CN113121573A - Purine derivatives and their use in medicine - Google Patents

Purine derivatives and their use in medicine Download PDF

Info

Publication number
CN113121573A
CN113121573A CN202011615924.4A CN202011615924A CN113121573A CN 113121573 A CN113121573 A CN 113121573A CN 202011615924 A CN202011615924 A CN 202011615924A CN 113121573 A CN113121573 A CN 113121573A
Authority
CN
China
Prior art keywords
methyl
chloro
amino
purin
dihydro
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
CN202011615924.4A
Other languages
Chinese (zh)
Inventor
魏用刚
许学珍
楚洪柱
何吕学
雷飞全
何阳
苏桂转
王美微
刘兵
孙毅
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.)
Chengdu Baiyu Pharmaceutical Co Ltd
Original Assignee
Chengdu Baiyu Pharmaceutical 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 Chengdu Baiyu Pharmaceutical Co Ltd filed Critical Chengdu Baiyu Pharmaceutical Co Ltd
Publication of CN113121573A publication Critical patent/CN113121573A/en
Pending legal-status Critical Current

Links

Classifications

    • 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
    • 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
    • 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
    • C07D473/02Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6
    • C07D473/18Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6 one oxygen and one nitrogen atom, e.g. guanine
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • 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

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The application discloses triazole derivatives and application thereof in medicine, and particularly relates to pyrimidine derivatives shown as a general formula (I), or stereoisomers, solvates, prodrugs, deuterons, metabolites, pharmaceutically acceptable salts or co-crystals thereof, a pharmaceutical composition containing the same and application of the compounds or the composition in the field of preparation of DNA-PK inhibitors, wherein definitions of substituents in the general formula (I) are the same as those in the specification

Description

Purine derivatives and their use in medicine
Technical Field
The application relates to purine derivatives shown in a general formula (I), or stereoisomers, solvates, prodrugs, deuterons, metabolites, pharmaceutically acceptable salts or co-crystals thereof, pharmaceutical compositions thereof and application of the purine derivatives in preparation of DNA-PK inhibitors.
Background
DNA-dependent protein kinase (DNA-PK) is a DNA-PK enzyme complex composed of a Ku70/Ku80 heterodimer and a DNA-dependent protein kinase catalytic subunit (DNA-PKcs). The enzyme complex needs to be activated in the presence of DNA to perform the corresponding function (George et al, 2019). As a serine/threonine protein kinase, DNA-PK belongs to PIKK (phosphoribosyl 3-kinase-related kinase) family members, has important functions in the process of repairing intracellular DNA Double Strand Breaks (DSBs) and cell DNA recombination or antibody DNA rearrangement (V (D) J recombination), and also participates in physiological processes such as chromosome modification, transcription regulation, telomere maintenance and the like.
During normal physiological processes, a number of factors may contribute to the development of DSBs in DNA: for example, DSBs often appear as intermediates during somatic DNA recombination, a physiological process important for the development of a functional immune system in all vertebrates; the replication fork in DNA replication encounters damaged bases and may also cause single or double strand breaks; DNA may also produce DSBs (Cannan & Pederson,2016) as a result of attack by Reactive Oxygen Species (ROS) during normal metabolic processes. In addition, there are a number of extrinsic factors that can cause DSBs, such as Ionizing Radiation (IR) and chemotherapeutic agents (e.g., topoisomerase II inhibitors), among others (George et al, 2019). If the DSBs are not repaired or incorrectly repaired, mutations and/or chromosomal aberrations will occur, eventually leading to cell death. To address the hazards posed by DSBs, eukaryotic cells have evolved multiple mechanisms to repair damaged DNA to maintain cell viability and genomic stability. In eukaryotic cells, the most prominent mode of DNA repair is non-homologous end joining (NHEJ). This direct joining of the fragmented DNA does not require the participation of homologous DNA fragments and can occur at any stage of the cell cycle. NHEJ is a dynamic process mediated by DNA-PK requiring multiple proteins to participate together with signaling pathways, the basic process being as follows: (1) the Ku70/Ku80 heterodimer recognizes and binds to the double-stranded DNA break end; (2) recruiting proteins such as DNA-PKcs, XRCC4-DNA ligase IV complex to both sides of the double strand of DNA break; (3) the DNA-PKcs self-phosphorylates and activates the kinase activity of the DNA-PKcs; (4) the DNA-PKcs is used as an adhesive to connect two ends of the broken DNA so as to prevent the degradation of the DNA by exonuclease; (5) processing the DNA to remove non-ligatable ends or other forms of damage at the break; (6) the XRCC4-DNA ligase IV complex repairs DNA ends (in some cases, DNA polymerase may be required to synthesize new ends prior to ligation). When the DNA-PKcs is phosphorylated, the change of protein conformation can be induced, and the activity of various proteins (such as Artemis, Ku70, Ku80 and DNA ligase) in the NHEJ process is regulated, which is important for the DNA repair process. Therefore, phosphorylated DNA-PKcs (pDNA-PKcs) often serve as markers for cellular DSBs.
It has been shown that DNA-PK activity is involved in the development of a variety of tumors: for example, DNA-PKcs in melanoma can promote angiogenesis and tumor metastasis; the expression level of DNA-PKcs in multiple myeloma is obviously up-regulated; the content of Ku protein in radiotherapy-resistant thyroid tumors is significantly increased (Ihara, Ashizawa, Shichijo, & Kudo, 2019). Thus, it is contemplated that a DNA-PK inhibitor may be used in combination with an anti-tumor therapy (e.g., IR, chemotherapeutic agent, etc.) which causes DNA damage to enhance the effect. The use of DNA-PK inhibitors may interfere to some extent with the DNA repair function of normal cells, however, there are many DNA repair pathways in normal cells as complement, while tumor cells are under strong DNA replication stress and lack an effective DNA repair approach. The killing effect of other antitumor drugs on tumor cells can be improved by inhibiting the activity of the tumor cell DNA-PK.
Through many years of research, a plurality of DNA-PK inhibitors have been discovered at present. The earliest compound with DNA-PK kinase inhibitory activity was found to be a fungal metabolite, Wortmannin, IC50(DNA-PK) at about 15nM, which compound plays an important role in both acetylation and phosphorylation of p53 protein (Sarkaria et al, 1998); the quercetin derivative LY294002 was reported later to also have DNA-PK inhibitory activity (Maira, Stauffer, Schnell,&Garcia-Echeverria, 2009); later, new generation DNA-PK inhibitors such as NU7026, NU7441 and the like are developed based on LY294002 structure. Although these compounds have been proved to have good killing effect on tumor cells, they have the problems of high toxicity, poor selectivity and the like, and cannot enter clinical development (Maira et al, 2009). Other DNA-PK inhibitors have also been reported, such as small molecule compounds like OK1035, SU11752, PP121, KU-0060648, but these compoundsThe substance also has the defects of low specificity to DNA-PK (George et al, 2019). Therefore, the development of highly active, highly specific, and low-toxicity DNA-PK inhibitors is still needed to better meet clinical needs.
Disclosure of Invention
One or more embodiments of the present application provide purine derivatives, or stereoisomers, solvates, prodrugs, deuterons, metabolites, pharmaceutically acceptable salts or co-crystals thereof, pharmaceutical compositions thereof and uses thereof in the preparation of DNA-PK inhibitors.
In one or more embodiments of the present application, the compounds have high inhibitory activity and/or high selectivity for DNA-PK, and can be used as chemo-and radiosensitizers to effectively treat cancer, improve the efficacy of the prior art, and reduce toxic side effects.
One or more embodiments of the present invention provide a compound represented by general formula (I), or a stereoisomer, solvate, prodrug, deuterode, metabolite, pharmaceutically acceptable salt, or co-crystal thereof:
Figure BDA0002876667980000021
wherein:
R1selected from H, -OH, cyano, halogen, -NH2、C1-6Alkyl or C1-6Alkoxy radical, said C1-6Alkyl is optionally further substituted with 1-3 substituents selected from D or halogen;
R2selected from H or C1-6An alkyl group;
R3is selected from C1-6Alkyl radical, C3-12Carbocyclyl, C3Heterocyclic group, C4-12Heterocyclyl radical, -C1-6alkylene-C3-12Carbocyclyl, -C1-6alkylene-C3Heterocyclyl or-C1-6alkylene-C4-12Heterocyclic group, said C3Heterocyclyl and C4-12Heterocyclyl contains 1 to 3 heteroatoms selected from N, O or S, said C1-6Alkyl radical, C3-12Carbocyclyl, C3Heterocyclyl and C4-12Heterocyclic group, C1-6Alkylene is optionally further substituted by 1 or more groups selected from-OH, carboxyl, halogen, cyano, ═ O, C1-6Alkyl radical, C1-6Heteroalkyl group, C2-6Alkenyl radical, C2-6Alkynyl, -NRa1Ra2、-C(=O)OC1-6Alkyl, -C (═ O) NRa1Ra2、C3-12Cycloalkyl radical, C3Heterocycloalkyl radical, C4-12Heterocycloalkyl radical, C6-12Aryl or C5-12Heteroaryl, or a substituted heteroaryl; and said C in said substituent1-6Alkyl radical, C1-6Heteroalkyl group, C2-6Alkenyl or C2-6Alkynyl is optionally further substituted by 1 or more groups selected from-OH, carboxy, cyano, halo, -O-Ra1、-NRa1Ra2、C3-12Cycloalkyl radical, C3Heterocycloalkyl radical, C4-12Heterocycloalkyl radical, C6-12Aryl or C5-12Heteroaryl, or a substituted heteroaryl;
with the following conditions: when R is3When cyclohexyl, the cyclohexyl is substituted by at least one cyano group; and R is3Is not tetrahydrofuranyl or oxacyclohexyl; alternatively, the conditions are: r3Is not cyclohexyl, tetrahydrofuranyl or oxacyclohexyl;
Ra1、Ra2each independently selected from H, C1-6Alkyl, -C (═ O) Ra3or-C (═ O) NRa4Ra5Wherein said C1-6The alkyl is optionally further substituted by 1 or more groups selected from OH, halogen, C1-6Alkyl radical, C1-6Alkoxy radical, C6-12Aryl radical, C5-12Heteroaryl group, C3-12Cycloalkyl radical, C3Heterocycloalkyl or C4-12Substituted with a substituent of heterocycloalkyl;
Ra3is selected from C1-6Alkyl radical, C1-6Alkoxy or C6-12An aryl group;
Ra4、Ra5each independently selected from H or C1-6An alkyl group; or Ra4And Ra5And N atom formA 3-to 8-membered heterocyclic ring, said 3-to 8-membered heterocyclic ring comprising 1 to 4 heteroatoms selected from N, O or S;
p is selected from 0, 1,2 or 3;
optionally, formula (I) may be optionally substituted with 1 or more D atoms.
One or more embodiments of the present application provide a compound, or a stereoisomer, solvate, prodrug, deuteride, metabolite, pharmaceutically acceptable salt or co-crystal thereof, wherein the compound is selected from a compound represented by the general formula (II):
Figure BDA0002876667980000031
wherein:
R1selected from halogen or C1-6Alkyl radical, said C1-6Alkyl is optionally further substituted with 1-3 substituents selected from D or halogen;
R3is selected from C1-6Alkyl radical, C3-12Carbocyclyl, C3Heterocyclic group, C4-12Heterocyclyl radical, -C1-6alkylene-C3-12Carbocyclyl, -C1-6alkylene-C3Heterocyclyl or-C1-6alkylene-C4-12Heterocyclic group, said C3Heterocyclyl and C4-12Heterocyclyl contains 1 to 3 heteroatoms selected from N, O or S, said C1-6Alkyl radical, C3-12Carbocyclyl, C3Heterocyclyl and C4-12Heterocyclic group, C1-6Alkylene is optionally further substituted by 1 or more groups selected from-OH, carboxyl, halogen, cyano, ═ O, C1-6Alkyl radical, C1-6Heteroalkyl group, C2-6Alkenyl radical, C2-6Alkynyl, -NRa1Ra2、-C(=O)OC1-6Alkyl, -C (═ O) NRa1Ra2、C3-12Cycloalkyl radical, C3Heterocycloalkyl radical, C4-12Heterocycloalkyl radical, C6-12Aryl or C5-12Heteroaryl, or a substituted heteroaryl; and said C in said substituent1-6Alkyl radical, C1-6Heteroalkyl group, C2-6Alkenyl or C2-6Alkynyl is optionally further substituted by 1 or more groups selected from-OH, carboxy, cyano, halo, -O-Ra1、-NRa1Ra2、C3-12Cycloalkyl radical, C3Heterocycloalkyl radical, C4-12Heterocycloalkyl radical, C6-12Aryl or C5-12Heteroaryl, or a substituted heteroaryl;
with the following conditions: when R is3When cyclohexyl, the cyclohexyl is substituted by at least one cyano group; and R is3Is not tetrahydrofuranyl or oxacyclohexyl; alternatively, the conditions are: r3Is not cyclohexyl, tetrahydrofuranyl or oxacyclohexyl;
Ra1、Ra2each independently selected from H, C1-6Alkyl, -C (═ O) Ra3or-C (═ O) NRa4Ra5Wherein said C1-6The alkyl is optionally further substituted by 1 or more groups selected from OH, halogen, C1-6Alkyl radical, C1-6Alkoxy radical, C6-12Aryl radical, C5-12Heteroaryl group, C3-12Cycloalkyl radical, C3Heterocycloalkyl or C4-12Substituted with a substituent of heterocycloalkyl;
Ra3is selected from C1-6Alkyl radical, C1-6Alkoxy or C6-12An aryl group;
Ra4、Ra5each independently selected from H or C1-6An alkyl group; or Ra4And Ra5And the N atom forms a 3-to 8-membered heterocyclic ring, said 3-to 8-membered heterocyclic ring comprising 1 to 4 heteroatoms selected from N, O or S;
p is selected from 0, 1,2 or 3.
One or more embodiments of the present application provide a compound of (I) or (II), or a stereoisomer, solvate, metabolite, prodrug, deutero-ent, pharmaceutically acceptable salt, or co-crystal thereof, wherein:
R1selected from halogen or C1-4Alkyl radical, said C1-6Alkyl is optionally further substituted with 1-3 substituents selected from D or halogen;
R3is selected from C1-4Alkyl radical, C3-8Carbocyclyl, C4-8Heterocyclyl radical, -C1-2alkylene-C3-8Carbocyclic group or-C1-2alkylene-C4-8Heterocyclic group, said C4-8Heterocyclyl contains 1 to 3 heteroatoms selected from N or O, said C1-4Alkyl radical, C3-8Carbocyclyl and C4-8Heterocyclic group, C1-2Alkylene is optionally further substituted by 1 or more groups selected from-OH, halogen, ═ O, cyano, or C1-4Alkyl is substituted by a substituent; and said C in said substituent1-4Alkyl is optionally further substituted with 1 or more substituents selected from-OH, cyano, or halogen; with the following conditions: when R is3When cyclohexyl, the cyclohexyl is substituted by at least one cyano group; and R is3Is not tetrahydrofuranyl or oxacyclohexyl; alternatively, the conditions are: r3Is not cyclohexyl, tetrahydrofuranyl or oxacyclohexyl;
p is selected from 1.
One or more embodiments of the present application provide a compound, or a stereoisomer, solvate, prodrug, deuteride, metabolite, pharmaceutically acceptable salt or co-crystal thereof, wherein the compound is selected from, but not limited to, the following structures:
Figure BDA0002876667980000041
Figure BDA0002876667980000051
Figure BDA0002876667980000052
or
Figure BDA0002876667980000053
One or more embodiments of the present application provide intermediates for the preparation of the compounds of the present invention, including but not limited to:
Figure BDA0002876667980000054
Figure BDA0002876667980000055
or
Figure BDA0002876667980000056
One or more embodiments of the present application provide a pharmaceutical composition comprising:
(1) the compound of the invention or a stereoisomer, solvate, prodrug, deuteron, metabolite, pharmaceutically acceptable salt or eutectic crystal thereof;
(2) optionally one or more other active ingredients; and
(3) a pharmaceutically acceptable carrier and/or excipient.
One or more embodiments of the present application provide a use of the pharmaceutical composition described above or the above-described compound of the present invention or a stereoisomer, solvate, prodrug, deuteron, metabolite, pharmaceutically acceptable salt or co-crystal thereof in the manufacture of a medicament for the treatment of cancer.
One or more embodiments of the present application provide a use of the pharmaceutical composition described above or the above-described compound of the present invention or a stereoisomer, solvate, prodrug, deuteron, metabolite, pharmaceutically acceptable salt or co-crystal thereof in the preparation of a DNA-PK inhibitor.
One or more embodiments of the present application provide a compound of the present application for use as a medicament.
One or more embodiments of the present application provide compounds of the present application for use as DNA-PK inhibitors.
One or more embodiments of the present application provide a compound of the present application for use in a method of treating, preventing, or inhibiting cancer.
One or more embodiments of the present application provide a compound of the present application for use in a method of inhibiting DNA-PK.
One or more embodiments of the present application provide methods of treating, preventing, or inhibiting cancer, comprising administering to a subject in need thereof a compound of the present application.
One or more embodiments of the present application provide a method of inhibiting DNA-PK comprising administering to a subject in need thereof a compound of the present application.
Unless stated to the contrary, the terms used in the specification and claims have the following meanings.
Where carbon, hydrogen, oxygen, sulfur, nitrogen or F, Cl, Br, I are involved in the radicals and compounds of the invention, including their isotopes, and where carbon, hydrogen, oxygen, sulfur or nitrogen are involved in the radicals and compounds of the invention, optionally further substituted with one or more of their corresponding isotopes, where isotopes of carbon include12C、13C and14c, isotopes of hydrogen including protium (H), deuterium (D, also called deuterium), tritium (T, also called deuterium), isotopes of oxygen including16O、17O and18isotopes of O, sulfur including32S、33S、34S and36isotopes of S, nitrogen include14N and15isotopes of N, F include17F and19isotopes of F, chlorine including35Cl and37cl, isotopes of bromine including79Br and81Br。
"alkyl" refers to a straight or branched chain saturated aliphatic hydrocarbon group of 1 to 20 carbon atoms, preferably an alkyl group of 1 to 8 (e.g., 1,2,3,4, 5, 6, 7, 8) carbon atoms, more preferably an alkyl group of 1 to 6 carbon atoms, and further preferably an alkyl group of 1 to 4 carbon atoms. Non-limiting examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, neo-butyl, tert-butyl, n-pentyl, isopentyl, neopentyl, n-hexyl, and various branched isomers thereof; when the alkyl group is substituted, it may be optionally further substituted with 1 or more substituents.
"alkoxy" refers to a group formed by replacement of at least 1 carbon atom in an alkyl group with an oxygen atom. Non-limiting examples include methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, sec-butoxy, tert-butoxy, n-pentoxy, n-hexoxy, cyclopropoxy, and cyclobutoxy. The alkyl group is defined as the "alkyl" group as defined above.
"alkenyl" means a straight or branched chain unsaturated aliphatic hydrocarbon group containing 1 to 10 (e.g., 1,2,3,4, 5, 6, 7,8, 9, 10) carbon-carbon double bonds, consisting of 2 to 20 carbon atoms, preferably 2 to 12 (e.g., 2,3,4, 5, 6, 7,8, 9, 10, 11, 12) carbon atoms, more preferably 2 to 8 carbon atoms, and still more preferably 2 to 6 carbon atoms. Non-limiting examples include vinyl, propen-2-yl, buten-2-yl, penten-4-yl, hexen-2-yl, hexen-3-yl, hepten-2-yl, hepten-3-yl, hepten-4-yl, octen-3-yl, nonen-3-yl, decen-4-yl and undecen-3-yl. The alkenyl group may be optionally further substituted with 1 or more substituents.
"alkynyl" means a straight or branched chain unsaturated aliphatic hydrocarbon group containing from 1 to 10 (e.g., 1,2,3,4, 5, 6, 7,8, 9, or 10) carbon-carbon triple bonds, consisting of from 2 to 20 carbon atoms, preferably an alkynyl group of from 2 to 12 (e.g., 2,3,4, 5, 6, 7,8, 9, 10, 11, or 12) carbon atoms, more preferably an alkynyl group of from 2 to 8 carbon atoms, and even more preferably an alkynyl group of from 2 to 6 carbon atoms. Non-limiting examples include ethynyl, propyn-1-yl, propyn-2-yl, butyn-1-yl, butyn-2-yl, butyn-3-yl, 3-dimethylbutyn-2-yl, pentyn-1-yl, pentyn-2-yl, hexyn-1-yl, 1-heptyn-1-yl, heptyn-3-yl, heptyn-4-yl, octyn-3-yl, nonyn-3-yl, decyn-4-yl, undec-3-yl, dodecyn-4-yl. The alkynyl group may be optionally further substituted with 1 to more substituents.
"aryl" means a substituted or unsubstituted aromatic ring which may be a5 to 8 membered (e.g., 5, 6, 7,8 membered) monocyclic, 5 to 12 membered (e.g., 5, 6, 7,8, 9, 10, 11, 12 membered) bicyclic, or 10 to 15 membered (e.g., 10, 11, 12, 13, 14, 15 membered) tricyclic ring system which may be bridged or spiro, non-limiting examples of which include phenyl, naphthyl. The aryl group may be optionally further substituted with 1 or more substituents.
"heteroaryl" refers to a substituted or unsubstituted aromatic ring, which can be a 3 to 8 membered (e.g., 3,4, 5, 6, 7,8 membered) monocyclic, 5 to 12 membered (e.g., 5, 6, 7,8, 9, 10, 11, 12 membered) bicyclic, or 10 to 15 membered (e.g., 10, 11, 12, 13, 14, 15 membered) tricyclic ring system, and contains 1 to 6 (e.g., 1,2,3,4, 5, 6) heteroatoms selected from N, O or S, preferably 5 to 8 membered heteroaryl, wherein the optionally substituted 1 to 4 (e.g., 1,2,3, 4) N, S in the ring of the heteroaryl can be oxidized to various oxidation states. The heterocyclic group may be attached at a heteroatom or carbon atom, the heteroaryl group may be a bridged or spiro ring, non-limiting examples include cyclic pyridyl, furyl, thienyl, pyranyl, pyrrolyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, piperidinyl benzimidazolyl, benzopyridyl, pyrrolopyridyl. Heteroaryl is optionally further substituted with 1 or more substituents.
"carbocyclyl" or "carbocycle" refers to a saturated or unsaturated aromatic or non-aromatic ring. When aromatic, it is as defined above for "aryl"; when non-aromatic, it may be a 3 to 10 membered (e.g., 3,4, 5, 6, 7,8, 9, 10 membered) monocyclic, 4 to 12 membered (e.g., 4, 5, 6, 7,8, 9, 10, 11, 12 membered) bicyclic, or 10 to 15 membered (e.g., 10, 11, 12, 13, 14, 15 membered) tricyclic ring system, which may be bridged or spiro, non-limiting examples of which include cyclopropyl, cyclobutyl, cyclopentyl, 1-cyclopentyl-1-alkenyl, 1-cyclopentyl-2-alkenyl, 1-cyclopentyl-3-alkenyl, cyclohexyl, 1-cyclohexyl-2-alkenyl, 1-cyclohexyl-3-alkenyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, cycloundecyl, cyclododecyl, or a pharmaceutically acceptable salt thereof,
Figure BDA0002876667980000071
Figure BDA0002876667980000072
Said "carbocyclyl" or "carbocycle" is optionally further substituted with 1 or moreSubstituted by a substituent.
"Heterocyclyl" or "heterocycle" refers to a saturated or unsaturated aromatic or non-aromatic heterocycle, which when aromatic is defined as "heteroaryl" as defined above; when a non-aromatic heterocycle, it may be a 3 to 10 membered (e.g. 3,4, 5, 6, 7,8, 9, 10 membered) monocyclic, 4 to 12 membered (e.g. 4, 5, 6, 7,8, 9, 10, 11, 12 membered) bicyclic or 10 to 15 membered (e.g. 10, 11, 12, 13, 14, 15 membered) tricyclic ring system and contains 1 to 4 (e.g. 1,2,3, 4) heteroatoms selected from N, O or S, preferably a 3 to 8 membered heterocyclyl. From 1 to 4 (e.g., 1,2,3, 4) N, S optionally substituted in the ring of the "heterocyclyl" or "heterocycle" can be oxidized to various oxidation states; "heterocyclyl" or "heterocycle" may be attached at a heteroatom or carbon atom; "Heterocyclyl" or "heterocycle" may be a bridged or spiro ring. Non-limiting examples of "heterocyclyl" or "heterocycle" include epoxyethyl, epoxypropyl, aziridinyl, oxetanyl, azetidinyl, thietanyl, 1, 3-dioxolanyl, 1, 4-dioxolanyl, 1, 3-dioxanyl, azepinyl, oxepinyl, thiepinyl, oxazepinyl, diazepinyl, thiazepinyl, pyridyl, piperidyl, homopiperidinyl, furyl, thienyl, pyranyl, N-alkylpyrrolyl, pyrimidinyl, pyrazinyl, pyridazinyl, piperazinyl, homopiperazinyl, imidazolyl, piperidyl, morpholinyl, thiomorpholinyl, thialkyl, 1, 3-dithianyl, dihydrofuranyl, dithianyl, tetrahydrofuranyl, tetrahydrothienyl, tetrahydropyranyl, tetrahydrothiopyranyl, tetrahydropyrrolyl, Tetrahydroimidazolyl, tetrahydrothiazolyl, tetrahydropyranyl, benzimidazolyl, benzopyridyl, pyrrolopyridyl, chromanyl, 2-pyrrolinyl, 3-pyrrolinyl, indolinyl, 2H-pyranyl, 4H-pyranyl, dioxacyclohexyl, 1, 3-dioxolanyl, pyrazolinyl, dithianyl, dithienoalkyl, dihydrothienyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, 1,2,3, 4-tetrahydroisoquinolinyl, 3-azabicyclo [3.1.0] hexyl, 3-azabicyclo [4.1.0] heptyl, azabicyclo [2.2.2] hexyl, 3H-indolinylquinolizinyl, N-pyridylurea, 1-dioxothiomorpholinyl, azabicyclo [3.2.1] octyl, azabicyclo [5.2.0] nonyl, oxatricyclo [5.3.1.1] dodecyl, Azaadamantyl and oxaspiro [3.3] heptanyl. Said "heterocyclyl" or "heterocycle" may optionally be further substituted with 1 or more substituents.
"cycloalkyl" means a saturated cyclic hydrocarbon group, the ring of which may be a 3 to 10-membered (e.g., 3,4, 5, 6, 7,8, 9, 10-membered) monocyclic ring, a 4 to 12-membered (e.g., 4, 5, 6, 7,8, 9, 10, 11, 12-membered) bicyclic ring, or a 10 to 20-membered (e.g., 4, 5, 6, 7,8, 9, 10, 11, 12-membered) polycyclic ring system, the ring carbon atoms are preferably 3 to 10 carbon atoms, and more preferably 3 to 8 carbon atoms. Non-limiting examples of "cycloalkyl" include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl, cycloheptenyl, 1, 5-cyclooctadienyl, 1, 4-cyclohexadienyl, cycloheptatrienyl, and the like. When the cycloalkyl group is substituted, it may be optionally further substituted with 1 or more substituents.
"heterocycloalkyl" refers to a substituted or unsubstituted saturated nonaromatic cyclic group that can be a 3 to 8 membered (e.g., 3,4, 5, 6, 7,8 membered) monocyclic, 4 to 12 membered (e.g., 4, 5, 6, 7,8, 9, 10, 11, 12 membered) bicyclic, or 10 to 15 membered (e.g., 10, 11, 12, 13, 14, 15 membered) tricyclic ring system and contains 1,2, or 3 heteroatoms selected from N, O or S, preferably a 3 to 8 membered heterocyclic group. The optionally substituted N, S in the ring of "heterocycloalkyl" can be oxidized to various oxidation states; "heterocycloalkyl" can be attached to a heteroatom or a carbon atom; "heterocycloalkyl" can be a bridged or spiro ring. Non-limiting examples of "heterocycloalkyl" include oxiranyl, aziridinyl, oxetanyl, azetidinyl, 1, 3-dioxolanyl, 1, 4-dioxolanyl, 1, 3-dioxanyl, azepinyl, piperidinyl, perinyl, morpholinyl, thiomorpholinyl, 1, 3-dithianyl, tetrahydrofuranyl, tetrahydropyranyl, tetrahydropyrrolyl, tetrahydroimidazolyl, thiazolidinyl, tetrahydropyranyl, azabicyclo [3.2.1] octanyl, azabicyclo [5.2.0] nonanyl, oxatricyclo [5.3.1.1] dodecyl, azaadamantyl, and oxaspiro [3.3] heptanyl.
When "alkyl", "alkoxy", "alkenyl", "alkynyl", "aryl", "heteroaryl", "carbocyclyl", "carbocycle", "heterocyclyl", "heterocycle", "cycloalkyl", "heterocycloalkyl", or "heterocyclyl" as described above is substituted, it may optionally be further substituted with 0, 1,2,3,4, 5, 6, 7,8, 9, or 10 substituents selected from F, Cl, Br, I, hydroxy, mercapto, nitro, cyano, amino, C1-6Alkylamino group, ═ O, C1-6Alkyl radical, C1-6Alkoxy radical, C2-6Alkenyl radical, C2-6Alkynyl, -NRq4Rq5、=NRq6、-C(=O)OC1-6Alkyl, -OC (═ O) C1-6Alkyl, -C (═ O) NRq4Rq5、C3-12Cycloalkyl radical, C3Heterocycloalkyl radical C4-12Heterocycloalkyl radical, C6-12Aryl radical, C5-12Heteroaryl, -C (═ O) OC6-12Aryl, -OC (═ O) C6-12Aryl, -OC (═ O) C5-12Heteroaryl, -C (═ O) OC5-12Heteroaryl, -OC (═ O) C3Heterocycloalkyl, -OC (═ O) C4-12Heterocycloalkyl, -C (═ O) OC3Heterocycloalkyl, -C (═ O) OC4-12Heterocycloalkyl, -OC (═ O) C3-12Cycloalkyl, -C (═ O) OC3-12Cycloalkyl, -NHC (═ O) C3Heterocycloalkyl, -NHC (═ O) C4-12Heterocycloalkyl, -NHC (═ O) C6-12Aryl, -NHC (═ O) C5-12Heteroaryl, -NHC (═ O) C3-12Cycloalkyl, -NHC (═ O) C2-6Alkenyl or-NHC (═ O) C2-6Alkynyl and wherein said substituent C1-6Alkyl radical, C1-6Alkoxy radical, C2-6Alkenyl radical, C2-6Alkynyl, C3-12Cycloalkyl radical, C3Heterocycloalkyl radical, C4-12Heterocycloalkyl radical, C6-12Aryl radical, C5-12Heteroaryl, -NHC (═ O) C6-12Aryl, -NHC (═ O) C5-12Heteroaryl, -NHC (═ O) C3Heterocycloalkyl, -NHC (═ O) C4-12Heterocycloalkyl or-NHC (═ O) C3-12The cycloalkyl group is optionally further substituted by 1 to 3 substituents selected from OH, F, Cl, Br, I、C1-6Alkyl radical, C1-6Alkoxy, -NRq4Rq5Or substituted with a substituent of ═ O; rq1Is selected from C1-6Alkyl radical, C1-6Alkoxy or C6-12An aryl group; rq2、Rq3Selected from H or C1-6An alkyl group; wherein R isq4、Rq5Selected from H, C1-6Alkyl, -NH (C ═ NR)q1)NRq2Rq3、-S(=O)2NRq2Rq3、-C(=O)Rq1or-C (═ O) NRq2Rq3Wherein said C1-6The alkyl is optionally further substituted by 1 or more groups selected from OH, F, Cl, Br, I, C1-6Alkyl radical, C1-6Alkoxy radical, C6-12Aryl radical, C5-12Heteroaryl group, C3-12Cycloalkyl radical, C3Heterocycloalkyl or C4-12Substituted with a substituent of heterocycloalkyl; or Rq4And Rq5And the N atom forms a 3 to 8 membered heterocyclic ring which may contain 1 or more heteroatoms selected from N, O or S.
By "pharmaceutically acceptable salt" or "pharmaceutically acceptable salt thereof" is meant a salt of a compound of the invention that retains the biological effectiveness and properties of the free acid or free base obtained by reaction with a non-toxic inorganic or organic base, and the free base obtained by reaction with a non-toxic inorganic or organic acid.
"pharmaceutical composition" refers to a mixture of one or more compounds of the present invention, pharmaceutically acceptable salts or prodrugs thereof, and other chemical components, wherein "other chemical components" refers to pharmaceutically acceptable carriers, excipients, and/or one or more other therapeutic agents.
By "carrier" is meant a material that does not cause significant irritation to an organism and does not abrogate the biological activity and properties of the administered compound.
"excipient" refers to an inert substance added to a pharmaceutical composition to facilitate administration of a compound. Non-limiting examples include calcium carbonate, calcium phosphate, sugars, starches, cellulose derivatives (including microcrystalline cellulose), gelatin, vegetable oils, polyethylene glycols, diluents, granulating agents, lubricants, binders, and disintegrating agents.
By "prodrug" is meant a compound of the invention that is metabolically convertible in vivo to a biologically active compound. Prodrugs of the invention are prepared by modifying an amino or carboxyl group in a compound of the invention, which modification may be removed by routine manipulation or in vivo, to yield the parent compound. When a prodrug of the present invention is administered to a mammalian subject, the prodrug is cleaved to form a free amino or carboxyl group.
"cocrystal" refers to a crystal of an Active Pharmaceutical Ingredient (API) and a cocrystal former (CCF) bound by hydrogen bonding or other non-covalent bonds, wherein the API and CCF are both solid in their pure state at room temperature and a fixed stoichiometric ratio exists between the components. A co-crystal is a multi-component crystal that contains both a binary co-crystal formed between two neutral solids and a multicomponent co-crystal formed between a neutral solid and a salt or solvate.
"stereoisomers" refers to isomers resulting from the different arrangement of atoms in a molecule, including cis, trans isomers, enantiomers and conformational isomers.
"optional" or "optionally" or "selective" or "selectively" means that the subsequently described event or circumstance may, but need not, occur, and that the description includes instances where the event or circumstance occurs and instances where it does not. For example, "a heterocyclic group optionally substituted with an alkyl group" means that the alkyl group may, but need not, be present, and the description includes the case where the heterocyclic group is substituted with an alkyl group, and the case where the heterocyclic group is not substituted with an alkyl group.
Detailed Description
The following examples illustrate the technical solutions of the present invention in detail, but the scope of the present invention includes but is not limited thereto.
The structure of the compounds is determined by Nuclear Magnetic Resonance (NMR) or (and) Mass Spectrometry (MS). NMR shift (. delta.) of 10-6The units in (ppm) are given. NMR was measured using (Bruker Avance III 400 and Bruker Avance 300) nuclear magnetic spectrometers in deuterated dimethyl sulfoxide (DMSO-d)6) DeuteriumChloroform (CDCl)3) Deuterated methanol (CD)3OD), internal standard Tetramethylsilane (TMS);
MS measurement (Agilent 6120B (ESI)) and Agilent 6120B (APCI));
HPLC was carried out using an Agilent 1260DAD high pressure liquid chromatograph (Zorbax SB-C18100X 4.6mm, 3.5. mu.M);
the thin layer chromatography silica gel plate adopts HSGF254 of tobacco yellow sea or GF254 of Qingdao, the specification of silica gel plate used by Thin Layer Chromatography (TLC) is 0.15mm-0.20mm, and the specification of thin layer chromatography separation and purification product is 0.4mm-0.5 mm;
the column chromatography generally uses 200-mesh and 300-mesh silica gel of the Tibet yellow sea silica gel as a carrier;
the known starting materials of the present invention can be synthesized by methods known in the art or can be purchased from companies such as Tatan technology, Annaiji chemistry, Shanghai Demer, Chengdong chemical industry, Shaoshanghi chemical technology, and Bailingwei technology;
the nitrogen atmosphere refers to that the reaction bottle is connected with a nitrogen balloon with the volume of about 1L;
the hydrogen atmosphere refers to a hydrogen balloon with the volume of about 1L connected with a reaction bottle;
the hydrogenation reaction is usually vacuumized, filled with hydrogen and repeatedly operated for 3 times;
in the examples, the reaction was carried out under a nitrogen atmosphere, unless otherwise specified;
in the examples, unless otherwise specified, the solution means an aqueous solution;
in the examples, the reaction temperature is room temperature, and the optimum reaction temperature is 20 ℃ to 30 ℃;
DCM: dichloromethane;
EA: ethyl acetate;
HCl: hydrochloric acid;
THF: tetrahydrofuran;
DMF: n, N-dimethylformamide;
PE is petroleum ether;
TLC, thin layer chromatography;
SFC: supercritical fluid chromatography;
NCS: n-chlorosuccinimide;
Pd(dppf)Cl2: [1,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride;
DMSO, DMSO: dimethyl sulfoxide;
DTT: dithiothreitol;
ATP: adenosine triphosphate;
DNA: a deoxyribonucleotide;
IC50: refers to the concentration of the compound at which the activity of the DNA-PK kinase is 50% inhibited.
Examples
Example 1
7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -9- (Oxetadin-3-ylmethyl) -7, 9-dihydro-8H-purin-8-one (Compound 1)
7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-9-(oxetan-3-ylmethyl)-7,9-dihydro-8H-purin-8-one
Figure BDA0002876667980000101
The first step is as follows:
2-chloro-4- ((oxetan-3-ylmethyl) amino) pyrimidine-5-carboxylic acid ethyl ester (1b)
ethyl 2-chloro-4-((oxetan-3-ylmethyl)amino)pyrimidine-5-carboxylate
Ethyl 2, 4-dichloropyrimidine-5-carboxylate 1a (5g,22.6mmol) and potassium carbonate (6.2g,44.8mmol) were dissolved in acetonitrile (20mL), and 3-aminomethyloxetane (1.9g, 22.6mmol) was added at 0 ℃ and the mixture was stirred at room temperature for 20 hours. TLC was monitored to completion of the reaction, 30mL of water was added, extracted three times with 60mL of ethyl acetate, washed once with saturated brine, the organic phase was concentrated, and the residue was purified by silica gel column chromatography (petroleum ether: ethyl acetate (v/v) ═ 10:1 to 1:10) to give the title compound, ethyl 2-chloro-4- ((oxetan-3-ylmethyl) amino) pyrimidine-5-carboxylate 1b (white solid, 3.3g, 63% yield).
1H NMR(400MHz DMSO)δ8.66(t,1H),8.60(s,1H),4.62(q,2H),4.37-4.27(m,4H),3.75(d,2H),3.26-3.19(m,1H),1.30(t,3H)。
The second step is that:
2-chloro-4- ((oxetan-3-ylmethyl) amino) pyrimidine-5-carboxylic acid (1c)
2-chloro-4-((oxetan-3-ylmethyl)amino)pyrimidine-5-carboxylic acid
Ethyl 2-chloro-4- ((oxetan-3-ylmethyl) amino) pyrimidine-5-carboxylate 1b (3.3g,12.1mmol) was dissolved in 10mL of tetrahydrofuran and 5mL of water, and lithium hydroxide (582mg,24.2mmol) was added and the mixture was stirred at room temperature for 1 hour. TLC monitored to completion, dried tetrahydrofuran, adjusted pH to 4-5, precipitated a white solid, filtered, and the filter cake was washed twice with petroleum ether/ethyl acetate (v/v ═ 10/1), filtered, and dried to give the title compound, 2-chloro-4- ((oxetan-3-ylmethyl) amino) pyrimidine-5-carboxylic acid 1c (white solid, 2.4g, 82% yield), which was directly subjected to the next experiment.
The third step:
2-chloro-9- (oxetan-3-ylmethyl) -7, 9-dihydro-8H-purin-8-one (1d)
2-chloro-9-(oxetan-3-ylmethyl)-7,9-dihydro-8H-purin-8-one
2-chloro-4- ((oxetan-3-ylmethyl) amino) pyrimidine-5-carboxylic acid 1c (1.3g,5.3mmol) was dissolved in dimethylacetamide (20mL), triethylamine (585mg,5.8mmol), diphenylphosphorylazide (1.6g,5.8mmol) were added, and stirring was carried out at room temperature for 1h, followed by stepwise warming to 120 ℃ and stirring for 1.5 h. TLC was monitored to completion of the reaction, the reaction solution was concentrated, and the residue was purified by silica gel column chromatography (petroleum ether: ethyl acetate (v/v) ═ 5:1 to 1:10) to give the title compound, 2-chloro-9- (oxetan-3-ylmethyl) -7, 9-dihydro-8H-purin-8-one 1d (white solid, 780mg, yield 35%).
1H NMR(400MHz DMSO)δ8.13(s,1H),4.63(q,2H),4.39(t,2H),4.08(d,2H),3.46-3.15(m,3H)。
The fourth step:
2-chloro-7-methyl-9- (oxetan-3-ylmethyl) -7, 9-dihydro-8H-purin-8-one (1e)
2-chloro-7-methyl-9-(oxetan-3-ylmethyl)-7,9-dihydro-8H-purin-8-one
2-chloro-9- (oxetan-3-ylmethyl) -7, 9-dihydro-8H-purin-8-one 1d (800mg, 3.3mmol) was dissolved in dimethylformamide (10mL), dimethyl sulfate (420mg, 3.3mmol) and cesium carbonate (1.5g,4.8mmol) were added at 0 deg.C, and stirring was continued for 1H at 0 deg.C. TLC monitored the reaction to completion, then 10mL water was added, extracted 3 times with ethyl acetate, the organic phase was dried over anhydrous sodium sulfate, concentrated and a solid precipitated which was filtered to give the title compound, 2-chloro-7-methyl-9- (oxetan-3-ylmethyl) -7, 9-dihydro-8H-purin-8-one 1e (white solid, 423mg, 63% yield).
1H NMR(400MHz DMSO)δ8.33(s,1H),4.62(t,2H),4.40(t,2H),4.12(d,2H),3.36(s,3H),2.25(d,1H)。
The fifth step:
7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -9- (Oxetadin-3-ylmethyl) -7, 9-dihydro-8H-purin-8-one (Compound 1)
7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-9-(oxetan-3-ylmethyl)-7,9-dihydro-8H-purin-8-one
2-chloro-9- (3-aminomethyloxetane) -7-methyl-7, 9-dihydro-8H-purin-8-one 1e (423mg, 1.6mmol), 7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-ylamine (132mg, 0.3mmol), cesium carbonate (1g,1.2mmol), tris (dibenzylideneacetone) dipalladium (146mg, 0.16mmol),2,2 '-bis (diphenylphosphino) -1,1' -binaphthyl (200mg, 0.32mmol) were dissolved in dioxane, purged with nitrogen and purged, and stirred at 100 ℃ for 4H. TLC was monitored to completion of the reaction, the reaction solution was concentrated, and the residue was purified by silica gel column chromatography (dichloromethane: methanol (v/v) ═ 30/1) to give the title compound, 7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -9- (oxetan-3-ylmethyl) -7, 9-dihydro-8H-purin-8-one compound 1 (white solid, 12.6mg, 3.2% yield).
1H NMR(400MHz DMSO)δ9.13(s,1H),8.70(s,1H),8.38(s,1H),8.08(s,1H),7.71(s,1H),4.62(q,2H),4.41(t,2H),4.05(d,2H),3.35-3.30(m,4H),2.38(s,3H)。
LC-MS m/z(ESI)=367.10[M+1]。
Example 2
7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -9- ((3-methyloxoheterocyclin-3-yl) methyl) -7, 9-dihydro-8H-purin-8-one (Compound 2)
7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-9-((3-methyloxetan-3-yl)methyl)-7,9-dihydro-8H-purin-8-one
Figure BDA0002876667980000121
The first step is as follows:
2-chloro-4- ((3-methyl-oxa-3-ylmethyl) -amino) -pyrimidine-5-carboxylic acid ethyl ester (2a)
ethyl 2-chloro-4-[(3-methyl-oxetan-3-ylmethyl)-amino]-pyrimidine-5-carboxylic
Ethyl 2, 4-dichloropyrimidine-5-carboxylate 1a (5.0g,22.62mmol), (carbo-3-methyloxetan-3-yl) methylamine (2.29g,22.62mmol) was dissolved in acetonitrile (600mL), stirred, potassium carbonate (4.69g,33.93mmol) was added in portions, and stirred at room temperature for 4 h. TLC after completion of the reaction was monitored and filtered, the filter residue was washed with ethyl acetate (300mL), the filtrate was concentrated to give a crude product which was purified by column separation (n-hexane: ethyl acetate (v/v) ═ 5:1) to give the title compound, ethyl 2-chloro-4- ((3-methyl-oxa-3-ylmethyl) -amino) -pyrimidine-5-carboxylate 2a (white solid, 3.8g, 77.4% yield).
1H NMR(400MHz DMSO)δ8.68(d,1H),8.63(s,1H),4.44(d,2H),4.33-4.29(m,2H),4.23(d,2H),3.69(d,2H),1.31(t,3H),1.26(s,3H)。
The second step is that:
2-chloro-4- (((3-methyl-oxa-3-ylmethyl) -amino) -pyrimidine-5-carboxylic acid (2b)
2-chloro-4-(((3-methyloxetan-3-yl)methyl)amino)pyrimidine-5-carboxylic acid
Ethyl 2-chloro-4- ((3-methyl-oxa-3-ylmethyl) -amino) -pyrimidine-5-carboxylate 2a (3.8g, 13.30mmol) was dissolved in tetrahydrofuran (20mL), water (20mL), and lithium hydroxide (636.9mg,20.60mmol) was added and stirred at room temperature for 1 h. TLC to completion, concentrated to remove tetrahydrofuran, adjusted to pH 5 with 6N hydrochloric acid and a white solid precipitated, filtered, the filter cake was washed twice with petroleum ether and the solid collected to give the title compound, 2-chloro-4- [ ((3-methyl-oxa-3-ylmethyl) -amino ] -pyrimidine-5-carboxylic acid 2b (white solid, 3.0g, 87.54% yield), which was directly subjected to the next step of the experiment.
1H NMR(400MHz DMSO)δ8.87(t,1H),8.59(s,1H),4.43(d,2H),4.23(d,2H),3.68(d,2H),1.26(s,3H)。
The third step:
2-chloro-9- (3-methyl-oxetan-3-ylmethyl) -7, 9-dihydropurin-8-one (2c)
2-chloro-9-(3-methyl-oxetan-3-ylmethyl)-7,9-dihydro-purin-8-one
2-chloro-4- ((3-methyl-oxa-3-ylmethyl) -amino) -pyrimidine-5-carboxylic acid 2b (3.0g, 58.21mmol) was dissolved in dimethylacetamide (40mL), triethylamine (1.18g,11.64mmol), diphenylphosphorylazide (3.2g,11.64mmol) were added, followed by gradual warming to 120 ℃ and stirring for 1 h. TLC was monitored to completion and the reaction was poured into ice water, the solid collected by filtration, washed 3 times with water, concentrated and dried in vacuo to give the title compound, 2-chloro-9- (3-methyl-oxetan-3-ylmethyl) -7, 9-dihydropurin-8-one 2c (white solid, 2.0g, 67.45% yield).
1H NMR(400MHz DMSO)δ11.70(s,1H),8.15(s,1H),4.62(d,2H),4.22(d,2H),3.99(s,2H),1.23(s,3H)。
The fourth step:
2-chloro-7-methyl-9- ((3-methyloxetan-3-yl) methyl) -7, 9-dihydro-8H-purin-8-one (2d)
2-chloro-7-methyl-9-((3-methyloxetan-3-yl)methyl)-7,9-dihydro-8H-purin-8-one
2-chloro-9- (3-methyl-oxetan-3-ylmethyl) -7, 9-dihydropurin-8-one 2c (1g, 3.93mmol) was dissolved in dimethylformamide (20mL), dimethyl sulfate (495mg, 3.93mmol) and cesium carbonate (1.92g,5.89mmol) were added at 0 deg.C, and stirring was continued for 1h at 0 deg.C. TLC monitored to completion of the reaction, the reaction was poured into ice water and a solid precipitated which was filtered and collected to give the title compound, 2-chloro-7-methyl-9- ((3-methyloxoazetidin-3-yl) methyl) -7, 9-dihydro-8H-purin-8-one 2d (white solid, 0.6g, 56.87% yield).
1H NMR(400MHz DMSO)δ8.40(d,1H),4.65(d,2H),4.25(d,2H),4.06(s,2H),3.41(s,3H),1.27(s,3H)。
The fifth step:
7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -9- ((3-methyloxoheterocyclin-3-yl) methyl) -7, 9-dihydro-8H-purin-8-one (Compound 2)
7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-9-((3-methyloxetan-3-yl)methyl)-7,9-dihydro-8H-purin-8-one
2-chloro-7-methyl-9- (((3-methyloxetan-3-yl) methyl) -7, 9-dihydro-8H-purin-8-one 2d (0.6g, 2.23mmol), 7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-ylamine (297mg, 2.01mmol), cesium carbonate (1.45g,4.46mmol), tris (dibenzylideneacetone) dipalladium (204mg, 0.2mmol) and 2,2 '-bis (diphenylphosphino) -1,1' -binaphthyl (277mg, 0.4mmol) were dissolved in dioxane, protected with nitrogen and purged, stirred at 100 ℃ for 1 h.TLC until the reaction was completed, the reaction solution was poured into ice water, the solid was collected, and the solid was column chromatographed (dichloromethane: methanol (v/v) ═ 30/1), the title compound, 7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -9- ((3-methyloxobutan-3-yl) methyl) -7, 9-dihydro-8H-purin-8-one compound 2 (white solid, 0.15g, 17.66% yield) was obtained.
1H NMR(400MHz DMSO)δ9.08(s,1H),8.66(s,1H),8.37(s,1H),8.11(s,1H),7.70(s,1H),4.61(d,2H),4.17(d,2H),3.93(s,2H),3.35(s,2H),2.36(s,3H),1.22(s,3H)。
LC-MS m/z(ESI)=381.10[M+1]。
Example 3
7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -9- ((tetrahydrofuran-2-yl) methyl) -7, 9-dihydro-8H-purin-8-one (Compound 3)
7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-9-((tetrahydrofuran-2-yl)methyl)-7,9-dihydro-8H-purin-8-one
Figure BDA0002876667980000131
Figure BDA0002876667980000141
The first step is as follows:
2-chloro-4- (((tetrahydrofuran-2-yl) methyl) amino) pyrimidine-5-carboxylic acid ethyl ester (3a)
ethyl 2-chloro-4-(((tetrahydrofuran-2-yl)methyl)amino)pyrimidine-5-carboxylate
Ethyl 2, 4-dichloropyrimidine-5-carboxylate 1a (5g,22.6mmol) and potassium carbonate (6.2g,44.8mmol) were dissolved in acetonitrile (20mL), and carbon- (tetrahydrofuran-2-yl) methylamine (2.3g,22.6mmol) was added at 0 ℃ and the mixture was stirred at room temperature for 20 hours. TLC was monitored to the end of the reaction, 30mL of water was added, extracted three times with 60mL of ethyl acetate, the organic layer was washed once with saturated brine, dried over anhydrous sodium sulfate, and the residue after concentration was purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 10:1) to give the title compound, ethyl 2-chloro-4- (((tetrahydrofuran-2-yl) methyl) amino) pyrimidine-5-carboxylate 3a (white solid, 4.7g, 87% yield).
1H NMR(400MHz DMSO)δ8.62(s,1H),8.57(s,1H),4.33-4.28(m,2H),4.04-4.01(m,1H),3.82-3.77(m,1H),3.68-3.58(m,1H),3.47-3.42(m,1H),1.96-1.81(m,2H),1.59-1.54(m,1H),1.32-1.29(m,3H)。
LC-MS m/z(ESI)=286.20[M+1]。
The second step is that:
2-chloro-4- (((tetrahydrofuran-2-yl) methyl) amino) pyrimidine-5-carboxylic acid (3b)
2-chloro-4-(((tetrahydrofuran-2-yl)methyl)amino)pyrimidine-5-carboxylic acid
Ethyl 2-chloro-4- (((tetrahydrofuran-2-yl) methyl) amino) pyrimidine-5-carboxylate 3a (4.7g,16.4mmol) was dissolved in 10mL of tetrahydrofuran and 5mL of water, and lithium hydroxide (788mg,32.8mmol) was added and the mixture was stirred at room temperature for 1 h. TLC monitored to the end of the reaction, dried over tetrahydrofuran, adjusted to pH 4-5 and a white solid precipitated, filtered, and the filter cake was washed twice with petroleum ether/ethyl acetate (v/v ═ 10/1) and concentrated to give the title compound, 2-chloro-4- (((tetrahydrofuran-2-yl) methyl) amino) pyrimidine-5-carboxylic acid 3b (white solid, 3.8g, 83% yield).
1H NMR(400MHz DMSO)δ13.75(s,1H),8.74(t,1H),8.58(s,1H),4.40-4.01(m,1H),3.82-3.76(m,1H),3.69-3.58(m,2H),3.46-3.40(m,1H),1.95-1.90(m,1H),1.86-1.81(m,1H),1.58-1.53(m,1H)。
LC-MS m/z(ESI)=259.20[M+1]。
The third step:
2-chloro-9- ((tetrahydrofuran-2-yl) methyl) -7, 9-dihydro-8H-purin-8-one (3c)
2-chloro-9-((tetrahydrofuran-2-yl)methyl)-7,9-dihydro-8H-purin-8-one
2-chloro-4- (((tetrahydrofuran-2-yl) methyl) amino) pyrimidine-5-carboxylic acid 3b (3.8g, 14.7mmol) was dissolved in dimethylacetamide (20mL), triethylamine (1.8g, 17.6mmol), diphenylphosphorylazide (4.4g, 16.2mmol) were added, followed by gradual temperature rise to 120 ℃ and stirring for 1.5 h. The reaction solution was concentrated, and the residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 5:1 to 1:10) to give the title compound, 2-chloro-9- ((tetrahydrofuran-2-yl) methyl) -7, 9-dihydro-8H-purin-8-one 3c (white solid, 1.3g, yield 46%).
LC-MS m/z(ESI)=255.20[M+1]。
The fourth step:
2-chloro-7-methyl-9- ((tetrahydrofuran-2-yl) methyl) -7, 9-dihydro-8H-purin-8-one (3d)
2-chloro-7-methyl-9-((tetrahydrofuran-2-yl)methyl)-7,9-dihydro-8H-purin-8-one
2-chloro-9- ((tetrahydrofuran-2-yl) methyl) -7, 9-dihydro-8H-purin-8-one 3c (1.3g, 5.1mmol) was dissolved in dimethylformamide (10mL), dimethyl sulfate (644mg, 5.1mmol) and cesium carbonate (2.5g,7.6mmol) were added at 0 deg.C, and stirring was continued for 1H at 0 deg.C. TLC was monitored to the end of the reaction and 10mL water was added and a solid precipitated which was filtered to give the title compound, 2-chloro-7-methyl-9- ((tetrahydrofuran-2-yl) methyl) -7, 9-dihydro-8H-purin-8-one 3d (white solid, 600mg, 45% yield).
1H NMR(400MHz DMSO)δ8.35(s,1H),4.24-4.20(m,1H),3.90-3.85(m,1H),3.79-3.73(m,2H),3.60(q,1H),3.37(s,3H),1.95-1.77(m,3H),1.70-1.63(m,1H)。
LC-MS m/z(ESI)=268.20[M+1]。
The fifth step:
7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -9- ((tetrahydrofuran-2-yl) methyl) -7, 9-dihydro-8H-purin-8-one (Compound 3)
7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-9-((tetrahydrofuran-2-yl)methyl)-7,9-dihydro-8H-purin-8-one
Dissolving 2-chloro-7-methyl-9- ((tetrahydrofuran-2-yl) methyl) -7, 9-dihydro-8H-purin-8-one 3d (200mg, 0.7mmol), 7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-ylamine (105mg, 0.7mmol), cesium carbonate (456mg,1.4mmol), methanesulfonic acid (2-dicyclohexylphosphine-3, 6-dimethoxy-2 ',4',6 '-triisopropyl-1, 1' -biphenyl) (2 '-amino-1, 1' -biphenyl-2-yl) palladium (II) (63.4mg, 0.07mmol) in dioxane (3mL), stirring at 100 deg.C for 4h under nitrogen protection and ventilation. TLC was monitored to the end of the reaction, the reaction solution was concentrated, and the residue was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 30/1) to give the title compound, 7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -9- ((tetrahydrofuran-2-yl) methyl) -7, 9-dihydro-8H-purin-8-one compound 3 (white solid, 30mg, 31% yield).
1H NMR(400MHz DMSO)δ9.15(s,1H),8.67(s,1H),8.36(s,1H),8.09(s,1H),7.69(s,1H),4.27-4.22(m,3H),3.84-3.67(m,3H),3.62-3.57(m,1H),2.38(s,3H),1.94-1.77(m,3H),1.69-1.62(m,1H)。
LC-MS m/z(ESI)=381.20[M+1]。
Example 4
7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -9- ((tetrahydrofuran-3-yl) methyl) -7, 9-dihydro-8H-purin-8-one (Compound 4)
7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-9-((tetrahydrofuran-3-yl)methyl)-7,9-dihydro-8H-purin-8-one
Figure BDA0002876667980000151
The first step is as follows:
2-chloro-4- (((tetrahydrofuran-3-yl) methyl) amino) pyrimidine-5-carboxylic acid ethyl ester (4a)
ethyl 2-chloro-4-(((tetrahydrofuran-3-yl)methyl)amino)pyrimidine-5-carboxylate
Ethyl 2, 4-dichloropyrimidine-5-carboxylate 1a (5g,22.6mmol) and potassium carbonate (6.2g,44.8mmol) were dissolved in acetonitrile (20mL), and (tetrahydrofuran-3-yl) methylamine (2.3g,22.6mmol) was added at 0 ℃ and stirred at room temperature for 20 h. TLC was monitored to the end of the reaction, 30mL of water was added, extracted three times with 60mL of ethyl acetate, the organic layer was washed once with saturated brine, dried over anhydrous sodium sulfate, and the residue after concentration was purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 10:1) to give the title compound, ethyl 2-chloro-4- (((tetrahydrofuran-3-yl) methyl) amino) pyrimidine-5-carboxylate 4a (white solid, 4.4g, 85% yield).
1H NMR(400MHz DMSO)δ8.61-8.58(m,2H),4.31(q,2H),3.78-3.73(m,1H),3.69-3.58(m,2H),3.48-3.44(m,3H),2.57-2.53(m,1H),1.98-1.91(m,1H),1.62-1.58(m,1H),1.31(t,3H)。
LC-MS m/z(ESI)=286.20[M+1]。
The second step is that:
2-chloro-4- (((tetrahydrofuran-3-yl) methyl) amino) pyrimidine-5-carboxylic acid (4b)
2-chloro-4-(((tetrahydrofuran-3-yl)methyl)amino)pyrimidine-5-carboxylic acid
Ethyl 2-chloro-4- (((tetrahydrofuran-3-yl) methyl) amino) pyrimidine-5-carboxylate 4a (4.4g,15.3mmol) was dissolved in 10mL of tetrahydrofuran and 5mL of water, and lithium hydroxide (736mg,30.6mmol) was added and the mixture was stirred at room temperature for 1 h. TLC monitored to the end of the reaction, dried over tetrahydrofuran, adjusted to pH 4-5 and a white solid precipitated, filtered, and the filter cake was washed twice with petroleum ether/ethyl acetate (v/v ═ 10/1) and concentrated to give the title compound, 2-chloro-4- (((tetrahydrofuran-3-yl) methyl) amino) pyrimidine-5-carboxylic acid 4b (white solid, 3.4g, 80% yield).
1H NMR(400MHz DMSO)δ13.75(s,1H),8.75(s,1H),8.57(s,1H),3.78-3.73(m,3H),3.47-3.43(m,3H),2.58-2.52(m,1H),1.98-1.89(m,3H),1.63-1.55(m,1H)。
LC-MS m/z(ESI)=259.20[M+1]。
The third step:
2-chloro-9- ((tetrahydrofuran-3-yl) methyl) -7, 9-dihydro-8H-purin-8-one (4c)
2-chloro-9-((tetrahydrofuran-3-yl)methyl)-7,9-dihydro-8H-purin-8-one
2-chloro-4- ((tetrahydrofuran-3-yl) methyl) amino) pyrimidine-5-carboxylic acid 4b (3.4g, 13.0mmol) was dissolved in dimethylacetamide (20mL), triethylamine (1.58g,13mmol) and diphenylphosphoryl azide (3.6g,13mmol) were added, followed by gradual warming to 120 ℃ and stirring for 1.5 h. TLC was monitored to the end of the reaction, the reaction was concentrated, and the residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 5:1 to 1:10) to give the title compound, 2-chloro-9- ((tetrahydrofuran-3-yl) methyl) -7, 9-dihydro-8H-purin-8-one 4c (white solid, 1.46g, 50% yield).
1H NMR(400MHz DMSO)δ11.65(s,1H),8.14(s,1H),3.80-3.74(m,3H),3.66-3.57(m,2H),3.52-3.49(m,1H),2.73-2.69(m,1H),1.98-1.89(m,1H),1.67-1.60(m,1H)。
LC-MS m/z(ESI)=255.20[M+1]。
The fourth step:
2-chloro-7-methyl-9- ((tetrahydrofuran-3-yl) methyl) -7, 9-dihydro-8H-purin-8-one (4d)
2-chloro-7-methyl-9-((tetrahydrofuran-3-yl)methyl)-7,9-dihydro-8H-purin-8-one
2-chloro-9- ((tetrahydrofuran-3-yl) methyl) -7, 9-dihydro-8H-purin-8-one 4c (1.6g, 5.2mmol) was dissolved in dimethylformamide (10mL), dimethyl sulfate (663mg, 5.2mmol) and cesium carbonate (2.4g,7.7mmol) were added at 0 deg.C, and stirring was continued for 1H at 0 deg.C. TLC was monitored to the end of the reaction and 10mL water was added and a solid precipitated which was filtered to give the title compound, 2-chloro-7-methyl-9- ((tetrahydrofuran-3-yl) methyl) -7, 9-dihydro-8H-purin-8-one 4d (white solid, 1.2g, 80% yield).
1H NMR(400MHz DMSO)δ8.35(s,1H),3.81-3.75(m,3H),3.65-3.57(m,2H),3.52-3.49(m,1H),3.37(s,3H),2.73-2.70(m,1H),1.96-1.91(m,1H),1.67-1.61(m,1H)。
LC-MS m/z(ESI)=268.20[M+1]。
The fifth step:
7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -9- ((tetrahydrofuran-3-yl) methyl) -7, 9-dihydro-8H-purin-8-one (Compound 4)
7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-9-((tetrahydrofuran-3-yl)methyl)-7,9-dihydro-8H-purin-8-one
2-chloro-9- ((tetrahydrofuran-3-yl) methyl) -7, 9-dihydro-8H-purin-8-one 4d (200mg, 0.7mmol), 7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-ylamine (105mg, 0.7mmol), cesium carbonate (456mg,1.4mmol), methanesulfonic acid (2-dicyclohexylphosphine-3, 6-dimethoxy-2 ',4',6 '-triisopropyl-1, 1' -biphenyl) (2 '-amino-1, 1' -biphenyl-2-yl) palladium (II) (CAS: 1470372-59-8) (63.4mg, 0.07mmol) were dissolved in dioxane (3mL), nitrogen protected and purged, stirring was carried out at 100 ℃ for 4 h. TLC was monitored to the end of the reaction, the reaction solution was concentrated, and the residue was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 30/1) to give the title compound 7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -9- ((tetrahydrofuran-3-yl) methyl) -7, 9-dihydro-8H-purin-8-one compound 4 (white solid, 30mg, 31% yield).
1H NMR(400MHz DMSO)δ9.14(s,1H),8.69(s,1H),8.37(s,1H),8.09(s,1H),7.70(s,1H),3.78-3.72(m,3H),3.66-3.57(m,2H),3.53-3.50(m,1H),3.32(s,3H),2.79-2.72(m,1H),2.38(s,3H),1.96-1.87(m,1H),1.69-1.60(m,1H)。
LC-MS m/z(ESI)=381.20[M+1]。
Example 5
7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -9- ((tetrahydro-2H-pyran-4-yl) methyl) -7, 9-dihydro-8H-purin-8-one (Compound 5)
7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-9-((tetrahydro-2H-pyran-4-yl)methyl)-7,9-dihydro-8H-purin-8-one
Figure BDA0002876667980000171
The first step is as follows:
2-chloro-4- (((tetrahydro-2H-pyran-4-yl) methyl) amino ] pyrimidine-5-carboxylic acid ethyl ester (5a)
ethyl 2-chloro-4-(((tetrahydro-2H-pyran-4-yl)methyl)amino)pyrimidine-5-carboxylate
Ethyl 2, 4-dichloropyrimidine-5-carboxylate 1a (5g,22.6mmol), potassium carbonate (6.2g,44.8mmol) were dissolved in acetonitrile (20mL), and carbon- (tetrahydropyran-4-yl) -methylamine (2.6g,22.6mmol) was added at 0 ℃ and stirred at room temperature for 20 h. TLC was monitored to the end of the reaction, 30mL of water was added, extracted three times with 60mL of ethyl acetate, the organic layer was washed once with saturated brine, dried over anhydrous sodium sulfate, and the residue after concentration was purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 10:1) to give the title compound, ethyl 2-chloro-4- (((tetrahydro-2H-pyran-4-yl) methyl) amino) pyrimidine-5-carboxylate 5a (white solid, 3.6g, 70% yield).
1H NMR(400MHz DMSO)δ8.60(s,1H),8.54(t,1H),4.31(q,2H),3.84(dd,2H),3.38(t,2H),3.29-3.23(m,2H),1.87-1.82(m,1H),1.55(dd,2H),1.30(t,3H),1.25-1.17(m,2H)。
LC-MS m/z(ESI)=301.20[M+1]。
The second step is that:
2-chloro-4- (((tetrahydro-2H-pyran-4-yl) methyl) amino) pyrimidine-5-carboxylic acid (5b)
2-chloro-4- (((tetrahydro-2H-pyran-4-yl) methyl) amino) pyrimidine-5-carboxlic acid Ethyl 2-chloro-4- (((tetrahydro-2H-pyran-4-yl) methyl) amino ] pyrimidine-5-carboxylate 5a (4.6g,15.3mmol) was dissolved in 10mL of tetrahydrofuran, 5mL of water, lithium hydroxide (736mg,30.6mmol) was added, and the mixture was stirred at room temperature for 1H. TLC monitored to the end of the reaction, dried over tetrahydrofuran, adjusted to PH 4-5 and a white solid precipitated, filtered, and the filter cake was washed twice with petroleum ether/ethyl acetate (v/v ═ 10/1) and concentrated to give the title compound, 2-chloro-4- (((tetrahydro-2H-pyran-4-yl) methyl) amino) pyrimidine-5-carboxylic acid 5b (white solid, 4.0g, 83% yield).
1H NMR(400MHz DMSO)δ13.72(s,1H),8.71(s,1H),8.56(s,1H),3.84(dd,2H),3.37(t,2H),3.29-3.23(m,2H),1.87-1.80(m,1H),1.54(dd,2H),1.26-1.17(m,2H)。
LC-MS m/z(ESI)=273.20[M+1]。
The third step:
2-chloro-9- ((tetrahydro-2H-pyran-4-yl) methyl) -7, 9-dihydro-8H-purin-8-one (5c)
2-chloro-9-((tetrahydro-2H-pyran-4-yl)methyl)-7,9-dihydro-8H-purin-8-one
2-chloro-4- (((tetrahydro-2H-pyran-4-yl) methyl) amino) pyrimidine-5-carboxylic acid 5b (4g, 14.6mmol) was dissolved in dimethylacetamide (20mL), triethylamine (2.9g,29mmol) and diphenylphosphoryl azide (5.2g,19mmol) were added, followed by gradual warming to 120 ℃ and stirring for 1.5H. TLC was monitored to the end of the reaction, the reaction was concentrated, and the residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 5:1 to 1:10) to give the title compound, 2-chloro-9- ((tetrahydro-2H-pyran-4-yl) methyl) -7, 9-dihydro-8H-purin-8-one 5c (white solid, 1.46g, 50% yield).
1H NMR(400MHz DMSO)δ11.63(s,1H),8.13(s,1H),3.81(dd,2H),3.66(d,2H),3.25-3.15(m,2H),2.07-2.00(m,1H),1.50(dd,2H),1.29-1.21(m,2H)。
LC-MS m/z(ESI)=269.20[M+1]。
The fourth step:
2-chloro-7-methyl-9- ((tetrahydro-2H-pyran-4-yl) methyl) -7, 9-dihydro-8H-purin-8-one (5d)
2-chloro-7-methyl-9-((tetrahydro-2H-pyran-4-yl)methyl)-7,9-dihydro-8H-purin-8-one
2-chloro-9- ((tetrahydro-2H-pyran-4-yl) methyl) -7, 9-dihydro-8H-purin-8-one 5c (1.4g,5.4mmol) was dissolved in dimethylformamide (10mL), dimethyl sulfate (688mg, 5.4mmol) and cesium carbonate (2.6g,8.0mmol) were added at 0 deg.C, and stirring was continued for 1H at 0 deg.C. TLC was monitored to the end of the reaction and 10mL water was added and a solid precipitated which was filtered to give the title compound, 2-chloro-7-methyl-9- ((tetrahydro-2H-pyran-4-yl) methyl) -7, 9-dihydro-8H-purin-8-one 5d (white solid, 1.2g, 80% yield).
1H NMR(400MHz DMSO)δ8.34(s,1H),3.81(dd,2H),3.70(d,2H),3.36(s,3H),3.25-3.19(m,2H),2.07-2.00(m,1H),1.51(dd,2H),1.29-1.19(m,2H)。
LC-MS m/z(ESI)=283.20[M+1]。
The fifth step:
7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -9- ((tetrahydro-2H-pyran-4-yl) methyl) -7, 9-dihydro-8H-purin-8-one (Compound 5)
7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-9-((tetrahydro-2H-pyran-4-yl)methyl)-7,9-dihydro-8H-purin-8-one
Dissolving 2-chloro-7-methyl-9- ((tetrahydro-2H-pyran-4-yl) methyl) -7, 9-dihydro-8H-purin-8-one 5d (200mg, 0.7mmol), 7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-ylamine (105mg, 0.7mmol), cesium carbonate (456mg,1.4mmol), methanesulfonic acid (2-dicyclohexylphosphine-3, 6-dimethoxy-2 ',4',6 '-triisopropyl-1, 1' -biphenyl) (2 '-amino-1, 1' -biphenyl-2-yl) palladium (II) (63.4mg, 0.07mmol) in dioxane (3mL), nitrogen protection and purging, stirring was carried out at 100 ℃ for 4 h. TLC was monitored to the end of the reaction, the reaction solution was concentrated, and the residue was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 30/1) to give the title compound, 7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -9- ((tetrahydro-2H-pyran-4-yl) methyl) -7, 9-dihydro-8H-purin-8-one compound 5 (white solid, 12.6mg, 3.2% yield).
1H NMR(400MHz DMSO)δ9.23(s,1H),9.21(s,1H),8.51(s,1H),8.12(s,1H),7.79(s,1H),3.84-3.81(m,2H),3.65(d,2H),3.32(s,3H),3.26-3.20(m,2H),2.40(s,3H),2.10-2.04(m,1H),1.50(d,2H),1.28-1.17(m,2H)。
LC-MS m/z(ESI)=395.20[M+1]。
Example 6
9- (3-Hydroxycyclopentyl) -7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -7, 9-dihydro-
8H-purin-8-one (Compound 6)
9-(3-hydroxycyclopentyl)-7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-7,9-dihydro-8H-purin-8-one
Figure BDA0002876667980000191
The first step is as follows:
(3-Hydroxycyclopentyl) carbamic acid tert-butyl ester (6b)
tert-butyl(3-oxocyclopentyl)carbamate
Tert-butyl (3-oxocyclopentyl) carbamate 6a (10g, 50.19mmol) was dissolved in 60mL THF and sodium borohydride (2.85g, 75.28mmol) was added slowly under ice bath. After 3h, the reaction was monitored by TLC to be complete. Saturated NaCl solution was slowly added until no air bubbles appeared, EA was added for extraction, the organic phase was collected, and the organic solvent was removed by rotary evaporation to give the title compound, tert-butyl (3-hydroxycyclopentyl) carbamate 6b (yellow oily liquid, 10.10g, 99% yield).
LC-MS m/z(ESI)=202.20[M+1]。
The second step is that:
3-Aminocyclopentane-1-ol hydrochloride (6c)
3-aminocyclopentan-1-ol hydrochloride
Tert-butyl (3-hydroxycyclopentyl) carbamate 6b (10.10g, 50.18mmol) was added to 20mL of 1, 4-dioxane solution and stirred at room temperature until completely dissolved, the reaction was placed in an ice bath, 4M hydrochloric acid-1, 4-dioxane solution (8.75g, 239.87mmol) was slowly added and stirring continued until a small amount of solid appeared, and after 2h, TLC monitored for completion of the reaction, filtration and concentration gave the title compound, 3-aminocyclopentane-1-alkoxide 6c (black oil, 6.91g, 100% yield).
LC-MS m/z(ESI)=138.10[M+1]。
The third step:
2-chloro-4- ((3-hydroxycyclopentyl) amino) pyrimidine-5-carboxylic acid ethyl ester (6d)
ethyl 2-chloro-4-((3-hydroxycyclopentyl)amino)pyrimidine-5-carboxylate
3-Aminocyclopentane-1-ol hydrochloride 6c (6.91g, 50.22mmol) and ethyl 2, 4-dichloropyrimidine-5-carboxylate (13.32g, 60.26mmol) were added to 75mL of ACN, and after stirring at room temperature until ethyl 2, 4-dichloropyrimidine-5-carboxylate was dissolved, potassium carbonate (17.35g, 125.54mmol) was slowly added under ice bath. After stirring at room temperature for 10 hours, the reaction was completed, the reaction mixture was filtered, the organic phase was collected, extracted with EA (3 × 100mL), the organic phase was collected and concentrated, and the residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 1/1). The title compound, ethyl 2-chloro-4- ((3-hydroxycyclopentyl) amino) pyrimidine-5-carboxylate 6d (yellow oily liquid, 7.78g, 54.2% yield) was obtained.
LC-MS m/z(ESI)=286.10[M+1]。
The fourth step:
2-chloro-4- ((3-hydroxycyclopentyl) amino) pyrimidine-5-carboxylic acid (6e)
2-chloro-4-((3-hydroxycyclopentyl)amino)pyrimidine-5-carboxylic acid
Ethyl 2-chloro-4- ((3-hydroxycyclopentyl) amino) pyrimidine-5-carboxylate 6d (7.78g, 27.23mmol) was dissolved in 50mL THF, lithium hydroxide (1.3g, 54.46mmol) was dissolved in 50mL water, lithium hydroxide aqueous solution was slowly added to the THF solution under ice bath to cause turbidity, after 2h, TLC monitoring of the reaction was complete, THF was spun dry, hydrochloric acid solution was added to adjust pH to 4, the solid was collected by filtration, and the filtrate was extracted with EA (3 × 50 mL). The organic phase was combined with the filter residue and concentrated to give the title compound, 2-chloro-4- ((3-hydroxycyclopentyl) amino) pyrimidine-5-carboxylic acid 6e (pale yellow solid, 6.78g, 96% yield).
LC-MS m/z(ESI)=258.10[M+1]。
The fifth step:
2-chloro-9- (3-hydroxycyclopentyl) -7, 9-dihydro-8H-purin-8-one (6f)
2-chloro-9-(3-hydroxycyclopentyl)-7,9-dihydro-8H-purin-8-one
2-chloro-4- ((3-hydroxycyclopentyl) amino) pyrimidine-5-carboxylic acid 6e (6.0g, 23.29mmol) was dissolved in 60mL DMAc under ice-bath, diphenyl phosphorazidate (6.41g, 23.29mmol) was slowly added, followed by triethylamine (2.36g, 23.29 mmol). After stirring at room temperature for 1H, heating in an oil bath at 90 ℃ for 5H, monitoring by TLC for completion of the reaction, dropping the reaction solution into 200mL of water to precipitate a solid, adding EA (3 × 100mL) for extraction, collecting the organic phase, washing with a saturated ammonium chloride solution, drying with anhydrous sodium sulfate, and concentrating to obtain the title compound, 2-chloro-9- (3-hydroxycyclopentyl) -7, 9-dihydro-8H-purin-8-one 6f (yellow oily liquid, 5.47g, 92% yield).
LC-MS m/z(ESI)=255.10[M+1]。
And a sixth step:
2-chloro-9- (3-hydroxycyclopentyl) -7-methyl-7, 9-dihydro-8H-purin-8-one (6g)
2-chloro-9-(3-hydroxycyclopentyl)-7-methyl-7,9-dihydro-8H-purin-8-one
2-chloro-9- (3-hydroxycyclopentyl) -7, 9-dihydro-8H-purin-8-one 6f (2.5g, 9.82mmol) and cesium carbonate (4.16g, 12.76mmol) were dissolved in 40mL DMF, stirred in an ice bath for 10min, then dimethyl sulfate (1.24g, 9.82mmol) was slowly added, and after 1H, the reaction was monitored by TLC for completion. To the reaction solution was added 100mL of water, solids were precipitated, EA (3 × 50mL) was added for extraction, the organic phase was washed with saturated brine, the organic phase was collected and concentrated, and the residue was purified by column chromatography to give the title compound, 2-chloro-9- (3-hydroxycyclopentyl) -7-methyl-7, 9-dihydro-8H-purin-8-one, 6g (orange solid, 900mg, 34% yield).
1H NMR(400MHz,DMSO-d6)δ8.35(s,1H),4.90(d,1H),4.71–4.62(m,1H),4.16–4.08(m,1H),3.35(s,3H),2.29–2.23(m,1H),2.17–2.10(m,2H),1.92–1.77(m,3H)。
LC-MS m/z(ESI)=269.10[M+1]。
The seventh step:
9- (3-Hydroxycyclopentyl) -7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -7, 9-dihydro-8H-purin-8-one (Compound 6)
9-(3-hydroxycyclopentyl)-7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)7,9-dihydro-8H-purin-8-one
Mixing 6g (300mg, 1.12mmol) of 2-chloro-9- (3-hydroxycyclopentyl) -7-methyl-7, 9-dihydro-8H-purin-8-one and 7-methyl- [1,2, 4%]Triazolo [1,5-a]Pyridine-6-amine (165mg, 1.12mmol), cesium carbonate (727mg, 2.23mmol), methanesulfonic acid (2-dicyclohexylphosphine-3, 6-dimethoxy-2 ',4',6 '-triisopropyl-1, 1' -biphenyl) (2 '-amino-1, 1' -biphenyl-2-yl) palladium (R) ((R) ())II) (101mg, 0.11mmol) was dissolved in 1, 4-dioxane (5mL), purged with nitrogen, stirred at 110 ℃ for 4.5h and monitored by TLC to the end of the reaction. The reaction solution was concentrated, and the residue was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 20/1), and subjected to Pre-HPLC to give the title compound, 9- (3-hydroxycyclopentyl) -7-methyl-2- ((7-methyl- [1,2, 4-methyl- [ 7 ])]Triazolo [1,5-a]Pyridin-6-yl) amino) -7, 9-dihydro-8H-purin-8-one (compound 6, i.e. compound 6-1 and compound 6-2), two white solids, compound 6-1(69mg, yield 16%, RT ═ 5.15min, dr ═ 99.11%), compound 6-2(48mg, yield 11%, RT ═ 5.53min, dr ═ 99.04%), Pre-hplc (oz), mobile phase CO, mobile phase270/30,/isopropanol; the column temperature is 35 ℃; column pressure 80 bar; the flow rate is 1 mL/min; a detector signal channel is 220nm @4 nm; the wavelength of the diode array detector is 200-400 nm.
Compound 6-1:
1H NMR(400MHz,DMSO-d6)δ9.10(s,1H),8.64(s,1H),8.38(s,1H),8.10(s,1H),7.70(s,1H),4.97–4.89(m,1H),4.58(d,1H),4.23(s,1H),3.29(s,3H),2.38(s,3H),2.33–2.31(m,1H),2.14–2.06(m,2H),1.81–1.76(m,1H),1.49–1.46(m,1H),0.86–0.82(m,1H)。
compound 6-2:
1H NMR(400MHz,DMSO-d6)δ9.10(s,1H),8.64(s,1H),8.38(s,1H),8.10(s,1H),7.70(s,1H),4.97–4.89(m,1H),4.58(d,1H),4.24(s,1H),3.29(s,3H),2.38(s,3H),2.31–2.24(m,1H),2.05–1.99(m,2H),1.81–1.76(m,1H),1.50–1.44(m,1H),0.86–0.82(m,1H)。
LC-MS m/z(ESI)=383.40[M+1]。
example 7
1-methyl-4- (7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) cyclohexane-1-carbonitrile (Compound 7)
1-methyl-4-(7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-8-oxo-7,8-dihydro-9H-purin-9-yl)cyclohexane-1-carbonitrile
Figure BDA0002876667980000211
Figure BDA0002876667980000221
The first step is as follows:
1-methyl-4-oxocyclohexane-1-carbonitrile (7b)
2-1-methyl-4-oxocyclohexane-1-carbonitrile
1, 4-dioxaspiro [4.5] decane-8-carbonitrile 7a (5.00g,29.9mmol) was dissolved in THF (60mL), lithium bistrifluoromethanesulfonylimide (9.87mL,34.38mmol) was slowly added dropwise at 0 ℃ and the reaction stirred, after 1h iodomethane (4.24g,29.9mmol) was slowly added and the reaction stirred for 1 h. TLC was carried out until the reaction was completed, and the mixture was extracted 2 times with a saturated ammonium chloride solution and with bromine water and ethyl acetate, dried over anhydrous magnesium sulfate, filtered and concentrated to obtain an organic layer as intermediate 8-methyl-1, 4-dioxaspiro [4.5] decane-8-carbonitrile, which was dissolved in THF (60mL) and then 3M HCl (60mL) was added to the reaction solution and the mixture was heated to 50 ℃ for 5 hours to react. TLC monitored the reaction to completion, cooled to room temperature and adjusted to pH 7-8 with 3M NaOH, extracted 3 times with dichloromethane, dried over anhydrous sodium sulfate and concentrated the organic phase to give the title compound, 1-methyl-4-oxocyclohexane-1-carbonitrile 7b (yellow liquid, 3.48g, 84.84% yield).
LC-MS m/z(ESI)=138.10[M+1]。
The second step is that:
4-amino-1-methylcyclohexane-1-carbanitrile (7c)
4-amino-1-methylcyclohexane-1-carbonitrile
Dissolving 1-methyl-4-oxocyclohexane-1-carbonitrile 7b (3.48g, 25.37mmol) in anhydrous methanol (50mL), adding ammonium formate (16.00g,253.70mmol) under stirring at normal temperature for reaction for 4h, adding sodium cyanoborohydride (1.88g,30.44mmol) and stirring for reaction for 3h, monitoring by TLC until the reaction is finished, adding 2N HCl to adjust the pH value to 1-2 and stirring for reaction for 0.5h, adding ethyl acetate to extract for 2 times, adjusting the pH value to about 10 with 2N NaOH solution, extracting with dichloromethane for 3 times, drying over anhydrous magnesium sulfate, and concentrating the organic phase to obtain the title compound, 4-amino-1-methylcyclohexane-1-carbonitrile 7c (pale yellow liquid, 2.80g, 79.85% yield).
LC-MS m/z(ESI)=139.10[M+1]。
The third step:
2-chloro-4- ((4-cyano-4-methylcyclohexyl) amino) pyrimidine-5-carboxylic acid ethyl ester (7d)
ethyl 2-chloro-4-((4-cyano-4-methylcyclohexyl)amino)pyrimidine-5-carboxylate
Ethyl 2, 4-dichloro-5-pyrimidinecarboxylate 1a (6.72g,30.39mmol) was dissolved in acetonitrile (30mL), potassium carbonate (8.40g, 60.78mmol) was added with stirring at 0 ℃, a solution of 4-amino-1-methylcyclohexane-1-carbonitrile 7c (2.80g, 20.26mmol) in acetonitrile (10mL) was slowly added dropwise and the reaction was stirred for 1h, monitored by TLC until the reaction was complete, filtered over celite and purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 20:1) to give the title compound, ethyl 2-chloro-4- ((4-cyano-4-methylcyclohexyl) amino) pyrimidine-5-carboxylate 7d (white solid, 2.63g, 40.22% yield).
1H NMR(400MHz,Chloroform-d)δ8.68(d,1H),4.38(p,2H),2.23–1.82(m,4H),1.79–1.49(m,6H),1.46–1.36(m,6H)。
LC-MS m/z(ESI)=323.10[M+1]。
The fourth step:
2-chloro-4- ((4-cyano-4-methylcyclohexyl) amino) pyrimidine-5-carboxylic acid (7e)
2-chloro-4-((4-cyano-4-methylcyclohexyl)amino)pyrimidine-5-carboxylic acid
Ethyl 2-chloro-4- ((4-cyano-4-methylcyclohexyl) amino) pyrimidine-5-carboxylate 7d (2.63g, 8.15mmol) was dissolved in tetrahydrofuran/water (20mL/10mL), lithium hydroxide monohydrate (1.00g, 24.45mmol) was added, the reaction was stirred at room temperature for 2h, monitored by TLC to the end of the reaction, concentrated to evaporate tetrahydrofuran, 2N HCl was added to adjust the pH to about 3-4, a white solid precipitated, filtered, and the filter cake was washed twice with water and petroleum ether/ethyl acetate (v/v ═ 10/1) to give the title compound, 2-chloro-4- ((4-cyano-4-methylcyclohexyl) amino) pyrimidine-5-carboxylic acid 7e (white solid, 2.20g, yield 91.59%).
1H NMR(400MHz,DMSO-d6)δ13.75(s,1H),8.59(s,1H),8.52(d,1H),3.97(m,1H),2.06–1.92(m,4H),1.63–1.47(m,4H),1.35(s,3H)。
LC-MS m/z(ESI)=295.10[M+1]。
The fifth step:
4- (2-chloro-8-oxo-7, 8-dihydro-9H-purin-9-yl) -1-methylcyclohexane-1-carba-nitrile (7f)
4-(2-chloro-8-oxo-7,8-dihydro-9H-purin-9-yl)-1-methylcyclohexane-1-carbonitrile
2-chloro-4- ((4-cyano-4-methylcyclohexyl) amino) pyrimidine-5-carboxylic acid 7e (2.20g, 7.46mmol) was dissolved in N, N-dimethylacetamide (15mL), and triethylamine (1.04mL,7.46mmol) and diphenylphosphorylazide (1.61mL,7.46mmol) were added under stirring at room temperature for 2 hours, followed by heating to 110 ℃ and reflux reaction for 2.5 hours. TLC was monitored to the end of the reaction, extracted 3 times with water and ethyl acetate, and concentrated organic layer silica gel column chromatography (dichloromethane/methanol (v/v) ═ 80/1) to give the title compound, 4- (2-chloro-8-oxo-8, 9-dihydro-7 h-purin-9-yl) -1-methylcyclohexane-1-carbonitrile 7f (white solid, 0.9g, 41.35% yield).
1H NMR(400MHz,DMSO-d6)δ11.64(s,1H),8.13(s,1H),4.31–4.20(m,1H),2.45–2.33(m,2H),2.01–1.94(m,4H),1.71–1.64(m,2H),1.49(s,3H)。
LC-MS m/z(ESI)=292.10[M+1]。
And a sixth step:
4- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) -1-methylcyclohexane-1-carbanitrile (7g)
4-(2-chloro-7-methyl-8-oxo-7,8-dihydro-9H-purin-9-yl)-1-methylcyclohexane-1-carbonitrile
4- (2-chloro-8-oxo-8, 9-dihydro-7 h-purin-9-yl) -1-methylcyclohexane-1-carbonitrile 7f (0.90g,3.08mmol) was dissolved in N, N-dimethylformamide (10mL) and dimethyl sulfate (0.29mL,3.08mmol) and cesium carbonate (1.51g,4.62mmol) were added with stirring at 0 ℃ for 2 h. TLC monitored to completion of the reaction, the reaction mixture was slowly added dropwise to ice water and stirred to precipitate a white solid, which was washed with water and petroleum ether 3 times and filtered to obtain 7g (white solid, 0.85g, 90.26% yield) of the title compound, 4- (2-chloro-7-methyl-8-oxo-8, 9-dihydro-7 h-purin-9-yl) -1-methylcyclohexane-1-carbonitrile.
1H NMR(400MHz,DMSO-d6)δ8.35(s,1H),4.35–4.20(m,1H),3.36(s,3H),2.48–2.31(m,2H),2.06–1.95(m,3H),1.83–1.76(m,1H),1.71–1.57(m,2H),1.48(s,1H),1.36(s,2H)。
LC-MS m/z(ESI)=306.10[M+1]。
The seventh step:
1-methyl-4- (7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) cyclohexane-1-carbonitrile (Compound 7-1 and Compound 7-2)
1-methyl-4-(7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-8-oxo-7,8-dihydro-9H-purin-9-yl)cyclohexane-1-carbonitrile
7g (0.15g,0.49mmol) of 4- (2-chloro-7-methyl-8-oxo-8, 9-dihydro-7 h-purin-9-yl) -1-methylcyclohexane-1-carbonitrile was dissolved in 1, 4-dioxane (12mL), and 7-methyl- [1,2,4] was added thereto under stirring at room temperature]Triazolo [1,5-a]Pyridine-6-amine (0.11G,0.73mmol), cesium carbonate (0.24G,0.73mmol), Brettphos-G3-Pd (0.05G,0.04mmol), purged with nitrogen, and heated to 110 deg.C for 4h under reflux. TLC monitored to the end of the reaction, the reaction was concentrated and the residue was purified by silica gel column chromatography (dichloromethane/methanol (v/v): 30/1) to give title compound 7 (i.e., compound 7-1 and compound 7-2), two white solids, compound 7-1(19mg, 9.26% (structurally suspected) RT ═ 5.068min, DR%: 99.85%), compound 7-2(28mg, 13.64% (structurally suspected) RT ═ 5.900min, DR%: 99.38%). Pre-HPLC (OZ), mobile phase CO2(50% isopropanol/acetonitrile solution with 0.3% diethylamine) ═ 60/40; the column temperature is 35 ℃; column pressure 80 bar; the flow rate is 1 mL/min; a detector signal channel is 215nm @4.8 nm; the wavelength of the diode array detector is 200-400 nm.
Compound 7-11H NMR(400MHz,DMSO-d6)δ8.97(s,1H),8.73(s,1H),8.40(s,1H),8.11(s,1H),7.73(s,1H),4.19–4.10(m,1H),3.29(s,3H),2.32(s,3H),2.29–2.20(m,2H),1.90–1.75(m,4H),1.58–1.50(m,2H),0.84(s,3H)。
LC-MS m/z(ESI)=418.20[M+1]。
Compound 7-21H NMR(400MHz,DMSO-d6)δ9.06(s,1H),8.54(s,1H),8.34(s,1H),8.09(s,1H),7.67(s,1H),4.22–4.13(m,1H),3.30(s,3H),2.47–2.41(m,2H),2.36(s,3H),2.03–1.95(m,2H),1.80–1.73(m,2H),1.55–1.45(m,2H),1.33(s,3H)。
LC-MS m/z(ESI)=418.20[M+1]。
Example 8
Trans-4- (7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) cyclohexane-1-carbonitrile (Compound 8)
trans-4-(7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-8-oxo-7,8-dihydro-9H-purin-9-yl)cyclohexane-1-carbonitrile
Figure BDA0002876667980000241
Figure BDA0002876667980000251
The first step is as follows:
tert-butyl (trans-4-carbamoylcyclohexyl) carbamate (8b)
tert-butyl(trans-4-carbamoylcyclohexyl)carbamate
Trans-4- ((tert-butoxycarbonyl) amino) cyclohexane-1-carboxylic acid 8a (5.0g,20.5mmol), O- (7-azabenzotriazolyl) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (9.4g,24.7mmol) were dissolved in dichloromethane (15mL), stirred at 0 ℃ for 20min, N, N diisopropylethylamine (10.5g,82mmol) and ammonium chloride (3.3g,61.5mmol) were added, and stirred at room temperature for 4 h. TLC monitored to the end of the reaction, 10mL of water was added to the reaction, the organic phase was separated, washed once with saturated brine, dried over anhydrous sodium sulfate and stirred with silica gel, the product was passed through a normal phase column chromatography to obtain the title compound tert-butyl (trans-4-carbamoylcyclohexyl) carbamate 8b (white solid, 4.4g, 83% yield).
LC-MS m/z(ESI)=243.30[M+1]。
The second step is that:
tert-butyl (trans-4-cyanocyclohexyl) carbamate (8c)
tert-butyl(trans-4-cyanocyclohexyl)carbamate
Tert-butyl (trans-4-carbamoylcyclohexyl) carbamate 8b (4.4g,18.0mmol) was dissolved in 70mL pyridine, cooled in an ice bath, and then phosphorus oxychloride (7.7mL) was added dropwise to the reaction mixture, and stirred in an ice bath for 1 h. TLC monitored the reaction was complete, added 20mL of water under ice bath, extracted 4 times with ethyl acetate, then the organic phase was washed 7 times with acid water, finally washed twice with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated to dryness to give the compound tert-butyl (trans-4-cyanocyclohexyl) carbamate 8c (yellow solid, 1.2g, 30% yield).
1H NMR(400MHz DMSO)δ6.80(m,1H),3.24-3.22(m,1H),2.63-2.55(m,1H),1.99-1.95(m,2H),1.77-1.73(m,2H),1.56-1.46(m,2H),1.36(s,9H),1.21-1.11(m,2H)。
LC-MS m/z(ESI)=225.30[M+1]。
The third step:
trans-4-aminocyclohexane-1-carbonitrile (8d)
trans-4-aminocyclohexane-1-carbonitrile
Tert-butyl (trans-4-cyanocyclohexyl) carbamate 8c (1.2g, 9.6mmol) was dissolved in ethyl acetate hydrochloride solution (20mL), heated to 45 ℃ and stirred for 2 h. TLC monitored the reaction to completion and a white solid precipitated which was concentrated and dried to give the title compound trans-4-aminocyclohexane-1-carbonitrile 8d (white solid, 660mg, 60% yield).
LC-MS m/z(ESI)=125.30[M+1]。
The fourth step:
2-chloro-4- (trans-4-cyanocyclohexyl) amino) pyrimidine-5-carboxylic acid ethyl ester (8e)
ethyl 2-chloro-4-(trans-4-cyanocyclohexyl)amino)pyrimidine-5-carboxylate
Ethyl 2, 4-dichloropyrimidine-5-carboxylate 1a (1.4g,5.35mmol) and potassium carbonate (1.4g,10.7mmol) were dissolved in acetonitrile (10mL), and trans-4-aminocyclohexane-1-carbonitrile 8d (660mg,5.35mmol) was added at 0 ℃ and stirred at room temperature for 20 h. TLC was monitored to the end of the reaction, and the reaction solution was extracted three times with water and ethyl acetate, washed once with saturated brine, dried over anhydrous sodium sulfate and purified by silica gel column chromatography (n-hexane/ethyl acetate (v/v) ═ 10:1) to give the title compound ethyl 2-chloro-4- (trans-4-cyanocyclohexyl) amino) pyrimidine-5-carboxylate 8e (white solid, 810mg, yield 62%).
1H NMR(400MHz DMSO)δ8.62(s,1H),8.27(d,1H),4.30(q,2H),4.04-3.96(m,1H),4.04-3.67(m,1H),2.76-2.70(m,1H),2.04-2.00(m,2H),1.96-1.92(m,2H),1.72-1.62(m,2H),1.47-1.37(m,2H),1.30(t,3H)。
LC-MS m/z(ESI)=310.20[M+1]。
The fifth step:
2-chloro-4- ((trans-4-cyanocyclohexyl) amino) pyrimidine-5-carboxylic acid (8f)
2-chloro-4-((trans-4-cyanocyclohexyl)amino)pyrimidine-5-carboxylic acide
Ethyl 2-chloro-4- ((trans-4-cyanocyclohexyl) amino) pyrimidine-5-carboxylate 8e (810mg,2.6mmol) was dissolved in tetrahydrofuran/water (4mL/4mL), lithium hydroxide (247mg,5.2mmol) was added, and the mixture was stirred at room temperature for 1 h. TLC to completion of the reaction, dried tetrahydrofuran was spun off, pH adjusted to 4-5 and a white solid precipitated, filtered, the filter cake was washed twice with petroleum ether/ethyl acetate (v/v ═ 10/1) and concentrated to give the title compound 2-chloro-4- ((trans-4-cyanocyclohexyl) amino) pyrimidine-5-carboxylic acid 8f (white solid, 790mg, 86% yield)
1H NMR(400MHz DMSO)δ8.58(s,1H),8.49(d,1H),4.01-3.87(m,1H),2.76-2.71(m,1H),2.03-1.93(m,4H),1.72-1.63(m,2H),1.44-1.35(m,2H)。
LC-MS m/z(ESI)=282.30[M+1]。
And a sixth step:
trans-4- (2-chloro-8-oxo-7, 8-dihydro-9H-purin-9-yl) cyclohexane-1-carbonitrile (8g)
trans-4-(2-chloro-8-oxo-7,8-dihydro-9H-purin-9-yl)cyclohexane-1-carbonitrile
2-chloro-4- (trans-4-cyanocyclohexyl) amino) pyrimidine-5-carboxylic acid 8f (730mg, 2.6mmol) was dissolved in dimethylacetamide (10mL), triethylamine (263mg,2.6mmol) and azido diphenyl phosphate (715mg,2.6mmol) were added, followed by gradual temperature rise to 110 ℃ and stirring for 1.5 h. TLC to the end of the reaction, the reaction was concentrated and the residue was chromatographed on silica gel column (petroleum ether/ethyl acetate (v/v) ═ 1:10) to give the title compound trans-4- (2-chloro-8-oxo-7, 8-dihydro-9H-purin-9-yl) cyclohexane-1-carbonitrile 8g (white solid, 553mg, 68% yield).
1H NMR(400MHz DMSO)δ11.63(s,1H),8.12(s,1H),4.23-4.16(m,1H),2.78-2.72(m,1H),2.25-2.12(m,4H),1.82-1.68(m,4H)。
LC-MS m/z(ESI)=278.30[M+1]。
The seventh step:
trans-4- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) cyclohexane-1-carbanitrile (8H)
trans-4-(2-chloro-7-methyl-8-oxo-7,8-dihydro-9H-purin-9-yl)cyclohexane-1-carbonitrile
8g (553mg, 2mmol) of trans-4- (2-chloro-8-oxo-7, 8-dihydro-9H-purin-9-yl) cyclohexane-1-carbonitrile was dissolved in dimethylformamide (5mL), dimethyl sulfate (252mg, 2mmol) and cesium carbonate (977mg,3mmol) were added at 0 deg.C, and stirring was carried out at 0 deg.C for 30 min. TLC monitored the reaction to completion, 10mL of water was added to the reaction mixture, a solid precipitated, and filtered to give the title compound trans-4- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) cyclohexane-1-carbonitrile 8H (yellow solid, 420mg, 72% yield).
1H NMR(400MHz DMSO)δ8.34(s,1H),4.28-4.21(m,1H),2.79-2.72(m,1H),2.22-2.13(m,4H),1.82-1.70(m,4H)。
LC-MS m/z(ESI)=292.30[M+1]。
Eighth step:
trans-4- (7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) cyclohexane-1-carbonitrile (Compound 8)
trans-4-(7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-8-oxo-7,8-dihydro-9H-purin-9-yl)cyclohexane-1-carbonitrile
Trans-4- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) cyclohexane-1-carbonitrile 8H (200mg, 0.68mmol), 7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-amine (101mg, 0.68mmol), cesium carbonate (391mg,1.2mmol), methanesulfonic acid (2-dicyclohexylphosphine-3, 6-dimethoxy-2 ',4',6 '-triisopropyl-1, 1' -biphenyl) (2 '-amino-1, 1' -biphenyl-2-yl) palladium (II) (62mg, 0.068mmol) was dissolved in dioxane 3mL, protected with nitrogen and purged, after stirring at 110 ℃ for 4H, the reaction was monitored by TLC for completion, the reaction solution was concentrated, and the residue was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 60/1), and by Pre-HPLC, the title compound, trans-4- (7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) cyclohexane-1-carbonitrile compound 8 (white solid, 57mg, 20.2% yield) was obtained.
1H NMR(400MHz DMSO)δ9.22(s,1H),8.65(s,1H),8.38(s,1H),8.11(s,1H),7.72(s,1H),4.25-4.19(m,1H),3.30(s,3H),2.67-2.61(m,1H),2.41(s,3H),2.32-2.24(m,2H),2.15-2.12(m,2H),1.82-1.79(m,2H),1.73-1.64(m,2H)。
LC-MS m/z(ESI)=404.20[M+1]。
Example 9
Cis-4- (7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) cyclohexane-1-carbonitrile (Compound 9)
cis-4-(7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-8-oxo-7,8-dihydro-9H-purin-9-yl)cyclohexane-1-carbonitrile
Figure BDA0002876667980000271
Figure BDA0002876667980000281
The first step is as follows:
tert-butyl (cis-4-carbamoylcyclohexyl) carbamate (9b)
tert-butyl(cis-4-carbamoylcyclohexyl)carbamate
Cis-4- ((tert-butoxycarbonyl) amino) cyclohexane-1-carboxylic acid 9a (5.0g,20.5mmol), O- (7-azabenzotriazolyl) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (9.4g,24.7mmol) were dissolved in dichloromethane (15mL), stirred at 0 ℃ for 20min, N, N diisopropylethylamine (10.5g,82mmol) and ammonium chloride (3.3g,61.5mmol) were added, and stirred at room temperature for 4 h. TLC monitored to the end of the reaction, 10mL of water was added to the reaction mixture, the organic phase was separated, washed once with saturated brine, dried over anhydrous sodium sulfate and stirred with silica gel, the product was passed through a normal phase column chromatography to obtain the title compound tert-butyl (cis-4-carbamoylcyclohexyl) carbamate 9b (white solid, 3.5g, 71% yield).
LC-MS m/z(ESI)=243.30[M+1]。
The second step is that:
tert-butyl (cis-4-cyanocyclohexyl) carbamate (9c)
tert-butyl(cis-4-cyanocyclohexyl)carbamate
Tert-butyl (cis-4-carbamoylcyclohexyl) carbamate 9b (3.5g,14.4mmol) was dissolved in 70mL of pyridine, cooled in an ice bath, and then phosphorus oxychloride (7.7mL) was added dropwise to the reaction mixture, and stirred in an ice bath for 1 h. TLC monitored the reaction was complete, added 20mL of water under ice bath, extracted 4 times with ethyl acetate, then the organic phase was washed 7 times with acid water, finally washed twice with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated to dryness to give the compound tert-butyl (cis-4-cyanocyclohexyl) carbamate 9c (yellow solid, 1.5g, 36% yield).
1H NMR(400MHz DMSO)δ6.90(d,1H),3.27-3.25(m,1H),3.01-2.99(m,1H),1.86-1.81(m,2H),1.71-1.67(m,2H),1.62-1.54(m,2H),1.43-1.40(m,1H),1.37(s,3H),1.34-1.32(m,1H)。
LC-MS m/z(ESI)=225.30[M+1]。
The third step:
cis-4-aminocyclohexane-1-carbonitrile (9d)
cis-4-aminocyclohexane-1-carbonitrile
Tert-butyl (trans-4-cyanocyclohexyl) carbamate 9c (1.5g, 6.7mmol) was dissolved in ethyl acetate hydrochloride solution (20mL), heated to 45 ℃ and stirred for 2 h. TLC monitored the reaction to completion, a white solid precipitated, which was concentrated and dried to give the title compound cis-4-aminocyclohexane-1-carbonitrile 9d (white solid, 1g, 90% yield).
LC-MS m/z(ESI)=125.30[M+1]。
The fourth step:
2-chloro-4- (cis-4-cyanocyclohexyl) amino) pyrimidine-5-carboxylic acid ethyl ester (9e)
ethyl 2-chloro-4-(cis-4-cyanocyclohexyl)amino)pyrimidine-5-carboxylate
Ethyl 2, 4-dichloropyrimidine-5-carboxylate 1a (1.48g,6.7mmol) and potassium carbonate (1.38g,10mmol) were dissolved in acetonitrile (10mL), and cis-4-aminocyclohexane-1-carbonitrile 9d (1g,6.7mmol) was added at 0 ℃ and stirred at room temperature for 20 h. TLC was monitored to the end of the reaction, and the reaction solution was extracted three times with water and ethyl acetate, washed once with saturated brine, dried over anhydrous sodium sulfate and purified by silica gel column chromatography (n-hexane/ethyl acetate (v/v) ═ 10:1) to give the title compound ethyl 2-chloro-4- (cis-4-cyanocyclohexyl) amino) pyrimidine-5-carboxylate 9e (white solid, 589mg, yield 60%).
1H NMR(400MHz DMSO)δ8.63(s,1H),8.38(d,1H),4.35-4.29(m,2H),4.07-3.99(m,1H),3.11-3.09(m,1H),1.92-1.73(m,6H),1,63-1.54(m,2H),1.47(t,3H)。
LC-MS m/z(ESI)=310.20[M+1]。
The fifth step:
2-chloro-4- ((cis-4-cyanocyclohexyl) amino) pyrimidine-5-carboxylic acid (9f)
2-chloro-4-((cis-4-cyanocyclohexyl)amino)pyrimidine-5-carboxylic acide
Ethyl 2-chloro-4- ((cis-4-cyanocyclohexyl) amino) pyrimidine-5-carboxylate 9e (589mg,1.9mmol) was dissolved in tetrahydrofuran/water (4mL/4mL), lithium hydroxide (160mg,3.8mmol) was added, and the mixture was stirred at room temperature for 1 h. TLC monitored to the end of the reaction, dried tetrahydrofuran was spun down, adjusted to pH 4-5 and a white solid precipitated, filtered, and the filter cake was washed twice with petroleum ether/ethyl acetate (v/v ═ 10/1) and concentrated to give the title compound 2-chloro-4- ((cis-4-cyanocyclohexyl) amino) pyrimidine-5-carboxylic acid 9f (white solid, 490mg, 86% yield).
1H NMR(400MHz DMSO)δ13.83(s,1H),8.62-8.60(d,2H),4.05-3.98(m,1H),3.10(t,1H),1.94-1.73(m,6H),1.60-1.56(m,2H)。
LC-MS m/z(ESI)=282.30[M+1]。
And a sixth step:
cis-4- (2-chloro-8-oxo-7, 8-dihydro-9H-purin-9-yl) cyclohexane-1-carbonitril (9g)
cis-4-(2-chloro-8-oxo-7,8-dihydro-9H-purin-9-yl)cyclohexane-1-carbonitrile
2-chloro-4- (cis-4-cyanocyclohexyl) amino) pyrimidine-5-carboxylic acid 9f (490mg, 1.75mmol) was dissolved in dimethylacetamide (10mL), triethylamine (176mg,1.75mmol) and diphenylphosphoryl azide (481mg,1.75mmol) were added, followed by gradual warming to 110 ℃ and stirring for 1.5 h. TLC to the end of the reaction, the reaction was concentrated and the residue was chromatographed on silica gel column (petroleum ether/ethyl acetate (v/v) ═ 1:10) to give the title compound cis-4- (2-chloro-8-oxo-7, 8-dihydro-9H-purin-9-yl) cyclohexane-1-carbonitrile 9g (white solid, 436mg, 68% yield).
1H NMR(400MHz DMSO)δ11.64(s,1H),8.13(s,1H),4.23-4.16(m,1H),3.22-3.16(m,1H),2.44-2.39(m,2H),2.00(d,2H),1.79-1.72(m,4H)。
LC-MS m/z(ESI)=278.30[M+1]。
The seventh step:
cis-4- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) cyclohexane-1-carbanion (9H)
cis-4-(2-chloro-7-methyl-8-oxo-7,8-dihydro-9H-purin-9-yl)cyclohexane-1-carbonitrile
Cis-4- (2-chloro-8-oxo-7, 8-dihydro-9H-purin-9-yl) cyclohexane-1-carbonitrile 9g (1.6g, 5.77mmol) was dissolved in dimethylformamide (10mL), dimethyl sulfate (728mg, 5.77mmol) and cesium carbonate (2.8g,8.65mmol) were added at 0 deg.C, and stirring was carried out at 0 deg.C for 30 min. TLC monitored the reaction to completion, 10mL of water was added to the reaction mixture, a solid precipitated, and filtered to give the title compound cis-4- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) cyclohexane-1-carbonitrile 9H (yellow solid, 1.25g, 75% yield).
1H NMR(400MHz DMSO)δ8.35(s,1H),4.28-4.19(m,1H),3.34(s,3H),3.23-3.17(m,1H),2.46-2.39(m,2H),2.00(d,2H),1.80-1.73(m,4H)。LC-MS m/z(ESI)=292.30[M+1]The seventh step:
cis-4- (7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) cyclohexane-1-carbonitrile (Compound 9)
cis-4-(7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-8-oxo-7,8-dihydro-9H-purin-9-yl)cyclohexane-1-carbonitrile
Cis-4- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) cyclohexane-1-carbonitrile 9H (200mg, 0.68mmol), 7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-amine (101mg, 0.68mmol), cesium carbonate (391mg,1.2mmol), methanesulfonic acid (2-dicyclohexylphosphine-3, 6-dimethoxy-2 ',4',6 '-triisopropyl-1, 1' -biphenyl) (2 '-amino-1, 1' -biphenyl-2-yl) palladium (II) (62mg, 0.068mmol) was dissolved in dioxane 3mL, protected with nitrogen and purged, stirred at 110 ℃ for 4 h. The reaction solution was concentrated, and the residue was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 60/1), and the title compound, cis-4- (7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) cyclohexane-1-carbonitrile compound 9 (white solid, 131mg, yield 41.2%), was obtained by Pre-HPLC.
1H NMR(400MHz DMSO)δ9.08(s,1H),8.57(s,1H),8.34(s,1H),8.09(s,1H),7.67(s,1H),4.23-4.15(m,1H),3.30(s,3H),3.14(s,1H),2.51-2.49(m,2H),2.36(s,3H),1.98(d,2H),1.76(d,2H),1.71-1.66(m,2H)。
LC-MS m/z(ESI)=404.20[M+1]。
Example 10
7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -9- (1-methylpiperidin-4-yl) -7, 9-dihydro-8H-purin-8-one (Compound 10)
7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-9-(1-methylpiperidin-4-yl)-7,9-dihydro-8H-purin-8-one
Figure BDA0002876667980000301
The first step is as follows:
4- ((1- (tert-Butoxycarbonyl) piperidin-4-yl) amino) -2-chloropyrimidine-5-carboxylic acid ethyl ester (10b)
ethyl 4-((1-(tert-butoxycarbonyl)piperidin-4-yl)amino)-2-chloropyrimidine-5-carboxylate
Ethyl 2, 4-dichloropyrimidine-5-carboxylate 1a (5g,22.6mmol) and potassium carbonate (6.2g,45.2mmol) were dissolved in acetonitrile (20mL), and (4-aminopiperidine-1-carboxylic acid tert-butyl ester 10a (4.5g,22.6mmol) was added at 0 ℃, stirred at room temperature for 20h, TLC monitored for completion, 30mL of water was added, extracted three times with ethyl acetate 60mL, washed once with saturated brine, dried over anhydrous sodium sulfate, and purified by silica gel column chromatography (n-hexane: ethyl acetate ═ 10:1) to give the title compound, ethyl 4- ((1- (tert-butoxycarbonyl) piperidin-4-yl) amino) -2-chloropyrimidine-5-carboxylate 10b (white solid, 8.2g, 95% yield).
1H NMR(400MHz DMSO)δ8.63(s,1H),8.32(d,1H),4.33-4.28(m,2H),4.17-4.14(m,1H),3.85(d,2H),2.94(s,2H),1.88-1.80(m,2H),1.51-1.44(m,2H),1.41(s,9H),1.32-1.29(m,3H)。
LC-MS m/z(ESI)=385.10[M+1]。
The second step is that:
4- ((1- (tert-Butoxycarbonyl) piperidin-4-yl) amino) -2-chloropyrimidine-5-carboxylic acid (10c)
4-((1-(tert-butoxycarbonyl)piperidin-4-yl)amino)-2-chloropyrimidine-5-carboxylic acid
Ethyl 4- ((1- (tert-butoxycarbonyl) piperidin-4-yl) amino) -2-chloropyrimidine-5-carboxylate 10b (8.2g, 21.3mmol) was dissolved in 10mL of tetrahydrofuran and 5mL of water, lithium hydroxide (1.8g,42.7mmol) was added, the mixture was stirred at room temperature for 1h, TLC monitored to completion, tetrahydrofuran was spun dry, pH was adjusted to 4-5, a white solid was precipitated, filtered, and the cake was washed twice with petroleum ether/ethyl acetate (v/v ═ 10/1), and concentrated to give the title compound, 4- ((1- (tert-butoxycarbonyl) piperidin-4-yl) amino) -2-chloropyrimidine-5-carboxylic acid 10c (white solid, 7g, yield 86%).
1H NMR(400MHz DMSO)δ8.59(s,1H),8.54(d,1H),4.20-4.10(m,1H),3.87-3.84(m,2H),2.96(s,3H),1.91-1.73(m,3H),1.41(s,9H)。
LC-MS m/z(ESI)=357.10[M+1]。
The third step
4- (2-chloro-8-oxo-7, 8-dihydro-9H-purin-9-yl) piperidine-1-carboxylic acid tert-butyl ester (10d)
tert-butyl 4-(2-chloro-8-oxo-7,8-dihydro-9H-purin-9-yl)piperidine-1-carboxylate
4- ((1- (tert-Butoxycarbonyl) piperidin-4-yl) amino) -2-chloropyrimidine-5-carboxylic acid 10c (7g, 19.6mmol) was dissolved in dimethylacetamide (10mL), triethylamine (1.96g,19.6mmol) was added, diphenyl phosphorazidate (5.4g,19.6mmol) was then gradually warmed to 110 ℃, stirred for 1.5H, TLC monitored for completion of the reaction, concentrated the reaction solution, and purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 5:1 to 1:10) to give the title compound, tert-butyl 4- (2-chloro-8-oxo-7, 8-dihydro-9H-purin-9-yl) piperidine-1-carboxylate 10d (white solid, 6.4g, 87% yield).
1H NMR(400MHz DMSO)δ8.14(s,1H),4.41-4.33(m,1H),4.06(d,2H),2.88(s,2H),2.30-2.20(m,2H),1.84-1.71(m,2H),1.43(s,9H)。
LC-MS m/z(ESI)=354.10[M+1]。
The fourth step
4- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) piperidine-1-carboxylic acid tert-butyl ester (10e)
tert-butyl4-(2-chloro-7-methyl-8-oxo-7,8-dihydro-9H-purin-9-yl)piperidine-1-carboxylate
Tert-butyl 4- (2-chloro-8-oxo-7, 8-dihydro-9H-purin-9-yl) piperidine-1-carboxylate 10d (6.4g, 18.1mmol) was dissolved in dimethylformamide (10mL), dimethyl sulfate (2.28g, 18.1mmol) and cesium carbonate (8.5g,27.1mmol) were added at 0 deg.C, stirred at 0 deg.C for 30min, TLC monitored to completion, followed by addition of 30mL water with solid precipitation, and filtered to give the title compound, tert-butyl 4- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) piperidine-1-carboxylate 10e (white solid, 5.4g, 79% yield).
1H NMR(400MHz DMSO)δ8.36(s,1H),4.46-4.37(m,1H),4.05(d,2H),3.35(s,3H),2.87(s,2H),2.33-2.19(m,2H),1.74-1.72(m,2H),1.43(s,9H)。
LC-MS m/z(ESI)=368.10[M+1]。
The fifth step
2-chloro-7-methyl-9- (piperidin-4-yl) -7, 9-dihydro-8H-purin-8-one (10f)
2-chloro-7-methyl-9-(piperidin-4-yl)-7,9-dihydro-8H-purin-8-one
Tert-butyl 4- (2-chloro-8-oxo-7, 8-dihydro-9H-purin-9-yl) piperidine-1-carboxylate 10e (2g, 5.4mmol) was added to a 100mL one-necked flask, followed by addition of ethyl acetate hydrochloride solution 2M (10mL) at ambient temperature, stirring at ambient temperature for 4H, TLC monitoring to completion, and concentration of the reaction solution to give the title compound, hydrochloride 10f of 2-chloro-7-methyl-9- (piperidin-4-yl) -7, 9-dihydro-8H-purin-8-one (white solid, 1.4g, 91% yield).
1H NMR(400MHz DMSO)δ8.39(s,1H),6.42(s,1H),4.59-4.53(m,1H),3.39(s,2H),3.36(s,3H),3.16-3.04(m,2H),2.62-2.50(m,2H),2.07-1.93(m,2H)。
LC-MS m/z(ESI)=268.10[M+1]。
And a sixth step:
2-chloro-7-methyl-9- (1-methylpiperidin-4-yl) -7, 9-dihydro-8H-purin-8-one (10g)
2-chloro-7-methyl-9-(1-methylpiperidin-4-yl)-7,9-dihydro-8H-purin-8-one
Dissolve 2-chloro-7-methyl-9- (piperidin-4-yl) -7, 9-dihydro-8H-purin-8-one 10f (1.4g,5.4mmol) in methanol (20mL), add 100mg of 4A molecular sieve, then add paraformaldehyde (783mg,27mmol), stir at room temperature for 6H, then add sodium cyanoborohydride (1g,16.2mmol), monitor by TLC to completion, filter and concentrate to give the title compound, 2-chloro-7-methyl-9- (1-methylpiperidin-4-yl) -7, 9-dihydro-8H-purin-8-one 10g (white solid, 600mg, 62% yield).
1H NMR(400MHz DMSO)δ8.35(s,1H),4.21-4.13(m,1H),3.35(s,3H),2.92(d,2H),2.45-2.41(m,2H),2.23(s,3H),2.09-2.03(m,2H),1.70-1.67(m,2H)。
LC-MS m/z(ESI)=282.10[M+1]。
The seventh step:
7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -9- (1-methylpiperidin-4-yl) -7, 9-dihydro-8H-purin-8-one (Compound 10)
7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-9-(1-methylpiperidin-4-yl)-7,9-dihydro-8H-purin-8-one
10g of 2-chloro-7-methyl-9- (1-methylpiperidin-4-yl) -7, 9-dihydro-8H-purin-8-one (150mg, 0.53mmol), 7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-amine (79mg, 0.53mmol), cesium carbonate (518mg,1.59mmol), methanesulfonic acid (2-dicyclohexylphosphine-3, 6-dimethoxy-2 ',4',6 '-triisopropyl-1, 1' -biphenyl)) 2 '-amino-1, 1' -biphenyl-2-yl) palladium (II) (48mg, 0.053mmol) was dissolved in dioxane (3mL), purged with nitrogen and stirred at 100 ℃ for 4 h. TLC was monitored to completion, the reaction solution was concentrated, and the residue was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 20/1), and subjected to Pre-HPLC to give the title compound, 7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -9- (1-methylpiperidin-4-yl) -7, 9-dihydro-8H-purin-8-one compound 10 (white solid, 20mg, 18% yield).
1H NMR(400MHz DMSO)δ9.23(s,1H),8.33(s,1H),8.29(s,1H),8.12(s,1H),7.69(s,1H),4.53-4.33(m,1H),3.50(d,2H),2.86-2.77(m,4H),2.43(s,3H),1.99(d,2H)。
LC-MS m/z(ESI)=394.20[M+1]。
Example 11
7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -9- (piperidin-4-yl) -7, 9-dihydro-8H-purin-8-one (Compound 11)
7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-9-(piperidin-4-yl)-7,9-dihydro-8H-purin-8-one
Figure BDA0002876667980000321
Figure BDA0002876667980000331
The first step is as follows:
tert-butyl 4- (7-methyl-2- (((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -8-oxo-7, 8-dihydro-
9H-purin-9-yl) piperidine-1-carboxylic acid (11a)
tert-butyl4-(7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-8-oxo-7,8-dihydro-9H-purin-9-yl)piperidine-1-carboxylate
Tert-butyl 4- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) piperidine-1-carboxylate 10e (200mg, 0.54mmol), 7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-amine (148mg, 0.54mmol), cesium carbonate (528mg,1.62mmol), methanesulfonic acid (2-dicyclohexylphosphine-3, 6-dimethoxy-2 ',4',6 '-triisopropyl-1, 1' -biphenyl)) 2 '-amino-1, 1' -biphenyl-2-yl palladium (II) (50mg, 0.054mmol) was dissolved in dioxane (3mL), nitrogen protected and purged, stirring was carried out at 100 ℃ for 4 h. TLC was monitored to completion, the reaction was concentrated, and the residue was purified by silica gel column chromatography) dichloromethane/methanol (v/v) ═ 60/1) to give the title compound, tert-butyl 4- (7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) piperidine-1-carboxylic acid 11a (white solid, 120mg, 62% yield).
1H NMR(400MHz DMSO)δ9.08(s,1H),8.57(s,1H),8.36(s,1H),8.08(s,1H),7.69-7.68(m,1H),4.38-4.30(m,1H),4.06-4.10(m,2H),3.29(s,3H),2.81(s,2H),2.40(s,3H),2.37-2.31(m,2H),1.75-1.72(m,2H),1.39(s,9H)。
LC-MS m/z(ESI)=480.20[M+1]。
The second step is that:
7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -9- (piperidin-4-yl) -7, 9-dihydro-8H-purin-8-one (Compound 11)
7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-9-(piperidin-4-yl)-7,9-dihydro-8H-purin-8-one
Tert-butyl 4- (7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) piperidine-1-carboxylic acid 11a (120mg, 0.25mmol) was added to a 50mL one-necked flask, and 2M ethyl acetate hydrochloride (10mL) was added thereto, and stirred at ordinary temperature for 4 hours. TLC was monitored to completion, the reaction was concentrated, and the residue was subjected to Pre-HPLC to give the title compound, 7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -9- (piperidin-4-yl) -7, 9-dihydro-8H-purin-8-one compound 11 (white solid, 32mg, 23% yield).
1H NMR(400MHz DMSO)δ9.20(s,1H),8.62(s,1H),8.37(s,1H),8.09(s,1H),7.70(s,1H),4.30-4.22(m,1H),3.30(s,3H),3.10-3.07(m,2H),2.67-2.58(m,2H),2.41(s,3H),2.38-2.32(m,2H),1.69-1.65(m,2H)。
LC-MS m/z(ESI)=380.20[M+1]。
Example 12
9- (trans-3-Hydroxycyclobutyl) -7-methyl-2- (((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -7, 9-dihydro-8H-purin-8-one (Compound 12)
9-(trans-3-hydroxycyclobutyl)-7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-7,9-dihydro-8H-purin-8-one
Figure BDA0002876667980000341
The first step is as follows:
2-chloro-4- ((trans-3-hydroxycyclobutyl) amino) pyrimidine-5-carboxylic acid ethyl ester (12a)
ethyl 2-chloro-4-((trans-3-hydroxycyclobutyl)amino)pyrimidine-5-carboxylate
(trans-3-aminocyclobutane-1-ol hydrochloride (3.00g,34.48mmol) was dissolved in acetonitrile (25mL), potassium carbonate (14.30g,103.45mmol), ethyl 2, 4-dichloro-5-pyrimidinecarboxylate 1a (11.43g,51.72mmol) were added with stirring at 0 deg.C, the reaction was stirred at room temperature for 1h, TLC was performed until the reaction was completed, and after filtration through celite, purification by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 2:1) gave the title compound ethyl 2-chloro-4- ((trans-3-hydroxycyclobutyl) amino) pyrimidine-5-carboxylate (12a) (white solid, 2.9g, yield 30.95%).
1H NMR(400MHz,DMSO-d6)δ8.61(s,1H),8.48(d,1H),5.17-5.11(m,1H),4.61–4.51(m,1H),4.36–4.27(m,3H),2.32-2.20(m,4H),1.32(t,3H)。
LC-MS m/z(ESI)=272.00[M+1]。
The second step is that:
4- ((trans-3- ((tert-butyldiphenylsilyl) oxy) cyclobutyl) amino) -2-chloropyrimidine-5-carboxylic acid ethyl ester (12b)
ethyl 4-((trans-3-((tert-butyldiphenylsilyl)oxy)cyclobutyl)amino)-2-chloropyrimidine-5-carboxylate
Ethyl 2-chloro-4- ((trans-3-hydroxycyclobutyl) amino) pyrimidine-5-carboxylate 12a (3.10g,11.41mmol) was dissolved in dichloromethane (20mL), tert-butyldiphenylchlorosilane (4.45mL,17.11mmol), imidazole (1.94g,28.52mmol) were added, the reaction was stirred at room temperature for 2h under nitrogen protection and with aeration, TLC monitored to the end of the reaction, and purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 10:1) to give the title compound ethyl 4- (trans-3- ((tert-butyldiphenylsilyl) oxy) cyclobutyl) amino) -2-chloropyrimidine-5-carboxylate (12b) (colorless liquid, 4.18g, yield 64.26%).
LC-MS m/z(ESI)=510.20[M+1]。
The third step:
4- ((trans-3- ((tert-butyldiphenylsilyl) oxy) cyclobutyl) amino) -2-chloropyrimidine-5-carboxylic acid (12c)
4-((trans-3-((tert-butyldiphenylsilyl)oxy)cyclobutyl)amino)-2-chloropyrimidine-5-carboxylic acid
Ethyl 4- ((trans-3- ((tert-butyldiphenylsilyl) oxy) cyclobutyl) amino) -2-chloropyrimidine-5-carboxylate 12b (4.18g,8.19mmol) was dissolved in tetrahydrofuran/water (30mL/15mL), lithium hydroxide monohydrate (1.03g,24.58mmol) was added, the reaction was stirred at ordinary temperature for 2 hours, TLC was monitored to end the reaction, after evaporation of tetrahydrofuran by concentration, 2N HCl was added to adjust pH to 3-4, water and ethyl acetate were added to extract twice, the organic layer was concentrated and purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 20:1) to obtain the title compound 4- ((trans-3- ((tert-butyldiphenylsilyl) oxy) cyclobutyl) amino) -2-chloropyrimidine-5-carboxylate (12c) (white solid, 3.87g, 97.97% yield).
LC-MS m/z(ESI)=482.20[M+1]。
The fourth step:
9- (trans-3- ((tert-butyldiphenylsilyl) oxy) cyclobutyl) -2-chloro-7, 9-dihydro-8H-purin-8-one (12d)
9-(trans-3-((tert-butyldiphenylsilyl)oxy)cyclobutyl)-2-chloro-7,9-dihydro-8H-purin-8-one
4- (trans-3- ((tert-butyldiphenylsilyl) oxy) cyclobutyl) amino) -2-chloropyrimidine-5-carboxylic acid 12c (3.87g,8.03mmol) was dissolved in N, N-dimethylacetamide (38mL), and triethylamine (1.11mL,8.03mmol) and diphenylphosphorylazide (1.73mL,8.03mmol) were added under stirring at ordinary temperature to react for 2 hours, followed by heating to 110 ℃ and refluxing for 2.5 hours. TLC was monitored to completion of the reaction, water and ethyl acetate were added to the reaction solution to extract, and the concentrated organic layer was purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 1:1) to give the title compound 9- (trans-3- ((tert-butyldiphenylsilyl) oxy) cyclobutyl) -2-chloro-7, 9-dihydro-8H-purin-8-one (12d) (white solid, 1.81g, 47.06% yield).
1H NMR(400MHz,DMSO-d6)δ11.59(s,1H),8.09(s,1H),7.65–7.62(m,2H),7.62–7.60(m,2H),7.47-7.39(m,6H),5.05–4.97(m,1H),4.96–4.89(m,1H),2.98-2.84(m,2H),2.54-2.46(m,2H),1.02(s,9H)。
LC-MS m/z(ESI)=479.20[M+1]。
The fifth step:
9- (trans-3- ((tert-butyldiphenylsilyl) oxy) cyclobutyl) -2-chloro-7-methyl-7, 9-dihydro-8H-purin-8-one (12e)
9-(trans-3-((tert-butyldiphenylsilyl)oxy)cyclobutyl)-2-chloro-7-methyl-7,9-dihydro-8H-purin-8-one
9- (trans-3- ((tert-butyldiphenylsilyl) oxy) cyclobutyl) -2-chloro-7, 9-dihydro-8H-purin-8-one 12d (1.81g,3.78mmol) was dissolved in N, N-dimethylformamide (20mL) and dimethyl sulfate (0.36mL,3.78mmol) and cesium carbonate (2.47g,7.57mmol) were added with stirring at 0 ℃ for 2H. TLC was monitored to completion of the reaction, the reaction solution was slowly added dropwise to ice water and stirred, ethyl acetate was added to extract, and the concentrated organic layer was purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 3:1) to give the title compound 9- (trans-3- ((tert-butyldiphenylsilyl) oxy) cyclobutyl) -2-chloro-7-methyl-7, 9-dihydro-8H-purin-8-one (12e) (white solid, 1.80g, 96.62% yield).
LC-MS m/z(ESI)=493.20[M+1]。
And a sixth step:
9- (trans-3- ((tert-butyldiphenylsilyl) oxy) cyclobutyl) -7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -7, 9-dihydro-8H-purin-8-one (12f)
9-(trans-3-((tert-butyldiphenylsilyl)oxy)cyclobutyl)-7-methyl-2-((7-methyl-
[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-7,9-dihydro-8H-purin-8-one
Reacting 9- (trans-3- ((tert-butyldiphenylsilyl) oxy) cyclobutyl) -2-chloro-7-methyl-7, 9-dihydro-8H-purin-8-one 12e (200mg, 0.41mmol), 7-methyl- [1,2, 4%]Triazolo [1,5-a]Pyridin-6-amine (123mg, 0.82mmol), cesium carbonate (264mg, 0.82mmol) and Brettphos G3 Pd (37mg, 0.041mmol) were added to a dry reaction flask, N2After three replacements, dry 1, 4-dioxane (1mL) was added and the reaction was allowed to proceed for 5h at 110 ℃. Cooled to room temperature, filtered and the filtrate concentrated to give the crude product (12f) which was taken to the next step without purification.
The seventh step:
9- (trans-3-Hydroxycyclobutyl) -7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -7, 9-dihydro-8H-purin-8-one (Compound 12)
9-(trans-3-hydroxycyclobutyl)-7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-7,9-dihydro-8H-purin-8-one
The crude product 12f obtained in the sixth step was dissolved in 1, 4-dioxane (5mL), followed by dropwise addition of concentrated hydrochloric acid (1mL) and reaction at room temperature for 2 hours. TLC monitored to the end of the reaction, adjusted pH to around 7 with saturated sodium bicarbonate solution, extracted three times with dichloromethane, combined organic phases, concentrated and purified using silica gel column chromatography (dichloromethane/methanol (v/v ═ 40:1)) to give the title compound 9- ((trans-3-hydroxycyclobutyl) -7-methyl-2- ((6-methyl-2, 3-dihydrobenzofuran-5-yl) amino) -7, 9-dihydro-8H-purin-8-one (compound 12) (white solid, 54mg, 36.23% yield).
1H NMR(400MHz,DMSO-d6)δ8.89(s,1H),8.24(s,1H),7.97(s,1H),7.22(s,1H),6.61(s,1H),,4.40–4.33(m,1H),3.25(s,3H),3.11(t,2H),3.02-2.92(m,2H),2.19-2.13(m,2H),2.12(s,3H)。
LC-MS m/z(ESI)=368.10[M+1]。
Example 13
9- (cis-3-Hydroxycyclobutyl) -7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -7, 9-dihydro-8H-purin-8-one (Compound 13)
9-(cis-3-hydroxycyclobutyl)-7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-7,9-dihydro-8H-purin-8-one
Figure BDA0002876667980000361
The first step is as follows:
2-chloro-4- ((cis-3-hydroxycyclobutyl) amino) pyrimidine-5-carboxylic acid ethyl ester (13a)
ethyl 2-chloro-4-((cis-3-hydroxycyclobutyl)amino)pyrimidine-5-carboxylate
(1s,3s) -3-aminocyclobutane-1-ol hydrochloride (3.00g,34.48mmol) was dissolved in acetonitrile (25mL), potassium carbonate (14.30g,103.45mmol), ethyl 2, 4-dichloro-5-pyrimidinecarboxylate 1a (11.43g,51.72mmol) were added with stirring at 0 ℃, the reaction was stirred at room temperature for 1h, TLC was monitored to the end of the reaction, and after filtration through celite, purification by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 2:1) gave title compound ethyl 2-chloro-4- ((cis-3-hydroxycyclobutyl) amino) pyrimidine-5-carboxylate (13a) (white solid, 3.3g, 32.91% yield).
1H NMR(400MHz,DMSO-d6)δ8.60(s,1H),8.41(d,1H),4.96(s,1H),4.31(q,2H),4.08–3.97(m,1H),3.94–3.84(m,1H),2.71-2.63(m,2H),1.88–1.80(m,2H),1.33–1.29(m,3H)。
LC-MS m/z(ESI)=272.00[M+1]。
The second step is that:
2-chloro-4- ((cis-3-hydroxycyclobutyl) amino) pyrimidine-5-carboxylic acid (13b)
2-chloro-4-((cis-3-hydroxycyclobutyl)amino)pyrimidine-5-carboxylate
Ethyl 2-chloro-4- ((1s,3s) -3-hydroxycyclobutyl) amino) pyrimidine-5-carboxylate 13a (3.30g,12.14mmol) was dissolved in tetrahydrofuran/water (30mL/15mL), lithium hydroxide monohydrate (1.53g,36.44mmol) was added, the reaction was stirred at room temperature for 2h, TLC monitored to end the reaction, the tetrahydrofuran was concentrated to evaporate, 2N HCl was added to adjust the pH to about 3-4, a white solid precipitated, filtered, and the cake was washed twice with water and petroleum ether/ethyl acetate (v/v ═ 10/1) to give the title compound 2-chloro-4- ((cis-3-hydroxycyclobutyl) amino) pyrimidine-5-carboxylic acid (13b) (white solid, 2.44g, yield 82.45%).
1H NMR(400MHz,DMSO-d6)δ8.61(d,1H),8.57(s,1H),4.43-4.20(m,1H),4.07–3.96(m,1H),3.93–3.83(m,1H),2.72-2.63(m,2H),1.87–1.77(m,2H)。
LC-MS m/z(ESI)=244.00[M+1]。
The third step:
2-chloro-9- (cis-3-hydroxycyclobutyl) -7, 9-dihydro-8H-purin-8-one (13c)
2-chloro-9-(cis-3-hydroxycyclobutyl)-7,9-dihydro-8H-purin-8-one
2-chloro-4- (((1s,3s) -3-hydroxycyclobutyl) amino) pyrimidine-5-carboxylic acid 13b (2.44g,10.01mmol) was dissolved in N, N-dimethylacetamide (20mL), triethylamine (1.39mL,10.01mmol) and diphenylphosphorylazide (2.16mL,10.01mmol) were added under stirring at room temperature for reaction for 2h, and the mixture was heated to 110 ℃ and refluxed for reaction for 2.5 h. TLC monitored the reaction was complete, water and dichloromethane were added to the reaction solution for extraction, and the organic layer was concentrated to afford the title compound 2-chloro-9- (cis-3-hydroxycyclobutyl) -7, 9-dihydro-8H-purin-8-one (13c) (white solid, 0.90g, 37.35% yield).
1H NMR(400MHz,DMSO-d6)δ11.59(s,1H),8.12(s,1H),4.31–4.20(m,1H),4.00-3.92(m,1H),3.80-3.54(m,1H),2.83–2.73(m,2H),2.58–2.51(m,2H)。
LC-MS m/z(ESI)=241.00[M+1]。
The fourth step:
2-chloro-9- (cis-3-hydroxycyclobutyl) -7-methyl-7, 9-dihydro-8H-purin-8-one (13d)
2-chloro-9-(cis-3-hydroxycyclobutyl)-7-methyl-7,9-dihydro-8H-purin-8-one
2-chloro-9- ((1s,3s) -3-hydroxycyclobutyl) -7, 9-dihydro-8H-purin-8-one 13c (0.70g,1.25mmol) was dissolved in N, N-dimethylformamide (10mL) and dimethyl sulfate (0.28mL,1.25mmol) and cesium carbonate (1.42g,4.36mmol) were added with stirring at 0 ℃ for 2H. TLC was monitored to completion of the reaction, the reaction solution was slowly added dropwise to ice water and stirred, ethyl acetate was added to extract, and the concentrated organic layer was purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 1:1) to give the title compound 2-chloro-9- (cis-3-hydroxycyclobutyl) -7-methyl-7, 9-dihydro-8H-purin-8-one (13d) (white solid, 0.32g, 41.20% yield).
1H NMR(400MHz,DMSO-d6)δ8.33(s,1H),5.31(d,1H),4.34–4.23(m,1H),4.02-3.91(m,1H),3.34(s,3H),2.81-2.72(m,2H),2.58–2.51(m,2H)。
LC-MS m/z(ESI)=255.00[M+1]。
The fifth step:
9- (cis-3-Hydroxycyclobutyl) -7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -7, 9-dihydro-8H-purin-8-one (Compound 13)
9-(cis-3-hydroxycyclobutyl)-7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-7,9-dihydro-8H-purin-8-one
Mixing 2-chloro-9- ((1s,3s) -3-hydroxycyclobutyl) -7-methyl-7, 9-dihydro-8H-purin-8-one 13d (50mg, 0.20mmol), 7-methyl- [1,2, 4%]Triazolo [1,5-a]Pyridin-6-amine (59mg, 0.40mmol), cesium carbonate (128mg,0.4mmol) and Brettphos G3 Pd (19mg, 0.020mmol) were added to a dry reaction tube, N2After three replacements, dried 1, 4-dioxane (1mL) was added and reacted at 110 ℃ for 5 h. TLC was monitored to the end of the reaction, cooled to room temperature and purified using silica gel column chromatography (dichloromethane/methanol (v/v ═ 40:1)) to give the title compound 9- ((1s,3s) -3-hydroxycyclobutyl) -7-methyl-2- ((6-methyl-2, 3-dihydrobenzofuran-5-yl) amino) -7, 9-dihydro-8H-purin-8-one (compound 13) (light yellow solid, 22mg, 30.50% yield).
1H NMR(400 MHz,DMSO-d6)δ8.65(s,1H),8.14(s,1H),7.95(s,1H),7.26(s,1H),3.94-3.88(m,1H),3.25(s,3H),3.13(t,2H),2.82–2.73(m,2H),2.54-2.44(m,2H),2.12(s,3H)。
LC-MS m/z(ESI)=368.10[M+1]。
Example 14
9- (3-Hydroxybicyclo [3.2.1] octyl-8-yl) -7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -7, 9-dihydro-8H-purin-8-one (Compound 14)
9-(3-hydroxybicyclo[3.2.1]octan-8-yl)-7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-7,9-dihydro-8H-purin-8-one
Figure BDA0002876667980000381
The first step is as follows:
8-Aminobicyclo [3.2.1] octyl-3-one hydrochloride (14b)
8-aminobicyclo[3.2.1]octan-3-one hydrochloride
Tert-butyl 3-oxobicyclo [3.2.1] oct-8-yl) carbamate 14a (1.0g,4.18mmol) was dissolved in 4N dioxane hydrochloride (40mL) and reacted at room temperature for 1 h. TLC was used to monitor the completion of the reaction, and the reaction mixture was directly concentrated to dryness to obtain the objective compound 8-aminobicyclo [3.2.1] octan-3-one hydrochloride 14b (white solid, 730mg, yield 99.42%).
1H NMR(401MHz,DMSO-d6)δ8.77(s,2H),3.36(m,2H),2.80-2.77(m,2H),2.16-2.13(m,2H),1.87-1.85(m,2H),1.43-1.42(m,2H)。
LC-MS m/z(ESI)=176.20[M+1]。
The second step is that:
2-chloro-4- ((3-oxobicyclo [3.2.1] oct-8-yl) amino) pyrimidine-5-carboxylic acid ethyl ester (14c)
ethyl 2-chloro-4-((3-oxobicyclo[3.2.1]octan-8-yl)amino)pyrimidine-5-carboxylate
Ethyl 2, 4-dichloropyrimidine-5-carboxylate 1a (1.38g,6.26mmol), 8-aminobicyclo [3.2.1] octyl-3-one hydrochloride 14b (730mg,4.17mmol) and potassium carbonate (1.73g,12.52mmol) were dissolved in acetonitrile (20mL), and the reaction mixture was reacted at room temperature for 16 hours. TLC, monitored reaction was complete, filtered and the solid was washed with a small amount of acetonitrile, the filtrates were combined and concentrated, and the crude product was isolated by column chromatography (petroleum ether: ethyl acetate ═ 1:1) to afford title compound 2-chloro-4- ((3-oxobicyclo [3.2.1] oct-8-yl) amino) pyrimidine-5-carboxylic acid ethyl ester 14c (white solid, 1.0g, 74.01% yield).
1H NMR(400MHz,DMSO-d6)δ8.86(d,2H),8.68(s,1H),4.36-4.30(q,2H),4.20-4.16(m,1H),2.66-2.65(m,2H),2.56-2.55(m,2H),2.21-2.17(m,2H),1.97-1.93(m,2H),1.53-1.33(m,2H),1.31(t,3H)。
LC-MS m/z(ESI)=324.10[M+1]。
The third step:
2-chloro-4- ((3-oxobicyclo [3.2.1] oct-8-yl) amino ] pyrimidine-5-carboxylic acid (14d)
2-chloro-4-((3-oxobicyclo[3.2.1]octan-8-yl)amino)pyrimidine-5-carboxylic acid
Ethyl 2-chloro-4- ((3-oxobicyclo [3.2.1] oct-8-yl) amino) pyrimidine-5-carboxylate 14c (1.0g, 3.09mmol) was dissolved in 20mL of tetrahydrofuran, water 20mlL, and lithium hydroxide (259mg,6.18mmol) was added, followed by stirring at room temperature for 1 h. TLC to monitor the reaction to completion, concentrate to remove tetrahydrofuran, adjust pH to 5 with 2N hydrochloric acid, precipitate a white solid, filter, wash the filter cake twice with petroleum ether, and collect the solid to give the title compound 2-chloro-4- ((3-3-oxobicyclo [3.2.1] octan-8-yl)
Amino ] pyrimidine-5-carboxylic acid 14d (white solid, 800mg, yield 87.59%).
1H NMR(400MHz,DMSO-d6)δ13.93(s,1H),9.20(d,2H),8.64(s,1H),4.20-4.16(m,1H),2.66-2.64(m,2H),2.54-2.53(m,2H),2.21-2.16(m,2H),1.96-1.93(m,2H),1.53-1.47(m,2H)。
LC-MS m/z(ESI)=296.10[M+1]。
The fourth step:
2-chloro-9- (3-oxobicyclo [3.2.1] oct-8-yl) -7, 9-dihydro-8H-purin-8-one (14e)
2-chloro-9-(3-oxobicyclo[3.2.1]octan-8-yl)-7,9-dihydro-8H-purin-8-one
14d (800mg,2.71mmol) of 2-chloro-4- ((3-3-oxobicyclo [3.2.1] oct-8-yl) amino) pyrimidine-5-carboxylic acid was dissolved in dimethylacetamide (20mL), and triethylamine (0.37mL,2.7mmol) and diphenylphosphorylazide (0.58mL,2.7mmol) were added, followed by gradual warming to 90 ℃ and stirring for 2 hours. TLC monitored completion of the reaction, the reaction was poured into ice water, filtered to collect the solid, washed 3 times with water, concentrated and dried in vacuo to give the title compound 2-chloro-9- (3-oxobicyclo [3.2.1] oct-8-yl) -7, 9-dihydro-8H-purin-8-one 14e (white solid, 630mg, 94.71% yield).
1H NMR(400MHz,DMSO-d6)δ11.66(s,1H),8.13(s,1H),4.01-3.99(m,1H),3.65-3.64(m,2H),2.66-2.61(m,2H),2.12-2.08(m,2H),1.89-1.86(m,2H),1.54-1.49(m,2H)。
LC-MS m/z(ESI)=293.00[M+l]。
The fifth step:
2-chloro-7-methyl-9- (3-oxobicyclo [3.2.1] oct-8-yl) -7, 9-dihydro-8H-purin-8-one (14f)
2-chloro-7-methyl-9-(3-oxobicyclo[3.2.1]octan-8-yl)-7,9-dihydro-8H-purin-8-one
2-chloro-9- (3-oxobicyclo [3.2.1] oct-8-yl) -7, 9-dihydro-8H-purin-8-one 14e (630mg, 2.15mmol) was dissolved in dimethylformamide (10mL), and dimethyl sulfate (0.2mL, 2.15mmol) and cesium carbonate (1.4g,4.3mmol) were added at 0 ℃ and stirred at 0 ℃ for 1H. TLC monitored the completion of the reaction, and the reaction mixture was poured into ice water to precipitate a solid, which was then filtered and collected to obtain the title compound, 2-chloro-7-methyl-9- (3-oxobicyclo [3.2.1] oct-8-yl) -7, 9-dihydro-8H-purin-8-one 14f (white solid, 490mg, 74.22% yield).
1H NMR(400MHz,DMSO-d6)δ8.37(s,1H),4.03-4.01(m,1H),3.66-3.65(m,2H),3.36(s,3H),2.65-2.59(m,2H),2.13-2.08(m,2H),1.89-1.87(m,2H),1.55-1.50(m,2H)。
LC-MS m/z(ESI)=307.10[M+l]。
And a sixth step:
2-chloro-9- (3-hydroxybicyclo [3.2.1] oct-8-yl) -7-methyl-7, 9-dihydro-8H-purin-8-one (14g)
2-chloro-9-(3-hydroxybicyclo[3.2.1]octan-8-yl)-7-methyl-7,9-dihydro-8H-purin-8-one
2-chloro-7-methyl-9- (3-oxobicyclo [3.2.1] oct-8-yl) -7, 9-dihydro-8H-purin-8-one 14f (490mg, 1.6mmol) was dissolved in tetrahydrofuran (30mL) and methanol (30mL), and sodium borohydride (181.30mg, 4.79mmol) was added at 0 ℃. The reaction was monitored by TLC, and the reaction mixture was poured into ice water to precipitate a solid, which was then filtered and collected to obtain 14g (white solid, 380mg, 77.04% yield) of the title compound, 2-chloro-9- (3-hydroxybicyclo [3.2.1] oct-8-yl) -7-methyl-7, 9-dihydro-8H-purin-8-one.
1H NMR(400MHz,DMSO-d6)δ8.37(s,1H),4.28-4.20(m,1H),3.74-3.66(m,2H),3.45-3.43(m,2H),3.37(s,3H),2.14-2.07(m,1H),1.74-1.50(m,5H),1.28-1.22(m,2H)。
LC-MS m/z(ESI)=309.10[M+l]。
The seventh step:
9- (3-Hydroxybicyclo [3.2.1] octyl-8-yl) -7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -7, 9-dihydro-8H-purin-8-one (Compound 14)
9-(3-hydroxybicyclo[3.2.1]octan-8-yl)-7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-7,9-dihydro-8H-purin-8-one
14G (100mg,0.32mmol) of 2-chloro-9- (3-hydroxybicyclo [3.2.1] n-oct-8-yl) -7-methyl-7, 9-dihydro-8H-purin-8-one, 7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-amine (97mg, 0.64mmol), cesium carbonate (228mg, 0.64mmol) and Brettphos G3 Pd (29mg,0.032mmol) were added to a dry reaction tube, replaced with nitrogen three times, then 1, 4-dioxane (2mL) was added, and reacted at 110 ℃ for 5 hours. The reaction solution was concentrated and purified by silica gel column chromatography (DCM: MeOH ═ 20:1) to give the title compound, 9- (3-hydroxybicyclo [3.2.1] oct-8-yl) -7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -7, 9-dihydro-8H-purin-8-one compound 14 (white solid, 50mg, yield 37.16%).
1H NMR(400MHz,DMSO-d6)δ9.03(s,1H),8.57(d,1H),8.37(s,1H),8.09(d,1H),7.69(s,1H),4.30(d,1H),3.75–3.64(m,1H),3.54(d,2H),3.30(s,3H),2.38(s,3H),1.69–1.40(m,6H),1.30(t,2H)。
LC-MS m/z(ESI)=421.20[M+1]。
Example 15
9- (3-hydroxy-3-methylcyclohexyl) -7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -7, 9-dihydro-8H-purin-8-one (Compound 15)
9-(3-hydroxy-3-methylcyclohexyl)-7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-7,9-dihydro-8H-purin-8-one
Figure BDA0002876667980000401
The first step is as follows:
tert-butyl (3-hydroxy-3-methylcyclohexyl) carbamate (15b)
tert-butyl(3-hydroxy-3-methylcyclohexyl)carbamate
Tert-butyl (3-oxocyclohexyl) carbamate 15a (5.00g,23.4mmol) was dissolved in tetrahydrofuran (80mL) and pre-cooled at-78 deg.C, then 1.6M methyllithium-diethyl ether solution (61.5mL,98.28mmol) was slowly added dropwise over 45min, the reaction mixture was stirred at-78 deg.C for 1h, the remaining 1.6M methyllithium-diethyl ether solution (61.5mL,98.28mmol) was added, and the reaction mixture was stirred at-78 deg.C for 1 h. TLC was monitored to the end of the reaction, saturated ammonium chloride solution was added to quench the reaction, water and ethyl acetate were added to extract, and the concentrated organic layer was purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 8:1) to give the title compound tert-butyl (3-hydroxy-3-methylcyclohexyl) carbamate 15b (white solid, 2.07g, 38.53% yield).
1H NMR(400MHz,Chloroform-d)δ4.30(s,1H),3.68(s,1H),1.90(d,3H),1.67(q,1H),1.59–1.48(m,2H),1.37(s,9H),1.19–1.12(m,4H),1.06(t,1H),0.96–0.84(m,1H)。
LC-MS m/z(ESI)=230.20[M+1]。
The second step is that:
3-amino-1-methylcyclohexane-1-ol hydrochloride (15c)
3-amino-1-methylcyclohexan-1-ol hydrochloride
Tert-butyl (3-hydroxy-3-methylcyclohexyl) carbamate 15b (2.07g,9.03mmol) was dissolved in 4M hydrogen chloride-1, 4-dioxane (10mL), and the reaction was stirred at room temperature. TLC monitored to the end of the reaction, concentrated to evaporate the 1, 4-dioxane solution to give the title compound 3-amino-1-methylcyclohexane-1-ol hydrochloride 15c (pale yellow solid, crude, 1.49g, 99.60% yield).
LC-MS m/z(ESI)=130.20[M+1]。
The third step:
2-chloro-4- ((3-hydroxy-3-methylcyclohexyl) amino) pyrimidine-5-carboxylic acid ethyl ester (15d)
ethyl 2-chloro-4-((3-hydroxy-3-methylcyclohexyl)amino)pyrimidine-5-carboxyl-ate
Ethyl 2, 4-dichloropyrimidine-5-carboxylate 1a (2.99g,13.54mmol) and 3-amino-1-methylcyclohexane-1-ol hydrochloride 15c (1.49g,9.03mmol) were dissolved in acetonitrile (20mL), and potassium carbonate (3.74g,27.08mmol) was added with stirring and stirred at room temperature for 4 hours. TLC was monitored to the end of the reaction and concentrated and purified by column separation (petroleum ether: ethyl acetate (v/v) ═ 4:1) to afford the title compound ethyl 2-chloro-4- ((3-hydroxy-3-methylcyclohexyl) amino) pyrimidine-5-carboxylate 15d (white solid, 2.23g, 78.74% yield).
1H NMR(400MHz,Chloroform-d)δ8.57(s,1H),8.19(d,1H),4.43–4.32(m,1H),4.27(q,2H),2.07–1.95(m,2H),1.83–1.69(m,2H),1.64–1.56(m,2H),1.31(t,4H),1.26(d,1H),1.21(s,3H),1.10–1.05(m,1H)。
LC-MS m/z(ESI)=314.10[M+1]。
The fourth step:
2-chloro-4- ((3-hydroxy-3-methylcyclohexyl) amino) pyrimidine-5-carboxylic acid (15e)
2-chloro-4-((3-hydroxy-3-methylcyclohexyl)amino)pyrimidine-5-carboxylate
Ethyl 2-chloro-4- ((3-hydroxy-3-methylcyclohexyl) amino) pyrimidine-5-carboxylate 15d (2.23g,7.11mmol) was dissolved in tetrahydrofuran/water (15mL/15mL), and lithium hydroxide (895mg,21.32mmol) was added and stirred at room temperature for 1 h. TLC monitored to the end of the reaction, concentrated to remove tetrahydrofuran, adjusted to pH 3-4 with 2N hydrochloric acid and a white solid precipitated, filtered, and the filter cake was washed twice with petroleum ether to collect the solid to give the title compound 2-chloro-4- ((3-hydroxy-3-methylcyclohexyl) amino) pyrimidine-5-carboxylic acid 15e (white solid, 1.6g, 78.79% yield).
LC-MS m/z(ESI)=286.10[M+1]。
The fifth step:
2-chloro-9- (3-hydroxy-3-methylcyclohexyl) -7, 9-dihydro-8H-purin-8-one (15f)
2-chloro-9-(3-hydroxy-3-methylcyclohexyl)-7,9-dihydro-8H-purin-8-one
2-chloro-4- ((3-hydroxy-3-methylcyclohexyl) amino) pyrimidine-5-carboxylic acid 29d (1.38g,4.83mmol) was dissolved in dimethylacetamide (20mL), triethylamine (0.67mL,4.83mmol), diphenylphosphorylazide (1.04mL,4.83mmol) were added, followed by gradual warming to 90 ℃ and stirring for 2 h. TLC was carried out until the reaction was completed, and the reaction mixture was poured into ice water, filtered to collect the solid, washed with water 3 times, concentrated and dried in vacuo to give the objective compound 2-chloro-9- (3-hydroxy-3-methylcyclohexyl) -7, 9-dihydro-8H-purin-8-one 15f (white solid, 1.35g, yield 98.86%).
1H NMR(400MHz,DMSO-d6)δ11.60(s,1H),8.10(s,1H),4.62-4.54(m,1H),4.35(s,1H),2.19(t,1H),2.10-1.99(m,1H),1.70-1.53(m,5H),1.30-1.22(m,1H),1.16(s,3H)。
LC-MS m/z(ESI)=283.10[M+1]。
And a sixth step:
2-chloro-9- (3-hydroxy-3-methylcyclohexyl) -7-methyl-7, 9-dihydro-8H-purin-8-one (15g)
2-chloro-9-(3-hydroxy-3-methylcyclohexyl)-7-methyl-7,9-dihydro-8H-purin-8-one
2-chloro-9- (3-hydroxy-3-methylcyclohexyl) -7, 9-dihydro-8H-purin-8-one 15f (1.35g, 4.77mmol) was dissolved in dimethylformamide (10mL), and dimethyl sulfate (0.45mL, 4.77mmol) and cesium carbonate (1.56g,4.77mmol) were added at 0 ℃ and the reaction stirred for 1H. TLC was carried out until the reaction was completed, and the reaction mixture was poured into ice water to precipitate a solid, which was then filtered to collect the solid to obtain 15g (white solid, 873mg, 61.61% yield) of the title compound, 2-chloro-9- (3-hydroxy-3-methylcyclohexyl) -7-methyl-7, 9-dihydro-8H-purin-8-one.
1H NMR(400MHz,DMSO-d6)δ8.34(s,1H),4.66-4.58(m,1H),4.37(s,1H),3.35(s,3H),2.20(t,1H),2.11-2.00(m,1H),1.72-1.54(m,5H),1.32-1.24(m,1H),1.17(s,3H)。
LC-MS m/z(ESI)=297.10[M+1]。
The seventh step:
9- (3-hydroxy-3-methylcyclohexyl) -7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -7, 9-dihydro-8H-purin-8-one (Compound 15)
9-(3-hydroxy-3-methylcyclohexyl)-7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-7,9-dihydro-8H-purin-8-one
15G (200mg, 0.67mmol) of 2-chloro-9- (3-hydroxy-3-methylcyclohexyl) -7-methyl-7, 9-dihydro-8H-purin-8-one, 15G (200mg, 0.67mmol) of 7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-amine (200mg, 1.34mmol), cesium carbonate (473mg, 1.34mmol) and Brettphos G3 Pd (61mg, 0.067mmol) were added to a dry reaction tube, replaced with nitrogen three times, then 1, 4-dioxane (2mL) was added, and reacted at 110 ℃ for 5H. The reaction solution was concentrated and purified by silica gel column chromatography (DCM: MeOH ═ 20:1) to give the title compound, 9- (3-hydroxy-3-methylcyclohexyl) -7-methyl-2- ((-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -7, 9-dihydro-8H-purin-8-one compound 15 (pale yellow solid, 200mg, yield 73.08%).
1H NMR(400MHz,DMSO-d6)δ9.15(s,1H),8.65(s,1H),8.37(s,1H),8.09(s,1H),7.70(d,1H),4.61-4.53(m,1H),4.27(s,1H),3.29(s,3H),2.39(d,3H),2.27(t,1H),2.06–1.93(m,1H),1.76–1.45(m,5H),1.23-1.15(m,1H),1.10(s,3H)。
LC-MS m/z(ESI)=409.20[M+1]。
Example 16
7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -9- (2-oxospiro [3.3] hept-6-yl) -7, 9-dihydro-8H-purin-8-one (Compound 16)
7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-9-(2-oxaspiro[3.3]heptan-6-yl)-7,9-dihydro-8H-purin-8-one
Figure BDA0002876667980000421
Figure BDA0002876667980000431
The first step is as follows:
4- ((2-oxaspiro [3.3] hept-6-yl) amino) -2-chloropyrimidine-5-carboxylic acid ethyl ester (16a)
ethyl 4-((2-oxaspiro[3.3]heptan-6-yl)amino)-2-chloropyrimidine-5-carboxylate
Ethyl 2, 4-dichloropyrimidine-5-carboxylate 1a (4.43g,20.05mmol), 2-oxaspiro [3.3] heptan-6-amine hydrochloride (2.0g,13.37mmol) and potassium carbonate (5.54g, 40.10mmol) were dissolved in acetonitrile (60mL), and the reaction mixture was reacted at room temperature for 16 hours. TLC, after the reaction was completed, the filtrate was washed with a small amount of acetonitrile, the filtrates were combined and concentrated, and the crude product was separated by column chromatography (petroleum ether: ethyl acetate 1:1) to give the target compound, ethyl 4- ((2-oxaspiro [3.3] hept-6-yl) amino) -2-chloropyrimidine-5-carboxylate 16a (white solid, 3.0g, yield 75.38%).
1H NMR(400MHz,DMSO-d6)δ8.60(s,1H),8.47(d,1H),4.63(s,2H),4.49(s,2H),4.39-4.33(m,1H),4.30-4.27(q,2H),2.66-2.61(m,2H),2.31-2.26(m,2H),1.30(t,3H)。
LC-MS m/z(ESI)=298.10[M+1]。
The second step is that:
4- ((2-oxaspiro [3.3] hept-6-yl) amino) -2-chloropyrimidine-5-carboxylic acid (16b)
4-((2-oxaspiro[3.3]heptan-6-yl)amino)-2-chloropyrimidine-5-carboxylic acid
Ethyl 4- ((2-oxaspiro [3.3] hept-6-yl) amino) -2-chloropyrimidine-5-carboxylate 16a (3.0g, 10.08mmol) was dissolved in tetrahydrofuran/water (30mL/30mL), and lithium hydroxide (845mg,20.15mmol) was added, followed by stirring at room temperature for 1 h. TLC monitored to the end of the reaction, concentrated to remove tetrahydrofuran, adjusted to pH 5 with 2N hydrochloric acid and a white solid precipitated, filtered, and the filter cake was washed twice with petroleum ether to collect the solid to give the title compound 4- ((2-oxaspiro [3.3] hept-6-yl) amino) -2-chloropyrimidine-5-carboxylic acid 16b (white solid, 1.8g, 66.24% yield).
1H NMR(400MHz,DMSO-d6)δ13.76(s,1H),8.64(d,1H),8.57(s,1H),4.64(s,2H),4.49(s,2H),4.40-4.30(m,1H),2.67-2.61(m,2H),2.28-2.23(m,2H)。
LC-MS m/z(ESI)=270.20[M+1]。
The third step:
2-chloro-9- (2-oxaspiro [3.3] hept-6-yl) -7, 9-dihydro-8H-purin-8-one (16c)
2-chloro-9-(2-oxaspiro[3.3]heptan-6-yl)-7,9-dihydro-8H-purin-8-one
4- ((2-Oxaspiro [3.3] hept-6-yl) amino) -2-chloropyrimidine-5-carboxylic acid 16b (1.8g,6.67mmol) was dissolved in dimethylacetamide (40mL), triethylamine (0.92mL,6.67mmol), diphenylphosphorylazide (1.4mL,6.67mmol) were added, followed by stepwise warming to 90 ℃ and stirring for reaction for 2 h. TLC was monitored to completion, and the reaction mixture was poured into ice water, filtered to collect the solid, washed with water 3 times, concentrated and dried in vacuo to give 2-chloro-9- (2-oxaspiro [3.3] hept-6-yl) -7, 9-dihydro-8H-purin-8-one 16c (white solid, 1.3g, 73.03% yield).
1H NMR(400MHz,DMSO-d6)δ11.61(s,1H),8.11(s,1H),4.71-4.63(m,5H),3.02-2.94(m,2H),2.69-2.66(m,2H)。
LC-MS m/z(ESI)=267.10[M+1]。
The fourth step:
2-chloro-7-methyl-9- (2-oxaspiro [3.3] hept-6-yl) -7, 9-dihydro-8H-purin-8-one (16d)
2-chloro-7-methyl-9-(2-oxaspiro[3.3]heptan-6-yl)-7,9-dihydro-8H-purin-8-one
2-chloro-9- (2-oxaspiro [3.3] hept-6-yl) -7, 9-dihydro-8H-purin-8-one 16c (1.3g, 4.87mmol) was dissolved in dimethylformamide (10mL), dimethyl sulfate (0.46mL, 4.87mmol) and cesium carbonate (3.18g,9.75mmol) were added at 0 deg.C, and the reaction was stirred for 1H. TLC was monitored to the end of the reaction, and the reaction mixture was poured into ice water to precipitate a solid, which was then filtered and collected to obtain the title compound, 2-chloro-7-methyl-9- (2-oxaspiro [3.3] hept-6-yl) -7, 9-dihydro-8H-purin-8-one 16d (white solid, 875mg, 63.94% yield).
1H NMR(400MHz,DMSO-d6)δ8.35(s,1H),4.76-4.69(m,1H),4.66-4.63(m,4H),3.33(s,3H),3.01-2.95(m,2H),2.71-2.66(m,2H)。
LC-MS m/z(ESI)=281.10[M+1]。
The fifth step:
7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -9- (2-oxospiro [3.3] hept-6-yl) -7, 9-dihydro-8H-purin-8-one (Compound 16)
7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-9-(2-oxaspiro[3.3]heptan-6-yl)-7,9-dihydro-8H-purin-8-one
2-chloro-7-methyl-9- (2-oxaspiro [3.3] heptyl-6-yl) -7, 9-dihydro-8H-purin-8-one 16d (200mg, 0.71mmol), 7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-amine (212mg, 1.42mmol), cesium carbonate (502mg, 1.42mmol) and Brettphos G3 Pd (65mg, 0.071mmol) were added to a dry reaction tube, replaced with nitrogen three times, then 1, 4-dioxane (2mL) was added, and reacted at 110 ℃ for 5H. The reaction solution was concentrated and purified by silica gel column chromatography (DCM: MeOH ═ 20:1) to give the title compound, 7-methyl-2- (((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -9- (2-oxospiro [3.3] hept-6-yl) -7, 9-dihydro-8H-purin-8-one compound 16 (white solid, 160mg, yield 57.42%).
1H NMR(400MHz,DMSO-d6)δ9.05(s,1H),8.65(s,1H),8.39(s,1H),8.11(s,1H),7.73(s,1H),4.60(d,3H),4.16(s,2H),3.27(s,3H),2.97–2.91(m,2H),2.56–2.51(m,2H),2.36(d,3H)。
LC-MS m/z(ESI)=393.10[M+1]。
Example 17
9- (6-Hydroxyspiro [3.3] hept-2-yl) -7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -7, 9-dihydro-8H-purin-8-one (Compound 17)
9-(6-hydroxyspiro[3.3]heptan-2-yl)-7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-7,9-dihydro-8H-purin-8-one
Figure BDA0002876667980000441
Figure BDA0002876667980000451
The first step is as follows:
6-Aminospiro [3.3] heptane-2-ol hydrochloride (17b)
6-aminospiro[3.3]heptan-2-ol hydrochloride
Tert-butyl (6-hydroxyspiro [3.3] heptan-2-yl) carbamate 17a (2g,8.80mmol) was dissolved in 4M hydrogen chloride-1, 4-dioxane (15mL), and the reaction was stirred at room temperature for 2 h. TLC monitored to the end of the reaction, concentrated to evaporate the 1, 4-dioxane solution to afford the title compound 6-aminospiro [3.3] heptane-2-ol hydrochloride 17b (pale yellow solid, crude, 1.44g, 100% yield).
LC-MS m/z(ESI)=163.10[M+1]。
The second step is that:
2-chloro-4- ((6-hydroxyspiro [3.3] hept-2-yl) amino) pyrimidine-5-carboxylic acid ethyl ester (17c)
Ethyl 2-chloro-4- ((3-hydroxy-3-methycyclohexenyl) amino) pyrimidine-5-carboxyl-ate Ethyl 2, 4-dichloropyrimidine-5-carboxylate 1a (2.92g,13.20mmol), 6-aminospiro [3.3] heptane-2-ol hydrochloride 17b (1.44g,8.80mmol) were dissolved in acetonitrile (20mL), potassium carbonate (3.65g,26.40mmol) was added with stirring, and stirred at room temperature for 4 h. TLC was monitored to the end of the reaction and concentrated and purified by column separation (petroleum ether: ethyl acetate (v/v) ═ 4:1) to afford the title compound ethyl 2-chloro-4- ((3-hydroxy-3-methylcyclohexyl) amino) pyrimidine-5-carboxylate 17c (white solid g, 1.36g, 49.57% yield).
1H NMR(400MHz,DMSO-d6)δ8.59(s,1H),8.43(d,1H),4.91(s,1H),4.44–4.34(m,1H),4.31(q,2H),4.02–3.93(m,1H),2.43–2.35(m,2H),2.34–2.27(m,1H),2.23–2.16(m,1H),2.07–1.99(m,2H),1.90–1.80(m,2H),1.31(t,3H)。
LC-MS m/z(ESI)=312.10[M+1]。
The third step:
2-chloro-4- ((6-hydroxyspiro [3.3] hept-2-yl) amino) pyrimidine-5-carboxylic acid (17d)
2-chloro-4-((6-hydroxyspiro[3.3]heptan-2-yl)amino)pyrimidine-5-carboxylic acid
Ethyl 2-chloro-4- ((6-hydroxyspiro [3.3] hept-2-yl) amino) pyrimidine-5-carboxylate 17c (1.0g, 3.21mmol) was dissolved in 10mL of tetrahydrofuran and 10mL of water, and lithium hydroxide (269mg,6.42mmol) was added thereto, followed by stirring at room temperature for 1 hour. TLC monitored reaction to completion, concentrated to remove tetrahydrofuran, adjusted to pH 5 with 2N hydrochloric acid and a white solid precipitated, filtered, and the filter cake was washed twice with petroleum ether to collect the solid to give the title compound 2-chloro-4- ((6-hydroxyspiro [3.3] hept-2-yl) amino) pyrimidine-5-carboxylic acid 17d (760mg, white solid, 83.52% yield).
1H NMR(400MHz,DMSO-d6)δ13.75(s,1H),8.62(d,1H),8.56(s,1H),4.90(s,1H),4.42-4.32(m,1H),4.00-3.93(m,1H),2.42-2.35(m,2H),2.33-2.27(m,1H),2.22-2.16(m,1H),2.02-1.97(m,2H),1.88-1.80(m,2H)。
LC-MS m/z(ESI)=284.10[M+1]。
The fourth step:
2-chloro-9- (6-hydroxyspiro [3.3] heptan-2-yl) -7, 9-dihydro-8H-purin-8-one (17e)
2-chloro-9-(6-hydroxyspiro[3.3]heptan-2-yl)-7,9-dihydro-8H-purin-8-one
17d (760mg,2.68mmol) of 2-chloro-4- ((6-hydroxyspiro [3.3] hept-2-yl) amino) pyrimidine-5-carboxylic acid was dissolved in dimethylacetamide (10mL), triethylamine (0.37mL,2.68mmol) and diphenylphosphorylazide (0.57mL,2.68mmol) were added, followed by stepwise warming to 90 ℃ and stirring for 2 hours. TLC monitored the reaction to completion, the reaction was poured into ice water, the solid collected by filtration, washed 3 times with water, concentrated and dried in vacuo to give the title compound 2-chloro-9- (6-hydroxyspiro [3.3] heptan-2-yl) -7, 9-dihydro-8H-purin-8-one 17e (580mg, white solid, 77.13% yield).
1H NMR(400MHz,DMSO-d6)δ11.60(s,1H),8.10(s,1H),4.96(d,1H),4.70-4.60(m,1H),2.92-2.87(m,2H),2.46-2.43(m,2H),2.32-2.21(m,3H),1.92-1.87(m,2H)。
LC-MS m/z(ESI)=281.10[M+1]。
The fifth step:
2-chloro-9- (6-hydroxyspiro [3.3] heptan-2-yl) -7-methyl-7, 9-dihydro-8H-purin-8-one (17f)
2-chloro-9-(6-hydroxyspiro[3.3]heptan-2-yl)-7-methyl-7,9-dihydro-8H-purin-8-one
2-chloro-9- (6-hydroxyspiro [3.3] heptan-2-yl) -7, 9-dihydro-8H-purin-8-one 17e (580mg, 2.07mmol) was dissolved in dimethylformamide (5mL), and dimethyl sulfate (260mg, 2.07mmol) and cesium carbonate (1.3g,4.13mmol) were added at 0 ℃ and the reaction stirred for 1 hour. TLC monitored the reaction was complete, the reaction mixture was poured into ice water, a solid precipitated, and the solid was collected by filtration to give the title compound 2-chloro-9- (6-hydroxyspiro [3.3] heptan-2-yl) -7-methyl-7, 9-dihydro-8H-purin-8-one 17f (380mg, white solid, 62.40% yield).
1H NMR(400MHz,DMSO-d6)δ8.33(s,1H),4.97(d,1H),4.70-4.66(m,1H),4.05-3.96(m,1H),3.33(s,3H),2.95-2.87(m,2H),2.51-2.43(m,1H),2.31-2.25(m,3H),1.93-1.87(m,2H)。
LC-MS m/z(ESI)=295.10[M+1]。
And a sixth step:
9- (6-Hydroxyspiro [3.3] hept-2-yl) -7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -7, 9-dihydro-8H-purin-8-one (Compound 17)
9-(6-hydroxyspiro[3.3]heptan-2-yl)-7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-7,9-dihydro-8H-purin-8-one
2-chloro-9- (6-hydroxyspiro [3.3] hept-2-yl) -7-methyl-7, 9-dihydro-8H-purin-8-one 17f (140mg, 0.47mmol), 7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-amine (142mg, 0.94mmol), cesium carbonate (335mg, 0.94mmol) and Brettphos G3 Pd (43mg, 0.047mmol) were added to a dry reaction tube, replaced with nitrogen three times, then 1, 4-dioxane (2mL) was added, and reacted at 110 ℃ for 5H. The reaction solution was concentrated and purified by silica gel column chromatography (DCM: MeOH ═ 20:1) to give the title compound, 9- (6-hydroxyspiro [3.3] hept-2-yl) -7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -7, 9-dihydro-8H-purin-8-one compound 17 (white solid, 110mg, yield 57.57%).
1H NMR(400MHz,DMSO-d6)δ9.05(s,1H),8.67(s,1H),8.37(s,1H),8.09(s,1H),7.71(s,1H),4.90(d,1H),4.65-4.56(m,1H),3.93-3.84(m,1H),3.27(s,3H),2.87-2.79(m,2H),2.37(s,4H),2.20-2.05(m,2H),1.86-1.78(m,2H),1.64-1.59(m,1H)。
LC-MS m/z(ESI)=407.20[M+1]。
Example 18
9- (8-Oxabicyclo [3.2.1] octyl-3-yl) -7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -7, 9-dihydro-8H-purin-8-one (Compound 18)
9-(8-oxabicyclo[3.2.1]octan-3-yl)-7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-7,9-dihydro-8H-purin-8-one
Figure BDA0002876667980000471
The first step is as follows:
8-oxabicyclo [3.2.1] octan-3-one oxime (18b)
8-oxabicyclo[3.2.1]octan-3-one oxime
8-Oxabicyclo [3.2.1] octan-3-one 18a (1.4g, 11.1mmol), hydroxylamine hydrochloride (925mg,13.3mmol) and potassium carbonate (3.1g,22.2mmol) were dissolved in a mixed solvent of ethanol/water (10mL/5mL) and reacted at 80 ℃ for 2 hours. TLC monitored the reaction was complete, and the reaction was quenched with water, extracted with ethyl acetate, dried over anhydrous sodium sulfate, filtered and concentrated to give the title compound, 8-oxabicyclo [3.2.1] octan-3-one oxime 18b (light yellow solid, 1.56g, 100% yield).
LC-MS m/z(ESI)=142.10[M+1]。
The second step is that:
8-oxabicyclo [3.2.1] octan-3-amine (18c)
8-oxabicyclo[3.2.1]octan-3-amine
8-Oxabicyclo [3.2.1] octan-3-one oxime 18b (1.5g, 10.64mmol) was dissolved in methanol (30mL), and nickel chloride hexahydrate (2.53g, 10.64mmol) was added at room temperature to react for 0.5h at room temperature. Cooling the reaction solution to-30 ℃, slowly adding sodium borohydride (6.0g, 159.6mmol), slowly heating to room temperature after the addition is finished, and reacting overnight; TLC monitored the reaction was complete, and the reaction was quenched with water, extracted with ethyl acetate, dried over anhydrous sodium sulfate, filtered and concentrated to give the title compound, 8-oxabicyclo [3.2.1] octan-3-amine 18c (pale yellow oil, 639mg, 45% yield).
LC-MS m/z(ESI)=128.20[M+1]。
The third step:
4- ((8-oxabicyclo [3.2.1] octan-3-yl) amino) -2-chloropyrimidine-5-carboxylic acid ethyl ester (18d)
ethyl 4-((8-oxabicyclo[3.2.1]octan-3-yl)amino)-2-chloropyrimidine-5-carboxylate
Ethyl 2, 4-dichloropyrimidine-5-carboxylate 1a (1.1g, 5.03mmol) and potassium carbonate (1.74g, 12.58mmol) were dissolved in acetonitrile (20mL), and 8-oxabicyclo [3.2.1] oct-3-amine 18c (639mg, 5.03mmol) was added at 0 ℃ and the mixture was stirred at room temperature for 20 hours. TLC monitored to completion of the reaction, 30mL of water was added to the reaction mixture, a solid precipitated, filtered and washed with water 3 times, and concentrated to give the title compound, ethyl 4- ((8-oxabicyclo [3.2.1] octan-3-yl) amino) -2-chloropyrimidine-5-carboxylate 18d (white solid, 1.01g, 64.3% yield).
1H NMR(400MHz,DMSO)δ8.91(d,1H),8.64(s,1H),4.38–4.31(m,4H),4.30–4.24(m,1H),2.20–2.08(m,2H),2.02–1.90(m,4H),1.66(d,2H),1.32(t,3H)。
The fourth step:
4- ((8-oxabicyclo [3.2.1] oct-3-yl) amino) -2-chloropyrimidine-5-carboxylic acid (18e)
4-((8-oxabicyclo[3.2.1]octan-3-yl)amino)-2-chloropyrimidine-5-carboxylic acid
Ethyl 4- ((8-oxabicyclo [3.2.1] octan-3-yl) amino) -2-chloropyrimidine-5-carboxylate (1.0g, 3.21mmol)18d was dissolved in 10mL of tetrahydrofuran and 5mL of water, and lithium hydroxide (308mg, 12.83mmol) was added thereto, followed by stirring at room temperature for 1 hour. TLC monitored to the end of the reaction, tetrahydrofuran was spun down, pH adjusted to 4-5 with 2N HCl and a white solid precipitated, filtered, and the filter cake was washed twice with petroleum ether/ethyl acetate (v/v ═ 10/1) and concentrated to give the title compound, 4- ((8-oxabicyclo [3.2.1] oct-3-yl) amino) -2-chloropyrimidine-5-carboxylic acid 18e (white solid, 808mg, 87.9% yield).
1H NMR(400MHz,DMSO)δ13.83(s,1H),9.17(d,1H),8.59(s,1H),4.32(s,2H),4.29–4.22(m,1H),2.17–2.07(m,2H),2.00–1.89(m,4H),1.65(d,2H)。
The fifth step:
9- (8-oxabicyclo [3.2.1] oct-3-yl) -2-chloro-7, 9-dihydro-8H-purin-8-one (18f)
9-(8-oxabicyclo[3.2.1]octan-3-yl)-2-chloro-7,9-dihydro-8H-purin-8-one
18e (808mg, 2.85mmol) of 4- ((8-oxabicyclo [3.2.1] oct-3-yl) amino) -2-chloropyrimidine-5-carboxylic acid was dissolved in dimethylacetamide (20mL), and triethylamine (288mg, 2.85mmol), diphenyl phosphorazidate (784mg, 2.85mmol) were added, followed by gradual warming to 120 ℃ and stirring for 1.5 h. TLC was monitored to completion of the reaction, the reaction solution was concentrated, and the residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 5:1 to 1:10) to give the title compound, 9- (8-oxabicyclo [3.2.1] oct-3-yl) -2-chloro-7, 9-dihydro-8H-purin-8-one 18f (white solid, 653mg, 71% yield).
1H NMR(600MHz,DMSO)δ11.64(s,1H),8.12(s,1H),4.47–4.44(m,2H),4.42–4.40(m,1H),2.28–2.20(m,2H),2.07–2.01(m,2H),1.96–1.90(m,2H),1.81–1.75(m,2H)。
And a sixth step:
9- (8-oxabicyclo [3.2.1] oct-3-yl) -2-chloro-7-methyl-7, 9-dihydro-8H-purin-8-one (18g)
9-(8-oxabicyclo[3.2.1]octan-3-yl)-2-chloro-7-methyl-7,9-dihydro-8H-purin-8-one
9- (8-Oxabicyclo [3.2.1] oct-3-yl) -2-chloro-7, 9-dihydro-8H-purin-8-one 18f (653mg, 2.33mmol) was dissolved in dimethylformamide (10mL), dimethyl sulfate (293mg, 2.33mmol) and cesium carbonate (1.52g,4.65mmol) were added at 0 ℃ and stirred at 0 ℃ for 1H. TLC was monitored to the end of the reaction, 10mL of water was then added, extracted 3 times with ethyl acetate, and the organic phase was dried over anhydrous sodium sulfate and concentrated to give the title compound, 9- (8-oxabicyclo [3.2.1] oct-3-yl) -2-chloro-7-methyl-7, 9-dihydro-8H-purin-8-one, 18g (white solid, 533mg, 81.6% yield).
1H NMR (400MHz, DMSO). delta.8.36-8.34 (m,1H),4.51-4.47(m,2H), 4.45-4.42 (m,1H),3.34(s,3H), 2.31-2.22 (m,2H), 2.07-1.98 (m,2H), 1.98-1.90 (m,2H),1.79(t, 2H). The seventh step:
9- (8-Oxabicyclo [3.2.1] octyl-3-yl) -7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -7, 9-dihydro-8H-purin-8-one (Compound 18)
9-(8-oxabicyclo[3.2.1]octan-3-yl)-7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-7,9-dihydro-8H-purin-8-one
18G (100mg, 0.34mmol) of 9- (8-oxabicyclo [3.2.1] oct-3-yl) -2-chloro-7-methyl-7, 9-dihydro-8H-purin-8-one, 7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-amine (51mg, 0.34mmol), cesium carbonate (221mg,0.68mmol), Brettphos Pd G3(31mg, 0.034mmol) were dissolved in dioxane, purged with nitrogen and stirred at 100 ℃ for 4H. TLC was monitored to the end of the reaction, the reaction solution was concentrated, and the residue was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 30/1) to give the title compound 9- (8-oxabicyclo [3.2.1] octyl-3-yl) -7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -7, 9-dihydro-8H-purin-8-one compound 18 (white solid, 41mg, yield 28.9%).
1H NMR(400MHz,DMSO-d6)δ9.01(s,1H),8.70(s,1H),8.39(s,1H),8.11(s,1H),7.74(s,1H),4.41–4.27(m,3H),3.29(s,3H),2.34(s,3H),2.17–2.07(m,2H),1.91(t,2H),1.62(s,2H),1.28(d,2H)。
Example 19
7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -9- (2-oxaspiro [3.5] nonyl-7-yl) -7, 9-dihydro-8H-purin-8-one (Compound 19)
7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-9-(2-oxaspiro[3.5]nonan-7-yl)-7,9-dihydro-8H-purin-8-one
Figure BDA0002876667980000491
Figure BDA0002876667980000492
The first step is as follows:
2-oxaspiro [3.5] nonane-7-ketoxime (19b)
2-oxaspiro[3.5]nonan-7-one oxime
19a (1.5g,10.7mmol), hydroxylamine hydrochloride (744mg,10.7mmol) and potassium carbonate (2.95g,21.4mmol) were dissolved in a mixed solvent of ethanol/water (10mL/5mL), reacted at 80 ℃ for 2 hours, TLC monitored until the reaction was completed, quenched by adding water to the reaction solution, extracted with ethyl acetate, dried over anhydrous sodium sulfate, filtered and concentrated to give the title compound, 2-oxaspiro [3.5] non-7-ketoxime 19b (pale yellow solid, 1.45g, 89% yield).
The second step is that:
2-oxaspiro [3.5] nonan-7-amine (19c)
2-oxaspiro[3.5]nonan-7-amine
2-Oxaspiro [3.5] non-7-ketoxime 19b (1.45g, 9.34mmol) was dissolved in methanol (30mL), nickel chloride hexahydrate (2.22g, 9.34mmol) was added at room temperature, and after reaction at room temperature for 0.5h, the reaction mixture was cooled to-30 ℃ and sodium borohydride (5.3g, 140.1mmol) was slowly added, and after the addition, the temperature was slowly raised to room temperature and reacted overnight. TLC monitored the reaction was complete, and the reaction was quenched with water, extracted with ethyl acetate, dried over anhydrous sodium sulfate, filtered and concentrated to give the title compound, 2-oxaspiro [3.5] nonan-7-amine 19c (pale yellow oil, 481mg, 37.6% yield).
The third step:
4- ((2-oxaspiro [3.5] non-7-yl) amino) -2-chloropyrimidine-5-carboxylic acid ethyl ester (19d)
ethyl 4-((2-oxaspiro[3.5]nonan-7-yl)amino)-2-chloropyrimidine-5-carboxylate
Ethyl 2, 4-dichloropyrimidine-5-carboxylate 1a (753mg, 3.41mmol) and potassium carbonate (940mg, 6.81mmol) were dissolved in acetonitrile (20mL), and 2-oxaspiro [3.5] nonan-7-amine 19c (481mg, 3.41mmol) was added thereto at 0 ℃ and the mixture was stirred at room temperature for 20 hours. TLC monitored to the end of the reaction, 30mL of water was added to the reaction solution, a solid precipitated, filtered and washed with water 3 times, and concentrated to give the title compound, ethyl 4- ((2-oxaspiro [3.5] nonan-7-yl) amino) -2-chloropyrimidine-5-carboxylate 19d (white solid, 640mg, 57.6% yield).
1H NMR(400MHz,DMSO)δ8.61(s,1H),8.29(d,1H),4.34–4.29(m,4H),4.24(s,2H),3.98–3.89(m,1H),2.05–1.92(m,2H),1.86–1.75(m,2H),1.65–1.56(m,2H),1.45–1.35(m,2H),1.34–1.27(m,3H)。
The fourth step:
4- ((2-oxaspiro [3.5] nonyl-7-yl) amino) -2-chloropyrimidine-5-carboxylic acid (19e)
4-((2-oxaspiro[3.5]nonan-7-yl)amino)-2-chloropyrimidine-5-carboxylic acid
Ethyl 4- ((2-oxaspiro [3.5] non-7-yl) amino) -2-chloropyrimidine-5-carboxylate 19d (640mg, 1.96mmol) was dissolved in 10mL of tetrahydrofuran and 5mL of water, and lithium hydroxide (189mg, 7.86mmol) was added and the mixture was stirred at room temperature for 1 h. TLC monitored to the end of the reaction and tetrahydrofuran was spun down, adjusted to pH 4-5 and a white solid precipitated, filtered, and the filter cake was washed twice with petroleum ether/ethyl acetate (v/v ═ 10/1) and concentrated to give the title compound, 4- ((2-oxaspiro [3.5] nonyl-7-yl) amino) -2-chloropyrimidine-5-carboxylic acid 19e (white solid, 518mg, 88.5% yield).
1H NMR(400MHz,DMSO)δ13.74(s,1H),8.57(s,1H),8.48(d,1H),4.32(s,2H),4.24(s,2H),3.97–3.84(m,1H),1.99(d,2H),1.86–1.76(m,2H),1.66–1.54(m,2H),1.42–1.26(m,2H)。
The fifth step:
2-chloro-9- (2-oxaspiro [3.5] non-7-yl) -7, 9-dihydro-8H-purin-8-one (19f)
2-chloro-9-(2-oxaspiro[3.5]nonan-7-yl)-7,9-dihydro-8H-purin-8-one
4- ((2-oxaspiro [3.5] nonyl-7-yl) amino) -2-chloropyrimidine-5-carboxylic acid 19e (518mg, 1.74mmol) was dissolved in dimethylacetamide (20mL), triethylamine (175mg, 1.74mmol), diphenyl azidophosphate (479mg, 1.74mmol) were added, followed by stepwise warming to 120 ℃ and stirring for 1.5 h. TLC was monitored to completion of the reaction, the reaction solution was concentrated, and the residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 5:1 to 1:10) to give the title compound, 2-chloro-9- (2-oxaspiro [3.5] non-7-yl) -7, 9-dihydro-8H-purin-8-one 19f (white solid, 353mg, 72.8% yield).
1H NMR(600MHz,DMSO)δ11.61(s,1H),8.11(s,1H),4.41(s,2H),4.24(s,2H),4.15–4.08(m,1H),2.24–2.08(m,4H),1.69(d,2H),1.64–1.51(m,2H)。
And a sixth step:
2-chloro-7-methyl-9- (2-oxaspiro [3.5] non-7-yl) -7, 9-dihydro-8H-purin-8-one (19g)
2-chloro-7-methyl-9-(2-oxaspiro[3.5]nonan-7-yl)-7,9-dihydro-8H-purin-8-one
2-chloro-9- (2-oxaspiro [3.5] non-7-yl) -7, 9-dihydro-8H-purin-8-one 19f (353mg, 1.18mmol) was dissolved in dimethylformamide (10mL), dimethyl sulfate (150mg, 1.18mmol) and cesium carbonate (464mg,3.36mmol) were added at 0 deg.C, and stirred at 0 deg.C for 1H. TLC monitored the reaction to completion, then 10mL of water was added, extracted 3 times with ethyl acetate, the organic phase was dried over anhydrous sodium sulfate, concentrated and a solid precipitated which was filtered to give the title compound, 2-chloro-7-methyl-9- (2-oxaspiro [3.5] non-7-yl) -7, 9-dihydro-8H-purin-8-one, 19g (white solid, 313mg, 85.5% yield).
1H NMR(400MHz,DMSO)δ8.34(s,1H),4.41(s,2H),4.25(s,2H),4.22–4.08(m,1H),3.34(s,3H),2.22–2.05(m,4H),1.69(d,2H),1.64–1.50(m,2H)。
The seventh step:
7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -9- (2-oxaspiro [3.5] nonyl-7-yl) -7, 9-dihydro-8H-purin-8-one (Compound 19)
7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-9-(2-oxaspiro[3.5]nonan-7-yl)-7,9-dihydro-8H-purin-8-one
19G (60mg, 0.194mmol) of 2-chloro-7-methyl-9- (2-oxaspiro [3.5] non-7-yl) -7, 9-dihydro-8H-purin-8-one, 19G (29mg, 0.194mmol) of 7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-amine (29mg, 0.194mmol), cesium carbonate (127mg, 0.39mmol), Brettphos Pd G3(17mg, 0.019mmol) were dissolved in dioxane, purged with nitrogen and stirred at 100 ℃ for 4H. The reaction solution was concentrated, and the residue was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 30/1) to give the title compound 9- (8-oxabicyclo [3.2.1] octyl-3-yl) -7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -7, 9-dihydro-8H-purin-8-one compound 19 (white solid, 38mg, yield 44.7%).
1H NMR(400MHz,DMSO-d6)δ8.99(s,1H),8.66(s,1H),8.38(s,1H),8.10(s,1H),7.73(s,1H),4.15(s,2H),4.05(s,1H),3.92(s,2H),3.29(s,3H),2.34(s,3H),2.09–2.00(m,4H),1.63–1.56(m,2H),1.51–1.44(m,2H)。
Example 20
9- (6- (hydroxymethyl) spiro [3.3] hept-2-yl) -7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -7, 9-dihydro-8H-purin-8-one (Compound 20)
9-(6-(hydroxymethyl)spiro[3.3]heptan-2-yl)-7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-7,9-dihydro-8H-purin-8-one
Figure BDA0002876667980000511
Figure BDA0002876667980000512
The first step is as follows:
(6-Aminospiro [3.3] heptan-2-yl) methanol (20b)
(6-aminospiro[3.3]heptan-2-yl)methanol
6-amino spiro [3.3] heptane-2-carboxylic acid methyl ester 20a (2.0g,11.8mmol) is dissolved in tetrahydrofuran (15mL), the mixed solution is cooled to 0 ℃, 1N lithium aluminum hydride solution (23.6mL, 1mol/L tetrahydrofuran solution) is slowly dripped, and after the dripping is finished, the temperature is slowly raised to room temperature and stirring is carried out for 2 h. TLC monitored to the end of the reaction, quenched by addition of methanol, concentrated under reduced pressure to evaporate the solvent, slurried with acetonitrile (50mL), filtered and the filtrate was rotary dried to give the title compound (6-aminospiro [3.3] heptan-2-yl) methanol 20b (white solid, 1.3g, 77.8% yield).
LC-MS m/z(ESI)=142.10[M+1]。
The second step is that:
2-chloro-4- ((6- (hydroxymethyl) spiro [3.3] heptan-2-yl) amino) pyrimidine-5-carboxylic acid ethyl ester (20c)
ethyl 2-chloro-4-((6-(hydroxymethyl)spiro[3.3]heptan-2-yl)amino)pyrimidine-5-carboxylate
Ethyl 2, 4-dichloropyrimidine-5-carboxylate 1a (2.03g,9.19mmol), (6-aminospiro [3.3] heptan-2-yl) methanol 20b (1.3g,9.19mmol), potassium carbonate (1.27g,9.19mmol) were dissolved in acetonitrile (25mL) and the reaction was stirred at room temperature for 16 h. TLC to the end of the reaction, the filtrate was concentrated and purified by silica gel column chromatography (petroleum ether: ethyl acetate (v/v) ═ 1:1) to give the title compound ethyl 2-chloro-4- ((6- (hydroxymethyl) spiro [3.3] heptan-2-yl) amino) pyrimidine-5-carboxylate 20c (white solid, 1.56g, 52.2% yield).
1H NMR(400MHz,Chloroform-d)δ8.65(s,1H),8.54(d,1H),4.59–4.48(m,1H),4.35(q,2H),3.59(dd,2H),2.66–2.58(m,1H),2.51–2.38(m,2H),2.27–2.19(m,1H),2.11–2.04(m,2H),2.03–1.91(m,2H),1.91–1.85(m,1H),1.84–1.76(m,1H),1.39(t,3H)。
LCMS m/z(ESI)=326.10[M+1]。
The third step:
2-chloro-4- ((6- (hydroxymethyl) spiro [3.3] heptan-2-yl) amino) pyrimidine-5-carboxylic acid (20d)
2-chloro-4-((6-(hydroxymethyl)spiro[3.3]heptan-2-yl)amino)pyrimidine-5-carboxylic acid
Ethyl 2-chloro-4- ((6- (hydroxymethyl) spiro [3.3] heptan-2-yl) amino) pyrimidine-5-carboxylate 20c (1.07g,3.28mmol) was dissolved in tetrahydrofuran/water (10mL/10mL), lithium hydroxide monohydrate (0.41g,9.85mmol) was added, and the reaction was stirred at ordinary temperature for 1 h. TLC to the end of the reaction, concentrated to evaporate tetrahydrofuran, then adjusted the pH to 3-4 by addition of 2N HCl and a white solid precipitated, filtered and the filter cake was washed twice with water and petroleum ether/ethyl acetate (v/v ═ 10/1) to afford the title compound 2-chloro-4- ((6- (hydroxymethyl) spiro [3.3] heptan-2-yl) amino) pyrimidine-5-carboxylic acid 20d (white solid, 918mg, 93.88% yield).
1H NMR(400MHz,DMSO-d6)δ12.69(s,1H),9.67(d,1H),8.35(s,1H),4.42–4.31(m,1H),3.30(s,1H),2.55–2.46(m,3H),2.40–2.31(m,1H),2.28–2.19(m,1H),2.10–2.00(m,2H),2.00–1.91(m,2H),1.83–1.69(m,2H)。
LC-MS m/z(ESI)=298.10[M+1]。
The fourth step:
2-chloro-9- (6- (hydroxymethyl) spiro [3.3] heptan-2-yl) -7, 9-dihydro-8H-purin-8-one (20e)
2-chloro-9-(6-(hydroxymethyl)spiro[3.3]heptan-2-yl)-7,9-dihydro-8H-purin-8-one
Dissolving 20d (0.91g and 3.06mmol) of 2-chloro-4- ((6- (hydroxymethyl) spiro [3.3] heptane-2-yl) amino) pyrimidine-5-carboxylic acid with N, N-dimethylacetamide (12mL), adding triethylamine (0.42mL and 3.06mmol) and diphenylphosphorylazide (0.66mL and 10.01mmol) under stirring at normal temperature for reaction for 2h, and heating to 110 ℃ for reflux reaction for 2.5 h. TLC was monitored to the end of the reaction, water and ethyl acetate were added to the reaction solution for extraction, and the concentrated organic layer was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 20:1) to give the title compound 2-chloro-9- (6- (hydroxymethyl) spiro [3.3] heptan-2-yl) -7, 9-dihydro-8H-purin-8-one 20e (white solid, 516mg, 57.28% yield).
1H NMR(400MHz,DMSO-d6)δ11.60(s,1H),8.11(s,1H),4.68–4.57(m,1H),4.46(t,1H),3.37–3.32(m,2H),2.95–2.81(m,2H),2.42–2.35(m,1H),2.32–2.19(m,2H),2.19–2.12(m,1H),2.06–1.98(m,1H),1.89–1.77(m,2H)。
LC-MS m/z(ESI)=295.10[M+1]。
The fifth step:
2-chloro-9- (6- (hydroxymethyl) spiro [3.3] heptan-2-yl) -7-methyl-7, 9-dihydro-8H-purin-8-one (20f)
2-chloro-9-(6-(hydroxymethyl)spiro[3.3]heptan-2-yl)-7-methyl-7,9-dihydro-8H-purin-8-one
2-chloro-9- (6- (hydroxymethyl) spiro [3.3] heptan-2-yl) -7, 9-dihydro-8H-purin-8-one 20e (516mg,1.75mmol) was dissolved in N, N-dimethylformamide (6mL), and dimethyl sulfate (0.17mL,1.75mmol) and cesium carbonate (1.14g,3.50mmol) were added with stirring at 0 ℃ for 1H. TLC monitored to completion, the reaction mixture was slowly added dropwise to ice water with stirring, ethyl acetate was added for extraction, and the organic layer was concentrated to give the title compound 2-chloro-9- (6- (hydroxymethyl) spiro [3.3] heptan-2-yl) -7-methyl-7, 9-dihydro-8H-purin-8-one 20f (white solid, 470mg, 86.95% yield).
1H NMR(400MHz,DMSO-d6)δ8.33(s,1H),4.71–4.60(m,1H),4.10–3.96(m,2H),3.35(s,1H),3.33(s,3H),2.93–2.81(m,2H),2.44–2.36(m,1H),2.32–2.21(m,2H),2.20–2.13(m,1H),2.07–1.99(m,1H),1.89–1.78(m,2H)。
LC-MS m/z(ESI)=309.10[M+1]。
And a sixth step:
9- (6- (hydroxymethyl) spiro [3.3] hept-2-yl) -7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -7, 9-dihydro-8H-purin-8-one (Compound 20)
9-(6-(hydroxymethyl)spiro[3.3]heptan-2-yl)-7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-7,9-dihydro-8H-purin-8-one
2-chloro-9- (6- (hydroxymethyl) spiro [3.3] heptan-2-yl) -7-methyl-7, 9-dihydro-8H-purin-8-one 20f (100mg,0.32mmol) was dissolved in 1, 4-dioxane (6mL), and 7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-amine (72mg,0.49mmol), cesium carbonate (158mg,0.49mmol), Brettphos-G3-Pd (29mg,0.032mmol) was added with stirring at room temperature, with nitrogen protection and purging, followed by heating to 110 ℃ and refluxing for 4H. TLC was monitored to the end of the reaction, the reaction was concentrated, and the residue was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 20:1) to give title compound 20 (white solid, 60mg, 44.59% yield).
1H NMR(400MHz,DMSO-d6)δ9.05(s,1H),8.66(s,1H),8.36(s,1H),8.09(s,1H),7.71(s,1H),4.66–4.54(m,1H),4.40(t,1H),3.29–3.23(m,5H),2.85–2.77(m,2H),2.37(s,3H),2.30–2.22(m,1H),2.19–2.12(m,1H),2.12–2.04(m,2H),1.81–1.74(m,1H),1.66–1.59(m,1H),1.49–1.42(m,1H)。
LC-MS m/z(ESI)=421.20[M+1]。
Example 21
9- (3- (hydroxymethyl) bicyclo [1.1.1] pent-1-yl) -7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -7, 9-dihydro-8H-purin-8-one (Compound 21)
9-(3-(hydroxymethyl)bicyclo[1.1.1]pentan-1-yl)-7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-7,9-dihydro-8H-purin-8-one
Figure BDA0002876667980000531
Figure BDA0002876667980000532
The first step is as follows:
(3-Aminobicyclo [1.1.1] pentane-1-yl) methanol (21b)
(3-aminobicyclo[1.1.1]pentan-1-yl)methanol
The starting material, 3- (tert-butoxycarbonyl) amino) bicyclo [1.1.1] pentane-1-carboxylic acid 21a (2.0g,8.80mmol) was dissolved in dichloromethane (10mL), and further, trifluoroacetic acid (10mL) was added, and the mixture was stirred at room temperature for 2 hours. TLC monitoring till the reaction is finished, decompressing and drying the solvent, dissolving the obtained light yellow solid in tetrahydrofuran (15mL), cooling the mixed solution to 0 ℃, slowly dropwise adding 1N lithium aluminum hydride solution (16.6mL, 1mol/L tetrahydrofuran solution), slowly heating to room temperature and stirring for 2h after the dropwise adding is finished, adding methanol to quench the reaction after the reaction is finished, decompressing and drying the solvent, adding acetonitrile (50mL) for pulping, filtering and drying the filtrate to obtain the title compound (3-amino bicyclo [1.1.1] pentane-1-yl) methanol 21b (white solid, 890mg, yield 89.4%).
LCMS m/z(ESI)=114.10[M+1]。
The second step is that:
2-chloro-4- (3- (hydroxymethyl) bicyclo [1.1.1] pent-1-yl) aminopyrimidine-5-carboxylic acid ethyl ester (21c)
ethyl 2-chloro-4-((3-(hydroxymethyl)bicyclo[1.1.1]pentan-1-yl)amino)-pyrimi-dine-5-carboxylate
Ethyl 2, 4-dichloropyrimidine-5-carboxylate 1a (1.74g,7.86mmol), (3-aminobicyclo [1.1.1] pentan-1-yl) methanol 21b (890mg,7.86mmol) and potassium carbonate (1.08g,7.86mmol) were dissolved in acetonitrile (15mL), and the reaction mixture was reacted at room temperature for 16 hours. TLC monitored the reaction was complete, filtered and the solid was washed with a small amount of acetonitrile, the filtrates were combined and concentrated, and the crude product was purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 1:1) to give the title compound ethyl 2-chloro-4- (3- (hydroxymethyl) bicyclo [1.1.1] pent-1-yl) aminopyrimidine-5-carboxylate 21c (light yellow solid, 1.11g, 50.0% yield).
LCMS m/z(ESI)=284.10[M+1]。
The third step:
2-chloro-4- (3- (hydroxymethyl) bicyclo [1.1.1] pent-1-yl) aminopyrimidine-5-carboxylic acid (21d)
2-chloro-4-((3-(hydroxymethyl)bicyclo[1.1.1]pentan-1-yl)amino)-pyrimidine-5-carboxylate
Ethyl 2-chloro-4- (3- (hydroxymethyl) bicyclo [1.1.1] pent-1-yl) aminopyrimidine-5-carboxylate 21c (1.11g,3.73mmol) was dissolved in tetrahydrofuran/water (10mL/10mL), and lithium hydroxide monohydrate (0.47g,11.18mmol) was added, followed by reaction with stirring at room temperature for 1 h. TLC to completion, concentrated to evaporate tetrahydrofuran and then adjusted to pH 3-4 by addition of 2N HCl, a white solid precipitated, filtered and the filter cake was washed twice with water and petroleum ether/ethyl acetate (v/v ═ 10/1) to afford the title compound 2-chloro-4- (3- (hydroxymethyl) bicyclo [1.1.1] pent-1-yl) aminopyrimidine-5-carboxylic acid 21d (white solid, 534mg, 53.11% yield).
LC-MS m/z(ESI)=270.10[M+1]。
The fourth step:
2-chloro-9- (3- (hydroxymethyl) bicyclo [1.1.1] pent-1-yl) -7, 9-dihydro-8H-purin-8-one (21e)
2-chloro-9-(3-(hydroxymethyl)bicyclo[1.1.1]pentan-1-yl)-7,9-dihydro-8H-purin-8-one
21d (0.50g,1.85mmol) of 2-chloro-4- (3- (hydroxymethyl) bicyclo [1.1.1] pent-1-yl) aminopyrimidine-5-carboxylic acid was dissolved in N, N-dimethylacetamide (12mL), triethylamine (0.40mL,1.85mmol) and diphenylphosphorylazide (0.26mL,1.85mmol) were added under stirring at room temperature to react for 2 hours, and the mixture was heated to 110 ℃ and refluxed for 2.5 hours. TLC was monitored to completion of the reaction, water and ethyl acetate were added to the reaction solution to extract, and the concentrated organic layer was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 20:1) to give the title compound 2-chloro-9- (3- (hydroxymethyl) bicyclo [1.1.1] pentan-1-yl) -7, 9-dihydro-8H-purin-8-one 21e (white solid, 183mg, 37.09% yield).
1H NMR(400MHz,DMSO-d6)δ11.58(s,1H),8.10(s,1H),4.66(t,1H),3.54(d,2H),2.24(s,6H)。
LCMS m/z(ESI)=267.10[M+1]。
The fifth step:
2-chloro-9- (3- (hydroxymethyl) bicyclo [1.1.1] pent-1-yl) -7-methyl-7, 9-dihydro-8H-purin-8-one (21f)
2-chloro-9-(3-(hydroxymethyl)bicyclo[1.1.1]pentan-1-yl)-7-methyl-7,9-dihydro-8H-purin-8-one
2-chloro-9- (3- (hydroxymethyl) bicyclo [1.1.1] pent-1-yl) -7, 9-dihydro-8H-purin-8-one 21e (180mg,0.67mmol) was dissolved in N, N-dimethylformamide (4mL), and dimethyl sulfate (64. mu.L, 0.67mmol) and cesium carbonate (440mg,1.35mmol) were added with stirring at 0 ℃ and reacted for 1H. TLC was monitored to completion of the reaction, the reaction solution was slowly added dropwise to ice water and stirred, ethyl acetate was added to the mixture for extraction, and the concentrated organic layer was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 20:1) to give the title compound 2-chloro-9- (3- (hydroxymethyl) bicyclo [1.1.1] pent-1-yl) -7-methyl-7, 9-dihydro-8H-purin-8-one 21f (white solid, 80mg, 42.54% yield).
LC-MS m/z(ESI)=281.10[M+1]。
And a sixth step:
9- (3- (hydroxymethyl) bicyclo [1.1.1] pent-1-yl) -7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -7, 9-dihydro-8H-purin-8-one (Compound 21)
9-(3-(hydroxymethyl)bicyclo[1.1.1]pentan-1-yl)-7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-7,9-dihydro-8H-purin-8-one
2-chloro-9- (3- (hydroxymethyl) bicyclo [1.1.1] pent-1-yl) -7-methyl-7, 9-dihydro-8H-purin-8-one 21f (80mg,0.28mmol) was dissolved in 1, 4-dioxane (6mL), and 7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-amine (63mg,0.43mmol), cesium carbonate (139mg,0.43mmol), Brettphos-G3-Pd (26mg,0.028mmol) was added with stirring at room temperature, with nitrogen blanketing and purging, followed by heating to 110 ℃ and reflux reaction for 4H. TLC was monitored to the end of the reaction, the reaction solution was concentrated, and the residue was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 15:1) to give the title compound 9- (3- (hydroxymethyl) bicyclo [1.1.1] pent-1-yl) -7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -7, 9-dihydro-8H-purin-8-one compound 21 (white solid, 35mg, yield 31.85%).
1H NMR(400MHz,DMSO-d6)δ9.14(s,1H),8.61(s,1H),8.37(s,1H),8.09(s,1H),7.70(s,1H),4.65(t,1H),3.50(d,2H),3.26(s,3H),2.39(s,3H),2.20(s,6H)。
LC-MS m/z(ESI)=393.20[M+1]。
Example 22:
9- (4, 4-Difluorocyclohexyl) -7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -7, 9-dihydro-8H-purin-8-one (Compound 22)
9-(4,4-difluorocyclohexyl)-7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-7,9-dihydro-8H-purin-8-one
Figure BDA0002876667980000551
Figure BDA0002876667980000561
The first step is as follows:
2-chloro-4- ((4, 4-difluorocyclohexyl) amino) pyrimidine-5-carboxylic acid ethyl ester (22a)
ethyl-2-chloro-4-((4,4-difluorocyclohexyl)amino)pyrimidine-5-carboxylate
Ethyl 2, 4-dichloropyrimidine-5-carboxylate 1a (6.5g,29.41mmol) and 4, 4-difluorocyclohex-1-ylamine hydrochloride (5.0g,29.41mmol) were dissolved in acetonitrile (100mL), and potassium carbonate (10.16g,73.52mmol) was added under stirring at room temperature for 4 h. After completion of the reaction monitored by TLC, filtration was performed, the residue was washed with ethyl acetate, and the filtrate was concentrated and purified by silica gel column chromatography (n-hexane/ethyl acetate (v/v) ═ 1:1) to give the title compound ethyl 2-chloro-4- ((tetrahydro-2H-pyran-4-yl) amino) pyrimidine-5-carboxylate 22a (white solid, 8.0g, yield 85.10%).
1H NMR(400MHz,DMSO-d6)δ8.85(s,1H),8.11(d,1H),4.34-4.25(m,2H),4.21-4.10(m,1H),2.11-1.91(m,6H),1.71-1.57(m,2H),1.23(t,3H)。
LC-MS m/z(ESI)=320.10[M+l]。
The second step is that:
2-chloro-4- ((4, 4-difluorocyclohexyl) amino) pyrimidine-5-carboxylic acid (22b)
2-chloro-4-((4,4-difluorocyclohexyl)amino)pyrimidine-5-carboxylic acid
Ethyl 2-chloro-4- ((4, 4-difluorocyclohexyl) amino) pyrimidine-5-carboxylate 22a (4g, 12.47mmol) was dissolved in tetrahydrofuran/water (50ml/50ml), and lithium hydroxide (597.22mg,24.94mmol) was added and stirred at room temperature for 1 h. TLC monitored to completion, tetrahydrofuran was spun off, pH 5 adjusted with 2N hydrochloric acid, white solid precipitated, filtered, and the filter cake was washed twice with petroleum ether to collect the solid to give the title compound 2-chloro-4- ((4, 4-difluorocyclohexyl) amino) pyrimidine-5-carboxylic acid 22b (white solid, 3.4g, 91.53% yield).
1H NMR(400MHz,DMSO-d6)δ13.79(s,1H),8.60(s,1H),8.55(d,1H),4.22-4.06(m,1H),2.12-1.89(m,6H),1.72-1.56(m,2H)。
LC-MS m/z(ESI)=292.00[M+l]。
The third step:
2-chloro-9- (4, 4-difluorocyclohexyl) -7, 9-dihydro-8H-purin-8-one (22c)
2-chloro-9-(4,4-difluorocyclohexyl)-7,9-dihydro-8H-purin-8-one
2-chloro-4- ((tetrahydro-2H-pyran-4-yl) amino) pyrimidine-5-carboxylic acid 22b (3.34g,11.45mmol) was dissolved in dimethylacetamide (50mL), triethylamine (1.6mL,11.45mmol), diphenylphosphorylazide (1.22mL,11.45mmol) were added, followed by gradual warming to 120 ℃ and stirring for 1.5H. The reaction was monitored by TLC, the reaction was poured into ice water, the solid collected by filtration, washed 3 times with water, concentrated and dried in vacuo to afford the title compound 2-chloro-9- (4, 4-difluorocyclohexyl) -7, 9-dihydro-8H-purin-8-one 22c (white solid, 2.2g, 78.5% yield).
1H NMR(400MHz,DMSO-d6)δ11.66(s,1H),8.13(s,1H),4.46-4.36(m,1H),2.17-1.94(m,6H),1.87-1.80(m,2H)。
LC-MS m/z(ESI)=289.10[M+l]。
The fourth step:
2-chloro-7-methyl-9- (4, 4-difluorocyclohexyl) -7, 9-dihydro-8H-purin-8-one (22d)
2-chloro-9-(4,4-difluorocyclohexyl)-7-methyl-7,9-dihydro-8H-purin-8-one
2-chloro-9- (4, 4-difluorocyclohexyl) -7, 9-dihydro-8H-purin-8-one 22c (2.62g, 9.08mmol) was dissolved in dimethylformamide (50mL), dimethyl sulfate (1.14g, 9.08mmol) and cesium carbonate (4.44g,13.61mmol) were added at 0 deg.C, and stirring was continued for 1H at 0 deg.C. The reaction was monitored by TLC and the reaction mixture was poured into ice water to precipitate a solid, which was then filtered and collected to obtain the title compound 2-chloro-7-methyl-9- (4, 4-difluorocyclohexyl) -7, 9-dihydro-8H-purin-8-one 22d (white solid, 2.2g, 80% yield).
1H NMR(400MHz,DMSO-d6)δ8.37(s,1H),4.55-4.39(m,1H),3.33(s,3H),2.21-2.01(m,6H),1.90-1.79(m,2H)。
LC-MS m/z(ESI)=303.10[M+l]。
The fifth step:
9- (4, 4-Difluorocyclohexyl) -7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -7, 9-dihydro-8H-purin-8-one
9-(4,4-difluorocyclohexyl)-7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-7,9-dihydro-8H-purin-8-one
2-chloro-7-methyl-9- (4, 4-difluorocyclohexyl) -7, 9-dihydro-8H-purin-8-one 22d (150mg,0.50mmol) was dissolved in 1, 4-dioxane (6mL), and 7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-amine (73mg,0.50mmol), cesium carbonate (323mg,0.99mmol), methanesulfonic acid (2-dicyclohexylphosphine) -3, 6-dimethoxy-2 ',4',6 '-triisopropyl-1, 1' -biphenyl) (2 '-amino-1, 1' -biphenyl-2-yl) palladium (II) (45mg, 0.05mmol) was added with stirring at normal temperature, with nitrogen protection and aeration, then the temperature is increased to 110 ℃ and the reaction is refluxed for 4 h. TLC to the end of the reaction, the reaction was concentrated, and the residue was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 20:1) to give title compound 22 (white solid, 30mg, 15% yield).
1H NMR(600MHz,DMSO-d6)δ9.02(s,1H),8.65(s,1H),8.37(s,1H),8.08(s,1H),7.69(s,1H),4.40–4.32(m,1H),3.30(s,3H),2.36(s,3H),2.11–1.93(m,6H),1.82–1.77(m,2H)。
LC-MS m/z(ESI)=415.20[M+l]。
Example 23:
4- (7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) -8-oxo 7, 8-dihydro-9H-purin-9-ylbicyclo [2.2.2] octane-1-carbonitrile (Compound 23)
4-(7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-8-oxo-7,8-dihydro-9H-purin-9-yl)bicyclo[2.2.2]octane-1-carbonitrile
Figure BDA0002876667980000571
Figure BDA0002876667980000581
The first step is as follows:
4-carbamoyl-bicyclo [2.2.2] octane-1-carboxylic acid methyl ester (23b)
methyl 4-carbamoylbicyclo[2.2.2]octane-1-carboxylate
4- (methoxycarbonyl) bicyclo [2.2.2] octane-1-carboxylic acid 23a (10.0g, 47.12mmol) was dissolved in anhydrous dichloromethane (200mL), 2- (7-azabenzotriazole) -N, N, N ', N' -tetramethylurea hexafluorophosphate (18.81g,49.47mmol) was added under ice-bath, the temperature was maintained for reaction for 30min, N, N-diisopropylethylamine (24.62mL,141.35mmol) was added, and ammonium chloride solid (3.78g,70.67mmol) was added slowly in multiple portions. The reaction was gradually returned to room temperature and reacted overnight. After the reaction was completed by TLC, the reaction mixture was directly treated with 100mL of 0.5N hydrochloric acid solution, and the organic phase after separation was successively treated with 100mL of each of water and saturated brine, dried and concentrated to obtain the desired compound methyl 4-carbamoylbicyclo [2.2.2] octane-1-carboxylate 23b (white solid, 21g, crude product).
1H NMR(400MHz,DMSO-d6)δ6.94(s,1H),6.73(s,1H),3.57(s,3H),1.72-1.63(m,12H)。
LC-MS m/z(ESI)=212.10[M+1]。
The second step is that:
4-Cyanobicyclo [2.2.2] octane-1-carboxylic acid methyl ester (23c)
methyl 4-cyanobicyclo[2.2.2]octane-1-carboxylate
Methyl 4-carbamoylbicyclo [2.2.2] octane-1-carboxylate 23b (21g,99.40mmol) was dissolved in dichloromethane (200mL), pyridine (16.02mL,198.81mmol), trifluoroacetic anhydride (21.00mL,149.10mmol) were added in an ice bath, and the reaction was continued for 1h while maintaining the temperature. The completion of the reaction was monitored by TLC, and the reaction mixture was directly filtered, and the filter cake was washed with 100mL of methylene chloride, and the organic phases were combined, followed by 100mL each of 1N hydrochloric acid solution, water and saturated brine, drying, concentration and column chromatography to obtain the desired product methyl 4-cyanobicyclo [2.2.2] octane-1-carboxylate 23c (white solid, 5.3g,60mg, two-step yield 58.21%).
1H NMR(400MHz,CDCl3-d6)δ3.66(s,3H),1.98-1.94(m,6H),1.86-1.82(m,6H)。
LC-MS m/z(ESI)=194.10[M+1]。
The third step:
4-Cyanobicyclo [2.2.2] octane-1-carboxylic acid (23d)
4-cyanobicyclo[2.2.2]octane-1-carboxylic acid
Methyl 4-cyanobicyclo [2.2.2] octane-1-carboxylate 23c (5.3g, 27.43mmol) was dissolved in 50mL of tetrahydrofuran and 50mL of water, and lithium hydroxide (1.73g,41.14mmol) was added thereto and the mixture was stirred at room temperature for 1 hour. The reaction was monitored by TLC for completion, concentrated to remove tetrahydrofuran, adjusted to pH 5 with 2N hydrochloric acid to precipitate a white solid, filtered, and the filter cake was washed twice with petroleum ether to collect the solid to give the title compound 4-cyanobicyclo [2.2.2] octane-1-carboxylic acid 23d (white solid, 5.0g, crude).
1H NMR(400MHz,DMSO-d6)δ12.23(s,1H),1.89-1.85(m,6H),1.72-1.68(m,6H)。
LC-MS m/z(ESI)=180.10[M+1]。
The fourth step:
4-Aminobicyclo [2.2.2] octane-1-carbonitrile hydrochloride (23e)
4-aminobicyclo[2.2.2]octane-1-carbonitrile hydrochloride
4-Cyanobicyclo [2.2.2] octane-1-carboxylic acid 23d (5.0g,27.90mmol) was dissolved in toluene (60mL), diphenyl phosphorazidate (6.01mL,27.90mmol) and triethylamine (3.88mL,27.90mmol) were added in an ice bath, and the reaction mixture was stirred at room temperature for 1 hour, then warmed to 90 ℃ and reacted for 3 hours. TLC monitored to completion of the reaction, cooled to room temperature, poured slowly into 100mL of 1N hydrochloric acid solution to give a large amount of solid, filtered, collected solid, slurried with ethyl acetate (150mL), and dried in vacuo to give the title compound 4-aminobicyclo [2.2.2] octane-1-carbonitrile hydrochloride 23e (white solid, 7.3g, crude).
1H NMR(401MHz,DMSO-d6)δ8.45(s,2H),2.01–1.97(m,6H),1.81-1.76(m,6H)。
LC-MS m/z(ESI)=151.20[M+1]。
The fifth step:
2-chloro-4- ((4-cyanobicyclo [2.2.2] octan-1-yl) amino) pyrimidine-5-carboxylic acid ethyl ester (23f)
ethyl 2-chloro-4-((4-cyanobicyclo[2.2.2]octan-1-yl)amino)pyrimidine-5-carboxylate
Ethyl 2, 4-dichloropyrimidine-5-carboxylate 1a (8.74g,39.53mmol), 4-aminobicyclo [2.2.2] octane-1-carbonitrile hydrochloride 23e (7.38g,39.53mmol, 60% purity), and potassium carbonate (21.85g,158.13mmol) were dissolved in acetonitrile (200mL), and the reaction mixture was reacted at room temperature for 16 hours. TLC monitored the reaction was complete, filtered and the solid was washed with a small amount of acetonitrile, the filtrates were combined and concentrated, and the crude product was chromatographed on silica gel (petroleum ether/ethyl acetate (v/v) ═ 1:1) to afford the title compound ethyl 2-chloro-4- ((4-cyanobicyclo [2.2.2] octan-1-yl) amino) pyrimidine-5-carboxylate 23f (white solid, 4.0g, 30.23% yield).
1H NMR(400MHz,DMSO-d6)δ8.63(s,1H),8.29(s,1H),4.30(q,2H),2.10–2.00(m,12H),1.30(t,3H)。
LC-MS m/z(ESI)=335.10[M+1]。
And a sixth step:
2-chloro-4- ((4-cyanobicyclo [2.2.2] octan-1-yl) amino) pyrimidine-5-carboxylic acid (23g)
2-chloro-4-((4-cyanobicyclo[2.2.2]octan-1-yl)amino)pyrimidine-5-carboxylic acid
Ethyl 2-chloro-4- ((4-cyanobicyclo [2.2.2] octan-1-yl) amino) pyrimidine-5-carboxylate 23f (4g, 11.95mmol) was dissolved in 50mL of tetrahydrofuran and 50mL of water, and lithium hydroxide (1.01g,23.90mmol) was added thereto and the mixture was stirred at room temperature for 1 hour. The reaction was monitored by TLC for completion, concentrated to remove tetrahydrofuran, adjusted to pH 5 with 2N hydrochloric acid, white solid precipitated, filtered, the filter cake was washed twice with petroleum ether and the solid was collected to give 23g (3.6g, white solid, 98.23% yield) of the title compound 2-chloro-4- ((4-cyanobicyclo [2.2.2] octan-1-yl) amino) pyrimidine-5-carboxylic acid, which was directly subjected to the next experiment.
1H NMR(600MHz,DMSO-d6)δ13.85(s,1H),8.59(s,1H),8.56(s,1H),2.07–1.98(m,12H)。
LCMS m/z(ESI)=307.10[M+l]。
The seventh step:
4- (2-chloro-8-oxo-7, 8-dihydro-9H-purin-9-yl) bicyclo [2.2.2] octane-1-carbonitrile (23H)
4-(2-chloro-8-oxo-7,8-dihydro-9H-purin-9-yl)bicyclo[2.2.2]octane-1-carbonitrile
23g (3.8g,12.39mmol) of 2-chloro-4- ((4-cyanobicyclo [2.2.2] octan-1-yl) amino) pyrimidine-5-carboxylic acid was dissolved in dimethylacetamide (50mL), and triethylamine (1.72mL,12.39mmol) and diphenylphosphorylazide (2.67mL,12.39mmol) were added, followed by gradual warming to 120 ℃ and stirring for 1.5 hours. TLC was used to monitor the completion of the reaction, and the reaction mixture was poured into ice water, filtered to collect the solid, washed with water 3 times, concentrated and dried in vacuo to give 4- (2-chloro-8-oxo-7, 8-dihydro-9H-purin-9-yl) bicyclo [2.2.2] octane-1-carbonitrile for 23H (3.3g, white solid, 87.7% yield).
1H NMR(400MHz,DMSO-d6)δ11.61(s,1H),8.09(s,1H),2.48–2.40(m,6H),2.09–2.03(m,6H)。
LC-MS m/z(ESI)=304.20[M+l]。
Eighth step:
4- (2-chloro-7-methyl-8-oxo 7, 8-dihydro-9H-purin-9-yl) bicyclo [2.2.2] octane-1-carbonitrile (23i)
4-(2-chloro-7-methyl-8-oxo-7,8-dihydro-9H-purin-9-yl)bicyclo[2.2.2]octane-1-carbonitrile
4- (2-chloro-8-oxo-7, 8-dihydro-9H-purin-9-yl) bicyclo [2.2.2] octane-1-carbonitrile (3.3g, 11.43mmol) was dissolved in dimethylformamide (50mL) for 23H, dimethyl sulfate (1.44g, 11.43mmol) and cesium carbonate (5.59g,17.15mmol) were added at 0 ℃ and stirred at 0 ℃ for 1H. The reaction was monitored by TLC, and the reaction mixture was poured into ice water to precipitate a solid, which was then filtered to collect a solid to obtain the title compound, 4- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) bicyclo [2.2.2] octane-1-carbonitrile 23i (3.1g, white solid, 89.59% yield).
1H NMR(401MHz,DMSO-d6)δ8.33(s,1H),3.29(s,3H),2.48–2.41(m,6H),2.10–2.04(m,6H)。
LC-MS m/z(ESI)=318.20[M+l]。
The ninth step:
4- (7-methyl-2- ((7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) amino) 8-oxo 7, 8-dihydro-9H-purin-9-yl) bicyclo [2.2.2] octane-1-carbonitrile
4-(7-methyl-2-((7-methyl-[1,2,4]triazolo[1,5-a]pyridin-6-yl)amino)-8-oxo-7,8-dihydro-9H-purin-9-yl)bicyclo[2.2.2]octane-1-carbonitrile
4- (2-chloro-7-methyl-8-oxo 7, 8-dihydro-9H-purin-9-yl) bicyclo [2.2.2] octane-1-carbonitrile 23i (150mg,0.47mmol) was dissolved in 1, 4-dioxane (10mL), and 7-methyl- [1,2,4] triazolo [1,5-a ] pyridin-6-amine (70mg,0.47mmol), cesium carbonate (307mg,0.94mmol), methanesulfonic acid (2-dicyclohexylphosphine) -3, 6-dimethoxy-2 ',4',6 '-triisopropyl-1, 1' -biphenyl) (2 '-amino-1, 1' -biphenyl-2-yl) palladium (II) (43mg, 0.047mmol), nitrogen blanket and aeration, followed by heating to 110 ℃ and reflux for 4 h. TLC to the end of the reaction, the reaction was concentrated, and the residue was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 20:1) to give title compound 23 (white solid, 60mg, 29.60% yield).
1H NMR(401MHz,DMSO-d6)δ9.13(s,1H),8.61(s,1H),8.39(s,1H),8.09(s,1H),7.71(s,1H),3.24(s,3H),2.42-2.46(m,6H),2.39(s,3H),1.98-2.02(m,6H)。
LC-MS m/z(ESI)=430.20[M+l]。
Biological assay
1. DNA-PK kinase inhibition assay
The inhibitory activity of the compound against DNA-PK kinase was examined by using DNA-PK kinase assay kit (purchased from Promega, cat # V4107, lot # 0000366495). The results were quantified using chemiluminescence as follows:
i. constructing ADP-fluorescence standard curves with different concentrations according to the kit instructions;
preparing 5. mu.L reaction in 384-well white plates, adding 1. mu.L of compound (concentration gradient set at 1. mu.M, 200nM, 40nM, 8nM, 1.6nM, 0.32nM, 0.064nM, 0.013nM, respectively), 20units of DNA-PK kinase, 0.2. mu.g/. mu.L substrate, 10. mu.g/. mu.L DNA, 50. mu.M ATP, 1% DMSO, respectively, to each well;
mixing, centrifuging (1000rpm, 30s), and incubating at 37 ℃ for 60 min;
add 5. mu.L ADP-GloTMReagent stops reaction, mixes evenly, centrifuges (1000rpm, 30s) and incubate for 40min at room temperature;
v. adding 10 mu LKING ase Detection Reagent, shaking and mixing evenly, centrifuging (1000rpm, 30s), and incubating for 30min at room temperature;
fluorescence was measured using a microplate reader (Thermo fisher, Varioskan LUX). IC Using GraphPad Pris m 850The results of the calculation of (2) are shown in Table 1.
TABLE 1 DNA-PK kinase inhibitory Activity of the Compounds of the invention
Compound numbering IC50(nM)
1 C
2 B
3 B
4 B
5 C
6-1 B
6-2 B
7-1 A
7-2 A
8 A
9 A
11 C
12 B
13 B
14 B
15 C
16 B
17 B
18 B
19 B
20 B
21 C
22 B
23 B
Comparative example D
Note: the control example is reference J.Med.chem (2020),63(7), 3461-.
0<A≤1nM;
1nM<B≤5nM;
5nM<C≤50nM;
50nM<D≤100nM;
100nM<E。
And (4) conclusion: the results show that the compound has more remarkable inhibition effect on DNA-PK kinase compared with the control example.
2. Proliferation inhibition assay
2.1 Experimental materials
A549 cells (purchased from ATCC), Doxorubicin hydrochloride (Doxorubicin, purchased from Shanghai ceramic Biotechnology Co., Ltd., product No. T1020), DMEM (10% FBS) medium, 96-well white plate, cell viability assay kit (purchased from Promega, product No. G9241).
2.2 Experimental procedures
Counting after resuspending A549 cells, and laying 96-well plates at 500 cells/well (80 μ L); the culture was carried out overnight.
Configure 10 × drug concentration gradient (1: 5): initial concentration was 100 μ M, with medium 1: the 5-fold dilution was 20. mu.M, 4. mu.M, 800nM, 160nM, 32nM, 6.4nM, 1.3nM (final concentration 10. mu.M, 2. mu.M, 0.4. mu.M, 80nM, 16nM, 3.2nM, 0.64nM, 0.13 nM).
3. Adding 10 mu L of the compound to be detected into each well, repeating each concentration gradient for 2 times, and setting a single medicine group (the compound to be detected or Doxorubicin), a combined group and a culture medium control group:
a single drug group: adding the test compound (final concentration 10. mu.M, 2. mu.M, 0.4. mu.M, 80nM, 16nM, 3.2nM, 0.64nM, 0.13nM) or only doxorubicin (final concentration 10. mu.M, 2. mu.M, 0.4. mu.M, 80nM, 16nM, 3.2nM, 0.64nM, 0.13nM), and further culturing at 37 ℃ in an incubator for 120 h;
combination group: adding test compound (final concentration 10 μ M,2 μ M, 0.4 μ M, 80nM, 16nM, 3.2nM, 0.64nM, 0.13nM), incubating at 37 deg.C for 1h, adding 10nM Doxorubicin (10 μ L/well) to each well, and further culturing at 37 deg.C for 120 h;
control group: add 20. mu.L of medium and incubate at 37 ℃ for 120 h.
4. The cell culture plate was removed, and an equal volume of the test solution (100. mu.L of the medium, 100. mu.L of the test solution) was added to the cell culture plate.
5. Shaking the plate shaking machine for 2min to lyse the cells.
6. The mixture was left at room temperature for 10min to stabilize the signal.
7. Chemiluminescence was measured with a microplate reader (Thermo fisher; Varioskan LUX).
And (4) conclusion: the compound disclosed by the invention and Doxorubicin are combined to have a more obvious proliferation inhibition effect on A549 cells, and the cytotoxicity is reduced.
3. Experiment for inhibiting transplanted tumor
3.1 Experimental materials: a549 cells (purchased from ATCC); doxorubicin liposome (Dox) (Lipo Doxorubicin, trade name "Libaoduo", available from Shanghai Compound Dangjiang biomedical corporation); female nude mice (weight 18-20g) at 6 weeks of age (Beijing Wittingle laboratory animal technology Co., Ltd.) were treated with 10 mice each.
3.2 determination of the inhibition effect of Doxorubicin on A549 transplantable tumors in combination with the compound to be screened:
3.2.1 collecting A549 cells in a logarithmic phase of growth, washing for 2 times by precooling PBS for later use;
3.2.2Balb/c nude mouse laboratory acclimation for 3 days, inoculating A549 cells subcutaneously in right costal region, the inoculation cell amount is 5 × 106Only, when the tumor grows to 200mm3Performing a drug effect experiment on the left and right;
3.2.3 mice with successful tumor growth are randomly grouped, and a single Doxorubicin (Dox) group, a compound to be tested, a Dox combination group and a control group (Vehicle) are arranged and are administrated for 21 days; mice were gavaged (i.g.) 2 times daily (BID, 5mL/kg dosing volume; solvent 5% DMSO + 30% 2-hydroxypropyl-. beta. -cyclodextrin). After 1h of intragastric administration in the morning, Lipo Doxorubicin (2.5mg/kg) was injected via the tail vein; 1 time per week (QW, administration volume 5 mL/kg;);
3.2.4 mice were weighed 2 times per week and tumor volumes were determined simultaneously: tumor volume (V) was calculated as: v1/2 xlLong and long×LShort length 2And calculating a tumor inhibition rate (%) - (D21 tumor volume (Vehicle) -D21 tumor volume (administration group))/D21 tumor volume (Vehicle) × 100;
3.2.5 the tumors were isolated and weighed 21 days after administration, and the rate of change in body weight (%) (D21 body weight-D0 body weight)/D0 body weight × 100 was calculated.
And (4) conclusion: experimental results show that the compound and the Doxorubicin are combined to remarkably improve the tumor inhibition effect of the Doxorubicin, and obvious weight loss is not caused.
While the present invention has been described in detail with respect to the specific embodiments thereof, it will be understood by those skilled in the art that the above embodiments are illustrative and not to be construed as limiting the present invention, and that various changes and modifications can be made therein by those skilled in the art without departing from the spirit of the invention and within the scope of the appended claims.

Claims (8)

1. A compound of formula (I), or a stereoisomer, solvate, prodrug, deuterode, metabolite, pharmaceutically acceptable salt or co-crystal thereof:
Figure FDA0002876667970000011
wherein:
R1selected from H, -OH, cyano, halogen, -NH2、C1-6Alkyl or C1-6Alkoxy radical, said C1-6Alkyl is optionally further substituted with 1-3 substituents selected from D or halogen;
R2selected from H or C1-6An alkyl group;
R3is selected from C1-6Alkyl radical, C3-12Carbocyclyl, C3Heterocyclic group, C4-12Heterocyclyl radical, -C1-6alkylene-C3-12Carbocyclyl, -C1-6alkylene-C3Heterocyclyl or-C1-6alkylene-C4-12Heterocyclic group, said C3Heterocyclyl and C4-12Heterocyclyl contains 1 to 3 heteroatoms selected from N, O or S, said C1-6Alkyl radical, C3-12Carbocyclyl, C3Heterocyclyl and C4-12Heterocyclic group, C1-6Alkylene is optionally further substituted by 1 or more groups selected from-OH, carboxyl, halogen, cyano, ═ O, C1-6Alkyl radical, C1-6Heteroalkyl group, C2-6Alkenyl radical, C2-6Alkynyl, -NRa1Ra2、-C(=O)OC1-6Alkyl, -C (═ O) NRa1Ra2、C3-12Cycloalkyl radical, C3Heterocycloalkyl radical, C4-12Heterocycloalkyl radical, C6-12Aryl or C5-12Heteroaryl, or a substituted heteroaryl; and said C in said substituent1-6Alkyl radical, C1-6Heteroalkyl group, C2-6Alkenyl or C2-6Alkynyl is optionally further substituted by 1 or more groups selected from-OH, carboxy, cyano, halo, -O-Ra1、-NRa1Ra2、C3-12Cycloalkyl radical, C3Heterocycloalkyl radical, C4-12Heterocycloalkyl radical, C6-12Aryl or C5-12Heteroaryl, or a substituted heteroaryl;
with the following conditions: when R is3When cyclohexyl, the cyclohexyl is substituted by at least one cyano group; and R is3Is not tetrahydrofuranyl or oxacyclohexyl;
Ra1、Ra2each independently selected from H, C1-6Alkyl, -C (═ O) Ra3or-C (═ O) NRa4Ra5Wherein said C1-6The alkyl is optionally further substituted by 1 or more groups selected from OH, halogen, C1-6Alkyl radical, C1-6Alkoxy radical, C6-12Aryl radical, C5-12Heteroaryl group, C3-12Cycloalkyl radical, C3Heterocycloalkyl, or C4-12Substituted with a substituent of heterocycloalkyl;
Ra3is selected from C1-6Alkyl radical, C1-6Alkoxy or C6-12An aryl group;
Ra4、Ra5each independently selected from H or C1-6An alkyl group; or Ra4And Ra5And the N atom forms a 3-to 8-membered heterocyclic ring, said 3-to 8-membered heterocyclic ring comprising 1 to 4 heteroatoms selected from N, O or S;
p is selected from 0, 1,2 or 3;
optionally, formula (I) is substituted with 1 or more D atoms.
2. The compound of claim 1, or a stereoisomer, solvate, prodrug, deuterode, metabolite, pharmaceutically acceptable salt, or co-crystal thereof, wherein the compound is selected from compounds represented by the general formula (II):
Figure FDA0002876667970000012
wherein:
R1selected from halogen or C1-6Alkyl radical, said C1-6Alkyl is optionally further substituted with 1-3 substituents selected from D or halogen;
R3is selected from C1-6Alkyl radical, C3-12Carbocyclyl, C3Heterocyclic group, C4-12Heterocyclyl radical, -C1-6alkylene-C3-12Carbocyclyl, -C1-6alkylene-C3Heterocyclyl or-C1-6alkylene-C4-12Heterocyclic group, said C3Heterocyclyl and C4-12Heterocyclyl contains 1 to 3 heteroatoms selected from N, O or S, said C1-6Alkyl radical, C3-12Carbocyclyl, C3Heterocyclyl and C4-12Heterocyclic group, C1-6Alkylene is optionally further substituted by 1 or more groups selected from-OH, carboxyl, halogen, cyano, ═ O, C1-6Alkyl radical, C1-6Heteroalkyl group, C2-6Alkenyl radical, C2-6Alkynyl, -NRa1Ra2、-C(=O)OC1-6Alkyl, -C (═ O) NRa1Ra2、C3-12Cycloalkyl radical, C3Heterocycloalkyl radical, C4-12Heterocycloalkyl radical, C6-12Aryl or C5-12Heteroaryl, or a substituted heteroaryl; and said C in said substituent1-6Alkyl radical, C1-6Heteroalkyl group, C2-6Alkenyl or C2-6Alkynyl is optionally further substituted by 1 or more groups selected from-OH, carboxy, cyano, halo, -O-Ra1、-NRa1Ra2、C3-12Cycloalkyl radical, C3Heterocycloalkyl radical, C4-12Heterocycloalkyl radical, C6-12Aryl or C5-12Heteroaryl, or a substituted heteroaryl;
with the following conditions: when R is3When cyclohexyl, the cyclohexyl is substituted by at least one cyano group; and R is3Is not tetrahydrofuranyl or oxacyclohexyl;
Ra1、Ra2each independently selected from H, C1-6Alkyl, -C (═ O) Ra3or-C (═ O) NRa4Ra5Wherein said C1-6The alkyl is optionally further substituted by 1 or more groups selected from OH, halogen, C1-6Alkyl radical, C1-6Alkoxy radical, C6-12Aryl radical, C5-12Heteroaryl group, C3-12Cycloalkyl radical, C3Heterocycloalkyl or C4-12Substituted with a substituent of heterocycloalkyl;
Ra3is selected from C1-6Alkyl radical, C1-6Alkoxy or C6-12An aryl group;
Ra4、Ra5each independently selected from H or C1-6An alkyl group; or Ra4And Ra5And the N atom forms a 3-to 8-membered heterocyclic ring, said 3-to 8-membered heterocyclic ring comprising 1 to 4 heteroatoms selected from N, O or S;
p is selected from 0, 1,2 or 3.
3. The compound of claim 2, or a stereoisomer, solvate, prodrug, deuterode, metabolite, pharmaceutically acceptable salt, or co-crystal thereof, wherein:
R1selected from halogen or C1-4Alkyl radical, said C1-6Alkyl is optionally further substituted with 1-3 substituents selected from D or halogen;
R3is selected from C1-4Alkyl radical, C3-8Carbocyclyl, C4-8Heterocyclyl radical, -C1-2alkylene-C3-8Carbocyclic group or-C1-2alkylene-C4-8Heterocyclic group, said C4-8Heterocyclyl contains 1 to 3 heteroatoms selected from N or O, said C1-4Alkyl radical, C3-8Carbocyclyl and C4-8Heterocyclic group, C1-2Alkylene is optionally further substituted by 1 or more groups selected from-OH, halogen, ═ O, cyano, or C1-4Alkyl is substituted by a substituent; and said C in said substituent1-4Alkyl is optionally further substituted with 1 or more substituents selected from-OH, cyano, or halogen; with the following conditions: when R is3When cyclohexyl, the cyclohexyl is substituted by at least one cyano group; and R is3Is not tetrahydrofuranyl or oxacyclohexyl;
p is selected from 1.
4. The compound according to any one of claims 1-3, or a stereoisomer, solvate, prodrug, deuteride, metabolite, pharmaceutically acceptable salt or co-crystal thereof, wherein the compound is selected from one of the following structures:
Figure FDA0002876667970000021
Figure FDA0002876667970000031
Figure FDA0002876667970000032
or
Figure FDA0002876667970000033
5. An intermediate for the preparation of a compound according to any one of claims 1 to 4, said intermediate being selected from the group consisting of:
Figure FDA0002876667970000034
Figure FDA0002876667970000041
Figure FDA0002876667970000042
or
Figure FDA0002876667970000043
6. A pharmaceutical composition comprising:
(1) a compound of any one of claims 1 to 5 or a stereoisomer, solvate, prodrug, deutero-ent, metabolite, pharmaceutically acceptable salt or co-crystal thereof;
(2) optionally one or more other active ingredients; and
(3) a pharmaceutically acceptable carrier and/or excipient.
7. Use of a compound of any one of claims 1-4, or a stereoisomer, solvate, prodrug, deutero-compound, metabolite, pharmaceutically acceptable salt or co-crystal thereof, or a pharmaceutical composition of claim 6, in the manufacture of a medicament for the treatment of cancer.
8. Use of a compound of any one of claims 1-4, or a stereoisomer, solvate, prodrug, deutero-compound, metabolite, pharmaceutically acceptable salt or co-crystal thereof, or a pharmaceutical composition of claim 6, in the preparation of a DNA-PK inhibitor.
CN202011615924.4A 2019-12-31 2020-12-30 Purine derivatives and their use in medicine Pending CN113121573A (en)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
CN201911423498 2019-12-31
CN2019114234981 2019-12-31
CN202010165176 2020-03-11
CN2020101651768 2020-03-11
CN2020106788678 2020-07-15
CN202010678867 2020-07-15

Publications (1)

Publication Number Publication Date
CN113121573A true CN113121573A (en) 2021-07-16

Family

ID=76686510

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202011615924.4A Pending CN113121573A (en) 2019-12-31 2020-12-30 Purine derivatives and their use in medicine

Country Status (2)

Country Link
CN (1) CN113121573A (en)
WO (1) WO2021136461A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
CN114249753A (en) * 2020-09-22 2022-03-29 山东轩竹医药科技有限公司 Triazolopyridine kinase inhibitors

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TW202229281A (en) * 2020-12-24 2022-08-01 大陸商江蘇恆瑞醫藥股份有限公司 Purinone compounds, their preparation methods and medical use

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140275024A1 (en) * 2013-03-12 2014-09-18 Vertex Pharmaceuticals Incorporated Dna-pk inhibitors
CN105566290A (en) * 2014-10-30 2016-05-11 康朴生物医药技术(上海)有限公司 Isoindoline derivative and intermediate, preparation method, pharmaceutical composition and application thereof
CN110177791A (en) * 2016-12-20 2019-08-27 阿斯利康(瑞典)有限公司 Amino-triazolopyridine compound and its purposes in treating cancer
WO2019201283A1 (en) * 2018-04-20 2019-10-24 Xrad Therapeutics, Inc. Dual atm and dna-pk inhibitors for use in anti-tumor therapy
WO2019238929A1 (en) * 2018-06-15 2019-12-19 Astrazeneca Ab Purinone compounds and their use in treating cancer
CN114573605A (en) * 2020-12-01 2022-06-03 武汉光谷亚太医药研究院有限公司 DNA-dependent protein kinase inhibitors and uses thereof
CN114728969A (en) * 2019-11-25 2022-07-08 南京明德新药研发有限公司 Pyrimido imidazoles as DNA-PK inhibitors

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140275024A1 (en) * 2013-03-12 2014-09-18 Vertex Pharmaceuticals Incorporated Dna-pk inhibitors
CN105566290A (en) * 2014-10-30 2016-05-11 康朴生物医药技术(上海)有限公司 Isoindoline derivative and intermediate, preparation method, pharmaceutical composition and application thereof
CN110177791A (en) * 2016-12-20 2019-08-27 阿斯利康(瑞典)有限公司 Amino-triazolopyridine compound and its purposes in treating cancer
WO2019201283A1 (en) * 2018-04-20 2019-10-24 Xrad Therapeutics, Inc. Dual atm and dna-pk inhibitors for use in anti-tumor therapy
WO2019238929A1 (en) * 2018-06-15 2019-12-19 Astrazeneca Ab Purinone compounds and their use in treating cancer
CN114728969A (en) * 2019-11-25 2022-07-08 南京明德新药研发有限公司 Pyrimido imidazoles as DNA-PK inhibitors
CN114573605A (en) * 2020-12-01 2022-06-03 武汉光谷亚太医药研究院有限公司 DNA-dependent protein kinase inhibitors and uses thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
JACQUELINE H.L. FOK,等: "AZD7648 is a potent and selective DNA-PK inhibitor that enhances radiation, chemotherapy and olaparib activity", 《NATURE COMMUNICATIONS》 *
庄亮,等: "DNA-PK作为放化疗增敏靶点的研究进展", 《中华肿瘤防治杂志》 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
CN114249753A (en) * 2020-09-22 2022-03-29 山东轩竹医药科技有限公司 Triazolopyridine kinase inhibitors

Also Published As

Publication number Publication date
WO2021136461A1 (en) 2021-07-08

Similar Documents

Publication Publication Date Title
CN109715634B (en) Fused bicyclic inhibitors of MENIN-MLL interaction
CN113121573A (en) Purine derivatives and their use in medicine
CN113121538B (en) Furan derivative and application thereof in medicine
CN112513029A (en) Nitrogen-containing aryl phosphorus oxide derivative, preparation method and application thereof
CN113795483A (en) Carboxamide-pyrimidine derivatives as SHP2 antagonists
EP3444250A1 (en) Fgfr4 inhibitor, preparation method therefor, and applications thereof
CN113121574A (en) Purine derivatives and their use in medicine
CN114315834A (en) Imidazolidinone derivatives and their use in medicaments
KR20120105468A (en) N-9-substituted purine compounds, compositions and methods of use
EP3145512A1 (en) Substituted ethynyl heterobicyclic compounds as tyrosine kinase inhibitors
EP3498707A1 (en) Fgfr4 inhibitor and preparation method and use thereof
WO2022165513A1 (en) Cdk2 inhibitors and methods of using the same
EP4269402A1 (en) Aromatic heterocyclic compound, pharmaceutical composition and use thereof
CN117295736A (en) Benzo [ C ] [2,6] naphthyridine derivatives, compositions thereof and therapeutic uses thereof
CN113387962A (en) Pyrazolo [3,4-d ] pyrimidine-3-one derivative, pharmaceutical composition and application thereof
KR20210129607A (en) 3-azabicycloalkyl derivatives and pharmaceutical composition comprising the same
CN115477640A (en) Pyridazinones as PARP7 inhibitors
CN113825757B (en) Substituted fused bicyclic derivatives, preparation method thereof and application thereof in medicine
CN112778183A (en) Nitrogen-containing ring derivative regulator, preparation method and application thereof
EP4289835A1 (en) Cdk inhibitor
JP2019530740A (en) Pyridine and 5-membered aromatic ring compounds, their production and use
CN114728938B (en) Tetrahydroisoquinoline spiro compounds as PRMT5 inhibitors
EP4308569A1 (en) Selective modulators of ataxia telangiectasia mutated (atm) kinase and uses thereof
TW202225163A (en) Aromatic heterocyclic compound, and pharmaceutical composition and application thereof
US20240034731A1 (en) Aza-quinazoline compounds and methods of use

Legal Events

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

Ref country code: HK

Ref legal event code: DE

Ref document number: 40046824

Country of ref document: HK

RJ01 Rejection of invention patent application after publication

Application publication date: 20210716

RJ01 Rejection of invention patent application after publication