CN111892579A - Kinase inhibitor - Google Patents

Kinase inhibitor Download PDF

Info

Publication number
CN111892579A
CN111892579A CN202010883627.1A CN202010883627A CN111892579A CN 111892579 A CN111892579 A CN 111892579A CN 202010883627 A CN202010883627 A CN 202010883627A CN 111892579 A CN111892579 A CN 111892579A
Authority
CN
China
Prior art keywords
amino
methyl
ethyl
pyrimidin
acrylamide
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.)
Granted
Application number
CN202010883627.1A
Other languages
Chinese (zh)
Other versions
CN111892579B (en
Inventor
李同双
赵兴东
田强
张卫鹏
刘洪彬
王宪龙
谭浩瀚
谭锐
刘启洪
姜立花
刘研新
令狐莉
林敏�
孙婧
王为波
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.)
Shanghai Allist Medicine Polytron Technologies Inc
Original Assignee
Shanghai Fochon Pharmaceutical Co Ltd
Fochon Pharmaceuticals Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=54832920&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=CN111892579(A) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Shanghai Fochon Pharmaceutical Co Ltd, Fochon Pharmaceuticals Ltd filed Critical Shanghai Fochon Pharmaceutical Co Ltd
Priority to CN202010883627.1A priority Critical patent/CN111892579B/en
Publication of CN111892579A publication Critical patent/CN111892579A/en
Application granted granted Critical
Publication of CN111892579B publication Critical patent/CN111892579B/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
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems

Abstract

The invention relates to a class of mutated EGFR selective inhibitors, pharmaceutical compositions and methods of use.

Description

Kinase inhibitor
This application is a divisional application of chinese patent application No. 201580031253.8 entitled "class of kinase inhibitors" filed on 12/6/2015.
Technical Field
The present invention relates to a novel class of aminopyrimidine ring derivatives and pharmaceutically acceptable salts thereof which selectively inhibit the activity of mutant Epidermal Growth Factor (EGFR) kinase and are useful as medicaments for the treatment of proliferative diseases, such as cancer, in mammals, especially humans. The invention also relates to a use of said compounds in the treatment of cancer, and to pharmaceutical preparations containing said compounds.
Background
Protein tyrosine kinases catalyze the transfer of a phosphate group from ATP or GTP to a tyrosine residue on a protein substrate. Protein tyrosine kinases can be classified as either receptor (e.g., EGFR, HER-2, VEGFR, FGFR) or non-receptor (e.g., Src, Jak, Abl). Receptor tyrosine kinases function to activate secondary signaling factors through phosphorylation, which leads signals from outside the cell to inside the cell. A variety of cellular processes are initiated by these signals, including proliferation, carbohydrate utilization, protein synthesis, angiogenesis, cell growth, and cell survival.
Inappropriate or uncontrolled activation of tyrosine kinase activity, such as overexpression or mutation of the kinase, has been shown to result in uncontrolled cell growth. There is a lot of evidence that EGFR is involved in the development of human tumors, more than 60% of all solid tumors expressing at least one EGFR or its ligand. Overexpression of EGFR is common in breast, lung, head and neck, and bladder cancers.
The first generation of reversible, ATP-competitive EGFR kinase inhibitors, such as gefitinib and erlotinib, are effective clinical treatments for non-small cell lung cancer carrying mutations in the EGFR kinase domain activity (Nature,2009,462, 1070-107). Although first generation EGFR inhibitors show encouraging clinical efficacy, over time almost all patients develop resistance to these inhibitors, e.g., the gatekeeper mutation T790M, with about half of the resistant mutations of gefitinib and erlotinib developing the T790M mutation. (proc.natl.acad.sci.u.s.a.2008,105,2070-2075) furthermore, the T790M mutation was also inherently present and the T790M mutation may have an independent oncogenic effect.
Currently developed drugs, including second generation covalent inhibitors such as afatinib, neratinib, canertinib, and dacatinib, are effective against T790M resistance mutations, but exhibit dose-dependent toxicity such as diarrhea and rash due to simultaneous inhibition of wild-type EGFR.
Thus, there remains a pressing need for selective inhibitors of mutant EGFR kinases. Although mutant EGFR selective inhibitors have been reported in the literature, e.g., WO 2013014448 and WO 2012061299, there is still a strong need for novel EGFR mutant selective inhibitors that have advantages in at least one of therapeutic efficacy, stability, selectivity, safety, pharmacodynamic and pharmacokinetic profile in the treatment of aberrantly proliferating diseases. The invention relates to a novel EGFR mutation selective inhibitor.
Disclosure of Invention
The invention relates to novel aminopyrimidine ring derivatives, pharmaceutical compositions thereof, and their use as medicaments.
In one aspect, the present invention provides at least one compound of formula (I):
Figure BDA0002654899220000021
and/or at least one pharmaceutically acceptable salt thereof,
wherein:
q is selected from aryl and heteroaryl;
x is selected from N and C;
y is selected from N and C;
R1selected from: hydrogen, C1-10Alkyl radical, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, heterocyclyl and heterocyclyl-C1-4Alkyl, wherein each alkyl, cycloalkyl and heterocyclyl is unsubstituted or substituted with at least one, such as 1,2,3 or 4, independently selected from R6aSubstituted with the substituent(s);
R1’selected from hydrogen and halogen;
R2and R3Are respectively selected from: hydrogen, halogen, hydroxy, CN, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl and heteroaryl-C1-4Alkyl, wherein each alkyl, alkenyl, alkynyl, cycloalkyl and heterocyclyl is unsubstituted or substituted with at least one, such as 1,2,3 or 4, independently selected from R6aWherein each aryl and heteroaryl is unsubstituted or substituted with at least one, such as 1,2,3 or 4, independently selected from R6bSubstituted with the substituent(s); or R2And R3Together with the carbon atom to which they are attached form a 5-6 membered ring containing 0, 1,2 or 3 heteroatoms independently selected from oxygen, sulfur and nitrogen, which ring may be unsubstituted or substituted with 1 or 2 heteroatoms selected from R6aSubstituted with the substituent(s);
each R4Independently selected from: hydrogen, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl, -OR8、-NR7S(O)rR8、-NO2-halogen, -S (O)rR7、-SR8、-S(O)2OR7、-OS(O)2R8、-S(O)rNR7R8、-NR7R8、-O(CR9R10)tNR7R8、-C(O)R7、-CO2R8、-CO2(CR9R10)tCONR7R8、-OC(O)R7、-CN、-C(O)NR7R8、-NR7C(O)R8、-OC(O)NR7R8、-NR7C(O)OR8、-NR7C(O)NR7R8and-CR7(N-OR8) (ii) a Wherein each C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl and C3-10Cycloalkyl is unsubstituted or substituted by at least one, such as 1,2,3 or 4, independently selected from R6aSubstituted with the substituent(s);
each R5Independently selected from: hydrogen, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl and heteroaryl-C1-4Alkyl, -OR8、-NR7S(O)rR8、-NO2Halogen, -S (O)rR7、-SR8、-S(O)2OR7、-OS(O)2R8、-S(O)rNR7R8、-NR7R8、-O(CR9R10)tNR7R8、-C(O)R7、-CO2R8、-CO2(CR9R10)tCONR7R8、-OC(O)R7、-CN、-C(O)NR7R8、-NR7C(O)R8、-OC(O)NR7R8、-NR7C(O)OR8、-NR7C(O)NR7R8and-CR7(N-OR8) Wherein each C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl and heterocyclyl are unsubstituted or substituted by at least one, such as 1,2,3 or 4, independently selected from R6aSubstituted with the substituent(s); each aryl and heteroaryl is unsubstituted or substituted with at least one, such as 1,2,3 or 4, independently selected from R6bSubstituted with the substituent(s);
each R6aIndependently selected from: c1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl, -OR8、-NR7S(O)rR8、-NO2Halogen, -S (O)rR7、-SR8、-S(O)2OR7、-OS(O)2R8、-S(O)rNR7R8、-NR7R8、-(CR9R10)tOR8、-(CR9R10)tNR7R8、-(CR9R10)tSR8、-(CR9R10)tS(O)rR8、-(CR9R10)tCO2R8、-(CR9R10)tCONR7R8、-(CR9R10)tNR7CO2R8、-(CR9R10)tOCONR7R8、-(CR9R10)tNR7CONR7R8、-(CR9R10)tNR7SO2NR7R8、-O(CR9R10)tNR7R8、-C(O)R7、-C(O)(CR9R10)tOR8、-C(O)(CR9R10)tNR7R8、-C(O)(CR9R10)tSR8、-C(O)(CR9R10)tS(O)rR8、-CO2R8、-CO2(CR9R10)tCONR7R8、-OC(O)R7、-CN、-C(O)NR7R8、-NR7C(O)R8、-OC(O)NR7R8、-NR7C(O)OR8、-NR7C(O)NR7R8and-CR7(N-OR8);
Each R6bIndependently selected from: r6aAryl, aryl-C1-4Alkyl, heteroaryl and heteroaryl-C1-4An alkyl group;
each R7And each R8Independently selected from: hydrogen, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, cycloalkyl-C1-4Alkyl, heterocyclic radical C1-4Alkyl, aryl, heteroaryl,Heteroaryl, aryl-C1-4Alkyl, heteroaryl and heteroaryl-C1-4An alkyl group; wherein each alkyl, alkenyl, alkynyl, cycloalkyl and heterocyclyl is unsubstituted or substituted with at least one, such as 1,2,3 or 4, independently selected from R6aWherein each aryl and heteroaryl is unsubstituted or substituted with at least one, such as 1,2,3 or 4, independently selected from R6bSubstituted with the substituent(s); or R7And R8Together with the atom or atoms to which they are attached form a 4-12 membered heterocyclic ring containing 0, 1 or 2 additional heteroatoms independently selected from oxygen, sulfur and nitrogen, which ring may be unsubstituted or substituted with 1 or 2 heteroatoms selected from R6bSubstituted with the substituent(s);
each R9And each R10Independently selected from: hydrogen, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, cycloalkyl-C1-4Alkyl, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl and heteroaryl-C1-4An alkyl group; or R9And R10Together with the carbon atom or atoms to which they are attached form a 3-7 membered ring containing 0, 1 or 2 heteroatoms independently selected from oxygen, sulfur and nitrogen, which ring may be unsubstituted or substituted by 1 or 2 heteroatoms selected from R6aSubstituted with the substituent(s);
m is independently selected from 0, 1, 2and 3;
n is independently selected from 0, 1, 2and 3;
each r is independently selected from 1 and 2;
each t is independently selected from 1, 2and 3.
In another aspect, the present invention provides a pharmaceutical composition comprising at least one compound of formula (I) and/or at least one pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier.
In another aspect, the present invention provides a method for modulating mutant EGFR, the method comprising administering to a system or subject in need thereof a therapeutically effective amount of at least one compound of formula (I) and/or at least one pharmaceutically acceptable salt or pharmaceutical composition, thereby modulating mutant EGFR.
In another aspect, the invention also provides a method of treating, ameliorating or preventing a disorder responsive to inhibition of mutated EGFR comprising administering to a system or subject in need thereof an effective amount of at least one compound of formula (I) and/or at least one pharmaceutically acceptable salt or pharmaceutical composition, or in combination with another therapeutic agent, for treating the above disorder.
In another aspect, the invention provides the use of at least one compound of formula (I) and/or at least one pharmaceutically acceptable salt for the manufacture of a medicament for the treatment of a mutated EGFR-mediated disorder. The compounds can be used alone or in combination with another therapeutic agent to treat a mutant EGFR-mediated disorder, wherein the disorder is an autoimmune disease, a transplant disease, an infectious disease, or a cell proliferation disorder.
Furthermore, the present invention provides a method of treating a cell proliferative disorder, the method comprising administering to a system or subject in need thereof an effective amount of at least one compound of formula (I) and/or at least one pharmaceutically acceptable salt or pharmaceutical composition, or in combination with another therapeutic agent, to treat the above disorder.
Alternatively, the present invention provides the use of at least one compound of formula (I) and/or at least one pharmaceutically acceptable salt for the manufacture of a medicament for the treatment of a cell proliferative disorder. In particular embodiments, the compounds can be used alone or in combination with another therapeutic agent to treat a cell proliferative disorder, wherein the disorder includes, but is not limited to, lymphoma, osteosarcoma, melanoma, or a tumor of the breast, kidney, prostate, colorectal, thyroid, ovary, pancreas, neuron, lung, uterus, or gastrointestinal tract.
In the above methods of using the compounds of the present invention, at least one compound of formula (I) and/or at least one pharmaceutically acceptable salt may be used in a system consisting of cells or tissues, or in a mammalian, such as human or animal, subject.
Detailed Description
The terms used herein are defined as follows:
"alkyl" refers to branched and straight chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms. Unless otherwise indicated, "alkyl" means C1-C6An alkyl group. For example, "C1-C6C in alkyl1-C6"refers to a linear or branched arrangement of groups having 1,2,3,4, 5 or 6 carbon atoms. For example, "C1-C8Alkyl groups "include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, isobutyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl, and the like.
"cycloalkyl" refers to a saturated aliphatic cyclic hydrocarbon group having a specified number of carbon atoms. "cycloalkyl" means C, unless otherwise indicated3-C10A cycloalkyl group. For example, "cycloalkyl" includes, but is not limited to, cyclopropyl, methyl-cyclopropyl, 2-dimethyl-cyclobutyl, 2-ethyl-cyclopentyl, and cyclohexyl.
"alkenyl" means a non-aromatic straight, branched or cyclic hydrocarbon group containing 2 to 10 carbon atoms and having at least one carbon-carbon double bond. In some embodiments, 1 carbon-carbon double bond is present, and up to 4 non-aromatic carbon-carbon double bonds may be present. Thus, "C2-6Alkenyl "means alkenyl containing 2 to 6 carbon atoms. Alkenyl groups include, but are not limited to, ethenyl, propenyl, butenyl, 2-methylbutenyl, and cyclohexenyl. The straight, branched or cyclic portion of the alkenyl group may contain a double bond, and substituted alkenyl groups, if indicated, may be substituted.
"alkynyl" refers to a straight, branched or cyclic hydrocarbon group containing 2 to 10 carbon atoms and at least one carbon-carbon triple bond. In some embodiments, there may be 3 carbon-carbon triple bonds. Thus, "C2-6Alkynyl "refers to alkynyl groups containing 2-6 carbon atoms. Alkynyl groups include, but are not limited to, ethynyl, propynyl, butynyl, 3-methylbutynyl, and the like. The straight, branched or cyclic portion of the alkynyl group may contain a triple bond, and a substituted alkynyl group, if indicated, may be substituted.
"aryl" includes: 5-and 6-membered aromatic carbocyclic rings, such as phenyl; bicyclic rings having at least one aromatic carbon ring, such as naphthyl, indane and 1,2,3, 4-tetrahydroquinoline, and tricyclic rings having at least one aromatic carbon ring, such as fluorene. Aryl substituents are considered to be linked through an aromatic ring if they are bicyclic or tricyclic and at least one of the rings is non-aromatic.
For example, aryl includes 5-and 6-membered aromatic carbocyclic rings fused to a 5-to 7-membered heterocyclic ring containing one or more heteroatoms selected from N, O and S, provided that the site of attachment is an aromatic carbocyclic ring. Divalent radicals, which are formed from substituted benzene derivatives and have free valence electrons on the ring atoms, are named substituted phenylene radicals. Divalent radicals derived from monovalent polycyclic hydrocarbon radicals whose name ends with "-yl", which are obtained by removing one more hydrogen atom from a carbon atom containing the valence electron of the radical, are named after the name of the monovalent radical plus "-idene", for example, naphthyl, which has two attachment sites, is called naphthylene. The definition of aryl, however, does not include, nor overlap with, heteroaryl, and is defined individually as follows. Thus, if one or more aromatic carbocyclic rings are fused to an aromatic ring of a heterocyclic ring, the ring system formed should be considered heteroaryl rather than aryl as defined herein.
"halogen" refers to fluorine, chlorine, bromine and iodine.
"heteroaryl" means
A 5-to 8-membered aromatic monocyclic ring, which contains 1 to 4, and in certain embodiments 1 to 3, heteroatoms selected from N, O and S, the remainder being carbon atoms;
an 8-to 12-membered bicyclic ring containing 1 to 4, and in certain embodiments 1 to 3, heteroatoms selected from N, O and S, the remainder being carbon atoms, and wherein at least one heteroatom is present in the aromatic ring; and
11-to 14-membered tricyclic rings. The ring contains from 1 to 4, and in certain embodiments from 1 to 3, heteroatoms selected from N, O and S, the remainder being carbon atoms, and at least one of the heteroatoms is present in the aromatic ring.
When the total number of S and O in the heteroaryl group is greater than 1, these heteroatoms are not adjacent to each other. In some embodiments, the total number of S and O in the heteroaryl is no greater than 2. In some embodiments, the total number of S and O in the heteroaryl is no greater than 1.
Examples of heteroaryl groups include, but are not limited to (the numbering of the attachment site is preferred, as in position 1), 2-pyridyl, 3-pyridyl, 4-pyridyl, 2, 3-pyrazinyl, 3, 4-pyrazinyl, 2, 4-pyrimidinyl, 3, 5-pyrimidinyl, 1-pyrazolyl, 2, 3-pyrazolyl, 2, 4-imidazolinyl, isoxazolyl, oxazolyl, thiazolyl, thiadiazo, tetrazolyl, thienyl, benzothienyl, furyl, benzofuranyl, benzimidazolinyl, dihydroindolyl, pyrazinyl, triazolyl, quinolinyl, pyrazolyl, and 5,6,7, 8-tetrahydroisoquinolinyl.
Further, heteroaryl groups include, but are not limited to, pyrrolyl, isothiazolyl, triazinyl, pyrazinyl, pyridazinyl, indolyl, benzotriazolyl, quinolyl, and isoquinolyl groups. As defined below for heterocyclic groups, "heteroaryl" includes N-oxide derivatives of nitrogen-containing heteroaryl groups.
The nomenclature of monovalent heteroaryl radicals ends with the "radical", which derives a divalent radical derived by removing a further hydrogen atom from the carbon atom containing the valence electron, by adding "idene" to the name of the monovalent radical, for example: the pyridyl group having two attachment sites is called a pyridylidene. The definition of heteroaryl does not include, nor overlap with, aryl as defined above.
If the heteroaryl substituent is a bicyclo or tricyclic ring, and at least one of the rings is non-aromatic or contains no heteroatoms, it is generally considered to be linked via an aromatic ring or a heteroatom-containing ring, respectively.
"heterocycle" (and derivatives thereof such as "heterocyclic" or "heterocyclyl") refers broadly to a single cyclic aliphatic hydrocarbon, typically having from 3 to 12 ring atoms, containing at least 2 carbon atoms, and further containing from 1 to 3 heteroatoms independently selected from oxygen, sulfur, and nitrogen, and also to combinations comprising at least one of the foregoing heteroatoms. Alternatively, the heterocyclic ring as defined above may be a polycyclic ring system (e.g. bicyclic) wherein two or more rings are connected by fusion or bridging, wherein at least one ring contains one or more heteroatoms independently selected from oxygen, sulphur, nitrogen. "heterocycle" also refers to a 5-to 7-membered heterocyclic ring fused to 5-and 6-membered aromatic carbocyclic rings containing one or more heteroatoms selected from nitrogen, oxygen, and sulfur, provided that the site of attachment is on the heterocyclic ring. Heterocycles may be saturated or contain one to more double bonds (i.e., partially unsaturated). The heterocyclic ring may be substituted with oxo (oxo). Either the carbon atom or the heteroatom of the heterocycle may be the attachment site, provided that a stable structure is formed. When a substituent is present on the heterocycle, the substituent may be attached to any heteroatom or carbon atom on the heterocycle, provided that a stable chemical structure is formed. The heterocyclic and heteroaryl definitions described herein do not overlap.
Suitable heterocycles include, for example (attachment site is preferably 1) 1-pyrrolidinyl, 2, 4-imidazolidinyl, 2, 3-pyrazolidinonyl, 1-piperidinyl, 2-piperidinyl, 3-piperidinyl, 4-piperidinyl and 2, 5-piperazinyl. Also included are 2-morpholino and 3-morpholino (oxygen atom position numbering is preferably 1). Substituted heterocycles also include ring systems substituted with one or more oxo groups, such as piperidinyl-N-oxide, morpholinyl-N-oxide, 1-oxo-1-thiomorpholinyl, and 1, 1-dioxo-1-thiomorpholinyl. The bis-heterocyclic compounds include, for example:
Figure BDA0002654899220000051
as used herein, "aralkyl" refers to an aryl-substituted alkyl group. Exemplary aralkyl groups include benzyl, phenethyl, naphthylmethyl, and the like. In some embodiments, aralkyl groups contain 7 to 20 or 7 to 11 carbon atoms. When using "aryl radicals C1-4Alkyl "in which" C1-4"refers to the number of carbon atoms in the alkyl portion rather than the aryl portion. Likewise, when "aryl C" is used1-10Alkyl "in which" C1-10"refers to the number of carbon atoms in the alkyl portion rather than the aryl portion.
As used herein, "heterocyclylalkyl" refers to a heterocyclyl-substituted alkyl group. When using "heterocyclyl C1-6Alkyl "in which" C1-6"refers to the number of carbon atoms in the alkyl moiety rather than in the heterocyclyl moiety.
As used herein, "cycloalkylalkyl" refers to cycloalkyl-substituted alkyl groups. When using "C3-10Cycloalkylalkyl radicals "In which is "C3-10"refers to the number of carbon atoms in the cycloalkyl moiety rather than the alkyl moiety. When using "C3-7Cycloalkylalkyl "in which" C3-7"refers to the number of carbon atoms in the cycloalkyl moiety rather than the alkyl moiety. When using "C3-8Cycloalkylalkyl "in which" C3-8"refers to the number of carbon atoms in the cycloalkyl moiety rather than the alkyl moiety. When using "cycloalkyl radicals C1-10Alkyl "in which" C1-10"refers to the number of carbon atoms in the alkyl moiety rather than the cycloalkyl moiety.
As used herein, "heteroarylalkyl" refers to a heteroaryl-substituted alkyl group. When using "heteroaryl C1-4Alkyl "in which" C1-4"refers to the number of carbon atoms in the cycloalkyl moiety rather than in the heteroaryl moiety. Likewise, when "heteroaryl C" is used1-10Alkyl "in which" C1-10"refers to the number of carbon atoms in the alkyl moiety rather than in the heteroaryl moiety.
To avoid ambiguity, for example: when alkyl, cycloalkyl, heterocyclylalkyl, aryl and/or heteroaryl substitutions are mentioned, it is meant that each of these groups is substituted individually or that these groups are mixed. That is: if R is1Is aralkyl, the aryl moiety may be unsubstituted or substituted by at least one, such as 1,2,3 or 4, independently selected from R6bMay also be unsubstituted or substituted by at least one, such as 1,2,3 or 4, independently selected from R6aA substituent of (1).
"pharmaceutically acceptable salt" refers to salts with pharmaceutically acceptable non-toxic bases or acids, including inorganic or organic bases and inorganic or organic acids. The salt of an inorganic base may be selected, for example, from: aluminum, ammonium, calcium, copper, iron, ferrous iron, lithium, magnesium, manganese, manganous, potassium, sodium and zinc salts. Further, the salt of a pharmaceutically acceptable inorganic base may be selected from ammonium, calcium, magnesium, potassium and sodium salts. One or more crystal structures may be present in the solid salt, as well as in the form of hydrates. The pharmaceutically acceptable salts of organic non-toxic bases may be selected, for example, from: primary, secondary and tertiary amine salts, the substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins such as arginine, betaine, caffeine, choline, N-dibenzylethylenediamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethylmorpholine, N-ethylpiperidine, glucosamine, histidine, hydrabamine, isopropylamine, lysine, methylglucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine and tripropylamine, tromethamine.
When the compound referred to in this patent is a base, it is necessary to prepare a salt thereof with at least one pharmaceutically acceptable non-toxic acid selected from inorganic and organic acids. For example, selected from the group consisting of acetic acid, benzenesulfonic acid, benzoic acid, camphorsulfonic acid, citric acid, ethanesulfonic acid, fumaric acid, gluconic acid, glutamic acid, hydrobromic acid, hydrochloric acid, isethionic acid, lactic acid, maleic acid, malic acid, mandelic acid, methanesulfonic acid, mucic acid, nitric acid, pamoic acid, pantothenic acid, phosphoric acid, succinic acid, sulfuric acid, tartaric acid, and p-toluenesulfonic acid. In some embodiments, these acids may be selected, for example: citric, hydrobromic, hydrochloric, maleic, phosphoric, sulfuric, fumaric and tartaric acids.
"protecting group" (Pg) refers to a class of substituents used to block or protect a particular functional group by reacting with other functional groups on a compound. For example, "amino protecting group" refers to a substituent attached to an amino group that blocks or protects the amino functionality on a compound. Suitable amino protecting groups include acetyl, trifluoroacetyl, tert-Butoxycarbonyl (BOC), benzyloxycarbonyl (CBZ) and 9-fluorenylmethyloxycarbonyl (Fmoc). Similarly, "hydroxy protecting group" refers to a class of hydroxy substituents that are effective in blocking or protecting the hydroxy function. Suitable protecting groups include, but are not limited to, acetyl and silyl groups. "carboxy protecting group" refers to a class of carboxy substituents that function effectively to block or protect a carboxy group. Common carboxyl protecting groups include, but are not limited to, -CH2CH2SO2Ph, cyanoethyl, 2- (trimethylsilyl) ethyl, 2- (trimethylsilyl) ethoxymethyl, 2- (p-toluenesulfonyl) ethyl, 2- (p-nitrophenylthio) ethyl, 2- (diphenylphosphino) -ethyl, nitroethyl and the like. General description and use for protecting groupsSee, T.W.Greene, Protective Groups in Organic Synthesis, John Wiley&Sons,New York,1991。
By "administering" or "administration" of at least one compound and/or at least one pharmaceutically acceptable salt thereof is meant providing a compound of the invention or a pharmaceutically acceptable salt thereof to a subject in need thereof.
An "effective amount" is an amount of a compound of interest, or a pharmaceutically acceptable salt thereof, that is capable of eliciting a biological or medical response in a tissue, system, animal or human that is observable by a researcher, veterinarian, clinician or other clinician.
The "composition" includes: the invention may take the form of a kit, article of manufacture, or any combination thereof. The pharmaceutical composition comprises: products comprising the active ingredient and an inert ingredient as a carrier, as well as products produced by any two or more of the ingredients, directly or indirectly, by combination, complexation or aggregation, or by dissociation of one or more of the ingredients, or by other types of reactions or interactions of one or more of the ingredients.
By "pharmaceutically acceptable" is meant compatible with the other ingredients of the formulation and not unacceptably toxic to the user.
1. The present invention provides at least one compound of formula (I):
Figure BDA0002654899220000071
and/or at least one pharmaceutically acceptable salt thereof,
wherein:
q is selected from aryl and heteroaryl;
x is selected from N and C;
y is selected from N and C;
R1selected from: hydrogen, C1-10Alkyl radical, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, heterocyclyl and heterocyclyl-C1-4An alkyl group, wherein each alkyl group,Cycloalkyl and heterocyclyl are unsubstituted or substituted by at least one, such as 1,2,3 or 4, independently selected from R6aSubstituted with the substituent(s);
R1’selected from hydrogen and halogen;
R2and R3Are respectively selected from: hydrogen, halogen, hydroxy, CN, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl and heteroaryl-C1-4Alkyl, wherein each alkyl, alkenyl, alkynyl, cycloalkyl and heterocyclyl is unsubstituted or substituted with at least one, such as 1,2,3 or 4, independently selected from R6aWherein each aryl and heteroaryl is unsubstituted or substituted with at least one, such as 1,2,3 or 4, independently selected from R6bSubstituted with the substituent(s); or R2And R3Together with the carbon atom to which they are attached form a 5-6 membered ring containing 0, 1,2 or 3 heteroatoms independently selected from oxygen, sulfur and nitrogen, which ring may be unsubstituted or substituted with 1 or 2 heteroatoms selected from R6aSubstituted with the substituent(s);
each R4Independently selected from: hydrogen, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl, -OR8、-NR7S(O)rR8、-NO2-halogen, -S (O)rR7、-SR8、-S(O)2OR7、-OS(O)2R8、-S(O)rNR7R8、-NR7R8、-O(CR9R10)tNR7R8、-C(O)R7、-CO2R8、-CO2(CR9R10)tCONR7R8、-OC(O)R7、-CN、-C(O)NR7R8、-NR7C(O)R8、-OC(O)NR7R8、-NR7C(O)OR8、-NR7C(O)NR7R8and-CR7(N-OR8) (ii) a Wherein each C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl and C3-10Cycloalkyl is unsubstituted or substituted by at least one, such as 1,2,3 or 4, independently selected from R6aSubstituted with the substituent(s);
each R5Independently selected from: hydrogen, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl and heteroaryl-C1-4Alkyl, -OR8、-NR7S(O)rR8、-NO2Halogen, -S (O)rR7、-SR8、-S(O)2OR7、-OS(O)2R8、-S(O)rNR7R8、-NR7R8、-O(CR9R10)tNR7R8、-C(O)R7、-CO2R8、-CO2(CR9R10)tCONR7R8、-OC(O)R7、-CN、-C(O)NR7R8、-NR7C(O)R8、-OC(O)NR7R8、-NR7C(O)OR8、-NR7C(O)NR7R8and-CR7(N-OR8) Wherein each C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl and heterocyclyl are unsubstituted or substituted by at least one, such as 1,2,3 or 4, independently selected from R6aSubstituted with the substituent(s); each aryl and heteroaryl is unsubstituted or substituted with at least one, such as 1,2,3 or 4, independently selected from R6bSubstituted with the substituent(s);
each R6aIndependently selected from: c1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl, -OR8、-NR7S(O)rR8、-NO2Halogen, -S (O)rR7、-SR8、-S(O)2OR7、-OS(O)2R8、-S(O)rNR7R8、-NR7R8、-(CR9R10)tOR8、-(CR9R10)tNR7R8、-(CR9R10)tSR8、-(CR9R10)tS(O)rR8、-(CR9R10)tCO2R8、-(CR9R10)tCONR7R8、-(CR9R10)tNR7CO2R8、-(CR9R10)tOCONR7R8、-(CR9R10)tNR7CONR7R8、-(CR9R10)tNR7SO2NR7R8、-O(CR9R10)tNR7R8、-C(O)R7、-C(O)(CR9R10)tOR8、-C(O)(CR9R10)tNR7R8、-C(O)(CR9R10)tSR8、-C(O)(CR9R10)tS(O)rR8、-CO2R8、-CO2(CR9R10)tCONR7R8、-OC(O)R7、-CN、-C(O)NR7R8、-NR7C(O)R8、-OC(O)NR7R8、-NR7C(O)OR8、-NR7C(O)NR7R8and-CR7(N-OR8);
Each R6bIndependently selected from: r6aAryl, aryl-C1-4Alkyl, heteroaryl and heteroaryl-C1-4An alkyl group;
each R7And each R8Independently selected from: hydrogen, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, cycloalkaneAlkyl, cycloalkyl-C1-4Alkyl, heterocyclic radical C1-4Alkyl, aryl, heteroaryl, aryl-C1-4Alkyl, heteroaryl and heteroaryl-C1-4An alkyl group; wherein each alkyl, alkenyl, alkynyl, cycloalkyl and heterocyclyl is unsubstituted or substituted with at least one, such as 1,2,3 or 4, independently selected from R6aWherein each aryl and heteroaryl is unsubstituted or substituted with at least one, such as 1,2,3 or 4, independently selected from R6bSubstituted with the substituent(s); or R7And R8Together with the atom or atoms to which they are attached form a 4-12 membered heterocyclic ring containing 0, 1 or 2 additional heteroatoms independently selected from oxygen, sulfur and N, which ring may be unsubstituted or substituted with 1 or 2 heteroatoms selected from R6bSubstituted with the substituent(s);
each R9And each R10Independently selected from: hydrogen, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, cycloalkyl-C1-4Alkyl, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl and heteroaryl-C1-4An alkyl group; or R9And R10Together with the carbon atom or atoms to which they are attached form a 3-7 membered ring containing 0, 1 or 2 heteroatoms independently selected from oxygen, sulfur and nitrogen, which ring may be substituted with 1-2 heteroatoms selected from R6aSubstituted with the substituent(s);
m is independently selected from 0, 1, 2and 3;
n is independently selected from 0, 1, 2and 3;
each r is independently selected from 1 and 2;
each t is independently selected from 1, 2and 3.
2.1, and/or at least one pharmaceutically acceptable salt thereof, wherein X is selected from N and C and Y is selected from C.
At least one compound of any one of claims 1-2, and/or at least one pharmaceutically acceptable salt thereof, wherein X is selected from N and Y is selected from C.
4.1-2, and/or at least one pharmaceutically acceptable salt thereof, wherein X and Y are both C, and,
n is 2and one R5Is (3R) -3- (dimethylamino) pyrrol-1-yl, (3S) -3- (dimethylamino) pyrrol-1-yl, 3- (dimethylamino) azetidin-1-yl, [2- (dimethylamino) ethyl]- (methyl) amino, [2- (methylamino) ethyl](methyl) amino, 5-methyl-2, 5-diazaspiro [3.4 ]]Sunflower-2-yl, (3aR,6aR) -5-methylhexa-hydro-pyrrole [3,4-b]Pyrrol-1 (2H) -yl, 1-methyl-1, 2,3, 6-tetrahydropyridin-4-yl, 4-methylpiperazin-1-yl, 4- [2- (dimethylamino) -2-oxyethyl]Piperazin-1-yl, methyl [2- (4-methylpiperazin-1-yl) ethyl]Amino, methyl [2- (morpholin-4-yl) ethyl]Amino, 1-amino-1, 2,3, 6-tetrahydropyridin-4-yl or 4- [ (2S) -2-aminopropionyl]Piperazin-1-yl, another R5Is a methoxy group or a methyl group,
R1、R1’and R2Is hydrogen, and R3When hydrogen, fluorine, chlorine, methyl or cyano,
q is not 4,5,6, 7-tetrahydropyrrolo [1,5-a ] pyridin-3-yl, pyrazolo [1,5-a ] pyridin-3-yl or 1H-indol-3-yl. .
At least one compound of any one of claims 1 to 4, and/or at least one pharmaceutically acceptable salt thereof, wherein the general formula is
Figure BDA0002654899220000081
6.1-5, and/or at least one pharmaceutically acceptable salt thereof, wherein R1’Selected from hydrogen.
7.1-5, and/or at least one pharmaceutically acceptable salt thereof, wherein R1’Selected from fluorine.
8.1-7, and/or at least one pharmaceutically acceptable salt thereof, wherein R1Selected from hydrogen.
9.1-8, and/or at least one pharmaceutically acceptable salt thereof, wherein R2Selected from hydrogen.
10.1 to 9, and/or at least one thereofA pharmaceutically acceptable salt, wherein R3Selected from hydrogen, halogen and C1-10An alkyl group.
11.10, and/or at least one pharmaceutically acceptable salt thereof, wherein R3Selected from hydrogen.
At least one compound of any one of claims 1-11, and/or at least one pharmaceutically acceptable salt thereof, wherein Q is selected from heteroaryl.
13.12, and/or at least one pharmaceutically acceptable salt thereof, wherein Q is selected from
Figure BDA0002654899220000091
14.1-13, and/or at least one pharmaceutically acceptable salt thereof, wherein R4Selected from hydrogen, C1-10Alkyl, halogen and cyano, wherein C1-10Alkyl is unsubstituted or substituted by at least one, such as 1,2,3 or 4, independently selected from R6aIs substituted with the substituent(s).
15.14, and/or at least one pharmaceutically acceptable salt thereof, wherein R4Selected from the group consisting of hydrogen, methyl, halogen and cyano, wherein methyl is unsubstituted or substituted with at least one group independently selected from R6aIs preferably R6aIs fluorine.
16.1-15, and/or at least one pharmaceutically acceptable salt thereof, wherein R5Independently selected from OR8Heterocyclic group, NR7R8Wherein heterocyclyl is unsubstituted or substituted by at least one, such as 1,2,3 or 4, independently selected from R6aIs substituted with the substituent(s).
17.16, and/or at least one pharmaceutically acceptable salt thereof, wherein R5Independently selected from methoxy, ethoxy, isopropoxy, piperazin-1-yl, 4-methylpiperazin-1-yl, methyl (2- (methylamino) ethyl) amino, (2- (dimethylamino) ethyl) (methyl) amino, (2- (ethyl) (methyl) amino) ethyl) (methyl) amino and (methyl (2- (N-methyl) aminoAcetylamino) ethyl) amino).
18. At least one compound is selected from:
n- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide,
N- (6-methoxy-2- (methyl (2- (methylamino) ethyl) amino) -5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide,
N- (2- ((2- (ethyl (methyl) amino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide,
N- (6-methoxy-2- (methyl (2- (N-methylacetamido) ethyl) amino) -5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide,
N- (6-ethoxy-2- (methyl (2- (methylamino) ethyl) amino) -5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-ethoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide,
N- (6-isopropoxy-2- (methyl (2- (N-methylacetamido) ethyl) amino) -5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-isopropoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide,
N- (6-methoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- (piperazin-1-yl) pyridin-3-yl) acrylamide,
N- (6-methoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- (4-methylpiperidin-1-yl) pyridin-3-yl) acrylamide,
N- (6-methoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- (piperidin-1-yl) pyridin-3-yl) acrylamide,
N- (6-ethoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- (4-methylpiperidin-1-yl) pyridin-3-yl) acrylamide,
N- (6-isopropoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- (piperidin-1-yl) pyridin-3-yl) acrylamide,
N- (6-isopropoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- (4-methylpiperidin-1-yl) pyridin-3-yl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -5- ((4- (7-fluoro-1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -6-methoxypyridin-3-yl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -5- ((4- (6-fluoro-1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -6-methoxypyridin-3-yl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -5- ((4- (5-fluoro-1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -6-methoxypyridin-3-yl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -5- ((4- (4-fluoro-1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -6-methoxypyridin-3-yl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -5- ((5-fluoro-4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -6-methoxypyridin-3-yl) acrylamide,
N- (5- ((5-chloro-4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxypyridin-3-yl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((5-methyl-4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide,
N- (3- ((2- (dimethylamino) ethyl) (methyl) amino) -5-methoxy-6- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) pyridin-2-yl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) -2-fluoroacrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) phenyl) -2-fluoroacrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (1-methylindazin-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5- ((4- (1-methylindazin-3-yl) pyrimidin-2-yl) amino) phenyl) acrylamide,
N- (5- ((4- (1-cyanoindolizin-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxypyridin-3-yl) acrylamide,
N- (5- ((4- (1-cyanoindolizin-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -5- ((4- (indolizin-3-yl) pyrimidin-2-yl) amino) -6-methoxypyridin-3-yl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -5- ((4- (indolizin-3-yl) pyrimidin-2-yl) amino) -4-methoxyphenyl) acrylamide,
N- (5- ((4- (1-chloroindolizin-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxypyridin-3-yl) acrylamide,
N- (5- ((4- (1-chloroindolizin-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (3-methylindazin-1-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5- ((4- (3-methylindazin-1-yl) pyrimidin-2-yl) amino) phenyl) acrylamide,
N- (5- ((4- (3-cyanoindolizin-1-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxypyridin-3-yl) acrylamide,
N- (5- ((4- (3-cyanoindolizin-1-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -5- ((4- (indolizin-1-yl) pyrimidin-2-yl) amino) -6-methoxypyridin-3-yl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -5- ((4- (indolizin-1-yl) pyrimidin-2-yl) amino) -4-methoxyphenyl) acrylamide,
N- (5- ((4- (3-chloroindolizin-1-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxypyridin-3-yl) acrylamide,
N- (5- ((4- (3-chloroindolizin-1-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (3-methyl-1H-indol-1-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5- ((4- (3-methyl-1H-indol-1-yl) pyrimidin-2-yl) amino) phenyl) acrylamide,
N- (6-methoxy-5- ((4- (3-methyl-1H-indol-1-yl) pyrimidin-2-yl) amino) -2- (4-methylpiperazin-1-yl) pyridin-3-yl) acrylamide,
N- (4-methoxy-5- ((4- (3-methyl-1H-indol-1-yl) pyrimidin-2-yl) amino) -2- (4-methylpiperazin-1-yl) phenyl) acrylamide,
N- (5- ((4- (3-cyano-1H-indol-1-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxypyridin-3-yl) acrylamide,
N- (5- ((4- (3-cyano-1H-indol-1-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide,
N- (5- ((4- (1-chloroimidazo [1,5-a ] pyrid-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxypyridin-3-yl) acrylamide,
N- (5- ((4- (1-chloroimidazo [1,5-a ] pyrid-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (1- (trifluoromethyl) imidazo [1,5-a ] pyridin-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5- ((4- (1- (trifluoromethyl) imidazo [1,5-a ] pyridin-3-yl) pyrimidin-2-yl) amino) phenyl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (1-methylimidazol [1,5-a ] pyrid-in-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5- ((4- (1- (methylimidazol [1,5-a ] pyrid-in-3-yl) pyrimidin-2-yl) amino) phenyl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (1-methylimidazol [1,5-a ] pyrid-in-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) -2-fluoroacrylamide,
N- (5- ((4- (1, 2-dihydropyrrolo [3,2,1-hi ] indol-5-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxypyridin-3-yl) acrylamide,
N- (5- ((4- (1, 2-dihydropyrrolo [3,2,1-hi ] indol-5-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (3-methyl-1H-indazol-1-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (3-methyl-1H-indazol-1-yl) pyrimidin-2-yl) amino) pyridin-3-yl) -2-fluoroacrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (1-methyl-1H-indazol-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (1-methyl-1H-indazol-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) -2-fluoroacrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (3-methylimidazol [1,5-a ] pyrid-in-1-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (3-methylimidazo [1,5-a ] pyrid-in-1-yl) pyrimidin-2-yl) amino) pyridin-3-yl) -2-fluoroacrylamide,
and pharmaceutically acceptable salts thereof.
In another aspect, the invention provides a pharmaceutical combination comprising at least one compound of any one of 1-18 and pharmaceutically acceptable salts thereof, wherein the route of administration of the combination includes oral, parenteral, intraperitoneal, intravenous, intraarterial, transdermal, sublingual, intramuscular, rectal, buccal, intranasal, liposomal administration, via inhalation, vaginal, intraocular, by topical administration (e.g., through a catheter or stent), subcutaneous, intraadipose, intraarticular, or intrathecal.
In another aspect, the invention provides a kit comprising at least one compound of any one of the above 1-18 and pharmaceutically acceptable salts thereof; and instructions including one or more of the following: instructions for what disease the composition applies to, composition storage information, dosage information, and how to use the composition. In a particular variation, the kit comprises the compound in a multiple dose form.
In another aspect, the present invention provides a preparation comprising at least one compound of any one of the above 1-18 and pharmaceutically acceptable salts thereof; and a packaging material. In one variation, the packaging material comprises a container. In one particular variation, the container includes a label that identifies one or more of the following: instructions for what disease the compound applies to, stored information, dosage information, and/or how to use the composition. In another variation, the article of manufacture comprises the compound in a multiple dose form.
In another aspect, the invention provides a method of treatment comprising administering to a subject at least one compound of any one of 1-18 and pharmaceutically acceptable salts thereof.
In another aspect, the present invention provides a method of inhibiting mutant EGFR by the action of at least one compound of any one of 1-18 and pharmaceutically acceptable salts thereof with mutant EGFR.
In another aspect, the invention provides a method of inhibiting mutant EGFR, comprising causing at least one compound of any one of 1-18 and pharmaceutically acceptable salts thereof to be present in a body, to inhibit the activity of mutant EGFR in the body.
In another aspect, the invention provides a method of treating a disease state for which mutated EGFR activity contributes to the pathology and/or symptomology of the disease, the method comprising causing a therapeutically effective amount of at least one compound of any one of 1-18 and pharmaceutically acceptable salts thereof to be present in a subject to ameliorate the disease state.
In variations of each of the above methods, the disease state is selected from: cancerous proliferative diseases (e.g., brain, lung, squamous cell, bladder, stomach, pancreas, breast, head, neck, kidney, ovary, prostate, colon, epidermis, esophagus, testicular, gynecological, or thyroid cancer); non-cancerous proliferative diseases (e.g., benign skin hyperplasia (e.g., psoriasis), restenosis, and Benign Prostatic Hypertrophy (BPH)); pancreatitis; kidney disease; pain; preventing blast implantation; treating diseases associated with angiogenesis or vasculogenesis (e.g., tumor angiogenesis, acute and chronic infectious diseases such as rheumatoid arthritis, atherosclerosis, inflammatory bowel disease, skin diseases such as psoriasis, eczema, and scleroderma, diabetes, diabetic retinopathy, retinopathy of prematurity, age-related macular degeneration, hemangioma, glioma, melanoma, kaposi's sarcoma and ovarian cancer, breast cancer, lung cancer, pancreatic cancer, prostate cancer, colon cancer, and epidermoid cancer); asthma; neutrophil chemotaxis (e.g., reperfusion injury from myocardial infarction and stroke and inflammatory arthritis); septic shock; t cell-mediated diseases where immunosuppression is valuable (e.g., prevention of organ transplant rejection, graft versus host disease, systemic lupus erythematosus, multiple sclerosis, and rheumatoid arthritis); atherosclerosis; inhibiting keratinocytes responsive to the growth factor mixture; chronic Obstructive Pulmonary Disease (COPD) and other diseases.
In another aspect, the invention provides a method of treating a disease state for which a mutation in the EGFR gene contributes to the pathology and/or symptomology of the disease, such as melanoma, lung cancer, colon cancer and other types of tumors.
In another aspect, the present invention relates to the use of at least one compound of any one of 1 to 18 and pharmaceutically acceptable salts thereof as a medicament. In another aspect, the invention relates to the use of any one of the compounds of at least one of 1-18 and pharmaceutically acceptable salts thereof for the manufacture of a medicament for inhibiting mutant EGFR.
In another aspect, the invention relates to the manufacture of at least one compound of any one of 1-18 and pharmaceutically acceptable salts thereof as a medicament for treating pathological and/or symptomatic disease states resulting from mutated EGFR activity.
Administration and pharmaceutical compositions
Generally, the compounds of the present invention will be administered in a therapeutically effective amount via any of the usual and acceptable means known in the art, alone or in combination with one or more therapeutic agents. The therapeutically effective amount may vary widely depending on the severity of the disease, age and relative health of the subject, the potency of the compound used and other general skills known in the art. For example, for the treatment of neoplastic diseases and immune system diseases, the required dosage will vary depending upon the mode of administration, the particular condition being treated and the desired effect.
In general, satisfactory results are achieved at daily dosages of from 0.001 to 100mg/kg body weight, in particular from about 0.03 to 2.5mg/kg body weight. Daily doses for larger mammals, such as humans, may be administered in a convenient form, for example in divided doses up to four times a day or in sustained release form, from about 0.5mg to about 2000mg, or more specifically, from 0.5mg to 1000 mg. Suitable unit dosage forms for oral administration contain from about 1 to 50mg of the active ingredient.
The compounds of the invention may be administered in the form of pharmaceutical compositions, by any conventional route; e.g., enterally, e.g., orally, e.g., in the form of tablets or capsules, parenterally, e.g., in the form of injectable solutions or suspensions; or topically, e.g., in the form of a lotion, gel, ointment, or cream, or in the form of a nasal or suppository.
Pharmaceutical compositions comprising a compound of the invention in free base or pharmaceutically acceptable salt form in association with at least one pharmaceutically acceptable carrier or diluent may be manufactured in conventional manner by means of mixing, granulating, coating, dissolving or lyophilizing processes. For example, pharmaceutical compositions comprising a compound of the invention in combination with at least one pharmaceutically acceptable carrier or diluent may be formulated in conventional manner by admixture with a pharmaceutically acceptable carrier or diluent. Unit dosage forms for oral administration contain, for example, from about 0.1mg to about 500mg of active substance.
In one embodiment, the pharmaceutical composition is a solution of the active ingredient, including a suspension or dispersion, such as an isotonic aqueous solution. In the case of lyophilized compositions comprising the active ingredient alone or in admixture with a carrier such as mannitol, the dispersion or suspension may be replenished prior to use. The pharmaceutical compositions may be sterilized and/or contain adjuvants, such as preserving, stabilizing, wetting or emulsifying agents, solution promoters, salts for regulating the osmotic pressure and/or buffers. Suitable preservatives include, but are not limited to, antioxidants such as ascorbic acid, microbicides such as sorbic acid or benzoic acid. The solution or suspension may also contain viscosity increasing agents including, but not limited to, sodium carboxymethylcellulose, dextran, polyvinylpyrrolidone, gelatin, or solubilizing agents such as tween 80 (polyoxyethylene (20) sorbitan monooleate).
Suspensions may contain vegetable oils, synthetic or semi-synthetic oils, as the oily component in the oil, commonly used for injection purposes. Examples include liquid fatty acid esters containing as the acid component a long chain fatty acid having from 8 to 22 carbon atoms, or in some embodiments, from 12 to 22 carbon atoms. Suitable liquid fatty acid esters include, but are not limited to, lauric acid, tridecylic acid, myristic acid, pentadecylic acid, palmitic acid, margaric acid, stearic acid, arachidic acid, behenic acid or corresponding unsaturated acids, such as oleic acid, elaidic acid, erucic acid, brassidic acid and linoleic acid, if desired, with antioxidants such as vitamin E, 3-carotene or 3, 5-di-tert-butylhydroxytoluene. The alcohol component of these fatty acid esters may have six carbon atoms and may be monovalent or polyvalent, such as mono-, di-or trivalent alcohols. Suitable alcohol components include, but are not limited to, methanol, ethanol, propanol, butanol or pentanol or isomers thereof, ethylene glycol and glycerol.
Other suitable fatty acid esters include, but are not limited to, ethyl oleate, isopropyl myristate, isopropyl palmitate, isopropyl myristate, isopropyl palmitate, isopropyl myristate,
Figure BDA0002654899220000131
m2375 (polyoxyethylene glycerol),
Figure BDA0002654899220000132
M1944 CS (unsaturated polyglycolyzed glycerides and mixtures containing glycerides and polyethylene glycol esters from almond oil by alcoholysis), LABRASOLTM(saturated polyglycolized glycerides prepared by alcoholysis of TCM and containing glycerides and polyethylene glycol esters; both available from GaKefosse, France), and/or
Figure BDA0002654899220000133
812 (triglycerides of saturated fatty acids with chain lengths of C8 to C12, from huls AG, germany), and vegetable oils such as cottonseed oil, almond oil, olive oil, castor oil, sesame oil, soybean oil or peanut oil.
Pharmaceutical compositions for oral administration may be obtained, for example, by mixing the active ingredient with one or more solid carriers, if desired granulating a resulting mixture, and processing the mixture or granules by adding further excipients, in the form of tablets or tablet cores.
Suitable carriers include, but are not limited to, fillers, for example sugars, such as lactose, sucrose, mannitol or sorbitol, cellulose preparations and/or calcium phosphates, for example tricalcium phosphate or calcium hydrogen phosphate, and also binders, for example starches, such as corn, wheat, rice or potato starch, methylcellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose and/or polyvinylpyrrolidone, and/or, if desired, disintegrants, such as the above-mentioned starches, carboxymethyl starch, crosslinked polyvinylpyrrolidone, alginic acid or a salt thereof, such as sodium alginate. Additional excipients include flow-regulating agents and lubricants, for example silicic acid, talc, stearic acid or salts thereof, such as magnesium or calcium stearate, and/or polyethylene glycol, or derivatives thereof.
The tablet cores may be provided with a suitable, optionally enteric, coating by using, inter alia, a concentrated sugar solution, which may comprise gum arabic, talc, polyvinylpyrrolidone, polyethylene glycol and/or titanium dioxide, or a coating solution of a suitable organic solvent or solvent mixture, or, for enteric coatings, the preparation of suitable cellulose preparations, such as solutions of acetylcellulose phthalate or hydroxypropylmethylcellulose phthalate. Dyes or pigments may be added to the tablets or tablet coatings, for example for identification purposes or to indicate different doses of the active ingredient.
Pharmaceutical compositions for oral administration may also include hard capsules, including gelatin or soft, sealed capsules containing gelatin and a plasticizer, such as glycerol or sorbitol. Hard capsules may contain the active ingredients in the form of granules, for example in admixture with fillers such as corn starch, binders and/or glidants such as talc or magnesium stearate, and optionally stabilizers. In soft capsules, the active ingredient may be dissolved or suspended in suitable liquid excipients, such as fatty oils, paraffin oils or liquid polyethylene glycols or fatty acid esters of ethylene glycol or propylene glycol, to which stabilizers and detergents, for example of the fatty acid ester type of polyoxyethylene sorbitol, are added.
Pharmaceutical compositions suitable for rectal administration, for example suppositories, comprise a combination of the active ingredient and a suppository base. Suitable suppository bases are, for example, natural or synthetic triglycerides, paraffin hydrocarbons, polyethylene glycols or higher alkanols.
Pharmaceutical compositions suitable for parenteral administration may comprise the active ingredient in water-soluble form, for example as a water-soluble salt or as an aqueous injection suspension comprising a viscosity-increasing substance, for example sodium carboxymethylcellulose, an aqueous solution of sorbitol and/or dextran, if desired, and a stabilizer. The active ingredient, optionally together with excipients, may also be in a lyophilized form and may be prepared as a solution by addition of a suitable solvent prior to parenteral administration. The solutions used, for example for parenteral administration, can also be used as infusion solutions. Injectable preparations are generally prepared under sterile conditions, and filled, for example, in ampoules or vials, and in sealed containers.
The compounds of the invention may be administered as the sole active ingredient or with other drugs useful in immunomodulating therapy or useful against neoplastic diseases. For example, the compounds of the present invention may be used in combination with a pharmaceutical composition effective for each of the above-mentioned diseases, for example, the compounds of the present invention may be used in combination with cyclophosphamide, 5-fluorouracil, fludarabine, gemcitabine, cisplatin, carboplatin, vincristine, vinblastine, etoposide, irinotecan, paclitaxel, docetaxel, rituximab, doxorubicin, gefitinib or imatinib; or also with cyclosporins, rapamycin, ascomycins or their immunosuppressive analogues, e.g. cyclosporins a, cyclosporins G, FK-506, sirolimus and everolimus, glucocorticoids such as: prednisone, cyclophosphamide, azathioprine, methotrexate, gold salts, sulfasalazine, antimalarials, brequinar, leflunomide, mizoribine, mycophenolic acid, phenolate and 15-deoxyspergualin, immunosuppressive monoclonal antibodies, such as monoclonal antibody leukocyte receptors, for example MHC, CD2, CD3, CD4, CD7, CD25, CD28, I CD40, CD45, CD58, CD80, CD86, CD152, CD137, CD154, ICOS, LFA-1, VLA-4 or their ligands, or other immunomodulatory compounds, such as CTLA41 g.
The invention also provides a pharmaceutical combination, e.g. a kit, comprising a) a compound as disclosed herein, in free form or in pharmaceutically acceptable salt form, and b) at least one co-drug. The kit may contain instructions for its use.
Examples
There are various methods for synthesizing the compound or at least one pharmaceutically acceptable salt thereof, and representative methods are exemplified in this example. It is to be noted, however, that at least one compound of formula I or at least one pharmaceutically acceptable salt thereof may also be obtained synthetically by other synthetic routes.
In certain compounds of formula (I), the linkage between an atom and another atom may result in the presence of a particular stereoisomer (e.g., a chiral center). The synthesis of at least one compound of formula (I) or at least one pharmaceutically acceptable salt thereof may result in a mixture of different isomers (enantiomers, diastereomers). Unless a particular configuration is specified, all recited compounds include different stereoisomers that may exist.
At least one compound of formula (I) may also be prepared as a pharmaceutically acceptable acid addition salt, for example, by reacting the free base form of the compound of the invention with a pharmaceutically acceptable inorganic or organic acid. Or reacting at least one compound of formula (I) in free acid form with a pharmaceutically acceptable inorganic or organic base to form a pharmaceutically acceptable base addition salt. Inorganic and organic acids and bases suitable for the preparation of pharmaceutically acceptable salts of the compounds of formula (I) are described in the definitions section of this application. In addition, salt forms of the compounds of formula (I) can also be prepared by using salts of the starting materials or intermediates.
The free acid or base of the compound of formula (I) may be prepared from the corresponding base addition salt or acid addition salt thereof. The acid addition salt forms of the compounds of formula (I) may be converted to the corresponding free base, for example by treatment with a suitable base such as ammonium hydroxide solution, sodium hydroxide and the like. The base addition salt forms of the compounds of formula (I) may be converted to the corresponding free acids, for example by treatment with a suitable acid such as hydrochloric acid and the like.
The N-oxide of at least one compound of formula (I) or at least one pharmaceutically acceptable salt thereof may be prepared by methods known in the art. For example, the N-oxide may be obtained by reacting a non-oxidized form of the compound of formula (I) with an oxidizing agent (e.g., trifluoroperacetic acid, peroxymaleic acid, peroxybenzoic acid, peroxyacetic acid, m-chloroperoxybenzoic acid, and the like) in an inert organic solvent (e.g., a halogenated hydrocarbon such as dichloromethane) at a temperature near 0 ℃. Alternatively, the N-oxides of the compounds of formula (I) may also be prepared from the N-oxides of the starting materials.
The non-oxidized compound of formula I can be prepared by reacting N-oxide thereof with a reducing agent (such as sulfur, sulfur dioxide, triphenylphosphine, lithium borohydride, sodium borohydride, phosphorus trichloride, phosphorus tribromide and the like) in a corresponding inert organic solvent (such as acetonitrile, ethanol, hydrated dioxane and the like) at 0-80 ℃.
Protected derivatives of compounds of formula I may be prepared by methods well known to those skilled in the art. For a detailed technical description of the addition and removal of protecting groups see: greene, Protecting Groups in organic Synthesis,3rd edition, John Wiley & Sons, Inc.1999.
The labels and common sense, charts and examples used in these reactions are consistent with the current scientific literature, e.g., the journal of the American chemical Association or the journal of biochemistry. Unless otherwise indicated, standard single or three letter abbreviations generally refer to L-amino acid residues. All starting materials used were purchased from commercial suppliers and used without further purification unless otherwise indicated. For example, the following abbreviations are used in the examples and throughout the specification: g (g), mg (mg), L (L), mL (mL), μ L (μ L), psi (pounds per square inch), M (mol), mM (mmol), i.v. (i.v.), Hz (Hz), MHz (megahertz), mol (mol), mmol (mmol), RT (ambient temperature), min (min), h (h), mp (melting point), TLC (thin layer chromatography), Rr (retention time), RP (reversed phase), MeOH (methanol), i-PrOH (isopropanol), TEA (triethylamine), TFA (trifluoroacetic acid), TFAA (trifluoroacetic anhydride), THF (tetrahydrofuran), DMSO (dimethyl sulfoxide), EtOAc (ethyl acetate), DME (1, 2-dimethyl ether), DCM (dichloromethane), DCE (dichloroethane), DMF (N, N-dimethylformamide), DMPU (N, N-dimethylpropylurea), CDI (1, 1-carbonyldiimidazole), IBCF (isobutyl chloroformate), HOAc (acetic acid), HOSu (N-hydroxysuccinimide), HOBT (1-hydroxybenzotriazole), Et2O (diethyl ether), EDCI (1- (3-dimethylaminopropyl) 3-ethylcarbodiimide hydrochloride), BOC (tert-butyloxycarbonyl),FMOC (9-fluorenylmethoxycarbonyl), DCC (dicyclohexylcarbodiimide), CBZ (benzyloxycarbonyl), Ac (acetyl), atm (atmospheric pressure), TMSE (2- (trimethylsilyl) ethyl), TMS (trimethylsilyl), TIPS (triisopropylsilane), TBS (t-butyldimethylsilyl), DMAP (dimethylaminopyridine), Me (methyl), OMe (methoxy), Et (ethyl), tBu (t-butyl), HPLC (high performance liquid chromatography), BOP (bis (2-oxo-3-oxazolidinyl) hypophosphoryl chloride), TBAF (tetrabutylammonium fluoride), mCPBA (m-chloroperoxybenzoic acid).
Ether or Et2O is diethyl ether; brine is then a saturated aqueous NaCl solution. Unless otherwise indicated, all temperatures refer to degrees Celsius (degrees Celsius) and all reactions are carried out in an inert atmosphere at room temperature.
1H NMR spectra were recorded using a Bruker Avance 400 NMR spectrometer. Chemical shifts are expressed in ppm. The coupling constants are all in hertz (Hz). Apparent diversity is described in the split mode and is assigned as s (singlet), d (doublet), t (triplet), q (quartet), m (multiplet) and br (generalized peak)
Low resolution Mass Spectrometry (MS) and compound purity data were from a Waters ZQ mass spectrometry single-pole system equipped with an electrospray ion detector (ESI), ultraviolet detectors (220 and 254nm) and Evaporative Light Scattering Detector (ELSD). Thin layer chromatography was performed using 0.25mm E.Merck silica gel plate (60F-254), 5% phosphomolybdic acid in ethanol, ninhydrin or p-phenylene oxide solution and observing under ultraviolet lamp. The flash column chromatography used was silica gel (230- "400 mesh, Merck).
Synthetic route
At least one compound of formula (I) and/or at least one pharmaceutically acceptable salt thereof may be synthesized by various methods, some exemplary methods are provided below and in the examples. Other synthetic methods can be readily suggested by those skilled in the art based on the information disclosed herein.
It may be necessary to protect reactive groups in the reactions described below in order to prevent these reactive groups from participating in other undesired reactions: such as hydroxyl, amino, imino, mercapto or carboxyl groups, which are contained in the final product. Commonly used protecting Groups are referred to T.W.Greene and P.G.M.Wuts in "Protective Groups in organic Chemistry" John Wiley and Sons, 1991.
The synthetic routes for all compounds of the invention are illustrated by the following routes and examples. The starting materials used are commercially available or may be prepared according to established procedures or by methods exemplified herein.
The intermediates listed in the following figures were either obtained from the literature or synthesized according to established analogous synthetic methods.
The compounds of formula I of the present invention may be prepared by the route shown in figure 1. The intermediate of formula III is coupled with amino aromatic hydrocarbon of formula IV through Buchwald ammoniation reaction or other ammoniation conditions provided by literature to obtain intermediate VII or VI, the nitro group of intermediate VII is reduced by iron powder/ammonium chloride in single solvent or mixed solvent such as ethanol-water to obtain amine VIII, and the intermediate VIII can also be obtained by removing the protecting group on the amino group of intermediate VI. Intermediate VI can be obtained by coupling intermediate III with amino-arene V. The amine VIII is condensed with an acid IX or an acid chloride X to give the compound I. The compounds I can also be obtained by reacting the amines VIII first with the acid chlorides XI and then in aqueous solution of a base such as sodium hydroxide, the elimination reaction takes place.
Figure BDA0002654899220000161
An alternative synthetic route to the key intermediate amine VIII is shown in FIG. 2. Amine XIII is coupled with halogenated XIV or XV under similar conditions as shown in FIG. 1 or converted to amine VIII from the literature known nitro compound VII or intermediate VI under conditions as shown in FIG. 1.
Figure BDA0002654899220000171
As shown in fig. 3, in some cases, the compound of formula I is obtained by coupling intermediate III with intermediate VIa under the conditions shown in fig. 1 or fig. 2.
Figure BDA0002654899220000172
The preparation method of VIaa is shown in FIG. 4. The intermediate VIaa is obtained by taking commercially provided dichloropyrimidine as a starting material and sequentially carrying out chlorine regioselective substitution reaction, bromination, amination, deprotection, acetylation and reduction reaction.
Figure BDA0002654899220000181
As an illustration of the preparation of intermediate XIV, the synthetic method of XIVa is shown in fig. 5. Commercially available dichloropyrimidine reacted with an amine followed by a sodium alkyl alkoxide and subsequent halogenation gave the intermediate XIVa.
Figure BDA0002654899220000182
In some cases, the order of carrying out the reactions shown in the above schemes may be adjusted to ensure that the reactions proceed or to avoid side reactions. The following examples are for a full understanding of the present invention and are not to be construed as limiting the invention in any way.
Example 1
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- (4- (1-methyl-1H-indole-3-) Yl) pyrimidin-2-ylamino) pyridin-3-yl) acrylamide (1)
6-chloro-2-methoxy-3-nitropyridine (1a)
To a solution of 2, 6-dichloro-3-nitropyridine (5.79g, 30.0mmol) in dry tetrahydrofuran (30mL) was added methanol (1.15mL, 28.5mmol) at 0 ℃ followed by 4 portions of sodium hydride (60%, 1.20g, 30.0 mmol). After stirring at 0 ℃ for 1h, the mixture was stirred at room temperature for 1 h. The mixture was diluted with water (50mL) and extracted with ethyl acetate (2X 30mL). The extract was washed with brine and dried over anhydrous magnesium sulfate. The solvent was evaporated to dryness and the residue was recrystallized from n-hexane to give 6-chloro-2-methoxy-3-nitropyridine (1a) as a pale yellow solid (4.04g, 71%).
1 1 2 2N- (6-methoxy-5-nitro-compound)Pyridino-2-yl) -N, N, N-trimethylethane-1, 2-diamine (1b)
6-chloro-2-methoxy-3-nitropyridine (1a) (3.77g, 20.0mmol) and N1,N1,N2Heating and refluxing a solution of (2.24g, 22.0mmol) of trimethylethane-1, 2-diamine in ethanol (45mL) for 3h, evaporating off the solvent under reduced pressure, and purifying the residue by column chromatography on silica gel using dichloromethane/methanol/aqueous ammonia (92:6:2) as eluent to give N1- (6-methoxy-5-nitropyridin-2-yl) -N1,N2,N2-trimethylethane-1, 2-diamine (1b) (2.40g, 47%). MS-ESI (M/z) 255[ M +1]]+
1 1 2 2N- (3-bromo-6-methoxy-5-nitropyridin-2-yl) -N, N, N-trimethylethane-1, 2-diamine (1c)
In N1- (6-methoxy-5-nitropyridin-2-yl) -N1,N2,N2A suspension of (1.33g, 5.24mmol) of (1b) trimethylethane-1, 2-diamine in acetonitrile (30mL) was added N-bromosuccinimide (1.40g,7.82mmol) at 0 deg.C and stirred at 0 deg.C for 1 h. The solvent was evaporated under reduced pressure and the residue was purified by column chromatography over silica gel eluting with dichloromethane/methanol/aqueous ammonia (92:6:2) to give N as a yellow solid1- (3-bromo-6-methoxy-5-nitropyridin-2-yl) -N1,N2,N2-trimethylethane-1, 2-diamine (1c) (1.47g, 84%). MS-ESI (M/z):333and 335[1:1, M +1]]+
2 3 2N- (2- (dimethylamino) ethyl) -N- (diphenylmethylene) -6-methoxy-N-methyl-5-nitropyridine-2, 3- Diamine (1d)
Will N1- (3-bromo-6-methoxy-5-nitropyridin-2-yl) -N1,N2,N2-trimethylethane-1, 2-diamine (1c) (666mg,2.00mmol), Pd2(dba)3A mixture of (55mg,0.060mmol), (+/-) -BINAP (75mg,0.120mmol), benzophenone imine (471mg,2.60mmol) and sodium tert-butoxide (250mg,2.60mmol) in toluene (8mL) was heated to 110 ℃ under nitrogen for 2h and then cooled to room temperature. The solid was removed by filtration through celite and washed with ethyl acetate. Distilling the filtrate under reduced pressure to remove the solvent, purifying the residue by silica gel column chromatographyDissolving (eluent is 20-40% ethyl acetate/N-hexane) to obtain N2- (2- (dimethylamino) ethyl) -N3- (diphenylmethanone) -6-methoxy-N2Methyl-5-nitropyridine-2, 3-diamine (1d) as a yellow solid (227mg, 26%). MS-ESI (M/z):434[ M +1 [)]+
2 2N- (2- (dimethylamino) ethyl) -6-methoxy-N-methyl-5-nitropyridine-2, 3-diamine (1e)
At room temperature under N2- (2- (dimethylamino) ethyl) -N3- (diphenylmethanone) -6-methoxy-N2A solution of-methyl-5-nitropyridine-2, 3-diamine (1d) (194mg,0.45mmol) in methanol (5mL) was added concentrated hydrochloric acid (1 mL). After the mixture was stirred at room temperature for 1h, it was diluted with saturated sodium bicarbonate solution and extracted with ethyl acetate (3 ×). The extract was washed with water and brine, and dried over anhydrous magnesium sulfate. The solvent was evaporated under reduced pressure and the residue was purified by silica gel column chromatography (eluent 3-10% methanol/dichloromethane) to give N2- (2- (dimethylamino) ethyl) -6-methoxy-N2Methyl-5-nitropyridine-2, 3-diamine (1e) as a yellow solid (12.4mg, 10%). MS-ESI (M/z) 270[ M +1]]+
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5-nitropyridin-3-yl) acrylamide (1f)
At 0 ℃ to N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-5-nitropyridine-2, 3-diamine (1e) (12.4mg,0.046mmol) in tetrahydrofuran-water (10:1,1mL) was added 3-chloropropionyl chloride (14mg,0.11mmol), stirred at 0 ℃ for 0.5h, added sodium hydroxide (20mg,0.50mmol), the mixture was heated to 60 ℃ for reaction for 3h, cooled to room temperature, diluted with water, extracted with ethyl acetate (2 ×), and the extract solvent was evaporated to dryness under reduced pressure. The residue was purified by silica gel column chromatography (eluent 5% methanol/dichloromethane) to yield N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5-nitropyridin-3-yl) acrylamide (1f) (6.7mg, 45%). MS-ESI (M/z) 324.2[ M +1]]+
N- (5-amino-2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxypyridin-3-yl) acrylamide (1g)
Adding N- (2-((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5-nitropyridin-3-yl) acrylamide (1f) (6.7mg,0.021mmol), iron powder (7.0mg,0.12mmol) and ammonium chloride (1.1mg,0.021mmol) in ethanol-water (3:1,2mL) mixture was heated under reflux for 6h, the solvent was evaporated under reduced pressure, and the residue was purified by silica gel column chromatography with dichloromethane/methanol/aqueous ammonia (92:6:2) as eluent to give N- (5-amino-2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxypyridin-3-yl) acrylamide (1g) (3.6mg, 59%). MS-ESI (M/z):294[ M +1]+
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- (4- (1-methyl-1H-indole-3-) Yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (1)
A mixture of N- (5-amino-2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxypyridin-3-yl) acrylamide (1g) (3.6mg,0.012mmol), 3- (2-chloropyrimidin-4-yl) -1-methyl-1H-indole (1H) (6.0mg,0.025mmol) (prepared according to document WO 2013/14448) and p-toluenesulfonic acid monohydrate (4.7mg,0.025mmol) in 2-pentanol (0.5mL) was heated to 105 ℃ for 2H, the solvent was evaporated under reduced pressure, the residue was purified by silica gel column chromatography with dichloromethane/methanol (95:5) as eluent to give N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- (4- (1-methyl-1H- Indol-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (1) (5.0mg, 82%). MS-ESI (M/z) 500[ M +1]]+
Example 2
N- (6-methoxy-2- (methyl (2- (methylamino) ethyl) amino) -5- ((4- (1-methyl-1H-indole-3-) Yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (2)
Tert-butyl 2- ((6-methoxy-5-nitropyridin-2-yl) (methyl) amino) ethyl (methyl) carbamate (2b)
A solution of 1a (4.00g,21.3mmol) and tert-butylmethyl (2- (methylamino) ethyl) carbamate (2a,4.38g,22.3mmol) (prepared according to the literature, J.Med.chem.,1992,35,565) in ethanol (47mL) was heated under reflux for 3h, the solvent was evaporated under reduced pressure, the residue was purified by silica gel column chromatography with an eluent of 15-30% ethyl acetate/n-hexane to give tert-butyl 2- ((6-methoxy-5-nitropyridin-2-yl) (methyl) amino) Ethyl (methyl) carbamate (2b) (4.65g, 63%) as a yellow solid. MS-ESI (M/z):341[ M +1 [)]+
Tert-butyl 2- ((3-bromo-6-methoxy-5-nitropyridin-2-yl) (methyl) amino) ethyl (methyl) carbamic acid Ester (2c)
To a suspension of tert-butyl 2- ((6-methoxy-5-nitropyridin-2-yl) (methyl) amino) ethyl (methyl) carbamate (2b) (4.32g,12.7mmol) in acetonitrile was added N-bromosuccinimide (3.40g,19.1mmol) at 0 ℃, the mixture was stirred for 1h at 0 ℃, the solvent was evaporated under reduced pressure and the residue was purified by column chromatography on silica gel with 20-30% ethyl acetate/N-hexane as eluent to give tert-butyl 2- ((3-bromo-6-methoxy-5-nitropyridin-2-yl) (methyl) amino) ethyl (methyl) carbamate (2c) as a yellow solid (5.00g, 93%). MS-ESI (M/z):419and 421[1:1, M +1 and M +3 ]]+
Tert-butyl 2- ((3- (diphenylmethylene) -6-methoxy-5-nitropyridin-2-yl) (methyl) amino) ethyl (methyl) Yl) carbamic acid ester (2d)
2- ((3-bromo-6-methoxy-5-nitropyridin-2-yl) (methyl) amino) ethyl (methyl) carbamate (2c) (2.095g,5.00mmol), Pd2(dba)3A mixture of (137mg,0.150mmol), (+/-) -BINAP (187mg,0.300mmol), benzophenone imine (1.177g,6.50mmol) and sodium tert-butoxide (624mg,6.50mmol) in toluene (20mL) was heated to 110 ℃ under nitrogen for 3h, cooled to room temperature, filtered off the solid over celite, washed with ethyl acetate, evaporated to dryness under reduced pressure and the residue was purified by column chromatography over silica gel with 20-40% ethyl acetate/n-hexane as eluent to give tert-butyl 2- ((3- (benzhydrylidene) -6-methoxy-5-nitropyridin-2-yl) (methyl) amino) ethyl (methyl) carbamate (2d) as a light yellow solid (600mg, 23%). MS-ESI (M/z):520[ M +1]]+
Tert-butyl 2- ((3-amino-6-methoxy-5-nitropyridin-2-yl) (methyl) amino) ethyl (methyl) carbamic Acid ester (2e)
To a solution of tert-butyl 2- ((3- (diphenylmethylene) -6-methoxy-5-nitropyridin-2-yl) (methyl) amino) ethyl (methyl) carbamate (2d) (580mg,1.12mmol) in ethanol (20mL) at room temperature was addedHydrochloric acid (1N,0.5mL) was added and the reaction was heated to 50 ℃ for 7 h. The mixture was diluted with saturated sodium bicarbonate solution, extracted with ethyl acetate (3 ×), the extracts washed with water and brine, dried over magnesium sulfate, the solvent evaporated under reduced pressure and the residue purified by column chromatography on silica gel eluting with 20-40% ethyl acetate/hexanes to give tert-butyl 2- ((3-amino-6-methoxy-5-nitropyridin-2-yl) (methyl) amino) ethyl (methyl) carbamate (2e) as an orange solid (280mg, 68%). MS-ESI (M/z):356[ M +1]]+
Tert-butyl 2- ((3-acrylamido-6-methoxy-5-nitropyridin-2-yl) (methyl) amino) ethyl (methyl) Carbamate (2f)
To a solution of tert-butyl 2- ((3-amino-6-methoxy-5-nitropyridin-2-yl) (methyl) amino) ethyl (methyl) carbamate (2e) (280mg,0.789mmol) in tetrahydrofuran-water (10:1,5.5mL) at 0 deg.C was added 3-chloropropionyl chloride (120mg,0.946mmol), stirred at 0 deg.C for 0.5h, added sodium hydroxide NaOH (126mg,3.76mmol), the mixture was heated to 65 deg.C for 1h and cooled to room temperature. The mixture was diluted with water and extracted with ethyl acetate (2 ×). The solvent was evaporated under reduced pressure and the residue was purified by silica gel column chromatography eluting with 30-50% ethyl acetate/hexanes to give tert-butyl 2- ((3-acrylamido-6-methoxy-5-nitropyridin-2-yl) (methyl) amino) ethyl (methyl) carbamate (2f) (185mg, 57%). MS-ESI (M/z) 410.3[ M +1]]+
Tert-butyl 2- ((3-acrylamido-5-amino-6-methoxypyridin-2-yl) (methyl) amino) ethyl (methyl) Carbamate (2g)
Tert-butyl 2- ((3-acrylamido-6-methoxy-5-nitropyridin-2-yl) (methyl) amino) ethyl (methyl) carbamate (2f) (185mg,0.452mmol), iron powder (152mg,2.71mmol), ammonium chloride (24.2mg,0.452mmol) in ethanol-water (3:1,15mL) was heated at reflux for 1 h. The solid was removed by filtration through celite and washed with ethyl acetate. The filtrate was diluted with ethyl acetate, washed with water and brine, and the solvent was evaporated to dryness under reduced pressure to give crude tert-butyl 2- ((3-acrylamido-5-amino-6-methoxypyridin-2-yl) (methyl) amino) ethyl (methyl) carbamate (2g) (149mg, 87%) as a brown oil. MS-ESI (M/z) 380[ M +1]]+
N- (6-methoxy-2- (methyl (2- (methylamino) ethyl) amino) -5- ((4- (1-methyl-1H-indol-3-yl) Pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (2)
A mixture of tert-butyl 2- ((3-acrylamido-5-amino-6-methoxypyridin-2-yl) (methyl) amino) ethyl (methyl) carbamate (2g) (94.0mg,0.248mmol), 3- (2-chloropyrimidin-4-yl) -1-methyl-1H-indole (1H,72.5mg,0.298mmol) and p-toluenesulfonic acid monohydrate (56.6mg,0.298mmol) in 2-pentanol (1.5mL) was heated to 105 ℃ for 2H. The solvent was evaporated under reduced pressure and the residue was purified by silica gel column chromatography eluting with dichloromethane/methanol/ammonia (92:7:1) to give N- (6-methoxy-2- (methyl (2- (methylamino) ethyl) amino) -5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (2) (100.0mg, 83%). MS-ESI (M/z):487.4[ M +1]]+
Example 3
N- (2- ((2- (ethyl (methyl) amino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (1-methyl-1H-) Indol-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (3)
Acetaldehyde (40% in water, 9.0mg,0.082mmol) and sodium triacetoxyborohydride (8.7mg,0.041mmol) were added to a solution of N- (6-methoxy-2- (methyl (2- (methylamino) ethyl) amino) -5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (2) (15.4mg,0.0317mmol) in 1, 2-dichloroethane (1mL) at room temperature, after stirring the mixture at room temperature for 1H, a saturated aqueous sodium bicarbonate solution (5mL) was added to dilute it, dichloromethane was extracted (2X 5mL), the extract was dried over sodium sulfate, the solvent was evaporated under reduced pressure, the residue was purified by silica gel column chromatography with dichloromethane/methanol/ammonia as eluent (95:4:1), n- (2- ((2- (ethyl (methyl) amino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (3) was obtained as a brown solid (12.5mg, 77%). MS-ESI (M/z) 515[ M +1]]+
Example 4
N- (6-methoxy-2- (methyl (2- (N-methylacetamido) ethyl) amino) -5- (4- (1-methyl-1H-indole) Indole-3-yl) pyrimidine-2-Yl) amino) pyridin-3-yl) acrylamide (4)
To a solution of N- (6-methoxy-2- (methyl (2- (methylamino) ethyl) amino) -5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (2) (28.3mg,0.058mmol) in dichloromethane (1mL) was added DIPEA (15.2L,0.087mmol), cooled to 0 deg.C, and a solution of acetic anhydride (5.9 mg in 0.20mL of dichloromethane) was added dropwise. The mixture was stirred at rt for 1H, diluted with saturated aqueous sodium bicarbonate (5mL), extracted with dichloromethane (2 × 5mL), the extracts dried over sodium sulfate, the solvent evaporated under reduced pressure and the residue purified by silica gel column chromatography eluting with 2-3% methanol in dichloromethane to give N- (6-methoxy-2- (methyl (2- (N-methylacetamido) ethyl) amino) -5- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (4) as a yellow solid (15.1mg, 49%). MS-ESI (M/z) 528.4[ M +1]]+
Example 5
N- (6-ethoxy-2- (methyl (2- (methylamino) ethyl) amino) -5- (4- (1-methyl-1H-indol-3-yl) pyrimidine Pyridin-2-yl) amino) pyridin-3-yl) acrylamide (5)
6-chloro-2-ethoxy-3-nitropyridine (5a)
Preparation of 6-chloro-2-ethoxy-3-nitropyridine (5a) methanol was replaced by ethanol according to the synthesis of 1 a. MS-ESI (M/z) 203[ M +1]]+
N- (6-ethoxy-2- (methyl (2- (methylamino) ethyl) amino) -5- (4- (1-methyl-1H-indol-3-yl) pyrimidine Pyridin-2-yl) amino) pyridin-3-yl) acrylamide (5)
Preparation of the title compound N- (6-ethoxy-2- (methyl (2- (methylamino) ethyl) amino) -5- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (5) (36.1mg) 6-chloro-2-methoxy-3-nitropyridine (1a) was replaced with 6-chloro-2-ethoxy-3-nitropyridine (5a) according to the synthesis of example 2. MS-ESI (M/z):501[ M +1]]+
Example 6
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-ethoxy-5- ((4- (1-methyl-1H-indole-3-) Yl) pyrimidin-2-yl) amino) pyridine-3-yl) acrylamide (6)
To a solution of N- (6-ethoxy-2- (methyl (2- (methylamino) ethyl) amino) -5- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (5) (10.1mg,0.020mmol) in 1, 2-dichloroethane (0.6mL) was added acetaldehyde (37% in water, 17mg) and sodium triacetoxyborohydride (6.4mg,0.030mmol) at room temperature, the mixture was stirred at room temperature for 1H, then diluted with saturated aqueous sodium bicarbonate (2mL), dichloromethane was extracted (2 × 2mL), the extract was dried over sodium sulfate, the solvent was evaporated under reduced pressure, the residue was purified by silica gel column chromatography with an eluent of 5-10% dichloromethane/methanol, n- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-ethoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (6) was obtained as a gray solid (8.5mg, 82%). MS-ESI (M/z) 515[ M +1]]+
Example 7
N- (6-isopropoxy-2- (methyl (2- (methylamino) ethyl) amino) -5- (4- (1-methyl-1H-indol-3-yl) Pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (7)
6-chloro-2-isopropoxy-3-nitropyridine (7a)
Preparation of 6-chloro-2-isopropoxy-3-nitropyridine (7a) methanol was replaced by isopropanol according to the synthesis of 1 a. MS-ESI (M/z):217[ M +1]+
N- (6-isopropoxy-2- (methyl (2- (methylamino) ethyl) amino) -5- (4- (1-methyl-1H-indol-3-yl) Pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (7)
Preparation of the title compound 7(36.1mg) 6-chloro-2-methoxy-3-nitropyridine (1a) was replaced with 6-chloro-2-isopropoxy-3-nitropyridine (7a) according to the synthesis of example 2. MS-ESI (M/z) 515[ M +1]]+
Example 8
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-isopropoxy-5- (4- (1-methyl-1H-indole-) 3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (8)
Preparation of the title compound 8(6.3mg, 75%) the synthesis of example 6 was followedProcess for the replacement of N- (6-ethoxy-2- (methyl (2- (methylamino) ethyl) amino) -5- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (5) with N- (6-isopropoxy-2- (methyl (2- (methylamino) ethyl) amino) -5- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (7). MS-ESI (M/z):529[ M +1]+
Example 9
N- (6-methoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- (piperazin-1-yl) Pyridin-3-yl) acrylamide (9)
Preparation of the title compound 9(25mg) tert-butyl (2- (methylamino) ethyl) carbamate (2a) was replaced with tert-butylpiperazine-1-carbamate according to the synthesis method of example 2. MS-ESI (M/z) 485[ M +1]]+
Example 10
N- (6-methoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- (4-methylpiperazine- 1-yl) pyridin-3-yl) acrylamide (10)
Preparation of title compound 10(5.2mg) N- (6-ethoxy-2- (methyl (2- (methylamino) ethyl) amino) -5- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (5) was replaced with N- (6-methoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- (piperazin-1-yl) pyridin-3-yl) acrylamide (9) according to the synthesis of example 6. MS-ESI (M/z):499[ M +1]]+
Example 11
N- (6-ethoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- (piperazin-1-yl) Pyridin-3-yl) acrylamide (11)
Preparation of title compound 11(55mg) according to the synthetic method of example 2, methyl (2- (methylamino) ethyl) carbamate (2a) was replaced with tert-butylpiperazine-1-carboxylate and 6-chloro-2-methoxy-3-nitropyridine (1a) was replaced with 6-chloro-2-ethoxy-3-nitropyridine (5a), respectively. MS-ESI (M/z):499[ M +1]]+
Example 12
N- (6-ethoxy-5- ((4- (1-)methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- (4-methylpiperazine- 1-yl) pyridin-3-yl) acrylamide (12)
Preparation of title compound 12(7.2mg) N- (6-ethoxy-2- (methyl (2- (methylamino) ethyl) amino) -5- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (5) was replaced with N- (6-ethoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- (piperazin-1-yl) pyridin-3-yl) acrylamide (11) according to the synthesis of example 6. MS-ESI (M/z) 513[ M +1]]+
Example 13
N- (6-isopropoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- (piperazin-1- Yl) pyridin-3-yl) acrylamide (13)
Title compound 13(75mg) was prepared according to the synthesis method of example 2 substituting tert-butylmethyl (2- (methylamino) ethyl) carbamate (2a) for tert-butylpiperazine-1-carbamate and 6-chloro-2-methoxy-3-nitropyridine (1a) for 6-chloro-2-isopropoxy-3-nitropyridine (7a), respectively. MS-ESI (M/z) 513[ M +1]]+
Example 14
N- (6-isopropoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- (4-methylpiper Oxazin-1-yl) pyridin-3-yl) acrylamide (14)
Preparation of title compound 14(7.4mg) N- (6-ethoxy-2- (methyl (2- (methylamino) ethyl) amino) -5- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (5) was replaced with N- (6-isopropoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- (piperazin-1-yl) pyridin-3-yl) acrylamide (13) according to the synthesis of example 6. MS-ESI (M/z):527[ M +1 [)]+
Example 15
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -5- ((4- (7-fluoro-1-methyl-1H-indol-3-yl) Pyrimidin-2-yl) amino) -6-methoxypyridin-3-yl) acrylamide (15)
1 1 2 2N- (6-chloro-3-nitropyridin-2-yl)) -N, N, N-trimethylethane-1, 2-diamine (15a)
To a solution of 2, 6-chloro-3-nitropyridine (19.3g,100.0mmol) and triethylamine (20.2g,200.0mmol) in dry dichloromethane (200mL) at 0 deg.C was slowly added N dropwise1,N1,N2A solution of trimethylethane-1, 2-diamine (10.2g,100.0mmol) in dry dichloromethane (20ml) over 0.5h, the reaction mixture was stirred at 0 ℃ for 1.5 h. The mixture was diluted with water (200mL) and extracted with dichloromethane (100 mL. times.3). The combined organic layers were washed with brine (200mL), dried over sodium sulfate, filtered to remove solids, and the filtrate was concentrated to give N1- (6-chloro-3-nitropyridin-2-yl) -N1,N2,N2Trimethylethane-1, 2-diamine (15a) as a yellow oil (23.8g, 91%). MS-ESI (M/z):259[ M +1]+
1 1 2 2N- (6-methoxy-3-nitropyridin-2-yl) -N, N, N-trimethylethane-1, 2-diamine (15b)
At 0-5 ℃ to N1- (6-chloro-3-nitropyridin-2-yl) -N1,N2,N2-trimethylethane-1, 2-diamine (15a) (23.8g,91.9mmol) in methanol (200mL) was added sodium methoxide (10.8g,183.8mmol), the mixture was heated under reflux for 2h, cooled to room temperature, the methanol was evaporated to dryness, diluted with water (200mL), and extracted with dichloromethane (200 mL. times.2). The organic layers were combined, washed with water (200 mL. times.2) and brine (200mL), and dried over sodium sulfate. Filtering to remove solid, concentrating the filtrate under reduced pressure, purifying the crude product by silica gel column chromatography with ethyl acetate/petroleum ether (1:1) as eluent to obtain N1- (6-methoxy-3-nitropyridin-2-yl) -N1,N2,N2Trimethylethane-1, 2-diamine (15b) as a yellow oil (18.0g, 77%). MS-ESI (M/z) 255[ M +1]]+
1 1 2 2N- (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N, N, N-trimethylethane-1, 2-diamine (15c)
At 0-5 ℃ to N1- (6-methoxy-3-nitropyridin-2-yl) -N1,N2,N2-trimethylethane-1, 2-diamine (15b) (11.0g,43.1mmol) in glacial acetic acid (120mL) was added N-iodosuccinimide in 3 portions(11.6g,51.8mmol), and the mixture was stirred at room temperature for 4.5 h. The solvent was evaporated under reduced pressure, diluted with water (300mL), extracted with ethyl acetate (200 mL. times.3), the organic layers were combined, washed with water (200 mL. times.2) and brine (200mL), and dried over sodium sulfate. Filtering to remove solid, concentrating the filtrate under reduced pressure, purifying the crude product by silica gel column chromatography with ethyl acetate/petroleum ether (1:1) as eluent to obtain N1- (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N1,N2,N2Trimethylethane-1, 2-diamine (15c) as a yellow oil (14.0g, 88%). MS-ESI (M/z):381[ M +1]]+
1 1 2 2N- (5- ((diphenylmethylene) amino) -6-methoxy-3-nitropyridin-2-yl) -N, N, N-trimethylethan Alkane-1, 2-diamine (15d)
Under inert nitrogen, adding N1- (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N1,N2,N2A mixture of-trimethylethane-1, 2-diamine (15c) (10.0g,26.2mmol), tris (dibenzylideneacetone) dipalladium (0) (1.2g,1.3mmol), (+/-) -2,2 '-bis (diphenylphosphino) -1,1' -binaphthyl (1.6g,2.6mmol), benzophenone imine (7.1g,39.4mmol) and sodium tert-butoxide (3.8g,39.4mmol) in toluene (150mL) was heated to 110 ℃ for 16 h. Cooling to room temperature, filtering with diatomaceous earth, removing solid, washing with ethyl acetate, evaporating solvent under reduced pressure, purifying the residue by silica gel column chromatography, eluting with ethyl acetate to obtain N1- (5- ((diphenylmethylene) amino) -6-methoxy-3-nitropyridin-2-yl) -N1,N2,N2Trimethylethane-1, 2-diamine (15d) as red oil (2.2g, 19%). MS-ESI (M/z):434[ M +1 [)]+
2 2N- (2- (dimethylamino) ethyl) -6-methoxy-N-methyl-3-nitropyridine-2, 5-diamine (15e)
At room temperature, to N1- (5- ((diphenylmethylene) amino) -6-methoxy-3-nitropyridin-2-yl) -N1,N2,N2A solution of (2.2g,5.1mmol) of (15d) trimethylethane-1, 2-diamine in tetrahydrofuran (150mL) was stirred at room temperature for 1.5h with 0.5N hydrochloric acid (24 mL). The tetrahydrofuran is evaporated to dryness under reduced pressure, and the pH value is adjusted to 9-10 by potassium carbonate. The resulting mixture was treated with dichloromethaneAlkane extraction (30 mL. times.4), combined organic layers, washed with brine (200mL), dried over sodium sulfate. Filtering off solids, concentrating the filtrate under reduced pressure, purifying the residue by silica gel column chromatography with an eluent of dichloromethane/methanol (20: 1-10: 1) to obtain N2- (2- (dimethylamino) ethyl) -6-methoxy-N2Methyl-3-nitropyridine-2, 5-diamine (15e) as a red oil (0.7g, 51%). MS-ESI (M/z) 270[ M +1]]+
7-fluoro-1-methyl-1H-indole (15f)
To a solution of 7-fluoro-1H-indole (1.0g,7.4mmol) in N, N-dimethylformamide (10mL) was added sodium hydride (60% in mineral oil, 0.44g,11.1mmol) at 0-5 deg.C, the resulting suspension was stirred at room temperature for 20 minutes, iodomethane (0.7mL,11.1mmol) was added at room temperature and stirred at room temperature for 1H. The reaction was quenched with water (40mL), the mixture was extracted with ethyl acetate (30 mL. times.3), the organic layers were combined, washed with water (30 mL. times.3) and brine (30mL), dried over sodium sulfate. The solid was filtered off, the filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography eluting with ethyl acetate/petroleum ether (1:20) to give 7-fluoro-1-methyl-1H-indole (15f) as a pale yellow liquid (1.0g, 91%). MS-ESI (M/z) 150[ M +1]]+
3- (2-Chloropyrimidin-4-yl) -7-fluoro-1-methyl-1H-indole (15g)
A solution of 7-fluoro-1-methyl-1H-indole (15f) (0.60g,4.0mmol), 2, 4-dichloropyrimidine (0.60g,4.0mmol), anhydrous iron (III) chloride (0.65g,4.0mmol) in 1, 2-ethanediol dimethyl ether (12mL) was stirred at 60 ℃ for 7H. The reaction was quenched with water (40mL), and the resulting mixture was extracted with ethyl acetate (30 mL. times.3), the organic layers were combined, washed with water (30 mL. times.3) and brine (30mL), and dried over sodium sulfate. The solid was filtered off, the filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography eluting with ethyl acetate/petroleum ether (1: 5-1: 2) to give 3- (2-chloropyrimidin-4-yl) -7-fluoro-1-methyl-1H-indole (15g) as a brown solid (0.50g, 48%). MS-ESI (M/z):262[ M +1 [)]+
2 5N- (2- (dimethylamino) ethyl) -N- (4- (7-fluoro-1-methyl-1H-indol-3-yl) pyrimidin-2-yl) -6-methyl 2oxy-N-methyl-3-nitropyridine-2, 5-diamine (15h)
A reaction of 3 to (2-Chloropyrimidin-4-yl) -7-fluoro-1-methyl-1H-indole (15g) (30.0mg,0.11mmol), N2- (2- (dimethylamino) ethyl) -6-methoxy-N2A mixture of (E) -methyl-3-nitropyridine-2, 5-diamine (15e) (31.0mg,0.11mmol), cesium carbonate (112.0mg,0.33mmol), tris (dibenzylideneacetone) dipalladium (0) (21.0mg,0.022mmol), (+/-) -2,2 '-bis (diphenylphosphino) -1,1' -binaphthyl (29.0mg,0.044mmol) in dioxane (3mL) was stirred at 100 ℃ for 7h and cooled to room temperature. The reaction was quenched with water (40mL), and the resulting mixture was extracted with ethyl acetate (30 mL. times.3), the organic layers were combined, washed with water (30 mL. times.3) and brine (30mL), and dried over sodium sulfate. The solid was filtered off, the filtrate was concentrated under reduced pressure and the residue was purified by column chromatography on silica gel using dichloromethane/methanol/ammonia (100:5:0.5) as eluent to give N2- (2- (dimethylamino) ethyl) -N5- (4- (7-fluoro-1-methyl-1H-indol-3-yl) pyrimidin-2-yl) -6-methoxy-N2Methyl-3-nitropyridine-2, 5-diamine (15h) as a yellow solid (34.0mg, 60%). MS-ESI (M/z):495[ M +1 [)]+
2 5N- (2- (dimethylamino) ethyl) -N- (4- (7-fluoro-1-methyl-1H-indol-3-yl) pyrimidin-2-yl) -6-methyl 2oxy-N-methylpyridine-2, 3, 5-triamine (15i)
Will N2- (2- (dimethylamino) ethyl) -N5- (4- (7-fluoro-1-methyl-1H-indol-3-yl) pyrimidin-2-yl) -6-methoxy-N2A mixture of methyl-3-nitropyridine-2, 5-diamine (15h) (34.0mg,0.07mmol), iron powder (41.0mg,0.73mmol), ammonium chloride (13.0mg,0.24mmol) in ethanol (12mL) and water (3mL) was stirred at 80 ℃ for 1.5 h. The solid was filtered off, washed with ethanol (5 mL. times.3), and the filtrate was concentrated under reduced pressure. Purifying the residue by silica gel column chromatography, eluting with dichloromethane/methanol/ammonia water (100:5: 0.5-100: 10:0.5) to obtain N2- (2- (dimethylamino) ethyl) -N5- (4- (7-fluoro-1-methyl-1H-indol-3-yl) pyrimidin-2-yl) -6-methoxy-N2-methyl-N22,3, 5-Triamine (15i) as a yellow oil (28.0mg, 88%). MS-ESI (M/z):465[ M +1]]+
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -5- ((4- (7-fluoro-1-methyl-1H-indol-3-yl) Pyrimidin-2-yl) amino) -6-methoxypyridin-3-yl) propenesAmide (15)
At 0 ℃ to N2- (2- (dimethylamino) ethyl) -N5- (4- (7-fluoro-1-methyl-1H-indol-3-yl) pyrimidin-2-yl) -6-methoxy-N2-methyl-N2To a solution of (15i) (28.0mg,0.06mmol) of (E) -methylpyridine-2, 3, 5-triamine in anhydrous dichloromethane (1mL) was added acryloyl chloride (5.5mg,0.06mmol), and the mixture was stirred at 0 ℃ for 1 h. The reaction was quenched with saturated aqueous sodium bicarbonate (20mL) and the resulting mixture was extracted with dichloromethane (20 mL. times.3). The organic layers were combined, washed with water (20 mL. times.3) and brine (20mL), and dried over sodium sulfate. The solid was filtered off, the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography eluting with dichloromethane/methanol/ammonia (100:5: 0.5-100: 10:0.5) to give N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -5- ((4- (7-fluoro-1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -6-methoxypyridin-3-yl) acrylamide (15) as a yellow solid (20.0mg, 65%). MS-ESI (M/z):519[ M +1]]+
Example 16
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -5- ((4- (6-fluoro-1-methyl-1H-indol-3-yl) Pyrimidin-2-yl) amino) -6-methoxypyridin-3-yl) acrylamide (16)
Preparation of the title compound 16(20.0mg) 7-fluoro-1H-indole was replaced with 6-fluoro-1H-indole according to the synthesis of example 15. MS-ESI (M/z):519[ M +1]]+
Example 17
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -5- ((4- (5-fluoro-1-methyl-1H-indol-3-yl) Pyrimidin-2-yl) amino) -6-methoxypyridin-3-yl) acrylamide (17)
Preparation of the title compound 17(25.0mg) 7-fluoro-1H-indole was replaced with 5-fluoro-1H-indole according to the synthesis of example 15. MS-ESI (M/z):519[ M +1]]+
Example 18
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -5- ((4- (4-fluoro-1-methyl-1H-indol-3-yl) Pyrimidin-2-yl) amino) -6-methoxypyridin-3-yl) acrylamide (18)
The title compound 18 (23)0mg) 7-fluoro-1H-indole was replaced with 4-fluoro-1H-indole according to the synthesis of example 15. MS-ESI (M/z):519[ M +1]]+
Example 19
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -5- ((5-fluoro-4- (1-methyl-1H-indol-3-yl) Pyrimidin-2-yl) amino) -6-methoxypyridin-3-yl) acrylamide (19)
3- (2-chloro-5-fluoropyrimidin-4-yl) -1-methyl-1H-indole (19a)
A solution of 1-methyl-1H-indole (0.50g,3.8mmol), 2, 4-dichloro-5-fluoropyrimidine (0.64g,3.8mmol) and anhydrous iron (III) chloride (0.62g,3.8mmol) in 1, 2-ethanediol dimethyl ether (7.0mL) was stirred at 60 ℃ for 7H. The reaction was quenched with water (40mL), and the resulting mixture was extracted with ethyl acetate (30 mL. times.3), the organic layers were combined, washed with water (30 mL. times.3) and brine (30mL), and dried over sodium sulfate. The solid was filtered off, the filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography eluting with ethyl acetate/petroleum ether (1: 5-1: 2) to give 3- (2-chloro-5-fluoropyrimidin-4-yl) -1-methyl-1H-indole (19a) as a brown solid (0.54g, 54%). MS-ESI (M/z):262[ M +1 [)]+
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -5- ((5-fluoro-4- (1-methyl-1H-indol-3-yl) Pyrimidin-2-yl) amino) -6-methoxypyridin-3-yl) acrylamide (19)
Preparation of the title compound 19(23.0mg) 3- (2-chloropyrimidin-4-yl) -7-fluoro-1-methyl-1H-indole (15g) was replaced with 3- (2-chloro-5-fluoropyrimidin-4-yl) -1-methyl-1H-indole (19a) according to the synthesis of example 15. MS-ESI (M/z):519[ M +1]]+
Example 20
N- (5- ((5-chloro-4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl Yl) (methyl) amino) -6-methoxypyridin-3-yl) acrylamide (20)
3- (2, 5-dichloropyrimidin-4-yl) -1-methyl-1H-indole (20a)
A solution of 1-methyl-1H-indole (0.50g,3.8mmol), 2,4, 5-trichloropyrimidine (0.64g,3.8mmol) and anhydrous iron (III) chloride (0.62g,3.8mmol) in 1, 2-ethylene glycol dimethyl ether (7.0mL) was addedStirred at 60 ℃ for 7 h. The reaction was quenched with water (40mL), and the resulting mixture was extracted with ethyl acetate (30 mL. times.3), the organic layers were combined, washed with water (30 mL. times.3) and brine (30mL), and dried over sodium sulfate. The solid was filtered off, the filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography eluting with ethyl acetate/petroleum ether (1: 5-1: 2) to give 3- (2, 5-dichloropyrimidin-4-yl) -1-methyl-1H-indole (20a) as a brown solid (0.56g, 53%). MS-ESI (M/z):278[ M +1]+
N- (5- ((5-chloro-4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl Yl) (methyl) amino) -6-methoxypyridin-3-yl) acrylamide (20)
Preparation of the title compound 20(20.0mg) 3- (2-chloropyrimidin-4-yl) -7-fluoro-1-methyl-1H-indole (15g) was replaced with 3- (2, 5-dichloropyrimidin-4-yl) -1-methyl-1H-indole (20a) according to the synthesis of example 15. MS-ESI (M/z):535[ M +1]]+
Example 21
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((5-methyl-4- (1-methyl-1H-) Indol-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (21)
3- (2-chloro-5-methylpyrimidin-4-yl) -1-methyl-1H-indole (21a)
A solution of 1-methyl-1H-indole (0.50g,3.8mmol), 2, 4-dichloro-5-methylpyrimidine (0.64g,3.8mmol) and anhydrous iron (III) chloride (0.62g,3.8mmol) in 1, 2-ethylene glycol dimethyl ether (7.0mL) was stirred at 60 ℃ for 7H. The reaction was quenched with water (40mL), and the resulting mixture was extracted with ethyl acetate (30 mL. times.3), the organic layers were combined, washed with water (30 mL. times.3) and brine (30mL), and dried over sodium sulfate. The solid was filtered off, the filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography eluting with ethyl acetate/petroleum ether (1: 5-1: 2) to give 3- (2-chloro-5-methylpyrimidin-4-yl) -1-methyl-1H-indole (21a) as a brown solid (0.50g, 47%). MS-ESI (M/z):258[ M +1]]+
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((5-methyl-4- (1-methyl-1H-) Indol-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (21)
Preparation of the title compound 21(26.0mg) 3- (2-chloropyrimidin-4-yl) -7-fluoro-1-methyl-1H-indole (15g) was replaced with 21a according to the synthesis of example 15. MS-ESI (M/z) 515[ M +1]]+
Example 22
N- (3- ((2- (dimethylamino) ethyl) (methyl) amino) -5-methoxy-6- ((4- (1-methyl-1H-indole-3-) Yl) pyrimidin-2-yl) amino) pyridin-2-yl) acrylamide (22)
5-bromo-3-methoxy-2-nitropyridine (22a)
Preparation of 5-bromo-3-methoxy-2-nitropyridine (22a) the synthesis described in US2006/84665 was followed.
Tert-butyl (2- ((5-methoxy-6-nitropyridin-3-yl) (methyl) amino) ethyl) (methyl) carbamate (22b)
A mixture of 5-bromo-3-methoxy-2-nitropyridine (22a) (2.8g,12.02mmol), tert-butylmethyl (2- (methylamino) ethyl) carbamate (2.67g,13.22mmol), palladium acetate (0.27g,1.20mmol), Xantphos (0.69g,1.20mmol), and cesium carbonate (11.7g,35.91mmol) in toluene (56mL) was stirred at 100 ℃ for 2.5h under nitrogen. The mixture was cooled to room temperature, quenched with water (200mL), and extracted with ethyl acetate (3X 50 mL). The extract was washed with brine, dried over sodium sulfate, concentrated and purified by column chromatography on silica gel with 50% ethyl acetate/hexanes as eluent to give tert-butyl (2- ((5-methoxy-6-nitropyridin-3-yl) (methyl) amino) ethyl) (methyl) carbamate (22b) as a yellow solid (3.0g, 73%). MS-ESI (M/z):341[ M +1 [)]+
Tert-butyl (2- ((2-bromo-5-methoxy-6-nitropyridin-2-yl) (methyl) amino) ethyl (methyl) carbamic acid methyl ester Acid ester (22c)
To a mixture of tert-butyl (2- ((5-methoxy-6-nitropyridin-3-yl) (methyl) amino) ethyl) (methyl) carbamate (22b) (1.70g,5.00mmol) in glacial acetic acid (30mL) at 0 deg.C was added a solution of bromine (0.88g,5.50mmol) in glacial acetic acid (5 mL). The mixture was stirred at 0 ℃ for 0.5h, at room temperature, diluted with water (150mL) and extracted with ethyl acetate (3X). The extract was washed with saturated aqueous sodium bicarbonate and brine, and dried over sodium sulfate. Evaporating the solvent under reduced pressure, and subjecting the residue to silica gel column chromatographyPurification with 20-40% ethyl acetate/hexanes eluent to give tert-butyl (2- ((2-bromo-5-methoxy-6-nitropyridin-2-yl) (methyl) amino) ethyl (methyl) carbamate (22c) as a yellow solid (1.53g, 73%). MS-ESI (M/z):419.2and 421.2[1:1, M +1 and M +3 ]]+
Tert-butyl (2- ((2- ((diphenylmethylene) amino) -5-methoxy-6-nitropyridin-3-yl) (methyl) amino) Ethyl (methyl) carbamate (22d)
Tert-butyl (2- ((2-bromo-5-methoxy-6-nitropyridin-2-yl) (methyl) amino) ethyl (methyl) carbamate (22c) (419mg,1.00mmol), Pd2(dba)3A mixture of (27.5mg,0.030mmol), (+/-) -BINAP (37.4mg,0.060mmol), benzophenone imine (199mg,1.10mmol) and sodium tert-butoxide (192mg,2.00mmol) in toluene (5mL) was heated to 80 ℃ under nitrogen for 4 h. Cooled to room temperature and the solvent was evaporated under reduced pressure. Tetrahydrofuran (14mL) and 0.5N hydrochloric acid (14mL) were added successively, the mixture stirred at room temperature for 1.5h, diluted with sodium bicarbonate (50mL), and extracted with ethyl acetate (2X). The extract was washed with brine, dried over magnesium sulfate, the solvent evaporated under reduced pressure and the residue purified by silica gel column chromatography eluting with 20-50% ethyl acetate/n-hexane to give tert-butyl (2- ((2- ((diphenylmethylene) amino) -5-methoxy-6-nitropyridin-3-yl) (methyl) amino) ethyl (methyl) carbamate (22d) as a yellow solid (76mg, 21%). MS-ESI (M/z):356.2[ M +1] ethyl (methyl) carbamate (22 d): 356.2]+
Tert-butyl (2- ((2-acrylamido-5-methoxy-6-nitropyridin-3-yl) (methyl) amino) ethyl (methyl) Yl) carbamic acid ester (22e)
To tert-butyl (2- ((2- ((diphenylmethylene) amino) -5-methoxy-6-nitropyridin-3-yl) (methyl) amino) ethyl (methyl) carbamate (22d) at 0 deg.C)(75mg,0.211mmol) in acetonitrile (3mL) was added 3-chloropropionyl chloride (53.6mg,0.422mmol), DMAP (51.5mg,0.422mmol) and DIPEA (81.7mg,0.633 mmol). The mixture was stirred at 0 ℃ for 2.5h, and sodium hydroxide (10N,0.8mL) was added. The mixture was stirred at rt for 1.5h, diluted with water and extracted with ethyl acetate (2 ×). The solvent was evaporated under reduced pressure and the residue was purified by silica gel column chromatography eluting with 5% methanol/dichloromethane to give tert-butyl (2- ((2-propene) 2)Amido-5-methoxy-6-nitropyridin-3-yl) (methyl) amino) ethyl (methyl) carbamate (22e) (28.5mg, 33%). MS-ESI (M/z) 410.2[ M +1]]+
Tert-butyl (2- ((2-acrylamido-6-amino-5-methoxypyridin-3-yl) (methyl) amino) ethyl (methyl) Carbamate (22f)
A mixture of tert-butyl (2- ((2-acrylamido-5-methoxy-6-nitropyridin-3-yl) (methyl) amino) ethyl (methyl) carbamate (22e) (28.5mg,0.070mmol), iron powder (19.5mg,0.348mmol) and ammonium chloride (3.7mg,0.070mmol) in ethanol-water (3:1,15mL) was heated under reflux for 2h, the solvent was evaporated under reduced pressure, the residue was purified by silica gel column chromatography with eluent of 92:6:2 dichloromethane-methanol-ammonia to give tert-butyl (2- ((2-acrylamido-6-amino-5-methoxypyridin-3-yl) (methyl) amino) ethyl (methyl) carbamate (22f) (9.0mg, 34%). MS-ESI (m/z):380.2[ M +1] +.
Tert-butyl (2- ((2-acrylamido-5-methoxy-6- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-) Yl) amino) pyridin-3-yl) (methyl) amino) ethyl (methyl) carbamate (22g)
Tert-butyl (2- ((2-acrylamido-6-amino-5-methoxypyridin-3-yl) (methyl) amino) ethyl (methyl) carbamate (22f) (8.6mg,0.023mmol), 3- (2-chloropyrimidin-4-yl) -1-methyl-1H-indole (1H,6.6mg,0.027mmol), Pd2(dba)3A mixture of (3.4mg,0.0036mmol), BINAP (2.2mg,0.0036mmol) and cesium carbonate (11mg,0.034mmol) in dioxane (0.5mL) was heated to 100 deg.C for 2.5 h. Diluting with water, and extracting with dichloromethane. The solvent was evaporated under reduced pressure and the residue was purified by silica gel column chromatography eluting with 95:5 dichloromethane-methanol to give tert-butyl (2- ((2-acrylamido-5-methoxy-6- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) (methyl) amino) ethyl (methyl) carbamate (22g) (3.3mg, 25%). MS-ESI (M/z):586.3[ M +1] M]+
N- (3- ((2- (dimethylamino) ethyl) (methyl) amino) -5-methoxy-6- ((4- (1-methyl-1H-indole-3-) Yl) pyrimidin-2-yl) amino) pyridin-2-yl) acrylamide (22)
To tert-butyl (2) at room temperature- ((2-acrylamido-5-methoxy-6- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) (methyl) amino) ethyl (methyl) carbamate (22g) (3.3mg) in dichloromethane (0.5mL) trifluoroacetic acid (0.5mL) was added. The mixture was stirred at room temperature for 40 minutes. The solvent is evaporated to dryness, NaBH (OAc) is added3(3.5mg), 1, 2-dichloroethane (0.3mL) and formaldehyde (37% water, 15 mg). The mixture was stirred at room temperature for 1h, diluted with sodium bicarbonate and extracted with dichloromethane. The crude product was purified by column chromatography on silica gel eluting with 95:4:1 dichloromethane-methanol-ammonia to give N- (3- ((2- (dimethylamino) ethyl) (methyl) amino) -5-methoxy-6- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) pyridin-2-yl) acrylamide (22) (2.5mg, 89%). MS-ESP 501.4[ M +1]]+
Example 23
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (1-methyl-1H-indole-3-) Yl) pyrimidin-2-yl) amino) pyridin-3-yl) -2-fluoroacrylamide (23)
2 2 5N- (2- (dimethylamino) ethyl) -6-methoxy-N-methyl-N- (4- (1-methyl-1H-indol-3-yl) pyrimidine Pyridin-2-yl) -3-nitropyridine-2, 5-diamine (23a)
Preparation of the title compound 23a the synthesis of example 2 was followed, adding N1- (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N1,N2,N2Replacement of-trimethylethane-1, 2-diamine (2g) by N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-3-nitropyridine-2, 5-diamine (15 e). MS-ESI (M/z) 477[ M +1]]+
2 2 5N- (2- (dimethylamino) ethyl) -6-methoxy-N-methyl-N- (4- (1-methyl-1H-indol-3-yl) pyrimidine Pyridin-2-yl) pyridine-2, 3, 5-triamine (23b)
Preparation of the title Compound 23b according to the procedure for the synthesis of 15i, N1- (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N1,N2,N2Replacement of-trimethylethane-1, 2-diamine (15h) by N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-N5- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) -3-nitropyridine-2, 5-diamine (23 a). MS-ESI (M/z):447[ M +1]]+
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (1-methyl-1H-indole-3-) Yl) pyrimidin-2-yl) amino) pyridin-3-yl) -2-fluoroacrylamide (23)
Will N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-N5A mixture of (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) pyridine-2, 3, 5-triamine (23b) (20mg,0.045mmol), HOBT (18mg,0.135mmol), EDCI (26mg,0.135), triethylamine (14mg,0.135mmol) in DMF (2ml) was stirred at 35 ℃ for 3H. The reaction was cooled to room temperature and quenched with water (5 mL). The mixture was adjusted to pH 9-10 with sodium carbonate and ethyl acetate (8 mL. times.3). The organic layers were combined, washed with brine (20mL) and dried over sodium sulfate. The solid was removed by filtration, the filtrate was concentrated under low pressure and the residue was purified by column chromatography over silica gel with dichloromethane/methanol (10:1) as eluent to give N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) -2-fluoroacrylamide (23). MS-ESI (M/z):518[ M +1 [)]+
Example 24
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5- ((4- (1-methyl-1H-indole-3-) Yl) pyrimidin-2-yl) amino) phenyl) -2-fluoroacrylamide (24)
N1- (2- (dimethylamino) ethyl) -5-methoxy-N1-methyl-2-nitrobenzene-1, 4-diamine (24a)
4-fluoro-2-methoxy-5-nitroaniline (3.0g,16.0mmol) (prepared according to document WO 2013/14448), N1,N1,N2A mixture of (E) -trimethylethane-1, 2-diamine (2.0g,19.7mmol) and DIPEA (2.7g,20.8mmol) in DMA was stirred at 80 ℃ for 5h and cooled to room temperature. The DMA was evaporated, diluted with water (50mL) and extracted with dichloromethane (50 mL. times.3). Combining the organic layers, washing with water (100mL) and brine (100mL), drying over sodium sulfate, filtering to remove solids, concentrating the filtrate under reduced pressure, and purifying the residue by silica gel column chromatography using dichloromethane/methanol (20: 1-10: 1) as eluent to give N1- (2- (dimethylamino) ethyl) -5-methoxy-N1-methyl-2-nitrobenzene-1, 4-diamine (24 a). MS-ESI (M/z):269[ M + 1%]+
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5- ((4- (1-methyl-1H-indole-3-) Yl) pyrimidin-2-yl) amino) phenyl) -2-fluoroacrylamide (24)
Preparation of the title compound 24 according to the synthesis of example 23, N2- (2- (methoxy) ethyl) -6-methoxy-N2Replacement of-methyl-3-nitropyridine-2, 5-diamine (15e) by N1- (2- (dimethylamino) ethyl) -5-methoxy-N1-methyl-2-nitrobenzene-1, 4-diamine (24 a). MS-ESI (M/z):517[ M + 1%]+
Example 25
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (1-methylindozin-3-yl) Pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (25)
1- (2-ethoxy-2-oxyethyl) -2-ethylpyridine-1-bromide (25a)
To a mixture of 2-ethylpyridine (10g,0.09mol) in ethanol (200mL) was added ethyl 2-bromoacetate (23g,0.14mmol), heated under reflux for 16h, then cooled to room temperature and concentrated under reduced pressure to give crude 1- (2-ethoxy-2-oxyethyl) -2-ethylpyridine-1-carbonyl bromide (25a) which was used in the next reaction without further purification. MS-ESI (M/z):194[ M [)]+
Ethyl 1-methylindoxazine-3-carboxylate (25b)
To a solution of 1- (2-ethoxy-2-oxyethyl) -2-ethylpyridine-1-bromide (25a) (10g,0.036mol) and potassium carbonate (15g,0.10mol) in ethanol (100mL) was added (E) -ethyl 2-cyano-3-ethoxyacrylate (25g,0.14 mol). The mixture was heated under reflux for 15h, then cooled to room temperature, concentrated under reduced pressure, and the residue was extracted with ethyl acetate (50 mL. times.3). The combined organic layers were washed with brine, dried over sodium sulfate, filtered and concentrated under reduced pressure. Purifying the residue by silica gel column chromatography, wherein an eluent is n-hexane/ethyl acetate (50: 1-20: 1) to obtain ethyl 1-methylindoxazine-3-carboxylate (25 b). MS-ESI (M/z) 275[ M +1]]+
N-methoxy-N, 1-dimethylindolizine-3-amide (25c)
To a solution of ethyl 1-methylindoxazine-3-carboxylate (25b) (3.0g,0.014mol) and N, O-dimethylhydroxylamine hydrochloride (3.6g,0.037mol) in tetrahydrofuran (50mL) at 0 deg.C was added isopropylmagnesium chloride (2M in Et2O,35mL,0.07 mol). The mixture was stirred for 1h, quenched with water and extracted with ethyl acetate (50 mL. times.3). The organic layers are combined, washed with brine, dried over sodium sulfate, filtered, concentrated under reduced pressure, and the residue is purified by silica gel column chromatography with N-hexane/ethyl acetate (20: 1-10: 1) as an eluent to obtain N-methoxy-N, 1-dimethylindolizine-3-amide (25 c). MS-ESI (M/z) 219[ M +1]]+
1- (1-methylindozin-3-yl) ethanone (25d)
To a solution of N-methoxy-N, 1-dimethylindolizine-3-amide (25c) (2.6g,0.012mol) in tetrahydrofuran (50mL) at 0 deg.C was added methylmagnesium bromide (3M diethyl ether, 8.0mL,0.024 mol). The mixture was stirred for 1h, quenched with water and extracted with ethyl acetate (50 mL. times.3). The organic layers were combined, washed with brine, dried over sodium sulfate, filtered, and concentrated under reduced pressure to give crude 1- (1-methylindazin-3-yl) ethanone (25d), which was used in the next reaction without further purification. MS-ESI (M/z):174[ M +1]+
(E) -1- (1-methylindozin-3-yl) -3- (pyrrolidin-1-yl) prop-2-en-1-one (25e)
Pyrrolidine (10mL,0.12mol) was added to a solution of 1- (1-methylindozin-3-yl) ethanone (25d) (2.0g,0.012mol) in DMF-DMA (25mL) at 25 ℃. After the mixture was stirred at 90 ℃ for 1 hour, it was extracted with ethyl acetate (50 mL. times.3). The organic layer was washed with brine, dried over sodium sulfate, filtered and concentrated under reduced pressure. Purifying the residue by silica gel column chromatography, wherein an eluent is n-hexane/ethyl acetate (10: 1-2: 1) to obtain (E) -1- (1-methylindozin-3-yl) -3- (pyrrolidine-1-yl) prop-2-ene-1-one (25E). MS-ESI (M/z) 255[ M +1]]+
4- (1-Methylidazin-3-yl) pyrimidin-2-amine (25f)
Guanidine hydrochloride (1.1g,0.19mol) was added to a solution of (E) -1- (1-methylindozin-3-yl) -3- (pyrrolidin-1-yl) prop-2-en-1-one (25E) (1.0g,0.0039mol) and potassium carbonate (2.7g,0.012mol) in isopropanol (10mL) at 25 ℃. After the mixture was stirred at 130 ℃ for 16h, it was cooled to room temperature and extracted with ethyl acetate (50 mL. times.3). Salt for organic layerWashed with water, dried over sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by column chromatography on silica gel eluting with dichloromethane/methanol (30:1) to give 4- (1-methylindozin-3-yl) pyrimidin-2-amine (25 f). MS-ESI (M/z) 225[ M +1]]+
2 2 5N- (2- (dimethylamino) ethyl) -6-methoxy-N-methyl-N- (4- (1-methylindazin-3-yl) pyrimidine-2- Yl) -3-Nitropyridine-2, 5-diamine (25g)
To 4- (1-methylindozin-3-yl) pyrimidin-2-amine (25f) (150mg,0.67mmol), N at 25 deg.C1- (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N1,N2,N2-trimethylethane-1, 2-diamine (15c) (382mg,0.33 mmol) and cesium carbonate (0.87g,2.67mmol) in toluene (5mL) and 1, 4-dioxane (2.5mL) was added Xantphos (193mg,0.33mmol) palladium acetate (75mg,0.33mmol) and the mixture was stirred at 110 ℃ for 3h, cooled to room temperature, ethyl acetate extracted (50mL x 3). organic layer was washed with brine, dried over sodium sulfate, filtered, concentrated under reduced pressure and purified by column chromatography on silica gel with dichloromethane/methanol (40: 1-20: 1) as eluent to give N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-N5- (4- (1-methylindozin-3-yl) pyrimidin-2-yl) -3-nitropyridine-2, 5-diamine (25 g). MS-ESI (M/z) 477[ M +1]]+
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (1-methylindozin-3-yl) Pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (25)
Preparation of the title compound 25 the synthesis of example 15 was followed, adding N1- (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N1,N2,N2Replacement of-trimethylethane-1, 2-diamine (15c) by N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-N5- (4- (1-methylindozin-3-yl) pyrimidin-2-yl) -3-nitropyridine-2, 5-diamine (25 g). MS-ESI (M/z):501[ M +1]]+
Example 26
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5- ((4- (1-methylindozin-3-yl) Pyrimidin-2-yl) Amino) phenyl) acrylamide (26)
N1- (4-iodo-5-methoxy-2-nitrophenyl) -N1, N2, N2-trimethylethane-1, 2-diamine (26a)
At 0 ℃ to N1- (2- (dimethylamino) ethyl) -5-methoxy-N1To a solution of (3.2g,11.9mmol) of (24a) methyl-2-nitrobenzene-1, 4-diamine in (50mL) 1N sulfuric acid was added sodium nitrite (1.28g,17.9 mmol). The mixture was stirred at 0 ℃ for 15 minutes, then potassium iodide (4.0g,23.8mmol) was added. The mixture is stirred for 1h at 90 ℃, cooled to room temperature, and adjusted to pH 9-10 by sodium carbonate. The resulting mixture was extracted with ethyl acetate (20 mL. times.3), the organic layers were combined, washed with brine (20mL), dried over sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by column chromatography on silica gel eluting with dichloromethane/methanol (20:1) to give N1- (4-iodo-5-methoxy-2-nitrophenyl) -N1,N2,N2-trimethylethane-1, 2-diamine (26 a). MS-ESI (M/z) 380[ M +1]]+
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5- ((4- (1-methylindozin-3-yl) Pyrimidin-2-yl) amino) phenyl) acrylamide (26)
Preparation of the title compound 26 the synthesis of example 25 was followed, adding N1- (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N1,N2,N2Replacement of-trimethylethane-1, 2-diamine (15c) by N1- (4-iodo-5-methoxy-2-nitrophenyl) -N1,N2,N2-trimethylethane-1, 2-diamine (26 a). MS-ESI (M/z) 500[ M +1]]+
Example 27
N- (5- ((4- (1-cyanoindolizin-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) Amino) -6-methoxypyridin-3-yl acrylamide (27)
1- (2-ethoxy-2-oxyethyl) pyridine-1-bromide (27a)
To a mixture of pyridine (10g,0.09mol) in ethanol (200mL) was added ethyl 2-bromoacetate (23g,0.14mmol), heated at reflux for 16h, then cooled to room temperature and concentrated under reduced pressure to give crude 1- (2-ethoxy-2-oxyethyl) pyridine-1-bromide (27)a) Used in the next reaction without further purification. MS-ESI (M/z):167[ M ]]+
Ethyl 1-cyanoindolizine-3-carboxylic acid ester (27b)
To a solution of 1- (2-ethoxy-2-oxyethyl) pyridine-1-bromide (27a) (10g,0.04mol) and triethylamine (29mL,0.2mol) in ethanol (250mL) was added (E) -3-methacrylonitrile (10g,0.12 mol). The mixture was heated under reflux for 15h, then cooled to room temperature, concentrated under reduced pressure, and the residue was extracted with ethyl acetate (50 mL. times.3). The combined organic layers were washed with brine, dried over sodium sulfate, filtered and concentrated under reduced pressure. Purifying the residue by silica gel column chromatography, wherein an eluent is n-hexane/ethyl acetate (50: 1-20: 1), and obtaining the ethyl 1-cyanoindolizine-3-carboxylic ester (27 b). MS-ESI (M/z):215[ M +1]]+
N- (5- ((4- (1-cyanoindolizin-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) Amino) -6-methoxypyridin-3-yl acrylamide (27)
Preparation of the title compound 27 according to the synthesis method of example 25, ethyl 1-methylindolizine-3-carboxylate (25b) was replaced with ethyl 1-cyanoindolizine-3-carboxylate (27 b). MS-ESI (M/z):501[ M +1]]+
Example 28
N- (5- ((4- (1-cyanoindolizin-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) Amino) -4-methoxyphenyl) acrylamide (28)
Preparation of the title compound 28 according to the procedure for the synthesis of example 27, N1- (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N1,N2,N2Replacement of-trimethylethane-1, 2-diamine (15c) by N1- (4-iodo-5-methoxy-2-nitrophenyl) -N1,N2,N2-trimethylethane-1, 2-diamine (26 a). MS-ESI (M/z) 511[ M +1]]+
Example 29
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -5- ((4- (indolizin-3-yl) pyrimidin-2-yl) amino) - 6-methoxypyridin-3-yl) acrylamide (29)
Preparation of the title compound 29 according to example 25The synthesis method of (1) is to replace 2-ethylpyridine by 2-methylpyridine. MS-ESI (M/z):487[ M +1]]+
Example 30
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -5- ((4- (indolizin-3-yl) pyrimidin-2-yl) amino) - 4-methoxyphenyl) acrylamide (30)
Preparation of the title compound 30 according to the synthesis of example 29, N1- (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N1,N2,N2Replacement of-trimethylethane-1, 2-diamine (15c) by N1- (4-iodo-5-methoxy-2-nitrophenyl) -N1,N2,N2-trimethylethane-1, 2-diamine (26 a). MS-ESI (M/z):486[ M +1 [)]+
Example 31
N- (5- ((4- (1-Chloroindazin-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino 6-methoxypyridin-3-yl acrylamide (31)
Ethyl indolizine-3-carboxylic acid ester (31a)
Preparation of the title compound, ethyl indolizine-3-carboxylate (31a) 2-ethylpyridine was replaced with 2-methylpyridine according to the synthetic procedure for compound 25 b. MS-ESI (M/z) 190[ M +1]]+
Ethyl 1-chloridazine-3-carboxylate (31b)
To a solution of ethyl indolizine-3-carboxylate (31a) (0.4g,2.2mmol) in DMF (5mL) at 0 ℃ was added N-chlorosuccinimide (0.3g,2.2mmol), the mixture was stirred at 25 ℃ for 1h, extracted with ethyl acetate (50mL × 3), the combined organic layers were washed with brine, dried over sodium sulfate, and concentrated under reduced pressure. The residue was purified by column chromatography on silica gel using n-hexane/ethyl acetate (50:1) as eluent to give ethyl 1-chloroindolizine-3-carboxylate (31 b). MS-ESI (M/z):224[ M +1 [)]+
N- (5- ((4- (1-Chloroindazin-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino 6-methoxypyridin-3-yl) acrylamide(31)
Preparation of the title compound 31 according to the synthesis of example 25, ethyl 1-methylindoxazine-3-carboxylate (25b) is replaced by ethyl 1-chloroindolizine-3-carboxylate (31 b). MS-ESI (M/z):521[ M +1]]+
Example 32
N- (5- ((4- (1-Chloroindazin-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino Yl) -4-methoxyphenyl) acrylamide (32)
Preparation of the title compound 32 the synthesis of example 31 was followed, adding N1- (5-iodo-6-methoxy-3-nitropyridine-2-
Radical) -N1,N2,N2Replacement of-trimethylethane-1, 2-diamine (15c) by N1- (4-iodo-5-methoxy-2-nitrophenyl) -N1,N2,N2-trimethylethane-1, 2-diamine (26 a). MS-ESI (M/z):520[ M +1]]+
Example 33
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (3-methylindozin-1-yl) Pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (33)
3-methylindolizine-1-carbonitrile (33a)
A mixture of 2-bromopropanal (7.26g,53.4mmol) and 2-pyridineacetonitrile (6.3g,53.4mmol) in tetrahydrofuran (70mL) was heated at 70 ℃ for 24 h. The mixture was concentrated under reduced pressure, the residue was diluted with ethyl acetate and hydrochloric acid (20mL,2M), and the organic layer was washed with saturated sodium bicarbonate solution and brine, dried over sodium sulfate, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography using petroleum ether/ethyl acetate (10:1 to 5:1) as eluent to give 3-methylindoxazine-1-carbonitrile (33 a). MS-ESI (M/z) 157[ M +1]]+
1- (3-methylindozin-1-yl) ethane-1-imine (33b)
To a solution of 3-methylindoxazine-1-carbonitrile (33a) (1.6g,10.3mmol) in tetrahydrofuran was added 3M methylmagnesium bromide (18mL) in an ice-water bath, and the mixture was stirred at room temperature for 15 h. The mixture was quenched with saturated ammonium chloride solution and extracted with dichloromethane. The organic layer was washed with brine, dried over sodium sulfate, and concentrated under reduced pressure to give 1- (3-methylindozin-1-yl) ethane-1-imine (33 b). MS-ESI (M/z):173[ M +1]]+
1- (3-Methylidazin-1-yl) ethan-1-one (33c)
To a solution of 1- (3-methylindazin-1-yl) ethane-1-imine (33b) (1.0g,5.8mmol) in tetrahydrofuran was added 6N sulfuric acid (60mL) at room temperature, heated to 55 ℃ for 24h, and cooled to room temperature. The reaction mixture was diluted with water and dichloromethane, the organic layer was separated, washed with saturated sodium bicarbonate solution and brine, dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by column chromatography on silica gel to give 1- (3-methylindozin-1-yl) ethan-1-one (33 c). MS-ESI (M/z):174[ M +1]+
1- (3-methylindozin-1-yl) -3- (pyrrolidin-1-yl) prop-2-en-1-one (33d)
To a solution of 1- (3-methylindozin-1-yl) ethan-1-one (33c) (110mg,0.7mmol) in DMF at 25 ℃ was added DMF-DMA (170mg,1.4mmol) and pyrrolidine (50mg,0.7 mmol). The mixture was stirred for 1.5h at 90 ℃. After cooling to room temperature, extraction was carried out with ethyl acetate. The organic layer was washed with brine, dried over sodium sulfate and concentrated under reduced pressure to give crude 1- (3-methylindozin-1-yl) -3- (pyrrolidin-1-yl) prop-2-en-1-one (33 d). MS-ESI (M/z) 255[ M +1]]+
4- (3-Methylidazin-1-yl) pyrimidin-2-amine (33e)
To a solution of 1- (3-methylindozin-1-yl) -3- (pyrrolidin-1-yl) prop-2-en-1-one (33d) (300mg,1.2mmol) in isopropanol was added guanidine hydrochloride (337mg,3.5mmol) and potassium carbonate (662mg,4.8 mmol). The mixture was stirred in a sealed tube at 120 ℃ for 20 h. Cooled to room temperature and extracted with ethyl acetate. The organic layer was washed with brine, dried over sodium sulfate, filtered, and concentrated under reduced pressure to give 4- (3-methylindazin-1-yl) pyrimidin-2-amine (33 e). MS-ESI (M/z) 225[ M +1]]+
2 2 5N- (2- (dimethylamino) ethyl) -6-methoxy-N-methyl-N- (4- (3-methylindazin-3-yl) pyrimidine-2- Yl) -3-nitropyridine-2, 5-diamine (33f)
A mixture of 4- (3-methylindozin-1-yl) pyrimidin-2-amine (33e) (30.0mg,0.13mmol), 25c (76.0mg,0.20mmol), palladium acetate (15.0mg,0.07mmol), Xantphos (38.0mg,0.07mmol) and cesium carbonate (170mg,0.52mmol) in toluene (1mL) and dioxane (0.5mL) was heated to 110 ℃ under nitrogen to reactAnd 3 h. Cooling to room temperature, adding water for dilution, and extracting with ethyl acetate. The organic layer was washed with brine, dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by column chromatography on silica gel eluting with 9% methanol in dichloromethane to give N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-N5- (4- (3-methylindozin-3-yl) pyrimidin-2-yl) -3-nitropyridine-2, 5-diamine (33 f). MS-ESI (M/z) 477[ M +1]]+
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (3-methylindozin-1-yl) Pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (33)
Preparation of the title compound 33 according to the synthesis of example 15, N1- (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N1,N2,N2Replacement of-trimethylethane-1, 2-diamine (15c) by N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-N5- (4- (3-methylindozin-3-yl) pyrimidin-2-yl) -3-nitropyridine-2, 5-diamine (33 f). MS-ESI (M/z):501[ M +1]]+.
Example 34
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5- ((4- (3-methylindozin-1-yl) Pyrimidin-2-yl) amino) phenyl) acrylamide (34)
Preparation of the title compound 34 the synthesis of example 33 was followed, N1- (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N1,N2,N2Replacement of-trimethylethane-1, 2-diamine (15c) by N1- (4-iodo-5-methoxy-2-nitrophenyl) -N1,N2,N2-trimethylethane-1, 2-diamine (26 a). MS-ESI (M/z) 500[ M +1]]+
Example 35
N- (5- ((4- (3-Cyanoindolizin-1-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) Amino) -6-methoxypyridin-3-yl acrylamide (35)
1-Acetylindolizine-3-carbonitrile (35a)
1- (cyanomethyl) pyridine-1-bromide (10g,50.3mmol), 4-methoxybutaneA mixture of (5.1g,50.3mmol) of (E) -3-en-2-one and (10g,100.6mmol) of triethylamine in ethanol (100mL) was heated at 80 ℃ for 7 h. The reaction mixture was concentrated under reduced pressure, the residue was diluted with dichloromethane and water, the organic layer was separated, washed with brine, dried over sodium sulfate and concentrated. The residue was stirred in isopropanol for 0.5h, filtered and dried to give 1-acetylindolizine-3-carbonitrile (35a) as a yellow solid. MS-ESI (M/z) 185[ M +1]]+
1- (3- (pyrrolidin-1-yl) acryloyl) indolizine-3-carbonitrile (35b)
To a solution of 1- (3-methylindozin-1-yl) ethan-1-one (33c) (110mg,0.7mmol) in DMF was added DMF-DMA (170mg,1.4mmol) and pyrrolidine (50mg,0.7 mmol). The mixture was stirred for 1.5h at 90 ℃. After cooling to room temperature, extraction was carried out with ethyl acetate. The organic layer was washed with brine, dried over sodium sulfate and concentrated under reduced pressure to give crude 1- (3-methylindozin-1-yl) -3- (pyrrolidin-1-yl) prop-2-en-1-one (33 d). To a solution of 1-acetylindolizine-3-carbonitrile (35a) (400mg,2.17mmol) in DMF was added DMF-DMA (516mg,4.34mmol) and pyrrolidine (308mg,4.34 mmol). The mixture was stirred for 1.5h at 90 ℃. After cooling to room temperature, extraction was carried out with ethyl acetate. The organic layer was washed with brine, dried over sodium sulfate, and concentrated under reduced pressure to give crude 1- (3- (pyrrolidin-1-yl) acryloyl) indolizine-3-carbonitrile (35 b). MS-ESI (M/z) 266[ M +1]]+
1- (2-Aminopyrimidin-4-yl) indolizine-3-carbonitrile (35c)
To a solution of 1- (3- (pyrrolidin-1-yl) acryloyl) indolizine-3-carbonitrile (35b) (637mg,2.4mmol) in 1-butanol was added guanidine hydrochloride (690mg,7.2mmol) and potassium carbonate (1.3g,9.6 mmol). The mixture was stirred for 20h at 115 ℃. After cooling to room temperature, water was added for dilution, and extraction was performed with ethyl acetate. The organic layer was washed with brine, dried over sodium sulfate, and concentrated under reduced pressure to give crude 1- (2-aminopyridin-4-yl) indolizine-3-carbonitrile (35 c). MS-ESI (M/z) 236[ M +1]]+
N- (5- ((4- (3-Cyanoindolizin-1-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) Amino) -6-methoxypyridin-3-yl acrylamide (35)
Preparation of the title compound 35 according to the procedure for the synthesis of example 33, 4- (3-methylindozin-1-yl) pyrimidin-2-amine (33e) was replaced with 1- (2-aminopyridin-4-yl)) Indolizine-3-carbonitrile (35 c). MS-ESI (M/z):512[ M +1 [)]+
Example 36
N- (5- ((4- (3-Cyanoindolizin-1-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) Amino) -4-methoxyphenyl) acrylamide (36)
Preparation of the title compound 36 the synthesis of example 35 was followed, adding N1- (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N1,N2,N2Replacement of-trimethylethane-1, 2-diamine (15c) by N1- (4-iodo-5-methoxy-2-nitrophenyl) -N1,N2,N2-trimethylethane-1, 2-diamine (26 a). MS-ESI (M/z) 511[ M +1]]+
Example 37
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -5- ((4- (indolizin-1-yl) pyrimidin-2-yl) amino) - 6-methoxypyridin-3-yl) acrylamide (37)
1- (2-ethoxy-2-oxyethyl) pyridine-1-bromide (37a)
To a mixture of pyridine (10g,0.09mol) in ethanol (200mL) was added ethyl 2-bromoacetate (23g,0.14mmol), heated at reflux for 16h, then cooled to room temperature to give crude 1- (2-ethoxy-2-oxyethyl) pyridine-1-bromide (27a) which was used in the next reaction without further purification. MS-ESI (M/z):167[ M ]]+. A solution of pyridine (2.00g,25.30mmol) and ethyl 2-bromoacetate (6.3g,38.00mmol) in ethanol (20mL) was heated at 70 ℃ for 1h under nitrogen. Concentrated under reduced pressure, the residue was filtered through celite and washed with petroleum ether to give 6.9 g of 1- (2-ethoxy-2-oxyethyl) pyridine-1-carbonyl bromide (37 a). MS-ESI (M/z) 167[ M +1]]+
Ethyl 1-acetylindolizine-3-carboxylate (37b)
To a solution of 1- (2-ethoxy-2-oxyethyl) pyridine-1-bromide (27a) (10g,0.04mol) and triethylamine (29mL,0.2mol) in ethanol (250mL) was added (E) -3-methacrylonitrile (10g,0.12 mol). The mixture was heated at reflux for 15h, concentrated under reduced pressure, and the residue (50 mL. times.3). The combined organic layers were washed with brine, dried over sodium sulfate, filtered and concentrated under reduced pressure. Purifying the residue by silica gel column chromatography with eluent ofN-hexane/ethyl acetate (50: 1-20: 1) to obtain ethyl 1-cyanoindolizine-3-carboxylate. To a solution of 1- (2-ethoxy-2-oxyethyl) pyridine-1-bromide (37a) (5.50g,15.10mmol) and 4-methoxybut-3-en-2-one (2.3g,22.3mmol) in ethanol (65mL) at 20 deg.C was added triethylamine (4.50g,44.60 mmol). The mixture was heated at 80 ℃ overnight, then cooled to room temperature, diluted with water and extracted with ethyl acetate (3X 50 mL). The organic layer was concentrated under reduced pressure. The residue was purified by column chromatography on silica gel to give ethyl 1-acetylindolizine-3-carboxylate (37b) (2.65 g). MS-ESI (M/z) 232[ M +1]]+
1- (indolizin-1-yl) ethan-1-one (37c)
To a solution of ethyl 1-acetylindolizine-3-carboxylate (37b) (2.60g,11.3mmol) in methanol (30mL) at 20 deg.C was added 10% sodium hydroxide (2.0g,50.6m mol). The mixture was heated at 65 ℃ for 1h, the solvent was removed by concentration under reduced pressure, polyphosphoric acid (10.0g) and methanol (40ml) were added, and the mixture was heated at 65 ℃ for 40 minutes. Cooled to room temperature, diluted with water, extracted with dichloromethane (3X 50mL), washed with sodium bicarbonate and brine, dried over sodium sulfate, and concentrated to give 1- (indolizin-1-yl) ethan-1-one (37c) (1.23 g). MS-ESI (M/z) 160[ M +1]]+
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -5- ((4- (indolizin-1-yl) pyrimidin-2-yl) amino) - 6-methoxypyridin-3-yl) acrylamide (37)
Preparation of the title compound 37 following the procedure for the synthesis of example 35, 1-acetylindolizine-3-carbonitrile (35a) was replaced with 1- (indolizin-1-yl) ethan-1-one (37 c). MS-ESI (M/z):487[ M +1]]+
Example 38
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -5- ((4- (indolizin-1-yl) pyrimidin-2-yl) amino) - 4-methoxyphenyl) acrylamide (38)
Preparation of the title compound 38 the procedure for the synthesis of example 37 was followed, mixing N1- (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N1,N2,N2Replacement of-trimethylethane-1, 2-diamine (15c) by N1- (4-iodo-5-methoxy-2-nitrophenyl) -N1,N2,N2-trimethylethane-1, 2-diamine (26 a). MS (Mass Spectrometry)-ESI(m/z):486[M+1]+
Example 39
N- (5- ((4- (3-Chloroindolizin-1-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino 6-methoxypyridin-3-yl acrylamide (39)
1- (3-Chlorindolizin-1-yl) ethan-1-one (39a)
A mixture of 1- (indolizin-1-yl) ethan-1-one (37c) and cuprous chloride (962mg,5.7mmol) in acetonitrile (6mL) was stirred at room temperature for 4h, diluted with water (10mL) and extracted with dichloromethane (3X 10 mL). The extract was dried over sodium sulfate and concentrated under reduced pressure. Purifying the residue by silica gel column chromatography, wherein the eluent is petroleum ether/ethyl acetate (8: 1-6: 1) to obtain 1- (3-chloridazin-1-yl) ethane-1-ketone (39 a). MS-ESI (M/z):194[ M +1]]+
N- (5- ((4- (3-Chloroindolizin-1-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino 6-methoxypyridin-3-yl acrylamide (39)
Preparation of the title compound 39 following the procedure for the synthesis of example 35, 1-acetylindolizine-3-carbonitrile (35a) was replaced with 1- (3-chloroindolizin-1-yl) ethan-1-one (39 a). MS-ESI (M/z):521[ M +1]]+
Example 40
N- (5- ((4- (3-Chloroindolizin-1-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino Yl) -4-methoxyphenyl) acrylamide (40)
Preparation of the title compound 40 according to the procedure for the synthesis of example 39, N1- (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N1,N2,N2Replacement of-trimethylethane-1, 2-diamine (15c) by N1- (4-iodo-5-methoxy-2-nitrophenyl) -N1,N2,N2-trimethylethane-1, 2-diamine (26 a). MS-ESI (M/z):520[ M +1]]+
EXAMPLE 41
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (3-methyl-1H-indole-1-) Yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (41)
1- (2-Chloropyrimidin-4-yl) -3-methyl-1H-indole (41a)
To a solution of 2, 4-dichloropyrimidine (2.89g,20.0mmol) and 3-methyl-1H-indole (1.31g,10.0mmol) in DMA (50mL) at room temperature were added HOBT (306mg,2.0mmol) and potassium carbonate (1.93g,14.0mmol), the mixture was stirred at 70 ℃ for 24H, diluted with water (50mL), extracted with ethyl acetate (2X 30mL), the extract washed with brine, dried over sodium sulfate, and concentrated under reduced pressure. The residue was purified by column chromatography on silica gel eluting with petroleum ether/ethyl acetate (10: 1-5: 1) to give 1- (2-chloropyrimidin-4-yl) -3-methyl-1H-indole (41a) as a yellow solid (1g, 40%).
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (3-methyl-1H-indole-1-) Yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (41)
Preparation of the title compound 41(17.3mg) 3- (2-chloropyrimidin-4-yl) -1-methyl-1H-indole (1H) was replaced with 1- (2-chloropyrimidin-4-yl) -3-methyl-1H-indole (41a) as in 1. MS-ESI (M/z):501[ M +1]]+
Example 42
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5- ((4- (3-methyl-1H-indole-1-) Yl) pyrimidin-2-yl) amino) phenyl) acrylamide (42)
N- (4-fluoro-2-methoxy-5-nitrophenyl) -4- (3-methyl-1H-indol-1-yl) pyrimidin-2-amine (42a)
A mixture of 1- (2-chloropyrimidin-4-yl) -3-methyl-1H-indole (41a) (278mg,1.14mmol), 4-fluoro-2-methoxy-5-nitroaniline (213mg,1.14mmol) (prepared according to document WO 2013/14448), and p-toluenesulfonic acid monohydrate (260mg,1.37mmol) in 2-pentanol (4mL) was heated at 105 ℃ for 2H, filtered, and concentrated under reduced pressure to give N- (4-fluoro-2-methoxy-5-nitrophenyl) -4- (3-methyl-1H-indol-1-yl) pyrimidin-2-amine (42a) (320mg, 72%). MS-ESI (M/z):394[ M +1 [)]+
1 1 4N- (2- (dimethylamino) ethyl) -5-methoxy-N-methyl-N- (4- (3-methyl-1H-indol-1-yl) pyrimidine Pyridin-2-yl) -2-nitrophenyl-1, 4-diamine (42b)
Reacting N- (4-fluoro-2-methoxy-5-nitrophenyl) -4- (3-methyl-1)H-indol-1-yl) pyrimidin-2-amine (42a) (160mg,0.407mmol), N1,N1,N2A mixture of trimethylethane-1, 2-diamine (0.064mL,0.489mmol) and DIPEA (0.088mL,0.529mmol) in DMA was heated at 85 ℃ for 3 h. Concentrating under reduced pressure, purifying the residue by silica gel column chromatography eluting with dichloromethane/methanol/ammonia (92:6:2) to obtain N1- (2- (dimethylamino) ethyl) -5-methoxy-N1-methyl-N4- (4- (3-methyl-1H-indol-1-yl) pyrimidin-2-yl) -2-nitrophenyl-1, 4-diamine (42b) (139mg, 73%). MS-ESI (M/z):476[ M +1 [)]+
1 1 4N- (2- (dimethylamino) ethyl) -5-methoxy-N-methyl-N- (4- (3-methyl-1H-indol-1-yl) pyrimidine Pyridin-2-yl) -2-nitrophenyl-1, 4-diamine (42c)
Will N1- (2- (dimethylamino) ethyl) -5-methoxy-N1-methyl-N4- (4- (3-methyl-1H-indol-1-yl) pyrimidin-2-yl) -2-nitrobenzene-1, 4-diamine (42b) (139mg,0.293mmol), iron powder (98mg,1.76mmol) and ammonium chloride (22.0mg,0.293mmol) in ethanol-water (3:1,15mL) mixture was heated at reflux for 1H. The pad was filtered through celite, washed with ethyl acetate, the washings diluted with ethyl acetate, washed with water and brine. The solvent was evaporated under reduced pressure and the residue was purified by silica gel column chromatography eluting with dichloromethane/methanol/ammonia (92:6:2) to give N1- (2- (dimethylamino) ethyl) -5-methoxy-N1-methyl-N4- (4- (3-methyl-1H-indol-1-yl) pyrimidin-2-yl) -2-nitrophenyl-1, 4-diamine (42c) (120mg, 92%). MS-ESI (M/z):446[ M +1]]+
3-chloro-N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5- ((4- (3-methyl-1H-indole) Indol-1-yl) pyrimidin-2-yl) amino) phenyl) propionamide (42d)
At 0 ℃ to N1- (2- (dimethylamino) ethyl) -5-methoxy-N1-methyl-N4- (4- (3-methyl-1H-indol-1-yl) pyrimidin-2-yl) -2-nitrobenzene-1, 4-diamine (42c) (120mg,0.27mmol) in tetrahydrofuran-water (10:1,3mL) was added 3-chloropropionyl chloride (0.031mL,0.32mmol), stirred at 0 ℃ for 2H, added ammonia (0.5mL), diluted with water and extracted with ethyl acetate. Evaporating the solvent, purifying the residue by silica gel column chromatographyEluent 2-5% methanol/dichloromethane to give 3-chloro-N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5- ((4- (3-methyl-1H-indol-1-yl) pyrimidin-2-yl) amino) phenyl) propanamide (42d) (100mg, 69%). MS-ESI (M/z):536[ M +1]+
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5- ((4- (3-methyl-1H-indole-1-) Yl) pyrimidin-2-yl) amino) phenyl) acrylamide (42)
To a solution of 3-chloro-N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5- ((4- (3-methyl-1H-indol-1-yl) pyrimidin-2-yl) amino) phenyl) propionamide (42d) (100mg,0.187mmol) in tetrahydrofuran-water (10:1,2mL) was added sodium hydroxide (60mg,1.5mmol) at room temperature, stirred for 2H, diluted with water and extracted with ethyl acetate. The solvent was evaporated under reduced pressure and the residue was purified by silica gel column chromatography eluting with 1-2% methanol/dichloromethane to give N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5- ((4- (3-methyl-1H-indol-1-yl) pyrimidin-2-yl) amino) phenyl) acrylamide (42) (60mg, 65%). MS-ESI (M/z) 500[ M +1]]+
Example 43
N- (6-methoxy-5- ((4- (3-methyl-1H-indol-1-yl) pyrimidin-2-yl) amino) -2- (4-methylpiperazine- 1-yl) pyridin-3-yl) acrylamide (43)
N- (4-methoxy-5- ((4- (3-methyl-1H-indol-1-yl) pyrimidin-2-yl) amino) -2- (piperazin-1-yl) Phenyl) acrylamide (43a)
Preparation of the title compound 43a (10mg) according to the synthetic method of example 41, N1,N1,N2-trimethylethane-1, 2-diamine with tert-butylpiperazine-1-carboxylate. MS-ESI (M/z) 485[ M +1]]+
N- (6-methoxy-5- ((4- (3-methyl-1H-indol-1-yl) pyrimidin-2-yl) amino) -2- (4-methylpiperazine- 1-yl) pyridin-3-yl) acrylamide (43)
Preparation of title compound 43(4.9mg) according to the procedure for the synthesis of example 6, N- (6-ethoxy-2- (methyl (2- (methylamino) ethyl) amino) -5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) pyridine-3-yl) acrylamide (5) was replaced by N- (4-methoxy-5- ((4- (3-methyl-1H-indol-1-yl) pyrimidin-2-yl) amino) -2- (piperazin-1-yl) phenyl) acrylamide (43 a). MS-ESI (M/z):499[ M +1]]+
Example 44
N- (4-methoxy-5- ((4- (3-methyl-1H-indol-1-yl) pyrimidin-2-yl) amino) -2- (4-methylpiperazine- 1-yl) phenyl) acrylamide (44)
Preparation of the title compound 44 the procedure was followed for the synthesis of example 42, coupling N1,N1,N2-trimethylethane-1, 2-diamine is replaced by 1-methylpiperazine. MS-ESI (M/z) 498[ M +1]]+
Example 45
N- (5- ((4- (3-cyano-1H-indol-1-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (meth) amino) -6-methoxypyridin-3-yl acrylamide (45)
1- (2-Chloropyrimidin-4-yl) -1H-indole-3-carbonitrile (45b)
Preparation of the title compound, 1- (2-chloropyrimidin-4-yl) -1H-indole-3-carbonitrile (45b) (220mg) according to the synthetic method of 41a, 3-methyl-1H-indole was replaced with 1H-indole-3-carbonitrile (45a) (preparation of this reagent according to the reference: JOC 1958,23, 1178). MS-ESI (M/z) 255[ M +1]]+
N- (5- ((4- (3-cyano-1H-indol-1-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (meth) amino) -6-methoxypyridin-3-yl acrylamide (45)
Preparation of the title compound 45(7.0mg) according to the synthesis of example 15, 3- (2-chloropyrimidin-4-yl) -7-fluoro-1-methyl-1H-indole (15g) was replaced with 1- (2-chloropyrimidin-4-yl) -1H-indole-3-carbonitrile (45 b). MS-ESI (M/z):512[ M +1 [)]+
Example 46
N- (5- ((4- (3-cyano-1H-indol-1-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (meth) amino) -4-methoxyphenyl acrylamide (46)
Preparation of the title compound 46(5mg) according to the synthetic method of example 45, N2- (2- (two)Methylamino) ethyl) -6-methoxy-N2Replacement of-methyl-3-nitropyridine-2, 5-diamine (15e) by N1- (2- (dimethylamino) ethyl) -5-methoxy-N1-methyl-2-nitropyridine-1, 4-diamine (24 a). MS-ESI (M/z) 511[ M +1]]+
Example 47
N- (5- ((4- (1-chloroimidazole [1,5-a ]))]And pyridine-3-yl pyrimidine-2-yl) amino) -2- ((2- (dimethylamino) Ethyl) (methyl) amino) -6-methoxypyridin-3-yl) acrylamide (47)
2- (methylthio) -N- (pyridin-2-yl) pyrimidine-4-amide (47a)
A mixture of pyridin-2-ylmethylamine (900mg,8.3mmol),2- (methylthio) pyrimidine-4-carboxylic acid (1g,5.9mmol) (prepared according to document WO 2006/117368), HOBT (2.4g,17.7mmol), EDCI (3.4g,17.7mmol), triethylamine (1.78g,17.7mmol) in DMF (10ml) was stirred at room temperature for 2 h. The reaction is cooled to room temperature, quenched by adding water (10mL), adjusted to pH 9-10 by sodium carbonate, and extracted by ethyl acetate (15 mL. times.3). The combined organic layers were washed with brine (20mL), dried over sodium sulfate, filtered to remove solids, the filtrate was concentrated, and the residue was purified by silica gel column chromatography using petroleum ether/ethyl acetate (2: 1-1: 1) as eluent to give 2- (methylthio) -N- (pyridin-2-yl) pyrimidine-4-amide (47 a). MS-ESI (M/z):247[ M + 1[ ]]+
3- (2- (methylthio) pyrimidin-4-yl) imidazole [1,5-a]Pyridine (47b)
A solution of 2- (methylthio) -N- (pyridin-2-yl) pyrimidine-4-amide (47a) (370mg,1.5mmol) in acetonitrile (2mL) was added to phosphorus oxychloride (2.5mL), and the mixture was stirred at 85 ℃ for 20 h. Cooled to room temperature and quenched with water (10 mL). The pH was adjusted to 8-9 with sodium hydroxide and extracted with dichloromethane (8 mL. times.3). The combined organic layers were washed with brine (20mL), dried over sodium sulfate and concentrated under reduced pressure to give crude 3- (2- (methylthio) pyrimidin-4-yl) imidazo [1,5-a]And pyridine (47 b). MS-ESI (M/z):243[ M +1 [)]+
1-chloro-3- (2- (methylthio) pyrimidin-4-yl) imidazo [1,5-a]Pyridine (47c)
To 3- (2- (methylthio) pyrimidin-4-yl) imidazo [1,5-a ] at room temperature]Pyridine (200mg,0.83mmol) in DMF (2mL) was added N-chlorosuccinimide (144mg,1.08mmol), stirred at room temperature for 2h, addedWater quench (5 mL). The resulting mixture was extracted with dichloromethane (5 mL. times.2), the organic layers were combined, washed with brine (10mL), dried over sodium sulfate, and concentrated under reduced pressure to give crude 1-chloro-3- (2- (methylthio) pyrimidin-4-yl) imidazo [1,5-a]And pyridine (47 c). MS-ESI (M/z):277[ M +1]]+
4- (1-chloroimidazole [1,5-a ]]And pyridin-3-yl) pyrimidin-2-ol (47d)
To 1-chloro-3- (2- (methylthio) pyrimidin-4-yl) imidazo [1,5-a]To a solution of the pyridine (47c) (150mg,0.54mmol) in NMP (5mL) was added 10% sodium hydroxide (2mL) and the mixture was stirred at 100 ℃ for 24 h. The reaction was cooled to room temperature and the pH was adjusted to 5-6 with 2N hydrochloric acid. The resulting mixture was extracted with dichloromethane (10 mL. times.3), the organic layers were combined, washed with brine (20mL), dried over sodium sulfate, and concentrated to give crude 4- (1-chloroimidazo [1,5-a ] product]And pyridin-3-yl) pyrimidin-2-ol (47 d). MS-ESI (M/z):247[ M + 1[ ]]+
1-chloro-3- (2-chloropyrimidin-4-yl) imidazo [1,5-a]Pyridine (47e)
4- (1-chloroimidazole [1,5-a ]]Pyridopyridin-3-yl) pyrimidin-2-ol (47d) (140mg,0.57mmol) in acetonitrile (1mL) phosphorus oxychloride (1mL) was added and the mixture stirred at 80 ℃ for 1.5h, cooled to room temperature and quenched with water (5 mL). The pH was adjusted to 8-9 with sodium hydroxide and extracted with dichloromethane (8 mL. times.3). The combined organic layers were washed with brine (20mL), dried over sodium sulfate and concentrated under reduced pressure to give crude 1-chloro-3- (2-chloropyrimidin-4-yl) imidazo [1,5-a]And pyridine (47 e). MS-ESI (M/z):265[ M +1 [)]+
1 4N- (4- (1-chloroimidazole [1,5-a ]]Pyridin-3-yl) pyrimidin-2-yl-N- (2- (dimethylamino) ethyl) -2- 4methoxy-N-methyl-5-nitrobenzene-1, 4-diamine (47f)
1-chloro-3- (2-chloropyrimidin-4-yl) imidazole [1,5-a ] is reacted with a catalyst]Pyridine (47e) (54mg,0.20mmol), N2- (2- (dimethylamino) ethyl) -6-methoxy-N2A mixture of methyl-3-nitropyridine-2, 5-diamine (15e) (61mg,0.22mmol) and methanesulfonic acid (27mg,0.24mmol) in isopropanol (2.5mL) was stirred at 90 ℃ for 3h, the reaction was cooled to room temperature, quenched with water (10mL), adjusted to pH 8-9 with sodium carbonate, and extracted with ethyl acetate (30 mL. times.3). Combine the organic layers, wash with water (30mL) and brine (30mL), dry over sodium sulfate, filter to remove solids, concentrate the filtrate, and leaveThe residue was purified by silica gel column chromatography eluting with dichloromethane/methanol (10:1) to give N1- (4- (1-chloroimidazo [1,5-a ]]And pyridin-3-yl) pyrimidin-2-yl) -N4- (2- (dimethylamino) ethyl) -2-methoxy-N4-methyl-5-nitrobenzene-1, 4-diamine (47 f). MS-ESI (M/z) 498[ M +1]]+
N- (5- ((4- (1-chloroimidazole [1,5-a ]))]And pyridine-3-yl pyrimidine-2-yl) amino) -2- ((2- (dimethylamino) Ethyl) (methyl) amino) -6-methoxypyridin-3-yl) acrylamide (47)
Preparation of the title compound (47) according to the synthesis of example 15, N2- (2- (dimethylamino) ethyl) -N5- (4- (7-fluoro-1-methyl-1H-indol-3-yl) pyrimidin-2-yl) -6-methoxy-N2Replacement of-methyl-3-nitropyridine-2, 5-diamine (15h) by N1- (4- (1-chloroimidazo [1,5-a ]]And pyridin-3-yl) pyrimidin-2-yl) -N4- (2- (dimethylamino) ethyl) -2-methoxy-N4-methyl-5-nitrobenzene-1, 4-diamine (47 f). MS-ESI (M/z):468[ M +1 [)]+
Example 48
N- (5- ((4- (1-chloroimidazole [1,5-a ]))]And pyridine-3-yl pyrimidine-2-yl) amino) -2- ((2- (dimethylamino) Ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide (48)
Preparation of the title compound 48 according to the procedure for the synthesis of example 47, N2- (2- (dimethylamino) ethyl) -6-methoxy-N2Replacement of-methyl-3-nitropyridine-2, 5-diamine (15e) by N1- (2- (dimethylamino) ethyl) -5-methoxy-N1-methyl-2-nitropyridine-1, 4-diamine (24 a). MS-ESI (M/z):521[ M +1]]+
Example 49
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (1- (trifluoromethyl) imidazole) [1,5-a]And pyridin-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (49)
1-iodo-3- (2- (methylthio) pyrimidin-4-yl) imidazole [1,5-a]Pyridine (49a)
To 3- (2- (methylthio) pyrimidin-4-yl) imidazo [1,5-a ] at room temperature]And pyridine (47b) (300mg,1.24 mmo)l) to a solution of DMF (5mL) was added N-iodosuccinimide (362mg,1.61mmol) and the mixture was stirred at room temperature for 2 h. The reaction was quenched with water (5mL) and the resulting mixture was extracted with dichloromethane (8 mL. times.2). The combined organic layers were washed with brine (10mL), dried over sodium sulfate, and concentrated under reduced pressure to give 1-iodo-3- (2- (methylthio) pyrimidin-4-yl) imidazo [1,5-a]And pyridine (49 a). MS-ESI (M/z):369[ M +1]]+
3- (2- (methylthio) pyrimidin-4-yl) -1- (trifluoromethyl) imidazole [1,5-a]Pyridine (49b)
1-iodo-3- (2- (methylmercapto) pyrimidine-4-yl) imidazole [1,5-a]A mixture of bipyridine (49a) (200mg,0.54mmol), (trifluoromethyl) trimethylsilane (153mg,1.08mmol), phenanthroline (194mg,1.08mmol), trimethylborate (112mg,1.08mmol), potassium fluoride (63mg,1.08mmol), and copper (I) iodide (154mg,0.81mmol) in dimethylsulfoxide (5mL) was stirred at 60 deg.C overnight. The reaction was cooled to room temperature, quenched with water (10mL), and extracted with ethyl acetate (10 mL. times.3). The combined organic layers were washed with water (20mL) and brine (20mL), dried over sodium sulfate, filtered to remove the solid, the filtrate was concentrated, and the residue was purified by column chromatography on silica gel using ethyl acetate/petroleum ether (1:2) as eluent to give 3- (2- (methylthio) pyrimidin-4-yl) -1- (trifluoromethyl) imidazo [1,5-a]And pyridine (49 b). MS-ESI (M/z):311[ M +1 [)]+
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (1- (trifluoromethyl) imidazole) [1,5-a]And pyridin-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (49)
Preparation of the title compound 49 according to the procedure for the synthesis of example 47, 1-chloro-3- (2- (methylthio) pyrimidin-4-yl) imidazo [1,5-a]Replacement of the pyridopyridine (47c) with 3- (2- (methylthio) pyrimidin-4-yl) -1- (trifluoromethyl) imidazole [1,5-a ]]And pyridine (49 b). MS-ESI (M/z):556[ M +1 [)]+
Example 50
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5- ((4- (1- (trifluoromethyl) imidazole) [1,5-a]And pyridin-3-yl) pyrimidin-2-yl) amino) phenyl) acrylamide (50)
Preparation of the title compound 50 according to the procedure for the synthesis of example 47, N2-(2- (dimethylamino) ethyl) -6-methoxy-N2Replacement of-methyl-3-nitropyridine-2, 5-diamine (15e) by N1- (2- (dimethylamino) ethyl) -5-methoxy-N1-methyl-2-nitropyridine-1, 4-diamine (24 a). MS-ESI (M/z) 555[ M +1]]+
Example 51
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (1-methylimidazol [1, 5-a)] And pyridin-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (51)
1-methyl-3- (2- (methylmercapto) pyrimidin-4-yl) imidazole [1,5-a]Pyridine (51a)
Preparation of the title compound 51a pyridin-2-ylmethylamine was replaced with 1- (pyridin-2-yl) ethan-1-amine according to the synthesis of 47 b. MS-ESI (M/z):257[ M +1]]+
3- (2-Chloropyrimidin-4-yl) -1-methylimidazol [1,5-a]Pyridine (51b)
Preparation of the title Compound 51b according to the procedure for the synthesis of 47e, 1-chloro-3- (2- (methylthio) pyrimidin-4-yl) imidazole [1,5-a]Replacement of the pyridopyridine (47c) with 1-methyl-3- (2- (methylthio) pyrimidin-4-yl) imidazo [1,5-a]And pyridine (51 a). MS-ESI (M/z):245[ M +1 [)]+
2 2 5N- (2- (dimethylamino) ethyl) -6-methoxy-N-methyl-N- (4- (1-methylimidazol [1, 5-a)]Pyridine- 3-yl) pyrimidin-2-yl) pyridine-2, 3, 5-triamine (51c)
Preparation of the title compound (51c) according to the procedure for the synthesis of 15i, 3- (2-chloropyrimidin-4-yl) -7-fluoro-1-methyl-1H-indole (15g) was replaced with 3- (2-chloropyrimidin-4-yl) -1-methylimidazol [1, 5-a)]And pyridine (51 b). MS-ESI (M/z):448[ M +1]]+
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (1-methylimidazol [1, 5-a)] And pyridin-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (51)
Preparation of the title compound 51 the synthesis of example 15 was followed, adding N2- (2- (dimethylamino) ethyl) -N5- (4- (7-fluoro-1-methyl-1H-indol-3-yl) pyrimidin-2-yl) -6-methoxy-N2-methyl radical-N2Replacement of-methylpyridine-2, 3, 5-triamine (15i) by N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-N5- (4- (1-methylimidazole [1,5-a ]]And-pyridin-3-yl) pyrimidin-2-yl) pyridine-2, 3, 5-triamine (51 c). MS-ESI (M/z):502[ M +1 [)]+
Example 52
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5- ((4- (1- (methylimidazole [1, 5-) a]And pyridin-3-yl) pyrimidin-2-yl) amino) phenyl) acrylamide (52)
Preparation of the title compound 52 according to the synthesis of example 51, N2- (2- (dimethylamino) ethyl) -6-methoxy-N2Replacement of-methyl-3-nitropyridine-2, 5-diamine (15e) by N1- (2- (dimethylamino) ethyl) -5-methoxy-N1-methyl-2-nitropyridine-1, 4-diamine (24 a). MS-ESI (M/z):501[ M +1]]+.
Example 53
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (1-methylimidazol [1, 5-a)] And pyridin-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) -2-fluoroacrylamide (53)
Preparation of the title compound (53) according to the synthesis of example 23, N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-N5Replacement of- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) pyridine-2, 3, 5-triamine (23b) by N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-N5- (4- (1-methylimidazole [1,5-a ]]And-pyridin-3-yl) pyrimidin-2-yl) pyridine-2, 3, 5-triamine (51 c). MS-ESI (M/z):520[ M +1]]+
Example 54
N- (5- ((4- (1, 2-dihydropyrrole [3,2, 1-hi)]Indol-5-yl pyrimidin-2-yl) amino) -2- ((2- (dimethylin) Amino) ethyl) (methyl) amino) -6-methoxypyridin-3-yl) acrylamide (54)
2- (2-Nitrophenyl) ethanol (54a)
A mixture of 1-methyl-2-nitrobenzene (48g,0.35mol), formaldehyde (10.4g,0.35mol), 20% sodium hydroxide (3.6mL) and dimethyl sulfoxide was heated at 50 ℃ for 2h, then the mixture was cooled to room temperature, diluted with water (200mL), and extracted with ethyl acetate (300 mL. times.3). The combined organic layers were washed with water (200 mL. times.2) and brine (200mL), dried over sodium sulfate, filtered to remove solids, the filtrate was concentrated, and the residue was purified by column chromatography over silica gel using ethyl acetate/petroleum ether (1:1) as eluent to give 2- (2-nitrophenyl) ethanol (54a) as a brown oil (14.4g, 25%).
Tert-butyldimethyl (2-nitrophenylethoxy) silane (54b)
To a solution of 2- (2-nitrophenyl) ethanol (54a) (1.7g,0.01mol) in dry dichloromethane (15ml) were added imidazole (0.88g,0.013mol) and tert-butyldimethylchlorosilane (2.1g,0.014mol), and the mixture was stirred at room temperature for 2 h. The mixture was then filtered, diluted with dichloromethane (30mL), washed with 1N hydrochloric acid (30mL), water and aqueous sodium bicarbonate, the organic layers combined, dried over sodium sulfate, the solid removed by filtration, and the filtrate concentrated to give tert-butyldimethyl (2-nitrophenylethoxy) silane (54b) as a brown oil (2.8g, 97%).
7- (2- ((tert-butyldimethylsilyl) oxy) ethyl) -1H-indole (54c)
To a solution of tert-butyldimethyl (2-nitrophenylethoxy) silane (54b) (4.5g,16mmol) in dry tetrahydrofuran (50ml) was added vinyl magnesium bromide (56ml,56mmol) dropwise at-70 ℃ under nitrogen. The mixture was slowly warmed to room temperature, poured into aqueous ammonium chloride, and extracted with ethyl acetate (50 mL). The combined organic layers were dried over sodium sulfate, filtered to remove solids, the filtrate was concentrated and the residue was purified by silica gel column chromatography eluting with ethyl acetate/petroleum ether (1:100) to give 7- (2- ((tert-butyldimethylsilyl) oxy) ethyl) -1H-indole (54c) as a yellow oil (2.6g, 59%). MS-ESI (M/z):276[ M +1 [)]+
2- (3- (2-Chloropyrimidin-4-yl) -1H-indol-7-yl) ethanol (54d)
Preparation of the title compound 54d (0.33g, 30%, brown solid) 7-fluoro-1-methyl-1H-indole (15f) was replaced with 7- (2- ((tert-butyldimethylsilyl) oxy) ethyl) -1H-indole (54c) according to 15g of the synthesis. MS-ESI (M/z):274[ M +1]]+
7- (2-chloroethyl) -3- (2-chloropyrimidin-4-yl) -1H-Indole (54e)
A suspension of 2- (3- (2-chloropyrimidin-4-yl) -1H-indol-7-yl) ethanol (54d) (50mg,0.18mmol) in dry CAN (5ml) was added dropwise to sulfinyl chloride (0.44mg,0.36mmol) at 0 ℃. The resulting mixture was stirred at room temperature overnight, cooled to room temperature, extracted with aqueous sodium bicarbonate (20mL) and extracted with ethyl acetate (20 mL). The combined organic layers were washed with water (20 mL. times.2) and brine (200mL), dried over anhydrous sodium sulfate, filtered to remove the solid, the filtrate was concentrated, and the residue was purified by silica gel column chromatography using ethyl acetate/petroleum ether (1:3) as eluent to give 7- (2-chloroethyl) -3- (2-chloropyrimidin-4-yl) -1H-indole (54e) as a brown solid (0.30g, 50%). MS-ESI (M/z) 292[ M +1]]+
5- (2-Chloropyrimidin-4-yl) -1, 2-dihydropyrrolo [3,2,1-hi]Indole (54f)
To a solution of 7- (2-chloroethyl) -3- (2-chloropyrimidin-4-yl) -1H-indole (54e) (33mg,0.11mmol) in DMF (3ml) at room temperature was added cesium carbonate (78mg,0.22mmol) and the mixture was heated at 60 ℃ for 10H. The mixture was cooled to room temperature, quenched with water (20mL), and extracted with ethyl acetate (20 mL). The combined organic layers were washed with water (20 mL. times.2) and brine (200mL), dried over anhydrous sodium sulfate, filtered to remove solids, and the filtrate was concentrated to give 5- (2-chloropyrimidin-4-yl) -1, 2-dihydropyrrolo [3,2,1-hi]Indole (54f) as a brown solid (0.30g, 100%). MS-ESI (M/z) 256[ M +1]]+
N- (5- ((4- (1, 2-dihydropyrrole [3,2, 1-hi)]Indol-5-yl pyrimidin-2-yl) amino) -2- ((2- (dimethylin) Amino) ethyl) (methyl) amino) -6-methoxypyridin-3-yl) acrylamide (54)
Preparation of the title compound 54(5.0mg) according to the procedure for the synthesis of 15, 3- (2-chloropyrimidin-4-yl) -7-fluoro-1-methyl-1H-indole (15g) was replaced with 5- (2-chloropyrimidin-4-yl) -1, 2-dihydropyrrolo [3,2,1-hi]Indole (54 f). MS-ESI (M/z) 513[ M +1]]+
Example 55
N- (5- ((4- (1, 2-dihydropyrrole [3,2, 1-hi)]Indol-5-yl pyrimidin-2-yl) amino) -2- ((2- (dimethylin) Amino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide (55)
Preparation of the title compound (55) according to the synthesis of example 54N1- (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N1,N2,N2Replacement of-trimethylethane-1, 2-diamine (15c) by N1- (4-iodo-5-methoxy-2-nitrophenyl) -N1,N2,N2-trimethylethane-1, 2-diamine (24 a). MS-ESI (M/z):512[ M +1 [)]+
Example 56
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (3-methyl-1H-indazol-1-) Yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (56)
3-methyl-1H-indazole (56a)
To a solution of 1- (2-fluorophenyl) ethanone (20g,0.14mol) in ethane-1, 2-diol (70ml) was added hydrazine hydrate (4.7ml,0.15mol) at room temperature, the temperature was maintained for 2h, and then the mixture was heated to 150 ℃ overnight. The mixture was cooled to room temperature, quenched with water (200mL) and extracted with dichloromethane (200 mL). The combined organic layers were washed with water (200 mL. times.2) and brine (200mL), dried over anhydrous sodium sulfate, filtered to remove the solid, the filtrate was concentrated, and the residue was purified by column chromatography over silica gel using ethyl acetate/petroleum ether (1:20) as eluent to give 3-methyl-1H-indazole (56a) as a brown solid (7g, 37%). MS-ESI (M/z) 133[ M +1]]+
1- (2-Chloropyrimidin-4-yl) -3-methyl-1H-indazole (56b)
To a solution of 3-methyl-1H-indazole (56a) (1.1g,8.3mmol) in dry DMF at 0 deg.C was added sodium hydride, the temperature was maintained for 2H, then 2, 4-dichloropyrimidine (1.24g,8.3mmol) was added and the resulting mixture stirred at room temperature overnight. Quenched by addition of aqueous ammonium chloride (20mL) and extracted with ethyl acetate (50 mL). The combined organic layers were washed with water (50 mL. times.2) and brine (50mL), dried over anhydrous sodium sulfate, filtered to remove the solid, the filtrate was concentrated, and the residue was purified by silica gel column chromatography using ethyl acetate/petroleum ether (1:20) as eluent to give 1- (2-chloropyrimidin-4-yl) -3-methyl-1H-indazole (56b) as a white solid (0.85g, 43%). MS-ESI (M/z):245[ M +1 [)]+
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (3-methyl-1H-indazol-1-) Yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamides(56)
Preparation of title compound 56(8.0mg) according to the procedure for the synthesis of example 15, 3- (2-chloropyrimidin-4-yl) -7-fluoro-1-methyl-1H-indole (15g) was replaced with 1- (2-chloropyrimidin-4-yl) -3-methyl-1H-indazole (56 b). MS-ESI (M/z):502[ M +1 [)]+
Example 57
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (3-methyl-1H-indazol-1-) Yl) pyrimidin-2-yl) amino) pyridin-3-yl) -2-fluoroacrylamide (57)
Preparation of the title compound 57(5.0mg) 3- (2-chloropyrimidin-4-yl) -1-methyl-1H-indole (1H) was replaced with 1- (2-chloropyrimidin-4-yl) -3-methyl-1H-indazole (56b) according to the synthesis of example 23. MS-ESI (M/z):520[ M +1]]+
Example 58
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (1-methyl-1H-indazol-3-) Yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (58)
1- (1-methyl-1H-indazol-3-yl) ethanone (58a)
The title compound 58a was prepared according to the literature: heterocylic chem.,2013,50: E221.
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (1-methyl-1H-indazol-3-) Yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (58)
Preparation of the title compound 58 following the synthesis of example 25, 1- (1-methylindozin-3-yl) ethanone (25d) was replaced with 1- (1-methyl-1H-indazol-3-yl) ethanone (58 a). MS-ESI (M/z):502[ M +1 [)]+
Example 59
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (1-methyl-1H-indazol-3-) Yl) pyrimidin-2-yl) amino) pyridin-3-yl) -2-fluoroacrylamide (59)
2 2N- (2- (dimethylamino) ethyl) -6-methoxy-N-methyl-N5- (4- (1-methyl-1H-indazol-3-yl) pyrimidine Pyridin-2-yl) -3-nitropyridine-2, 5-diamine (59a)
Preparation of compound 59a 1- (1-methyl-indolizin-3-yl) ethanone (25d) was replaced with 1- (1-methyl-1H-indazol-3-yl) ethanone (58a) according to the synthesis of example 25. MS-ESI (M/z):448[ M +1]]+
2 2N- (2- (dimethylamino) ethyl) -6-methoxy-N-methyl-N5- (4- (1-methyl-1H-indazol-3-yl) pyrimidine Pyridin-2-yl) pyridine-2, 3, 5-triamine (59b)
Preparation of the title compound 59b according to the synthesis of 15i, N1- (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N1,N2,N2Replacement of-trimethylethane-1, 2-diamine (15h) by N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-N5- (4- (1-methyl-1H-indazol-3-yl) pyrimidin-2-yl) -3-nitropyridine-2, 5-diamine (59 a). MS-ESI (M/z):447[ M +1]]+
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (1-methyl-1H-indazol-3-) Yl) pyrimidin-2-yl) amino) pyridin-3-yl) -2-fluoroacrylamide (59)
Preparation of the title compound (59) according to the synthesis of example 23, N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-N5Replacement of- (4- (1-methyl-1H-indazol-3-yl) pyrimidin-2-yl) pyridine-2, 3, 5-triamine (23b) by N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-N5- (4- (1-methyl-1H-indazol-3-yl) pyrimidin-2-yl) pyridine-2, 3, 5-triamine (59 b). MS-ESI (M/z):520[ M +1]]+
Example 60
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (3-methylimidazol [1, 5-a)] And pyridin-1-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (60)
1- (3-methylimidazol [1,5-a ]]And pyridin-1-yl) ethan-1-one (60a)
The title compound 60a was prepared according to the literature synthesis: journal of Chemical society, 1955,2834.
N- (2- ((2- (dimethylamino) ethyl) ((meth) acrylic acid)Methyl) amino) -6-methoxy-5- ((4- (3-methylimidazol [1, 5-a)] And pyridin-1-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (60)
Preparation of the title compound 60 according to the procedure for the synthesis of example 25, 1- (1-methylindozin-3-yl) ethanone (25d) was replaced with 1- (3-methylimidazol [1,5-a ]]And-pyridin-1-yl) ethan-1-one (60 a). MS-ESI (M/z):502[ M +1 [)]+
Example 61
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (3-methylimidazol [1, 5-a)] And pyridin-1-yl) pyrimidin-2-yl) amino) pyridin-3-yl) -2-fluoroacrylamide (61)
2 2 5N- (2- (dimethylamino) ethyl) -6-methoxy-N-methyl-N- (4- (3-methylimidazol [1, 5-a)]Pyridine- 1-yl) pyrimidin-2-yl) pyridine-2, 3, 5-triamine (61a)
Preparation of the title compound 61a 1- (1-methylindozin-3-yl) ethanone (25d) was replaced with 1- (3-methylimidazol [1,5-a ] according to the synthesis method of example 25]And-pyridin-1-yl) ethan-1-one (60 a). MS-ESI (M/z):448[ M +1]]+
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (3-methylimidazol [1, 5-a)] And pyridin-1-yl) pyrimidin-2-yl) amino) pyridin-3-yl) -2-fluoroacrylamide (61)
Preparation of the title compound 61 the synthesis of example 23 was followed, adding N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-N5Replacement of- (4- (1-methyl-1H-indazol-3-yl) pyrimidin-2-yl) pyridine-2, 3, 5-triamine (23b) by N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-N5- (4- (3-methylimidazol [1,5-a ]]And-pyridin-1-yl) pyrimidin-2-yl) pyridine-2, 3, 5-triamine. MS-ESI (M/z):520[ M +1]]+
Cell proliferation assay
The inhibition of L858R/T790M EGFR by compounds was tested by determining their inhibition of H1975 cell proliferation. In this experiment, compounds were identified by examining their inhibitory effect on H1975 cell proliferationInhibition of L858R/T790M EGFR. H1975 cells were cultured in RPMI-1640 medium containing 40-80% fetal bovine serum. H1975 cells in logarithmic growth phase are inoculated into 96-well culture plates at a density of 3000/well and at 37 ℃ with 5% CO2Incubate overnight. The compounds were added to 96-well plates at different concentrations (final concentrations 10000, 3333.3, 1111.1, 270.4, 123.5, 41.2, 13.7, 4.6 and 1.5nM) at 37 ℃ in 5% CO2Incubate for 72 hours. The medium was discarded, and 20. mu.l MTS/100. mu.l medium was added to each well. After 1.5h incubation, the reaction was stopped by adding 25. mu.l of 10% SDS per well. The absorbance at 490nm and 650nm was measured with a microplate reader. IC calculation Using GraphPad Prism 5.050
The inhibition of wild-type EGFR by compounds was examined by measuring the inhibition of a431 cell proliferation by compounds. In this experiment, the inhibitory effect of a compound on wild-type EGFR was determined by examining the inhibitory effect of a compound on a431 cell proliferation. A431 cells were cultured in RPMI-1640 medium containing 40-80% fetal bovine serum. H1975 cells in logarithmic growth phase are inoculated into 96-well culture plates at a density of 3000/well and at 37 ℃ with 5% CO2Incubate overnight. The compounds were added to 96-well plates at different concentrations (final concentrations 10000, 3333.3, 1111.1, 270.4, 123.5, 41.2, 13.7, 4.6 and 1.5nM) at 37 ℃ in 5% CO2Incubate for 72 hours. The medium was discarded, and 20. mu.l MTS/100. mu.l medium was added to each well. After 1.5h incubation, the reaction was stopped by adding 25. mu.l of 10% SDS per well. The absorbance at 490nm and 650nm was measured with a microplate reader. IC calculation Using GraphPad Prism 5.050
Selected compounds prepared as described above were tested according to the biological methods described herein. The results are shown in Table 1.
TABLE 1
Examples H1975 IC50(nM) A431 IC50(nM)
1 112 >1000
15 156 /
16 193 >1000
17 128 1213
18 183 /
19 76 /
20 48 /
21 69 /
23 184 /
24 219 /
25 219 >1000
27 324 >1000
32 190 >1000
36 244 1193
38 183 /
41 350 /
50 476 /
51 348 /
53 452 /
54 481 /

Claims (3)

1. A compound selected from:
n- (6-methoxy-2- (methyl (2- (methylamino) ethyl) amino) -5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide,
N- (2- ((2- (ethyl (methyl) amino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide,
N- (6-methoxy-2- (methyl (2- (N-methylacetamido) ethyl) amino) -5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide,
N- (6-ethoxy-2- (methyl (2- (methylamino) ethyl) amino) -5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-ethoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide,
N- (6-isopropoxy-2- (methyl (2- (N-methylacetamido) ethyl) amino) -5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-isopropoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide,
N- (6-methoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- (piperazin-1-yl) pyridin-3-yl) acrylamide,
N- (6-methoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- (4-methylpiperidin-1-yl) pyridin-3-yl) acrylamide,
N- (6-methoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- (piperidin-1-yl) pyridin-3-yl) acrylamide,
N- (6-ethoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- (4-methylpiperidin-1-yl) pyridin-3-yl) acrylamide,
N- (6-isopropoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- (piperidin-1-yl) pyridin-3-yl) acrylamide,
N- (6-isopropoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- (4-methylpiperidin-1-yl) pyridin-3-yl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -5- ((4- (7-fluoro-1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -6-methoxypyridin-3-yl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -5- ((4- (6-fluoro-1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -6-methoxypyridin-3-yl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -5- ((4- (5-fluoro-1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -6-methoxypyridin-3-yl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -5- ((4- (4-fluoro-1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -6-methoxypyridin-3-yl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -5- ((5-fluoro-4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -6-methoxypyridin-3-yl) acrylamide,
N- (5- ((5-chloro-4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxypyridin-3-yl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((5-methyl-4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide,
N- (3- ((2- (dimethylamino) ethyl) (methyl) amino) -5-methoxy-6- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) pyridin-2-yl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (1-methylindazin-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5- ((4- (1-methylindazin-3-yl) pyrimidin-2-yl) amino) phenyl) acrylamide,
N- (5- ((4- (1-cyanoindolizin-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxypyridin-3-yl) acrylamide,
N- (5- ((4- (1-cyanoindolizin-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -5- ((4- (indolizin-3-yl) pyrimidin-2-yl) amino) -6-methoxypyridin-3-yl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -5- ((4- (indolizin-3-yl) pyrimidin-2-yl) amino) -4-methoxyphenyl) acrylamide,
N- (5- ((4- (1-chloroindolizin-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxypyridin-3-yl) acrylamide,
N- (5- ((4- (1-chloroindolizin-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (3-methylindazin-1-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5- ((4- (3-methylindazin-1-yl) pyrimidin-2-yl) amino) phenyl) acrylamide,
N- (5- ((4- (3-cyanoindolizin-1-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxypyridin-3-yl) acrylamide,
N- (5- ((4- (3-cyanoindolizin-1-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -5- ((4- (indolizin-1-yl) pyrimidin-2-yl) amino) -6-methoxypyridin-3-yl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -5- ((4- (indolizin-1-yl) pyrimidin-2-yl) amino) -4-methoxyphenyl) acrylamide,
N- (5- ((4- (3-chloroindolizin-1-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxypyridin-3-yl) acrylamide,
N- (5- ((4- (3-chloroindolizin-1-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (3-methyl-1H-indol-1-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5- ((4- (3-methyl-1H-indol-1-yl) pyrimidin-2-yl) amino) phenyl) acrylamide,
N- (6-methoxy-5- ((4- (3-methyl-1H-indol-1-yl) pyrimidin-2-yl) amino) -2- (4-methylpiperazin-1-yl) pyridin-3-yl) acrylamide,
N- (4-methoxy-5- ((4- (3-methyl-1H-indol-1-yl) pyrimidin-2-yl) amino) -2- (4-methylpiperazin-1-yl) phenyl) acrylamide,
N- (5- ((4- (3-cyano-1H-indol-1-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxypyridin-3-yl) acrylamide,
N- (5- ((4- (3-cyano-1H-indol-1-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (3-methyl-1H-indazol-1-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (1-methyl-1H-indazol-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide,
N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ((4- (3-methylimidazol [1,5-a ] pyrid-in-1-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide,
or a pharmaceutically acceptable salt thereof.
2. A pharmaceutical composition comprising a compound of claim 1 and/or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier.
3. A method of treating cell proliferation disorders comprising administering to a patient in need thereof an effective amount of a compound of claim 1 and/or a pharmaceutically acceptable salt thereof, optionally in combination with another therapeutic agent.
CN202010883627.1A 2014-06-12 2015-06-12 Kinase inhibitors Active CN111892579B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202010883627.1A CN111892579B (en) 2014-06-12 2015-06-12 Kinase inhibitors

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
US201462011109P 2014-06-12 2014-06-12
US62/011109 2014-06-12
US201462090360P 2014-12-11 2014-12-11
US62/090360 2014-12-11
PCT/CN2015/081332 WO2015188777A1 (en) 2014-06-12 2015-06-12 Certain protein kinase inhibitors
CN201580031253.8A CN106660993B (en) 2014-06-12 2015-06-12 Kinase inhibitor
CN202010883627.1A CN111892579B (en) 2014-06-12 2015-06-12 Kinase inhibitors

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
CN201580031253.8A Division CN106660993B (en) 2014-06-12 2015-06-12 Kinase inhibitor

Publications (2)

Publication Number Publication Date
CN111892579A true CN111892579A (en) 2020-11-06
CN111892579B CN111892579B (en) 2023-07-25

Family

ID=54832920

Family Applications (3)

Application Number Title Priority Date Filing Date
CN201580031253.8A Active CN106660993B (en) 2014-06-12 2015-06-12 Kinase inhibitor
CN202010883613.XA Active CN111875585B (en) 2014-06-12 2015-06-12 Kinase inhibitors
CN202010883627.1A Active CN111892579B (en) 2014-06-12 2015-06-12 Kinase inhibitors

Family Applications Before (2)

Application Number Title Priority Date Filing Date
CN201580031253.8A Active CN106660993B (en) 2014-06-12 2015-06-12 Kinase inhibitor
CN202010883613.XA Active CN111875585B (en) 2014-06-12 2015-06-12 Kinase inhibitors

Country Status (2)

Country Link
CN (3) CN106660993B (en)
WO (1) WO2015188777A1 (en)

Families Citing this family (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105315259B (en) 2014-07-29 2018-03-09 上海艾力斯医药科技有限公司 Pyridine amine pyrimidine derivates, its preparation method and application
CN117402143A (en) * 2014-10-11 2024-01-16 上海翰森生物医药科技有限公司 EGFR inhibitor and preparation and application thereof
PL3604294T3 (en) * 2014-10-13 2021-11-02 Yuhan Corporation Compounds and compositions for modulating egfr mutant kinase activities
CN111170998B (en) 2014-11-05 2023-04-11 益方生物科技(上海)股份有限公司 Pyrimidine or pyridine compound, preparation method and medical application thereof
CN113563332A (en) * 2014-12-23 2021-10-29 达纳-法伯癌症研究所公司 Novel pyrimidines as EGFR inhibitors and methods of treating disorders
CN106117185B (en) * 2015-08-31 2017-11-07 广州必贝特医药技术有限公司 2,4 2 nitrogen-containing group substituted uracil compounds and its preparation method and application
US10787428B2 (en) 2015-12-24 2020-09-29 Kyowa Kirin Co., Ltd. α,β-unsaturated amide compound
CN106928150B (en) * 2015-12-31 2020-07-31 恩瑞生物医药科技(上海)有限公司 Acrylamide aniline derivative and pharmaceutical application thereof
CN109328059B (en) * 2016-01-07 2021-08-17 Cs制药技术有限公司 Selective inhibitors of clinically important mutants of EGFR tyrosine kinase
CN106279160B (en) * 2016-03-18 2017-09-26 海南越康生物医药有限公司 The amino-metadiazine compound preparation method of N phenyl 2 and purposes
SG10202102920QA (en) 2016-05-26 2021-05-28 Recurium Ip Holdings Llc Egfr inhibitor compounds
CN106957304B (en) * 2017-04-25 2017-12-01 孔令廷 A kind of graphene-supported FeCl3The preparation method of catalyst and its purposes in preparation Anti-cancer medicament intermediate
CA3068158A1 (en) * 2017-06-23 2018-12-27 Kyowa Kirin Co., Ltd. .alpha.,.beta.-unsaturated amide compound
CN107266437B (en) 2017-07-19 2019-08-30 海南越康生物医药有限公司 The crystal form of N- phenyl -2- amino-metadiazine compound, salt form and preparation method thereof
MD3658552T2 (en) 2017-07-28 2024-02-29 Yuhan Corp Process for preparing N-(5-((4-(4-((dimethylamino)methyl)-3-phenyl-1H-pyrazol-1-yl)pyrimidin-2-yl)amino)-4-methoxy-2-morpholinophenyl)acrylamide by reacting the corresponding amine with a 3-halo-propionyl chloride
CN107827875B (en) * 2017-09-25 2021-07-09 文韬创新药物研究(北京)有限责任公司 Application of benzimidazole derivative as cyclin-dependent kinase 4/6 inhibitor
US11918592B2 (en) 2018-01-16 2024-03-05 Syros Pharmaceuticals, Inc. Inhibitors of cyclin dependent kinase 7 (CDK7)
US11311542B2 (en) 2018-01-16 2022-04-26 Syros Pharmaceuticals, Inc. Inhibitors of cyclin dependent kinase 7 (CDK7)
CN110857292A (en) * 2018-08-22 2020-03-03 上海艾力斯医药科技有限公司 EGFR kinase inhibitor and preparation method and application thereof
CR20210287A (en) 2018-11-01 2022-02-15 Syros Pharmaceuticals Inc Inhibitors of cyclin-dependent kinase 7 (cdk7)
SG11202108311RA (en) 2019-02-26 2021-09-29 Janssen Biotech Inc Combination therapies and patient stratification with bispecific anti-egfr/c-met antibodies
CN110041302B (en) * 2019-03-01 2021-11-30 南方医科大学 2-amino-4-substituted pyridine derivative and synthesis method and application thereof
CN111747950B (en) * 2019-03-29 2024-01-23 深圳福沃药业有限公司 Pyrimidine derivatives for the treatment of cancer
US11879013B2 (en) 2019-05-14 2024-01-23 Janssen Biotech, Inc. Combination therapies with bispecific anti-EGFR/c-Met antibodies and third generation EGFR tyrosine kinase inhibitors
JP2023052711A (en) * 2020-03-04 2023-04-12 国立研究開発法人理化学研究所 Method for releasing compound
CN111303123B (en) * 2020-03-31 2021-08-31 南京雷正医药科技有限公司 2- (2,4, 5-substituted anilino) pyrimidine compound and application thereof
CA3224994A1 (en) * 2021-06-22 2022-12-29 Dana-Farber Cancer Institute, Inc. (1h-pyrrolo[2,3-b]pyridin-1-yl)pyrimidin-2-yl-amino-phenyl--acrylamide inhibitors of egfr for use in the treatment of brain tumors
CN115701429B (en) * 2021-08-02 2024-03-12 上海和誉生物医药科技有限公司 4- (1H-indol-1-yl) pyrimidine-2-amino derivative, and preparation method and application thereof

Citations (3)

* 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
CN103702990A (en) * 2011-07-27 2014-04-02 阿斯利康(瑞典)有限公司 2-(2,4,5-substituted -anilino) pyrimidine derivatives as egfr modulators useful for treating cancer
CN105085489A (en) * 2014-11-05 2015-11-25 上海页岩科技有限公司 Pyrimidine or pyridine compound, and preparation method and pharmaceutical application thereof

Patent Citations (3)

* 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
CN103702990A (en) * 2011-07-27 2014-04-02 阿斯利康(瑞典)有限公司 2-(2,4,5-substituted -anilino) pyrimidine derivatives as egfr modulators useful for treating cancer
CN105085489A (en) * 2014-11-05 2015-11-25 上海页岩科技有限公司 Pyrimidine or pyridine compound, and preparation method and pharmaceutical application thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
ASTRA ZENECA等: "Structure- and Reactivity-Based Development of Covalent Inhibitors of the Activating and Gatekeeper Mutant Forms of the Epidermal Growth Factor Receptor (EGFR)", 《J. MED. CHEM.》 *

Also Published As

Publication number Publication date
CN111892579B (en) 2023-07-25
CN111875585B (en) 2023-06-23
CN106660993B (en) 2020-09-11
WO2015188777A1 (en) 2015-12-17
CN106660993A (en) 2017-05-10
CN111875585A (en) 2020-11-03

Similar Documents

Publication Publication Date Title
CN106660993B (en) Kinase inhibitor
JP7058636B2 (en) Inhibitor of cyclin-dependent kinase 7 (CDK7)
US9757364B2 (en) Naphthyridine derivative compounds
CN110372720B (en) Kinase inhibitor
KR102072869B1 (en) Quinolines as fgfr kinase modulators
KR102341202B1 (en) Quinazolinone derivatives useful as fgfr kinase modulators
EP2124951B1 (en) 5-cyan0-4- (pyrrolo[2, 3b]pyridine-3-yl) -pyrimidine derivatives useful as protein kinase inhibitors
US10022374B2 (en) Certain protein kinase inhibitors
KR20180134983A (en) Heterocyclic compounds as RET kinase inhibitors
JP6599983B2 (en) Carbazole derivatives
EP1658289B1 (en) 3-substituted imidazopyridine-derivatives as c-kit inhibitors
CN110294761B (en) Substituted pyrazolo [1,5-a ] pyrimidine compounds as Trk kinase inhibitors
AU2013344049A1 (en) ALK kinase inhibitors
WO2016011979A1 (en) 2,4-disubstituted 7h-pyrrolo[2,3-d]pyrimidine derivative, preparation method and medicinal use thereof
EP3807266B1 (en) Pyrazole derivatives as malt1 inhibitors
KR20200115583A (en) 2H-indazole derivatives as CDK4 and CDK6 inhibitors and their therapeutic use
CN113166110B (en) 2-aminopyrimidine compound and application thereof
CN107207504B (en) Phthalazinone derivatives, preparation method and use thereof
CN113677680A (en) EGFR inhibitor and composition and application thereof
JP2018502141A (en) Quinazoline and quinoline compounds and uses thereof
CN107207441B (en) Protein kinase inhibitors
CN113661164A (en) CDK kinase inhibitor and application thereof
CN111164084A (en) Triazolotriazine derivatives useful as A2A receptor antagonists
WO2021057867A1 (en) Class of cdk inhibitor based on organic arsine, preparation method and application thereof
CN111875583A (en) Triazole derivative and preparation method and application thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
TA01 Transfer of patent application right
TA01 Transfer of patent application right

Effective date of registration: 20210713

Address after: 201210 5th floor, building 1, 1118 Halley Road, 1227 zhangheng Road, Pudong New Area pilot Free Trade Zone, Shanghai

Applicant after: Shanghai ALLIST medicine Polytron Technologies Inc.

Address before: Room 512, A Building, 1289 Yishan Road, Xuhui District, Shanghai, 2003

Applicant before: CHINA SHANGHAI FOCHON PHARMACEUTICAL Co.,Ltd.

Applicant before: FOCHON PHARMACEUTICALS, Ltd.

CB02 Change of applicant information
CB02 Change of applicant information

Address after: No. 268 Lingxiaohua Road, Zhoupu Town, Pudong New Area, Shanghai, 201318

Applicant after: Shanghai ALLIST medicine Polytron Technologies Inc.

Address before: 201210 5th floor, building 1, 1118 Halley Road, 1227 zhangheng Road, Pudong New Area pilot Free Trade Zone, Shanghai

Applicant before: Shanghai ALLIST medicine Polytron Technologies Inc.

GR01 Patent grant
GR01 Patent grant