CN117940437A - DNA-PK selective inhibitor and preparation method and application thereof - Google Patents

DNA-PK selective inhibitor and preparation method and application thereof Download PDF

Info

Publication number
CN117940437A
CN117940437A CN202280060488.XA CN202280060488A CN117940437A CN 117940437 A CN117940437 A CN 117940437A CN 202280060488 A CN202280060488 A CN 202280060488A CN 117940437 A CN117940437 A CN 117940437A
Authority
CN
China
Prior art keywords
alkyl
membered
methyl
dihydro
halogen
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202280060488.XA
Other languages
Chinese (zh)
Inventor
陈坤成
张凯
任仁
刘志华
陈曦
雷永珂
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.)
Capital Pharmaceutical Holdings Beijing Co ltd
Original Assignee
Capital Pharmaceutical Holdings Beijing 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 Capital Pharmaceutical Holdings Beijing Co ltd filed Critical Capital Pharmaceutical Holdings Beijing Co ltd
Publication of CN117940437A publication Critical patent/CN117940437A/en
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D473/00Heterocyclic compounds containing purine ring systems
    • C07D473/26Heterocyclic compounds containing purine ring systems with an oxygen, sulphur, or nitrogen atom directly attached in position 2 or 6, but not in both
    • C07D473/32Nitrogen atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/22Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains four or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Abstract

The application relates to a DNA-PK selective inhibitor shown in a formula (II), a preparation method and application thereof. The use includes the use of a compound of formula (II) in the manufacture of a medicament for the treatment of a DNA-PK related disorder. In the preparation process, the compound of the application is obtained through a series of reactions such as substitution, reduction, cyclization, alkylation and the like.

Description

DNA-PK selective inhibitor and preparation method and application thereof
Cross reference
The present application claims priority from chinese patent application No. 202111046420.X, chinese patent application No. 202111267966.8, chinese patent application No. 2022, chinese patent application No. 202210029171.1, chinese patent application No. 2022, chinese patent application No. 3, chinese patent application No. 202210315734.3, the disclosure of which is incorporated herein by reference in its entirety.
Technical Field
The present invention relates to compounds that selectively inhibit the activity of DNA-PK proteins, to methods of preparing such compounds and salts thereof, and to methods of treating DNA-PK mediated diseases, including cancer, using such compounds and salts.
Background
A DNA-dependent protein kinase (DNA-PK) is a serine, threonine protein kinase that is activated when bound to DNA. DNA-PK is a trimer of heterodimers consisting of one catalytic subunit DNA-PKcs (size 470 kD) and 2 regulatory subunits Ku70, ku80 polymerized after exposure to the cleaved DNA. Among them, DNA-PKcs are one of members of the phosphatidylinositol-3 kinase family, involved in various biochemical processes including repair of DNA breaks into double strands (double strand breaks, DSBs), signaling of programmed death, gene monitoring, maintenance of telomere structure, etc. (FASEB J.,2005,19 (7): 704-715). The radioactive rays and a plurality of anticancer drugs can directly or indirectly act on DNA or DNA metabolic processes, thereby causing DNA damage, initiating a series of cell reactions such as damaged DNA repair and the like, and the repair result is to improve the survival of cells, which is one of the mechanisms of resisting the tumor cells to radiotherapy and chemotherapy. The sensitivity of cells to chemoradiotherapy can be improved by inhibiting the repair of these DNA lesions (int. J. Hyperthermia,2008,24 (1): 17-29). In DNA damage, DNA double strand breaks (DNA double strand break, DSB) are the most deadly, while repair of DSB is mainly by DNA-dependent protein kinase DNA-PK dominated DNA non-homologous end joining (nonhomologous end joining, NHEJ) (Cell res.,2008,18 (1): 114-124). In addition to playing a major role in the repair of DSBs, DNA-PK functions in other ways:
1) The V (D) J chain rearrangements of immunoglobulins and T cell receptors, such as deletion of DNA-PKcs or Ku proteins, the mammalian cells can exhibit severe combined immunodeficiency (severe combined immunodeficiency, SCID);
2) Maintenance of stable telomere structure, lack of Ku or DNA-PKcs can lead to genomic instability, cell growth retardation and premature senescence;
3) DNA-PKcs is a serine/threonine kinase, a member of the PI-3-K (Phospha tidy lino sito l-3-kinase) kinase family (which also includes ATM, ATR, etc.), and plays a role in cell signaling and cell cycle functions after DNA damage (int. J. Radiation. Oncol. Biol. Phys.,2005,61 (3): 915-921).
A number of factors can induce DNA double strand breaks including chemotherapy, radiation therapy and PARP inhibitors such as olaparib. DNA-PK inhibitors are likely to aid in these therapies. DNA-PK inhibitors can also be used as effective monotherapy, especially for endogenous DNA damage where other DNA repair pathways are deleted in tumor cells. At present, a plurality of DNA-PK selective inhibitors enter a clinical stage, wherein two medicaments enter a clinical second stage, but related medicaments are not marketed so far, and the requirements of the related medicaments are not met. The DNA-PK selective inhibitor provided by the invention has high activity, strong drug resistance and small clinical side effect, can effectively enhance the sensitivity of radiotherapy and chemotherapy in tumor treatment, and has good economic value and application prospect.
Disclosure of Invention
The invention provides a DNA-PK selective inhibitor which is a compound shown in a general formula (II) or pharmaceutically acceptable salt, solvate, polymorph or isomer thereof. The invention also provides a series of compounds represented by the general formula (II), pharmaceutically acceptable salts, solvates, polymorphs, or isomers thereof, pharmaceutical compositions containing these compounds, and methods of treating diseases using such compounds.
In one aspect, the present invention provides a compound of formula (II) or a pharmaceutically acceptable salt, solvate, polymorph or isomer thereof,
Wherein,
Ring A is a 6-10 membered aryl or a 5-12 membered heteroaryl,
Ring B is a 3-12 membered carbocycle or a 4-12 membered heterocycle, C and S on ring B may optionally be oxidized,
Z is-N (R) -, O or S,
Y is N or CR 20, and the total number of the catalyst is N,
R 20 is H, halogen, or C 1-6 alkyl,
X 2 is CR 2 or N,
X 1 is CRR 4, O, S, or NR 6,
R 1 is H, C 1-6 alkyl, C 3-8 cycloalkyl, or 3-8 membered heterocycloalkyl,
R 7 and R 8 are each independently selected from halogen, CN, -OH, -NH 2、C 1-6 alkyl, 3-8 membered cycloalkyl, 3-8 membered heterocycloalkyl, -O-C 1-6 alkyl and-NR-C 1-6 alkyl,
M and n are each independently 0, 1, 2, or 3,
R 3 is R 5 or-X 3-R 5,
R 4 is R 6 or-X 3-R 6,
X 3 is each independently-O-, -S-, or-NR-,
R 5 and R 6 are each independently selected from H, halogen, -CN, -OH, -NH 2、C 1-6 alkyl, 3-8 membered cycloalkyl, 3-8 membered heterocycloalkyl, 5-12 membered heteroaryl, -O-C 1-6 alkyl, or-NR-C 1-6 alkyl, which alkyl, cycloalkyl, heterocycloalkyl, or heteroaryl may be optionally substituted with halogen, -CN, -OH, -NH 2、C 1-6 alkyl, 3-8 membered cycloalkyl, 3-8 membered heterocycloalkyl, 5-12 membered heteroaryl, -O-C 1-6 alkyl, or-NR-C 1-6 alkyl, or
R 5 and R 6 are joined to form- (CH 2) p-X-(CH 2) q -, wherein X is a bond, -CR=CR-, -CO-CR=CR-, -C≡C-, -CO-C≡C-, -O-, -S (O) 2-、-S(O) 2 NR- -N (R) -, -CO-, -C (O) NR-, -C (O) O-, 6-10 membered arylene, 5-12 membered heteroarylene, 3-12 membered carbocycle, or 3-12 membered heterocycle, and- (CH 2 in CH 2) p-X-(CH 2) q -, and the arylene, heteroarylene, carbocycle, and heterocycle may optionally be substituted with halogen, -CN, -OH, -NH 2、-S(O)R、-S(O) 2R、-S(O) 2NR-C 1-6 alkyl,C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, 3-12 membered cycloalkyl, 3-12 membered heterocycloalkyl, 6-10 membered aryl, 5-12 membered heteroaryl, -O-C 1-6 alkyl, or-NR-C 1-6 alkyl substitution,
P and q are each independently 0, 1,2,3, or 4, and p+q is 1,2,3,4, 5, or 6,
R 2 is selected from the group consisting of H, halogen, CH 2F、CHF 2、CF 3、-OH、-NH 2、CN、C 1-6 alkyl, -O-C 1-6 alkyl, - (CH 2) 1-6-CN、-(CH 2) 1-6-O-C 1-6 alkyl, - (CH 2) 1-3 -OH and-NR-C 1-6 alkyl,
R is each independently H, C 1-6 alkyl, 3-8 membered cycloalkyl, or 3-8 membered heterocycloalkyl, which alkyl, cycloalkyl, and heterocycloalkyl may be optionally substituted with halogen, -CN, -OH, -NH 2、-O-C 1-6 alkyl, or-NH-C 1-6 alkyl;
in certain embodiments, the B ring is a 3-12 membered carbocyclic ring or a 4-12 membered heterocyclic ring, and S on the B ring may optionally be oxidized;
in certain embodiments, Y is N or CH, preferably N;
In certain embodiments, R 5 and R 6 are each independently selected from H, halogen, -CN, -OH, -NH 2、C 1-6 alkyl, 3-8 membered cycloalkyl, 3-8 membered heterocycloalkyl, 5-12 membered heteroaryl, -O-C 1-6 alkyl, or-NR-C 1-6 alkyl;
In certain embodiments, R 5 and R 6 are taken together to form- (CH 2) p-X-(CH 2) q -, wherein X is a bond, -O-, -S-, -N (R) -, -CO-, -C (O) NR-, -C (O) O-, 6-10 membered arylene, 5-12 membered heteroarylene, 3-12 membered carbocycle, or 3-12 membered heterocycle, and- (CH 2 in CH 2) p-X-(CH 2) q -and the arylene, heteroarylene, carbocycle and heterocycle may be optionally substituted with halogen, -CN, -OH, -NH 2、C 1-6 alkyl, 3-12 membered cycloalkyl, 3-12 membered heterocycloalkyl, 6-10 membered aryl, 5-12 membered heteroaryl, -O-C 1-6 alkyl, or-NR-C 1-6 alkyl, p, q, R being as defined above;
In certain embodiments, R 5 and R 6 are taken together to form- (CH 2) p-X-(CH 2) q -, wherein X is a bond, -O-, -S-, -N (R) -, -CO-, -C (O) NR-, -C (O) O-, 6-10 membered arylene, 5-12 membered heteroarylene, 3-12 membered carbocycle, or 3-12 membered heterocycle, and- (CH 2 in CH 2) p-X-(CH 2) q -and the arylene, heteroarylene, carbocycle and heterocycle optionally substituted by halogen, -CN, -OH, -NH 2, or C 1-6 alkyl, p, q, R being as defined above;
In certain embodiments, R 2 is selected from H, halogen, CH 2F、CHF 2、CF 3、-OH、-NH 2, CN, and C 1-6 alkyl;
In certain embodiments, R 1 is C 1-6 alkyl;
In certain embodiments, R 1 is CD 3;
In another aspect, the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt, solvate, polymorph or isomer thereof,
Wherein,
Ring A is a 6-10 membered aryl or a 5-12 membered heteroaryl,
Ring B is a 3-12 membered carbocycle or a 4-12 membered heterocycle, S on ring B may optionally be oxidized,
Z is-N (R) -, O or S,
X 2 is CR 2 or N,
X 1 is CRR 4, O, S, or NR 6,
R 1 is H, C 1-6 alkyl, C 3-8 cycloalkyl, or 3-8 membered heterocycloalkyl,
R 7 and R 8 are each independently selected from halogen, CN, -OH, -NH 2、C 1-6 alkyl, 3-8 membered cycloalkyl, 3-8 membered heterocycloalkyl, -O-C 1-6 alkyl and-NR-C 1-6 alkyl,
M and n are each independently 0, 1, 2, or 3,
R 3 is R 5 or-X 3-R 5,
R 4 is R 6 or-X 3-R 6,
X 3 is each independently-O-, -S-, or-NR-,
R 5 and R 6 are each independently selected from H, halogen, -CN, -OH, -NH 2、C 1-6 alkyl, 3-8 membered cycloalkyl, 3-8 membered heterocycloalkyl, -O-C 1-6 alkyl, or-NR-C 1-6 alkyl, or
R 5 and R 6 are joined to form- (CH 2) p-X-(CH 2) q -, wherein X is a bond, -CR=CR-, -CO-CR=CR-, -C≡C-, -CO-C≡C-, -O-, -S (O) 2-、-S(O) 2 NR- -N (R) -, -CO-, -C (O) NR-, -C (O) O-, 6-10 membered arylene, 5-12 membered heteroarylene, 3-12 membered carbocycle, or 3-12 membered heterocycle, and- (CH 2 in CH 2) p-X-(CH 2) q -, and the arylene, heteroarylene, carbocycle, and heterocycle may optionally be substituted with halogen, -CN, -OH, -NH 2、-S(O)R、-S(O) 2R、-S(O) 2NR-C 1-6 alkyl,C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, 3-12 membered cycloalkyl, 3-12 membered heterocycloalkyl, 6-10 membered aryl, 5-12 membered heteroaryl, -O-C 1-6 alkyl, or-NR-C 1-6 alkyl substitution,
P and q are each independently 0, 1,2,3, or 4, and p+q is 1,2,3,4, 5, or 6,
R 2 is selected from the group consisting of H, halogen, CH 2F、CHF 2、CF 3、-OH、-NH 2、CN、C 1-6 alkyl, -O-C 1-6 alkyl, - (CH 2) 1-6-CN、-(CH 2) 1-6-O-C 1-6 alkyl, - (CH 2) 1-3 -OH and-NR-C 1-6 alkyl,
R is each independently H, C 1-6 alkyl, 3-8 membered cycloalkyl, or 3-8 membered heterocycloalkyl, which alkyl, cycloalkyl, and heterocycloalkyl may be optionally substituted with halogen, -CN, -OH, -NH 2、-O-C 1-6 alkyl, or-NH-C 1-6 alkyl;
in certain embodiments, R 5 and R 6 are taken together to form- (CH 2) p-X-(CH 2) q -, wherein X is a bond, -O-, -S-, -N (R) -, -CO-, -C (O) NR-, -C (O) O-, 6-10 membered arylene, 5-12 membered heteroarylene, 3-12 membered carbocycle, or 3-12 membered heterocycle, and- (CH 2 in CH 2) p-X-(CH 2) q -and the arylene, heteroarylene, carbocycle and heterocycle may be optionally substituted with halogen, -CN, -OH, -NH 2、C 1-6 alkyl, 3-12 membered cycloalkyl, 3-12 membered heterocycloalkyl, 6-10 membered aryl, 5-12 membered heteroaryl, -O-C 1-6 alkyl, or-NR-C 1-6 alkyl, p, q, R being as defined above;
In certain embodiments, R 5 and R 6 are taken together to form- (CH 2) p-X-(CH 2) q -, wherein X is a bond, -O-, -S-, -N (R) -, -CO-, -C (O) NR-, -C (O) O-, 6-10 membered arylene, 5-12 membered heteroarylene, 3-12 membered carbocycle, or 3-12 membered heterocycle, and- (CH 2 in CH 2) p-X-(CH 2) q -and the arylene, heteroarylene, carbocycle and heterocycle optionally substituted by halogen, -CN, -OH, -NH 2, or C 1-6 alkyl, p, q, R being as defined above;
In certain embodiments, R 2 is selected from H, halogen, CH 2F、CHF 2、CF 3、-OH、-NH 2, CN, and C 1-6 alkyl;
In certain embodiments, R 1 is C 1-6 alkyl;
In certain embodiments, R 1 is CD 3;
in some embodiments of the invention, the compounds of the invention are selected from:
or a pharmaceutically acceptable salt, solvate, polymorph or isomer thereof.
It is noted that the compounds and salts described in this specification can exist in solvated and unsolvated forms; the atoms of the compounds and salts described herein may exist as isotopes thereof; furthermore, the compounds and salts described in this specification may exist in optically active or racemic forms via one or more asymmetric carbon atoms.
In another aspect, the invention provides a pharmaceutical composition comprising a compound of the invention, or a pharmaceutically acceptable salt, solvate, polymorph, or tautomer thereof. In some embodiments, the pharmaceutical compositions of the present invention further comprise pharmaceutically acceptable excipients.
In another aspect, the invention provides a method of treating a DNA-PK related disease comprising administering to a subject an effective amount of a compound of the invention, or a pharmaceutically acceptable salt, solvate, polymorph or isomer thereof, or a pharmaceutical composition thereof;
In another aspect, the invention provides the use of a compound of the invention, or a pharmaceutically acceptable salt, solvate, polymorph, or tautomer thereof, or a pharmaceutical composition thereof, in the manufacture of a medicament for the treatment of a disease associated with DNA-PK.
In some embodiments of the invention, the DNA-PK related disease is cancer; preferably, the cancer is colorectal cancer, glioblastoma, gastric cancer, ovarian cancer, diffuse large B-cell lymphoma, chronic lymphocytic leukemia, acute myelogenous leukemia, head and neck squamous cell carcinoma, breast cancer, prostate cancer, bladder cancer, hepatocellular carcinoma, small-cell lung cancer, or non-small-cell lung cancer.
Detailed Description
In the following detailed description of the invention, exemplary embodiments are set forth that utilize the principles of the present invention. The features and advantages of the present invention may be better understood by reference to the following summary.
It is to be understood that the scope of the various aspects of the invention is defined by the claims, and methods and structures within the scope of these claims, as well as equivalent methods and structures, are within the scope of the claims.
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the claimed subject matter belongs. All patents, patent applications, and publications cited herein are hereby incorporated by reference in their entirety unless otherwise indicated.
It is to be understood that both the foregoing general description and the following detailed description are exemplary and explanatory only and are not restrictive of any subject matter of the invention. The use of the singular also includes the plural unless specifically stated otherwise. The use of "or" means "and/or" unless stated otherwise. Furthermore, the terms "include," as well as other forms, such as "comprising," "including," and "containing," are not limiting.
Certain chemical terms
The terms "optional," "optional," or "optionally" mean that the subsequently described event or circumstance may or may not occur, and that the description includes instances where the event or circumstance occurs and instances where it does not. For example, "optionally substituted alkyl" means "unsubstituted alkyl" or "substituted alkyl". Also, the optionally substituted group may be unsubstituted (e.g.,: -CH 2CH 3), fully substituted (e.g.,: -CF 2CF 3), monosubstituted (e.g.,: -CH 2CH 2 F), or any level between monosubstituted and fully substituted (e.g.,: -CH 2CHF 2、-CF 2CH 3、-CFHCHF 2, etc.). It will be appreciated by those skilled in the art that for any group comprising one or more substituents, no substitution or pattern of substitution is introduced that is sterically impossible and/or synthetic.
Unless otherwise indicated, conventional methods within the skill of the art, such as mass spectrometry, nuclear magnetism, high performance liquid chromatography, infrared and ultraviolet/visible spectrometry, and pharmacological methods are employed. Unless specifically defined otherwise, the relevant terms and experimental procedures and techniques herein in analytical chemistry, organic synthetic chemistry, and pharmaceutical and medicinal chemistry are known in the art. Standard techniques may be used in chemical synthesis, chemical analysis, pharmaceutical preparation, formulation and delivery, and treatment of patients. For example, the reaction and purification can be carried out using the manufacturer's instructions for the kit, or in a manner well known in the art or in accordance with the teachings of the present invention. The techniques and methods described above may generally be practiced according to conventional methods well known in the art, based on a number of general and more specific descriptions in the literature cited and discussed in this specification. In this specification, groups and substituents thereof can be selected by one skilled in the art to provide stable moieties and compounds.
When substituents are described by conventional formulas written from left to right, the substituents also include chemically equivalent substituents obtained when writing formulas from right to left. For example, -CH 2 O-is equivalent to-OCH 2 -.
The terms "group", "chemical group" as used herein refer to a particular moiety or functional group of a molecule. Chemical groups are often considered as chemical entities that are embedded or attached to a molecule.
Some of the chemical groups named herein may be represented by shorthand notations for the total number of carbon atoms. For example, C 1- 6 alkyl describes an alkyl group, as defined below, having a total of 1 to 6 carbon atoms. The total number of carbon atoms indicated by the shorthand notation does not include carbon atoms on a possible substituent.
The term "halogen", "halo" or "halide" refers to bromine, chlorine, fluorine or iodine.
The compounds of the invention may comprise one or more (e.g., one, two, three or four) isotopic substitutions. For example, in the compounds, H may be in any isotopic form, including 1H、 2 H (D or deuterium) and 3 H (T or tritium); c may be in any isotopic form, including 12C、 13 C and 14 C; o may be in any isotopic form, including 16 O, 18 O, and the like.
The terms "aromatic", "aromatic ring", "aromatic ring" as used herein refer to a planar ring or ring portion of multiple rings having a delocalized electron conjugated system of 4n+2 electrons, where n is an integer. The aromatic ring may be formed from 5, 6, 7, 8, 9 or more than 9 atoms. The aromatic compound may be optionally substituted and may be monocyclic or polycyclic with fused rings.
The term "heteroatom" or "hetero" as used herein alone or as part of other ingredients refers to atoms other than carbon and hydrogen. The heteroatoms are independently selected from oxygen, nitrogen, sulfur, phosphorus, silicon, selenium, and tin, but are not limited to these atoms. In embodiments where two or more heteroatoms are present, the two or more heteroatoms may be the same as one another, or some or all of the two or more heteroatoms may be different from one another.
The term "fused" or "fused ring" as used herein, alone or in combination, refers to a cyclic structure in which two or more rings share one or more bonds.
The term "spiro" or "spiro" as used herein, alone or in combination, refers to a cyclic structure in which two or more rings share one or more atoms.
The term "alkyl" as used herein alone or in combination refers to an optionally substituted straight or optionally substituted branched monovalent saturated hydrocarbon having from 1 to 12 carbon atoms, preferably from 1 to 8 carbon atoms, more preferably from 1 to 6 carbon atoms, attached to the rest of the molecule by single bonds, such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, n-hexyl, n-heptyl, 2-methylhexyl, 3-methylhexyl, n-octyl, n-nonyl, n-decyl, and the like.
The term "alkenyl" as used herein, alone or in combination, refers to an optionally substituted straight or optionally substituted branched monovalent hydrocarbon radical having one or more c=c double bonds and having from 2 to about 10 carbon atoms, more preferably from 2 to about 6 carbon atoms. The double bonds in these groups may be in either cis or trans conformation and should be understood to include both isomers. Examples include, but are not limited to, vinyl (ch=ch 2), 1-propenyl (CH 2CH=CH 2), isopropenyl (C (CH 3)=CH 2), butenyl, and 1, 3-butadienyl, and the like, where alkenyl groups as defined herein appear in numerical ranges, for example, "C 2-C 6 alkenyl" or "C 2- 6 alkenyl" refers to alkenyl groups that may consist of 2 carbon atoms, 3 carbon atoms, 4 carbon atoms, 5 carbon atoms, or 6 carbon atoms, and alkenyl groups herein also encompass instances where numerical ranges are not specified.
The term "alkynyl", as used herein alone or in combination, refers to an optionally substituted straight or branched chain monovalent hydrocarbon radical having one or more c≡c triple bonds and having from 2 to about 10 carbon atoms, more preferably from 2 to about 6 carbon atoms. Examples include, but are not limited to, ethynyl, 2-propynyl, 2-butynyl, 1, 3-butadiynyl, and the like. Where alkynyl groups are defined herein to have a numerical range, for example "C 2-C 6 alkynyl" or "C 2- 6 alkynyl" refers to alkynyl groups that may be composed of 2 carbon atoms, 3 carbon atoms, 4 carbon atoms, 5 carbon atoms or 6 carbon atoms, and alkynyl groups herein also encompass instances where no numerical range is specified.
The term "aryl" refers to an all-carbon monocyclic or fused ring having a fully conjugated pi-electron system, which has 6 to 14 carbon atoms, preferably 6 to 12 carbon atoms, and most preferably 6 carbon atoms. Aryl groups may be unsubstituted or substituted with one or more substituents, examples of which include, but are not limited to, alkyl, alkyloxy, aryl, aralkyl, amino, halogen, hydroxy, sulfonyl, sulfinyl, phosphoryl, and heteroalicyclic. Non-limiting examples of unsubstituted aryl groups include, but are not limited to, phenyl, naphthyl, and anthracenyl.
The term "arylene" as used herein, alone or in combination, refers to a divalent group derived from a monovalent aryl group as defined above.
The term "heteroaryl" refers to a monocyclic or fused ring of 5 to 12 ring atoms having 5, 6, 7, 8, 9, 10, 11 or 12 ring atoms containing 1,2, 3 or 4 ring atoms selected from N, O, S, the remaining ring atoms being C and having a fully conjugated pi-electron system. Heteroaryl groups may be unsubstituted or substituted, and the substituents include, but are not limited to, alkyl, alkyloxy, aryl, aralkyl, amino, halogen, hydroxy, cyano, nitro, carbonyl, and heteroalicyclic. Non-limiting examples of unsubstituted heteroaryl groups include, but are not limited to, pyrrolyl, furanyl, thienyl, imidazolyl, oxazolyl, pyrazolyl, pyridyl, pyrimidinyl, pyrazinyl, quinolinyl, isoquinolinyl, tetrazolyl, triazinyl.
The term "heteroarylene" as used herein, alone or in combination, refers to a divalent group derived from a monovalent heteroaryl group as defined above.
The term "cycloalkyl" as used herein, alone or in combination, refers to an optionally substituted monovalent saturated hydrocarbon ring containing from 3 to about 15 ring-forming carbon atoms or from 3 to about 10 ring-forming carbon atoms, and may also include other non-ring-forming carbon atoms as substituents (e.g., methylcyclopropyl).
The term "carbocycle" refers to a structure covalently closed by a carbon, which may be saturated or partially unsaturated. Carbocycles may be formed from 3, 4,5,6, 7, 8, 9 or more than 9 atoms. The distinction between the terms carbocycle and heterocycle is that the ring backbone of a heterocycle contains at least one atom different from carbon. "carbocycles" herein may be monocyclic or polycyclic, and polycyclic carbocycles include spiro, fused and bridged rings. The carbocycle may be optionally substituted. "carbocycle" herein preferably comprises about 5 to about 20 or 5 to 10 or 5-8 or 5-6 backbone ring atoms.
The terms "heterocycle", "heterocycloalkyl", as used herein, alone or in combination, refer to an aliphatic heterocycle, which may be saturated or partially unsaturated. Where the number of carbon atoms of a heterocycle is referred to herein (e.g., a C 3- 6 heterocycle), there must be at least one non-carbon atom (heteroatom) in the ring. For example, the designation "C 3- 6 heterocycle" refers only to the number of carbon atoms in the ring, and not to the total number of atoms in the ring. The designation "4-6 membered heterocyclic ring" refers to the total number of atoms contained in the ring (i.e., a four, five, or six membered ring in which at least one atom is a carbon atom, at least one atom is a heteroatom, and the remaining 2-4 atoms are carbon atoms or heteroatoms). For heterocycles having two or more heteroatoms, the two or more heteroatoms may be the same as or different from each other. The "heterocycle" herein may be a single ring or multiple rings, and the multiple ring heterocycle includes spiro rings, condensed rings and bridged rings. The heterocycle may be optionally substituted. The "heterocycle" herein preferably contains from about 5 to about 20 or 5 to 10 or 5-8 or 5-6 backbone ring atoms.
The term "polymorph" or "polymorphism" as used herein means that the compounds of the present invention have a variety of lattice morphologies. Some compounds of the invention may have more than one crystal form, and the invention encompasses all polymorphs or mixtures thereof.
Intermediate compounds of the present invention and polymorphs thereof are also within the scope of the present invention.
Unless otherwise specified, olefinic double bonds contained in the compounds of the present invention include the E and Z isomers.
It will be appreciated that the compounds of the present invention may contain asymmetric centers. These asymmetric centers may independently be in the R or S configuration. Some of the compounds of the present invention may also exhibit cis-trans isomerism, as will be apparent to those skilled in the art. It is to be understood that the compounds of the present invention include their individual geometric isomers and stereoisomers as well as mixtures thereof, including racemic mixtures. These isomers may be separated from their mixtures by performing or modifying known methods, such as chromatography techniques and recrystallization techniques, or they may be prepared separately from the appropriate isomers of their intermediates.
The term "pharmaceutically acceptable salt" as used herein includes both acid and base addition salts.
"Pharmaceutically acceptable salts of acids" refers to those salts that retain the biological effectiveness and properties of the free base of the compound, are not biologically or otherwise undesirable, are formed with inorganic acids such as, but not limited to, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like, or organic acids such as, but not limited to, acetic acid, 2-dichloroacetic acid, adipic acid, alginic acid, ascorbic acid, aspartic acid, benzenesulfonic acid, benzoic acid, decanoic acid, hexanoic acid, carbonic acid, cinnamic acid, citric acid, and the like. By "pharmaceutically acceptable salts of bases" is meant those salts which retain the biological effectiveness and properties of the free acid of the compound, are not biologically or otherwise undesirable. These salts are prepared by reacting the free acid with an inorganic or organic base. Salts formed by reaction with inorganic bases include, but are not limited to, sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts, and the like. Preferred inorganic salts are ammonium, sodium, potassium, calcium, and manganese salts.
The organic bases forming salts include, but are not limited to, primary, secondary, tertiary, cyclic amines and the like, such as ammonia, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, diethanolamine, ethanolamine, dicyclohexylamine, ethylenediamine, purine, piperazine, piperidine, choline, caffeine and the like. Particularly preferred organic bases are isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline and caffeine.
Crystallization often yields solvates of the compounds of the present invention. The term "solvate" as used herein refers to a complex of one or more molecules of a compound of the invention and one or more molecules of a solvent.
The solvent may be water, in which case the solvate is a hydrate. In addition, an organic solvent is also possible. Thus, the compounds of the present invention may exist as hydrates, including monohydrate, dihydrate, hemihydrate, trihydrate, tetrahydrate, and the like, as well as the corresponding solvated forms. The compounds of the invention may be true solvates, but in other cases the compounds of the invention may only occasionally retain water or a mixture of water with some other solvent. The compounds of the invention may be reacted in a solvent or precipitated or crystallized in a solvent. Solvates of the compounds of the present invention are also included within the scope of the present invention.
The term "pharmaceutical composition" as used herein refers to a formulation that is mixed with a compound of the present invention and a medium that is generally accepted in the art for delivery of a biologically active compound to a mammal, such as a human. Such a medium comprises all pharmaceutically acceptable carriers.
The term "acceptable" in relation to a formulation, composition or ingredient as used herein means that there is no sustained detrimental effect on the overall health of the subject being treated.
The term "pharmaceutically acceptable" as used herein refers to a material (e.g., carrier or diluent) that does not affect the biological activity or properties of the compounds of the present invention, and is relatively non-toxic, i.e., the material can be administered to an individual without causing an adverse biological reaction or interacting in an adverse manner with any of the components contained in the composition.
"Pharmaceutically acceptable carrier" includes, but is not limited to, adjuvants, carriers, excipients, adjuvants, deodorants, diluents, preservatives, dyes/colorants, flavor enhancers, surfactants and wetting agents, dispersing agents, suspending agents, stabilizers, isotonic agents, solvents, or emulsifiers that have been approved by the relevant government administration for use in humans and domestic animals.
The terms "subject," "patient," "subject," or "individual" as used herein refer to an individual having a disease, disorder, or condition, and the like, including mammals and non-mammals. Examples of mammals include, but are not limited to, any member of the class mammalia: human, non-human primates (e.g., chimpanzees and other apes and monkeys); livestock, such as cattle, horses, sheep, goats, pigs; domestic animals such as rabbits, dogs, and cats; laboratory animals, including rodents, such as rats, mice, guinea pigs, and the like. Examples of non-human mammals include, but are not limited to, birds, fish, and the like. In one embodiment of the related methods and compositions provided herein, the mammal is a human.
The term "treatment" as used herein refers to the treatment of a related disease or condition in a mammal, particularly a human, including
(I) Preventing a disease or condition in a mammal, particularly a mammal that has been previously exposed to a disease or condition but has not been diagnosed with the disease or condition, from developing the corresponding disease or condition;
(ii) Inhibiting the disease or disorder, i.e., controlling its progression;
(iii) Alleviating the disease or condition, i.e., causing regression of the disease or condition;
(iv) Relieving symptoms caused by diseases or symptoms.
The terms "disease" and "disorder" as used herein may be used interchangeably or differently and, because some specific diseases or disorders have not yet been known to cause a disease (and therefore the cause of the disease is not yet known), they cannot be considered as a disease but rather can be considered as an unwanted condition or syndrome, more or less specific symptoms of which have been confirmed by clinical researchers.
The term "effective amount," "therapeutically effective amount," or "pharmaceutically effective amount" as used herein refers to an amount of at least one agent or compound that is sufficient to alleviate one or more symptoms of the disease or disorder being treated to some extent after administration. The result may be a reduction and/or alleviation of signs, symptoms, or causes of a disease, or any other desired alteration of a biological system. For example, an "effective amount" for treatment is the amount of a composition comprising a compound disclosed herein that is required to provide clinically significant relief from a disorder. Effective amounts suitable in any individual case can be determined using techniques such as a dose escalation test.
The terms "administering," "administering," and the like as used herein refer to a method capable of delivering a compound or composition to a desired site for biological action. These methods include, but are not limited to, oral routes, duodenal routes, parenteral injection (including intravenous, subcutaneous, intraperitoneal, intramuscular, intraarterial injection or infusion), topical administration, and rectal administration. In preferred embodiments, the compounds and compositions discussed herein are administered orally.
The anti-cancer treatments described herein may be useful as monotherapy or may include conventional surgery, radiation therapy or chemotherapy in addition to administration of a compound having formula (I); or a combination of such additional therapies. Such conventional surgery, radiation therapy or chemotherapy may be administered simultaneously, sequentially or separately with the compound having formula (I) for treatment.
Preparation of the Compounds of the invention
The following schemes show the methods for preparing the compounds of the present invention.
It will be appreciated that in the following description, combinations of substituents and/or variables of the formula are only allowed in the case of stable compounds.
Those skilled in the art will also appreciate that in the schemes described below, the functional groups of the intermediate compounds may need to be protected by suitable protecting groups. These functional groups include hydroxyl, amino, mercapto and carboxyl groups. Suitable hydroxy protecting groups include trialkylsilyl or diarylalkylsilyl (e.g., t-butylmethylsilyl, t-butyldiphenylsilyl or trimethylsilyl), tetrahydropyranyl, benzyl, and the like. Suitable amino, amidino and guanidine protecting groups include t-butoxycarbonyl, benzyloxycarbonyl, and the like. Suitable protecting groups for mercapto groups include-C (O) -R "(R" means alkyl, aryl or arylalkyl), p-methoxybenzyl, trityl, and the like. Suitable carboxyl protecting groups include alkyl, aryl or arylalkyl esters. The protecting groups may be added or removed by standard techniques known to those skilled in the art.
Examples
The following non-limiting examples are illustrative only and do not limit the invention in any way.
Unless otherwise indicated, temperatures are degrees celsius. Reagents were purchased from commercial suppliers of national pharmaceutical group chemicals Beijing Co., ltd, alfaAesar (Alfaaesar), or Beijing carboline technologies Co., ltd, and these reagents were used directly without further purification unless otherwise indicated.
Unless otherwise indicated, the following reactions were carried out at room temperature, in anhydrous solvents, under positive pressure of nitrogen or argon, or using dry tubes; the reaction flask is provided with a rubber diaphragm, so that a substrate and a reagent are added through a syringe; glassware drying and/or heat drying.
Column chromatography purification uses 200-300 mesh silica gel from the Qingdao marine chemical plant unless otherwise indicated; preparation of thin layer chromatography A thin layer chromatography silica gel prefabricated plate (HSGF 254) manufactured by Kagaku chemical industry research institute of tobacco, inc.; the MS was determined using a Thermo LCQ sheet type (ESI) liquid chromatograph-mass spectrometer; the polarimeter SGW-3 was used for polarimeter, shanghai Shen Guang instruments, inc.
Nuclear magnetic data (1 HNMR) were run at 400MHz using a Varian device. As the solvent used for the nuclear magnetic data, CDCl 3、CD 3OD、D 2 O, DMSO-d6 and the like were mentioned, based on tetramethylsilane (0.00 ppm) or based on the residual solvent (CDCl 3:7.26ppm;CD 3OD:3.31ppm;D 2 O:4.79ppm; d6-DMSO:2.50 ppm). When peak shape diversity is indicated, the following abbreviations represent the different peak shapes: s (singlet), d (doublet), t (triplet), q (quartet), m (multiplet), br (broad), dd (doublet), dt (doublet). If the coupling constant is given, it is in Hertz (Hz).
Abbreviations:
intermediate 1: 7-methyl-6-nitroquinolin-4-ol
3-Methyl-4-nitroaniline (1.52 g) and 5-ethoxymethylaenyl-2, 2-dimethyl-1, 3-dioxane-4, 6-dione (2.01 g) were dissolved in absolute ethanol (50 mL), and the reaction mixture was heated to 85℃and stirred for 5 hours. The reaction solution was cooled to room temperature, and the precipitate was filtered and dried in vacuo. Subsequently suspended in diphenyl ether (50 mL) and heated to 250 ℃ for 4 hours, after the reaction solution cooled, the precipitate was filtered and the resulting solid was purified by silica gel column chromatography (eluent: dichloromethane: methanol=20:1 (V: V)) to give 7-methyl-6-nitroquinolin-4-ol (600 mg).
Intermediate 2: 2-chloro-7-methyl-9- (piperidin-4-yl) -7, 9-dihydro-8H-purin-8-one
Step 1:4- (2-chloro-5-nitropyrimidin-4-yl) amino) piperidine-1-carboxylic acid tert-butyl ester
2, 4-Dichloro-5-nitropyrimidine (0.97 g) and triethylamine (1.01 g) were dissolved in tetrahydrofuran (50 mL), and tert-butyl 4-aminopiperidine-1-carboxylate (1.00 g) was slowly added while ice-cooling. The reaction was stirred at room temperature for 2 hours. The reaction solution was poured into a saturated aqueous ammonium chloride solution (200 mL), extracted with ethyl acetate (100 mL), and the extract was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: petroleum ether: ethyl acetate=5:1 (V: V)) to give a yellow solid (1.5 g).
Step 2:4- (5-amino-2-chloropyrimidin-4-yl) amino) piperidine-1-carboxylic acid tert-butyl ester
To a solution of tert-butyl 4- ((2-chloro-5-nitropyrimidin-4-yl) amino) -piperidine-1-carboxylate (1.5 g) in acetic acid (50 mL) was added reduced iron powder (1.9 g), and the reaction mixture was stirred at room temperature for 2 hours, filtered through celite, the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: dichloromethane: methanol=10:1 (V: V)) to give a white solid (1.2 g).
Step 3:4- (2-chloro-8-oxo-7, 8-dihydro-9H-purin-9-yl) piperidine-1-carboxylic acid tert-butyl ester
The tert-butyl 4- ((5-amino-2-chloropyrimidin-4-yl) amino) piperidine-1-carboxylate (1.2 g) and CDI (1.5 g) of step 2 were dissolved in tetrahydrofuran (50 mL) under nitrogen, heated to 65 ℃ and stirred for 2 hours, cooled to room temperature, the reaction solution was poured into water (100 mL), the reaction solution was extracted with ethyl acetate, the extract was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: petroleum ether: ethyl acetate=1:1 (V: V)) to give a white solid (1.1 g).
Step 4:4- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) piperidine-1-carboxylic acid tert-butyl ester
To a solution of tert-butyl 4- (2-chloro-8-oxo-7, 8-dihydro-9H-purin-9-yl) piperidine-1-carboxylate (1.1 g) obtained in step 3 in DMF (20 mL) was slowly added 60% (mineral oil) sodium hydride (200 mg) at 0deg.C, and stirring was continued at 0deg.C for 10 min after the addition. Methyl iodide (1.0 g) was then slowly added to the reaction solution, and after 1 hour, the reaction solution was poured into a saturated aqueous ammonium chloride solution and stirred continuously, and white precipitate was generated, and the precipitate was filtered and dried to give the objective compound (900 mg).
Step 5: 2-chloro-7-methyl-9- (piperidin-4-yl) -7, 9-dihydro-8H-purin-8-one
To a solution of tert-butyl 4- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) piperidine-1-carboxylate (900 mg) obtained in step 4 in dioxane (30 mL) was slowly added 4M hydrochloric acid (30 mL) at 0℃and stirring was continued for 10 hours after the addition. The reaction solution was then neutralized to pH 8 with saturated aqueous sodium bicarbonate solution, white precipitate was generated, and the solid was filtered and dried to give the objective compound (400 mg).
Intermediate 3:4- (2-Bromoethoxy) -7-methyl-6-nitroquinoline
DIAD (306 mg) was slowly added dropwise to a solution of 7-methyl-6-nitroquinolin-4-ol (204 mg), 2-bromoethanol (340 mg) and triphenylphosphine (526 mg) in THF (30 mL) at 0deg.C, and the reaction was warmed to room temperature and stirred for a further 12 hours after the addition. The filtrate was concentrated, and the residue was purified by silica gel column chromatography (eluent: dichloromethane: methanol=20:1 (V: V)) to give a pale yellow solid (290 mg).
Intermediate 4: 2-chloro-7-methyl-9- (4-oxopiperidin-1-yl) -7, 9-dihydro-8H-purin-8-one
Step 1: 8-nitroso-1, 4-dioxa-8-azaspiro [4.5] decane
To a solution of 1, 4-dioxa-8-azaspiro [4.5] decane (1.43 g) in acetic acid (15 mL) and water (5 mL) at 0deg.C was slowly added dropwise sodium nitrite solution (620 mg in 1mL water), and after the addition was completed, the mixture was warmed to room temperature and stirring was continued for 2 hours. Ethyl acetate was added for extraction, the extract was concentrated under reduced pressure, and the residue was used in the next step without purification.
Step 2:1, 4-dioxa-8-azaspiro [4.5] dec-8-amine
To a solution of 8-nitroso-1, 4-dioxa-8-azaspiro [4.5] decane (1.1 g) in acetic acid (10 mL) and methanol (10 mL) at room temperature was added reduced zinc powder (2 g), followed by stirring at 25℃for 2 hours. The reaction solution was filtered through celite, and the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: dichloromethane: methanol=3:1 (V: V)) to give the title compound (350 mg).
Step 3: n- (2-chloro-5-nitropyrimidin-4-yl) -1, 4-dioxa-8-azaspiro [4.5] dec-8-amine
2, 4-Dichloro-5-nitropyrimidine (970 mg) and triethylamine (2.01 g) were dissolved in tetrahydrofuran (20 mL), and 1, 4-dioxa-8-azaspiro [4.5] dec-8-amine (350 mg) was slowly added under ice-bath. The reaction mixture was stirred at room temperature for 5 hours. The reaction solution was poured into a saturated aqueous sodium chloride solution (100 mL), extracted with ethyl acetate (50 mL), and the extract was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: petroleum ether: ethyl acetate=5:1 (V: V)) to give a yellow solid (500 mg).
Step 4: 2-chloro-N4- (1, 4-dioxa-8-azaspiro [4.5] dec-8-yl) pyrimidine-4, 5-diamine
To a solution of N- (2-chloro-5-nitropyrimidin-4-yl) -1, 4-dioxa-8-azaspiro [4.5] decan-8-amine (500 mg) in acetic acid (30 mL) at room temperature was added reduced iron powder (1.5 g), and the reaction mixture was stirred at room temperature for 2 hours, filtered through celite, and the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: dichloromethane: methanol=10:1 (V: V)) to give a white solid (350 mg).
Step 5: 2-chloro-9- (1, 4-dioxa-8-azaspiro [4.5] decan-8-yl) -7, 9-dihydro-8H-purin-8-one
2-Chloro-N4- (1, 4-dioxa-8-azaspiro [4.5] dec-8-yl) pyrimidine-4, 5-diamine (350 mg) and CDI (500 mg) in step 4 were dissolved in acetonitrile (10 mL), heated to 85℃under nitrogen and stirred for 12 hours, cooled to room temperature, the reaction solution was poured into water (100 mL), extracted with ethyl acetate, the extract was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: petroleum ether: ethyl acetate=1:1 (V: V)) to give a white solid (250 mg).
Step 6: 2-chloro-7-methyl-9- (1, 4-dioxa-8-azaspiro [4.5] decan-8-yl) -7, 9-dihydro-8H-purin-8-one
To a solution of 2-chloro-9- (1, 4-dioxa-8-azaspiro [4.5] decan-8-yl) -7, 9-dihydro-8H-purin-8-one (250 mg) obtained in step 5 in DMF (10 mL) was slowly added 60% (mineral oil) sodium hydride (80 mg) at 0deg.C, and stirring was continued at 0deg.C for 10 min after the addition. Methyl iodide (200 mg) was then slowly added to the reaction solution, stirred for 1 hour, and the reaction solution was poured into a saturated aqueous ammonium chloride solution, stirred constantly, and white precipitate was generated, filtered and dried to give the objective compound (200 mg).
Step 7: 2-chloro-7-methyl-9- (4-oxopiperidin-1-yl) -7, 9-dihydro-8H-purin-8-one
To a solution of 2-chloro-7-methyl-9- (1, 4-dioxa-8-azaspiro [4.5] decan-8-yl) -7, 9-dihydro-8H-purin-8-one (200 mg) obtained in step 6 in tetrahydrofuran (10 mL) at 0℃was slowly added 4M hydrochloric acid (10 mL), and stirring was continued for 10 hours after the addition. The reaction solution was then neutralized to pH 8 with saturated aqueous sodium bicarbonate, a white precipitate was formed, and the solid was filtered and dried to give the title compound (130 mg).
Intermediate 5:4- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Step 1:4- ((2-chloro-5-nitropyrimidin-4-yl) amino) tetrahydro-2H-pyran-4-carbonitrile
2, 4-Dichloro-5-nitropyrimidine (1.94 g) and triethylamine (1.01 g) were dissolved in tetrahydrofuran (50 mL), and 4-aminotetralin-2H-pyran-4-carbonitrile (1.26 g) was slowly added to the solution in an ice bath. The reaction was allowed to warm to room temperature and stirred overnight. The reaction solution was poured into a saturated aqueous ammonium chloride solution (200 mL), extracted with ethyl acetate (100 mL), and the extract was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: petroleum ether: ethyl acetate=3:1 (V: V)) to give a white solid (1.9 g).
Step 2:4- ((5-amino-2-chloropyrimidin-4-yl) amino) tetrahydro-2H-pyran-4-carbonitrile
To a solution of 4- ((2-chloro-5-nitropyrimidin-4-yl) amino) tetrahydro-2H-pyran-4-carbonitrile (1.9 g) in acetic acid (80 mL) at room temperature was added reduced iron powder (1.5 g), and the reaction was then warmed to 45℃and stirred for 2 hours. Cooled to room temperature, the reaction mixture was poured into ice water and extracted with ethyl acetate (100 mL); the extract was washed with saturated brine, dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: dichloromethane: methanol=20:1 (V: V)) to give a white solid (1.3 g).
Step 3:4- (2-chloro-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
4- ((5-Amino-2-chloropyrimidin-4-yl) amino) tetrahydro-2H-pyran-4-carbonitrile (1.3 g) and CDI (1.3 g) from step 2 were dissolved in tetrahydrofuran (50 mL), heated to 65℃under nitrogen and stirred for 12 hours, cooled to room temperature, the reaction solution was poured into water (150 mL), extracted with methylene chloride, the extract was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: petroleum ether: ethyl acetate=3:1 (V: V)) to give a pale yellow solid (1 g).
Step 4:4- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
To a solution of 4- (2-chloro-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile (1 g) obtained in step 3 in DMF (20 mL) was slowly added 60% (mineral oil) of sodium hydride (200 mg) at 0deg.C, and stirring was continued at 0deg.C for 0.5 hours after the addition. Methyl iodide (1.41 g) was then slowly added to the reaction solution, and after completion of the reaction, the reaction solution was poured into a saturated aqueous ammonium chloride solution with continuous stirring, and yellow precipitate was generated, filtered and dried to give a yellow solid (900 mg).
Intermediate 6: 2-chloro-7-cyclopropyl-9- (4-oxocyclohexyl) -7, 9-dihydro-8H-purin-8-one
Step 1: 2-chloro-7-cyclopropyl-9- (1, 4-dioxaspiro [4.5] decan-8-yl) -7, 9-dihydro-8H-purin-8-one
To a solution of 2-chloro-9- (1, 4-dioxaspiro [4.5] decan-8-yl) -7, 9-dihydro-8H-purin-8-one (311 mg) in 1, 2-dichloroethane (20 mL) was added cyclopropylboronic acid (100 mg), copper acetate (20 mg) and pyridine (160 mg) in this order at room temperature, and stirring was continued with opening for 12 hours after the addition. Pouring the reaction solution into saturated ammonium chloride aqueous solution after the reaction is finished, and extracting by ethyl acetate; the extract was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: dichloromethane: methanol=20:1 (V: V)) to give the title compound (200 mg).
Step 2: 2-chloro-7-cyclopropyl-9- (4-oxocyclohexyl) -7, 9-dihydro-8H-purin-8-one
To a solution of 2-chloro-7-cyclopropyl-9- (1, 4-dioxaspiro [4.5] decan-8-yl) -7, 9-dihydro-8H-purin-8-one (250 mg) in dioxane (20 mL) was added 6N hydrochloric acid (5 mL) at room temperature, and the mixture was stirred for 12 hours after the addition. Pouring the reaction solution into saturated sodium bicarbonate aqueous solution after the reaction is finished, and extracting by ethyl acetate; the extract was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: dichloromethane: methanol=20:1 (V: V)) to give the title compound (130 mg).
Intermediate 7: 6-chloro-3-methyl-1- (4-oxocyclohexyl) -1, 3-dihydro-2H-imidazo [4,5-c ] pyridin-2-one
Intermediate 7 was synthesized following the synthetic procedure for intermediate 4 using 2, 4-dichloro-5-nitropyridine instead of 2, 4-dichloro-5-nitropyrimidine.
Intermediate 8: 6-chloro-3-methyl-1- (tetrahydro-2H-pyran-4-yl) -1, 3-dihydro-2H-imidazo [4,5-c ] pyridin-2-one
Step 1: 2-chloro-5-nitro-N- (tetrahydro-2H-pyran-4-yl) pyridin-4-amine
2, 4-Dichloro-5-nitropyridine (1.93 g) and triethylamine (1.01 g) were dissolved in tetrahydrofuran (50 mL), and 4-aminotetrahydropyran (1.01 g) was slowly added under an ice bath. The reaction was allowed to warm to room temperature and stirred overnight. The reaction solution was poured into a saturated aqueous ammonium chloride solution (100 mL), extracted with ethyl acetate (100 mL), the extract was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: petroleum ether: ethyl acetate=3:1 (V: V)) to give a white solid (1.5 g).
Step 2: 6-chloro-N 4 - (tetrahydro-2H-pyran-4-yl) pyridine-3, 4-diamine
To a solution of 2-chloro-5-nitro-N- (tetrahydro-2H-pyran-4-yl) pyridin-4-amine (1.5 g) in ethanol (30 mL) and water (10 mL) at room temperature was added ammonium chloride (1.5 g) and reduced iron powder (1.6 g), and the reaction was then warmed to 80℃and stirred for 1 hour. Cooled to room temperature, the reaction mixture was poured into ice water and extracted with ethyl acetate (100 mL); the extract was washed with saturated brine, dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: dichloromethane: methanol=20:1 (V: V)) to give a white solid (1.2 g).
Step 3: 6-chloro-1- (tetrahydro-2H-pyran-4-yl) -1, 3-dihydro-2H-imidazo [4,5-c ] pyridin-2-one
6-Chloro-N 4 - (tetrahydro-2H-pyran-4-yl) pyridine-3, 4-diamine (1.2 g) and CDI (1.6 g) in step 2 were dissolved in tetrahydrofuran (50 mL), heated to 65℃under nitrogen protection and stirred for 2 hours, cooled to room temperature, the reaction solution was poured into water (150 mL), the reaction solution was extracted with methylene chloride, the extract was washed with saturated common salt, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: petroleum ether: ethyl acetate=3:1 (V: V)) to give pale yellow solid (1 g).
Step 4: 6-chloro-3-methyl-1- (tetrahydro-2H-pyran-4-yl) -1, 3-dihydro-2H-imidazo [4,5-c ] pyridin-2-one
To a solution of 6-chloro-1- (tetrahydro-2H-pyran-4-yl) -1, 3-dihydro-2H-imidazo [4,5-c ] pyridin-2-one (1 g) in DMF (20 mL) obtained in step 3 was slowly added 60% (mineral oil) sodium hydride (240 mg) at 0deg.C, and stirring was continued at 0deg.C for 0.5 hours after the addition. Methyl iodide (1.41 g) was then slowly added to the reaction solution and stirring was continued for 1 hour, and after completion of the reaction, the reaction solution was poured into a saturated aqueous ammonium chloride solution and stirred continuously, and yellow precipitate was generated, filtered and dried to give a yellow solid (920 mg).
Intermediate 9: 6-chloro-3-methyl-1-morpholin-1, 3-dihydro-2H-imidazo [4,5-c ] pyridin-2-one
Intermediate 9 was synthesized from 4-aminomorpholine following the synthesis of intermediate 8.
Intermediate 10: 2-chloro-7- (methyl-d 3) -9- (4-oxocyclohexyl) -7, 9-dihydro-8H-purin-8-one
To a solution of 2-chloro-9- (4-oxocyclohexyl) -7, 9-dihydro-8H-purin-8-one (2.67 g) in DMF (100 mL) at 0deg.C was slowly added 60% (mineral oil) sodium hydride (600 mg) and stirring was continued at 0deg.C for 0.5H after addition. Methyl iodide (1.41 g) was then slowly added to the reaction solution, and after completion of the reaction, the reaction solution was poured into a saturated aqueous ammonium chloride solution with the white precipitate being generated with continuous stirring, filtered and dried to give a white solid (2.5 g).
Example 1: (1 1S,1 4s,7 1S,7 3s)-2 7,4 7 -dimethyl-2 8,2 9 -dihydro-2 7 H-5-oxa-3, 8-diaza-4 (6, 4) -quinolin-2 (9, 2) -purin-1 (1, 4) -cyclohexane-7 (1, 3) -cyclobutyl-octan-2 8 -one
Step 1: tert-butyl ((1 s,3 s) -3- ((7-methyl-6-nitroquinolin-4-yl) oxy) methyl) cyclobutyl) carbamate
To a solution of 7-methyl-6-nitroquinolin-4-ol (204 mg), tert-butyl ((1 s,3 s) -3- (hydroxymethyl) cyclobutyl) carbamate (302 mg) and triphenylphosphine (526 mg) in THF (30 mL) at 0 ℃ was slowly added dropwise DIAD (306 mg), and after the addition the reaction was warmed to room temperature and stirring was continued for 12 hours. Filtration, concentration of the filtrate, and purification of the residue by silica gel column chromatography (eluent: dichloromethane: methanol=20:1 (V: V)) gave a pale yellow solid (320 mg).
Step 2: (1 s,3 s) -3- ((7-methyl-6-nitroquinolin-4-yl) oxy) methyl) cyclobutan-1-amine trifluoroacetate salt
Tert-butyl ((1 s,3 s) -3- ((7-methyl-6-nitroquinolin-4-yl) oxy) methyl) cyclobutyl carbamate (320 mg) was dissolved in dichloromethane (10 mL) at 0 ℃ and trifluoroacetic acid (3 mL) was slowly added thereto. The reaction mixture was stirred at room temperature for 1 hour, the reaction solution was concentrated under reduced pressure, and the residue was used in the next reaction without purification (300 mg).
Step 3: 2-chloro-7-methyl-9- ((1 s,4 s) -4- ((1 s,3 s) -3- ((7-methyl-6-nitroquinolin-4-yl) oxy) methyl) cyclobutyl) amino) cyclohexyl) -7, 9-dihydro-8H-purin-8-one
2-Chloro-7-methyl-9- (4-oxocyclohexyl) -7, 9-dihydro-8H-purin-8-one (141 mg) and (1 s,3 s) -3- ((7-methyl-6-nitroquinolin-4-yl) oxy) methyl) cyclobutan-1-amine trifluoroacetate (300 mg) were dissolved in THF (50 mL) at 0℃and sodium triacetoxyborohydride (400 mg) was added thereto. The reaction mixture was stirred at room temperature for 12 hours, the reaction solution was poured into a saturated aqueous sodium hydrogencarbonate solution, ethyl acetate was added for extraction, the extract was concentrated under reduced pressure, and the residue was purified by column chromatography (eluent: dichloromethane: methanol=10:1 (V: V)) to give the title compound (150 mg).
Step 4:9- ((1S, 4 s) -4- ((1 s,3 s) -3- ((6-amino-7-methylquinolin-4-yl) oxy) methyl) cyclobutyl) amino) cyclohexyl) -2-chloro-7-methyl-7, 9-dihydro-8H-purin-8-one
To a solution of 2-chloro-7-methyl-9- ((1 s,4 s) -4- ((1 s,3 s) -3- ((7-methyl-6-nitroquinolin-4-yl) oxy) methyl) cyclobutyl) amino) cyclohexyl) -7, 9-dihydro-8H-purin-8-one (150 mg) in acetic acid (15 mL) at room temperature was added reduced iron powder (0.2 g), followed by stirring at 25 ℃ for 2 hours. The reaction solution was filtered through celite, and the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: dichloromethane: methanol=5:1 (V: V)) to give the title compound (100 mg).
Step 5: (1 1S,1 4s,7 1S,7 3s)-2 7,4 7 -dimethyl-2 8,2 9 -dihydro-2 7 H-5-oxa-3, 8-diaza-4 (6, 4) -quinolin-2 (9, 2) -purin-1 (1, 4) -cyclohexane-7 (1, 3) -cyclobutyl-octan-2 8 -one
The above 9- ((1 s,4 s) -4- ((1 s,3 s) -3- ((6-amino-7-methylquinolin-4-yl) oxy) methyl) cyclobutyl) amino) cyclohexyl) -2-chloro-7-methyl-7, 9-dihydro-8H-purin-8-one (52 mg), ruPhosPdG (9 mg) and cesium carbonate (65 mg) were dissolved in dioxane (10 mL) under nitrogen, heated to 100 ℃ and stirred for 3 hours. The reaction solution was cooled to room temperature, filtered, and the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developer: dichloromethane: methanol=15:1 (V: V)) to give the objective compound (31 mg).
1H NMR(400MHz,DMSO-d 6)8.54(d,J=5.2Hz,1H),8.30(s,1H),8.20(s,1H),8.14(s,1H),7.78(s,1H),6.96(d,J=5.2Hz,1H),4.39(s,2H),3.96-4.06(m,1H),3.29(s,3H),2.81-3.02(m,1H),2.61-2.78(m,1H),2.24-2.47(m,8H),1.67-1.88(m,2H),1.54-1.64(m,2H),1.30-1.49(m,4H),0.88-1.12(br,1H).
Example 2: (1 1s,1 4s)-2 7,4 7, 7-tetramethyl-2 8,2 9 -dihydro-2 7 H-5-oxa-3, 9-diaza-4 (6, 4) -quinolin-2 (9, 2) -purin-1 (1, 4) -cyclohexylcyclononan-2 8 -one
The present compound (20 mg) was synthesized following the synthesis procedure of example 1 using tert-butyl (3-hydroxy-2, 2-dimethylpropyl) carbamate as an alternative starting material.
1H NMR(400MHz,CDCl 3)8.60(d,J=4.8Hz,1H),8.47(s,1H),7.89(s,1H), 7.88(s,1H),6.94(s,1H),6.72(d,J=4.8Hz,1H),4.16-4.26(m,1H),4.01(s,2H),3.41(s,3H),2.71-2.84(m,3H),2.64(s,2H),2.53(s,3H),1.80-1.88(m,2H),1.49-1.70(m,4H),1.17(s,6H).
Example 3: (1 1R,1 4S,6 1R,6 3S)-2 7,4 7 -dimethyl-2 8,2 9 -dihydro-2 7 H-5-oxa-3, 7-diaza-4 (6, 4) -quinolin-2 (9, 2) -purin-1 (1, 4) -cyclohexane-6 (1, 3) -cyclopentylcycloheptane-2- 8 -one
The synthesis of the present compound (19 mg) was performed according to the synthesis method of example 1 using tert-butyl ((1R, 3R) -3-hydroxycyclopentyl) carbamate as a substitute.
1H NMR(400MHz,CDCl 3)8.59(d,J=5.2Hz,1H),8.45(s,1H),7.89(s,1H),7.87(s,1H),6.88(s,1H),6.67(d,J=5.2Hz,1H),4.96-5.01(m,1H),4.18-4.28(m,1H),3.41(s,3H),3.31-3.36(m,1H),2.87-2.92(m,1H),2.48-2.72(m,5H),2.12-2.32(m,3H),1.84-2.10(m,4H),1.52-1.80(m,5H).
Example 4: (1 1S,1 4S,6 1R,6 3R)-2 7,4 7 -dimethyl-2 8,2 9 -dihydro-2 7 H-5-oxa-3, 7-diaza-4 (6, 4) -quinolin-2 (9, 2) -purin-1 (1, 4) -cyclohexane-6 (1, 3) -cyclopentylcycloheptane-2- 8 -one
The present compound (11 mg) was synthesized following the synthesis procedure of example 1 using tert-butyl ((1R, 3S) -3-hydroxycyclopentyl) carbamate as a substitute starting material.
MS(ESI)m/z 486.11(M+H) +
Example 5: (1 1S,1 4S,7R)-27,4 7, 7-trimethyl-2 8,2 9 -dihydro-2 7 H-5-oxa-3, 9-diaza-4 (6, 4) -quinolin-2 (9, 2) -purin-1 (1, 4) -cyclohexylcyclononan-2 8 -one
The present compound (23 mg) was synthesized following the synthesis procedure of example 1 using tert-butyl (R) - (3-hydroxy-2-methylpropyl) carbamate as an alternative starting material.
1H NMR(400MHz,DMSO-d 6)8.52(d,J=5.2Hz,1H),8.41(s,1H),8.30(s,1H),8.18(s,1H),7.74(s,1H),6.83(d,J=5.2Hz,1H),4.38-4.48(m,1H),4.28-4.36(m,1H),4.12-4.20(m,1H),3.95-4.05(m,1H),3.36(s,3H),2.52-2.70(m,4H),2.46(s,3H),2.34-2.44(m,1H),1.94-2.14(m,2H),1.58-1.68(m,2H),1.36-1.54(m,2H),0.97(d,J=6.4Hz,3H).
Example 6: (1 1S,1 4s,6 1R,6 3r)-2 7,4 7 -dimethyl-2 8,2 9 -dihydro-2 7 H-5-oxa-3, 8-diaza-4 (6, 4) -quinolin-2 (9, 2) -purin-1 (1, 4) -cyclohexane-6 (1, 3) -cyclobutyl-octan-2 8 -one
The synthesis of the present compound (9 mg) was performed following the synthesis procedure of example 1 using tert-butyl ((1 s,3 s) -3-hydroxycyclobutyl) methyl carbamate as a substitute.
MS(ESI)m/z486.12(M+H) +
Example 7: (1's, 4's) -7',7' -dimethyl-spiro [ cyclopropane-1, 7 '-5-oxo-3, 9-diaza-4 (6, 4) -quinolin-2 (9, 2) -purin-1 (1, 4) -cyclohexylcyclononane ] -8' -one
The present compound (23 mg) was synthesized following the synthesis procedure of example 1 using tert-butyl ((1- (hydroxymethyl) cyclopropyl) methyl) carbamate as an alternative starting material.
MS(ESI)m/z 486.10(M+H) +
Example 8: (1's, 4's) -7',7' -dimethyl-spiro [ cyclopropane-1, 8 '-5-oxo-3, 9-diaza-4 (6, 4) -quinolin-2 (9, 2) -purin-1 (1, 4) -cyclohexylcyclononane ] -8' -one
The present compound (23 mg) was synthesized following the synthesis procedure of example 1 using tert-butyl (1- (2-hydroxyethyl) cyclopropyl) carbamate as an alternative starting material.
1H NMR(400MHz,CDCl 3)8.81(s,1H),8.58(d,J=4.8Hz,1H),7.92(s,1H),7.88(s,1H),7.05(s,1H),6.76(d,J=4.8Hz,1H),4.58(t,J=5.6Hz,2H),4.25-4.35(m,1H),3.43(s,3H),2.88-2.93(m,1H),2.65-2.76(m,2H),2.54(s,3H),2.02(t,J=5.6Hz,2H),1.82-1.90(m,2H),1.56-1.76(m,4H),0.58-0.62(m,2H),0.35-0.38(m,2H).
Example 9:2 7,4 7 -dimethyl-2 8,2 9 -dihydro-2 7 H-5-oxa-3, 8-diaza-4 (6, 4) -quinolin-2 (9, 2) -purin-1 (4, 1) -piperidinecyclooctan-2- 8 -one
Step 1: 2-chloro-7-methyl-9- (1-nitrosopiperidin-4-yl) -7, 9-dihydro-8H-purin-8-one
To a solution of 2-chloro-7-methyl-9- (piperidin-4-yl) -7, 9-dihydro-8H-purin-8-one (267 mg) in acetic acid (5 mL) and water (1 mL) was slowly added dropwise sodium nitrite solution (62 mg in 0.1mL of water) at 0deg.C, and the mixture was stirred at room temperature for 2 hours after the completion of the addition. Ethyl acetate was added for extraction, the extract was concentrated under reduced pressure, and the residue was purified by column chromatography (eluent: dichloromethane: methanol=20:1 (V: V)) to give the title compound (200 mg).
Step 2:9- (1-aminopiperidin-4-yl) -2-chloro-7-methyl-7, 9-dihydro-8H-purin-8-one
To a solution of 2-chloro-7-methyl-9- (1-nitrosopiperidin-4-yl) -7, 9-dihydro-8H-purin-8-one (200 mg) in acetic acid (10 mL) and methanol (10 mL) at room temperature was added zinc reduction powder (1 g), followed by stirring at 25℃for 2 hours. The reaction solution was filtered through celite, and the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: dichloromethane: methanol=3:1 (V: V)) to give the title compound (150 mg).
Step 3: 2-chloro-7-methyl-9- (1- ((2- ((7-methyl-6-nitroquinolin-4-yl) oxy) ethyl) amino) piperidin-4-yl) -7, 9-dihydro-8H-purin-8-one
The above 9- (1-aminopiperidin-4-yl) -2-chloro-7-methyl-7, 9-dihydro-8H-purin-8-one (150 mg), 4- (2-bromoethoxy) -7-methyl-6-nitroquinoline (200 mg) and cesium carbonate (326 mg) were dissolved in acetonitrile (10 mL) under nitrogen, heated to 80℃and stirred for 3 hours. The reaction solution was cooled to room temperature, filtered, and the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developer: dichloromethane: methanol=15:1 (V: V)) to give the objective compound (110 mg).
Step 4:9- (1- ((2- ((6-amino-7-methylquinolin-4-yl) oxy) ethyl) amino) piperidin-4-yl) -2-chloro-7-methyl-7, 9-dihydro-8H-purin-8-one
To a solution of 2-chloro-7-methyl-9- (1- ((2- ((7-methyl-6-nitroquinolin-4-yl) oxy) ethyl) amino) piperidin-4-yl) -7, 9-dihydro-8H-purin-8-one (110 mg) in acetic acid (10 mL) at room temperature was added reduced iron powder (110 mg), followed by stirring at 25 ℃ for 3 hours. The reaction solution was filtered through celite, and the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: dichloromethane: methanol=5:1 (V: V)) to give the title compound (70 mg).
Step 5:2 7,4 7 -dimethyl-2 8,2 9 -dihydro-2 7 H-5-oxa-3, 8-diaza-4 (6, 4) -quinolin-2 (9, 2) -purin-1 (4, 1) -piperidinecyclooctan-2- 8 -one
The above 9- (1- ((2- ((6-amino-7-methylquinolin-4-yl) oxy) ethyl) amino) piperidin-4-yl) -2-chloro-7-methyl-7, 9-dihydro-8H-purin-8-one (48 mg), ruPhos Pd G (9 mg) and cesium carbonate (65 mg) were dissolved in dioxane (10 mL) under nitrogen, heated to 100 ℃ and stirred for 3 hours. The reaction solution was cooled to room temperature, filtered, and the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developer: dichloromethane: methanol=15:1 (V: V)) to give the objective compound (21 mg).
MS(ESI)m/z 447.15(M+H) +
Example 10:2 7,4 7 -dimethyl-2 8,2 9 -dihydro-2 7 H-5-oxa-3, 8-diaza-4 (6, 4) -quinolin-2 (9, 2) -purin-1 (1, 4) -piperidinecyclooctan-2- 8 -one
Step 1: tert-butyl (2- ((7-methyl-6-nitroquinolin-4-yl) oxy) ethyl) carbamate
DIAD (306 mg) was slowly added dropwise to a solution of 7-methyl-6-nitroquinolin-4-ol (204 mg), tert-butyl (2-hydroxyethyl) carbamate (320 mg) and triphenylphosphine (526 mg) in THF (30 mL) at 0℃and stirred at room temperature for 12 hours after the completion of the dropwise addition. Filtration, concentration of the filtrate, and purification of the residue by silica gel column chromatography (eluent: dichloromethane: methanol=20:1 (V: V)) gave a pale yellow solid (300 mg).
Step 2:2- ((7-methyl-6-nitroquinolin-4-yl) oxy) ethane-1-amine trifluoroacetate salt
Tert-butyl (2- ((7-methyl-6-nitroquinolin-4-yl) oxy) ethyl) carbamate (300 mg) was dissolved in dichloromethane (10 mL) at 0 ℃ and trifluoroacetic acid (3 mL) was slowly added thereto. The reaction mixture was stirred at room temperature for 1 hour, the reaction solution was concentrated under reduced pressure, and the residue was used in the next reaction (250 mg) without purification.
Step 3: 2-chloro-7-methyl-9- (4- ((2- ((7-methyl-6-nitroquinolin-4-yl) oxy) ethyl) amino) piperidin-1-yl) -7, 9-dihydro-8H-purin-8-one
2- ((7-Methyl-6-nitroquinolin-4-yl) oxy) ethane-1-amine trifluoroacetate (250 mg) and 2-chloro-7-methyl-9- (4-oxopiperidin-1-yl) -7, 9-dihydro-8H-purin-8-one (260 mg) were dissolved in THF (50 mL) at 0 ℃ and sodium triacetoxyborohydride (600 mg) was added thereto. The reaction mixture was stirred at room temperature for 12 hours, the reaction solution was poured into a saturated aqueous sodium bicarbonate solution, ethyl acetate was added for extraction, the extract was concentrated under reduced pressure, and the residue was purified by column chromatography (eluent: dichloromethane: methanol=10:1 (V: V)) to give the title compound (250 mg).
Step 4:9- (4- ((2- ((6-amino-7-methylquinolin-4-yl) oxy) ethyl) amino) piperidin-1-yl) -2-chloro-7-methyl-7, 9-dihydro-8H-purin-8-one
To a solution of 2-chloro-7-methyl-9- (4- ((2- ((7-methyl-6-nitroquinolin-4-yl) oxy) ethyl) amino) piperidin-1-yl) -7, 9-dihydro-8H-purin-8-one (250 mg) in acetic acid (15 mL) at room temperature was added reduced iron powder (500 mg), followed by stirring at 25 ℃ for 2 hours. The reaction solution was filtered through celite, and the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: dichloromethane: methanol=5:1 (V: V)) to give the title compound (150 mg).
Step 5:2 7,4 7 -dimethyl-2 8,2 9 -dihydro-2 7 H-5-oxa-3, 8-diaza-4 (6, 4) -quinolin-2 (9, 2) -purin-1 (1, 4) -piperidinecyclooctan-2- 8 -one
The above 9- (4- ((2- ((6-amino-7-methylquinolin-4-yl) oxy) ethyl) amino) piperidin-1-yl) -2-chloro-7-methyl-7, 9-dihydro-8H-purin-8-one (48 mg), ruPhos Pd G (9 mg) and cesium carbonate (65 mg) were dissolved in dioxane (10 mL) under nitrogen, heated to 100 ℃ and stirred for 3 hours. The reaction solution was cooled to room temperature, filtered, and the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developer: dichloromethane: methanol=15:1 (V: V)) to give the objective compound (24 mg).
MS(ESI)m/z 447.17(M+H) +
Example 11:4- (7-methyl-2- ((7-methyl-2- (trifluoromethyl) - [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Step 1: 2-trifluoromethyl-7-methyl-6-nitro- [1,2,4] triazolo [1,5-a ] pyridine
2-Chloro-4-methyl-5-nitropyridine (172 mg), 5-trifluoromethyl-1, 3, 4-thiadiazol-2-amine (168 mg) and DIEA (390 mg) were dissolved in toluene (10 mL) under nitrogen, and the mixture was stirred for 48 hours after heating to 150 ℃. The reaction solution was cooled to room temperature, concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: dichloromethane: methanol=20:1 (V: V)) to give the title compound (30 mg).
Step 2: 2-trifluoromethyl-7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-amine
To an acetic acid (5 mL) solution of 2-trifluoromethyl-7-methyl-6-nitro- [1,2,4] triazolo [1,5-a ] pyridine (30 mg) at room temperature was added reduced iron powder (0.2 g), and the mixture was stirred at 40℃for 1 hour. After cooling, the reaction solution was poured into 50mL of water and extracted with ethyl acetate; the extract was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: dichloromethane: methanol=20:1 (V: V)) to give the title compound (18 mg).
Step 3:4- (7-methyl-2- ((-methyl-2- (trifluoromethyl) - [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
2-Trifluoromethyl-7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-amine (18 mg), 4- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile (25 mg), ruPhos Pd G (9 mg) and cesium carbonate (65 mg) were dissolved in dioxane (10 mL) under nitrogen, heated to 100℃and stirred for 2 hours. The reaction solution was cooled to room temperature, filtered, and the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developer: dichloromethane: methanol=20:1 (V: V)) to give the objective compound (13 mg).
1H NMR(400MHz,DMSO-d 6)9.31(s,1H),8.91(s,1H),8.22(s,1H),7.85(s,1H),3.87-3.96(m,2H),3.52-3.62(m,2H),3.30(s,3H),2.61-2.76(m,4H),2.43(s,3H).
Example 12:4- (2- ((2- (difluoromethyl) -7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
The present compound (15 mg) was synthesized following the synthesis procedure of example 11 using 5- (difluoromethyl) thiazol-2-amine as a starting material.
1H NMR(400MHz,DMSO-d 6)9.20(s,1H),8.86(s,1H),8.20(s,1H),7.77(s,1H),7.22(t,J=52.8Hz,1H),3.88-3.94(m,2H),3.52-3.60(m,2H),3.29(s,3H),2.67-2.76(m,2H),2.59-2.66(m,2H),2.40(s,3H).
Example 13:4- (2- ((2-amino-7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Step 1:1- (4-methyl-5-nitropyridin-2-yl) guanidine
2-Chloro-4-methyl-5-nitropyridine (1.72 g), guanidine hydrochloride (10 g) and potassium carbonate (30 g) were dissolved in t-butanol (100 mL) under nitrogen, heated to 90℃and stirred for 48 hours. Cooled to room temperature, the reaction solution was poured into 150mL of water, and the solid was filtered and dried and used directly in the next step.
Step 2: 7-methyl-6-nitro- [1,2,4] triazolo [1,5-a ] pyridin-2-amine
To a solution of 1- (4-methyl-5-nitropyridin-2-yl) guanidine (390 mg) in methanol (50 mL) at room temperature was added NCS (300 mg), followed by stirring at 40℃for 0.5 hours. Then, a potassium carbonate solution (600 mg in 10mL of water) was added at this temperature, stirring was continued for 1 hour, and then, the mixture was cooled to room temperature, and the reaction solution was poured into 50mL of water and extracted with ethyl acetate; the extract was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: dichloromethane: methanol=15:1 (V: V)) to give the title compound (180 mg).
Step 3: 7-methyl- [1,2,4] triazolo [1,5-a ] pyridine-2, 6-diamine
To a solution of 7-methyl-6-nitro- [1,2,4] triazolo [1,5-a ] pyridin-2-amine (180 mg) in acetic acid (15 mL) at room temperature was added reduced iron powder (0.5 g), followed by stirring at 40℃for 1 hour. The reaction solution was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: dichloromethane: methanol=10:1 (V: V)) to give the title compound (100 mg).
Step 4:4- (2- ((2-amino-7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
The above 7-methyl- [1,2,4] triazolo [1,5-a ] pyridine-2, 6-diamine (16 mg), 4- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile (29 mg), ruPhos Pd G (9 mg) and cesium carbonate (65 mg) were dissolved in dioxane (10 mL) under nitrogen, heated to 100℃and stirred for 2 hours. The reaction solution was cooled to room temperature, filtered, and the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developer: dichloromethane: methanol=15:1 (V: V)) to give the objective compound (14 mg).
MS(ESI)m/z 421.13(M+H) +
Example 14: (1 1s,1 4 s) -7, 7-difluoro-2 7,4 7 -dimethyl-2 8,2 9 -dihydro-2 7 H-5-oxa-3, 9-diaza-4 (6, 4) -quinolin-2 (9, 2) -purin-1 (1, 4) -cyclohexacyclononan-2 8 -one
Step 1: tert-butyl (2, 2-difluoro-3- ((7-methyl-6-nitroquinolin-4-yl) oxy) propyl) carbamate
DIAD (306 mg) was slowly added dropwise to a solution of 7-methyl-6-nitroquinolin-4-ol (204 mg), tert-butyl (2, 2-difluoro-3-hydroxypropyl) carbamate (422 mg) and triphenylphosphine (526 mg) in THF (30 mL) at 0℃and the reaction was warmed to room temperature after the addition and stirring was continued for 12 hours. Filtration, concentration of the filtrate, and purification of the residue by silica gel column chromatography (eluent: dichloromethane: methanol=20:1 (V: V)) gave a pale yellow solid (310 mg).
Step 2:2, 2-difluoro-3- ((7-methyl-6-nitroquinolin-4-yl) oxy) propan-1-amine trifluoroacetate
Tert-butyl (2, 2-difluoro-3- ((7-methyl-6-nitroquinolin-4-yl) oxy) propyl) carbamate (310 mg) was dissolved in dichloromethane (10 mL) at 0 ℃ and trifluoroacetic acid (3 mL) was slowly added thereto. The reaction mixture was stirred at room temperature for 1 hour, the reaction solution was concentrated under reduced pressure, and the residue was used in the next reaction without purification (290 mg).
Step 3: 2-chloro-9- ((1 s,4 s) -4- ((2, 2-difluoro-3- ((7-methyl-6-nitroquinolin-4-yl) oxy) propyl) amino) cyclohexyl) -7-methyl-7, 9-dihydro-8H-purin-8-one
2-Chloro-7-methyl-9- (4-oxocyclohexyl) -7, 9-dihydro-8H-purin-8-one (141 mg) and 2, 2-difluoro-3- ((7-methyl-6-nitroquinolin-4-yl) oxy) propan-1-amine trifluoroacetate (290 mg) were dissolved in THF (50 mL) at 0℃and sodium triacetoxyborohydride (500 mg) was added thereto. The reaction mixture was stirred at room temperature for 12 hours, the reaction solution was poured into a saturated aqueous sodium bicarbonate solution, ethyl acetate was added for extraction, the extract was concentrated under reduced pressure, and the residue was purified by column chromatography (eluent: dichloromethane: methanol=10:1 (V: V)) to give the title compound (170 mg).
Step 4:9- ((1 s,4 s) -4- ((3- ((6-amino-7-methylquinolin-4-yl) oxy) -2, 2-difluoropropyl) amino) cyclohexyl) -2-chloro-7-methyl-7, 9-dihydro-8H-purin-8-one
To a solution of 2-chloro-9- ((1 s,4 s) -4- ((2, 2-difluoro-3- ((7-methyl-6-nitroquinolin-4-yl) oxy) propyl) amino) cyclohexyl) -7-methyl-7, 9-dihydro-8H-purin-8-one (170 mg) in acetic acid (30 mL) at room temperature was added reduced iron powder (0.3 g), followed by stirring at 25 ℃ for 2 hours. The reaction solution was filtered through celite, and the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: dichloromethane: methanol=5:1 (V: V)) to give the title compound (110 mg).
Step 5: (1 1s,1 4 s) -7, 7-difluoro-2 7,4 7 -dimethyl-2 8,2 9 -dihydro-2 7 H-5-oxa-3, 9-diaza-4 (6, 4) -quinolin-2 (9, 2) -purin-1 (1, 4) -cyclohexacyclononan-2 8 -one
The above 9- ((1 s,4 s) -4- ((3- ((6-amino-7-methylquinolin-4-yl) oxy) -2, 2-difluoropropyl) amino) cyclohexyl) -2-chloro-7-methyl-7, 9-dihydro-8H-purin-8-one (53 mg), ruPhos Pd G (9 mg) and cesium carbonate (65 mg) were dissolved in dioxane (10 mL) under nitrogen, heated to 100 ℃ and stirred for 3 hours. The reaction solution was cooled to room temperature, filtered, and the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developer: dichloromethane: methanol=15:1 (V: V)) to give the objective compound (21 mg).
1H NMR(400MHz,DMSO-d 6)8.55(d,J=4.8Hz,1H),8.42(s,1H),8.18-8.21(m,2H),7.77(s,1H),7.02(d,J=5.6Hz,1H),4.74(t,J=7.2Hz,2H),3.97-4.07(m,1H),3.31(s,3H),3.21-3.28(m,1H),2.98-3.11(m,2H),2.70-2.76(m,1H),2.40-2.57(m,5H),1.74-1.85(m,2H),1.54-1.62(m,2H),1.39-1.50(m,2H).
Example 15: (1 1R,1 4 R, 7S) -7- (tert-butyl) -2 7,4 7 -dimethyl-2 8,2 9 -dihydro-2 7 H-5-oxa-3, 8-diaza-4 (6, 4) -quinolin-2 (9, 2) -purin-1 (1, 4) -cyclohexanecyclooctan-2 8 -one
The present compound (30 mg) was synthesized following the synthesis procedure of example 14 using tert-butyl (S) - (1-hydroxy-3, 3-dimethylbutan-2-yl) carbamate as an alternative starting material.
1H NMR(400MHz,CDCl 3)8.79(s,1H),8.61(d,J=5.2Hz,1H),7.88(s,2H),7.41(s,1H),6.76(d,J=5.2Hz,1H),4.18-4.40(m,6H),3.40(s,3H),3.04-3.16(m,2H),2.78(d,J=5.2Hz,1H),2.55(s,3H),2.00-2.24(m,3H),1.71-1.98(m,4H),1.46-1.58(m,1H),0.99(s,6H).
Example 16: (1 1R,1 4S,6 1R,6 2S)-2 7,4 7 -dimethyl-2 8,2 9 -dihydro-2 7 H-5-oxa-3, 7-diaza-4 (6, 4) -quinolin-2 (9, 2) -purin-1 (1, 4) -cyclohexane-6 (1, 2) -cyclopentanyl-cycloheptan-2 8 -one
The present compound (22 mg) was synthesized following the synthesis method of example 14 using tert-butyl ((1 s,2 s) -2-hydroxycyclopentyl) carbamate as a substitute starting material.
1H NMR(400MHz,CDCl 3)8.60(s,1H),8.55(d,J=5.2Hz,1H),7.92(s,1H),7.86(s,1H),7.30(s,1H),6.71(d,J=5.6Hz,1H),4.84-4.88(m,1H),4.34-4.43(m,1H),3.41-3.48(m,4H),2.99-3.04(m,1H),2.81-2.92(m,1H),2.58(s,3H),2.37-2.48(m,1H),2.23-2.33(m,1H),2.07-2.15(m,1H),1.96-2.06(m,2H),1.51-1.94(m,9H).
Example 17: (1 1S,1 4 S, 7R) -7- (tert-butyl) -2 7,4 7 -dimethyl-2 8,2 9 -dihydro-2 7 H-5-oxa-3, 8-diaza-4 (6, 4) -quinolin-2 (9, 2) -purin-1 (1, 4) -cyclohexanecyclooctan-2 8 -one
The present compound (15 mg) was synthesized following the synthesis procedure of example 1 using tert-butyl (R) - (1-hydroxy-3, 3-dimethylbutan-2-yl) carbamate as an alternative starting material.
1H NMR(400MHz,CDCl 3)8.86(s,1H),8.61(d,J=6.0Hz,1H),7.99-8.05(br,1H),7.92(s,1H),6.83(d,J=6.0Hz,1H),4.39(d,J=4.8Hz,2H),4.23-4.33(m,1H),3.42(s,3H),3.05-3.18(m,2H),2.81(t,J=5.2Hz,1H),2.49-2.63(m,4H),2.18-2.26(m,1H),2.06-2.14(m,1H),1.92-2.03(m,2H),1.50-1.86(m,4H),1.00(s,9H).
Example 18: (1 1s,1 4s)-2 7,4 7 -dimethyl-2 8,2 9 -dihydro-2 7 H-5-thia-3, 8-diaza-4 (6, 4) -quinolin-2 (9, 2) -purin-1 (1, 4) -cyclohexanecyclooctan-2- 8 -one
Step 1: tert-butyl (2- ((7-methyl-6-nitroquinolin-4-yl) thio) ethyl) carbamate
Sodium hydride (160 mg,60% mineral oil) was added to a solution of tert-butyl (2-mercaptoethyl) carbamate (354 mg) in DMF (30 mL) at 0deg.C, and maintained at 0deg.C for 10 minutes after the addition was complete, then 4-chloro-7-methyl-6-nitroquinoline (223 mg) was added, the reaction was warmed to room temperature and stirring was continued for 2 hours. The reaction solution was poured into water to cause precipitation. The solid was filtered and washed with water to give a pale yellow solid (250 mg).
Steps 2, 3, 4, 5 the procedure of steps 2, 3, 4, 5 in the synthesis of example 14 was followed to synthesize the compound (23 mg).
1H NMR(400MHz,CDCl 3)8.92(s,1H),8.60(d,J=4.8Hz,1H),7.92(s,1H), 7.90(s,1H),7.36(d,J=4.8Hz,1H),6.98(s,1H),4.23-4.32(m,1H),3.38-3.46(m,4H),2.85-2.92(m,3H),2.60-2.74(m,2H),2.53(s,3H),1.84-1.92(m,2H),1.68-1.76(m,2H),1.52-1.67(m,4H).
Example 19: (1 1s,1 4s)-2 7 -cyclopropyl-4 7 -methyl-2 8,2 9 -dihydro-2 7 H-5-oxo-3, 8-diaza-4 (6, 4) -quinolin-2 (9, 2) -purin-1 (1, 4) -cyclohexanecyclooctan-2 8 -one
The present compound (40 mg) was synthesized following the synthesis procedure of example 14 using tert-butyl (2-hydroxyethyl) carbamate and intermediate 6 as alternatives.
1H NMR(400MHz,CDCl 3)8.45-8.72(m,2H),8.09(s,1H),7.87(s,1H),7.32(s,1H),6.71(d,J=4.4Hz,1H),4.34-4.46(m,3H),3.05-3.12(m,3H),2.89-2.94(m,1H),2.49-2.62(m,5H),2.11-2.20(m,2H),1.60-1.78(m,5H),1.08-1.15(m,2H),1.01-1.06(m,2H).
Example 20: (1 1R,1 4R,6 1S,6 2S)-2 7,4 7 -dimethyl-2 8,2 9 -dihydro-2 7 H-5-oxa-3, 7-diaza-4 (6, 4) -quinolin-2 (9, 2) -purin-1 (1, 4) -cyclohexane-6 (1, 2) -cyclopentanyl-cycloheptan-2 8 -one
The present compound (45 mg) was synthesized following the synthesis method of example 14 using tert-butyl (1S, 2R) -2-hydroxycyclopentyl carbamate (CAS: 913631-66-0) as a substitute starting material.
1H NMR(400MHz,DMSO-d 6)8.49(d,J=5.6Hz,1H),8.48(s,1H),8.22(s,1H),7.96(s,1H),7.76(s,1H),6.89(d,J=5.6Hz,1H),4.16-4.28(m,2H),3.32(s,3H),3.20-3.28(m,1H),2.96-3.02(m,1H),2.46-2.58(m,4H),2.14-2.37(m,2H),1.36-1.98(m,11H),1.10-1.22(m,1H).
Example 21: (1 1s,1 4s)-2 7,4 7 -dimethyl-2 8,2 9 -dihydro-2 7 H-3, 8-diaza-4 (6, 4) -quinolin-2 (9, 2) -purin-1 (1, 4) -cyclohexanecyclooctan-5-yn-2 8 -one
Step 1: tert-butyl (3- (7-methyl-6-nitroquinolin-4-yl) prop-2-yn-1-yl) carbamate
A mixture of 4-chloro-7-methyl-6-nitroquinoline (223 mg), N-Boc-aminopropyne (620 mg), palladium acetate (22 mg), BINAP (125 mg), potassium carbonate (276 mg) and toluene (30 mL) was heated to reflux and stirred for 12 hours under nitrogen. Cooled to room temperature, filtered, the filtrate concentrated, and the residue purified by silica gel column chromatography (eluent: dichloromethane: methanol=20:1 (V: V)) to give a pale yellow solid (170 mg).
Step 2:3- (7-methyl-6-nitroquinolin-4-yl) propyl-2-yn-1-amine trifluoroacetate salt
Tert-butyl (3- (7-methyl-6-nitroquinolin-4-yl) prop-2-yn-1-yl) carbamate (170 mg) was dissolved in dichloromethane (10 mL) at 0 ℃ and trifluoroacetic acid (3 mL) was slowly added thereto. The mixture was stirred at room temperature for 1 hour, concentrated under reduced pressure, and the residue was used in the next reaction (180 mg) without purification.
Step 3: 2-chloro-7-methyl-9- ((1 s,4 s) -4- ((3- (7-methyl-6-nitroquinolin-4-yl) prop-2-yn-1-yl) amino) cyclohexyl) -7, 9-dihydro-8H-purin-8-one
2-Chloro-7-methyl-9- (4-oxocyclohexyl) -7, 9-dihydro-8H-purin-8-one (141 mg) and 3- (7-methyl-6-nitroquinolin-4-yl) propyl-2-yn-1-amine trifluoroacetate (180 mg) were dissolved in THF (50 mL) at 0deg.C, to which was added sodium triacetoxyborohydride (400 mg). The mixture was stirred at room temperature for 12 hours, poured into a saturated aqueous sodium bicarbonate solution, extracted with ethyl acetate, and the extract was concentrated under reduced pressure, and the residue was purified by column chromatography (eluent: dichloromethane: methanol=10:1 (V: V)) to give the title compound (200 mg).
Step 4:9- ((1 s,4 s) -4- ((3- (6-amino-7-methylquinolin-4-yl) prop-2-yn-1-yl) amino) cyclohexyl) -2-chloro-7-methyl-7, 9-dihydro-8H-purin-8-one
To a solution of 2-chloro-7-methyl-9- ((1 s,4 s) -4- ((3- (7-methyl-6-nitroquinolin-4-yl) prop-2-yn-1-yl) amino) cyclohexyl) -7, 9-dihydro-8H-purin-8-one (200 mg) in acetic acid (15 mL) at room temperature was added reduced iron powder (0.4 g), followed by stirring at 25 ℃ for 2 hours. Filtration through celite, concentration of the filtrate under reduced pressure, and purification of the residue by silica gel column chromatography (eluent: dichloromethane: methanol=5:1 (V: V)) gave the title compound (150 mg).
Step 5: (1 1s,1 4s)-2 7,4 7 -dimethyl-2 8,2 9 -dihydro-2 7 H-3, 8-diaza-4 (6, 4) -quinolin-2 (9, 2) -purin-1 (1, 4) -cyclohexanecyclooctan-5-yn-2 8 -one
9- ((1 S,4 s) -4- ((3- (6-amino-7-methylquinolin-4-yl) prop-2-yn-1-yl) amino) cyclohexyl) -2-chloro-7-methyl-7, 9-dihydro-8H-purin-8-one (48 mg), ruPhos Pd G (9 mg) and cesium carbonate (65 mg) were dissolved in dioxane (10 mL) under nitrogen, heated to 100℃and stirred for 3 hours. The reaction solution was cooled to room temperature, filtered, and the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developer: dichloromethane: methanol=15:1 (V: V)) to give the objective compound (21 mg).
1H NMR(400MHz,CDCl 3)8.69(s,1H),8.64(d,J=4.4Hz,1H),7.91(s,2H),7.37(d,J=4.4Hz,1H),7.20(s,1H),4.17-4.28(m,1H),3.92(s,2H),3.34-3.48(m,4H),3.13(s,1H),2.72-2.86(m,2H),2.59(s,3H),2.18-2.34(m,2H),1.96-2.14(m,4H).
Example 22: (1 1S,1 4s)-2 7,4 7 -dimethyl-2 8,2 9 -dihydro-2 7 H-3, 8-diaza-4 (6, 4) -quinolin-2 (9, 2) -purin-1 (1, 4) -cyclohexanecyclooctane-2 8 -one
To a solution of (1 1s,1 4s)-2 7,4 7 -dimethyl-2 8,2 9 -dihydro-2 7 H-3, 8-diaza-4 (6, 4) -quinolin-2 (9, 2) -purin-1 (1, 4) -cyclohexanecyclooctan-5-yn-2- 8 -one (20 mg) in methanol (5 mL) was added 10% palladium on charcoal (20 mg) at room temperature, followed by stirring in a hydrogen atmosphere at 2 atmospheres for 24 hours.
1H NMR(400MHz,CDCl 3)8.84(s,1H),8.64(d,J=4.4Hz,1H),7.92(s,1H),7.90(s,1H),7.21(d,J=4.4Hz,1H),7.16(s,1H),4.33-4.43(m,1H),3.43(s,3H),3.39(t,J=7.2Hz,2H),2.88-3.00(m,3H),2.72-2.75(m,2H),2.57(s,3H),1.83-1.98(m,4H),1.60-1.71(m,4H).
Example 23:1 7,3 7 -dimethyl-3 8,3 9 -dihydro-3 7 H-8-oxa-2-aza-1 (6, 4) -quinolin-3 (2, 9) -purinocyclooctan-3- 8 -one
Step 1:4- (4-bromobutoxy) -7-methyl-6-nitroquinoline
DIAD (408 mg) was slowly added dropwise to a solution of 7-methyl-6-nitroquinolin-4-ol (204 mg), 4-bromo-n-butanol (306 mg) and triphenylphosphine (516 mg) in toluene (20 mL) at 0℃and, after the addition was completed, the mixture was warmed to room temperature and stirred for 12 hours. Filtration, concentration of the filtrate under reduced pressure, and purification of the residue by silica gel column chromatography (eluent: petroleum ether: ethyl acetate=2:1 (V: V)) gave a pale yellow oil (300 mg).
Step 2: 2-chloro-7-methyl-9- (4- ((7-methyl-6-nitroquinolin-4-yl) oxy) butyl) -7, 9-dihydro-8H-purin-8-one
4- (4-Bromobutoxy) -7-methyl-6-nitroquinoline (300 mg), 2-chloro-7-methyl-7, 9-dihydro-8H-purin-8-one (184 mg) and potassium carbonate (276 mg) were dissolved in DMF (10 mL), heated to 100℃and stirred for 3 hours. Cooled to room temperature, the reaction solution was poured into ice water to give a solid, which was used in the next step (250 mg) without purification after filtration and drying.
Step 3:9- (4- ((6-amino-7-methylquinolin-4-yl) oxy) butyl) -2-chloro-7-methyl-7, 9-dihydro-8H-purin-8-one
To a solution of 2-chloro-7-methyl-9- (4- ((7-methyl-6-nitroquinolin-4-yl) oxy) butyl) -7, 9-dihydro-8H-purin-8-one (250 mg) in acetic acid (15 mL) at room temperature was added reduced iron powder (0.5 g), followed by stirring at 35℃for 1 hour. Cooling to room temperature, pouring the reaction solution into 50mL of water, and extracting with ethyl acetate; the extract was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: dichloromethane: methanol=20:1 (V: V)) to give a pale yellow solid (200 mg).
Step 4:1 7,3 7 -dimethyl-3 8,3 9 -dihydro-3 7 H-8-oxa-2-aza-1 (6, 4) -quinolin-3 (2, 9) -purinocyclooctan-3- 8 -one
9- (4- ((6-Amino-7-methylquinolin-4-yl) oxy) butyl) -2-chloro-7-methyl-7, 9-dihydro-8H-purin-8-one (41 mg), ruPhos Pd G (9 mg) and cesium carbonate (65 mg) were dissolved in dioxane (10 mL) under nitrogen, heated to 100℃and stirred for 2 hours. Cooled to room temperature, filtered, and the filtrate concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developer: dichloromethane: methanol=30:1 (V: V)) to give the objective compound (15 mg).
1H NMR(400MHz,CDCl 3)9.45(s,1H),8.65(d,J=5.2Hz,1H),7.91(s,1H),7.85(s,1H),7.07(s,1H),6.90(d,J=5.2Hz,1H),4.32(t,J=5.6Hz,2H),3.90-3.94(m,2H),3.43(s,3H),2.54(s,3H),2.29-2.37(m,2H),1.92-1.98(m,2H).
Example 24: (1 1S,1 4S)-2 7,4 7 -dimethyl-2 8,2 9 -dihydro-2 7 H-5-mercapto-3, 9-diaza-4 (6, 4) -quinolin-2 (9, 2) -purin-1 (1, 4) -cyclohexanecyclononan-2 8 -one
Step 1: tert-butyl (2- ((7-methyl-6-nitroquinolin-4-yl) thio) propyl) carbamate
Sodium hydride (80 mg,60% mineral oil) was added to a solution of tert-butyl (2-mercaptopropyl) carbamate (191 mg) in DMF (10 mL) at 0deg.C, and maintained at 0deg.C for 10 minutes after the addition was complete, then 4-chloro-7-methyl-6-nitroquinoline (223 mg) was added, the reaction was warmed to room temperature and stirring was continued for 2 hours. The reaction solution was poured into water to cause precipitation. Filtered, washed with water and dried to give a pale yellow solid (350 mg).
Steps 2, 3, 4, 5 the procedure of steps 2, 3, 4, 5 in the synthesis of example 21 was followed to synthesize the compound (23 mg).
1H NMR(400MHz,CDCl 3)8.65(d,J=4.4Hz,1H),8.52(s,1H),7.95(s,2H),7.11(d,J=4.4Hz,1H),6.98(s,1H),4.19-4.28(m,1H),3.38-3.46(m,5H),3.10-3.34(m,3H),2.54(s,3H),2.24-2.52(m,6H),1.95-2.06(m,2H),1.78-1.90(m, 2H).
Example 25: (1 1s,1 4s)-2 7,4 7 -dimethyl-2 8,2 9 -dihydro-2 7 H-5-thia-5, 5-dioxo-3, 8-diaza-4 (6, 4) -quinolin-2 (9, 2) -purin-1 (1, 4) -cyclohexanecyclooctan-2 8 -one
(1 1s,1 4s)-2 7,4 7 -Dimethyl-2 8,2 9 -dihydro-2 7 H-5-thia-3, 8-diaza-4 (6, 4) -quinolin-2 (9, 2) -purin-1 (1, 4) -cyclohexanecyclooctan-2- 8 -one (46 mg) was dissolved in methylene chloride (20 mL) at room temperature, m-chloroperoxybenzoic acid (40 mg) was slowly added and stirred for 2 hours.
1H NMR(400MHz,CDCl 3)8.88(d,J=4.4Hz,1H),8.57(s,1H),7.98-8.01(m,2H),7.96(s,1H),7.21(s,1H),5.26-5.38(br,1H),4.49-4.58(m,1H),3.66-3.73(m,1H),3.46(s,3H),3.07-3.21(m,3H),2.93-2.98(m,1H),2.82-2.88(m,1H),2.69-2.81(m,1H),2.61(s,3H),2.38-2.47(m,1H),2.18-2.28(m,1H),1.78-1.93(m,2H),1.52-1.66(m,2H).
Example 26: (1 1r,1 4r)-2 7,4 7, 7-trimethyl-2 8,2 9 -dihydro-2 7 H-5-oxa-3, 7-diaza-4 (6, 4) -quinolin-2 (9, 2) -purin-1 (1, 4) -cyclohexane-6 (1, 3) -cyclobutanecycloheptan-2 8 -one
(1 1r,1 4r)-2 7,4 7 -Dimethyl-2 8,2 9 -dihydro-2 7 H-5-oxa-3, 7-diaza-4 (6, 4) -quinolin-2 (9, 2) -purin-1 (1, 4) -cyclohexan-6 (1, 3) -cyclobutanecycloheptan-2 8 -one (47 mg) was dissolved in methanol (20 mL) at room temperature, aqueous formaldehyde solution (0.05 mL, 37%) and sodium triacetoxyborohydride (50 mg) were slowly added and stirred for 2 hours.
1H NMR(400MHz,CDCl 3)8.60(d,J=5.2Hz,1H),8.50(s,1H),7.94(s,1H),7.90(s,1H),6.94(s,1H),6.48(d,J=5.2Hz,1H),4.84-4.90(m,1H),4.29-4.39(m,1H),3.54-3.73(m,2H),3.41(s,3H),3.08-3.24(m,2H),2.64-2.78(m,2H),2.57(s,3H),2.42-2.54(m,5H),1.86-1.95(m,2H),1.54-1.77(m,4H).
Example 27: (1 1s,1 4s)-2 7,4 7, 8-trimethyl-2 8,2 9 -dihydro-2 7 H-5-oxa-3, 8-diaza-4 (6, 4) -quinolin-2 (9, 2) -purin-1 (1, 4) -cyclohexanecyclooctan-2- 8 -one
(1 1s,1 4s)-2 7,4 7 -Dimethyl-2 8,2 9 -dihydro-2 7 H-5-oxa-3, 8-diaza-4 (6, 4) -quinolin-2 (9, 2) -purin-1 (1, 4) -cyclohexanecyclooctan-2- 8 -one (45 mg) was dissolved in methanol (20 mL) at room temperature, an aqueous formaldehyde solution (0.05 mL, 37%) and sodium triacetoxyborohydride (50 mg) were slowly added and stirred for 2 hours.
1H NMR(400MHz,CDCl 3)8.95(s,1H),8.57(d,J=5.2Hz,1H),7.93(s,1H),7.88(s,1H),7.32(s,1H),6.72(d,J=5.2Hz,1H),4.41-4.50(m,1H),4.31(t,J=5.2Hz,2H),3.44(s,3H),2.76-2.92(m,2H),2.58(s,3H),2.26-2.39(m,3H),2.19(s,3H),1.58-1.72(m,6H).
Example 28: (1 1r,1 4r)-2 7,4 7, 8-trimethyl-2 8,2 9 -dihydro-2 7 H-5-oxa-3, 8-diaza-4 (6, 4) -quinolin-2 (9, 2) -purin-1 (1, 4) -cyclohexanecyclooctan-2- 8 -one
(1 1r,1 4r)-2 7,4 7 -Dimethyl-2 8,2 9 -dihydro-2 7 H-5-oxa-3, 8-diaza-4 (6, 4) -quinolin-2 (9, 2) -purin-1 (1, 4) -cyclohexanecyclooctan-2- 8 -one (45 mg) was dissolved in methanol (20 mL) at room temperature, an aqueous formaldehyde solution (0.05 mL, 37%) and sodium triacetoxyborohydride (50 mg) were slowly added and stirred for 2 hours.
1H NMR(400MHz,CDCl 3)8.97(s,1H),8.59(d,J=5.2Hz,1H),7.97(s,1H),7.94(s,1H),7.35(s,1H),6.76(d,J=5.2Hz,1H),4.41-4.50(m,1H),4.34(t,J=4.8Hz,2H),3.44(s,3H),2.74-2.92(m,2H),2.59(s,3H),2.26-2.39(m,3H),2.19(s,3H),1.58-1.72(m,6H).
Example 29: (1 1s,1 4s)-2 7,4 5 -dimethyl-2 8,2 9 -dihydro-2 7H-4 1 H-5-oxa-3, 8-diaza-2 (9, 2) -purin-4 (6, 1) -benzo [ d ] [1,2,3] triazol-1 (1, 4) -cyclohexanecyclooctan-2 8 -one
Step 1: 5-methyl-6-nitro-1H-benzo [ d ] [1,2,3] triazol-1-ol
To a solution of 5-chloro-2, 4-dinitrotoluene (216 mg) in ethanol (10 mL) at 0deg.C was slowly added dropwise 89% hydrazine hydrate solution (0.5 mL), and the mixture was heated to 80deg.C and stirred for 6 hours. Cooled to room temperature, filtered, the filtrate concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: dichloromethane: methanol=20:1 (V: V)) to give a pale yellow solid (97 mg).
Step 2: tert-butyl (2- ((5-methyl-6-nitro-1H-benzo [ d ] [1,2,3] triazol-1-yl) oxy) carbamic acid ethyl ester
DIAD (204 mg) was slowly added dropwise to a solution of 5-methyl-6-nitro-1H-benzo [ d ] [1,2,3] triazol-1-ol (97 mg), tert-butyl (2-hydroxyethyl) carbamate (162 mg) and triphenylphosphine (263 mg) in THF (30 mL) at 0℃and stirred at room temperature for 12 hours after the completion of the dropwise addition. Filtration, concentration of the filtrate under reduced pressure, and purification of the residue by silica gel column chromatography (eluent: dichloromethane: methanol=20:1 (V: V)) gave a pale yellow solid (150 mg)
Steps 3, 4, 5, 6 the procedure of steps 2, 3, 4, 5 in the synthesis of example 21 was followed to synthesize the compound (23 mg).
1H NMR(400MHz,CDCl 3)8.68(s,1H),7.95(s,1H),7.77(s,1H),7.32(s,1H),4.62-4.69(m,2H),4.37-4.47(m,1H),3.44(s,3H),3.02-3.08(m,2H),2.85-3.01(m,3H),2.52(s,3H),1.92-2.03(m,2H),1.63-1.78(m,4H).
Example 30: (1 1s,1 4s)-2 7,4 7 -dimethyl-2 8,2 9 -dihydro-2 7 H-5-oxa-3, 8-diaza-4 (6, 4) -quinolin-2 (9, 2) -purin-1 (1, 4) -cyclohexanecyclooctane-2 8 -thione
To a solution of (1 1s,1 4s)-2 7,4 7 -dimethyl-2 8,2 9 -dihydro-2 7 H-5-oxa-3, 8-diaza-4 (6, 4) -quinolin-2 (9, 2) -purin-1 (1, 4) -cyclohexanecyclooctan-2- 8 -one (45 mg) in toluene (15 mL) was added a lawsen reagent (120 mg) at room temperature, followed by stirring at 130 ℃ for 2 hours in a microwave reactor, methanol (5 mL) was added, followed by filtration through celite, the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developer: dichloromethane: methanol=20:1 (V: V)) to give the title compound (5 mg).
1H NMR(400MHz,CDCl 3)8.78(s,1H),8.65(s,1H),8.58(d,J=5.2Hz,1H),7.87(s,1H),7.50(s,1H),6.71(d,J=5.2Hz,1H),4.37(t,J=4.8Hz,2H),4.07-4.15(m,1H),3.10-3.14(m,3H),2.69-2.80(m,5H),2.60(s,3H),2.19-2.26(m,2H),1.79-1.87(m,2H),1.65-1.76(m,2H).
Example 31: (1 1s,1 4s)-2 3,4 7 -dimethyl-2 2,2 3 -dihydro-2 1 H-5-oxa-3, 8-diaza-4 (6, 4) -quinolin-2 (1, 6) -imidazo [4,5-c ] pyridin-1 (1, 4) -cyclohexanecyclooctan-2 2 -one
Step 1: tert-butyl (2- ((7-methyl-6-nitroquinolin-4-yl) oxy) ethyl) carbamate
DIAD (306 mg) was slowly added dropwise to a solution of 7-methyl-6-nitroquinolin-4-ol (204 mg), tert-butyl (2-hydroxyethyl) carbamate (320 mg) and triphenylphosphine (526 mg) in THF (30 mL) at 0℃and the reaction was warmed to room temperature after the dropwise addition and stirring was continued for 12 hours. The filtrate was concentrated, and the residue was purified by silica gel column chromatography (eluent: dichloromethane: methanol=20:1 (V: V)) to give a pale yellow solid (300 mg).
Step 2:2- ((7-methyl-6-nitroquinolin-4-yl) oxy) ethane-1-amine trifluoroacetate salt
Tert-butyl (2- ((7-methyl-6-nitroquinolin-4-yl) oxy) ethyl) carbamate (300 mg) was dissolved in dichloromethane (10 mL) at 0 ℃ and trifluoroacetic acid (3 mL) was slowly added thereto. The reaction mixture was stirred at room temperature for 1 hour, the reaction solution was concentrated under reduced pressure, and the residue was used in the next reaction (250 mg) without purification.
Step 3: 6-chloro-3-methyl-1- ((1 s,4 s) -4- ((2- ((7-methyl-6-nitroquinolin-4-yl) oxy) ethyl) amino) cyclohexyl) -1, 3-dihydro-2H-imidazo [4,5-c ] pyridin-2-one
2- ((7-Methyl-6-nitroquinolin-4-yl) oxy) ethane-1-amine trifluoroacetate (250 mg) and intermediate 76-chloro-3-methyl-1- (4-oxocyclohexyl) -1, 3-dihydro-2H-imidazo [4,5-c ] pyridin-2-one (280 mg) were dissolved in THF (50 mL) at 0 ℃ and sodium triacetoxyborohydride (600 mg) was added thereto. The reaction mixture was stirred at room temperature for 12 hours, the reaction solution was poured into a saturated aqueous sodium bicarbonate solution, ethyl acetate was added for extraction, the extract was concentrated under reduced pressure, and the residue was purified by column chromatography (eluent: dichloromethane: methanol=10:1 (V: V)) to give the title compound (230 mg).
Step 4:1- ((1 s,4 s) -4- ((2- ((6-amino-7-methylquinolin-4-yl) oxy) ethyl) amino) cyclohexyl) -6-chloro-3-methyl-1, 3-dihydro-2H-imidazo [4,5-c ] pyridin-2-one
To a solution of 6-chloro-3-methyl-1- ((1 s,4 s) -4- ((2- ((7-methyl-6-nitroquinolin-4-yl) oxy) ethyl) amino) cyclohexyl) -1, 3-dihydro-2H-imidazo [4,5-c ] pyridin-2-one (230 mg) in acetic acid (15 mL) at room temperature was added reduced iron powder (500 mg), followed by stirring to 25 ℃ for 2 hours. The reaction solution was filtered through celite, and the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: dichloromethane: methanol=5:1 (V: V)) to give the title compound (160 mg).
Step 5: (1 1s,1 4s)-2 3,4 7 -dimethyl-2 2,2 3 -dihydro-2 1 H-5-oxa-3, 8-diaza-4 (6, 4) -quinolin-2 (1, 6) -imidazo [4,5-c ] pyridin-1 (1, 4) -cyclohexanecyclooctan-2 2 -one
1- ((1 S,4 s) -4- ((2- ((6-amino-7-methylquinolin-4-yl) oxy) ethyl)) amino) cyclohexyl) -6-chloro-3-methyl-1, 3-dihydro-2H-imidazo [4,5-c ] pyridin-2-one (47 mg), ruPhos Pd G (9 mg) and cesium carbonate (65 mg) were dissolved in dioxane (10 mL) under nitrogen, heated to 100℃and stirred for 3 hours. The reaction solution was cooled to room temperature, filtered, and the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developer: dichloromethane: methanol=15:1 (V: V)) to give the objective compound (34 mg).
1H NMR(400MHz,CDCl 3)8.58(d,J=4.8Hz,1H),8.04(s,1H),7.89(s,1H),7.88(s,1H),7.47(s,1H),7.03(s,1H),6.70(d,J=5.2Hz,1H),4.49-4.58(m,1H),4.35-4.39(m,2H),3.45(s,3H),3.09-3.14(m,3H),2.56(s,3H),2.30-2.42(m,2H),2.06-2.14(m,2H),1.68-1.80(m,4H).
Example 32: (2 1s,2 4s)-1 7,5 7 -dimethyl-1 8,1 9 -dihydro-1 7 H-4-oxa-6-aza-5 (4, 6) -quinolin-1 (9, 2) -purin-3 (1, 3) -aza-2 (1, 4) -cyclohexanecyclohexane-1 8 -one
The procedure of example 31 was followed using 3-hydroxyazetidine-1-carboxylic acid tert-butyl ester and 2-chloro-7-methyl-9- (4-oxocyclohexyl) -7, 9-dihydro-8H-purin-8-one as alternatives to give the title compound (14 mg).
1H NMR(400MHz,CDCl 3)8.77(s,1H),8.68(d,J=5.6Hz,1H),7.94(s,1H),7.87(s,1H),7.42(s,1H),6.82(d,J=5.6Hz,1H),5.19(t,J=3.6Hz,1H),4.28-4.38(m,1H),3.41(s,3H),3.34-3.39(m,2H),3.02-3.09(m,2H),2.79-2.91(m,2H),2.57(s,3H),2.44-2.48(m,1H),1.66-1.75(m,2H),1.41-1.52(m,4H).
Example 33:4- (7-methyl-2- ((4-methyl-6- (1H-pyrazol-1-yl) pyridin-3-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Step 1: 4-methyl-5-nitro-2- (1H-pyrazol-1-yl) pyridine
2-Chloro-4-methyl-5-nitropyridine (172 mg), pyrazole (136 mg) and potassium carbonate (415 mg) were dissolved in acetonitrile (10 mL) under nitrogen, heated to 80℃and stirred for 12 hours. The reaction solution was cooled to room temperature, concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: dichloromethane: methanol=20:1 (V: V)) to give the title compound (150 mg).
Step 2: 4-methyl-6- (1H-pyrazol-1-yl) pyridin-3-amine
To a solution of 4-methyl-5-nitro-2- (1H-pyrazol-1-yl) pyridine (150 mg) in acetic acid (15 mL) at room temperature was added reduced iron powder (0.5 g), and the mixture was stirred at 40℃for 1 hour. Pouring the reaction solution into 50mL of water, and extracting with ethyl acetate; the extract was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: dichloromethane: methanol=20:1 (V: V)) to give the title compound (110 mg).
Step 3:4- (7-methyl-2- ((4-methyl-6- (1H-pyrazol-1-yl) pyridin-3-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
4-Methyl-6- (1H-pyrazol-1-yl) pyridin-3-amine (17 mg), 4- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile (29 mg), ruPhos Pd G (9 mg) and cesium carbonate (65 mg) were dissolved in dioxane (10 mL) under nitrogen, heated to 100℃and stirred for 2 hours. The reaction solution was cooled to room temperature, filtered, and the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developer: dichloromethane: methanol=20:1 (V: V)) to give the objective compound (25 mg).
1H NMR(400MHz,CDCl 3)8.79(s,1H),8.52(dd,J=2.8Hz,0.8Hz,1H),7.91(s,1H),7.87(s,1H),7.70-7.72(m,1H),6.59(s,1H),6.45-6.47(m,1H),4.02-4.08(m,2H),3.82-3.89(m,2H),3.39(s,3H),2.72-2.82(m,4H),2.39(s,3H).
Example 34:4- (2- ((4- (1- (difluoromethyl) -1H-pyrazol-4-yl) -2-methylphenyl) amino) -7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Step 1:4- (1- (difluoromethyl) -1H-pyrazol-4-yl) -2-methylaniline
4-Iodo-2-methylaniline (233 mg), (1- (difluoromethyl) -1H-pyrazol-4-yl) boronic acid (200 mg), palladium tetraphenylphosphine (112 mg) and potassium carbonate (278 mg) were dissolved in dioxane (10 mL) and water (2 mL) under nitrogen, and heated to 100℃and stirred for 2 hours. The reaction solution was cooled to room temperature, concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: dichloromethane: methanol=15:1 (V: V)) to give the title compound (120 mg).
Step 2:4- (2- ((4- (1- (difluoromethyl) -1H-pyrazol-4-yl) -2-methylphenyl) amino) -7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
4- (1- (Difluoromethyl) -1H-pyrazol-4-yl) -2-methylaniline (22 mg), 4- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile (29 mg), ruPhos Pd G (9 mg) and cesium carbonate (65 mg) were dissolved in dioxane (10 mL) under nitrogen, heated to 100℃and stirred for 2 hours. The reaction solution was cooled to room temperature, filtered, and the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developer: dichloromethane: methanol=20:1 (V: V)) to give the objective compound (27 mg).
1H NMR(400MHz,DMSO-d 6)9.00(s,1H),8.31(s,1H),8.11(s,1H),7.92(s,1H),7.38(s,1H),7.21(t,J=60.4Hz,1H),6.60(s,1H),4.02-4.09(m,2H),3.82-3.89(m,2H),3.39(s,3H),2.73-2.82(m,4H),2.36(s,3H).
Example 35: (1 1s,1 4s)-2 7,4 6 -dimethyl-2 8 -oxo-2 8,2 9 -dihydro-2 7 H-5-oxo-3, 8-diaza-2 (9, 2) -purine-4 (1, 3) -benzo-1 (1, 4) -cyclohexanone cyclooctane-4 4 -carboxamide
Step 1: tert-butyl (2- (2-bromo-4-methyl-5-nitrophenoxy) ethyl) carbamate
To a solution of tert-butyl 2-bromo-4-methyl-5-nitrophenol (232 mg), (3-hydroxyethyl) carbamate (243 mg) and triphenylphosphine (526 mg) in toluene (10 mL) at 0deg.C was slowly added DIAD (306 mg) dropwise, and the mixture was stirred at room temperature for 12 hours after completion of the dropwise addition. The filtrate was concentrated, and the residue was purified by silica gel column chromatography (eluent: petroleum ether: ethyl acetate=3:1 (V: V)) to give a pale yellow solid (350 mg).
Step 2:2- (2- ((tert-Butoxycarbonyl) amino) ethoxy) -5-methyl-4-nitrobenzoic acid
Tert-butyl (2- (2-bromo-4-methyl-5-nitrophenoxy) ethyl) carbamate (350 mg), [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride (70 mg) and triethylamine (1 mL) were dissolved in methanol (20 mL), and heated to 60 ℃ under a carbon monoxide atmosphere at one atmosphere of pressure and stirred for 12 hours. Cooled to room temperature, 1M aqueous lithium hydroxide (10 mL) was added to the reaction solution and stirred for 5 hours. The reaction mixture was washed with ethyl acetate, and the aqueous phase was adjusted to pH 3 with dilute hydrochloric acid to precipitate. The solid was filtered and dried to give the title compound (120 mg).
Step 3: tert-butyl (2- (2-carbamoyl-4-methyl-5-nitrophenoxy) ethyl) formate
2- (2- ((T-Butoxycarbonyl) amino) ethoxy) -5-methyl-4-nitrobenzoic acid (120 mg), HATU (200 mg) and ammonium chloride (108 mg) were dissolved in DMF (10 mL) at room temperature, DIEA (1 mL) was added and stirred for 6 h. The reaction mixture was poured into water (30 mL) to precipitate out. The solid was filtered, washed with water and dried to give the title compound (90 mg).
Steps 4, 5, 6, 7 the procedure of steps 2, 3, 4, 5 in the synthesis of example 21 was followed to synthesize the compound (11 mg).
1H NMR(400MHz,CDCl3)8.65(s,1H),7.87-7.96(m,3H),7.16(s,1H),5.62(s,1H),4.32-4.46(m,3H),3.42(s,3H),2.95-3.16(m,5H),2.34(s,3H),1.84-1.94(m,2H),1.60-1.70(m,4H).
Example 36: (5 1s,5 4s)-2 7,4 7 -dimethyl-4 8,4 9 -dihydro-4 7 H-3, 6-diaza-2 (4, 6) -quinolin-4 (2, 9) -purin-1 (1, 3) -nitrogen mustard-5 (1, 4) -cyclohexanecycloheptane-4 8 -one
Step 1: tert-butyl ((1- (7-methyl-6-nitroquinolin-4-yl) azetidin-3-yl) methyl) carbamate
Sodium hydride (80 mg,60% mineral oil) was added to a solution of 3-Boc-aminomethylazetidine (186 mg) in DMF (10 mL) at 0deg.C, stirred at 0deg.C for 10 minutes after the completion of the dropwise addition, then 4-chloro-7-methyl-6-nitroquinoline (223 mg) was added, and stirred at room temperature for 1 hour. The reaction solution was poured into water to cause precipitation. The solid was filtered, washed with water and purified by column chromatography to give a pale yellow solid (250 mg).
Steps 2, 3, 4, 5 the procedure of steps 2, 3, 4, 5 in the synthesis of example 21 was followed to synthesize the compound (23 mg).
1H NMR(400MHz,CDCl 3)8.35(s,1H),8.27(s,1H),8.15(d,J=6.4Hz,1H),7.92(s,1H),6.99(s,1H),6.14(d,J=6.4Hz,1H),5.52-5.65(m,1H),4.44-4.64(m,2H),4.30-4.43(m,1H),3.88-4.02(m,1H),3.42(s,3H),2.95-3.22(m,3H),2.90(s,2H),2.51(s,3H),1.54-2.16(m,8H).
Example 37: 7-methyl-2- ((4-methyl-6- (1H-pyrazol-1-yl) pyridin-3-yl) amino) -9- (tetrahydro-2H-pyran-4-yl) -7, 9-dihydro-8H-purin-8-one
4-Methyl-6- (1H-pyrazol-1-yl) pyridin-3-amine (17 mg), 2-chloro-7-methyl-9- (tetrahydro-2H-pyran-4-yl) -7, 9-dihydro-8H-purin-8-one (27 mg), ruPhos Pd G (9 mg) and cesium carbonate (65 mg) were dissolved in dioxane (10 mL) under nitrogen, and heated to 100℃and stirred for 2 hours. The reaction solution was cooled to room temperature, filtered, and the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developer: dichloromethane: methanol=20:1 (V: V)) to give the objective compound (24 mg).
1H NMR(400MHz,CDCl 3)9.01(s,1H),8.52(d,J=2.8Hz,1H),7.86(s,1H),7.84(s,1H),7.71(d,J=1.6Hz,1H),6.58(s,1H),6.44-6.45(m,1H),4.48-4.56(m,1H),4.12(dd,J=11.6Hz,4.4Hz,2H),3.53(t,J=12.0Hz,2H),3.39(s,3H),2.69-2.80(m,2H),2.41(s,3H),1.70-1.74(m,2H).
Example 38: 3-methyl-6- ((7-methylquinoxalin-6-yl) amino) -1- (tetrahydro-2H-pyran-4-yl) -1, 3-dihydro-2H-imidazo [4,5-c ] pyridin-2-one
7-Methylquinoxalin-6-amine (16 mg), 6-chloro-3-methyl-1- (tetrahydro-2H-pyran-4-yl) -1, 3-dihydro-2H-imidazo [4,5-c ] pyridin-2-one (27 mg), ruPhos Pd G (9 mg) and cesium carbonate (65 mg) are dissolved in dioxane (10 mL) under nitrogen, heated to 100deg.C and stirred for 5 hours. The reaction solution was cooled to room temperature, filtered, and the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developer: dichloromethane: methanol=20:1 (V: V)) to give the objective compound (20 mg).
1H NMR(400MHz,CDCl 3)8.91(s,1H),8.69(s,1H),8.60(s,1H),8.16(s,1H),8.08(s,1H),7.82(s,1H),7.37(s,1H),4.38-4.48(m,1H),4.00(dd,J=12.0Hz, 4.0Hz,2H),3.48(t,J=12.0Hz,2H),3.33(s,3H),2.56(s,3H),2.26-2.36(m,2H),1.66-1.70(m,2H).
Example 39: (1 1S,1 4s,7 1S,7 3s)-2 3,4 7 -dimethyl-2 2,2 3 -dihydro-2 1 H-5-oxa-3, 8-diaza-4 (6, 4) -quinolin-2 (1, 6) -imidazo [4,5-c ] pyridin-1 (1, 4) -cyclohexanone-7 (1, 3) -cyclobutanecyclooctane-2 2 -one
The synthesis procedure of example 31 was followed using cis-3-hydroxymethyl cyclobutyl carbamic acid tert-butyl ester as a substitute to give the title compound (16 mg).
1H NMR(400MHz,CDCl 3)8.60(d,J=5.2Hz,1H),8.23(s,1H),7.95(s,1H),7.84(s,1H),7.01(s,1H),6.66(d,J=5.2Hz,1H),6.49(s,1H),4.30-4.44(m,3H),3.41(s,3H),3.05-3.12(m,1H),2.86-2.94(m,1H),2.43-2.60(m,4H),2.25-2.34(m,4H),1.90-1.98(m,2H),1.73-1.76(m,2H),1.47-1.62(m,4H).
Example 40: 3-methyl-6- ((7-methylquinazolin-6-yl) amino) -1- (tetrahydro-2H-pyran-4-yl) -1, 3-dihydro-2H-imidazo [4,5-c ] pyridin-2-one
7-Methylquinazolin-6-amine (16 mg), 6-chloro-3-methyl-1- (tetrahydro-2H-pyran-4-yl) -1, 3-dihydro-2H-imidazo [4,5-c ] pyridin-2-one (27 mg), ruPhos Pd G (9 mg) and cesium carbonate (65 mg) are dissolved in dioxane (10 mL) under nitrogen, heated to 100deg.C and stirred for 5 hours. The reaction solution was cooled to room temperature, filtered, and the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developer: dichloromethane: methanol=20:1 (V: V)) to give the objective compound (10 mg).
1H NMR(400MHz,CDCl 3)9.29(s,1H),9.01(s,1H),8.78(s,1H),8.16(s,1H),8.05(s,1H),7.80(s,1H),7.30(s,1H),4.38-4.47(m,1H),4.00(dd,J=11.2Hz,4.8Hz,2H),3.48(t,J=12.0Hz,2H),3.33(s,3H),2.56(s,3H),2.24-2.35(m,2H),1.66-1.70(m,2H).
Example 41: (1 1R,1 4R,7S)-2 3,4 7, 7-trimethyl-2 2,2 3 -dihydro-2 1 H-5-oxa-3, 8-diaza-4 (6, 4) -quinolin-2 (1, 6) -imidazo [4,5-c ] pyridin-1 (1, 4) -cyclohexanecyclooctan-2 2 -one
The procedure of example 31 was followed using tert-butylBOC- (S) -2-amino-1-propanol as a substitute to give the title compound (19 mg).
1H NMR(400MHz,CDCl 3)8.60(d,J=5.2Hz,1H),7.99-8.03(m,2H),7.90(s,1H),7.50(s,1H),7.12(s,1H),6.77(d,J=5.6Hz,1H),4.48-4.56(m,1H),4.29(dd,J=8.8Hz,1.6Hz,1H),3.97(t,J=9.6Hz,1H),3.52-3.61(m,1H),3.45(s,3H),3.29-3.35(m,1H),2.56(s,3H),2.22-2.36(m,3H),1.90-1.98(m,1H),1.79-1.87(m,1H),1.56-1.72(m,3H),1.18(d,J=6.4Hz,3H).
Example 42: (1 1s,1 4s)-2 3,4 7 -dimethyl-2 2,2 3 -dihydro-2 1 H-5-oxa-3, 9-diaza-4 (6, 4) -quinolin-2 (1, 6) -imidazo [4,5-c ] pyridin-1 (1, 4) -cyclohexylcyclononan-2 2 -one
The procedure of example 31 was followed using tert-butyl (3-hydroxypropyl) carbamate as a substitute to give the title compound (19 mg).
1H NMR(400MHz,CDCl 3)8.63(d,J=5.2Hz,1H),8.14(s,1H),7.95(s,1H),7.84(s,1H),7.52(s,1H),6.72(d,J=5.6Hz,1H),4.52-4.54(m,2H),4.34-4.42(m,1H),3.43(s,3H),2.83-2.90(m,3H),2.53(s,3H),2.22-2.34(m,2H),2.12-2.19(m,2H),1.93-2.01(m,2H),1.61-1.78(m,4H).
Example 43: 3-methyl-6- ((7-methylquinoxalin-6-yl) amino) -1-morpholino-1, 3-dihydro-2H-imidazo [4,5-c ] pyridin-2-one
7-Methylquinoxalin-6-amine (16 mg), intermediate 9: 6-chloro-3-methyl-1-morpholin-1, 3-dihydro-2H-imidazo [4,5-c ] pyridin-2-one (27 mg), ruPhos Pd G (9 mg) and cesium carbonate (65 mg) are dissolved in dioxane (10 mL), heated to 100deg.C and stirred for 5 hours. The reaction solution was cooled to room temperature, filtered, and the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developer: dichloromethane: methanol=20:1 (V: V)) to give the objective compound (29 mg).
1H NMR(400MHz,CDCl 3)8.70(d,J=2.0Hz,1H),8.63(d,J=2.0Hz,1H),8.29(s,1H),7.91(s,1H),7.90(s,1H),6.98(s,1H),6.51(s,1H),3.58-4.14(m,6H),3.41(s,3H),2.77-3.08(m,2H),2.55(s,3H).
Example 44: 3-methyl-6- ((6-methylbenzo [ c ] [1,2,5] thiadiazol-5-yl) amino) -1- (tetrahydro-2H-pyran-4-yl) -1, 3-dihydro-2H-imidazo [4,5-c ] pyridin-2-one
6-Toluo [ c ] [1,2,5] thiadiazol-5-amine (17 mg), 6-chloro-3-methyl-1- (tetrahydro-2H-pyran-4-yl) -1, 3-dihydro-2H-imidazo [4,5-c ] pyridin-2-one (27 mg), ruPhos Pd G (9 mg) and cesium carbonate (65 mg) were dissolved in dioxane (10 mL) under nitrogen, heated to 100℃and stirred for 5 hours. The reaction solution was cooled to room temperature, filtered, and the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developer: dichloromethane: methanol=20:1 (V: V)) to give the objective compound (19 mg).
1H NMR(400MHz,CDCl 3)8.23(s,1H),7.96(s,1H),7.80(s,1H),6.91(s,1H),6.36(s,1H),4.50-4.59(m,1H),4.12(dd,J=12.0Hz,4.4Hz,2H),3.54(td,J=12.0Hz,2.0Hz,2H),3.45(s,3H),2.53(s,3H),2.35-2.46(m,2H),1.77-1.81(m,2H).
Example 45: 3-methyl-6- ((6-methylbenzo [ d ] [1,3] dioxin-5-yl) amino) -1- (tetrahydro-2H-pyran-4-yl) -1, 3-dihydro-2H-imidazo [4,5-c ] pyridin-2-one
6-Methylbenzo [ d ] [1,3] dioxin-5-amine (15 mg), 6-chloro-3-methyl-1- (tetrahydro-2H-pyran-4-yl) -1, 3-dihydro-2H-imidazo [4,5-c ] pyridin-2-one (27 mg), ruPhos Pd G (9 mg) and cesium carbonate (65 mg) were dissolved in dioxane (10 mL) under nitrogen, and heated to 100℃and stirred for 5 hours. The reaction solution was cooled to room temperature, filtered, and the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developer: dichloromethane: methanol=20:1 (V: V)) to give the objective compound (14 mg).
1H NMR(400MHz,CDCl 3)7.75(s,1H),6.87(s,1H),6.74(s,1H),6.24(s,1H),6.06(s,1H),5.96(s,2H),4.38-4.46(m,1H),4.08(dd,J=11.6Hz,4.4Hz,2H),3.50(t,J=11.6Hz,2H),3.38(s,3H),2.23-2.34(m,2H),2.17(s,3H),1.68-1.72(m,2H).
Example 46: 3-methyl-6- ((7-methylquinoxalin-6-yl) amino) -1- (4-oxocyclohexyl) -1, 3-dihydro-2H-imidazo [4,5-c ] pyridin-2-one
7-Methylquinoxalin-6-amine (16 mg), intermediate 7: 6-chloro-3-methyl-1- (4-oxocyclohexyl) -1, 3-dihydro-2H-imidazo [4,5-c ] pyridin-2-one (28 mg), ruPhos Pd G (9 mg) and cesium carbonate (65 mg) are dissolved in dioxane (10 mL), heated to 100deg.C and stirred for 5 hours. The reaction solution was cooled to room temperature, filtered, and the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developer: dichloromethane: methanol=20:1 (V: V)) to give the objective compound (20 mg).
1H NMR(400MHz,CDCl 3)8.69(d,J=2.0Hz,1H),8.64(d,J=2.0Hz,1H),8.25(s,1H),7.96(s,1H),7.91(s,1H),6.86(s,1H),6.47(s,1H),4.67-4.76(m,1H),3.46(s,3H),2.52-2.65(m,9H),2.17-2.24(m,2H).
Example 47: 3-methyl-6- ((6-methylbenzo [ c ] [1,2,5] thiadiazol-5-yl) amino) -1- (4-oxocyclohexyl) -1, 3-dihydro-2H-imidazo [4,5-c ] pyridin-2-one
6-Toluo [ c ] [1,2,5] thiadiazol-5-amine (16 mg), intermediate 7: 6-chloro-3-methyl-1- (4-oxocyclohexyl) -1, 3-dihydro-2H-imidazo [4,5-c ] pyridin-2-one (28 mg), ruPhos Pd G (9 mg) and cesium carbonate (65 mg) are dissolved in dioxane (10 mL), heated to 100deg.C and stirred for 5 hours. The reaction solution was cooled to room temperature, filtered, and the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developer: dichloromethane: methanol=20:1 (V: V)) to give the objective compound (20 mg).
1H NMR(400MHz,CDCl 3)8.18(s,1H),7.97(s,1H),7.80(s,1H),6.82(s,1H),6.39(s,1H),4.68-4.77(m,1H),3.46(s,3H),2.54-2.64(m,6H),2.52(s,3H),2.17-2.23(m,2H).
Example 48: (1 1R,1 4r,7 1S,7 3s)-2 3,4 7 -dimethyl-2 2,2 3 -dihydro-2 1 H-5-oxa-3, 8-diaza-4 (6, 4) -quinolin-2 (1, 6) -imidazo [4,5-c ] pyridin-1 (1, 4) -cyclohexanone-7 (1, 3) -cyclobutanecyclooctane-2 2 -one
The synthesis procedure of example 31 was followed using cis-3-hydroxymethyl cyclobutyl carbamic acid tert-butyl ester as a substitute to give the title compound (26 mg).
1H NMR(400MHz,CDCl 3)8.65(d,J=5.6Hz,1H),8.23(s,1H),7.94(s,1H),7.80(s,1H),6.92(s,1H),6.57(d,J=5.2Hz,1H),6.30(s,1H),4.34-4.46(m,3H),3.46-3.56(m,1H),3.39(s,3H),2.32-2.57(m,5H),1.94-2.17(m,6H),1.73-1.79(m,4H),1.20-1.30(m,2H).
Example 49: (1 1S,1 4s,7 1S,7 3s)-4 7 -methyl-2 7 - (methyl-d 3)-2 8,2 9 -dihydro-2 7 H-5-oxa-3, 8-diaza-4 (6, 4) -quinolin-2 (9, 2) -purin-1 (1, 4) -cyclohexane-7 (1, 3) -cyclobutyl-octan-2 8 -one
The synthesis procedure of example 1 was followed using intermediate 10 as a substitute to give the title compound (21 mg).
1H NMR(400MHz,CDCl 3)8.60(d,J=5.2Hz,1H),8.42(s,1H),7.90(s,1H),7.89(s,1H),6.68(d,J=5.2Hz,1H),6.58(s,1H),4.37(s,2H),4.17-4.26(m,1H),3.00-3.08(m,1H),2.84-2.88(m,1H),2.36-2.57(m,8H),1.91-1.99(m,2H),1.73-1.80(m,2H),1.48-1.68(m,5H).
Biological Activity assay
DNA-PK compound biological activity test method
1. In vitro enzymatic Activity assay of Compounds on DNA-PK
The compounds of this patent were used to determine the IC50 value for inhibition of enzymatic activity of DNA-PK using time resolved fluorescence resonance energy transfer (TR-FRET). Compounds were diluted 10-fold in a gradient from 1mM using 100% DMSO (7 total concentrations), and 2. Mu.L of each concentration was added to 48. Mu.L of reaction buffer (50mM HEPES pH7.5, 10mM MgCl2,1mM DTT,0.01%Brij-35 and 1mM EGTA pH 8.0) and diluted and mixed. 2.5. Mu.L of the diluted compound was added to 384 well plates (OptiPlate-384, purchased from Perkinelmer), followed by 5. Mu.L of DNA-PK (Full-length, final concentration 5 nM), centrifuged and mixed, and 2.5. Mu.L of ATP (final concentration 2. Mu.M) and Fluorescein-p53 Substrate mixed solution (final concentration 2. Mu.M, purchased from Thermo) were added to initiate a reaction, and the total reaction volume was 10. Mu.L. 384 well plates were placed in an incubator at 23℃for 2 hours and 10. Mu.L was then addedTb-anti-phospho-p 53[ pSer15] anti-body (final concentration 2nM, purchased from Thermo) and EDTA cocktail (final concentration 10 mM) were used to terminate the reaction. After a further 1 hour incubation in an incubator, the fluorescence values (340 nm excitation, detection of light emitted at 520nm and 490nm, the ratio of which is the signal of the activity of the enzyme) were read on an Envision (purchased from PerkinElmer). The enzymatic activity signal of DNA-PK was measured at 7 concentrations for each compound, and the IC50 value of the compound was calculated using GRAPHPAD PRISM software. The enzymatic activities of some examples are shown in Table 1.
TABLE 1 part of the example enzymatic Activity data
Numbering of compounds DNA-PK Enzymatic Activity IC 50(nM)
Example 1 0.242
Example 18 0.117
Example 19 0.115
Example 22 0.166
Example 24 0.121
2. Determination of proliferation Activity of Compounds in MDA-MB-468 cells
The invention relates to a detection method for inhibiting cell proliferation, which is established under the combined condition of a DNA-PK inhibitor established in human breast cancer cells MDA-MB-468 and a chemotherapeutic drug Doxorubicin. The specific method comprises the following steps: human breast cancer cells MDA-MB-468 cells were cultured using RPMI-1640 medium (purchased at Biological Industries, BI), with 10% fetal bovine serum (FBS, purchased at Hyclone) and 1% penicillin/streptomycin diabody (P/S, purchased at Life Techonology) at 37℃under 5% CO 2. The day before compound detection, MDA-MB-468 cells were plated in 96-well plates (# 3917, purchased from Corning) at a concentration of 1000 cells/190. Mu.L/well. After 24 hours, doxorubicin was added to a final concentration of 10nM (final DMSO concentration of 0.1%), the test compound was subjected to 3-fold gradient dilutions (total 10 concentrations) with 100% DMSO starting at 10mM, then 2 μl of each concentration was added to 48 μl of RPMI-1640 medium for dilution, and 5 μl of each of the diluted test compounds at different concentrations was added to the plated cell suspension. After co-incubation of the compounds with cells in the Cell incubator for 120h (5 days), 25. Mu.L of Cell-Titer Glo (G7570, purchased from Promega) reagent was added after the media was drained and incubated for another 5-10 minutes. Fluorescence values were then read on CLARIO starPlus (available from BMG) microplate reader and data were used to calculate IC50 values for inhibition of cell proliferation by the compound using GRAPHPAD PRISM software.
TABLE 2 Activity of some of the example compounds IC 50 (nM) in MDA-MB-468 cells
3. Pharmacokinetic data for the compounds:
Male SD rats were derived from Peking Veitz laboratory animal technologies Inc., rats were grouped into groups of 3, each group being orally perfused with a suspension of the test sample (5 mg/kg, 0.5%HPMC,0.1%Tween 80 in H 2 O suspension). Animals were fasted overnight prior to the experiment, with a time of fasting ranging from 10 hours prior to dosing to 4 hours post dosing. Blood was collected at 0.25, 0.5, 1, 2, 4, 6, 8, and 24 hours after dosing, respectively. After isoflurane anesthesia using a small animal anesthesia machine, 0.3mL of whole blood was collected through the fundus venous plexus, placed in a heparin anticoagulation tube, the sample was centrifuged at 4000rpm for 5min at 4 ℃, and the plasma was transferred to a centrifuge tube and stored at-80 ℃ until analysis. Samples from plasma were extracted using protein precipitation and the extracts were analyzed by LC/MS.
TABLE 3 pharmacokinetic data for partial Compounds
4. In vivo pharmacodynamics of compound on human non-small cell lung cancer NCI-H1703 cell subcutaneous xenograft tumor BALB/c nude mouse model
Experimental animals: female BALB/c-nude mice, 6-8 weeks old, weighing 18-22 g.
The experimental method comprises the following steps:
1) Cell culture
Human non-small cell lung cancer NCI-H1703 cells were cultured in vitro, 10% fetal bovine serum, 100U/mL penicillin and 100. Mu.g/mL streptomycin were added to RPM11640 medium, and incubated in 5% CO2 incubator at 37 ℃. Passaging was performed twice a week with conventional digestion treatments with pancreatin-EDTA. When the saturation of the cells is 80% -90% and the number reaches the requirement, the cells are collected, counted and inoculated.
2) Tumor inoculation
0.1ML (5X 106) NCI-H1703 cells (30% matrigel added) were inoculated subcutaneously on the left and right back of each mouse and the group administration was started when the average tumor volume reached about 150mm 3.
3) Preparation of the test substance:
examples 1 and 3 were each formulated as a 3mg/mL suspension with 0.5% HPMC+0.1% Tween80 as vehicle.
4) Tumor measurement and experimental index
Tumor diameters were measured twice weekly with vernier calipers. The calculation formula of the tumor volume is: v=0.5a×b 2, a and b represent the long and short diameters of the tumor, respectively. The tumor-inhibiting effect of the compound was evaluated by TGI (%) or relative tumor proliferation rate T/C (%). Relative tumor proliferation rate T/C (%) =t RTV/C RTV×100%(T RTV: RTV mean of treatment group; c RTV: RTV mean of control). The Relative Tumor Volume (RTV) was calculated from the results of the tumor measurements, given by the formula rtv=vt/Vo, where V 0 is the tumor volume measured on the group administration (i.e. day 0), vt is the tumor volume measured on one measurement, and T RTV and C RTV take the same day data.
TGI (%) reflects the tumor growth inhibition rate. TGI (%) = [1- (average tumor volume at the end of the administration of a certain treatment group-average tumor volume at the start of the administration of the treatment group)/(average tumor volume at the end of the treatment of the solvent control group-average tumor volume at the start of the treatment of the solvent control group) ]x100%.
5) Statistical analysis
Statistical analysis was performed using SPSS software based on RTV data at the end of the test. The treatment group showed the best treatment effect at day 18 after dosing at the end of the trial, so the statistical analysis was performed to evaluate the inter-group differences based on this data. The comparison between the two groups was analyzed with T-test. p <0.05 was considered a significant difference.
6) Experimental results and conclusions
Example 1 the results of the experiment at a dose of 30mg/kg (twice a day administration) are shown in Table 4. The results of the experiment of example 31 at a dose of 30mg/kg (twice daily administration) are shown in Table 5.
TABLE 4 in vivo pharmacodynamic data for example 1
TABLE 5 in vivo pharmacodynamic data for example 31
Note that:
a. Mean ± SEM, n=5;
b. tumor growth inhibition was calculated from T/C and TGI (%) = [1- (T 18-T 0)/(V 18-V 0) ]x100);
c.p values were obtained by analysis of tumor volume relative values (RTV) using t-test;
Conclusion: in the experiment, compared with a control group, the compound in the embodiment of the invention has obvious tumor inhibiting effect, and tumor-bearing mice show good tolerance to the compound.
Industrial applicability
The invention provides a DNA-PK selective inhibitor, and preparation and application thereof. The invention also provides a series of compounds represented by the general formula (I), pharmaceutically acceptable salts, solvates, polymorphs, or isomers thereof, pharmaceutical compositions containing these compounds, and methods of treating diseases using such compounds. The DNA-PK selective inhibitor provided by the invention has high activity, strong drug resistance and small clinical side effect, can effectively enhance the sensitivity of radiotherapy and chemotherapy in tumor treatment, and has good economic value and application prospect.

Claims (12)

  1. A compound of formula (II) or a pharmaceutically acceptable salt, solvate, polymorph or isomer thereof,
    Wherein,
    Ring A is a 6-10 membered aryl or a 5-12 membered heteroaryl,
    Ring B is a 3-12 membered carbocycle or a 4-12 membered heterocycle, C and S on ring B may optionally be oxidized,
    Z is-N (R) -, O or S,
    Y is N or CR 20, and the total number of the catalyst is N,
    R 20 is H, halogen, or C 1-6 alkyl,
    X 2 is CR 2 or N,
    X 1 is CRR 4, O, S, or NR 6,
    R 1 is H, C 1-6 alkyl, C 3-8 cycloalkyl, or 3-8 membered heterocycloalkyl,
    R 7 and R 8 are each independently selected from halogen, CN, -OH, -NH 2、C 1-6 alkyl, 3-8 membered cycloalkyl, 3-8 membered heterocycloalkyl, -O-C 1-6 alkyl and-NR-C 1-6 alkyl,
    M and n are each independently 0, 1, 2, or 3,
    R 3 is R 5 or-X 3-R 5,
    R 4 is R 6 or-X 3-R 6,
    X 3 is each independently-O-, -S-, or-NR-,
    R 5 and R 6 are each independently selected from H, halogen, -CN, -OH, -NH 2、C 1-6 alkyl, 3-8 membered cycloalkyl, 3-8 membered heterocycloalkyl, 5-12 membered heteroaryl, -O-C 1-6 alkyl, or-NR-C 1-6 alkyl, which alkyl, cycloalkyl, heterocycloalkyl, or heteroaryl may be optionally substituted with halogen, -CN, -OH, -NH 2、C 1-6 alkyl, 3-8 membered cycloalkyl, 3-8 membered heterocycloalkyl, 5-12 membered heteroaryl, -O-C 1-6 alkyl, or-NR-C 1-6 alkyl, or
    R 5 and R 6 are joined to form- (CH 2) p-X-(CH 2) q -, wherein X is a bond, -CR=CR-, -CO-CR=CR-, -C≡C-, -CO-C≡C-, -O-, -S (O) 2-、-S(O) 2 NR- -N (R) -, -CO-, -C (O) NR-, -C (O) O-, 6-10 membered arylene, 5-12 membered heteroarylene, 3-12 membered carbocycle, or 3-12 membered heterocycle, and- (CH 2 in CH 2) p-X-(CH 2) q -, and the arylene, heteroarylene, carbocycle, and heterocycle may optionally be substituted with halogen, -CN, -OH, -NH 2、-S(O)R、-S(O) 2R、-S(O) 2NR-C 1-6 alkyl,C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, 3-12 membered cycloalkyl, 3-12 membered heterocycloalkyl, 6-10 membered aryl, 5-12 membered heteroaryl, -O-C 1-6 alkyl, or-NR-C 1-6 alkyl substitution,
    P and q are each independently 0, 1,2,3, or 4, and p+q is 1,2,3,4, 5, or 6,
    R 2 is selected from the group consisting of H, halogen, CH 2F、CHF 2、CF 3、-OH、-NH 2、CN、C 1-6 alkyl, -O-C 1-6 alkyl, - (CH 2) 1-6-CN、-(CH 2) 1-6-O-C 1-6 alkyl, - (CH 2) 1-3 -OH and-NR-C 1-6 alkyl,
    R is each independently H, C 1-6 alkyl, 3-8 membered cycloalkyl, or 3-8 membered heterocycloalkyl, which alkyl, cycloalkyl, and heterocycloalkyl may be optionally substituted with halogen, -CN, -OH, -NH 2、-O-C 1-6 alkyl, or-NH-C 1-6 alkyl.
  2. The compound of claim 1, or a pharmaceutically acceptable salt, solvate, polymorph or isomer thereof, wherein ring B is a 3-12 membered carbocyclic ring or a 4-12 membered heterocyclic ring, and S on ring B is optionally oxidized.
  3. The compound according to claim 1, or a pharmaceutically acceptable salt, solvate, polymorph or isomer thereof, having a structure represented by formula (I)
    Wherein,
    Ring A is a 6-10 membered aryl or a 5-12 membered heteroaryl,
    Ring B is a 3-12 membered carbocycle or a 4-12 membered heterocycle, S on ring B may optionally be oxidized,
    Z is-N (R) -, O or S,
    X 2 is CR 2 or N,
    X 1 is CRR 4, O, S, or NR 6,
    R 1 is H, C 1-6 alkyl, C 3-8 cycloalkyl, or 3-8 membered heterocycloalkyl,
    R 7 and R 8 are each independently selected from halogen, CN, -OH, -NH 2、C 1-6 alkyl, 3-8 membered cycloalkyl, 3-8 membered heterocycloalkyl, -O-C 1-6 alkyl and-NR-C 1-6 alkyl,
    M and n are each independently 0, 1, 2, or 3,
    R 3 is R 5 or-X 3-R 5,
    R 4 is R 6 or-X 3-R 6,
    X 3 is each independently-O-, -S-, or-NR-,
    R 5 and R 6 are each independently selected from H, halogen, -CN, -OH, -NH 2、C 1-6 alkyl, 3-8 membered cycloalkyl, 3-8 membered heterocycloalkyl, -O-C 1-6 alkyl, or-NR-C 1-6 alkyl, or
    R 5 and R 6 are joined to form- (CH 2) p-X-(CH 2) q -, wherein X is a bond, -CR=CR-, -CO-CR=CR-, -C≡C-, -CO-C≡C-, -O-, -S (O) 2-、-S(O) 2 NR- -N (R) -, -CO-, -C (O) NR-, -C (O) O-, 6-10 membered arylene, 5-12 membered heteroarylene, 3-12 membered carbocycle, or 3-12 membered heterocycle, and- (CH 2 in CH 2) p-X-(CH 2) q -, and the arylene, heteroarylene, carbocycle, and heterocycle may optionally be substituted with halogen, -CN, -OH, -NH 2、-S(O)R、-S(O) 2R、 -S(O) 2NR-C 1-6 alkyl,C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, 3-12 membered cycloalkyl, 3-12 membered heterocycloalkyl, 6-10 membered aryl, 5-12 membered heteroaryl, -O-C 1-6 alkyl, or-NR-C 1-6 alkyl substitution,
    P and q are each independently 0, 1,2,3, or 4, and p+q is 1,2,3,4, 5, or 6,
    R 2 is selected from the group consisting of H, halogen, CH 2F、CHF 2、CF 3、-OH、-NH 2、CN、C 1-6 alkyl, -O-C 1-6 alkyl, - (CH 2) 1-6-CN、-(CH 2) 1-6-O-C 1-6 alkyl, - (CH 2) 1-3 -OH and-NR-C 1-6 alkyl,
    R is each independently H, C 1-6 alkyl, 3-8 membered cycloalkyl, or 3-8 membered heterocycloalkyl, which alkyl, cycloalkyl, and heterocycloalkyl may be optionally substituted with halogen, -CN, -OH, -NH 2、-O-C 1-6 alkyl, or-NH-C 1-6 alkyl.
  4. The compound of claim 1, or a pharmaceutically acceptable salt, solvate, polymorph or isomer thereof, wherein R 5 and R 6 are taken together to form- (CH 2) p-X-(CH 2) q -, wherein X is a bond, -O-, -S-, -N (R) -, -CO-, -C (O) NR-, -C (O) O-, 6-10 membered arylene, 5-12 membered heteroarylene, 3-12 membered carbocyclic ring, or 3-12 membered heterocyclic ring, and- (CH 2 in CH 2) p-X-(CH 2) q -, and the arylene, heteroarylene, carbocyclic ring, and heterocyclic ring are optionally substituted with halogen, -CN, -OH, -NH 2、C 1-6 alkyl, 3-12 membered cycloalkyl, 3-12 membered heterocycloalkyl, 6-10 membered aryl, 5-12 membered heteroaryl, -O-C 1-6 alkyl, or-NR-C 1-6 alkyl, p, q, R being as defined in claim 1.
  5. The compound of claim 1, or a pharmaceutically acceptable salt, solvate, polymorph or isomer thereof, wherein R 5 and R 6 are taken together to form- (CH 2) p-X-(CH 2) q -, wherein X is a bond, -O-, -S-, -N (R) -, -CO-, -C (O) NR-, -C (O) O-, 6-10 membered arylene, 5-12 membered heteroarylene, 3-12 membered carbocycle, or 3-12 membered heterocycle, and CH 2 in- (CH 2) p-X-(CH 2) q -, and the arylene, heteroarylene, carbocycle, and heterocycle are optionally substituted with halogen, -CN, -OH, -NH 2, or C 1-6 alkyl, p, q, R being as defined in claim 1.
  6. The compound of claim 1, or a pharmaceutically acceptable salt, solvate, polymorph or isomer thereof, wherein R 2 is selected from H, halogen, CH 2F、CHF 2、CF 3、-OH、-NH 2, CN, and C 1-6 alkyl.
  7. The compound of claim 1, or a pharmaceutically acceptable salt, solvate, polymorph or isomer thereof, wherein R 1 is C 1-6 alkyl.
  8. The following compounds:
    or a pharmaceutically acceptable salt, solvate, polymorph or isomer thereof.
  9. A pharmaceutical composition comprising a compound according to any one of claims 1-8, or a pharmaceutically acceptable salt, solvate, polymorph, or isomer thereof.
  10. Use of a compound according to any one of claims 1-8, or a pharmaceutically acceptable salt, solvate, polymorph, or isomer thereof, or a pharmaceutical composition according to claim 9, in the manufacture of a medicament for treatment of a DNA-PK related disease.
  11. The use according to claim 10, wherein the DNA-PK related disease is cancer.
  12. The use of claim 10, wherein the DNA-PK related disease is colorectal cancer, glioblastoma, gastric cancer, ovarian cancer, diffuse large B-cell lymphoma, chronic lymphocytic leukemia, acute myelogenous leukemia, head and neck squamous cell carcinoma, breast cancer, prostate cancer, bladder cancer, hepatocellular carcinoma, small-cell lung cancer, or non-small-cell lung cancer.
CN202280060488.XA 2021-09-07 2022-09-07 DNA-PK selective inhibitor and preparation method and application thereof Pending CN117940437A (en)

Applications Claiming Priority (9)

Application Number Priority Date Filing Date Title
CN202111046420X 2021-09-07
CN202111046420 2021-09-07
CN2021112679668 2021-10-27
CN202111267966 2021-10-27
CN202210029171 2022-01-11
CN2022100291711 2022-01-11
CN2022103157343 2022-03-25
CN202210315734 2022-03-25
PCT/CN2022/117405 WO2023036156A1 (en) 2021-09-07 2022-09-07 Dna-pk selective inhibitor, and preparation method therefor and use thereof

Publications (1)

Publication Number Publication Date
CN117940437A true CN117940437A (en) 2024-04-26

Family

ID=85506080

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202280060488.XA Pending CN117940437A (en) 2021-09-07 2022-09-07 DNA-PK selective inhibitor and preparation method and application thereof

Country Status (2)

Country Link
CN (1) CN117940437A (en)
WO (1) WO2023036156A1 (en)

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI820146B (en) * 2018-06-15 2023-11-01 瑞典商阿斯特捷利康公司 Purinone compounds and their use in treating cancer
WO2021197159A1 (en) * 2020-03-31 2021-10-07 南京明德新药研发有限公司 Macrocyclic compounds that serve as dna-pk inhibitors
CN114605414A (en) * 2020-12-08 2022-06-10 首药控股(北京)股份有限公司 DNA-PK selective inhibitor and preparation method and application thereof
CN114573584A (en) * 2020-11-18 2022-06-03 首药控股(北京)股份有限公司 DNA-PK selective inhibitor and preparation method and application thereof
CN113956272A (en) * 2020-07-20 2022-01-21 首药控股(北京)有限公司 DNA-PK selective inhibitor and preparation method and application thereof
CN114195805A (en) * 2020-09-18 2022-03-18 首药控股(北京)股份有限公司 DNA-PK selective inhibitor and preparation method and application thereof
CN114907384A (en) * 2021-02-09 2022-08-16 首药控股(北京)股份有限公司 DNA-PK selective inhibitor and preparation method and application thereof
WO2022017368A1 (en) * 2020-07-20 2022-01-27 首药控股(北京)股份有限公司 Dna-pk selective inhibitor, preparation method therefor and use thereof
WO2022177302A1 (en) * 2021-02-17 2022-08-25 보령제약 주식회사 Pyrimidine-fused ring compound with dna-pk inhibition activity and use thereof
WO2022199547A1 (en) * 2021-03-22 2022-09-29 成都赜灵生物医药科技有限公司 7,9-dihydropurine derivative and pharmaceutical purpose thereof

Also Published As

Publication number Publication date
WO2023036156A1 (en) 2023-03-16

Similar Documents

Publication Publication Date Title
ES2619125T3 (en) Heterocyclic compounds fused as protein kinase inhibitors
WO2022061251A1 (en) Compounds and methods for kras modulation and indications therefor
CN114394966A (en) Pyridopyrimidinone CDK2/4/6 inhibitors
JP2018507214A (en) Bicyclic heterocycles as FGFR4 inhibitors
CA2894126A1 (en) Prmt5 inhibitors and uses thereof
TW202334137A (en) Pi3k inhibitors and methods of treating cancer
JP6888006B2 (en) Pyrazolo, pyrazolo, imidazole-pyrimidine and pyridine compounds that inhibit MNK1 and MNK2
KR20220130168A (en) Pyrimidine-4(3H)-ketone heterocyclic compounds, methods for their preparation, and their use in medicine and pharmacology
TW202102509A (en) Compounds targeting prmt5
KR20220061958A (en) Heterobicyclic amides as inhibitors of CD38
JP7384535B2 (en) Quinazoline compounds and their preparation, use and pharmaceutical compositions
CN116406272A (en) DNA-PK selective inhibitor and preparation method and application thereof
KR20190092549A (en) CDK4 / 6 inhibitor
EP3936509A1 (en) Fgfr4 kinase inhibitor, preparation method therefor and use thereof
CN113956272A (en) DNA-PK selective inhibitor and preparation method and application thereof
JP2017526720A (en) Compounds and compositions as kinase inhibitors
TW202043223A (en) 5-azaindazole derivatives as adenosine receptor antagonists
JP2020525471A (en) Novel Substituted Azaindoline Derivatives As NIK Inhibitors
BR112020010004A2 (en) pyrazolopyridinone compounds
CN114195805A (en) DNA-PK selective inhibitor and preparation method and application thereof
CN117940437A (en) DNA-PK selective inhibitor and preparation method and application thereof
ES2256506T3 (en) TRICYCLE AGONISTS OF THE CRF RECEIVER.
CN114573584A (en) DNA-PK selective inhibitor and preparation method and application thereof
CN114634521A (en) DNA-PK selective inhibitor and preparation method and application thereof
CN114605414A (en) DNA-PK selective inhibitor and preparation method and application thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination