the chemical changes of the present invention and the reagents and materials required therefor. In order to obtain the compounds of the present invention, the synthesis steps or reaction schemes need to be modified or selected by those skilled in the art as necessary based on the existing practice.
Pharmaceutically acceptable salts of the compounds of the present invention include acid addition salts and base addition salts thereof. Suitable acid addition salts are formed from acids which form pharmaceutically acceptable salts. Examples include hydrochloride, acetate, aspartate, benzoate, bicarbonate/carbonate, glucoheptonate, gluconate, nitrate, orotate, palmitate and other similar salts. Suitable base addition salts are formed from bases which form pharmaceutically acceptable salts. Examples include aluminum salts, arginine salts, choline salts, magnesium salts, and other similar salts. For reviews of suitable salts see, for example, "Remington's Pharmaceutical Sciences", mack publishing Company, Easton, Pa., (2005); and "handbook of pharmaceutically acceptable salts: properties, Selection and application "(handbook of Pharmaceutical Salts: Properties, Selection, and Use), Stahl and Wermuth (Wiley-VCH, Weinheim, Germany, 2002). Methods for preparing pharmaceutically acceptable salts of the compounds of the present invention are known to those skilled in the art.
The compounds of the present invention may exist in specific geometric or stereoisomeric forms. The present invention contemplates all such compounds, including cis and trans isomers, (-) -and (+) -enantiomers, (R) -and (S) -enantiomers, diastereomers, (D) -isomers, (L) -isomers, as well as racemic and other mixtures thereof, such as enantiomerically or diastereomerically enriched mixtures, all of which are within the scope of the present invention. Additional asymmetric carbon atoms may be present in substituents such as alkyl groups. All such isomers, as well as mixtures thereof, are included within the scope of the present invention. In certain embodiments, preferred compounds are those that exhibit more advantageous biological activity as isomers. Purified or partially purified isomers and stereoisomers, or racemic or diastereomeric mixtures of the compounds of the invention are also included within the scope of the invention. Purification and isolation of such materials can be accomplished by standard techniques known in the art.
Optically pure enantiomers can be obtained by resolution of the racemic mixture according to conventional methods, for example by formation of diastereomeric salts using optically active acids or bases, or by formation of covalent diastereomers. Mixtures of diastereomers may be separated into the individual diastereomers by methods known in the art (e.g., by chromatography or fractional crystallization) based on their physical and/or chemical differences. The optically active enantiomeric base or acid is then released from the separated diastereomeric salt. Another method of separating racemic enantiomers may use chiral chromatography (e.g., a chiral HPLC column), and the separated chiral isomers may be subjected to conventional derivatization prior to separation or may not be derivatized, depending on which method may achieve more efficient separation of chiral isomers. Enzymatic methods can also be used to separate derivatized or underivatized chiral isomers. Likewise, the optically pure compound of the present invention can be obtained by chiral synthesis using an optically active raw material.
In addition, the compounds of the present invention may exist in tautomeric forms. The invention includes all possible tautomers of the compounds of the invention, also including the forms of a single tautomer or any mixture of said tautomers in any ratio.
The compounds of the invention may be present in the form of solvates, preferably hydrates, wherein the compounds of the invention comprise as structural element of the crystal lattice of the compound a polar solvent, such as in particular water, methanol or ethanol. The amount of polar solvent, particularly water, may be present in stoichiometric or non-stoichiometric proportions.
The invention also encompasses all possible crystalline forms or polymorphs of the compounds of the invention, which may be single polymorphs or mixtures of more than one polymorph in any ratio.
The invention also includes all pharmaceutically acceptable isotopically-labeled compounds, 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.
Also included within the scope of the present invention are metabolites of the compounds of the present invention, i.e., substances formed in vivo upon administration of the compounds of the present invention. Metabolites of a compound can be identified by techniques well known in the art, and their activity can be characterized by assay methods. Such products may result, for example, from oxidation, reduction, hydrolysis, amidation, deamidation, esterification, enzymatic hydrolysis, etc. of the administered compound. Accordingly, the present invention includes metabolites of the compounds of the present invention, including compounds made by the process of contacting the compounds of the present invention with a mammal for a time sufficient to produce a metabolite thereof.
The present invention further includes within its scope prodrugs of the compounds of the present invention which are certain derivatives of the compounds of the present invention which may themselves have little or no pharmacological activity which, when administered into or onto the body, may be converted to the compounds of the present 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 on the use of prodrugs can be found in "Pro-drugs as Novel Delivery Systems", volume 14, ACS Symposium Series (t.higuchi and v.stella). Prodrugs of the invention may be prepared, for example, by substituting certain moieties known to those skilled in the art as "pro-moieties" (e.g., "Design of Prodrugs", described in h. bundgaard (Elsevier, 1985)) for appropriate functional groups present in compounds of the invention.
The invention also encompasses compounds of the invention containing a protecting group. In 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 concerned, thereby forming a chemically protected form of the compounds of the present invention. This may be achieved by conventional protecting Groups, such as those described in t.w.greene & p.g.m.wuts, Protective Groups in Organic Synthesis, John Wiley & Sons,2006, which references are incorporated herein by reference. The protecting group may be removed at a suitable subsequent stage using methods known in the art.
The terms "administration" or "administering" and the like refer to a method that can enable a compound or composition to be delivered to a desired site of biological action. These methods include, but are not limited to, parenteral (including intravenous, subcutaneous, intraperitoneal, intramuscular, intravascular injection or infusion), topical, rectal administration, and the like.
As used herein, the term "treating" includes alleviating, or ameliorating a disease or condition, preventing other conditions, ameliorating or preventing underlying metabolic factors of a condition, inhibiting a disease or condition, e.g., arresting the development of a disease or condition, alleviating a disease or condition, promoting remission of a disease or condition, or arresting signs of a disease or condition, and extends to include prevention. "treating" also includes achieving a therapeutic benefit and/or a prophylactic benefit. Therapeutic benefit refers to eradication or amelioration of the condition being treated. In addition, therapeutic benefit is achieved by eradicating or ameliorating one or more physiological signs associated with the underlying disease, and amelioration of the disease in the patient is observed, although the patient may still be suffering from the underlying disease. Prophylactic benefit refers to the use of a composition by a patient to prevent the risk of a disease, or the administration of a composition by a patient presenting with one or more physiological conditions of a disease, although the disease has not yet been diagnosed.
The term "effective amount" (e.g., "therapeutically effective amount" or "prophylactically effective amount") as used herein refers to an amount of active ingredient that will achieve the desired effect to some extent upon administration, e.g., to alleviate one or more symptoms of the treated condition or to prevent the occurrence of the condition or symptoms thereof.
As used herein, "individual" includes a human or non-human animal. Exemplary human individuals include human individuals (referred to as patients) having a disease (e.g., a disease described herein) or normal individuals. "non-human animals" in the context of the present invention include all vertebrates, such as non-mammals (e.g., birds, amphibians, reptiles) and mammals, such as non-human primates, livestock and/or domesticated animals (e.g., sheep, dogs, cats, cows, pigs, etc.).
The following detailed description is intended to illustrate non-limiting embodiments and to enable others skilled in the art to more fully understand the teachings of the present invention, its principles, and its practical application, so that others skilled in the art may modify and implement the invention in various forms, which are best suited to the requirements of a particular use.
Compounds of the invention
In one aspect, the present invention provides a compound of formula (I):
Figure BDA0001836596920000101
or a pharmaceutically acceptable salt, stereoisomer, solvate, polymorph, tautomer, isotopic compound, metabolite, or prodrug thereof;
wherein:
x is selected from O, S and NRx;RxSelected from H, C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with one or more RASubstitution;
y is selected from O, S, N, or Y is absent;
z is selected from N, O, S, C and CH;
q is selected from O, S and NH;
when Y is N, R1Selected from H, hydroxyl, sulfydryl, amino, halogen, nitro, cyano, C1-C8Alkyl radical, C2-C8Alkenyl radical, C2-C8Alkynyl, C6-C10Aryl and five to ten membered heteroaryl; wherein said alkyl, alkenyl, alkynyl, aryl or heteroaryl is optionally substituted with one or more RBSubstitution;
R2selected from H, hydroxyl, sulfydryl, amino, halogen, nitro, cyano, C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with one or more RCSubstitution;
L1is C1-C8A hydrocarbon chain;
ring a is absent, or ring a is selected from: c3-C10Cycloalkyl radical, C3-C10Heterocyclic group, C6-C10Aryl and five to ten membered heteroaryl; wherein said cycloalkyl, heterocyclyl, aryl, heteroaryl is optionally substituted with one or more RDSubstitution;
L2is absent, or L2Is C1-C8A hydrocarbon chain;
R3is H, C3-C10Cycloalkyl radical, C6-C10Aryl, five-to ten-membered heteroaryl or-WR6R7Wherein W is N or CH, wherein
R6And R7Each independently selected from H, C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with one or more RESubstitution; or
R6And R7Together with the N or C atom to which they are attached form a three-to ten-membered heterocyclic ring; said heterocycle optionallyBy one or more RESubstitution; and optionally, R6And R7The three-to ten-membered heterocyclic ring formed together with the N or C atom to which it is attached may optionally be fused to the a ring;
R4is selected from C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group; wherein said alkyl, alkenyl or and alkynyl are each optionally substituted with one or more RFSubstitution;
ring B is absent, or ring B is selected from C3-C10Cycloalkyl radical, C3-C10Heterocyclic group, C6-C10Aryl and five to ten membered heteroaryl; wherein said cycloalkyl, heterocyclyl, aryl and heteroaryl are each optionally substituted with one or more RGSubstitution;
R5selected from H, C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with one OR more groups selected from-OR8、-SR8and-NR8R8' is substituted with a substituent;
R8and R8' each is independently selected from H, C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with one or more RHSubstitution; or R8And R8' together with the N atom to which it is attached, form a three-to ten-membered heterocyclic ring, preferably a four-to six-membered heterocyclic ring;
RA、RB、RC、RD、RE、RF、RG、RHeach occurrence is independently selected from halogen, cyano, nitro, -Ra、-ORa、=O、-SRa、-NRaRb、=NRa-C (halogen)3-CR (halogen)2、-CR2(halogen), -OCN, -SCN, -N ═ C ═ O, -NCS, -NO2、-N(Ra)C(=O)Rb、-N(Ra)C(=O)ORb、-N(Ra)C(=O)NRbRc、-C(=O)NRaRb、-C(=O)ORa、-OC(=O)NRaRb、-OC(=O)Ra、-OC(=O)ORa、-C(=O)Ra、-S(=O)2ORa、-S(=O)2Ra、-OS(=O)2Ra、-OS(=O)2ORa、-S(=O)2NRaRb、-S(=O)Ra、-N(Ra)S(=O)2Rb、-N(Ra)S(=O)2NRbRc、-N(Ra)S(=O)2ORb、-OP(=O)(ORa)ORb、-P(=O)(ORa)ORb、-C(=O)Ra、-C(=S)Ra、-C(=O)ORa、-C(=S)ORa、-C(=O)SRa、-C(=S)SRa、-C(=O)NRaRb、-C(=S)NRaRb、-C(=NRa)NRbRcand-NRaC(=NRb)NRcRd;Ra、Rb、RcAnd RdEach occurrence is independently selected from H, C1-C8Alkyl radical, C3-C8Cycloalkyl, a three-to eight-membered heterocyclic group, C6-C10Aryl, five-to ten-membered heteroaryl, C6-C10aryl-C1-C4Alkyl and five-to ten-membered heteroaryl-C1-C4An alkyl group; wherein said alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, aryl-C1-C4Alkyl, heteroaryl-C1-C4Each alkyl group is optionally substituted by one or more halogen, ═ O, -OH, -NH2-SH substitution;
wherein when R isA、RB、RC、RD、RE、RF、RGOr RHEach independently is-NRaRbWhen R isa、RbOptionally together with the N atom to which they are attached form a three-to ten-membered heterocyclic ringThe ring is preferably a four-to six-membered heterocyclic ring.
In one embodiment, Q, which is attached by a double bond to the heterocyclic ring consisting of the pyrimidine-5-carbon atom, the pyrimidine-6-carbon atom, the carbon atom between N, Z, N and Z, and optionally Y, is selected from O and S.
In one embodiment, X is O. In another embodiment, X is S. In yet another embodiment, X is NRxAnd R isxSelected from H, C1-C4Alkyl radical, C2-C4Alkenyl and C2-C4An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with one or more RAAnd (4) substitution. In another embodiment, X is N, and R isxSelected from H and C1-C4An alkyl group; wherein said alkyl is optionally substituted with one or more RAAnd (4) substitution.
In one embodiment, Y is O or S, and R1Is absent.
In one embodiment, Y is N, and R is1Selected from H, C1-C8Alkyl radical, C2-C8Alkenyl radical, C2-C8Alkynyl, C6-C10Aryl and five to ten membered heteroaryl; wherein said alkyl, alkenyl, alkynyl, aryl and heteroaryl are each optionally substituted with one or more RBSubstitution; wherein the five-to ten-membered heteroaryl contains 1-3 heteroatoms each independently selected from O, N and S. In a preferred embodiment, Y is N, and R is1Selected from H, C1-C4Alkyl radical, C3-C4Alkenyl radical, C3-C4Alkynyl, C6-C10Aryl and five to ten membered heteroaryl; wherein said alkyl, alkenyl, alkynyl, aryl and heteroaryl are each optionally substituted with one or more RBSubstitution; wherein the five-to ten-membered heteroaryl contains 1-3 heteroatoms each independently selected from O, N and S. In a more preferred embodiment, R1Is H or C1-C4An alkyl group; preferably H or C1-C3An alkyl group; further preferred is C1-C2An alkyl group; wherein said alkyl is optionally substituted with one or more RBAnd (4) substitution. More preferably, R1Is H or C1-C2An alkyl group; wherein said alkyl is optionally substituted with one or more halogens. In a further preferred embodiment, R1Is H, CH3、CF3Or CH2CF3(ii) a Particularly preferred is H or CH3
In another embodiment, Y and R1Absent, i.e., the N-Y structure is actually N.
In one embodiment, Z is O or S, and R2Is absent.
In one embodiment, Z is N, and R is2Selected from H, hydroxy, mercapto, amino, C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with one or more RCAnd (4) substitution. In another embodiment, Z is N, and R is2Selected from H, hydroxy, mercapto, amino, C1-C4Alkyl radical, C2-C4Alkenyl and C2-C4An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with one or more RCAnd (4) substitution. In a preferred embodiment, Z is N, and R is2Selected from H, hydroxy, mercapto, C1-C4Alkyl radical, C3-C4Alkenyl and C3-C4An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with one or more RCAnd (4) substitution. In a more preferred embodiment, R2Is H, hydroxy, mercapto or C1-C4An alkyl group; preferably H, hydroxy, mercapto or C1-C3An alkyl group; wherein said alkyl is optionally substituted with one or more RCAnd (4) substitution. More preferably, R2Is H, hydroxy or methyl; wherein said methyl is optionally substituted with one or more halogens. In a further preferred embodiment, R2Is H, hydroxy or methyl; h or hydroxyl is particularly preferred.
In one embodiment, Z is C or CH, and R2Selected from H, hydroxyl, sulfydryl, amino, halogen, nitro, cyano, C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group; wherein alkyl, alkenyl and alkynyl are each optionally substituted with one or more RCAnd (4) substitution. In yet another embodiment, Z is C or CH, and R2Selected from H, hydroxy, mercapto, halogen, C1-C4Alkyl radical, C2-C4Alkenyl and C2-C4An alkynyl group; wherein alkyl, alkenyl and alkynyl are each optionally substituted with one or more RCAnd (4) substitution. In another embodiment, Z is CH, and R2Selected from H, hydroxy, mercapto, halogen, C1-C4Alkyl radical, C2-C4Alkenyl and C2-C4An alkynyl group; wherein alkyl, alkenyl and alkynyl are each optionally substituted with one or more RCAnd (4) substitution. In a preferred embodiment, Z is CH, and R is2Is H, hydroxy, mercapto, halogen or C1-C4An alkyl group; preferably H, hydroxy, mercapto, halogen or C1-C3An alkyl group; wherein said alkyl is optionally substituted with one or more RCAnd (4) substitution. More preferably, Z is CH, and R2Is H, hydroxy, halogen or methyl; wherein said methyl is optionally substituted with one or more halogens. In a further preferred embodiment, Z is CH, and R is2H, F, hydroxy or methyl, particularly preferably H or hydroxy.
In one embodiment, L1Is- (CH)2)n-; wherein n is 1,2, 3,4, 5,6, 7 or 8; preferably 1,2, 3,4 or 5, more preferably 1,2 or 3; particularly preferably 1.
In one embodiment, ring A, ring L2And R3At least one of which is present in the general formula (I). When present, ring A, L2And R3Contains at least one N atom.
In one embodiment, ring A is absent and L is1And L2Are directly connected together. In another embodiment, the a ring is selected from phenyl, pyridine, furan, thiophene, and pyrrole; and each optionally substituted with one or more RDAnd (4) substitution. In another embodiment, the A ring is selected fromCyclohexane, cyclopentane, cyclobutane, and cyclopropane; and each optionally substituted with one or more RDAnd (4) substitution. In yet another embodiment, the a ring is selected from the group consisting of: bicyclo [2.2.1]Heptane, with 1-carbon and 4-carbon atoms attached to the rest of the molecule; bicyclo [1.1.1]Pentane, attached to the rest of the molecule at 1-carbon and 3-carbon atoms; 7-Oxabicyclo [2.2.1]Heptane, with 1-carbon and 4-carbon atoms attached to the rest of the molecule; wherein said bicyclo [2.2.1]Heptane, bicyclo [1.1.1]Pentane and 7-oxabicyclo [2.2.1 ]]Heptane; and each optionally substituted with one or more RDAnd (4) substitution. In a preferred embodiment, the a ring is selected from phenyl and pyridine; and each optionally substituted with one or more RDSubstitution; wherein R isDHalogen is preferred, and F is particularly preferred. In a further preferred embodiment, the a ring is phenyl, and is optionally substituted with one or more RDSubstitution; wherein R isDHalogen is preferred, and F is particularly preferred.
In one embodiment, L2Is selected from- (CH)2)n-; wherein n is 1,2, 3,4 or 5,6, 7 or 8; preferably 1,2, 3,4 or 5, more preferably 1,2 or 3; particularly preferably 1.
In one embodiment, R3Is H.
In yet another embodiment, R3is-WR6R7Wherein W is CH, i.e. R3is-CH (R)6)R7
Wherein R is6And R7Each independently selected from H, C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with one or more RESubstitution; or
R6And R7Together with the C atom to which they are attached form a three-to ten-membered heterocyclic ring; wherein said heterocycle contains 1-3 heteroatoms independently selected from O, S and N, and is optionally substituted with one or more RESubstitution;
R6and R7Together with the C atom to which they are attached form a triad toThe ten-membered heterocyclic ring may optionally be fused to the A ring.
In another embodiment, R3is-WR6R7Wherein W is N, i.e. R3is-NR6R7
Wherein R is6And R7Each independently selected from H, C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with one or more RESubstitution; or
R6And R7Together with the N atom to which they are attached form a three-to ten-membered heterocyclic ring; wherein said heterocycle contains 0-2 additional heteroatoms independently selected from O, S and N, and is optionally substituted with one or more RESubstitution;
R6and R7The three-to ten-membered heterocyclic ring formed together with the N atom to which it is attached may optionally be fused to the a ring.
In a preferred embodiment, R3Is selected from-N (C)1-C4Alkyl) (C1-C4Alkyl), pyrrolidine, piperidine, morpholine, and piperazine; wherein said alkyl, pyrrolidine, piperidine, morpholine and piperazine are each optionally substituted with one or more REAnd (4) substitution. In a further preferred embodiment, R3Is selected from-N (CH)3)2Pyrrolidin-1-yl, piperidin-1-yl, morpholin-1-yl and 4-C1-C3Alkyl-piperazin-1-yl; wherein said CH3Pyrrolidin-1-yl, piperidin-1-yl, morpholin-1-yl and piperazin-1-yl each optionally substituted with one or more REAnd (4) substitution. In a further preferred embodiment, R3Is pyrrolidin-1-yl or piperidin-1-yl; wherein said pyrrolidin-1-yl and piperidin-1-yl are each optionally substituted with one or more REAnd (4) substitution.
In one embodiment, L2And ring A is absent, R3Directly with L1And (4) connecting.
In one embodiment, R4Is C1-C4Alkyl, wherein the alkyl is optionally monoA plurality of RFAnd (4) substitution. In a preferred embodiment, R4Is C1-C3Alkyl, especially C3An alkyl group; wherein said alkyl is optionally substituted with one or more RFAnd (4) substitution. In another preferred embodiment, R4is-CH2-、-CH(CH3)-、-CH(C2H5)-、-CH(C3H7-n)-、-CH(CH2ORa)-、-CH(CH2SRa)-、-C(CH2ORa)(Rb)-、-C(CH2ORa)(ORb)-、-CH(C(=O)ORa)-、-CH(S(=O)Ra)-、-CH(S(=O)2Ra)-、-CH(S(=O)2ORa)-、-CH(OC(=O)Ra)-、-CH(OS(=O)2Ra)-、-C(C(=O)ORa)(Rb)-、-C(S(=O)Ra)(Rb)-、-C(S(=O)2Ra)(Rb)-、-C(S(=O)2ORa)(Rb)-、-C(OC(=O)Ra)(Rb)-、-C(OS(=O)2Ra)(Rb)-、-C(C(=O)ORa)(ORb)-、-C(S(=O)Ra)(ORb)-、-C(S(=O)2Ra)(ORb)-、-C(S(=O)2ORa)(ORb)-、-CH(CH2CH2ORa)-、-CH(CH2CH2SRa)-、-C(CH2CH2ORa)(Rb)-、-C(CH2CH2ORa)(ORb)-、-CH(CH2C(=O)ORa)-、-CH(CH2S(=O)Ra)-、-CH(CH2S(=O)2Ra)-、-CH(CH2S(=O)2ORa)-、-CH(CH2OC(=O)Ra)-、-CH(CH2OS(=O)2Ra)-、-C(CH2C(=O)ORa)(Rb)-、-C(CH2S(=O)Ra)(Rb)-、-C(CH2S(=O)2Ra)(Rb)-、-C(CH2S(=O)2ORa)(Rb)-、-C(CH2OC(=O)Ra)(Rb)-、-C(CH2OS(=O)2Ra)(Rb)-、-C(CH2C(=O)ORa)(ORb)-、-C(CH2S(=O)Ra)(ORb)-、-C(CH2S(=O)2Ra)(ORb)-、-C(CH2S(=O)2ORa)(ORb)-、-C(CH2OC(=O)Ra)(ORb)-、-C(CH2OS(=O)2Ra)(ORb)-、-CH(CH2CH2S(=O)Ra)-、-CH(CH2CH2S(=O)2Ra)-、-CH(CH2CH2S(=O)2ORa)-、-CH(CH2CH2OC(=O)Ra)-、-CH(CH2CH2OS(=O)2Ra)-、-C(CH2CH2S(=O)Ra)(Rb)-、-C(CH2CH2S(=O)2Ra)(Rb)-、-C(CH2CH2S(=O)2ORa)(Rb)-、-C(CH2CH2OC(=O)Ra)(Rb)-、-C(CH2CH2OS(=O)2Ra)(Rb)-、-C(CH2CH2S(=O)Ra)(ORb)-、-C(CH2CH2S(=O)2Ra)(ORb)-、-C(CH2CH2S(=O)2ORa)(ORb)-、-C(CH2CH2OC(=O)Ra)(ORb)-、-C(CH2CH2OS(=O)2Ra)(ORb)-、-CH(CH2NRaRb)-、-C(CH2NRaRb)(Rc)-、-C(CH2NRaRb)(ORc)-、-CH(CH2CH2NRaRb)-、-C(CH2CH2NRaRb)(Rc)-、-C(CH2CH2NRaRb)(ORc)-、-CH(CH2C(=O)NRaRb)-、-CH(CH2S(=O)2NRaRb)-、-C(CH2C(=O)NRaRb)(Rc)-、-C(CH2S(=O)2NRaRb)(Rc)-、-C(CH2C(=O)NRaRb)(ORc)-、-C(CH2S(=O)2NRaRb)(ORc)-、-CH(CH2N(Ra)C(=O)Rb)-、-CH(CH2N(Ra)S(=O)2Rb)-、-CH(CH2CH2S(=O)2NRaRb)-、-CH(CH2CH2N(Ra)C(=O)Rb)-、-CH(CH2CH2N(Ra)S(=O)2Rb)-、-CH(CH2N(Ra)C(=O)Rb)(Rc)-、-CH(CH2N(Ra)S(=O)2Rb)(Rc)-、-CH(CH2CH2S(=O)2NRaRb)(Rc)-、-CH(CH2CH2N(Ra)C(=O)Rb)(Rc)-、-CH(CH2CH2N(Ra)S(=O)2Rb)(Rc)-、-CH(CH2N(Ra)C(=O)Rb)(ORc)-、-CH(CH2N(Ra)S(=O)2Rb)(ORc)-、-CH(CH2CH2S(=O)2NRaRb)(ORc)-、-CH(CH2CH2N(Ra)C(=O)Rb)(ORc) -or-CH (CH)2CH2N(Ra)S(=O)2Rb)(ORc) -; more preferably-CH (C)3H7-n)-、-CH(CH2ORa)-、-CH(CH2SRa)-、-CH(CH2CH2ORa)-、-CH(CH2CH2SRa)-、-CH(CH2C(=O)ORa)-、-CH(CH2S(=O)2Ra)-、-CH(CH2S(=O)2ORa)-、-CH(CH2OC(=O)Ra)-、-CH(CH2OS(=O)2Ra)-、-CH(CH2CH2OS(=O)2Ra)-、-CH(CH2CH2OC(=O)Ra)-、-CH(CH2CH2N(Ra)S(=O)2Rb)-、-CH(CH2NRaRb)-、-CH(CH2CH2NRaRb)-、-CH(CH2CH2N(Ra)C(=O)Rb)-、-CH(CH2C(=O)NRaRb) -or-CH (CH)2S(=O)2NRaRb) -; further preferred is-CH (C)3H7-n)-、-CH(CH2CH2ORa)-、-CH(CH2CH2SRa)-、-CH(CH2C(=O)ORa)-、-CH(CH2S(=O)2Ra)-、-CH(CH2S(=O)2ORa)-、-CH(CH2OC(=O)Ra)-、-CH(CH2OS(=O)2Ra)-、-CH(CH2CH2OS(=O)2Ra)-、-CH(CH2CH2OC(=O)Ra)-、-CH(CH2CH2N(Ra)S(=O)2Rb)-、-CH(CH2CH2NRaRb)-、-CH(CH2CH2N(Ra)C(=O)Rb)-、-CH(CH2C(=O)NRaRb) -or-CH (CH)2S(=O)2NRaRb) -; particularly preferred is-CH (CH)2CH2ORa)-、-CH(CH2CH2OS(=O)2Ra)-、-CH(CH2CH2OC(=O)Ra)-、-CH(CH2CH2N(Ra)S(=O)2Rb)-、-CH(CH2C(=O)ORa)-、-CH(CH2S(=O)2ORa)-、-CH(CH2CH2NRaRb)-、-CH(CH2CH2N(Ra)C(=O)Rb)-、-CH(CH2C(=O)NRaRb) -or-CH (CH)2S(=O)2NRaRb)-。
In one embodiment, ring B is absent and R is4And R5Are directly connected together. In another embodiment, the B ring is selected from phenyl, pyridine, furan, thiophene, pyrrole, thiazole, oxazole and pyran; and each optionally substituted with one or more RGAnd (4) substitution. In yet another embodiment, the B ring is selected from the group consisting of pyrrolidine, piperidine, tetrahydrofuran, dihydropyran, tetrahydropyran, and propylene oxide; and each optionally substituted with one or more RGAnd (4) substitution. In another embodiment, ring B is selected from cyclohexane, cyclopentane, cyclobutane, and cyclopropane; and each optionally substituted with one or more RGAnd (4) substitution. In a preferred embodiment, the B ring is phenyl or pyridine; and are each optionally substituted by one or more substituents each independently selected from halogen, cyano, nitro, -Ra、-ORa、-SRaand-NRaRbIs substituted with a group (b).
In one embodiment, R5Is H. In another embodiment, R5Is C3-C6Alkyl, and optionally substituted by one OR more-OR8、-SR8or-NR8R8' substitution; wherein
In one embodiment, ring B is absent and R is5Directly with R4And (4) connecting.
R8And R8' each is independently selected from H and C1-C4An alkyl group; preferably C1-C2An alkyl group; wherein said alkyl is optionally substituted with one or more RHSubstitution;
or R8And R8' connected to, -NR8R8' formation of a four-to eight-membered heterocyclic ring; wherein said heterocycle contains 0-1 additional heteroatoms independently selected from O, S and N, and is optionally substituted with one or more RHAnd (4) substitution. In a preferred embodiment, -NR8R8' formation of a four-to six-membered heterocyclic ring; wherein said heterocyclic ring contains 0-1 independently selectedAn additional heteroatom from O, S and N, and optionally substituted with one or more RHAnd (4) substitution.
In a preferred embodiment of the present invention, embodiments of the compound of formula (I) may include compounds of formulae (I-1) to (I-7).
In one embodiment of the present invention, the present invention provides a compound of formula (I-1) or a pharmaceutically acceptable salt, stereoisomer, solvate, polymorph, tautomer, isotopic compound, metabolite or prodrug thereof:
Figure BDA0001836596920000161
wherein R is1、R2、R3、R4、R5、L1、L2X, Q, A and B are as defined for formula (I).
In a preferred embodiment, X and Q are each independently selected from O and S.
In a preferred embodiment, R1Is H or C1-C4An alkyl group; preferably H or C1-C3An alkyl group; further preferred is C1-C2An alkyl group; wherein said alkyl is optionally substituted with one or more RBAnd (4) substitution. More preferably, R1Is H or C1-C2An alkyl group; wherein said alkyl is optionally substituted with one or more halogens. In a further preferred embodiment, R1Is H, CH3、CF3Or CH2CF3(ii) a Particularly preferred is H or CH3
In a preferred embodiment, R2Selected from H, hydroxy, mercapto, amino, C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with one or more RCAnd (4) substitution. In another preferred embodiment, R2Selected from H, hydroxy, mercapto, amino, C1-C4Alkyl radical, C2-C4Alkenyl and C2-C4An alkynyl group;wherein said alkyl, alkenyl and alkynyl are each optionally substituted with one or more RCAnd (4) substitution. In a further preferred embodiment, R2Selected from H, hydroxy, mercapto, C1-C4Alkyl radical, C3-C4Alkenyl and C3-C4An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with one or more RCAnd (4) substitution. In a more preferred embodiment, R2Is H, hydroxy, mercapto or C1-C4An alkyl group; preferably H, hydroxy, mercapto or C1-C3An alkyl group; wherein said alkyl is optionally substituted with one or more RCAnd (4) substitution. More preferably, R2Is H, hydroxy or methyl; wherein said methyl is optionally substituted with one or more halogens. In a further preferred embodiment, R2Is H, hydroxy or methyl; h or hydroxyl is particularly preferred.
In a preferred embodiment, L1Is- (CH)2)n-; wherein n is 1,2 or 3; particularly preferably 1.
In a preferred embodiment, ring A, ring L2And R3At least one of which is present in the general formula (I). When present, ring A, L2And R3Contains at least one N atom.
In a preferred embodiment, the a ring is selected from the group consisting of phenyl, pyridine, furan, thiophene, and pyrrole; and each optionally substituted with one or more RDAnd (4) substitution. In a more preferred embodiment, the a ring is selected from phenyl and pyridine; and each optionally substituted with one or more RDSubstitution; wherein R isDHalogen is preferred, and F is particularly preferred. In a further preferred embodiment, the a ring is phenyl, and is optionally substituted with one or more RDSubstitution; wherein R isDHalogen is preferred, and F is particularly preferred.
In a preferred embodiment, L2Is selected from- (CH)2)n-; wherein n is 1,2 or 3; particularly preferably 1.
In a preferred embodiment, R3Is a five-to ten-membered heteroaryl or-WR6R7Wherein W is N or CH, wherein
R6And R7Each independently selected from H, C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with one or more RESubstitution; or
R6And R7Together with the N or C atom to which they are attached form a three-to ten-membered heterocyclic ring; said heterocycle containing 1-3 heteroatoms independently selected from O, S and N, and optionally substituted with one or more REAnd (4) substitution.
In a more preferred embodiment, R3is-NR6R7
Wherein R is6And R7Each independently selected from H, C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with one or more RESubstitution; or
R6And R7Together with the N atom to which they are attached form a three-to ten-membered heterocyclic ring; wherein said heterocycle contains 0-2 additional heteroatoms independently selected from O, S and N, and is optionally substituted with one or more REAnd (4) substitution.
In a preferred embodiment, R3Is selected from-N (C)1-C4Alkyl) (C1-C4Alkyl), pyrrolidine, piperidine, morpholine, and piperazine, wherein the alkyl, pyrrolidine, piperidine, morpholine, and piperazine are each optionally substituted with one or more REAnd (4) substitution. In a further preferred embodiment, R3Is selected from-N (CH)3)2Pyrrolidin-1-yl, piperidin-1-yl, morpholin-1-yl and 4-C1-C3Alkyl-piperazin-1-yl; wherein said CH3Pyrrolidin-1-yl, piperidin-1-yl, morpholin-1-yl and piperazin-1-yl each optionally substituted with one or more REAnd (4) substitution. In a further preferred embodiment, R3Is pyrrolidin-1-yl or piperidin-1-yl; wherein said pyrrolidin-1-yl and piperidin-1-yl are each optionally monoA plurality of REAnd (4) substitution.
In a preferred embodiment, R4Is C1-C4Alkyl, especially C3An alkyl group; wherein said alkyl is optionally substituted with one or more RFAnd (4) substitution. In another preferred embodiment, R4is-CH (C)3H7-n)-、-CH(CH2ORa)-、-CH(CH2SRa)-、-CH(CH2CH2ORa)-、-CH(CH2CH2SRa)-、-CH(CH2C(=O)ORa)-、-CH(CH2S(=O)2Ra)-、-CH(CH2S(=O)2ORa)-、-CH(CH2OC(=O)Ra)-、-CH(CH2OS(=O)2Ra)-、-CH(CH2CH2OS(=O)2Ra)-、-CH(CH2CH2OC(=O)Ra)-、-CH(CH2CH2N(Ra)S(=O)2Rb)-、-CH(CH2NRaRb)-、-CH(CH2CH2NRaRb)-、-CH(CH2CH2N(Ra)C(=O)Rb)-、-CH(CH2C(=O)NRaRb) -or-CH (CH)2S(=O)2NRaRb) -; particularly preferred is-CH (CH)2CH2ORa)-、-CH(CH2CH2OS(=O)2Ra)-、-CH(CH2CH2OC(=O)Ra)-、-CH(CH2CH2N(Ra)S(=O)2Rb)-、-CH(CH2C(=O)ORa)-、-CH(CH2S(=O)2ORa)-、-CH(CH2CH2NRaRb)-、-CH(CH2CH2N(Ra)C(=O)Rb)-、-CH(CH2C(=O)NRaRb) -or-CH (CH)2S(=O)2NRaRb)-。
In a preferred embodiment, ring B is absent and R is4And R5Are directly connected together.
In a preferred embodiment, R5Is H.
In another embodiment of the present invention, the present invention provides a compound of formula (I-2), or a pharmaceutically acceptable salt, stereoisomer, solvate, polymorph, tautomer, isotopic compound, metabolite or prodrug thereof:
Figure BDA0001836596920000181
wherein R is1、R2、R3、R4、R5、L1、L2X, A and B are as defined for formula (I).
In a preferred embodiment, X is selected from O and S.
In a preferred embodiment, R1Is H or C1-C4An alkyl group; preferably H or C1-C3An alkyl group; further preferred is C1-C2An alkyl group; wherein said alkyl is optionally substituted with one or more RBAnd (4) substitution. More preferably, R1Is H or C1-C2An alkyl group; wherein said alkyl is optionally substituted with one or more halogens. In a further preferred embodiment, R1Is H, CH3、CF3Or CH2CF3(ii) a Particularly preferred is H or CH3
In a preferred embodiment, R2Selected from H, hydroxy, mercapto, amino, C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with one or more RCAnd (4) substitution. In another preferred embodiment, R2Selected from H, hydroxy, mercapto, amino, C1-C4Alkyl radical, C2-C4Alkenyl and C2-C4An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with one or more RCAnd (4) substitution. In a further preferred embodiment, R2Is selected fromH. Hydroxy, mercapto, C1-C4Alkyl radical, C3-C4Alkenyl and C3-C4An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with one or more RCAnd (4) substitution. In a more preferred embodiment, R2Is H, hydroxy, mercapto or C1-C4An alkyl group; preferably H, hydroxy, mercapto or C1-C3An alkyl group; wherein said alkyl is optionally substituted with one or more RCAnd (4) substitution. More preferably, R2Is H, hydroxy or methyl; wherein said methyl is optionally substituted with one or more halogens. In a further preferred embodiment, R2Is H, hydroxy or methyl; h or hydroxyl is particularly preferred.
In a preferred embodiment, L1Is- (CH)2)n-; wherein n is 1,2 or 3; particularly preferably 1.
In a preferred embodiment, ring A, ring L2And R3At least one of which is present in the general formula (I). When present, ring A, L2And R3Contains at least one N atom.
In a preferred embodiment, the a ring is selected from the group consisting of phenyl, pyridine, furan, thiophene, and pyrrole; and each optionally substituted with one or more RDAnd (4) substitution. In a more preferred embodiment, the a ring is selected from phenyl and pyridine; and each optionally substituted with one or more RDSubstitution; wherein R isDHalogen is preferred, and F is particularly preferred. In a further preferred embodiment, the a ring is phenyl, and is optionally substituted with one or more RDSubstitution; wherein R isDHalogen is preferred, and F is particularly preferred.
In a preferred embodiment, L2Is selected from- (CH)2)n-; wherein n is 1,2 or 3; particularly preferably 1.
In a preferred embodiment, R3Is a five-to ten-membered heteroaryl or-WR6R7Wherein W is N or CH, wherein
R6And R7Each independently selected from H, C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with one or more RESubstitution; or
R6And R7Together with the N or C atom to which they are attached form a three-to ten-membered heterocyclic ring; said heterocycle containing 1-3 heteroatoms independently selected from O, S and N, and optionally substituted with one or more REAnd (4) substitution.
In a more preferred embodiment, R3is-NR6R7
Wherein R is6And R7Each independently selected from H, C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with one or more RESubstitution; or
R6And R7Together with the N atom to which they are attached form a three-to ten-membered heterocyclic ring; wherein said heterocycle contains 0-2 additional heteroatoms independently selected from O, S and N, and is optionally substituted with one or more REAnd (4) substitution.
In a preferred embodiment, R3Is selected from-N (C)1-C4Alkyl) (C1-C4Alkyl), pyrrolidine, piperidine, morpholine, and piperazine, wherein the alkyl, pyrrolidine, piperidine, morpholine, and piperazine are each optionally substituted with one or more REAnd (4) substitution. In a further preferred embodiment, R3Is selected from-N (CH)3)2Pyrrolidin-1-yl, piperidin-1-yl, morpholin-1-yl and 4-C1-C3Alkyl-piperazin-1-yl; wherein said CH3Pyrrolidin-1-yl, piperidin-1-yl, morpholin-1-yl and piperazin-1-yl each optionally substituted with one or more REAnd (4) substitution. In a further preferred embodiment, R3Is pyrrolidin-1-yl or piperidin-1-yl; wherein said pyrrolidin-1-yl and piperidin-1-yl are each optionally substituted with one or more REAnd (4) substitution.
In a preferred embodiment, R4Is C1-C4Alkyl, especially C3An alkyl group; wherein said alkyl is optionally substituted with one or more RFAnd (4) substitution. In another preferred embodiment, R4is-CH (C)3H7-n)-、-CH(CH2ORa)-、-CH(CH2SRa)-、-CH(CH2CH2ORa)-、-CH(CH2CH2SRa)-、-CH(CH2C(=O)ORa)-、-CH(CH2S(=O)2Ra)-、-CH(CH2S(=O)2ORa)-、-CH(CH2OC(=O)Ra)-、-CH(CH2OS(=O)2Ra)-、-CH(CH2CH2OS(=O)2Ra)-、-CH(CH2CH2OC(=O)Ra)-、-CH(CH2CH2N(Ra)S(=O)2Rb)-、-CH(CH2NRaRb)-、-CH(CH2CH2NRaRb)-、-CH(CH2CH2N(Ra)C(=O)Rb)-、-CH(CH2C(=O)NRaRb) -or-CH (CH)2S(=O)2NRaRb) -; particularly preferred is-CH (CH)2CH2ORa)-、-CH(CH2CH2OS(=O)2Ra)-、-CH(CH2CH2OC(=O)Ra)-、-CH(CH2CH2N(Ra)S(=O)2Rb)-、-CH(CH2C(=O)ORa)-、-CH(CH2S(=O)2ORa)-、-CH(CH2CH2NRaRb)-、-CH(CH2CH2N(Ra)C(=O)Rb)-、-CH(CH2C(=O)NRaRb) -or-CH (CH)2S(=O)2NRaRb)-。
In a preferred embodiment, ring B is absent and R is4And R5Are directly connected together.
In a preferred embodiment, R5Is H.
In yet another embodiment of the present invention, the present invention provides a compound of formula (I-3), or a pharmaceutically acceptable salt, stereoisomer, solvate, polymorph, tautomer, isotopic compound, metabolite or prodrug thereof:
Figure BDA0001836596920000201
wherein R is1、R2、R3、RF、R5、L1、L2X, A and B are as defined for formula (I).
In a preferred embodiment, X is selected from O and S.
In a preferred embodiment, R1Is H or C1-C4An alkyl group; preferably H or C1-C3An alkyl group; further preferred is C1-C2An alkyl group; wherein said alkyl is optionally substituted with one or more RBAnd (4) substitution. More preferably, R1Is H or C1-C2An alkyl group; wherein said alkyl is optionally substituted with one or more halogens. In a further preferred embodiment, R1Is H, CH3、CF3Or CH2CF3(ii) a Particularly preferred is H or CH3
In a preferred embodiment, R2Selected from H, hydroxy, mercapto, amino, C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with one or more RCAnd (4) substitution. In another preferred embodiment, R2Selected from H, hydroxy, mercapto, amino, C1-C4Alkyl radical, C2-C4Alkenyl and C2-C4An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with one or more RCAnd (4) substitution. In a further preferred embodiment, R2Selected from H, hydroxy, mercapto, C1-C4Alkyl radical, C3-C4Alkenyl and C3-C4An alkynyl group; wherein said alkyl groupAlkenyl and alkynyl are each optionally substituted with one or more RCAnd (4) substitution. In a more preferred embodiment, R2Is H, hydroxy, mercapto or C1-C4An alkyl group; preferably H, hydroxy, mercapto or C1-C3An alkyl group; wherein said alkyl is optionally substituted with one or more RCAnd (4) substitution. More preferably, R2Is H, hydroxy or methyl; wherein said methyl is optionally substituted with one or more halogens. In a further preferred embodiment, R2Is H, hydroxy or methyl; h or hydroxyl is particularly preferred.
In a preferred embodiment, L1Is- (CH)2)n-; wherein n is 1,2 or 3; particularly preferably 1.
In a preferred embodiment, ring A, ring L2And R3At least one of which is present in the general formula (I). When present, ring A, L2And R3Contains at least one N atom.
In a preferred embodiment, the a ring is selected from the group consisting of phenyl, pyridine, furan, thiophene, and pyrrole; and each optionally substituted with one or more RDAnd (4) substitution. In a more preferred embodiment, the a ring is selected from phenyl and pyridine; and each optionally substituted with one or more RDSubstitution; wherein R isDHalogen is preferred, and F is particularly preferred. In a further preferred embodiment, the a ring is phenyl, and is optionally substituted with one or more RDSubstitution; wherein R isDHalogen is preferred, and F is particularly preferred.
In a preferred embodiment, L2Is selected from- (CH)2)n-; wherein n is 1,2 or 3; particularly preferably 1.
In a preferred embodiment, R3Is a five-to ten-membered heteroaryl or-WR6R7Wherein W is N or CH, wherein
R6And R7Each independently selected from H, C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with one or moreRESubstitution; or
R6And R7Together with the N or C atom to which they are attached form a three-to ten-membered heterocyclic ring; said heterocycle containing 1-3 heteroatoms independently selected from O, S and N, and optionally substituted with one or more REAnd (4) substitution.
In a more preferred embodiment, R3is-NR6R7
Wherein R is6And R7Each independently selected from H, C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with one or more RESubstitution; or
R6And R7Together with the N atom to which they are attached form a three-to ten-membered heterocyclic ring; wherein said heterocycle contains 0-2 additional heteroatoms independently selected from O, S and N, and is optionally substituted with one or more REAnd (4) substitution.
In a preferred embodiment, R3Is selected from-N (C)1-C4Alkyl) (C1-C4Alkyl), pyrrolidine, piperidine, morpholine, piperazine, wherein said alkyl, pyrrolidine, piperidine, morpholine and piperazine are each optionally substituted with one or more REAnd (4) substitution. In a further preferred embodiment, R3Is selected from-N (CH)3)2Pyrrolidin-1-yl, piperidin-1-yl, morpholin-1-yl and 4-C1-C3Alkyl-piperazin-1-yl; wherein said CH3Pyrrolidin-1-yl, piperidin-1-yl, morpholin-1-yl and piperazin-1-yl each optionally substituted with one or more REAnd (4) substitution. In a further preferred embodiment, R3Is pyrrolidin-1-yl or piperidin-1-yl; wherein said pyrrolidin-1-yl and piperidin-1-yl are each optionally substituted with one or more REAnd (4) substitution.
In a preferred embodiment, RFIs H, -ORa、-SRa、-C(=O)ORa、-S(=O)2Ra、-S(=O)2ORa、-OC(=O)Ra、-OS(=O)2Ra、-N(Ra)S(=O)2Rb、-NRaRb、-N(Ra)C(=O)Rb、-C(=O)NRaRbor-S (═ O)2NRaRb(ii) a Particularly preferred is-ORa、-OS(=O)2Ra、-OC(=O)Ra、-N(Ra)S(=O)2Rb、-C(=O)ORa、-S(=O)2ORa、-NRaRb、-N(Ra)C(=O)Rb、-C(=O)NRaRbor-S (═ O)2NRaRb
In a preferred embodiment, ring B is absent and R is4And R5Are directly connected together.
In a preferred embodiment, R5Is H.
In still another embodiment of the present invention, the present invention provides a compound of formula (I-4) or a pharmaceutically acceptable salt, stereoisomer, solvate, polymorph, tautomer, isotopic compound, metabolite or prodrug thereof:
Figure BDA0001836596920000211
wherein R is2、R3、R4、R5、L1、L2X, Q, A and B are as defined for formula (I).
In a preferred embodiment, X and Q are each independently selected from O and S.
In a preferred embodiment, R2Selected from H, hydroxy, mercapto, amino, C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with one or more RCAnd (4) substitution. In another preferred embodiment, R2Selected from hydroxy, mercapto, amino, C1-C4Alkyl radical, C2-C4Alkenyl and C2-C4An alkynyl group; wherein saidAlkyl, alkenyl and alkynyl each optionally substituted with one or more RCAnd (4) substitution. In a further preferred embodiment, R2Selected from hydroxy, mercapto, C1-C4Alkyl radical, C3-C4Alkenyl and C3-C4An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with one or more RCAnd (4) substitution. In a more preferred embodiment, R2Is hydroxy, mercapto or C1-C4An alkyl group; preferably hydroxy, mercapto or C1-C3An alkyl group; wherein said alkyl is optionally substituted with one or more RCAnd (4) substitution. More preferably, R2Is hydroxy or methyl; wherein said methyl is optionally substituted with one or more halogens. In a further preferred embodiment, R2Is hydroxy or methyl; particularly preferred is a hydroxyl group.
In a preferred embodiment, L1Is- (CH)2)n-; wherein n is 1,2 or 3; particularly preferably 1.
In a preferred embodiment, ring A, ring L2And R3At least one of which is present in the general formula (I). When present, ring A, L2And R3Contains at least one N atom.
In a preferred embodiment, the a ring is selected from the group consisting of phenyl, pyridine, furan, thiophene, and pyrrole; and each optionally substituted with one or more RDAnd (4) substitution. In a more preferred embodiment, the a ring is selected from phenyl and pyridine; and each optionally substituted with one or more RDSubstitution; wherein R isDHalogen is preferred, and F is particularly preferred. In a further preferred embodiment, the a ring is phenyl, and is optionally substituted with one or more RDSubstitution; wherein R isDHalogen is preferred, and F is particularly preferred.
In a preferred embodiment, L2Is selected from- (CH)2)n-; wherein n is 1,2 or 3; particularly preferably 1.
In a preferred embodiment, R3Is a five-to ten-membered heteroaryl or-WR6R7Wherein W is N or CH, wherein
R6And R7Each independently selected from H, C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with one or more RESubstitution; or
R6And R7Together with the N or C atom to which they are attached form a three-to ten-membered heterocyclic ring; said heterocycle containing 1-3 heteroatoms independently selected from O, S and N, and optionally substituted with one or more REAnd (4) substitution.
In a more preferred embodiment, R3is-NR6R7
Wherein R is6And R7Each independently selected from H, C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with one or more RESubstitution; or
R6And R7Together with the N atom to which they are attached form a three-to ten-membered heterocyclic ring; wherein said heterocycle contains 0-2 additional heteroatoms independently selected from O, S and N, and is optionally substituted with one or more REAnd (4) substitution.
In a preferred embodiment, R3Is selected from-N (C)1-C4Alkyl) (C1-C4Alkyl), pyrrolidine, piperidine, morpholine, piperazine, wherein said alkyl, pyrrolidine, piperidine, morpholine and piperazine are each optionally substituted with one or more REAnd (4) substitution. In a further preferred embodiment, R3Is selected from-N (CH)3)2Pyrrolidin-1-yl, piperidin-1-yl, morpholin-1-yl and 4-C1-C3Alkyl-piperazin-1-yl; wherein said CH3Pyrrolidin-1-yl, piperidin-1-yl, morpholin-1-yl and piperazin-1-yl each optionally substituted with one or more REAnd (4) substitution. In a further preferred embodiment, R3Is pyrrolidin-1-yl or piperidin-1-yl; wherein said pyrrolidin-1-yl and piperidin-1-yl are each optionally substituted with one or more REAnd (4) substitution.
In a preferred embodiment, R4Is C1-C4Alkyl, especially C3An alkyl group; wherein said alkyl is optionally substituted with one or more RFAnd (4) substitution. In another preferred embodiment, R4is-CH (C)3H7-n)-、-CH(CH2ORa)-、-CH(CH2SRa)-、-CH(CH2CH2ORa)-、-CH(CH2CH2SRa)-、-CH(CH2C(=O)ORa)-、-CH(CH2S(=O)2Ra)-、-CH(CH2S(=O)2ORa)-、-CH(CH2OC(=O)Ra)-、-CH(CH2OS(=O)2Ra)-、-CH(CH2CH2OS(=O)2Ra)-、-CH(CH2CH2OC(=O)Ra)-、-CH(CH2CH2N(Ra)S(=O)2Rb)-、-CH(CH2NRaRb)-、-CH(CH2CH2NRaRb)-、-CH(CH2CH2N(Ra)C(=O)Rb)-、-CH(CH2C(=O)NRaRb) -or-CH (CH)2S(=O)2NRaRb) -; particularly preferred is-CH (CH)2CH2ORa)-、-CH(CH2CH2OS(=O)2Ra)-、-CH(CH2CH2OC(=O)Ra)-、-CH(CH2CH2N(Ra)S(=O)2Rb)-、-CH(CH2C(=O)ORa)-、-CH(CH2S(=O)2ORa)-、-CH(CH2CH2NRaRb)-、-CH(CH2CH2N(Ra)C(=O)Rb)-、-CH(CH2C(=O)NRaRb) -or-CH (CH)2S(=O)2NRaRb)-。
In a preferred embodiment, ring B is absent and R is4And R5Are directly connected together.
In a preferred embodiment, R5Is H.
In yet another embodiment of the present invention, the present invention provides a compound of formula (I-5), or a pharmaceutically acceptable salt, stereoisomer, solvate, polymorph, tautomer, isotopic compound, metabolite or prodrug thereof:
Figure BDA0001836596920000231
wherein R is3、R4、R5、L1、L2X, Q, A and B are as defined for formula (I).
In a preferred embodiment, X and Q are each independently selected from O and S.
In a preferred embodiment, L1Is- (CH)2)n-; wherein n is 1,2 or 3; particularly preferably 1.
In a preferred embodiment, ring A, ring L2And R3At least one of which is present in the general formula (I). When present, ring A, L2And R3Contains at least one N atom.
In a preferred embodiment, the a ring is selected from the group consisting of phenyl, pyridine, furan, thiophene, and pyrrole; and each optionally substituted with one or more RDAnd (4) substitution. In a more preferred embodiment, the a ring is selected from phenyl and pyridine; and each optionally substituted with one or more RDSubstitution; wherein R isDHalogen is preferred, and F is particularly preferred. In a further preferred embodiment, the a ring is phenyl, and is optionally substituted with one or more RDSubstitution; wherein R isDHalogen is preferred, and F is particularly preferred.
In a preferred embodiment, L2Is selected from- (CH)2)n-; wherein n is 1,2 or 3; particularly preferably 1.
In a preferred embodiment, R3is-NR6R7In a preferred embodiment, R3Is a five-to ten-membered heteroaryl or-WR6R7Wherein W is N or CH, wherein
R6And R7Each independently selected from H, C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group; wherein
Each of said alkyl, alkenyl and alkynyl is optionally substituted with one or more RESubstitution; or
R6And R7Together with the N or C atom to which they are attached form a three-to ten-membered heterocyclic ring; said heterocycle containing 1-3 heteroatoms independently selected from O, S and N, and optionally substituted with one or more REAnd (4) substitution.
In a more preferred embodiment, R3is-NR6R7
Wherein R is6And R7Each independently selected from H, C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with one or more RESubstitution; or
R6And R7Together with the N atom to which they are attached form a three-to ten-membered heterocyclic ring; wherein said heterocycle contains 0-2 additional heteroatoms independently selected from O, S and N, and is optionally substituted with one or more REAnd (4) substitution.
In a preferred embodiment, R3Is selected from-N (C)1-C4Alkyl) (C1-C4Alkyl), pyrrolidine, piperidine, morpholine, piperazine, wherein said alkyl, pyrrolidine, piperidine, morpholine and piperazine are each optionally substituted with one or more REAnd (4) substitution. In a further preferred embodiment, R3Is selected from-N (CH)3)2Pyrrolidin-1-yl, piperidin-1-yl, morpholin-1-yl and 4-C1-C3Alkyl-piperazin-1-yl; wherein said CH3Pyrrolidin-1-yl, piperidin-1-yl, morpholin-1-yl and piperazin-1-yl each optionally substituted with one or more REAnd (4) substitution. In a further preferred embodiment, R3Is pyrrolidin-1-yl or piperidin-1-yl; whereinSaid pyrrolidin-1-yl and piperidin-1-yl each optionally substituted with one or more REAnd (4) substitution.
In a preferred embodiment, R4Is C1-C4Alkyl, especially C3An alkyl group; wherein said alkyl is optionally substituted with one or more RFAnd (4) substitution. In another preferred embodiment, R4is-CH (C)3H7-n)-、-CH(CH2ORa)-、-CH(CH2SRa)-、-CH(CH2CH2ORa)-、-CH(CH2CH2SRa)-、-CH(CH2C(=O)ORa)-、-CH(CH2S(=O)2Ra)-、-CH(CH2S(=O)2ORa)-、-CH(CH2OC(=O)Ra)-、-CH(CH2OS(=O)2Ra)-、-CH(CH2CH2OS(=O)2Ra)-、-CH(CH2CH2OC(=O)Ra)-、-CH(CH2CH2N(Ra)S(=O)2Rb)-、-CH(CH2NRaRb)-、-CH(CH2CH2NRaRb)-、-CH(CH2CH2N(Ra)C(=O)Rb)-、-CH(CH2C(=O)NRaRb) -or-CH (CH)2S(=O)2NRaRb) -; particularly preferred is-CH (CH)2CH2ORa)-、-CH(CH2CH2OS(=O)2Ra)-、-CH(CH2CH2OC(=O)Ra)-、-CH(CH2CH2N(Ra)S(=O)2Rb)-、-CH(CH2C(=O)ORa)-、-CH(CH2S(=O)2ORa)-、-CH(CH2CH2NRaRb)-、-CH(CH2CH2N(Ra)C(=O)Rb)-、-CH(CH2C(=O)NRaRb) -or-CH (CH)2S(=O)2NRaRb)-。
In a preferred embodimentIn embodiments, ring B is absent and R is4And R5Are directly connected together.
In a preferred embodiment, R5Is H.
In yet another embodiment of the present invention, the present invention provides a compound of formula (I-6), or a pharmaceutically acceptable salt, stereoisomer, solvate, polymorph, tautomer, isotopic compound, metabolite or prodrug thereof:
Figure BDA0001836596920000241
wherein R is3、R4、R5、L1、L2X, A and B are as defined for formula (I).
In a preferred embodiment, X is selected from O and S.
In a preferred embodiment, L1Is- (CH)2)n-; wherein n is 1,2 or 3; particularly preferably 1.
In a preferred embodiment, ring A, ring L2And R3At least one of which is present in the general formula (I). When present, ring A, L2And R3Contains at least one N atom.
In a preferred embodiment, the a ring is selected from the group consisting of phenyl, pyridine, furan, thiophene, and pyrrole; and each optionally substituted with one or more RDAnd (4) substitution. In a more preferred embodiment, the a ring is selected from phenyl and pyridine; and each optionally substituted with one or more RDSubstitution; wherein R isDHalogen is preferred, and F is particularly preferred. In a further preferred embodiment, the a ring is phenyl, and is optionally substituted with one or more RDSubstitution; wherein R isDHalogen is preferred, and F is particularly preferred.
In a preferred embodiment, L2Is selected from- (CH)2)n-; wherein n is 1,2 or 3; particularly preferably 1.
In a preferred embodiment, R3Is a five-to ten-membered heteroaryl or-WR6R7Wherein W is N or CH, wherein
R6And R7Each independently selected from H, C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with one or more RESubstitution; or
R6And R7Together with the N or C atom to which they are attached form a three-to ten-membered heterocyclic ring; said heterocycle containing 1-3 heteroatoms independently selected from O, S and N, and optionally substituted with one or more REAnd (4) substitution.
In a more preferred embodiment, R3is-NR6R7
Wherein R is6And R7Each independently selected from H, C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with one or more RESubstitution; or
R6And R7Together with the N atom to which they are attached form a three-to ten-membered heterocyclic ring; wherein said heterocycle contains 0-2 additional heteroatoms independently selected from O, S and N, and is optionally substituted with one or more REAnd (4) substitution.
In a preferred embodiment, R3Is selected from-N (C)1-C4Alkyl) (C1-C4Alkyl), pyrrolidine, piperidine, morpholine, piperazine, wherein said alkyl, pyrrolidine, piperidine, morpholine and piperazine are each optionally substituted with one or more REAnd (4) substitution. In a further preferred embodiment, R3Is selected from-N (CH)3)2Pyrrolidin-1-yl, piperidin-1-yl, morpholin-1-yl and 4-C1-C3Alkyl-piperazin-1-yl; wherein said CH3Pyrrolidin-1-yl, piperidin-1-yl, morpholin-1-yl and piperazin-1-yl each optionally substituted with one or more REAnd (4) substitution. In a further preferred embodiment, R3Is pyrrolidin-1-yl or piperidin-1-yl; wherein said pyrrolidin-1-yl and piperidin-1-yl are each optionallyOptionally substituted by one or more REAnd (4) substitution.
In a preferred embodiment, R4Is C1-C4Alkyl, especially C3An alkyl group; wherein said alkyl is optionally substituted with one or more RFAnd (4) substitution. In another preferred embodiment, R4is-CH (C)3H7-n)-、-CH(CH2ORa)-、-CH(CH2SRa)-、-CH(CH2CH2ORa)-、-CH(CH2CH2SRa)-、-CH(CH2C(=O)ORa)-、-CH(CH2S(=O)2Ra)-、-CH(CH2S(=O)2ORa)-、-CH(CH2OC(=O)Ra)-、-CH(CH2OS(=O)2Ra)-、-CH(CH2CH2OS(=O)2Ra)-、-CH(CH2CH2OC(=O)Ra)-、-CH(CH2CH2N(Ra)S(=O)2Rb)-、-CH(CH2NRaRb)-、-CH(CH2CH2NRaRb)-、-CH(CH2CH2N(Ra)C(=O)Rb)-、-CH(CH2C(=O)NRaRb) -or-CH (CH)2S(=O)2NRaRb) -; particularly preferred is-CH (CH)2CH2ORa)-、-CH(CH2CH2OS(=O)2Ra)-、-CH(CH2CH2OC(=O)Ra)-、-CH(CH2CH2N(Ra)S(=O)2Rb)-、-CH(CH2C(=O)ORa)-、-CH(CH2S(=O)2ORa)-、-CH(CH2CH2NRaRb)-、-CH(CH2CH2N(Ra)C(=O)Rb)-、-CH(CH2C(=O)NRaRb) -or-CH (CH)2S(=O)2NRaRb)-。
In a preferred embodiment, ring B is absent and R is4And R5Are directly connected together.
In a preferred embodiment, R5Is H.
In yet another embodiment of the present invention, the present invention provides a compound of formula (I-7), or a pharmaceutically acceptable salt, stereoisomer, solvate, polymorph, tautomer, isotopic compound, metabolite or prodrug thereof:
Figure BDA0001836596920000261
wherein R is3、RF、R5、L1、L2X, A and B are as defined for formula (I).
In a preferred embodiment, X is selected from O and S.
In a preferred embodiment, L1Is- (CH)2)n-; wherein n is 1,2 or 3; particularly preferably 1.
In a preferred embodiment, ring A, ring L2And R3At least one of which is present in the general formula (I). When present, ring A, L2And R3Contains at least one N atom.
In a preferred embodiment, the a ring is selected from the group consisting of phenyl, pyridine, furan, thiophene, and pyrrole; and each optionally substituted with one or more RDAnd (4) substitution. In a more preferred embodiment, the a ring is selected from phenyl and pyridine; and each optionally substituted with one or more RDSubstitution; wherein R isDHalogen is preferred, and F is particularly preferred. In a further preferred embodiment, the a ring is phenyl, and is optionally substituted with one or more RDSubstitution; wherein R isDHalogen is preferred, and F is particularly preferred.
In a preferred embodiment, L2Is selected from- (CH)2)n-; wherein n is 1,2 or 3; particularly preferably 1.
In a preferred embodiment, R3Is a five-to ten-membered heteroaryl or-WR6R7Wherein W is N or CH, wherein
R6And R7Each independently selected from H, C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with one or more RESubstitution; or
R6And R7Together with the N or C atom to which they are attached form a three-to ten-membered heterocyclic ring; said heterocycle containing 1-3 heteroatoms independently selected from O, S and N, and optionally substituted with one or more REAnd (4) substitution.
In a more preferred embodiment, R3is-NR6R7
Wherein R is6And R7Each independently selected from H, C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group;
wherein said alkyl, alkenyl and alkynyl are each optionally substituted with one or more RESubstitution; or
R6And R7Together with the N atom to which they are attached form a three-to ten-membered heterocyclic ring; wherein said heterocycle contains 0-2 additional heteroatoms independently selected from O, S and N, and is optionally substituted with one or more REAnd (4) substitution.
In a preferred embodiment, R3Is selected from-N (C)1-C4Alkyl) (C1-C4Alkyl), pyrrolidine, piperidine, morpholine, piperazine, wherein said alkyl, pyrrolidine, piperidine, morpholine and piperazine are each optionally substituted with one or more REAnd (4) substitution. In a further preferred embodiment, R3Is selected from-N (CH)3)2Pyrrolidin-1-yl, piperidin-1-yl, morpholin-1-yl and 4-C1-C3Alkyl-piperazin-1-yl; wherein said CH3Pyrrolidin-1-yl, piperidin-1-yl, morpholin-1-yl and piperazin-1-yl each optionally substituted with one or more REAnd (4) substitution. In a further preferred embodiment, R3Is pyrrolidin-1-yl or piperidin-1-yl; wherein said pyrrolidin-1-yl and piperidin-1-yl are each optionally monoA plurality of REAnd (4) substitution.
In a preferred embodiment, RFIs H, -ORa、-SRa、-C(=O)ORa、-S(=O)2Ra、-S(=O)2ORa、-OC(=O)Ra、-OS(=O)2Ra、-N(Ra)S(=O)2Rb、-NRaRb、-N(Ra)C(=O)Rb、-C(=O)NRaRbor-S (═ O)2NRaRb(ii) a Particularly preferred is-ORa、-OS(=O)2Ra、-OC(=O)Ra、-N(Ra)S(=O)2Rb、-C(=O)ORa、-S(=O)2ORa、-NRaRb、-N(Ra)C(=O)Rb、-C(=O)NRaRbor-S (═ O)2NRaRb
In a preferred embodiment, ring B is absent and R is4And R5Are directly connected together.
In a preferred embodiment, R5Is H.
In certain embodiments, the present invention provides the following compounds, or a pharmaceutically acceptable salt, stereoisomer, solvate, polymorph, tautomer, isotopic compound, metabolite, or prodrug thereof:
Figure BDA0001836596920000271
Figure BDA0001836596920000281
it is to be understood that substituents and substitution patterns on the compounds provided herein can be selected by one of ordinary skill in the art to provide compounds that are chemically stable and can be synthesized by techniques known in the art as well as those described herein.
Pharmaceutical compositions, formulations and kits
The present invention also provides a pharmaceutical composition comprising a compound of the present invention, or a pharmaceutically acceptable salt, stereoisomer, solvate, polymorph, tautomer, isotopic compound, metabolite, or prodrug thereof, and at least one pharmaceutically acceptable carrier. The carrier may include excipients, diluents, or mixtures thereof.
The compound of the present invention can be administered to a patient orally or parenterally in the form of conventional formulations such as capsules, microcapsules, tablets, granules, powders, lozenges, pills, suppositories, injections, suspensions, syrups, patches, creams, lotions, ointments, gels, sprays, solutions and emulsions. Suitable formulations can be prepared by a method generally employed using conventional organic or inorganic additives, such as excipients (e.g., sucrose, starch, mannitol, sorbitol, lactose, glucose, cellulose, talc, calcium phosphate, or calcium carbonate), binders (e.g., cellulose, methyl cellulose, hydroxymethyl cellulose, polypropylpyrrolidone, polyvinylpyrrolidone, gelatin, acacia, polyethylene glycol, sucrose, or starch), disintegrants (e.g., starch, carboxymethyl cellulose, hydroxypropyl starch, low-substituted hydroxypropyl cellulose, sodium bicarbonate, calcium phosphate, or calcium citrate), lubricants (e.g., magnesium stearate, light anhydrous silicic acid, talc, or sodium lauryl sulfate), flavoring agents (e.g., citric acid, menthol, glycine, or orange powder), preservatives (e.g., sodium benzoate, sodium bisulfite, methyl or propyl paraben), stabilizers (e.g., citric acid, sodium citrate or acetic acid), suspending agents (e.g., methylcellulose, polyvinylpyrrolidone or aluminum stearate), dispersing agents (e.g., hydroxypropylmethylcellulose), diluents (e.g., water), and waxes (e.g., cocoa butter, white petrolatum or polyethylene glycol). For example, an effective amount of the compound in a pharmaceutical composition can be an amount that achieves the desired effect; for example, from about 0.005mg/kg of subject body weight to about 10mg/kg of subject body weight in a unit dose for oral and parenteral administration.
In one embodiment, the invention provides a capsule containing a compound of the invention without an additional carrier.
The pharmaceutical compositions of the present invention may be in the form of tablets, chewable tablets, capsules, solutions, parenteral solutions, lozenges, suppositories, suspensions and the like. The compositions may be formulated to contain the daily dose or a suitable fraction of the daily dose in a dosage unit which may be a single tablet or capsule or a suitable volume of liquid. In one embodiment, the solution is prepared from a water soluble salt, such as a hydrochloride salt. In general, all compositions are prepared according to known methods in pharmaceutical chemistry. Capsules may be prepared by mixing the compound with a suitable carrier or diluent and filling the capsules with an appropriate amount of the mixture. Commonly used carriers and diluents include, but are not limited to, inert powdered materials such as various starches, powdered cellulose, especially crystalline and microcrystalline cellulose, sugars such as fructose, mannitol and sucrose, cereal flour and similar edible powders.
Tablets may be prepared by direct compression, wet granulation or dry granulation. The preparation thereof usually contains a diluent, a binder, a lubricant and a disintegrant as well as the compound. Typical diluents include, for example, various types of starch, lactose, mannitol, kaolin, calcium phosphate or sulfate, inorganic salts (such as sodium chloride) and powdered sugar. Powdered cellulose derivatives are also useful. Typical tablet binders are substances such as starch, gelatin and sugars (e.g. lactose, fructose, glucose, etc.). Natural and synthetic gums are also suitable, including acacia, alginate, methylcellulose, polyvinylpyrrolidone, and the like. Polyethylene glycol, ethyl cellulose, and waxes may also serve as binders.
A lubricant may be required in the tablet formulation to prevent the tablet and punch from sticking in the die. Lubricants may be selected from such slippery solids as talc, magnesium and calcium stearate, stearic acid and hydrogenated vegetable oils. Tablet disintegrants are compounds that swell when wetted to break the tablet and release the compound. They include starches, clays, celluloses, algins and gums. More particularly, it is possible to use, for example, corn and potato starch, methylcellulose, agar, bentonite, wood cellulose, powdered natural sponge, anion exchange resins, alginic acid, guar gum, citrus pulp and carboxymethylcellulose, and sodium lauryl sulfate. The tablets may be coated with sugar as a flavoring and sealing agent, or with a film-forming protecting agent to optimize the dissolution properties of the tablet. The compositions may also be formulated as chewable tablets, for example by incorporating substances such as mannitol into the formulation.
Typical bases may be used when administration as a suppository is desired. Cocoa butter is a traditional suppository base that may be modified by adding waxes to raise its melting point slightly. Water-miscible suppository bases, particularly including polyethylene glycols of various molecular weights, are widely used.
The effect of the compounds can be delayed or prolonged by suitable formulations. For example, slowly dissolving pellets of the compound may be prepared and incorporated into a tablet or capsule or as a slow release implantable device. The technique also involves preparing pellets of several different dissolution rates and filling the capsule with a mixture of pellets. The tablet or capsule may be coated with a film that resists dissolution for a predictable period of time. Even parenteral formulations can be prepared to be long acting by dissolving or suspending the compound in an oily or emulsified vehicle which allows it to be slowly dispersed in serum.
It is a further object of the invention to provide an article of manufacture, for example in the form of a kit. The articles of manufacture of the invention comprise the pharmaceutical compositions of the invention and optionally include packaging and instructions.
Methods of treatment and uses
In another aspect, the invention also provides a compound of the invention, or a pharmaceutically acceptable salt, stereoisomer, solvate, polymorph, tautomer, isotopic compound, metabolite or prodrug thereof, or a pharmaceutical composition of the invention, for use in the preparation of a medicament for treating a TLR7Use in the manufacture of a medicament for a disease responsive to activation of a receptor. In one embodiment, the pair of TLRs7The disease responsive to receptor activation is selected from liver-related diseases, tumor or HIV infection。
In one embodiment, the liver-related disease is selected from viral hepatitis, autoimmune liver disease, drug-toxic liver disease, liver injury from liver disease, hepatic failure, chronic severe hepatitis, cirrhosis, liver abscess, fatty liver, primary liver cancer, preferably the liver-related disease is hepatitis b and hepatitis c. In one embodiment, the tumor is selected from leukemia, lymphoma, melanoma, non-small cell lung cancer.
In one embodiment, the present invention also provides a pharmaceutical combination comprising component (1) a compound of formula (I) or a pharmaceutically acceptable salt, stereoisomer, solvate, polymorph, tautomer, isotopic compound, metabolite or prodrug thereof, or a pharmaceutical composition, and component (2) a PD-1 antibody, a PD-L1 antibody, or a PD-1 inhibitor, a PD-L1 inhibitor, or a PD-1/PD-L1 inhibitor. The invention also provides application of the medicine composition in preparing a medicine for treating tumors. The tumor type is preferably selected from leukemia, lymphoma, melanoma, or non-small cell lung cancer.
The dosage of the compound to be administered to a subject is variable to a considerable extent and can be subject to the judgment of a health care professional.
For convenience reasons, the compounds of the invention may be administered orally. In one embodiment, the compound is administered with water or with a meal when administered orally. In another embodiment, the compound is dispersed in water or fruit juice (e.g., apple juice or orange juice) and administered orally as a suspension.
The compounds may also be administered intradermally, intramuscularly, intraperitoneally, transdermally, intravenously, subcutaneously, intranasally, intradurally, sublingually, intracerebrally, intravaginally, transdermally, rectally, transmucosally, by inhalation or topically to the ear, nose, eye or skin. The mode of administration is at the discretion of the health care professional and may depend in part on the site of the medical condition.
Advantageous effects
The compounds of the invention have higher TLRs7Agonistic activity, and excellent relative to TLRs8Of (2)7The compound has good safety while having activity, for example, the exposure of the compound in a systemic blood system is extremely low, and the compound can induce the expression of IFN- α in blood plasma and liver under low concentration, so that the compound has lower toxicity and can realize better curative effect.
Examples
For clarity, the invention is further illustrated by examples. The examples are given solely for the purpose of illustration and are not intended to limit the scope of the invention, but to enable those skilled in the art to clearly understand and practice the invention. It will be appreciated by those skilled in the art that the order of some of the reaction steps may be adjusted or one or more steps may be omitted, as well as protection/deprotection reaction steps added or omitted, depending on the desired product structure to be obtained, in order to prepare the compounds of the present invention, and this is also within the scope of the present invention.
All solvents used in the present invention are commercially available and can be used without further purification. Unless otherwise stated, the reaction is generally carried out in an anhydrous solvent under an inert gas atmosphere. Proton nmr data were recorded on an AVANCE III HD (300MHz) spectrometer with chemical shifts expressed as (ppm) at tetramethylsilane low field. Mass spectra were measured on an Agilent 1200 series plus 6110 (G1956A). The LC/MS or Shimadzu MS contains a DAD SPD-M20A (LC) and Shimadzu Micromass 2020 detector. The mass spectrometer was equipped with an electrospray ion source (ESI) operating in either positive or negative ion mode.
The invention employs the following abbreviations:
aq is aqueous;
SEMCl (2- (chloromethoxy) ethyl) trimethylsilane;
eq equivalents;
1,3-DPPP 1, 3-bis (diphenylphosphino) propane;
DCM dichloromethane;
PE petroleum ether;
DMA N, N-dimethylacetamide;
DMF N, N-dimethylformamide;
NMP N-methylpyrrolidone;
EtOAc ethyl acetate;
i-PrOH isopropanol;
EtOH ethanol;
MeOH with methanol;
n-BuOH n-butanol;
THF tetrahydrofuran;
boc-t-butoxycarbonyl;
HOAc acetic acid;
NaCNBH3sodium cyanoborohydride;
sodium borohydride acetate STAB;
NaBH(OAc)3sodium borohydride acetate;
CDI N, N' -carbonyldiimidazole;
LAH lithium aluminum hydride;
9-BBN 9-borabicyclononane;
MsCl methanesulfonyl chloride;
RT room temperature;
o.n. overnight;
Boc2o di-tert-butyl dicarbonate;
TFA trifluoroacetic acid;
TFAA trifluoroacetic anhydride;
TEA triethylamine;
DIBAL-H diisobutylaluminum hydride;
NBS bromosuccinimide;
AIBN azobisisobutyronitrile;
DPPF 1,1' -bis (diphenylphosphino) ferrocene;
Ph3p triphenylphosphine;
Pd(OAc)2palladium acetate;
Pd(PPh3P)2Cl2bis (triphenylphosphine) palladium chloride;
Pd2(dba)3tris (benzylidene acetone) dipalladium;
n-BuLi n-butyllithium.
Compounds were named either manually or by Chem Draw software and commercially available compounds were given the supplier catalog name.
HPLC analysis was performed using Shimadzu SIL-20A autosampler and Shimadzu LC20AB system with a Shimadzu SIL-20A autosampler and a Nippon Shimadzu DAD: SPD-M20A detector, using a Shim-pack XR-ODS (2.2 μ M packing, 2.0X50mm) column. 5-100 AB-8 min method, start elution with 95% A (A is 0.05% TFA in water) and end with 100% B (B is MeCN) using a linear gradient, 8 min for the entire process, then 2 min with 100% B. The column was equilibrated to 100:5 for an additional 0.5 min for a total run time of 12.5 min. 10-100 AB-3.4 min method, apply a linear gradient, start elution with 90% A (A is 0.05% TFA in water) and end elution with 100% B (B is acetonitrile), the entire process is 2.2 min, then 0.7 min with 100% B. The column was equilibrated to 90:10 for an additional 0.2 minutes for a total run time of 3.4 minutes. The column temperature was 50 ℃ and the flow rate was 0.8 mL/min. The scanning wavelength of the diode array detector is 200-400 nm.
Thin Layer Chromatography (TLC) was performed on a Sanpont-group silica gel GF254, spots were detected by irradiation with a UV light lamp, and in some cases by other methods, in these cases iodine (about 1g iodine was added to 10g silica gel and mixed thoroughly), vanillin (about 1g vanillin dissolved in 100mL 10% H)2SO4Prepared in (r)), ninhydrin (available from Aldrich) or special color developer (mixed thoroughly (NH)4)6Mo7O24·4H2O、5g(NH4)2Ce(IV)(NO3)6、450mLH2O and 50mL concentrated H2S04Prepared) thin layer plates were spread and the compounds were examined. Still, w.c. was used; kahn, m.; a similar method to that disclosed in the art and Mitra, M.Jouma1of Organic Chemistry,1978,43,2923-Spectra. Common solvents for flash or thin layer chromatography are dichloromethane/methanol, ethyl acetate/methanol and hexane/ethyl acetate mixtures. Performing preparative chromatography on a Gilson _281Prep LC322 system by using a Gilson UV/VIS-156 detector, wherein the adopted chromatographic column is AgellaVenusil ASB Prep C18, 150 x21.2mm; CHIRALPAKAD-3, 0.46cm by 10cm,3 μm; x select C18, 19mm X150 mm; phenomenex Gemini C18, 5 μm, 150x30 mm; or Phenomenex synergy C18, 4 μm, 150x30 mm. Eluting the compound with a low gradient of acetonitrile/water containing 0.05% TFA, 0.25% HCOOH or 0.5% NH at a flow rate of about 25mL/min3·H2O, total run time 8-15 minutes.
Synthetic route
The compound of formula I-1 of the present invention can be prepared by the following synthetic scheme 1.
Figure BDA0001836596920000331
Synthesis scheme 1
Figure BDA0001836596920000332
Starting from 2,4, 6-trichloro-5-nitropyrimidine or an analogue (a commercial reagent), an intermediate 1 is obtained through nucleophilic substitution reaction in the reaction step 1. Subsequently, it is subjected to nucleophilic substitution reaction with ammonia via step 2 to give a 2-chloro-pyrimidin-6-amine compound (intermediate 2). Then, in step 3, chlorine on the 2-position of the pyrimidine reacts with sodium alkoxide, potassium alkoxide, hydrosulphate or organic amine to obtain an intermediate 3. Then, via step 4: 1) reducing nitro group with zinc powder, 2) removing Boc, 3) reacting with amphiphilic reagent and closing ring to obtain product, namely R2A compound of formula I-1 when H.
R2When H, compounds of formula I-1 can also be prepared by synthetic scheme 2, via intermediate 3 of synthetic scheme 1:
synthesis scheme 2
Figure BDA0001836596920000341
Intermediate 3 can be obtained by the same procedure as in scheme 12A compound of formula I-1 when H.
The compounds of the general formulae I-2, I-3 according to the invention can be prepared by the skilled person by selecting suitable starting materials via synthetic schemes 1 or 2.
The compounds of formula I-6 of the present invention can be prepared by synthetic scheme 3.
Figure BDA0001836596920000342
Synthesis scheme 3
Figure BDA0001836596920000343
Starting from the intermediate compound 1', i.e. 2-substituted-4, 6-dichloro-5-nitropyrimidine or an analogue (commercial reagent or prepared according to the method of WO2016/44183 a 1), a nucleophilic substitution reaction with ammonia takes place, via step 1 ", to give the 6-chloro-pyrimidin-4-amine compound (intermediate 2'). Then, step 2 "is reacted with an organic amine to provide intermediate 3'. Then, via step 3 ": 1) reducing nitro with zinc powder, 2) carrying out oxidation reaction on the intermediate obtained in the step 1) and hydrogen peroxide to selectively obtain a 5-position hydroxylamine compound, and 3) reacting the 5-position hydroxylamine compound obtained in the step 2) with an amphiphilic reagent and closing a ring to obtain a product I-6.
The compounds of the general formulae I-4, I-5, I-7 according to the invention can be prepared by the skilled person by selecting suitable starting materials with reference to scheme 3.
Preparation examples
The following specific experimental examples are set forth in order to provide those skilled in the art with a clear understanding and appreciation of the invention. They should not be considered as limiting the scope of the invention, but merely as being illustrative and representative thereof.
Example 1: 5-amino-7-butoxy-2-methyl-1- (3- (pyrrolidin-1-ylmethyl) benzyl) -1, 4-dihydropyrimido [5,4-e ] [1,2,4] triazin-3 (2H) -one
Figure BDA0001836596920000351
Synthetic schemes
Figure BDA0001836596920000361
Step A: preparation of tert-butyl 2- (2-butoxy-6-chloro-5-nitropyrimidin-4-yl) -1-methylhydrazine-1-carboxylate
A250 ml round-bottom three-necked flask was taken, and 2-butoxy-4, 6-dichloro-5-nitropyrimidine (9.97 g, 37.6 mmol) and triethylamine (10.36 g, 102.6 mmol) were added to tetrahydrofuran (50.0 ml). At 0 ℃, a solution of tert-butyl 1-methylhydrazine-1-carboxylate (5.00 g, 34.2 mmol) in tetrahydrofuran (30.0 ml) was slowly added dropwise, and after completion of the addition, the mixture was stirred at room temperature for 2 hours.
The solid residue obtained by rotary evaporation under reduced pressure was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate-5/1) to give 7.5 g of tert-butyl 2- (2-butoxy-6-chloro-5-nitropyrimidin-4-yl) -1-methylhydrazine-1-carboxylate as a yellow oily liquid (yield 53%).
MS(ESI)M/Z:376[M+H+]。
And B: preparation of tert-butyl 2- (2-butoxy-6-chloro-5-nitropyrimidin-4-yl) -2- (3-formylbenzyl) -1-methylhydrazine-1-carboxylate
A500 ml round bottom three-necked flask was taken and added with tert-butyl 2- (2-butoxy-6-chloro-5-nitropyrimidin-4-yl) -1-methylhydrazine-1-carboxylate (7.50 g, 19.94 mmol), acetonitrile (100 ml), potassium carbonate (8.27 g, 59.84 mmol) and 3- (bromomethyl) benzaldehyde (4.33 g, 21.94 mmol) in this order at room temperature. After stirring and mixing uniformly, the mixture is heated for 1 hour at 50 ℃ in an oil bath.
The reaction solution was filtered through celite with suction, the filter cake was rinsed with ethyl acetate (50 ml × 2 times), and the filtrate was collected and concentrated by rotary evaporation under reduced pressure. The solid residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate-10/1) to give 8.00 g of 2- (2-butoxy-6-chloro-5-nitropyrimidin-4-yl) -2- (3-formylbenzyl) -1-methylhydrazine-1-carboxylic acid tert-butyl ester as a yellow oily liquid (yield 81%).
MS(ESI)M/Z:494[M+H+]。
And C: preparation of tert-butyl 2- (6-amino-2-butoxy-5-nitropyrimidin-4-yl) -2- (3-formylbenzyl) -1-methylhydrazine-1-carboxylate
A250 ml autoclave was used to heat 2- (2-butoxy-6-chloro-5-nitropyrimidin-4-yl) -2- (3-formylbenzyl) -1-methylhydrazine-1-carboxylic acid tert-butyl ester (8.00 g, 16.19 mmol) in a methanol solution of ammonia (7.0M,100 ml, 700 mmol) in an oil bath at 60 ℃ overnight.
The solvent was removed by concentration under reduced pressure to give 8.50 g of tert-butyl 2- (6-amino-2-butoxy-5-nitropyrimidin-4-yl) -2- (3-formylbenzyl) -1-methylhydrazine-1-carboxylate as a yellow solid. The crude product was used directly in the next step without purification.
MS(ESI)M/Z:475[M+H+]。
Step D: preparation of tert-butyl 2- (6-amino-2-butoxy-5-nitropyrimidin-4-yl) -1-methyl-2- (3- (pyrrolidin-1-ylmethyl) benzyl) hydrazine-1-carboxylate
A250 ml round bottom three-necked flask was taken, and tert-butyl 2- (6-amino-2-butoxy-5-nitropyrimidin-4-yl) -2- (3-formylbenzyl) -1-methylhydrazine-1-carboxylate (8.50 g, 17.89 mmol), pyrrolidine (15.2 g, 214.1 mmol), dichloromethane (50.0 ml) and
Figure BDA0001836596920000371
molecular sieve grade (4.0 g). After stirring for 30 minutes, sodium borohydride acetate (7.58 g, 35.75 mmol) was added slowly in portions at zero degrees centigrade and after addition was complete the reaction was allowed to proceed overnight at room temperature.
The reaction solution was diluted with water (100 ml). The mixture was filtered through celite and the filter cake was washed with dichloromethane (50 ml × 2 times). The filtrate was collected and separated. The organic phase obtained is washed with saturated sodium chloride solution (100 ml × 1 times), then dried over anhydrous sodium sulfate and finally concentrated under reduced pressure. The concentrate was purified by column chromatography on silica gel (eluent: dichloromethane/methanol ═ 20/1), yielding 7.01 g of tert-butyl 2- (6-amino-2-butoxy-5-nitropyrimidin-4-yl) -1-methyl-2- (3- (pyrrolidin-1-ylmethyl) benzyl) hydrazine-1-carboxylate as a yellow liquid (yield 73.7%).
MS(ESI)M/Z:530[M+H+]。
Step E: 2- (5, 6-diamino-2-butoxypyrimidin-4-yl) -1-methyl-2- (3- (pyrrolidin-1-ylmethyl) benzyl) hydrazine-1-carboxylic acid tert-butyl ester acetate
To a 100ml three-necked flask, zinc powder (7.00 g, 107.6 mmol) was added to a solution of tert-butyl 2- (6-amino-2-butoxy-5-nitropyrimidin-4-yl) -1-methyl-2- (3- (pyrrolidin-1-ylmethyl) benzyl) hydrazine-1-carboxylate (7.00 g, 13.2 mmol) in acetic acid (70.0 ml). Stir at room temperature overnight.
And (4) carrying out suction filtration on the reaction solution, and collecting filtrate. The filter cake was washed with ethyl acetate (20 ml × 2 times) and the organic phases were combined. The organic phase is concentrated under reduced pressure to give 7.95 g of brown liquid tert-butyl 2- (5, 6-diamino-2-butoxypyrimidin-4-yl) -1-methyl-2- (3- (pyrrolidin-1-ylmethyl) benzyl) hydrazine-1-carboxylate acetate. The crude product was used directly in the next reaction without purification.
MS(ESI)M/Z:500[M+H+]。
Step F: preparation of 2-butoxy-6- (2-methyl-1- (3- (pyrrolidin-1-ylmethyl) benzyl) hydrazino) pyrimidine-4, 5-diamine trifluoroacetate salt
A100 ml three-necked flask was taken, and trifluoroacetic acid (40.0 ml) was slowly added to a dichloromethane solution (40.0 ml) of tert-butyl 2- (5, 6-diamino-2-butoxypyrimidin-4-yl) -1-methyl-2- (3- (pyrrolidin-1-ylmethyl) benzyl) hydrazine-1-carboxylate (7.90 g, 15.8 mmol)) and stirred at room temperature for 3 hours.
Concentration under reduced pressure gave 11.6 g of brown liquid 2-butoxy-6- (2-methyl-1- (3- (pyrrolidin-1-ylmethyl) benzyl) hydrazino) pyrimidine-4, 5-diamine trifluoroacetate. The crude product was used directly in the next reaction without purification.
MS(ESI)M/Z:400[M+H+]。
Step G: preparation of 5-amino-7-butoxy-2-methyl-1- (3- (pyrrolidin-1-ylmethyl) benzyl) -1, 4-dihydropyrimido [5,4-e ] [1,2,4] triazin-3 (2H) -one trifluoroacetate salt
To a mixed solution of 2-butoxy-6- (2-methyl-1- (3- (pyrrolidin-1-ylmethyl) benzyl) hydrazino) pyrimidine-4, 5-diamine (2.00 g, 5.0 mmol) in tetrahydrofuran (10.0 ml) and dichloromethane (10.0 ml) at 0 degrees celsius was added N, N' -carbonyldiimidazole (1.62 g, 10.0 mmol) and triethylamine (1.52 g, 15.0 mmol) in that order. The addition was complete and the reaction solution was allowed to warm to 0 ℃ and stirring was continued for 1 hour.
The reaction solution was concentrated under reduced pressure to give a brown solid (2.10 g) which was dissolved in dimethyl sulfoxide (5.0 ml) to clarify and purified by preparative high performance liquid chromatography. The purification conditions were as follows, column: x select C1819 mm × 150 mm; mobile phase: water (containing 0.05% ammonia) and acetonitrile; flow rate 25 ml/min; gradient: acetonitrile rose from 10% to 80% in 7 minutes; detection wavelength: 254 nm. The product was collected and lyophilized under reduced pressure. 306 mg of 5-amino-7-butoxy-2-methyl-1- (3- (pyrrolidin-1-ylmethyl) benzyl) -1, 4-dihydropyrimido [5,4-e ] [1,2,4] triazin-3 (2H) -one trifluoroacetate are obtained as a pale yellow solid (Compound 1, yield 64%).
MS(ESI)M/Z:426[M+H+]。
1H NMR(300MHz,DMSO-d6)δ8.07(s,1H),7.47–7.37(m,2H),7.19–7.16(m,2H),6.58(br,s,2H),4.38–4.29(m,4H),4.01(t,J=6.6Hz,2H),3.27–3.23(m,2H),3.06(s,3H),2.98–2.86(m,2H),2.07–1.84(m,4H),1.66–1.59(m,2H),1.43–1.38(m,2H),0.93(t,J=7.2Hz,3H)。
Example 2: 5-amino-7-butoxy-1- (4-fluoro-3- (pyrrolidin-1-ylmethyl) benzyl) -2-methyl-1, 4-dihydropyrimido [5,4-e ] [1,2,4] triazin-3 (2H) -one
Figure BDA0001836596920000381
Synthetic schemes
Figure BDA0001836596920000391
Step A: preparation of 5- (bromomethyl) -2-fluorobenzoic acid
Bromosuccinimide (31.9 g, 179 mmol) and azobisisobutyronitrile (2.60 g, 16.2 mmol) were added sequentially to a solution of 2-fluoro-5-methylbenzoic acid (25.00 g, 162 mmol) in carbon tetrachloride (1.50 l) under a nitrogen atmosphere. After stirring and dissolving, the reaction solution was put in an oil bath and heated at 90 ℃ for 2 hours.
After the reaction system was cooled to room temperature, 500 ml of a saturated sodium carbonate solution was added to the reaction solution to dilute it. Standing the obtained mixed solution for layering. The organic phase is separated off and the aqueous phase is extracted with dichloromethane (500 ml. times.3 times). The combined organic phases are washed with saturated sodium chloride solution (500 ml × 2 times). Then dried over anhydrous sodium sulfate and finally concentrated under reduced pressure. The resulting solid residue was purified by column on silica gel (eluent: ethyl acetate/petroleum ether: 1/3) to give 25.6 g of 5- (bromomethyl) -2-fluorobenzoic acid as a white solid (yield 67.7%).
The reaction showed no MS signal and was used directly in the next reaction.
Step B-C: preparation of tert-butyl 2- (2-butoxy-6-chloro-5-nitropyrimidin-4-yl) -2- (4-fluoro-3- (hydroxymethyl) benzyl) -1-methylhydrazine-1-carboxylate
5- (bromomethyl) -2-fluorobenzoic acid (25.60 g, 110 mmol) was dissolved in borane in tetrahydrofuran (2.0 mol/L, 500 ml, 1.0 mol) and stirred at room temperature for 16 hours. Subsequently, the resulting solution was concentrated in vacuo. The resulting solid residue was purified by column on silica gel (eluent: ethyl acetate/petroleum ether-1/3) to give 12.50 g of (5- (bromomethyl) -2-fluorophenyl) methanol as a brown liquid (yield 51.9%). The reaction was not stable enough to give no MS signal. Directly used for the next reaction.
Tert-butyl 2- (2-butoxy-6-chloro-5-nitropyrimidin-4-yl) -1-methylhydrazine-1-carboxylate (2.06 g, 5.5 mmol), (5- (bromomethyl) -2-fluorophenyl) methanol (1.75 g, 8.0 mmol) and anhydrous potassium carbonate (2.20 g, 15.9 mmol) were dissolved in succession in acetonitrile (50 ml). After stirring and mixing uniformly, the reaction solution was heated overnight at 50 ℃ in an oil bath.
And (3) post-treatment: after the reaction solution was cooled to room temperature, 100ml of pure water was added to dilute the reaction solution. The resulting mixture was concentrated under reduced pressure to remove acetonitrile. The remaining aqueous phase was extracted with ethyl acetate (50 ml × 3 times) and the organic phases were combined. The organic phase is washed with saturated sodium chloride solution (50 ml × 2 times), then dried over anhydrous sodium sulfate and finally concentrated under reduced pressure. The residue was purified by column on silica gel (eluent: ethyl acetate/petroleum ether ═ 1/1) to give 1.90 g of tert-butyl 2- (2-butoxy-6-chloro-5-nitropyrimidin-4-yl) -2- (4-fluoro-3- (hydroxymethyl) benzyl) -1-methylhydrazine-1-carboxylate as a yellow solid (yield, 68.8%).
MS(ESI)M/Z:514[M+H+]。
Step D: 2- (2-butoxy-6-chloro-5-nitropyrimidin-4-yl) -2- (4-fluoro-3-formylbenzyl) -1-methylhydrazine-1-carboxylic acid tert-butyl ester
To a solution of tert-butyl 2- (2-butoxy-6-chloro-5-nitropyrimidin-4-yl) -2- (4-fluoro-3- (hydroxymethyl) benzyl) hydrazine-1-carboxylate (1.90 g, 3.70 mmol) in dichloromethane (50 ml) was added manganese dioxide (1.92 g, 22.07 mmol). Stir at 40 degrees celsius overnight.
The reaction mixture was filtered through celite under reduced pressure and the filter cake was rinsed with dichloromethane (30 ml × 2 times). The filtrate was collected and concentrated under reduced pressure. The residue was purified by column on silica gel (eluent: ethyl acetate/petroleum ether ═ 1/1) to give 1.50 g of tert-butyl 2- (2-butoxy-6-chloro-5-nitropyrimidin-4-yl) -2- (4-fluoro-3-formylbenzyl) -1-methylhydrazine-1-carboxylate as a yellow solid (yield 79.4%).
MS(ESI)M/Z:514[M+H+]。
Step E: 2- (6-amino-2-butoxy-5-nitropyrimidin-4-yl) -2- (4-fluoro-3-formylbenzyl) -1-methylhydrazine-1-carboxylic acid tert-butyl ester
Tert-butyl 2- (2-butoxy-6-chloro-5-nitropyrimidin-4-yl) -2- (4-fluoro-3-formylbenzyl) -1-methylhydrazine-1-carboxylate (1.50 g, 3.01 mmol) was dissolved in a solution of ammonia in isopropanol (2.0 mol/l, 50.0 ml, 100.0 ml) in a 100ml autoclave. After stirring and mixing uniformly, the reaction solution is heated for 16 hours at 60 ℃.
And (3) after the reaction system is cooled to room temperature, concentrating the reaction solution under reduced pressure. Pure water (20 ml) was added to the residue. Suction filtration was carried out under reduced pressure, and the filter cake was washed with pure water (20 ml. times.2 times). The filter cake was collected and dried to yield 1.40 g of tert-butyl 2- (6-amino-2-butoxy-5-nitropyrimidin-4-yl) -2- (4-fluoro-3-formylbenzyl) -1-methylhydrazine-1-carboxylate as a yellow solid (yield, 97.0%).
MS(ESI)M/Z:493[M+H+]。
Step F: preparation of tert-butyl 2- (6-amino-2-butoxy-5-nitropyrimidin-4-yl) -2- (4-fluoro-3- (pyrrolidin-1-ylmethyl) benzyl) -1-methylhydrazine-1-carboxylate
Tert-butyl 2- (6-amino-2-butoxy-5-nitropyrimidin-4-yl) -2- (4-fluoro-3-formylbenzyl) -1-methylhydrazine-1-carboxylate (1.40 g, 2.84 mmol), pyrrolidine (2.42 g, 34.08 mmol) were dissolved in succession in dichloromethane (50 ml) under nitrogen. Sodium triacetoxyborohydride (1.20 g, 5.66 mmol) was added slowly in portions in an ice-water bath. The addition was complete and the reaction apparatus was moved to room temperature and stirred overnight.
Pure water (50 ml) was added to dilute the reaction solution. The mixture was extracted with dichloromethane (50 ml × 2 times) and the organic phases were combined. The organic phase was first backwashed with saturated brine (50 ml. times.2 times), then dried over anhydrous sodium sulfate, and finally concentrated under reduced pressure. The residue was purified by column on silica gel (eluent: ethyl acetate/petroleum ether ═ 1/1) to give 510 mg of solid tert-butyl 2- (6-amino-2-butoxy-5-nitropyrimidin-4-yl) -2- (4-fluoro-3- (pyrrolidin-1-ylmethyl) benzyl) -1-methylhydrazine-1-carboxylate (yield, 32.7%).
MS(ESI)M/Z:548[M+H+]。
Step G: preparation of tert-butyl 2- (5, 6-diamino-2-butoxypyrimidin-4-yl) -2- (4-fluoro-3- (pyrrolidin-1-ylmethyl) benzyl) -1-methylhydrazine-1-carboxylate
Tert-butyl 2- (6-amino-2-butoxy-5-nitropyrimidin-4-yl) -2- (4-fluoro-3- (pyrrolidin-1-ylmethyl) benzyl) -1-methylhydrazine-1-carboxylate (510 mg, 0.96 mmol) was dissolved in acetic acid (5 ml), followed by addition of zinc powder (510 mg) and stirring at room temperature overnight.
And (5) carrying out suction filtration under reduced pressure, and collecting filtrate. The filter cake was rinsed with dichloromethane (10 ml × 2). The filtrates were combined and concentrated under reduced pressure to give 450 mg of tert-butyl 2- (5, 6-diamino-2-butoxypyrimidin-4-yl) -2- (4-fluoro-3- (pyrrolidin-1-ylmethyl) benzyl) -1-methylhydrazine-1-carboxylate as a yellow liquid (yield, 93.6%)
MS(ESI)M/Z:518[M+H+]。
Step H: preparation of 2-butoxy-6- (1- (4-fluoro-3- (pyrrolidin-1-ylmethyl) benzyl) -2-methylhydrazino) pyrimidine-4, 5-diamine trifluoroacetate salt
Tert-butyl 2- (5, 6-diamino-2-butoxypyrimidin-4-yl) -2- (4-fluoro-3- (pyrrolidin-1-ylmethyl) benzyl) -1-methylhydrazine-1-carboxylate (450 mg, 0.89 mmol) was dissolved in dichloromethane (10 ml). Trifluoroacetic acid (3.0 ml) was slowly added dropwise under an ice-water bath. After the completion of the dropwise addition, the reaction apparatus was moved to room temperature and stirred for 1 hour.
The reaction solution was concentrated under reduced pressure to give 300 mg of brown liquid, 2-butoxy-6- (1- (4-fluoro-3- (pyrrolidin-1-ylmethyl) benzyl) -2-methylhydrazino) pyrimidine-4, 5-diamine trifluoroacetate. The product is used for the next reaction without purification.
MS(ESI)M/Z:418[M+H+]。
Step I: preparation of 5-amino-7-butoxy-1- (4-fluoro-3- (pyrrolidin-1-ylmethyl) benzyl) -2-methyl-1, 4-dihydropyrimido [5,4-e ] [1,2,4] triazin-3 (2H) -one trifluoroacetate salt
2-butoxy-6- (1- (4-fluoro-3- (pyrrolidin-1-ylmethyl) benzyl) -2-methylhydrazino) pyrimidine-4, 5-diamine (300 mg, 0.718 mmol) was dissolved in a mixed solution of tetrahydrofuran (5.0 ml) and dichloromethane (5.0 ml). Triethylamine (350 mg, 2.16 mmol) and N, N' -carbonyldiimidazole (350 mg, 2.16 mmol) were added successively under an ice-water bath. Subsequently, the reaction solution was transferred to room temperature and stirred for 1 hour.
The reaction mixture was concentrated under reduced pressure. The residue was dissolved in N, N-dimethylformamide (4.0 ml) to clarify. The crude product was purified by preparative high performance liquid chromatography. The separation conditions were as follows, column: x select C1819 mm × 150 mm; mobile phase: water (containing 0.05% TFA) and acetonitrile; flow rate: 25 ml/min; gradient: acetonitrile rose from 10% to 80% in 7 minutes; detection wavelength: 254 nm. Lyophilization under reduced pressure afforded 88.0 mg of a brown semisolid 5-amino-7-butoxy-1- (4-fluoro-3- (pyrrolidin-1-ylmethyl) benzyl) -2-methyl-1, 4-dihydropyrimido [5,4-e ] [1,2,4] triazin-3 (2H) -one trifluoroacetate (Compound 2, yield 27.7%).
MS(ESI)M/Z:444[M+H+]。
1H NMR(300MHz,CD3OD,ppm)δ7.53–7.44(m,1H),7.33–7.18(m,2H),4.50(s,2H),4.44(s,2H),4.42–4.29(m,2H),3.68–3.42(m,2H),3.18(s,5H),2.30–2.04(m,4H),1.81–1.66(m,2H),1.56–1.43(m,2H),1.02–0.90(m,3H)。
19F NMR(300MHz,CD3OD,ppm),δ-76.82,-117.53。
Example 3: 6-amino-2-butoxy-7-hydroxy-9- (4- (pyrrolidin-1-ylmethyl) benzyl) -7H-purin-8 (9H) -one
Figure BDA0001836596920000421
Synthetic schemes
Figure BDA0001836596920000422
Step A: 2-butoxy-5-nitro-N4Preparation of (4- (pyrrolidin-1-ylmethyl) benzyl) pyrimidine-4, 6-diamine
To a 500 ml three-necked round-bottomed flask, under ice-water bath, were added 2-butoxy-6-chloro-5-nitropyrimidin-4-amine (19.18 g, 77.96 mmol), tetrahydrofuran (200 ml) and triethylamine (15.75 g, 155.92 mmol) in this order. After stirring for 5 minutes, (4- (pyrrolidin-1-ylmethyl) phenyl) methylamine (14.81 g, 77.96 mmol) was slowly added dropwise. After the dropwise addition, the reaction apparatus was moved to room temperature and stirring was continued for 1 hour. A large amount of white solid precipitated. TLC monitoring found disappearance of starting material.
And (4) carrying out suction filtration on the reaction solution, and collecting filtrate. The filter residue was washed with ethyl acetate (50 ml. times.3 times). The filtrates were combined and concentrated under reduced pressure to give 30.82 g of 2-butoxy-5-nitro-N4- (4- (pyrrolidin-1-ylmethyl) benzyl) pyrimidine-4, 6-diamine. The crude product was used directly in the next reaction without purification.
MS(ESI)M/Z:401[M+H+]。
And B: 2-butoxy-N4- (4- (pyrrolidin-1-ylmethyl) benzyl) pyrimidin-4Preparation of 5, 6-triamines
To a 500 ml three-necked round-bottomed flask was added 2-butoxy-5-nitro-N in sequence4- (4- (pyrrolidin-1-ylmethyl) benzyl) pyrimidine-4, 6-diamine (30.82 g, crude), ethanol (240 ml) and water 60 (ml). After stirring to dissolve, iron powder (4.31 g, 77.05 mmol) and ammonium chloride (4.08 g, 77.05 mmol) were added to the mixture. Subsequently, the reaction apparatus was heated in an oil bath at 60 ℃. After 12 hours, LC-MS monitors the disappearance of the starting material.
The reaction solution was filtered through celite and the filtrate was collected. The filter residue was washed with ethyl acetate (50 ml. times.3 times). The filtrates were combined and concentrated under reduced pressure to give 29.81 g of 2-butoxy-N4- (4- (pyrrolidin-1-ylmethyl) benzyl) pyrimidine-4, 5, 6-triamine. The crude product was used directly in the next reaction without purification.
MS(ESI)M/Z:371[M+H+]。
And C: 2-butoxy-5- (hydroxyamino) -N4Preparation of (4- (pyrrolidin-1-ylmethyl) benzyl) pyrimidine-4, 6-diamine
To a 100ml three-necked round-bottomed flask, 2-butoxy-N was sequentially added4- (4- (pyrrolidin-1-ylmethyl) benzyl) pyrimidine-4, 5, 6-triamine (3.00 g, crude) and N-methylpyrrolidone (30.0 ml). Then, 30% hydrogen peroxide (3.00 ml) and an aqueous sodium hydroxide solution (6.0N, 0.30 ml, 1.8 mmol) were slowly added dropwise in this order in an ice-water bath. After the dropwise addition, the temperature of the reaction solution was controlled at 0 ℃. After 5 hours, LC-MS detects the formation of by-products. And (4) carrying out suction filtration on the reaction mixed solution, and purifying the filtrate by using preparative high performance liquid chromatography. The separation conditions were as follows, column: x select C1819 mm × 150 mm; mobile phase: water and acetonitrile; flow rate: 25 ml/min; gradient: acetonitrile rose from 10% to 70% in 7 minutes; detection wavelength: 254 nm. The product was collected and lyophilized under reduced pressure to give 0.42 g of 2-butoxy-5- (hydroxyamino) -N4- (4- (pyrrolidin-1-ylmethyl) benzyl) pyrimidine-4, 6-diamine.
MS(ESI)M/Z:387[M+H+]。
Step D: preparation of 6-amino-2-butoxy-7-hydroxy-9- (4- (pyrrolidin-1-ylmethyl) benzyl) -7H-purin-8 (9H) -one trifluoroacetate and 2-butoxy-7-hydroxy-6- ((4- (pyrrolidin-1-ylmethyl) benzyl) amino) -7H-purin-8 (9H) -one trifluoroacetate
Taking a 50ml round-bottom three-neck flask, adding 2-butoxy-5- (hydroxyamino) -N under ice-water bath4To a solution of (4- (pyrrolidin-1-ylmethyl) benzyl) pyrimidine-4, 6-diamine (300 mg, 0.78 mmol) in dichloromethane (5.00 ml) were added N, N' -carbonyldiimidazole (1.26 g, 7.8 mmol) and triethylamine (1.58 g, 15.6 mmol) in that order. After the addition was complete, the reaction apparatus was moved to room temperature and stirred for 45 minutes.
After the reaction solution was concentrated under reduced pressure, a brown solid was obtained and dissolved in dimethyl sulfoxide (5.0 ml) to be clear. Then, it was purified by preparative high performance liquid chromatography. The purification conditions were as follows, column: x select C1819 mm × 150 mm; mobile phase: water (containing 0.05% TFA) and acetonitrile; flow rate 25 ml/min; gradient: acetonitrile rose from 10% to 70% in 7 minutes; detection wavelength: 254 nm. The product was collected and lyophilized at low temperature under reduced pressure to give 70.8 mg of yellow solid 6-amino-2-butoxy-7-hydroxy-9- (4- (pyrrolidin-1-ylmethyl) benzyl) -7H-purin-8 (9H) -one trifluoroacetate (Compound 3, yield 21.9%) and 2.5 mg of off-white solid 2-butoxy-7-hydroxy-6- ((4- (pyrrolidin-1-ylmethyl) benzyl) amino) -7H-purin-8 (9H) -one trifluoroacetate (yield 0.8%).
6-amino-2-butoxy-7-hydroxy-9- (4- (pyrrolidin-1-ylmethyl) benzyl) -7H-purin-8 (9H) -one trifluoroacetate (Compound 3)
MS(ESI)M/Z:413[M+H+]。
19F NMR(300MHz,DMSO-d6)δ-73.72。
1H NMR(300MHz,DMSO-d6)δ7.63–7.48(m,2H),7.47–7.37(m,2H),6.53(br,s,2H),4.92(s,2H),4.82(s,2H),4.20–4.10(m,2H)3.85-3.67(m,2H),3.67-3.50(m,2H),2.11–2.01(m,4H),1.75–1.54(m,2H),1.47–1.30(m,2H),0.91(t,J=7.5Hz,3H)。
2-butoxy-7-hydroxy-6- ((4- (pyrrolidin-1-ylmethyl) benzyl) amino) -7H-purin-8 (9H) -one trifluoroacetate
MS(ESI)M/Z:413[M+H+]。
19F NMR(300MHz,DMSO-d6):δ-74.18。
1H NMR(300MHz,DMSO-d6)δ11.18(s,1H),9.74(s,1H),7.54–7.38(m,4H),7.18–7.08(m,1H),4.72–4.58(m,2H),4.34(s,2H),4.09(t,J=6.6Hz,2H),3.70–3.40(m,2H),3.17–2.96(m,2H),2.15–1.74(m,4H),1.68–1.50(m,2H),1.45–1.25(m,2H),0.88(t,J=7.2Hz,3H)。
Example 4: 6-amino-2-butoxy-7-hydroxy-9- (3- (pyrrolidin-1-ylmethyl) benzyl) -7H-purine-8 (9H) -thione
Figure BDA0001836596920000441
Synthetic schemes
Figure BDA0001836596920000442
Step A: preparation of 6-amino-2-butoxy-7-hydroxy-9- (3- (pyrrolidin-1-ylmethyl) benzyl) -7H-purine-8 (9H) -thione
Taking a 50ml round-bottom three-neck flask, adding 2-butoxy-5- (hydroxyamino) -N under ice-water bath4To a solution of (3- (pyrrolidin-1-ylmethyl) benzyl) pyrimidine-4, 6-diamine (200 mg, 0.52 mmol) in dichloromethane (6.00 ml) were added N, N' -thiocarbonyldiimidazole (0.92 g, 5.2 mmol) and triethylamine (1.05 g, 10.4 mmol) in that order. After the addition was complete, the reaction apparatus was moved to room temperature and stirred for 1 hour.
The reaction mixture was concentrated under reduced pressure. The resulting brown solid was dissolved in dimethyl sulfoxide (3.0 ml) until clear. Then, the crude product is purified by preparative high performance liquid chromatography. The purification conditions were as follows, column: x select C1819 mm × 150 mm; mobile phase: water (containing 0.05% TFA) and acetonitrile; flow rate 25 ml/min; gradient: acetonitrile rose from 15% to 65% in 6 minutes; detection wavelength: 254 nm. The product was collected and lyophilized at low temperature under reduced pressure to give 2.2 mg of white solid 6-amino-2-butoxy-7-hydroxy-9- (3- (pyrrolidin-1-ylmethyl) benzyl) -7H-purine-8 (9H) -thione trifluoroacetate (Compound 4, yield 1.0%).
MS(ESI)M/Z:429[M+H+]。
1H NMR(300MHz,CD3OD)δ7.71–7.50(m,4H),5.46(s,2H),4.82(s,2H),4.31(t,J=6.6Hz,2H),3.83–3.69(m,4H),2.40–2.18(m,4H),1.79–1.70(m,2H),1.55–1.48(m,2H),0.99(t,J=7.4Hz,3H)。
19F NMR(300MHz,CD3OD):δ-76.89。
Example 5: 6-amino-2-butoxy-7-hydroxy-9- (3- (pyrrolidin-1-ylmethyl) benzyl) -7H-purin-8 (9H) -one
Figure BDA0001836596920000451
Synthetic schemes
Figure BDA0001836596920000452
Step A: 2-butoxy-5-nitro-N4Preparation of (3- (pyrrolidin-1-ylmethyl) benzyl) pyrimidine-4, 6-diamine
To a 500 ml three-necked round-bottomed flask, under ice-water bath, were added 2-butoxy-6-chloro-5-nitropyrimidin-4-amine (19.16 g, 77.96 mmol), tetrahydrofuran (200 ml) and triethylamine (15.68 g, 155.92 mmol) in this order. After stirring for 7 minutes, (3- (pyrrolidin-1-ylmethyl) phenyl) methylamine (14.78 g, 77.96 mmol) was slowly added dropwise. After the dropwise addition, the reaction apparatus was moved to room temperature and stirring was continued for 1.5 hours.
And (4) carrying out suction filtration on the reaction solution, and collecting filtrate. The filter residue was washed with ethyl acetate (50 ml. times.3 times). The filtrates were combined and concentrated under reduced pressure to give 30.84 g of 2-butoxy-5-nitro-N4- (3- (pyrrolidin-1-ylmethyl) benzyl) pyrimidine-4, 6-diamine. The crude product was used directly in the next reaction without purification.
MS(ESI)M/Z:401[M+H+]。
And B: 2-butoxy-N4Preparation of (3- (pyrrolidin-1-ylmethyl) benzyl) pyrimidine-4, 5, 6-triamine
To a 500 ml three-necked round-bottomed flask was added 2-butoxy-5-nitro-N in sequence4- (3- (pyrrolidin-1-ylmethyl) benzyl) pyrimidine-4, 6-diamine (30.78 g, crude), ethanol (240 ml) and water 60 (ml). After stirring to dissolve, iron powder (4.28 g, 77.05 mmol) and ammonium chloride (4.06 g, 77.05 mmol) were added to the mixture. Subsequently, the reaction apparatus was heated in an oil bath at 60 ℃. After 12 hours, LC-MS monitors the disappearance of the starting material.
The reaction solution was filtered through celite and the filtrate was collected. The filter residue was washed with ethyl acetate (50 ml. times.3 times). The filtrates were combined and concentrated under reduced pressure to give 29.78 g of 2-butoxy-N4-(3- (pyrrolidin-1-ylmethyl) benzyl) pyrimidine-4, 5, 6-triamine. The crude product was used directly in the next reaction without purification.
MS(ESI)M/Z:371[M+H+]。
And C: 2-butoxy-5- (hydroxyamino) -N4Preparation of (3- (pyrrolidin-1-ylmethyl) benzyl) pyrimidine-4, 6-diamine
To a 100ml three-necked round-bottomed flask, 2-butoxy-N was sequentially added4- (3- (pyrrolidin-1-ylmethyl) benzyl) pyrimidine-4, 5, 6-triamine (3.08 g, crude) and N-methylpyrrolidone (30.0 ml). Then, 30% hydrogen peroxide (3.00 ml) and an aqueous sodium hydroxide solution (6.0N, 0.30 ml, 1.8 mmol) were slowly added dropwise in this order in an ice-water bath. After the dropwise addition, the temperature of the reaction solution is controlled at 0 ℃, and after 5 hours, LC-MS detects that byproducts are generated. And (4) carrying out suction filtration on the reaction mixed solution, and purifying the filtrate by using preparative high performance liquid chromatography. The separation conditions were as follows, column: x select C1819 mm × 150 mm; mobile phase: water (0.05% ammonia) and acetonitrile; flow rate: 25 ml/min; gradient: acetonitrile rose from 10% to 70% in 7 minutes; detection wavelength: 254 nm. Freeze-drying at low temperature to give 0.43 g of 2-butoxy-5- (hydroxyamino) -N4- (3- (pyrrolidin-1-ylmethyl) benzyl) pyrimidine-4, 6-diamine.
MS(ESI)M/Z:387[M+H+]。
Step D: 6-amino-2-butoxy-7-hydroxy-9- (3- (pyrrolidin-1-ylmethyl) benzyl) -7H-purin-8 (9H) -one
Taking a 50ml round-bottom three-neck flask, adding 2-butoxy-5- (hydroxyamino) -N under ice-water bath4- (3- (pyrrolidine)-1-ylmethyl) benzyl) pyrimidine-4, 6-diamine (300 mg, 0.78 mmol) in dichloromethane (5.00 ml) was added N, N' -carbonyldiimidazole (1.26 g, 7.8 mmol) and triethylamine (1.58 g, 15.6 mmol) in that order. After the addition was complete, the reaction apparatus was moved to room temperature and stirred for 1 hour.
After the reaction solution was concentrated under reduced pressure, a brown solid was obtained and dissolved in dimethyl sulfoxide (5.0 ml) to be clear. Then, it was purified by preparative high performance liquid chromatography. The purification conditions were as follows, column: x select C1819 mm × 150 mm; mobile phase: water (containing 0.05% TFA) and acetonitrile; flow rate 25 ml/min; gradient: acetonitrile rose from 10% to 70% in 7 minutes; detection wavelength: 254 nm. The product was collected and lyophilized at low temperature under reduced pressure to give 27.8 mg of yellow semi-solid 6-amino-2-butoxy-7-hydroxy-9- (3- (pyrrolidin-1-ylmethyl) benzyl) -7H-purin-8 (9H) -one trifluoroacetate (Compound 5, yield 8.7%).
MS(ESI)M/Z:413[M+H+]。
1H NMR(300MHz,DMSO-d6)δ11.87(s,1H),7.47–7.43(m,4H),6.55(br,s,2H),4.92(s,2H),4.82(s,2H),4.13(t,J=7.6Hz,2H),3.81–3.72(m,2H),3.60–3.52(m,2H),2.17–2.05(m,4H),1.65–1.57(m,2H),1.40–1.33(m,2H),0.90(t,J=7.4Hz,3H)。
19F NMR(300MHz,DMSO-d6)δ-74.50。
Example 6: 6-amino-2-butoxy-7-hydroxy-9- (3- (piperidin-1-ylmethyl) benzyl) -7H-purin-8 (9H) -one
Figure BDA0001836596920000471
Synthetic schemes
Figure BDA0001836596920000472
Step A: 2-butoxy-5-nitro-N4Preparation of (3- (piperidin-1-ylmethyl) benzyl) pyrimidine-4, 6-diamine
To a 500 ml three-necked round-bottomed flask, under ice-water bath, were added 2-butoxy-6-chloro-5-nitropyrimidin-4-amine (19.18 g, 77.96 mmol), tetrahydrofuran (200 ml) and triethylamine (15.75 g, 155.92 mmol) in this order. After stirring for 5 minutes ((3- (piperidin-1-ylmethyl) phenyl) methylamine (15.98 g, 77.96 mmol) was slowly added dropwise, the reaction apparatus was moved to room temperature and stirred for 1 hour, a large amount of white solid precipitated, and the starting material was found to disappear by TLC monitoring.
And (4) carrying out suction filtration on the reaction solution, and collecting filtrate. The filter residue was washed with ethyl acetate (50 ml. times.3 times). The filtrates were combined and concentrated under reduced pressure to give 29.02 g of 2-butoxy-5-nitro-N4- (3- (piperidin-1-ylmethyl) benzyl) pyrimidine-4, 6-diamine. The crude product was used directly in the next reaction without purification.
MS(ESI)M/Z:415[M+H+]。
And B: 2-butoxy-N4Preparation of (3- (piperidin-1-ylmethyl) benzyl) pyrimidine-4, 5, 6-triamine
To a 500 ml three-necked round-bottomed flask was added 2-butoxy-5-nitro-N in sequence4- (3- (piperidin-1-ylmethyl) benzyl) pyrimidine-4, 6-diamine (29.02 g crude), ethanol (240 ml) and water (60.0 ml). After stirring to dissolve, iron powder (4.31 g, 77.05 mmol) and ammonium chloride (4.08 g, 77.05 mmol) were added to the reaction mixture. After the feeding, the mixture is heated in an oil bath at 60 ℃. After 12 hours, the disappearance of the starting material was observed by LC-MS monitoring.
The reaction solution was filtered with suction, the filtrate was collected, and the residue was washed with ethyl acetate (50 ml. times.3 times). The filtrates were combined and concentrated under reduced pressure to give 24.1 g of 2-butoxy-N4- (3- (piperidin-1-ylmethyl) benzyl) pyrimidine-4, 5, 6-triamine. The crude product was used directly in the next reaction without purification.
MS(ESI)M/Z:385[M+H+]。
And C: 2-butoxy-5- (hydroxyamino) -N4Preparation of (3- (piperidin-1-ylmethyl) benzyl) pyrimidine-4, 6-diamine
To a 100ml three-necked round bottom flask, 2-butoxy-N4- (3- (piperidin-1-ylmethyl) benzyl) pyrimidine-4, 5, 6-triamine (3.09 g, 8.05 mmol) and N-methylpyrrolidinone (30.0 ml) were added sequentially. Then, 30% hydrogen peroxide is slowly dropped in sequence under ice water bath(3.00 ml) and aqueous sodium hydroxide (6.0N, 0.30 ml). After the dropwise addition, the temperature of the reaction solution was controlled at 0 ℃. After 5 hours, LC-MS detects the formation of by-products. The reaction mixture was filtered off with suction. The filtrate was purified by preparative high performance liquid chromatography. The separation conditions were as follows, column: x select C1819 mm × 150 mm; mobile phase: water (0.05% ammonia) and acetonitrile; flow rate: 25 ml/min; gradient: acetonitrile rose from 10% to 70% in 7 minutes; detection wavelength: 254 nm. The product was collected and lyophilized under reduced pressure to give 0.51 g of 2-butoxy-5- (hydroxyamino) -N4- (3- (piperidin-1-ylmethyl) benzyl) pyrimidine-4, 6-diamine.
MS(ESI)M/Z:401[M+H+]。
Step D: preparation of 6-amino-2-butoxy-7-hydroxy-9- (3- (piperidin-1-ylmethyl) benzyl) -7H-purin-8 (9H) -one trifluoroacetate
To the solution of 2-butoxy-5- (hydroxyamino) -N in an ice-water bath4To a solution of (3- (piperidin-1-ylmethyl) benzyl) pyrimidine-4, 6-diamine (0.30 g, 0.75 mmol) in dichloromethane (5.00 ml) were added N, N' -carbonyldiimidazole (1.26 g, 7.8 mmol) and triethylamine (1.58 g, 15.6 mmol) in that order. After the addition was complete, the reaction apparatus was moved to room temperature and stirred for 45 minutes.
The reaction solution was concentrated under reduced pressure. The brown solid was dissolved in dimethyl sulfoxide (5.0 ml) until clear and purified by preparative high performance liquid chromatography. The purification conditions were as follows, column: x select C1819 mm × 150 mm; mobile phase: water (containing 0.05% TFA) and acetonitrile; flow rate 25 ml/min; gradient: acetonitrile rose from 10% to 70% in 7 minutes; detection wavelength: 254 nm. The product was collected and lyophilized at low temperature under reduced pressure to give 17.6 mg of 6-amino-2-butoxy-7-hydroxy-9- (3- (piperidin-1-ylmethyl) benzyl) -7H-purin-8 (9H) -one trifluoroacetate as a pale yellow solid (Compound 6, yield 5.3%).
MS(ESI)M/Z:427[M+H+]。
19F NMR(300MHz,DMSO-d6)δ-74.25。
1H NMR(300MHz,DMSO-d6)δ11.75(s,1H),7.58–7.34(m,4H),6.52(br,s,2H),4.92(s,2H),4.74(s,2H),4.12(t,J=6.6Hz,2H),3.59–3.52(m,2H),3.52–3.40(m,2H),1.87–1.74(m,4H),1.66–1.56(m,3H),1.42–1.30(m,3H),0.90(t,J=7.5Hz,3H)。
Example 7: 5-amino-7-butoxy-2-methyl-1- ((4- (pyrrolidin-1-ylmethyl) pyridin-2-yl) methyl) -1, 4-dihydropyrimido [5,4-e ] [1,2,4] triazin-3 (2H) -one
Figure BDA0001836596920000491
Synthetic schemes
Figure BDA0001836596920000501
Step A: preparation of methyl 2-methylisonicotinate
2-methylisonicotinic acid (35.0 g, 255.5 mmol) was dissolved in methanol (520 ml). And under the ice-water bath, controlling the temperature of the reaction liquid not to exceed 10 ℃. Thionyl chloride (63.8 g, 536.5 mmol) was slowly added dropwise with stirring. After the dropwise addition, the reaction solution was heated to 60 ℃ and reacted for 6 hours.
The reaction solution was concentrated under reduced pressure, and the residue was dissolved in water (500 ml) and extracted with ethyl acetate (100 ml. times.1). The aqueous phase was adjusted to pH 9 to 10 with potassium carbonate and extracted with ethyl acetate (200 ml. times.3). The resulting organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure to obtain 35.2 g of methyl 2-methylisonicotinate as a white solid (yield 91.2%).
MS(ESI)M/Z:152[M+H+]。
And B: preparation of methyl 2- (bromomethyl) isonicotinate
To a1 l three-necked flask, methyl 2-methylisonicotinate (35.2 g, 233.1 mmol), carbon tetrachloride (400.0 ml), N-bromosuccinimide (41.5 g, 233.1 mmol), and azobisisobutyronitrile (3.8 g, 23.2 mmol) were sequentially added under a nitrogen atmosphere. Subsequently, the reaction liquid oil bath was heated to 60 degrees celsius and stirred overnight.
After the reaction solution was cooled to room temperature, the reaction solution was concentrated under reduced pressure. The resulting solid residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate 8/1). The product was collected and concentrated under reduced pressure to give 18.2 g of methyl 2- (bromomethyl) isonicotinate as a purple solid (yield 34.1%).
MS(ESI)M/Z:230.232[M+H+]。
And C: preparation of (2- (bromomethyl) pyridin-4-yl) methanol
Methyl 2- (bromomethyl) isonicotinate (2.00 g, 8.7 mmol) was dissolved in toluene (30 ml) under nitrogen. After the raw materials were stirred and dissolved, the reaction apparatus was placed in a salt-ice bath, and the temperature of the reaction solution was controlled to be maintained between-10 and 0 ℃, and a toluene solution of diisobutylaluminum hydride (6.9 ml, 1.5M, 10.4 mmol) was slowly added dropwise. After the addition was complete, stirring was continued at 0 ℃ for 2 hours.
Saturated sodium potassium tartrate solution (50 ml) was slowly added dropwise to the reaction solution in an ice water bath. Suction filtration was carried out under reduced pressure, and the aqueous phase was extracted with ethyl acetate (20 ml. times.2 times). The organic phases were combined. The organic phase was dried over anhydrous sodium sulfate and then concentrated under reduced pressure. 1.27 g of (2- (bromomethyl) pyridin-4-yl) methanol are obtained as a pale red solid. The crude product was used directly in the next reaction without purification.
Step D: preparation of tert-butyl 2- (2-butoxy-6-chloro-5-nitropyrimidin-4-yl) -2- ((4- (hydroxymethyl) pyridin-2-yl) methyl) -1-methylhydrazine-1-carboxylate
To a 100ml three-necked flask, tert-butyl 2- (2-butoxy-6-chloro-5-nitropyrimidin-4-yl) -1-methylhydrazine-1-carboxylate (1.2 g, 3.2 mmol), acetonitrile (50 ml), 2- (bromomethyl) pyridin-4-yl) methanol (643 mg, 3.2 mmol) and potassium carbonate (1.32 g, 9.6 mmol) were added in this order at room temperature. The reaction mixture was stirred at room temperature overnight.
The reaction solution was concentrated under reduced pressure to obtain a solid residue. The crude product was then purified by column chromatography on silica gel (petroleum ether/ethyl acetate 8/1) to give 820 mg of tert-butyl 2- (2-butoxy-6-chloro-5-nitropyrimidin-4-yl) -2- ((4- (hydroxymethyl) pyridin-2-yl) methyl) -1-methylhydrazine-1-carboxylate as a pale yellow solid (yield 51.5%).
MS(ESI)M/Z:497[M+H+]。
Step E: preparation of tert-butyl 2- (6-amino-2-butoxy-5-nitropyrimidin-4-yl) -2- ((4- (hydroxymethyl) pyridin-2-yl) methyl) -1-methylhydrazine-1-carboxylate
To a 30 ml autoclave, tert-butyl 2- (2-butoxy-6-chloro-5-nitropyrimidin-4-yl) -2- ((4- (hydroxymethyl) pyridin-2-yl) methyl) -1-methylhydrazine-1-carboxylate (670 mg, 1.35 mmol) and a solution of ammonia in tetrahydrofuran (20 ml, 2.0M,40.0 ml) were added in this order. After sealing, the mixture was heated at 50 ℃ for 2 hours.
After the reaction solution was cooled to room temperature, the reaction solution was concentrated under reduced pressure to give 520 mg of yellow solid tert-butyl 2- (6-amino-2-butoxy-5-nitropyrimidin-4-yl) -2- ((4- (hydroxymethyl) pyridin-2-yl) methyl) -1-methylhydrazine-1-carboxylate. The crude product was used directly in the next reaction without purification.
MS(ESI)M/Z:478[M+H+]。
Step F: preparation of tert-butyl 2- (6-amino-2-butoxy-5-nitropyrimidin-4-yl) -2- ((4-formylpyridin-2-yl) methyl) -1-methylhydrazine-1-carboxylate
To a 100ml three-necked flask, tert-butyl 2- (6-amino-2-butoxy-5-nitropyrimidin-4-yl) -2- ((4- (hydroxymethyl) pyridin-2-yl) methyl) -1-methylhydrazine-1-carboxylate (520 mg, 1.09 mmol), manganese dioxide (948 mg, 10.9 mmol), and chloroform (40.0 ml) were added in this order at room temperature. Subsequently, the reaction solution was heated at 50 ℃ for 5 hours.
After the reaction solution was cooled to room temperature, it was filtered with celite under reduced pressure. The filtrate was concentrated under reduced pressure and dissolved in N, N-dimethylformamide (2 ml) until clear. Then purified by preparative high performance liquid chromatography (column: X select C1819 mm X150 mm mobile phase: water (0.05% TFA)/acetonitrile; flow rate: 25 ml/min; gradient: acetonitrile from 10% to 90% in 7 min; detection wavelength: 254 nm. the product was collected and lyophilized under reduced pressure to give 180 mg of 2- (6-amino-2-butoxy-5-nitropyrimidin-4-yl) -2- ((4-formylpyridin-2-yl) methyl) -1-methylhydrazine-1-carboxylic acid tert-butyl ester as a yellow solid (34.8% yield).
MS(ESI)M/Z:496[M+H+]。
Step G: preparation of tert-butyl 2- (6-amino-2-butoxy-5-nitropyrimidin-4-yl) -1-methyl-2- ((4- (pyrrolidin-1-ylmethyl) pyridin-2-yl) methyl) hydrazine-1-carboxylate
To a 50ml three-necked flask was added tert-butyl 2- (6-amino-2-butoxy-5-nitropyrimidin-4-yl) -2- ((4-formylpyridin-2-yl) methyl) -1-methylhydrazine-1-carboxylate (180 mg, 0.38 mmol), dichloromethane (15 ml) and pyrrolidine (81 mg, 1.14 mmol) at 0 ℃ under nitrogen. After stirring at 0 to 10 ℃ for 10 minutes, sodium triacetoxyborohydride (97 mg, 0.46 mmol) was added. Subsequently, the reaction apparatus was moved to room temperature and stirred overnight.
The reaction solution was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (dichloromethane/methanol ═ 10/1) to give 120 mg of yellow semisolid tert-butyl 2- (6-amino-2-butoxy-5-nitropyrimidin-4-yl) -1-methyl-2- ((4- (pyrrolidin-1-ylmethyl) pyridin-2-yl) methyl) hydrazine-1-carboxylate (yield 59.6%).
MS(ESI)M/Z:531[M+H+]。
Step H: preparation of tert-butyl 2- (5, 6-diamino-2-butoxypyrimidin-4-yl) -1-methyl-2- ((4- (pyrrolidin-1-ylmethyl) pyridin-2-yl) methyl) hydrazine-1-carboxylate acetate
To a 50ml three-necked flask, tert-butyl 2- (6-amino-2-butoxy-5-nitropyrimidin-4-yl) -1-methyl-2- ((4- (pyrrolidin-1-ylmethyl) pyridin-2-yl) methyl) hydrazine-1-carboxylate (100 mg, 0.19 mmol), acetic acid (10 ml) and zinc powder (100 mg, 1.53 mmol) were added in this order at room temperature. Subsequently, the reaction mixture was stirred at room temperature for 3 hours.
And (4) carrying out suction filtration on the reaction solution under reduced pressure, and collecting filtrate. The filter cake was rinsed with ethyl acetate (5 ml × 2). The filtrates were combined and concentrated under reduced pressure to give 80 mg of yellow semisolid tert-butyl 2- (5, 6-diamino-2-butoxypyrimidin-4-yl) -1-methyl-2- ((4- (pyrrolidin-1-ylmethyl) pyridin-2-yl) methyl) hydrazine-1-carboxylate acetate. The crude product was used directly in the next reaction without purification.
MS(ESI)M/Z:501[M+H+]。
Step I: preparation of tert-butyl 2- (5, 6-diamino-2-butoxypyrimidin-4-yl) -1-methyl-2- ((4- (pyrrolidin-1-ylmethyl) pyridin-2-yl) methyl) hydrazine-1-carboxylate hydrochloride
To a 25ml three-necked flask, tert-butyl 2- (5, 6-diamino-2-butoxypyrimidin-4-yl) -1-methyl-2- ((4- (pyrrolidin-1-ylmethyl) pyridin-2-yl) methyl) hydrazine-1-carboxylate acetate (87 mg, 0.16 mmol) and 1, 4-dioxane (5.0 ml) were added. Stir at 0 ℃ for 5 minutes. Subsequently, a hydrochloric acid solution of 1, 4-dioxane (5.0 ml, 4.0M,20.0 mmol) was added. Stir at 0 ℃ for 1 hour.
The reaction solution was concentrated under reduced pressure to give 83 mg of yellow solid tert-butyl 2- (5, 6-diamino-2-butoxypyrimidin-4-yl) -1-methyl-2- ((4- (pyrrolidin-1-ylmethyl) pyridin-2-yl) methyl) hydrazine-1-carboxylate hydrochloride. The crude product was used directly in the next reaction without purification.
MS(ESI)M/Z:401[M+H+]。
Step J: preparation of 5-amino-7-butoxy-2-methyl-1- ((4- (pyrrolidin-1-ylmethyl) pyridin-2-yl) methyl) -1, 2-dihydropyrimido [5,4-e ] [1,2,4] triazin-3 (4H) -one trifluoroacetate
To a 25ml three-necked flask was added a mixed solution of tert-butyl 2- (5, 6-diamino-2-butoxypyrimidin-4-yl) -1-methyl-2- ((4- (pyrrolidin-1-ylmethyl) pyridin-2-yl) methyl) hydrazine-1-carboxylate hydrochloride (83 mg, 0.21 mmol), dichloromethane (4.0 ml) and tetrahydrofuran (4.0 ml). Triethylamine (106 mg, 1.05 mmol), N' -carbonyldiimidazole (102 mg, 0.63 mmol) were added sequentially with stirring at 0 ℃. Subsequently, the reaction solution was stirred for 1 hour while controlling the temperature at 0 ℃.
The reaction mixture was concentrated under reduced pressure. The crude product was dissolved in N, N-dimethylformamide (2.0 ml) until clear and the crude product was purified by preparative high performance liquid chromatography. The separation conditions were as follows, column: x select C1819 mm × 150 mm; mobile phase: water (containing 0.05% trifluoroacetic acid) and acetonitrile; flow rate: 25 ml/min; gradient: acetonitrile rose from 10% to 90% in 7 minutes; detection wavelength: 254 nm. The product was collected and lyophilized at low temperature to give 11.1 mg of 5-amino-7-butoxy-2-methyl-1- ((4- (pyrrolidin-1-ylmethyl) pyridin-2-yl) methyl) -1, 2-dihydropyrimido [5,4-e ] [1,2,4] triazin-3 (4H) -one trifluoroacetate as a brown oil (compound 7, yield 9.8%).
MS(ESI)M/Z:427[M+H+]。
1H NMR(300MHz,Methanol-d4)δ8.57(d,J=2.7Hz,1H),7.55-7.43(m,2H),4.62(s,2H),4.43(s,2H),4.27(t,J=7.5Hz,2H),3.63–3.30(m,4H),3.18(s,3H),2.25–1.95(m,4H),1.78–1.68(m,2H),1.54-1.42(m,2H),0.98(t,J=7.5Hz,3H)。
19F NMR(282MHz,Chloroform-d)δ-75.61。
Example 8: 5-amino-2-methyl-1- (3- (pyrrolidin-1-ylmethyl) benzyl) -7- (1- (tetrahydro-2H-pyran-4-yl) ethoxy) -1, 4-dihydropyrimido [5,4-e ] [1,2,4] triazin-3 (2H) -one
Figure BDA0001836596920000531
Synthetic schemes
Figure BDA0001836596920000541
Step A: preparation of 2- (6-amino-2-hydroxy-5-nitropyrimidin-4-yl) -1-methyl-2- (3- (pyrrolidin-1-ylmethyl) benzyl) hydrazine-1-carboxylic acid tert-butyl ester
Tert-butyl 2- (6-amino-2-butoxy-5-nitropyrimidin-4-yl) -1-methyl-2- (3- (pyrrolidin-1-ylmethyl) benzyl) hydrazine-1-carboxylate (2.00 g, 3.77 mmol) was dissolved in 50ml of tert-butanol, and 4 mol/l aqueous sodium hydroxide solution (9.4 ml) was added. The reaction was carried out at 70 ℃ overnight. LCMS monitored disappearance of starting material.
The reaction was concentrated under reduced pressure to remove the solvent, and the filter residue was dissolved in 10ml of N, N-dimethylformamide until clear. Purifying by preparative high performance liquid chromatography. The separation conditions were as follows, column: x select C1819 mm × 150 mm; mobile phase: water and acetonitrile; flow rate: 25 ml/min; gradient: acetonitrile rose from 0% to 70% in 30 minutes; detection wavelength: 254 nm. The product was collected and lyophilized to give 1.00 g of tert-butyl 2- (6-amino-2-hydroxy-5-nitropyrimidin-4-yl) -1-methyl-2- (3- (pyrrolidin-1-ylmethyl) benzyl) hydrazine-1-carboxylate (yield 82%).
MS(ESI)M/Z:474[M+H+]。
Steps B1 to B2: preparation of 1- (tetrahydro-2H-pyran-4-yl) ethyl methanesulfonate
1- (tetrahydro-2H-pyran-4-yl) ethan-1-one (10.00 g, 78.13 mmol) was dissolved in dry methanol (100 mL) under an ice-water bath. Then, sodium borohydride (1.50 g, 39.47 mmol) was added to the reaction solution in portions. After stirring for half an hour, TLC monitored and found that the starting material disappeared.
Ice water (50 ml) was slowly added to the reaction. The mixture was concentrated under reduced pressure to remove methanol. The aqueous phase was extracted with ethyl acetate (50 ml. times.3 times) and the organic phases were combined. The organic phase is washed once with 50ml of saturated sodium chloride solution, then dried over anhydrous sodium sulfate and concentrated under reduced pressure. 10.02 g of 1- (tetrahydro-2H-pyran-4-yl) -ethan-1-ol are obtained. The product is used for the next reaction without purification.
1- (tetrahydro-2H-pyran-4-yl) ethan-1-ol (5.00 g, 38.46 mmol), triethylamine (7.80 g, 70.08 mmol) was dissolved in 20 ml of dichloromethane under an ice-water bath. Methanesulfonyl chloride (5.30 g, 46.09 mmol) was then added dropwise. After the dropwise addition, the reaction solution was naturally warmed to room temperature, and then stirred at room temperature for 4 hours. TLC monitoring found disappearance of starting material.
Ice water (20 ml) was slowly added to the reaction. The mixture was extracted with dichloromethane (20 ml × 3 times) and the organic phases were combined. The organic phase is washed first with 1 mol/l dilute hydrochloric acid (20 ml × 2 times), then with saturated sodium bicarbonate solution (20 ml × 2 times) and finally with saturated sodium chloride solution (20 ml × 2 times). The washed organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure to give 7.50 g of 1- (tetrahydro-2H-pyran-4-yl) -ethyl methanesulfonate. The compound is unstable and does not need to be purified, and the crude product is directly used for the next reaction.
And C: preparation of 2- (6-amino-5-nitro-2- (1- (tetrahydro-2H-pyran-4-yl) ethoxy) pyrimidin-4-yl) -1-methyl-2- (3- (pyrrolidine) -1-ylmethyl) benzyl) hydrazine-1-carboxylic acid tert-butyl ester
Tert-butyl 2- (6-amino-2-hydroxy-5-nitropyrimidin-4-yl) -1-methyl-2- (3- (pyrrolidin-1-ylmethyl) benzyl) hydrazine-1-carboxylate (1.00 g, 2.11 mmol) and 1- (tetrahydro-2H-pyran-4-yl) ethyl methanesulfonate (7.50 g, 36.06 mmol) were dissolved in 100ml of acetonitrile followed by addition of cesium carbonate (2.00 g, 6.13 mmol). After stirring and mixing well, the reaction was carried out overnight at 80 ℃.
And (4) carrying out suction filtration on the reaction solution, and collecting filtrate. The filter residue was washed with dichloromethane (20 ml × 2 times). The filtrates were combined and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (eluent: methanol/dichloromethane ═ 1/20) to give 458 mg of tert-butyl 2- (6-amino-5-nitro-2- (1- (tetrahydro-2H-pyran-4-yl) ethoxy) pyrimidin-4-yl) -1-methyl-2- (3- (pyrrolidine) -1-ylmethyl) benzyl) hydrazine-1-carboxylate as a white solid (yield 37%).
MS(ESI)M/Z:586[M+H+]。
Step D: preparation of 2- (5, 6-diamino-2- (1- (tetrahydro-2H-pyran-4-yl) ethoxy) pyrimidin-4-yl) -1-methyl-2- (3- (pyrrolidin-1-ylmethyl) benzyl) hydrazine-1-carboxylic acid tert-butyl ester acetate
Tert-butyl 2- (6-amino-5-nitro-2- (1- (tetrahydro-2H-pyran-4-yl) ethoxy) pyrimidin-4-yl) -1-methyl-2- (3- (pyrrolidine) -1-ylmethyl) benzyl) hydrazine-1-carboxylate (458 mg, 0.78 mmol) was dissolved in 5.00 ml of glacial acetic acid. Then, zinc powder (101.4 mg, 1.56 mmol) was added to the reaction solution. Stir at room temperature overnight.
The reaction was filtered, the filtrate was collected, and the residue was washed with ethyl acetate (20 ml × 2 times). The filtrates were combined and concentrated under reduced pressure to give 400 mg of tert-butyl 2- (5, 6-diamino-2- (1- (tetrahydro-2H-pyran-4-yl) ethoxy) pyrimidin-4-yl) -1-methyl-2- (3- (pyrrolidin-1-ylmethyl) benzyl) hydrazine-1-carboxylate acetate as a yellow oil. The product is used for the next reaction without purification.
MS(ESI)M/Z:556[M+H+]。
Step E: preparation of 6- (2-methyl-1- (3- (pyrrolidin-1-ylmethyl) benzyl) hydrazino) -2- (1- (tetrahydro-2H-pyran-4-yl) ethoxy) pyrimidine-4, 5-diamine trifluoroacetate salt
2- (5, 6-diamino-2- (1- (tetrahydro-2H-pyran-4-yl) ethoxy) pyrimidin-4-yl) -1-methyl-2- (3- (pyrrolidin-1-ylmethyl) benzyl) hydrazine-1-carboxylic acid tert-butyl ester acetate (400 mg, crude) was dissolved in dichloromethane (10.0 ml) under an ice-water bath. Trifluoroacetic acid (3.0 ml) was then added dropwise. After the dropwise addition, the reaction apparatus was moved to room temperature and stirred for 3 hours.
The reaction solution was concentrated to dryness under reduced pressure to give 350 mg of 6- (2-methyl-1- (3- (pyrrolidin-1-ylmethyl) benzyl) hydrazino) -2- (1- (tetrahydro-2H-pyran-4-yl) ethoxy) pyrimidine-4, 5-diamine trifluoroacetate as a yellow liquid.
MS(ESI)M/Z:456[M+H+]。
Step F: preparation of 5-amino-2-methyl-1- (3- (pyrrolidin-1-ylmethyl) benzyl) -7- (1- (tetrahydro-2H-pyran-4-yl) ethoxy) -1, 4-dihydropyrimido [5,4-e ] [1,2,4] triazin-3 (2H) -one trifluoroacetate
6- (2-methyl-1- (3- (pyrrolidin-1-ylmethyl) benzyl) hydrazino) -2- (1- (tetrahydro-2H-pyran-4-yl) ethoxy) pyrimidine-4, 5-diamine trifluoroacetate (350 mg, crude), triethylamine (775 mg, 7.66 mmol) and N, N' -carbonyldiimidazole (373.4 mg, 2.30 mmol) were added successively to a mixed solvent of dichloromethane (5.0 ml) and tetrahydrofuran (5.0 ml) under an ice-water bath. After stirring and mixing uniformly, the reaction solution was naturally warmed to room temperature, and stirring was continued at room temperature for 1 hour.
The reaction was concentrated under reduced pressure, and the residue was dissolved in N, N-dimethylformamide (5.0 ml) until clear, and purified by preparative high performance liquid chromatography. The purification conditions were as follows, column: x select C1819 mm × 150 mm; mobile phase: water (containing 0.05% TFA) and acetonitrile; the flow rate is 20 ml/min; gradient: acetonitrile rose from 10% to 60% in 15 minutes; detection wavelength: 254 nm. The product was collected and lyophilized under reduced pressure. This gave 11.1 mg of 5-amino-2-methyl-1- (3- (pyrrolidin-1-ylmethyl) benzyl) -7- (1- (tetrahydro-2H-pyran-4-yl) ethoxy) -1, 4-dihydropyrimido [5,4-e ] [1,2,4] triazin-3 (2H) -one trifluoroacetate as a yellow solid. (Compound 8, three-step yield 2.4%)
MS(ESI)M/Z:482[M+H+]。
19F NMR(300MHz,Methanol-d4)δ-76.98。
1H NMR(300MHz,Methanol-d4)δ7.65–7.48(m,2H),7.41(d,J=7.8Hz,1H),7.27–7.18(m,1H),5.13–4.97(m,1H),4.51(s,2H),4.37(s,2H),4.09–3.85(m,2H),3.60–3.39(m,4H),3.25–3.08(m,5H),2.30–2.11(m,2H),2.10–1.96(m,2H),1.93–1.73(m,2H),1.73–1.58(m,1H),1.58–1.39(m,2H),1.34(d,J=6.3Hz,3H)。
Example 9: 5-amino-7-butoxy-2-methyl-1- (3- ((2-methylpyrrolidin-1-yl) methyl) benzyl) -1, 4-dihydropyrimido [5,4-e ] [1,2,4] triazin-3 (2H) -one
Figure BDA0001836596920000561
Synthetic schemes
Figure BDA0001836596920000571
Step A: preparation of tert-butyl 2- (6-amino-2-butoxy-5-nitropyrimidin-4-yl) -1-methyl-2- (3- (2-methylpyrrolidin-1-ylmethyl) benzyl) hydrazine-1-carboxylate
To a solution of tert-butyl 2- (6-amino-2-butoxy-5-nitropyrimidin-4-yl) -2- (3-formylbenzyl) -1-methylhydrazine-1-carboxylate (150 mg, 0.316 mmol) in 1, 2-dichloroethane (2.0 ml) under a nitrogen atmosphere was added 2-methylpyrrolidine (150 mg, 0.316 mmol). Half an hour later, sodium borohydride acetate (150 mg, 0.316 mmol) was added slowly in portions at zero degrees centigrade. Stirred at room temperature for three hours.
And (3) post-treatment: the reaction was diluted with water (5 ml) and extracted with dichloromethane (10 ml. times.3 times). The combined organic phases were dried over anhydrous sodium sulfate, concentrated under reduced pressure and the concentrate was purified over a silica gel column (eluent: methanol/dichloromethane ═ 1/10) to give 2- (6-amino-2-butoxy-5-nitropyrimidin-4-yl) -1-methyl-2- (3- (2-methylpyrrolidin-1-ylmethyl) benzyl) hydrazine-1-carboxylic acid tert-butyl ester as a pale yellow solid (150 mg yield 87%).
MS(ESI)M/Z:544[M+H+]。
And B: 2- (5, 6-diamino-2-butoxypyrimidin-4-yl) -1-methyl-2- (3- (2-methylpyrrolidin-1-ylmethyl) benzyl) hydrazine-1-carboxylic acid tert-butyl ester acetate
To a solution of tert-butyl 2- (6-amino-2-butoxy-5-nitropyrimidin-4-yl) -1-methyl-2- (3- (2-methylpyrrolidin-1-ylmethyl) benzyl) hydrazine-1-carboxylate (150 mg, 0.276 mmol) in acetic acid (1.5 ml) was added zinc powder (180 mg, 2.81 mmol). Stirred at room temperature for 2 hours.
And (3) post-treatment: filtration and washing of the filter cake with ethyl acetate (5 ml. times.3) the filtrate was collected and concentrated under reduced pressure to give 2- (5, 6-diamino-2-butoxypyrimidin-4-yl) -1-methyl-2- (3- (2-methylpyrrolidin-1-ylmethyl) benzyl) hydrazine-1-carboxylic acid tert-butyl ester acetate as a pale yellow oil (120 mg) which was used without purification in the next reaction.
MS(ESI)M/Z:514[M+H+]
And C: preparation of 2-butoxy-6- (2-methyl-1- (3- (2-methylpyrrolidin-1-ylmethyl) benzyl) hydrazino) pyrimidine-4, 5-diamine trifluoroacetate salt
A100 ml three-necked flask was taken, and trifluoroacetic acid (0.5 ml) was slowly added to a dichloromethane solution (1 ml) of tert-butyl 2- (5, 6-diamino-2-butoxypyrimidin-4-yl) -1-methyl-2- (3- (2-methylpyrrolidin-1-ylmethyl) benzyl) hydrazine-1-carboxylate acetate (120 mg, 0.234 mmol), and the mixture was stirred at room temperature for 3 hours.
And (3) post-treatment: concentration under reduced pressure gave 2-butoxy-6- (2-methyl-1- (3- (2-methylpyrrolidin-1-ylmethyl) benzyl) hydrazino) pyrimidine-4, 5-diamine trifluoroacetate (160 mg) as a pale yellow oil which was used in the next reaction without purification.
MS(ESI)M/Z:414[M+H+]。
Step D: preparation of 5-amino-7-butoxy-2-methyl-1- (3- (2-methylpyrrolidin-1-ylmethyl) benzyl) -1, 4-dihydropyrimido [5,4-e ] [1,2,4] triazin-3 (2H) -one trifluoroacetate
To a mixed solution of 2-butoxy-6- (2-methyl-1- (3- (2-methylpyrrolidin-1-ylmethyl) benzyl) hydrazino) pyrimidine-4, 5-diamine trifluoroacetate (97 mg, 0.233 mmol) in tetrahydrofuran (2 ml) and dichloromethane (2 ml) was added triethylamine (235 mg, 2.33 mmol) and N, N' -carbonyldiimidazole (112 mg, 0.70 mmol) sequentially at 0 ℃. The addition was complete and the reaction solution was allowed to warm to 0 ℃ and stirring was continued for 1 hour.
And (3) post-treatment: the reaction was concentrated under reduced pressure, and the residue was dissolved in N, N-dimethylformamide (2.0 ml) to be clear. The crude product was purified by preparative high performance liquid chromatography. The purification conditions were as follows, column: x-select C1819X 150; mobile phase: water (0.05% TFA) and acetonitrile; flow rate 25 ml/min; gradient: acetonitrile from 10% to 80% in 8 minutes; detection wavelength: 254 nm. Lyophilization under reduced pressure at low temperature gave 5.0 mg of a pale yellow semisolid 5-amino-7-butoxy-2-methyl-1- (3- (2-methylpyrrolidin-1-ylmethyl) benzyl) -1, 4-dihydropyrimido [5,4-e ] [1,2,4] triazin-3 (2H) -one trifluoroacetate (Compound 9, yield 5%).
MS(ESI)M/Z:440[M+H+]。
1H-NMR:(300MHz,Methanol-d4):δ7.51–7.35(m,3H),7.22(d,J=6.9Hz,1H),4.58–4.53(m,1H),4.48(s,2H),4.28(t,J=6.0Hz,2H),4.15–4.11(m,1H),3.56–3.49(m,1H),3.18–3.09(m,4H),2.41–2.34(m,1H),2.12–1.96(m,2H),1.79–1.70(m,3H),1.56–1.37(m,6H),0.97(t,J=7.2Hz,3H)。
19F NMR(282MHz,Methanol-d4)δ-76.88。
Example 10: 5-amino-7-butoxy-1- (3- (pyrrolidin-1-ylmethyl) benzyl) -1, 4-dihydropyrimido [5,4-e ] [1,2,4] triazin-3 (2H) -one
Figure BDA0001836596920000581
Synthetic schemes
Figure BDA0001836596920000591
Step A: preparation of methyl 2- (2-butoxy-6-amino-5-nitropyrimidin-4-yl) hydrazine-1-carboxylate
To a solution of 2-butoxy-6-chloro-5-nitropyrimidin-4-amine (2 g, 8.1 mmol) in tetrahydrofuran (20 ml) were added triethylamine (1.63 g, 16.2 mmol) and methoxycarbonylhydrazide (1.02 g, 15.04 mmol) in this order, followed by stirring at room temperature overnight.
And (3) post-treatment: concentration under reduced pressure gave a solid residue which was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate ═ 2/1) to give methyl 2- (2-butoxy-6-amino-5-nitropyrimidin-4-yl) hydrazine-1-carboxylate (1.44 g, 60% yield) as a yellow solid.
MS(ESI)M/Z:301[M+H+]。
And B: preparation of methyl 2- (2-butoxy-6-amino-5-nitropyrimidin-4-yl) -2- (3-formylbenzyl) hydrazine-1-carboxylate
To a solution of methyl 2- (2-butoxy-6-amino-5-nitropyrimidin-4-yl) hydrazine-1-carboxylate (1.44 g, 4.80 mmol) in acetonitrile (20 ml) was added potassium carbonate (746.3 mg, 5.4 mmol). Then stirred at room temperature for 8 hours.
And (3) post-treatment: concentration under reduced pressure gave a solid residue which was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate ═ 3/1) to give methyl 2- (2-butoxy-6-amino-5-nitropyrimidin-4-yl) -2- (3-formylbenzyl) hydrazine-1-carboxylate (1.18 g, 60% yield) as a yellow solid.
And C: preparation of methyl 2- (2-butoxy-6-amino-5-nitropyrimidin-4-yl) -2- (3- (pyrrolidin-1-ylmethyl) benzyl) hydrazine-1-carboxylate
To a solution of methyl 2- (2-butoxy-6-amino-5-nitropyrimidin-4-yl) -2- (3-formylbenzyl) hydrazine-1-carboxylate (700 mg, 1.68 mmol) in dichloromethane (25 ml) was added pyrrolidine (600 mg, 8.36 mmol), and the solution was stirred at room temperature for 5 minutes. Sodium borohydride acetate (925 mg, 4.37 mmol) was then added and stirred at room temperature for 5 hours.
And (3) post-treatment: concentration under reduced pressure gave a solid residue which was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate ═ 2/1) to give methyl 2- (2-butoxy-6-chloro-5-nitropyrimidin-4-yl) -2- (3-formylbenzyl) hydrazine-1-carboxylate (450 mg, 57% yield) as a yellow solid.
MS(ESI)M/Z:474[M+H+]
Step D: preparation of methyl 2- (5, 6-diamino-2-butoxypyrimidin-4-yl) -2- (3- (pyrrolidin-1-ylmethyl) benzyl) hydrazine-1-carboxylate
To a solution of methyl 2- (2-butoxy-6-amino-5-nitropyrimidin-4-yl) -2- (3-formylbenzyl) hydrazine-1-carboxylate (250 mg, 0.53 mmol) in ethanol (10 ml) and water (2.5 ml) was added iron powder (249.3 mg, 4.45 mmol) and ammonium chloride (83 mg, 1.5 mmol). Subsequently, the reaction apparatus was stirred at 60 ℃ for 12 hours.
And (3) post-treatment: filtering and collecting filtrate. The filter residue was washed with ethyl acetate (50 ml. times.3). The filtrates are combined and concentrated under reduced pressure,
the residue was purified by preparative high performance liquid chromatography. The separation conditions were as follows, column: x select C1819 mm × 150 mm; mobile phase: water (0.05% ammonium bicarbonate) and acetonitrile; flow rate: 25 ml/min; gradient: acetonitrile rose from 0% to 90% in 25 minutes; detection wavelength: 254 nm. The product was collected and lyophilized under reduced pressure to give methyl 2- (5, 6-diamino-2-butoxypyrimidin-4-yl) -2- (3- (pyrrolidin-1-ylmethyl) benzyl) hydrazine-1-carboxylate (45 mg, 19% yield) as a yellow solid.
MS(ESI)M/Z:444[M+H+]。
Step E: 5-amino-7-butoxy-1- (3- (pyrrolidin-1-ylmethyl) benzyl) -1, 4-dihydropyrimido [5,4-e ] [1,2,4] triazin-3 (2H) -one
Methyl 2- (5, 6-diamino-2-butoxypyrimidin-4-yl) -2- (3- (pyrrolidin-1-ylmethyl) benzyl) hydrazine-1-carboxylate (45 mg, 0.1 mmol) was dissolved in N, N-dimethylacetamide (2 ml) and reacted at 120 ℃ for 1 hour.
And (3) post-treatment: the reaction solution was filtered and purified by preparative high performance liquid chromatography. The purification conditions were as follows, column: xselect C1819 mm × 150 mm; mobile phase: water (containing 0.05% trifluoroacetic acid) and acetonitrile; flow rate: 25 ml/min; gradient: acetonitrile rose from 5% to 80% in 20 minutes; detection wavelength: 254 nm. Lyophilization under reduced pressure gave 8 mg of 5-amino-7-butoxy-1- (3- (pyrrolidin-1-ylmethyl) benzyl) -1, 4-dihydropyrimido [5,4-e ] [1,2,4] triazin-3 (2H) -one trifluoroacetate as a brown solid (compound 10, 1.4% yield)
MS(ESI)M/Z:412[M+H+]。
1H NMR(300MHz,Methanol-d4):δ7.69–7.42(m,4H),4.66(s,2H),4.45–4.27(m,4H),3.50(s,2H),3.16(s,2H),2.17–1.81(m,4H),1.81–1.28(m,4H),0.99–0.94(m,3H)。
19F NMR(282MHz,Methanol-d4)δ-77.03。
Example 11: 5-amino-7-butoxy-1- (2-fluoro-5- (pyrrolidin-1-ylmethyl) benzyl) -2-methyl-1, 4-dihydropyrimido [5,4-e ] [1,2,4] triazin-3 (2H) -one
Figure BDA0001836596920000611
Synthetic schemes
Figure BDA0001836596920000612
Step A-B: preparation of (3- (bromomethyl) -4-fluorophenyl) methanol
To a1 l single neck round bottom flask were added 4-fluoro-3-methylbenzoic acid (10.0 g, 64.9 mmol), carbon tetrachloride (300 ml), N-bromosuccinimide (11.5 g, 64.9 mmol), and azobisisobutyronitrile (200 mg, catalytic amount) in that order. After stirring and mixing uniformly, heating at 90 ℃ for 3 hours. LC-MS monitoring indicated completion of the reaction.
After the reaction mixture was cooled to room temperature, saturated aqueous ammonium chloride (100 ml) was added to the reaction system to quench the reaction. And (4) carrying out suction filtration on the reaction solution, and collecting filtrate. Standing and layering the filtrate, and separating to obtain an organic phase. The aqueous phase is extracted with dichloromethane (100 ml × 2 times). The organic phases are combined, washed with water (50 ml × 2 times), then dried over anhydrous sodium sulfate and finally concentrated under reduced pressure. The concentrate was purified by silica gel column (eluent: ethyl acetate/petroleum ether: 3/2). The product was collected and concentrated under reduced pressure to give 6.80 g of 3- (bromomethyl) -4-fluorobenzoic acid as a white solid (yield, 45.1%). The crude product was used directly in the next reaction.
To a 500 ml single neck round bottom flask were added 3- (bromomethyl) -4-fluorobenzoic acid (6.30 g, 19.69 mmol) and tetrahydrofuran (100 ml) in that order. Under ice-bath, borane-tetrahydrofuran complex (1.0M, 200 ml, 200 mmol) was slowly added dropwise. After the addition was complete the ice bath was removed and the reaction was stirred at room temperature for an additional 12 hours and TLC monitoring indicated the disappearance of the starting material.
The reaction was slowly added to a5 l plastic beaker containing ice water (300 ml). The mixture was extracted with ethyl acetate (500 ml. times.3 times) and the organic phases were combined. The organic phase was dried over anhydrous sodium sulfate and then concentrated under reduced pressure. 5.0 g of (3- (bromomethyl) -4-fluorophenyl) methanol as a white solid were obtained (yield, 84.5%). The reaction mixture was used in the next reaction without further purification.
And C: preparation of tert-butyl 2- (2-butoxy-6-chloro-5-nitropyrimidin-4-yl) -2- (2-fluoro-5- (hydroxymethyl) benzyl) -1-methylhydrazine-1-carboxylate
To a 50ml single neck round bottom flask were added sequentially (3- (bromomethyl) -4-fluorophenyl) methanol (630 mg, 2.89 mmol), acetonitrile (15.0 ml), tert-butyl 2- (2-butoxy-6-chloro-5-nitropyrimidin-4-yl) -1-methylhydrazine-1-carboxylate (1.08 g, 2.89 mmol) and anhydrous potassium carbonate (1.10 g, 7.98 mmol). Stir at 60 ℃ for 3 hours. After the reaction system was cooled to room temperature, water (20.0 ml) was added to dilute the reaction solution. The mixture was extracted with ethyl acetate (50 ml. times.3 times) and the organic phases were combined. The organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure to give 1.24 g of tert-butyl 2- (2-butoxy-6-chloro-5-nitropyrimidin-4-yl) -2- (2-fluoro-5- (hydroxymethyl) benzyl) -1-methylhydrazine-1-carboxylate as a pale yellow oil. The product was used in the next reaction without further purification.
MS(ESI)M/Z:514[M+H+]。
Step D: preparation of tert-butyl 2- (2-butoxy-6-chloro-5-nitropyrimidin-4-yl) -2- (2-fluoro-5-formylbenzyl) -1-methylhydrazine-1-carboxylate
To a 25ml single-necked flask, tert-butyl 2- (2-butoxy-6-chloro-5-nitropyrimidin-4-yl) -2- (2-fluoro-5- (hydroxymethyl) benzyl) -1-methylhydrazine-1-carboxylate (1.24 g, 2.42 mmol) and dichloromethane (10 ml) were added. After stirring to dissolve, manganese dioxide (585 mg, 6.72 mmol) was added. Stir at 40 degrees celsius overnight.
And (4) carrying out suction filtration on the reaction solution, and collecting filtrate. The filtrate was concentrated under reduced pressure. The concentrate was purified by silica gel column (eluent: ethyl acetate/petroleum ether: 1/1). The product was collected and concentrated under reduced pressure to give 580 mg of tert-butyl 2- (2-butoxy-6-chloro-5-nitropyrimidin-4-yl) -2- (2-fluoro-5-formylbenzyl) -1-methylhydrazine-1-carboxylate as a pale yellow solid (yield, 43.3%).
MS(ESI)M/Z:512[M+H+]。
Step E: preparation of tert-butyl 2- (6-amino-2-butoxy-5-nitropyrimidin-4-yl) -2- (2-fluoro-5-formylbenzyl) -1-methylhydrazine-1-carboxylate
To a 10ml autoclave reactor were added tert-butyl 2- (2-butoxy-6-chloro-5-nitropyrimidin-4-yl) -2- (2-fluoro-5-formylbenzyl) -1-methylhydrazine-1-carboxylate (580 mg, 0.978 mmol) and a solution of ammonia in isopropanol (2.0M,3.0 ml, 6.0 mmol) in that order. After stirring and mixing uniformly, the reaction solution was heated in an oil bath at 60 ℃ for 12 hours. After completion of the reaction as monitored by LC-MS, the reaction solution was poured into water (5 ml) and extracted with ethyl acetate (10 ml × 3 times). The organic phases were combined and dried over anhydrous sodium sulfate. Concentration under reduced pressure gave 390 mg of crude 2- (6-amino-2-butoxy-5-nitropyrimidin-4-yl) -2- (2-fluoro-5-formylbenzyl) -1-methylhydrazine-1-carboxylic acid tert-butyl ester as a pale yellow oil. The compound was used directly in the next reaction without purification.
MS(ESI)M/Z:493[M+H+]。
Step F: preparation of tert-butyl 2- (6-amino-2-butoxy-5-nitropyrimidin-4-yl) -2- (2-fluoro-5- (pyrrolidin-1-ylmethyl) benzyl) -1-methylhydrazine-1-carboxylate
To a 25ml three-necked flask were added tert-butyl 2- (6-amino-2-butoxy-5-nitropyrimidin-4-yl) -2- (2-fluoro-5-formylbenzyl) -1-methylhydrazine-1-carboxylate (390 mg, 0.793 mmol), dichloromethane (5.0 ml) and pyrrolidine (750 mg, 1.59 mmol) in that order. After stirring at room temperature for half an hour, sodium triacetoxyborohydride (750 mg, 1.58 mmol) was added slowly in portions at 0 ℃. After the addition was complete, the reaction was allowed to stir at room temperature for three hours. LC-MS monitoring indicated complete reaction.
Water (10 ml) was added to dilute the reaction. The mixture was extracted with dichloromethane (20 ml × 3 times). The organic phases were combined. The organic phase was dried over anhydrous sodium sulfate and then concentrated under reduced pressure, and the resulting residue was purified over a silica gel column (eluent: methanol/dichloromethane ═ 1/10) to give 270 mg of tert-butyl 2- (6-amino-2-butoxy-5-nitropyrimidin-4-yl) -2- (2-fluoro-5- (pyrrolidin-1-ylmethyl) benzyl) -1-methylhydrazine-1-carboxylate as a pale yellow solid (yield, 62.3%).
MS(ESI)M/Z:548[M+H+]。
Step G: preparation of tert-butyl 2- (5, 6-diamino-2-butoxypyrimidin-4-yl) -2- (2-fluoro-5- (pyrrolidin-1-ylmethyl) benzyl) -1-methylhydrazine-1-carboxylate
To a 25ml single neck round bottom flask was added tert-butyl 2- (6-amino-2-butoxy-5-nitropyrimidin-4-yl) -2- (2-fluoro-5- (pyrrolidin-1-ylmethyl) benzyl) -1-methylhydrazine-1-carboxylate (270 mg, 0.494 mmol), reduced zinc powder (360 mg, 5.62 mmol) and acetic acid (3 ml) in that order. After stirring at room temperature for 2 hours, LC-MS monitoring indicated complete reaction.
The reaction solution was filtered through celite. The filtrate was collected and the filter cake was rinsed with ethyl acetate (20 ml × 2 times). The combined filtrates were concentrated under reduced pressure to give 240 mg of crude 2- (5, 6-diamino-2-butoxypyrimidin-4-yl) -2- (2-fluoro-5- (pyrrolidin-1-ylmethyl) benzyl) -1-methylhydrazine-1-carboxylic acid tert-butyl ester as a pale yellow oil, which was used in the next reaction without purification.
MS(ESI)M/Z:518[M+H+]。
Step H: preparation of 2-butoxy-6- (1- (2-fluoro-5- (pyrrolidin-1-ylmethyl) benzyl) -2-methylhydrazino) pyrimidine-4, 5-diamine trifluoroacetate salt
To a 25ml single neck round bottom flask was added tert-butyl 2- (5, 6-diamino-2-butoxypyrimidin-4-yl) -2- (2-fluoro-5- (pyrrolidin-1-ylmethyl) benzyl) -1-methylhydrazine-1-carboxylate (240 mg, 0.464 mmol), dichloromethane (2.0 ml) and trifluoroacetic acid (1.0 ml) in that order. After stirring at room temperature for 2 hours, LC-MS monitoring indicated complete reaction.
The reaction solution was concentrated under reduced pressure to give 320 mg of 2-butoxy-6- (1- (2-fluoro-5- (pyrrolidin-1-ylmethyl) benzyl) -2-methylhydrazino) pyrimidine-4, 5-diamine as a pale yellow oil. The compound was used directly in the next reaction without purification.
MS(ESI)M/Z:418[M+H+]。
Step I: preparation of 5-amino-7-butoxy-1- (2-fluoro-5- (pyrrolidin-1-ylmethyl) benzyl) -2-methyl-1, 4-dihydropyrimido [5,4-e ] [1,2,4] triazin-3 (2H) -one trifluoroacetate to a 25mL single neck round bottom flask were added 2-butoxy-6- (1- (2-fluoro-5- (pyrrolidin-1-ylmethyl) benzyl) -2-methylhydrazinyl) pyrimidine-4, 5-diamine (320 mg, 0.767 mmol), tetrahydrofuran (3.0 mL) and dichloromethane (3.0 mL) in that order. After stirring to dissolve, triethylamine (773 mg, 7.67 mmol) and N, N' -carbonyldiimidazole (368 mg, 2.30 mmol) were added sequentially to an ice-water bath. After 1 hour, LC-MS monitoring indicated complete reaction.
The reaction solution was concentrated under reduced pressure, and the resulting residue was dissolved in N, N-dimethylformamide (3.0 ml) to be clear. Purifying by preparative high performance liquid chromatography. The purification conditions were as follows, column: x select C1819 mm × 150 mm; mobile phase: water (containing 0.05% TFA) and acetonitrile; flow rate 25 ml/min; gradient: acetonitrile rose from 10% to 80% in 7 minutes; detection wavelength: 254 nm. The product was collected and lyophilized under reduced pressure to give 139.8 mg of a yellow semi-solid, 5-amino-7-butoxy-1- (2-fluoro-5- (pyrrolidin-1-ylmethyl) benzyl) -2-methyl-1, 4-dihydropyrimido [5,4-e ] [1,2,4] triazin-3 (2H) -one trifluoroacetate salt (Compound 11, yield 41.1%).
MS(ESI)M/Z:444[M+H+]。
1H-NMR(300MHz,CD3OD,ppm):δ7.58–7.55(m,1H),7.45(d,J=6.6Hz,1H),7.19(t,J=9.0Hz,1H),4.59(s,2H),4.39–4.34(m,4H),3.46(s,2H),3.23(s,3H),3.12(s,2H),2.23–1.92(m,4H),1.81–1.70(m,2H),1.55–1.40(m,2H),0.99(t,J=7.5Hz,3H)。
19F NMR(300MHz,CD3OD):δ-117.77。
Effects of the embodiment
An experimental instrument: envision (Perkinelmer; model: 2104); MSD (MSD; model: MESO SECTORS600system)
Cells and reagents:
HEK-Blue hTLR7 and HEK-Blue hTLR8 cells (InvivoGen)
HEK-BlueTMDetection(InvivoGen)
DMEM(Gibco)
blasticidin(Invitrogen)
ZeocinTM(InvivoGen)
NormocinTM(InvivoGen)
Penicillin-Streptomycin(Gibco)
Fetal Bovine Serum(Gibco)
Phosphate Buffered Saline(PBS)(Gibco)
Abbreviations:
DMSO:Dimethyl Sulfoxide(Sigma)
MSD:Meso Scale Detection
QC:quality control
IS:Internal Standard
effect example 1: HEK-Blue hTLR7And HEK-Blue hTLR8Assay for drug receptor affinity activity in cells
Experimental procedure
The method comprises the following steps: cell plating:
a) HEK-Blue is brought in advanceTMDetection reagent and PBS placed in 37 degrees C preheating.
b) The medium in the flask was discarded and washed once with pre-warmed PBS.
c) An appropriate amount of PBS was added to the flask and left at 37 ℃ for 5 min.
d) The flask was tapped to exfoliate the cells, and the digestion reaction was terminated with a medium containing 10% FBS.
e) The cell suspension was transferred to a 15mL sterile centrifuge tube and centrifuged at 1000rpm for 5 min.
f) Discarding supernatant, and using HEK-BlueTMDetection reagent heavy suspension cells, and count.
g) In HEK-BlueTMDilution of cell density to 2.2X10 in Detection reagent5/mL。
h) 45 μ L of diluted cell suspension, i.e. 10000 cells per well, was seeded into 384 well plates.
Step two: and (3) drug treatment:
a) test compounds were made up in 10mM stock in DMSO.
b) Preparation of compounds
The control compound GS-9620 has the following structure:
Figure BDA0001836596920000651
test and control compounds GS-9620:
the maximum concentration was diluted from 10mM to 2mM, followed by 3-fold dilution, 10 gradients, DMSO as a negative control. mu.L of the diluted compound was added to 38. mu.L of HEK-BlueTMDetection reagents were diluted 20-fold intermediately.
c) Treatment of cells with compounds
5 μ L of the intermediate diluted compound was added to a 384 well plate seeded with 45 μ L of cells and the drug was diluted 10 fold to a final DMSO concentration of 0.5%.
The final effect concentrations of test compound and control compound were 10000,3333,1111,370,123,41,13,4.6,1.5,0.5,0.0 nM.
d) Placing 384-well plate at 37 deg.C and 5% CO2The incubator of (1) was incubated for 16 hours.
Step three: detection of
The light absorption of secreted alkaline phosphatase (SEAP) at 620nm was detected using an instrument Envision. The receptor affinity activity of each test compound and control compound GS-9620 was calculated and is shown in Table 1.
Effect example 2: TLRs in human peripheral blood mononuclear cells7/8Agitation test
The experimental steps are as follows:
the method comprises the following steps: human blood collection
1. 15mL of blood from one donor was collected aseptically.
2.15mL of blood can be tested in duplicate wells for 3 compounds.
Step two: isolation of human peripheral blood mononuclear cells
Isolation of PBMCs should be isolated within 2 hours after blood collection.
2.6 mL of human blood isolate was added to a 15mL sterile centrifuge tube and allowed to equilibrate at room temperature.
3. Slowly adding 5mL of blood adherent into 6mL of separation solution to allow the separation solution to be naturally layered, and avoiding adding the separation solution below the liquid level.
4. Centrifuge at room temperature for 30 minutes at 400 g.
5. After centrifugation, the centrifuge tube was slowly removed and the uppermost plasma layer was carefully removed.
6. After removal of plasma, the buffy coat containing the monocytes was transferred to a new sterile 15mL centrifuge tube.
7. 10mL of PBS was added to the centrifuge tube containing the monocytes and mixed well.
8. Centrifuge at room temperature for 10 minutes at a centrifuge speed of 300 g.
9. The supernatant was discarded, and the cells were resuspended in 10mL PBS and mixed well.
10. Centrifuge at room temperature for 10 minutes at a centrifuge speed of 300 g.
11. Repeat steps 9, 10, discard the supernatant, resuspend cell counts with 1mL of medium RPMI1640+ 10% FBS + 1% PS.
12. The density of the inoculated cells is 1.5X106Per mL, 96-well plates were seeded with 100. mu.L of cell suspension, 15 ten thousand cells per well.
13. The 96-well plate was placed at 37 ℃ in 5% CO2An incubator.
Step three: dilution of test Compounds
1. Test compounds were made up in 10mM stock in DMSO.
2. The test compound was diluted in 3-fold gradient from 10mM to prepare 9 concentrations.
3. Control compound 14 concentrations (3-fold dilution or 1.5-fold dilution from stock).
DMSO negative control.
5. 10-fold intermediate dilutions were made in medium (5 μ L gradient dilution compound to 45 μ L medium).
Step four: treatment of cells with compounds
1. Each well was supplemented with 98. mu.L of medium to make the total volume 198. mu.L.
2.2 μ L of the intermediate diluted compound (step three) was taken to the corresponding 96-well plate, with final DMSO concentration of 0.1% and final test compound concentrations of 10000,3333,1111,370,123,41,13,4.6,1.5, 0.0 nM. Control compounds were used at 10000,3333, 2222, 1481, 987, 658, 219, 146, 98, 65, 43,29, 19, 13, 0nM final concentrations.
Step five: cell incubation
The 96-well plate with the compound added was placed at 37 ℃ in 5% CO2Incubate for 20 hours.
Step six: MSD test detection
1. The 96-well plate was centrifuged at 1500rpm for 5 minutes at room temperature.
2. The supernatant was aspirated for MSD experiments. The results are shown in Table 2.
TNF- α detection procedure in MSD experiments
The method comprises the following steps: preparation of standards and samples
1. Preparing a standard substance: adding 250 mu L of Diluent 2 into the standard dry powder, fully and uniformly mixing, and using after 5 minutes. Then a gradient dilution was performed, 4-fold dilution with 7 gradients, with dilution 2 as zero.
2. Sample preparation: the sample was centrifuged at room temperature for 5 minutes at 1500rpm and the supernatant was aspirated for use.
3. Preparing an antibody: the antibody stock concentration was 50X and the concentration used was 1X. Antibodies were diluted with Diluent 3.
4. Preparation of detection liquid: stock solution was 4X in concentration and 2X in concentration, diluted with distilled water.
5. Preparation of a washing liquid: stock solution was 20X, use concentration 1X, dilute with distilled water.
Step two, using step of detecting TNF- α by using MSD kit
1. Adding a sample: 50 μ L of sample, standard or control, sealed with a membrane seal, incubated for 2 hours in a room temperature with an oscillator at 650 rpm.
2. Washing the plate: wash the plate 3 times with 300 μ L1X wash per well and clean the last wash.
3. Adding an antibody: add 25. mu.L of the prepared antibody to each well, seal the plate with membrane, incubate for 2 hours in room temperature with an oscillator at 650 rpm.
4. Washing the plate: wash the plate 3 times with 300 μ L1X wash per well and clean the last wash.
5. And (3) detection: add 150. mu.L of 2X detection solution to each well and detect using MSD instrument. It should be noted that the assay can be performed without incubation after the addition of the assay solution.
Detection step of IFN- α in MSD experiment
The method comprises the following steps: preparation of standards and samples
1. Preparing a standard substance: the standard stock was 1. mu.g/mL, the effect starting concentration was 2500pg/mL, followed by a gradient dilution, 4-fold dilution, 7 gradients, with Diluent 2 as zero point.
2. Sample preparation: the sample was centrifuged at room temperature for 5 minutes at 1500rpm and the supernatant was aspirated for use.
3. Preparing an antibody: the antibody stock concentration was 50X and the concentration used was 1X. Antibodies were diluted with Diluent 3.
4. Preparation of detection liquid: stock solution was 4X in concentration and 2X in concentration, diluted with distilled water.
5. Preparation of a washing liquid: stock solution was 20X, use concentration 1X, dilute with distilled water.
Step two, using step of detecting IFN- α by MSD kit
1. Adding Diluent 2: add 25. mu.L of Diluent 2 to each well and shake at 650rpm for 30 minutes at room temperature.
2. Adding a sample: add 25 μ L of sample or standard into each well, seal with membrane, incubate for 2 hours in room temperature with oscillator at 650 rpm.
3. Washing the plate: wash the plate 3 times with 300 μ L1X wash per well and clean the last wash.
4. Adding an antibody: add 25. mu.L of the prepared antibody to each well, seal the plate with membrane, incubate for 2 hours in room temperature with an oscillator at 650 rpm.
5. Washing the plate: wash the plate 3 times with 300 μ L1X wash per well and clean the last wash.
6. And (3) detection: add 150. mu.L of 2X detection solution to each well and detect using MSD instrument. It should be noted that the assay can be performed without incubation after the addition of the assay solution.
Table 1 results of receptor affinity activity assay:
Figure BDA0001836596920000681
table 2 hPBMC activity test results:
Figure BDA0001836596920000682
the data in tables 1 and 2 show that the compounds of the invention have high TLRs7Activity and relation to TLRs8High TLR of7And (4) selectivity. Its high activity was also confirmed in the hPBMC assay.
PD experiments of Compounds in mice
The experimental steps are as follows:
a) dosage: 10mg/kg
Test article: GS-9620 (control Compound), Compound 3
b) The test substance was orally administered to three C57 mice by gavage. After administration, blood was collected via the tail vein at 1,2 and 4 hours, respectively. Whole blood is collected in a pre-filled container2Plasma was obtained in EDTA tubes and centrifuged and stored in a-80 ℃ refrigerator.
c) Plasma samples were lysed and analyzed for IFN- α and TNF- α.
Analysis of IFN- α and TNF- α IFN- α and TNF- α concentrations were tested in accordance with the MSD assay described above for IFN- α and TNF- α.
d) Quantitative analysis of plasma and liver compounds
LC-MS/MS device
High Pressure Liquid Chromatography (HPLC), Prominence (Degasser DGU-20A 5R); liquidchromagraph LC-30 AD; communications Bus Module CBM-20A, Auto Sampler SIL-30 AC; rack changer II.
Chromatography column Eclipse XDB-Phenyl 2.1X 50mm 3.5 μm.
Mass Spectrometer (MS): AB Sciex Triple quant 5500 LC/MS/MS.
HPLC conditions:
mobile phase
Solution A5% acetonitrile Water (0.1% formic acid)
Solution B95% acetonitrile Water (0.1% formic acid)
Plasma sample preparation:
1) preparing a standard substance: mu.L of working solution (containing 1,2,4,10,20,100,200,1000,2000ng/mL of compound respectively) was added to 10. mu.L of blank C57 mouse plasma or liver homogenate to obtain 0.5-1000 ng/mL (0.5,1,2,5,10,50,100,500,1000ng/mL) standard in a total volume of 15. mu.L.
2) Quality control sample (QC): 5 plasma quality control samples with the concentration of 1.5ng/mL,3ng/mL,6ng/mL,50ng/mL and 800ng/mL are independently prepared.
3) Test sample 5. mu.L of white liquor + 10. mu.L of test plasma sample.
Preparing a liver sample:
liver samples (grams) were homogenized in pbs (ml) at a ratio of 1: 4. Subsequently, 5. mu.L of the blank solution was added to 10. mu.L of the liver sample after homogenization to obtain a liver sample.
Preparing a supernatant fluid:
mu.L of the standard sample, 15. mu.L of the QC sample, and 15. mu.L of the unknown sample were added to 200. mu.L of IS-containing acetonitrile, respectively, for precipitation of proteins. The sample was then vortex mixed for 30 minutes and then spun centrifugally at 4 degrees over a 15 minute period. 10 μ L of the supernatant was taken for LC/MS/MS quantitative analysis.
Results and conclusions:
the exposure of the compound and the expression levels of IFN- α and TNF- α are shown in figure 1. As can be seen from figure 1, the exposure of the compound 3 in the plasma and liver of mice is lower than the lower limit of the LC/MS/MS analysis quantification, however, the compound 3 can still induce the expression of IFN- α in the plasma and liver, which indicates that the compound 3 can greatly reduce the toxic and side effects caused by the compound and metabolites thereof relative to GS-9620.

Claims (21)

1. A compound of formula (I), or a pharmaceutically acceptable salt, stereoisomer, solvate, polymorph tautomer, isotopic compound, metabolite or prodrug thereof:
Figure FDA0001836596910000011
wherein:
x is selected from O, S and NRx;RxSelected from H, C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with 1,2, 3 or 4RASubstitution;
y is selected from O, S, N, or Y is absent;
z is selected from N, O, S, C and CH;
q is selected from O, S and NH;
when Y is N, R1Selected from H, hydroxyl, sulfydryl, amino, halogen, nitro, cyano, C1-C8Alkyl radical, C2-C8Alkenyl radical, C2-C8Alkynyl, C6-C10Aryl and five to ten membered heteroaryl; wherein said alkyl, alkenyl, alkynyl, aryl and heteroaryl are optionally substituted with 1,2, 3 or 4RBSubstitution;
R2selected from H, hydroxyl, sulfydryl, amino, halogen, nitro, cyano, C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with 1,2, 3 or 4RCSubstitution;
L1is C1-C8A hydrocarbon chain;
ring a is absent, or ring a is selected from: c3-C10Cycloalkyl radical, C3-C10Heterocyclic group, C6-C10Aryl and five to ten membered heteroaryl; wherein said cycloalkyl, heterocyclyl, aryl and heteroaryl are optionally substituted with 1,2, 3 or 4RDSubstitution;
L2is absent, or L2Is C1-C8A hydrocarbon chain;
R3is H, C3-C10Cycloalkyl radical, C6-C10Aryl, five-to ten-membered heteroarylor-WR6R7Wherein W is N or CH, wherein
R6And R7Each independently selected from H, C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with 1,2, 3 or 4RESubstitution; or
R6And R7Together with the N or C atom to which they are attached form a three-to ten-membered heterocyclic ring; said heterocycle being optionally substituted by 1,2, 3 or 4RESubstitution; and optionally, R6And R7The three-to ten-membered heterocyclic ring formed together with the N or C atom to which it is attached may optionally be fused to the a ring;
R4is selected from C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group; wherein said alkyl, alkenyl or and alkynyl are each optionally substituted with 1,2, 3 or 4RFSubstitution;
ring B is absent, or ring B is selected from C3-C10Cycloalkyl radical, C3-C10Heterocyclic group, C6-C10Aryl and five to ten membered heteroaryl; wherein said cycloalkyl, heterocyclyl, aryl and heteroaryl are each optionally substituted with 1,2, 3 or 4RGSubstitution;
R5selected from H, C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with 1,2, 3 OR 4 substituents selected from-OR8、-SR8and-NR8R8' is substituted with a substituent;
R8and R8' each is independently selected from H, C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with 1,2, 3 or 4RHSubstitution; or R8And R8' together with the N atom to which they are attached form a three-to ten-membered heterocyclic ring;
RA、RB、RC、RD、RE、RF、RG、RHeach occurrence is independently selected from halogen, cyano, nitro, -Ra、-ORa、=O、-SRa、-NRaRb、=NRa-C (halogen)3-CR (halogen)2、-CR2(halogen), -OCN, -SCN, -N ═ C ═ O, -NCS, -NO2、-N(Ra)C(=O)Rb、-N(Ra)C(=O)ORb、-N(Ra)C(=O)NRbRc、-C(=O)NRaRb、-C(=O)ORa、-OC(=O)NRaRb、-OC(=O)Ra、-OC(=O)ORa、-C(=O)Ra、-S(=O)2ORa、-S(=O)2Ra、-OS(=O)2Ra、-OS(=O)2ORa、-S(=O)2NRaRb、-S(=O)Ra、-N(Ra)S(=O)2Rb、-N(Ra)S(=O)2NRbRc、-N(Ra)S(=O)2ORb、-OP(=O)(ORa)ORb、-P(=O)(ORa)ORb、-C(=O)Ra、-C(=S)Ra、-C(=O)ORa、-C(=S)ORa、-C(=O)SRa、-C(=S)SRa、-C(=O)NRaRb、-C(=S)NRaRb、-C(=NRa)NRbRcand-NRaC(=NRb)NRcRd;Ra、Rb、RcAnd RdEach occurrence is independently selected from H, C1-C8Alkyl radical, C3-C8Cycloalkyl, a three-to eight-membered heterocyclic group, C6-C10Aryl, five-to ten-membered heteroaryl, C6-C10aryl-C1-C4Alkyl and five-to ten-membered heteroaryl-C1-C4An alkyl group; wherein said alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, aryl-C1-C4Alkyl and heteroarylradical-C1-C4Each alkyl group is optionally substituted by 1,2, 3 or 4 halogen, ═ O, -OH, -NH2-SH substitution;
wherein when R isA、RB、RC、RD、RE、RF、RGOr RHEach independently is-NRaRbWhen R isa、RbOptionally together with the N atom to which they are attached form a three-to ten-membered heterocyclic ring.
2. The compound of claim 1, or a pharmaceutically acceptable salt, stereoisomer, solvate, polymorph tautomer, isotopic compound, metabolite or prodrug thereof, wherein X is O or S, preferably O.
3. The compound of any one of claims 1-2, or a pharmaceutically acceptable salt, stereoisomer, solvate, polymorph tautomer, isotopic compound, metabolite, or prodrug thereof, wherein Y and R1Is absent, or
Y is N, and R1Selected from H, C1-C8Alkyl radical, C2-C8Alkenyl radical, C2-C8Alkynyl, C6-C10Aryl and five to ten membered heteroaryl; wherein said alkyl, alkenyl, alkynyl, aryl and heteroaryl are each optionally substituted with 1,2, 3 or 4RBSubstitution; wherein the five-to ten-membered heteroaryl contains 1-3 heteroatoms each independently selected from O, N and S; preferably, Y is N, and R1Is H or C1-C4An alkyl group; preferably H or C1-C3An alkyl group; further preferred is C1-C2An alkyl group; wherein said alkyl is optionally substituted with 1,2, 3 or 4RBSubstitution; more preferably, Y is N, and R1Is H or C1-C2An alkyl group; wherein said alkyl is optionally substituted with 1,2, 3 or 4 halogens; wherein R is1Further preferably H, CH3、CF3Or CH2CF3Particularly preferably H or CH3
4. The compound of any one of claims 1-3, or a pharmaceutically acceptable salt, stereoisomer, solvate, polymorph tautomer, isotopic compound, metabolite, or prodrug thereof, wherein Z is N, and R is2Selected from H, hydroxy, mercapto, amino, C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with 1,2, 3 or 4RCSubstitution; preferably, Z is N, and R2Selected from H, hydroxy, mercapto, amino, C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with 1,2, 3 or 4RCSubstitution; more preferably, Z is N, and R2Is H, hydroxy or methyl, wherein the methyl is optionally substituted with 1,2 or 3 halogens; wherein R is2Further preferably H, hydroxy or methyl, particularly preferably H or hydroxy; or
Z is CH, and R2Selected from H, hydroxyl, sulfydryl, amino, halogen, nitro, cyano, C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group; wherein alkyl, alkenyl and alkynyl are each optionally substituted with 1,2, 3 or 4RCSubstitution; r2Preferably H, hydroxy, mercapto, halogen or C1-C4An alkyl group; preferably H, hydroxy, mercapto, halogen or C1-C3An alkyl group; wherein said alkyl is optionally substituted with 1,2, 3 or 4RCSubstitution; r2More preferably H, hydroxy, halogen or methyl, wherein the methyl is optionally substituted with 1,2 or 3 halogens; more preferably, R2H, F, hydroxy or methyl, particularly preferably H or hydroxy.
5. The compound of any one of claims 1-4, or a pharmaceutically acceptable salt, stereoisomer, solvate, polymorph tautomer, isotopic compound, metabolite, or prodrug thereof, wherein L1Is- (CH)2)n-; wherein n is 1,2, 3,4, 5,6, 7 or 8; preferably 1,2, 3,4 or 5, more preferably 1,2 or 3; particularly preferably 1.
6. The compound of any one of claims 1-5, or a pharmaceutically acceptable salt, stereoisomer, solvate, polymorph tautomer, isotopic compound, metabolite, or prodrug thereof, wherein ring a is selected from the group consisting of: a phenyl group; pyridine; furan; thiophene; pyrrole; cyclohexane; cyclopentane; cyclobutane; cyclopropane; bicyclo [2.2.1]Heptane, with 1-carbon and 4-carbon atoms attached to the rest of the molecule; bicyclo [1.1.1]Pentane, attached to the rest of the molecule at 1-carbon and 3-carbon atoms; 7-Oxabicyclo [2.2.1]Heptane, with 1-carbon and 4-carbon atoms attached to the rest of the molecule; wherein the phenyl, pyridine, furan, thiophene, pyrrole, cyclohexane, cyclopentane, cyclobutane, cyclopropane, bicyclo [2.2.1]Heptane, bicyclo [1.1.1]Pentane and 7-oxabicyclo [2.2.1 ]]Each heptane is optionally substituted with 1,2, 3 or 4RDSubstitution; preferably, the a ring is selected from phenyl and pyridine; and are each optionally substituted by 1,2, 3 or 4RDSubstitution; wherein R isDHalogen is preferred, and F is particularly preferred.
7. The compound of any one of claims 1-6, or a pharmaceutically acceptable salt, stereoisomer, solvate, polymorph tautomer, isotopic compound, metabolite, or prodrug thereof, wherein L2Is selected from- (CH)2)n-; wherein n is 1,2, 3,4 or 5,6, 7 or 8; preferably 1,2, 3,4 or 5, more preferably 1,2 or 3; particularly preferably 1.
8. The compound of any one of claims 1-7, or a pharmaceutically acceptable salt, stereoisomer, solvate, polymorph tautomer, isotopic compound, metabolite, or prodrug thereof, wherein R is3is-NR6R7Optionally substituted by 1,2, 3 or 4RESubstitution; preferably, R3Is selected from-N (C)1-C4Alkyl) (C1-C4Alkyl), pyrrolidine, piperidine, morpholine, and piperazine, wherein the alkyl, pyrrolidine, piperidine, morpholine, and piperazine are each optionally substituted with 1,2, 3, or 4RESubstitution; more preferably, R3Is selected from-N (CH)3)2Pyrrolidin-1-yl, piperidin-1-yl, morpholin-1-yl and 4-C1-C3Alkyl-piperazin-1-yl; wherein said CH3Pyrrolidin-1-yl, piperidin-1-yl, morpholin-1-yl and piperazin-1-yl are each optionally substituted with 1,2, 3 or 4RESubstitution; further preferably, R3Selected from pyrrolidin-1-yl and piperidin-1-yl; wherein said pyrrolidin-1-yl and piperidin-1-yl are each optionally substituted with 1,2, 3 or 4REAnd (4) substitution.
9. The compound of any one of claims 1-8, or a pharmaceutically acceptable salt, stereoisomer, solvate, polymorph tautomer, isotopic compound, metabolite, or prodrug thereof, wherein R is4Is C1-C4Alkyl, wherein said alkyl is optionally substituted with 1,2, 3 or 4RFSubstitution; preferably, R4Is C1-C3Alkyl, especially C3An alkyl group; wherein said alkyl is optionally substituted with 1,2, 3 or 4RFSubstitution; more preferably, R4is-CH2-、-CH(CH3)-、-CH(C2H5)-、-CH(C3H7-n)-、-CH(CH2ORa)-、-CH(CH2SRa)-、-C(CH2ORa)(Rb)-、-C(CH2ORa)(ORb)-、-CH(C(=O)ORa)-、-CH(S(=O)Ra)-、-CH(S(=O)2Ra)-、-CH(S(=O)2ORa)-、-CH(OC(=O)Ra)-、-CH(OS(=O)2Ra)-、-C(C(=O)ORa)(Rb)-、-C(S(=O)Ra)(Rb)-、-C(S(=O)2Ra)(Rb)-、-C(S(=O)2ORa)(Rb)-、-C(OC(=O)Ra)(Rb)-、-C(OS(=O)2Ra)(Rb)-、-C(C(=O)ORa)(ORb)-、-C(S(=O)Ra)(ORb)-、-C(S(=O)2Ra)(ORb)-、-C(S(=O)2ORa)(ORb)-、-CH(CH2CH2ORa)-、-CH(CH2CH2SRa)-、-C(CH2CH2ORa)(Rb)-、-C(CH2CH2ORa)(ORb)-、-CH(CH2C(=O)ORa)-、-CH(CH2S(=O)Ra)-、-CH(CH2S(=O)2Ra)-、-CH(CH2S(=O)2ORa)-、-CH(CH2OC(=O)Ra)-、-CH(CH2OS(=O)2Ra)-、-C(CH2C(=O)ORa)(Rb)-、-C(CH2S(=O)Ra)(Rb)-、-C(CH2S(=O)2Ra)(Rb)-、-C(CH2S(=O)2ORa)(Rb)-、-C(CH2OC(=O)Ra)(Rb)-、-C(CH2OS(=O)2Ra)(Rb)-、-C(CH2C(=O)ORa)(ORb)-、-C(CH2S(=O)Ra)(ORb)-、-C(CH2S(=O)2Ra)(ORb)-、-C(CH2S(=O)2ORa)(ORb)-、-C(CH2OC(=O)Ra)(ORb)-、-C(CH2OS(=O)2Ra)(ORb)-、-CH(CH2CH2S(=O)Ra)-、-CH(CH2CH2S(=O)2Ra)-、-CH(CH2CH2S(=O)2ORa)-、-CH(CH2CH2OC(=O)Ra)-、-CH(CH2CH2OS(=O)2Ra)-、-C(CH2CH2S(=O)Ra)(Rb)-、-C(CH2CH2S(=O)2Ra)(Rb)-、-C(CH2CH2S(=O)2ORa)(Rb)-、-C(CH2CH2OC(=O)Ra)(Rb)-、-C(CH2CH2OS(=O)2Ra)(Rb)-、-C(CH2CH2S(=O)Ra)(ORb)-、-C(CH2CH2S(=O)2Ra)(ORb)-、-C(CH2CH2S(=O)2ORa)(ORb)-、-C(CH2CH2OC(=O)Ra)(ORb)-、-C(CH2CH2OS(=O)2Ra)(ORb)-、-CH(CH2NRaRb)-、-C(CH2NRaRb)(Rc)-、-C(CH2NRaRb)(ORc)-、-CH(CH2CH2NRaRb)-、-C(CH2CH2NRaRb)(Rc)-、-C(CH2CH2NRaRb)(ORc)-、-CH(CH2C(=O)NRaRb)-、-CH(CH2S(=O)2NRaRb)-、-C(CH2C(=O)NRaRb)(Rc)-、-C(CH2S(=O)2NRaRb)(Rc)-、-C(CH2C(=O)NRaRb)(ORc)-、-C(CH2S(=O)2NRaRb)(ORc)-、-CH(CH2N(Ra)C(=O)Rb)-、-CH(CH2N(Ra)S(=O)2Rb)-、-CH(CH2CH2S(=O)2NRaRb)-、-CH(CH2CH2N(Ra)C(=O)Rb)-、-CH(CH2CH2N(Ra)S(=O)2Rb)-、-CH(CH2N(Ra)C(=O)Rb)(Rc)-、-CH(CH2N(Ra)S(=O)2Rb)(Rc)-、-CH(CH2CH2S(=O)2NRaRb)(Rc)-、-CH(CH2CH2N(Ra)C(=O)Rb)(Rc)-、-CH(CH2CH2N(Ra)S(=O)2Rb)(Rc)-、-CH(CH2N(Ra)C(=O)Rb)(ORc)-、-CH(CH2N(Ra)S(=O)2Rb)(ORc)-、-CH(CH2CH2S(=O)2NRaRb)(ORc)-、-CH(CH2CH2N(Ra)C(=O)Rb)(ORc) -or-CH (CH)2CH2N(Ra)S(=O)2Rb)(ORc) -; more preferably-CH (C)3H7-n)-、-CH(CH2ORa)-、-CH(CH2SRa)-、-CH(CH2CH2ORa)-、-CH(CH2CH2SRa)-、-CH(CH2C(=O)ORa)-、-CH(CH2S(=O)2Ra)-、-CH(CH2S(=O)2ORa)-、-CH(CH2OC(=O)Ra)-、-CH(CH2OS(=O)2Ra)-、-CH(CH2CH2OS(=O)2Ra)-、-CH(CH2CH2OC(=O)Ra)-、-CH(CH2CH2N(Ra)S(=O)2Rb)-、-CH(CH2NRaRb)-、-CH(CH2CH2NRaRb)-、-CH(CH2CH2N(Ra)C(=O)Rb)-、-CH(CH2C(=O)NRaRb) -or-CH (CH)2S(=O)2NRaRb) -; further preferred is-CH (C)3H7-n)-、-CH(CH2CH2ORa)-、-CH(CH2CH2SRa)-、-CH(CH2C(=O)ORa)-、-CH(CH2S(=O)2Ra)-、-CH(CH2S(=O)2ORa)-、-CH(CH2OC(=O)Ra)-、-CH(CH2OS(=O)2Ra)-、-CH(CH2CH2OS(=O)2Ra)-、-CH(CH2CH2OC(=O)Ra)-、-CH(CH2CH2N(Ra)S(=O)2Rb)-、-CH(CH2CH2NRaRb)-、-CH(CH2CH2N(Ra)C(=O)Rb)-、-CH(CH2C(=O)NRaRb) -or-CH (CH)2S(=O)2NRaRb) -; particularly preferred is-CH (CH)2CH2ORa)-、-CH(CH2CH2OS(=O)2Ra)-、-CH(CH2CH2OC(=O)Ra)-、-CH(CH2CH2N(Ra)S(=O)2Rb)-、-CH(CH2C(=O)ORa)-、-CH(CH2S(=O)2ORa)-、-CH(CH2CH2NRaRb)-、-CH(CH2CH2N(Ra)C(=O)Rb)-、-CH(CH2C(=O)NRaRb) -or-CH (CH)2S(=O)2NRaRb)-。
10. The compound of any one of claims 1-9, or a pharmaceutically acceptable salt, stereoisomer, solvate, polymorph tautomer, isotopic compound, metabolite, or prodrug thereof, wherein ring B is absent; or
Ring B is selected from phenyl, pyridine, furan, thiophene, pyrrole, thiazole, oxazole, pyran, pyrrolidine, piperidine, tetrahydrofuran, dihydropyran, tetrahydropyran, propylene oxide, cyclohexane, cyclopentane, cyclobutane, and cyclopropane, and each is optionally substituted with 1,2, 3, or 4RGSubstitution; preferably, ring B is phenyl or pyridine; and are each optionally substituted by 1,2, 3 or 4 radicals each independently selected from halogen, cyano, nitro, -Ra、-ORa、-SRaand-NRaRbIs substituted with a group (b).
11. Claim 1A compound of any one of-10, or a pharmaceutically acceptable salt, stereoisomer, solvate, polymorph tautomer, isotopic compound, metabolite or prodrug thereof, wherein R is5Selected from H and C3-C6Alkyl, wherein the alkyl is optionally substituted by 1,2, 3 OR 4-OR8、-SR8or-NR8R8' substitution; wherein
R8And R8' each is independently selected from H and C1-C4An alkyl group; preferably C1-C2An alkyl group; wherein said alkyl is optionally substituted with 1,2, 3 or 4RHSubstitution;
or R8And R8' connected to, -NR8R8' formation of a four-to eight-membered heterocyclic ring; wherein said heterocycle contains 0-1 additional heteroatoms independently selected from O, S and N, and is optionally substituted with 1,2, 3, or 4RHSubstitution; preferably, -NR8R8' formation of a four-to six-membered heterocyclic ring; wherein said heterocycle contains 0-1 additional heteroatoms independently selected from O, S and N, and is optionally substituted with 1,2, 3, or 4RHAnd (4) substitution.
12. A compound of formula (I-1), formula (I-2) or formula (I-3), or a pharmaceutically acceptable salt, stereoisomer, solvate, polymorph tautomer, isotopic compound, metabolite or prodrug thereof:
Figure FDA0001836596910000061
wherein, X, Q, R1、R2、L1、A、L2、R3、R4、RF、B、R5As defined in any one of claims 1 to 11.
13. A compound of formula (I-4), or a pharmaceutically acceptable salt, stereoisomer, solvate, polymorph tautomer, isotopic compound, metabolite or prodrug thereof:
Figure FDA0001836596910000062
wherein, X, Q, L1、A、L2、R3、R4、RF、B、R5As defined in any one of claims 1 to 11;
R2selected from H, hydroxy, mercapto, amino, C1-C8Alkyl radical, C2-C8Alkenyl and C2-C8An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with one or more RCSubstitution;
preferably, R2Selected from hydroxy, mercapto, amino, C1-C4Alkyl radical, C2-C4Alkenyl and C2-C4An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with one or more RCSubstitution;
more preferably, R2Selected from hydroxy, mercapto, C1-C4Alkyl radical, C3-C4Alkenyl and C3-C4An alkynyl group; wherein said alkyl, alkenyl and alkynyl are each optionally substituted with one or more RCSubstitution;
further preferably, R2Is hydroxy, mercapto or C1-C4An alkyl group; preferably hydroxy, mercapto or C1-C3An alkyl group; wherein said alkyl is optionally substituted with one or more RCSubstitution;
more preferably, R2Is hydroxy or methyl; wherein said methyl is optionally substituted with one or more halo;
further preferably, R2Is hydroxy or methyl; particularly preferred is a hydroxyl group.
14. A compound of formula (I-5), formula (I-6) or formula (I-7), or a pharmaceutically acceptable salt, stereoisomer, solvate, polymorph tautomer, isotopic compound, metabolite or prodrug thereof:
Figure FDA0001836596910000071
wherein, X, Q, L1、A、L2、R3、R4、RF、B、R5As defined in any one of claims 1 to 11.
15. The compound of any one of claims 1-14, or a pharmaceutically acceptable salt, stereoisomer, solvate, polymorph tautomer, isotopic compound, metabolite, or prodrug thereof, wherein the compound is selected from the group consisting of:
Figure FDA0001836596910000081
Figure FDA0001836596910000091
16. a pharmaceutical composition comprising a compound of any one of claims 1-15, or a pharmaceutically acceptable salt, stereoisomer, solvate, polymorph, tautomer, isotopic compound, metabolite, or prodrug thereof, and at least one pharmaceutically acceptable carrier.
17. Use of a compound of any one of claims 1-15, or a pharmaceutically acceptable salt, stereoisomer, solvate, polymorph, tautomer, isotopic compound, metabolite, or prodrug thereof, or a pharmaceutical composition of claim 16 for the preparation of a medicament for treating a TLR7Use in the manufacture of a medicament for a disease responsive to activation of a receptor.
18. Use of a compound of any one of claims 1 to 15, or a pharmaceutically acceptable salt, stereoisomer, solvate, polymorph, tautomer, isotopic compound, metabolite or prodrug thereof, or a pharmaceutical composition of claim 16, for the manufacture of a medicament for the treatment of a liver-related disease selected from the group consisting of viral hepatitis, autoimmune liver disease, drug-toxic liver disease, hepatic injury from the liver, hepatic failure from the liver, chronic severe hepatitis, cirrhosis, liver abscess, fatty liver, primary liver cancer, preferably the liver-related disease is hepatitis b and hepatitis c.
19. Use of a compound of any one of claims 1-15, or a pharmaceutically acceptable salt, stereoisomer, solvate, polymorph, tautomer, isotopic compound, metabolite, or prodrug thereof, or a pharmaceutical composition of claim 16 in the manufacture of a medicament for the treatment of a tumor.
20. A pharmaceutical combination comprising
(1) A compound of any one of claims 1-15 or a pharmaceutically acceptable salt, stereoisomer, solvate, polymorph, tautomer, isotopic compound, metabolite or prodrug thereof, or a pharmaceutical composition of claim 16, and
(2) a PD-1 antibody, a PD-L1 antibody or a PD-1 inhibitor, a PD-L1 inhibitor or a PD-1/PD-L1 inhibitor.
21. Use of a compound according to any one of claims 1 to 15, or a pharmaceutically acceptable salt, stereoisomer, solvate, polymorph, tautomer, isotopic compound, metabolite or prodrug thereof, or a pharmaceutical composition according to claim 16, for the manufacture of a medicament for the treatment of an HIV infection, preferably said HIV infection is aids.
CN201811228693.4A 2018-10-22 2018-10-22 Five-membered or six-membered heterocyclic pyrimidine compound and application thereof Pending CN111072667A (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CN201811228693.4A CN111072667A (en) 2018-10-22 2018-10-22 Five-membered or six-membered heterocyclic pyrimidine compound and application thereof
PCT/CN2019/111625 WO2020083089A1 (en) 2018-10-22 2019-10-17 Compound of 5- or 6-membered heterocyclic pyrimidines and use thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201811228693.4A CN111072667A (en) 2018-10-22 2018-10-22 Five-membered or six-membered heterocyclic pyrimidine compound and application thereof

Publications (1)

Publication Number Publication Date
CN111072667A true CN111072667A (en) 2020-04-28

Family

ID=70309672

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201811228693.4A Pending CN111072667A (en) 2018-10-22 2018-10-22 Five-membered or six-membered heterocyclic pyrimidine compound and application thereof

Country Status (2)

Country Link
CN (1) CN111072667A (en)
WO (1) WO2020083089A1 (en)

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007034817A1 (en) * 2005-09-22 2007-03-29 Dainippon Sumitomo Pharma Co., Ltd. Novel adenine compound
US20090105212A1 (en) * 2005-09-22 2009-04-23 Dainippon Sumitomo Pharma Co., Ltd. a corporation of Japan Novel adenine compound
PE20081887A1 (en) * 2007-03-20 2009-01-16 Dainippon Sumitomo Pharma Co NEW ADENINE COMPOUND
TWI434849B (en) * 2007-06-29 2014-04-21 Gilead Sciences Inc Modulators of toll-like receptor 7
KR101787309B1 (en) * 2008-12-09 2017-10-18 길리애드 사이언시즈, 인코포레이티드 Modulators of toll-like receptors
NZ598933A (en) * 2009-10-22 2013-04-26 Gilead Sciences Inc Derivatives of purine or deazapurine useful for the treatment of (inter alia) viral infections
MY182353A (en) * 2015-05-08 2021-01-20 Hoffmann La Roche Novel sulfonimidoylpurinone compounds and derivatives for the treatment and prophylaxis of virus infection

Also Published As

Publication number Publication date
WO2020083089A1 (en) 2020-04-30

Similar Documents

Publication Publication Date Title
AU2017204007B2 (en) Heteroaryl compounds and methods of use thereof
CN107849040B (en) Tricyclic derivative compound, preparation method thereof, and pharmaceutical composition containing the same
CN107759587B (en) [1,2,4] triazolo [1,5-a ] pyridine compound and preparation method and medical application thereof
IL293962A (en) Kras mutant protein inhibitors
DE60315615T2 (en) TRICYCLIC COMPOUNDS BASED ON THIOPHES AND MEDICAMENTS COMPRISING THEM
JP2022550436A (en) antiviral heterocyclic compound
WO2019120234A2 (en) Compound functioning as bromodomain protein inhibitor, and composition
EP3318561A1 (en) Heteroaryl compounds and methods of use thereof
JP6786566B2 (en) Heteroaryl compounds and how to use them
JP6302480B2 (en) Triazolo-pyrazine derivatives useful for the treatment of central nervous system diseases
TW202246212A (en) Novel compounds useful as sting agonists and uses thereof
EP3534888B1 (en) Substituted bicyclic heteroaryl allosteric modulators of nicotinic acetylcholine receptors
CN111072667A (en) Five-membered or six-membered heterocyclic pyrimidine compound and application thereof
JP2011504878A (en) Method for producing imidazoazepinone compound
WO2019196918A1 (en) Five-membered heterocyclo-pyrimidine compound, pharmaceutical composition and use thereof
WO2019079649A1 (en) Substituted pyrrolopyridines as inhibitors of activin receptor-like kinase
WO2023220247A1 (en) Lrrk2 inhibitors
EP3720860A1 (en) Positive allosteric modulators of the muscarinic acetylcholine receptor m4

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
WD01 Invention patent application deemed withdrawn after publication
WD01 Invention patent application deemed withdrawn after publication

Application publication date: 20200428