CN113087718B - Thienopyrimidinone compounds and medical application thereof - Google Patents

Thienopyrimidinone compounds and medical application thereof Download PDF

Info

Publication number
CN113087718B
CN113087718B CN202010022368.3A CN202010022368A CN113087718B CN 113087718 B CN113087718 B CN 113087718B CN 202010022368 A CN202010022368 A CN 202010022368A CN 113087718 B CN113087718 B CN 113087718B
Authority
CN
China
Prior art keywords
compound
alkyl
pharmaceutically acceptable
methyl
mmol
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN202010022368.3A
Other languages
Chinese (zh)
Other versions
CN113087718A (en
Inventor
陈寿军
宋帅
蒋小玲
田强
宋宏梅
薛彤彤
王晶翼
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Sichuan Kelun Biotech Biopharmaceutical Co Ltd
Original Assignee
Sichuan Kelun Biotech Biopharmaceutical Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Sichuan Kelun Biotech Biopharmaceutical Co Ltd filed Critical Sichuan Kelun Biotech Biopharmaceutical Co Ltd
Priority to CN202010022368.3A priority Critical patent/CN113087718B/en
Publication of CN113087718A publication Critical patent/CN113087718A/en
Application granted granted Critical
Publication of CN113087718B publication Critical patent/CN113087718B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis

Abstract

The invention relates to thienopyrimidinone compounds and medical application thereof. The invention belongs to the field of medicines, and particularly relates to a compound shown in a formula I, pharmaceutically acceptable salts, esters, solvates, stereoisomers, tautomers, prodrugs, any crystal forms, metabolites or mixtures thereof. The invention also relates to medical application of the compound shown in the formula I, pharmaceutically acceptable salts, esters, solvates, stereoisomers, tautomers, prodrugs, any crystal forms, metabolites or mixtures thereof. The compounds of the present invention are useful for inhibiting the activity of deubiquitinase USP7 and for preventing or treating diseases or disorders associated with USP7 modulation (e.g. cancer).

Description

Thienopyrimidinone compounds and medical application thereof
Technical Field
The invention belongs to the field of medicines, and particularly relates to thienopyrimidinone compounds and medical application of the compounds.
Background
Ubiquitin-protease system (UPS) is a basic physiological regulatory process in cells, and proteins are degraded by proteases after being ubiquitinated and modified through a series of cascade reactions. Abnormalities in UPS are closely related to diseases such as tumors, neurodegenerative diseases, viral infections, and the like. Drugs are currently developed mainly for five classes of targets, protease, E1 activating enzyme, E2 binding enzyme, E3 ligase, deubiquitinases (DUBs) in UPS systems.
Deubiquitinase is capable of specifically cleaving an isopeptide bond formed between a glycine residue at the carbon terminus of ubiquitin and a target protein, allowing ubiquitin to be detached from the target protein, so that the target protein is protected from degradation, relocation, activation, or the like.
Currently there are nearly 100 DUBs in humans, where ubiquitin-specific proteases (ubiquitins-specific proteases, USPs) are the largest family members of DUBs, including about 85 members, an isopeptidase belonging to the cysteine protease family [1] . It has now been found that over 40 members of the USPs family have been associated with the development and progression of tumors.
USP7 is located in the nucleus and is a key deubiquitinase in UPS, and can specifically cut off the isopeptide bond formed between ubiquitin carbon terminal and target protein, so that ubiquitin can be obtainedDetachment of target protein so that target protein is prevented from degradation, repositioning or activation, etc [2]
USP7 plays an important role in the wide distribution of human tissues, in neural development, cell cycle regulation, epigenetic regulation, DNA damage repair, and immune response. Studies have shown that USP7 is overexpressed in hepatocellular carcinoma, multiple myeloma, colon cancer, lung cancer, prostate cancer, bladder cancer, and the like, and that this overexpression is directly related to tumor invasion and poor prognosis [3]
USP7 has a rich substrate variety and is mostly protein related to cell cycle regulation, immune response, apoptosis, DNA damage repair, etc., such as MDM2, p53, ERCC6, foxp3, PTEN, FOXO4, etc [4] . MDM2 is overexpressed in some tumor cells, USP7 protects MDM2 from ubiquitination, and MDM2 promotes ubiquitination and degradation of p53 protein after binding to p53 protein, promoting tumor growth.
USP7 may also exert a tumorigenic effect by directly modulating the expression of tumor suppressor proteins (e.g., p53, PTEN, FOXO4, p114ARF, p16INK 4) and tumor promoter proteins (e.g., N-MYC, REST), up-regulating the expression of tumor-associated factors (e.g., HIF-1), and modulating tumor-associated signaling pathways (e.g., SHH signaling pathway, wnt/β -catenin signaling pathway, androgen receptor signaling pathway, DNA damage repair signaling pathway) [5]
In addition, USP7 also plays a role in tumor immunity monitoring escape by regulating Treg cell upstream signal molecules (such as transcription factor FOXP3 and epigenetic regulatory factor Tip 60), up-regulating Treg cell activity and inhibiting Teff cell (CD8+T cell) activity [6]
Development of inhibitors of USP7 is one of the hot spots in the field of tumor research. Currently, no drug is marketed worldwide against the USP7 target, and all compounds under investigation are in preclinical research.
Although the companies of hybrid SA, forma Therapeutics, inc., les Laboratoires Servier, almac Discovery Limited have all made corresponding researches on USP7 inhibitors and related patent publications, there is still a need in the art for new USP7 inhibitors, particularly USP7 inhibitors having high activity and other excellent properties.
Disclosure of Invention
Through extensive research, we have surprisingly found that a thienopyrimidinone compound, which has a strong inhibitory effect on the activity of deubiquitinase USP7, can be used for preventing or treating diseases or disorders associated with USP7 modulation (e.g. cancer). On the basis, the invention also provides medical application of the thienopyrimidinone compound.
A first aspect of the invention relates to a compound of formula I or a pharmaceutically acceptable salt, ester, solvate (e.g., hydrate), stereoisomer, tautomer, prodrug, any crystal form, metabolite, or mixture thereof:
wherein,
R 1 selected from hydrogen, C 1-6 Alkyl, C 6-10 Aryl, 5-10 membered heteroaryl,Halogen and cyano, wherein the C 1-6 Alkyl, C 6-10 Aryl and 5-10 membered heteroaryl are each independently optionally substituted with one or more of the following: hydrogen, halogen, cyano, -OR a 、-NR b R c 、-C 1-6 alkylene-NR b R c 、C 3-8 Cycloalkyl, 3-8 membered heterocycloalkyl, optionally halogenated C 1-6 Alkyl, -C (O) R 6 、-COOR 7 、-C(O)NR b R c 、-S(O) q R 8 、-S(O) q NR b R c 、-O-C 2-6 alkylene-NR b R c 、-NR a -C 2-6 alkylene-NR b R c
R 2 Selected from hydrogen, halogen, cyano, optionally halogenated C 1-6 Alkyl, C 3-8 Cycloalkyl;
R 3 selected from-C (O) R 9 、-COOR 10 、-C(O)NR e R f 、-S(O) q R 11 and-S (O) q NR e R f
The A ring and the B ring are independently selected from C 3-8 Cycloalkyl, C 3-8 Cycloalkenyl, 3-8 membered heterocycloalkyl, 3-8 membered heterocycloalkenyl, C 6-10 Aryl and 5-10 membered heteroaryl;
wherein R R groups are the same or different, m R groups 4 The groups are the same group or different groups;
r is selected from hydrogen, optionally halogenated C 1-6 Alkyl, halogen and cyano;
R 4 selected from hydrogen, oxo, C 1-6 Alkyl, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocycloalkyl, C 6-10 Aryl, 5-10 membered heteroaryl, -OR a 、-NR b R c Halogen, cyano, -C (O) R 6 、-COOR 7 、-C(O)NR b R c 、-O-C 2-6 alkylene-NR b R c and-NR d -C 2-6 alkylene-NR b R c Wherein the C 1-6 Alkyl, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocycloalkyl, C 6-10 Aryl and 5-10 membered heteroaryl are each independently optionally substituted with one or more of the following: hydrogen, halogen, cyano, C 1-6 Alkyl, hydroxy, amino, -N (C) 1-6 Alkyl group 2 、-NH(C 1-6 An alkyl group);
R a 、R b 、R c and R is d Each independently selected from hydrogen, C 1-6 Alkyl, C 3-8 Cycloalkyl, 3-8 membered heterocycloalkyl, C 6-10 Aryl, 5-10 membered heteroaryl, -C (O) R 6 and-S (O) q R 8 Wherein the C 1-6 Alkyl, C 3-8 Cycloalkyl, 3-8 membered heterocycloalkyl, C 6-10 Aryl and 5-10 membered heteroaryl are each independently optionally substituted with one or more of the following: hydrogen, halogen, hydroxy, amino, cyano, optionally halogenated C 1-6 Alkyl, C 3-8 Cycloalkyl; alternatively, R b 、R c Forms a 3-6 membered heterocyclic group with the nitrogen atom to which they are attached;
R 6 、R 7 and R is 8 Each independently selected from hydrogen, C 1-6 Alkyl and C 3-8 Cycloalkyl, wherein, the C 1-6 Alkyl and C 3-8 Cycloalkyl groups are each independently optionally substituted with one or more of the following groups: hydrogen, halogen, cyano, amino, hydroxy;
R 9 、R 10 、R 11 、R e and R is f Each independently selected from hydrogen, C 1-6 Alkyl, C 2-6 Alkenyl, C 3-8 Cycloalkyl, 3-8 membered heterocycloalkyl, C 6-10 Aryl, 5-10 membered heteroaryl, or R e 、R f Forms a 3-6 membered heterocyclic group with the nitrogen atom to which they are attached; wherein the C 1-6 Alkyl, C 2-6 Alkenyl, C 3-8 Cycloalkyl, 3-8 membered heterocycloalkyl, C 6-10 Aryl, 5-10 membered heteroaryl, and 3-6 membered heterocycle are each independently optionally substituted with one or more substituents selected from the group consisting of: hydrogen, cyano, halogen, -OH, C 1-6 Alkoxy, -NR g R h 、C 1-6 Alkyl, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocycloalkyl, C 6-10 Aryl, 5-10 membered heteroaryl, and C in the substituents 1-6 Alkyl, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocycloalkyl, C 6-10 Aryl and 5-10 membered heteroaryl are each independently optionally substituted with one or more of the following: hydrogen, halogen, cyano, optionally halogenated C 1-6 Alkyl, C 6-10 Aryl, 5-10 membered heteroaryl;
R g 、R h each independently selected from hydrogen, C 1-6 Alkyl, C 2-6 Alkenyl, C 3-8 Cycloalkyl, 3-8 membered heterocycloalkyl;
m is selected from 0, 1, 2, 3, 4, 5, 6, 7 and 8;
q is selected from 1 and 2;
r is selected from 0, 1, 2 and 3.
In the present invention, in the case of the formula I,the representation is: the group on the A ring can be substituted by R R groups, and the R R groups can take the same group or different groups; the radicals on the B ring being able to be replaced by m R 4 Substituted by radicals, m R 4 The groups may take the same group or different groups.
In some embodiments of the first aspect of the invention, R 1 Selected from C 1-6 Alkyl, C 6-10 Aryl groupWherein the C 1-6 Alkyl and C 6-10 Aryl groups are each independently optionally substituted with one or more substituents selected from the group consisting of: hydrogen, halogen, cyano, -OR a 、-NR b R c 、-C 1-6 alkylene-NR b R c
In some embodiments of the first aspect of the invention, R 1 Selected from C 1-6 Alkyl, C 6-10 Aryl groupWherein the C 1-6 Alkyl and C 6-10 Aryl groups are each independently optionally substituted with one or more of the following groups: hydrogen, halogen, -OR a 、-C 1-6 alkylene-NR b R c
The A ring and the B ring are independently selected from C 6-10 Aryl, 5-10 membered heteroaryl, 3-8 membered heterocyclenyl, 3-8 membered heterocycloalkyl, C 3-8 Cycloalkenyl and C 3-8 Cycloalkyl;
r is selected from hydrogen, optionally halogenated C 1-6 Alkyl, halogen and cyano (preferably, R is selected from hydrogen and halogen);
r is selected from 0, 1 and 2;
R 4 selected from hydrogen, -NR b R c 、C 1-6 Alkyl and-OR a The method comprises the steps of carrying out a first treatment on the surface of the m is selected from 0, 1 and 2;
R a 、R b and R is c Each independently selected from hydrogen and C 1-6 An alkyl group.
In some embodiments of the first aspect of the invention, R 1 Selected from C 1-6 Alkyl, C 6-10 Aryl groupWherein the C 1-6 Alkyl and C 6-10 Aryl groups are each independently optionally substituted with one or more of the following groups: hydrogen, hydroxy, NH 2 -C 1-6 Alkylene-, halogen, amino, cyano;
the A ring and the B ring are independently selected from C 3-8 Cycloalkyl, C 3-8 Cycloalkenyl, 3-8 membered heterocycloalkyl, 3-8 membered heterocycloalkenyl, C 6-10 Aryl and 5-10 membered heteroaryl;
r is selected from hydrogen, optionally halogenated C 1-6 Alkyl, halogen and cyano;
R 4 selected from hydrogen, amino, C 1-6 alkyl-NH- (C) 1-6 Alkyl group 2 N-, optionally halogenated C 1-6 Alkyl, oxo, C 3-8 Cycloalkyl, hydroxy and cyano;
m is selected from 0, 1, 2 and 3;
r is selected from 0, 1 and 2.
In some embodiments of the first aspect of the invention, R 1 Selected from C 1-6 Alkyl, C 6-10 Aryl groupWherein the C 6-10 Aryl is optionally substituted with one or more of the following: hydrogen, hydroxy, NH 2 -C 1-6 Alkylene-, halogen, amino, cyano;
the A ring and the B ring are independently selected from C 6-10 Aryl, 5-10 membered heteroaryl, 3-8 membered heterocyclenyl, 3-8 membered heterocycloalkyl, C 3-8 Cycloalkenyl and C 3-8 Cycloalkyl (you)Optionally, the A ring and the B ring are each independently selected from C 6-10 Aryl, 5-10 membered heteroaryl, 3-8 membered heterocycloalkyl and C 3-8 Cycloalkyl);
r is selected from hydrogen, optionally halogenated C 1-6 Alkyl, halogen and cyano (preferably, R is selected from hydrogen and halogen);
r is selected from 0, 1 and 2;
R 4 selected from hydrogen, amino, C 1-6 alkyl-NH- (C) 1-6 Alkyl group 2 N-、C 1-6 Alkyl and hydroxy;
m is selected from 0, 1 and 2.
In some embodiments of the first aspect of the invention, R 1 Selected from C 1-6 Alkyl, C 6-10 Aryl groupWherein the C 6-10 Aryl is optionally substituted with one or more of the following: hydroxy, NH 2 -C 1-6 Alkylene-, halogen;
the A and B rings are each independently selected from C 6-10 Aryl, 5-10 membered heteroaryl, 3-8 membered heterocyclenyl, 3-8 membered heterocycloalkyl, C 3-8 Cycloalkenyl and C 3-8 Cycloalkyl (preferably, the A and B rings are each independently selected from C 6-10 Aryl, 5-10 membered heteroaryl, 3-8 membered heterocycloalkyl and C 3-8 Cycloalkyl);
R is selected from hydrogen, optionally halogenated C 1-6 Alkyl, halogen and cyano (preferably, R is selected from hydrogen and halogen);
r is selected from 0, 1 and 2;
R 4 selected from hydrogen, amino, C 1-6 alkyl-NH- (C) 1-6 Alkyl group 2 N-、C 1-6 Alkyl and hydroxy;
m is selected from 0, 1 and 2.
In some embodiments of the first aspect of the invention, R 1 Selected from C 1-6 Alkyl, C 6-10 Aryl groupWherein the C 6-10 Aryl is optionally substituted with one or more of the following: hydroxy, NH 2 -C 1-6 Alkylene-, halogen;
ring A is C 6-10 An aryl group;
the B ring is selected from 5-10 membered heteroaryl, 3-8 membered heterocyclenyl, 3-8 membered heterocycloalkyl, C 3-8 Cycloalkenyl and C 3-8 Cycloalkyl groups.
In some embodiments of the first aspect of the invention, R 1 Selected from C 1-6 Alkyl, C 6-10 Aryl groupWherein the C 6-10 Aryl is optionally substituted with one or more of the following: hydroxy, NH 2 -C 1-6 Alkylene-, halogen;
ring A is phenyl;
ring B is selected from pyrazolyl, 3-pyrrolinyl, pyrrolidinyl, cyclopentenyl and cyclopentenyl, preferably, ring B is selected from pyrazolyl, pyrrolidinyl and cyclopentenyl;
r is selected from hydrogen and halogen;
r is selected from 0, 1 and 2;
R 4 selected from hydrogen, amino, C 1-6 alkyl-NH- (C) 1-6 Alkyl group 2 N-、C 1-6 Alkyl and hydroxy;
m is selected from 0, 1 and 2.
In some embodiments of the first aspect of the invention, R 1 Selected from methyl, phenyl andwherein the phenyl group is optionally substituted with one or more of the following groups: halogen, hydroxy and NH 2 -CH 2 -;
Ring A is phenyl;
the B ring is selected from pyrazolyl, 3-pyrrolinyl, pyrrolidinyl, cyclopentenyl and cyclopentenyl;
r is selected from hydrogen and halogen, preferably R is selected from hydrogen, F and Cl;
r is selected from 0 and 1;
R 4 selected from hydrogen, amino, CH 3 -NH-、(CH 3 ) 2 N-, methyl and hydroxy;
m is selected from 0, 1 and 2.
In some embodiments of the first aspect of the invention, R 1 A phenyl group optionally substituted with one or more substituents selected from the group consisting of: hydrogen, fluorine, chlorine, -OH, -CH 2 NH 2
In some embodiments of the first aspect of the invention, R 1 Is thatWherein the A ring is selected from C 6-10 Aryl and 5-10 membered heteroaryl; r is selected from hydrogen, optionally halogenated C 1-6 Alkyl and halogen; r is selected from 1, 2 and 3.
In some embodiments of the first aspect of the invention, R 1 Is thatWherein ring A is C 6-10 An aryl group; r is selected from hydrogen, optionally halogenated C 1-6 Alkyl and halogen; r is selected from 1, 2 and 3.
In some embodiments of the first aspect of the invention, R 1 Is thatWherein ring A is C 6-10 An aryl group; r is selected from hydrogen and halogen; r is selected from 1, 2 and 3.
In some embodiments of the first aspect of the invention, R 1 Is thatWherein, the ring A is benzene ring; r is selected from hydrogen, fluorine and chlorine; r is selected from 1 and 2.
In some embodiments of the first aspect of the invention, R 1 Is thatWherein the B ring is selected from C 3-8 Cycloalkyl, 3-8 membered heterocycloalkyl, and 5-10 membered heteroaryl; r is R 4 Selected from hydrogen, optionally halogenated C 1-6 Alkyl, oxo, -OH, -NH 2 、-NH(C 1-6 Alkyl), -N (C) 1-6 Alkyl group 2 、C 3-8 Cycloalkyl and cyano; m is selected from 1, 2 and 3.
In some embodiments of the first aspect of the invention, R 1 Is thatWherein the B ring is selected from C 3-8 Cycloalkyl, 3-8 membered heterocycloalkyl, and 5-10 membered heteroaryl; r is R 4 Selected from hydrogen, optionally halogenated C 1-6 Alkyl, -OH, -NH 2 、-NH(C 1-6 Alkyl) and-N (C) 1-6 Alkyl group 2 The method comprises the steps of carrying out a first treatment on the surface of the m is selected from 1, 2 and 3.
In some embodiments of the first aspect of the invention, R 1 Is thatWherein the B ring is selected from C 4-6 Cycloalkyl, 4-6 membered heterocycloalkyl, and 5-10 membered heteroaryl; r is R 4 Selected from hydrogen, methyl, -NH 2 、-NHCH 3 -OH and-N (CH) 3 ) 2 The method comprises the steps of carrying out a first treatment on the surface of the m is selected from 1 and 2.
In some embodiments of the first aspect of the invention, R 1 Is thatWherein the B ring is selected from pyrazolyl, 3-pyrrolinyl, pyrrolidinyl, cyclopentenyl and cyclopentenyl; r is R 4 Selected from hydrogen, methyl, -NH 2 、-NHCH 3 -OH and-N (CH) 3 ) 2 The method comprises the steps of carrying out a first treatment on the surface of the m is selected from 1 and 2.
In some embodiments of the first aspect of the invention, R 1 Is thatWherein the B ring is pyrrolidinyl; r is R 4 Selected from hydrogen, methyl, -NH 2 、-NHCH 3 -OH and-N (CH) 3 ) 2 The method comprises the steps of carrying out a first treatment on the surface of the m is selected from 1 and 2.
In some embodiments of the first aspect of the invention, R 1 Selected from methyl group,
In some embodiments of the first aspect of the invention, R 1 Selected from methyl group,
In some embodiments of the first aspect of the invention, R 1 Selected from C 6-10 Aryl groupWherein the C 6-10 Aryl is optionally substituted with one or more of the following: halogen and-C 1-6 alkylene-NR b R c
The A ring and the B ring are independently selected from C 6-10 Aryl, 3-8 membered heterocycloalkenyl, 3-8 membered heterocycloalkyl, C 3-8 Cycloalkenyl and C 3-8 Cycloalkyl;
r is selected from hydrogen and halogen; r is selected from 0, 1 and 2;
R 4 selected from hydrogen, -NR b R c 、C 1-6 Alkyl and-OR a The method comprises the steps of carrying out a first treatment on the surface of the m is selected from 0, 1 and 2;
R a 、R b and R is c Each independently selected from hydrogen and C 1-6 An alkyl group.
In some embodiments of the first aspect of the invention, R 1 Selected from C 6-10 Aryl groupWherein the C 6-10 Aryl is optionally substituted with one or more of the following: halogen and NH 2 -C 1-6 An alkylene group;
the A ring and the B ring are independently selected from C 6-10 Aryl, 3-8 membered heterocycloalkenyl, 3-8 membered heterocycloalkyl, C 3-8 Cycloalkenyl and C 3-8 Cycloalkyl;
r is selected from hydrogen and halogen; r is selected from 0, 1 and 2;
R 4 selected from hydrogen, amino, C 1-6 alkyl-NH- (C) 1-6 Alkyl group 2 N-、C 1-6 Alkyl and hydroxy; m is selected from 0, 1 and 2.
In some embodiments of the first aspect of the invention, R 1 Selected from phenyl and Wherein the phenyl group is optionally substituted with one or more of the following groups: halogen and NH 2 -CH 2 -;
Ring A is phenyl;
the B ring is selected from 3-pyrrolinyl, pyrrolidinyl, cyclopentenyl and cyclopentenyl;
r is selected from hydrogen and halogen; r is selected from 0 and 1;
R 4 selected from hydrogen, amino, CH 3 -NH-、(CH 3 ) 2 N-, methyl and hydroxy; m is selected from 0, 1 and 2.
In some embodiments of the first aspect of the invention, R 1 Selected from the group consisting of
In some embodiments of the first aspect of the invention, R 2 Selected from hydrogen, halogen, optionally halogenated C 1-6 Alkyl and C 3-8 Cycloalkyl groups.
In some embodiments of the first aspect of the invention, R 2 Selected from hydrogen, fluorine, methyl, trifluoromethyl; preferably, the method comprises the steps of,R 2 is hydrogen.
In some embodiments of the first aspect of the invention, R 3 Selected from-C (O) R 9 and-C (O) NR e R f ;R 9 、R e 、R f Each independently selected from hydrogen, C 1-6 Alkyl, C 2-6 Alkenyl, C 3-8 Cycloalkyl and 3-8 membered heterocycloalkyl, wherein said C 1-6 Alkyl, C 2-6 Alkenyl, C 3-8 Cycloalkyl and 3-8 membered heterocycloalkyl are each independently optionally substituted with one or more substituents selected from the group consisting of: halogen, optionally halogenated C 1-6 Alkyl, C 3-8 Cycloalkyl, C 6-10 Aryl and 5-10 membered heteroaryl; and, the C 6-10 Aryl and 5-10 membered heteroaryl are each independently optionally substituted with one or more of the following: hydrogen, halogen, optionally halogenated C 1-6 An alkyl group.
In some embodiments of the first aspect of the invention, R 3 is-C (O) R 9 ;R 9 Selected from hydrogen, C 1-6 Alkyl, C 2-6 Alkenyl, C 3-8 Cycloalkyl and 3-8 membered heterocycloalkyl, wherein said C 1-6 Alkyl, C 2-6 Alkenyl, C 3-8 Cycloalkyl and 3-8 membered heterocycloalkyl are each independently optionally substituted with one or more substituents selected from the group consisting of: halogen, optionally halogenated C 1-6 Alkyl, C 3-8 Cycloalkyl, C 6-10 Aryl and 5-10 membered heteroaryl; and, the C 6-10 Aryl and 5-10 membered heteroaryl are each independently optionally substituted with one or more of the following: hydrogen, halogen, optionally halogenated C 1-6 An alkyl group.
In some embodiments of the first aspect of the invention, R 3 is-C (O) R 9 ;R 9 Selected from hydrogen, C 1-6 Alkyl, C 2-6 Alkenyl and C 3-8 Cycloalkyl, wherein, the C 1-6 Alkyl, C 2-6 Alkenyl and C 3-8 Cycloalkyl groups are each independently optionally substituted with one or more substituents as follows: halogen, C 1-6 Alkyl, C 3-8 Cycloalkyl, C 6-10 Aryl and 5-10 membered heteroaryl; and is also provided withC in the substituent 1-6 Alkyl, C 3-8 Cycloalkyl, C 6-10 Aryl and 5-10 membered heteroaryl are each independently optionally substituted with one or more of the following: hydrogen, halogen, C 1-6 An alkyl group.
In some embodiments of the first aspect of the invention, R 3 is-C (O) R 9 ;R 9 Selected from hydrogen, C 1-6 Alkyl and C 2-6 Alkenyl group, wherein the C 1-6 Alkyl and C 2-6 Alkenyl groups are each independently optionally substituted with one or more substituents selected from the group consisting of: halogen, C 1-6 Alkyl, C 3-8 Cycloalkyl, C 6-10 Aryl and 5-10 membered heteroaryl; and, C in the substituent 1-6 Alkyl, C 3-8 Cycloalkyl, C 6-10 Aryl and 5-10 membered heteroaryl are each independently optionally substituted with one or more of the following: hydrogen, halogen, C 1-6 An alkyl group.
In some embodiments of the first aspect of the invention, R 3 is-C (O) R 9 ;R 9 Is C optionally substituted with one or more substituents 1-6 Alkyl: halogen, optionally halogenated C 1-6 Alkyl, C 3-8 Cycloalkyl, C 6-10 Aryl, 5-10 membered heteroaryl, wherein the C 6-10 Aryl and 5-10 membered heteroaryl are each independently optionally substituted with a C selected from hydrogen, halogen and optionally halogenated 1-6 One or more groups in the alkyl group are substituted.
In some embodiments of the first aspect of the invention, R 3 is-C (O) R 9 ;R 9 Is C optionally substituted with one or more substituents 1-6 Alkyl: optionally halogenated C 1-6 Alkyl, C 3-8 Cycloalkyl, C 6-10 Aryl, 5-10 membered heteroaryl, wherein the C 6-10 Aryl and 5-10 membered heteroaryl are each independently optionally substituted with one or more halogens.
In some embodiments of the first aspect of the invention, R 3 is-C (O) R 9 ;R 9 Is C optionally substituted with one or more substituents 1-6 Alkyl: methyl groupDifluoromethyl, trifluoromethyl, cyclopropyl, optionally halogenated phenyl (e.g. phenyl substituted by a fluoro), optionally halogenated pyrazolyl (e.g. pyrazolyl substituted by a fluoro or chloro).
In some embodiments of the first aspect of the invention, R 3 Is C (O) R 9 ,R 9 Selected from the following groups:
the second aspect of the present invention relates to a compound of formula ii, a pharmaceutically acceptable salt, ester, solvate, stereoisomer, tautomer, prodrug, any form, metabolite, or mixture thereof:
wherein,
R 1 、R 2 is as defined in the first aspect of the invention;
R 13 is O;
R 14 selected from C 6-10 Aryl and 5-8 membered heteroaryl, wherein C 6-10 Aryl and 5-8 membered heteroaryl are each independently optionally substituted with one or more substituents as follows: hydrogen, halogen, hydroxy, C 1-6 Alkyl, C 1-6 Alkoxy, amino, cyano;
R 15 selected from H, C 1-6 Alkyl and C 3-8 Cycloalkyl group, wherein C 1-6 Alkyl and C 3-8 Cycloalkyl groups are each independently optionally substituted with one or more substituents as follows: halogen, hydroxy, C 1-6 Alkyl, C 1-6 Alkoxy, amino, cyano;
n is selected from 1, 2, 3, 4, 5 and 6; preferably, n is selected from 1, 2 and 3, more preferably n is 1.
In some embodiments of the second aspect of the invention, R 14 Selected from C 6-10 Aryl and 5-8 membered heteroaryl, wherein C 6-10 Aryl and 5-8 membered heteroaryl are each independently optionally substituted with one or more halogens.
In some embodiments of the second aspect of the invention, R 14 Selected from C 6-10 Aryl and 5-8 membered nitrogen containing heteroaryl groups, wherein C 6-10 Aryl and 5-8 membered nitrogen containing heteroaryl groups are optionally substituted with one or more halogens.
In some embodiments of the second aspect of the invention, R 14 Selected from phenyl and pyrazolyl, wherein the phenyl and pyrazolyl are each independently optionally substituted with one or more halogens.
In some embodiments of the second aspect of the invention, R 14 Selected from phenyl group,
In some embodiments of the second aspect of the invention, R 15 Selected from C 1-6 Alkyl and C 3-8 Cycloalkyl group, wherein C 1-6 The alkyl group is optionally substituted with one or more halogens.
In some embodiments of the second aspect of the invention, R 15 Selected from methyl, difluoromethyl, trifluoromethyl and cyclopropyl.
In some embodiments of the second aspect of the invention, R 15 Selected from methyl, difluoromethyl and cyclopropyl.
In some embodiments of the second aspect of the invention, R 1 Selected from methyl group,/>
R 2 Selected from hydrogen, fluorine, methyl and trifluoromethyl;
R 14 selected from phenyl group,
R 15 Selected from methyl, difluoromethyl and cyclopropyl; and is also provided with
n is selected from 1, 2 and 3.
In some embodiments of the first aspect of the invention, the compound is selected from compounds 1-44 in the following table.
In some embodiments of the second aspect of the invention, the compound is selected from compounds 1-44, R in the following table 1 、R 14 And R is 15 As shown in the table, R 2 Is H, R 13 O and n is 1.
/>
/>
/>
/>
/>
Any atom in the compounds of the present invention may be replaced by its isotope. For example 12 C can be isotopically substituted with C 13 C or 14 C is substituted; 1 h can be covered by 2 H (D, deuterium) or 3 H (T, tritium) substitution, and the like. Isotopically-labeled compounds obtained by substituting any atom in the compounds encompassed by the present invention with their isotopes.
The third aspect of the present invention relates to a pharmaceutical composition comprising a compound according to the first or second aspect of the present invention, a pharmaceutically acceptable salt, ester, solvate, stereoisomer, tautomer, prodrug, any crystal form, metabolite or mixture thereof, and optionally a pharmaceutically acceptable adjuvant.
In some embodiments of the third aspect of the invention, the pharmaceutical composition is a tablet, pill, capsule, lozenge, hard candy, powder, spray, cream, ointment, suppository, gel, paste, lotion, ointment, aqueous suspension, injectable solution, elixir or syrup.
In some embodiments of the third aspect of the invention, the pharmaceutical composition may be administered parenterally, topically, intravenously, orally, subcutaneously, intra-arterially, intradermally, transdermally, rectally, intracranially, intraperitoneally, intranasally, intramuscularly or as an inhalant, and may also be administered by any of the forms of use known to those skilled in the art of medicine. The compounds of the present invention or pharmaceutically acceptable salts, esters, solvates, stereoisomers, tautomers, prodrugs, any crystal forms, metabolites, or mixtures thereof, may be formulated into a variety of suitable dosage forms depending on the route of administration.
In some embodiments of the third aspect of the invention, the pharmaceutical composition further comprises an additional active ingredient that inhibits the activity of deubiquitinase USP7 or an additional active ingredient that prevents or treats cancer.
In some embodiments of the third aspect of the invention, the pharmaceutical composition further comprises an additional pharmaceutically active ingredient for preventing or treating a disease or disorder associated with modulation of deubiquitinase USP 7.
In some embodiments of the third aspect of the invention, the disease or disorder associated with modulation of deubiquitinase USP7 is selected from cancer, neurodegenerative diseases (e.g., alzheimer's disease, parkinson's disease), diabetes, bone and joint diseases, arthritic disorders, osteoporosis, immune disorders, cardiovascular diseases, ischemic diseases, viral infectious diseases, and bacterial infectious diseases.
In some embodiments of the third aspect of the invention, the pharmaceutical composition or suitable dosage form may contain from 0.01mg to 1000mg of a compound of the invention.
The fourth aspect of the invention also relates to a pharmaceutical product comprising a compound according to the first or second aspect of the invention, a pharmaceutically acceptable salt, ester, solvate, stereoisomer, tautomer, prodrug, any crystal form, metabolite or mixture thereof, or a pharmaceutical composition according to the third aspect of the invention, and optionally a product package (e.g. the product package comprises a product instruction).
In a fifth aspect the present invention relates to a method for inhibiting the activity of deubiquitinase USP7 in vitro or for inhibiting the proliferation of tumor cells in vitro comprising administering to a subject an effective amount of a compound according to the first or second aspect of the present invention, a pharmaceutically acceptable salt, ester, solvate, stereoisomer, tautomer, prodrug, any crystal form, metabolite or mixture thereof, or a pharmaceutical composition according to the third aspect of the present invention.
A sixth aspect of the present invention relates to the use of a compound according to the first or second aspect of the present invention, a pharmaceutically acceptable salt, ester, solvate, stereoisomer, tautomer, prodrug, any form, metabolite or mixture thereof, or a pharmaceutical composition according to the third aspect of the present invention, for the manufacture of a deubiquitinase USP7 inhibitor or for the manufacture of a medicament for the prevention or treatment of a disease or condition associated with the modulation of deubiquitinase USP 7.
In some embodiments of the sixth aspect of the invention, the disease or disorder associated with modulation of deubiquitinase USP7 is selected from cancer, neurodegenerative diseases, diabetes, bone and joint diseases, arthritic disorders, osteoporosis, immune disorders, cardiovascular diseases, ischemic diseases, viral infectious diseases, and bacterial infectious diseases.
In some embodiments of the sixth aspect of the invention, the cancer is selected from at least one of hepatocellular carcinoma, multiple myeloma, colon cancer, lung cancer, prostate cancer, and bladder cancer.
A seventh aspect of the present invention relates to a method for preventing or treating a disease or condition associated with modulation of deubiquitinase USP7, comprising administering to a subject in need thereof an effective amount of a compound according to the first or second aspect of the present invention, a pharmaceutically acceptable salt, ester, solvate, stereoisomer, tautomer, prodrug, any crystal form, metabolite or mixture thereof, or a pharmaceutical composition according to the third aspect of the present invention.
In some embodiments of the seventh aspect of the invention, the disease or disorder associated with modulation of deubiquitinase USP7 is selected from cancer, neurodegenerative diseases, diabetes, bone and joint diseases, arthritic disorders, osteoporosis, immune disorders, cardiovascular diseases, ischemic diseases, viral infectious diseases, and bacterial infectious diseases.
In some embodiments of the seventh aspect of the invention, the cancer is selected from at least one of hepatocellular carcinoma, multiple myeloma, colon cancer, lung cancer, prostate cancer, and bladder cancer.
A compound according to the first or second aspect of the invention, a pharmaceutically acceptable salt, ester, solvate, stereoisomer, tautomer, prodrug, any crystal form, metabolite, or mixture thereof, or a pharmaceutical composition according to the third aspect of the invention, for use in inhibiting deubiquitinase USP7 activity, or for use in the prevention or treatment of a disease or disorder associated with deubiquitinase USP7 modulation.
In some embodiments of the first or second aspect of the invention, the disease or disorder associated with modulation of deubiquitinase USP7 is selected from cancer, neurodegenerative diseases, diabetes, bone and joint diseases, arthritic disorders, osteoporosis, immune disorders, cardiovascular diseases, ischemic diseases, viral infectious diseases, and bacterial infectious diseases.
In some embodiments of the first or second aspect of the invention, the cancer is selected from at least one of hepatocellular carcinoma, multiple myeloma, colon cancer, lung cancer, prostate cancer, and bladder cancer.
The compounds of the first or second aspect of the invention may be synthesised using the methods described below, synthetic methods known in the art of synthetic organic chemistry or well known variations thereof. Preferred methods include, but are not limited to, the methods described below.
A process for preparing a compound as described in the first or second aspect of the invention, comprising:
step one: reacting the compound a with a compound e to obtain a compound b;
step two: removing the protecting group PG from the compound b through deprotection reaction to obtain a compound c;
step three: reacting the compound c with a compound f to obtain a compound d;
step four: the compound d is coupled with the compound g to obtain the compound shown in the formula I or the second aspect of the invention.
Wherein R is 1 、R 2 、R 3 Is as defined in the first aspect of the invention; x is selected from halogen and-OTf, preferably X is chlorine or bromine; PG is selected from C 1-6 Alkyloxycarbonyl, halogen substituted C 1-6 Alkyl oxycarbonyl, C 2-6 Alkenyl-alkoxycarbonyl, cbz, benzyl and 9-fluorenylmethoxycarbonyl, preferably PG is selected from Boc; r is R 17 Selected from hydroxyl and halogen, preferably hydroxyl; r is R 18 Selected from boric acid, C 1-6 Alkyl substituted boric acids and five membered oxaboranes substituted with one or more methyl groups.
In some embodiments of the preparation process of the present invention, the reaction in step one is carried out in an organic solvent selected from DMSO, DMF, DMA, alcohols (e.g. methanol, ethanol, isopropanol, t-butanol), ethers (e.g. anisole, diethyl ether, THF, 1, 4-dioxane, etc.), halogenated hydrocarbons (e.g. dichloromethane, chloroform, carbon tetrachloride) and acetonitrile, preferably DMSO, DMF.
In some embodiments of the preparation process of the present invention, the reaction in step one is carried out in the presence of a base selected from the group consisting of triethylamine, DIPEA, pyridine, NMM, DMAP, sodium acetate, potassium acetate, ammonium acetate, potassium carbonate, sodium bicarbonate, cesium carbonate, potassium phosphate, potassium hydroxide, sodium hydroxide, preferably the base is selected from the group consisting of potassium carbonate, cesium carbonate and pyridine.
In some embodiments of the preparation process of the present invention, the reaction in step one is carried out at a temperature of from 0℃to 200℃and preferably at a temperature of from 50℃to 150 ℃.
In some embodiments of the process of the present invention, the deprotection reaction in step two is carried out in a solvent selected from the group consisting of water, DMF, DMA, N-methylpyrrolidone, alcohols (e.g., methanol, ethanol, isopropanol, etc.), ethers (e.g., diethyl ether, THF, 1, 4-dioxane, etc.), halogenated hydrocarbons (e.g., dichloromethane, chloroform, carbon tetrachloride, 1, 2-dichloroethane, etc.), acetonitrile and ethyl acetate, preferably dichloromethane, methanol, tetrahydrofuran.
In some embodiments of the process of the present invention, the deprotection reaction in step two is performed in the presence of an acid selected from trifluoroacetic acid, hydrochloric acid/1, 4-dioxane solution, hydrochloric acid/ethyl acetate solution, hydrobromic acid.
In some embodiments of the process of the present invention, the deprotection reaction in step two is carried out at a temperature of from 0℃to 50℃and preferably at a temperature of from 0℃to 25 ℃.
In some embodiments of the process of the present invention, the condensation reaction in step three is performed in an organic solvent selected from the group consisting of halogenated hydrocarbons (e.g., dichloromethane, chloroform, 1, 2-dichloroethane, etc.), nitriles (e.g., acetonitrile, etc.), N-methylpyrrolidone, DMF, DMA, 1, 4-dioxane, DMSO, and any combination thereof, preferably dichloromethane, DMF.
In some embodiments of the process of the present invention, the condensation reaction in step three is performed in the presence of a condensing agent selected from the group consisting of ethyl chloroformate, isopropyl chloroformate, HATU, HBTU, EEDQ, DEPC, DCC, DIC, EDC, BOP, pyAOP, and PyBOP, preferably the condensing agent is HATU, EDC.
In some embodiments of the process according to the invention, the condensation reaction in step three is carried out in the presence of a base selected from triethylamine, DIPEA, NMM and DMAP, preferably DIPEA.
In some embodiments of the process of the present invention, the condensation reaction in step three is carried out at a temperature of from 0 ℃ to 100 ℃, preferably from 15 ℃ to 50 ℃.
In some embodiments of the preparation process of the present invention, the coupling reaction of step four is performed in an organic solvent selected from the group consisting of halogenated hydrocarbons (e.g., dichloromethane, chloroform, 1, 2-dichloroethane, etc.), methanol, ethanol, DMF, acetonitrile, ethers (e.g., ethylene glycol dimethyl ether, tetrahydrofuran, 1, 4-dioxane), aromatic hydrocarbons (e.g., toluene, benzene, xylene), water, and any combination thereof, preferably 1, 4-dioxane/water, toluene/water.
In some embodiments of the preparation process of the present invention, the coupling reaction of step four is performed in the presence of a base, which is an organic base or an inorganic base; preferably, the organic base is selected from triethylamine, DIPEA, NMM, sodium tert-butoxide, potassium acetate, sodium acetate, and the inorganic base is selected from potassium carbonate, sodium bicarbonate, cesium carbonate, potassium phosphate, and potassium dihydrogen phosphate; preferably, the inorganic base is selected from potassium carbonate, potassium phosphate, potassium acetate, sodium carbonate, sodium bicarbonate.
In some embodiments of the preparation process of the present invention, the coupling reaction of step four is performed under the catalysis of a catalyst selected from the group consisting of palladium tetraphenylphosphine, palladium acetate, pd 2 (dba) 3 、Pd(PPh 3 ) 2 Cl 2 、Pd(PPh 3 ) 2 Cl 2 Dichloromethane complex, pd (dppf) Cl 2 And Pd (amphos) Cl 2 Preferably palladium tetraphenylphosphine, pd (dppf) Cl 2 、Pd(amphos)Cl 2
In some embodiments of the preparation process of the present invention, the coupling reaction of step four is carried out at a temperature of from 0℃to 200℃and preferably at a temperature of from 50℃to 150 ℃.
The present invention encompasses all possible crystalline forms or polymorphs of the compounds of the present invention, which may be single polymorphs or mixtures of multiple polymorphs in any ratio.
The compounds of the present invention or pharmaceutically acceptable salts thereof may also form solvates, such as alcohol complexes and the like.
The compounds of the invention may also be in the form of prodrugs or in the form of releasing the active ingredient upon metabolic changes in the body. The selection and preparation of the appropriate prodrug derivatives is well known to those skilled in the art.
The compounds of the invention may also be in chemically protected form, the protecting group being capable of protecting against an active group (e.g. amino group) of the compound, the protecting group being capable of metabolising in vivo to release the active ingredient. The selection and preparation of the appropriate chemoprotective forms is well known to those skilled in the art.
It will also be appreciated that certain compounds of the invention may exist in free form for use in therapy or, where appropriate, in the form of pharmaceutically acceptable derivatives thereof. According to the present invention, pharmaceutically acceptable derivatives include, but are not limited to, pharmaceutically acceptable salts, solvates, metabolites or prodrugs which, upon administration to a patient in need thereof, are capable of providing the compounds of the present invention or metabolites or residues thereof, either directly or indirectly.
The compounds of the invention may exist in the form of hydrates or solvates, wherein the compounds of the invention comprise a polar solvent as a structural element of the compound lattice, in particular, for example, water, methanol or ethanol. The polar solvent, in particular water, may be present in stoichiometric or non-stoichiometric amounts.
Also included within the scope of the invention are metabolites of the compounds of the invention, i.e., compounds that form in vivo upon administration of the drug.
In the present invention, unless otherwise specified, wherein:
the term "alkyl" is defined as a straight or branched chain saturated aliphatic hydrocarbon group. For example, the term "C 1-6 Alkyl "means a straight or branched chain radical having 1 to 6 carbon atoms (e.g., methyl, ethyl, n-propyl, isopropyl, n-butyl)Isobutyl, sec-butyl, tert-butyl, n-pentyl or n-hexyl) optionally substituted with one or more (such as 1 to 3) suitable substituents, such as halogen (this group is referred to as "haloalkyl", for example, -CF) 3 、-C 2 F 5 、-CHF 2 、-CH 2 F、-CH 2 CF 3 、-CH 2 Cl or-CH 2 CH 2 CF 3 Etc.).
The term "cycloalkyl" refers to a saturated monocyclic or bicyclic hydrocarbon group such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl. For example, as used herein, the term "C 3-8 Cycloalkyl "refers to a saturated monocyclic or bicyclic hydrocarbon group of 3 to 8 ring-forming carbon atoms (e.g., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl) optionally substituted with one or more (such as 1 to 3) suitable substituents, e.g., methyl-substituted cyclopropyl.
The term "cycloalkenyl" refers to an unsaturated, non-aromatic, monocyclic or multicyclic (such as bicyclic) hydrocarbon ring containing at least one carbon-carbon double bond. For example, as used herein, the term "C 3-8 Cycloalkenyl "refers to an unsaturated, non-aromatic, monocyclic or multicyclic (such as bicyclic) hydrocarbon ring of 3 to 8 carbon atoms having at least one carbon-carbon double bond, optionally substituted with one or more (such as 1 to 3) suitable substituents, for example methyl substituted cyclobutenyl.
The term "alkoxy" refers to an "alkyl" or "cycloalkyl" as defined above, attached to the parent molecular moiety through an oxygen atom, e.g., C 1-6 Alkoxy, C 1-3 Alkoxy or C 3-8 Cycloalkoxy, for example methoxy, ethoxy, n-propoxy, isopropoxy, cyclopropoxy, n-butoxy, isobutoxy, tert-butoxy, sec-butoxy, cyclobutoxy, pentoxy, isopentoxy or n-hexoxy, or isomers thereof.
The term "heterocycloalkyl" refers to a saturated mono-or bicyclic group having 2, 3, 4, 5, 6, 7, 8 or 9 carbon atoms in the ring and one or more (e.g., one, two, three or four) selected from C (=o), O, S, N, S (=o), S (=O) 2 A heteroatom-containing group; the heterocycloalkyl group may be attached to the remainder of the molecule through any one of the carbon atoms or a heteroatom (if present). For example, as used herein, the term 3-8 membered heterocycloalkyl, 5-6 membered heterocycloalkyl, or 3-7 membered heterocycloalkyl is having 3-8, 5-6, or 3-7 ring atoms in the ring, and containing at least one heteroatom (which may be the same or different, e.g., oxygen, nitrogen, or sulfur). Such as, but not limited to, oxiranyl, aziridinyl, azetidinyl, oxetanyl, tetrahydrofuranyl, pyrrolidinyl, pyrrolidinonyl, imidazolidinyl, pyrazolidinyl, pyrrolinyl, tetrahydropyranyl.
The term "heterocycloalkenyl" refers to an unsaturated, non-aromatic, monocyclic or polycyclic (such as bicyclic) group containing at least one carbon-carbon double bond having 2, 3, 4, 5, 6, 7, 8, or 9 carbon atoms in the ring and one or more (e.g., one, two, three, or four) carbon atoms selected from C (=o), O, S, N, S (=o), S (=o) 2 A heteroatom-containing group; the heterocycloalkenyl group may be attached to the remainder of the molecule through any one of the carbon atoms or a heteroatom (if present). For example, as used herein, the term 3-8 membered heterocycloalkenyl is one having 3-8 ring atoms in the ring and which contains at least one heteroatom (which is, for example, oxygen, nitrogen, or sulfur) that may be the same or different. Such as dioxolyl (dioxanyl).
The term "aryl" refers to an all-carbon monocyclic or fused-ring polycyclic aromatic group having a conjugated pi-electron system. For example, as used herein, the term "C 6-10 An aromatic ring "means an aromatic group containing 6 to 10 carbon atoms, such as phenyl or naphthyl. Aryl is optionally substituted with 1 or more (such as 1 to 3) suitable substituents, for example alkyl (-CH) 3 、-C 2 H 5 ) Halogen (F, cl, br) substitution.
The term "heteroaryl" refers to a monocyclic, bicyclic or tricyclic aromatic ring system having 5, 6, 8, 9, 10, 11, 12, 13 or 14 ring atoms, for example having 5 to 10 ring atoms, in particular 1 or 2 or 3 or 4 or 5 or 6 or 9 or 10 ring atomsA carbon atom, and which contains at least one heteroatom (which may be the same or different, for example, oxygen, nitrogen or sulfur), and which may additionally be benzo-fused in each case. For example, as used herein, the term "5-10 membered heteroaryl" means a monocyclic, bicyclic or tricyclic aromatic ring system having 5-10 ring atoms, and which contains at least one heteroatom (which is, for example, oxygen, nitrogen or sulfur) that may be the same or different. In particular, heteroaryl is selected from thienyl, furyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, and the like, and the benzo derivatives thereof; or pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, and the like, and their benzo derivatives. Heteroaryl groups are optionally substituted with 1 or more (such as 1 to 3) suitable substituents, for example alkyl (-CH) 3 、-C 2 H 5 ) Halogen (F, cl, br) substitution. As used herein, the term "halo" or "halogen" group is defined to include F, cl, br or I.
The term "halogen" refers to fluorine, chlorine, bromine, iodine.
The term "C 1-6 Alkylene "refers to a divalent radical remaining from a C1-6 alkyl radical with one hydrogen atom removed, where" C 1-6 Alkyl "is defined above. Such as C1-4 alkylene, C1-2 alkylene, C1 alkylene, C2 alkylene, C3 alkylene, C4 alkylene, C5 alkylene, C6 alkylene.
The term "mercapto" is also known as a hydrosulfuryl or thiol group and has the structural formula-SH.
The carbon and nitrogen atoms in the term "cyano" are linked by a triple bond and have the formula-CN.
The term "nitro" refers to the residue of nitric acid after removal of a hydroxyl group, having the formula-NO 2
The term "alkenyl" refers to an aliphatic hydrocarbon group containing at least one carbon-carbon double bond and one hydrogen atom of which is replaced by a bond. Alkenyl groups may be straight or branched and contain from about 2 to about 15 carbon atoms. For example, "C" herein 2-6 Alkenyl "is a radical containing 2 to 6Alkenyl groups of carbon atoms. Non-limiting examples of alkenyl groups include ethenyl, propenyl, n-butenyl, 3-methylbut-2-enyl, n-pentenyl, octenyl and decenyl. Alkenyl may be unsubstituted alkenyl or alkenyl substituted with one or more identical or different substituents, each substituent being independently selected from the group consisting of halogen, alkenyl, alkynyl, aryl, cycloalkyl, cyano, hydroxy, -O-alkyl, -O-aryl, -alkylene-O-alkyl, alkylthio, -NH 2 -NH (alkyl), -N (alkyl) 2 -NH (cycloalkyl), -O-C (O) -alkyl, -O-C (O) -aryl, -O-C (O) -cycloalkyl, -C (O) OH and-C (O) O-alkyl.
The term "alkynyl" refers to a hydrocarbon group having one or more c≡c triple bonds. The alkynyl group has, but is not limited to, 2 to 18 carbon atoms, for example, it has 2 to 10 carbon atoms, for example, 2 to 6 carbon atoms. "lower alkynyl" herein refers to lower alkynyl groups, e.g., having 2 to 8 carbon atoms, e.g., 2 to 6 carbon atoms, e.g., 2 to 4 carbon atoms. Examples of alkynyl groups herein include, but are not limited to, ethynyl, 2-propynyl, 2-butynyl, 1, 3-butadiynyl, and the like. Alkynyl groups as defined herein are present in the numerical range, e.g. "C 2-6 Alkynyl "refers to an alkynyl group that may be composed of 2 carbon atoms, 3 carbon atoms, 4 carbon atoms, 5 carbon atoms, or 6 carbon atoms, and alkynyl herein also encompasses cases where no numerical range is specified.
The term "amino" refers to substituted or unsubstituted "-NH- 2 ". For example, representative amino groups include-NH 2 、-NHCH 3 、-N(CH 3 ) 2 、-NHC 1-3 Alkyl, -N (C) 1-3 Alkyl group 2 Etc. Unless otherwise indicated, compounds of the invention containing an amino moiety may include protected derivatives thereof. Suitable protecting groups for the amino moiety include acetyl, t-butoxycarbonyl, benzyloxycarbonyl, and the like.
The term "substituted" means that one or more (e.g., one, two, three, or four) hydrogens on the designated atom are replaced with a selection from the indicated group, provided that the designated atom's normal valency is not exceeded, and that the substitution forms a stable compound. Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.
The term "optionally substituted with …" means optionally substituted or partially substituted or unsubstituted with a particular group, radical.
When a group is described as "optionally substituted with one or more substituents," the group may be (1) unsubstituted or (2) substituted. If a carbon on a group is described as optionally substituted with one or more substituents, one or more hydrogens on the carbon (to the extent any hydrogens are present) may be independently and/or together substituted with independently selected substituents or unsubstituted. If the nitrogen on a group is described as optionally substituted with one or more substituents, then one or more hydrogens on the nitrogen (to the extent any hydrogens are present) may each be substituted or unsubstituted with an independently selected substituent.
When the bond of a substituent is shown as a bond through the ring connecting two atoms, then such substituent may be bonded to any ring-forming atom in the substitutable ring.
The term "stereoisomer" refers to an isomer formed as a result of at least one asymmetric center. In compounds having one or more (e.g., one, two, three, or four) asymmetric centers, it may result in racemates, racemic mixtures, single enantiomers, diastereomeric mixtures, and individual diastereomers. Specific individual molecules may also exist as geometric isomers (cis/trans). Similarly, the compounds of the invention may exist as a mixture of two or more structurally distinct forms (commonly referred to as tautomers) in rapid equilibrium. Representative examples of tautomers include keto-enol tautomers, phenol-keto tautomers, nitroso-oxime tautomers, imine-enamine tautomers, and the like. It is to be understood that the scope of the present application encompasses all such isomers in any ratio (e.g., 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%) or mixtures thereof.
The term "pharmaceutically acceptable" means that the substance or composition must be chemically and/or toxicologically compatible with the other components of the formulation and/or the mammal being treated therewith.
The term "pharmaceutically acceptable salts" includes conventional salts with pharmaceutically acceptable inorganic or organic acids, or inorganic or organic bases.
For a review of suitable salts, see Stahl and Wermuth, "Handbook of Pharmaceutical Salts: properties, selection, and Use" (Wiley-VCH, 2002). Methods for preparing pharmaceutically acceptable salts of the compounds of the invention are known to those skilled in the art.
The term "pharmaceutical composition" includes products comprising a therapeutically effective amount of a compound of the invention, as well as any product that results, directly or indirectly, from a combination of compounds of the invention.
The term "effective amount" refers to an amount sufficient to achieve the desired therapeutic effect, e.g., to achieve a reduction in symptoms associated with the disease to be treated.
The term "treatment" means any administration of a compound of the invention, including:
(1) Preventing disease in an animal that may be predisposed to the disease but has not undergone or displayed disease pathology or symptomology;
(2) Inhibiting the disease (i.e., preventing further development of pathology and/or symptomology) in an animal experiencing or exhibiting disease pathology or symptomology; or alternatively
(3) Disease improvement (i.e., reversal of pathology and/or symptomology) occurs in animals that are experiencing or exhibiting pathology or symptomology of the disease.
The purpose of "treatment" is to reduce or eliminate the disease state or condition for which it is intended. A subject is successfully "treated" if the subject has received a therapeutic amount of a compound, an optical isomer thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, as per the methods described herein, and the subject exhibits an observable and/or detectable decrease or improvement in one or more indications and symptoms. It is also to be understood that the treatment of the disease state or condition described includes not only complete treatment, but also less than complete treatment, but achieves some biologically or medically relevant result.
The term "room temperature" refers to 20 ℃ + -5 ℃.
The term "about" when used to modify a certain value or range of values refers to the value or range of values and ranges of errors that are acceptable to those skilled in the art, e.g., the ranges of errors are + -10%, + -5%, + -4%, + -3%, + -2%, + -1%, + -0.5%, etc.
The invention has the following beneficial effects:
the compound or the pharmaceutical composition has a strong inhibition effect on the activity of the deubiquitinase USP7, and can be used for preventing or treating diseases or symptoms related to USP7 regulation, particularly cancers.
Detailed Description
Embodiments of the present invention will be described in detail below with reference to examples, but it will be understood by those skilled in the art that the following examples are only for illustrating the present invention and should not be construed as limiting the scope of the present invention. The specific conditions are not noted in the examples and are carried out according to conventional conditions or conditions recommended by the manufacturer. The reagents or apparatus used are conventional products available on the market without the manufacturer's attention.
In the conventional synthesis methods, examples, and examples of intermediate synthesis, the meanings of the abbreviations are shown in the following table.
/>
The structures of the compounds described in the examples below were prepared by nuclear magnetic resonance 1 H NMR) or Mass Spectrometry (MS), the instrumentation and operating conditions for nuclear magnetic resonance, mass spectrometry are as follows:
(1) Nuclear magnetic resonance 1 H NMR) was a Bruker 400MHz NMR. The solvent was determined to be deuterated Methanol (Methanol-d) 4 ) Deuterated Chloroform (CD)Cl 3 ) Hexadeuterated dimethyl sulfoxide (DMSO-d) 6 ) Deuterated water (D) 2 O). The internal standard substance is Tetramethylsilane (TMS).
Abbreviations in Nuclear Magnetic Resonance (NMR) spectra represent the following meanings:
s: unimodal (single); d: dual peaks (douplets); t: triplet (triplet); q: quartet (quaternion); dd: double doublets (double doublets); qd: four doublets (quartet douplets); ddd: a double doublet (double double doublet); ddt: double triplet (double double triplet); dddd: double peak (double double double doublet); m: multiple peaks (multiplets); br: broad peak (broad); delta: chemical shift; j: coupling constants; hz: hertz.
All delta values are expressed in ppm values.
(2) The measuring instrument of the Mass Spectrum (MS) is an Agilent (ESI) mass spectrometer, and the model is Agilent6120B.
The thin layer chromatography used in the following examples was a thin layer chromatography silica gel plate (thickness 1mm, specification 200x 200mm, binder sodium carboxymethylcellulose) produced by Nicotiana Xinnuo chemical Co., ltd. The detection instrument was a ZF-20D dark box ultraviolet analyzer, and the detection wavelength was 254nm and 365nm.
The apparatus and operating conditions for preparative high performance liquid chromatography used in the following examples were:
method A:
chromatographic column: waters SunFire Prep C18 OBD 5 μm 19X 150mm;
Mobile phase a: acetonitrile; mobile phase B: water (containing 0.05wt% formic acid);
method B:
chromatographic column: waters SunFire Prep C18 OBD 5 μm 19X 150mm;
mobile phase a: acetonitrile; mobile phase B: water (containing 0.05wt% formic acid);
method C:
chromatographic column: waters SunFire Prep C18 OBD 5 μm 19X 150mm;
mobile phase a: acetonitrile; mobile phase B: water (containing 0.05wt% formic acid);
method D:
chromatographic column: waters SunFire Prep C18 OBD 5 μm 19X 150mm;
mobile phase a: acetonitrile; mobile phase B: water (containing 0.05wt% formic acid);
method E:
chromatographic column: waters SunFire Prep C18 OBD 5 μm 19X 150mm;
mobile phase a: acetonitrile; mobile phase B: water (containing 0.05wt% formic acid);
method F:
chromatographic column: waters XBridge Prep C18 OBD 5 μm 19X 150mm;
mobile phase a: acetonitrile; mobile phase B: water (containing 0.05wt% formic acid);
/>
method G:
chromatographic column: waters SunFire Prep C18 OBD 5 μm 19X 150mm;
mobile phase a: acetonitrile; mobile phase B: water (containing 0.05wt% trifluoroacetic acid);
method I:
chromatographic column: waters SunFire Prep C18 OBD 5 μm 19X 150mm;
mobile phase a: acetonitrile; mobile phase B: water (containing 0.05wt% formic acid);
method J:
chromatographic column: waters SunFire Prep C18 OBD 5 μm 19X 150mm;
Mobile phase a: acetonitrile; mobile phase B: water (containing 0.05wt% trifluoroacetic acid);
method K:
chromatographic column: waters SunFire Prep C18 OBD 5 μm 19X 150mm;
mobile phase a: acetonitrile; mobile phase B: water (containing 0.05wt% formic acid);
method L:
chromatographic column: waters SunFire Prep C18 OBD 5 μm 19X 150mm;
mobile phase a: acetonitrile; mobile phase B: water (containing 0.05wt% formic acid);
method M:
chromatographic column: waters SunFire Prep C18 OBD 5 μm 19X 150mm;
mobile phase a: acetonitrile; mobile phase B: water (containing 0.05wt% trifluoroacetic acid);
method N:
chromatographic column: waters SunFire Prep C18 OBD 5 μm 19X 150mm;
mobile phase a: acetonitrile; mobile phase B: water (containing 0.05wt% trifluoroacetic acid);
method O:
chromatographic column: waters XBridge Prep C18 OBD 5 μm 19X 150mm;
mobile phase a: acetonitrile; mobile phase B: water (containing 0.05wt% formic acid);
method P:
chromatographic column: waters SunFire Prep C18 OBD 5 μm 19X 150mm;
mobile phase a: acetonitrile; mobile phase B: water (containing 0.05wt% formic acid);
/>
example 1 (R) -3- ((4-hydroxy-1- (3-phenylbutyryl) piperidin-4-yl) methyl) -7- (1H-indazol-6- Base) thieno [3,4-d]Synthesis of pyrimidin-4 (3H) -one (compound 1)
Step one: synthesis of (R) -7-bromo-3- ((4-hydroxy-1- (3-phenylbutyryl) piperidin-4-yl) methyl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 1-2)
Compounds 1-1 (0.13 g,0.56 mmol) were reacted at room temperature,(R) -3-phenyl-1- (1-oxa-6-aza-spiro [ 2.5)]Octyl-6-yl) butanone (synthetic method reference WO 2018073602) (0.15 g,0.56 mmol) was dissolved in DMF (4 mL), potassium carbonate (0.16 g,1.13 mmol) was added and the reaction was warmed to 80℃and reacted for 8h. The reaction mixture was cooled to room temperature, purified by preparative high performance liquid chromatography (method D), and lyophilized to give the title compound (0.14 g). ESI-MS (m/z): 490.1,492.1[ M+H ]] +
Step two: synthesis of (R) -3- ((4-hydroxy-1- (3-phenylbutyryl) piperidin-4-yl) methyl) -7- (1H-indazol-6-yl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 1)
Compounds 1-2 (40 mg,0.08 mmol), (1H-indazol-6-yl) boronic acid (16 mg,0.10 mmol), potassium carbonate (31 mg,0.24 mmol) were added sequentially to the reaction flask at room temperature, followed by 1, 4-dioxane (2 mL) and water (0.5 mL), and finally tetrakis triphenylphosphine palladium (8.0 mg,0.008 mmol) was added and replaced with nitrogen three times. The mixture was heated to 80℃under nitrogen and stirred for 4h. The reaction solution was cooled to room temperature, the reaction solution was concentrated under reduced pressure, and the purified product was isolated by preparative high performance liquid chromatography (method D) and lyophilized to give the title compound (32 mg).
The structural characterization is as follows:
1 H NMR(400MHz,DMSO-d 6 )δ13.18(s,1H),8.51(s,1H),8.45(s,1H),8.12-8.04(m,2H),7.84(d,J=8.0Hz,1H),7.69(d,J=8.0Hz,1H),7.29-7.10(m,5H),4.91(d,J=4.0Hz,1H),4.09-3.85(m,3H),3.72-3.61(m,1H),3.26-3.12(m,2H),2.94-2.81(m,1H),2.66-2.53(m,2H),1.59-1.27(m,4H),1.24-1.16(m,3H).ESI-MS(m/z):528.3[M+H] +
example two (R) -3- ((4-hydroxy-1- (3-phenylbutyryl) piperidin-4-yl) methyl) -7- (4-hydroxyphenyl) Base) thieno [3,4-d]Synthesis of pyrimidin-4 (3H) -one (compound 2)
Compounds 1-2 (30 mg,0.06 mmol), 4-hydroxyphenylboronic acid (20 mg,0.09 mmol), potassium carbonate (25 mg,0.18 mmol) were added sequentially to the reaction flask at room temperature, followed by 1, 4-dioxane (4 mL) and water (0.8 mL). Finally, tetrakis triphenylphosphine palladium (7.1 mg, 0.006mmol) was added and replaced three times with nitrogen. The mixture was heated to 80℃under nitrogen and stirred for 4h. The reaction solution was cooled to room temperature, and the reaction solution was concentrated under reduced pressure, and purified by preparative high performance liquid chromatography (method D), and freeze-dried to give the title compound (20 mg).
The structural characterization is as follows:
1 H NMR(400MHz,DMSO-d 6 )δ9.74(s,1H),8.34(s,1H),7.97(d,J=10.4Hz,1H),7.91-7.83(m,2H),7.28-7.22(m,4H),7.20-7.09(m,1H),6.91-6.82(m,2H),4.89(d,J=4.8Hz,1H),4.10-3.99(m,1H),3.99-3.80(m,2H),3.70-3.60(m,1H),3.28-3.09(m,2H),2.87-2.81(m,1H),2.71-2.52(m,2H),1.57-1.29(m,3H),1.25-1.16(m,4H).ESI-MS(m/z):504.2[M+H] +
example III (R) -7- (4- (aminomethyl) phenyl) -3- ((4-hydroxy-1- (3-phenylbutyryl) piperidine-4-) Methyl) thieno [3,4-d ]]Synthesis of pyrimidin-4 (3H) -one (compound 3)
Step one: synthesis of tert-butyl (R) -4- (3- ((4-hydroxy-1- (3-phenylbutyryl) piperidin-4-yl) methyl-4-oxo-3, 4-dihydrothieno [3,4-d ] pyrimidin-7-yl) benzyl carbamate (compound 3-1)
According to the operation described in example II, using compounds 1-2 (20 mg,0.04 mmol) and 4- (N-t-butoxycarbonylaminomethyl) phenylboronic acid (15 mg,0.06 mmol) as reaction materials in place of compounds 1-2 (30 mg,0.06 mmol) and 4-hydroxyphenylboronic acid, respectively, the reaction solution was concentrated under reduced pressure to obtain crude products of the title compound, 21mg, which was directly used in the next reaction. ESI-MS (m/z): 617.3[ M+H ] ] +
Step two: synthesis of (R) -7- (4- (aminomethyl) phenyl) -3- ((4-hydroxy-1- (3-phenylbutyryl) piperidin-4-yl) methyl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 3)
To crude compound 3-1 (21 mg,0.03 mmol) was added hydrochloric acid/1, 4-dioxane solution (5 mL, hydrochloric acid concentration: 4 mol/L), and the mixture was reacted at room temperature for 1h. Preparation type high performance liquid chromatography separation and purification (method F), adding dilute hydrochloric acid (0.5 mL,1 mol/L) into the preparation, and freeze drying to obtain hydrochloride of the title compound, 10mg.
The structural characterization is as follows:
1 H NMR(400MHz,DMSO-d 6 )δ8.53(s,1H),8.37(s,3H),8.17-8.02(m,3H),7.58(d,J=8.4Hz,2H),7.29-7.25(m,4H),7.17-7.12(m,1H),4.10-3.86(m,5H),3.71-3.62(m,2H),3.26-3.12(m,2H),2.91-2.84(m,1H),2.69-2.54(m,2H),1.58-1.23(m,4H),1.20(d,J=6.2Hz,3H).ESI-MS(m/z):517.3[M+H] +
example tetra 7- (1-amino-2, 3-dihydro-1H-inden-5-yl) -3- ((4-hydroxy-1- ((R) -3-phenylbutyryl) Group) piperidin-4-yl) methyl) thieno [3,4-d]Synthesis of pyrimidin-4 (3H) -one (compound 4)
Step one: synthesis of tert-butyl (5- (3- ((4-hydroxy-1- ((R) -3-phenylbutyryl) piperidin-4-yl) methyl) -4-oxo-3, 4-dihydrothieno [3,4-d ] pyrimidin-7-yl) -2, 3-dihydro-1H-inden-1-yl) carbamate (Compound 4-1)
According to the procedure described in example one, step two, the reaction was carried out using compound 1-2 (40 mg,0.11 mmol) and tert-butyl 5- (4, 5-tetramethyl-1, 3, 2-dioxaborane-2-yl) -2, 3-dihydro-1H-inden-1-yl) carbamate (46 mg,0.09 mmol) as reaction starting materials instead of compound 1-2 and (1H-indazol-6-yl) boric acid, respectively, and the reaction mixture was concentrated under reduced pressure and purified by silica gel column chromatography (eluent: dichloromethane/methanol=15/1 (v/v) to give the title compound, 45mg. ESI-MS (m/z): 643.4[ M+H ] ] +
Step two: synthesis of 7- (1-amino-2, 3-dihydro-1H-inden-5-yl) -3- ((4-hydroxy-1- ((R) -3-phenylbutyryl) piperidin-4-yl) methyl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 4)
According to the procedure described in step two of example three, the reaction was carried out using compound 4-1 (30 mg,0.05 mmol) as a reaction starting material instead of compound 3-1, to prepare a high performance liquid chromatography for separation and purification (method C), and the preparation was concentrated under reduced pressure and freeze-dried to give the formate salt of the title compound, 20mg.
The structural characterization is as follows:
1 H NMR(400MHz,DMSO-d 6 )δ8.47(s,1H),8.43(s,1H),8.02(d,J=12.0Hz,1H),7.97(s,1H),7.92(d,J=8.0Hz,1H),7.58(d,J=8.0Hz,1H),7.30-7.21(m,4H),7.19-7.11(m,1H),4.59(t,J=6.8Hz,1H),4.10-3.98(m,1H),3.94(d,J=8.0Hz,1H),3.88(d,J=14.4Hz,1H),3.82(d,J=14.0Hz,1H),3.30-3.01(m,3H),2.98-2.78(m,2H),2.72-2.55(m,2H),2.49-2.42(m,1H),2.00-1.85(m,1H),1.46-1.10(m,4H),1.21(dd,J=6.8,4.0Hz,3H).ESI-MS(m/z):543.3[M+H] +
example five 3- ((4-hydroxy-1- ((R) -3-phenylbutyryl) piperidin-4-yl) methyl) -7- (1- (methamine) Phenyl) -2, 3-dihydro-1H-inden-5-yl thieno [3,4-d]Synthesis of pyrimidin-4 (3H) -one (compound 5)
Step one: synthesis of N-methyl-5- (4, 5-tetramethyl-1, 3, 2-dioxaborane-2-yl) -2, 3-dihydro-1H-inden-1-amine (Compound 5-2)
Compound 5-1 (2.00 g,6.59 mmol) was dissolved in methanol (80 mL), a solution of methylamine in ethanol (7 mL,33% wt.) was added, the reaction was carried out at room temperature for 12h, formic acid (0.80 mL,21.08 mmol) was added to adjust pH.apprxeq.5, sodium cyanoborohydride (1.24 g,19.76 mmol) was further added, the reaction was continued at room temperature for 12h, after concentrating the reaction solution under reduced pressure, the filtrate was concentrated under reduced pressure to give crude formate of the title compound, 2.08g, which was directly used for the next reaction. ESI-MS (m/z): 274.3[ M+H ] ] +
Step two: synthesis of tert-butyl (4, 5-tetramethyl-1, 3, 2-dioxaborane-2-yl) -2, 3-dihydro-1H-inden-1-yl) carbamate (Compound 5-3)
The crude formate salt of Compound 5-2 (2.03 g,6.36 mmol) was dissolved in tetrahydrofuranTo the furan (40 mL), saturated aqueous sodium hydrogencarbonate (6.36 mmol,40 mL) and di-tert-butyl dicarbonate (1.67 g,7.63 mmol) were added in this order, and the mixture was reacted at room temperature for 6 hours. The reaction mixture was extracted with ethyl acetate (3×100 ml), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated under reduced pressure, and then purified by column chromatography on silica gel (eluent: petroleum ether/ethyl acetate=9/1 (v/v)) to give the title compound, 0.87g. ESI-MS (m/z): 318.2[ M-56+H] +
Step three: synthesis of tert-butyl (5- (3- ((4-hydroxy-1- ((R) -3-phenylbutyryl) piperidin-4-yl) methyl) -4-oxo-3, 4-dihydrothieno [3,4-d ] pyrimidin-7-yl) -2, 3-dihydro-1H-inden-1-yl) (methyl) carbamate (Compound 5-4)
According to the procedure described in example II, the reaction was carried out using the compound 5-3 (57 mg,0.15 mmol) as a reaction starting material instead of 4-hydroxyphenylboronic acid, and the reaction solution was separated and purified by preparative high performance liquid chromatography (method E) and freeze-dried to give the title compound, 60mg. ESI-MS (m/z): 657.4[ M+H ] ] +
Step four: synthesis of 3- ((4-hydroxy-1- ((R) -3-phenylbutyryl) piperidin-4-yl) methyl) -7- (1- (methylamino) -2, 3-dihydro-1H-inden-5-yl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 5)
According to the procedure described in step two of example three, compound 5-4 (45 mg,0.07 mmol) was used as a reaction starting material instead of compound 3-1 to conduct a reaction, preparative high performance liquid chromatography was used for separation and purification (method C), diluted hydrochloric acid (0.5 mL,1 mol/L) was added to the preparation, and freeze-drying was performed to obtain hydrochloride of the title compound, 40mg.
The structural characterization is as follows:
1 H NMR(400MHz,DMSO-d 6 )δ9.45(br,1H),9.30(br,1H),8.52(s,1H),8.09(d,J=10.3Hz,1H),8.04(s,1H),7.98(d,J=8.2Hz,1H),7.78(d,J=8.0Hz,1H),7.33-7.20(m,4H),7.20-7.09(m,1H),4.72(br,1H),4.09-3.83(m,3H),3.72-3.59(m,1H),3.30-3.11(m,3H),3.02-2.83(m,2H),2.68-2.53(m,5H),2.49-2.39(m,2H),2.26-2.16(m,1H),1.58-1.22(m,4H),1.20(d,J=5.9Hz,3H).ESI-MS(m/z):557.4[M+H] +
example six 7- (2-amino-2, 3-dihydro-1H-inden-5-yl) -3- ((4-hydroxy-1- ((R) -3-phenylbutyryl) Group) piperidin-4-yl) methyl) thieno [3,4-d]Synthesis of pyrimidin-4 (3H) -one (compound 6)
Step one: synthesis of tert-butyl (5- (3- ((4-hydroxy-1- ((R) -3-phenylbutyryl) piperidin-4-yl) methyl) -4-oxo-3, 4-dihydrothieno [3,4-d ] pyrimidin-7-yl) -2, 3-dihydro-1H-inden-2-yl) carbamate (Compound 6-1)
According to the procedure described in step two of example one, the reaction was carried out using tert-butyl 5- ((4, 5-tetramethyl-1, 3, 2-dioxaborane-2-yl) -2, 3-dihydro-1H-inden-2-yl) carbamate (33 mg,0.09 mmol) as starting material instead of (1H-indazol-6-yl) boronic acid, and the reaction solution was concentrated under reduced pressure to give crude product of the title compound, 39mg, which was directly used in the next reaction. ESI-MS (m/z): 643.3[ M+H ] ] +
Step two: synthesis of 7- (2-amino-2, 3-dihydro-1H-inden-5-yl) -3- ((4-hydroxy-1- ((R) -3-phenylbutyryl) piperidin-4-yl) methyl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 6)
According to the procedure described in step two of example three, the reaction was carried out using crude compound 6-1 (39 mg,0.06 mmol) as a reaction starting material instead of compound 3-1, the reaction mixture was purified by preparative liquid phase separation (method F), diluted hydrochloric acid (0.5 mL,1 mol/L) was added to the preparation, and freeze-dried to give hydrochloride of the title compound, 21mg.
The structural characterization is as follows:
1 H NMR(400MHz,DMSO-d 6 )δ8.46(s,1H),8.39(d,J=5.6Hz,3H),8.10(d,J=10.4Hz,1H),7.95(s,1H),7.93-7.88(m,1H),7.38(d,J=8.0Hz,1H),7.29-7.25(m,4H),7.20-7.11(m,1H),4.11-3.81(m,4H),3.73-3.59(m,1H),3.33(td,J=16.5,7.6Hz,2H),3.27-3.13(m,2H),3.06(td,J=16.3,5.3Hz,2H),2.91-2.84(m,1H),2.69-2.53(m,2H),1.58-1.22(m,4H),1.20(d,J=6.7Hz,3H).ESI-MS(m/z):543.3[M+H] +
example seven 3- ((4-hydroxy-1- ((R) -3-phenylbutyryl) piperidin-4-yl) methyl) -7- (2- (methylamino) Phenyl) -2, 3-dihydro-1H-inden-5-yl thieno [3,4-d]Synthesis of pyrimidin-4 (3H) -one (compound 7)
Step one: synthesis of tert-butyl 5-bromo-2, 3-dihydro-1H-indene-2-methylcarbamate (Compound 7-2)
Compound 7-1 (700 mg,2.24 mmol) was weighed out and dissolved in DMF (14 mL), cooled to 0℃and sodium hydride (135 mg,3.36mmol,60% purity) was added and the reaction was maintained at this temperature for 1h. Methyl iodide (382 mg,2.69 mmol) was added thereto, and the mixture was slowly warmed to room temperature and reacted for 16 hours. The reaction mixture was quenched with saturated aqueous ammonium chloride (100 mL), extracted with ethyl acetate (3X 50 mL), and the organic phases were combined and washed with saturated brine (3X 50 mL). The organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated to give the title compound, 600mg, which was used directly in the next reaction. ESI-MS (m/z): 270.0,272.0[ M-56+H ] ] +
Step two: synthesis of tert-butyl (5- (4, 5-tetramethyl-1, 3, 2-dioxaborane-2-yl) -2, 3-dihydro-1H-inden-2-yl) -methylcarbamate (Compound 7-3)
Compound 7-2 (200 mg,0.61 mmol), pinacol biborate (187 mg,0.74 mmol), potassium acetate (181 mg,1.84 mmol) were weighed into 1, 4-dioxane (5 mL), nitrogen was replaced 3 times, and Pd (dppf) Cl was added 2 (22 mg,0.03 mmol), nitrogen was substituted three times, and the temperature was raised to 80℃under a nitrogen atmosphere to react for 16 hours. After the reaction solution was cooled to room temperature, the reaction solution was concentrated under reduced pressure, and the resultant was separated and purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate=10/1 (v/v)), whereby 200mg of the title compound was obtained. ESI-MS (m/z): 318.2[ M-56+H] +
Step three: synthesis of tert-butyl (5- (3- ((4-hydroxy-1- ((R) -3-phenylbutyryl) piperidin-4-yl) methyl) -4-oxo-3, 4-dihydrothieno [3,4-d ] pyrimidin-7-yl) -2, 3-dihydro-1H-inden-2-yl) (methyl) carbamate (Compound 7-4)
According to one step of the embodimentThe procedure described in II was followed using 7-3 (23 mg,0.06 mmol) as a starting material instead of (1H-indazol-6-yl) boronic acid, and the reaction mixture was purified by preparative thin layer chromatography (developer: dichloromethane/methanol=15/1 (v/v)) to give the title compound, 10mg. ESI-MS (m/z): 657.3[ M+H ] ] +
Step four: synthesis of 3- ((4-hydroxy-1- ((R) -3-phenylbutyryl) piperidin-4-yl) methyl) -7- (2- (methylamino) -2, 3-dihydro-1H-inden-5-yl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 7)
According to the procedure described in step two of example three, the reaction was carried out using compound 7-4 (10 mg,0.02 mmol) as a reaction starting material instead of compound 3-1, the reaction solution was separated and purified by preparative high performance liquid chromatography (method C), diluted hydrochloric acid (0.5 mL,1 mol/L) was added to the preparation solution, and freeze-drying was carried out to obtain hydrochloride of the title compound, 8mg.
The structural characterization is as follows:
1 H NMR(400MHz,DMSO-d 6 )δ9.47-9.21(m,2H),8.46(s,1H),8.09(d,J=10.4Hz,1H),7.95(s,1H),7.90(dd,J=7.9,1.7Hz,1H),7.39(t,J=8.2Hz,1H),7.32-7.21(m,4H),7.19-7.12(m,1H),4.12-3.79(m,4H),3.66(t,J=11.7Hz,1H),3.34(td,J=16.1,7.8Hz,2H),3.28-3.07(m,4H),2.86(t,J=11.2Hz,1H),2.59(m,4H),2.48-2.52(m,1H),1.57-1.22(m,4H),1.20(dd,J=7.0,1.7Hz,3H).ESI-MS(m/z):557.3[M+H] +
example eight 7- (2- (dimethylamino) -2, 3-dihydro-1H-inden-5-yl) -3- ((4-hydroxy-1- ((R) -3-benzene Dibutyryl) piperidin-4-yl) methyl) thieno [3,4-d]Synthesis of pyrimidin-4 (3H) -one (compound 8)
Step one: synthesis of 5- (4, 5-tetramethyl-1, 3, 2-dioxaborane-2-yl) -1H-inden-2 (3H) -one (Compound 8-2)
According to the procedure described in example seven, step two, the reaction was carried out using compound 8-1 (1.00 g,4.74 mmol) as starting material instead of compound 7-2Purification by column chromatography on silica gel (eluent: petroleum ether/ethyl acetate=10/1 (v/v)) afforded the title compound, 1.10g. ESI-MS (m/z): 259.1[ M+H ]] +
Step two: synthesis of 2- ((dimethylamino) -2, 3-dihydro-1H-inden-5-yl) boronic acid (Compound 8-3)
To methanol (5 mL) was added compound 8-2 (100 mg,0.39 mmol) and a methanol solution of dimethylamine (5 mL, 2M), and after reacting at room temperature for 20 hours, sodium borohydride (43 mg,1.16 mmol) was added, and the reaction was continued for 2 hours. The reaction solution was purified by preparative high performance liquid chromatography (method C) and freeze-dried to give the formate of the title compound, 10mg. ESI-MS (m/z): 206.2[ M+H ]] +
Step three: synthesis of 7- (2- (dimethylamino) -2, 3-dihydro-1H-inden-5-yl) -3- ((4-hydroxy-1- ((R) -3-phenylbutyryl) piperidin-4-yl) methyl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 8)
According to the procedure described in step two of example one, the reaction was carried out using formate salt of compound 8-3 (10 mg,0.05 mmol) as the starting material instead of (1H-indazol-6-yl) boric acid, and the reaction mixture was purified by preparative high performance liquid chromatography (method C) and freeze-dried to give formate salt of the title compound, 11mg.
The structural characterization is as follows:
1 H NMR(400MHz,DMSO-d 6 )δ8.43(s,1H),8.21(s,1H),8.01(d,J=10.3Hz,1H),7.87(s,1H),7.80(d,J=7.8Hz,1H),7.31-7.21(m,5H),7.17-7.12(m,1H),4.92(s,2H),4.08-4.01(m,1H),4.00-3.89(m,1H),3.88-3.83(m,1H),3.69-3.63(m,1H),3.27-3.14(m,2H),3.10-3.00(m,2H),2.90-2.76(m,3H),2.66-2.56(m,2H),2.22(s,6H),1.56-1.26(m,4H),1.20(d,J=6.8Hz,3H).ESI-MS(m/z):571.3[M+H] +
example nine (R) -3- ((4-hydroxy-1- (3-phenylbutyryl) piperidin-4-yl) methyl) -7- (isoindoline ] 5-yl) thieno [3,4-d]Synthesis of pyrimidin-4 (3H) -one (compound 9)
Step one: synthesis of tert-butyl (R) -5- (3- ((4-hydroxy-1- (3-phenylbutyryl) piperidin-4-yl) methyl) -4-oxo-3, 4-dihydrothieno [3,4-d ] pyrimidin-7-yl) isoindoline-2-carboxylate (compound 9-1)
According to the procedure described in example one, step two, using tert-butyl 5- (4, 5-tetramethyl-1, 3, 2-dioxaborane-2-yl) isoindoline-2-carboxylate (32 mg,0.09 mmol) as a reaction starting material instead of (1H-indazol-6-yl) boronic acid, the reaction solution was separated and purified by preparative thin layer chromatography (developing reagent: dichloromethane/methanol=10/1 (v/v)) to give the title compound, 16mg. ESI-MS (m/z): 629.3[ M+H ]] +
Step two: synthesis of (R) -3- ((4-hydroxy-1- (3-phenylbutyryl) piperidin-4-yl) methyl) -7- (isoindolin-5-yl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 9)
According to the procedure described in step two of example seven, the reaction was carried out using compound 9-1 (16 mg,0.03 mmol) as a reaction starting material instead of compound 7-2, the reaction solution was separated and purified by preparative high performance liquid chromatography (method C), diluted hydrochloric acid (0.5 mL,1 mol/L) was added to the preparation solution, and freeze-drying was carried out to obtain hydrochloride of the title compound, 12mg.
The structural characterization is as follows:
1 H NMR(400MHz,DMSO-d 6 )δ9.72(t,J=5.8Hz,2H),8.52(s,1H),8.11(s,1H),8.07(d,J=8Hz,2H),7.51(d,J=8.1Hz,1H),7.25(p,J=3.1Hz,4H),7.15(dt,J=5.4,2.6Hz,1H),4.93(s,1H),4.58(t,J=5.6Hz,2H),4.54(t,J=5.6Hz,2H),4.03(t,J=8.4Hz,1H),3.98-3.83(m,2H),3.65(s,1H),3.28-3.13(m,2H),2.87(t,J=11.4Hz,1H),2.68-2.53(m,2H),1.58-1.24(m,4H),1.20(dd,J=6.9,1.6Hz,3H).ESI-MS(m/z):529.3[M+H] +
examples ten 3- ((1- (4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butanoyl) -4-hydroxypiperidine-4- Methyl) -7- (1- (methylamino) -2, 3-dihydro-1H-inden-5-yl) thieno [3,4-d]Pyrimidin-4 (3H) -ones (compounds 10 ) synthesis of
Step one: synthesis of tert-butyl 4- ((7-bromo-4-oxothieno [3,4-d ] pyrimidin-3 (4H) -yl) methyl) -4-hydroxypiperidine-1-carboxylate (compound 10-1)
According to the procedure described in example one, step one, 1-oxa-6-aza-spiro [2.5]Octyl-6-carboxylic acid tert-butyl ester (1.38 g,6.49 mmol) as reaction starting material instead of compound (R) -3-phenyl-1- (1-oxa-6-aza-spiro [ 2.5)]Octyl-6-yl) butanone was reacted, the reaction solution was poured into water (100 mL), a solid was precipitated, suction filtration was performed, and the cake was washed with petroleum ether (3X 10 mL) to give the title compound, 1.45g. ESI-MS (m/z): 388.0,390.0[ M-56+H ]] +
Step two: synthesis of 7-bromo-3- ((4-hydroxypiperidin-4-yl) methyl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 10-2)
Compound 10-1 (200 mg,0.45 mmol) was dissolved in dichloromethane (2 mL), and a hydrochloric acid/ethyl acetate solution (2 mL, hydrochloric acid concentration: 4 mol/L) was added dropwise to react at room temperature for 2h. After concentrating the reaction solution under reduced pressure, 150mg of the hydrochloride of the title compound was obtained. The reaction mixture was used in the next reaction without purification. ESI-MS (m/z): 344.0,346.0[ M+H ]] +
Step three: synthesis of 7-bromo-3- ((1- (4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butanoyl) -4-hydroxypiperidin-4-yl) methyl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 10-3)
To a solution of 4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butyric acid (102 mg,0.49 mmol) in methylene chloride (5 mL) were added HATU (178 mg,0.47 mmol) and DIPEA (173 mg,1.34 mmol) successively, and after 5 minutes of reaction, hydrochloride (0.17 g,0.45 mmol) of compound 10-2 was added, and after 2 hours of reaction at room temperature, the reaction mixture was concentrated under reduced pressure, and the preparative high performance liquid chromatography was purified (method B) and freeze-dried to give the title compound, 127mg. ESI-MS (m/z): 534.0,536.0[ M+H ] ] +
Step four: synthesis of tert-butyl (5- (3- ((1- (4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butanoyl-4-hydroxypiperidin-4-yl) methyl) -4-oxo-3, 4-dihydrothieno [3,4-d ] pyrimidin-7-yl) -2, 3-dihydro-1H-inden-1-yl) (methyl) carbamate (Compound 10-4)
According to the procedure described in step two of example one, using compound 10-3 (60 mg,0.07 mmol) and compound 5-3 (50 mg,0.13 mmol) as reaction starting materials, instead of compounds 1-2 and (1H-indazol-6-yl) boric acid, respectively, the title compound was obtained as 80mg by separating and purifying by silica gel column chromatography (eluent: dichloromethane/methanol=19/1 (v/v)). ESI-MS (m/z): 701.2[ M+H ]] +
Step five: synthesis of 3- ((1- (4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butanoyl) -4-hydroxypiperidin-4-yl) methyl) -7- (1- (methylamino) -2, 3-dihydro-1H-inden-5-yl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 10)
According to the procedure described in the tenth step of example, compound 10-4 (80 mg,0.05 mmol) was used as a reaction starting material instead of compound 10-1, and the reaction mixture was purified by preparative high performance liquid chromatography (method C), diluted hydrochloric acid (0.5 mL,1 mol/L) was added to the preparation, and freeze-dried to give hydrochloride of the title compound, 48mg.
The structural characterization is as follows:
1 H NMR(400MHz,DMSO-d 6 )δ9.56(s,1H),9.41(s,1H),8.51(d,J=1.6Hz,1H),8.13(d,J=3.9Hz,1H),8.04(d,J=1.6Hz,1H),7.98(dd,J=8.0,1.7Hz,1H),7.85-7.78(m,2H),6.26(td,J H-F =55.2Hz,J H-H =3.8Hz,1H),6.05-5.95(m,1H),4.78-4.98(m,1H),4.08-3.88(m,3H),3.69(d,J=13.6Hz,1H),3.35-3.15(m,3H),3.0-2.84(m,3H),2.57(t,J=5.3Hz,3H),2.50-2.42(m,2H),2.28-2.15(m,1H),1.65-1.30(m,4H).ESI-MS(m/z):601.2[M+H] +
example eleven 3- ((1- (4, 4-difluoro-3-phenylbutyryl) -4-hydroxypiperidin-4-yl) methyl) -7- (1- (methylamino) -2, 3-dihydro-1H-inden-5-yl) thieno [3,4-d]Synthesis of pyrimidin-4 (3H) -one (compound 11)
Step one: synthesis of tert-butyl 4-hydroxy-4- ((4-oxo-7- (1-oxo-2, 3-dihydro-1H-inden-5-yl) thieno [3,4-d ] pyrimidin-3 (4H) -yl) methyl) piperidine-1-carboxylate (compound 11-1)
According to the procedure described in example one, step two, starting from compound 10-1 (378 mg,0.85 mmol) and 5- (4, 5-tetramethyl-1, 3, 2-dioxaborane-2-yl) -2, 3-dihydro-1H-inden-1-one (329 mg,1.28 mmol) in place of compounds 1-2 and (1H-indazol-6-yl) boronic acid, respectively, the title compound was obtained by separation and purification by silica gel column chromatography (eluent: ethyl acetate/methanol=25/1 (v/v)). ESI-MS (m/z): 496.1[ M+H ]] +
Step two: synthesis of 3- ((4-hydroxypiperidin-4-yl) methyl) -7- (1-oxo-2, 3-dihydro-1H-inden-5-yl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 11-2)
According to the procedure described in step two of example three, the reaction was carried out using compound 11-1 (420 mg,0.85 mmol) as a reaction starting material instead of compound 3-1, the reaction solution was concentrated under reduced pressure, ethyl acetate was added, stirred at room temperature, and suction-filtered to give the hydrochloride of the title compound, 360mg. ESI-MS (m/z): 396.1[ M+H ] ] +
Step three: synthesis of 3- ((1- (4, 4-difluoro-3-phenylbutyryl) -4-hydroxypiperidin-4-yl) methyl) -7- (1-oxo-2, 3-dihydro-1H-inden-5-yl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 11-3)
4, 4-difluoro-3-phenylbutyric acid (15 mg,0.07 mmol) was weighed and dissolved in DMF (1 mL), HATU (28 mg,0.07 mmol), compound 11-2 hydrochloride (30 mg,0.07 mmol) and DIPEA (27 mg,0.21 mmol) were added sequentially and reacted at room temperature for 1h. The reaction solution was purified by preparative high performance liquid chromatography (method D) and freeze-dried to give the title compound (34 mg). ESI-MS (m/z): 578.2[ M+H ]] +
Step four: synthesis of 3- ((1- (4, 4-difluoro-3-phenylbutyryl) -4-hydroxypiperidin-4-yl) methyl) -7- (1- (methylamino) -2, 3-dihydro-1H-inden-5-yl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 11)
Compound 11-3 (16 mg,0.03 mmol) was dispersed in methanol (5 mL) and methylene chloride (1 mL), and an ethanol solution of methylamine (26 mg,0.83 mmol) was added thereto and reacted at room temperature for 16h. Sodium cyanoborohydride (5 mg,0.08 mmol) was added, glacial acetic acid was added dropwise to adjust the pH to approximately 5, and the reaction was continued at room temperature for 16h. The reaction mixture was concentrated under reduced pressure, and then purified by preparative high performance liquid chromatography (method J), and freeze-dried to give the title compound as trifluoroacetate salt, 11mg.
The structural characterization is as follows:
1 H NMR(400MHz,DMSO-d 6 )δ8.90-8.72(m,2H),8.56-8.49(m,1H),8.11-7.96(m,3H),7.71-7.62(m,1H),7.35-7.25(m,5H),6.23(td,J H- F=56.6Hz,J H-H =3.9Hz,1H),4.92(s,1H),4.82-4.72(m,1H),4.06-3.92(m,2H),3.87(s,1H),3.76-3.58(m,2H),3.25-3.08(m,2H),3.01-2.87(m,3H),2.86-2.75(m,1H),2.69-2.62(m,3H),2.23-2.12(m,1H),1.61-1.14(m,5H).ESI-MS(m/z):593.2[M+H] +
example twelve (3- (1- (4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butanoyl) -4-hydroxypiperidine-4-) Methyl) -7- (2- (methylamino) -2, 3-dihydro-1H-inden-5-yl) thieno [3,4-d]Pyrimidin-4 (3H) -ones (compounds 12 ) synthesis of
Step one: synthesis of tert-butyl (5- (3- ((1- (4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butanoyl) -4-hydroxypiperidin-4-yl) methyl) -4-oxo-3, 4-dihydrothieno [3,4-d ] pyrimidin-7-yl) -2, 3-dihydro-1H-inden-2-yl) (methyl) carbamate (Compound 12-1)
According to the procedure described in step two of example one, using compound 10-3 (20 mg,0.04 mmol) and compound 7-3 (21 mg,0.06 mmol) as reaction starting materials, instead of compound 1-2 and (1H-indazol-6-yl) boric acid, respectively, the reaction solution was concentrated under reduced pressure, and then purified by preparative thin layer chromatography (eluent: dichloromethane/methanol=20/1 (v/v)) to give the title compound, 14mg. ESI-MS (m/z): 701.3[ M+H ]] +
Step two: synthesis of (3- (1- (4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butanoyl) -4-hydroxypiperidin-4-yl) methyl) -7- (2- (methylamino) -2, 3-dihydro-1H-inden-5-yl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 12)
According to the procedure described in step two of example ten, the reaction was carried out using compound 12-1 (14 mg,0.02 mmol) as a reaction starting material instead of compound 10-1, and after concentrating the reaction solution under reduced pressure, it was separated and purified by preparative high performance liquid chromatography (method A), and diluted hydrochloric acid (0.5 mL,1 mol/L) was added to the prepared solution, and freeze-dried to give the hydrochloride of the title compound, 5mg.
The structural characterization is as follows:
1 H NMR(400MHz,DMSO-d 6 )δ9.03(t,J=7.0Hz,2H),8.46(d,J=1.5Hz,1H),8.05(d,J=4.3Hz,1H),7.96(s,1H),7.91(dd,J=7.8,1.7Hz,1H),7.81(dt,J=4.1,2.5Hz,1H),7.38(d,J=8.0Hz,1H),6.26(td,J H - F =56.0Hz,J H-H =4.0H z,1H),5.98(dd,J=5.9,2.5Hz,1H),5.09-4.86(m,2H),4.01(p,J=6.3Hz,2H),3.93(d,J=10.1Hz,2H),3.69(d,J=13.6Hz,1H),3.39-3.31(m,2H),3.31-3.20(m,2H),3.13(td,J=17.3,6.1Hz,2H),2.98-2.83(m,2H),2.62(t,J=5.4Hz,3H),1.66-1.50(m,1H),1.50-1.29(m,3H).ESI-MS(m/z):601.2[M+H] +
example thirteen 3- ((4-hydroxy-1- (4, 4-trifluoro-3-phenylbutyryl) piperidin-4-yl) methyl) -7- (2- (methylamino) -2, 3-dihydro-1H-inden-5-yl) thieno [3,4-d]Synthesis of pyrimidin-4 (3H) -one (compound 13)
Step one: synthesis of 7-bromo-3- ((4-hydroxy-1- (4, 4-trifluoro-3-phenylbutyryl) piperidin-4-yl) methyl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 13-1)
According to the procedure described in step three of example eleven, using the hydrochloride salt of compound 10-2 (200 mg,0.53 mmol) and 4, 4-trifluoro-3-phenylbutyric acid (138 mg,0.63 mmol) as reaction starting materials, the reaction mixture was poured into water (20 mL), extracted with ethyl acetate (3X 15 mL), backwashed with saturated aqueous sodium chloride solution, dried over anhydrous sodium sulfate, suction filtered, and the filtrate concentrated under reduced pressure to give a crude product, followed byPurification by column chromatography on silica gel (eluent: dichloromethane/ethyl acetate=1/1 (v/v)) afforded the title compound, 250mg. ESI-MS (m/z): 544.0,546.0[ M+H ]] +
Step two: synthesis of tert-butyl 5- ((3- ((4-hydroxy-1- (4, 4-trifluoro-3-phenylbutyryl) piperidin-4-yl) methyl) -4-oxo-3, 4-dihydrothieno [3,4-d ] pyrimidin-7-yl) -2, 3-dihydro-1H-inden-2-yl) carbamate (compound 13-2)
According to the procedure described in step two of example one, using compound 13-1 (30 mg,0.06 mmol) and compound 7-3 (31 mg,0.08 mmol) as reaction starting materials in place of compound 1-2 and (1H-indazol-6-yl) boric acid, respectively, the reaction solution was concentrated under reduced pressure and then separated and purified by preparative thin layer chromatography (developer: dichloromethane/ethyl acetate=3/1 (v/v)) to give the title compound, 30mg. ESI-MS (m/z): 711.3[ M+H ]] +
Step three: synthesis of 3- ((4-hydroxy-1- (4, 4-trifluoro-3-phenylbutyryl) piperidin-4-yl) methyl) -7- (2- (methylamino) -2, 3-dihydro-1H-inden-5-yl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 13)
According to the procedure described in the tenth step of example, compound 13-2 (30 mg,0.04 mmol) was used as a reaction starting material instead of compound 10-1, and the reaction mixture was concentrated under reduced pressure, and then purified by preparative high performance liquid chromatography (method C), and diluted hydrochloric acid (0.5 mL,1 mol/L) was added to the preparation, followed by freeze-drying to give hydrochloride of the title compound, 21mg.
The structural characterization is as follows:
1 H NMR(400MHz,DMSO-d 6 )δ9.22(s,2H),8.46(s,1H),8.06(d,J=8.8Hz,1H),7.96(s,1H),7.90(d,J=8.0Hz,1H),7.43-7.26(m,6H),4.95(s,1H),4.17-4.06(m,1H),4.02-3.92(m,3H),3.87(d,J=9.0,2.1Hz,1H),3.82-3.76(m,1H),3.40-3.29(m,2H),3.28-3.21(m,1H),3.21-3.09(m,3H),3.01-2.79(m,2H),2.61(t,J=5.4Hz,3H),1.66-1.14(m,4H).ESI-MS(m/z):611.3[M+H] +
examples fourteen 3- ((1- (4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butanoyl) -4-hydroxypiperidine-4- Methyl) -7- (4-fluorophenyl) thieno [3,4-d]Pyrimidine-4%3H) Synthesis of Ketone (Compound 14)
Step one: synthesis of tert-butyl 4- ((7- (4-fluorobenzene) -4-oxothiophene [3,4-d ] pyrimidin-3 (4H) -yl) methyl) -4-hydroxypiperidine-1-carboxylate (compound 14-1)
4-Fluorophenylboronic acid (319 mg,4.42 mmol) and Compound 10-1 (1.31 g,2.95 mmol) were dissolved in water (4 mL) and 1, 4-dioxane (20 mL) at room temperature, potassium carbonate (1.22 g,8.84 mmol) and tetraphenylphosphine palladium (3411 mg,0.30 mmol) were added in this order, and after three nitrogen substitutions, the temperature was raised to 80℃for 14h. The reaction solution was cooled to room temperature, concentrated under reduced pressure to give a crude product, and purified by silica gel column chromatography (eluent: dichloromethane/ethyl acetate=2/1 (v/v)) to give the title compound, 1.30g. ESI-MS (m/z): 460.2[ M+H ]] +
Step two: synthesis of 7- (4-fluorophenyl) -3- ((4-hydroxypiperidin-4-yl) methyl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 14-2)
Compound 14-1 (1.30 g,2.83 mmol) was dissolved in hydrochloric acid/1, 4-dioxane solution (20 mL,4 mol/L) at room temperature and reacted at room temperature for 1h. The reaction was filtered off with suction, the filter cake was rinsed with ethyl acetate (2X 30 mL), the filter cake was collected and dried to give the hydrochloride salt of the title compound, 1.10g. ESI-MS (m/z): 360.1[ M+H ]] +
Step three: synthesis of 3- ((1- (4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butyryl) -4-hydroxypiperidin-4-yl) methyl) -7- (4-fluorophenyl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 14)
The hydrochloride salt of compound 14-2 (20 mg,0.05 mmol), 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butyric acid (12 mg,0.06 mmol) and HATU (20 mg,0.05 mmol) were weighed out at room temperature and dissolved in dichloromethane (3 mL), DIPEA (20 mg,0.15 mmol) was added and reacted at room temperature for 4 hours. The reaction solution was concentrated under reduced pressure, and then purified by preparative high performance liquid chromatography (method D), and the preparation solution was concentrated under reduced pressure and freeze-dried to give the title compound (19 mg).
The structural characterization is as follows:
1 H NMR(400MHz,DMSO-d 6 )δ8.49(d,J=1.4Hz,1H),8.19-8.06(m,2H),8.04(d,J=4.6Hz,1H),7.81(dd,J=4.3,2.4Hz,1H),7.41-7.27(m,2H),6.12-6.39(td,J H-F =56.0Hz,J H-H =3.6Hz,1H),5.98(ddd,J=6.0,2.6,1.5Hz,1H),4.99(d,J=12.7Hz,1H),4.95(d,J=2.7Hz,1H),4.09-3.89(m,3H),3.68(d,J=13.6Hz,1H),3.31-3.19(m,2H),2.90(ddd,J=19.1,16.7,4.2Hz,2H),1.64-1.50(m,1H),1.50-1.31(m,3H).ESI-MS(m/z):550.1[M+H] +
example pentadecyl (3- (1- (4, 4' -difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butanoyl) -4-hydroxypiperidine- 4-yl) methyl) - (4-fluorophenyl) thieno [3,4-d]Pyrimidin-4 (3H) -one-A (compound 14-A) and- (3- (1- (4, 4') Difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butyryl) -4-hydroxypiperidin-4-yl) methyl) - (4-fluorophenyl) thieno [3,4 ] d]Synthesis of pyrimidin-4 (3H) -one-B (compound 14-B)
Compound 14 (300 mg,0.55 mmol) was isolated by chiral high performance liquid chromatography to give compound 14-A (135 mg) and compound 14-B (112 mg). Chiral high performance liquid chromatography conditions for compound 14-A and compound 14-B were as follows: chiral column: CHIRALPAK AY-3 (AY 30CD-TJ 004) (0.46 cm I.D..times.15 cm L); sample injection amount: 2. Mu.L; mobile phase: etoh=100%; flow rate: 0.5mL/min; wavelength: UV 214nm; temperature: 35 ℃; high performance liquid chromatography apparatus: shimadzu LC-20AD CP-HPLC-05; wherein the retention time of compound 14-a is: 5.383min, the retention time of Compound 14-B was 8.216min.
The structural characterization is as follows:
compound 14-a: 1 H NMR(400MHz,DMSO-d 6 )δ8.49(d,J=1.4Hz,1H),8.15-8.07(m,2H),8.04(d,J=4.4Hz,1H),7.81(dt,J=4.8,2.8Hz,1H),7.38-7.28(m,2H),6.25(td,J H-F =54.8Hz,J H-H =2.8Hz,1H),5.98(dt,J=6.0,2.0Hz,1H),5.01-4.95(m,2H),4.04-3.98(m,1H),3.95-3.90(m,2H),3.68(d,J=13.6Hz,1H),3.30-3.20(m,2H),2.98-2.84(m,2H),1.67-1.50(m,1H),1.49-1.32(m,3H).ESI-MS(m/z):550.0[M+H] +
compound 14-B: 1 H NMR(400MHz,DMSO-d 6 )δ8.49(d,J=1.4Hz,1H),8.16-8.07(m,2H),8.04(d,J=4.4Hz,1H),7.81(dt,J=4.8,2.8Hz,1H),7.39-7.28(m,2H),6.26(td,J H-F =54.8Hz,J H-H =2.6Hz,1H),5.98(dt,J=6.0,2.0Hz,1H),5.03-4.95(m,2H),4.04-3.98(m,1H),3.95-3.90(m,2H),3.68(d,J=13.6Hz,1H),3.30-3.20(m,2H),2.96-2.85(m,2H),1.67-1.51(m,1H),1.48-1.32(m,3H).ESI-MS(m/z):550.0[M+H] +
examples sixteen 3- ((1- (4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butanoyl) -4-hydroxypiperidine-4- Methyl) -7-methylthiophene [3,4-d ]]Synthesis of pyrimidin-4 (3H) -one (compound 15)
Step one: synthesis of tert-butyl 4-hydroxy-4- ((7-methyl-4-oxothieno [3,4-d ] pyrimidin-3 (4H) -yl) methyl) piperidine-1-carboxylate (compound 15-1)
According to the procedure described in step two of example one, using compound 10-1 (100 mg,0.23 mmol) and trimethylcyclotriboroxine (113 mg,0.90 mmol) as reaction starting materials in place of compound 1-2 and (1H-indazol-6-yl) boric acid, respectively, the reaction solution was concentrated under reduced pressure and then separated and purified by silica gel column chromatography (eluent: dichloromethane/methanol=13/1 (v/v)) to give the title compound, 72mg. ESI-MS (m/z): 380.1[ M+H ]] +
Step two: synthesis of 3- ((4-hydroxypiperidin-4-yl) methyl) -7-methylthiophene [3,4-d ] pyrimidin-4 (3H) -one (compound 15-2)
According to the procedure described in step two of example ten, the reaction was carried out using compound 15-1 (72 mg,0.19 mmol) as a reaction starting material instead of compound 10-1, and the reaction solution was concentrated under reduced pressure to give the hydrochloride of the title compound, 60mg. The reaction mixture was used in the next reaction without purification. ESI-MS (m/z): 280.1[ M+H ] ] +
Step three: synthesis of 3- ((1- (4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butyryl) -4-hydroxypiperidin-4-yl) methyl) -7-methylthiophene [3,4-d ] pyrimidin-4 (3H) -one (compound 15)
According to the procedure described in step three of example eleven, using the hydrochloride salt of compound 15-2 (20 mg,0.06 mmol) and 4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butyric acid (15 mg,0.07 mmol) as reaction starting materials in place of compound 11-2 hydrochloride salt and 4, 4-difluoro-3-phenylbutyric acid, respectively, the reaction mixture was isolated and purified by preparative high performance liquid chromatography (method C), and freeze-dried to give the title compound, 10mg.
The structural characterization is as follows:
1 H NMR(400MHz,DMSO-d 6 )δ8.22(d,J=1.8Hz,1H),7.93(d,J=4.7Hz,1H),7.80(q,J=2.8Hz,1H),6.40-6.10(m,1H),5.98(dd,J=5.9,2.5Hz,1H),5.10-4.95(m,1H),4.05-3.95(m,1H),3.92-3.85(m,2H),3.71-3.62(m,1H),3.33-3.17(m,2H),2.97-2.80(m,2H),2.60(s,3H),1.63-1.28(m,4H).ESI-MS(m/z):470.1[M+H] +
example seventeen 3- ((1- (4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butanoyl) -4-hydroxypiperidine-4- Methyl) -7- (2-methyl-2- (methylamino) -2, 3-dihydro-1H-inden-5-yl) thieno [3,4-d]Pyrimidin-4 (3H) -ones Synthesis of (Compound 16)
Step one: synthesis of tert-butyl (5-bromo-2-methyl-2, 3-dihydro-1H-inden-2-yl) carbamate (Compound 16-2)
Compound 16-1 (30 mg,0.13 mmol) was suspended in dichloromethane (1.5 mL) at room temperature, DIPEA (34 mg,0.27mmol,44 uL) and di-tert-butyl dicarbonate (29 mg,0.13mmol,30 uL) were added, and the reaction was stirred at room temperature for 5h. Concentrating under reduced pressure to obtain crude title compound (40 mg). ESI-MS (m/z): 270.2,272.2[ M-56+H ] ] +
Step two: synthesis of tert-butyl (5-bromo-2-methyl-2, 3-dihydro-1H-inden-2-yl) (methyl) carbamate (Compound 16-3)
According to the procedure described in step one of example seven, using compound 16-2 (40 mg,0.12 mmol) as a reaction starting material instead of compound 7-1, the title compound was obtained as 29mg by preparative thin layer chromatography for purification (developer: petroleum ether/ethyl acetate=5/1 (v/v)). ESI-MS (m/z): 240.0,242.0[ M-100+H ]] +
Step three: synthesis of tert-butyl methyl (2-methyl-5- (4, 5-tetramethyl-1, 3, 2-dioxaborane-2-yl) -2, 3-dihydro-1-hydro-inden-2-yl) carbamate (Compound 16-4)
According to the procedure described in step two of example seven, using compound 16-3 (34 mg,0.10 mmol) as a reaction starting material instead of compound 7-2, the title compound was obtained as 18mg by preparative thin layer chromatography for purification (developer: petroleum ether/ethyl acetate=10/1 (v/v)). ESI-MS (m/z): 288.2[ M-100+H] +
Step four: synthesis of tert-butyl (5- (3- ((1- (4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butanoyl) -4-hydroxypiperidin-4-yl) methyl) -4-oxo-3, 4-dihydrothieno [3,4-d ] pyrimidin-7-yl) -2-methyl-2, 3-dihydro-1H-inden-2-yl) (methyl) carbamate (Compound 16-5)
According to the procedure described in step two of example one, using 10-3 (20 mg,0.04 mmol) and compound 16-4 (18 mg,0.05 mmol) and starting materials for the reaction in place of compounds 1-2 and (1H-indazol-6-yl) boronic acid, respectively, purification by preparative thin layer chromatography (developer: dichloromethane/methanol=20/1 (v/v)) afforded the title compound, 26mg. ESI-MS (m/z): 715.3[ M+H ]] +
Step five: 3- ((1- (4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butyryl) -4-hydroxypiperidin-4-yl) methyl) -7- (2-methyl-2- (methylamino) -2, 3-dihydro-1H-inden-5-yl) thiophene [3,4-d ] pyrimidin-4 (3H) -one (synthesis of Compound 16)
Compound 16-5 (27 mg,0.04 mmol) was dissolved in ethyl acetate (2 mL) at room temperature, and an ethyl acetate solution of hydrochloric acid (2 mL,4 mol/L) was added thereto, followed by stirring at 20℃for 3h. Concentrating under reduced pressure, adding ethyl acetate, precipitating solid, vacuum filtering, washing filter cake with methyl tert-butyl ether, and drying. After dissolution in water, freeze drying afforded the hydrochloride salt of the title compound, 23mg.
The structural characterization is as follows:
1 H NMR(400MHz,D 2 O)δ8.14(d,J=4.8Hz,1H),7.81(d,J=18.8Hz,1H),7.60-7.44(m,3H),7.28(d,J=3.6Hz,1H),6.10(td,J H-F =54.4Hz,J H-H =3.2Hz,1H),5.84(ddd,J=44.4,5.6,2.8Hz,1H),5.00-4.80(m,1H),4.11-3.65(m,4H),3.44-3.18(m,4H),3.17-3.06(m,2H),3.04-2.80(m,2H),2.64(s,3H),1.73-1.54(m,1H),1.50(s,3H),1.48-1.18(m,3H).ESI-MS(m/z):615.2[M+H] +
example eighteen 3- ((1- (3-chloro-1H-pyrazol-1-yl) -4, 4-difluorobutyryl) -4-hydroxypiperidin-4-yl) Methyl) -7- (1- (methylamino) -2, 3-dihydro-1H-inden-5-yl) thieno [3,4-d]Pyrimidin-4 (3H) -one (compound 17) Is synthesized by (a)
Step one: synthesis of 3- ((1- (3-chloro-1H-pyrazol-1-yl) -4, 4-difluorobutyryl) -4-hydroxypiperidin-4-yl) methyl) -7- (1-oxo-2, 3-dihydro-1H-inden-5-yl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 17-1)
According to the procedure described in step three of example eleven, 3- (3-chloro-1H-pyrazol-1-yl) -4, 4-difluorobutyric acid (30 mg,0.69 mmol) was used as a reaction starting material instead of 4, 4-difluoro-3-phenylbutyric acid, water (5 mL) was added to the reaction solution, ethyl acetate (3X 5 mL) was extracted, and the organic phase was washed with saturated brine (5 mL), dried over anhydrous sodium sulfate and concentrated to give crude title compound, 41mg. ESI-MS (m/z): 601.7,603.7[ M+H ]] +
Step two: synthesis of 3- ((1- (3-chloro-1H-pyrazol-1-yl) -4, 4-difluorobutyryl) -4-hydroxypiperidin-4-yl) methyl) -7- (1- (methylamino) -2, 3-dihydro-1H-inden-5-yl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 17)
The crude product of compound 17-1 (41 mg) was dissolved in methanol (10 mL) at room temperature, and a methylamine/ethanol solution (4 mL,33% wt) was added thereto, followed by stirring at room temperature for 18h. After the reaction solution was concentrated, the concentrate was dissolved in methanol (10 mL), and sodium cyanoborohydride (21 mg,0.034 mmol) and acetic acid (20 mg,0.034 mmol) were added thereto, followed by stirring at room temperature for 2 hours. The reaction mixture was concentrated and purified by preparative high performance liquid chromatography (method I) and freeze-dried to give the formate of the title compound (10 mg).
The structural characterization is as follows:
1 H NMR(400MHz,DMSO-d 6 )δ8.45(d,J=2.4Hz,1H),8.03(d,J=4.4Hz,1H),7.97-7.90(m,2H),7.82(d,J=8.0Hz,1H),7.46(d,J=8.0Hz,1H),6.34(d,J=2.4Hz,1H),6.29(t,J H-F =56.0Hz,1H),5.10(br,1H),4.96(br,1H),4.22(t,J=6.4Hz,1H),4.06-3.97(m,1H),3.96-3.89(m,2H),3.72-3.65(m,1H),3.07-2.79(m,6H),2.43-2.33(m,4H),1.92-1.82(m,1H),1.66-1.31(m,5H);ESI-MS(m/z):616.7,618.7[M+H] +
example nineteen 3- ((1- (3-chloro-1H-pyrazol-1-yl) butyryl) -4-hydroxypiperidin-4-yl) methyl) -7- (1- (methylamino) -2, 3-dihydro-1H-inden-5-yl) thieno [3,4-d]Synthesis of pyrimidin-4 (3H) -one (compound 18)
Step one: synthesis of 3- ((1- (3-chloro-1H-pyrazol-1-yl) butanoyl) -4-hydroxypiperidin-4-yl) methyl) -7- (1-oxo-2, 3-dihydro-1H-inden-5-yl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 18-1)
According to the procedure described in step three of example eleven, 3- (3-chloro-1H-pyrazol-1-yl) butyric acid (30 mg,0.69 mmol) was used as a reaction starting material instead of 4, 4-difluoro-3-phenylbutyric acid, water (5 mL) was added to the reaction solution, ethyl acetate (3X 5 mL) was extracted, and the organic phase was washed with saturated brine (5 mL), dried over anhydrous sodium sulfate and concentrated to give crude title compound (39 mg). ESI-MS (m/z): 565.8,567.8[ M+H ]] +
Step two: synthesis of 3- ((1- (3-chloro-1H-pyrazol-1-yl) butanoyl) -4-hydroxypiperidin-4-yl) methyl) -7- (1- (methylamino) -2, 3-dihydro-1H-inden-5-yl) thiophene [3,4-d ] pyrimidin-4 (3H) -one (compound 18)
According to the procedure described in eighteenth step of example, crude product (39 mg) of compound 18-1 was used as a reaction starting material instead of crude product of compound 17-1 to carry out a reaction, the reaction solution was separated and purified by preparative high performance liquid chromatography (method I), and the preparation solution was concentrated under reduced pressure and freeze-dried to give formate salt of the title compound, 14mg.
The structural characterization is as follows:
1 H NMR(400MHz,DMSO-d 6 )δ8.45(d,J=2.4Hz,1H),8.34(s,1H),8.03(d,J=5.2Hz,1H),7.92(s,1H),7.87(d,J=8.0Hz,1H),7.80(dd,J=3.6,2.4Hz,1H),7.46(d,J=8.0Hz,1H),6.23(t,J=2.0Hz,1H),4.95(brs,1H),4.79-4.68(m,1H),4.21(t,J=6.8Hz,1H),4.08-3.99(m,1H),3.96-3.88(m,2H),3.67-3.61(m,1H),3.30-3.11(m,1H),3.03-2.91(m,2H),2.90-2.78(m,2H),2.77-2.65(m,1H),3.39(s,3H),2.42-2.33(m,1H),1.93-1.82(m,1H),1.47-1.38(m,7H);ESI-MS(m/z):580.8,582.8[M+H] +
example twenty 3- ((1- (3-cyclopropyl-3-phenylpropionyl) -4-hydroxypiperidin-4-yl) methyl) -7- (1- (methylamino) -2, 3-dihydro-1H-inden-5-yl) thieno [3,4-d]Synthesis of pyrimidin-4 (3H) -one (compound 19)
Step one: synthesis of 7-bromo-3- ((1- (3-cyclopropyl-3-phenylpropionyl) -4-hydroxypiperidin-4-yl) methyl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 19-1)
According to the procedure described in step three of example ten, the reaction was carried out using 3-cyclopropyl-3-phenylpropionic acid (150 mg,0.79 mmol) as a reaction raw material instead of the compound 4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butyric acid, and purification by silica gel column chromatography (eluent: ethyl acetate=100%) gave the title compound, 270mg. ESI-MS (m/z): 516.0,518.0[ M+H ]] +
Step two: synthesis of tert-butyl 5- ((3- ((1- (3-cyclopropyl-3-phenylpropionyl) -4-hydroxypiperidin-4-yl) methyl) -4-oxo-3, 4-dihydrothiophene [3,4-d ] pyrimidin-7-yl) -2, 3-dihydro-1H-inden-1-yl) (methyl) carbamate (compound 19-2)
According to the procedure described in step two of example one, using compound 19-1 (30 mg,0.06 mmol) and compound 5-3 (26 mg,0.07 mmol) as reaction starting materials in place of compound 1-2 and (1H-indazol-6-yl) boric acid, respectively, the reaction solution was concentrated under reduced pressure, and then, thin layer chromatography was performed to isolate and purify (developer: ethyl acetate=100%) to give the title compound, 35mg. ESI-MS (m/z): 683.3[ M+H ] ] +
Step three: synthesis of 3- ((1- (3-cyclopropyl-3-phenylpropionyl) -4-hydroxypiperidin-4-yl) methyl) -7- (1- (methylamino) -2, 3-dihydro-1H-inden-5-yl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 19)
Compound 19-2 (35 mg,0.05 mmol) was weighed into a reaction flask at room temperature, and ethyl acetate hydrochloride solution (2 mL,4 mol/L) was added thereto, followed by stirring at room temperature for reaction for 1h. The solvent was removed by concentration under reduced pressure, and the preparation was purified by HPLC (method C), diluted hydrochloric acid (0.5 mL,1 mol/L) was added to the preparation, and the mixture was lyophilized to give the hydrochloride of the title compound (17 mg).
The structural characterization is as follows:
1 H NMR(400MHz,Methanol-d 4 ):δ8.81-8.17(m,2H),7.94-7.67(m,3H),7.38-7.15(m,5H),4.99-4.93(m,1H),4.87-4.78(m,1H),4.27-3.84(m,3H),3.74(t,J=14.8Hz,1H),3.29-3.23(m,1H),3.20-2.91(m,3H),2.88-2.71(m,4H),2.70-2.60(m,1H),2.37-2.22(m,2H),1.73-1.52(m,1H),1.51-1.41(m,1H),1.40-1.31(m,1H),1.31-1.18(m,1H),1.18-0.70(m,1H),0.69-0.56(m,1H),0.48-0.38(m,1H),0.37-0.26(m,1H),0.16-0.07(m,1H).ESI-MS(m/z):583.4[M+H] +
example twenty-one 3- ((1- (3-cyclopropyl-3-phenylpropionyl) -4-hydroxypiperidin-4-yl) methyl) -7- (2- (methylamino) -2, 3-dihydro-1H-inden-5-yl) thieno [3,4-d]Synthesis of pyrimidin-4 (3H) -one (compound 20)
Step one: synthesis of tert-butyl 5- (3- ((1- (3-cyclopropyl-3-phenylpropionyl) -4-hydroxypiperidin-4-yl) methyl) -4-oxo-3, 4-dihydrothieno [3,4-d ] pyrimidin-7-yl) -2, 3-dihydro-1H-inden-2-yl) (methyl) carbamate (compound 20-1)
According to the procedure described in step two of example one, using compound 19-1 (30 mg,0.06 mmol) and compound 7-3 (26 mg,0.07 mmol) as reaction starting materials in place of compound 1-2 and (1H-indazol-6-yl) boric acid, respectively, the reaction solution was concentrated under reduced pressure, and then, thin layer chromatography was performed to isolate and purify (developer: ethyl acetate=100%) to give the title compound, 35mg. ESI-MS (m/z): 683.3[ M+H ] ] +
Step two: synthesis of 3- ((1- (3-cyclopropyl-3-phenylpropionyl) -4-hydroxypiperidin-4-yl) methyl) -7- (2- (methylamino) -2, 3-dihydro-1H-inden-5-yl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 20)
According to the procedure described in step three of example twenty, the reaction was carried out using compound 20-1 (35 mg,0.05 mmol) as a reaction starting material instead of compound 19-2, and the reaction solution was concentrated under reduced pressure, and then purified by preparative high performance liquid chromatography (method C), and the preparation solution was added with dilute hydrochloric acid (0.5 mL,1 mol/L) and freeze-dried to give the hydrochloride of the title compound, 21mg.
The structural characterization is as follows:
1 H NMR(400MHz,Methanol-d 4 ):δ8.73-8.56(m,2H),7.73(s,1H),7.63(d,J=8.4Hz,1H),7.48(d,J=7.8Hz,1H),7.41-7.13(m,5H),4.31-4.02(m,3H),4.02-3.89(m,1H),3.75(t,J=14.0Hz,1H),3.57-3.46(m,2H),3.37-3.31(m,1H),3.28-3.18(m,2H),3.08-2.89(m,2H),2.88-2.69(m,4H),2.33-2.23(m,1H),1.73-1.53(m,1H),1.50-1.41(m,1H),1.37-1.30(m,1H),1.28-1.17(m,1H),1.16-0.70(m,1H),0.67-0.59(m,1H),0.47-0.38(m,1H),0.36-0.27(m,1H),0.15-0.07(m,1H).ESI-MS(m/z):583.3[M+H] +
examples twenty two 7- (4-chloro-1- (methylamino) -2, 3-dihydro-1H-inden-5-yl) -3- ((1- (4, 4-difluoro-) -1-) 3- (3-fluoro-1H-pyrazol-1-yl) butyryl) -4-hydroxypiperidin-4-yl) methyl) thieno [3,4-d]Pyrimidin-4 (3H) -ones Synthesis of Compound 21
Step one: synthesis of 7- (4-chloro-1-oxo-2, 3-dihydro-1H-inden-5-yl) -3- ((1- (4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butyryl) -4-hydroxypiperidin-4-yl) methyl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 21-1)
5-bromo-4-chloro-2, 3-dihydro-1H-inden-1-one (60 mg,0.24 mmol), pinacol diboronate (68 mg,0.27 mmol) and [1,1' -bis (diphenylphosphino) ferrocene at room temperature ]Palladium dichloride (18 mg,0.02 mmol) and potassium acetate (48 mg,0.49 mmol) were reacted in 1, 4-dioxane (5 mL), the reaction system was replaced with nitrogen three times, and the temperature was raised to 80℃for 16h. The reaction solution was cooled to room temperature, and Compound 10-3 (40 mg,0.07 mmol), potassium carbonate (31 mg,0.22 mmol), tetrakis (triphenylphosphine) palladium (9 mg, 0.0070 mmol) and water (1 mL) were added, nitrogen was substituted three times, and the temperature was raised to 100℃for reaction for 4 hours. After cooling to room temperature and concentrating under reduced pressure, the product was purified by preparative thin layer chromatography (developer: ethyl acetate=100%) to give the title compound, 25mg. ESI-MS (m/z): 620.1,622.1[ M+H ]] +
Step two: synthesis of 7- (4-chloro-1- (methylamino) -2, 3-dihydro-1H-inden-5-yl) -3- ((1- (4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butyryl) -4-hydroxypiperidin-4-yl) methyl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 21)
Compound 21-1 (25 mg,0.04 mmol) was weighed into a reaction flask at room temperature, and a methylamine/ethanol solution (2 mL,33% wt) was added thereto, followed by stirring at room temperature for reaction for 5 hours. The solvent was removed by concentration under reduced pressure, dissolved in methanol (2 mL), and sodium borohydride (3 mg,0.08 mmol) was added and stirred at room temperature for 2h. The reaction solution was purified by preparative high performance liquid chromatography (method J) and lyophilized to give the title compound as trifluoroacetate salt (8 mg).
The structural characterization is as follows:
1 H NMR(400MHz,Methanol-d 4 ):δ8.54(d,J=1.4Hz,1H),7.99(d,J=7.8Hz,1H),7.67-7.51(m,3H),6.12(td,J H-F =55.4Hz,J H-H =4.4Hz,1H),5.91-5.84(m,1H),4.99-4.90(m,2H),4.19-4.09(m,1H),4.05(d,J=3.4Hz,1H),4.00(d,J=1.8Hz,1H),3.84-3.75(m,1H),3.49-3.36(m,2H),3.29-3.22(m,1H),3.18-3.10(m,1H),3.10-3.00(m,1H),2.96-2.83(m,1H),2.79(s,3H),2.73-2.64(m,1H),2.38-2.29(m,1H),1.77-1.46(m,4H).ESI-MS(m/z):635.2,637.2[M+H] +
examples twenty-three 3- ((1- (4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butanoyl) -4-hydroxypiperidine- 4-yl) methyl) -7- (4-fluoro-1- (methylamino) -2, 3-dihydro-1H-inden-5-yl) thieno [3,4-d]Pyrimidin-4 (3H) -ones Synthesis of Compound 22
Step one: synthesis of 3- ((1- (4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butyryl) -4-hydroxypiperidin-4-yl) methyl) -7- (4-fluoro-1-oxo-2, 3-dihydro-1H-inden-5-yl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 22-1)
According to the procedure described in example twenty-two step one, using 5-bromo-4-fluoro-2, 3-dihydro-1H-inden-1-one (100 mg,0.44 mmol) as a reaction starting material instead of 5-bromo-4-chloro-2, 3-dihydro-1H-inden-1-one, the reaction solution was concentrated under reduced pressure and then separated and purified by preparative thin layer chromatography (developer: ethyl acetate=100%) to give the title compound, 35mg. ESI-MS (m/z): 604.2[ M+H ]] +
Step two: synthesis of 3- ((1- (4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butyryl) -4-hydroxypiperidin-4-yl) methyl) -7- (4-fluoro-1- (methylamino) -2, 3-dihydro-1H-inden-5-yl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 22)
According to the procedure described in example twenty-second step, using compound 22-1 (35 mg,0.06 mmol) as a reaction starting material instead of compound 21-1, the reaction solution was purified by preparative high performance liquid chromatography (method C) and freeze-dried to give the formate salt of the title compound, 28mg.
The structural characterization is as follows:
1 H NMR(400MHz,Methanol-d 4 ):δ8.54(s,1H),8.49(d,J=1.6Hz,1H),8.13(t,J=7.2Hz,1H),8.03(d,J=7.8Hz,1H),7.59(dt,J=5.2,2.4Hz,1H),7.43(d,J=7.8Hz,1H),6.12(td,J H-F =55.4Hz,J H-H =4.4Hz,1H),5.91-5.84(m,1H),5.01-4.93(m,1H),4.70(dd,J=7.8,4.4Hz,1H),4.19-4.09(m,1H),4.05(d,J=3.6Hz,1H),4.00(s,1H),3.84-3.75(m,1H),3.48-3.36(m,2H),3.26-3.17(m,1H),3.12-3.02(m,2H),2.96-2.83(m,1H),2.69(s,3H),2.67-2.58(m,1H),2.30-2.21(m,1H),1.76-1.47(m,4H).ESI-MS(m/z):619.2[M+H] +
example twenty-four 7- (4-chloro-1- (methylamino) -2, 3-dihydro-1H-inden-5-yl) -3- ((1- (3-cyclopropyl-) - 3-phenylpropionyl) -4-hydroxypiperidin-4-yl methyl) thieno [3,4-d]Synthesis of pyrimidin-4 (3H) -one (compound 23)
Step one: synthesis of tert-butyl (5-bromo-4-chloro-2, 3-dihydro-1H-inden-1-yl) (methyl) carbamate (Compound 23-2)
Compound 23-1 (100 mg,0.41 mmol) was dissolved in methanol (2 mL), a methylamine/ethanol solution (2 mL,33% wt) was added, the reaction mixture was dried by spinning at room temperature for 16h, the residue was dissolved in methanol (2 mL), sodium borohydride (30 mg,0.81 mmol) was added, and after 1h at room temperature, di-tert-butyl dicarbonate (444 mg,2.04 mmol) was added, and the reaction was continued at room temperature for 1h. The solvent was drained under reduced pressure and purified by column chromatography on silica gel (eluent: petroleum ether/ethyl acetate=10/1 (v/v)) to give 130mg of the title compound. ESI-MS (m/z): 304.3, 306.3[ M-56+H ]] +
Step two: synthesis of tert-butyl 4-chloro-5- (4, 5-tetramethyl-1, 3, 2-dioxaborane-2-yl) -2, 3-dihydro-1H-inden-1-yl) (methyl) carbamate (Compound 23-3)
According to the procedure described in step two of example seven, the reaction was carried out using compound 23-2 (130 mg,0.36 mmol) as a reaction starting material instead of compound 7-2, and the concentrated reaction solution was separated and purified by preparative thin layer chromatography (developing solvent: petroleum ether/acetic acid) Ethyl ester=15/1 (v/v)) to give the title compound, 100mg. ESI-MS (m/z): 352.2,354.2[ M-56+H ]] +
Step three: synthesis of tert-butyl 4-chloro-5- (3- ((1- (3-cyclopropyl-3-phenylpropionyl) -4-hydroxypiperidin-4-yl) methyl) -4-oxo-3, 4-dihydrothieno [3,4-d ] pyrimidin-7-yl) -2, 3-dihydro-1H-inden-1-yl) (methyl) carbamate (Compound 23-4)
According to the procedure described in step two of example one, using compound 19-1 (20 mg,0.04 mmol) and compound 23-4 (17 mg,0.04 mmol) as reaction starting materials in place of compound 1-2 and (1H-indazol-6-yl) boric acid, respectively, the reaction solution was concentrated under reduced pressure and then purified by preparative thin layer chromatography (developer: ethyl acetate/methanol=60/1 (v/v)) to give the title compound, 15mg. ESI-MS (m/z): 717.3,719.3[ M+H ]] +
Step four: synthesis of 7- (4-chloro-1- (methylamino) -2, 3-dihydro-1H-inden-5-yl) -3- ((1- (3-cyclopropyl-3-phenylpropionyl) -4-hydroxypiperidin-4-yl) methyl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 23)
According to the procedure described in step three of example twenty, the reaction was carried out using compound 23-4 (15 mg,0.02 mmol) as a reaction starting material instead of compound 19-2, and the reaction solution was concentrated under reduced pressure and then purified by preparative high performance liquid chromatography (method K) and freeze-dried to give the formate salt of the title compound, 8mg.
The structural characterization is as follows:
1 H NMR(400MHz,Methanol-d 4 )δ8.48(d,J=7.0Hz,1H),7.92(s,0.4H),7.86(s,0.6H),7.55-7.39(m,2H),7.32-7.08(m,5H),4.65-4.54(m,1H),4.22-3.59(m,4H),3.30(s,1H),3.20-2.76(m,5H),2.65-2.51(m,1H),2.58(s,3H),2.30-2.08(m,2H),1.64-1.04(m,5H),0.63-0.49(m,1H),0.43-0.29(m,2H),0.31-0.19(m,1H),0.14-0.02(m,1H).ESI-MS(m/z):617.3,619.3[M+H] +
examples twenty-five 7- (6-chloro-1- (methylamino) -2, 3-dihydro-1H-inden-5-yl) -3- ((1- (4, 4-difluoro-) -1-) 3- (3-fluoro-1H-pyrazol-1-yl) butyryl) -4-hydroxypiperidin-4-yl) methyl) thieno [3,4-d]Pyrimidin-4 (3H) -ones Synthesis of (Compound 24)
Step one: synthesis of 6-chloro-5- (4, 5-tetramethyl-1, 3, 2-dioxaborane-2-yl) -2, 3-dihydro-1H-inden-1-one (Compound 24-2)
Compound 24-1 (100 mg,0.41 mmol), pinacol biborate (207 mg,0.25 mmol) and [1,1' -bis (diphenylphosphino) ferrocene were reacted at room temperature]Palladium dichloride (15 mg,0.02 mmol) and potassium acetate (120 mg,1.22 mmol) were dissolved in 1, 4-dioxane (2 mL), and after three times of nitrogen substitution, the reaction was warmed to 80℃and stirred for 16h. The reaction solution was filtered and concentrated to give crude title compound (119 mg). ESI-MS (m/z): 293.1,295.1[ M+H ]] +
Step two: synthesis of 7- (6-chloro-1-oxo-2, 3-dihydro-1H-inden-5-yl) -3- ((1- (4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butyryl) -4-hydroxypiperidin-4-yl) methyl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 24-3)
According to the procedure described in step two of example one, the reaction was carried out using compound 10-3 (35 mg,0.07 mmol) and crude compound 24-2 (23 mg) as reaction starting materials instead of compound 1-2 and (1H-indazol-6-yl) boric acid, respectively, and the reaction solution was filtered and concentrated to give crude title compound, 41mg. ESI-MS (m/z): 620.0,622.0[ M+H ] ] +
Step three: synthesis of 7- (6-chloro-1-oxo-2, 3-dihydro-1H-inden-5-yl) -3- ((1- (4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butyryl) -4-hydroxypiperidin-4-yl) methyl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 24)
According to the procedure described in example twenty-second step, the crude product (41 mg) of Compound 24-3 was used as a reaction starting material instead of Compound 21-1, and the reaction mixture was purified by preparative high performance liquid chromatography (method J) and freeze-dried to give the trifluoroacetate salt of the title compound, 4mg.
The structural characterization is as follows:
1 H NMR(400MHz,Methanol-d 4 )δ8.54(s,1H),7.99(d,J=8.0Hz,1H),7.75(s,1H),7.62(s,1H),7.61-7.56(m,1H),6.11(td,J H-F =55.2Hz,J H-H =4.4Hz,1H),5.91-5.84(m,1H),5.00-4.90(m,1H),4.85-4.79(m,1H),4.19-4.09(m,1H),4.07-3.98(m,2H),3.85-3.75(m,1H),3.45-3.35(m,2H),3.23-3.15(m,1H),3.09-3.05(m,1H),2.96-2.84(m,1H),2.79(s,3H),2.71-2.63(m,1H),2.34-2.26(m,1H),1.79-1.45(m,4H).ESI-MS(m/z):635.2,637.2[M+H] +
example twenty-six 7- (6-chloro-1- (methylamino) -2, 3-dihydro-1H-inden-5-yl) -3- ((1- (3-cyclopropyl-) - 3-phenylpropionyl) -4-hydroxypiperidin-4-yl methyl) thieno [3,4-d]Synthesis of pyrimidin-4 (3H) -one (compound 25)
Step one: synthesis of 7- (6-chloro-1-oxo-2, 3-dihydro-1H-inden-5-yl) -3- ((1- (3-cyclopropyl-3-phenylpropionyl) -4-hydroxypiperidin-4-yl) methyl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 25-1)
According to the procedure described in step two of example one, using compound 19-1 (30 mg,0.06 mmol) and compound 24-2 (13 mg,0.06 mmol) as reaction starting materials in place of compound 1-2 and (1H-indazol-6-yl) boric acid, respectively, the reaction solution was concentrated under reduced pressure and then separated and purified by preparative thin layer chromatography (developer: ethyl acetate/methanol=30/1 (v/v)) to give the title compound, 20mg. ESI-MS (m/z): 602.2,604.2[ M+H ] ] +
Step two: synthesis of 7- (6-chloro-1- (methylamino) -2, 3-dihydro-1H-inden-5-yl) -3- ((1- (3-cyclopropyl-3-phenylpropionyl) -4-hydroxypiperidin-4-yl) methyl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 25)
According to the procedure described in example twenty-second step, a reaction was performed using compound 25-1 (20 mg,0.03 mmol) as a reaction starting material instead of compound 21-1, and the reaction solution was separated and purified by preparative high performance liquid chromatography (method C), and freeze-dried to give the title compound, 8mg.
The structural characterization is as follows:
1 H NMR(400MHz,Methanol-d 4 ):δ8.50(d,J=7.2Hz,1H),7.92(d,J=24.4Hz,1H),7.62(s,1H),7.48(s,1H),7.34-7.24(m,4H),7.22-7.15(m,1H),4.38(t,J=6.4Hz,1H),4.25-4.11(m,1H),4.00(q,J=14.0Hz,1H),3.92-3.76(m,1H),3.75-3.65(m,1H),3.29-3.14(m,1H),3.12-3.03(m,1H),3.02-2.84(m,3H),2.83-2.64(m,1H),2.53(s,3H),2.52-2.41(m,1H),2.32-2.23(m,1H),2.11-1.99(m,1H),1.63-1.26(m,4H),1.24-1.11(m,1H),0.66-0.55(m,1H),0.46-0.36(m,1H),0.35-0.25(m,1H),0.14-0.04(m,1H).ESI-MS(m/z):617.3,619.3[M+H] +
examples twenty-seven 7- (7-chloro-1- (methylamino) -2, 3-dihydro-1H-inden-5-yl) -3- ((1- (4, 4-difluoro-) -1-) 3- (3-fluoro-1H-pyrazol-1-yl) butyryl) -4-hydroxypiperidin-4-yl) methyl) thieno [3,4-d]Pyrimidin-4 (3H) -ones Synthesis of (Compound 26)
Step one: synthesis of 7-chloro-5- (4, 5-tetramethyl-1, 3, 2-dioxaborane-2-yl) -2, 3-dihydro-1H-inden-1-one (Compound 26-2)
According to the procedure described in the twenty-fifth step one of example, the reaction was conducted using compound 26-1 (20 mg,0.08 mmol) as a reaction starting material instead of compound 24-1, and the reaction solution was filtered and concentrated to give a crude product of the title compound, 54mg. ESI-MS (m/z): 293.1,295.1[ M+H ]] +
Step two: synthesis of 7- (7-chloro-1-oxo-2, 3-dihydro-1H-inden-5-yl) -3- ((1- (4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butyryl) -4-hydroxypiperidin-4-yl) methyl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 26-3)
According to the procedure described in step two of example one, the reaction was carried out using crude compounds 10-3 (44 mg,0.08 mmol) and 26-2 (54 mg) as reaction starting materials instead of compounds 1-2 and (1H-indazol-6-yl) boric acid, respectively, and the reaction solution was filtered and concentrated to give crude title compound, 51mg. ESI-MS (m/z): 620.0,622.0[ M+H ]] +
Step three: synthesis of 7- (7-chloro-1- (methylamino) -2, 3-dihydro-1H-inden-5-yl) -3- ((1- (4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butyryl) -4-hydroxypiperidin-4-yl) methyl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 26)
According to the procedure described in example twenty-second step, the crude product (51 mg) of compound 24-3 was used as a reaction starting material instead of compound 21-1, and the reaction mixture was purified by preparative high performance liquid chromatography (method J) and freeze-dried to give the trifluoroacetate salt of the title compound, 6mg.
The structural characterization is as follows:
1 H NMR(400MHz,Methanol-d 4 )δ8.44(d,J=1.2Hz,1H),8.20(s,1H),8.07(d,J=8.0Hz,1H),7.96(s,1H),7.61-7.56(m,1H),6.11(td,J H-F =55.6Hz,J H-H =4.4Hz,1H),5.87(td,J=6.0Hz,2.4Hz,1H),4.97-4.93(m,2H),4.19-4.09(m,1H),4.05(s,1H),4.01(s,1H),3.85-3.75(m,1H),3.45-3.35(m,2H),3.18-3.10(m,1H),3.09-3.02(m,1H),2.96-2.82(m,1H),2.77(s,3H),2.64-2.56(m,1H),2.43-2.35(m,1H),1.79-1.44(m,4H),1.40-1.34(m,1H);ESI-MS(m/z):635.2,637.2[M+H] +
example twenty-eight 7- (7-chloro-1- (methylamino) -2, 3-dihydro-1H-inden-5-yl) -3- ((1- (3-cyclopropyl-) - 3-phenylpropionyl) -4-hydroxypiperidin-4-yl methyl) thieno [3,4-d]Synthesis of pyrimidin-4 (3H) -one (compound 27)
Step one: synthesis of 7- (7-chloro-1-oxo-2, 3-dihydro-1H-inden-5-yl) -3- ((1- (3-cyclopropyl-3-phenylpropionyl) -4-hydroxypiperidin-4-yl) methyl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 27-1)
According to the procedure described in step two of example one, using compound 19-1 (40 mg,0.08 mmol) and compound 26-2 (18 mg,0.09 mmol) as reaction starting materials in place of compound 1-2 and (1H-indazol-6-yl) boric acid, respectively, the reaction solution was concentrated under reduced pressure and then separated and purified by preparative thin layer chromatography (developer: ethyl acetate/methanol=30/1 (v/v)) to give the title compound, 20mg. ESI-MS (m)/z):602.2,604.2[M+H] +
Step two: synthesis of 7- (7-chloro-1- (methylamino) -2, 3-dihydro-1H-inden-5-yl) -3- ((1- (3-cyclopropyl-3-phenylpropionyl) -4-hydroxypiperidin-4-yl) methyl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 27)
Compound 27-1 (20 mg,0.03 mmol) was weighed into methanol (20 mL) and dichloromethane (10 mL) at room temperature, a methylamine/ethanol solution (2 mL,33% wt) was added, and the reaction was stirred at room temperature for 20h. The solvent was removed by concentration under reduced pressure, dissolved in methanol (20 mL) and methylene chloride (10 mL), and sodium borohydride (2 mg,0.07 mmol) was added thereto and stirred at room temperature for 1h. The reaction mixture was purified by preparative HPLC (method J), and the preparation was lyophilized with diluted hydrochloric acid (0.5 mL,1 mol/L) to give the hydrochloride of the title compound (11 mg).
The structural characterization is as follows:
1 H NMR(400MHz,Methanol-d 4 ):δ8.53(d,J=8.8Hz,1H),8.27(d,J=17.6Hz,1H),8.09(s,1H),7.89(s,1H),7.41-7.10(m,5H),4.97(dd,J=7.6,1.8Hz,1H),4.26-4.13(m,1H),4.13-4.01(m,1H),3.97-3.82(m,1H),3.79-3.69(m,1H),3.43-3.31(m,2H),3.20-3.11(m,1H),3.07-2.89(m,2H),2.88-2.69(m,4H),2.67-2.56(m,1H),2.47-2.39(m,1H),2.32-2.23(m,1H),1.69-1.30(m,4H),1.25-1.13(m,1H),0.66-0.56(m,1H),0.48-0.36(m,1H),0.36-0.25(m,1H),0.16-0.05(m,1H).ESI-MS(m/z):617.2,619.2[M+H] +
example twenty-nine 3- ((1- (4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butanoyl) -4-hydroxypiperidine ] 4-yl) methyl) -7- (3- (methylamino) -2, 3-dihydro-1H-inden-5-yl) thieno [3,4-d]Pyrimidin-4 (3H) -ones (compounds Synthesis of 28)
Step one: synthesis of 3- ((1- (4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butanoyl) -4-hydroxypiperidin-4-yl) methyl) -7- (3-oxo-2, 3-dihydro-1H-inden-5-yl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 28-1)
According to the procedure described in step two of example one, starting from compound 10-3 (25 mg,0.05 mmol) and (3-oxo-2, 3-dihydro-1H-inden-5-yl) boronic acid (14 mg,0.06 mmol) in place of compounds 1-2 and (1H-indazol-6-yl) boronic acid, respectively, the reaction was concentrated and purified by preparative thin layer chromatography (developer: ethyl acetate=100%) to give the title compound, 24mg. ESI-MS (m/z): 586.2[ M+H ]] +
Step two: synthesis of 3- ((1- (4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butanoyl) -4-hydroxypiperidin-4-yl) methyl) -7- (3- (methylamino) -2, 3-dihydro-1H-inden-5-yl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 28)
According to the procedure described in example twenty-second step, the reaction was carried out using 28-1 (24 mg,0.04 mmol) as a reaction starting material instead of 21-1, and the reaction solution was purified by preparative high performance liquid chromatography (method C) and freeze-dried to give the formate of the title compound, 13mg.
The structural characterization is as follows:
1 H NMR(400MHz,DMSO-d 6 ):δ8.44(d,J=1.6Hz,1H),8.34(s,1H),8.03(d,J=4.8Hz,1H),8.00(s,1H),7.97(d,J=7.6Hz,1H),7.81(dt,J=4.8,2.8Hz,1H),7.35(d,J=7.8Hz,1H),6.32(dd,J H-F =54.8Hz,J H-H =3.6Hz,1H),6.01-5.95(m,1H),5.23-4.79(m,2H),4.26(t,J=6.6Hz,1H),4.06-3.99(m,1H),3.97-3.89(m,2H),3.69(d,J=13.6Hz,1H),3.31-3.21(m,2H),3.02-2.96(m,1H),2.95-2.85(m,2H),2.85-2.75(m,1H),2.41(s,3H),2.39-2.32(m,1H),1.93-1.82(m,1H),1.65-1.31(m,4H).ESI-MS(m/z):601.2[M+H] +
example thirty-7- (7-chloro-3- (methylamino) -2, 3-dihydro-1H-inden-5-yl) -3- ((1- (4, 4-difluoro-3-) (3-fluoro-1H-pyrazol-1-yl) butyryl) -4-hydroxypiperidin-4-yl) methyl) thieno [3,4-d]Pyrimidin-4 (3H) -ones Synthesis of Compound 29)
Step one: synthesis of 7- (7-chloro-3-oxo-2, 3-dihydro-1H-inden-5-yl) -3- ((1- (4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butyryl) -4-hydroxypiperidin-4-yl) methyl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 29-1)
According to the procedure described in example twenty-two step one, the reaction was performed using the compound 6-bromo-4-chloro-2, 3-dihydro-1H-inden-1-one (50 mg,0.20 mmol) as a reaction starting material instead of 5-bromo-4-chloro-2, 3-dihydro-1H-inden-1-one, and the reaction solution was concentrated under reduced pressure and then separated and purified by preparative thin layer chromatography (developing solvent: ethyl acetate=100%) to give the title compound, 30mg. ESI-MS (m/z): 620.2,622.2[ M+H ]] +
Step two: synthesis of 7- (7-chloro-3- (methylamino) -2, 3-dihydro-1H-inden-5-yl) -3- ((1- (4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butyryl) -4-hydroxypiperidin-4-yl) methyl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 29)
According to the procedure described in example twenty-second step, the reaction was carried out using compound 29-1 (30 mg,0.05 mmol) as a reaction starting material instead of compound 21-1, and the reaction solution was purified by preparative high performance liquid chromatography (method C) and freeze-dried to give the formate salt of the title compound, 13mg.
The structural characterization is as follows:
1 H NMR(400MHz,DMSO-d 6 ):δ8.51(d,J=1.6Hz,1H),8.29(s,1H),8.17(d,J=1.4Hz,1H),8.08(d,J=4.8Hz,1H),7.87(s,1H),7.81(q,J=2.8Hz,1H),6.26(td,J H-F =55.2Hz,J H-H =4.4Hz,1H),5.98(dd,J=6.0,2.4Hz,1H),5.15-4.86(m,2H),4.28(t,J=6.8Hz,1H),4.04-3.98(m,1H),3.96-3.89(m,2H),3.69(d,J=13.6Hz,1H),3.31-3.21(m,2H),3.03-2.87(m,3H),2.86-2.77(m,1H),2.61-2.52(m,1H),2.43-2.32(m,4H),1.93-1.83(m,1H),1.60-1.30(m,4H).ESI-MS(m/z):635.2,637.2[M+H] +
example thirty-one 7- (4-chloro-3- (methylamino) -2, 3-dihydro-1H-inden-5-yl) -3- ((1- (4, 4-difluoro-) -1-) 3- (3-fluoro-1H-pyrazol-1-yl) butyryl) -4-hydroxypiperidin-4-yl) methyl) thieno [3,4-d]Pyrimidin-4 (3H) -ones Synthesis of Compound 30
Step one: synthesis of 7-chloro-6- (4, 5-tetramethyl-1, 3, 2-dioxaborane-2-yl) -2, 3-dihydro-1H-inden-1-one (Compound 30-2)
According to the procedure described in step two of example seven, the reaction was carried out using compound 30-1 (450 mg,1.83 mmol) as a reaction starting material instead of compound 7-2, the reaction liquid was cooled to room temperature, concentrated under reduced pressure, and then separated and purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate=6/1 (v/v)) to give the title compound, 130mg. ESI-MS (m/z): 293.1,295.1[ M+H ]] +
Step two: synthesis of 7- (4-chloro-3-oxo-2, 3-dihydro-1H-inden-5-yl) -3- ((1- (4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butyryl) -4-hydroxypiperidin-4-yl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 30-3)
According to the procedure described in step two of example one, using compound 10-3 (50 mg,0.09 mmol) and compound 30-2 (41 mg,0.14 mmol) as reaction starting materials in place of compound 1-2 and (1H-indazol-6-yl) boric acid, respectively, the reaction solution was cooled to room temperature, filtered through celite, and the filtrate was concentrated under reduced pressure, and then purified by preparative thin layer chromatography (developing solvent: dichloromethane/methanol=10/1 (v/v)) to give the title compound, 25mg. ESI-MS (m/z): 620.1,622.1[ M+H ] ] +
Step three: synthesis of 7- (4-chloro-3- (methylamino) -2, 3-dihydro-1H-inden-5-yl) -3- ((1- (4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butyryl) -4-hydroxypiperidin-4-yl) methyl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 30)
Compound 30-3 (25 mg,0.04 mmol) was dissolved in a methylamine/ethanol solution (2 mL,33% wt) and reacted at room temperature for 17 hours, followed by concentration under reduced pressure, and further methanol (2 mL), sodium borohydride (3 mg,0.08 mmol) and reaction at room temperature for 1 hour were sequentially added. The reaction mixture was concentrated under reduced pressure, and then purified by preparative high performance liquid chromatography (method G), and lyophilized to give the title compound as trifluoroacetate salt (8 mg).
The structural characterization is as follows:
1 H NMR(400MHz,Methanol-d 4 )δ8.52(d,J=1.2Hz,1H),7.97(d,J=7.7Hz,1H),7.63(d,J=7.8Hz,1H),7.60-7.53(m,1H),7.44(d,J=7.8Hz,1H),6.09(td,J H-F =55.4Hz,J H-H =4.4Hz,1H),5.89-5.81(m,1H),4.97(d,J=6.7Hz,1H),4.94-4.88(m,1H),4.18-4.04(m,1H),4.00(d,J=16.6Hz,2H),3.82-3.73(m,1H),3.48-3.28(m,4H),3.20-3.07(m,2H),3.09-2.98(m,1H),2.94-2.81(m,1H),2.77(s,3H),2.67-2.55(m,1H),2.42(dd,J=15.1,7.7Hz,1H),1.76-1.42(m,4H).ESI-MS(m/z):635.2,637.2[M+H] +
example thirty-two 7- (2-chloro-4-fluorophenyl) -3- ((1- (3-fluoro-1H-pyrazol-1-yl) butanoyl) -4-hydroxy Piperidin-4-yl) methyl) thieno [3,4-d]Synthesis of pyrimidin-4 (3H) -one (compound 32)
Compound 10-3 (40 mg,0.08 mmol) was dissolved in 1, 4-dioxane (2 mL), and (2-chloro-4-fluorobenzene) boric acid (15 mg,0.08 mmol) and water (0.1 mL) were added under nitrogen protection, after nitrogen protection, triphenylphosphine palladium (9 mg,0.007 mmol) and sodium bicarbonate (21 mg,0.23 mmol) were added in sequence, after three nitrogen substitutions, reacted at 85℃for 5h, cooled to room temperature, concentrated under reduced pressure, and purified by preparative thin layer chromatography (developing solvent: dichloromethane/methanol=15/1 (v/v)) to give a crude product of the title compound, which was purified by preparative high performance liquid chromatography (method C), freeze-dried to give the title compound, 20mg.
The structural characterization is as follows:
1 H NMR(400MHz,Methanol-d 4 )δ8.52(t,J=1.4Hz,1H),7.97(d,J=7.2Hz,1H),7.63(dd,J=8.8,6.0Hz,1H),7.59(dt,J=6.4,2.4Hz,1H),7.42(ddd,J=8.8,2.8,1.2Hz,1H),7.26-7.19(m,1H),6.12(td,J H-F =55.4Hz,J H-H =4.4Hz,1H),5.87(ddd,J=10.4,5.6,2.4Hz,1H),5.00-4.90(m,1H),4.14(t,J=12.8Hz,1H),4.07-3.97(m,2H),3.84-3.75(m,1H),3.48-3.35(m,2H),3.10-3.00(m,1H),2.98-2.82(m,1H),1.77-1.47(m,4H).ESI-MS(m/z):584.1,586.1[M+H] +
example thirty-three 7- (4-chloro-1- (methylamino) -2, 3-dihydro-1H-inden-5-yl) -3- ((4-hydroxy-1-) ((R) -3-Benzobutyryl) piperidin-4-yl) methyl) thieno [3,4-d]Synthesis of pyrimidin-4 (3H) -one (compound 33)
Step one: synthesis of tert-butyl (4-chloro-5- (3- ((4-hydroxy-1- ((R) -3-phenylbutyryl) piperidin-4-yl) methyl) -4-oxo-3, 4-dihydrothieno [3,4-d ] pyrimidin-7-yl) -2, 3-dihydro-1H-inden-1-yl) (methyl) carbamate (Compound 33-1)
According to the procedure described in step two of example one, the reaction was carried out using compound 23-3 (30 mg,0.07 mmol) as a reaction starting material instead of (1H-indazol-6-yl) boric acid, and the reaction solution was concentrated under reduced pressure and then purified by preparative thin layer chromatography (developing solvent: dichloromethane/methanol=20/1 (v/v)) to give the title compound, 30mg. ESI-MS (m/z): 691.3,693.3[ M+H ]] +
Step two: synthesis of 7- (4-chloro-1- (methylamino) -2, 3-dihydro-1H-inden-5-yl) -3- ((4-hydroxy-1- ((R) -3-phenylbutyryl) piperidin-4-yl) methyl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 33)
According to the procedure described in seventeenth step of example, compound 33-1 (30 mg,0.04 mmol) was used as a reaction starting material instead of compound 16-5 to conduct a reaction, and the reaction solution was concentrated under reduced pressure, and was separated and purified by preparative high performance liquid chromatography (method L), and freeze-dried to give formate salt of the title compound, 18mg.
The structural characterization is as follows:
1 H NMR(400MHz,DMSO-d 6 )δ8.61(s,1H),7.94(d,J=10.4Hz,1H),7.48(d,J=7.8Hz,1H),7.40(d,J=8.0Hz,1H),7.32-7.22(m,4H),7.15(m,1H),4.89(brs,1H),4.24(t,J=6.8Hz,1H),4.09-3.99(m,1H),3.90(q,J=14.0Hz,1H),3.88-3.78(m,1H),3.72-3.58(m,1H),3.27-3.10(m,3H),3.07-2.98(m,1H),2.92-2.78(m,2H),2.69-2.53(m,2H),2.43-2.36(m,1H),2.35(s,3H),1.95-1.87(m,1H),1.58-1.15(m,4H),1.20(dd,J=7.2,2.0Hz,3H).ESI-MS(m/z):591.3,593.2[M+H] +
example thirty-four 7- (4-chloro-2- (methylamino) -2, 3-dihydro-1H-inden-5-yl) -3- ((1- (4, 4-difluoro-) -1-) 3- (3-fluoro-1H-pyrazol-1-yl) butyryl) -4-hydroxypiperidin-4-yl) methyl) thieno [3,4-d]Pyrimidin-4 (3H) -ones Synthesis of Compound 34
Step one: synthesis of 5-bromo-4-chloro-1H-indene (Compound 34-1)
Compound 30-1 (2.00 g,8.15 mmol) was dissolved in methanol (100 mL) at room temperature, sodium borohydride (0.60 g,16.29 mmol) was added in portions and stirred at room temperature for 2h. The solvent was drained under reduced pressure, quenched with dilute hydrochloric acid (20 mL,1 mol/L), extracted with ethyl acetate, dried over anhydrous sodium sulfate, filtered, the filtrate concentrated under reduced pressure, the concentrate dissolved in toluene (100 mL), p-toluenesulfonic acid (139 mg,0.81 mmol) was added and reacted at 120℃for 3h. The solvent was drained under reduced pressure and purified by column chromatography on silica gel (eluent: petroleum ether=100%) to give the title compound, 1.70g. The structural characterization is as follows:
1 H NMR(400MHz,CDCl 3 )δ7.35(d,J=8.0Hz,1H),7.13(d,J=8.0Hz,1H),6.96-6.89(m,1H),6.57(dt,J=5.6,2.0Hz,1H),3.38-3.36(m,2H).
step two: synthesis of 3-bromo-2-chloro-6, 6 a-dihydro-1 aH-indeno [1,2-b ] oxirane (Compound 34-2)
Compound 34-1 (1.50 g,6.54 mmol) was dissolved in dichloromethane (160 mL) at room temperature, and m-chloroperoxybenzoic acid (3.98 g,19.61 mmol) was added and reacted at room temperature for 18h. The solvent was drained under reduced pressure and purified by column chromatography on silica gel (eluent: petroleum ether/dichloromethane=1/1 (v/v)) to give the title compound, 900mg.
The structural characterization is as follows:
1 H NMR(400MHz,CDCl 3 )δ7.48(d,J=8.0Hz,1H),6.98(d,J=8.0Hz,1H),4.50-4.45(m,1H),4.12(s,1H),3.22(d,J=18.4Hz,1H),6.98(dd,J=18.4,2.4Hz,1H).
step three: synthesis of tert-butyl (5-bromo-4-chloro-2, 3-dihydro-1H-inden-2-yl) (methyl) carbamate (Compound 34-3)
Compound 34-2 (900 mg,3.67 mmol) was dissolved in toluene (50 mL) at room temperature, and silica gel (2.70 g) was added and reacted at 120℃for 6h. The filtrate was filtered, dried under reduced pressure, and the residue was dissolved in methanol (2.6 mL), aqueous methylamine (1 mL,40% wt.) was added, and after stirring for 10min, sodium borohydride (94 mg,2.54 mmol) was added, and after reacting at room temperature for 1h, di-tert-butyl dicarbonate (3.24 g,14.83 mmol) was added, and the reaction was continued at room temperature for 1h. The solvent was drained under reduced pressure and purified by column chromatography on silica gel (eluent: petroleum ether/ethyl acetate=30/1 (v/v)) to give the title compound, 500mg. ESI-MS (m/z): 304.3,306.3[ M-56+H ]] +
Step four: synthesis of tert-butyl (4-chloro-5- (4, 5-tetramethyl-1, 3, 2-dioxaborane-2-yl) -2, 3-dihydro-1H-inden-2-yl) (methyl) carbamate (Compound 34-4)
According to the procedure described in step two of example seven, using compound 34-3 (200 mg,0.65 mmol) as a reaction starting material instead of compound 7-2, the reaction solution after concentration was separated and purified by preparative thin layer chromatography (developer: petroleum ether/ethyl acetate=60/1 (v/v)) to give the title compound, 80mg. ESI-MS (m/z): ESI-MS (m/z): 352.3,354.3[ M-56+H ] ] +
The structural characterization is as follows:
1 H NMR(400MHz,CDCl 3 )δ7.52(d,J=7.2Hz,1H),7.08(d,J=7.2Hz,1H),5.08(brs,1H),3.30-3.15(m,2H),2.99(td,J=15.2,6.0Hz,2H),2.68(s,3H),1.46(s,9H),1.36(s,12H).ESI-MS(m/z):352.3,354.3[M-56+H] +
step five: synthesis of tert-butyl (4-chloro-5- (3- ((1- (4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butyryl) -4-hydroxypiperidin-4-yl) methyl) -4-oxo-3, 4-dihydrothieno [3,4-d ] pyrimidin-7-yl) -2, 3-dihydro-1H-inden-2-yl) (methyl) carbamate (compound 34-5)
According to the procedure described in example one, step two, compound 10-3 (15 mg,0.03 mmol) and the compound were usedThe reaction was carried out using 34-4 (12 mg,0.03 mmol) as a reaction starting material instead of the compounds 1-2 and (1H-indazol-6-yl) boric acid, respectively, and the reaction solution after concentration under reduced pressure was separated and purified by preparative thin layer chromatography (eluent: ethyl acetate/methanol=60/1 (v/v)) to give the title compound, 10mg. ESI-MS (m/z): 735.3,737.3[ M+H ]] +
Step six: synthesis of 7- (4-chloro-2- (methylamino) -2, 3-dihydro-1H-inden-5-yl) -3- ((1- (4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butyryl) -4-hydroxypiperidin-4-yl) methyl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 34)
According to the procedure described in the tenth step of example, compound 34-5 (10 mg,0.01 mmol) was used as a reaction starting material instead of compound 10-1, and the reaction mixture was concentrated under reduced pressure and purified by preparative high performance liquid chromatography (method C), followed by freeze-drying to give the formate salt of the title compound, 2mg.
The structural characterization is as follows:
1 H NMR(400MHz,Methanol-d 4 )δ8.73-8.45(br,1H),8.49(d,J=1.2Hz,1H),7.96(d,J=7.6Hz,1H),7.59(dt,J=6.0,2.8Hz,1H),7.43(d,J=7.6Hz,1H),7.30(d,J=7.6Hz,1H),6.11(td,J H-F =55.2Hz,J H-H =4.4Hz,1H),5.87(ddd,J=9.6,6.0,2.8Hz,1H),5.02-4.93(m,1H),4.21-4.08(m,1H),4.03(d,J=4.0Hz,1H),3.99(s,1H),3.94-3.85(m,1H),3.84-3.73(m,1H),3.54-3.34(m,4H),3.18-2.98(m,3H),2.88(ddd,J=30.0,16.4,4.0Hz,1H),2.63(s,3H),1.80-1.44(m,4H).ESI-MS(m/z):635.2,637.2[M+H] +
example thirty-five 7- (4-chloro-2- (methylamino) -2, 3-dihydro-1H-inden-5-yl) -3- ((1- (3-cyclopropyl-) -o 3-phenylpropionyl) -4-hydroxypiperidin-4-yl methyl) thieno [3,4-d]Synthesis of pyrimidin-4 (3H) -one (compound 35)
Step one: synthesis of tert-butyl (4-chloro-5- (3- ((1- (3-cyclopropyl-3-phenylpropionyl) -4-hydroxypiperidin-4-yl) methyl) -4-oxo-3, 4-dihydrothieno [3,4-d ] pyrimidin-7-yl) -2, 3-dihydro-1H-inden-2-yl) (methyl) carbamate (compound 35-1)
According to the procedure described in step two of example one, using compound 19-1 (20 mg,0.04 mmol) and compound 34-5 (16 mg,0.04 mmol) as reaction starting materials in place of compound 1-2 and (1H-indazol-6-yl) boric acid, respectively, the reaction solution was concentrated under reduced pressure and then separated and purified by preparative thin layer chromatography (developer: ethyl acetate/methanol=60/1 (v/v)) to give the title compound, 12mg. ESI-MS (m/z): 717.3,719.3[ M+H ]] +
Step two: synthesis of 7- (4-chloro-2- (methylamino) -2, 3-dihydro-1H-inden-5-yl) -3- ((1- (3-cyclopropyl-3-phenylpropionyl) -4-hydroxypiperidin-4-yl) methyl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 35)
According to the procedure described in step two of example ten, the reaction was carried out using compound 35-1 (12 mg,0.02 mmol) as a reaction starting material instead of compound 10-1, and the reaction solution was purified by preparative high performance liquid chromatography (method C) and freeze-dried to give the formate salt of the title compound, 8mg.
The structural characterization is as follows:
1 H NMR(400MHz,Methanol-d 4 )δ8.65-8.53(brs,1H),8.50(d,J=7.1Hz,1H),7.96(s,0.4H),7.90(s,0.6H),7.45(d,J=7.7Hz,1H),7.38-7.22(m,5H),7.22-7.14(m,1H),4.24-3.96(m,3H),3.93-3.69(m,2H),3.52(dt,J=15.5,7.5Hz,2H),3.22-3.10(m,2H),3.10-2.78(m,3H),2.71(s,3H),2.33-2.24(m,1H),1.65-1.12(m,5H),0.66-0.56(m,1H),0.46-0.36(m,1H),0.34-0.25(m,1H),0.13-0.04(m,1H).ESI-MS(m/z):617.3,619.3[M+H] +
example thirty-six 7- (7-chloro-2- (methylamino) -2, 3-dihydro-1H-inden-5-yl) -3- ((1- (4, 4-difluoro-) -1-) 3- (3-fluoro-1H-pyrazol-1-yl) butyryl) -4-hydroxypiperidin-4-yl) methyl) thieno [3,4-d]Pyrimidin-4 (3H) -ones Synthesis of (Compound 36)
Step one: synthesis of 5-bromo-7-chloro-1H-indene (Compound 36-2)
According to the procedure described in thirty-four steps one of example, a reaction was performed using compound 36-1 (12 mg,0.02 mmol) as a reaction raw material instead of compound 30-1, the reaction solution was concentrated under reduced pressure to remove the solvent, the crude product was washed with petroleum ether, filtered, and the filtrate was collected and concentrated under reduced pressure to give the title compound, 560mg.
The structural characterization is as follows:
1 H NMR(400MHz,CDCl 3 ):δ7.44(d,J=1.6Hz,1H),7.33(d,J=1.5Hz,1H),6.82(dt,J=5.5,1.9Hz,1H),6.65(dt,J=5.5,2.0Hz,1H),3.38(t,J=1.9Hz,2H).
step two: synthesis of 3-bromo-5-chloro-6, 6 a-dihydro-1 aH-indeno [1,2-b ] oxirane (Compound 36-3)
According to the procedure described in thirty-four steps two of example, using 36-2 (560 mg,2.44 mmol) as a reaction starting material instead of 34-1, the title compound was obtained by separation and purification by silica gel column chromatography (eluent: petroleum ether/dichloromethane=2/1 (v/v)).
The structural characterization is as follows:
1 H NMR(400MHz,CDCl 3 ):δ7.53(d,J=1.7Hz,1H),7.42(d,J=1.7Hz,1H),4.26(dd,J=2.7,1.4Hz,1H),4.15(t,J=2.8Hz,1H),3.20(dd,J=18.6,1.6Hz,1H),2.89(dd,J=18.6,3.0Hz,1H).
step three: synthesis of tert-butyl (6-bromo-4-chloro-2, 3-dihydro-1H-inden-2-yl) (methyl) carbamate (Compound 36-4)
According to the procedure described in thirty-four steps three of example, using 36-3 (400 mg,1.63 mmol) as a reaction starting material instead of 34-2, the title compound, 310mg, was obtained by separation and purification by silica gel column chromatography (eluent: petroleum ether/tetrahydrofuran=30/1 (v/v)).
The structural characterization is as follows:
1 H NMR(400MHz,CDCl 3 ):δ7.32(s,1H),7.24(s,1H),5.26-4.93(m,1H),3.25-3.12(m,2H),3.05-2.87(m,2H),2.72(s,3H),1.46(s,9H).
step four: synthesis of tert-butyl (3- ((1- (4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butyryl) -4-hydroxypiperidin-4-yl) methyl) -4-oxo-3, 4-dihydrothieno [3,4-d ] pyrimidin-7-yl) -2, 3-dihydro-1H-inden-2-yl) (methyl) carbamate (Compound 36-5)
According to the procedure described in example twenty-two step one, starting from compound 36-4 (30 mg,0.08 mmol) instead of 5-bromo-4-chloro-2, 3-dihydro-1H-inden-1-one, the title compound was obtained as 40mg by preparative thin layer chromatography separation and purification (developer: ethyl acetate/methanol=60/1 (v/v)). ESI-MS (m/z): 735.2,737.2[ M+H ]] +
Step five: synthesis of 7- (7-chloro-2- (methylamino) -2, 3-dihydro-1H-inden-5-yl) -3- ((1- (4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butyryl) -4-hydroxypiperidin-4-yl) methyl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 36)
According to the procedure described in the tenth step of example, compound 36-5 (35 mg,0.05 mmol) was used as a reaction starting material instead of compound 10-1, and the reaction mixture was purified by preparative high performance liquid chromatography (method C), diluted hydrochloric acid (0.5 mL,1 mol/L) was added to the preparation, and freeze-dried to give hydrochloride of the title compound, 20mg.
The structural characterization is as follows:
1 H NMR(400MHz,Methanol-d 4 ):δ8.55(s,1H),8.45(d,J=5.6Hz,1H),7.87(s,1H),7.73(s,1H),7.63-7.56(m,1H),6.12(td,J H-F =55.2Hz,J H-H =4.4Hz,1H),5.92-5.84(m,1H),5.00-4.94(m,1H),4.22-4.04(m,4H),3.85-3.75(m,1H),3.64-3.53(m,2H),3.48-3.36(m,2H),3.35-3.32(m,1H),3.26-3.19(m,1H),3.11-3.02(m,1H),2.97-2.84(m,1H),2.80(s,3H),1.79-1.46(m,4H).ESI-MS(m/z):635.3,637.3[M+H] +
example thirty-seven 7- (7-chloro-2- (methylamino) -2, 3-dihydro-1H-inden-5-yl) -3- ((1- (3-cyclopropyl-) -o 3-phenylpropionyl) -4-hydroxypiperidin-4-yl methyl) thieno [3,4-d]Synthesis of pyrimidin-4 (3H) -one (compound 37)
Step one: synthesis of tert-butyl (4-chloro-6- (3- ((1- (3-cyclopropyl-3-phenylpropionyl) -4-hydroxypiperidin-4-yl) methyl) -4-oxo-3, 4-dihydrothieno [3,4-d ] pyrimidin-7-yl) -2, 3-dihydro-1H-inden-2-yl) (methyl) carbamate (compound 37-1)
According to the procedure described in example twenty-second step one, starting from compound 19-1 (38 mg,0.07 mmol) and compound 36-4 (30 mg,0.08 mmol) in place of compound 10-3 and 5-bromo-4-chloro-2, 3-dihydro-1H-inden-1-one, respectively, the title compound was obtained by separation and purification by preparative thin layer chromatography (developer: ethyl acetate/methanol=60/1 (v/v)). ESI-MS (m/z): 717.4,719.4[ M+H ]] +
Step two: synthesis of 7- (7-chloro-2- (methylamino) -2, 3-dihydro-1H-inden-5-yl) -3- ((1- (3-cyclopropyl-3-phenylpropionyl) -4-hydroxypiperidin-4-yl) methyl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 37)
According to the procedure described in the tenth step of example, compound 37-1 (50 mg,0.07 mmol) was used as a reaction starting material instead of compound 10-1, and the reaction mixture was purified by preparative high performance liquid chromatography (method C), diluted hydrochloric acid (0.5 mL,1 mol/L) was added to the preparation, and freeze-dried to give hydrochloride of the title compound, 28mg.
The structural characterization is as follows:
1 H NMR(400MHz,Methanol-d 4 ):δ8.57(d,J=9.6Hz,1H),8.44(d,J=15.2Hz,1H),7.86(s,1H),7.72(s,1H),7.38-7.16(m,5H),4.33-4.12(m,2H),4.13-4.04(m,1H),3.99-3.85(m,1H),3.81-3.69(m,1H),3.65-3.52(m,2H),3.36-3.32(m,1H),3.30-3.16(m,2H),3.08-2.70(m,6H),2.34-2.23(m,1H),1.71-1.27(m,3.5H),1.26-1.13(m,1H),0.82-0.73(m,0.5H),0.67-0.58(m,1H),0.48-0.38(m,1H),0.36-0.26(m,1H),0.15-0.06(m,1H).ESI-MS(m/z):617.3,619.3[M+H] +
examples thirty-eight 7- (6-chloro-2- (methylamino) -2, 3-dihydro-1H-inden-5-yl) -3- ((1- (4, 4-difluoro-) -1-) 3- (3-fluoro-1H-pyrazol-1-yl) butyryl) -4-hydroxypiperidin-4-yl) methylThieno [3,4-d]Pyrimidin-4 (3H) -ones Synthesis of (Compound 38)
Step one: synthesis of 5-bromo-6-chloro-2- (hydroxyimino) -2, 3-dihydro-1H-inden-1-one (Compound 38-1)
Compound 24-1 (200 mg,0.81 mmol) and trimethylchlorosilane (106 mg,0.98 mmol) were dissolved in methyl tert-butyl ether (20 mL) at room temperature, isoamyl nitrite (115 mg,0.98 mmol) was slowly added dropwise, and the reaction was stirred at room temperature for 2h. The reaction was filtered and the filter cake was washed with methyl tert-butyl ether (2X 10 mL) to give crude title compound, 165mg. ESI-MS (m/z): 274.0,276.0[ M+H ]] +
Step two: synthesis of 2-amino-5-bromo-6-chloro-2, 3-dihydro-1H-inden-1-ol (Compound 38-2)
Compound 38-1 (160 mg,0.58 mmol) was dissolved in dry tetrahydrofuran (20 mL) at room temperature, sodium borohydride (86 mg,2.33 mmol) and boron trifluoride etherate (331 mg,2.33 mmol) were added, and the reaction was warmed to 65℃and stirred for 5h. The reaction mixture was cooled to room temperature, quenched with ethanol (20 mL), concentrated to give a crude product, which was purified by preparative HPLC (method C), and lyophilized to give the formate of the title compound (80 mg). ESI-MS (m/z): 262.3,264.3[ M+H ] ] +
Step three: synthesis of 5-bromo-6-chloro-2, 3-dihydro-1H-inden-2-amine (Compound 38-3)
Formate (67 mg,0.22 mmol) of compound 38-2 was dissolved in 1, 2-dichloroethane (20 mL) at room temperature, triethylsilane (6 mL) and boron trifluoride etherate (6 mL) were added, and the reaction was warmed to 85℃and stirred for 18h. The reaction solution was concentrated, and the concentrate was purified by preparative high performance liquid chromatography (method C) and freeze-dried to give the formate of the title compound, 43mg. ESI-MS (m/z): 246.3,248.3[ M+H ]] +
Step four: synthesis of tert-butyl (5-bromo-6-chloro-2, 3-dihydro-1H-inden-2-yl) carbamate (Compound 38-4)
The formazan of compound 38-3 is prepared at room temperatureThe acid salt (43 mg,0.15 mmol) was dissolved in dichloromethane (10 mL), triethylamine (30 mg,0.29 mmol) and di-tert-butyl dicarbonate (218 mg,0.29 mmol) were added, and the mixture was stirred at room temperature for 4h. The reaction solution was concentrated, and the concentrate was purified by preparative thin layer chromatography (developer: petroleum ether/ethyl acetate=8/1 (v/v)) to give the title compound, 40mg. ESI-MS (m/z): 290.3,292.3[ M-56+H ]] +
Step five: synthesis of tert-butyl (5-bromo-6-chloro-2, 3-dihydro-1H-inden-2-yl) (methyl) carbamate (38-5)
According to the procedure described in step one of example seven, using 38-4 (40 mg,0.12 mmol) as a reaction starting material instead of 7-1, the reaction solution was separated and purified by preparative thin layer chromatography (developer: petroleum ether/ethyl acetate=8/1 (v/v)) to give the title compound, 39mg. ESI-MS (m/z): 304.3,306.3[ M-56+H ] ] +
Step six: synthesis of tert-butyl (5-chloro-6- (4, 5-tetramethyl-1, 3, 2-dioxaborane-2-yl) -2, 3-dihydro-1H-inden-2-yl) (methyl) carbamate (Compound 38-6)
According to the procedure described in step two of example seven, the reaction was carried out using compound 38-5 (40 mg,0.11 mmol) as a reaction starting material instead of compound 7-2, and the reaction solution was filtered and concentrated to give a crude title compound, 45mg. ESI-MS (m/z): 352.2,354.2[ M-56+H ]] +
Step seven: synthesis of tert-butyl (5-chloro-6- (3- ((1- (4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butyryl) -4-hydroxypiperidin-4-yl) methyl) -4-oxo-3, 4-dihydrothieno [3,4-d ] pyrimidin-7-yl) -2, 3-dihydro-1H-inden-2-yl) (methyl) carbamate (Compound 38-7)
According to the procedure described in step two of example one, the reaction was carried out using compound 10-3 (9 mg,0.02 mmol) and crude compound 38-6 (6 mg) as reaction starting materials instead of compound 1-2 and (1H-indazol-6-yl) boric acid, respectively, and the reaction solution was filtered and concentrated to give crude title compound, 9mg. ESI-MS (m/z): 735.4,737.4[ M+H ]] +
Step eight: synthesis of 7- (6-chloro-2- (methylamino) -2, 3-dihydro-1H-inden-5-yl) -3- ((1- (4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butyryl) -4-hydroxypiperidin-4-yl) methyl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 38)
According to the procedure described in step two of example ten, the crude product (9 mg) of Compound 38-7 was used as a reaction starting material instead of Compound 10-1, and the reaction mixture was purified by preparative high performance liquid chromatography (method C) and the preparation was lyophilized to give the formate salt of the title compound, 2mg.
The structural characterization is as follows:
1 H NMR(400MHz,Methanol-d 4 )δ8.53(s,1H),8.51(s,1H),7.98(d,J=8.0Hz,1H),7.62-7.57(m,1H),7.50(d,J=8.0Hz,2H),6.12(td,J H-F =55.2Hz,J H-H =4.4Hz,1H),5.91-5.83(m,1H),5.01-4.94(m,1H),4.17-4.09(m,1H),4.07-3.96(m,3H),3.85-3.75(m,1H),3.50-3.35(m,4H),3.17-3.03(m,3H),2.96-2.87(m,1H),2.72(s,3H),1.68-1.46(m,4H).ESI-MS(m/z):635.2,637.2[M+H] +
examples thirty-nine 7- (2-amino-6-chloro-1-hydroxy-2, 3-dihydro-1H-inden-5-yl) -3- ((1- (4, 4-di- Fluoro-3- (3-fluoro-1H-pyrazol-1-yl) butyryl) -4-hydroxypiperidin-4-yl) methyl) thieno [3,4-d]Pyrimidine-4 (3H) Synthesis of Ketone (Compound 39)
Step one: synthesis of tert-butyl (5-bromo-1- ((tert-butoxycarbonyl) oxy) -6-chloro-2, 3-dihydro-1H-inden-2-yl) carbamate (Compound 39-1)
Compound 38-2 (20 mg,0.076 mmol) was dissolved in methylene chloride (10 mL) at room temperature, and triethylamine (16 mg,0.23 mmol) and di-tert-butyl dicarbonate (50 mg,0.23 mmol) were added to react at room temperature for 4h. The reaction solution was concentrated, and the residue was purified by preparative thin layer chromatography (developer: petroleum ether/ethyl acetate=8/1 (v/v)) to give the title compound, 30mg. ESI-MS (m/z): 461.1,463.1[ M+H ]] +
Step two: synthesis of tert-butyl (5-chloro-6- (4, 5-tetramethyl-1, 3, 2-dioxaborane-2-yl) -2, 3-dihydro-1H-inden-2-yl) (methyl) carbamate (Compound 39-2)
According to the procedure described in step two of example seven, the reaction was carried out using compound 39-1 (30 mg,0.06 mmol) as a reaction starting material instead of compound 7-2, and the reaction solution was filtered and concentrated to give a crude title compound, 30mg. ESI-MS (m/z): 509.5,511.5[ M+H ]] +
Step three: synthesis of tert-butyl (1- ((tert-butoxycarbonyl) oxy) -6-chloro-5- (3- ((1- (4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butyryl) -4-hydroxypiperidin-4-yl) methyl) -4-oxo-3, 4-dihydrothieno [3,4-d ] pyrimidin-7-yl) -2, 3-dihydro-1H-inden-2-yl) carbamate (Compound 39-3)
According to the procedure described in step two of example one, using compound 10-3 (31 mg,0.06 mmol) and crude compound 39-2 (30 mg,0.05 mmol) as reaction starting materials, instead of compound 1-2 and (1H-indazol-6-yl) boric acid, respectively, the reaction solution was filtered and concentrated, and the concentrate was purified by preparative thin layer chromatography (developer: dichloromethane/methanol=20/1 (v/v)) to give the title compound, 15mg. ESI-MS (m/z): 837.2,839.3[ M+H ]] +
Step four: synthesis of 7- (2-amino-6-chloro-1-hydroxy-2, 3-dihydro-1H-inden-5-yl) -3- ((1- (4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butyryl) -4-hydroxypiperidin-4-yl) methyl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 39)
According to the procedure described in step two of example ten, the reaction was carried out using compound 39-3 (15 mg, 0.020mmol) as a reaction starting material instead of compound 10-1, and the reaction solution was separated and purified by preparative high performance liquid chromatography (method J), and the preparation was freeze-dried to give the trifluoroacetate salt of the title compound, 6mg.
The structural characterization is as follows:
1 H NMR(400MHz,Methanol-d 4 )δ8.53(d,J=4.0Hz,1H),8.01-7.96(m,1H),7.64-7.57(m,2H),7.54-7.50(m,1H),6.12(td,J H-F =55.2Hz,J H-H =4.4Hz,1H),5.91-5.84(m,1H),5.31(d,J=4.0Hz,0.3H),5.17(d,J=4.0Hz,0.7H),5.01-4.94(m,1H),4.19-4.05(m,4H),3.94(s,3H),3.85-3.75(m,1H),3.59-3.36(m,4H),3.27-3.13(m,2H),3.10-3.01(s,1H),2.97-2.84(m,1H),2.80(s,3H),1.77-1.47(m,4H).ESI-MS(m/z):637.3,639.2[M+H] +
example forty 7- (6-chloro-2- (methylamino) -2, 3-dihydro-1H-inden-5-yl) -3- ((1- (3-cyclopropyl-3-) Phenylpropionyl) -4-hydroxypiperidin-4-yl methyl) thieno [3,4-d]Synthesis of pyrimidin-4 (3H) -one (compound 40)
Step one: synthesis of tert-butyl (5-chloro-6- (3- ((1- (3-cyclopropyl-3-phenylpropionyl) -4-hydroxypiperidin-4-yl) methyl) -4-oxo-3, 4-dihydrothieno [3,4-d ] pyrimidin-7-yl) -2, 3-dihydro-1H-inden-2-yl) (methyl) carbamate (Compound 40-1)
According to the procedure described in step two of example one, using compound 19-1 (30 mg,0.06 mmol) and compound 38-6 (20 mg,0.05 mmol) as reaction starting materials, the reaction was performed in place of compound 1-2 and (1H-indazol-6-yl) boric acid, respectively, and the title compound, 20mg, was obtained by preparative high performance liquid chromatography separation and purification (method M). ESI-MS (m/z): 717.3,719.3[ M+H ]] +
Step two: synthesis of 7- (6-chloro-2- (methylamino) -2, 3-dihydro-1H-inden-5-yl) -3- ((1- (3-cyclopropyl-3-phenylpropionyl) -4-hydroxypiperidin-4-yl) methyl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 40)
According to the procedure described in step two of example ten, the reaction was carried out using compound 40-1 (20 mg,0.03 mmol) as a reaction starting material instead of compound 10-1, and the reaction solution was concentrated under reduced pressure, purified water was added and then freeze-dried to give the hydrochloride salt of the title compound, 14mg.
The structural characterization is as follows:
1 H NMR(400MHz,Methanol-d 4 ):δ8.56(d,J=8.0Hz,1H),8.06(d,J=20.0Hz,1H),7.54(s,1H),7.51(s,1H),7.35-7.14(m,5H),4.24-4.01(m,3H),3.96-3.72(m,2H),3.55-3.43(m,2H),3.22-3.18(m,1H),3.17-3.11(m,1H),2.98-2.82(m,2H),2.78(s,3H),2.72-2.64(m,1H),2.33-2.24(m,1H),1.65-1.54(m,1H),1.50-1.41(m,1H),1.35-1.29(m,2H),1.23-1.15(m,1H),0.82-0.72(m,1H),0.46-0.36(m,1H),0.35-0.26(m,1H),0.14-0.06(m,1H).ESI-MS(m/z):617.3,619.3[M+H] +
example forty-7- (4-chloro-2- (methylamino) -2, 3-dihydro-1H-inden-5-yl) -3- ((1- (3-cyclopropyl-) - 3- (4-fluorophenyl) propionyl) -4-hydroxypiperidin-4-yl) methyl thieno [3,4-d]Pyrimidin-4 (3H) -one (compound 41) Is synthesized by (a)
Step one: synthesis of 7-bromo-3- ((1- (3-cyclopropyl-3- (4-fluorophenyl) propionyl) -4-hydroxypiperidin-4-yl) methyl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 41-2)
According to the procedure described in step three of example ten, the reaction was performed using compound 41-1 (127 mg,0.61 mmol) as a reaction raw material instead of compound 4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butyric acid, and the reaction solution was separated and purified by silica gel column chromatography (eluent: dichloromethane/methanol=19/1 (v/v)) after concentration under reduced pressure to obtain the title compound, 285mg. ESI-MS (m/z): 534.2,536.2[ M+H ]] +
Step two: synthesis of tert-butyl (4-chloro-5- (3- ((1- (3-cyclopropyl-3- (4-fluorophenyl) propionyl) -4-hydroxypiperidin-4-yl) methyl) -4-oxo-3, 4-dihydrothieno [3,4-d ] pyrimidin-7-yl) -2, 3-dihydro-1H-inden-2-yl) (methyl) carbamate (Compound 41-3)
According to the procedure described in step two of example one, using compound 41-2 (26 mg,0.05 mmol) and compound 34-4 (20 mg,0.05 mmol) as reaction starting materials in place of compound 1-2 and (1H-indazol-6-yl) boric acid, respectively, the reaction solution was concentrated under reduced pressure and then separated and purified by preparative thin layer chromatography (eluent: ethyl acetate/methanol=60/1 (v/v)) to give the title compound, 15mg. ESI-MS (m/z): 735.3,737.3[ M+H ]] +
Step three: synthesis of 7- (4-chloro-2- (methylamino) -2, 3-dihydro-1H-inden-5-yl) -3- ((1- (3-cyclopropyl-3- (4-fluorophenyl) propionyl) -4-hydroxypiperidin-4-yl) methyl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 41)
According to the procedure described in step two of example ten, the reaction was carried out using compound 41-3 (15 mg,0.02 mmol) as a reaction starting material instead of compound 10-1, and the reaction solution was purified by preparative high performance liquid chromatography (method C) and freeze-dried to give the formate salt of the title compound, 9mg.
The structural characterization is as follows:
1 H NMR(400MHz,MeOH-d 4 )δ8.55(brs,1H),8.49(d,J=4.0Hz,1H),7.94(d,J=14.8Hz,1H),7.43(d,J=7.6Hz,1H),7.38-7.18(m,3H),7.06(t,J=8.8Hz,1H),6.98(t,J=8.4Hz,1H),4.24-4.10(m,1H),4.09-3.82(m,3H),3.82-3.70(m,1H),3.58-3.40(m,2H),3.30-3.02(m,3H),3.02-2.65(m,3H),2.65(s,3H),2.40-2.20(m,1H),1.70-1.35(m,3H),1.22-0.95(m,2H),0.66-0.54(m,1H),0.47-0.35(m,1H),0.35-0.23(m,1H),0.16-0.04(m,1H).ESI-MS(m/z):635.3,637.3[M+H] +
example forty-two 7- (6-chloro-2-methyl-2- (methylamino) -2, 3-dihydro-1H-inden-5-yl) -3- ((1- (4), 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butyryl) -4-hydroxypiperidin-4-yl) methyl) thieno [3,4-d]Pyrimidine-4 (3H) Synthesis of Ketone (Compound 42)
Step one: synthesis of 5-bromo-6-chloro-2-methyl-2, 3-dihydro-1H-inden-2-amine (Compound 42-1)
Compound 16-1 (125 mg,0.46 mmol) was weighed and dissolved in hydrochloric acid (10 mL,6 mol/L) at room temperature, N-chlorosuccinimide (64 mg,0.48 mmol) was added after stirring and dissolution, and the temperature was raised to 60℃and the reaction was stirred for 8 hours. The reaction solution was then cooled to room temperature, adjusted to pH 9 with saturated aqueous sodium bicarbonate, extracted with ethyl acetate (3X 10 mL), and the organic phases were combined, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The concentrated solution was purified by preparative high performance liquid chromatography (method N) and freeze-dried to give the title compound as trifluoroacetate salt, 66mg. ESI-MS (m/z): 260.0,262.0[ M+H ]] +
Step two: synthesis of tert-butyl (5-bromo-6-chloro-2-methyl-2, 3-dihydro-1H-inden-2-yl) carbamate (Compound 42-2)
The trifluoroacetate salt (30 mg,0.13 mmol) of compound 42-1 was suspended in methylene chloride (4 mL) at room temperature, N-diisopropylethylamine (70 mg,0.54 mmol) and di-tert-butyl dicarbonate (68 mg,0.18 mmol) were added, and reacted at room temperature for 5 hours. The reaction solution was concentrated under reduced pressure to give the title compound, 65mg. ESI-MS (m/z): 304.1,306.1[ M-56+H ]] +
Step three: synthesis of tert-butyl (5-bromo-6-chloro-2-methyl-2, 3-dihydro-1H-inden-2-yl) (methyl) carbamate (Compound 42-3)
According to the procedure described in step one of example seven, the reaction was carried out using compound 42-2 (65 mg,0.16 mmol) as a reaction starting material instead of compound 7-1, and the reaction solution was purified by preparative high performance liquid chromatography (method O) and freeze-dried to give the title compound, 48mg. ESI-MS (m/z): 274.0,276.0[ M-100+H ]] +
Step four: synthesis of tert-butyl (5-chloro-6- (3- ((1- (4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butyryl) -4-hydroxypiperidin-4-yl) methyl) -4-oxo-3, 4-dihydrothieno [3,4-d ] pyrimidin-7-yl) -2-methyl-2, 3-dihydro-1H-inden-2-yl) (methyl) carbamate (Compound 42-4)
Compound 42-3 (63 mg,0.17 mmol) and pinacol biborate (64 mg,0.25 mmol) were dissolved in 1, 4-dioxane (4 mL) at room temperature, potassium acetate (50 mg,0.50 mmol) was added, and after displacement of nitrogen, pd (dppf) Cl was added 2 (12 mg,0.02 mol), nitrogen was replaced again for 5 times, and the reaction was carried out at 80℃for 12 hours under nitrogen protection. Stopping the reaction, and cooling the reaction liquid to room temperature. Then, 10-3 (30 mg,0.06 mmol), 1, 4-dioxane (0.5 mL), water (0.5 mL) and potassium carbonate (23 mg,0.17 mmol) were added in this order, and after replacing nitrogen, pd (PPh) was added 3 ) 4 (7 mg,0.01 mmol). The reaction was carried out at 80℃for 14h under nitrogen protection. The reaction was stopped, cooled to room temperature, and the reaction solution was purified by preparative thin layer chromatography (developer: dichloromethane/methanol=20/1 (v/v)) to give the title compound, 21mg. ESI-MS (m/z): 749.3,751.3[ M+H ] ] +
Step five: synthesis of 7- (6-chloro-2-methyl-2- (methylamino) -2, 3-dihydro-1H-inden-5-yl) -3- ((1- (4, 4-difluoro-3- (3-fluoro-1H-pyrazol-1-yl) butyryl) -4-hydroxypiperidin-4-yl) methyl) thieno [3,4-d ] pyrimidin-4 (3H) -one (compound 42)
Compound 42-4 (21 mg,0.03 mmol) was dissolved in ethyl acetate (1 mL) at room temperature, and hydrochloric acid/ethyl acetate solution (2 mL,4 mol/L) was added. The reaction was carried out at room temperature for 2 hours. The reaction solution was subjected to preparative high performance liquid phase separation and purification (method P), and freeze-dried to give the formate of the title compound, 8mg.
The structural characterization is as follows:
1 H NMR(400MHz,DMSO-d 6 )δ8.60(d,J=1.2Hz,1H),7.95(d,J=4.8Hz,1H),7.84-7.78(m,1H),7.44(d,J=2.4Hz,2H),6.26(td,J H-F =55.2Hz,J H-H =3.6Hz,1H),5.99(ddd,J=6.0,2.8,1.2Hz,1H),5.07-4.86(m,2H),4.06-3.98(m,1H),3.96-3.85(m,2H),3.72-3.65(m,1H),3.30-3.22(m,2H),3.03(t,J=16.8Hz,2H),2.97-2.76(m,4H),2.31(s,3H),1.64-1.33(m,4H),1.26(s,3H).ESI-MS(m/z):649.3,651.3[M+H] +
pharmacological Activity test
Test example 1: inhibition assay of in vitro enzymatic Activity of USP7 (protease)
Test system:
kit USP7 Inhibitor Screening Assay Kit, (BPS catalyst: 79256) containing:
protease USP7 HisFLAGtags enzyme, (BPS catalyst: 80395),
the Substrate Ub-AMC Substrate, (BPS catalyst: 81150),
buffer 5 XUSP 7 Assay Buffer, (BPS catalyst: 79274).
Test instrument:
BMG PHERAstar FS。
test parameters:
USP7 concentration: 3nM; ub-AMC concentration: 100nM;
buffer system: 1.25X105 USP7 Assay Buffer;0.06% bsa;1mM DTT; ddH 2 O;
Compound and enzyme incubation time: 20min;
enzyme kinetic reaction time: 20min;
Fluorescence: excitation wavelength is 350nm, emission wavelength is 460nm.
Parameters of the enzyme-labeled instrument: BMG PHERAstar FS Fluorescence, excitation wavelength 350nm and emission wavelength 460nm.
The test method comprises the following steps:
the test was performed according to the kit instructions, the procedure is as follows,
test group: incubating a mixture of a compound to be tested and protease USP7 for 20min at room temperature in a buffer system, adding a substrate Ub-AMC to start a reaction, and reading the fluorescence value of each well in each cycle (1 min) by adopting an enzyme kinetic method, and reading 20 cycles.
Negative group: the test compounds were replaced with 0.2% dmso aqueous solution and the experimental procedure was the same as the test group.
Blank group: the test compounds were replaced with 0.2% dmso aqueous solution and no protease USP7 was added, the experimental procedure was the same as for the test group.
And (3) data processing:
the relative inhibition activity of each concentration group was calculated with inhibition ratio = 100% - (fluorescence value of test group-fluorescence value of blank group)/(fluorescence value of negative group-fluorescence value of blank group) ×100%. The half maximal Inhibitory Concentration (IC) of the compound was calculated by fitting a curve according to a four parameter model 50 )。
Test results:
inhibition of USP7 activity by the compounds of the present invention was determined as described above and the results are shown in table 1.
TABLE 1 results of USP7 enzyme Activity inhibition assay
/>
Conclusion:
in the enzyme activity inhibition test of USP7, the compound of the invention shows strong inhibition activity, especially the compounds of examples 3, 4, 6, 7, 8, 10, 17, 18, 23, 31, 39 and 42 have extremely strong inhibition activity on USP 7.
Test example 2: test for inhibiting proliferation Activity of Compounds on MM.1S cells
Test system:
cell name/manufacturer: MM.1S/Nanjing Corp Bai
Kit name/manufacturer:Luminescent Cell Viability Assay,Promega。
test instrument:
BMG PHERAstar FS。
test parameters:
cell number: 3000 cells/well
Plating medium: MM.1S 1640+10% FBS
Dosing medium: MM.1S 1640+10% FBS
Compound incubation conditions: 37 ℃,5% CO 2
Incubation time: 5d
Detecting the temperature: RT (reverse transcription) method
Chemiluminescence: luminescent
The test method comprises the following steps:
culturing cells in a medium containing 10% fetal bovine serum, and standing at 37deg.C with 5% CO 2 Culturing under culture conditions. Appropriate amount of cells were plated into 96-well plates and cultured overnight in an incubator. The following day, complete medium containing pre-diluted compounds was added and incubated for 5d at 37 ℃. On the fifth day, detection reagent CellTiter-GLo was added to each well and the Relative Luminescence Units (RLU) of each well was detected by chemiluminescence.
And (3) data processing:
background values were obtained using CellTiter-Glo without cell-containing medium. Cell viability = (sample RLU-background RLU)/(vehicle RLU-background RLU) ×100%, maximum inhibition = 100% -cell viability Maximum concentration of The half maximal Inhibitory Concentration (IC) of the compound was calculated by fitting a curve according to a four parameter model 50 )。
Test results:
the inhibitory activity of the compounds on mm.1s cell proliferation was measured using the methods described above, and the results are shown in table 2.
TABLE 2 inhibition of MM.1S cell proliferation activity by Compounds
/>
Conclusion:
in the test of inhibiting the proliferation activity of the compounds on MM.1S cells, the compounds of the invention show strong inhibition activity on cell proliferation, in particular, the compounds 14-B, 36 and 38 in examples 3, 5, 6, 7, 10, 12 and 21 and example 15 have extremely strong inhibition activity on the proliferation of the MM.1S cells.
Test example 3: biochemical hERG inhibition assay
Test system:
the kit comprises: preconductor TM hERG Fluorescence Polarization Assay, (ThermoFisher Catalog: PV 5365), contains:
positive control compound E4031;
hERG cell membrane;
affinity Tracer tracker; and
hERG buffer.
Test instrument:
BMG PHERAstar FS。
test parameters:
hERG concentration: 1X
Tracer concentration: 1nM
Incubation time: 2h
Fluorescence polarization: excitation wavelength: 540nm; emission wavelength: 590nm
The test method comprises the following steps:
the test was performed according to the kit instructions, the steps were as follows:
test group: the compounds to be tested were added at different concentrations to microwells containing hERG cell membranes, and a Tracer with high hERG affinity was added to each well, and after incubating the microwells for 2 hours at room temperature, the change in fluorescence polarization (excitation wavelength: 540nm; emission wavelength: 590 nm) was detected using a multifunctional microplate reader.
Positive control group: the test compound was replaced with 30. Mu.M positive control compound E4031, and the experimental procedure was the same as that of the test group.
Blank control group: the test compounds were replaced with hERG buffer and hERG cell membranes were not added, and the experimental procedure was the same as for the test group.
And (3) data processing:
based on the data ratio, the percent inhibition (%) of the compounds of the invention against hERG at various concentrations was calculated, and the half Inhibitory Concentration (IC) of the compounds was determined 50 ) Is not limited in terms of the range of (a).
Percentage inhibition ratio (%) = (1- (fluorescence polarization value of test compound-fluorescence polarization value of positive control group)/(fluorescence polarization value of blank control group-fluorescence polarization value of positive control group)) ×100
Test results:
inhibition of hERG by the compounds was determined using the methods described above and the results are shown in table 3.
TABLE 3 hERG inhibition assay results
Examples numbering IC 50 (μM)
10 >10
12 >10
34 >10
38 >10
39 >10
The test results show that the compounds of examples 10, 12, 34, 38, 39 of the invention have low affinity for hERG and compete with the affinity Tracer for IC 50 All > 10. Mu.M.
Test example 4: biochemical CYP enzyme (cytochrome P450) inhibition assay
Test system:
P450-Glo TM CYP1A2 Screening System,(Promega Catalog:V9770);
P450-Glo TM CYP2D6 Screening System,(Promega Catalog:V9890);
P450-Glo TM CYP3A4 Screening System,(Promega Catalog:V9920)。
test instrument:
BMG PHERAstar FS。
the test method comprises the following steps:
the test was performed according to the kit instructions, respectively, as follows:
4.1 inhibition of CYP1 A2:
Test group: the test compounds were added to the microwell plates at different concentrations (10. Mu.M/1. Mu.M), and Luciferin-ME (100. Mu.M), KPO were added to each well 4 (100 mM) and CYP1A2 (0.01 pmol/. Mu.L) were pre-incubated at room temperature for 10min, then added to the NADPH regeneration system to react at room temperature for 30min, and finally added with an equal volume of detection buffer to incubate at room temperature for 20min, and then subjected to chemiluminescent detection.
Negative control group: the experimental procedure was the same as for the test group except that no test compound was added.
Blank control group: the experimental procedure was the same as for the test group except that no test compound was added and CYP1A2 was replaced with CYP1A2 membrane (0.01 pmol/. Mu.L).
4.2 inhibition of CYP2D 6:
test group: the test compounds were added to the microwell plates at different concentrations (10. Mu.M/1. Mu.M), and Luciferin-ME EGE (30. Mu.M), KPO were added to each well 4 (100 mM) and CYP2D6 (5 nM) were pre-incubated at room temperature for 10min, then reacted at 37℃for 30min with the addition of NADPH regeneration system, and finally incubated at room temperature for 20min with an equal volume of detection buffer for chemiluminescent detection.
Negative control group: the experimental procedure was the same as for the test group except that no test compound was added.
Blank control group: the experimental procedure was identical to that of the test group except that no test compound was added and CYP2D6 was replaced with CYP2D6 membrane (5 nM).
4.3 inhibition of CYP3 A4:
test group: the test compounds were added to the microwell plates at different concentrations (10. Mu.M/1. Mu.M), and Luciferin-IPA (3. Mu.M), KPO were added to each well 4 (100 mM) and CYP3A4 (2 nM) were pre-incubated at room temperature for 10min, then added to the NADPH regeneration system to react at room temperature for 30min, and finally added with an equal volume of detection buffer to incubate at room temperature for 20min for chemiluminescent detection.
Negative control group: the experimental procedure was the same as for the test group except that no test compound was added.
Blank control group: the experimental procedure was identical to that of the test group except that no test compound was added and CYP3A4 was replaced with CYP3A4 membrane (2 nM).
And (3) data processing:
percentage inhibition (%) = (1- (chemiluminescent value of test compound concentration group-chemiluminescent value of blank control group)/(chemiluminescent value of negative control group-chemiluminescent value of blank control group)) ×100.
Based on the inhibition of CYP enzyme by the compound at various concentrations, half inhibition concentration (IC 50 ) Or range. IC (integrated circuit) 50 =x× (1-percent inhibition (%))/percent inhibition (%), where X is the compound assayThe concentration was tested.
Test results:
inhibition of three CYPs by the compounds of the present invention was determined as described above and the results are shown in table 4.
TABLE 4 CYPs inhibition test results
Conclusion:
the results show that the compounds of examples 12 and 17 have no obvious inhibition effect on 3 main CYP subtypes, and the potential drug interaction possibility is relatively low, so that the compounds have good patent drug properties.
Test example 5: rat Pharmacokinetic (PK) studies
The pharmacokinetic profile was examined by administering the compounds of the present application to male SD rats Intravenously (IV) and intragastrically (PO), respectively. IV and PO were administered at 1mg/kg and 5mg/kg, respectively, with the IV vehicle being 10% DMSO:10% Solutol (polyethylene glycol-15 hydroxystearate): 80% saline and the PO vehicle being 10% DMSO:10% Solutol (polyethylene glycol-15 hydroxystearate): 80% saline. Blood was collected at various time points after IV and PO dosing (0 h before dosing collected, 0.083, 0.25, 0.5, 1, 2, 4, 6, 8 and 24h venous blood samples after dosing; 0h before dosing collected, 0.25, 0.5, 1, 2, 4, 6, 8 and 24h oral blood samples after dosing). Blood was EDTA.K 2 Anticoagulation, centrifugation to obtain plasma sample, and storing at-80 ℃. Plasma samples were subjected to LC-MS/MS analysis after treatment with precipitated proteins. Pharmacokinetic parameters were calculated using the non-compartmental model using WinNonlin 6.3 software and the results are shown in table 5.
TABLE 5 pharmacokinetic parameters of Compounds administered by IV and PO in rat plasma
The bioavailability of 14-B given orally (PO group) to example 15 was 32.3% in rats compared to intravenous administration. It was shown that 14-B in example 15 of the present application is better absorbed in vehicle 10% DMSO:10% Solutol (polyethylene glycol-15 hydroxystearate): 80% physiological saline.
Test example 6: mouse Pharmacokinetic (PK) studies
The pharmacokinetic profile was examined by administering the compounds of the present application to female balb/c mice Intravenously (IV) and intragastrically (PO), respectively. IV and PO were administered at 1mg/kg and 10mg/kg, respectively, with 5% DMSO to 5% Solutol (polyethylene glycol-15 hydroxystearate) to 90% saline and 10% DMSO to 10% Solutol (polyethylene glycol-15 hydroxystearate) to 80% saline. Collecting venous blood samples 0h before administration, 0.083, 0.25, 0.5, 1, 2, 4, 6, 8 and 24h after administration; blood samples were taken 0h before dosing, 0.25, 0.5, 1, 2, 4, 6, 8 and 24h after dosing, and blood was taken EDTA.K 2 Anticoagulation, centrifugation to obtain plasma sample, and storing at-80 ℃. Plasma samples were subjected to LC-MS/MS analysis after treatment with precipitated proteins. Pharmacokinetic parameters were calculated using the non-compartmental model using WinNonlin 6.3 software, and the results are shown in table 6.
TABLE 6 pharmacokinetic parameters of Compounds administered by IV and PO in plasma in mice
In comparison to intravenous administration, the bioavailability of 14-B given orally (PO group) to example 15 by mice was 23.1%. It was shown that 14-B in example 15 of the present application is better absorbed in vehicle 10% DMSO:10% Solutol (polyethylene glycol-15 hydroxystearate): 80% physiological saline.
Various modifications of the invention, in addition to those described herein, will be apparent to those skilled in the art from the foregoing description. Such modifications are also intended to fall within the scope of the appended claims. Each reference cited in this application (including all patents, patent applications, journal articles, books, and any other publications) is incorporated herein by reference in its entirety.
Reference is made to:
[1]Wu and Kumar,Journal of Medicinal Chemistry,2018,61:422-443.
[2]Turnbull and Ioannidis,Nature,2017,550,481-486.
[3]Pozhidaeva and Bezsonova,DNA Repair,2019,76,30-39.
[4]Chauhan and Tian,Cancer Cell,2012,22,345-358.
[5]Zhou and Wang,Medicinal Chemistry,2018,14,3-18.
[6]Wang and Wu,PLoS One,2017,12,1-23.

Claims (32)

1. a compound of formula II, a pharmaceutically acceptable salt, stereoisomer, or mixture thereof:
wherein,
R 1 is C 1-6 Alkyl, C 6-10 Aryl, 5-to 10-membered heteroarylWherein:
a) The C is 6-10 Aryl and 5-10 membered heteroaryl are each independently optionally substituted with one or more of the following: hydrogen, halogen, -OR a 、-NR b R c 、-C 1-6 alkylene-NR b R c And optionally halogenated C 1-6 An alkyl group;
b) The C is 1-6 The alkyl group is optionally substituted with one or more of the following groups: hydrogen, halogen, -OR a 、-NR b R c and-C 1-6 alkylene-NR b R c
R 2 Selected from hydrogen, halogen and optionally halogenated C 1-6 An alkyl group;
ring A is selected from C 6-10 An aryl group;
ring B is selected from C 3-8 Cycloalkyl, C 3-8 Cycloalkenyl, 3-8 membered heterocycloalkyl, 3-8 membered heterocycloalkenyl, and 5-6 membered heteroaryl;
wherein R R groups are the same or different, m R groups 4 The groups are the same group or different groups;
r is selected from hydrogen, optionally halogenated C 1-6 Alkyl, halogen;
R 4 selected from hydrogen, C 1-6 Alkyl, -OR a 、-NR b R c Halogen and cyano;
R a 、R b 、R c each independently selected from hydrogen and C 1-6 An alkyl group;
R 13 is O;
R 14 selected from C 6-10 Aryl and 5-8 membered heteroaryl, wherein C 6-10 Aryl and 5-8 membered heteroaryl are each independently optionally substituted with one or more substituents as follows: hydrogen, halogen, hydroxy, C 1-6 Alkyl, C 1-6 Alkoxy, amino, cyano;
R 15 selected from H, C 1-6 Alkyl and C 3-8 Cycloalkyl group, wherein C 1-6 Alkyl and C 3-8 Cycloalkyl groups are each independently optionally substituted with one or more substituents as follows: halogen, hydroxy, C 1-6 Alkyl, C 1-6 Alkoxy, amino, cyano;
n is selected from 1, 2, 3, 4, 5 and 6;
m is selected from 0, 1, 2, 3 and 4;
r is selected from 0, 1, 2 and 3.
2. The compound, pharmaceutically acceptable salt, stereoisomer, or mixture thereof according to claim 1,
R 1 selected from C 1-6 Alkyl, C 6-10 Aryl groupWherein the C 1-6 Alkyl and C 6-10 Aryl groups are each independently optionally substituted with one or more of the following groups: hydrogen, halogen, -OR a 、-C 1-6 alkylene-NR b R c
r is selected from 0, 1 and 2;
R 4 selected from hydrogen, -NR b R c 、C 1-6 Alkyl and-OR a
m is selected from 0, 1 and 2.
3. The compound, pharmaceutically acceptable salt, stereoisomer, or mixture thereof according to claim 2, wherein,
r is selected from hydrogen and halogen.
4. A compound, pharmaceutically acceptable salt, stereoisomer, or mixture thereof, as claimed in claim 2 or 3, wherein,
R 1 selected from C 1-6 Alkyl, C 6-10 Aryl groupWherein the C 6-10 Aryl is optionally substituted with one or more of the following: hydrogen, hydroxy, NH 2 -C 1-6 Alkylene-, halogen;
R 4 selected from hydrogen, amino, C 1-6 alkyl-NH- (C) 1-6 Alkyl group 2 N-、C 1-6 Alkyl and hydroxy.
5. The compound, pharmaceutically acceptable salt, stereoisomer, or mixture thereof according to claim 4,
R 1 selected from C 1-6 Alkyl, C 6-10 Aryl groupWherein the C 6-10 Aryl groups are optionally substituted as followsSubstituted by one or more groups of: hydroxy, NH 2 -C 1-6 Alkylene-, halogen;
ring A is C 6-10 Aryl groups.
6. The compound, pharmaceutically acceptable salt, stereoisomer, or mixture thereof according to claim 4,
R 4 selected from hydrogen, amino, CH 3 -NH-、(CH 3 ) 2 N-, methyl and hydroxy.
7. The compound, pharmaceutically acceptable salt, stereoisomer, or mixture thereof, according to any one of claims 1-3, 5-6, wherein,
ring A is phenyl;
the B ring is selected from pyrazolyl, 3-pyrrolinyl, pyrrolidinyl, cyclopentenyl and cyclopentenyl.
8. The compound, pharmaceutically acceptable salt, stereoisomer, or mixture thereof according to claim 4,
ring A is phenyl;
the B ring is selected from pyrazolyl, 3-pyrrolinyl, pyrrolidinyl, cyclopentenyl and cyclopentenyl.
9. The compound, pharmaceutically acceptable salt, stereoisomer, or mixture thereof according to claim 1,
R 1 selected from methyl group,
10. The compound, pharmaceutically acceptable salt, stereoisomer, or mixture thereof according to claim 9,
R 1 selected from methyl group,
11. The compound, pharmaceutically acceptable salt, stereoisomer, or mixture thereof, according to any one of claims 1-3, 5-6, 8-10, wherein,
R 2 Selected from hydrogen, fluorine, methyl and trifluoromethyl.
12. The compound, pharmaceutically acceptable salt, stereoisomer, or mixture thereof according to claim 11, wherein,
R 2 is hydrogen.
13. The compound of any one of claims 1-3, 5-6, 8-10, 12, pharmaceutically acceptable salts, stereoisomers, or mixtures thereof, wherein,
R 14 selected from C 6-10 Aryl and 5-8 membered heteroaryl, wherein C 6-10 Aryl and 5-8 membered heteroaryl are each independently optionally substituted with one or more halogens.
14. The compound, pharmaceutically acceptable salt, stereoisomer, or mixture thereof according to claim 13, wherein,
R 14 selected from C 6-10 Aryl and 5-8 membered nitrogen containing heteroaryl groups, wherein C 6-10 Aryl and 5-8 membered nitrogen containing heteroaryl groups are optionally substituted with one or more halogens.
15. The compound, pharmaceutically acceptable salt, stereoisomer, or mixture thereof according to claim 14, wherein,
R 14 selected from phenyl and pyrazolyl, wherein the phenyl and pyrazolyl are each independently optionally substituted with one or more halogens.
16. The compound, pharmaceutically acceptable salt, stereoisomer, or mixture thereof according to claim 15, wherein,
R 14 selected from phenyl group,
17. The compound of any one of claims 1-3, 5-6, 8-10, 12, pharmaceutically acceptable salts, stereoisomers, or mixtures thereof, wherein,
R 15 selected from C 1-6 Alkyl and C 3-8 Cycloalkyl group, wherein C 1-6 The alkyl group is optionally substituted with one or more halogens.
18. The compound, pharmaceutically acceptable salt, stereoisomer, or mixture thereof according to claim 17, wherein,
R 15 selected from methyl, difluoromethyl, trifluoromethyl and cyclopropyl.
19. The compound, pharmaceutically acceptable salt, stereoisomer, or mixture thereof according to claim 18, wherein,
R 15 selected from methyl, difluoromethyl and cyclopropyl.
20. The compound of any one of claims 1-3, 5-6, 8-10, 12, pharmaceutically acceptable salts, stereoisomers, or mixtures thereof, wherein,
n is selected from 1, 2 and 3.
21. The compound, pharmaceutically acceptable salt, stereoisomer, or mixture thereof according to claim 20, wherein,
n is 1.
22. The compound of any one of claims 1-3, 5-6, 8-10, 12, pharmaceutically acceptable salts, stereoisomers, or mixtures thereof, wherein,
selected from the following groups:
23. a compound according to claim 1, a pharmaceutically acceptable salt, stereoisomer, or mixtures thereof, wherein the compound is selected from the group consisting of:
24. A pharmaceutical composition comprising a compound of any one of claims 1 to 23, a pharmaceutically acceptable salt, stereoisomer, or mixture thereof, and optionally a pharmaceutically acceptable adjuvant.
25. The pharmaceutical composition of claim 24, wherein the pharmaceutical composition further comprises other active ingredients that inhibit the activity of deubiquitinase USP7 or other active ingredients that prevent or treat diseases or disorders associated with the modulation of deubiquitinase USP 7.
26. The pharmaceutical composition of claim 25, wherein the disease or disorder associated with modulation of deubiquitinase USP7 is selected from cancer, neurodegenerative diseases, diabetes, bone and joint diseases, immune disorders, cardiovascular diseases, viral infectious diseases, and bacterial infectious diseases.
27. The pharmaceutical composition of claim 25, wherein the disease or disorder associated with modulation of deubiquitinase USP7 is selected from the group consisting of arthritic disorders, osteoporosis, ischemic diseases.
28. A pharmaceutical product comprising a compound according to any one of claims 1 to 23, a pharmaceutically acceptable salt, stereoisomer or mixture thereof, or a pharmaceutical composition according to any one of claims 24 to 27, and optionally a product package.
29. Use of a compound of any one of claims 1 to 23, a pharmaceutically acceptable salt, stereoisomer, or mixture thereof, or a pharmaceutical composition of any one of claims 24-27, in the manufacture of a deubiquitinase USP7 inhibitor or in the manufacture of a medicament for the prevention or treatment of a disease or disorder associated with deubiquitinase USP7 modulation.
30. The use of claim 29, wherein the disease or disorder associated with deubiquitinase USP7 modulation is selected from cancer, neurodegenerative diseases, diabetes, bone and joint diseases, immune disorders, cardiovascular diseases, viral infectious diseases, and bacterial infectious diseases.
31. The use of claim 29, wherein the disease or disorder associated with modulation of deubiquitinase USP7 is selected from the group consisting of arthritic disorders, osteoporosis offices, ischemic diseases.
32. The use of claim 30, wherein the cancer is selected from at least one of hepatocellular carcinoma, multiple myeloma, colon cancer, lung cancer, prostate cancer, and bladder cancer.
CN202010022368.3A 2020-01-09 2020-01-09 Thienopyrimidinone compounds and medical application thereof Active CN113087718B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202010022368.3A CN113087718B (en) 2020-01-09 2020-01-09 Thienopyrimidinone compounds and medical application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202010022368.3A CN113087718B (en) 2020-01-09 2020-01-09 Thienopyrimidinone compounds and medical application thereof

Publications (2)

Publication Number Publication Date
CN113087718A CN113087718A (en) 2021-07-09
CN113087718B true CN113087718B (en) 2024-02-09

Family

ID=76663502

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010022368.3A Active CN113087718B (en) 2020-01-09 2020-01-09 Thienopyrimidinone compounds and medical application thereof

Country Status (1)

Country Link
CN (1) CN113087718B (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114805367B (en) * 2022-05-24 2023-04-07 郑州大学 Triazole pyrimidine derivative and preparation method and application thereof

Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090105227A1 (en) * 2005-05-09 2009-04-23 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti Spa Thienopyrroles As Antiviral Agents
CN101516891A (en) * 2006-07-06 2009-08-26 阵列生物制药公司 Dihydrothieno pyrimidines as AKT protein kinase inhibitors
CN101675053A (en) * 2006-12-07 2010-03-17 健泰科生物技术公司 Phosphoinositide 3-kinase inhibitor compounds and methods of use
US20130281474A1 (en) * 2010-09-28 2013-10-24 Merck Sharp & Dohme Corp. Fused tricyclic inhibitors of mammalian target of rapamycin
CN103930415A (en) * 2011-09-02 2014-07-16 海布里詹尼克斯股份公司 Selective and reversible inhibitors of ubiquitin specific protease 7
CN108495852A (en) * 2016-09-08 2018-09-04 四川科伦博泰生物医药股份有限公司 2- acylaminothiazole analog derivatives and preparation method thereof and purposes
US20180369245A1 (en) * 2015-12-09 2018-12-27 Cadent Therapeutics, Inc. Thienopyrimidinone nmda receptor modulators and uses thereof
CN109311888A (en) * 2016-06-10 2019-02-05 法国施维雅药厂 New acyclic derivatives, preparation method and the pharmaceutical composition containing them
CN109563094A (en) * 2016-06-10 2019-04-02 法国施维雅药厂 Acyclic derivatives, their preparation method and the pharmaceutical composition containing them that new (miscellaneous) aryl replaces
WO2019105963A1 (en) * 2017-11-29 2019-06-06 Les Laboratoires Servier New piperidinyl derivatives as inhibitors of ubiquitin specific protease 7
CN110240593A (en) * 2018-03-09 2019-09-17 四川科伦博泰生物医药股份有限公司 Substituted aromatic amines compound and its preparation method and application

Patent Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090105227A1 (en) * 2005-05-09 2009-04-23 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti Spa Thienopyrroles As Antiviral Agents
CN101516891A (en) * 2006-07-06 2009-08-26 阵列生物制药公司 Dihydrothieno pyrimidines as AKT protein kinase inhibitors
CN101675053A (en) * 2006-12-07 2010-03-17 健泰科生物技术公司 Phosphoinositide 3-kinase inhibitor compounds and methods of use
US20130281474A1 (en) * 2010-09-28 2013-10-24 Merck Sharp & Dohme Corp. Fused tricyclic inhibitors of mammalian target of rapamycin
CN103930415A (en) * 2011-09-02 2014-07-16 海布里詹尼克斯股份公司 Selective and reversible inhibitors of ubiquitin specific protease 7
US20180369245A1 (en) * 2015-12-09 2018-12-27 Cadent Therapeutics, Inc. Thienopyrimidinone nmda receptor modulators and uses thereof
CN109311888A (en) * 2016-06-10 2019-02-05 法国施维雅药厂 New acyclic derivatives, preparation method and the pharmaceutical composition containing them
CN109563094A (en) * 2016-06-10 2019-04-02 法国施维雅药厂 Acyclic derivatives, their preparation method and the pharmaceutical composition containing them that new (miscellaneous) aryl replaces
CN108495852A (en) * 2016-09-08 2018-09-04 四川科伦博泰生物医药股份有限公司 2- acylaminothiazole analog derivatives and preparation method thereof and purposes
WO2019105963A1 (en) * 2017-11-29 2019-06-06 Les Laboratoires Servier New piperidinyl derivatives as inhibitors of ubiquitin specific protease 7
CN110240593A (en) * 2018-03-09 2019-09-17 四川科伦博泰生物医药股份有限公司 Substituted aromatic amines compound and its preparation method and application

Also Published As

Publication number Publication date
CN113087718A (en) 2021-07-09

Similar Documents

Publication Publication Date Title
CN113365988B (en) SHP2 inhibitor and application thereof
EP3523277B1 (en) Substituted pyrrolidines as cftr modulators
CN108623615B (en) Macrocyclic derivatives of pyrazolo [3,4-d ] pyrimidin-3-one, pharmaceutical compositions and uses thereof
CN106999479B (en) Heteroaryl compounds useful as inhibitors of SUMO activating enzyme
CN114292272A (en) Nucleoside compound and application thereof
RU2676259C2 (en) Dna-pk inhibitors
CN114174299A (en) Degradation of Bruton's Tyrosine Kinase (BTK) by conjugation of BTK inhibitors to E3 ligase ligands and methods of use
EP3608321A1 (en) 1h-imidazo[4,5-h]quinazoline compound as protein kinase inhibitor
EP4129996A1 (en) Novel aminopyrimidine egfr inhibitor
WO2014134772A1 (en) Compounds inhibiting leucine-rich repeat kinase enzyme activity
CA3201793A1 (en) Macrocycles containing a 1,3,4-oxadiazole ring for use as modulators of cystic fibrosis transmembrane conductance regulator
WO2022076621A1 (en) Modulators of cystic fibrosis transmembrane conductance regulator
CN101679418A (en) [2,6] naphthyridines compounds as protein kinase inhibitors
EP4137484A1 (en) Tricyclic compounds as egfr inhibitors
CA3172498A1 (en) Degradation of bruton's tyrosine kinase (btk) by conjugation of btk inhibitors with e3 ligase ligand and methods of use
WO2021180103A1 (en) Degradation of bruton's tyrosine kinase (btk) by conjugation of btk inhibitors with e3 ligase ligand and methods of use
EP2900656B1 (en) Spiro ring compounds as hepatitis c virus (hcv) inhibitors
EP3381919A1 (en) Substituted triazolopiperazine parp inhibitor, preparation method therefor, and use thereof
TW202003496A (en) Novel phosphoinositide 3-kinase inhibitor and preparation method and use thereof
CA3024831C (en) Novel 5h-pyrrolo[2,3-d]pyrimidin-6(7h)-one derivative
CN113087718B (en) Thienopyrimidinone compounds and medical application thereof
JP2023535096A (en) Compounds having kinase inhibitory activity
EP4151634A1 (en) Preparation of biaryl ring-linked aromatic heterocyclic derivative as immunomodulator and use thereof
AU2017341999A1 (en) Inhibitors of mutant isocitrate dehydrogenases and compositions and methods thereof
EP3992193A1 (en) Pyrazolopyrimidine compound, preparation method for same, and applications thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant