CN113493439B - Substituted acrylamide derivative, composition and application thereof - Google Patents

Substituted acrylamide derivative, composition and application thereof Download PDF

Info

Publication number
CN113493439B
CN113493439B CN202110293647.8A CN202110293647A CN113493439B CN 113493439 B CN113493439 B CN 113493439B CN 202110293647 A CN202110293647 A CN 202110293647A CN 113493439 B CN113493439 B CN 113493439B
Authority
CN
China
Prior art keywords
compound
pharmaceutically acceptable
tautomer
stereoisomer
acceptable salt
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
CN202110293647.8A
Other languages
Chinese (zh)
Other versions
CN113493439A (en
Inventor
王义汉
赵九洋
邢青峰
李焕银
艾义新
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.)
Shenzhen Targetrx Inc
Original Assignee
Shenzhen Targetrx Inc
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 Shenzhen Targetrx Inc filed Critical Shenzhen Targetrx Inc
Publication of CN113493439A publication Critical patent/CN113493439A/en
Application granted granted Critical
Publication of CN113493439B publication Critical patent/CN113493439B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4545Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
    • 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
    • A61K31/4985Pyrazines or piperazines ortho- or peri-condensed with heterocyclic ring 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/53Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with three nitrogens as the only ring hetero atoms, e.g. chlorazanil, melamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Abstract

The invention provides a substituted acrylamide derivative, a composition containing the compound and application of the compound, wherein the substituted acrylamide derivative is a compound shown as a formula (I) or a tautomer, a stereoisomer, a prodrug, a crystal form, a pharmaceutically acceptable salt, a hydrate or a solvate of the compound. The compounds of the invention and compositions thereof are useful for the treatment and/or prevention of wild-type and/or mutant EGFR and/or HER2 kinase mediated tumors.

Description

Substituted acrylamide derivative, composition and application thereof
Technical Field
The invention belongs to the technical field of medicines, and particularly relates to substituted acrylamide derivatives with an inhibitory effect on wild and/or mutant EGFR and/or HER2, pharmaceutical compositions containing the same, and preparation methods and applications of the same.
Background
EGFR is a receptor tyrosine kinase that exerts its physiological function in normal tissues by binding to epidermal growth factor (hereinafter also referred to as EGF) as a ligand, and contributes to growth and apoptosis inhibition of epithelial tissues. Somatic mutations in the EGFR gene are known to be carcinogenic: for example, EGFR lacking 746 th to 750 th amino acids in the exon 19 region (hereinafter also referred to as "exon 19 deletion mutation") and EGFR lacking the leucine th to arginine in the exon 21 region (hereinafter also referred to as "L858R mutation") continuously induce EGF-independent kinase activity and cause growth and survival of cancer cells. For example, these mutations are observed in about 30% -50% of non-small cell lung cancers in east asia, and also in about 10% of non-small cell lung cancers in europe and the united states, and thus are considered to be one of the causes of cancer.
Therefore, research and development of EGFR inhibitors as antitumor agents have been actively conducted and applied to the treatment of EGFR mutation-positive lung cancer. For example, gefitinib, erlotinib and afatinib have high antitumor effects on exon 19 deletion-mutated and L858R-mutated EGFR-positive lung cancers, but they cause side effects such as digestive tract diseases and skin diseases when used in therapeutic doses thereof.
Recent studies have found that EGFR of some cancers has a novel mutation in which one or more amino acids are inserted into the exon 20 region (hereinafter also referred to as "exon 20 insertion mutation"), and these cancers have low sensitivity to previously known EGFR inhibitors.
On the other hand, several rare EGFR mutations, such as point mutation or deletion mutation of exon 18 and point mutation of exon 21, have been reported. For example, a novel EGFR point-mutated lung cancer has been found in which glycine at position 719 in the exon 18 region is substituted with an arbitrary amino acid (hereinafter referred to as G719X mutation), and leucine at position 861 in the exon 21 region is substituted with glutamine (hereinafter referred to as L861Q mutation).
HER2 (also known as ErbB 2) is a receptor tyrosine kinase belonging to the ErbB2 family. HER2 is considered as a protooncogene, and gene amplification, mutation, overexpression, and the like of HER2 have been reported in various cancers. In these cancer cells accompanied by HER2 gene abnormality and overexpression, signals of survival, proliferation, and the like of the cancer cells are enhanced by HER2 and signal activation of downstream pathways.
HER2 mutation is one of the common driver mutations in lung cancer, and is mainly manifested by gene amplification, point mutation, exon 20 insertion mutation, and other mutation types (such as deletion insertion mutation, frameshift mutation, etc.), wherein the insertion mutation of exon 20 is the most common. For example, HER2 mutants comprise a YVMA insertion into exon 20 (hereinafter referred to as ex20 insYVMA). Mutant HER2 activates signaling, phosphorylates EGFR, induces tumor formation and spread more efficiently than wild-type HER2.
Therefore, it is presumed that an inhibitor capable of controlling the kinase activity of HER2 exerts an antitumor effect by inhibiting HER 2in cancer cells and the signal transduction of downstream pathways, and thus is considered to be effective as a cancer therapeutic agent.
Therefore, there is a need to further develop new EGFR inhibitors and HER2 inhibitors in order to expect that wild EGFR and/or exon 20 insertion-mutated EGFR, exon 18 point-mutated EGFR, exon 21 point-mutated EGFR, wild HER2 and/or mutated HER2 can be effectively inhibited.
Summary of The Invention
The invention provides a novel acrylamide derivative, a composition containing the same and application thereof, wherein the acrylamide derivative has better inhibitory activity and high selectivity on exon 20 insertion (exon 20 ins) mutant EGFR, exon 18 point mutant EGFR, exon 21 point mutant EGFR, exon 19 deletion (exon 19 del) mutant EGFR, L858R mutant EGFR, exon 19 deletion/T790M mutant EGFR, L858R/T790M mutant EGFR and the like, and has inhibitory activity on wild HER2 and/or mutated HER2, so that an antitumor drug with low toxic and side effects is provided.
In contrast, the invention adopts the following technical scheme:
in one aspect, the present invention relates to a compound of formula (I), or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof:
Figure BDA0002983446160000021
wherein the content of the first and second substances,
ring A is an aromatic ring;
ring C being C 6-10 Aryl or 5 to 10 membered heteroaryl;
A 1 is CR A1 Or N;
A 2 is C or N;
A 4 is CR A4 Or N;
A 5 is C or N;
wherein R is A1 And R A4 Each independently selected from H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Alkoxy, and optionally substituted with one or more R ";
B 1 is CR 1 Or N;
B 2 is CR 2 Or N;
B 3 is CR 3 Or N;
B 4 is CR 4 Or N;
wherein R is 1 、R 2 、R 3 And R 4 Each independently selected from H, D, halogen, -CN, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, -C (O) R a 、-C(O)OR a 、-C(O)NR b R c 、-NR b R c 、-NR a C(O)R b 、-NR a C(O)OR b 、-NR a C(O)NR b R c 、-OR a 、-OC(O)R a 、-OC(O)OR a 、-OC(O)NR b R c 、C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5-to 10-membered heteroaryl, and optionally substituted with one or more R ″;
L is O, S or NR L
Wherein R is L Is selected from H or C 1-6 Alkyl, and optionally substituted with one or more R;
v is (CR) V1 R V2 ) o
Wherein R is V1 And R V2 Each independently selected from H, D, halogen or C 1-6 Alkyl, and optionally substituted with one or more R;
o =1, 2,3, 4,5 or 6;
R 6 is H, D, halogen, -CN, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5-to 10-membered heteroaryl, optionally substituted with one or more R;
R 5 and R 7 Each independently selected from H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Haloalkyl, and the aforementioned groups are optionally substituted with one or more R; or, R 5 And R 7 Together with the double bond to which they are attached form a triple bond;
each R is independently selected from H, D, halogen, -CN, = O, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, -C (O) R a 、-C(O)OR a 、-C(O)NR b R c 、-NR b R c 、-NR a C(O)R b 、-NR a C(O)OR b 、-NR a C(O)NR b R c 、-OR a 、-OC(O)R a 、-OC(O)OR a 、-OC(O)NR b R c 、C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5 to 10 membered heteroaryl; alternatively, two R groups on the same atom or on adjacent atoms may together form C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5 to 10 membered heteroaryl; wherein each group in the definition of R is optionally substituted with one or more D, up to complete deuteration;
m =0, 1,2, 3,4, 5,6, 7, 8 or 9;
r' is independently selected from H, D, halogen, -CN, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, -C (O) R a 、-C(O)OR a 、-C(O)NR b R c 、-NR b R c 、-NR a C(O)R b 、-NR a C(O)OR b 、-NR a C(O)NR b R c 、-OR a 、-OC(O)R a 、-OC(O)OR a 、-OC(O)NR b R c 、C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5 to 10 membered heteroaryl, and optionally substituted with one or more R ";
n =0, 1,2, 3 or 4;
p =0, 1 or 2;
each R' is independently selected from H, D, halogen, -CN, = O, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, -C (O) R a 、-C(O)OR a 、-C(O)NR b R c 、-NR b R c 、-NR a C(O)R b 、-NR a C(O)OR b 、-NR a C(O)NR b R c 、-OR a 、-OC(O)R a 、-OC(O)OR a 、-OC(O)NR b R c 、C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5-to 10-membered heteroaryl, or two R' groups on the same atom or on adjacent atoms may together form C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5 to 10 membered heteroaryl; wherein each group in the definition of R "is optionally substituted with one or more D, up to complete deuteration;
r is independently selected from H, D, halogen, -CN, = O, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, -C (O) R a 、-C(O)OR a 、-C(O)NR b R c 、-NR b R c 、-NR a C(O)R b 、-NR a C(O)OR b 、-NR a C(O)NR b R c 、-OR a 、-OC(O)R a 、-OC(O)OR a 、-OC(O)NR b R c 、C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5 to 10 membered heteroaryl; alternatively, two R groups on the same atom or on adjacent atoms may together form C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5 to 10 membered heteroaryl; wherein each group in the definition of R is optionally substituted with one or more D, up to complete deuteration;
R a 、R b and R c Each independently selected from H, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5-to 10-membered heteroaryl, or R b And R c Together with the N atom to which they are attached form a 3-to 7-membered heterocyclyl or 5-to 10-membered heteroaryl; wherein R is a 、R b And R c Each group in the definition is optionally substituted with one or more D, up to complete deuteration.
In another aspect, the present invention provides a pharmaceutical composition comprising a compound of the present invention, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, and a pharmaceutically acceptable excipient. In particular embodiments, the compounds of the present invention are provided in a therapeutically effective amount. In particular embodiments, the compounds of the present invention are provided in a prophylactically effective amount.
In another aspect, the present invention provides a use of a compound of the present invention or a tautomer, stereoisomer, prodrug, crystal form, pharmaceutically acceptable salt, hydrate, or solvate thereof, or a pharmaceutical composition of the present invention, for the preparation of a medicament for the treatment and/or prevention of a wild-type or mutant EGFR kinase-mediated tumor.
In another aspect, the present invention provides a method of treating and/or preventing a disease in a subject, such as a wild-type and/or mutant EGFR kinase-mediated tumor, comprising administering to the subject a compound of the present invention, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, or a pharmaceutical composition of the present invention.
In specific embodiments, the mutant EGFR is selected from the group consisting of exon 20 insertion mutant EGFR, exon 18 point mutant EGFR, exon 21 point mutant EGFR, exon 19 deletion mutant EGFR, or L858R mutant EGFR.
In a specific embodiment, the exon 20 insertion mutation is a mutation wherein one or more amino acids are inserted into the exon 20 region. In a specific embodiment, the exon 20 insertion mutation is a mutation wherein 1 to 7 amino acids are inserted into the exon 20 region. In a specific embodiment, the exon 20 insertion mutation is a mutation wherein 1 to 4 amino acids are inserted into the exon 20 region. In a specific embodiment, the exon 20 insertion mutation is A763_ Y764insFQEA, V769_ D770insASV, D770_ N771insSVD, D770_ N771insNPG, D770_ N771insG, D770> GY, N771_ P772insN, P772_ R773insPR, H773_ V774insNPH, H773_ V774insPH, H773_ V774insAH, H773_ V774insH, H774_ C774insHV, A761_ E762insEAFQ. In specific embodiments, the exon 20 insertion mutation is V769_ D770insASV, D770_ N771insSVD, D770_ N771insNPG, H773_ V774insNPH, or H773_ V774insPH.
In a specific embodiment, said exon 18 point mutation is selected from the group consisting of a G719X mutation of exon 18 or an E709X mutation of exon 18. In a specific embodiment, the G719X mutation is at least one mutation selected from the group consisting of G719A, G S and G719C. In a specific embodiment, the E709X mutation is at least one mutation selected from the group consisting of E709K and E709A.
In a specific embodiment, said exon 21 point mutation is selected from the group consisting of the L861X mutation of exon 21. In a specific embodiment, the L861X mutation is an L861Q mutation.
In a specific embodiment, the mutant EGFR has a T790M mutation and has at least one mutation selected from the group consisting of an exon 20 insertion mutation, an exon 18 point mutation, an exon 21 point mutation, an exon 19 deletion mutation or a L858R mutation.
In a specific embodiment, the compounds of the present invention, or tautomers, stereoisomers, prodrugs, crystalline forms, pharmaceutically acceptable salts, hydrates or solvates thereof, are used for treating tumor patients expressing EGFR with an exon 20 insertion mutation.
In a specific embodiment, the compounds of the present invention, or tautomers, stereoisomers, prodrugs, crystalline forms, pharmaceutically acceptable salts, hydrates or solvates thereof, are used for treating tumor patients expressing EGFR with a T790M mutation and with an exon 20 insertion mutation.
In a specific embodiment, the compounds of the present invention, or tautomers, stereoisomers, prodrugs, crystalline forms, pharmaceutically acceptable salts, hydrates or solvates thereof, are used for treating a patient having a tumor expressing EGFR with an exon 18 point mutation.
In a specific embodiment, the compounds of the present invention, or tautomers, stereoisomers, prodrugs, crystalline forms, pharmaceutically acceptable salts, hydrates or solvates thereof, are used for treating a tumor patient expressing EGFR having a T790M mutation and having an exon 18 point mutation.
In a specific embodiment, a compound of the invention, or a tautomer, stereoisomer, prodrug, crystal form, pharmaceutically acceptable salt, hydrate, or solvate thereof, is used to treat a tumor patient expressing EGFR with an exon 21 point mutation.
In a specific embodiment, the compounds of the present invention, or tautomers, stereoisomers, prodrugs, crystalline forms, pharmaceutically acceptable salts, hydrates or solvates thereof, are used for treating cancer patients expressing EGFR having the T790M mutation and having an exon 21 point mutation.
In a specific embodiment, the compounds of the present invention, or tautomers, stereoisomers, prodrugs, crystalline forms, pharmaceutically acceptable salts, hydrates or solvates thereof, are used for treating a patient with a tumor expressing a mutant EGFR having an exon 19 deletion.
In a specific embodiment, the compounds of the present invention, or tautomers, stereoisomers, prodrugs, crystalline forms, pharmaceutically acceptable salts, hydrates or solvates thereof, are used to treat cancer patients expressing EGFR with the T790M mutation and with an exon 19 deletion mutant.
In a specific embodiment, the compounds of the present invention, or tautomers, stereoisomers, prodrugs, crystalline forms, pharmaceutically acceptable salts, hydrates or solvates thereof, are used for treating patients with tumors expressing EGFR with L858R mutation.
In a specific embodiment, the compounds of the present invention, or tautomers, stereoisomers, prodrugs, crystalline forms, pharmaceutically acceptable salts, hydrates or solvates thereof, are used for treating tumor patients expressing EGFR with the T790M mutation and with the L858R mutation.
In a specific embodiment, the present invention provides a use of a compound of the present invention or a tautomer, stereoisomer, prodrug, crystal form, pharmaceutically acceptable salt, hydrate, or solvate thereof, or a pharmaceutical composition of the present invention for the preparation of a medicament for the treatment and/or prevention of the following tumors, or a method for the treatment and/or prevention of the following tumors in a subject, comprising administering a compound of the present invention or a tautomer, stereoisomer, prodrug, crystal form, pharmaceutically acceptable salt, hydrate, or solvate thereof, or a pharmaceutical composition of the present invention to the subject: lung cancer, breast cancer, head and neck cancer, brain cancer, uterine cancer, hematopoietic cancer or skin cancer.
In another aspect, the present invention provides a use of a compound of the present invention or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, or a pharmaceutical composition of the present invention, for the preparation of a medicament for the treatment and/or prevention of a tumor mediated by wild type and/or mutant HER2 kinase.
In another aspect, the present invention provides a method of treating and/or preventing a disease, such as a wild-type and/or mutant HER2 kinase-mediated tumor, in a subject, comprising administering to the subject a compound of the present invention, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, or a pharmaceutical composition of the present invention.
In specific embodiments, the mutated HER2 is selected from G309A mutant HER2, S310F mutant HER2, R678Q mutant HER2, L775_ T759 deletion mutant HER2, D769H mutant HER2, V777L mutant HER2, V842I mutant HER2, R869C mutant HER2, L755S mutant HER2, or ex20 insymva mutant HER2.
In a specific embodiment, the ex20insYVMA mutant HER2 is selected from a775_ G776insYVMA mutant HER2 mutation.
In a specific embodiment, the present invention provides a use of a compound of the present invention or a tautomer, stereoisomer, prodrug, crystal form, pharmaceutically acceptable salt, hydrate, or solvate thereof, or a pharmaceutical composition of the present invention for the preparation of a medicament for the treatment and/or prevention of the following tumors, or a method for the treatment and/or prevention of the following tumors in a subject, comprising administering a compound of the present invention or a tautomer, stereoisomer, prodrug, crystal form, pharmaceutically acceptable salt, hydrate, or solvate thereof, or a pharmaceutical composition of the present invention to the subject: lung cancer, gastric cancer or breast cancer.
Other objects and advantages of the present invention will be apparent to those skilled in the art from the following detailed description, examples and claims.
Definition of
Chemical definition
The definitions of specific functional groups and chemical terms are described in more detail below.
When a range of values is recited, it is intended to include each value and every subrange within the range. E.g. "C 1-6 Alkyl "includes C 1 、C 2 、C 3 、C 4 、C 5 、C 6 、C 1-6 、C 1-5 、C 1-4 、C 1-3 、C 1-2 、C 2-6 、C 2-5 、C 2-4 、C 2-3 、C 3-6 、C 3-5 、C 3-4 、C 4-6 、C 4-5 And C 5-6 An alkyl group.
“C 1-6 Alkyl "refers to a straight chain having 1 to 6 carbon atoms orBranched saturated hydrocarbon groups, also referred to herein as "lower alkyl groups. In some embodiments, C 1-4 Alkyl groups are particularly preferred. Examples of such alkyl groups include, but are not limited to: methyl (C) 1 ) Ethyl (C) 2 ) N-propyl (C) 3 ) Isopropyl (C) 3 ) N-butyl (C) 4 ) Tert-butyl (C) 4 ) Sec-butyl (C) 4 ) Isobutyl (C) 4 ) N-pentyl group (C) 5 ) 3-pentyl radical (C) 5 ) Pentyl group (C) 5 ) Neopentyl (C) 5 ) 3-methyl-2-butyl (C) 5 ) Tert-amyl (C) 5 ) And n-hexyl (C) 6 ). Each of the alkyl groups is independently optionally substituted, whether or not the alkyl group is preceded by a "substituted", e.g., 1 to 5 substituents, 1 to 3 substituents, or 1 substituent, with appropriate substituents being defined below.
“C 2-6 Alkenyl "refers to a straight or branched hydrocarbon group having 2 to 6 carbon atoms and at least one carbon-carbon double bond. In some embodiments, C 2-4 Alkenyl groups are preferred. C 2-6 Examples of alkenyl groups include: vinyl radical (C) 2 ) 1-propenyl (C) 3 ) 2-propenyl (C) 3 ) 1-butenyl (C) 4 ) 2-butenyl (C) 4 ) Butadienyl radical (C) 4 ) Pentenyl (C) 5 ) Pentadienyl (C) 5 ) Hexenyl (C) 6 ) And so on. The term "C 2-6 Alkenyl "also includes heteroalkenyl groups in which one or more (e.g., 1,2, 3, or 4) carbon atoms are replaced with a heteroatom (e.g., oxygen, sulfur, nitrogen, boron, silicon, phosphorus). Each of the alkenyl groups is independently optionally substituted, e.g., 1 to 5 substituents, 1 to 3 substituents, or 1 substituent, with suitable substituents being defined below, whether or not the alkenyl group is modified "substituted" or not.
“C 2-6 Alkynyl "refers to a straight or branched hydrocarbon group having 2 to 6 carbon atoms, at least one carbon-carbon triple bond, and optionally one or more carbon-carbon double bonds. In some embodiments, C 2-4 Alkynyl groups are preferred. C 2-6 Examples of alkynyl groups include, but are not limited to: ethynyl (C) 2 ) 1-propynyl (C) 3 ) 2-propynyl (C) 3 ) 1-butynyl (C) 4 ) 2-butynyl (C) 4 ) Pentynyl radical (C) 5 ) Hexynyl (C) 6 ) And so on. The term "C 2-6 Alkynyl also includes heteroalkynyl in which one or more (e.g., 1,2, 3, or 4) carbon atoms are replaced with a heteroatom (e.g., oxygen, sulfur, nitrogen, boron, silicon, phosphorus). Each of the alkynyl groups is independently optionally substituted, whether or not the alkynyl group is modified by "substituted", for example, 1 to 5 substituents, 1 to 3 substituents, or 1 substituent, with appropriate substituents being defined below.
“C 1-6 Alkoxy "refers to the group-OR, where R is substituted OR unsubstituted C 1-6 An alkyl group. In some embodiments, C 1-4 Alkoxy groups are particularly preferred. Specific said alkoxy groups include, but are not limited to: methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, tert-butoxy, sec-butoxy, n-pentoxy, n-hexoxy and 1,2-dimethylbutoxy. Each of the alkoxy groups is independently optionally substituted, whether or not the alkoxy group is modified by "substituted", for example, 1 to 5 substituents, 1 to 3 substituents, or 1 substituent, with suitable substituents being defined below.
“C 1-6 Alkylamino "means a radical-NHR or-NR 2 Wherein R is substituted or unsubstituted C 1-6 An alkyl group. In some embodiments, C 1-4 Alkylamino is particularly preferred. Specific said alkylamino groups include, but are not limited to: methylamino, ethylamino, n-propylamino, isopropylamino, n-butylamino, dimethylamino, methylethylamino and diethylamino. Each of the alkylamino groups is independently optionally substituted, e.g., 1 to 5 substituents, 1 to 3 substituents, or 1 substituent, with suitable substituents being defined below, whether or not the alkylamino group is modified "substituted" before.
"halo" or "halogen" refers to fluoro (F), chloro (Cl), bromo (Br), and iodo (I). In some embodiments, the halogen group is F, cl or Br. In some embodiments, the halogen group is F or Cl. In some embodiments, the halogen group is F.
Thus, "C 1-6 Haloalkyl "and" C 1-6 "haloalkoxy" means toThe above-mentioned "C 1-6 Alkyl "and" C 1-6 Alkoxy ", which is substituted with one or more halo groups. In some embodiments, C 1-4 Haloalkyl is particularly preferred, more preferably C 1-2 A haloalkyl group. In some embodiments, C 1-4 Haloalkoxy is particularly preferred, more preferably C 1-2 A haloalkoxy group. Exemplary said haloalkyl groups include, but are not limited to: -CF 3 、-CH 2 F、-CHF 2 、-CHFCH 2 F、-CH 2 CHF 2 、-CF 2 CF 3 、-CCl 3 、-CH 2 Cl、-CHCl 2 2,2,2-trifluoro-1,1-dimethyl-ethyl, and the like. Exemplary said haloalkoxy groups include, but are not limited to: -OCH 2 F、-OCHF 2 、-OCF 3 And so on.
“C 3-10 Cycloalkyl "refers to a non-aromatic cyclic hydrocarbon group having 3 to 10 ring carbon atoms and zero heteroatoms. In some embodiments, C 3-7 Cycloalkyl is preferred, C 3-6 Cycloalkyl is particularly preferred, more preferably C 5-6 A cycloalkyl group. Cycloalkyl also includes ring systems in which the aforementioned cycloalkyl ring is fused to one or more aryl or heteroaryl groups, where the point of attachment is on the cycloalkyl ring, and in such cases the number of carbons continues to represent the number of carbons in the cycloalkyl system. Exemplary such cycloalkyl groups include, but are not limited to: cyclopropyl (C) 3 ) Cyclopropenyl group (C) 3 ) Cyclobutyl (C) 4 ) Cyclobutenyl radical (C) 4 ) Cyclopentyl (C) 5 ) Cyclopentenyl group (C) 5 ) Cyclohexyl (C) 6 ) Cyclohexenyl (C) 6 ) Cyclohexyldienyl (C) 6 ) Cycloheptyl (C) 7 ) Cycloheptenyl (C) 7 ) Cycloheptadienyl (C) 7 ) Cycloheptatrienyl (C) 7 ) Cyclooctyl (C) 8 ) Cyclooctenyl (C) 8 ) Bicyclo [2.2.1]Heptyl (C) 7 ) Bicyclo [2.2.2]Octyl radical (C) 8 ) Cyclononyl radical (C) 9 ) Cyclononenyl (C) 9 ) Cyclodecyl (C) 10 ) Cyclodecenyl (C) 10 ) octahydro-1H-indenyl (C) 9 ) Decahydronaphthyl (C) 10 ) Spiro [4.5 ]]Decyl (C) 10 ) And so on. Each of the cycloalkyl groups is independently optionally substituted, whether or not the cycloalkyl group is modified "substituted", for example, 1 to 5 substituents, 1 to 3 substituents, or 1 substituent, with appropriate substituents being defined below.
"3-to 10-membered heterocyclyl" is or refers to a group having a ring carbon atom and 1 to 4 ring heteroatoms in a 3-to 10-membered non-aromatic ring system, wherein each heteroatom is independently selected from nitrogen, oxygen, sulfur, boron, phosphorus, and silicon. In heterocyclic groups containing one or more nitrogen atoms, the point of attachment may be carbon or a nitrogen atom, as valency permits. In some embodiments, 3 to 7 membered heterocyclic groups are preferred, which are 3 to 7 membered non-aromatic ring systems having ring carbon atoms and 1 to 3 ring heteroatoms; in some embodiments, 3 to 6 membered heterocyclic groups are particularly preferred, which are 3 to 6 membered non-aromatic ring systems having ring carbon atoms and 1 to 3 ring heteroatoms; more preferably a5 to 6 membered heterocyclic group which is a5 to 6 membered non aromatic ring system having ring carbon atoms and 1 to 3 ring heteroatoms. Heterocyclyl also includes ring systems wherein the aforementioned heterocyclyl ring is fused to one or more cycloalkyl, aryl or heteroaryl groups, wherein the point of attachment is on the heterocyclyl ring; and in such cases the number of ring members continues to represent the number of ring members in the heterocyclyl ring system. Each of the heterocyclic groups is independently optionally substituted, e.g., 1 to 5 substituents, 1 to 3 substituents, or 1 substituent, with suitable substituents being defined below, whether or not the heterocyclic group is pre-modified with "substituted".
Exemplary 3-membered heterocyclic groups containing one heteroatom include, but are not limited to: aziridinyl, oxacyclopropaneyl, thienylyl. Exemplary 4-membered heterocyclic groups containing one heteroatom include, but are not limited to: azetidinyl, oxetanyl and thietanyl. Exemplary 5-membered heterocyclic groups containing one heteroatom include, but are not limited to: tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, dihydrothienyl, pyrrolidinyl, dihydropyrrolyl, and pyrrolyl-2,5-dione. Exemplary 5-membered heterocyclic groups containing two heteroatoms include, but are not limited to: dioxolanyl, oxathiolanyl (oxathiolanyl), dithiolanyl (disulphenyl) and oxathiolaneOxazolidin-2-one. Exemplary 5-membered heterocyclic groups containing three heteroatoms include, but are not limited to: triazolinyl, oxadiazolinyl and thiadiazolinyl. Exemplary 6-membered heterocyclic groups containing one heteroatom include, but are not limited to: piperidinyl, tetrahydropyranyl, dihydropyridinyl and thiacyclohexyl (thianyl). Exemplary 6-membered heterocyclic groups containing two heteroatoms include, but are not limited to: piperazinyl, morpholinyl, dithiacyclohexane, dioxanyl. Exemplary 6-membered heterocyclic groups containing three heteroatoms include, but are not limited to: hexahydrotriazinyl (triazinanyl). Exemplary 7-membered heterocyclic groups containing one heteroatom include, but are not limited to: azepane, oxepanyl and thiepane. Exemplary 8-membered heterocyclic groups containing one heteroatom include, but are not limited to: azacyclooctyl, oxocyclooctyl, and thiacyclooctyl. Exemplary with C 6 Aryl ring fused 5-membered heterocyclyl (also referred to herein as 5,6-bicyclic heterocyclyl) includes, but is not limited to: indolinyl, isoindolinyl, dihydrobenzofuranyl, dihydrobenzothienyl, benzoxazolonyl, and the like. Exemplary with C 6 Aryl ring fused 6-membered heterocyclyl (also referred to herein as 6,6-bicyclic heterocyclyl) includes, but is not limited to: tetrahydroquinolinyl, tetrahydroisoquinolinyl, and the like.
“C 6-14 Aryl "refers to a group having 6-14 ring carbon atoms and zero heteroatoms in a monocyclic or polycyclic (e.g., bicyclic or tricyclic) 4n +2 aromatic ring system (e.g., having 6, 10, or 14 pi electrons shared in a cyclic arrangement). In some embodiments, an aryl group has six ring carbon atoms ("C) 6 Aryl "; for example, phenyl). In some embodiments, an aryl group has ten ring carbon atoms ("C) 10 Aryl "; e.g., naphthyl, e.g., 1-naphthyl and 2-naphthyl). In some embodiments, an aryl group has fourteen ring carbon atoms ("C) 14 Aryl "; for example, an anthracene group). In some embodiments, C 6-10 Aryl is particularly preferred, more preferably C 6 And (4) an aryl group. Aryl also includes ring systems in which the above-mentioned aryl ring is fused to one or more cycloalkyl or heterocyclyl groups, and the point of attachment is on the aryl ring, in which caseNext, the number of carbon atoms continues to represent the number of carbon atoms in the aryl ring system. Each of the aryl groups is independently optionally substituted, whether or not the aryl group is preceded by a "substituted", e.g., 1 to 5 substituents, 1 to 3 substituents, or 1 substituent, with appropriate substituents being defined below.
"5-to 10-membered heteroaryl" refers to a group having a 5-10 membered monocyclic or bicyclic 4n +2 aromatic ring system (e.g., having 6 or 10 pi electrons shared in a cyclic arrangement) with ring carbon atoms and 1-4 ring heteroatoms, wherein each heteroatom is independently selected from nitrogen, oxygen, and sulfur. In heteroaryl groups containing one or more nitrogen atoms, the point of attachment may be a carbon or nitrogen atom, as valency permits. Heteroaryl bicyclic ring systems may include one or more heteroatoms in one or both rings. Heteroaryl also includes ring systems in which the aforementioned heteroaryl ring is fused to one or more cycloalkyl or heterocyclyl groups, and the point of attachment is on the heteroaryl ring, in which case the number of carbon atoms continues to represent the number of carbon atoms in the heteroaryl ring system. In some embodiments, 5 to 6 membered heteroaryl is particularly preferred, which is a 5-6 membered monocyclic or bicyclic 4n +2 aromatic ring system having ring carbon atoms and 1-4 ring heteroatoms. Each of the heteroaryl groups is independently optionally substituted, e.g., 1 to 5 substituents, 1 to 3 substituents, or 1 substituent, with suitable substituents being defined below, whether or not the heteroaryl group is modified "substituted" before.
Exemplary 5-membered heteroaryl groups containing one heteroatom include, but are not limited to: pyrrolyl, furanyl and thienyl. Exemplary 5-membered heteroaryl groups containing two heteroatoms include, but are not limited to: imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl and isothiazolyl. Exemplary 5-membered heteroaryl groups containing three heteroatoms include, but are not limited to: triazolyl, oxadiazolyl and thiadiazolyl. Exemplary 5-membered heteroaryl groups containing four heteroatoms include, but are not limited to: a tetrazolyl group. Exemplary 6-membered heteroaryl groups containing one heteroatom include, but are not limited to: a pyridyl group. Exemplary 6-membered heteroaryl groups containing two heteroatoms include, but are not limited to: pyridazinyl, pyrimidinyl and pyrazinyl. Exemplary 6-membered heteroaryl groups containing three or four heteroatoms include, but are not limited to: triazinyl and tetrazinyl. Exemplary 7-membered heteroaryl groups containing one heteroatom include, but are not limited to: azepinyl, oxacycloheptyl, and thiacycloheptyl trienyl groups. Exemplary 5,6-bicyclic heteroaryls include, but are not limited to: indolyl, isoindolyl, indazolyl, benzotriazolyl, benzothienyl, isobenzothienyl, benzofuranyl, benzisothiafuranyl, benzimidazolyl, benzoxazolyl, benzisoxazolyl, benzooxadiazolyl, benzothiazolyl, benzisothiazolyl, benzothiadiazolyl, indezinyl, and purinyl. Exemplary 6,6-bicyclic heteroaryls include, but are not limited to: naphthyridinyl, pteridinyl, quinolinyl, isoquinolinyl, cinnolinyl, quinoxalinyl, phthalazinyl and quinazolinyl.
"carbonyl" means a-C (O) -group.
Exemplary substituents on carbon atoms include, but are not limited to: halogen, -CN, -NO 2 、-N 3 、-SO 2 H、-SO 3 H、-OH、-OR aa 、-ON(R bb ) 2 、-N(R bb ) 2 、-N(R bb ) 3 + X - 、-N(OR cc )R bb 、-SH、-SR aa 、-SSR cc 、-C(=O)R aa 、-CO 2 H、-CHO、-C(OR cc ) 2 、-CO 2 R aa 、-OC(=O)R aa 、-OCO 2 R aa 、-C(=O)N(R bb ) 2 、-OC(=O)N(R bb ) 2 、-NR bb C(=O)R aa 、-NR bb CO 2 R aa 、-NR bb C(=O)N(R bb ) 2 、-C(=NR bb )R aa 、-C(=NR bb )OR aa 、-OC(=NR bb )R aa 、-OC(=NR bb )OR aa 、-C(=NR bb )N(R bb ) 2 、-OC(=NR bb )N(R bb ) 2 、-NR bb C(=NR bb )N(R bb ) 2 、-C(=O)NR bb SO 2 R aa 、-NR bb SO 2 R aa 、-SO 2 N(R bb ) 2 、-SO 2 R aa 、-SO 2 OR aa 、-OSO 2 R aa 、-S(=O)R aa 、-OS(=O)R aa 、-Si(R aa ) 3 、-OSi(R aa ) 3 、-C(=S)N(R bb ) 2 、-C(=O)SR aa 、-C(=S)SR aa 、-SC(=S)SR aa 、-SC(=O)SR aa 、-OC(=O)SR aa 、-SC(=O)OR aa 、-SC(=O)R aa 、-P(=O) 2 R aa 、-OP(=O) 2 R aa 、-P(=O)(R aa ) 2 、-OP(=O)(R aa ) 2 、-OP(=O)(OR cc ) 2 、-P(=O) 2 N(R bb ) 2 、-OP(=O) 2 N(R bb ) 2 、-P(=O)(NR bb ) 2 、-OP(=O)(NR bb ) 2 、-NR bb P(=O)(OR cc ) 2 、-NR bb P(=O)(NR bb ) 2 、-P(R cc ) 2 、-P(R cc ) 3 、-OP(R cc ) 2 、-OP(R cc ) 3 、-B(R aa ) 2 、-B(OR cc ) 2 、-BR aa (OR cc ) Alkyl, haloalkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl is independently substituted with 0, 1,2, 3,4, or 5R dd Substituted by groups;
or two geminal hydrogens on a carbon atom are replaced by a group = O, = S, = NN (R) bb ) 2 、=NNR bb C(=O)R aa 、=NNR bb C(=O)OR aa 、=NNR bb S(=O) 2 R aa 、=NR bb Or = NOR cc Substitution;
R aa each is independently selected from alkyl, haloalkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl, or two R aa The groups combine to form a heterocyclyl or heteroaryl ring in which each alkyl, alkenyl, alkynyl, carbocyclic ringIndependently by 0, 1,2, 3,4 or 5R dd Substituted by groups;
R bb each independently selected from: hydrogen, -OH, -OR aa 、-N(R cc ) 2 、-CN、-C(=O)R aa 、-C(=O)N(R cc ) 2 、-CO 2 R aa 、-SO 2 R aa 、-C(=NR cc )OR aa 、-C(=NR cc )N(R cc ) 2 、-SO 2 N(R cc ) 2 、-SO 2 R cc 、-SO 2 OR cc 、-SOR aa 、-C(=S)N(R cc ) 2 、-C(=O)SR cc 、-C(=S)SR cc 、-P(=O) 2 R aa 、-P(=O)(R aa ) 2 、-P(=O) 2 N(R cc ) 2 、-P(=O)(NR cc ) 2 Alkyl, haloalkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl, or two R bb The groups combine to form a heterocyclyl or heteroaryl ring, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl is independently substituted with 0, 1,2, 3,4, or 5R dd Substituted by groups;
R cc each is independently selected from the group consisting of hydrogen, alkyl, haloalkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl, or two R cc The groups combine to form a heterocyclyl or heteroaryl ring wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl is independently substituted with 0, 1,2, 3,4 or 5R dd Substituted by groups;
R dd each independently selected from: halogen, -CN, -NO 2 、-N 3 、-SO 2 H、-SO 3 H、-OH、-OR ee 、-ON(R ff ) 2 、-N(R ff ) 2 ,、-N(R ff ) 3 + X - 、-N(OR ee )R ff 、-SH、-SR ee 、-SSR ee 、-C(=O)R ee 、-CO 2 H、-CO 2 R ee 、-OC(=O)R ee 、-OCO 2 R ee 、-C(=O)N(R ff ) 2 、-OC(=O)N(R ff ) 2 、-NR ff C(=O)R ee 、-NR ff CO 2 R ee 、-NR ff C(=O)N(R ff ) 2 、-C(=NR ff )OR ee 、-OC(=NR ff )R ee 、-OC(=NR ff )OR ee 、-C(=NR ff )N(R ff ) 2 、-OC(=NR ff )N(R ff ) 2 、-NR ff C(=NR ff )N(R ff ) 2 、-NR ff SO 2 R ee 、-SO 2 N(R ff ) 2 、-SO 2 R ee 、-SO 2 OR ee 、-OSO 2 R ee 、-S(=O)R ee 、-Si(R ee ) 3 、-OSi(R ee ) 3 、-C(=S)N(R ff ) 2 、-C(=O)SR ee 、-C(=S)SR ee 、-SC(=S)SR ee 、-P(=O) 2 R ee 、-P(=O)(R ee ) 2 、-OP(=O)(R ee ) 2 、-OP(=O)(OR ee ) 2 Alkyl, haloalkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, heteroaryl, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl is independently substituted with 0, 1,2, 3,4 or 5R gg Substituted by radicals, or two geminal R dd Substituents may combine to form = O or = S;
R ee is independently selected from the group consisting of alkyl, haloalkyl, alkenyl, alkynyl, carbocyclyl, aryl, heterocyclyl and heteroaryl, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl is independently substituted with 0, 1,2, 3,4 or 5R gg Substitution of radicals;
R ff each independently selected from hydrogen, alkyl, haloalkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl, or two R ff The groups combine to form a heterocyclyl or heteroaryl ring wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclylAryl and heteroaryl are independently substituted with 0, 1,2, 3,4 or 5R gg Substituted by groups;
R gg is independently of each other: halogen, -CN, -NO 2 、-N 3 、-SO 2 H、-SO 3 H、-OH、-OC 1-6 Alkyl, -ON (C) 1-6 Alkyl radical) 2 、-N(C 1-6 Alkyl radical) 2 、-N(C 1-6 Alkyl radical) 3 + X - 、-NH(C 1-6 Alkyl radical) 2 + X - 、-NH 2 (C 1-6 Alkyl radical) + X - 、-NH 3 + X - 、-N(OC 1-6 Alkyl) (C 1-6 Alkyl), -N (OH) (C) 1-6 Alkyl), -NH (OH), -SH, -SC 1-6 Alkyl, -SS (C) 1-6 Alkyl), -C (= O) (C) 1-6 Alkyl), -CO 2 H、-CO 2 (C 1-6 Alkyl), -OC (= O) (C) 1-6 Alkyl), -OCO 2 (C 1-6 Alkyl), -C (= O) NH 2 、-C(=O)N(C 1-6 Alkyl radical) 2 、-OC(=O)NH(C 1-6 Alkyl), -NHC (= O) (C) 1-6 Alkyl), -N (C) 1-6 Alkyl) C (= O) (C 1-6 Alkyl), -NHCO 2 (C 1-6 Alkyl), -NHC (= O) N (C) 1-6 Alkyl radical) 2 、-NHC(=O)NH(C 1-6 Alkyl), -NHC (= O) NH 2 、-C(=NH)O(C 1-6 Alkyl), -OC (= NH) (C) 1-6 Alkyl), -OC (= NH) OC 1-6 Alkyl, -C (= NH) N (C) 1-6 Alkyl radical) 2 、-C(=NH)NH(C 1-6 Alkyl), -C (= NH) NH 2 、-OC(=NH)N(C 1-6 Alkyl radical) 2 、-OC(NH)NH(C 1-6 Alkyl), -OC (NH) NH 2 、-NHC(NH)N(C 1-6 Alkyl radical) 2 、-NHC(=NH)NH 2 、-NHSO 2 (C 1-6 Alkyl), -SO 2 N(C 1-6 Alkyl radical) 2 、-SO 2 NH(C 1-6 Alkyl), -SO 2 NH 2 、-SO 2 C 1-6 Alkyl, -SO 2 OC 1-6 Alkyl, -OSO 2 C 1-6 Alkyl, -SOC 1-6 Alkyl, -Si (C) 1-6 Alkyl radical) 3 、-OSi(C 1-6 Alkyl radical) 3 、-C(=S)N(C 1-6 Alkyl radical) 2 、C(=S)NH(C 1-6 Alkyl), C (= S) NH 2 、-C(=O)S(C 1-6 Alkyl), -C (= S) SC 1-6 Alkyl, -SC (= S) SC 1-6 Alkyl, -P (= O) 2 (C 1-6 Alkyl), -P (= O) (C) 1-6 Alkyl radical) 2 、-OP(=O)(C 1-6 Alkyl radical) 2 、-OP(=O)(OC 1-6 Alkyl radical) 2 、C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2 -C 6 Alkenyl radical, C 2 -C 6 Alkynyl, C 3 -C 7 Carbocyclyl, C 6 -C 10 Aryl radical, C 3 -C 7 Heterocyclic group, C 5 -C 10 A heteroaryl group; or two geminal R gg Substituents may combine to form = O or = S; wherein, X - Are counter ions.
Exemplary substituents on the nitrogen atom include, but are not limited to: hydrogen, -OH, -OR aa 、-N(R cc ) 2 、-CN、-C(=O)R aa 、-C(=O)N(R cc ) 2 、-CO 2 R aa 、-SO 2 R aa 、-C(=NR bb )R aa 、-C(=NR cc )OR aa 、-C(=NR cc )N(R cc ) 2 、-SO 2 N(R cc ) 2 、-SO 2 R cc 、-SO 2 OR cc 、-SOR aa 、-C(=S)N(R cc ) 2 、-C(=O)SR cc 、-C(=S)SR cc 、-P(=O) 2 R aa 、-P(=O)(R aa ) 2 、-P(=O) 2 N(R cc ) 2 、-P(=O)(NR cc ) 2 Alkyl, haloalkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl, or two R's attached to a nitrogen atom cc The groups combine to form a heterocyclyl or heteroaryl ring wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl is independently substituted with 0, 1,2, 3,4 or 5R dd Is substituted by radicals, and wherein R aa 、R bb 、R cc And R dd As described aboveThe following steps are carried out.
"deuterated" or "D" means that one or more hydrogens of the compound or group are replaced with deuterium; deuterium can be mono-, di-, poly-, or fully substituted. The terms "deuterated one or more" and "deuterated one or more" are used interchangeably.
"non-deuterated compound" refers to a compound containing deuterium at a ratio of deuterium atoms no greater than the natural isotopic content of deuterium (0.015%).
The deuterium isotope content of deuterium at the deuterated position is at least 0.015% greater than the natural deuterium isotope content, preferably greater than 30%, more preferably greater than 50%, more preferably greater than 75%, more preferably greater than 95%, more preferably greater than 99%.
The term "pharmaceutically acceptable salts" refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without excessive toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, the pharmaceutically acceptable salts described in detail in J.pharmaceutical Sciences (1977) 66. Pharmaceutically acceptable salts of the compounds of the present invention include salts derived from suitable inorganic and organic acids and bases. Examples of pharmaceutically acceptable non-toxic acid addition salts are salts with inorganic acids, such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid, or with organic acids, such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid. Salts formed using methods conventional in the art, e.g., ion exchange methods, are also included. Other pharmaceutically acceptable salts include: adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cypionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, gluconate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, laurylSulfates, malates, maleates, malonates, methanesulfonates, 2-naphthalenesulfonates, nicotinates, nitrates, oleates, oxalates, palmitates, pamoate, pectinates, persulfates, 3-phenylpropionates, phosphates, picrates, pivalates, propionates, stearates, succinates, sulfates, tartrates, thiocyanates, p-toluenesulfonate, undecanoates, valerates, and the like. Pharmaceutically acceptable salts derived from suitable bases include alkali metals, alkaline earth metals, ammonium and N + (C 1-4 Alkyl radical) 4 And (3) salt. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium salts, and the like. Other pharmaceutically acceptable salts include, if appropriate, non-toxic ammonium, quaternary ammonium and amine cations formed with counterions such as halide, hydroxide, formate, sulfate, phosphate, nitrate, lower alkyl sulfonate and aryl sulfonate.
The "subject" to which the drug is administered includes, but is not limited to: a human (i.e., a male or female of any age group, e.g., a pediatric subject (e.g., an infant, a child, an adolescent) or an adult subject (e.g., a young adult, a middle-aged adult, or an older adult)) and/or a non-human animal, e.g., a mammal, e.g., a primate (e.g., a cynomolgus monkey, a rhesus monkey), a cow, a pig, a horse, a sheep, a goat, a rodent, a cat, and/or a dog. In some embodiments, the subject is a human. In some embodiments, the subject is a non-human animal. The terms "human", "patient" and "subject" are used interchangeably herein.
"disease," "disorder," and "condition" are used interchangeably herein.
As used herein, unless otherwise specified, the term "treatment" includes effects that occur when a subject suffers from a particular disease, disorder or condition, which reduces the severity of the disease, disorder or condition, or delays or slows the progression of the disease, disorder or condition ("therapeutic treatment"), and also includes effects that occur before the subject begins to suffer from the particular disease, disorder or condition ("prophylactic treatment").
"combination" and related terms refer to the simultaneous or sequential administration of the therapeutic agents of the present invention. For example, a compound of the invention may be administered simultaneously or sequentially with another therapeutic agent in separate unit dosage forms, or simultaneously with another therapeutic agent in a single unit dosage form.
Detailed Description
Compound (I)
As used herein, "compounds of the invention" refers to compounds of formula (I) below (including a subset of each formula), or a pharmaceutically acceptable salt, hydrate, or solvate thereof.
In one embodiment, the present invention relates to a compound of formula (I), or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof:
Figure BDA0002983446160000131
wherein the content of the first and second substances,
ring A is an aromatic ring;
ring C is C 6-10 Aryl or 5 to 10 membered heteroaryl;
A 1 is CR A1 Or N;
A 2 is C or N;
A 4 is CR A4 Or N;
A 5 is C or N;
wherein R is A1 And R A4 Each independently selected from H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Alkoxy, and optionally substituted with one or more R ";
B 1 is CR 1 Or N;
B 2 is CR 2 Or N;
B 3 is CR 3 Or N;
B 4 is CR 4 Or N;
wherein R is 1 、R 2 、R 3 And R 4 Each independently selected from H, D, halogen, -CN, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, -C (O) R a 、-C(O)OR a 、-C(O)NR b R c 、-NR b R c 、-NR a C(O)R b 、-NR a C(O)OR b 、-NR a C(O)NR b R c 、-OR a 、-OC(O)R a 、-OC(O)OR a 、-OC(O)NR b R c 、C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5 to 10 membered heteroaryl, and optionally substituted with one or more R ";
l is O, S or NR L
Wherein R is L Is selected from H or C 1-6 Alkyl, and optionally substituted with one or more R;
v is (CR) V1 R V2 ) o
Wherein R is V1 And R V2 Each independently selected from H, D, halogen or C 1-6 Alkyl, and optionally substituted with one or more R;
o =1, 2,3, 4,5 or 6;
R 6 is H, D, halogen, -CN, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5 to 10 membered heteroaryl, optionally substituted with one or more R;
R 5 and R 7 Each independently selected from H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Haloalkyl, and the aforementioned groups are optionally substituted with one or more R; or, R 5 And R 7 Together with the double bond to which they are attached form a triple bond;
each R is independently selected from H, D, halogen, -CN, = O, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, -C (O) R a 、-C(O)OR a 、-C(O)NR b R c 、-NR b R c 、-NR a C(O)R b 、-NR a C(O)OR b 、-NR a C(O)NR b R c 、-OR a 、-OC(O)R a 、-OC(O)OR a 、-OC(O)NR b R c 、C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5 to 10 membered heteroaryl; alternatively, two R groups on the same atom or on adjacent atoms may together form C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5 to 10 membered heteroaryl; wherein each group in the definition of R is optionally substituted with one or more D, up to complete deuteration;
m =0, 1,2, 3,4, 5,6, 7, 8 or 9;
r' is independently selected from H, D, halogen, -CN, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, -C (O) R a 、-C(O)OR a 、-C(O)NR b R c 、-NR b R c 、-NR a C(O)R b 、-NR a C(O)OR b 、-NR a C(O)NR b R c 、-OR a 、-OC(O)R a 、-OC(O)OR a 、-OC(O)NR b R c 、C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5 to 10 membered heteroaryl, and optionally substituted with one or more R ";
n =0, 1,2, 3 or 4;
p =0, 1 or 2;
r' are each independently selected from H, D, halogen, -CN, = O, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, -C (O) R a 、-C(O)OR a 、-C(O)NR b R c 、-NR b R c 、-NR a C(O)R b 、-NR a C(O)OR b 、-NR a C(O)NR b R c 、-OR a 、-OC(O)R a 、-OC(O)OR a 、-OC(O)NR b R c 、C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5-to 10-membered heteroaryl, or two R' groups on the same atom or on adjacent atoms may together form C 3-7 Cycloalkyl, 3 to7-membered heterocyclic group, C 6-10 Aryl or 5 to 10 membered heteroaryl; wherein each group in the definition of R "is optionally substituted with one or more D, up to complete deuteration;
r is independently selected from H, D, halogen, -CN, = O, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, -C (O) R a 、-C(O)OR a 、-C(O)NR b R c 、-NR b R c 、-NR a C(O)R b 、-NR a C(O)OR b 、-NR a C(O)NR b R c 、-OR a 、-OC(O)R a 、-OC(O)OR a 、-OC(O)NR b R c 、C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5 to 10 membered heteroaryl; alternatively, two R groups on the same atom or on adjacent atoms may together form C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5 to 10 membered heteroaryl; wherein each group in the definition of R is optionally substituted with one or more D, up to complete deuteration;
R a 、R b and R c Each independently selected from H, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5-to 10-membered heteroaryl, or R b And R c Together with the N atom to which they are attached form a 3-to 7-membered heterocyclyl or 5-to 10-membered heteroaryl; wherein R is a 、R b And R c Each group in the definition is optionally substituted with one or more D, up to complete deuteration.
Ring C
In one embodiment, ring C is C 6-10 An aryl group; in another embodiment, ring C is 5 to 10 membered heteroaryl; in another embodiment, ring C is phenyl or 5 to 6 membered heteroaryl; in another embodiment, ring C is phenyl or a5 to 6 membered heteroaryl group containing 1-3 heteroatoms N, O or S; in another embodiment, ring C is phenyl, pyridyl, pyrimidinyl, pyridazinyl, pyrazinyl, pyrrolyl, pyrazolyl, or a pharmaceutically acceptable salt thereofImidazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, furyl or thienyl; in another embodiment, ring C is phenyl or pyridyl; in another embodiment, ring C is pyridyl.
A 1 、A 2 、A 4 And A 5
In one embodiment, A is 1 Is CR A1 (ii) a In another embodiment, A 1 Is CH; in another embodiment, A 1 Is N.
In one embodiment, A is 2 Is C; in another embodiment, A 2 Is N.
In one embodiment, A is 4 Is CR A4 (ii) a In another embodiment, A 4 Is CH; in another embodiment, A 4 Is N.
In one embodiment, A is 5 Is C; in another embodiment, A 5 Is N.
In one embodiment of the process of the present invention,
Figure BDA0002983446160000151
selected from the following structures:
Figure BDA0002983446160000152
preferably, the first and second electrodes are formed of a metal,
Figure BDA0002983446160000161
selected from the following structures:
Figure BDA0002983446160000162
preferably, the first and second electrodes are formed of a metal,
Figure BDA0002983446160000163
selected from the following structures:
Figure BDA0002983446160000164
B 1 、B 2 、B 3 and B 4
In one embodiment, B 1 Is CR 1 (ii) a In another embodiment, B 1 Is N.
In one embodiment, B 2 Is CR 2 (ii) a In another embodiment, B 2 Is N.
In one embodiment, B 3 Is CR 3 (ii) a In another embodiment, B 3 Is N.
In one embodiment, B 4 Is CR 4 (ii) a In another embodiment, B 4 Is N.
R 1 、R 2 、R 3 And R 4
In one embodiment, R 1 、R 2 、R 3 And R 4 Independently is H; in another embodiment, R 1 、R 2 、R 3 And R 4 Independently is D; in another embodiment, R 1 、R 2 、R 3 And R 4 Independently is halogen; in another embodiment, R 1 、R 2 、R 3 And R 4 Independently is-CN; in another embodiment, R 1 、R 2 、R 3 And R 4 Independently is C 1-6 An alkyl group; in another embodiment, R 1 、R 2 、R 3 And R 4 Independently is C 1-6 A haloalkyl group; in another embodiment, R 1 、R 2 、R 3 And R 4 Independently is C 2-6 An alkenyl group; in another embodiment, R 1 、R 2 、R 3 And R 4 Independently is C 2-6 Alkynyl; in another embodiment, R 1 、R 2 、R 3 And R 4 Independently is-C (O) R a (ii) a In another embodiment, R 1 、R 2 、R 3 And R 4 Independently is-C (O) OR a (ii) a In another embodiment, R 1 、R 2 、R 3 And R 4 Independently is-C (O) NR b R c (ii) a In another embodiment, R 1 、R 2 、R 3 And R 4 Independently is-NR b R c (ii) a In another embodiment, R 1 、R 2 、R 3 And R 4 Independently is-NR a C(O)R b (ii) a In another embodiment, R 1 、R 2 、R 3 And R 4 Independently is-NR a C(O)OR b (ii) a In another embodiment, R 1 、R 2 、R 3 And R 4 Independently is-NR a C(O)NR b R c (ii) a In another embodiment, R 1 、R 2 、R 3 And R 4 Independently is-OR a (ii) a In another embodiment, R 1 、R 2 、R 3 And R 4 Independently is-OC (O) R a (ii) a In another embodiment, R 1 、R 2 、R 3 And R 4 independently-OC (O) OR a (ii) a In another embodiment, R 1 、R 2 、R 3 And R 4 Independently is-OC (O) NR b R c (ii) a In another embodiment, R 1 、R 2 、R 3 And R 4 Independently is C 3-7 A cycloalkyl group; in another embodiment, R 1 、R 2 、R 3 And R 4 Independently is a 3-to 7-membered heterocyclyl; in another embodiment, R 1 、R 2 、R 3 And R 4 Independently is C 6-10 An aryl group; in another embodiment, R 1 、R 2 、R 3 And R 4 Independently a 5-to 10-membered heteroaryl group.
In another embodiment, R 1 、R 2 、R 3 And R 4 Independently selected from H, D, halogen, -CN, C 1-6 Alkyl radical, C 1-6 Alkoxy or C 3-7 A cycloalkyl group; in another embodimentIn, R 1 、R 2 、R 3 And R 4 Independently selected from H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 An alkoxy group; in another embodiment, R 1 、R 2 、R 3 And R 4 Independently selected from H, D, halogen or C 1-6 An alkyl group; in another embodiment, R 1 、R 2 、R 3 And R 4 Independently selected from H, D, F, cl or methyl; in another embodiment, R 1 、R 2 、R 3 And R 4 Independently selected from H or Cl. In another embodiment, R 2 Is halogen, preferably Cl.
In another embodiment, R 1 、R 2 、R 3 And R 4 Independently by one or more R ", for example by 1,2, 3,4, 5,6, 7, 8, 9, 10 or more R".
L
In one embodiment, L is O; in another embodiment, L is S; in another embodiment, L is NR L (ii) a In another embodiment, L is NH.
V
In one embodiment, V is (CR) V1 R V2 ) o (ii) a In another embodiment, V is (CR) V1 R V2 ) 1 、(CR V1 R V2 ) 2 、(CR V1 R V2 ) 3 、(CR V1 R V2 ) 4 、(CR V1 R V2 ) 5 Or (CR) V1 R V2 ) 6 (ii) a In another embodiment, R V1 And R V2 Each independently H, D, halogen or C 1-6 An alkyl group; in another embodiment, R V1 And R V2 Are all H. In another embodiment, V is (CH) 2 ) o (ii) a Preferably CH 2
R 6
In one embodiment, R 6 Is H; in another embodiment, R 6 Is D; in another embodiment, R 6 Is halogen; in another embodiment, R 6 is-CN; in another embodiment, R 6 Is C 1-6 An alkyl group; in another embodiment, R 6 Is C 1-6 A haloalkyl group; in another embodiment, R 6 Is C 3-7 A cycloalkyl group; in another embodiment, R 6 Is a3 to 7 membered heterocyclyl; in another embodiment, R 6 Is C 6-10 An aryl group; in another embodiment, R 6 Is a5 to 10 membered heteroaryl.
In another embodiment, R 6 Substituted with one or more R, for example with 1,2, 3,4, 5,6, 7, 8, 9, 10 or more R.
In another embodiment, R 6 Selected from H, halogen, -CN, C 1-6 Alkyl radical, C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5 to 10 membered heteroaryl; in another embodiment, R 6 Selected from H, halogen, -CN or C 1-6 An alkyl group; in another embodiment, R 6 Selected from H, F, cl, br, -CN, methyl, ethyl, isopropyl, -CH 2 N(CH 3 ) 2 、-CH 2 N(CH 3 )(CH 2 CH 3 )、-CH 2 N(CH 3 )(CH(CH 3 ) 2 )、-CH 2 N(CH 2 CH 3 ) 2 Or
Figure BDA0002983446160000181
In another embodiment, R 6 Selected from H, F, cl, methyl or-CH 2 N(CH 3 ) 2 (ii) a In another embodiment, R 6 Selected from H, methyl or-CH 2 N(CH 3 ) 2 (ii) a In another embodiment, R 6 Is H.
R 5 And R 7
In one embodiment, R 5 And R 7 Each independently is H; in another embodiment, R 5 And R 7 Each is independently D; in another embodiment, R 5 And R 7 Each independently is halogen; in another embodiment, R 5 And R 7 Each independently is-CN; in another embodiment, R 5 And R 7 Each independently is C 1-6 An alkyl group; in another embodiment, R 5 And R 7 Each independently is C 1-6 A haloalkyl group; in another embodiment, R 5 And R 7 Together with the double bond to which they are attached, form a triple bond.
In another embodiment, R 5 And R 7 Each independently selected from H, halogen, -CN or C 1-6 An alkyl group; in another embodiment, R 5 And R 7 Each independently selected from H, halogen or CN; in another embodiment, R 5 And R 7 Each independently selected from H, F, cl or CN; in another embodiment, R 5 And R 7 Each independently selected from H or CN; in another embodiment, R 5 And R 7 Is H.
R and m
In one embodiment, R is H; in another embodiment, R is D; in another embodiment, R is halogen; in another embodiment, R is-CN; in another embodiment, R is = O; in another embodiment, R is C 1-6 An alkyl group; in another embodiment, R is C 1-6 A haloalkyl group; in another embodiment, R is C 2-6 An alkenyl group; in another embodiment, R is C 2-6 An alkynyl group; in another embodiment, R is-C (O) R a (ii) a In another embodiment, R is-C (O) OR a (ii) a In another embodiment, R is-C (O) NR b R c (ii) a In another embodiment, R is-NR b R c (ii) a In another embodiment, R is-NH 2 (ii) a In another embodiment, R is-NR a C(O)R b (ii) a In another embodiment, R is-NR a C(O)OR b (ii) a In another embodiment, R is-NR a C(O)NR b R c (ii) a In another embodiment, R is-OR a (ii) a In another embodimentIn embodiments, R is-OH; in another embodiment, R is-OC (O) R a (ii) a In another embodiment, R is-OC (O) OR a (ii) a In another embodiment, R is-OC (O) NR b R c (ii) a In another embodiment, R is C 3-7 A cycloalkyl group; in another embodiment, R is a 3-to 7-membered heterocyclyl; in another embodiment, R is C 6-10 An aryl group; in another embodiment, R is 5 to 10 membered heteroaryl; in another embodiment, two R groups on the same atom or on adjacent atoms may together form C 3-7 A cycloalkyl group; in another embodiment, two R groups on the same atom or on adjacent atoms may together form a3 to 7 membered heterocyclyl; in another embodiment, two R groups on the same atom or on adjacent atoms may together form C 6-10 An aryl group; in another embodiment, two R groups on the same atom or on adjacent atoms may together form a5 to 10 membered heteroaryl; wherein each group in the definition of R is optionally substituted with one or more D, up to complete deuteration.
In one embodiment, m =0; in another embodiment, m =1; in another embodiment, m =2; in another embodiment, m =3; in another embodiment, m =4; in another embodiment, m =5; in another embodiment, m =6; in another embodiment, m =7; in another embodiment, m =8; in another embodiment, m =9.
p
In one embodiment, p =0; in another embodiment, p =1; in another embodiment, p =2.
In a further embodiment of the process of the present invention,
Figure BDA0002983446160000191
selected from the following structures:
Figure BDA0002983446160000192
preferably, the following components are used:
Figure BDA0002983446160000193
r' and n
In one embodiment, R' is H; in another embodiment, R' is D; in another embodiment, R' is halogen; in another embodiment, R' is — CN; in another embodiment, R' is C 1-6 An alkyl group; in another embodiment, R' is C 1-6 A haloalkyl group; in another embodiment, R' is C 2-6 An alkenyl group; in another embodiment, R' is C 2-6 Alkynyl; in another embodiment, R' is-C (O) R a (ii) a In another embodiment, R' is-C (O) OR a (ii) a In another embodiment, R' is-C (O) NR b R c (ii) a In another embodiment, R' is-NR b R c (ii) a In another embodiment, R' is-NR a C(O)R b (ii) a In another embodiment, R' is-NR a C(O)OR b (ii) a In another embodiment, R' is-NR a C(O)NR b R c (ii) a In another embodiment, R' is-OR a (ii) a In another embodiment, R' is-OC (O) R a (ii) a In another embodiment, R' is-OC (O) OR a (ii) a In another embodiment, R' is-OC (O) NR b R c (ii) a In another embodiment, R' is C 3-7 A cycloalkyl group; in another embodiment, R' is a3 to 7 membered heterocyclyl; in another embodiment, R' is C 6-10 An aryl group; in another embodiment, R' is a5 to 10 membered heteroaryl.
In another embodiment, R' is substituted with one or more R ", for example with 1,2, 3,4, 5,6, 7, 8, 9, 10 or more R".
In one embodiment, n =0; in another embodiment, n =1; in another embodiment, n =2; in another embodiment, n =3; in another embodiment, n =4.
R”
In one embodiment, R "is H; in another embodiment, R "is D; in another embodiment, R "is halogen; in another embodiment, R "is-CN; in another embodiment, R "is = O; in another embodiment, R' is C 1-6 An alkyl group; in another embodiment, R' is C 1-6 A haloalkyl group; in another embodiment, R' is C 2-6 An alkenyl group; in another embodiment, R' is C 2-6 An alkynyl group; in another embodiment, R' is-C (O) R a (ii) a In another embodiment, R' is-C (O) OR a (ii) a In another embodiment, R' is-C (O) NR b R c (ii) a In another embodiment, R' is-NR b R c (ii) a In another embodiment, R' is-NH 2 (ii) a In another embodiment, R' is-NR a C(O)R b (ii) a In another embodiment, R' is-NR a C(O)OR b (ii) a In another embodiment, R' is-NR a C(O)NR b R c (ii) a In another embodiment, R' is-OR a (ii) a In another embodiment, R "is-OH; in another embodiment, R' is-OC (O) R a (ii) a In another embodiment, R' is-OC (O) OR a (ii) a In another embodiment, R' is-OC (O) NR b R c (ii) a In another embodiment, R' is C 3-7 A cycloalkyl group; in another embodiment, R "is a3 to 7 membered heterocyclyl; in another embodiment, R' is C 6-10 An aryl group; in another embodiment, R "is a5 to 10 membered heteroaryl; in another embodiment, two R "groups on the same atom or on adjacent atoms may together form C 3-7 A cycloalkyl group; in another embodiment, two R "groups on the same atom or on adjacent atoms may together form a 3-to 7-membered heterocyclyl; in another embodiment, two R "groups on the same atom or on adjacent atoms may together form C 6-10 An aryl group; in another embodiment, the same atom or adjacent atomsThe two R "groups on (a) may together form a5 to 10 membered heteroaryl; wherein each group in the definition of R "is optionally substituted with one or more D, up to complete deuteration.
R*
In one embodiment, R is H; in another embodiment, R is D; in another embodiment, R is halogen; in another embodiment, R is-CN; in another embodiment, R is = O; in another embodiment, R is C 1-6 An alkyl group; in another embodiment, R is C 1-6 A haloalkyl group; in another embodiment, R is C 2-6 An alkenyl group; in another embodiment, R is C 2-6 An alkynyl group; in another embodiment, R is-C (O) R a (ii) a In another embodiment, R is-C (O) OR a (ii) a In another embodiment, R is-C (O) NR b R c (ii) a In another embodiment, R is-NR b R c (ii) a In another embodiment, R is-NH 2 (ii) a In another embodiment, R is-NR a C(O)R b (ii) a In another embodiment, R is-NR a C(O)OR b (ii) a In another embodiment, R is-NR a C(O)NR b R c (ii) a In another embodiment, R is-OR a (ii) a In another embodiment, R is-OH; in another embodiment, R is-OC (O) R a (ii) a In another embodiment, R is-OC (O) OR a (ii) a In another embodiment, R is-OC (O) NR b R c (ii) a In another embodiment, R is C 3-7 A cycloalkyl group; in another embodiment, R is a3 to 7 membered heterocyclyl; in another embodiment, R is C 6-10 An aryl group; in another embodiment, R is a5 to 10 membered heteroaryl; in another embodiment, two R groups on the same atom or on adjacent atoms may together form C 3-7 A cycloalkyl group; in another embodiment, two R groups on the same atom or on adjacent atoms may together form a3 to 7 membered heterocyclyl; in another embodiment, two R groups on the same atom or on adjacent atoms may together form C 6-10 An aryl group; in another embodiment, two R groups on the same atom or on adjacent atoms may together form a5 to 10 membered heteroaryl; wherein each group in the definition of R is optionally substituted with one or more D, up to complete deuteration.
R a 、R b And R c
In one embodiment, R a 、R b And R c Independently is H; in another embodiment, R a 、R b And R c Independently is C 1-6 An alkyl group; in one embodiment, R a 、R b And R c Independently is C 1-6 A haloalkyl group; in one embodiment, R a 、R b And R c Independently is C 3-7 A cycloalkyl group; in one embodiment, R a 、R b And R c Independently is a 3-to 7-membered heterocyclyl; in one embodiment, R a 、R b And R c Independently is C 6-10 An aryl group; in one embodiment, R a 、R b And R c Independently is a5 to 10 membered heteroaryl; wherein R is a 、R b And R c Each group in the definition is optionally substituted with one or more D, up to complete deuteration.
Any of the above embodiments, or any combination thereof, may be combined with any of the other embodiments, or any combination thereof. For example, ring C, A 1 、A 2 、A 4 、A 5 、B 1 、B 2 、B 3 、B 4 、L、V、R 5 、R 6 、R 7 Any technical scheme of R, m, R', n and p or any combination thereof. The present invention is intended to include all combinations of these solutions, limited to space, not listed one by one.
In a more specific embodiment, the present invention relates to a compound of formula (I) as described above, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein,
Figure BDA0002983446160000211
selected from the following structures:
Figure BDA0002983446160000212
preferably, the first and second electrodes are formed of a metal,
Figure BDA0002983446160000213
selected from the following structures:
Figure BDA0002983446160000214
preferably, the first and second electrodes are formed of a metal,
Figure BDA0002983446160000215
selected from the following structures:
Figure BDA0002983446160000221
in a more specific embodiment, the present invention relates to a compound of formula (I) as described above, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein ring C is phenyl or 5-to 6-membered heteroaryl; preferably phenyl, pyridyl, pyrimidinyl, pyridazinyl, pyrazinyl, pyrrolyl, pyrazolyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, furyl or thienyl; preferably phenyl or pyridyl.
In a more specific embodiment, the present invention relates to a compound of formula (I) as described above, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein V is (CH) 2 ) o (ii) a Preferably CH 2
In a more specific embodiment, the present invention relates to a compound of formula (I) as described above, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein L is O.
In a more particular embodiment, the present invention relates to a compound of formula (I) as described above, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein B is 1 Is CR 1 、B 2 Is CR 2 、B 3 Is CR 3 、B 4 Is CR 4 (ii) a Preferably, R 1 、R 2 、R 3 And R 4 Independently selected from H, D, halogen, -CN, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl or C 2-6 Alkynyl.
In a more specific embodiment, the present invention relates to a compound of formula (I) as described above, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein,
Figure BDA0002983446160000222
selected from the following structures:
Figure BDA0002983446160000223
preferably, the following components are used:
Figure BDA0002983446160000224
in a more specific embodiment, the present invention relates to a compound of formula (I) as described above, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, which is a compound of the general formula:
Figure BDA0002983446160000231
wherein each group is as defined above.
In a more specific embodiment, the present invention relates to a compound of formula (I), as described above, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, which is a compound of formula (II-1), (III-1), or (IV-1):
Figure BDA0002983446160000241
wherein, the first and the second end of the pipe are connected with each other,
ring A is an aromatic ring;
A 2 is C or N;
A 4 is CR A4 Or N;
A 5 is C or N;
wherein R is A4 Is H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Alkoxy, optionally substituted with one or more R ";
R 1 is H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Haloalkyl, and optionally substituted with one or more R ";
R 2 is H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Haloalkyl, and optionally substituted with one or more R ";
R 3 is H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Haloalkyl, and optionally substituted with one or more R ";
R 4 is H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Haloalkyl, and optionally substituted with one or more R ";
o =1, 2,3 or 4;
each R is independently selected from H, D, halogen, -CN, = O, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl or C 2-6 Alkynyl; wherein each group in the definition of R is optionally substitutedOne or more D substitutions, up to complete deuteration;
m =0, 1,2, 3,4 or 5;
p =0, 1 or 2;
r' is independently selected from H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Haloalkyl, and optionally substituted with one or more R ";
n =0, 1,2, 3 or 4;
R 5 、R 6 and R 7 Each independently selected from H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Haloalkyl, and the aforementioned groups are optionally substituted with one or more R; or, R 5 And R 7 Together with the double bond to which they are attached form a triple bond;
r is independently selected from H, D, halogen, -CN, = O, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl or C 2-6 Alkynyl; wherein each group in the definition of R is optionally substituted with one or more D up to complete deuteration;
each R' is independently selected from H, D, halogen, -CN, = O, -OR a or-NR b R c
R a 、R b And R c Each independently selected from H, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, or R b And R c Together with the N atom to which they are attached form a 3-to 7-membered heterocyclyl or 5-to 10-membered heteroaryl; wherein R is a 、R b And R c Each group in the definition is optionally substituted with one or more D, up to complete deuteration.
In a more specific embodiment, the present invention relates to a compound of formula (I), as described above, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, which is a compound of formula (II-2), (III-2), or (IV-2):
Figure BDA0002983446160000251
wherein the content of the first and second substances,
ring A is an aromatic ring;
A 2 is C or N;
A 4 is CR A4 Or N;
A 5 is C or N;
wherein R is A4 Is H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Alkoxy, optionally substituted with one or more R ";
R 1 is H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Haloalkyl, and optionally substituted with one or more R ";
R 2 is H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Haloalkyl, and optionally substituted with one or more R ";
o =1, 2,3 or 4;
each R is independently selected from H, D, halogen, -CN, = O, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl or C 2-6 An alkynyl group; wherein each group in the definition of R is optionally substituted with one or more D, up to complete deuteration;
m =0, 1,2, 3,4 or 5;
r' are each independently selected from H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Haloalkyl, and optionally substituted with one or more R ";
n =0, 1,2, 3 or 4;
each R' is independently selected from H, D, halogen, -CN, = O, -OR a or-NR b R c
R a 、R b And R c Each independently selected from H, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, or R b And R c Together with the N atom to which they are attached form a 3-to 7-membered heterocyclyl or 5-to 10-membered heteroaryl; wherein R is a 、R b And R c Each radical in the definition is optionally substituted by one or moreD substitution, until fully deuterated.
In a more specific embodiment, the present invention relates to a compound of formula (I), as described above, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, which is a compound of formula (V-1) or (VI-1):
Figure BDA0002983446160000261
wherein the content of the first and second substances,
A 1 is CR A1 Or N;
wherein R is A1 Is H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Alkoxy, optionally substituted with one or more R ";
R 1 is H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Haloalkyl, and optionally substituted with one or more R ";
R 2 is H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Haloalkyl, and optionally substituted with one or more R ";
R 3 is H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Haloalkyl, and optionally substituted with one or more R ";
R 4 is H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Haloalkyl, and optionally substituted with one or more R ";
o =1, 2,3 or 4;
each R is independently selected from H, D, halogen, -CN, = O, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl or C 2-6 An alkynyl group; wherein each group in the definition of R is optionally substituted with one or more D, up to complete deuteration;
m =0, 1,2, 3,4 or 5;
p =0, 1 or 2;
r' is independently selected from H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 HalogenatedAlkyl, and optionally substituted with one or more R ";
n =0, 1,2, 3 or 4;
R 5 、R 6 and R 7 Each independently selected from H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Haloalkyl, and the aforementioned groups are optionally substituted with one or more R; or, R 5 And R 7 Together with the double bond to which they are attached form a triple bond;
r is independently selected from H, D, halogen, -CN, = O, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl or C 2-6 Alkynyl; wherein each group in the definition of R is optionally substituted with one or more D, up to complete deuteration;
each R' is independently selected from H, D, halogen, -CN, = O, -OR a or-NR b R c
R a 、R b And R c Each independently selected from H, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, or R b And R c Together with the N atom to which they are attached form a 3-to 7-membered heterocyclyl or 5-to 10-membered heteroaryl; wherein R is a 、R b And R c Each group in the definition is optionally substituted with one or more D, up to complete deuteration.
In a more specific embodiment, the present invention relates to a compound of formula (I), as described above, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, which is a compound of formula (V-2) or (VI-2):
Figure BDA0002983446160000271
wherein the content of the first and second substances,
A 1 is CR A1 Or N;
wherein R is A1 Is H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Alkoxy optionally substituted by one or moreR' is substituted;
R 1 is H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Haloalkyl, and optionally substituted with one or more R ";
R 2 is H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Haloalkyl, and optionally substituted with one or more R ";
o =1, 2,3 or 4;
each R is independently selected from H, D, halogen, -CN, = O, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl or C 2-6 Alkynyl; wherein each group in the definition of R is optionally substituted with one or more D, up to complete deuteration;
m =0, 1,2, 3,4 or 5;
r' is independently selected from H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Haloalkyl, and optionally substituted with one or more R ";
n =0, 1,2, 3 or 4;
each R' is independently selected from H, D, halogen, -CN, = O, -OR a or-NR b R c
Each R a 、R b And R c Each independently selected from H, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, or R b And R c Together with the N atom to which they are attached form a 3-to 7-membered heterocyclyl or 5-to 10-membered heteroaryl; wherein R is a 、R b And R c Each group in the definition is optionally substituted with one or more D, up to complete deuteration.
In a more specific embodiment, the present invention relates to a compound of formula (I) as described above, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein the compound is selected from the group consisting of:
Figure BDA0002983446160000281
Figure BDA0002983446160000291
the compounds of the present invention may include one or more asymmetric centers, and thus may exist in a variety of stereoisomeric forms, e.g., enantiomeric and/or diastereomeric forms. For example, the compounds of the invention may be in the form of individual enantiomers, diastereomers, or geometric isomers (e.g., cis and trans isomers), or may be in the form of mixtures of stereoisomers, including racemic mixtures and mixtures enriched in one or more stereoisomers. Isomers may be separated from mixtures by methods known to those skilled in the art, including: chiral High Pressure Liquid Chromatography (HPLC) and the formation and crystallization of chiral salts; alternatively, preferred isomers may be prepared by asymmetric synthesis.
"tautomer" refers to a compound in which one functional group changes its structure to another functional isomer, and which rapidly interconverts into two isomers in dynamic equilibrium, the two isomers being referred to as tautomers.
One skilled in the art will appreciate that the organic compound may form a complex with the solvent in which it reacts or from which it precipitates or crystallizes. These complexes are referred to as "solvates". When the solvent is water, the complex is referred to as a "hydrate". The present invention encompasses all solvates of the compounds of the present invention.
The term "solvate" refers to a form of a compound or salt thereof that is combined with a solvent, typically formed by a solvolysis reaction. This physical association may include hydrogen bonding. Conventional solvents include water, methanol, ethanol, acetic acid, DMSO, THF, ether, and the like. The compounds described herein can be prepared, for example, in crystalline form, and can be solvated. Suitable solvates include pharmaceutically acceptable solvates and further include stoichiometric and non-stoichiometric solvates. In some cases, the solvate will be capable of isolation, for example, when one or more solvent molecules are incorporated into the crystal lattice of a crystalline solid. "solvate" includes solvates in solution and isolatable solvates. Representative solvates include hydrates, ethanolates, and methanolates.
The term "hydrate" refers to a compound that is associated with an aqueous phase. In general, the ratio of the number of water molecules contained in a hydrate of a compound to the number of molecules of the compound in the hydrate is determined. Thus, hydrates of the compounds can be used, for example, with the formula R. x H 2 O represents, wherein R is the compound, and x is a number greater than 0. A given compound may form more than one hydrate type, including, for example, monohydrate (x is 1), lower hydrates (x is a number greater than 0 and less than 1), e.g., hemihydrate (R0.5H) 2 O)) and polyhydrates (x is a number greater than 1, e.g. dihydrate (R.2H) 2 O) and hexahydrate (R.6H) 2 O))。
The compounds of the present invention may be in amorphous or crystalline form (crystalline or polymorphic). Furthermore, the compounds of the present invention may exist in one or more crystalline forms. Accordingly, the present invention includes within its scope all amorphous or crystalline forms of the compounds of the present invention. The term "polymorph" refers to a crystalline form of a compound (or a salt, hydrate, or solvate thereof) in a particular crystal packing arrangement. All polymorphs have the same elemental composition. Different crystalline forms typically have different X-ray diffraction patterns, infrared spectra, melting points, densities, hardness, crystal shape, optoelectronic properties, stability and solubility. Recrystallization solvent, crystallization rate, storage temperature, and other factors may result in a crystalline form being favored. Various polymorphs of a compound may be prepared by crystallization under different conditions.
The invention also includes isotopically-labeled compounds, which are identical to those recited in formula (I), but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes that can be incorporated into the compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine and chlorine, such as 2 H、 3 H、 13 C、 11 C、 14 C、 15 N、 18 O、 17 O、 31 P、 32 P、 35 S、 18 F and 36 and (4) Cl. Compounds of the present invention, prodrugs thereof, and pharmaceutically acceptable salts of said compounds or of said prodrugs which contain the aforementioned isotopes and/or other isotopes of other atoms are within the scope of this invention. Certain isotopically-labelled compounds of the invention, e.g. by incorporation of radioactive isotopes (e.g. by introducing 3 H and 14 c) Can be used in drug and/or substrate tissue distribution assays. Tritium, i.e. 3 H and carbon-14, i.e. 14 The C isotopes are particularly preferred because of their ease of preparation and detection. Further, by heavier isotopes, e.g. deuterium, i.e. 2 H, may be preferred in some cases because of the higher metabolic stability that may provide therapeutic benefits, such as increased in vivo half-life or reduced dosage requirements. Isotopically-labelled compounds of formula (I) of the present invention and prodrugs thereof can generally be prepared by substituting a readily available isotopically-labelled reagent for a non-isotopically-labelled reagent in the course of performing the procedures disclosed in the schemes and/or in the examples and preparations below.
In addition, prodrugs are also included within the context of the present invention. The term "prodrug" as used herein refers to a compound that is converted in vivo by hydrolysis, for example in the blood, into its active form, which has a medicinal effect. Pharmaceutically acceptable Prodrugs are described in t.higuchi and v.stella, produgs as Novel Delivery Systems, vol.14 of a.c.s.symposium Series, edward b.roche, ed., bioreversible Carriers in Drug Design, american Pharmaceutical Association and Pergamon Press,1987, and d.fleisher, s.ramon and h.barcra "Improved oral Delivery: solubility limits are provided for each of the users of the produgs ", advanced Drug Delivery Reviews (1996) 19 (2) 115-130, each of which is incorporated herein by reference.
A prodrug is any covalently bonded compound of the present invention that releases the parent compound in vivo when such prodrug is administered to a patient. Prodrugs are generally prepared by modifying functional groups in a manner such that the modification is effected by routine manipulation or in vivo cleavage to produce the parent compound. Prodrugs include, for example, compounds of the present invention wherein a hydroxy, amino, or sulfhydryl group is bonded to any group that, when administered to a patient, cleaves to form a hydroxy, amino, or sulfhydryl group. Thus, representative examples of prodrugs include, but are not limited to, acetate/amide, formate/amide, and benzoate/amide derivatives of hydroxy, mercapto, and amino functional groups of the compounds of formula (I). In addition, in the case of formic acid (-COOH), esters such as methyl ester, ethyl ester, and the like can be used. The ester itself may be active and/or may hydrolyze under in vivo conditions in the human body. Suitable pharmaceutically acceptable in vivo hydrolysable ester groups include those which readily break down in the human body to release the parent acid or salt thereof.
Treatment of
The present invention provides a method of treating and/or preventing a disease, such as wild-type and/or mutant EGFR kinase-mediated cancer, in a subject, comprising administering to the subject a compound of the present invention, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, or a pharmaceutical composition of the present invention.
In specific embodiments, the mutant EGFR is selected from the group consisting of exon 20 insertion mutant EGFR, exon 18 point mutant EGFR, exon 21 point mutant EGFR, exon 19 deletion mutant EGFR, or L858R mutant EGFR.
In particular embodiments, the mutant EGFR has a T790M mutation and has at least one mutation selected from an exon 20 insertion mutation, an exon 18 point mutation, an exon 21 point mutation, an exon 19 deletion mutation, or a L858R mutation.
As used herein, "EGFR" refers to the human epidermal growth factor receptor protein, also known as ErbB-1 or HER1.
Herein, "wild-type EGFR" refers to EGFR without somatic mutations.
Herein, "exon 20 insertion mutation" refers to a mutation in which one or more amino acids (preferably 1 to 7, more preferably 1 to 4) are inserted into the exon 20 region (e.g., amino acid sequence at position 761 to 823) of EGFR; preferably, the mutation is a mutation in which the amino acid sequence FQEA (in the order of phenylalanine, glutamine, glutamic acid and alanine from the N-terminus) is inserted between alanine at position 763 and tyrosine at position 764in the region of exon 20 (a 763_ Y764 insFQEA); preferably, the mutation is a mutation in which the amino acid sequence ASV (in the order of alanine, serine and valine from the N-terminus) is inserted between valine at position 769 and aspartic acid at position 770in the exon 20 region (V769 _ D770 insASV); preferably, the mutation is a mutation in which the amino acid sequence SVD (in the order of serine, valine and aspartic acid from the N-terminus) is inserted between asparagine at position 770 and asparagine at position 771in the exon 20 region (D770-N771 insSVD); preferably, the mutation is a mutation in which the amino acid sequence NPG (asparagine, proline and glycine in this order from the N-terminus) is inserted between aspartic acid at position 770 and asparagine at position 771in the exon 20 region (D770-N771 insNPG); preferably, the mutation is a mutation in which amino acid G (glycine) is inserted between aspartic acid at position 770 and asparagine at position 771 (D770 — N771 insG); preferably, the mutation is a mutation in which aspartic acid at position 770in the exon 20 region is deleted and thereby the amino acid sequence GY (glycine and tyrosine in this order from the N-terminus) is inserted (D770 > GY); preferably, the mutation is a mutation in which amino acid N (asparagine) is inserted between asparagine at position 771 and proline at position 772in the exon 20 region (N771 _ P772 insN); preferably, the mutation is a mutation in which the amino acid sequence PR (proline and arginine in this order from the N-terminus) is inserted between proline at position 772 and histidine at position 773in the region of exon 20 (P772 _ R773 insPR); preferably, the mutation is a mutation in which the amino acid sequence NPH (asparagine, proline and histidine in this order from the N-terminus) is inserted between the 773 rd histidine and the 774 th valine in the exon 20 region (H773V 774 insNPH); preferably, the mutation is a mutation in which the amino acid sequence PH (proline and histidine in this order from the N-terminus) is inserted between the 773 rd histidine and the 774 th valine in the exon 20 region (H773 _ V774 insPH); preferably, the mutation is a mutation in which the amino acid sequence AH (alanine and histidine in this order from the N-terminus) is inserted between the 773 rd histidine and the 774 th valine in the exon 20 region (H773 _ V774 insAH); preferably, the mutation is a mutation in which the amino acid H (histidine) is inserted between the 773 rd histidine and the 774 th valine in the exon 20 region (H773 _ V774 insH); preferably, the mutation is a mutation in which an amino acid sequence HV (in the order of histidine and valine from the N-terminus) is inserted between valine at position 774 and cysteine at position 775 in the region of exon 20 (V774 _ C774 insHV); preferably, the mutation is a mutation in which the amino acid sequence EAFQ (in the order of glutamic acid, alanine, phenylalanine and glutamine from the N-terminus) is inserted between alanine at position 761 and glutamic acid at position 762in the exon 20 region (A761 _ E762 insEAFQ). More preferably, the mutation is a mutation in which the amino acid sequence ASV (in the order of alanine, serine and valine from the N-terminus) is inserted between valine at position 769 and aspartic acid at position 770in the exon 20 region (V769 _ D770 insASV); more preferably, the mutation is a mutation wherein the amino acid sequence SVD is inserted (in this order of serine, valine and aspartic acid from the N-terminus) between aspartic acid at position 770 and asparagine at position 771in the region of exon 20 (D770-N771 insSVD); more preferably, the mutation is a mutation in which the amino acid sequence NPG (asparagine, proline and glycine in this order from the N-terminus) is inserted between aspartic acid at position 770 and asparagine at position 771in the exon 20 region (D770-N771 insNPG); more preferably, the mutation is a mutation in which an amino acid G (glycine) is inserted between aspartic acid at position 770 and asparagine at position 771in the region of exon 20 (D770-N771 insG); more preferably, the mutation is a mutation in which an amino acid sequence NPH (asparagine, proline and histidine in this order from the N-terminus) is inserted between the 773 rd histidine and the 774 th valine in the exon 20 region (H773 _ V774 insNPH); more preferably, the mutation is a mutation in which the amino acid sequence PH (proline and histidine in this order from the N-terminus) is inserted between the 773 rd histidine and the 774 th valine in the exon 20 region (H773 _ V774 insPH); more preferably, the mutation is a mutation in which the amino acid sequence SVD (in the order of serine, valine and aspartic acid from the N-terminus) is inserted between aspartic acid at position 770 and aspartic acid at position 771in the exon 20 region (D770-N771 insSVD); more preferably, the mutation is a mutation in which an amino acid G (glycine) is inserted between aspartic acid at position 770 and asparagine at position 771in the region of exon 20 (D770-N771 insG).
Herein, "cancer patient expressing EGFR having exon 20 insertion mutation" refers to a cancer patient expressing EGFR having exon 20 insertion mutation in at least a part of the exon 20 region of EGFR. EGFR may have exon 20 insertion mutations in two or more different portions, but preferably one of them. Furthermore, EGFR may also have other mutations (e.g., exon 19 deletion mutation, L858R mutation, or T790M mutation) in addition to the exon 20 insertion mutation.
In the present invention, the method for detecting an insertion mutation expressing EGFR exon 20in a cancer patient is not particularly limited as long as the method can detect the mutation, and any known detection method can be used. The detection target for detecting the exon 20 insertion mutation may be any one of the gene sequence of the EGFR gene, the transcription product of the EGFR gene, and the EGFR protein.
The sample for detecting the exon 20 insertion mutation is not particularly limited as long as the sample is a biological sample isolated from a cancer patient, particularly a sample obtained from a cancer patient and containing malignant tumor cells. Examples of biological samples include body fluids (e.g., blood, urine, etc.), tissues, extracts thereof, and cultures from which tissues are obtained. The method of isolating the biological sample may be appropriately selected depending on the type of the biological sample.
The biological sample is prepared by appropriate treatment according to the detection method. In addition, a reagent for detection (for example, a reagent containing a primer or a probe) can be prepared by a conventional method according to the detection method.
In one embodiment of the invention, the step of detecting the presence of an exon 20 insertion mutation of EGFR expressed in a patient with a malignant tumor may be performed prior to administering an anti-tumor agent to the cancer patient.
Herein, "exon 18 point mutation" means a point mutation in an amino acid in the exon 18 region of wild-type EGFR. Preferably, the mutation is a point mutation or a deletion mutation in which 1 amino acid in the exon 18 region is substituted; more preferably, the mutations are a point mutation wherein the glutamic acid encoded by codon 709 in exon 18 is substituted with an arbitrary amino acid (i.e., E790X), and a point mutation wherein the glycine encoded by codon 719 in exon 18 is substituted with an arbitrary amino acid (i.e., G719X). Specifically, E790X may, for example: a point mutation in which the glutamic acid encoded by codon 709 in the region of exon 18 is substituted with lysine (i.e., E709K), and a point mutation in which the glutamic acid encoded by codon 709 in the region of exon 18 is substituted with alanine (i.e., E709A). G719X may for example: a point mutation wherein the glycine encoded by codon 719 in the region of exon 18 is replaced with alanine (i.e., G719A), a point mutation wherein the glycine encoded by codon 719 in the region of exon 18 is replaced with serine (i.e., G719S), and a point mutation wherein the glycine encoded by codon 719 in the region of exon 18 is replaced with cysteine (i.e., G719C), with G719A being most common.
Herein, "exon 18-point mutant EGFR" refers to EGFR having at least 1 exon 18-point mutation; preferably the EGFR has more than 2 related exon 18 point mutations; more preferably, the EGFR has 1 exon 18 point mutation. Furthermore, the EGFR may have other mutations (e.g., exon 19 deletion mutation, L858R mutation, T790M mutation, etc.) other than the exon 18 point mutation.
Herein, "exon 21" refers to the region 824-875 in the amino acid sequence of wild-type EGFR.
Herein, "exon 21 point mutation" means a point mutation in amino acids of the exon 21 region of wild-type EGFR. Preferably, the exon 21 point mutation is a point mutation wherein 1 amino acid in the exon 21 region is replaced; more preferably, the exon 21 point mutation is a point mutation wherein the leucine encoded by codon 861 in the region of exon 21 is substituted with any amino acid (i.e.L861X), for example a point mutation wherein the leucine encoded by codon 861 in the region of exon 21 is substituted with glutamine (i.e.L861Q).
Herein, "exon 21 point mutant EGFR" means EGFR having at least 1 exon 21 point mutation; preferably the EGFR has more than 2 relevant exon 21 point mutations; more preferably, the EGFR has 1 exon 21 point mutation. Furthermore, the EGFR may have other mutations (e.g., exon 19 deletion mutation, L858R mutation, T790M mutation, etc.) other than the exon 21 point mutation.
In a specific embodiment, the mutant EGFR is a T790M mutation and has at least one mutation selected from the group consisting of an exon 20 insertion mutation, an exon 18 point mutation, an exon 21 point mutation, an exon 19 deletion mutation, and a L858R mutation.
Specifically, the T790M mutation-containing EGFR variant having an exon 18 point mutation and an exon 21 point mutation is any one of the following: mutant EGFR having the T790M mutation and having exon 18 regions E709X and/or G719X; has a T790M mutation and has exon 21 region L861X mutant EGFR. Specifically any of the following: EGFR with T790M mutation and with E709K or E709A mutation; EGFR mutant with T790M mutation and with G719A, G719S or G719C; EGFR mutant with T790M mutation and with L861Q; of these, EGFR mutants with the T790M mutation and with G719A and with the T790M mutation and with L861Q are more common.
Herein, the method for detecting that EGFR expressed by a cancer patient has exon 18 and/or exon 21 point mutation may be any method as long as the above mutation can be detected, and known detection methods may be used.
The sample for detecting the exon 18 and/or exon 21 point mutation is not particularly limited as long as the sample is a biological sample isolated from a cancer patient, particularly a sample obtained from a cancer patient and containing malignant tumor cells. Examples of biological samples include body fluids (e.g., blood, urine, etc.), tissues, extracts thereof, and cultures from which tissues are obtained. The method for isolating a biological sample may be appropriately selected depending on the type of the biological sample.
The biological sample is prepared by appropriate treatment according to the detection method. In addition, a reagent for detection (for example, a reagent containing a primer or a probe) can be prepared by a conventional method according to the detection method.
In one embodiment of the invention, the step of detecting the presence of exon 18 and/or exon 21 point mutations expressed in a patient with a malignant tumor may be performed prior to administering an anti-neoplastic agent to the cancer patient.
Specific examples of mutated EGFR kinase-mediated tumors of the invention include, but are not limited to: head and neck cancer, gastrointestinal cancer (esophageal cancer, gastric cancer, duodenal cancer, liver cancer, bile duct cancer (e.g., gallbladder and bile duct cancer), pancreatic cancer, colorectal cancer (e.g., colon cancer and rectal cancer), and the like), lung cancer (e.g., non-small cell lung cancer, and mesothelioma), breast cancer, genital cancer (ovarian cancer, uterine cancer (e.g., cervical cancer, and endometrial cancer), and the like), urinary tract cancer (e.g., renal cancer, bladder cancer, prostate cancer, and testicular cancer), hematopoietic tumors (e.g., leukemia, malignant lymphoma, and multiple myeloma), osteosarcoma, soft tissue sarcoma, skin cancer, brain tumor, and the like. Preferred examples include lung cancer, breast cancer, head and neck cancer, brain cancer, uterine cancer, hematopoietic cancer or skin cancer.
In specific embodiments, the mutant EGFR is selected from the group consisting of exon 20 insertion mutant EGFR, exon 18 point mutant EGFR, exon 21 point mutant EGFR, exon 19 deletion mutant EGFR, or L858R mutant EGFR.
In particular embodiments, the mutant EGFR has a T790M mutation and has a mutation selected from exon 18 point mutant EGFR, exon 21 point mutant EGFR, exon 19 deletion mutant EGFR, or L858R mutant EGFR.
The present invention also provides a method of treating a patient with a tumor comprising the step of administering to a patient expressing a tumor having an EGFR mutant selected from the group consisting of exon 20 insertion mutant EGFR, exon 18 point mutant EGFR, exon 21 point mutant EGFR, exon 19 deletion mutant EGFR and L858R mutant EGFR an effective amount of an antineoplastic agent comprising a compound of the present invention or a pharmaceutically acceptable salt thereof.
The invention also provides a compound of the invention, or a pharmaceutically acceptable salt thereof, for use in treating a patient expressing a tumor having an exon 20 insertion mutant EGFR, an exon 18 point mutant EGFR, an exon 21 point mutant EGFR, an exon 19 deletion mutant EGFR or an L858R mutant EGFR.
The invention also provides the use of a compound of the invention, or a pharmaceutically acceptable salt thereof, for the treatment of a patient having a tumor selected from the group consisting of exon 20 insertion mutant EGFR, exon 18 point mutant EGFR, exon 21 point mutant EGFR, exon 19 deletion mutant EGFR or L858R mutant EGFR.
The present invention also provides a method for predicting the effect of treatment with an antitumor agent, which is the compound of the present invention or a pharmaceutically acceptable salt thereof as an active ingredient, in a tumor patient, the method comprising the following steps (1) and (2):
(1) A step of detecting the presence or absence of a mutation of the EGFR gene contained in a biological sample obtained from the patient; and
(2) A step of predicting that chemotherapy is highly likely to exhibit a sufficient therapeutic effect on a patient when the result of the detection in step (1) reveals that the EGFR gene has a mutation selected from the group consisting of an exon 20 insertion mutation, an exon 18 point mutation, an exon 21 point mutation, an exon 19 deletion mutation and an L858R mutation.
The present invention also provides a method for treating a patient having a tumor, the method comprising the following steps (1) to (2):
(1) A step of detecting the presence or absence of a mutation of the EGFR gene contained in a biological sample obtained from the patient;
(2) A step of treating the patient with the compound of the present invention or a pharmaceutically acceptable salt thereof when the EGFR gene is found to have a mutation selected from the group consisting of an exon 20 insertion mutation, an exon 18 point mutation, an exon 21 point mutation, an exon 19 deletion mutation and an L858R mutation as a result of the examination in step (1).
In another aspect, the present invention provides a method of treating and/or preventing a disease, such as a wild-type and/or mutant HER2 kinase-mediated tumor, in a subject, comprising administering to the subject a compound of the present invention, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, or a pharmaceutical composition of the present invention.
In specific embodiments, the mutated HER2 is selected from G309A mutant HER2, S310F mutant HER2, R678Q mutant HER2, L775_ T759 deletion mutant HER2, D769H mutant HER2, V777L mutant HER2, V842I mutant HER2, R869C mutant HER2, L755S mutant HER2, or ex20 insymva mutant HER2.
In a specific embodiment, the ex20insYVMA mutant HER2 is selected from a775_ G776insYVMA mutant HER2 mutation.
As used herein, "HER2" includes HER2 of a human or non-human mammal. Also, the term "HER2" includes subtypes.
In the present invention, the HER2 kinase-mediated tumor is preferably a tumor having HER2 overexpression, HER2 gene amplification or HER2 mutation. The "tumor" is not particularly limited, and examples thereof include head and neck cancer, esophageal cancer, gastric cancer, colon cancer, rectal cancer, liver cancer, gallbladder-bile duct cancer, biliary tract cancer, pancreatic cancer, lung cancer, breast cancer, ovarian cancer, cervical cancer, uterine cancer, kidney cancer, bladder cancer, prostate cancer, testicular tumor, bone-soft tissue sarcoma, hematological cancer, multiple myeloma, skin cancer, brain tumor, and mesothelial cancer. Preferably, the cancer is breast cancer, gastric cancer, esophageal cancer, ovarian cancer, lung cancer, esophageal cancer, gallbladder-bile duct cancer, biliary tract cancer, bladder cancer or colon cancer, more preferably breast cancer, gastric cancer, esophageal cancer, biliary tract cancer, ovarian cancer, lung cancer or esophageal cancer, and further preferably breast cancer, gastric cancer or lung cancer.
In the methods of treatment of the present invention, an "effective amount" refers to an amount or dose sufficient to produce the desired therapeutic benefit in an individual in need of such treatment. An effective amount or dose of a compound of the invention can be determined by conventional methods (e.g., modeling, dose escalation, or clinical trials) and by conventional factors (e.g., the mode or route of drug delivery, the pharmacokinetics of the agent, the severity and course of the infection, the health and weight of the individual, and the judgment of the treating physician). Exemplary doses are in the range of about 0.1mg to 1g per day, or about 1mg to 50mg per day, or about 50mg to 250mg per day, or about 250mg to 1g per day. The total dose can be administered as a single dose or as separate dosage units (e.g., BID, TID, QID).
After the patient has developed an improvement in the disease, the dosage can be adjusted for prophylactic or maintenance treatment. For example, the dosage or frequency of administration, or both, can be reduced to an amount necessary to maintain the desired therapeutic or prophylactic effect, depending on the symptoms. Of course, if the symptoms have been alleviated to an appropriate degree, treatment may be discontinued. However, when either symptom recurs, the patient may require long-term intermittent treatment. Patients may also require chronic treatment for extended periods of time.
Pharmaceutical compositions, formulations and kits
In another aspect, the present invention provides a pharmaceutical composition comprising a compound of the present invention (also referred to as "active ingredient") and a pharmaceutically acceptable excipient. In some embodiments, the pharmaceutical composition comprises an effective amount of an active ingredient. In some embodiments, the pharmaceutical composition comprises a therapeutically effective amount of an active ingredient. In some embodiments, the pharmaceutical composition comprises a prophylactically effective amount of an active ingredient.
Pharmaceutically acceptable excipients for use in the present invention refer to non-toxic carriers, adjuvants or vehicles that do not destroy the pharmacological activity of the compounds formulated therewith. Pharmaceutically acceptable carriers, adjuvants, or vehicles that may be used in the compositions of the present invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins (such as human serum albumin), buffer substances (such as phosphates), glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes (such as protamine sulfate), disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, silica gel, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol, and wool fat.
The invention also includes kits (e.g., pharmaceutical packages). The provided kits can include a compound of the invention, an additional therapeutic agent, and first and second containers (e.g., vials, ampoules, bottles, syringes, and/or dispensable packages or other suitable containers) containing the compound of the invention, the additional therapeutic agent. In some embodiments, kits are provided that may also optionally include a third container containing a pharmaceutically acceptable excipient for diluting or suspending a compound of the invention and/or other therapeutic agent. In some embodiments, the compound of the present invention and the additional therapeutic agent provided in the first container and the second container are combined to form one unit dosage form.
The pharmaceutical compositions provided by the present invention may be administered by a number of routes including, but not limited to: oral, parenteral, inhalation, topical, rectal, nasal, buccal, vaginal, by implant or other modes of administration. For example, parenteral administration as used herein includes subcutaneous administration, intradermal administration, intravenous administration, intramuscular administration, intraarticular administration, intraarterial administration, intrasynovial administration, intrasternal administration, intracerebrospinal administration, intralesional administration, and intracranial injection or infusion techniques.
Typically, an effective amount of a compound provided herein is administered. The amount of compound actually administered can be determined by a physician, as the case may be, including the condition to be treated, the chosen route of administration, the compound actually administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like.
When used to prevent a condition according to the invention, a subject at risk of developing the condition is administered a compound provided herein, typically based on physician's advice and under the supervision of a physician, at a dosage level as described above. Subjects at risk of developing a particular disorder, typically include subjects with a family history of the disorder, or those determined to be particularly susceptible to developing the disorder by genetic testing or screening.
The pharmaceutical compositions provided herein may also be administered chronically ("chronic administration"). By long-term administration is meant administration of the compound or pharmaceutical composition thereof over a long period of time, e.g., 3 months, 6 months, 1 year, 2 years, 3 years, 5 years, etc., or may continue for an indefinite period of time, e.g., for the remainder of the subject's life. In some embodiments, chronic administration is intended to provide a constant level of the compound in the blood over a prolonged period of time, e.g., within the therapeutic window.
Various methods of administration may be used to further deliver the pharmaceutical compositions of the present invention. For example, in some embodiments, the pharmaceutical composition may be administered as a bolus, e.g., in order to rapidly increase the concentration of the compound in the blood to an effective level. The bolus dose depends on the targeted systemic level of the active ingredient, e.g., an intramuscular or subcutaneous bolus dose results in a slow release of the active ingredient, while a bolus delivered directly to the vein (e.g., by IV intravenous drip) can be delivered more rapidly, allowing the concentration of the active ingredient in the blood to rise rapidly to an effective level. In other embodiments, the pharmaceutical composition may be administered as a continuous infusion, e.g., by IV intravenous drip, to provide a steady state concentration of the active ingredient in the body of the subject. Furthermore, in other embodiments, a bolus dose of the pharmaceutical composition may be administered first, followed by continuous infusion.
Oral compositions may take the form of bulk liquid solutions or suspensions or bulk powders. More generally, however, the compositions are provided in unit dosage form for convenient administration of the precise dosage. The term "unit dosage form" refers to physically discrete units suitable as unitary dosages for human patients and other mammals, each unit containing a predetermined quantity of active material adapted to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient. Typical unit dosage forms include prefilled, pre-measured ampoules or syringes of liquid compositions, or in the case of solid compositions, pills, tablets, capsules and the like. In such compositions, the compound is typically a minor component (about 0.1 to about 50% by weight, or preferably about 1 to about 40% by weight), with the remainder being various carriers or excipients and processing aids useful in forming the desired form of administration.
For oral dosages, a representative regimen is one to five oral dosages, particularly two to four oral dosages, typically three oral dosages per day. Using these dosing modes, each dose provides about 0.01 to about 20mg/kg of a compound of the invention, with preferred doses each providing about 0.1 to about 10mg/kg, especially about 1 to about 5mg/kg.
In order to provide a blood level similar to, or lower than, the use of the injected dose, a transdermal dose is generally selected in an amount of from about 0.01 to about 20% by weight, preferably from about 0.1 to about 10% by weight, and more preferably from about 0.5 to about 15% by weight.
From about 1 to about 120 hours, especially 24 to 96 hours, the injection dosage level is in the range of about 0.1 mg/kg/hour to at least 10 mg/kg/hour. To obtain sufficient steady state levels, a preload bolus of about 0.1mg/kg to about 10mg/kg or more may also be administered. For human patients of 40 to 80kg, the maximum total dose cannot exceed about 2 g/day.
Liquid forms suitable for oral administration may include suitable aqueous or nonaqueous carriers, as well as buffers, suspending and dispersing agents, coloring and flavoring agents, and the like. Solid forms may include, for example, any of the following components, or compounds with similar properties: a binder, for example, microcrystalline cellulose, gum tragacanth or gelatin; excipients, for example, starch or lactose, disintegrants, for example, alginic acid, primogel or corn starch; lubricants, for example, magnesium stearate; glidants, e.g., colloidal silicon dioxide; sweetening agents, for example, sucrose or saccharin; or a flavoring agent, for example, peppermint, methyl salicylate, or orange flavoring.
Injectable compositions are typically based on sterile saline or phosphate buffered saline for injection, or other injectable excipients known in the art. As previously mentioned, in such compositions, the active compound is typically a minor component, often about 0.05 to 10% by weight, with the remainder being injectable excipients and the like.
Transdermal compositions are typically formulated as topical ointments or creams containing the active ingredient. When formulated as an ointment, the active ingredient is typically combined with a paraffinic or water-miscible ointment base. Alternatively, the active ingredient may be formulated as a cream with a cream base, for example of the oil-in-water type. Such transdermal formulations are well known in the art and typically include other components for enhancing stable skin penetration of the active ingredient or formulation. All such known transdermal formulations and components are included within the scope provided by the present invention.
The compounds of the present invention may also be administered by transdermal means. Thus, transdermal administration may be accomplished using a reservoir (reservoir) or porous membrane type, or a variety of solid matrix patches.
The above components of the compositions for oral, injection or topical administration are merely representative. Other materials and processing techniques are described in Remington's Pharmaceutical Sciences,17th edition,1985, mack Publishing company, easton, pennsylvania, section 8, which is incorporated herein by reference.
The compounds of the present invention may also be administered in sustained release form, or from a sustained release delivery system. A description of representative sustained release materials can be found in Remington's Pharmaceutical Sciences.
The invention also relates to pharmaceutically acceptable formulations of the compounds of the invention. In one embodiment, the formulation comprises water. In another embodiment, the formulation comprises a cyclodextrin derivative. The most common cyclodextrins are α -, β -and γ -cyclodextrins consisting of 6, 7 and 8 α -1,4-linked glucose units, respectively, which optionally include one or more substituents on the linked sugar moiety, including but not limited to: methylated, hydroxyalkylated, acylated, and sulfoalkyl ether substitutions. In some embodiments, the cyclodextrin is sulfoalkyl ether β -cyclodextrin, e.g., sulfobutyl ether β -cyclodextrin, also known as Captisol. See, e.g., U.S.5,376,645. In some embodiments, the formulation includes hexapropyl- β -cyclodextrin (e.g., 10-50% in water).
Pharmaceutical combination
The compounds of the invention described herein may be used in combination with one or more other active ingredients in pharmaceutical compositions or methods for the treatment of the diseases and conditions described herein. Other additional active ingredients include other therapeutic agents or agents that mitigate the adverse effects of treatment against the intended disease target. The combinations can be used to increase efficacy, ameliorate other disease symptoms, reduce one or more side effects, or reduce the required dose of the compounds of the invention. The additional active ingredients may be formulated as separate pharmaceutical compositions from the compound of the invention or may be included in a single pharmaceutical composition with the compound of the invention. The additional active ingredient may be administered simultaneously with, before or after the administration of the compound of the invention.
Combination agents include those active ingredients known or observed to be effective in treating the diseases and conditions described herein, including those effective against another target associated with the disease. For example, the compositions and formulations, and methods of treatment of the present invention may further comprise other drugs, such as other agents useful for treating or ameliorating a target disease or associated symptoms or conditions. For cancer indications, the other agents include, but are not limited to, kinase inhibitors, such as EGFR inhibitors (e.g., erlotinib, gefitinib); raf inhibitors (e.g., vemurafenib), VEGFR inhibitors (e.g., sunitinib); standard chemotherapeutic agents, such as alkylating agents, antimetabolites, antitumor antibiotics, topoisomerase inhibitors, platinum drugs, mitotic inhibitors, antibodies, hormonal therapy, or corticosteroids. For pain indications, suitable combination agents include anti-inflammatory agents, such as NSAIDs. The pharmaceutical compositions of the invention may additionally comprise one or more of said active agents, and the method of treatment may additionally comprise administering an effective amount of one or more of said active agents.
Examples
The invention will be further illustrated with reference to the following specific examples. It should be understood that these examples are for illustrative purposes only and are not intended to limit the scope of the present invention. The experimental procedures, in which specific conditions are not specified, in the following examples are generally carried out according to conventional conditions or according to conditions recommended by the manufacturers. Parts and percentages are parts and percentages by weight unless otherwise indicated.
In general, in the preparative schemes, each reaction is carried out in an inert solvent at temperatures ranging from room temperature to reflux temperature (e.g., 0 ℃ to 100 ℃, preferably 0 ℃ to 80 ℃). The reaction time is usually 0.1 to 60 hours, preferably 0.5 to 24 hours.
Abbreviations used herein have the following meanings:
Pd(PPh 3 ) 4 : tetrakis (triphenylphosphine) palladium
Na 2 CO 3 : sodium carbonate
EDCI: 1-Ethyl- (3-dimethylaminopropyl) carbodiimide hydrochloride
HOBT: 1-hydroxybenzotriazoles
NIS: n-iodosuccinimide
DMF: n, N-dimethylformamide
Pd(dppf)Cl 2 : [1,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride
MTBE: methyl tert-butyl ether
DME: ethylene glycol dimethyl ether
NBS: n-bromosuccinimide
Cbz: benzyloxycarbonyl group
TEA: triethylamine
DCM: methylene dichloride
ACN: acetonitrile
POCl 3 : phosphorus oxychloride
tert-Butyl nitrate: nitrous acid tert-butyl ester
TPP: triphenylphosphine
The DIAD: diisopropyl azodicarboxylate
NH 4 OAc: ammonium acetate
DMSO, DMSO: dimethyl sulfoxide
B 2 pin 2 : biboronic acid pinacol ester
KOAc (Koac): potassium acetate
A Dioxane: dioxane (dioxane)
NaNO 2 : sodium nitrite
EtOH: ethanol
NaOH: sodium hydroxide
DIPEA: n, N-diisopropylethylamine
TFAA: trifluoroacetic anhydride
H 2 O 2 : hydrogen peroxide solution
And (3) DBU:1,8-diazabicyclo [5.4.0] undec-7-ene
NaH: sodium hydride
THF: tetrahydrofuran (THF)
i-PrOH: isopropyl alcohol
PtO 2 : platinum dioxide
EA: acetic acid ethyl ester
EtOH: ethanol
Tosmic: p-methylbenzenesulfonylmethyliononitrile
PdCl 2 : palladium dichloride
TESiH: triethylsilane
NaIO 4 : sodium periodate
Intermediate A1 3- (7-amino-1H-pyrazolo [4,3-d]Pyrimidin-3-yl) -5,6-dihydropyridine-1 (2H) -carboxylic acid Preparation of tert-butyl ester
Figure BDA0002983446160000401
The following synthetic route was used:
Figure BDA0002983446160000411
adding 3-iodine-1H-pyrazolo [4,3-d into a reaction bottle]Pyrimidine-7-amine (1.2g, 4.6 mmol), 3- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl) -5,6-dihydropyridine-1 (2H) -carboxylic acid tert-butyl ester (2.1g, 6.9 mmol), tetrakis (triphenylphosphine) palladium (53mg, 0.05mmol) and sodium carbonate (975mg, 9.2mmol) were added, 20ml dioxane and 2ml water were added and the reaction was stirred under nitrogen to 90 ℃ for 6 hours, TLC monitored for completion of the reaction, the solvent was concentrated and purified by silica gel column chromatography to give 1.17g of product, yield: 80.7 percent. LC-MS (APCI) M/z =317.2 (M + 1) +
Intermediate A2- (7-amino-1H-pyrazolo [4,3-d]Preparation of pyrimidin-3-yl) piperidine-1-carboxylic acid tert-butyl ester
Figure BDA0002983446160000412
The following synthetic route was used:
Figure BDA0002983446160000413
adding 3- (7-amino-1H-pyrazolo [4,3-d) into a reaction bottle]Pyrimidin-3-yl) -5,6-dihydropyridine-1 (2H) -carboxylic acid tert-butyl ester (1.17g, 3.7 mmol), catalytic amount of palladium on carbon, dissolved in 20ml of methanol, charged with hydrogen balloon, stirred at room temperature for 5 hours, checked by TLC, the catalyst was removed by filtration, concentrated and purified by silica gel column chromatography to give 1.05g of product, yield: 89 percent. LC-MS (APCI) M/z =319.5 (M + 1) +
Intermediate A3 (R) -3- (8-amino-1-iodoimidazo [1,5-a]Process for preparing pyrazin-3-yl) piperidine-1-carboxylic acid tert-butyl ester Preparation of
Figure BDA0002983446160000414
The following synthetic route was used:
Figure BDA0002983446160000421
step 1 Synthesis of (R) -3- (((3-chloropyrazin-2-yl) methyl) carbamoyl) piperidine-1-carboxylic acid tert-butyl ester
Adding 2-aminomethyl-3-chloropyrazine hydrochloride (1.0g, 4.62mmol), (R) -1- (tert-butoxycarbonyl) piperidine-3-carboxylic acid (1.11g, 4.85mmol) and HOBT (0.94g, 6.93mmol) into a reaction bottle, dissolving with 20ml of anhydrous DMF, adding triethylamine (2.34g, 23.1mmol), cooling to 0 ℃ under the protection of nitrogen, adding EDCI (1.33g, 6.93mmol) in batches, heating to room temperature, stirring for reacting overnight, adding excessive water after TLC monitoring of the reaction, quenching the reaction, extracting with ethyl acetate for 3-4 times, combining organic phases, washing with saturated common salt water, concentrating, purifying by column chromatography, and drying in vacuum to obtain a colorless oily product with the yield of 1.11 g: 67.8 percent. LC-MS (APCI): m/z =355.3 (M+1) +
Step 2 Synthesis of (R) -3- (8-chloroimidazo [1,5-alpha ] pyrazin-3-yl) piperidine-1-carboxylic acid tert-butyl ester
Adding (R) -3- (((3-chloropyrazin-2-yl) methyl) carbamoyl) piperidine-1-carboxylic acid tert-butyl ester (1.0g, 2.82mmol) into a reaction bottle, adding 25ml of anhydrous ethyl acetate under the protection of nitrogen to dissolve, adding 3.7ml of anhydrous DMF, slowly dropwise adding phosphorus oxychloride (2.6 g,17.0 mmol) under an ice salt bath, after the addition is finished, heating to room temperature, stirring for reaction for 1h, monitoring the reaction by TLC, adding 20ml of saturated sodium carbonate aqueous solution under an ice bath to quench the reaction, separating out an organic phase, extracting an aqueous phase for 3-4 times by using ethyl acetate, combining the organic phase, washing by using saturated saline solution, and purifying by silica gel column chromatography (petroleum ether: ethyl acetate 2:1) after concentration to obtain 0.89g of oily product, wherein the yield is as follows: 93.8 percent. LC-MS (APCI) M/z =337.8 (M + 1) +
Step 3 Synthesis of tert-butyl 3 (R) -3- (8-chloro-1-iodoimidazo [1,5-alpha ] pyrazin-3-yl) piperidine-1-carboxylate
Adding (R) -3- (8-chloroimidazo [1,5-alpha) into a reaction bottle]Pyrazin-3-yl) piperidine-1-carboxylic acid tert-butyl ester (0.89g, 2.64mmol), dissolved in 10ml DMF, added NIS (0.62g, 2.77mmol) at room temperature, warmed to 50 ℃ and stirred for reaction overnight, monitored by TLC after the reaction is completed, slowly added saturated sodium bicarbonate solution to quench the reaction, extracted 3-4 times with ethyl acetate, the organic phases are combined, washed with saturated brine, concentrated and subjected to silica gel column chromatography (petroleum ether: ethyl acetate 2:1) gave 1.06g of a beige solid, yield: 86.9 percent. LC-MS (APCI) M/z =463.2 (M + 1) +
Step 4 Synthesis of tert-butyl 3- (8-amino-1-iodoimidazole [1,5-a ] pyrazin-3-yl) piperidine-1-carboxylate
Adding (R) -3- (8-chloro-1-iodoimidazo [1,5-alpha) into a reaction bottle]Pyrazin-3-yl) piperidine-1-carboxylic acid tert-butyl ester (1.06g, 2.29mmol), 8ml 2-butanol, ammonia (6.7ml, 176mmol), heating to 90 ℃ under sealed conditions for reaction overnight, cooling to room temperature, monitoring by TLC after the reaction is finished, diluting with excessive water, extracting with ethyl acetate for 3-4 times, combining the organic phases, washing with saturated saline, drying with anhydrous sodium sulfate, concentrating to remove the solvent, adding MTBE, pulping and purifying to obtain 0.89g product,yield: 87.6 percent. LC-MS (APCI) M/z =444.3 (M + 1) +
Intermediate A4- (4-amino-5-bromopyrrolo [2,1-f)][1,2,4]Triazin-7-yl) piperidine-1-carboxylic acid tert-butyl ester Preparation of esters
Figure BDA0002983446160000431
The following synthetic route was used:
Figure BDA0002983446160000432
step 1 Synthesis of tert-butyl 3- (4-Aminopyrrolo [2,1-f ] [1,2,4] triazin-7-yl) -5,6-dihydropyridine-1 (2H) -carboxylate
4-amino-7-bromopyrrolo [2,1-f][1,2,4]Triazine (4.69g, 13.6 mmol), 3- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl) -5,6-dihydropyridine-1 (2H) -carboxylic acid tert-butyl ester (5.04g, 16.3mmol), pd (dppf) Cl 2 (0.5g, 0.68mmol) and Na 2 CO 3 (4.35g, 41mmol) was added to 70mL DME and 15mL water, purged with nitrogen three times, and allowed to warm to 90 ℃ for reaction overnight. The reaction was cooled to room temperature, 100mL of water was added, extraction was performed with ethyl acetate (60ml x 3), and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and separated by a silica gel column to obtain 3.21g of a pale yellow solid with a yield of 75%. ESI-MS:316[ M ] + +1].
Step 2 Synthesis of tert-butyl 3- (4-Aminopyrrolo [2,1-f ] [1,2,4] triazin-7-yl) piperidine-1-carboxylate
3- (4-amino pyrrolo [2,1-f)][1,2,4]Triazin-7-yl) -5,6-dihydropyridine-1 (2H) -carboxylic acid tert-butyl ester (3.21g, 10.2mmol) was dissolved in 30mL of absolute ethanol, 300mg of 10% palladium on carbon was added, hydrogen was substituted three times, and the mixture was stirred under a hydrogen atmosphere of one atmosphere overnight. After the reaction is completed, palladium-carbon is filtered, the filtrate is concentrated, and a light yellow oily substance of 2.9g is obtained by silica gel column separation, with the yield of 90%. ESI-MS:318[ m ] + +1].
Step 3 Synthesis of tert-butyl 3- (4-amino-5-bromopyrrolo [2,1-f ] [1,2,4] triazin-7-yl) piperidine-1-carboxylate
Reacting 3- (4-aminopyrrolo [2,1-f)][1,2,4]Triazin-7-yl) piperidine-1-carboxylic acid tert-butyl ester (2.9 g, 9.169mol) was dissolved in 30mL of DMF, NBS (1.78g, 10mmol) was added in portions under ice bath, and the mixture was allowed to naturally warm to room temperature for overnight reaction. After completion of the reaction, 100mL of water was added to the reaction mixture, and the mixture was extracted with ethyl acetate (40ml. Multidot.3), and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and separated by a silica gel column to obtain 2.54g of a pale yellow solid with a yield of 70%. ESI-MS:398[ M ] + +2].
Intermediate A5 (R) -3- (4-amino-5-bromoimidazo [5,1-f][1,2,4]Triazin-7-yl) piperidine-1-carboxylic acid benzyl ester Preparation of the base esters
Figure BDA0002983446160000433
The following synthetic route was used:
Figure BDA0002983446160000441
step 1 Synthesis of (R) -piperidine-1,3-dicarboxylic acid (1-benzyl) (3-succinimidyl) ester
(R) -1- ((benzyloxy) carbonyl) piperidine-3-carboxylic acid (8g, 30.4mmol), N-hydroxysuccinimide (4.26g, 37mmol) and triethylamine (6.14g, 60.8mmol) were dissolved in 50mL of dichloromethane, EDCI (8.68g, 45.3mmol) was added under ice-cooling, and the reaction was allowed to proceed at room temperature overnight. The reaction mixture was diluted with 50mL of dichloromethane, washed with water, and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and separated by silica gel column to give 10.15g of a pale yellow oil with a yield of 93%. ESI-MS:361[ M ] + +1].
Step 2 Synthesis of benzyl (R) -3- (((3-amino-5-oxo-4,5-dihydro-1,2,4-triazin-6-yl) methyl) carbamoyl) piperidine-1-carboxylate
(R) -piperidine-1,3-dicarboxylic acid (1-benzyl) (3-succinimidyl) ester (10.15g, 28).2 mmol) and 3-amino-6- (aminomethyl) -1,2,4-triazin-5 (4H) -one acetate (5.67g, 28.2mmol) were dissolved in 100mL acetonitrile, triethylamine (8.54g, 84.6 mmol) was added, and the reaction was allowed to warm to 50 ℃ overnight. After the reaction was cooled, 150mL of water was added to dilute the reaction solution, and the mixture was extracted with ethyl acetate (80ml × 3), and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and separated by a silica gel column to obtain 8.07g of a pale yellow solid with a yield of 74%. ESI-MS:387[ M ]) + +1].
Step 3 Synthesis of benzyl 3 (R) -3- (2-amino-4-oxo-3,4-dihydroimidazo [5,1-f ] [1,2,4] triazin-7-yl) piperidine-1-carboxylate
Benzyl (R) -3- (((3-amino-5-oxo-4,5-dihydro-1,2,4-triazin-6-yl) methyl) carbamoyl) piperidine-1-carboxylate (8.07g, 20.9mmol) was dissolved in 80mL acetonitrile, phosphorus oxychloride (6.4 g, 41.8mmol) was added slowly and the temperature was raised to 60 ℃ for reaction overnight. The reaction was cooled and quenched with water, adjusted to PH =7 with 2N sodium hydroxide solution, extracted with ethyl acetate (60ml × 3), the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and separated by silica gel column to give 5.78g of light yellow solid with a yield of 75.2%. ESI-MS [ 369 ] M + +1].
Step 4 Synthesis of benzyl 3- (2-amino-5-bromo-4-oxo-3,4-dihydroimidazo [5,1-f ] [1,2,4] triazin-7-yl) piperidine-1-carboxylate
Mixing (R) -3- (2-amino-4-oxo-3,4-dihydroimidazo [5,1-f][1,2,4]Benzyl triazin-7-yl) piperidine-1-carboxylate (5.78g, 15.7 mmol) was dissolved in 40mL DMF, NBS (2.94g, 16.5 mmol) was added in portions under ice bath, and the mixture was allowed to warm to room temperature naturally overnight. After completion of the reaction, 100mL of water was added to the reaction mixture, and the mixture was extracted with ethyl acetate (50ml × 3), and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and separated by a silica gel column to obtain 5.77g of a pale yellow solid with a yield of 82%. ESI-MS:449[ M ] + +2].
Step 5 Synthesis of benzyl 3- (5-bromo-4-oxo-3,4-dihydroimidazo [5,1-f ] [1,2,4] triazin-7-yl) piperidine-1-carboxylate
Mixing (R) -3- (2-amino-5-bromo-4-oxo-3,4-dihydroimidazo [5,1-f][1,2,4]Triazin-7-yl) piperidine-1-carboxylic acid benzyl ester (5.77g, 12.9 mmol) was dissolved in 50mL tetrahydrofuran, and tert-butyl nitrite was added slowly(2g, 19.4 mmol) and the reaction was carried out at 60 ℃ for 3 hours. After completion of the reaction, 100mL of water was added to dilute the reaction mixture, and the mixture was extracted with ethyl acetate (100ml × 3), and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and separated by a silica gel column to obtain 4.79g of a pale yellow solid with a yield of 86%. ESI-MS:434[ m ] + +2].
Step 6 Synthesis of benzyl (R) -3- (4-amino-5-bromoimidazo [5,1-f ] [1,2,4] triazin-7-yl) piperidine-1-carboxylate
Mixing (R) -3- (5-bromo-4-oxo-3,4-dihydroimidazo [5,1-f][1,2,4]Benzyl triazin-7-yl) piperidine-1-carboxylate (4.79g, 11.1mmol) and 1,2,4-triazole (2.3 g,33.3 mmol) were dissolved in 40mL pyridine, phosphorus oxychloride (5.1g, 33.3 mmol) was slowly added dropwise under ice bath, and after the addition was completed, the mixture was allowed to move to room temperature for reaction for 2 hours. An ammonia-ethanol solution (15mL, 7mol/L) was slowly added dropwise to the reaction mixture in an ice bath, and the mixture was stirred at room temperature for 2 hours after completion of the dropwise addition. The reaction was completed by TLC, the reaction solution was quenched with water, extracted with ethyl acetate (80ml × 3), and the organic phase was washed with 2N hydrochloric acid to pH =5-6, then washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and separated by silica gel column to give 3.6g of pale yellow solid with a yield of 75.5%. ESI-MS:433[ m ] + +2].
Intermediate A6 (R) -3- (4-chloro-5-iodo-7H-pyrrolo [2,3-d]Pyrimidin-7-yl) piperidine-1-carboxylic acid tert-butyl ester Preparation of (2)
Figure BDA0002983446160000451
The following synthetic route was used:
Figure BDA0002983446160000452
4-chloro-5-iodo-7H-pyrrolo [2,3-d ] was added sequentially to a 50mL three-necked flask equipped with magnetic stirring]Pyrimidine (2.50g, 8.95mmol) and anhydrous tetrahydrofuran (40 mL), stirred to dissolve, tert-butyl (S) -3-hydroxypiperidine-1-carboxylate (1.80g, 8.95mmol) added, cooled to 0 ℃ under nitrogen, triphenylphosphine (3.05g, 11.63m, m. RTM.) are addedmol), stirred for 5min, DIAD (2.35g, 11.63mmol) was added slowly dropwise, the ice bath removed after addition, and the reaction stirred at room temperature under nitrogen overnight. The reaction mixture was diluted with ethyl acetate (100 mL), washed with water (50ml × 2), dried over anhydrous sodium sulfate, filtered, concentrated, and the residue was passed through a silica gel column to give 500mg of a white solid with a yield of 12.08%. LC-MS (APCI) M/z =463.1 (M + 1) + . 1 H NMR(400MHz,CDCl 3 )δ(ppm):8.64(s,1H),7.49(s,1H),4.83-4.76(m,1H),4.24-4.15(m,1H),3.97(d,J=13.2Hz,1H),3.35-3.29(m,1H),3.09-3.02(m,1H),2.21-2.04(m,2H),1.87-1.70(m,2H),1.48(s,9H).
Preparation of intermediate B1 tert-butyl 3- (1-cyano-2-methoxy-2-oxoethyl) piperidine-1-carboxylate
Figure BDA0002983446160000461
The following synthetic route was used:
Figure BDA0002983446160000462
step 1 Synthesis of tert-butyl 3- (1-cyano-2-methoxy-2-oxoethylidene) piperidine-1-carboxylate
3-Oxopiperidine-1-carboxylic acid tert-butyl ester (5.0g, 25mmol), 2-cyanoacetic acid methyl ester (2.5g, 25mmol) and ammonium acetate (0.25g, 3.25mmol) were charged into a reaction flask, 25ml of toluene was added, 0.5ml of glacial acetic acid was further added, the mixture was heated to reflux (water separation) and stirred for 7 hours, after the completion of the TLC monitoring reaction, the solvent was removed by concentration, and silica gel column chromatography purification gave 6.75g of a colorless oily liquid, yield: 96.4 percent. LC-MS (APCI) M/z =281.1 (M + 1) +
Step 2 Synthesis of tert-butyl 3- (1-cyano-2-methoxy-2-oxoethyl) piperidine-1-carboxylate (intermediate B1)
Adding 3- (1-cyano-2-methoxy-2-oxoethylene) piperidine-1-carboxylic acid tert-butyl ester (6.75g, 24mmol) and catalytic amount of palladium carbon into a reaction bottle, adding 100ml ethanol for dissolving, charging hydrogen balloon, heating to 50 deg.CThe reaction was stirred overnight, after completion of the TLC monitoring, the catalyst was removed by filtration, and the filtrate was concentrated and purified by silica gel column chromatography to give 6.51g of a pale yellow oily liquid with yield: 96.3 percent. LC-MS (APCI) M/z =283.4 (M + 1) +
Preparation of intermediate B2-chloro-4- (pyridin-2-ylmethoxy) aniline
Figure BDA0002983446160000463
The following synthetic route was used:
Figure BDA0002983446160000464
step 1 Synthesis of 2- ((2-chloro-4-nitrophenoxy) methyl) pyridine
Adding 2-chloro-4-nitrophenol (5.2g, 30mmol), 2-chloromethylpyridine hydrochloride (5.42g, 33mmol) and anhydrous sodium carbonate (7.95g, 75mmol) into a reaction bottle, adding 20ml of anhydrous DMF for dissolving, heating to 80 ℃, stirring for reacting for 4 hours, cooling to room temperature after TLC monitoring reaction, adding excessive water for dilution, extracting with ethyl acetate for 3-4 times, combining organic phases, washing with saturated saline solution, concentrating, and purifying by silica gel column chromatography to obtain 4.5g of a product, wherein the yield is as follows: 56.8 percent. LC-MS (APCI) M/z =265.7 (M + 1) +
Step 2 Synthesis of 3-chloro-4- (pyridin-2-ylmethoxy) aniline (intermediate B2)
Adding 2- ((2-chloro-4-nitrophenoxy) methyl) pyridine (4.5 g, 17mmol) and catalytic amount of palladium carbon into a reaction bottle, adding 50ml of ethanol for dissolving, filling hydrogen balloon, stirring at room temperature for reacting overnight, filtering to remove the catalyst after TLC monitoring reaction is finished, concentrating the filtrate, and purifying by silica gel column chromatography to obtain 3.76g of light yellow oily liquid, wherein the yield is as follows: 94.1 percent. LC-MS (APCI) M/z =235.4 (M + 1) +
Preparation of intermediate B3- ((4-bromo-2-chlorophenoxy) methyl) pyridine
Figure BDA0002983446160000471
The following synthetic route was used:
Figure BDA0002983446160000472
4-bromo-2-chlorophenol (3.4g, 16.4mmol) and 2-chloromethylpyridine hydrochloride (3.49g, 21.3mmol) were dissolved in 30mL of DMSO, triethylamine (5.05g, 50mmol) was added, and the reaction was heated at 80 ℃ for 3 hours. The reaction mixture was diluted with 100mL of water, extracted with ethyl acetate (50ml × 3), the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and separated by silica gel column to give 4g of pale yellow solid with a yield of 82%. ESI-MS of 300[ m ] + +2].
Intermediate B4- ((2-chloro-4- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl) phenoxy) Preparation of Yl) methyl) pyridine
Figure BDA0002983446160000473
The following synthetic route was used:
Figure BDA0002983446160000474
2- ((4-bromo-2-chlorophenoxy) methyl) pyridine (3.98g, 13.3mmol), pinacol ester diboron (4.06g, 169mol), pd (dppf) Cl 2 (0.49g, 0.67mmol) and potassium acetate (3.92g, 40mmol) were added to 60mL dioxane, replaced with nitrogen three times, and the temperature was raised to 90 ℃ for reaction for 3 hours. The reaction mixture was cooled to room temperature, 100mL of water was added, extraction was performed with ethyl acetate (60ml × 3), the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and separated by a silica gel column to obtain 3.45g of a pale yellow solid with a yield of 75%. ESI-MS:346[ m ] + +1].
Example 1- (1-Acrylopiperidin-3-yl) -4-amino-1- (3-chloro-4- (pyridin-2-ylmethoxy) benzene Preparation of 1H-pyrazole-5-carbonitrile
Figure BDA0002983446160000475
The following synthetic route was used:
Figure BDA0002983446160000481
step 1 Synthesis of tert-butyl 3- (1- (2- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) hydrazono) -2-methoxy-2-oxoethyl) piperidine-1-carboxylate
Adding the intermediate B2 (468mg, 2mmol) into a reaction bottle, adding 1N hydrochloric acid aqueous solution (5ml, 5mmol) for dissolving, adding sodium nitrite (230mg, 3.33mmol), and stirring at room temperature for half an hour to obtain a solution A; adding the intermediate B1 (470mg, 1.67mmol) into another reaction bottle, adding 10ml of ethanol for dissolving, adding 5ml of saturated ammonium chloride aqueous solution under ice bath, then slowly dropwise adding the obtained solution A, stirring at room temperature for reacting overnight, after TLC monitoring reaction, adding a small amount of water for diluting, extracting with ethyl acetate for 3-4 times, combining organic phases, washing with saturated ammonium chloride and saturated saline water once, and purifying by silica gel column chromatography after concentration to obtain 340mg of oily liquid, wherein the yield is as follows: 40.6 percent. LC-MS (APCI) M/z =503.1 (M + 1) +
Step 2 Synthesis of 2- (1- (tert-Butoxycarbonyl) piperidin-3-yl) -2- (2- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) hydrazono) acetic acid
Adding 3- (1- (2- (3-chloro-4- (pyridine-2-ylmethoxy) phenyl) hydrazono) -2-methoxy-2-oxoethyl) piperidine-1-carboxylic acid tert-butyl ester (340mg, 0.68mmol) into a reaction bottle, adding 15ml tetrahydrofuran for dissolving, adding 3ml aqueous solution of sodium hydroxide (1.2g, 30mmol), stirring at room temperature for reaction overnight, after TLC monitoring reaction, adding 1N diluted hydrochloric acid to adjust pH to acidity, extracting with ethyl acetate for 3-4 times, combining organic phases, washing with saturated saline, drying with anhydrous sodium sulfate, filtering, concentrating to obtain 310mg of product, and directly putting into the next reaction without purification.
Step 3 Synthesis of tert-butyl 3- (2-amino-1- (2- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) hydrazono) -2-oxoethyl) piperidine-1-carboxylate
2- (1- (tert-butoxycarbonyl) piperidin-3-yl) -2- (2- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) hydrazono) acetic acid (310mg, 0.63mmol), EDCI (243mg, 1.26mmol) and HOBT (171mg, 1.26mmol) were charged into a reaction flask, dissolved by 10ml of dichloromethane, added with ammonia (0.05ml, 1.26mmol) and DIPEA (0.31ml, 1.89mmol) under nitrogen protection, stirred at room temperature for 2 hours, after completion of TLC monitoring reaction, diluted by a small amount of dichloromethane, washed with 1N diluted hydrochloric acid, saturated sodium bicarbonate solution and saturated brine in order, and purified by silica gel column chromatography after concentration to give 301mg of the product in yield: 98 percent. LC-MS (APCI) M/z =488.5 (M + 1) +
Step 4 Synthesis of tert-butyl 3- (1-cyano-1- (2- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) hydrazono) methyl) piperidine-1-carboxylate
Adding 3- (2-amino-1- (2- (3-chloro-4- (pyridine-2-methoxy) phenyl) hydrazono) -2-oxoethyl) piperidine-1-carboxylic acid tert-butyl ester (392mg, 0.8mmol) and pyridine (0.5ml, 5.6 mmol) into a reaction bottle, adding 10ml dichloromethane for dissolving, cooling to 0 ℃ under the protection of nitrogen, slowly dropwise adding trifluoroacetic anhydride (0.6ml, 4.0mmol), stirring at room temperature after the addition for 1 hour for reaction, monitoring the reaction by TLC, adding a small amount of dichloromethane for dilution, washing with 1N diluted hydrochloric acid, saturated sodium bicarbonate solution and saturated saline solution in turn, drying anhydrous sodium sulfate, concentrating to dryness, and directly putting into the next step for reaction without purification.
Step 5 Synthesis of tert-butyl 3- (4-amino-5-cyano-1- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) -1H-pyrazol-3-yl) piperidine-1-carboxylate
Directly dissolving the intermediate obtained in the previous step in 15ml of tert-butyl alcohol, adding bromoacetonitrile (0.11ml, 1.61mmol), adding potassium tert-butoxide (270mg, 2.41mmol) in batches under the protection of nitrogen, stirring at room temperature for 2 hours, and adding small amount of sodium benzoate after TLC monitoring reactionQuenching the reaction with water, extracting with ethyl acetate for 3-4 times, combining organic phases, washing with saturated brine, concentrating, and purifying by silica gel column chromatography to obtain 250mg of light yellow solid with yield: 61.1 percent. LC-MS (APCI) M/z =509.7 (M + 1) +
Step 6 Synthesis of 3- (1-acryloylpiperidin-3-yl) -4-amino-1- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) -1H-pyrazole-5-carbonitrile
To a reaction flask, tert-butyl 3- (4-amino-5-cyano-1- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) -1H-pyrazol-3-yl) piperidine-1-carboxylate (120mg, 0.24mmol) was added, a 4N ethyl hydrogen chloride acetate solution (2.5 ml, 10mmol) was added under nitrogen protection, the reaction was stirred at room temperature for 1h, and after completion of the reaction, the mixture was concentrated to dryness and directly fed to the next step.
Adding 5ml of anhydrous dichloromethane and triethylamine (0.07ml, 0.47mmol) into the intermediate, dropwise adding acryloyl chloride (22.4 mg, 0.25mmol) under ice bath, stirring for reaction for 10 minutes after the addition is finished, monitoring the reaction completion by TLC, adding dichloromethane for dilution, washing with water and saturated saline in sequence, and purifying by silica gel column chromatography after concentration to obtain 80mg of off-white solid with yield: 73.4 percent. LC-MS (APCI) M/z =463.5 (M + 1) +1 H NMR(400MHz,DMSO-d 6 )δ8.53(d,J=2.3Hz,1H),8.13(t,J=4.6Hz,1H),7.95(s,1H),7.72(d,J=4.6Hz,1H),7.41(d,J=3.4Hz,1H),7.32(t,J=4.6Hz,1H),6.88(d,J=3.4Hz,1H),6.63(m,1H),6.12(d,J=6.8Hz,1H),6.04(s,2H),5.63(d,J=6.8Hz,1H),5.51(s,2H),3.81(m,1H),3.66(m,3H),3.04(m,1H),2.01(m,2H),1.66(m,2H).
Example 2- (1-Acrylopiperidin-3-yl) -4-amino-1- (3-chloro-4- (pyridin-2-ylmethoxy) benzene Preparation of 1H-pyrrole-5-carboxamides
Figure BDA0002983446160000501
The following synthetic route was used:
Figure BDA0002983446160000502
step 1 Synthesis of tert-butyl 3- (4-amino-5-carbamoyl-1- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) -1H-pyrazol-3-yl) piperidine-1-carboxylate
Adding 3- (4-amino-5-cyano-1- (3-chloro-4- (pyridine-2-ylmethoxy) phenyl) -1H-pyrazole-3-yl) piperidine-1-carboxylic acid tert-butyl ester (150mg, 0.29mmol) into a reaction bottle, adding 5ml of methanol for dissolving, adding 3ml of aqueous solution of sodium hydroxide (240mg, 6 mmol), dropwise adding 30% hydrogen peroxide (1 ml) under ice bath, heating to room temperature, stirring for reaction for 3 hours, adding ethyl acetate for extraction for 3-4 times after TLC monitoring reaction, combining organic phases, washing with saturated saline, concentrating, and purifying by silica gel column chromatography to obtain 113mg of target product, wherein the yield is as follows: 74.1 percent. LC-MS (APCI) M/z =527.9 (M + 1) +
Step 2 Synthesis of 3- (1-acryloylpiperidin-3-yl) -4-amino-1- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) -1H-pyrazole-5-carboxamide
To a reaction flask, tert-butyl 3- (4-amino-5-carbamoyl-1- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) -1H-pyrazol-3-yl) piperidine-1-carboxylate (113mg, 0.22mmol) was added, a 4N ethyl hydrogen chloride solution (2.5 ml, 10mmol) was added under nitrogen protection, the reaction was stirred at room temperature for 1H, and after completion of the tlc monitoring, the mixture was concentrated to dryness and directly charged to the next step.
Adding 5ml of anhydrous dichloromethane and triethylamine (0.06ml, 0.43mmol) into the intermediate, dropwise adding acryloyl chloride (20.4 mg, 0.23mmol) under ice bath, stirring for 10 minutes after the addition is finished, monitoring the reaction by TLC, adding dichloromethane for dilution, washing with water and saturated saline in sequence, concentrating, and purifying by silica gel column chromatography to obtain 82mg of off-white solid with yield: 79.5 percent. LC-MS (APCI) M/z =481.3 (M + 1) +1 H NMR(400MHz,DMSO-d 6 )δ10.47(s,2H),8.33(d,J=2.4Hz,1H),8.10(t,J=4.6Hz,1H),7.75(s,1H),7.70(d,J=4.6Hz,1H),7.41(d,J=3.6Hz,1H),7.30(t,J=4.6Hz,1H),6.81(d,J=3.4Hz,1H),6.63(m,1H),6.12(d,J=6.8Hz,1H),6.00(s,2H),5.63(d,J=6.8Hz,1H),5.52(s,2H),3.81(m,1H),3.66(m,3H),3.04(m,1H),1.98(m,1H),1.75(m,3H).
Example 3- (7-amino-1- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) -1H-pyrazolo [4,3- d]Preparation of pyrimidin-3-yl) -5,6-dihydropyridin-1 (2H) -yl) prop-2-en-1-one
Figure BDA0002983446160000511
The following synthetic route was used:
Figure BDA0002983446160000512
step 1 Synthesis of tert-butyl 3- (7-amino-1- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) -1H-pyrazolo [4,3-d ] pyrimidin-3-yl) -5,6-dihydropyridine-1 (2H) -carboxylate
Adding the intermediate A1 (60mg, 0.2mmol), the intermediate B3 (120mg, 0.4mmol), N-dimethylglycine (13mg, 0.12mmol) and cuprous iodide (8mg, 0.04mmol) into a reaction bottle, adding 3ml of anhydrous DMF under the protection of nitrogen, adding DBU (92mg, 0.6mmol), heating to 110 ℃ after adding, stirring for reaction for 20 hours, adding saturated ammonium chloride after TLC monitoring reaction, quenching reaction, extracting with ethyl acetate for 3-4 times, combining organic phases, washing with saturated salt water, concentrating, purifying by silica gel column chromatography to obtain 74mg of product, yield: 69.4 percent. LC-MS (APCI) M/z =534.7 (M + 1) +
Step 2 Synthesis of 1- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) -3- (1,2,5,6-tetrahydropyridin-3-yl) -1H-pyrazolo [4,3-d ] pyrimidin-7-amine
3- (7-amino-1- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) -1H-pyrazolo [4,3-d ] pyrimidin-3-yl) -5,6-dihydropyridine-1 (2H) -carboxylic acid tert-butyl ester (74mg, 0.14mmol) was added to a reaction flask, 4N hydrogen chloride dioxane solution (3ml, 12mmol) was added under nitrogen protection, the reaction was stirred at room temperature for 1h, and after the reaction was monitored by TLC, the mixture was concentrated to dryness and directly charged to the next step.
Step 3 Synthesis of 1- (3- (7-amino-1- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) -1H-pyrazolo [4,3-d ] pyrimidin-3-yl) -5,6-dihydropyridin-1 (2H) -yl) prop-2-en-1-one
Adding 5ml of anhydrous dichloromethane and triethylamine (42.5mg, 0.42mmol) into the intermediate, dropwise adding acryloyl chloride (12.7mg, 0.14mmol) under ice bath, stirring after the addition, reacting for 10 minutes, then monitoring the reaction by TLC, adding dichloromethane for dilution, washing with water and saturated saline in turn, concentrating, and purifying by silica gel column chromatography to obtain 41mg of light yellow solid, wherein the yield is as follows: 60.1 percent. LC-MS (APCI) M/z =488.1 (M + 1) +1 H NMR(400MHz,DMSO-d 6 )δ8.77(s,1H),8.64(d,J=3.6Hz,1H),8.23(t,J=4.2Hz,1H),8.05(s,1H),7.72(d,J=4.2Hz,1H),7.41(d,J=3.6Hz,1H),7.32(t,J=4.4Hz,1H),6.89(d,J=5.5Hz,1H),6.63(m,1H),6.21(d,J=7.7Hz,1H),6.11(s,1H),5.63(d,J=7.7Hz,1H),3.81(t,J=4.8Hz,2H),3.55(s,2H),2.07(t,J=4.8Hz,2H).
Example 4- (3- (7-amino-1- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) -1H-pyrazolo [4,3- d]Preparation of pyrimidin-3-yl) piperidin-1-yl) prop-2-en-1-one
Figure BDA0002983446160000521
The following synthetic route was used:
Figure BDA0002983446160000522
step 1 Synthesis of tert-butyl 3- (7-amino-1- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) -1H-pyrazolo [4,3-d ] pyrimidin-3-yl) piperidine-1-carboxylate
Adding intermediate A2 (432mg, 1.36mmol), intermediate B3 (807mg, 2.72mmol), N-dimethylglycine (84mg, 0.82mmol) and cuprous iodide (52mg, 0.27mmol) into a reaction flask, adding 5ml of anhydrous DMF under nitrogen protection, adding DBU (620mg, 4.08mmol), heating to 110 deg.C, stirring for reaction for 15 hr, monitoring by TLC, adding saturated ammonium chloride, quenching, extracting with ethyl acetate3-4 times, combining organic phases, washing with saturated saline solution, concentrating, and purifying by silica gel column chromatography to obtain 290mg of a product with the yield: 39.9 percent. LC-MS (APCI) M/z =536.7 (M + 1) +
Step 2 Synthesis of 1- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) -3- (piperidin-3-yl) -1H-pyrazolo [4,3-d ] pyrimidin-7-amine
3- (7-amino-1- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) -1H-pyrazolo [4,3-d ] pyrimidin-3-yl) piperidine-1-carboxylic acid tert-butyl ester (290mg, 0.54mmol) was added to a reaction flask, 4N hydrogen chloride dioxane solution (5 ml, 20mmol) was added under nitrogen protection, the reaction was stirred at room temperature for 1H, and after the reaction was monitored by TLC, the reaction was concentrated to dryness and directly fed to the next step.
Step 3 Synthesis of 1- (3- (7-amino-1- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) -1H-pyrazolo [4,3-d ] pyrimidin-3-yl) piperidin-1-yl) prop-2-en-1-one
Adding 10ml of anhydrous dichloromethane and triethylamine (189mg, 1.87mmol) into the intermediate, dropwise adding acryloyl chloride (49mg, 0.54mmol) in ice bath, stirring for reaction for 10 minutes after the addition, monitoring the reaction by TLC, adding dichloromethane for dilution, washing with water and saturated saline in turn, concentrating, and purifying by silica gel column chromatography to obtain 120mg of yellow solid, wherein the yield is as follows: 45.5 percent. LC-MS (APCI) M/z =490.6 (M + 1) +1 H NMR(400MHz,DMSO-d 6 )δ8.71(s,1H),8.68(d,J=4.8Hz,1H),8.15(t,J=4.4Hz,1H),7.92(s,1H),7.72(d,J=4.4Hz,1H),7.45(d,J=4.8Hz,1H),7.30(t,J=4.4Hz,1H),6.82(d,J=5.5Hz,1H),6.51(m,1H),6.00(d,J=8.4Hz,1H),5.52(d,J=8.4Hz,1H),5.37(s,2H),3.82(m,1H),3.79(m,3H),2.96(m,1H),2.07(m,1H),1.88(m,3H).
Example 5 (R) -1- (3- (7-amino-1- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) -1H-pyrazolo [4, 3-d]preparation of pyrimidin-3-yl) piperidin-1-yl) prop-2-en-1-one
Figure BDA0002983446160000531
120mg of the racemate of example 4 was dissolved in methanolIn the solution, 43mg of the objective product was isolated under the following chiral preparative column and chiral resolution conditions (retention time: 27.79min, defined as R configuration). LC-MS (APCI) M/z =490.6 (M + 1) +1 H NMR(400MHz,DMSO-d 6 )δ8.69(s,1H),8.52(d,J=4.8Hz,1H),8.13(t,J=4.4Hz,1H),7.92(s,1H),7.72(d,J=4.4Hz,1H),7.41(d,J=4.8Hz,1H),7.32(t,J=4.4Hz,1H),6.82(d,J=5.5Hz,1H),6.51(m,1H),6.01(d,J=8.4Hz,1H),5.52(d,J=8.4Hz,1H),5.37(s,2H),3.82(m,1H),3.79(m,3H),3.11(m,1H),2.07(m,1H),1.88(m,3H).
Chiral preparative chromatographic column: CHIRALPAK IC (trade name), 10mm × 250mm (inner diameter × length), 5 μm (filler particle size)
Column temperature: 30 deg.C
Flow rate: 4.0mL/min
Ultraviolet detection wavelength: 254nm
Mobile phase: dichloromethane: n-hexane: ethanol =70
Example 6 (S) -1- (3- (7-amino-1- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) -1H-pyrazolo [4, 3-d]preparation of pyrimidin-3-yl) piperidin-1-yl) prop-2-en-1-one
Figure BDA0002983446160000541
120mg of the racemic compound of example 4 was dissolved in a methanol solution, and separation was carried out under the following chiral preparative column and chiral resolution conditions to obtain 45mg of the objective product (retention time: 22.48min, defined as S configuration). LC-MS (APCI) M/z =490.6 (M + 1) +1 H NMR(400MHz,DMSO-d 6 )δ8.71(s,1H),8.69(d,J=4.8Hz,1H),8.16(t,J=4.4Hz,1H),7.94(s,1H),7.72(d,J=4.4Hz,1H),7.45(d,J=4.8Hz,1H),7.31(t,J=4.4Hz,1H),6.82(d,J=5.5Hz,1H),6.51(m,1H),6.00(d,J=8.4Hz,1H),5.52(d,J=8.4Hz,1H),5.37(s,2H),3.82(m,1H),3.79(m,3H),2.86(m,1H),2.17(m,1H),1.89(m,3H)
Chiral preparative chromatography column: CHIRALPAK IC (trade name), 10mm × 250mm (inner diameter × length), 5 μm (filler particle size)
Column temperature: 30 deg.C
Flow rate: 4.0mL/min
Ultraviolet detection wavelength: 254nm
Mobile phase: dichloromethane: n-hexane: ethanol =70
Example 7 (R) -1- (3- (8-amino-1- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) imidazo [1,5-a] Preparation of pyrazin-3-yl) piperidin-1-yl) prop-2-en-1-one
Figure BDA0002983446160000542
The following synthetic route was used:
Figure BDA0002983446160000551
step 1 Synthesis of (R) -3- (8-amino-1- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) imidazo [1,5-alpha ] pyrazin-3-yl) piperidine-1-carboxylic acid tert-butyl ester
Adding the intermediate A3 (220mg, 0.5 mmol), the intermediate B4 (207mg, 0.6 mmol), the tetrakis (triphenylphosphine) palladium (30mg, 0.025mmol) and the anhydrous sodium carbonate (106mg, 1.0 mmol) into a reaction bottle, adding 20ml of ethylene glycol dimethyl ether and 5ml of water under the protection of nitrogen, reacting overnight at 90 ℃, cooling to room temperature, after TLC monitoring reaction, adding excessive water to quench reaction, extracting for 3-4 times by ethyl acetate, combining organic phases, washing by saturated salt water, concentrating, purifying by silica gel column chromatography to obtain 170mg of yellow solid, and obtaining the yield: and (3.7). LC-MS (APCI) M/z =535.7 (M + 1) +
Step 2 Synthesis of (R) -1- (3- (8-amino-1- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) imidazo [1,5-a ] pyrazin-3-yl) piperidin-1-yl) prop-2-en-1-one
(R) -3- (8-amino-1- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) imidazo [1,5- α ] pyrazin-3-yl) piperidine-1-carboxylic acid tert-butyl ester (132mg, 0.25mmol) was added to a reaction flask, 4N ethyl hydrogen chloride solution (5 ml, 20mmol) was added thereto, the reaction was stirred at room temperature for 1h, and the reaction was monitored by TLC for completion, concentrated to dryness, and directly charged to the next reaction.
Adding 8ml of anhydrous dichloromethane into a reaction bottle in the previous step, adding triethylamine (0.1ml, 0.5mmol), cooling to 0 ℃ under the protection of nitrogen, dropwise adding acryloyl chloride (24.5mg, 0.27mmol), stirring for reaction for 0.5h after the addition is finished, adding a saturated ammonium chloride aqueous solution to quench the reaction after the TLC monitoring reaction is finished, extracting the aqueous phase for 2-3 times by dichloromethane, combining the organic phases, washing by saturated saline solution, and purifying by silica gel column chromatography after concentration to obtain 66mg of off-white solid, wherein the yield is as follows: 54.8 percent. LC-MS (APCI) M/z =489.5 (M + 1) +1 H NMR(400MHz,DMSO-d 6 )δ8.71(d,J=2.3Hz,1H),8.13(m,3H),7.82(m,2H),7.57(d,J=4.6Hz,1H),7.34(t,J=2.3Hz,1H),7.18(d,J=4.6Hz,1H),6.74(q,J=6.8Hz,1H),6.15(d,J=5.5Hz,1H),5.64(d,J=5.5Hz,1H),5.58(s,2H),4.01(m,1H),3.81(m,2H),3.72(t,J=7.4Hz,2H),3.02(m,1H),2.87(m,2H),2.15(m,2H).
Example 8- (3- (4-amino-5- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) pyrrolo [2,1-f)][1, 2,4]Preparation of triazin-7-yl) piperidin-1-yl) prop-2-en-1-one
Figure BDA0002983446160000561
The following synthetic route was used:
Figure BDA0002983446160000562
step 1 Synthesis of tert-butyl 3- (4-amino-5- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) pyrrolo [2,1-f ] [1,2,4] triazin-7-yl) piperidine-1-carboxylate
Intermediate A4 (645mg, 1.63mmol), intermediate B4 (677mg, 1.96mmol), pd (dppf) Cl 2 (58mg, 0.08mmol) and Na 2 CO 3 (520mg, 4.9 mmol) was added to 12mL of DME and 4mL of water, and the mixture was purged with nitrogen three times, and allowed to warm to 90 ℃ for reaction overnight. Reacting the reaction solutionAfter cooling to room temperature, 30mL of water was added, extraction was performed with ethyl acetate (20mL × 3), and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and separated by a silica gel column to obtain 540mg of a pale yellow solid with a yield of 62%. ESI-MS:536[ m ] +1] + .
Step 2 Synthesis of 5- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) -7- (piperidin-3-yl) pyrrolo [2,1-f ] [1,2,4] triazin-4-amine
Reacting 3- (4-amino-5- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) pyrrolo [2,1-f][1,2,4]Triazin-7-yl) piperidine-1-carboxylic acid tert-butyl ester (535mg, 1mmol) was dissolved in 10mL of dichloromethane, 3mL of trifluoroacetic acid was added, and the mixture was stirred at room temperature for 1 hour. The solvent was removed by rotary evaporation, the residue was dissolved in 30mL of dichloromethane, washed with saturated sodium bicarbonate solution and saturated brine, respectively, and the organic phase was dried over anhydrous sodium sulfate, concentrated, and separated by silica gel column to give 322mg of a pale yellow solid with a yield of 74%. ESI-MS:436[ m ] +1] + .
Step 3 Synthesis of 1- (3- (4-amino-5- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) pyrrolo [2,1-f ] [1,2,4] triazin-7-yl) piperidin-1-yl) prop-2-en-1-one
Reacting 5- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) -7- (piperidin-3-yl) pyrrolo [2,1-f][1,2,4]Triazine-4-amine (322mg, 0.74mmol) and triethylamine (0.15g, 1.48mmol) were dissolved in 10mL of dichloromethane, the temperature was reduced to-20 ℃ in an ice bath, acryloyl chloride (67mg, 0.74mmol) was slowly added thereto, the ice bath was removed after the dropwise addition, and the mixture was stirred at room temperature for 1 hour. Diluting with 20mL of water, extracting with dichloromethane (15mL. Multidot.3), washing the organic phase with saturated brine, drying over anhydrous sodium sulfate, concentrating, and separating with silica gel column to obtain 240mg of pale yellow solid with a yield of 65%. ESI-MS:500[ deg. ] M ] +1] + . 1 H NMR(400MHz,CDCl 3 )δ8.61(dt,J=4.8,1.5Hz,1H),7.94(s,1H),7.78(td,J=7.7,1.8Hz,1H),7.67(d,J=7.9Hz,1H),7.52(d,J=2.1Hz,1H),7.30–7.28(m,1H),7.07(d,J=8.4Hz,1H),6.69(dd,J=16.7,10.5Hz,1H),6.46(s,1H),6.29(d,J=16.9Hz,1H),5.67(d,J=10.1Hz,1H),5.37(s,1H),5.33(s,2H),4.86(d,J=13.1Hz,0.5H),4.58(d,J=13.3Hz,0.5H),4.39(d,J=13.1Hz,0.5H),4.00(d,J=13.2Hz,0.5H),3.44(s,1H),3.15(q,J=13.0,11.0Hz,1H),2.99(t,J=11.8Hz,0.5H),2.87(t,J=11.9Hz,0.5H),2.25(d,J=10.3Hz,1H),1.90(d,J=12.1Hz,2H),1.78–1.71(m,1H).
Example 9 (R) -1- (3- (4-amino-5- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) pyrrolo [2,1-f] [1,2,4]Preparation of triazin-7-yl) piperidin-1-yl) prop-2-en-1-one
Figure BDA0002983446160000571
100mg of the racemic compound of example 8 was dissolved in a methanol solution and separated under chiral preparative chromatography and chiral resolution conditions described below to give the desired product (retention time: 61.925min, relative content: 50.04%, named R configuration). ESI-MS:500[ deg. ] M ] +1] + . 1 H NMR(400MHz,CDCl 3 )δ8.61(dt,J=4.8,1.5Hz,1H),7.94(s,1H),7.78(td,J=7.7,1.8Hz,1H),7.67(d,J=7.9Hz,1H),7.52(d,J=2.1Hz,1H),7.30–7.28(m,1H),7.07(d,J=8.4Hz,1H),6.69(dd,J=16.7,10.5Hz,1H),6.46(s,1H),6.29(d,J=16.9Hz,1H),5.67(d,J=10.1Hz,1H),5.37(s,1H),5.33(s,2H),4.86(d,J=13.1Hz,0.5H),4.58(d,J=13.3Hz,0.5H),4.39(d,J=13.1Hz,0.5H),4.00(d,J=13.2Hz,0.5H),3.44(s,1H),3.15(q,J=13.0,11.0Hz,1H),2.99(t,J=11.8Hz,0.5H),2.87(t,J=11.9Hz,0.5H),2.25(d,J=10.3Hz,1H),1.90(d,J=12.1Hz,2H),1.78–1.71(m,1H).
Chiral preparative chromatographic column: CHIRALPAK IC (trade name), 10mm × 250mm (inner diameter × length), 5 μm (filler particle size)
Column temperature: 30 deg.C
Flow rate: 1.0mL/min
Ultraviolet detection wavelength: 254nm
Mobile phase: methyl tert-butyl ether n-hexane methanol isopropanol =20
Example 10 (S) -1- (3- (4-amino-5- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) pyrrolo [2,1- f][1,2,4]Preparation of triazin-7-yl) piperidin-1-yl) prop-2-en-1-one
Figure BDA0002983446160000581
100mg of the racemic compound of example 8 was dissolved in a methanol solution and separated under chiral preparative chromatography and chiral resolution conditions described below to give 43mg of the desired product (retention time: 65.193min, relative content: 49.96%, named S configuration). ESI-MS:500[ deg. ] M ] +1] + . 1 H NMR(400MHz,CDCl 3 )δ8.61(dt,J=4.8,1.5Hz,1H),7.94(s,1H),7.78(td,J=7.7,1.8Hz,1H),7.67(d,J=7.9Hz,1H),7.52(d,J=2.1Hz,1H),7.30–7.28(m,1H),7.07(d,J=8.4Hz,1H),6.69(dd,J=16.7,10.5Hz,1H),6.46(s,1H),6.29(d,J=16.9Hz,1H),5.67(d,J=10.1Hz,1H),5.37(s,1H),5.33(s,2H),4.86(d,J=13.1Hz,0.5H),4.58(d,J=13.3Hz,0.5H),4.39(d,J=13.1Hz,0.5H),4.00(d,J=13.2Hz,0.5H),3.44(s,1H),3.15(q,J=13.0,11.0Hz,1H),2.99(t,J=11.8Hz,0.5H),2.87(t,J=11.9Hz,0.5H),2.25(d,J=10.3Hz,1H),1.90(d,J=12.1Hz,2H),1.78–1.71(m,1H).
Chiral preparative chromatographic column: CHIRALPAK IC (trade name), 10mm × 250mm (inner diameter × length), 5 μm (filler particle size)
Column temperature: 30 deg.C
Flow rate: 1.0mL/min
Ultraviolet detection wavelength: 254nm
Mobile phase: methyl tert-butyl ether n-hexane methanol isopropanol =20
Example 11 (R) -1- (3- (4-amino-5- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) imidazo [5,1- f][1,2,4]Preparation of triazin-7-yl) piperidin-1-yl) prop-2-en-1-one
Figure BDA0002983446160000582
The following synthetic route was used:
Figure BDA0002983446160000591
step 1 Synthesis of benzyl 3- (4-amino-5- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) imidazo [5,1-f ] [1,2,4] triazin-7-yl) piperidine-1-carboxylate
Intermediate A5 (330mg, 0.76mmol), intermediate B4 (342mg, 0.99mmol), pd (dppf) Cl 2 (30mg, 0.04mmol) and Na 2 CO 3 (244mg, 2.3mmol) was added to 12mL of DME and 4mL of water, replaced with nitrogen three times, and the reaction was allowed to warm to 90 ℃ overnight. The reaction mixture was cooled to room temperature, 30mL of water was added, extraction was performed with ethyl acetate (20ml × 3), and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and separated by a silica gel column to obtain 285mg of a pale yellow solid with a yield of 66%. ESI-MS:571 2 [ M +1 ]] + .
Step 2 Synthesis of (R) -5- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) -7- (piperidin-3-yl) imidazo [5,1-f ] [1,2,4] triazin-4-amine
Reacting (R) -3- (4-amino-5- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) imidazo [5,1-f][1,2,4]Triazin-7-yl) piperidine-1-carboxylic acid benzyl ester (285mg, 0.5mmol) was dissolved in 15mL of anhydrous ethanol, 30mg of 10% palladium on carbon was added, replaced with hydrogen three times, and stirred under a hydrogen atmosphere of one atmosphere overnight. After the reaction is completed, palladium-carbon is filtered, the filtrate is concentrated, and the light yellow oily substance of 180mg is obtained by silica gel column separation, with the yield of 82%. ESI-MS:437[ 2 [ M ] + +1].
Step 3 Synthesis of (R) -1- (3- (4-amino-5- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) imidazo [5,1-f ] [1,2,4] triazin-7-yl) piperidin-1-yl) prop-2-en-1-one
Mixing (R) -5- (3-chloro-4- (pyridine-2-yl methoxy) phenyl) -7- (piperidine-3-yl) imidazo [5,1-f][1,2,4]Triazine-4-amine (180mg, 0.41mmol) and triethylamine (83mg, 0.82mmol) were dissolved in 10mL of dichloromethane, cooled to-20 ℃ in an ice bath, acryloyl chloride (37mg, 0.41mmol) was slowly added, the ice bath was removed after the addition, and the mixture was stirred at room temperature for 1 hour. Diluting with 20mL of water, extracting with dichloromethane (10mL × 3), washing the organic phase with saturated brine, drying over anhydrous sodium sulfate, concentrating, and separating with silica gel column to obtain light yellow solid 120mg with yield of 60%. ESI-MS:491[ M ]) + +1] + . 1 H NMR(400MHz,CDCl 3 )δ8.59(ddd,J=4.9,1.8,1.0Hz,1H),7.87(d,J=12.4Hz,1H),7.80–7.70(m,2H),7.63(dt,J=7.9,1.1Hz,1H),7.44(dd,J=8.4,2.2Hz,1H),7.28–7.22(m,1H),7.10(d,J=8.5Hz,1H),6.62(dd,J=16.8,10.6Hz,1H),6.27(t,J=14.6Hz,1H),5.67(dd,J=20.3,10.5Hz,1H),5.34(s,2H),4.89(d,J=13.1Hz,0.5H),4.61(d,J=13.1Hz,0.5H),4.27(d,J=12.5Hz,0.5H),4.03(d,J=14.0Hz,0.5H),3.59–3.40(m,2H),3.19(t,J=13.6Hz,0.5H),2.85(t,J=12.5Hz,0.5H),2.25(s,1H),2.04(t,J=6.2Hz,1H),1.92(d,J=13.8Hz,1H),1.69(d,J=14.4Hz,1H).
Example 12- (3- (4-amino-5- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) -7H-pyrrolo [2,3- d]Preparation of pyrimidin-7-yl) piperidin-1-yl) prop-2-en-1-one
Figure BDA0002983446160000601
The following synthetic route was used:
Figure BDA0002983446160000602
step 1 Synthesis of tert-butyl 3- (4-amino-5- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) -7H-pyrrolo [2,3-d ] pyrimidin-7-yl) piperidine-1-carboxylate
A50 mL single neck flask equipped with a magnetic stirrer and condenser was charged with intermediate A6 (350mg, 0.79mmol), intermediate B4 (300mg, 0.87mmol), sodium carbonate (251mg, 2.37mmol), ethylene glycol dimethyl ether (5 mL) and water (5 mL) in that order, tetrakis (triphenylphosphine) palladium (91mg, 0.079 mmol) was added under nitrogen, the temperature was raised to 100 ℃ under nitrogen, and the reaction was allowed to warm with stirring overnight. After cooling to room temperature, water (20 mL) and ethyl acetate (30 mL) were added, the organic phase was separated, the aqueous phase was extracted with ethyl acetate (30mL. Multidot.2), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, concentrated, and the residue was passed through a silica gel column to give 310mg of a yellow solid, 73.38% yield. LC-MS (APCI) M/z =535.2 (M + 1) + . 1 H NMR(500MHz,CDCl 3 )δ(ppm):8.62(d,J=5.0Hz,1H),8.33(s,1H),7.80-7.77(m,1H),7.68(d,J=10.0Hz,1H),7.54(d,J=2.0Hz,1H),7.31(dd,J=9.0Hz,J=2.0Hz,1H),7.29-7.26(m,1H),7.08(d,J=9.0Hz,1H),7.01(s,1H),5.34(s,2H),5.15(br s,2H),4.79-4.74(m,1H),4.28-4.22(m,1H),4.08-4.00(m,1H),3.29-3.24(m,1H),3.00-2.95(m,1H),2.22-2.19(m,1H),2.06-1.99(m,1H),1.77-1.68(m,2H),1.46(s,9H).
Step 2 Synthesis of (R) -5- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) -7- (piperidin-3-yl) -7H-pyrrolo [2,3-d ] pyrimidin-4-amino
To a 25mL single-necked flask equipped with magnetic stirring was added (R) -3- (4-amino-5- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) -7H-pyrrolo [2,3-d]Pyrimidin-7-yl) piperidine-1-carboxylic acid tert-butyl ester (310mg, 0.58mmol) and dichloromethane (5 mL), TFA (1 mL) was added and the reaction was stirred at room temperature under nitrogen for 2 hours. Concentrated to dryness under reduced pressure, dichloromethane (10 mL) and saturated aqueous sodium bicarbonate solution (10 mL) were added, the organic phase was separated, the aqueous phase was extracted with dichloromethane (20ml × 2), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and concentrated to dryness to give 252mg of a brown solid in 99.2% yield. LC-MS (APCI) M/z =435.2 (M + 1) + .
Step 3 Synthesis of 1- (3- (4-amino-5- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) -7H-pyrrolo [2,3-d ] pyrimidin-7-yl) piperidin-1-yl) prop-2-en-1-one
(R) -5- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) -7- (piperidin-3-yl) -7H-pyrrolo [2,3-d]Pyrimidine-4-amino (252mg, 0.57mmol) was dissolved in acetonitrile (6 mL) and water (5 mL), triethylamine (116mg, 1.15mmol) was added, cooled to 0 deg.C, a solution of acryloyl chloride (78mg, 0.86mmol) in acetonitrile (1 mL) was slowly added dropwise under nitrogen, and after dropwise addition, the reaction was stirred at 0 deg.C for 1 hour. Saturated aqueous sodium bicarbonate (10 mL) and ethyl acetate (20 mL) were added and stirred for 5 minutes to separate the organic phase, the aqueous phase was extracted with ethyl acetate (15ml × 2), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, concentrated and passed through a silica gel column to give 120mg of a pale yellow solid with a yield of 42.69%. LC-MS (APCI) M/z =489.2 (M + 1) + . 1 H NMR(400MHz,CDCl 3 )δ(ppm):8.63(d,J=5.2Hz,1H),8.34(s,1H),7.82-7.78(m,1H),7.69(d,J=8.0Hz,1H),7.55(d,J=2.0Hz,1H),7.33-7.27(m,2H),7.09(d,J=8.8Hz,1H),7.01(s,1H),6.67-6.60(m,1H),6.33(d,J=16.8Hz,1H),5.78-5.50(m,1H),5.35(s,2H),5.14(br s,2H),4.85-4.61(m,2H),4.35-4.02(m,1H),3.41-2.85(m,2H),2.30-2.14(m,2H),1.98-1.82(m,2H).
Example 13- (3- (4-amino-5- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) -5H-pyrrolo [3,2- d]Preparation of pyrimidin-7-yl) piperidin-1-yl) prop-2-en-1-one
Figure BDA0002983446160000611
The following synthetic route was used:
Figure BDA0002983446160000621
step 1 Synthesis of (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) boronic acid
A250 mL single-neck flask equipped with magnetic stirring was charged with intermediate B4 (3.0g, 8.7mmol), acetone (60 mL) and water (60 mL) in this order, the resulting mixture was stirred to dissolve, ammonium acetate (3.36g, 43.5 mmol) and sodium periodate (7.92g, 34.77mmol) were added in this order, and the reaction mixture was stirred at room temperature under nitrogen atmosphere for 3 hours. Acetone was evaporated under reduced pressure, extracted with ethyl acetate (50 mL x 3), the organic phases were combined, washed with saturated brine (60 mL), dried over colorless sodium sulfate, filtered, concentrated and passed through a silica gel column to give 2.1g of a white solid with a yield of 91.8%. LC-MS (APCI) M/z =264.1 (M + 1) + .
Step 2 Synthesis of tert-butyl 3- (1- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) -5-cyano-4- (1,3-dioxoisoindol-2-yl) -1H-pyrrol-3-yl) piperidine-1-carboxylate
To a 50mL three-necked flask equipped with magnetic stirring, tert-butyl 3- (5-cyano-4- (1,3-dioxoisoindol-2-yl) -1H-pyrrol-3-yl) piperidine-1-carboxylate (1.26g, 3.0 mmol), (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) boronic acid (1.58g, 6.0 mmol), and copper acetate (546 mg,3.0 mmol) were added, evacuated and replaced with an oxygen atmosphere three times, and anhydrous dichloromethane (40 mL) and triethylamine (909mg, 9.0 mmol) were added dropwise, and the reaction was stirred at room temperature under an oxygen atmosphere overnight after completion of the addition. Water (40 mL) and dichloromethane (4) were added0 mL), stirred for 10 minutes, the organic phase was separated, the aqueous phase was extracted with dichloromethane (20 mL x 2), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, concentrated and passed through a silica gel column to give 1.0g of a white solid with a yield of 52.3%. LC-MS (APCI) M/z =638.2 (M + 1) + . 1 H NMR(300MHz,CDCl 3 )δ(ppm):8.62-8.61(m,1H),8.01-7.98(m,2H),7.86-7.81(m,2H),7.79-7.76(m,1H),7.58(d,J=2.7Hz,1H),7.40-7.36(m,1H),7.30-7.28(m,1H),7.10(d,J=9.0Hz,1H),6.99(s,1H),5.34(s,2H),4.06-4.00(m,1H),2.85-2.80(m,2H),2.56-2.53(m,1H),2.05-1.97(m,1H),1.71-1.67(m,1H),1.54(s,9H),1.51-1.42(m,3H).
Step 3 Synthesis of tert-butyl 3- (4-amino-1- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) -5-cyano-1H-pyrrol-3-yl) piperidine-1-carboxylate
To a 50mL single neck flask equipped with magnetic stirring was added 3- (1- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) -5-cyano-4- (1,3-dioxoisoindol-2-yl) -1H-pyrrol-3-yl) piperidine-1-carboxylic acid tert-butyl ester (0.58g, 0.91mmol) and absolute ethanol (10 mL), hydrazine hydrate (0.14g, 2.92mmol) was added dropwise, and the reaction was stirred at room temperature under nitrogen for half an hour. The solvent was distilled off under reduced pressure, and the residue was passed through a silica gel column to give 350mg of a white solid in a yield of 65.9%. LC-MS (APCI) M/z =508.2 (M + 1) + .
Step Synthesis of tert-butyl 3- (4-amino-5- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) -5H-pyrrolo [3,2-d ] pyrimidin-7-yl) piperidine-1-carboxylate
To a 50mL single-neck flask equipped with a magnetic stirrer and condenser, tert-butyl 3- (4-amino-1- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) -5-cyano-1H-pyrrol-3-yl) piperidine-1-carboxylate (350mg, 0.6 mmol) and absolute ethanol were added, the mixture was stirred to dissolve, formamidine acetate (620mg, 6.0 mmol) was added, and the mixture was refluxed overnight under nitrogen. Cooled to room temperature, the solvent was evaporated under reduced pressure, and the residue was passed through a silica gel column to give 290mg of a white solid in a yield of 90.1%. LC-MS (APCI) M/z =535.2 (M + 1) + .
Step 5 Synthesis of 5- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) -7- (piperidin-3-yl) -5H-pyrrolo [3,2-d ] pyrimidin-4-amine
To a 50mL single-neck flask equipped with magnetic stirring was added 3- (4-amino-5- (3-chloro-4- (pyridin-2-ylmethyl)Oxy) phenyl) -5H-pyrrolo [3,2-d]Pyrimidin-7-yl) piperidine-1-carboxylic acid tert-butyl ester (290mg, 0.54mmol) and dichloromethane (20 mL), trifluoroacetic acid (4 mL) was added and the reaction stirred at room temperature under nitrogen for 2h. Concentrated to dryness under reduced pressure, dichloromethane (30 mL) and saturated aqueous sodium bicarbonate (20 mL) were added, the organic phase was separated, the aqueous phase was extracted with dichloromethane (30 mL x 2), the organic phases were combined, dried over anhydrous sodium sulfate, filtered and concentrated to dryness to give 225mg of a brown solid in 96.9% yield. LC-MS (APCI) M/z =435.2 (M + 1) + .
Step 6 Synthesis of 1- (3- (4-amino-5- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) -5H-pyrrolo [3,2-d ] pyrimidin-7-yl) piperidin-1-yl) prop-2-en-1-one
5- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) -7- (piperidin-3-yl) -5H-pyrrolo [3,2-d]Pyrimidin-4-amine (225mg, 0.52mmol) was dissolved in acetonitrile (8 mL) and water (4 mL), triethylamine (78mg, 0.78mmol) was added, cooled to 0 deg.C, a solution of acryloyl chloride (60mg, 0.67mmol) in acetonitrile (2 mL) was slowly added dropwise under nitrogen, and after dropwise addition, the reaction was stirred at 0 deg.C for 1 hour. Saturated aqueous sodium bicarbonate (30 mL) and ethyl acetate (60 mL) were added, stirred for 5 minutes, the organic phase was separated, the aqueous phase was extracted with ethyl acetate (15 mL x 2), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, concentrated and passed through a silica gel column to give 140mg of a pale yellow solid with a yield of 55.2%. LC-MS (APCI) M/z =489.2 (M + 1) + . 1 H NMR(500MHz,CDCl 3 )δ(ppm):8.62(d,J=5.0Hz,1H),8.32(s,1H),7.81-7.7.77(m,1H),7.71(d,J=2.5Hz,1H),7.67(d,J=8.0Hz,1H),7.54(dd,J=8.5Hz,J=2.5Hz,1H),7.28-7.25(m,1H),7.11(d,J=9.0Hz,1H),6.92(s,1H),6.69-6.64(m,1H),6.45-6.41(m,1H),6.34(br s,2H),5.83-5.80(m,1H),5.35(s,2H),4.81-4.77(m,1H),4.12-4.10(m,1H),3.28-3.22(m,1H),2.55-2.50(m,1H),2.32-2.29(m,1H),2.01-1.98(m,1H),1.94-1.91(m,1H),1.70-1.65(m,1H).
Example 14 (S) -1- (3- (4-amino-5- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) -5H-pyrrolo [3,2-d]PyrimidinesPreparation of (E) -7-yl) piperidin-1-yl) prop-2-en-1-one
Figure BDA0002983446160000641
140mg of the racemic compound of example 13 was dissolved in dichloromethane (10 mL) and separated under chiral preparative chromatography and chiral resolution conditions described below to give 51mg of the desired product (retention time: 25.955min, named S configuration). LC-MS (APCI) M/z =489.2 (M + 1) + . 1 H NMR(500MHz,CDCl 3 )δ(ppm):8.62(d,J=5.0Hz,1H),8.32(s,1H),7.81-7.7.77(m,1H),7.71(d,J=2.5Hz,1H),7.67(d,J=8.0Hz,1H),7.54(dd,J=8.5Hz,J=2.5Hz,1H),7.28-7.25(m,1H),7.11(d,J=9.0Hz,1H),6.92(s,1H),6.69-6.64(m,1H),6.45-6.41(m,1H),6.34(br s,2H),5.83-5.80(m,1H),5.35(s,2H),4.81-4.77(m,1H),4.12-4.10(m,1H),3.28-3.22(m,1H),2.55-2.50(m,1H),2.32-2.29(m,1H),2.01-1.98(m,1H),1.94-1.91(m,1H),1.70-1.65(m,1H).
Chiral preparative chromatography column: CHIRALPAK IC (trade name), 10mm × 250mm (inner diameter × length), 5 μm (filler particle size)
Column temperature: 25 deg.C
Flow rate: 1.0mL/min
Ultraviolet detection wavelength: 254nm
Mobile phase: dichloromethane: n-hexane: methanol: ethanol =45
Example 15 (R) -1- (3- (4-amino-5- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) -5H-pyrrolo [3,2-d]Preparation of pyrimidin-7-yl) piperidin-1-yl) prop-2-en-1-one
Figure BDA0002983446160000642
140mg of the racemic mixture of the compound of example 13 was dissolved in dichloromethane (10 mL) and separated on a chiral preparative column under chiral resolution conditions to give 48mg of the desired product (retention time: 21.520min, designated as R configuration). LC-MS (APCI) M/z =489.2 (M + 1) + . 1 H NMR(500MHz,CDCl 3 )δ(ppm):8.62(d,J=5.0Hz,1H),8.32(s,1H),7.81-7.7.77(m,1H),7.71(d,J=2.5Hz,1H),7.67(d,J=8.0Hz,1H),7.54(dd,J=8.5Hz,J=2.5Hz,1H),7.28-7.25(m,1H),7.11(d,J=9.0Hz,1H),6.92(s,1H),6.69-6.64(m,1H),6.45-6.41(m,1H),6.34(br s,2H),5.83-5.80(m,1H),5.35(s,2H),4.81-4.77(m,1H),4.12-4.10(m,1H),3.28-3.22(m,1H),2.55-2.50(m,1H),2.32-2.29(m,1H),2.01-1.98(m,1H),1.94-1.91(m,1H),1.70-1.65(m,1H).
Chiral preparative chromatographic column: CHIRALPAK IC (trade name), 10mm X250 mm (inner diameter X length), 50 μm (filler particle size)
Column temperature: 25 deg.C
Flow rate: 1.0mL/min
Ultraviolet detection wavelength: 254nm
Mobile phase: dichloromethane: n-hexane: methanol: ethanol =45
Biological activity assay
Biological example 1: kinase inhibition assay
1) EGFR (WT) and EGFR (D770 _ N771 insNPG) kinase Activity inhibition assay
The inhibitory activity of the test drug on EGFR (WT) and EGFR (D770-N771 insNPG) (SignalChem, E-10-132 GG) was determined using ADP-GloTM Kinase Assay kit (Promega, V9102).
The highest concentration of the drug to be detected is 1 mu M, and the drug is diluted by 3 times of gradient and has 12 concentrations. A384 well plate (Perkin Elmer, 6007290) was prepared by adding 0.1. Mu.L of each drug solution at various concentrations to each well, mixing with 5. Mu.L of EGFR (WT) or 5. Mu.L of EGFR (D770 _ N771 insNPG), respectively, and double-plating wells. After incubation for 15min at 25 ℃ the reaction was started by adding 5. Mu.L of substrate and incubated for 60min at 25 ℃. The final reaction concentrations in the system were: 0.5nM EGFR, 10. Mu.M ATP,0.03mg/mL Poly (4 2 10mM, brij35.01%. Then 10. Mu.L ADP Glo reagent was added and incubation continued for 40min at 25 ℃. After adding 20. Mu.L of detection reagent and incubating at 25 ℃ for 40min, reading on an Envision plate reader (Perkin Elmer, 2104) and calculating the inhibition rate of the compound on the enzyme at different concentrations. Data were analyzed using GraphPad Prism 6.0 software, fitted to data using non-linear curve regression to derive dose-response curves, and IC was calculated therefrom 50 The value is obtained.
2) HER2 (WT) and HER2 (A775 _ G776 insyVMA) kinase activity inhibition assay
The inhibitory activity of the test drug on HER2 (WT) and HER2 (A775 _ G776 insYVMA) (SignalChem, E27-13 BG) was determined using an ADP-GloTM Kinase Assay kit (Promega, V9102).
The highest concentration of the drug to be detected is 1 mu M, and the drug is diluted by 3 times of gradient and has 12 concentrations. A384 well plate (Perkin Elmer, 6007290) was prepared by adding 0.1. Mu.L of each concentration of drug solution per well and mixing with 5. Mu.L of HER2 (WT) or 5. Mu.L of HER2 (A775-G776 insYVMA) in duplicate wells. After incubation for 15min at 25 ℃ the reaction was started by adding 5. Mu.L of substrate and incubated for 60min at 25 ℃. The final reaction concentrations in the system were: 20nM HER2, 5. Mu.M ATP,0.03mg/mL Poly (4 2 10mM, brij35, 0.01%. Then 10. Mu.L ADP Glo reagent was added and incubation continued for 40min at 25 ℃. After 20. Mu.L of the detection reagent was added and incubated at 25 ℃ for 40min, the enzyme inhibition rate of the compound at different concentrations was calculated by reading on an Envision microplate reader (Perkin Elmer, 2104). Data were analyzed using GraphPad Prism 6.0 software, fitted to data using non-linear curve regression to derive dose-response curves, and IC was calculated therefrom 50 The value is obtained.
The compounds of the present invention were tested in the above kinase inhibition experiments and found to have potent activity against EGFR (WT), EGFR (D770 _ N771 insNPG) and HER2 (WT), HER2 (A775 _ G776 insYVMA) kinases. The results for representative example compounds are summarized in table 1 below.
TABLE 1
Figure BDA0002983446160000661
* : the control compound was CHMFL-EGFR-202: (R) -1- (3- (4-amino-3- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) piperidin-1-yl) prop-2-en-1-one.
Biological example 2: testing for growth inhibitory Activity against cell lines expressing wild-type and mutant EGFR
1) Test of growth inhibitory Activity of A431 cells, A549 cells, H1975 cells, and HCC827 cells
The a431 cells and the a549 cells are wild-type EGFR cells; h1975 cells are EGFR cells with L858R point mutation and with T790M point mutation; HCC827 cells are mutant EGFR cells with exon 19 deletion.
Adjusting the concentration of A431 (WT EGFR) cells, A549 cells (WT EGFR), H1975 cells (L858R/T790M EGFR) and HCC827 cells (Ex 19 del), adding 50. Mu.L of the cell suspension to a 384-well plate, respectively, at 37 ℃ and 5% CO 2 The culture was carried out overnight. A Tecan D300E program was set up. Adding the medicine by a Tecan D300E instrument, wherein the highest concentration of the medicine to be detected is 10 mu M, diluting by 3 times of gradient, carrying out 10 concentrations, carrying out double-hole dilution, and continuously culturing for 72h. The 384 well plates were taken out and equilibrated at room temperature for 30min, 30. Mu.L of CTG (Promega, G7573) reagent was added to each well, and after the signal had stabilized, the Luminessence values were read on an EnVision (Perkin Elmer 2104). Inhibition (%) = (1-Lum) Drug to be tested /Lum Negative control ) x100, negative control 0.667% DMSO. IC (integrated circuit) 50 XL-fit software was used for the calculation of (1).
The compounds of the present invention were tested in the cytotoxicity test described above, and found to have no inhibitory activity against a431 cells and a549 cells of wild-type EGFR and potent activity and high selectivity against H1975 cells and HCC827 cells of mutant EGFR, and thus it was found that the compounds of the present invention can inhibit mutant EGFR with exon 19 deletion and mutant EGFR of L858R/T790M with high specificity. The results for representative example compounds are summarized in table 2 below.
2) Test of growth inhibitory Activity of Ba/F3 parental, ba/F3 EGFR-D770-N771ins _ SVD, ba/F3EGFR-L858R, ba/F3EGFR-L858R/T790M, ba/F3 EGFR-Del19/T790M and Ba/F3 EGFR-V769_ D770insASV cells
Taking cells in logarithmic growth phase, detecting the cell viability by trypan blue exclusion method, and ensuring the cell viability to be more than 90%. Adjusting the concentration of Ba/F3 parental, ba/F3 EGFR-D770-N771ins _ SVD, ba/F3EGFR-L858R, ba/F3EGFR-L858R/T790M, ba/F3 EGFR-Del19/T790M and Ba/F3 EGFR-V769_ D770insASV cells, adding 90 μ L of the cell suspension to a 96-well plate, 37 ℃, 5% CO 2 The culture was carried out overnight. The highest concentration of the drug to be detected is 1 mu M, and the drug is diluted by 3.16 times of gradient and has 9 concentrations.10 μ L of each drug solution with various concentrations was added to each well of a 96-well plate, and the culture was continued for 72 hours in triplicate. And (3) taking out the 96-well plate, placing the 96-well plate at room temperature for balancing for 30min, adding equal volume of CTG reagent into each well, vibrating the 96-well plate on an orbital shaker for 5min to crack cells, placing the 96-well plate at room temperature for 20min to stabilize a cold light signal, and reading the cold light value on a SpectraMax multi-label micropore plate detector (MD, 2104-0010A). Cell viability (%) = (cold light value of test drug-cold light value of culture medium control)/(cold light value of cell control-cold light value of culture medium control) × 100%. Data were analyzed using GraphPad Prism 7.0 software, fitted to the data using non-linear sigmoidal regression to derive a dose-effect curve, and IC was calculated therefrom 50 The value is obtained.
The compound also has strong activity and high selectivity on Ba/F3 EGFR-D770-N771ins _ SVD, ba/F3EGFR-L858R, ba/F3EGFR-L858R/T790M, ba/F3 EGFR-Del19/T790M and Ba/F3 EGFR-V769_ D770insASV cells, so that the compound can inhibit mutant EGFR inserted by exon 20, mutant EGFR of L858R/T790M and mutant EGFR of Del19/T790M with high specificity. The results for representative example compounds are summarized in tables 2 and 3 below.
Table 2:
Figure BDA0002983446160000671
Figure BDA0002983446160000681
* : the control compound was CHMFL-EGFR-202: (R) -1- (3- (4-amino-3- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) piperidin-1-yl) prop-2-en-1-one.
Table 3:
Figure BDA0002983446160000682
* : the control compound was CHMFL-EGFR-202: (R) -1- (3- (4-amino-3- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) piperidin-1-yl) prop-2-en-1-one.
Biological example 3: testing for growth inhibitory Activity against cell lines expressing wild-type HER2
1) Test of growth inhibitory Activity of SK-BR-3 cells and NCI-N87 cells
SK-BR-3 cells and NCI-N87 cells are wild type HER2 cells. Adjusting the concentrations of SK-BR-3 cells and NCI-N87 cells, adding 50. Mu.L of the cell suspensions to 384-well plates, respectively, at 37 ℃ and 5% CO 2 The culture was carried out overnight. A Tecan D300E program is set up. Adding the medicine by a Tecan D300E instrument, wherein the highest concentration of the medicine to be detected is 10 mu M, diluting by 3 times of gradient, carrying out 10 concentrations, carrying out double-hole dilution, and continuously culturing for 72h. The 384 well plates were removed and equilibrated at room temperature for 30min, 30. Mu.L of CTG (Promega, G7573) reagent was added to each well, and after 10min at room temperature, the Luminescence values were read on an EnVision (Perkin Elmer 2104) after the signal had stabilized. Inhibition (%) = (1-Lum) Drug to be tested /Lum Negative control ) x100, negative control 0.667% DMSO. IC (integrated circuit) 50 XL-fit software was used for the calculation of (1).
The compounds of the present invention were tested in the above cytotoxicity experiments and found to have potent activities on SK-BR-3 cells and NCI-N87 cells of wild-type HER2, and thus it was found that the compounds of the present invention can inhibit wild-type HER2 with high specificity. The results for representative example compounds are summarized in table 4 below.
2)Ba/F 3 Parent and Ba/F 3 Test of growth inhibitory Activity of HER2-A775_ G776insYVMA cells
Taking cells in logarithmic growth phase, detecting the cell viability by trypan blue exclusion method, and ensuring the cell viability to be more than 90%. Adjusting the concentration of Ba/F3 parental and Ba/F3 HER2-A775_ G776insYVMA cells, adding 90. Mu.L of cell suspension to a 96-well plate, 37 ℃, 5% CO 2 The culture was carried out overnight. The highest concentration of the drug to be detected is 1 mu M, and the drug is diluted by 3.16 times of gradient and has 9 concentrations. 10 μ L of the drug solution was added to each well of the 96-well plate, and the culture was continued for 72 hours in triplicate. Taking out 96-well plate, balancing at room temperature for 30min, adding equal volume of CTG reagent into each well, and vibrating on fixed-track shaking tableAfter 5min to lyse the cells and 20min at room temperature to stabilize the luminescence signal, the luminescence values were read on a SpectraMax multi-labeled microplate detector (MD, 2104-0010A). Cell survival (%) = (cold light value of test agent-cold light value of culture medium control)/(cold light value of cell control-cold light value of culture medium control) × 100%. Data were analyzed using GraphPad Prism 7.0 software, fitted to the data using non-linear sigmoidal regression to derive a dose-effect curve, and IC was calculated therefrom 50 The value is obtained.
The compounds of the invention also have potent activity and high selectivity against Ba/F3 HER2-A775_ G776insYVMA cells, and the results for representative example compounds are summarized in Table 4 below.
Table 4:
Figure BDA0002983446160000691
* : the control compound was CHMFL-EGFR-202: (R) -1- (3- (4-amino-3- (3-chloro-4- (pyridin-2-ylmethoxy) phenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) piperidin-1-yl) prop-2-en-1-one.
Biological example 4: pharmacokinetic experiment of rat
6 male Sprague-Dawley rats, 7-8 weeks old, weighing about 210g, were divided into 2 groups of 3 per group and compared for pharmacokinetic differences by intravenous or oral administration of a single dose of compound (10 mg/kg oral).
Rats were fed with standard feed and given water. Fasting began 16 hours prior to the experiment. The drug was dissolved with PEG400 and dimethyl sulfoxide. Blood was collected from the orbit at 0.083 hr, 0.25 hr, 0.5 hr, 1 hr, 2 hr, 4 hr, 6 hr, 8 hr, 12 hr and 24 hr post-dose.
The rats were briefly anesthetized after ether inhalation and 300 μ L of blood was collected from the orbit into a test tube. The tube contains 30. Mu.L of 1% heparin sodium solution. Before use, the tubes were dried overnight at 60 ℃. After completion of blood collection at the last time point, rats were sacrificed after ether anesthesia.
Immediately after blood collection, the tube was gently inverted at least 5 times to ensure adequate mixing and placed on ice. The blood samples were centrifuged at 5000rpm for 5 minutes at 4 ℃ to separate the plasma from the red blood cells. Pipette 100 μ L of plasma into a clean plastic centrifuge tube, designating the name of the compound and the time point. Plasma was stored at-80 ℃ before analysis. The concentration of the compounds of the invention in plasma was determined by LC-MS/MS. Pharmacokinetic parameters were calculated based on the plasma concentration of each animal at different time points.
Experiments show that the compound has better pharmacokinetic property in animals, thereby having better pharmacodynamics and treatment effect.
The foregoing is a further detailed description of the invention in connection with specific preferred embodiments and it is not intended to limit the invention to the specific embodiments described. For those skilled in the art to which the invention pertains, several simple deductions or substitutions can be made without departing from the spirit of the invention, and all shall be considered as belonging to the protection scope of the invention.
In summary, the present invention relates to the following technical solutions:
1. a compound of formula (I), or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof:
Figure BDA0002983446160000701
wherein the content of the first and second substances,
ring A is an aromatic ring;
ring C is C 6-10 Aryl or 5 to 10 membered heteroaryl;
A 1 is CR A1 Or N;
A 2 is C or N;
A 4 is CR A4 Or N;
A 5 is C or N;
wherein R is A1 And R A4 Each independently selected from H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Alkoxy, and optionally substituted by oneOr multiple R' substitution;
B 1 is CR 1 Or N;
B 2 is CR 2 Or N;
B 3 is CR 3 Or N;
B 4 is CR 4 Or N;
wherein R is 1 、R 2 、R 3 And R 4 Each independently selected from H, D, halogen, -CN, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, -C (O) R a 、-C(O)OR a 、-C(O)NR b R c 、-NR b R c 、-NR a C(O)R b 、-NR a C(O)OR b 、-NR a C(O)NR b R c 、-OR a 、-OC(O)R a 、-OC(O)OR a 、-OC(O)NR b R c 、C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5 to 10 membered heteroaryl, and optionally substituted with one or more R ";
l is O, S or NR L
Wherein R is L Is selected from H or C 1-6 Alkyl, and optionally substituted with one or more R;
v is (CR) V1 R V2 ) o
Wherein R is V1 And R V2 Each independently selected from H, D, halogen or C 1-6 Alkyl, and optionally substituted with one or more R;
o =1, 2,3, 4,5 or 6;
R 6 is H, D, halogen, -CN, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5 to 10 membered heteroaryl, and optionally substituted with one or more R;
R 5 and R 7 Each independently selected from H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Haloalkyl, and optionally substituted with one or more R; or, R 5 And R 7 Together with the double bond to which they are attached form a triple bond;
each R is independently selected from H, D, halogen, -CN, = O, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, -C (O) R a 、-C(O)OR a 、-C(O)NR b R c 、-NR b R c 、-NR a C(O)R b 、-NR a C(O)OR b 、-NR a C(O)NR b R c 、-OR a 、-OC(O)R a 、-OC(O)OR a 、-OC(O)NR b R c 、C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5 to 10 membered heteroaryl; alternatively, two R groups on the same atom or on adjacent atoms may together form C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5 to 10 membered heteroaryl; wherein each group in the definition of R is optionally substituted with one or more D, up to complete deuteration;
m =0, 1,2, 3,4, 5,6, 7, 8 or 9;
r' are each independently selected from H, D, halogen, -CN, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, -C (O) R a 、-C(O)OR a 、-C(O)NR b R c 、-NR b R c 、-NR a C(O)R b 、-NR a C(O)OR b 、-NR a C(O)NR b R c 、-OR a 、-OC(O)R a 、-OC(O)OR a 、-OC(O)NR b R c 、C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5 to 10 membered heteroaryl, and optionally substituted with one or more R ";
n =0, 1,2, 3 or 4;
p =0, 1 or 2;
r' are each independently selected from H, D, halogen, -CN, = O, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, -C (O) R a 、-C(O)OR a 、-C(O)NR b R c 、-NR b R c 、-NR a C(O)R b 、-NR a C(O)OR b 、-NR a C(O)NR b R c 、-OR a 、-OC(O)R a 、-OC(O)OR a 、-OC(O)NR b R c 、C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5-to 10-membered heteroaryl, or two R' groups on the same atom or on adjacent atoms may together form C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5 to 10 membered heteroaryl; wherein each group in the definition of R "is optionally substituted with one or more D, up to complete deuteration;
r is independently selected from H, D, halogen, -CN, = O, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, -C (O) R a 、-C(O)OR a 、-C(O)NR b R c 、-NR b R c 、-NR a C(O)R b 、-NR a C(O)OR b 、-NR a C(O)NR b R c 、-OR a 、-OC(O)R a 、-OC(O)OR a 、-OC(O)NR b R c 、C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5 to 10 membered heteroaryl; alternatively, two R groups on the same atom or on adjacent atoms may together form C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5 to 10 membered heteroaryl; wherein each group in the definition of R is optionally substituted with one or more D, up to complete deuteration;
R a 、R b and R c Each independently selected from H, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl, C 6-10 Aryl or 5-to 10-membered heteroaryl, or R b And R c Together with the N atom to which they are attached form a 3-to 7-membered heterocyclyl or 5-to 10-membered heteroaryl; wherein R is a 、R b And R c Each group in the definition is optionally substituted with one or more D, up to complete deuteration.
2. A compound according to claim 1, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein,
Figure BDA0002983446160000721
selected from the following structures:
Figure BDA0002983446160000722
preferably, the first and second electrodes are formed of a metal,
Figure BDA0002983446160000723
selected from the following structures:
Figure BDA0002983446160000724
preferably, the first and second electrodes are formed of a metal,
Figure BDA0002983446160000725
selected from the following structures:
Figure BDA0002983446160000731
3. a compound according to claim 1 or 2, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein ring C is phenyl or 5-to 6-membered heteroaryl; preferably phenyl, pyridyl, pyrimidinyl, pyridazinyl, pyrazinyl, pyrrolyl, pyrazolyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, furyl or thienyl; preferably phenyl or pyridyl.
4. A compound according to any one of claims 1-3, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt thereofAcceptable salts, hydrates or solvates thereof, wherein V is (CH) 2 ) o (ii) a Preferably CH 2
5. A compound according to any one of claims 1-4, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein L is O.
6. A compound according to any one of claims 1-5, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein B is 1 Is CR 1 、B 2 Is CR 2 、B 3 Is CR 3 、B 4 Is CR 4 (ii) a Preferably, R 1 、R 2 、R 3 And R 4 Independently selected from H, D, halogen, -CN, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl or C 2-6 Alkynyl.
7. A compound according to any one of claims 1-6, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein,
Figure BDA0002983446160000732
selected from the following structures:
Figure BDA0002983446160000733
preferably, the following components are used:
Figure BDA0002983446160000734
8. a compound according to any one of claims 1-7, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, which is a compound of the general formula:
Figure BDA0002983446160000741
wherein each group is as defined in any one of claims 1 to 7.
9. The compound according to claim 8, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, which is a compound of formula (II-1), (III-1), or (IV-1):
Figure BDA0002983446160000751
wherein the content of the first and second substances,
ring A is an aromatic ring;
A 2 is C or N;
A 4 is CR A4 Or N;
A 5 is C or N;
wherein R is A4 Is H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Alkoxy, and optionally substituted with one or more R ";
R 1 is H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Haloalkyl, and optionally substituted with one or more R ";
R 2 is H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Haloalkyl, and optionally substituted with one or more R ";
R 3 is H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Haloalkyl, and optionally substituted with one or more R ";
R 4 is H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Haloalkyl, and optionally substituted with one or more R ";
o =1, 2,3 or 4;
each R is independently selected from H, D, halogen, -CN, = O, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl or C 2-6 An alkynyl group; whereinEach group in the definition of R is optionally substituted with one or more D, up to complete deuteration;
m =0, 1,2, 3,4 or 5;
p =0, 1 or 2;
r' is independently selected from H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Haloalkyl, and optionally substituted with one or more R ";
n =0, 1,2, 3 or 4;
R 5 、R 6 and R 7 Each independently selected from H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Haloalkyl, and optionally substituted with one or more R; or, R 5 And R 7 Together with the double bond to which they are attached form a triple bond;
r is independently selected from H, D, halogen, -CN, = O, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl or C 2-6 Alkynyl; wherein each group in the definition of R is optionally substituted with one or more D, up to complete deuteration;
each R' is independently selected from H, D, halogen, -CN, = O, -OR a or-NR b R c
R a 、R b And R c Each independently selected from H, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, or R b And R c Together with the N atom to which they are attached form a 3-to 7-membered heterocyclyl or 5-to 10-membered heteroaryl; wherein R is a 、R b And R c Each group in the definition is optionally substituted with one or more D, up to complete deuteration.
10. A compound according to claim 9, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, which is a compound of formula (II-2), (III-2), or (IV-2):
Figure BDA0002983446160000761
wherein, the first and the second end of the pipe are connected with each other,
ring A is an aromatic ring;
A 2 is C or N;
A 4 is CR A4 Or N;
A 5 is C or N;
wherein R is A4 Is H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Alkoxy, and optionally substituted with one or more R ";
R 1 is H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Haloalkyl, and optionally substituted with one or more R ";
R 2 is H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Haloalkyl, and optionally substituted with one or more R ";
o =1, 2,3 or 4;
each R is independently selected from H, D, halogen, -CN, = O, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl or C 2-6 Alkynyl; wherein each group in the definition of R is optionally substituted with one or more D, up to complete deuteration;
m =0, 1,2, 3,4 or 5;
r' is independently selected from H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Haloalkyl, and optionally substituted with one or more R ";
n =0, 1,2, 3 or 4;
each R' is independently selected from H, D, halogen, -CN, = O, -OR a or-NR b R c
R a 、R b And R c Each independently selected from H, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, or R b And R c Together with the N atom to which they are attached form a 3-to 7-membered heterocyclyl or 5-to 10-membered heteroaryl; wherein R is a 、R b And R c Each radical in the definition is optionally substituted by one or more DTo complete deuteration.
11. The compound according to claim 8, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, which is a compound of formula (V-1) or (VI-1):
Figure BDA0002983446160000771
wherein the content of the first and second substances,
A 1 is CR A1 Or N;
wherein R is A1 Is H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Alkoxy, optionally substituted with one or more R ";
R 1 is H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Haloalkyl, and optionally substituted with one or more R ";
R 2 is H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Haloalkyl, and optionally substituted with one or more R ";
R 3 is H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Haloalkyl, and optionally substituted with one or more R ";
R 4 is H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Haloalkyl, and optionally substituted with one or more R ";
o =1, 2,3 or 4;
each R is independently selected from H, D, halogen, -CN, = O, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl or C 2-6 An alkynyl group; wherein each group in the definition of R is optionally substituted with one or more D, up to complete deuteration;
m =0, 1,2, 3,4 or 5;
p =0, 1 or 2;
r' are each independently selected from H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Haloalkyl, and optionally substituted with one or more R ″;
n =0, 1,2, 3 or 4;
R 5 、R 6 and R 7 Each independently selected from H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Haloalkyl, and optionally substituted with one or more R; or, R 5 And R 7 Together with the double bond to which they are attached form a triple bond;
r is independently selected from H, D, halogen, -CN, = O, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl or C 2-6 An alkynyl group; wherein each group in the definition of R is optionally substituted with one or more D, up to complete deuteration;
each R' is independently selected from H, D, halogen, -CN, = O, -OR a or-NR b R c
R a 、R b And R c Each independently selected from H, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, or R b And R c Together with the N atom to which they are attached form a 3-to 7-membered heterocyclyl or 5-to 10-membered heteroaryl; wherein R is a 、R b And R c Each group in the definition is optionally substituted with one or more D, up to complete deuteration.
12. The compound according to claim 11, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, which is a compound of formula (V-2) or (VI-2):
Figure BDA0002983446160000781
wherein, the first and the second end of the pipe are connected with each other,
A 1 is CR A1 Or N;
wherein R is A1 Is H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Alkoxy, and optionally substituted with one or more R ";
R 1 is H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Haloalkyl, and optionally substituted with one or more R ";
R 2 is H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Haloalkyl, and optionally substituted with one or more R ";
o =1, 2,3 or 4;
each R is independently selected from H, D, halogen, -CN, = O, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl or C 2-6 An alkynyl group; wherein each group in the definition of R is optionally substituted with one or more D, up to complete deuteration;
m =0, 1,2, 3,4 or 5;
r' is independently selected from H, D, halogen, -CN, C 1-6 Alkyl or C 1-6 Haloalkyl, and optionally substituted with one or more R ";
n =0, 1,2, 3 or 4;
each R' is independently selected from H, D, halogen, -CN, = O, -OR a or-NR b R c
R a 、R b And R c Each independently selected from H, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, or R b And R c Together with the N atom to which they are attached form a 3-to 7-membered heterocyclyl or 5-to 10-membered heteroaryl; wherein R is a 、R b And R c Each group in the definition is optionally substituted with one or more D, up to complete deuteration.
13. A compound, or tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, selected from the group consisting of:
Figure BDA0002983446160000791
Figure BDA0002983446160000801
14. a pharmaceutical composition comprising a compound of any one of claims 1-13, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, and a pharmaceutically acceptable excipient.
15. Use of a compound of any one of claims 1-13, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, or a pharmaceutical composition of claim 14, in the manufacture of a medicament for the treatment and/or prevention of a wild-type and/or mutant EGFR kinase-mediated tumor;
preferably wherein the mutated EGFR is selected from the group consisting of exon 20 insertion mutant EGFR, exon 18 point mutant EGFR, exon 21 point mutant EGFR, exon 19 deletion mutant EGFR or L858R mutant EGFR;
preferably, wherein the exon 20 insertion mutation is selected from the group consisting of V769_ D770insASV, D770_ N771insSVD, D770_ N771insNPG, D770_ N771insG, H773_ V774insNPH, and H773_ V774insPH;
preferably, wherein said exon 18 point mutation is selected from at least one mutation in G719A, G719S, G719C, E K and E709A;
preferably, wherein said exon 21 point mutation is selected from the group consisting of the L861Q mutation;
preferably, wherein the mutant EGFR also simultaneously has a T790M mutation.
16. Use of a compound of any one of claims 1-13, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, or a pharmaceutical composition of claim 14, in the manufacture of a medicament for the treatment and/or prevention of: lung cancer, breast cancer, head and neck cancer, brain cancer, uterine cancer, hematopoietic cancer or skin cancer.
17. Use of a compound of any one of claims 1-13, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, or a pharmaceutical composition of claim 14, in the manufacture of a medicament for the treatment and/or prevention of a tumor mediated by wild type and/or mutant HER2 kinase;
preferably, wherein the mutated HER2 is selected from G309A mutant HER2, S310F mutant HER2, R678Q mutant HER2, L775_ T759 deletion mutant HER2, D769H mutant HER2, V777L mutant HER2, V842I mutant HER2, R869C mutant HER2, L755S mutant HER2, or ex20insYVMA mutant HER2;
preferably, wherein the ex20insYVMA mutant HER2 is selected from a775_ G776insYVMA mutant HER2 mutation.
18. Use of a compound of any one of claims 1-13, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, or a pharmaceutical composition of claim 14, in the manufacture of a medicament for the treatment and/or prevention of: lung cancer, gastric cancer or breast cancer.

Claims (39)

1. A compound of formula (I), or a tautomer, stereoisomer, pharmaceutically acceptable salt thereof:
Figure FDA0003778936200000011
wherein the content of the first and second substances,
ring A is an aromatic ring;
ring C is phenyl or 5 to 6 membered heteroaryl;
Figure FDA0003778936200000012
selected from the following structures:
Figure FDA0003778936200000013
B 1 is CR 1
B 2 Is CR 2
B 3 Is CR 3
B 4 Is CR 4
Wherein R is 1 、R 2 、R 3 And R 4 Each independently selected from H, D, halogen or-CN;
l is O;
v is (CR) V1 R V2 ) o
Wherein R is V1 And R V2 Each independently selected from H, D, halogen or C 1-6 An alkyl group;
o=1;
R 6 is H or D;
R 5 and R 7 Each is independently selected from H or D;
each R is independently selected from H or D;
m =0, 1,2, 3,4, 5,6, 7, 8 or 9;
each R' is independently selected from H or D;
n =0, 1,2, 3 or 4;
p=1。
2. the compound of claim 1, or a tautomer, stereoisomer, pharmaceutically acceptable salt thereof, wherein,
Figure FDA0003778936200000021
selected from the following structures:
Figure FDA0003778936200000022
3. the compound of claim 1, or a tautomer, stereoisomer, or pharmaceutically acceptable salt thereof, ring C is phenyl or pyridyl.
4. The compound of claim 1, or a tautomer, stereoisomer, or pharmaceutically acceptable salt thereof, V is CH 2
5. The compound of claim 1, or a tautomer, stereoisomer, pharmaceutically acceptable salt thereof, R 1 、R 2 、R 3 And R 4 Independently selected from H or D.
6. The compound of claim 1, or a tautomer, stereoisomer, or pharmaceutically acceptable salt thereof, wherein,
Figure FDA0003778936200000023
the structure is as follows:
Figure FDA0003778936200000024
7. the compound of claim 1, or a tautomer, stereoisomer, pharmaceutically acceptable salt thereof, wherein,
Figure FDA0003778936200000025
the structure is as follows:
Figure FDA0003778936200000026
8. the compound of claim 1, or a tautomer, stereoisomer, or pharmaceutically acceptable salt thereof, which is a compound of the general formula:
Figure FDA0003778936200000031
wherein each group is as defined in any one of claims 1 to 7.
9. The compound of claim 1, or a tautomer, stereoisomer, or pharmaceutically acceptable salt thereof, which is a compound of the general formula:
Figure FDA0003778936200000032
wherein each group is as defined in any one of claims 1 to 7.
10. The compound of claim 1, or a tautomer, stereoisomer, or pharmaceutically acceptable salt thereof, which is a compound of the general formula:
Figure FDA0003778936200000033
wherein each group is as defined in any one of claims 1 to 7.
11. The compound of claim 1, or a tautomer, stereoisomer, or pharmaceutically acceptable salt thereof, which is a compound of the general formula:
Figure FDA0003778936200000041
wherein each group is as defined in any one of claims 1 to 7.
12. The compound of claim 1, or a tautomer, stereoisomer, or pharmaceutically acceptable salt thereof, which is a compound of the general formula:
Figure FDA0003778936200000042
wherein each group is as defined in any one of claims 1 to 7.
13. The compound of claim 1, or a tautomer, stereoisomer, or pharmaceutically acceptable salt thereof, which is a compound of the general formula:
Figure FDA0003778936200000043
wherein each group is as defined in any one of claims 1 to 7.
14. The compound of claim 1, or a tautomer, stereoisomer, or pharmaceutically acceptable salt thereof, which is a compound of the general formula:
Figure FDA0003778936200000051
wherein each group is as defined in any one of claims 1 to 7.
15. The compound of claim 1, or a tautomer, stereoisomer, or pharmaceutically acceptable salt thereof, which is a compound of the general formula:
Figure FDA0003778936200000052
wherein each group is as defined in any one of claims 1 to 7.
16. The compound of claim 1, or a tautomer, stereoisomer, or pharmaceutically acceptable salt thereof, which is a compound of the general formula:
Figure FDA0003778936200000053
wherein each group is as defined in any one of claims 1 to 7.
17. The compound of claim 1, or a tautomer, stereoisomer, or pharmaceutically acceptable salt thereof, which is a compound of the general formula:
Figure FDA0003778936200000061
wherein each group is as defined in any one of claims 1 to 7.
18. The compound of claim 1, or a tautomer, stereoisomer, or pharmaceutically acceptable salt thereof, which is a compound of the general formula:
Figure FDA0003778936200000062
wherein each group is as defined in any one of claims 1 to 7.
19. The compound of claim 1, or a tautomer, stereoisomer, or pharmaceutically acceptable salt thereof, which is a compound of formula (II-1):
Figure FDA0003778936200000063
wherein the content of the first and second substances,
ring A is an aromatic ring;
Figure FDA0003778936200000064
selected from the following structures:
Figure FDA0003778936200000071
R 1 is H, D, halogen or-CN;
R 2 is H, D, halogen or-CN;
R 3 is H, D, halogen or-CN;
R 4 is H, D, halogen or-CN;
o =1; each R is independently selected from H or D;
m =0, 1,2, 3,4 or 5;
p =1; each R' is independently selected from H or D;
n =0, 1,2, 3 or 4;
R 5 、R 6 and R 7 Each independently selected from H or D.
20. The compound of claim 1, or a tautomer, stereoisomer, or pharmaceutically acceptable salt thereof, which is a compound of formula (III-1):
Figure FDA0003778936200000072
wherein the content of the first and second substances,
Figure FDA0003778936200000073
selected from the following structures:
Figure FDA0003778936200000074
the remaining groups are as defined in claim 19.
21. The compound of claim 2, or a tautomer, stereoisomer, or pharmaceutically acceptable salt thereof, which is a compound of formula (IV-1):
Figure FDA0003778936200000081
wherein the content of the first and second substances,
Figure FDA0003778936200000082
the structure is as follows:
Figure FDA0003778936200000083
the remaining groups are as defined in claim 19.
22. The compound of claim 2, or a tautomer, stereoisomer, or pharmaceutically acceptable salt thereof, which is a compound of formula (II-2):
Figure FDA0003778936200000084
wherein the content of the first and second substances,
ring A is an aromatic ring;
Figure FDA0003778936200000085
selected from the following structures:
Figure FDA0003778936200000086
R 1 is H, D, halogen or-CN;
R 2 is H, D, halogen or-CN;
o=1;
each R is independently selected from H or D;
m =0, 1,2, 3,4 or 5;
each R' is independently selected from H or D;
n =0, 1,2, 3 or 4.
23. The compound of claim 2, or a tautomer, stereoisomer, or pharmaceutically acceptable salt thereof, which is a compound of formula (III-2):
Figure FDA0003778936200000091
wherein the content of the first and second substances,
Figure FDA0003778936200000092
selected from the following structures:
Figure FDA0003778936200000093
the remaining groups are as defined in claim 22.
24. The compound of claim 2, or a tautomer, stereoisomer, or pharmaceutically acceptable salt thereof, which is a compound of formula (IV-2):
Figure FDA0003778936200000094
wherein the content of the first and second substances,
Figure FDA0003778936200000095
the structure is as follows:
Figure FDA0003778936200000101
the remaining groups are as defined in claim 22.
25. The compound of claim 1, or a tautomer, stereoisomer, or pharmaceutically acceptable salt thereof, which is a compound of formula (V-1):
Figure FDA0003778936200000102
wherein the content of the first and second substances,
A 1 is CH or N;
R 1 is H, D, halogen or-CN;
R 2 is H, D, halogen or-CN;
R 3 is H, D, halogen or-CN;
R 4 is H, D, halogen or-CN;
o=1;
each R is independently selected from H or D;
m =0, 1,2, 3,4 or 5;
p=1;
each R' is independently selected from H or D;
n =0, 1,2, 3 or 4;
R 5 、R 6 and R 7 Each independently selected from H or D.
26. The compound of claim 1, or a tautomer, stereoisomer, or pharmaceutically acceptable salt thereof, which is a compound of formula (VI-1):
Figure FDA0003778936200000111
wherein each group is as defined in claim 25.
27. The compound of claim 1, or a tautomer, stereoisomer, or pharmaceutically acceptable salt thereof, which is a compound of formula (V-2):
Figure FDA0003778936200000112
wherein the content of the first and second substances,
A 1 is CH or N;
R 1 is H, D, halogen or-CN;
R 2 is H, D, halogen or-CN;
o=1;
each R is independently selected from H or D;
m =0, 1,2, 3,4 or 5;
each R' is independently selected from H or D;
n =0, 1,2, 3 or 4.
28. The compound of claim 1, or a tautomer, stereoisomer, or pharmaceutically acceptable salt thereof, which is a compound of formula (VI-2):
Figure FDA0003778936200000121
wherein each group is as defined in claim 27.
29. A compound, or tautomer, stereoisomer, pharmaceutically acceptable salt thereof, selected from the group consisting of:
Figure FDA0003778936200000122
30. a pharmaceutical composition comprising a compound of any one of claims 1-29, or a tautomer, stereoisomer, pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.
31. Use of a compound of any one of claims 1-29, or a tautomer, stereoisomer, pharmaceutically acceptable salt thereof, or a pharmaceutical composition of claim 30, for the preparation of a medicament for the treatment and/or prevention of a wild type and/or mutant EGFR kinase-mediated tumor.
32. The use of claim 31, wherein the mutated EGFR is selected from the group consisting of exon 20 insertion mutant EGFR, exon 18 point mutant EGFR, exon 21 point mutant EGFR, exon 19 deletion mutant EGFR, or L858R mutant EGFR.
33. The use of claim 32, wherein the exon 20 insertion mutation is selected from V769_ D770insASV, D770_ N771insSVD, D770_ N771insNPG, D770_ N771insG, H773_ V774insNPH or H773_ V774insPH;
wherein the exon 18 point mutation is selected from at least one mutation of G719A, G719S, G719C, E K and E709A;
wherein the exon 21 point mutation is selected from the group consisting of the L861Q mutation.
34. The use of claim 32, wherein the mutant EGFR also simultaneously has a T790M mutation.
35. Use of a compound of any one of claims 1-29, or a tautomer, stereoisomer, pharmaceutically acceptable salt thereof, or a pharmaceutical composition of claim 30, for the preparation of a medicament for the treatment and/or prevention of: lung cancer, breast cancer, head and neck cancer, brain cancer, uterine cancer, hematopoietic cancer or skin cancer.
36. Use of a compound according to any one of claims 1 to 29, or a tautomer, stereoisomer, pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 30, for the preparation of a medicament for the treatment and/or prevention of a wild-type and/or mutant HER2 kinase-mediated tumor.
37. The use of claim 36, wherein the mutated HER2 is selected from G309A mutant HER2, S310F mutant HER2, R678Q mutant HER2, L775_ T759 deletion mutant HER2, D769H mutant HER2, V777L mutant HER2, V842I mutant HER2, R869C mutant HER2, L755S mutant HER2, or ex20 insymva mutant HER2.
38. The use of claim 37, wherein the ex20insYVMA mutant HER2 is selected from a775_ G776insYVMA mutant HER2 mutation.
39. Use of a compound of any one of claims 1-29, or a tautomer, stereoisomer, pharmaceutically acceptable salt thereof, or a pharmaceutical composition of claim 30, for the preparation of a medicament for the treatment and/or prevention of: lung cancer, gastric cancer or breast cancer.
CN202110293647.8A 2020-03-20 2021-03-19 Substituted acrylamide derivative, composition and application thereof Active CN113493439B (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN2020102003527 2020-03-20
CN202010200352 2020-03-20

Publications (2)

Publication Number Publication Date
CN113493439A CN113493439A (en) 2021-10-12
CN113493439B true CN113493439B (en) 2022-10-14

Family

ID=77770496

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110293647.8A Active CN113493439B (en) 2020-03-20 2021-03-19 Substituted acrylamide derivative, composition and application thereof

Country Status (2)

Country Link
CN (1) CN113493439B (en)
WO (1) WO2021185348A1 (en)

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8426428B2 (en) * 2008-12-05 2013-04-23 Principia Biopharma, Inc. EGFR kinase knockdown via electrophilically enhanced inhibitors
PE20151495A1 (en) * 2012-11-15 2015-10-23 Pharmacyclics Inc PYRROLOPYRIMIDINE COMPOUNDS AS KINASE INHIBITORS
US20180305350A1 (en) * 2015-06-24 2018-10-25 Principia Biopharma Inc. Tyrosine kinase inhibitors
CN106928231B (en) * 2015-12-31 2021-06-01 合肥中科普瑞昇生物医药科技有限公司 Novel EGFR wild type and mutant kinase inhibitors
CN109310671B (en) * 2016-01-21 2021-08-06 淄博百极常生制药有限公司 Bruton's tyrosine kinase inhibitors
WO2018033091A1 (en) * 2016-08-17 2018-02-22 深圳市塔吉瑞生物医药有限公司 Fused bicyclic compound for inhibiting activity of tyrosine kinase

Also Published As

Publication number Publication date
WO2021185348A1 (en) 2021-09-23
CN113493439A (en) 2021-10-12

Similar Documents

Publication Publication Date Title
EP3788040B1 (en) Pyridazinones as parp7 inhibitors
CN111153901B (en) Nitrogen-containing fused heterocyclic SHP2 inhibitor compound, preparation method and application
US11629148B2 (en) Substituted pyrrolo[3,4-d]imidazoles as JAK inhibitors
EP3978490A1 (en) Nitrogen-containing heterocyclic derivative regulator, preparation method therefor and application thereof
EP3230287B1 (en) Piperidine substituted pyrazolo[1,5-a]pyrimidine derivatives with inhibitory activity on the replication of the respiratory syncytial virus (rsv)
JP7353682B2 (en) Substituted fused aromatic ring derivatives, compositions thereof, and uses thereof
EP3889152A1 (en) Heteroaromatic derivatives for use as regulator, preparation method therefor and use thereof
EP4092019A1 (en) Heteroaryl derivative, preparation method therefor, and use thereof
CN114163454A (en) Pyridine-containing polycyclic derivative inhibitor, and preparation method and application thereof
CN109851638B (en) Substituted diaminopyrimidine compounds
TW202016120A (en) Tricyclic derivatives inhibitor, preparation method, and applications thereof
EP4129987A1 (en) Crystal form of free alkali of nitrogen-containing aromatic derivatives
WO2013016999A1 (en) Heteroaryl-pyrimidine derivatives, and preparation method therefor and use thereof
EP3459953B1 (en) Novel 5h-pyrrolo[2,3-d]pyrimidin-6(7h)-one derivative
EP4289835A1 (en) Cdk inhibitor
EP3887372B1 (en) Further heteroaromatic compounds having activity against rsv
EP3992193A1 (en) Pyrazolopyrimidine compound, preparation method for same, and applications thereof
CN113493439B (en) Substituted acrylamide derivative, composition and application thereof
CN112574208B (en) Substituted fused tricyclic derivatives, compositions and uses thereof
CN113336760B (en) Substituted amide derivatives, compositions and uses thereof
RU2811484C1 (en) Substituted aromatic derivative with condensed rings and composition including it and their use
CN115504980A (en) Pyrimido nitrogen-containing six-membered aromatic heterocyclic compound and application thereof
CN111574521A (en) Substituted aromatic fused ring derivatives, compositions and uses thereof
EP3885346A1 (en) Aminopyrimidine compound used for inhibiting activity of protein kinase
CN117003757A (en) Substituted bicyclic heteroaryl compounds as USP1 inhibitors

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