CN108137544B - Aminopyrimidines useful for inhibiting protein tyrosine kinase activity - Google Patents

Aminopyrimidines useful for inhibiting protein tyrosine kinase activity Download PDF

Info

Publication number
CN108137544B
CN108137544B CN201680061348.9A CN201680061348A CN108137544B CN 108137544 B CN108137544 B CN 108137544B CN 201680061348 A CN201680061348 A CN 201680061348A CN 108137544 B CN108137544 B CN 108137544B
Authority
CN
China
Prior art keywords
compound
group
cancer
compounds
egfr
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
CN201680061348.9A
Other languages
Chinese (zh)
Other versions
CN108137544A (en
Inventor
王义汉
李焕银
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 CN108137544A publication Critical patent/CN108137544A/en
Application granted granted Critical
Publication of CN108137544B publication Critical patent/CN108137544B/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
    • A61K31/495Heterocyclic 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
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/46Two or more oxygen, sulphur or nitrogen atoms
    • C07D239/48Two nitrogen atoms
    • 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
    • C07D401/02Heterocyclic 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 two hetero rings
    • C07D401/12Heterocyclic 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 two hetero rings linked by a chain containing hetero atoms as chain links

Abstract

The invention relates to aminopyrimidine compounds with an inhibiting effect on protein tyrosine kinase activity, and relates to preparation and application thereof. Specifically, the invention discloses an aminopyrimidine compound shown in a formula (I) and a pharmaceutical composition containing the compound, and pharmaceutically acceptable salts, stereoisomers, solvates, hydrates, crystal forms, prodrugs or isotopic variations thereof. The compounds of the present invention are useful for the treatment and/or prevention of protein tyrosine kinase related diseases, such as cell proliferative diseases, cancer, immunological diseases.

Description

Aminopyrimidines useful for inhibiting protein tyrosine kinase activity
Technical Field
The invention belongs to the field of medicine. In particular, the invention relates to aminopyrimidine compounds with inhibiting effect on protein tyrosine kinase activity, pharmaceutical compositions containing the aminopyrimidine compounds, and preparation methods and applications of the aminopyrimidine compounds and the pharmaceutical compositions. More particularly, the present invention relates to aminopyrimidines useful for treating diseases associated with dysregulation of protein tyrosine kinases (such as, but not limited to EGFR (including HER), ALK, PDGFR) metabolic pathways, such as tumors and related diseases, and pharmaceutical compositions and methods of use thereof.
Background
Protein Tyrosine Kinases (PTK) are kinases which catalyze the transfer of gamma-phosphate on ATP to protein tyrosine residues, can catalyze the phosphorylation of the tyrosine residues of various substrate proteins, and have important functions in cell growth, proliferation and differentiation. Because protein tyrosine kinases have an extremely profound effect on cells, their activity is highly regulated. Kinases turn on or off by phosphorylation (sometimes by autophosphorylation), by binding to activator proteins, repressor proteins, or small molecules, or by controlling their localization in cells. Dysfunction of kinase activity, which is caused by genetic abnormalities or environmental factors, is now known to be associated with a variety of diseases. Some serious pathological states, including cancer and chronic inflammation, are associated with intracellular signal stimulation, and since kinases actively transmit signals, their inhibition provides a powerful way to inhibit or control signal transduction cascades.
Protein tyrosine kinases can be classified into non-receptor type and membrane receptor type according to whether they are present in cell membrane receptors or not. Non-receptor tyrosine kinases (nrrtks) are represented by Src, and may include Yes, Fyn, Lck, Fgr, Lyn, Fps/Fes, Ab1, and the like. Receptor Tyrosine Kinases (RTKs) can be classified into 9 types according to their structure, among which the following 4 types are more common: the Epidermal Growth Factor Receptor (EGFR) family, the insulin receptor family, the PDGF/MCSF/SCF receptor family, and the Fibroblast Growth Factor Receptor (FGFR) family.
The epidermal growth factor receptor (human EGFR; ErbB-1; HER1) is a member of the ErbB receptor family, which is a subfamily of the four closely related receptor tyrosine kinases EGFR (ErbB-1), HER2/c-NEU (ErbB-2), HER 3(ErbB-3) and HER 4 (FrbB-4). EGFR is a cell surface receptor for members of the extracellular protein ligand epidermal growth factor family (EGF family). Mutations that affect EGFR expression or activity may lead to cancer. EGFR is reported to be dysregulated in most solid tumors, such as lung, breast and brain tumors. It is estimated that 30% of epithelial cancers are associated with mutations, amplifications, or dysregulation of EGFR or family members.
A Receptor Tyrosine Kinase Inhibitor (RTKI) is an inhibitor of receptor tyrosine kinase, and the inhibitor is usually a small molecular compound, and blocks an intracellular region activation signal after a cell surface receptor is combined with a ligand through inhibiting the activity (reversible or irreversible) of the receptor tyrosine kinase, so that the final biological effect of transferring the signal to a channel is inhibited.
In recent years, the field of research on receptor tyrosine kinase inhibitors has attracted attention. The first generation of receptor tyrosine kinase inhibitors are single target tyrosine kinase inhibitors, such as gefitinib, erlotinib and other drugs. Gefitinib is an EGFR tyrosine kinase inhibitor, is mainly used for non-small cell lung cancer (NSCLC), and has an effective rate of more than 80 percent on the mutant tumor of a tyrosine kinase gene coding region. Erlotinib is an EGFR tyrosine kinase inhibitor, and the effective rate of the drug on patients with non-small cell lung cancer relapse is about 10%.
However, acquired resistance to gefitinib or erlotinib becomes a major clinical problem after a period of treatment. Studies have shown that one of the major causes of resistance is due to the T790M mutation, which is the "gatekeeper" of EGFR. Then, second generation receptor tyrosine kinase inhibitors against T790M, such as afatinib, were developed, which are irreversible inhibitors of EGFR and show advantages in clinical trials. Although these inhibitors are effective in inhibiting the T790M mutation, they also inhibit wild-type egfr (wt egfr) at the same time, resulting in serious side effects, which limit clinical use.
The third generation and subsequent EGFR inhibitors, including AZD9291 and CO-1686, can irreversibly inhibit EGFR, and have higher efficiency for patients with T790M drug resistance mutation, but also have inhibition effect on wild type EGFR.
Therefore, there is a need to further develop novel EGFR inhibitors that not only effectively inhibit the T790M mutation, but also have high selectivity for the T790M mutation over the wild type.
Disclosure of Invention
The invention provides a novel class of EGFR inhibitors which have better pharmacodynamic/pharmacokinetic properties and are more selective for the T790M mutation, the L858R mutation and the L858R/T790M double mutation.
Accordingly, in one aspect, the present invention provides a compound of formula (I):
Figure GPA0000244730960000031
wherein the content of the first and second substances,
R1independently selected from H, halogen, -CN, -NO2、C1-6Alkyl radical, C1-6Haloalkyl, C1-6Alkoxy or C1-6A haloalkoxy group;
R2independently selected from H, halogen, -CN, -NO2、C1-6Alkyl radical, C1-6Haloalkyl, C1-6Alkoxy or C1-6A haloalkoxy group;
the linking group L is selected from
1) C, N, O or an S atom, as valency permits; wherein C and N may optionally be substituted by RaSubstitution;
2)C1-6alkylene radical, C2-6Alkenylene radical, C2-6Alkynylene, C3-10Carbocyclylene, 3-to 10-membered heterocyclylene, C6-14Arylene radicals or C5-10A heteroarylene group; wherein said group is optionally substituted with one or more RaSubstitution;
3)-C(=O)-、-C(=O)O-、-C(=O)N(Rb)-、-N(Rb)C(=O)-、-N(Rb)C(=O)O-、-N(Rb)C(=O)N(Rb)-、-N(Rb)S(=O)-、-N(Rb)S(=O)2-、-OC(=O)-、-OC(=O)N(Rb)-、-OS(=O)-、-OS(=O)2-、-S(=O)-、-S(=O)2-、-S(=O)2O-or-S (═ O)2N(Rb)-;
RaSelected from H, halogen, -CN, -NO2Oxo, C1-6Alkyl radical, C1-6Haloalkyl or C1-6An alkoxy group;
Rbselected from H, C1-6Alkyl or C1-6A haloalkyl group;
x is selected from C or N atoms, provided that valency permits;
ring A is selected from C3-6Carbocyclyl, 3-to 10-membered heterocyclyl, C6-14Aryl or C5-10A heteroaryl group;
R3selected from halogen, -CN, -NO2、-Rc、-C(=O)Rc、-C(=O)ORc、-C(=O)N(Rc)(Rc)、-N(Rc)(Rc)、-N(Rc)C(=O)Rc、-N(Rc)C(=O)ORc、-N(Rc)C(=O)N(Rc)(Rc)、-N(Rc)S(=O)Rc、-N(Rc)S(=O)N(Rc)(Rc)、-N(Rc)S(=O)2Rc、-N(Rc)S(=O)2N(Rc)(Rc)、-ORc、-OC(=O)Rc、-OC(=O)ORc、-OC(=O)N(Rc)(Rc)、-S(=O)Rc、-S(=O)ORc、-S(=O)N(Rc)(Rc)、-S(=O)2Rc、-S(=O)2ORc、-S(=O)2N(Rc)(Rc)、-SC(=O)Rc、-SC(=O)ORcor-SC (═ O) N (R)c)(Rc) As chemically permitted;
Rcselected from H, halogen, -CN, -NO2、C1-6Alkyl radical, C1-6Haloalkyl, C2-6Alkenyl radical, C2-6Alkynyl, C3-10Carbocyclyl, 3-to 10-membered heterocyclyl, C6-14Aryl or C5-10A heteroaryl group; wherein said C3-10Carbocyclyl, 3-to 10-membered heterocyclyl, C6-14Aryl or C5-10Heteroaryl is also optionally substituted by C1-6Alkyl radical, C1-6Haloalkyl or-C (═ O) C1-6Alkyl substitution;
m is 0, 1 or 2;
n is 1, 2 or 3;
p represents a single or double bond, as far as valency permits;
q represents a single or double bond, provided that valency permits;
R4、R5and R6Independently selected from H, halogen, -CN, C1-6Alkyl or C1-6A haloalkyl group;
w is selected from N, O or S atom, wherein N may be optionally substituted by RaSubstitution;
provided that, when m is 0, X is a C atom;
or a pharmaceutically acceptable salt, stereoisomer, solvate, hydrate, crystal form, prodrug, or isotopic variant thereof.
In another aspect, the invention provides a pharmaceutical composition comprising a compound of the invention and a pharmaceutically acceptable excipient. In a specific embodiment, the compounds of the present invention are provided in an effective amount in the pharmaceutical composition. 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 invention provides pharmaceutical compositions comprising a compound of the invention and a pharmaceutically acceptable excipient, which also contain an additional therapeutic agent.
In another aspect, the invention provides kits comprising a compound of the invention, a pharmaceutically acceptable salt, stereoisomer, solvate, hydrate, crystal form, prodrug, or isotopic variant thereof, and an additional therapeutic agent, as well as a pharmaceutically acceptable carrier, adjuvant, or vehicle.
In another aspect, the invention provides methods of treating a condition associated with EGFR-driven cancer (including EGFR mutation-driven cancer, e.g., cancer with a T790M mutation, a L858R mutation, and a L858R/T790M double mutation) in a subject in need thereof, the method comprising: administering to the subject an effective amount of a compound of the invention. In specific embodiments, the EGFR-driven cancer is selected from: non-small cell lung cancer, lung adenocarcinoma, lung squamous carcinoma, pancreatic cancer, breast cancer, prostate cancer, liver cancer, skin cancer, epithelial cell cancer, gastrointestinal stromal tumor, leukemia, histiocytic lymphoma, nasopharyngeal carcinoma, etc. In particular embodiments, the compound is administered orally, subcutaneously, intravenously, or intramuscularly. In particular embodiments, the compound is administered chronically.
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. "C1-6Alkyl "includes C1、C2、C3、C4、C5、C6、C1-6、C1-5、C1-4、C1-3、C1-2、C2-6、C2-5、C2-4、C2-3、C3-6、C3-5、C3-4、C4-6、C4-5And C5-6An alkyl group.
It is to be understood that any of the moieties defined below may be substituted with a number of substituents when described herein, and that the corresponding definitions are within their scope as set forth below, including such substituted moieties. Unless otherwise specified, the term "substituted" is as defined below.
“C1-6Alkyl "refers to a straight or branched chain saturated hydrocarbon group having 1 to 6 carbon atoms, also referred to herein as" lower alkyl ". In some embodiments, C1-4Alkyl 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). Unless otherwise specified, each of the alkyl groups is independently optionally substituted, i.e., unsubstituted (an "unsubstituted alkyl") or substituted (a "substituted alkyl") with one or more substituents; for example, 1 to 5 substituents, 1 to 3 substituents, or 1 substituent. In some embodiments, alkyl is unsubstituted C1-6Alkyl (e.g., -CH)3). In some embodiments, alkyl is substituted C1-6An alkyl group.
“C2-6Alkenyl "refers to a straight or branched hydrocarbon group having 2 to 6 carbon atoms and one or more carbon-carbon double bonds (e.g., 1, 2, or 3 carbon-carbon double bonds). One or more carbon-carbon double bonds may be internal (e.g., in a 2-butenyl group) or terminal (e.g., in a 1-butenyl group). In some embodiments, C2-4Alkenyl groups are particularly preferred. Examples of the alkenyl group include, but are not limited to: 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. Unless otherwise specified, each of the alkenyl groups is independently optionally substituted, i.e., unsubstituted (an "unsubstituted alkenyl") or substituted (a "substituted alkenyl") with one or more substituents; for example, 1 to 5 substituents, 1 to 3 substituents, or 1 substituent. In some embodiments, alkenyl is unsubstituted C2-6An alkenyl group. In some embodiments, alkenyl is substituted C2-6An alkenyl group.
“C2-6Alkynyl "refers to a straight or branched hydrocarbon group having 2 to 6 carbon atoms, one or more carbon-carbon triple bonds (e.g., 1, 2, or 3 carbon-carbon triple bonds), and optionally one or more carbon-carbon double bonds (e.g., 1, 2, or 3 carbon-carbon double bonds). In some embodiments, C2-4Alkynyl groups are particularly preferred. In some embodiments, alkynyl groups do not contain any double bonds. One or more carbon triple bonds may be internal (e.g., in 2-butynyl) or terminal (e.g., in 1-butynyl). Examples of the alkynyl groupIncluding but not limited to: ethynyl (C)2) 1-propynyl (C)3) 2-propynyl (C)3) 1-butynyl (C)4) 2-butynyl (C)4) Pentynyl group (C)5) Hexynyl (C)6) And so on. Unless otherwise specified, each of the alkynyl groups is independently optionally substituted, i.e., unsubstituted (an "unsubstituted alkynyl") or substituted (a "substituted alkynyl") with one or more substituents; for example, 1 to 5 substituents, 1 to 3 substituents, or 1 substituent. In some embodiments, alkynyl is unsubstituted C2-6Alkynyl. In some embodiments, alkynyl is substituted C2-6Alkynyl.
As used herein, "alkylene", "alkenylene" and "alkynylene" refer to divalent radicals of the above alkyl, alkenyl and alkynyl radicals, respectively. When a range or number of carbons is provided by the particular "alkylene," "alkenylene," and "alkynylene," it is understood that the range or number refers to the range or number of carbons in the linear divalent carbon chain. "alkylene," "alkenylene," and "alkynylene" may be substituted with one or more substituents described herein, or unsubstituted.
“C1-6Alkylene "means removal of C1-6One hydrogen of the alkyl group forms a divalent alkylene group and may be a substituted or unsubstituted alkylene group. In some embodiments, C1-4Alkylene groups are particularly preferred. Unsubstituted said alkylene groups include, but are not limited to: methylene (-CH)2-) ethylene (-CH2CH2-) propylene (-CH)2CH2CH2-) butylene (-CH)2CH2CH2CH2-) pentylene (-CH)2CH2CH2CH2CH2-) and hexylene (-CH2CH2CH2CH2CH2CH2-) and the like. Exemplary substituted said alkylene groups, for example, said alkylene groups substituted with one or more alkyl (methyl) groups, include, but are not limited to: substituted methylene (-CH (CH)3)-、-C(CH3)2-) substituted ethylene (-CH (CH)3)CH2-、-CH2CH(CH3)-、-C(CH3)2CH2-、-CH2C(CH3)2-) substituted propylene (-CH (CH)3)CH2CH2-、-CH2CH(CH3)CH2-、-CH2CH2CH(CH3)-、-C(CH3)2CH2CH2-、-CH2C(CH3)2CH2-、-CH2CH2C(CH3)2-) and the like.
“C2-6Alkenylene "means the removal of C2-6One hydrogen of an alkenyl group forms a divalent alkenylene group, and may be a substituted or unsubstituted alkenylene group. In some embodiments, C2-4Alkenylene is particularly preferred. Exemplary unsubstituted alkenylene groups include, but are not limited to: vinylidene (-CH-) and propenylene (e.g., -CH-) CHCH2-、-CH2-CH ═ CH —). Exemplary substituted alkenylene groups, for example, alkenylene substituted with one or more alkyl (methyl) groups, include, but are not limited to: substituted ethylene (-C (CH)3)=CH-、-CH=C(CH3) -) substituted propenylene (-C (CH)3)=CHCH2-、-CH=C(CH3)CH2-、-CH=CHCH(CH3)-、-CH=CHC(CH3)2-、-CH(CH3)-CH=CH-、-C(CH3)2-CH=CH-、-CH2-C(CH3)=CH-、-CH2-CH=C(CH3) -) and the like.
“C2-6Alkynylene "means removal of C2-6One hydrogen of the alkynyl group forms a divalent alkynylene group, and may be a substituted or unsubstituted alkynylene group. In some embodiments, C2-4Alkynylene groups are particularly preferred. Exemplary such alkynylene groups include, but are not limited to: ethynylene (-C [ identical to ] C-), substituted or unsubstituted propynyl (-C [ identical to ] CCH)2-) and the like.
“C1-6Alkoxy "refers to the group-OR, where R is substituted OR unsubstituted C1-6An alkyl group. In some embodiments of the present invention, the substrate is,C1-4alkoxy 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.
"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, "C1-6Haloalkyl "and" C1-6Haloalkoxy "means" C "as defined above1-6Alkyl "and" C1-6Alkoxy ", which is substituted with one or more halo groups. In some embodiments, C1-4Haloalkyl is particularly preferred, more preferably C1-2A haloalkyl group. In some embodiments, C1-4Haloalkoxy is particularly preferred, more preferably C1-2A haloalkoxy group. Exemplary said haloalkyl groups include, but are not limited to: -CF3、-CH2F、-CHF2、-CHFCH2F、-CH2CHF2、-CF2CF3、-CCl3、-CH2Cl、-CHCl22, 2, 2-trifluoro-1, 1-dimethyl-ethyl, and the like. Exemplary said haloalkoxy groups include, but are not limited to: -OCH2F、-OCHF2、-OCF3And so on.
“C3-10Carbocyclyl "refers to a non-aromatic cyclic hydrocarbon group having 3 to 10 ring carbon atoms and zero heteroatoms. In some embodiments, C3-6Carbocyclyl is particularly preferred, more preferably C5-6A carbocyclic group. Carbocyclyl also includes ring systems in which the aforementioned carbocyclyl ring is fused to one or more aryl or heteroaryl groups, wherein the point of attachment is on the carbocyclyl ring, and in such cases the number of carbons continues to represent the number of carbons in the carbocyclyl system. Exemplary such carbocyclyl 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 (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. Unless otherwise specified, each of the carbocyclyl groups is independently optionally substituted, i.e., unsubstituted (an "unsubstituted carbocyclyl") or substituted with one or more substituents (a "substituted carbocyclyl"). In some embodiments, carbocyclyl is unsubstituted C3-10A carbocyclic group. In some embodiments, carbocyclyl is substituted C3-10A carbocyclic group.
"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 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 a 5 to 6 membered heterocyclic group which is a 5 to 6 membered non aromatic ring system having ring carbon atoms and 1 to 3 ring heteroatoms. Unless otherwise specified, each of the heterocyclyl groups is independently optionally substituted, i.e., unsubstituted (an "unsubstituted heterocyclyl") or substituted (a "substituted heterocyclyl") with one or more substituents. In some embodiments, a heterocyclyl is an unsubstituted 3-10 membered heterocyclyl. In some embodiments, heterocyclyl is a substituted 3-10 membered heterocyclyl. Heterocyclyl also includes ring systems in which the above-mentioned heterocyclyl ring is fused to one or more carbocyclic groups in which the point of attachment is on the carbocyclic ring, or in which the above-mentioned heterocyclyl ring is fused to one or more aryl or heteroaryl groupsA fused ring system 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. 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 (disulphuryl), and oxazolidin-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, dithiinyl, 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 thietanyl. Exemplary with C6Aryl 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 C6Aryl 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.
“C6-14Aryl "refers to a group having 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) of 6 to 14 ring carbon atoms and zero heteroatoms. In some embodiments, an aryl group has six ring carbon atoms ("C)6Aryl "; for example, phenyl). In some embodiments, an aryl group has ten ring carbon atoms ("C)10Aryl "; e.g., naphthyl, e.g., 1-naphthyl and 2-naphthyl). In some embodiments, an aryl group has fourteen ring carbon atoms ("C)14Aryl "; for example, an anthracene group). In some embodiments, C6-10Aryl is particularly preferred, more preferably C6And (4) an aryl group. Aryl also includes ring systems in which the aforementioned aryl ring is fused to one or more carbocyclic or heterocyclic groups and the point of attachment is on the aryl ring, in which case the number of carbon atoms continues to represent the number of carbon atoms in the aryl ring system. Unless otherwise specified, each of the aryl groups is independently optionally substituted, i.e., unsubstituted (an "unsubstituted aryl") or substituted (a "substituted aryl") with one or more substituents. In some embodiments, aryl is unsubstituted C6-14And (4) an aryl group. In some embodiments, aryl is substituted C6-14And (4) an aryl group.
“C5-10Heteroaryl "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 carbocyclic or heterocyclic 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, C5-6Heteroaryl groups are particularly preferred, which are those having ring carbon atoms and 1 to 4 ring heteroatomsA 5-6 membered monocyclic or bicyclic 4n +2 aromatic ring system. Unless otherwise specified, each of the heteroaryl groups is independently optionally substituted, i.e., unsubstituted (an "unsubstituted heteroaryl") or substituted (a "substituted heteroaryl") with one or more substituents. In some embodiments, the heteroaryl is an unsubstituted 5-10 membered heteroaryl. In some embodiments, heteroaryl is substituted 5-10 membered heteroaryl. 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.
As used herein, "carbocyclylene", "heterocyclylene", "arylene" and "heteroarylene" refer to the divalent radicals of carbocyclyl, heterocyclyl, aryl and heteroaryl, respectively, described above, resulting from the removal of one hydrogen atom from the radical at any possible position to yield a divalent radical. "carbocyclylene", "heterocyclylene", "arylene" and "heteroarylene" may be substituted with one or more substituents described herein, or may be unsubstituted. Exemplary unsubstituted "carbocyclylene", "heterocyclylene", "arylene", and "heteroarylene" include, but are not limited to, the following:
Figure GPA0000244730960000081
wherein Z represents O, N or an S atom.
"oxo" refers to ═ O.
Other definitions
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, Berge et al, in j.pharmaceutical Sciences (1977) 66: 1-19. 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 of amino groups 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, or using methods used in the art, such as ion exchange methods. 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, and gluconateLauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, p-toluenesulfonate, undecanoate, valerate, and the like. Pharmaceutically acceptable salts derived from suitable bases include alkali metals, alkaline earth metals, ammonium and N+(C1-4Alkyl radical)4And (3) salt. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium salts, and the like. Further pharmaceutically acceptable salts include, if appropriate, non-toxic ammonium, quaternary ammonium and amine cations formed using counterions such as halide, hydroxide, carboxylate, 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 the effect that occurs when a subject has 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 the effect that occurs before the subject begins to have the particular disease, disorder or condition ("prophylactic treatment").
Generally, an "effective amount" of a compound is an amount sufficient to elicit a biological response of interest. As will be appreciated by those of ordinary skill in the art, the effective amount of a compound of the present invention may vary depending on the following factors: for example, biological goals, pharmacokinetics of the compound, the disease being treated, mode of administration, and the age, health, and condition of the subject. An effective amount includes both therapeutically and prophylactically therapeutically effective amounts.
As used herein, unless otherwise specified, a "therapeutically effective amount" of a compound is an amount sufficient to provide a therapeutic benefit in the treatment of a disease, disorder, or condition, or to delay or minimize one or more symptoms associated with a disease, disorder, or condition. A therapeutically effective amount of a compound refers to the amount of a therapeutic agent, alone or in combination with other therapies, that provides a therapeutic benefit in the treatment of a disease, disorder, or condition. The term "therapeutically effective amount" can include an amount that improves the overall treatment, reduces or avoids symptoms or causes of a disease or disorder, or enhances the therapeutic efficacy of other therapeutic agents.
As used herein, unless otherwise specified, a "prophylactically effective amount" of a compound is an amount sufficient to prevent a disease, disorder, or condition, or one or more symptoms associated with a disease, disorder, or condition, or to prevent recurrence of a disease, disorder, or condition. A prophylactically effective amount of a compound refers to the amount of a therapeutic agent, alone or in combination with other agents, that provides a prophylactic benefit in preventing a disease, disorder, or condition. The term "prophylactically effective amount" can include an amount that improves overall prophylaxis, or an amount that enhances the prophylactic efficacy of other prophylactic agents.
"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.
"EGFR-driven cancer" refers to cancers characterized by inappropriately high expression of the EGFR gene or by mutations in the EGFR gene that alter the biological activity of the EGFR nucleic acid molecule or polypeptide. EGFR-driven cancers can occur in any tissue, including brain, blood, connective tissue, liver, mouth, muscle, spleen, stomach, testis, and trachea. EGFR-driven cancers include, but are not limited to, non-small cell lung cancer, lung adenocarcinoma, lung squamous carcinoma, pancreatic cancer, breast cancer, prostate cancer, liver cancer, skin cancer, epithelial cell cancer, gastrointestinal stromal tumor, leukemia, histiocytic lymphoid cancer, nasopharyngeal carcinoma.
"EGFR mutation" or "EGFR mutant" includes one or more deletions, substitutions or additions in the amino acid or nucleotide sequence of the EGFR protein or EGFR coding sequence. EGFR mutations may also include one or more deletions, substitutions or additions, or fragments thereof, so long as the mutant retains or increases tyrosine kinase activity relative to wild-type EGFR. In particular EGFR mutations, kinase or phosphorylation activity can be increased or decreased (e.g., by at least 5%, 10%, 15%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or even 100%) relative to wild-type EGFR. Exemplary EGFR mutations include, but are not limited to, the T790M mutation, the L858R mutation, and the L858R/T790M double mutation.
Detailed Description
Compound (I)
Herein, "compound of the invention" refers to a compound of formula (I) -a compound of formula (VI) (including a subset of formulae, e.g., a compound of formula (VIf) and a compound of formula (VIf-1)), a pharmaceutically acceptable salt, a stereoisomer, a solvate, a hydrate, a crystalline form, a prodrug, or an isotopic variant thereof.
In one embodiment, the present invention relates to compounds of formula (I):
Figure GPA0000244730960000101
wherein the content of the first and second substances,
R1independently selected from H, halogen, -CN, -NO2、C1-6Alkyl radical, C1-6Haloalkyl, C1-6Alkoxy or C1-6A haloalkoxy group;
R2independently selected from H, halogen, -CN, -NO2、C1-6Alkyl radical, C1-6Haloalkyl, C1-6Alkoxy or C1-6A haloalkoxy group;
the linking group L is selected from
1) C, N, O or an S atom, as valency permits; wherein C and N may optionally be substituted by RaSubstitution;
2)C1-6alkylene radical, C2-6Alkenylene radical, C2-6Alkynylene, C3-10Carbocyclylene, 3-to 10-membered heterocyclylene, C6-14Arylene radicals or C5-10A heteroarylene group; wherein said group is optionally substituted with one or more RaSubstitution;
3)-C(=O)-、-C(=O)O-、-C(=O)N(Rb)-、-N(Rb)C(=O)-、-N(Rb)C(=O)O-、-N(Rb)C(=O)N(Rb)-、-N(Rb)S(=O)-、-N(Rb)S(=O)2-、-OC(=O)-、-OC(=O)N(Rb)-、-OS(=O)-、-OS(=O)2-、-S(=O)-、-S(=O)2-、-S(=O)2O-or-S (═ O)2N(Rb)-;
RaSelected from H, halogen, -CN, -NO2Oxo, C1-6Alkyl radical, C1-6Haloalkyl or C1-6An alkoxy group;
Rbselected from H, C1-6Alkyl or C1-6A haloalkyl group;
x is selected from C or N atoms, provided that valency permits;
ring A is selected from C3-6Carbocyclyl, 3-to 10-membered heterocyclyl, C6-14Aryl or C5-10A heteroaryl group;
R3selected from halogen, -CN, -NO2、-Rc、-C(=O)Rc、-C(=O)ORc、-C(=O)N(Rc)(Rc)、-N(Rc)(Rc)、-N(Rc)C(=O)Rc、-N(Rc)C(=O)ORc、-N(Rc)C(=O)N(Rc)(Rc)、-N(Rc)S(=O)Rc、-N(Rc)S(=O)N(Rc)(Rc)、-N(Rc)S(=O)2Rc、-N(Rc)S(=O)2N(Rc)(Rc)、-ORc、-OC(=O)Rc、-OC(=O)ORc、-OC(=O)N(Rc)(Rc)、-S(=O)Rc、-S(=O)ORc、-S(=O)N(Rc)(Rc)、-S(=O)2Rc、-S(=O)2ORc、-S(=O)2N(Rc)(Rc)、-SC(=O)Rc、-SC(=O)ORcor-SC (═ O) N (R)c)(Rc) As chemically permitted;
Rcselected from H, halogen, -CN, -NO2、C1-6Alkyl radical, C1-6Haloalkyl, C2-6Alkenyl radical, C2-6Alkynyl, C3-10Carbocyclyl, 3-to 10-membered heterocyclyl, C6-14Aryl or C5-10A heteroaryl group; wherein said C3-10Carbocyclyl, 3-to 10-membered heterocyclyl, C6-14Aryl or C5-10Heteroaryl is also optionally substituted by C1-6Alkyl radical, C1-6Haloalkyl or-C (═ O) C1-6Alkyl substitution;
m is 0, 1 or 2;
n is 1, 2 or 3;
p represents a single or double bond, as far as valency permits;
q represents a single or double bond, provided that valency permits;
R4、R5and R6Independently selected from H, halogen, -CN, C1-6Alkyl or C1-6A haloalkyl group;
w is selected from N, O or S atom, wherein N may be optionally substituted by RaSubstitution;
provided that, when m is 0, X is a C atom;
or a pharmaceutically acceptable salt, stereoisomer, solvate, hydrate, crystal form, prodrug, or isotopic variant thereof.
Preferably, in this embodiment, R1Independently selected from halogen, C1-6Alkyl radical, C1-6Alkyl halidesBase, C1-6Alkoxy or C1-6A haloalkoxy group. More preferably, R1Independently selected from halogen, C1-6Alkoxy or C1-6A haloalkoxy group. More preferably, R1Is C1-6Alkoxy groups, such as methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, tert-butoxy, sec-butoxy, n-pentoxy, n-hexoxy and 1, 2-dimethylbutoxy. More preferably, R1Is C1-4Alkoxy groups, such as methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, tert-butoxy and sec-butoxy. More preferably, R1Is halogen, such as Cl or Br. More preferably, R1Is Cl or methoxy. Most preferably, R1Is methoxy.
Preferably, in this embodiment, R2Independently selected from halogen, C1-6Alkyl or C1-6A haloalkyl group. More preferably, R2Is C1-6A haloalkyl group. More preferably, R2Is C1-4A haloalkyl group. More preferably, R2Is C1-2Haloalkyl radicals, e.g. CF3、-CH2F、-CHF2、-CHFCH2F、-CH2CHF2、-CF2CF3、-CCl3、-CH2Cl and-CHCl2. Most preferably, R2is-CF3
Preferably, in this embodiment, L is selected from: 1) c, N or an O atom; optionally substituted by RaSubstitution; 2) c1-6Alkylene radical, C3-10Carbocyclylene, 3-to 10-membered heterocyclylene, C6-14Arylene radicals or C5-10Heteroarylene group, more preferably, C1-6Alkylene radical, C3-10Carbocyclylene or 3 to 10 membered heterocyclylene, more preferably 3 to 6 membered heterocyclylene, more preferably 5 to 6 membered heterocyclylene; optionally substituted by RaSubstitution; 3) -C (═ O) -, -C (═ O) O-, -C (═ O) N (R)b)-、-N(Rb)C(=O)-、-N(Rb)S(=O)2-、-OC(=O)-、-OS(=O)-、-OS(=O)2-, -S (═ O) -, or-S (═ O)2-, evenPreferably, -C (═ O) -, -C (═ O) O-, -C (═ O) N (R)b)-、-N(Rb) C (═ O) -or-OC (═ O) -, more preferably, -C (═ O) -, -C (═ O) O-or-OC (═ O) -, most preferably, -C (═ O) -.
Preferably, in this embodiment, RaSelected from H, halogen, -CN, oxo, C1-6Alkyl or C1-6A haloalkyl group. More preferably, RaSelected from H, halogen or oxo. Most preferably, RaIs H.
Preferably, in this embodiment, RbIs selected from H or C1-6An alkyl group. More preferably, RbIs selected from H or C1-4An alkyl group.
Preferably, in this embodiment, ring A is selected from C3-6Carbocyclyl or 3-to 10-membered heterocyclyl. More preferably, ring A is selected from C5-6Carbocyclyl or 3-to 6-membered heterocyclyl. Most preferably, ring a is a 5-to 6-membered heterocyclyl.
Preferably, in this embodiment, R3Selected from halogen, -CN, -NO2、-Rc、-C(=O)Rc、-C(=O)ORc、-C(=O)N(Rc)(Rc)、-N(Rc)(Rc)、-N(Rc)C(=O)Rc、-N(Rc)S(=O)Rc、-N(Rc)S(=O)2Rc、-ORc、-OC(=O)Rcor-S (═ O)2RcAs chemically permitted. More preferably, R3Is selected from-Rc、-C(=O)Rc、-C(=O)ORc、-C(=O)N(Rc)(Rc) or-S (═ O)2RcAs chemically permitted.
Preferably, in this embodiment, RcSelected from H, halogen, -CN, -NO2、C1-6Alkyl radical, C1-6Haloalkyl, 3-to 10-membered heterocyclyl. More preferably, RcSelected from H, C1-6Alkyl radical, C1-6Haloalkyl, 5-to 6-membered heterocyclyl. In a preferred embodiment, RcThe heterocyclic group in (1) is optionally substituted by C1-6Alkyl radical, C1-6Haloalkyl or-C (═ O) C1-6Alkyl radical takingAnd (4) generation. In a preferred embodiment, RcThe heterocyclic group in (a) is optionally substituted by-C (═ O) C1-6Alkyl substitution.
Preferably, in this embodiment, m is 0 or 1. In a preferred embodiment, m is 0. In another preferred embodiment, m is 1.
Preferably, in this embodiment, n is 1 or 2. More preferably, n is 1.
Preferably, in this embodiment, both p and q represent a single bond. In a preferred embodiment, p represents a single bond and q represents a double bond. In another preferred embodiment, p represents a double bond and q represents a single bond.
Preferably, in this embodiment, R4、R5And R6Independently selected from H, halogen, -CN, C1-6An alkyl group. More preferably, R4、R5And R6Are all H.
Preferably, in this embodiment, W is selected from N or O atoms.
In another embodiment, the invention relates to compounds of formulae (IIa) to (IIe):
Figure GPA0000244730960000121
wherein Y is selected from C, N, O or S atom, R1-R6X, W, A, n and q are as defined above.
In another embodiment, the invention relates to compounds of formulae (IIIa) - (IIIf):
Figure GPA0000244730960000122
wherein Y is selected from C, N, O or S atom, R1-R6、RaX, W, A, n, p and q are as defined above.
In another embodiment, the invention relates to compounds of formulae (IVa) - (IVo):
Figure GPA0000244730960000131
wherein Y is selected from C, N, O or S atom, R1-R6、RaX, W, A, n, p and q are as defined above.
In another embodiment, the invention relates to compounds of formulae (Va) - (Vo):
Figure GPA0000244730960000132
Figure GPA0000244730960000141
wherein Y is selected from C, N, O or S atom, o is 0, 1, 2 or 3, R1-R6、RaX, W, A, n and q are as defined above.
In another embodiment, the invention relates to compounds of formulae (VIa) - (VIf):
Figure GPA0000244730960000142
wherein Y is selected from C, N, O or S atom, R1-R6W, A, n and q are as defined above.
In a more specific embodiment, the present invention relates to compounds of the formula:
Figure GPA0000244730960000143
wherein R is1Is Cl or methoxy, W is-O-or-NH-, R3And n is as defined above.
The compounds of the invention may include one or more asymmetric centers and may therefore exist in a variety of stereoisomeric forms, for example, enantiomeric and/or diastereomeric forms. For example, the compounds of the present invention may be 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.
The present invention also includes all suitable isotopic variations of the compounds of the present invention. Isotopic variations of the compounds of the present invention are defined as wherein at least one atom is replaced by an atom having the same atomic number but an atomic mass different from the atomic mass usually found in nature. Examples of isotopes that can be incorporated into the compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, fluorine and chlorine, for example each2H、3H、13C、14C、15N、17O、18O、18F、31P、32P、35S and36and (4) Cl. Some isotopic variations of the compounds of the present invention, for example, in which a radioactive isotope such as3H or14C, useful in drug and/or substrate tissue distribution studies. Tritiated (i.e. by tritiation)3H) And carbon-14 (i.e.14C) Isotopes are particularly preferred for their ease of preparation and detectability. Furthermore, with isotopes (e.g. deuterium, i.e.2H) Substitution may provide some therapeutic advantages due to greater metabolic stability, such as increased in vivo half-life or reduced dosage requirements, and thus may be preferred in some circumstances. Isotopic variations of the compounds of the present invention can generally be prepared by: conventional procedures are, for example, by illustrative methods or by preparation as described in the examples below, using appropriate isotopic variations of suitable reagents.
The inventors of the present invention have made studies and unexpectedly found that the deuterated diaminopyrimidine compound and the pharmaceutically acceptable salt thereof according to the present invention have equivalent or superior pharmacokinetic and/or pharmacodynamic properties to non-deuterated compounds, and thus are suitable as compounds for inhibiting EGFR kinase, and further are more suitable for preparing drugs for treating cancers and diseases related to EGFR kinase.
The compounds of the present invention or pharmaceutically acceptable salts thereof may be in amorphous or crystalline form. 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.
One skilled in the art will appreciate that many organic compounds can form complexes with a solvent in which they react or from which they precipitate or crystallize out. 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.
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, to its active form with a medicinal effect. Pharmaceutically acceptable Prodrugs are described in t.higuchi and v.stella, Prodrugs 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.bara "Improved oral Drug Delivery: solubility limits overview by the use of drivers, Advanced Drug Delivery Reviews (1996)19(2)115-130, each of which is incorporated herein by reference.
A prodrug is any covalently bonded carrier that releases a compound of formula (I) 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, amine, or sulfhydryl group is bonded to any group that, when administered to a patient, cleaves to form the hydroxy, amine, or sulfhydryl group. Thus, representative examples of prodrugs include, but are not limited to, acetate, formate and benzoate derivatives of alcohol, sulfhydryl and amine functional groups of compounds of formula (I). In addition, in the case of carboxylic acid (-COOH), esters such as methyl ester, ethyl ester, and the like may 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.
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, polyvinylpyrrolidone, 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, provided kits 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 following formulation examples illustrate representative pharmaceutical compositions that can be prepared according to the present invention. However, the present invention is not limited to the following pharmaceutical compositions.
Exemplary formulation 1-tablet: the compound of the invention in dry powder form may be mixed with the dry gel binder in a weight ratio of about 1: 2. A smaller amount of magnesium stearate was added as a lubricant. The mixture is shaped in a tablet press to 0.3-30mg tablets (each tablet containing 0.1-10mg of active compound).
Exemplary formulation 2-tablet: the compound of the invention in dry powder form may be mixed with the dry gel binder in a weight ratio of about 1: 2. A smaller amount of magnesium stearate was added as a lubricant. The mixture is shaped in a tablet press into 30-90mg tablets (each tablet containing 10-30mg of active compound).
Exemplary formulation 3-tablet: the compound of the invention in dry powder form may be mixed with the dry gel binder in a weight ratio of about 1: 2. A smaller amount of magnesium stearate was added as a lubricant. The mixture is shaped in a tablet press to form 90-150mg tablets (each tablet containing 30-50mg of active compound).
Exemplary formulation 4-tablet: the compound of the invention in dry powder form may be mixed with the dry gel binder in a weight ratio of about 1: 2. A smaller amount of magnesium stearate was added as a lubricant. The mixture is shaped in a tablet press into 150-240mg tablets (each containing 50-80mg of active compound).
Exemplary formulation 5-tablet: the compound of the invention in dry powder form may be mixed with the dry gel binder in a weight ratio of about 1: 2. A smaller amount of magnesium stearate was added as a lubricant. The mixture is shaped in a tablet press to 240-270mg tablets (each containing 80-90mg of active compound).
Exemplary formulation 6-tablet: the compound of the invention in dry powder form may be mixed with the dry gel binder in a weight ratio of about 1: 2. A smaller amount of magnesium stearate was added as a lubricant. The mixture is shaped in a tablet press into 270-450mg tablets (each containing 90-150mg of active compound).
Exemplary formulation 7-tablet: the compound of the invention in dry powder form may be mixed with the dry gel binder in a weight ratio of about 1: 2. A smaller amount of magnesium stearate was added as a lubricant. The mixture was shaped into 450-900mg tablets (each tablet containing 150-300mg of active compound) in a tablet press.
Exemplary formulation 8-capsule: the compound of the invention in dry powder form may be mixed with a starch diluent in a weight ratio of about 1: 1. The mixture is filled into 250mg capsules (each containing 125mg of active compound).
Exemplary formulation 9-liquid: the compound of the present invention (125mg) may be mixed with sucrose (1.75g) and xanthan gum (4mg), and the resulting mixture may be blended, passed through a No.10 mesh U.S. sieve, and then mixed with a previously prepared aqueous solution of microcrystalline cellulose and sodium carboxymethylcellulose (11: 89, 50 mg). Sodium benzoate (10mg), flavouring and colouring agents were diluted with water and added with stirring. Sufficient water may then be added to give a total volume of 5 mL.
Exemplary formulation 10-injection: the compounds of the present invention may be dissolved or suspended in aqueous media, which may be injected in buffered sterile saline, to a concentration of about 5 mg/mL.
Administration of drugs
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 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 through the body, e.g., intramuscular or subcutaneous bolus doses result in 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, resulting in a rapid rise in the concentration of the active ingredient in the blood 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 suitable for the purpose of producing 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 5 mg/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 compositions are included within the scope of the present invention.
The compounds of the present invention may also be administered by transdermal means. Thus, transdermal administration can be achieved using a reservoir (reservoir) or porous membrane type, or a patch of various solid matrices.
The above components of the compositions for oral, injectable 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 substitution. 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).
Combination of
The compounds of the present invention or compositions thereof may be administered in combination with other therapeutic agents to treat the disease. Examples of known therapeutic agents include, but are not limited to, Adriamycin (Adriamycin), dexamethasone (dexamethasone), vincristine (vincristine), cyclophosphamide (cyclophosphatimide), fluorouracil (fluorouracil), topotecan (topotecan), taxol (taxol), interferon, platinum derivatives, taxanes (taxanes) (e.g., paclitaxel (paclitaxel)), vinca alkaloids (e.g., vinblastine (vinblastine)), anthracyclines (anthracyclines) (e.g., doxorubicin (doxorubicin)), epidophyllotoxins (epidophyllotoxins) (e.g., etoposide (etoposide)), cisclin (cistatin), mTOR inhibitors (e.g., rapamycin (rapamycin)), methotrexate (methtrexex), cisplatin (actinomycin D), actimycin D (methacin 10), betamethacin (10), colchicine (gentamycin), clonorcipramine (trimethophylline), tranexamycin (trimethoprim), tranexamine (trimethoprim (paconitine), tranexamine (paconitine), tranine (paconitine), tranexamine (paconitine), paconitine (paconitine), paconitine (paclobulin), paclobulin (paclobulin), paclobulin (paclobulin), paclobulin (paclobulin), paclobulin (paclobulin), paclobulin (paclobulin), paclobulin), paclobulin (paclobulin), paclobulin (pactamide (paclobulin), pactamide (paclobulin (pactamide), paclobulin), pactamide (paclobulin), paclobulin (paclobulin), paclobulin (paclobulin), paclobulin (paclobulin), paclobulin (paclobulin), paclobulin (paclobulin), paclobulin (paclobulin), paclobulin, Amphotericin (amphotericin), alkylating agents (e.g. chlorambucil (chlorambucil)), 5-fluorouracil, camptothecin (camptothecin), cisplatin, metronidazole (metronidazole) and GleevecTM. In other embodiments, the inventionThe compounds of the invention are administered in combination with a biological agent such as Avastin (Avastin) or Victib (VECTIBIX).
In some embodiments, a compound of the invention or a composition thereof may be administered in combination with an antiproliferative or chemotherapeutic agent selected from any one or more of the following: abarelix (abarelix), aldesleukin (aldesleukin), alemtuzumab (alemtuzumab), alitretinoin (alitretinoil), allopurinol (allopurinol), altretamine (altretamine), amifostine (amifostine), anastrozole (anastrozole), arsenic trioxide, asparaginase, azacitidine (azacitidine), BCG Live, bevacizumab (bevacizumab), fluorouracil, bepotastine (berotene), bleomycin (bleomycin), bortezomib (bortezomib), busulfan (busufan), dimethyltestosterone (calsterone), capecitabine (capecitabine), camptothecin, carboplatin (carbomoplatin), carmustine (carmustine), celecoxib (actinomycin), mellituracil (alcazalide), melracanthine (alcazalide), mellituramycin (doxycycline), melphalan (doxycycline), mellitorine (doxycycline), melphalan (doxycycline), mellitorine (doxycycline), clorac (doxycycline), mellitorine (doxycycline), doxycycline (doxycycline), clovir), clorac (doxycycline), norbixin (doxycycline), clofibrate), clovir (doxycycline), clovir), norbixin (doxycycline), norbixin (doxycycline), norbixin (doxycycline), docetaxel (docetaxel), doxorubicin hydrochloride, dromostanolone propionate, epirubicin, epoetin alpha (epoetin alfa), erlotinib (erlotinib), estramustine (estramustine), etoposide phosphate, etoposide, exemestane (exemestane), filgrastim (filgrastim), floxuridine (floxuridine), fludarabine (fludarabine), fulvestrant (fulvestrant), gefitinib (gemcitabine), gemtuzumab (gemtuzumab), goserelin acetate (goserelin acetate), histidinolin acetate (histrelin acetate), hydroxyurea (hydroxyuride), ibritumomab (ibritumomab), daltepatin (idamycin), idarubicin (isomycin), ifosfamide (ifosfamide), interferon alpha (interferon 2-imidazole), interferon alpha (interferon), interferon (interferon alpha (interferon), etoposide (etoposimide, etoposide), etoposidrafosfatin (epirubicin), valsartan (valsartan), flutemustine (tamicidine), flunomilide (valbutine), flunomide (valbutine), flunaridine (valbutrin (vallate (valtretin), flunaridine), flunarine (valtretin (valtamide (valtretin), flunarine (valtretin (valtrexatilide), ritorine), rituximab (valtamide), flunarine (valtamide), flunarine), flunaride), flunarine (valtamide), flunaride), flunarine), flunaride (valtamsultamide (valtamsultamsultamsultamsultamide (valtamide (valtamsultamide (valtamide), valtamsultamide), valtamide), flunaride), valtamide (valtamsultamide (valtamide), valtamide (valtamsultamsultamide (valtamide), valtamide (valtamsultamsultamsultamide (valtamide), or valtamide (valtamide), valtamide (valtamsultamsultamsultamsultamsultamide), or (valtamsultamide (valtamide), or valtamide (valtamsultamsultamsultamsultamide), or valtamide), or valtamsultamide), or valtamsultamsultamsultamide (valtamsultamide), or valtamsultamide (valtamide), or valtamide (valtam, Leuprolide acetate, levamisole (levamisole), lomustine (lomustine), megestrol acetate (megestrol acetate), melphalan (melphalan), mercaptopurine (mertepurine), 6-MP, mesna (mesna), methotrexate, psoralen (methoxsalen), mitomycin C (mitomycin C), mitotane (mitotane), mitoxantrone (mitoxantrone), nandrolone (nandrolone), nelarabine (nellabine), norflumumab (nofetumomab), orelbumin (oprecin), oxaliplatin (oxaliplatin), paclitaxel, palifermin (palifermin), pamidronate (pamidronate), capecitase (pegamustine), pemetrexene (pegapterin), pegapterin (pegapterin), pegapterin (pegapterin), pegapterin (pegapterin), pegapterin (pegapterin), pegapterin (pegapterin), pegapterin (pegapterin), pegapterin (pegapterin), pegapterin (pegapterin), pegapterin (pegapterin), pegapterin (pegylate), pegapterin (pegapterin), pegapterin (pegylate), pegapterin (pegapterin), pegapterin (pegapterin), pegylate), pegapterin (pegapterin), peg, Labyrinase (rasburicase), rituximab (rituximab), sargrastim (sargramostim), sorafenib (sorafenib), streptozocin (streptazocin), sunitinib maleate (sunitinib maleate), talc, tamoxifen (tamoxifen), temozolomide (temozolomide), teniposide, VM-26, testolactone (testolactone), thioguanine (thioguanine), 6-TG, thiotepa (thiotepa), topotecan, toremifene (toremifene), tositumomab (tositumomab), trastuzumab (stuzuzumab), retinoic acid (tretinoin), ATRA, uracil mustard (uracil), valbixin (valrubicin), vinblastine (vincristine), vinorelbine (vinblastine), vinorelbine (vinzoline), or vinzolirtine (zoledrine).
Other examples of therapeutic agents that the compounds of the invention may also be combined include, but are not limited to: therapeutic agents for Alzheimer's Disease, such as donepezil hydrochloride (donepezil hydrochloride) and rivastigmine (rivastigmine); therapeutic agents for Parkinson's Disease, such as L-DOPA/carbidopa (carbidopa), entacapone (entacapone), ropinirole (roprole), pramipexole (pramipexole), bromocriptine (bromocriptine), pergolide (pergolide), trihexyphenidyl (trihexyphenyl) and amantadine (amantadine); therapeutic agents for Multiple Sclerosis (MS), such as interferon-beta, glatiramer acetate, and mitoxantrone; therapeutic agents for asthma, such as albuterol (albuterol) and montelukast; therapeutic agents for schizophrenia, such as repulper (zyprexa), rispedal (risperdal), selekang (seroquel), and haloperidol (haloperidol); anti-inflammatory agents, such as corticosteroids, TNF blockers, IL-1RA, azathioprine (azathioprine), cyclophosphamide, and sulfasalazine (sulfasalazine); immunomodulators and immunosuppressants, such as cyclosporin (cyclosporine), tacrolimus (tacrolimus), rapamycin, mycophenolate mofetil (mycophenolate mofetil), interferons, corticosteroids, cyclophosphamide, azathioprine, and sulfasalazine; neurotrophic factors such as acetylcholinesterase inhibitors, MAO inhibitors, interferons, anticonvulsants, ion channel blockers, riluzole (riluzole), and anti-parkinson agents; therapeutic agents for cardiovascular diseases, such as beta blockers, ACE inhibitors, diuretics, nitrates, calcium channel blockers and statins (statins); therapeutic agents for liver diseases, such as corticosteroids, cholestyramine (cholestyramine), interferon, and antiviral agents; therapeutic agents for blood disorders, such as corticosteroids, anti-leukemic agents, and growth factors; and therapeutic agents for immunodeficiency disorders, such as gamma globulin.
Those other active agents may be administered separately from the composition containing the compound of the present invention as part of a multiple dosing regimen. Alternatively, those active agents may be part of a single dosage form, mixed together with the compounds of the present invention in a single composition. If administered as part of a multiple dosing regimen, the two active agents may be provided simultaneously, sequentially or at intervals from one another (typically within 5 hours of one another).
Treatment of
The present invention provides a method of inhibiting a protein tyrosine kinase (e.g., EGFR kinase) or treating a disease (e.g., cancer, a cell proliferative disorder, inflammation, infection, an immunological disorder, organ transplantation, a viral disorder, a cardiovascular disorder or a metabolic disorder) comprising the steps of: administering to a subject in need of treatment a compound of the present invention, or a pharmaceutically acceptable salt, stereoisomer, solvate, hydrate, crystalline form, prodrug, or isotopic variant thereof, or administering a pharmaceutical composition as described herein.
The compounds of the invention are useful for treating EGFR-driven cancers. In particular, the compounds are useful for treating EGFR-driven cancers that express EGFR mutants and for treating EGFR-driven cancers that are refractory to RTKI therapy (e.g., erlotinib or gefitinib).
The compounds of the invention are inhibitors of at least one mutant of EGFR and are therefore useful in the treatment of one or more conditions associated with the activity of one or more EGFR mutants (e.g., deletion mutations, activation mutations, resistance mutations, or combinations thereof, specific examples include the T790M mutation, the L858R mutation, and the L858R/T790M double mutation). Thus, in a specific embodiment, the present invention provides a method of treating a mutant EGFR-mediated disorder, comprising the step of administering to a patient in need thereof a compound of the present invention, or a pharmaceutically acceptable salt, stereoisomer, solvate, hydrate, crystal form, prodrug or isotopic variant thereof, or a pharmaceutical composition described herein.
Cancers treatable by the compounds of the invention include, but are not limited to: hyperproliferative diseases such as non-small cell lung cancer (NSCLS), small cell lung cancer, lung adenocarcinoma, lung squamous carcinoma, pancreatic cancer, breast cancer, prostate cancer, liver cancer, skin cancer, epithelial cell cancer, gastrointestinal stromal tumor, leukemia, histiocytic lymphoma, and nasopharyngeal carcinoma. In addition, the compounds of the present invention may also be used to maintain the effect of preventing cancer recurrence in patients in need of such treatment.
An effective amount of a compound of the invention is generally in a mean daily dose of from 0.01mg to 50mg of compound per kilogram of body weight of the patient, preferably from 0.1mg to 25mg of compound per kilogram of body weight of the patient, in single or multiple administrations. Generally, the compounds of the present invention may be administered to such patients in need of such treatment in a daily dosage range of from about 1mg to about 3500mg, preferably from 10mg to 1000mg per patient. For example, the daily dose per patient may be 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250, 300, 350, 400, 500, 600, 700, 800, 900 or 1000 mg. Administration may be once or more daily, weekly (or at intervals of several days), or on an intermittent schedule. For example, the compound may be administered one or more times per day on a weekly basis (e.g., monday), indefinitely or for several weeks, e.g., 4-10 weeks. Alternatively, the administration may be continued daily for several days (e.g., 2-10 days), followed by several days (e.g., 1-30 days) without administration of the compound, with the cycle repeated indefinitely or for a given number of times, e.g., 4-10 cycles. For example, the compounds of the invention may be administered daily for 5 days, followed by 9 days, followed by 5 days, followed by 9 days, and so on, with the cycle repeated indefinitely or 4-10 times in total.
When RTKI (e.g., erlotinib or gefitinib) is used in combination with a compound of the present invention, the individual components of the combination therapy may be administered at the dosage levels and at the regimen of their monotherapy. For example, erlotinib, for the treatment of NSCLC, has been administered orally at 150mg per day, and for pancreatic cancer, has been administered orally at 100mg per day. In another example, gefitinib has been administered orally at 250mg per day for the treatment of NSCLC.
Preferably, when RTKI (e.g., erlotinib or gefitinib) is used in combination with a compound of the present invention, the dosage level of one or both components is reduced as compared to when used alone.
Examples
The following examples are put forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how the methods and compounds claimed herein are made, prepared, and evaluated, and are intended to be purely exemplary of the invention and are not intended to limit the scope of what the inventors regard as their invention.
Synthesis method
The compounds of the present invention may be prepared according to conventional methods in the art using appropriate reagents, starting materials and purification methods known to those skilled in the art.
Scheme 1
Figure GPA0000244730960000221
Scheme 1 illustrates the preparation of intermediates. Specifically, the intermediate may be prepared by the following steps: 5-substituted 2, 4-dichloropyrimidines, which are commercially available or obtained from known starting materials via reactions well known in the art, are coupled with a protected 3-substituted aniline under typical coupling conditions, the protecting group is removed, and then the intermediate is coupled with a substituted acrylic acid or a derivative thereof (e.g., acryloyl chloride) to provide an intermediate.
Scheme 2
Figure GPA0000244730960000222
Scheme 2 illustrates several preparations of another intermediate. Specifically, the intermediate may be prepared by the following steps: 2-substituted 4-halonitrobenzene (which is commercially available or obtained from known starting materials via reactions well known in the art) is subjected to a coupling reaction, reduction reaction to give an intermediate; or the 3-substituted 4-nitrobenzoic acid (ester) is reduced, halogenated, coupled and reduced to obtain an intermediate.
Scheme 3
Figure GPA0000244730960000231
Scheme 3 illustrates the preparation of compounds of formula (I). Specifically, intermediates prepared by schemes 1 and 2 are prepared via coupling reactions.
The process for the preparation of the compounds of formula (I) according to the invention is described in more detail below, but these particular processes do not set any limit to the invention. The compounds of the present invention may also be conveniently prepared by optionally combining various synthetic methods described in the present specification or known in the art, and such combinations may be readily carried out by those skilled in the art to which the present invention pertains.
In general, in the preparation, each reaction is usually carried out in an inert solvent at a temperature ranging from room temperature to reflux temperature (e.g., from 0 ℃ to 100 ℃, preferably from 0 ℃ to 80 ℃). The reaction time is usually 0.1 to 60 hours, preferably 0.5 to 24 hours.
Example 1
N- (3- ((2- ((4- (1-acetylpiperidin-4-yl) oxy) -2-methoxyphenyl) amino) -5- (trifluoromethyl) pyrimidine Pyridin-4-yl) amino) phenyl) acrylamide
Figure GPA0000244730960000232
Step 1:
Figure GPA0000244730960000233
4-hydroxypiperidine-1-carboxylic acid tert-butyl ester (4.70g, 23.3mmol), 4-fluoro-2-methoxynitrobenzene (2g, 11.6mmol), tetrabutylammonium bromide (0.754g, 2.34mmol) and aqueous potassium hydroxide solution in toluene were stirred at 60 ℃ overnight. The reaction mixture was allowed to return to room temperature, diluted with water and extracted with ethyl acetate. The collected organic phase was dried and purified by column chromatography to give tert-butyl 4- (3-methoxy-4-nitrophenoxy) piperidine-1-carboxylate (3.73g, 91% yield) as a yellow oily product.1H NMR(300MHz,CDCl3)(δ/ppm):8.01(d,J=9.6Hz,1H),6.56-6.49(m,2H),4.61-4.57(m,1H),3.96(s,3H),3.75-3.67(m,2H),3.44-3.36(m,2H),2.01-1.94(m,2H),1.84-1.76(m,2H),1.45(s,9H)。LC-MS(APCI):m/z=353.2(M+1)+Purity: 97.6 percent.
Step 2:
Figure GPA0000244730960000234
4- (3-methoxy-4-nitrophenoxy) piperidine-1-carboxylic acid tert-butyl ester (0.5g, 1.42mmol) was added to a three-necked flask containing a mixture of ethanol (15mL), water (5mL), then iron powder (0.48g, 8.51mmol) and ammonium chloride (38mg, 0.71mmol) were added successively and stirred for 1h, after ethanol removal by low pressure rotary evaporation, water was added and extraction was performed with dichloromethaneThe organic phase was collected and spin-dried to give tert-butyl 4- (4-amino-3-methoxy-phenoxy) piperidine-1-carboxylate (0.25g, 56.7% yield) as a light brown oil. LC-MS (APCI): m/z 323.2(M +1)+Purity: 90.5 percent.
And step 3:
Figure GPA0000244730960000241
is equipped with magnetic stirring, N2Adding N-butyl alcohol (9mL) into a 50mL three-necked bottle of a ball thermometer, cooling in an ice-water bath, keeping the internal temperature not higher than 5 ℃, adding tert-butyl 3-aminophenyl) carbamate (0.96g, 4.6mmol), slowly dropwise adding 2, 4-dichloro-5-trifluoromethylpyrimidine (1.0g, 4.6mmol), then slowly dropwise adding N, N-diisopropylethylamine (0.67g, 5.5mmol) into the reaction solution, stirring the mixed solution in the ice-water bath for 1h, stirring for 4h at normal temperature, filtering a large amount of generated white solid, washing with N-butyl alcohol (2mL), pumping, and drying in vacuum at 50 ℃ to obtain 1.4g of the white solid with the yield of 78.3%.1H NMR(400MHz,DMSO-d6)(δ/ppm):9.53(s,1H),9.46(s,1H),8.57(s,1H),7.58(s,1H),7.28-7.26(m,2H),7.04-7.01(m,1H),1.48(s,9H),LC-MS(APCI):m/z=389.1(M+1)+Purity: 97 percent.
And 4, step 4:
Figure GPA0000244730960000242
dichloromethane (15mL) was added to a 100mL three-necked flask, tert-butyl (3- ((2-chloro-5- (trifluoromethyl) pyrimidin-4-yl) amino) phenyl) carbamate (1.0g, 2.57mmol) was added with stirring, cooled in an ice-water bath, and trifluoroacetic acid (3mL) was added dropwise, the ice bath was removed, and N was added dropwise2The reaction is stirred for 1h at normal temperature under the atmosphere. Dichloromethane (50mL) was added again, triethylamine (5.6mL, 40.4mmol) was added dropwise to neutralize the trifluoroacetic acid while cooling in an ice salt bath, the mixture was cooled to-10 ℃, acryloyl chloride (0.27g, 3mmol) was added dropwise slowly under nitrogen atmosphere, and stirring was maintained for 5 min. Addition of H2The reaction was quenched with O (100mL), the organic layer was separated, and water (100 mL. times.2) Washed with 0.5M HCl (aq., 15mL), saturated aqueous sodium bicarbonate (15mL), dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated, and the residue was passed through a silica gel column (100 mesh 200 mesh; petroleum ether: ethyl acetate: 3: 1) to give the title compound 0.6g, in 68.1% yield over two steps.1H NMR(300MHz,DMSO-d6)δ(ppm):10.25(s,1H),9.59(s,1H),8.59(s,1H),7.80-1.79(m,1H),7.51(d,J=7.8Hz,1H),7.36(t,J=7.8Hz,1H),7.14(d,J=7.8Hz,1H),6.50-6.41(m,1H),6.30-6.23(m,1H),5.77(dd,J=9.3Hz,2.1Hz,1H).LC-MS(APCI):m/z=343.1(M+1)+Purity: 98 percent.
And 5:
Figure GPA0000244730960000243
n- (3- (2-chloro-5-trifluoromethylpyrimidin-4-ylamino) phenyl) acrylamide (0.21g, 0.62mmol) and trifluoroacetic acid (three drops) were added to a solution of tert-butyl 4- (4-amino-3-methoxy-phenoxy) piperidine-1-carboxylate (0.2g, 0.62mmol) in 1, 4-dioxane, brought to 60 ℃ and stirred for 3 h. After the reaction mixture was cooled to room temperature, 3 drops of triethylamine were added, the mixture was distilled under reduced pressure, and the crude product was separated by column chromatography (PE: EA: 4: 1) to give a white solid product (240mg, 61.3% yield).1H NMR(300MHz,DMSO-d6)(δ/ppm):10.15(s,1H),8.69(br s,1H),8.29(s,1H),8.16(s,1H),7.75(br s,1H),7.53-7.48(m,2H),7.25(t,J=7.5Hz,1H),7.16-7.15(m,1H),6.58(d,J=2.4Hz,1H),6.49-6.40(m,1H),8.29-8.22(m,2H),5.76(dd,J=9.9Hz,2.4Hz,1H),4.46-4.44(m,1H),3.75(s,3H),3.67-3.62(m,3H),3.30-3.18(m,2H),1.85-1.83(m,2H),1.50-1.45(m,2H),1.41(s,9H)。LC-MS(APCI):m/z=629.2(M+1)+Purity: 95.6 percent.
Step 6:
Figure GPA0000244730960000251
trifluoroacetic acid (2mL) was added to a solution of tert-butyl 4- (4- (4- (3-acrylamidophenylamino) -5-trifluoromethylpyrimidin-2-ylamino) -3-methoxyphenoxy) piperidine-1-carboxylate (200mg) in dichloromethane (15mL) and stirred for 2 h. Spin-dry and use directly in the next step.
And 7:
Figure GPA0000244730960000252
triethylamine (60mg, 0.6mmol) was added to a solution of N- (3- (2- (2-methoxy-4- (piperidin-4-yloxy) phenylamino) -5-trifluoromethylpyrimidin-4-ylamino) phenyl) acrylamide (0.1g, 0.19mmol) in dichloromethane (10mL), cooled to 0 deg.C, and Ac was added under a nitrogen atmosphere2O (25mg, 0.25mmol), stirred for 10min, added water, extracted with dichloromethane, dried over anhydrous sodium sulfate and spun dry to column to give the product N- (3- ((2- ((4- (1-acetylpiperidin-4-yl) oxy) -2-methoxyphenyl) amino) -5- (trifluoromethyl) pyrimidin-4-yl) amino) phenyl) acrylamide as a white solid (60mg, 55% yield).1H NMR(300MHz,DMSO-d6)(δ/ppm):10.16(s,1H),8.67(br s,1H),8.29(s,1H),8.18(s,1H),7.75(br s,1H),7.52-7.48(m,2H),7.25(t,J=8.4Hz,1H),7.16(br s,1H),8.59(d,J=2.7Hz,1H),6.49-6.40(m,1H),6.29-6.22(m,2H),6.76(dd,J=9.9Hz,2.4Hz,1H),4.51-4.49(m,1H),3.84-3.81(m,1H),3.77(s,3H),3.66-3.63(m,1H),3.30-3.23(m,2H),2.02(s,3H),1.86-1.81(m,2H),1.58-1.55(m,2H)。LC-MS(APCI):m/z=571.2(M+1)+Purity: 97.5 percent.
Example 2
N- (3- ((2- ((4- (1-acetylpiperidin-4-yl) amino) -2-methoxyphenyl) amino) -5- (trifluoromethyl) pyrimidine Pyridin-4-yl) amino) phenyl) acrylamide
Figure GPA0000244730960000261
Step 1:
Figure GPA0000244730960000262
the synthetic procedure was the same as in step 1 of example 1 to give the product tert-butyl 4- (3-methoxy-4-nitrophenylamino) piperidine-1-carboxylate (0.96g, yield: 93.6%). LC-MS (APCI): 352(M +1)+Purity: 95.1 percent.
Step 2:
Figure GPA0000244730960000263
the synthetic procedure was the same as in step 2 of example 1 to give tert-butyl 4- (4-amino-3-methoxyphenylamino) piperidine-1-carboxylate (0.19g, yield: 91%) as a dark blue oily product, which was used in the next step as it was.
And step 3:
Figure GPA0000244730960000264
the synthesis procedure was the same as in step 3 of example 1 to give tert-butyl 4- ((4- ((4- (3-acrylamidophenylamino) -5-trifluoromethylpyrimidin-2-yl) amino-3-methoxyphenyl) amino) piperidine-1-carboxylate (89mg, yield: 54.6%) as a white solid product.
And 4, step 4:
Figure GPA0000244730960000265
the synthesis procedure was the same as in step 4 of example 1 to give the product N- (3- (2- (2-methoxy-4- (piperidin-4-ylamino) phenyl) amino) -5-trifluoromethylpyrimidin-2-yl) amino) phenyl) acrylamide, which was used directly in the next step.
And 5:
Figure GPA0000244730960000271
the synthesis procedure was the same as in example 1, step 5, to give the product N- (3- ((2- ((4- (1-acetylpiperidin-4-yl) amino) -2-methoxyphenyl) amino) -5- (trifluoromethyl) pyrimidin-4-yl) amino) phenyl) acrylamide (48mg, yield: 60.3%).1H NMR(300MHz,DMSO-d6)(δ/ppm):10.25(s,1H),8.45(br s,1H),8.23(s,1H),8.10(s,1H),7.65(br s,1H),7.52-7.49(m,1H),7.26-7.22(m,3H),6.54-6.45(m,1H),6.29-6.22(m,2H),6.05-6.90(m,1H),5.77-5.74(m,1H),5.40-5.37(m,1H),4.22-4.18(m,1H),3.78-3.82(m,1H),3.68(s,3H),3.24-3.15(m,1H),2.86-2.79(m,1H),2.00(s,3H),1.93-1.85(m,2H),1.30-1.25(m,3H)。LC-MS(APCI):m/z=570.1(M+1)+Purity: 95.6 percent.
Example 3
N- (3- ((2- ((4- (1-acetyl-1, 2, 3, 6-tetrahydropyridin-4-yl) -2-methoxyphenyl) amino) -5- (trifluoromethyl) pyrimidin-4-yl) amino) phenyl) acrylamide
Figure GPA0000244730960000272
Step 1:
Figure GPA0000244730960000273
PdCl2(dppf)2(422mg, 0.052mmol) was added to 4-bromo-2-methoxynitrobenzene (2.0g, 8.62mmol), 4- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -1, 2, 5, 6-tetrahydropyridine-1-carboxylic acid tert-butyl ester and K2CO3(3.57g, 19.38mmol) in 1, 4-dioxane (40mL) was heated to 90 ℃ under a nitrogen atmosphere and stirred for 14 h. After pouring water (150mL), it was extracted with dichloromethane (50mL x 3), dried by spinning, and the product was tert-butyl 4- (3-methoxy-4-nitrophenyl) -1, 2, 5, 6-tetrahydropyridine-1-carboxylate (1.6g, yield: 56%) as a yellow solid.1H NMR(300MHz,CDCl3)(δ/ppm):7.87(d,J=9Hz,1H),7.03-7.00(m,2H),6.19(s,1H),4.13(t,J=3Hz,2H),3.99(s,3H),3.67(t,J=6Hz,2H),1.53(s,9H)。LC-MS(APCI):m/z=335.2(M+1)+Purity: 94.6 percent.
Step 2:
Figure GPA0000244730960000274
the specific operation steps are the same as the step 4 of the example 1, and the product 4- (3-methoxy-4-nitrophenyl) -1, 2, 3, 6-tetrahydropyridine is obtained without separation: LC-MS (APCI): m/z 235.3(M +1)+Purity: 93.4 percent.
And step 3:
Figure GPA0000244730960000281
triethylamine (181.4mg, 1.79mmol) and acetic anhydride (91.5mg, 896. mu. mol) were added to the reaction mixture in the previous step, stirred at room temperature for 30min, spun dry, and passed through a column (DCM: MeOH ═ 40: 1 to 20: 1) to give 1- (4- (3-methoxy-4-nitrophenyl) -1, 2, 5, 6-tetrahydropyridin-1-yl) ethanone (150mg, yield: 90.9%) as a yellow oily product.1H NMR(300MHz,CDCl3)(δ/ppm):7.89(d,J=8.7Hz,1H),7.05-7.00(m,2H),6.25-6.16(m,1H),4.32-4.18(m,2H),4.00(s,3H),3.88-3.69(m,2H),2.64-2.62(m,2H)。LC-MS(APCI):m/z=277.3(M+1)+Purity: 98.9 percent.
And 4, step 4:
Figure GPA0000244730960000282
1N aqueous HCl (3 drops) was added to a solution of 1- (4- (3-methoxy-4-nitrophenyl) -1, 2, 5, 6-tetrahydropyridin-1-yl) ethanone (150mg, 542.9. mu. mol) and iron powder (182mg, 3.26mmol) in ethanol (10mL), refluxed at 95 ℃ for 2h, and the rotary brown oil was dissolved in dichloromethane, washed with water, dried,The organic phase was collected by washing once with brine and collecting the product 1- (4- (3-methoxy-4-aminophenyl) -1, 2, 5, 6-tetrahydropyridin-1-yl) ethanone as a yellow-black oil which was spin-dried. LC-MS (APCl): m/z 247.2(M +1)+Purity: 94.6 percent.
And 5:
Figure GPA0000244730960000283
trifluoroacetic acid (three drops) was added to a solution of 1- (4- (3-methoxy-4-aminophenyl) -1, 2, 5, 6-tetrahydropyridin-1-yl) ethanone (133mg, 540 μmol) and N- (3- ((2-chloro-5-trifluoromethylpyrimidin-4-yl) amino) phenyl) acrylamide (185mg, 540 μmol) in 1, 4-dioxane (5mL), stirred at 60 ℃ for 2h, triethylamine was added dropwise to adjust to neutrality, after extraction with dichloromethane, the column was spin-dried (DCM: MeOH ═ 40: 1) to give the product N- (3- ((2- ((4- (1-acetyl-1, 2, 3, 6-tetrahydropyridin-4-yl) -2-methoxyphenyl) amino) -5- (trifluoromethyl) pyrimidine as a white solid Pyridin-4-yl) amino) phenyl) acrylamide (90mg, yield: 30.2%).1H NMR(300MHz,CDCl3)(δ/ppm):8.32(s,1H),8.18(s,1H),7.89-7.79(m,2H),7.64-7.51(m,2H),7.38-7.34(m,1H),7.24-7.20(m,1H),6.90-6.75(m,3H),6.43(d,J=7.5Hz,1H),6.22-6.15(m,1H),6.01-5.94(d,J=18Hz,1H),5.75(d,J=6.3Hz,1H),4.23-4.11(m,2H),3.90(s,3H),3.80(t,J=3.2Hz,1H),3.64(t,J=3.2Hz,1H),2.52(d,J=13.5Hz,2H),2.17-2.14(m,3H)。LC-MS(APCI):m/z=553.3(M+1)+Purity: 97.8 percent.
Example 4
N- (3- ((2- ((4- (1-acetylpiperidin-4-yl) -2-methoxyphenyl) amino) -5- (trifluoromethyl) pyrimidine-4-) Yl) amino) phenyl) acrylamide
Figure GPA0000244730960000291
Step 1:
Figure GPA0000244730960000292
Pd/C (9mg) was added to a solution of 1- (4- (3-methoxy-4-aminophenyl) -1, 2, 5, 6-tetrahydropyridin-1-yl) ethanone (90mg, 365 μmol) in ethanol (10mL) under a hydrogen atmosphere, stirred at room temperature for 2h, filtered and spun dry to give 1- (4- (3-methoxy-4-aminophenyl) piperidin-1-yl) ethanone (90mg, 99.2%) as a yellow oily product. LC-MS (APCI): m/z 249.4(M +1)+Purity: 95.1 percent.
Step 2:
Figure GPA0000244730960000293
the synthesis procedure was the same as in example 3, step 5, to give the product N- (3- ((2- ((4- (1-acetylpiperidin-4-yl) -2-methoxyphenyl) amino) -5- (trifluoromethyl) pyrimidin-4-yl) amino) phenyl) acrylamide as a white solid (60mg, yield: 29.8%).1H NMR(300MHz,CDCl3)(δ/ppm):8.32(s,1H),8.14(d,J=8.4Hz,1H),7.89(m,1H),7.76-7.71(m,2H),7.55(br s,1H),7.39-7.28(m,2H),6.91(s,1H),6.69(d,J=2.4Hz,1H),6.61(d,J=7.5Hz,1H),6.50-6.44(m,1H),6.30-6.21(dd,J=16.8Hz,10.2Hz,1H),5.80-5.76(dd,J=10.2Hz,1.5Hz,1H),4.81-4.76(m,1H),3.96-3.87(m,4H),3.21-3.11(m,1H),2.72-2.58(m,2H),2.14(s,3H),1.92-1.83(M,2H),1.66-1.56(m,2H)。LC-MS(APCI):m/z=555.4(M+1)+Purity: 95.5 percent.
Example 5
N- (3- ((2- ((2-methoxy-4- (4- (4-methylpiperazin-1-yl) piperidin-1-yl) phenylamino) -5-trifluoro Methyl) pyrimidin-4-yl) amino) phenyl) acrylamides
Figure GPA0000244730960000294
Step 1:
Figure GPA0000244730960000295
4-fluoro-2-methoxynitrobenzene (1.0g, 5.84mmol), 1-methyl-4- (4-piperidinyl) piperazine (0.9g, 7.0mmol), K2CO3(2.0g, 14.6mmol) were added successively to a three-necked flask containing 4mL of DMF, heated to 80 ℃ and stirred overnight. The reaction solution was cooled to room temperature, diluted with water, extracted with ethyl acetate, dried over anhydrous sodium sulfate and then spin-dried to pass through a column to give 1- (1- (3-methoxy-4-nitrophenyl) piperidin-4-yl) -4-methylpiperazine (1.0g, yield: 51.1%) as a yellow solid product.1H NMR(300MHz,CDCl3) (δ/ppm): 8.01(d, J ═ 9.3Hz, 1H), 6.43(dd, J ═ 9.6Hz, 2.7Hz, 1H), 6.31(d, J ═ 2.4Hz, 1H), 3.98-3.94(m, 2H), 3.87(s, 3H), 3.03-2.94(m, 2H), 2.65-2.62(m, 4H), 2.54-2.46(m, 5H), 2.32(s, 3H), 2.01-1.96(m, 2H), 1.65-1.60(m, 2H), LC-ms (apci): m/z 335.2(M +1) +; purity: 95.0 percent.
Step 2:
Figure GPA0000244730960000301
1- (1- (3-methoxy-4-nitrophenyl) piperidin-4-yl) -4-methylpiperazine (0.2g, 0.59mmol) was added to a three-necked flask containing a mixture of ethanol (6mL) and water (2mL), then iron powder (0.2g, 3.58mmol), ammonium chloride (16mg, 0.29mmol) were added again, refluxed for 2h, after the reaction solution was cooled to room temperature, ethanol was spin-dried, and saturated NaHCO was added3(aq., 5mL), extracted with dichloromethane, the organic phases were combined and dried over anhydrous sodium sulfate and spin dried to give the product 2-methoxy-4- (4- (4-methylpiperazin-1-yl) piperidine-1-aniline (180mg, yield: 100%) as a light brown oil, LC-MS (APCI) M/z ═ 305(M +1)+Purity: 90.3 percent.
And step 3:
Figure GPA0000244730960000302
the synthesis procedure was the same as in step 5 of example 3 to give the product as a white solid (40mg, yield: 11.1%).1H NMR(300MHz,DMSO-d6)(δ/ppm):10.25(s,1H),8.45(br s,1H),8.23(s,1H),8.10(s,1H),7.65(br s,1H),7.52-7.49(m,1H),7.26-7.22(m,3H),6.54-6.45(m,1H),6.29-6.22(m,2H),6.05-6.90(m,1H),5.77-5.74(m,1H),3.77-3.72(m,2H),2.82-2.61(m,11H),2.48-2.34(m,3H),2.03(s,3H),1.91-1.85(m,2H),1.59-1.52(m,2H)。LC-MS(APCI):m/z=611.1(M+1)+Purity: 94.5 percent.
Example 6
N- (3- ((2- ((2-methoxy-4- (4-morpholinopiperidin-1-yl) phenyl) amino) -5- (trifluoromethyl) pyrimidine- 4-yl) amino) phenyl) acrylamide
Figure GPA0000244730960000303
Step 1:
Figure GPA0000244730960000311
4-fluoro-2-methoxynitrobenzene (0.5g, 2.92mmol), 4-piperidyl morpholine (0.6g, 3.5mmol), K2CO3(0.54g, 3.8mmol) was added in turn to a three-necked flask containing 3mL of DMF, and the mixture was stirred at room temperature overnight under a nitrogen atmosphere. Diluted with cold water (50mL), filtered, and dried to give 4- (1- (3-methoxy-4-nitrophenyl) piperidin-4-yl) morpholine (0.5g, 53% yield) as a yellow solid. LC-MS (APCI): m/z 322.2(M +1)+Purity: 96.3 percent.
Step 2:
Figure GPA0000244730960000312
the synthesis procedure was the same as in example 5, step 2, to give the product as a yellow solid (0.12g, 67% yield). LC-MS (APCI): m/z 292.1(M +1)+Purity: 98 percent.
And step 3:
Figure GPA0000244730960000313
the synthesis procedure was the same as in example 3, step 5, to give a brown solid product (120mg, yield 67%).1H NMR(300MHz,CDCl3)(δ/ppm):8.29(s,1H),7.97(d,J=8.4Hz,1H),7.88(s,1H),7.49-7.53(m,2H),7.33(t,J=8.7Hz,1H),7.16-7.19(br,1H),6.87(s,1H),6.48-6.52(m,2H),6.37-6.42(m,2H),6.26(q,J=10.2Hz,1H),5.77(d,J=10.8Hz,1H),3.86(s,3H),3.77(s,4H),3.64(d,J=15.6Hz,2H),2.69(d,J=16Hz,2H),2.62(s,4H),2.33(t,J=7.8Hz,1H),1.96(d,J=6.3Hz,4H)。LC-MS(APCI):m/z=598.3(M+1)+Purity: 97 percent.
Example 7
N- (3- ((2- ((4- (4-acetylpiperazine-1-carbonyl) -2-methoxyphenyl) amino) -5- (trifluoromethyl) pyrimidine- 4-yl) amino) phenyl) acrylamide
Figure GPA0000244730960000314
Step 1:
Figure GPA0000244730960000321
a solution of oxalyl chloride in dichloromethane (2.8ml, 5.6mmol) was added dropwise to a solution of 3-methoxy-4 nitrobenzoic acid (1.0g, 5.1mmol) in tetrahydrofuran (14ml) at 0 deg.C, followed by several additional drops of DMF. The reaction mixture was stirred at this temperature under nitrogen for 4h, brought to room temperature, and spun dry to give 1.2g of yellow product, which was dissolved in 5mL of dry dichloromethane. A two-necked flask containing a dichloromethane solution (15mL) of 1-acetylpiperazine (0.85g, 6.6mmol) was cooled to 0 ℃, triethylamine (1mL, 7.7mmol) was added to the mixture, the dichloromethane solution obtained in the previous step was slowly added dropwise to the reaction solution, stirred for 30min, quenched with water (20mL), extracted with dichloromethane (15mL × 2), the organic phase was collected, dried over anhydrous sodium sulfate, and spin-dried to obtain the product N-1- (4- (3-methoxy-4-nitrobenzoyl) acetylpiperazine (1.1g, 70.2% yield), LC-ms (apci): M/z 308.1(M +1) +, purity 95.1%.
Step 2:
Figure GPA0000244730960000322
the synthesis procedure was the same as in example 1, step 2, to give N-1- (4- (4-amino-3-methoxybenzoyl) acetylpiperazine (0.13g, yield: 72%) as a yellow solid product, LC-ms (apcl) M/z 278.2(M +1) +, purity: 98.3%.
And step 3:
Figure GPA0000244730960000323
the synthesis procedure was the same as in example 3, step 5, to give the product N- (3- ((2- ((4- (4-acetylpiperazine-1-carbonyl) -2-methoxyphenyl) amino) -5- (trifluoromethyl) pyrimidin-4-yl) amino) phenyl) acrylamide (110mg, 43% yield) as a brown solid.1H NMR(300MHz,CDCl3) (δ/ppm): 10.23(s, 1H), 8.97(s, 1H), 8.39(s, 1H), 8.21(s, 1H), 7.86(t, J ═ 8.1Hz, 2H), 7.54(d, J ═ 8.4Hz, 1H), 7.35(t, J ═ 7.8Hz, 1H), 7.15(d, J ═ 1.2Hz, 1H), 7.02(d, J ═ 1.8Hz, 1H), 6.65(d, J ═ 6, 1H), 6.43(dd, J ═ 17.1Hz, 9.9Hz, 1H), 6.34(dd, J ═ 17.1Hz, 1.8Hz, 1H), 5.76(s, 1H), 3.78(s, 3H), 3.24(s, 3H), 2.04 (s, 3H), 4.04 (d, 4H), 23.4H, 1H), 4-1H). LC-MS (APCI): m/z 584.2(M +1) +, purity: 97.6 percent.
Example 8
N- (3- ((2- (4- (1-acetylpiperidine-4-methylene) -2-methoxyphenyl) amino) -5- (trifluoromethyl) pyrimidine Pyridin-4-yl) amino) phenyl) acrylamide
Figure GPA0000244730960000324
Step 1:
Figure GPA0000244730960000331
under a nitrogen atmosphere, 3-methoxy-4-nitrobenzoic acid (0.48g, 1.66mmol), methanol (5mL), sulfuric acid (3mL) were sequentially added to the single-necked flask and refluxed for 2 h. Water (30mL) was added for dilution, dichloromethane (30mL) was added and the mixture was stirred for 5min, extracted with dichloromethane, the organic phase was collected, dried over anhydrous sodium sulfate, and then spin-dried on a column to give the product as a yellow oil (0.32g, 91% yield). LC-MS (APCI): m/z 212(M +1)+Purity: 95.3 percent.
Step 2:
Figure GPA0000244730960000332
under the atmosphere of nitrogen, 3-methoxy-4-nitrobenzoic acid methyl ester and tetrahydrofuran are sequentially added into a 50mL single-neck flask, cooled to 0 ℃, and added with LiAlH4(0.16g), stirred for 30min, diluted with sodium sulfate decahydrate and extracted by spin drying to give the product (3-methoxy-4-nitrophenyl) methanol as a brown oil (2.5g, 81% yield). LC-MS (APCI): 184.1(M +1) M/z+Purity: 98.1 percent.
And step 3:
Figure GPA0000244730960000333
under a nitrogen atmosphere, (3-methoxy-4-nitrophenyl) methanol (0.5g, 2.73mmol) and methylene chloride (20mL) were added in this orderInto a stirred 25mL single-necked flask, cooled to 0 deg.C, slowly added phosphorus tribromide (960mg, 3.55mmol), and stirred at room temperature for 3 h. The reaction mixture was diluted with water (30mL) and dichloromethane (30mL), stirred for 5min, extracted with dichloromethane, the organic phase was collected, washed with water, dried over anhydrous sodium sulfate and then spun through a column to give 4-bromomethyl-2-methoxynitrobenzene (0.37g, 55% yield) as a yellow oily product. LC-MS (APCI): m/z 246.1(M +1)+Purity: 94.8 percent.
And 4, step 4:
Figure GPA0000244730960000334
4-bromomethyl-2-methoxynitrobenzene (100mg, 0.4mmol) and triethyl phosphite (68mg, 0.4mmol) were added to a 100mL single-necked flask, heated to 160 ℃ and stirred at that temperature for 4h before cooling to room temperature, stirred in an ice-water bath for 1h, the filtrate collected by filtration was rotary dried to give 123mg of product as a yellow oil, LC-MS (APCI): 304(M +1) +, purity: 96.6 percent.
And 5:
Figure GPA0000244730960000335
add NaH (1.52g, 15mmol) and MsCl (1.5g, 13mmol) sequentially to a solution of diethyl 3-methoxy-4-nitrophenylphosphate (3g, 10mmol) in dichloromethane (60mL) at 0 deg.C under nitrogen, stir at room temperature for 1h, quench with water (60mL) and dichloromethane (50mL), separate the organic phase, and add water (60mL), 0.5M HCl (aq., 20mL), saturated NaHCO sequentially3The solution (20mL) was washed, dried over anhydrous sodium sulfate and then spun dry to give the product tert-butyl N-4- (3-methoxy-4-nitrophenylmethylene) piperidinecarboxylate as a pale solid (1.5g, 43% yield).1H NMR(300MHz,CDCl3)(δ/ppm):7.87(d,J=8.4Hz,1H),6.85-6.88(m,2H),6.37(s,1H),3.97(s,3H),3.55(t,J=7.8Hz,2H),3.45(t,J=7.8Hz,2H),2.45(t,J=7.8Hz,2H),2.15(t,J=7.8Hz,2H),1.51(s,9H)。LC-MS(APCI):m/z=349(M+1)+Purity: 98.4 percent.
Step 6:
Figure GPA0000244730960000341
the synthesis procedure was the same as in example 3, steps 2 and 3, to give N- (4- (3-methoxy-4-nitrophenylmethylene) acetylpiperidine (0.66g, 79% yield) as a white solid product, LC-ms (apci): M/z ═ 291.2(M +1)+Purity: 98.6 percent.
And 7:
Figure GPA0000244730960000342
n- (4- (3-methoxy-4-aminobenzylidene) acetylpiperidine (0.2g, 0.69mmol), iron powder (0.21g, 3.75mmol), NH4Cl (33mg, 0.62mmol) was added sequentially to a stirred mixture of ethanol (10mL) and water (2mL), refluxed for 2h, cooled to room temperature, ethanol was spun off, water (20mL) and dichloromethane were added and extracted to give the product N- (4- (3-methoxy-4-nitrobenzylidene) acetylpiperidine (0.13g, 73% yield) as a yellow solid, LC-MS (APCI) M/z 261.3(M +1)+Purity: 93.6 percent.
And 8:
Figure GPA0000244730960000343
the synthesis procedure was the same as in example 3, step 5, to give the product N- (3- ((2- ((4- (1-acetylpiperidine-4-methylene) -2-methoxyphenyl) amino) -5- (trifluoromethyl) pyrimidin-4-yl) amino) phenyl) acrylamide as a white solid (90mg, 38% yield).1H NMR(300MHz,CDCl3)(δ/ppm):8.32(s,1H),7.78(d,J=5.7Hz,1H),7.36(q,J=6.6Hz,1H),6.89-6.92(m,2H),6.62-6.72(m,4H),6.43-6.49(m,2H),6.34(m,2H),5.79(d,J=10.2Hz,2H),3.89(d,J=4.8Hz,3H),3.41-3.69(m,4H),2.53(q,J=6.9Hz,2H),2.39(m,2H),2.16(d,J=4.5Hz,3H)。LC-MS(APCI):m/z=567.3(M+1)+Purity: 97.1 percent.
Example 9
N- (3- ((2- (4- (1-acetylpiperidine-4-methyl) -2-methoxyphenyl) amino) -5- (trifluoromethyl) pyrimidine- 4-yl) amino) phenyl) acrylamide
Figure GPA0000244730960000344
Step 1:
Figure GPA0000244730960000351
the synthesis procedure was the same as in example 4, step 1, to give 1- (4- (4-amino-3-methoxybenzyl) piperidin-1-yl) ethanone (90mg, yield 90%) as a brown oil. LC-MS (APCI): m/z 263.2(M +1)+Purity: 94.6 percent.
Step 2:
Figure GPA0000244730960000352
the synthesis procedure was the same as in example 3, step 5, to give the product N- (3- ((2- (4- (1-acetylpiperidin-4-methyl) -2-methoxyphenyl) amino) -5- (trifluoromethyl) pyrimidin-4-yl) amino) phenyl) acrylamide as a white solid (85mg, 37% yield).1H NMR(300MHz,CDCl3)(δ/ppm):8.33(s,1H),8.09(d,J=9Hz,1H),7.90(s,1H),7.36(d,J=18Hz,2H),7.49-7.54(br,2H),7.31-7.39(m,2H),6.91(s,2H),6.64(s,1H),6.57(d,J=6.9Hz,1H),6.50(d,J=1.5Hz,1H),6.44(d,J=1.8Hz,1H),6.21-6.30(m,1H),5.79(d,J=10.2Hz,2H),5.79(dd,J=9.9Hz,1.2Hz,1H),4.59(d,J=13.5Hz,1H),3.88(s,3H),3.76-3.80(m,2H),2.94-3.03(m,2H),2.43-2.51(m,4H),2.39(m,2H),2.09(s,3H),2.53(q,J=6.9Hz,2H),127-1.32(m, 2H). LC-MS (APCI): m/z 569.4(M +1) +, purity: 94.6 percent.
Example 10
N- (3- ((2- ((4- (4-acetylpiperazin-1-yl) methyl) -2-methoxyphenyl) amino) -5- (trifluoromethyl) pyrimidine Pyridin-4-yl) amino) phenyl) acrylamide
Figure GPA0000244730960000353
Step 1:
Figure GPA0000244730960000354
the synthesis procedure was the same as in step 2 of example 1 to give a yellow solid product (0.12g, yield 68%). LC-MS (APCI): m/z 322.2(M +1)+Purity: 95.6 percent.
Step 2:
Figure GPA0000244730960000361
the synthesis procedure was the same as in example 1, step 3, to give a brown solid product (110mg, 43% yield), LC-ms (apci): m/z 628.3(M +1)+Purity: 97.6 percent.
And step 3:
Figure GPA0000244730960000362
under the protection of nitrogen, a dichloromethane solution of 4- (4- ((4- ((3-acrylamidophenyl) amino) -5-trifluoromethylpyrimidin-2-yl) amino) -3-methoxyphenyl) -methylpiperazine-1-carboxylic acid tert-butyl ester is cooled to 0 ℃, trifluoroacetic acid is added dropwise, stirring is carried out for 3h at room temperature, dichloromethane is added, cooling is carried out under an ice salt bath, triethylamine is added for neutralization, acetyl chloride is slowly added dropwise, water is added for quenching, an organic phase is separated, and water (100mL x2), 0.5M HCl (aq., 15mL) and saturated water are sequentially used for the neutralizationAnd NaHCO3The solution (15mL) was washed, dried over anhydrous sodium sulfate and then spin-dried over a column to give the product N- (3- ((2- ((4- (4-acetylpiperazin-1-yl) methyl) -2-methoxyphenyl) amino) -5- (trifluoromethyl) pyrimidin-4-yl) amino) phenyl) acrylamide as a white solid (85mg, 47% yield).1H NMR(300MHz,CDCl3)(δ/ppm):8.30(s,1H),8.06(d,J=7.8Hz,1H),7.91-7.96(m,3H),7.43(d,J=0.6Hz,1H),7.91-7.96(m,3H),7.30-7.36(m,2H),6.89(dd,J=16.5Hz,1.5Hz,2H),6.69(d,J=6.6Hz,1H),6.21-6.41(m,2H),5.76(dd,J=9.9Hz,1.5Hz,1H),3.87(s,3H),3.64(t,J=4.2Hz,2H),3.45-3.53(m,4H),2.43-2.50(m,4H),1.25(s,3H)。LC-MS(APCI):m/z=570.1(M+1)+Purity: 96.6 percent.
Biological assay
(1) Kinase inhibition
Reagents and consumables:
WT EGFR (Carna, Cat 08-115), EGFR [ L858R/T790M](Carna, Cat No. 08-510), ATP (Sigma, Cat No. A7699-1G), DMSO (Sigma, Cat No. D2650), 96-well plate (Corning, Cat No. 3365), 384-well plate (Greiner, Cat No. 784076), HTRF Kinase TK kit (Cisbio, Cat No. 62TK0PEJ), erlotinib (Selleckchem, Cat No. S7787), EGFR [ D746-750 [](Life Technologies, Cat. No. PV6178), 5 Xkinase buffer A (Life Technologies, Cat. No. PV3186), kinase tracer 199(Life Technologies, Cat. No. PV5830),
Figure GPA0000244730960000363
Eu-anti-GST antibody (Life Technologies, Cat. No. PV 5594).
The specific experimental method comprises the following steps:
compound preparation: test compounds were dissolved in DMSO to make 20mM stock. Then, the cells were diluted ten times in DMSO with a gradient of 3-fold. When adding medicine, the medicine is diluted by 10 times by using buffer solution.
EGFR and EGFR [ L858R/T790M]And (3) kinase detection: EGFR or EGFR [ L858R/T790M in 5 Xkinase buffer A]The kinase was mixed with the compound prepared in pre-dilution at various concentrations for 10 minutes, each concentrationDouble-hole. Adding corresponding substrate and ATP, reacting for 20 minutes at room temperature (wherein negative and positive controls: negative is blank control, and positive is erlotinib). Adding a detection reagent (reagent in an HTRF Kinase TK kit) after the reaction is finished, incubating for 30 minutes at room temperature, detecting by an Evnvision microplate reader, determining the enzyme activity in the presence of the compound with various concentrations, calculating the inhibitory activity of the compound with different concentrations on the enzyme activity, fitting the inhibitory activity of the compound with different concentrations on the enzyme activity according to a four-parameter equation and Graphpad 5.0 software, and calculating IC50The value is obtained.
The results of kinase inhibition in the examples are summarized in Table 1 below.
TABLE 1
Figure GPA0000244730960000371
(2) Cytotoxicity test
The in vitro antiproliferative activity of the compound of the invention on 3 tumor cells cultured in vitro is detected by adopting an MTS method. The experimental result shows that the compound has the inhibiting effect on the in vitro proliferation of cancer cells cultured in vitro; wherein the inhibition of in vitro proliferation of lung cancer cells is stronger than the inhibition of in vitro proliferation of skin cancer cells.
Cell line:
skin cancer cell a431 (purchased from american standard biological collection (ATCC)); lung cancer cells NCI-H1975 (purchased from american college of standards living things (ATCC)) and HCC827 (purchased from american college of standards living things (ATCC)); all were cultured in RPMI1640 medium containing 10% fetal bovine serum, 100U/ml penicillin, 100. mu.g/ml streptomycin.
Reagents and consumables:
RPMI-1640(GIBCO, Cat. No. A10491-01); fetal bovine serum (GIBCO, catalog No. 10099141); 0.25% trypsin-EDTA (GIBCO, cat No. 25200); penicillin-streptomycin, liquid (GIBCO, catalog number 15140-; DMSO (Sigma, cat # D2650); MTS assay kit (Promega, catalog No. G3581), 96-well plate (Corning, catalog No. 3365).
The specific experimental method comprises the following steps:
compound preparation: test compounds were dissolved in DMSO to prepare a 20mM stock solution, which was stored at-20 ℃. Diluted 3-fold with DMSO gradient and 10-fold. When adding medicine, the medicine is diluted 4 times by using cell culture medium.
MTS cell viability assay: cells in the logarithmic growth phase were digested with 0.25% trypsin-EDTA, and 150. mu.l of the compound diluted 4-fold in culture medium was inoculated into a 96-well plate at the optimized density, and 50. mu.l/well (ten concentrations: 100, 33.3, 11.1, 3.70, 1.23, 0.412, 0.137, 0.0457, 0.0152, 0.00508. mu.M were generally selected) was added after 24 hours. Wells to which the same volume of 0.5% DMSO was added served as controls. After the cells were cultured for 72 hours, the MTS measured the cell viability.
The method comprises the following specific operations: adherent cells, medium was discarded and a mixture containing 20. mu.L MTS and 100. mu.l medium was added to each well. The culture was continued for 1 to 4 hours in an incubator and then OD490 was measured using OD650 as a reference. Dose-response curves were generated and IC calculated using GraphPad Prism software50
The results of the inhibition of in vitro proliferation of cancer cells in the examples are summarized in table 2 below.
TABLE 2
Figure GPA0000244730960000381

Claims (16)

1. A compound of formula (I)
Figure FDA0003320864480000011
Wherein the content of the first and second substances,
R1independently selected from C1-6An alkoxy group;
R2independently selected from C1-6A haloalkyl group;
the linking group L is selected from C or O atoms, as valency permits; wherein C may optionally be substituted by RaSubstitution;
Raselected from H, halo or oxo;
x is selected from C or N atoms, provided that valency permits;
ring a is selected from 5-to 6-membered heterocyclyl;
R3is selected from-C (═ O) Rc
RcSelected from H, C1-6Alkyl radical, C1-6Haloalkyl or 5 to 6 membered heterocyclyl;
m is 0 or 1;
n is 1;
p represents a single or double bond, as far as valency permits;
q represents a single or double bond, provided that valency permits;
R4、R5and R6Independently selected from H, halogen, -CN, C1-6Alkyl or C1-6A haloalkyl group;
w is selected from NH;
provided that, when m is 0, X is a C atom;
or a pharmaceutically acceptable salt, stereoisomer, crystal form, or isotopic variant thereof.
2. The compound of claim 1, wherein R1Is methoxy, R2Is CF3W is-NH-and R4、R5And R6Are all H.
3. The compound of claim 1 selected from the group consisting of compounds of the formula:
Figure FDA0003320864480000012
wherein R is1-R6、RaX, W, A, n, p and q are as defined in claim 1.
4. The compound of claim 3 selected from the group consisting of compounds of the formula:
Figure FDA0003320864480000013
wherein R is1-R6、RaX, W, A, n and q are as defined in claim 1.
5. The compound of claim 1 selected from the group consisting of compounds of the formula:
Figure FDA0003320864480000021
wherein Y is selected from a C or N atom, R1-R6、RaX, W, n, p and q are as defined in claim 1.
6. The compound of claim 1 selected from the group consisting of compounds of the formula:
Figure FDA0003320864480000022
wherein Y is selected from a C or N atom, R1-R6W, n and q are as defined in claim 1.
7. The compound of claim 1 selected from the group consisting of compounds of the formula:
Figure FDA0003320864480000023
Figure FDA0003320864480000031
wherein R is1Is methoxy, W is-NH-, R3And n is as defined in claim 1.
8. The compound of any one of claims 1-7, wherein
R3Is selected from-C (═ O) Rc
RcIs selected from C1-6Alkyl or C1-6A haloalkyl group.
9. The compound of claim 8, wherein R3Selected from-C (═ O) Me or-C (═ O) CF3
10. A compound selected from the following compounds:
Figure FDA0003320864480000032
11. a pharmaceutical composition comprising a compound of any one of claims 1-10, or a pharmaceutically acceptable salt, stereoisomer, crystal form, or isotopic variant thereof, and a pharmaceutically acceptable excipient.
12. The pharmaceutical composition of claim 11, further comprising an additional therapeutic agent.
13. Kit of parts comprising
A first container comprising a compound of any one of claims 1-10, or a pharmaceutically acceptable salt, stereoisomer, crystal, or isotopic variant thereof; and
optionally, a second container containing an additional therapeutic agent; and
optionally, a third container containing pharmaceutically acceptable excipients for diluting or suspending the compound and/or other therapeutic agent.
14. Use of a compound of any one of claims 1-10, or a pharmaceutically acceptable salt, stereoisomer, crystal form, or isotopic variant thereof, in the manufacture of a medicament for treating an EGFR-driven cancer.
15. The use of claim 14, wherein the EGFR-driven cancer is selected from the group consisting of: non-small cell lung cancer, lung adenocarcinoma, lung squamous carcinoma, pancreatic cancer, breast cancer, prostate cancer, liver cancer, skin cancer, epithelial cell cancer, gastrointestinal stromal tumor, leukemia, histiocytic lymphoma, and nasopharyngeal carcinoma.
16. The use of claim 15, wherein the EGFR-driven cancer is a T790M mutation, a L858R mutation, and a L858R/T790M double mutation of non-small cell lung cancer.
CN201680061348.9A 2015-12-10 2016-11-24 Aminopyrimidines useful for inhibiting protein tyrosine kinase activity Active CN108137544B (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
CN2015109167762 2015-12-10
CN201510916776 2015-12-10
PCT/CN2016/107072 WO2017097113A1 (en) 2015-12-10 2016-11-24 Aminopyrimidine compounds for inhibiting acitivity of protein tyrosine kinase

Publications (2)

Publication Number Publication Date
CN108137544A CN108137544A (en) 2018-06-08
CN108137544B true CN108137544B (en) 2022-01-04

Family

ID=59012681

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201680061348.9A Active CN108137544B (en) 2015-12-10 2016-11-24 Aminopyrimidines useful for inhibiting protein tyrosine kinase activity

Country Status (2)

Country Link
CN (1) CN108137544B (en)
WO (1) WO2017097113A1 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108610295B (en) * 2018-04-04 2023-01-10 大连医科大学附属第一医院 Pyrimidines, compositions and their use in the treatment of lymphoma leukemia
CN112601742A (en) * 2018-06-30 2021-04-02 北京生命科学研究所 Cathepsin C inhibitors

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102083800A (en) * 2008-06-27 2011-06-01 阿维拉制药公司 Heteroaryl compounds and uses thereof
WO2012064706A1 (en) * 2010-11-10 2012-05-18 Avila Therapeutics, Inc. Mutant-selective egfr inhibitors and uses thereof
CN102482277A (en) * 2009-05-05 2012-05-30 达纳-法伯癌症研究所有限公司 Egfr inhibitors and methods of treating disorders
WO2015006754A2 (en) * 2013-07-11 2015-01-15 Acea Biosciences Inc. Heterocyclic compounds and uses thereof

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105968056A (en) * 2016-05-28 2016-09-28 大连医科大学 Diarylpyrimidine compound, composition and application

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102083800A (en) * 2008-06-27 2011-06-01 阿维拉制药公司 Heteroaryl compounds and uses thereof
CN102482277A (en) * 2009-05-05 2012-05-30 达纳-法伯癌症研究所有限公司 Egfr inhibitors and methods of treating disorders
WO2012064706A1 (en) * 2010-11-10 2012-05-18 Avila Therapeutics, Inc. Mutant-selective egfr inhibitors and uses thereof
WO2015006754A2 (en) * 2013-07-11 2015-01-15 Acea Biosciences Inc. Heterocyclic compounds and uses thereof

Also Published As

Publication number Publication date
WO2017097113A1 (en) 2017-06-15
CN108137544A (en) 2018-06-08

Similar Documents

Publication Publication Date Title
CN113382991B (en) Aminopyrimidines useful for inhibiting protein tyrosine kinase activity
EP3889154A1 (en) Heterocyclic compound intermediate, preparation method therefor and application thereof
CN115135636A (en) Quinoxalinone derivatives as irreversible inhibitors of KRAS G12C mutant proteins
CN112047938B (en) 2,4, 6-trisubstituted pyrimidine compounds as ATR kinase inhibitors
JP7092405B2 (en) Di (hetero) aryl macrocycle to inhibit kinase activity
CN102892759A (en) Nitrogen-containing heterocyclic compound having kynurenine production inhibitory activity
EP2070924A1 (en) New 2 hetarylthiazol-4-carboxylic acid derivatives, their manufacture and use as medicine
EP3553057A1 (en) (hetero)arylamide compound for inhibiting protein kinase activity
WO2023134266A1 (en) 2-piperidyl or 2-pyrazolyl substituted pyrimidine compound serving as egfr inhibitor
CN108137544B (en) Aminopyrimidines useful for inhibiting protein tyrosine kinase activity
CN111518082B (en) Aminopyrimidine compound, composition containing aminopyrimidine compound and application of aminopyrimidine compound
WO2017190637A1 (en) Fused pyrimidine compound for inhibiting protein tyrosine kinase activity
WO2022048631A1 (en) Compound having antitumor activity and use thereof
TWI817955B (en) Aromatic derivatives, their preparation methods and their applications in medicine
WO2022217118A1 (en) Pyrimidine based ras modulators and uses thereof
CN115943145A (en) Pyrazolopyrimidine compounds as ATR kinase inhibitors
EP3853225B1 (en) N-(5-(3-(1-((thiazol-2-yl)amino)-1-oxopropan-2-yl)phenyl)pyridin-2-yl)acrylamide derivatives as cdk7 inhibitors for the treatment of cancer
CN115181104A (en) Substituted fused tricyclic derivatives, compositions and uses thereof
CN111349084B (en) Aminopyrimidines useful for inhibiting protein kinase activity
CN114874189B (en) Substituted heteroaryl derivatives, compositions and uses thereof
CN117402142A (en) (hetero) aryl amides for inhibiting protein kinase activity
CN116940581A (en) Novel protein degradation agent and application thereof

Legal Events

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