WO2015188777A1 - Certain protein kinase inhibitors - Google Patents

Certain protein kinase inhibitors Download PDF

Info

Publication number
WO2015188777A1
WO2015188777A1 PCT/CN2015/081332 CN2015081332W WO2015188777A1 WO 2015188777 A1 WO2015188777 A1 WO 2015188777A1 CN 2015081332 W CN2015081332 W CN 2015081332W WO 2015188777 A1 WO2015188777 A1 WO 2015188777A1
Authority
WO
WIPO (PCT)
Prior art keywords
amino
methyl
ethyl
pyrimidin
dimethylamino
Prior art date
Application number
PCT/CN2015/081332
Other languages
French (fr)
Inventor
Tongshuang Li
Xingdong ZHAO
Qiang Tian
Weipeng Zhang
Hongbin Liu
Xianlong WANG
Haohan TAN
Rui Tan
Qihong Liu
Lihua Jiang
Yanxin Liu
Li LINGHU
Min Lin
Jing Sun
Weibo Wang
Original Assignee
Shanghai Fochon Pharmaceutical Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=54832920&utm_source=***_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2015188777(A1) "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 filed Critical Shanghai Fochon Pharmaceutical Co Ltd
Priority to CN201580031253.8A priority Critical patent/CN106660993B/en
Priority to CN202010883613.XA priority patent/CN111875585B/en
Priority to CN202010883627.1A priority patent/CN111892579B/en
Publication of WO2015188777A1 publication Critical patent/WO2015188777A1/en

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

Definitions

  • This invention relates to novel amino pyrimidine ring derivatives and the pharmaceutically acceptable salts thereof.
  • the compounds of the present invention are mutant-selective epidermal growth factor receptor (EGFR) kinase inhibitors.
  • EGFR epidermal growth factor receptor
  • the compounds of this invention are useful for the treatment of hyper-proliferative diseases such as cancer, in mammals and especially in humans.
  • This invention also relates to the use of the compounds in the treatment of cancer and the pharmaceutical preparations containing them.
  • Protein tyrosine kinases catalyze the transfer of a phosphate group from ATP or GTP to a tyrosine residue located on a protein substrate.
  • Protein tyrosine kinase can be broadly classified as receptor (e.g. EGFR, HER-2, VEGFR, FGFR) or non-receptor (e.g. Src, Jak, Abl) .
  • Receptor tyrosine kinases act to transmit signals from the outside of a cell to the inside by activating secondary messaging effectors via a phosphorylation event. A variety of cellular processes are promoted by these signals, including proliferation, carbohydrate utilization, protein synthesis, angiogenesis, cell growth, and cell survival.
  • the first-generation reversible, ATP-competitive EGFR kinase inhibitors are effective clinical therapies for non-small cell lung cancers that harbor activating mutations in the EGFR kinase domain.
  • first-generation EGFR inhibitors showed encouraging clinical responses, almost all patients developed resistance to these inhibitors over time, such as by mutation of gatekeeper residue T790M, which accounts for about half of all resistance to gefitinib and erlotinib (Proc. Natl. Acad. Sci. U.S.A. 2008, 105, 2070-2075) .
  • T790M may also be pre-existing; there may be an independent, oncogenic role for the T790M mutation.
  • Second-generation covalent inhibitors such as afatinib, neratinib, canertinib and dacomitinib
  • second-generation covalent inhibitors such as afatinib, neratinib, canertinib and dacomitinib
  • mutant-selective EGFR kinase inhibitors useful as therapeutic agents.
  • mutant-selective EGFR inhibitors were disclosed in the arts, e.g., WO 2013014448 and WO 2012061299, there is still a need for new mutant-selective EGFR inhibitors that have at least one advantageous property selected from potency, stability, selectivity, toxicity and pharmacodynamics properties as an alternative for the treatment of hyper-proliferative diseases.
  • a novel class of mutant-selective EGFR inhibitors is provided herein.
  • Q is selected from aryl and heteroaryl
  • X is selected from N and C;
  • Y is selected from N and C;
  • R 1 is selected from hydrogen, C 1-10 alkyl, C 3-10 cycloalkyl, C 3-10 cycloalkyl-C 1-4 alkyl, heterocyclyl, and heterocyclyl-C 1-4 alkyl, wherein alkyl, cycloalkyl, and heterocyclyl are each unsubstituted or substituted with at least one substituent, such as one, two, three, or four substituents, independently selected from R 6a ;
  • R 1’ is selected from hydrogen and halogen
  • R 2 and R 3 are independently selected from hydrogen, halogen, hydroxyl, CN, C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl, C 3-10 cycloalkyl, C 3-10 cycloalkyl-C 1-4 alkyl, heterocyclyl, heterocyclyl-C 1-4 alkyl, aryl, aryl-C 1-4 alkyl, heteroaryl, and heteroaryl-C 1-4 alkyl, wherein alkyl, alkenyl, alkynyl, cycloalkyl, and heterocyclyl are each unsubstituted or substituted with at least one substituent, such as one, two, three, or four substituents independently selected from R 6a , and each aryl and heteroaryl is unsubstituted or substituted with at least one substituent, such as one, two, three, or four substituents, independently selected from R 6b ; or R 2 and R 3 together with the carbon atoms to which they are
  • each R 4 is independently selected from hydrogen, C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl, C 3-10 cycloalkyl, -OR 8 , -NR 7 S (O) r R 8 , -NO 2 , -halogen, -S (O) r R 7 , -SR 8 , -S (O) 2 OR 7 , -OS (O) 2 R 8 , -S (O) r NR 7 R 8 , -NR 7 R 8 , -O (CR 9 R 10 ) t NR 7 R 8 , -C (O) R 7 , -CO 2 R 8 , -CO 2 (CR 9 R 10 ) t CONR 7 R 8 , -OC (O) R 7 , -CN, -C (O) NR 7 R 8 , -NR 7 C (O) R 8 , -OC (O) NR 7 R 8 , -NR 7 C (O)
  • each R 5 is independently selected from hydrogen, C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl, C 3-10 cycloalkyl, C 3-10 cycloalkyl-C 1-4 alkyl, heterocyclyl, heterocyclyl-C 1-4 alkyl, aryl, aryl-C 1-4 alkyl, heteroaryl, and heteroaryl-C 1-4 alkyl, -OR 8 , -NR 7 S (O) r R 8 , -NO 2 , halogen, -S (O) r R 7 , -SR 8 , -S (O) 2 OR 7 , -OS (O) 2 R 8 , -S (O) r NR 7 R 8 , -NR 7 R 8 , -O (CR 9 R 10 ) t NR 7 R 8 , -C (O) R 7 , -CO 2 R 8 , -CO 2 (CR 9 R 10 ) t CONR 7
  • each R 6a is independently selected from C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl, C 3-10 cycloalkyl, -OR 8 , -NR 7 S (O) r R 8 , -NO 2 , halogen, -S (O) r R 7 , -SR 8 , -S (O) 2 OR 7 , -OS (O) 2 R 8 , -S (O) r NR 7 R 8 , -NR 7 R 8 , - (CR 9 R 10 ) t OR 8 , - (CR 9 R 10 ) t NR 7 R 8 , - (CR 9 R 10 ) t SR 8 , - (CR 9 R 10 ) t S (O) r R 8 , - (CR 9 R 10 ) t CO 2 R 8 , - (CR 9 R 10 ) t CONR 7 R 8 , - (CR 9 R 10 ) t NR 7
  • each R 6b is independently selected from R 6a , aryl, aryl-C 1-4 alkyl, heteroaryl, and heteroaryl-C 1-4 alkyl;
  • each R 7 and each R 8 are independently selected from hydrogen, C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl, cycloalkyl, cycloalkyl-C 1-4 alkyl, heterocyclyl, heterocyclyl-C 1-4 alkyl, aryl, aryl-C 1-4 alkyl, heteroaryl, and heteroaryl-C 1-4 alkyl, wherein alkyl, alkenyl, alkynyl, cycloalkyl, and heterocyclyl are each unsubstituted or substituted with at least one substituent, such as one, two, three, or four substituents independently selected from R 6a , and aryl and heteroaryl are each unsubstituted or substituted with at least one substituent, such as one, two, three, or four substituents independently selected from R 6b ; or R 7 and R 8 together with the atom (s) to which they are attached form a heterocyclic ring of 4 to 12 members
  • each R 9 and each R 10 are independently selected from hydrogen, C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl, cycloalkyl, cycloalkyl-C 1-4 alkyl, heterocyclyl, heterocyclyl-C 1-4 alkyl, aryl, aryl-C 1-4 alkyl, heteroaryl, and heteroaryl-C 1-4 alkyl; or R 9 and R 10 together with the carbon atom (s) to which they are attached form a ring of 3 to 7 members containing 0, 1, or 2 heteroatoms independently selected from oxygen, sulfur and nitrogen, and optionally substituted with 1or 2 R 6a groups;
  • n is independently selected from 0, 1, 2, and 3;
  • n is independently selected from 0, 1, 2, and 3;
  • each r is independently selected from 1 and 2;
  • each t is independently selected from 1, 2, and 3.
  • compositions comprising at least one compound of formula (I) and/or at least one pharmaceutically acceptable salt thereof and at least one pharmaceutically acceptable excipient.
  • a therapeutically effective amount of at least one compound of formula (I) and/or at least one pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof comprising administering to a system or a subject in need thereof, a therapeutically effective amount of at least one compound of formula (I) and/or at least one pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof, thereby modulating said mutant EGFR.
  • a condition which responds to inhibition of mutant EGFR comprising administering to a system or subject in need of such treatment an effective amount of at least one compound of formula (I) and/or at least one pharmaceutically acceptable salt thereof or pharmaceutical compositions thereof, and optionally in combination with a second therapeutic agent, thereby treating said condition.
  • a condition mediated by mutant EGFR a condition mediated by mutant EGFR.
  • the compounds of the disclosure may be used alone or in combination with a second therapeutic agent to treat a condition mediated by mutant EGFR, wherein said condition is an autoimmune disease, a transplantation disease, an infectious disease or a cell proliferative disorder.
  • a cell proliferative disorder comprising administering to a system or subject in need of such treatment an effective amount of at least one compound of formula (I) and/or at least one pharmaceutically acceptable salt thereof or pharmaceutical compositions thereof, and optionally in combination with a second therapeutic agent, thereby treating said condition.
  • the compounds of the disclosure may be used alone or in combination with a chemotherapeutic agent to treat a cell proliferative disorder, including but not limited to, lymphoma, osteosarcoma, melanoma, or a tumor of breast, renal, prostate, colorectal, thyroid, ovarian, pancreatic, neuronal, lung, uterine or gastrointestinal tumor.
  • a chemotherapeutic agent to treat a cell proliferative disorder, including but not limited to, lymphoma, osteosarcoma, melanoma, or a tumor of breast, renal, prostate, colorectal, thyroid, ovarian, pancreatic, neuronal, lung, uterine or gastrointestinal tumor.
  • At least one compound of formula (I) and/or at least one pharmaceutically acceptable salt thereof may be administered to a system comprising cells or tissues, or to a mammalian subject such as a human or animal subject.
  • alkyl refers to both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms. Unless otherwise specified, “alkyl” refers to C l -C 10 alkyl. For example, C 1-6 , as in “C l-6 alkyl” is defined to include groups having 1, 2, 3, 4, 5, or 6 carbons in a linear or branched arrangement. For example, “C l-8 alkyl” includes but is not limited to methyl, ethyl, n-propyl, i-propyl, n-butyl, t-butyl, i-butyl, pentyl, hexyl, heptyl, and octyl.
  • cycloalkyl means a saturated aliphatic cyclic hydrocarbon group having the specified number of carbon atoms. Unless otherwise specified, “cycloalkyl” refers to C 3-l0 cycloalkyl. For example, “cycloalkyl” includes but is not limited to cyclopropyl, methyl-cyclopropyl, 2, 2-dimethyl-cyclobutyl, 2-ethyl-cyclopentyl, and cyclohexyl.
  • alkenyl refers to a non-aromatic hydrocarbon radical, straight, branched or cyclic, containing from 2 to 10 carbon atoms and at least one carbon to carbon double bond. In some embodiments, one carbon to carbon double bond is present, and up to four non-aromatic carbon-carbon double bonds may be present.
  • C 2-6 alkenyl means an alkenyl radical having from 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 double bonds and may be substituted if a substituted alkenyl group is indicated.
  • alkynyl refers to a hydrocarbon radical straight, branched or cyclic, containing from 2 to 10 carbon atoms and at least one carbon to carbon triple bond. In some embodiments, up to three carbon-carbon triple bonds may be present.
  • C 2-6 alkynyl means an alkynyl radical having from 2 to 6 carbon atoms.
  • Alkynyl groups include but are not limited to ethynyl, propynyl, butynyl, and 3-methylbutynyl.
  • the straight, branched or cyclic portion of the alkynyl group may contain triple bonds and may be substituted if a substituted alkynyl group is indicated.
  • aryl encompasses 5-and 6-membered carbocyclic aromatic rings, for example, benzene; bicyclic ring systems wherein at least one ring is carbocyclic and aromatic, for example, naphthalene, indane, and 1, 2, 3, 4-tetrahydroquinoline; and tricyclic ring systems wherein at least one ring is carbocyclic and aromatic, for example, fluorene.
  • the aryl substituent is bicyclic or tricyclic and at least one ring is non-aromatic, it is understood that attachment is via the aromatic ring.
  • aryl includes 5-and 6-membered carbocyclic aromatic rings fused to a 5-to 7-membered heterocyclic ring containing one or more heteroatoms selected from N, O, and S, provided that the point of attachment is at the carbocyclic aromatic ring.
  • Bivalent radicals formed from substituted benzene derivatives and having the free valences at ring atoms are named as substituted phenylene radicals.
  • Bivalent radicals derived from univalent polycyclic hydrocarbon radicals whose names end in "-yl” by removal of one hydrogen atom from the carbon atom with the free valence are named by adding "-idene” to the name of the corresponding univalent radical, e.g., a naphthyl group with two points of attachment is termed naphthylidene.
  • Aryl does not encompass or overlap in any way with heteroaryl, separately defined below. Hence, if one or more carbocyclic aromatic rings are fused with a heterocyclic aromatic ring, the resulting ring system is heteroaryl, not aryl, as defined herein.
  • halogen refers to fluorine, chlorine, bromine and iodine.
  • heteroaryl refers to aryl
  • 8-to 12-membered bicyclic rings containing one or more, for example, from 1 to 4, or, in some embodiments, from 1 to 3, heteroatoms selected from N, O, and S, with the remaining ring atoms being carbon and wherein at least one heteroatom is present in an aromatic ring; and
  • 11-to 14-membered tricyclic rings containing one or more, for example, from 1 to 4, or in some embodiments, from 1 to 3, heteroatoms selected from N, O, and S, with the remaining ring atoms being carbon and wherein at least one heteroatom is present in an aromatic ring.
  • the total number of S and O atoms in the heteroaryl group exceeds 1, those heteroatoms are not adjacent to one another. In some embodiments, the total number of S and O atoms in the heteroaryl group is not more than 2. In some embodiments, the total number of S and O atoms in the aromatic heterocycle is not more than 1.
  • heteroaryl groups include, but are not limited to, (as numbered from the linkage position assigned priority 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, thiadiazolyl, tetrazolyl, thienyl, benzothienyl, furyl, benzofuryl, benzoimidazolinyl, indolinyl, pyridizinyl, triazolyl, quinolinyl, pyrazolyl, and 5, 6, 7, 8-tetrahydroisoquinoline.
  • heteroaryl groups include but are not limited to pyrrolyl, isothiazolyl, triazinyl, pyrazinyl, pyridazinyl, indolyl, benzotriazolyl, quinoxalinyl, and isoquinolinyl.
  • heteroaryl is also understood to include the N-oxide derivative of any nitrogen-containing heteroaryl.
  • Bivalent radicals derived from univalent heteroaryl radicals whose names end in "-yl” by removal of one hydrogen atom from the atom with the free valence are named by adding "-idene" to the name of the corresponding univalent radical, e.g., a pyridyl group with two points of attachment is a pyridylidene.
  • Heteroaryl does not encompass or overlap with aryl as defined above.
  • heteroaryl substituent is bicyclic or tricyclic and at least one ring is non-aromatic or contains no heteroatoms, it is understood that attachment is via the aromatic ring or via the heteroatom containing ring, respectively.
  • heterocycle broadly refers to a single aliphatic ring, usually with 3 to 12 ring atoms, containing at least 2 carbon atoms in addition to one or more, preferably one to three heteroatoms independently selected from oxygen, sulfur, and nitrogen, as well as combinations comprising at least one of the foregoing heteroatoms.
  • a heterocycle as defined above may be multicyclic ring system (e.g. bicyclic) in which two or more rings may be fused or bridged together, wherein at least one such ring contains one or more heteroatoms independently selected from oxygen, sulfur, and nitrogen.
  • Heterocycle also refers to 5-to 7-membered heterocyclic ring containing one or more heteroatoms selected from N, O, and S fused with 5-and 6-membered carbocyclic aromatic ring, provided that the point of attachment is at the heterocyclic ring.
  • the rings may be saturated or have one or more double bonds (i.e. partially unsaturated) .
  • the heterocycle can be substituted by oxo.
  • the point of the attachment may be carbon or heteroatom in the heterocyclic ring, provided that attachment results in the creation of a stable structure.
  • the heterocyclic ring has substituents, it is understood that the substituents may be attached to any atom in the ring, whether a heteroatom or a carbon atom, provided that a stable chemical structure results.
  • Heterocycle does not overlap with heteroaryl.
  • Suitable heterocycles include, for example (as numbered from the linkage position assigned priority 1) , 1-pyrrolidinyl, 2-pyrrolidinyl, 2, 4-imidazolidinyl, 2, 3-pyrazolidinyl, 1-piperidinyl, 2-piperidinyl, 3-piperidinyl, 4-piperidinyl, and 2, 5-piperazinyl.
  • Morpholinyl groups are also contemplated, including 2-morpholinyl and 3-morpholinyl (numbered wherein the oxygen is assigned priority 1) .
  • Substituted heterocycle also includes ring systems substituted with one or more oxo moieties, such as piperidinyl N-oxide, morpholinyl-N-oxide, 1-oxo-1-thiomorpholinyl and 1, 1-dioxo-1-thiomorpholinyl.
  • Bicyclic heterocycles include, for example:
  • arylalkyl refers to an alkyl moiety substituted by an aryl group.
  • Example arylalkyl groups include benzyl, phenethyl, and naphthylmethyl groups. In some embodiments, arylalkyl groups have from 7 to 20 or 7 to 11 carbon atoms.
  • arylC l-4 alkyl the term “C 1-4 ” refers to the alkyl portion of the moiety and does not describe the number of atoms in the aryl portion of the moiety.
  • arylC 1-10 alkyl refers to the alkyl portion of the moiety and does not describe the number of atoms in the aryl portion of the moiety.
  • heterocyclylalkyl refers to alkyl substituted by heterocyclyl.
  • C 1-6 alkyl refers to the alkyl portion of the moiety and does not describe the number of atoms in the heterocyclyl portion of the moiety.
  • cycloalkylalkyl refers to alkyl substituted by cycloalkyl.
  • C 3-10 cycloalkylalkyl refers to the cycloalkyl portion of the moiety and does not describe the number of atoms in the alkyl portion of the moiety.
  • C 3-7 cycloalkylalkyl refers to the cycloalkyl portion of the moiety and does not describe the number of atoms in the alkyl portion of the moiety.
  • C 3-8 cycloalkylalkyl refers to the cycloalkyl portion of the moiety and does not describe the number of atoms in the alkyl portion of the moiety.
  • C 1-10 alkyl refers to the alkyl portion of the moiety and does not describe the number of atoms in the cycloalkyl portion of the moiety.
  • heteroarylalkyl refers to alkyl substituted by heteroaryl.
  • C 1-4 refers to the alkyl portion of the moiety and does not describe the number of atoms in the heteroaryl portion of the moiety.
  • C 1-10 refers to the alkyl portion of the moiety and does not describe the number of atoms in the heteroaryl portion of the moiety.
  • substitution of alkyl, cycloalkyl, heterocyclyl, aryl, and/or heteroaryl refers to substitution of each of those groups individually as well as to substitutions of combinations of those groups. That is, if R 1 is arylalkyl, the aryl portion may be unsubstituted or substituted with at least one substituent, such as one, two, three, or four substituents, independently selected from R 6b and the alkyl portion may also be unsubstituted or substituted with at least one substituent, such as one, two, three, or four substituens, independently selected from R 6a .
  • salts derived from inorganic bases may be selected, for example, from aluminum, ammonium, calcium, copper, ferric, ferrous, lithium, magnesium, manganic, manganous, potassium, sodium, and zinc salts. Further, for example, the pharmaceutically acceptable salts derived from inorganic bases may be selected from ammonium, calcium, magnesium, potassium, and sodium salts. Salts in the solid form may exist in one or more crystal structures, and may also be in the form of hydrates.
  • Salts derived from pharmaceutically acceptable organic non-toxic bases may be selected, for example, from salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, and basic ion exchange resins, such as arginine, betaine, caffeine, choline, N, N'-dibenzylethylene-diamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethyl-morpholine, N-ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylamine, lysine, methylglucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine, and tripropylamine, tromethamine.
  • salts may be prepared using at least one pharmaceutically acceptable non-toxic acid, selected from inorganic and organic acids.
  • acid may be selected, for example, from acetic, benzenesulfonic, benzoic, camphorsulfonic, citric, ethanesulfonic, fumaric, gluconic, glutamic, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, nitric, pamoic, pantothenic, phosphoric, succinic, sulfuric, tartaric, and p-toluenesulfonic acids.
  • such acid may be selected, for example, from citric, hydrobromic, hydrochloric, maleic, phosphoric, sulfuric, fumaric, and tartaric acids.
  • protecting group refers to a substituent that can be commonly employed to block or protect a certain functionality while reacting other functional groups on the compound.
  • an "amino-protecting group” is a substituent attached to an amino group that blocks or protects the amino functionality in the compound. Suitable amino-protecting groups include but are not limited to acetyl, trifluoroacetyl, t-butoxycarbonyl (BOC) , benzyloxycarbonyl (CBZ) and 9-fluorenylmethylenoxycarbonyl (Fmoc) .
  • a "hydroxy-protecting group” refers to a substituent of a hydroxy group that blocks or protects the hydroxy functionality.
  • Suitable protecting groups include but are not limited to acetyl and silyl.
  • a "carboxy-protecting group” refers to a substituent of the carboxy group that blocks or protects the carboxy functionality. Common carboxy-protecting groups include --CH 2 CH 2 SO 2 Ph, cyanoethyl, 2- (trimethylsilyl) ethyl, 2- (trimethylsilyl) ethoxymethyl, 2- (p-toluenesulfonyl) ethyl, 2- (p-nitrophenylsulfenyl) ethyl, 2- (diphenylphosphino) -ethyl, nitroethyl and the like.
  • protecting groups and their use see T. W. Greene, Protective Groups in Organic Synthesis, John Wiley &Sons, New York, 1991.
  • administering at least one compound and/or at least one pharmaceutically acceptable salt should be understood to mean providing at least one compound and/or at least one pharmaceutically acceptable salt thereof to the individual in recognized need of treatment.
  • the term "effective amount” means the amount of the at least one compound and/or at least one pharmaceutically acceptable salt that will elicit the biological or medical response of a tissue, system, animal or human that is being sought by the researcher, veterinarian, medical doctor or other clinician.
  • composition as used herein is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts.
  • composition in relation to a pharmaceutical composition is intended to encompass a product comprising the active ingredient (s) , and the inert ingredient (s) that make up the carrier, as well as any product which results, directly or indirectly, from combination, complexation or aggregation of any two or more of the ingredients, or from dissociation of one or more of the ingredients, or from other types of reactions or interactions of one or more of the ingredients.
  • pharmaceutically acceptable it is meant compatible with the other ingredients of the formulation and not unacceptably deleterious to the recipient thereof.
  • Q is selected from aryl and heteroaryl
  • X is selected from N and C;
  • Y is selected from N and C;
  • R 1 is selected from hydrogen, C 1-10 alkyl, C 3-10 cycloalkyl, C 3-10 cycloalkyl-C 1-4 alkyl, heterocyclyl, and heterocyclyl-C 1-4 alkyl, wherein alkyl, cycloalkyl, and heterocyclyl are each unsubstituted or substituted with at least one substituent, such as one, two, three, or four substituents, independently selected from R 6a ;
  • R 1’ is selected from hydrogen and halogen
  • R 2 and R 3 are independently selected from hydrogen, halogen, hydroxyl, CN, C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl, C 3-10 cycloalkyl, C 3-10 cycloalkyl-C 1-4 alkyl, heterocyclyl, heterocyclyl-C 1-4 alkyl, aryl, aryl-C 1-4 alkyl, heteroaryl, and heteroaryl-C 1-4 alkyl, wherein alkyl, alkenyl, alkynyl, cycloalkyl, and heterocyclyl are each unsubstituted or substituted with at least one substituent, such as one, two, three, or four substituents independently selected from R 6a , and each aryl and heteroaryl is unsubstituted or substituted with at least one substituent, such as one, two, three, or four substituents, independently selected from R 6b ; or R 2 and R 3 together with the carbon atoms to which they are
  • each R 4 is independently selected from hydrogen, C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl, C 3-10 cycloalkyl, -OR 8 , -NR 7 S (O) r R 8 , -NO 2 , -halogen, -S (O) r R 7 , -SR 8 , -S (O) 2 OR 7 , -OS (O) 2 R 8 , -S (O) r NR 7 R 8 , -NR 7 R 8 , -O (CR 9 R 10 ) t NR 7 R 8 , -C (O) R 7 , -CO 2 R 8 , -CO 2 (CR 9 R 10 ) t CONR 7 R 8 , -OC (O) R 7 , -CN, -C (O) NR 7 R 8 , -NR 7 C (O) R 8 , -OC (O) NR 7 R 8 , -NR 7 C (O)
  • each R 5 is independently selected from hydrogen, C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl, C 3-10 cycloalkyl, C 3-10 cycloalkyl-C 1-4 alkyl, heterocyclyl, heterocyclyl-C 1-4 alkyl, aryl, aryl-C 1-4 alkyl, heteroaryl, and heteroaryl-C 1-4 alkyl, -OR 8 , -NR 7 S (O) r R 8 , -NO 2 , halogen, -S (O) r R 7 , -SR 8 , -S (O) 2 OR 7 , -OS (O) 2 R 8 , -S (O) r NR 7 R 8 , -NR 7 R 8 , -O (CR 9 R 10 ) t NR 7 R 8 , -C (O) R 7 , -CO 2 R 8 , -CO 2 (CR 9 R 10 ) t CONR 7
  • each R 6a is independently selected from C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl, C 3-10 cycloalkyl, -OR 8 , -NR 7 S (O) r R 8 , -NO 2 , halogen, -S (O) r R 7 , -SR 8 , -S (O) 2 OR 7 , -OS (O) 2 R 8 , -S (O) r NR 7 R 8 , -NR 7 R 8 , - (CR 9 R 10 ) t OR 8 , - (CR 9 R 10 ) t NR 7 R 8 , - (CR 9 R 10 ) t SR 8 , - (CR 9 R 10 ) t S (O) r R 8 , - (CR 9 R 10 ) t CO 2 R 8 , - (CR 9 R 10 ) t CONR 7 R 8 , - (CR 9 R 10 ) t NR 7
  • each R 6b is independently selected from R 6a , aryl, aryl-C 1-4 alkyl, heteroaryl, and heteroaryl-C 1-4 alkyl;
  • each R 7 and each R 8 are independently selected from hydrogen, C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl, cycloalkyl, cycloalkyl-C 1-4 alkyl, heterocyclyl, heterocyclyl-C 1-4 alkyl, aryl, aryl-C 1-4 alkyl, heteroaryl, and heteroaryl-C 1-4 alkyl, wherein alkyl, alkenyl, alkynyl, cycloalkyl, and heterocyclyl are each unsubstituted or substituted with at least one substituent, such as one, two, three, or four substituents independently selected from R 6a , and aryl and heteroaryl are each unsubstituted or substituted with at least one substituent, such as one, two, three, or four substituents independently selected from R 6b ; or R 7 and R 8 together with the atom (s) to which they are attached form a heterocyclic ring of 4 to 12 members
  • each R 9 and each R 10 are independently selected from hydrogen, C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl, cycloalkyl, cycloalkyl-C 1-4 alkyl, heterocyclyl, heterocyclyl-C 1-4 alkyl, aryl, aryl-C 1-4 alkyl, heteroaryl, and heteroaryl-C 1-4 alkyl; or R 9 and R 10 together with the carbon atom (s) to which they are attached form a ring of 3 to 7 members containing 0, 1, or 2 heteroatoms independently selected from oxygen, sulfur and nitrogen, and optionally substituted with 1or 2 R 6a groups;
  • n is independently selected from 0, 1, 2, and 3;
  • n is independently selected from 0, 1, 2, and 3;
  • each r is independently selected from 1 and 2;
  • each t is independently selected from 1, 2, and 3.
  • At least one compound of 1 and/or at least one pharmaceutically acceptable salt thereof wherein X is selected from N and C, and Y is C.
  • At least one compound of any one of 1-2 and/or at least one pharmaceutically acceptable salt thereof wherein: when both X and Y are C, and
  • one R 5 is (3R) -3- (dimethylamino) pyrrolidin-1-yl, (3S) -3- (dimethylamino) pyrrolidin-1-yl, 3- (dimethylamino) azetidin-1-yl, [2- (dimethylamino) ethyl] - (methyl) amino, [2- (methylamino) ethyl] (methyl) amino, 5-methyl-2, 5-diazaspiro [3.4] oct-2-yl, (3aR, 6aR) -5-methylhexa-hydro-pyrrolo [3, 4-b] pyrrol-1 (2H) -yl, 1-methyl-1, 2, 3, 6-tetrahydropyridin-4-yl, 4-methylpiperizin-1-yl, 4- [2- (dimethylamino) -2-oxoethyl] piperazin-1-yl, methyl [2- (4-methylpiperazin-1-yl)
  • R 1 , R 1’ a nd R 2 are hydrogen, and R 3 is H, F, Cl, CH 3 or CN,
  • Q is not 4, 5, 6, 7-tetrahydropyrazolo [1, 5-a] pyridine-3-yl, pyrazolo [1, 5-a] pyridin-3-yl or 1H-indol-3-yl.
  • At least one compound selected from
  • a pharmaceutical composition comprising at least one compound of any one of 1-18 and pharmaceutically acceptable salts thereof, wherein the composition is adapted for administration by a route selected from the group consisting of orally, parenterally, intraperitoneally, intravenously, intraarterially, transdermally, sublingually, intramuscularly, rectally, transbuccally, intranasally, liposomally, via inhalation, vaginally, intraoccularly, via local delivery (for example by catheter or stent) , subcutaneously, intraadiposally, intraarticularly, and intrathecally.
  • kits comprising at least one compound of any one of 1-18 and pharmaceutically acceptable salts thereof, and instructions which comprise one or more forms of information selected from the group consisting of indicating a disease state for which the composition is to be administered, storage information for the composition, dosing information and instructions regarding how to administer the composition.
  • the kit comprises the compound in a multiple dose form.
  • an article of manufacture comprising at least one compound of any one of 1-18 and pharmaceutically acceptable salts thereof, and packaging materials.
  • the packaging material comprises a container for housing the compound.
  • the container comprises a label indicating one or more members of the group consisting of a disease state for which the compound is to be administered, storage information, dosing information and/or instructions regarding how to administer the compound.
  • the article of manufacture comprises the compound in a multiple dose form.
  • a therapeutic method comprising administering to a subject at least one compound of any one of 1-18 and pharmaceutically acceptable salts thereof.
  • a method of inhibiting a mutant EGFR kinase comprising contacting the mutant EGFR with at least one compound of any one of 1-18 and pharmaceutically acceptable salts thereof.
  • a method of inhibiting a mutant EGFR comprising causing at least one compound of any one of 1-18 and pharmaceutically acceptable salts thereof to be present in a subject in order to inhibit the mutant EGFR in vivo.
  • a method of treating a disease state for which a mutant EGFR possesses activity that contributes to the pathology and/or symptomology of the disease state comprising causing at least one compound of any one of 1-18 and pharmaceutically acceptable salts thereof to be present in a subject in a therapeutically effective amount for the disease state.
  • the disease state is selected from the group consisting of cancerous hyperproliferative disorders (e.g., brain, lung, squamous cell, bladder, gastric, pancreatic, breast, head, neck, renal, kidney, ovarian, prostate, colorectal, epidermoid, esophageal, testicular, gynecological or thyroid cancer) ; non-cancerous hyperproliferative disorders (e.g., benign hyperplasia of the skin (e.g., psoriasis) , restenosis, and benign prostatic hypertrophy (BPH) ) ; pancreatitis; kidney disease; pain; preventing blastocyte implantation; treating diseases related to vasculogenesis or angiogenesis (e.g., tumor angiogenesis, acute and chronic inflammatory disease such as rheumatoid arthritis, atherosclerosis, inflammatory bowel disease, skin diseases such as psoriasis, exzema, and s
  • a method of treating a disease state for which a mutation in the mutant EGFR gene contributes to the pathology and/or symptomology of the disease state including, for example, melanomas, lung cancer, colon cancer and other tumor types.
  • the present invention relates to the use of at least one compound of any one of 1-18 and pharmaceutically acceptable salts thereof as a medicament. In yet another of its aspects, the present invention relates to the use of at least one compound of any one of 1-18 and pharmaceutically acceptable salts thereof in the manufacture of a medicament for inhibiting a mutant EGFR.
  • the present invention relates to the use of at least one compound of any one of 1-18 and pharmaceutically acceptable salts thereof in the manufacture of a medicament for treating a disease state for which a mutant EGFR possesses activity that contributes to the pathology and/or symptomology of the disease state.
  • compounds of the disclosure will be administered in therapeutically effective amounts via any of the usual and acceptable modes known in the art, either singly or in combination with one or more therapeutic agents.
  • a therapeutically effective amount may vary widely depending on the severity of the disease, the age and relative health of the subject, the potency of the compound used and other factors known to those of ordinary skill in the art.
  • the required dosage will also vary depending on the mode of administration, the particular condition to be treated and the effect desired.
  • an indicated daily dosage in the larger mammal may be in the range from about 0.5 mg to about 2000 mg, or more particularly, from about 0.5 mg to about 1000 mg, conveniently administered, for example, in divided doses up to four times a day or in retard form.
  • Suitable unit dosage forms for oral administration comprise from ca. 1 to 50 mg active ingredient.
  • Compounds of the disclosure may be administered as pharmaceutical compositions by any conventional route; for example, 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 lotions, gels, ointments or creams, or in a nasal or suppository form.
  • enterally e.g., orally, e.g., in the form of tablets or capsules
  • parenterally e.g., in the form of injectable solutions or suspensions
  • topically e.g., in the form of lotions, gels, ointments or creams, or in a nasal or suppository form.
  • compositions comprising a compound of the present disclosure in free form or in a pharmaceutically acceptable salt form in association with at least one pharmaceutically acceptable carrier or diluent may be manufactured in a conventional manner by mixing, granulating, coating, dissolving or lyophilizing processes.
  • pharmaceutical compositions comprising a compound of the disclosure in association with at least one pharmaceutical acceptable carrier or diluent may be manufactured in conventional manner by mixing with a pharmaceutically acceptable carrier or diluent.
  • Unit dosage forms for oral administration contain, for example, from about 0.1 mg to about 500 mg of active substance.
  • the pharmaceutical compositions are solutions of the active ingredient, including suspensions or dispersions, such as isotonic aqueous solutions.
  • suspensions or dispersions such as isotonic aqueous solutions.
  • dispersions or suspensions can be made up before 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, or microbicides, such as sorbic acid or benzoic acid.
  • solutions or suspensions may further comprise viscosity-increasing agents, including but not limited to, sodium carboxymethylcellulose, carboxymethylcellulose, dextran, polyvinylpyrrolidone, gelatins, or solubilizers, e.g. Tween 80 (polyoxyethylene (20) sorbitan mono-oleate) .
  • viscosity-increasing agents including but not limited to, sodium carboxymethylcellulose, carboxymethylcellulose, dextran, polyvinylpyrrolidone, gelatins, or solubilizers, e.g. Tween 80 (polyoxyethylene (20) sorbitan mono-oleate) .
  • Suspensions in oil may comprise as the oil component the vegetable, synthetic, or semi-synthetic oils customary for injection purposes.
  • oils customary for injection purposes.
  • Examples include liquid fatty acid esters that contain as the acid component a long-chained 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, for example oleic acid, elaidic acid, erucic acid, brassidic acid and linoleic acid, and if desired, may contain antioxidants, for example vitamin E, 3-carotene or 3, 5-di-tert-butyl-hydroxytoluene.
  • the alcohol component of these fatty acid esters may have six carbon atoms and may be monovalent or polyvalent, for example a mono-, di-or trivalent, alcohol. Suitable alcohol components include but are not limited to methanol, ethanol, propanol, butanol or pentanol or isomers thereof; glycol and glycerol.
  • Suitable fatty acid esters include but are not limited ethyl-oleate, isopropyl myristate, isopropyl palmitate, M 2375, (polyoxyethylene glycerol) , M 1944 CS (unsaturated polyglycolized glycerides prepared by alcoholysis of apricot kernel oil and comprising glycerides and polyethylene glycol ester) , LABRASOL TM (saturated polyglycolized glycerides prepared by alcoholysis of TCM and comprising glycerides and polyethylene glycol ester; all available from GaKefosse, France) , and/or 812 (triglyceride of saturated fatty acids of chain length C8 to C12 from Hüls AG, Germany) , and vegetable oils such as cottonseed oil, almond oil, olive oil, castor oil, sesame oil, soybean oil, or groundnut oil.
  • vegetable oils such as cottonseed oil, almond oil, olive oil, castor oil, ses
  • compositions for oral administration may be obtained, for example, by combining the active ingredient with one or more solid carriers, and if desired, granulating a resulting mixture, and processing the mixture or granules by the inclusion of additional excipients, to form tablets or tablet cores.
  • Suitable carriers include but are not limited to fillers, such as sugars, for example lactose, saccharose, mannitol or sorbitol, cellulose preparations, and/or calcium phosphates, for example tricalcium phosphate or calcium hydrogen phosphate, and also binders, such as starches, for example corn, wheat, rice or potato starch, methylcellulose, hydroxypropyl methylcellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone, and/or, if desired, disintegrators, such as the above-mentioned starches, carboxymethyl starch, crosslinked polyvinylpyrrolidone, alginic acid or a salt thereof, such as sodium alginate.
  • Additional excipients include flow conditioners 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.
  • Tablet cores may be provided with suitable, optionally enteric, coatings through the use of, inter alia, concentrated sugar solutions which may comprise gum arable, talc, polyvinylpyrrolidone, polyethylene glycol and/or titanium dioxide, or coating solutions in suitable organic solvents or solvent mixtures, or, for the preparation of enteric coatings, solutions of suitable cellulose preparations, such as 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 active ingredient.
  • concentrated sugar solutions which may comprise gum arable, talc, polyvinylpyrrolidone, polyethylene glycol and/or titanium dioxide, or coating solutions in suitable organic solvents or solvent mixtures, or, for the preparation of enteric coatings, solutions of suitable cellulose preparations, such as acetylcellulose phthalate or hydroxypropylmethylcellulose phthalate.
  • Dyes or pigments may be added to the tablets or tablet coatings,
  • compositions for oral administration may also include hard capsules comprising gelatin or soft-sealed capsules comprising gelatin and a plasticizer, such as glycerol or sorbitol.
  • the hard capsules may contain the active ingredient 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.
  • the active ingredient may be dissolved or suspended in suitable liquid excipients, such as fatty oils, paraffin oil or liquid polyethylene glycols or fatty acid esters of ethylene or propylene glycol, to which stabilizers and detergents, for example of the polyoxyethylene sorbitan fatty acid ester type, may also be added.
  • suitable liquid excipients such as fatty oils, paraffin oil or liquid polyethylene glycols or fatty acid esters of ethylene or propylene glycol, to which stabilizers and detergents, for example of the polyoxyethylene sorbitan fatty acid ester type, may also be added.
  • compositions suitable for rectal administration are, for example, suppositories comprising 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.
  • compositions suitable for parenteral administration may comprise aqueous solutions of an active ingredient in water-soluble form, for example of a water-soluble salt, or aqueous injection suspensions that contain viscosity-increasing substances, for example sodium carboxymethylcellulose, sorbitol and/or dextran, and, if desired, stabilizers.
  • the active ingredient optionally together with excipients, can also be in the form of a lyophilizate and can be made into a solution before parenteral administration by the addition of suitable solvents. Solutions such as are used, for example, for parenteral administration can also be employed as infusion solutions.
  • the manufacture of injectable preparations is usually carried out under sterile conditions, as is the filling, for example, into ampoules or vials, and the sealing of the containers.
  • the compounds of the disclosure may be administered as the sole active ingredient, or together with other drugs useful against neoplastic diseases or useful in immunomodulating regimens.
  • the compounds of the disclosure may be used in accordance with the disclosure in combination with pharmaceutical compositions effective in various diseases as described above, e.g.
  • cyclophosphamide 5-fluorouracil, fludarabine, gemcitabine, cisplatinum, carboplatin, vincristine, vinblastine, etoposide, irinotecan, paclitaxel, docetaxel, rituxan, doxorubicine, gefitinib, or imatinib; or also with cyclosporins, rapamycins, ascomycins or their immunosuppressive analogs, e.g. cyclosporin A, cyclosporin G, FK-506, sirolimus or everolimus, corticosteroids, e.g.
  • prednisone cyclophosphamide, azathioprene, methotrexate, gold salts, sulfasalazine, antimalarials, brequinar, leflunomide, mizoribine, mycophenolic acid, mycophenolate, mofetil, 15-deoxyspergualine, immuno-suppressive monoclonal antibodies, e.g. monoclonal antibodies to leukocyte receptors, e.g.
  • kits comprising a) a first agent which is a compound of the disclosure as disclosed herein, in free form or in pharmaceutically acceptable salt form, and b) at least one co-agent.
  • the kit can comprise instructions for its administration.
  • the at least one compound of formula (I) can also be prepared as a pharmaceutically acceptable acid addition salt by, for example, reacting the free base form of the at least one compound with a pharmaceutically acceptable inorganic or organic acid.
  • a pharmaceutically acceptable base addition salt of the at least one compound of formula (I) can be prepared by, for example, reacting the free acid form of the at least one compound with a pharmaceutically acceptable inorganic or organic base.
  • Inorganic and organic acids and bases suitable for the preparation of the pharmaceutically acceptable salts of compounds of formula (I) are set forth in the definitions section of this Application.
  • the salt forms of the compounds of formula (I) can be prepared using salts of the starting materials or intermediates.
  • the free acid or free base forms of the compounds of formula (I) can be prepared from the corresponding base addition salt or acid addition salt form.
  • a compound of formula (I) in an acid addition salt form can be converted to the corresponding free base thereof by treating with a suitable base (e.g., ammonium hydroxide solution, sodium hydroxide, and the like) .
  • a compound of formula (I) in a base addition salt form can be converted to the corresponding free acid thereof by, for example, treating with a suitable acid (e.g., hydrochloric acid, etc) .
  • N-oxides of the at least one compound of formula (I) and/or at least one pharmaceutically acceptable salt thereof can be prepared by methods known to those of ordinary skill in the art.
  • N-oxides can be prepared by treating an unoxidized form of the compound of formula (I) with an oxidizing agent (e.g., trifluoroperacetic acid, permaleic acid, perbenzoic acid, peracetic acid, meta-chloroperoxybenzoic acid, or the like) in a suitable inert organic solvent (e.g., a halogenated hydrocarbon such as dichloromethane) at approximately 0 to 80 °C.
  • an oxidizing agent e.g., trifluoroperacetic acid, permaleic acid, perbenzoic acid, peracetic acid, meta-chloroperoxybenzoic acid, or the like
  • a suitable inert organic solvent e.g., a halogenated hydrocarbon such as dichloromethane
  • Compounds of formula (I) in an unoxidized form can be prepared from N-oxides of compounds of formula (I) by, for example, treating with a reducing agent (e.g., sulfur, sulfur dioxide, triphenyl phosphine, lithium borohydride, sodium borohydride, phosphorus trichloride, tribromide, and the like) in an suitable inert organic solvent (e.g., acetonitrile, ethanol, aqueous dioxane, and the like) at 0 to 80 °C.
  • a reducing agent e.g., sulfur, sulfur dioxide, triphenyl phosphine, lithium borohydride, sodium borohydride, phosphorus trichloride, tribromide, and the like
  • an inert organic solvent e.g., acetonitrile, ethanol, aqueous dioxane, and the like
  • Protected derivatives of the compounds of formula (I) can be made by methods known to those of ordinary skill in the art. A detailed description of the techniques applicable to the creation of protecting groups and their removal can be found in T.W. Greene, Protecting Groups in Organic Synthesis, 3rd edition, John Wiley &Sons, Inc. 1999.
  • references to ether or Et 2 O are to diethyl ether; brine refers to a saturated aqueous solution of NaCl. Unless otherwise indicated, all temperatures are expressed in °C (degrees Centigrade) . All reactions were conducted under an inert atmosphere at RT unless otherwise noted.
  • MS mass spectra
  • ESI electrospray ionization
  • UV detector 220 and 254 nm
  • ELSD evaporative light scattering detector
  • Thin-layer chromatography was performed on 0.25 mm E. Merck silica gel plates (60F-254) , visualized with UV light, 5%ethanolic phosphomolybdic acid, ninhydrin, or p-anisaldehyde solution. Flash column chromatography was performed on silica gel (230-400 mesh, Merck) .
  • At least one compound of formula I and/or at least one pharmaceutically acceptable salt thereof may be synthesized according to a variety of reaction schemes. Some illustrative schemes are provided below and in the examples. Other reaction schemes could be readily devised by those skilled in the art in view of the present disclosure.
  • the compound of formula I of the present disclosure can be prepared as shown in Scheme 1. Coupling of intermediates such as those of formula III with amino-arene such as IV using such coupling conditions as Buchwald amination reaction or amination conditions known in the literature provides the intermediate VII or VI. Reduction of the nitro group in intermediate VII using conditions such as Fe/NH 4 Cl in a solvent or mixture of solvents such as EtOH and H 2 O provides amine VIII. Alternatively intermediate VIII can be prepared by cleavage of the protecting group on the amine of intermediate VI. Intermediate VI can be obtained by the coupling between intermediate III and amino-arene V. Coupling of amine VIII with acid IX or acyl chloride X provides compound I. Compound of formula I can also be synthesized via a two-step sequence by reacting amine VIII with acyl chloride XI followed by elimination reaction in the presence of a base such as NaOH in a solvent such as H 2 O.
  • compound of formula I can be prepared by coupling of intermediate III with intermediate VIa under the conditions as described in Scheme 1 or Scheme 2 as shown in Scheme 3.
  • VIaa One preparation of VIaa is illustrated in Scheme 4. Starting from the commercially available dichloropyridine 1, intermediate VIaa can be prepared via a sequence of region-selective displacement of chloride, bromination, amination, deprotection, acylation and reduction.
  • N 1 - (6-Methoxy-5-nitropyridin-2-yl) -N 1 , N 2 , N 2 -trimethylethane-1, 2-diamine (1b) (1.33 g, 5.24 mmol) in acetonitrile (30 mL) at 0°C was added NBS (1.40 g, 7.82 mmol) .
  • NBS (1.40 g, 7.82 mmol
  • 6-chloro-2-ethoxy-3-nitropyridine (5a) was prepared according to the synthetic method of 1a by replacing methanol with ethanol. MS-ESI (m/z) : 203 [M + 1] + .
  • the title compound 8 (6.3 mg, 75%) was prepared according to the synthetic method of Example 6 by replacing N- (6-Ethoxy-2- (methyl (2- (methylamino) ethyl) amino) -5- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-ylamino) 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-ylamino) pyridin-3-yl) acr ylamide (7) .
  • MS-ESI (m/z) 529 [M + 1] + .
  • the title compound 10 (5.2 mg) was prepared according to the synthetic method of Example 6 by replacing N- (6-Ethoxy-2- (methyl (2- (methylamino) ethyl) amino) -5- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-ylamino) pyridin-3-yl) acrylamide (5) 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) .
  • MS-ESI (m/z) : 499 [M + 1] + .
  • the title compound 11 (55 mg) was prepared according to the synthetic method of Example 2 by replacing methyl (2- (methylamino) ethyl) carbamate (2a) with tert-butyl piperazine-1-carboxylate and replacing 6-Chloro-2-methoxy-3-nitropyridine (1a) with 6-chloro-2-ethoxy-3-nitropyridine (5a) respectively.
  • MS-ESI (m/z) : 499 [M + 1] + .
  • the title compound 12 (7.2 mg) was prepared according to the synthetic method of Example 6 by replacing N- (6-Ethoxy-2- (methyl (2- (methylamino) ethyl) amino) -5- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-ylamino) pyridin-3-yl) acrylamide (5) 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) .
  • MS-ESI (m/z) : 513 [M + 1] + .
  • the title compound 13 (75 mg) was prepared according to the synthetic method of Example 2 by replacing tert-butyl methyl (2- (methylamino) ethyl) carbamate (2a) with tert-butyl piperazine-1-carboxylate and replacing 6-Chloro-2-methoxy-3-nitropyridine (1a) with 6-chloro-2-isopropoxy-3-nitropyridine (7a) respectively.
  • MS-ESI m/z
  • N 1 - (6-chloro-3-nitropyridin-2-yl) -N 1 , N 2 , N 2 -trimethylethane-1, 2-diamine (15a) (23.8 g, 91.9 mmol) in methanol (200 mL) was added sodium methoxide (10.8 g, 183.8 mmol) at 0-5°C.
  • the mixture was heated to reflux for 2 h.
  • the mixture was cooled to r.t. and methanol was evaporated, diluted with water (200 mL) and extracted with dichloromethane (200 mL ⁇ 2) .
  • N 1 - (6-methoxy-3-nitropyridin-2-yl) -N 1 , N 2 , N 2 -trimethylethane-1, 2-diamine (15b) (11.0 g, 43.1 mmol) in glacial acetic acid (120 mL) was added N-iodosuccinimide (11.6 g, 51.8 mmol) for 3 batches at 0-5°C, and the mixture was stirred at room temperature for 4.5 h. Then solvent was evaporated under reduced pressure, diluted with water (300 mL) and extracted with ethyl acetate (200 mL ⁇ 3) .
  • the reaction was quenched by saturated aqueous sodium bicarbonate (20 mL) , and the resulting mixture was extracted with dichloromethane (20 mL ⁇ 3) . The organic layers were combined, washed with water (20 mL ⁇ 3) and brine (20 mL) , and dried over Na 2 SO 4 . The solids were filtered out and the filtrate was concentrated in vacuum.
  • 5-bromo-3-methoxy-2-nitropyridine (22a) was prepared according to the synthetic method described in US2006/84665.
  • the title compound 23a was prepared according to the synthetic method of Example 2 by replacing N 1 - (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N 1 , N 2 , N 2 -trimethylethane-1, 2-diamine (2g) with N 2 - (2- (dimethylamino) ethyl) -6-methoxy-N 2 -methyl-3-nitropyridine-2, 5-diamine (15e) .
  • MS-ESI (m/z) 477 [M + 1] + .
  • the title compound 23b was prepared according to the synthetic method of 15i by replacing N 1 - (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N 1 , N 2 , N 2 -trimethylethane-1, 2-diamine (15h) with N 2 - (2- (dimethylamino) ethyl) -6-methoxy-N 2 -methyl-N 5 - (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) -3-nitropy9ridine-2, 5-diamine (23a) .
  • MS-ESI (m/z) 447 [M + 1] + .
  • the title compound 24 was prepared according to the synthetic method of Example 23 by replacing N 2 - (2- (dimethylamino) ethyl) -6-methoxy-N 2 -methyl-3-nitropyridine-2, 5-diamine (15e) with N 1 - (2- (dimethylamino) ethyl) -5-methoxy-N 1 -methyl-2-nitrobenzene-1, 4-diamine (24a) .
  • MS-ESI (m/z) 517 [M + 1] + .
  • the title compound 25 was prepared according to the synthetic method of Example 15 by replacing N 1 - (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N 1 , N 2 , N 2 -trimethylethane-1, 2-diamine (15h) with N 2 - (2- (dimethylamino) ethyl) -6-methoxy-N 2 -methyl-N 5 - (4- (1-methylindolizin-3-yl) pyrimidin-2-yl) -3-nitropyridine-2, 5-diamine (25g) .
  • MS-ESI (m/z) 501 [M + 1] + .
  • the title compound 26 was prepared according to the synthetic method of Example 25 by replacing N 1 - (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N 1 , N 2 , N 2 -trimethylethane-1, 2-diamine (15c) with N 1 - (4-iodo-5-methoxy-2-nitrophenyl) -N 1 , N 2 , N 2 -trimethylethane-1, 2-diamine (26a) .
  • MS-ESI (m/z) 500 [M + 1] + .
  • the title compound 27 was prepared according to the synthetic method of Example 25 by replacing ethyl 1-methylindolizine-3-carboxylate (25b) with ethyl 1-cyanoindolizine-3-carboxylate (27b) .
  • MS-ESI (m/z) 501 [M + 1] + .
  • the title compound 28 was prepared according to the synthetic method of Example 27 by replacing N 1 - (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N 1 , N 2 , N 2 -trimethylethane-1, 2-diamine (15c) with N 1 - (4-iodo-5-methoxy-2-nitrophenyl) -N 1 , N 2 , N 2 -trimethylethane-1, 2-diamine (26a) .
  • MS-ESI (m/z) 511 [M + 1] + .
  • the title compound 30 was prepared according to the synthetic method of Example 29 by replacing N 1 - (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N 1 , N 2 , N 2 -trimethylethane-1, 2-diamine (15c) with N 1 - (4-iodo-5-methoxy-2-nitrophenyl) -N 1 , N 2 , N 2 -trimethylethane-1, 2-diamine (26a) .
  • MS-ESI (m/z) 486 [M + 1] + .
  • the title compound 31 was prepared according to the synthetic method of Example 25 by replacing compound ethyl 1-methylindolizine-3-carboxylate (25b) with ethyl 1-chloroindolizine-3-carboxylate (31b) .
  • MS-ESI (m/z) 521 [M + 1] + .
  • the title compound 32 was prepared according to the synthetic method of Example 31 by replacing N 1 - (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N 1 , N 2 , N 2 -trimethylethane-1, 2-diamine (15c) with N 1 - (4-iodo-5-methoxy-2-nitrophenyl) -N 1 , N 2 , N 2 -trimethylethane-1, 2-diamine (26a) .
  • MS-ESI (m/z) 520 [M + 1] + .
  • the title compound 33 was prepared according to the synthetic method of Example 15 by replacing N 1 - (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N 1 , N 2 , N 2 -trimethylethane-1, 2-diamine (15h) with compound N 2 - (2- (dimethylamino) ethyl) -6-methoxy-N 2 -methyl-N 5 - (4- (3-methylindolizin-1-yl) pyrimidin-2-yl) -3-nitropyridine-2, 5-diamine (33f) .
  • MS-ESI (m/z) 501 [M + 1] + .
  • the title compound 34 was prepared according to the synthetic method of Example 33 by replacing N 1 - (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N 1 , N 2 , N 2 -trimethylethane-1, 2-diamine (15c) with N 1 - (4-iodo-5-methoxy-2-nitrophenyl) -N 1 , N 2 , N 2 -trimethylethane-1, 2-diamine (26a) .
  • MS-ESI (m/z) 500 [M + 1] + .
  • the title compound 35 was prepared according to the synthetic method of Example 33 by replacing 4- (3-methylindolizin-1-yl) pyrimidin-2-amine (33e) with 1- (2-aminopyrimidin-4-yl) indolizine-3-carbonitrile (35c) .
  • MS-ESI (m/z) 512 [M + 1] + .
  • the title compound 36 was prepared according to the synthetic method of Example 35 by replacing N 1 - (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N 1 , N 2 , N 2 -trimethylethane-1, 2-diamine (15c) with N 1 - (4-iodo-5-methoxy-2-nitrophenyl) -N 1 , N 2 , N 2 -trimethylethane-1, 2-diamine (26a) .
  • MS-ESI (m/z) 511 [M + 1] + .
  • the title compound 37 was prepared according to the synthetic method of Example 35 by replacing 1-acetylindolizine-3-carbonitrile (35a) with1- (indolizin-1-yl) ethan-1-one (37c) .
  • MS-ESI (m/z) 487 [M + 1] + .
  • the title compound 38 was prepared according to synthetic the method of Example 37 by replacing N 1 - (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N 1 , N 2 , N 2 -trimethylethane-1, 2-diamine (15c) with N 1 - (4-iodo-5-methoxy-2-nitrophenyl) -N 1 , N 2 , N 2 -trimethylethane-1, 2-diamine (26a) .
  • MS-ESI (m/z) 486 [M + 1] + .
  • the title compound 39 was prepared according to the synthetic method of Example 35 by replacing 1-acetylindolizine-3-carbonitrile (35a) with1- (3-chloroindolizin-1-yl) ethan-1-one (39a) .
  • MS-ESI (m/z) 521 [M + 1] + .
  • the title compound 40 was prepared according to the synthetic method of Example39by replacing N 1 - (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N 1 , N 2 , N 2 -trimethylethane-1, 2-diamine (15c) withN 1 - (4-iodo-5-methoxy-2-nitrophenyl) -N 1 , N 2 , N 2 -trimethylethane-1, 2-diamine (26a) .
  • MS-ESI (m/z) 520 [M + 1] + .
  • the title compound 43 (4.9 mg) was prepared according to the synthetic method of Example 6 by replacing N- (6-Ethoxy-2- (methyl (2- (methylamino) ethyl) amino) -5- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-ylamino) pyridin-3-yl) acrylamide (5) with N- (4-methoxy-5- ( (4- (3-methyl-1H-indol-1-yl) pyrimidin-2-yl) amino) -2- (piperazin-1-yl) phenyl) acrylamide (43a) .
  • MS-ESI (m/z) : 499 [M + 1] + .
  • the title compound 44 was prepared according to the synthetic method of Example 42 by replacing N 1 , N 1 , N 2 -trimethylethane-1, 2-diamine with 1-methylpiperazine. MS-ESI (m/z) : 498 [M + 1] + .
  • the title compound 46 (5 mg) was prepared according to the synthetic method of Example 45 by replacing N 2 - (2- (dimethylamino) ethyl) -6-methoxy-N 2 -methyl-3-nitropyridine-2, 5-diamine (15e) with N 1 - (2- (dimethylamino) ethyl) -5-methoxy-N 1 -methyl-2-nitrobenzene-1, 4-diamine (24a) .
  • MS-ESI (m/z) 511 [M + 1] + .
  • the title compound (47) was prepared according to the synthetic method of Example 15 by replacing N 2 - (2- (dimethylamino) ethyl) -N 5 - (4- (7-fluoro-1-methyl-1H-indol-3-yl) pyrimidin-2-yl) -6-methoxy-N 2 -methyl-3-nitropyridine-2, 5-diamine (15h) with N 1 - (4- (1-chloroimidazo [1, 5-a] pyridin-3-yl) pyrimidin-2-yl) -N 4 - (2- (dimethylamino) ethyl) -2-methoxy -N 4 -methyl-5-nitrobenzene-1, 4-diamine (47f) .
  • MS-ESI (m/z) 468 [M + 1] + .
  • the title compound (48) was prepared according to the synthetic method of Example 47 by replacing N 2 - (2- (dimethylamino) ethyl) -6-methoxy-N 2 -methyl-3-nitropyridine-2, 5-diamine (15e) with N 1 - (4-iodo-5-methoxy-2-nitrophenyl) -N 1 , N 2 , N 2 -trimethylethane-1, 2-diamine (24a) .
  • MS-ESI (m/z) 521 [M + 1] + .
  • the title compound 49 was prepared according to the synthetic method of Example 47 by replacing 1-chloro-3- (2- (methylthio) pyrimidin-4-yl) imidazo [1, 5-a] pyridine (47c) with 3- (2- (methylthio) pyrimidin-4-yl) -1- (trifluoromethyl) imidazo [1, 5-a] pyridine (49b) .
  • MS-ESI (m/z) 556 [M + 1] +
  • the title compound 50 was prepared according to the synthetic method of Example 47 by replacing N 2 - (2- (dimethylamino) ethyl) -6-methoxy-N 2 -methyl-3-nitropyridine-2, 5-diamine (15e) with N 1 - (2- (dimethylamino) ethyl) -5-methoxy-N 1 -methyl-2-nitrobenzene-1, 4-diamine (24a) .
  • MS-ESI (m/z) 555 [M + 1] + .
  • the title compound 51a was prepared according to the synthetic method of 47b by replacing pyridin-2-ylmethanamine with 1- (pyridin-2-yl) ethan-1-amine. MS-ESI (m/z) : 257 [M + 1] + .
  • the title compound 51b was prepared according to the synthetic method of 47e by replacing 1-chloro-3- (2- (methylthio) pyrimidin-4-yl) imidazo [1, 5-a] pyridine (47c) with 1-methyl-3- (2- (methylthio) pyrimidin-4-yl) imidazo [1, 5-a] pyridine (51a) .
  • MS-ESI (m/z) 245 [M + 1] + .
  • the title compound (51c) was prepared according to the synthetic method of 15i by replacing 3- (2-chloropyrimidin-4-yl) -7-fluoro-1-methyl-1H-indole (15g) with 3- (2-chloropyrimidin-4-yl) -1-methylimidazo [1, 5-a] pyridine (51b) .
  • MS-ESI (m/z) 448 [M + 1] + .
  • the title compound 51 was prepared according to the synthetic method of Example 15 by replacing N 2 - (2- (dimethylamino) ethyl) -N 5 - (4- (7-fluoro-1-methyl-1H-indol-3-yl) pyrimidin-2-yl) -6-methoxy-N 2 -methylpyridine-2, 3, 5-triamine (15i) with N 2 - (2- (dimethylamino) ethyl) -6-methoxy-N 2 -methyl-N 5 - (4- (1-methylimidazo [1, 5-a] pyridin-3-yl) pyrimidin-2-yl) pyridine-2, 3, 5-tri amine (51c) .
  • MS-ESI (m/z) 502 [M + 1] + .
  • the title compound 52 was prepared according to the synthetic method of Example 51 by replacing N 2 - (2- (dimethylamino) ethyl) -6-methoxy-N 2 -methyl-3-nitropyridine-2, 5-diamine (15e) with N 1 - (2- (dimethylamino) ethyl) -5-methoxy-N 1 -methyl-2-nitrobenzene-1, 4-diamine (24a) .
  • MS-ESI (m/z) 501 [M + 1] + .
  • the title compound (53) was prepared according to the method of Example 23 by replacing N 2 - (2- (dimethylamino) ethyl) -6-methoxy-N 2 -methyl-N 5 - (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) pyridine-2, 3, 5-triamine (23b) with N 2 - (2- (dimethylamino) ethyl) -6-methoxy -N 2 -methyl-N 5 - (4- (1-methylimidazo [1, 5-a] pyridin-3-yl) pyrimidin-2-yl) pyridine-2, 3, 5-triamine (51c) .
  • MS-ESI (m/z) 520 [M + 1] + .
  • the title compound 54 (5.0 mg) was prepared by using the same procedure as described for 15 by replacing 3- (2-chloropyrimidin-4-yl) -7-fluoro-1-methyl-1H-indole (15g) with 5- (2-chloropyrimidin-4-yl) -1, 2-dihydropyrrolo [3, 2, 1-hi] indole (54f) .
  • MS-ESI (m/z) : 513 [M + 1] + .
  • the title compound (55) was prepared according to the synthetic method of Example 54 by replacing N 1 - (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N 1 , N 2 , N 2 -trimethylethane-1, 2-diamine (15c) withN 1 - (4-iodo-5-methoxy-2-nitrophenyl) -N 1 , N 2 , N 2 -trimethylethane-1, 2-diamine (24a) .
  • MS-ESI (m/z) 512 [M + 1] + .
  • the title compound 58 was prepared according to the synthetic method of Example 25 by replacing 1- (1-methylindolizin-3-yl) ethanone (25d) with 1- (1-methyl-1H-indazol-3-yl) ethanone (58a) .
  • MS-ESI (m/z) 502 [M + 1] + .
  • the compound 59a was prepared according to the synthetic method of Example 25 by replacing 1- (1-methylindolizin-3-yl) ethanone (25d) with 1- (1-methyl-1H-indazol-3-yl) ethanone (58a) .
  • MS-ESI (m/z) 448 [M + 1] + .
  • the title compound 59b was prepared according to the synthetic method of 15i by replacing N 1 - (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N 1 , N 2 , N 2 -trimethylethane-1, 2-diamine (15h) with N 2 - (2- (dimethylamino) ethyl) -6-methoxy-N 2 -methyl-N 5 - (4- (1-methyl-1H-indazol-3-yl) pyrimidin-2-yl) -3-nitropyridine-2, 5-diamine (59a) .
  • MS-ESI (m/z) 447 [M + 1] + .
  • the title compound (59) was prepared according to the synthetic method of Example 23 by replacing N 2 - (2- (dimethylamino) ethyl) -6-methoxy-N 2 -methyl-N 5 - (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) pyridine-2, 3, 5-triamine (23b) with N 2 - (2- (dimethylamino) ethyl) -6-methoxy -N 2 -methyl-N 5 - (4- (1-methyl-1H-indazol-3-yl) pyrimidin-2-yl) pyridine-2, 3, 5-triamine (59b) .
  • MS-ESI (m/z) 520 [M + 1] + .
  • the title compound 60a was prepared according to the synthetic method described in literature: Journal of Chemical society., 1955, 2834.
  • the title compound 60 was prepared according to the synthetic method of Example 25 by replacing 1- (1-methylindolizin-3-yl) ethanone (25d) with 1- (3-methylimidazo [1, 5-a] pyridin-1-yl) ethan-1-one (60a) .
  • MS-ESI (m/z) 502 [M + 1] + .
  • the compound 61a was prepared according to the synthetic method of Example 25 by replacing 1- (1-methylindolizin-3-yl) ethanone (25d) with 1- (3-methylimidazo [1, 5-a] pyridin-1-yl) ethan-1-one (60a) .
  • MS-ESI (m/z) 448 [M + 1] + .
  • the title compound 61 was prepared according to the synthetic method of Example 23 by replacing N 2 - (2- (dimethylamino) ethyl) -6-methoxy-N 2 -methyl-N 5 - (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) pyridine-2, 3, 5-triamine (23b) with N 2 - (2- (dimethylamino) ethyl) -6-methoxy -N 2 -methyl-N 5 - (4- (3-methylimidazo [1, 5-a] pyridin-1-yl) pyrimidin-2-yl) pyridine-2, 3, 5-triamine (61a) .
  • MS-ESI (m/z) 520 [M + 1] + .
  • H1975 cells were cultured in culture flasks to 40-80%confluence in RPMI-1640 plus 10%fetal bovine serum. Cells were collected and plated onto 96-well plates at desired cell density (3000 cells/well) . Plates were incubated overnight at 37°C, with 5%CO 2 to adhere.
  • A431 cell was cultured in culture flasks to 40-80%confluence in DMEM plus 10%fetal bovine serum. Cells were collected and plated onto 96-well plates at desired cell density (3000 cells/well) . Plates were incubated overnight at 37°C, with 5%CO 2 to adhere. Compounds were added to the plates, the final compound concentrations were 10000, 3333.3, 1111.1, 270.4, 123.5, 41.2, 13.7, 4.6 and 1.5 nM.

Abstract

Provided are certain EGFR mutant selective inhibitors, pharmaceutical compositions thereof, and methods of use therefor.

Description

CERTAIN PROTEIN KINASE INHIBITORS FIELD OF THE INVENTION
This invention relates to novel amino pyrimidine ring derivatives and the pharmaceutically acceptable salts thereof. The compounds of the present invention are mutant-selective epidermal growth factor receptor (EGFR) kinase inhibitors. The compounds of this invention are useful for the treatment of hyper-proliferative diseases such as cancer, in mammals and especially in humans. This invention also relates to the use of the compounds in the treatment of cancer and the pharmaceutical preparations containing them.
BACKGROUND OF THE INVENTION
Protein tyrosine kinases catalyze the transfer of a phosphate group from ATP or GTP to a tyrosine residue located on a protein substrate. Protein tyrosine kinase can be broadly classified as receptor (e.g. EGFR, HER-2, VEGFR, FGFR) or non-receptor (e.g. Src, Jak, Abl) . Receptor tyrosine kinases act to transmit signals from the outside of a cell to the inside by activating secondary messaging effectors via a phosphorylation event. A variety of cellular processes are promoted by these signals, including proliferation, carbohydrate utilization, protein synthesis, angiogenesis, cell growth, and cell survival.
Inappropriate or uncontrolled activation of tyrosine kinase activity, for example over-expression or mutation of this kinase, has been shown to result in uncontrolled cell growth. There is strong precedent for involvement of the EGFR in human cancer because over 60%of all solid tumors overexpress at least one of these proteins or their ligands. Overexpression of EGFR is commonly found in breast, lung, head and neck, bladder tumors.
The first-generation reversible, ATP-competitive EGFR kinase inhibitors, gefitinib and erlotinib, are effective clinical therapies for non-small cell lung cancers that harbor activating  mutations in the EGFR kinase domain. (Nature, 2009, 462, 1070-1074) Although first-generation EGFR inhibitors showed encouraging clinical responses, almost all patients developed resistance to these inhibitors over time, such as by mutation of gatekeeper residue T790M, which accounts for about half of all resistance to gefitinib and erlotinib (Proc. Natl. Acad. Sci. U.S.A. 2008, 105, 2070-2075) . Moreover, T790M may also be pre-existing; there may be an independent, oncogenic role for the T790M mutation.
Current drugs in development, including second-generation covalent inhibitors, such as afatinib, neratinib, canertinib and dacomitinib, are effective against the resistance mutation but exhibit dose-limiting toxicities such as diarrhea and skin rash due to concurrent inhibition of WT EGFR.
Therefore, there remains a need to provide mutant-selective EGFR kinase inhibitors useful as therapeutic agents. Although mutant-selective EGFR inhibitors were disclosed in the arts, e.g., WO 2013014448 and WO 2012061299, there is still a need for new mutant-selective EGFR inhibitors that have at least one advantageous property selected from potency, stability, selectivity, toxicity and pharmacodynamics properties as an alternative for the treatment of hyper-proliferative diseases. In this regard, a novel class of mutant-selective EGFR inhibitors is provided herein.
DISCLOSURE OF THE INVENTION
Disclosed herein are certain novel amino pyrimidine ring derivatives and pharmaceutical compositions thereof, and their use as pharmaceuticals.
In one aspect, disclosed herein is at least one compound of formula (I) :
Figure PCTCN2015081332-appb-000001
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;
R1 is selected from hydrogen, C1-10alkyl, C3-10cycloalkyl, C3-10cycloalkyl-C1-4alkyl, heterocyclyl, and heterocyclyl-C1-4alkyl, wherein alkyl, cycloalkyl, and heterocyclyl are each unsubstituted or substituted with at least one substituent, such as one, two, three, or four substituents, independently selected from R6a
R1’ is selected from hydrogen and halogen;
R2 and R3 are independently selected from hydrogen, halogen, hydroxyl, CN, C1-10alkyl, C2-10alkenyl, C2-10alkynyl, C3-10cycloalkyl, C3-10cycloalkyl-C1-4alkyl, heterocyclyl, heterocyclyl-C1-4alkyl, aryl, aryl-C1-4alkyl, heteroaryl, and heteroaryl-C1-4alkyl, wherein alkyl, alkenyl, alkynyl, cycloalkyl, and heterocyclyl are each unsubstituted or substituted with at least one substituent, such as one, two, three, or four substituents independently selected from R6a, and each aryl and heteroaryl is unsubstituted or substituted with at least one substituent, such as one, two, three, or four substituents, independently selected from R6b; or R2 and R3 together with the carbon atoms 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, and optionally substituted with 1or 2 R6a groups;
each R4 is independently selected from hydrogen, C1-10alkyl, C2-10alkenyl, 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 (CR9R10tNR7R8, -C (O) R7, -CO2R8, -CO2 (CR9R10tCONR7R8, -OC (O) R7, -CN, -C (O) NR7R8, -NR7C (O) R8, -OC (O) NR7R8, -NR7C (O) OR8, -NR7C (O) NR7R8, and -CR7 (N-OR8) , wherein C1-10alkyl, C2-10alkenyl, C2-10alkynyl, and C3-10cycloalkyl are each unsubstituted or substituted with at least one substituent, such as one, two, three, or four substituents, independently selected from R6a
each R5 is independently selected from hydrogen, C1-10alkyl, C2-10alkenyl, C2-10alkynyl, C3-10cycloalkyl, C3-10cycloalkyl-C1-4alkyl, heterocyclyl, heterocyclyl-C1-4alkyl, aryl, aryl-C1-4alkyl, heteroaryl, and heteroaryl-C1-4alkyl, -OR8, -NR7S (O) rR8, -NO2, halogen, -S (O) rR7, -SR8, -S (O) 2OR7, -OS (O) 2R8, -S (O) rNR7R8, -NR7R8, -O (CR9R10tNR7R8, -C (O) R7, -CO2R8, -CO2 (CR9R10tCONR7R8, -OC (O) R7, -CN, -C (O) NR7R8, -NR7C (O) R8, -OC (O) NR7R8, -NR7C (O) OR8, -NR7C (O) NR7R8, and -CR7 (N-OR8) , wherein C1-10alkyl, C2-10alkenyl, C2-10alkynyl, C3-10cycloalkyl and heterocyclyl are each unsubstituted or substituted with at least one substituent, such as one, two, three, or four substituents independently selected from R6a, and each aryl and heteroaryl is unsubstituted or substituted with at least one substituent, such as one, two, three, or four substituents, independently selected from R6b
each R6a is independently selected from C1-10alkyl, C2-10alkenyl, C2-10alkynyl, C3-10cycloalkyl, -OR8, -NR7S (O) rR8, -NO2, halogen, -S (O) rR7, -SR8, -S (O) 2OR7, -OS (O) 2R8, -S (O) rNR7R8, -NR7R8, - (CR9R10tOR8, - (CR9R10tNR7R8, - (CR9R10tSR8, - (CR9R10tS (O) rR8, - (CR9R10tCO2R8, - (CR9R10tCONR7R8, - (CR9R10tNR7CO2R8, - (CR9R10tOCONR7R8, - (CR9R10tNR7CONR7R8, - (CR9R10tNR7SO2NR7R8, -O (CR9R10tNR7R8, -C (O) R7, -C (O) (CR9R10tOR8, -C (O) (CR9R10tNR7R8, -C (O) (CR9R10tSR8, -C (O) (CR9R10tS (O) rR8, -CO2R8, -CO2 (CR9R10tCONR7R8, -OC (O) R7, -CN, -C (O) NR7R8, -NR7C (O) R8, -OC (O) NR7R8, -NR7C (O) OR8, -NR7C (O) NR7R8, and -CR7 (N-OR8) ;
each R6b is independently selected from R6a, aryl, aryl-C1-4alkyl, heteroaryl, and heteroaryl-C1-4 alkyl;
each R7 and each R8 are independently selected from hydrogen, C1-10alkyl, C2-10alkenyl,  C2-10alkynyl, cycloalkyl, cycloalkyl-C1-4alkyl, heterocyclyl, heterocyclyl-C1-4alkyl, aryl, aryl-C1-4alkyl, heteroaryl, and heteroaryl-C1-4alkyl, wherein alkyl, alkenyl, alkynyl, cycloalkyl, and heterocyclyl are each unsubstituted or substituted with at least one substituent, such as one, two, three, or four substituents independently selected from R6a, and aryl and heteroaryl are each unsubstituted or substituted with at least one substituent, such as one, two, three, or four substituents independently selected from R6b; or R7 and R8 together with the atom (s) to which they are attached form a heterocyclic ring of 4 to 12 members containing 0, 1, or 2 additional heteroatoms independently selected from oxygen, sulfur and nitrogen, and optionally substituted with 1or 2 R6b groups;
each R9 and each R10 are independently selected from hydrogen, C1-10alkyl, C2-10alkenyl, C2-10alkynyl, cycloalkyl, cycloalkyl-C1-4alkyl, heterocyclyl, heterocyclyl-C1-4alkyl, aryl, aryl-C1-4alkyl, heteroaryl, and heteroaryl-C1-4alkyl; or R9 and R10 together with the carbon atom (s) to which they are attached form a ring of 3 to 7 members containing 0, 1, or 2 heteroatoms independently selected from oxygen, sulfur and nitrogen, and optionally substituted with 1or 2 R6a groups;
m is independently selected from 0, 1, 2, and 3;
n is independently selected from 0, 1, 2, and 3;
each r is independently selected from 1 and 2;
each t is independently selected from 1, 2, and 3.
In yet another aspect, disclosed herein are the pharmaceutical compositions comprising at least one compound of formula (I) and/or at least one pharmaceutically acceptable salt thereof and at least one pharmaceutically acceptable excipient.
In yet another aspect, disclosed herein are the methods for modulating mutant EGFR, comprising administering to a system or a subject in need thereof, a therapeutically effective amount of at least one compound of formula (I) and/or at least one pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof, thereby modulating said mutant EGFR.
In yet another aspect, disclosed herein are the methods to treat, ameliorate or prevent a condition which responds to inhibition of mutant EGFR comprising administering to a system or subject in need of such treatment an effective amount of at least one compound of formula (I) and/or at least one pharmaceutically acceptable salt thereof or pharmaceutical compositions thereof, and optionally in combination with a second therapeutic agent, thereby treating said condition.
In yet another aspect, disclosed herein are the uses of at least one compound of formula (I) and/or at least one pharmaceutically acceptable salt thereof in the manufacture of a medicament for treating a condition mediated by mutant EGFR. The compounds of the disclosure may be used alone or in combination with a second therapeutic agent to treat a condition mediated by mutant EGFR, wherein said condition is an autoimmune disease, a transplantation disease, an infectious disease or a cell proliferative disorder.
Furthermore, disclosed herein are the methods for treating a cell proliferative disorder, comprising administering to a system or subject in need of such treatment an effective amount of at least one compound of formula (I) and/or at least one pharmaceutically acceptable salt thereof or pharmaceutical compositions thereof, and optionally in combination with a second therapeutic agent, thereby treating said condition.
Alternatively, disclosed herein are the uses of at least one compound of formula (I) and/or at least one pharmaceutically acceptable salt thereof in the manufacture of a medicament for treating a cell-proliferative disorder. In particular examples, the compounds of the disclosure may be used alone or in combination with a chemotherapeutic agent to treat a cell proliferative disorder, including but not limited to, lymphoma, osteosarcoma, melanoma, or a tumor of breast, renal, prostate, colorectal, thyroid, ovarian, pancreatic, neuronal, lung, uterine or gastrointestinal tumor.
In the above methods for using the compounds of the disclosure, at least one compound of formula (I) and/or at least one pharmaceutically acceptable salt thereof may be administered to a system comprising cells or tissues, or to a mammalian subject such as a human or animal subject.
As used herein the following definitions are applicable.
The term "alkyl" refers to both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms. Unless otherwise specified, “alkyl” refers to Cl-C10alkyl. For example, C1-6, as in "Cl-6alkyl" is defined to include groups having 1, 2, 3, 4, 5, or 6 carbons in a linear or branched arrangement. For example, "Cl-8alkyl" includes but is not limited to methyl, ethyl, n-propyl, i-propyl, n-butyl, t-butyl, i-butyl, pentyl, hexyl, heptyl, and octyl.
The term "cycloalkyl" means a saturated aliphatic cyclic hydrocarbon group having the specified number of carbon atoms. Unless otherwise specified, “cycloalkyl” refers to C3-l0cycloalkyl. For example, "cycloalkyl" includes but is not limited to cyclopropyl, methyl-cyclopropyl, 2, 2-dimethyl-cyclobutyl, 2-ethyl-cyclopentyl, and cyclohexyl.
The term "alkenyl" refers to a non-aromatic hydrocarbon radical, straight, branched or cyclic, containing from 2 to 10 carbon atoms and at least one carbon to carbon double bond. In some embodiments, one carbon to carbon double bond is present, and up to four non-aromatic carbon-carbon double bonds may be present. Thus, "C2-6alkenyl" means an alkenyl radical having from 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 double bonds and may be substituted if a substituted alkenyl group is indicated.
The term "alkynyl" refers to a hydrocarbon radical straight, branched or cyclic, containing from 2 to 10 carbon atoms and at least one carbon to carbon triple bond. In some embodiments, up to three carbon-carbon triple bonds may be present. Thus, "C2-6alkynyl" means an alkynyl radical having from 2 to 6 carbon atoms. Alkynyl groups include but are not limited to ethynyl, propynyl, butynyl, and 3-methylbutynyl. The straight, branched or cyclic portion of the alkynyl group may contain triple bonds and may be substituted if a substituted alkynyl group is indicated.
The term "aryl" encompasses 5-and 6-membered carbocyclic aromatic rings, for example, benzene; bicyclic ring systems wherein at least one ring is carbocyclic and aromatic, for example, naphthalene, indane, and 1, 2, 3, 4-tetrahydroquinoline; and tricyclic ring systems wherein at least one ring is carbocyclic and aromatic, for example, fluorene. In cases where the aryl substituent is  bicyclic or tricyclic and at least one ring is non-aromatic, it is understood that attachment is via the aromatic ring.
For example, aryl includes 5-and 6-membered carbocyclic aromatic rings fused to a 5-to 7-membered heterocyclic ring containing one or more heteroatoms selected from N, O, and S, provided that the point of attachment is at the carbocyclic aromatic ring. Bivalent radicals formed from substituted benzene derivatives and having the free valences at ring atoms are named as substituted phenylene radicals. Bivalent radicals derived from univalent polycyclic hydrocarbon radicals whose names end in "-yl" by removal of one hydrogen atom from the carbon atom with the free valence are named by adding "-idene" to the name of the corresponding univalent radical, e.g., a naphthyl group with two points of attachment is termed naphthylidene. Aryl, however, does not encompass or overlap in any way with heteroaryl, separately defined below. Hence, if one or more carbocyclic aromatic rings are fused with a heterocyclic aromatic ring, the resulting ring system is heteroaryl, not aryl, as defined herein.
The term "halogen" (or "halo" ) refers to fluorine, chlorine, bromine and iodine.
The term “heteroaryl” refers to
5-to 8-membered aromatic, monocyclic rings containing one or more, for example, from 1 to 4, or, in some embodiments, from 1 to 3, heteroatoms selected from N, O, and S, with the remaining ring atoms being carbon;
8-to 12-membered bicyclic rings containing one or more, for example, from 1 to 4, or, in some embodiments, from 1 to 3, heteroatoms selected from N, O, and S, with the remaining ring atoms being carbon and wherein at least one heteroatom is present in an aromatic ring; and
11-to 14-membered tricyclic rings containing one or more, for example, from 1 to 4, or in some embodiments, from 1 to 3, heteroatoms selected from N, O, and S, with the remaining ring atoms being carbon and wherein at least one heteroatom is present in an aromatic ring.
When the total number of S and O atoms in the heteroaryl group exceeds 1, those heteroatoms are not adjacent to one another. In some embodiments, the total number of S and O  atoms in the heteroaryl group is not more than 2. In some embodiments, the total number of S and O atoms in the aromatic heterocycle is not more than 1.
Examples of heteroaryl groups include, but are not limited to, (as numbered from the linkage position assigned priority 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, thiadiazolyl, tetrazolyl, thienyl, benzothienyl, furyl, benzofuryl, benzoimidazolinyl, indolinyl, pyridizinyl, triazolyl, quinolinyl, pyrazolyl, and 5, 6, 7, 8-tetrahydroisoquinoline.
Further heteroaryl groups include but are not limited to pyrrolyl, isothiazolyl, triazinyl, pyrazinyl, pyridazinyl, indolyl, benzotriazolyl, quinoxalinyl, and isoquinolinyl. As with the definition of heterocycle below, "heteroaryl" is also understood to include the N-oxide derivative of any nitrogen-containing heteroaryl.
Bivalent radicals derived from univalent heteroaryl radicals whose names end in "-yl" by removal of one hydrogen atom from the atom with the free valence are named by adding "-idene" to the name of the corresponding univalent radical, e.g., a pyridyl group with two points of attachment is a pyridylidene. Heteroaryl does not encompass or overlap with aryl as defined above.
In cases where the heteroaryl substituent is bicyclic or tricyclic and at least one ring is non-aromatic or contains no heteroatoms, it is understood that attachment is via the aromatic ring or via the heteroatom containing ring, respectively.
The term "heterocycle" (and variations thereof such as "heterocyclic" , or "heterocyclyl" ) broadly refers to a single aliphatic ring, usually with 3 to 12 ring atoms, containing at least 2 carbon atoms in addition to one or more, preferably one to three heteroatoms independently selected from oxygen, sulfur, and nitrogen, as well as combinations comprising at least one of the foregoing heteroatoms. Alternatively , a heterocycle as defined above may be multicyclic ring system (e.g. bicyclic) in which two or more rings may be fused or bridged together, wherein at least one such ring contains one or more heteroatoms independently selected from oxygen, sulfur, and nitrogen. “Heterocycle” also refers to 5-to 7-membered heterocyclic ring containing one or more heteroatoms  selected from N, O, and S fused with 5-and 6-membered carbocyclic aromatic ring, provided that the point of attachment is at the heterocyclic ring. The rings may be saturated or have one or more double bonds (i.e. partially unsaturated) . The heterocycle can be substituted by oxo. The point of the attachment may be carbon or heteroatom in the heterocyclic ring, provided that attachment results in the creation of a stable structure. When the heterocyclic ring has substituents, it is understood that the substituents may be attached to any atom in the ring, whether a heteroatom or a carbon atom, provided that a stable chemical structure results. Heterocycle does not overlap with heteroaryl.
Suitable heterocycles include, for example (as numbered from the linkage position assigned priority 1) , 1-pyrrolidinyl, 2-pyrrolidinyl, 2, 4-imidazolidinyl, 2, 3-pyrazolidinyl, 1-piperidinyl, 2-piperidinyl, 3-piperidinyl, 4-piperidinyl, and 2, 5-piperazinyl. Morpholinyl groups are also contemplated, including 2-morpholinyl and 3-morpholinyl (numbered wherein the oxygen is assigned priority 1) . Substituted heterocycle also includes ring systems substituted with one or more oxo moieties, such as piperidinyl N-oxide, morpholinyl-N-oxide, 1-oxo-1-thiomorpholinyl and 1, 1-dioxo-1-thiomorpholinyl. Bicyclic heterocycles include, for example:
Figure PCTCN2015081332-appb-000002
Figure PCTCN2015081332-appb-000003
As used herein, “arylalkyl” refers to an alkyl moiety substituted by an aryl group. Example arylalkyl groups include benzyl, phenethyl, and naphthylmethyl groups. In some embodiments, arylalkyl groups have from 7 to 20 or 7 to 11 carbon atoms. When used in the phrase "arylCl-4alkyl” , the term "C1-4” refers to the alkyl portion of the moiety and does not describe the number of atoms in the aryl portion of the moiety. Likewise, when used in the phrase "arylC1-10alkyl” , the term "C1-10” refers to the alkyl portion of the moiety and does not describe the number of atoms in the aryl portion of the moiety.
As used herein, “heterocyclylalkyl” refers to alkyl substituted by heterocyclyl. When used in the phrase "heterocyclyl-C1-6alkyl” , the term "C1-6” refers to the alkyl portion of the moiety and does not describe the number of atoms in the heterocyclyl portion of the moiety.
As used herein, "cycloalkylalkyl" refers to alkyl substituted by cycloalkyl. When used in the phrase "C3-10cycloalkylalkyl” , the term "C3-10” refers to the cycloalkyl portion of the moiety and does not describe the number of atoms in the alkyl portion of the moiety. When used in the phrase "C3-7cycloalkylalkyl” , the term "C3-7” refers to the cycloalkyl portion of the moiety and does not describe the number of atoms in the alkyl portion of the moiety. When used in the phrase "C3-8cycloalkylalkyl” , the term "C3-8” refers to the cycloalkyl portion of the moiety and does not describe the number of atoms in the alkyl portion of the moiety. When used in the phrase "cycloalkyl-Cl-10alkyl” , the term "C1-10” refers to the alkyl portion of the moiety and does not describe the number of atoms in the cycloalkyl portion of the moiety.
As used herein, "heteroarylalkyl" refers to alkyl substituted by heteroaryl. When used in the phrase "heteroaryl-Cl-4alkyl” , the term "C1-4” refers to the alkyl portion of the moiety and does not describe the number of atoms in the heteroaryl portion of the moiety. Likewise, when used in the phrase "heteroaryl-Cl-10 alkyl” , the term "C1-10” refers to the alkyl portion of the moiety and does not describe the number of atoms in the heteroaryl portion of the moiety.
For avoidance of doubt, reference, for example, to substitution of alkyl, cycloalkyl, heterocyclyl, aryl, and/or heteroaryl refers to substitution of each of those groups individually as well as to substitutions of combinations of those groups. That is, if R1 is arylalkyl, the aryl portion may be unsubstituted or substituted with at least one substituent, such as one, two, three, or four substituents, independently selected from R6b and the alkyl portion may also be unsubstituted or substituted with at least one substituent, such as one, two, three, or four substituens, independently selected from R6a.
The term "pharmaceutically acceptable salts" refers to salts prepared from pharmaceutically acceptable non-toxic bases or acids including inorganic or organic bases and inorganic or organic acids. Salts derived from inorganic bases may be selected, for example, from aluminum, ammonium, calcium, copper, ferric, ferrous, lithium, magnesium, manganic, manganous, potassium, sodium, and zinc salts. Further, for example, the pharmaceutically acceptable salts derived from inorganic bases may be selected from ammonium, calcium, magnesium, potassium, and sodium salts. Salts in the solid form may exist in one or more crystal structures, and may also be in the form of hydrates. Salts derived from pharmaceutically acceptable organic non-toxic bases may be selected, for example, from salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, and basic ion exchange resins, such as arginine, betaine, caffeine, choline, N, N'-dibenzylethylene-diamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethyl-morpholine, N-ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylamine, lysine, methylglucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine, and tripropylamine, tromethamine.
When the compound disclosed herein is basic, salts may be prepared using at least one pharmaceutically acceptable non-toxic acid, selected from inorganic and organic acids. Such acid may be selected, for example, from acetic, benzenesulfonic, benzoic, camphorsulfonic, citric, ethanesulfonic, fumaric, gluconic, glutamic, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, nitric, pamoic, pantothenic, phosphoric, succinic, sulfuric, tartaric, and p-toluenesulfonic acids. In some embodiments, such acid may be selected, for example, from citric, hydrobromic, hydrochloric, maleic, phosphoric, sulfuric, fumaric, and tartaric acids.
The term "protecting group" or "Pg" refers to a substituent that can be commonly employed to block or protect a certain functionality while reacting other functional groups on the compound. For example, an "amino-protecting group" is a substituent attached to an amino group that blocks or protects the amino functionality in the compound. Suitable amino-protecting groups include but are not limited to acetyl, trifluoroacetyl, t-butoxycarbonyl (BOC) , benzyloxycarbonyl (CBZ) and 9-fluorenylmethylenoxycarbonyl (Fmoc) . Similarly, a "hydroxy-protecting group" refers to a substituent of a hydroxy group that blocks or protects the hydroxy functionality. Suitable protecting groups include but are not limited to acetyl and silyl. A "carboxy-protecting group" refers to a substituent of the carboxy group that blocks or protects the carboxy functionality. Common carboxy-protecting groups include --CH2CH2SO2Ph, cyanoethyl, 2- (trimethylsilyl) ethyl, 2- (trimethylsilyl) ethoxymethyl, 2- (p-toluenesulfonyl) ethyl, 2- (p-nitrophenylsulfenyl) ethyl, 2- (diphenylphosphino) -ethyl, nitroethyl and the like. For a general description of protecting groups and their use, see T. W. Greene, Protective Groups in Organic Synthesis, John Wiley &Sons, New York, 1991.
The terms "administration of" and or "administering" at least one compound and/or at least one pharmaceutically acceptable salt should be understood to mean providing at least one compound and/or at least one pharmaceutically acceptable salt thereof to the individual in recognized need of treatment.
The term "effective amount" means the amount of the at least one compound and/or at least one pharmaceutically acceptable salt that will elicit the biological or medical response of a tissue,  system, animal or human that is being sought by the researcher, veterinarian, medical doctor or other clinician.
The term "composition" as used herein is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts. Such term in relation to a pharmaceutical composition is intended to encompass a product comprising the active ingredient (s) , and the inert ingredient (s) that make up the carrier, as well as any product which results, directly or indirectly, from combination, complexation or aggregation of any two or more of the ingredients, or from dissociation of one or more of the ingredients, or from other types of reactions or interactions of one or more of the ingredients.
The term "pharmaceutically acceptable" it is meant compatible with the other ingredients of the formulation and not unacceptably deleterious to the recipient thereof.
1. Disclosed herein is at least one compound of formula (I) :
Figure PCTCN2015081332-appb-000004
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;
R1 is selected from hydrogen, C1-10alkyl, C3-10cycloalkyl, C3-10cycloalkyl-C1-4alkyl, heterocyclyl, and heterocyclyl-C1-4alkyl, wherein alkyl, cycloalkyl, and heterocyclyl are each unsubstituted or substituted with at least one substituent, such as one, two, three, or four substituents, independently selected from R6a
R1’is selected from hydrogen and halogen;
R2 and R3 are independently selected from hydrogen, halogen, hydroxyl, CN, C1-10alkyl, C2-10alkenyl, C2-10alkynyl, C3-10cycloalkyl, C3-10cycloalkyl-C1-4alkyl, heterocyclyl, heterocyclyl-C1-4alkyl, aryl, aryl-C1-4alkyl, heteroaryl, and heteroaryl-C1-4alkyl, wherein alkyl, alkenyl, alkynyl, cycloalkyl, and heterocyclyl are each unsubstituted or substituted with at least one substituent, such as one, two, three, or four substituents independently selected from R6a, and each aryl and heteroaryl is unsubstituted or substituted with at least one substituent, such as one, two, three, or four substituents, independently selected from R6b; or R2 and R3 together with the carbon atoms 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, and optionally substituted with 1or 2 R6a groups;
each R4 is independently selected from hydrogen, C1-10alkyl, C2-10alkenyl, 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 (CR9R10tNR7R8, -C (O) R7, -CO2R8, -CO2 (CR9R10tCONR7R8, -OC (O) R7, -CN, -C (O) NR7R8, -NR7C (O) R8, -OC (O) NR7R8, -NR7C (O) OR8, -NR7C (O) NR7R8, and -CR7 (N-OR8) , wherein C1-10alkyl, C2-10alkenyl, C2-10alkynyl, and C3-10cycloalkyl are each unsubstituted or substituted with at least one substituent, such as one, two, three, or four substituents, independently selected from R6a
each R5 is independently selected from hydrogen, C1-10alkyl, C2-10alkenyl, C2-10alkynyl, C3-10cycloalkyl, C3-10cycloalkyl-C1-4alkyl, heterocyclyl, heterocyclyl-C1-4alkyl, aryl, aryl-C1-4alkyl, heteroaryl, and heteroaryl-C1-4alkyl, -OR8, -NR7S (O) rR8, -NO2, halogen, -S (O) rR7, -SR8, -S (O) 2OR7, -OS (O) 2R8, -S (O) rNR7R8, -NR7R8, -O (CR9R10tNR7R8, -C (O) R7, -CO2R8, -CO2 (CR9R10tCONR7R8, -OC (O) R7, -CN, -C (O) NR7R8, -NR7C (O) R8, -OC (O) NR7R8, -NR7C (O) OR8, -NR7C (O) NR7R8, and  -CR7 (N-OR8) , wherein C1-10alkyl, C2-10alkenyl, C2-10alkynyl, C3-10cycloalkyl and heterocyclyl are each unsubstituted or substituted with at least one substituent, such as one, two, three, or four substituents independently selected from R6a, and each aryl and heteroaryl is unsubstituted or substituted with at least one substituent, such as one, two, three, or four substituents, independently selected from R6b
each R6a is independently selected from C1-10alkyl, C2-10alkenyl, C2-10alkynyl, C3-10cycloalkyl, -OR8, -NR7S (O) rR8, -NO2, halogen, -S (O) rR7, -SR8, -S (O) 2OR7, -OS (O) 2R8, -S (O) rNR7R8, -NR7R8, - (CR9R10tOR8, - (CR9R10tNR7R8, - (CR9R10tSR8, - (CR9R10tS (O) rR8, - (CR9R10tCO2R8, - (CR9R10tCONR7R8, - (CR9R10tNR7CO2R8, - (CR9R10tOCONR7R8, - (CR9R10tNR7CONR7R8, - (CR9R10tNR7SO2NR7R8, -O (CR9R10tNR7R8, -C (O) R7, -C (O) (CR9R10tOR8, -C (O) (CR9R10tNR7R8, -C (O) (CR9R10tSR8, -C (O) (CR9R10tS (O) rR8, -CO2R8, -CO2 (CR9R10tCONR7R8, -OC (O) R7, -CN, -C (O) NR7R8, -NR7C (O) R8, -OC (O) NR7R8, -NR7C (O) OR8, -NR7C (O) NR7R8, and -CR7 (N-OR8) ;
each R6b is independently selected from R6a, aryl, aryl-C1-4alkyl, heteroaryl, and heteroaryl-C1-4alkyl;
each R7 and each R8 are independently selected from hydrogen, C1-10alkyl, C2-10alkenyl, C2-10alkynyl, cycloalkyl, cycloalkyl-C1-4alkyl, heterocyclyl, heterocyclyl-C1-4alkyl, aryl, aryl-C1-4alkyl, heteroaryl, and heteroaryl-C1-4alkyl, wherein alkyl, alkenyl, alkynyl, cycloalkyl, and heterocyclyl are each unsubstituted or substituted with at least one substituent, such as one, two, three, or four substituents independently selected from R6a, and aryl and heteroaryl are each unsubstituted or substituted with at least one substituent, such as one, two, three, or four substituents independently selected from R6b; or R7 and R8 together with the atom (s) to which they are attached form a heterocyclic ring of 4 to 12 members containing 0, 1, or 2 additional heteroatoms independently selected from oxygen, sulfur and nitrogen, and optionally substituted with 1or 2 R6b groups;
each R9 and each R10 are independently selected from hydrogen, C1-10alkyl, C2-10alkenyl, C2-10alkynyl, cycloalkyl, cycloalkyl-C1-4alkyl, heterocyclyl, heterocyclyl-C1-4alkyl, aryl, aryl-C1-4alkyl, heteroaryl, and heteroaryl-C1-4alkyl; or R9 and R10 together with the carbon atom (s) to  which they are attached form a ring of 3 to 7 members containing 0, 1, or 2 heteroatoms independently selected from oxygen, sulfur and nitrogen, and optionally substituted with 1or 2 R6a groups;
m is independently selected from 0, 1, 2, and 3;
n is independently selected from 0, 1, 2, and 3;
each r is independently selected from 1 and 2;
each t is independently selected from 1, 2, and 3.
2. At least one compound of 1 and/or at least one pharmaceutically acceptable salt thereof, wherein X is selected from N and C, and Y is C.
3. At least one compound of any one of 1-2 and/or at least one pharmaceutically acceptable salt thereof, wherein X is N, and Y is C.
4. At least one compound of any one of 1-2 and/or at least one pharmaceutically acceptable salt thereof, wherein: when both X and Y are C, and
n = 2, one R5 is (3R) -3- (dimethylamino) pyrrolidin-1-yl, (3S) -3- (dimethylamino) pyrrolidin-1-yl, 3- (dimethylamino) azetidin-1-yl, [2- (dimethylamino) ethyl] - (methyl) amino, [2- (methylamino) ethyl] (methyl) amino, 5-methyl-2, 5-diazaspiro [3.4] oct-2-yl, (3aR, 6aR) -5-methylhexa-hydro-pyrrolo [3, 4-b] pyrrol-1 (2H) -yl, 1-methyl-1, 2, 3, 6-tetrahydropyridin-4-yl, 4-methylpiperizin-1-yl, 4- [2- (dimethylamino) -2-oxoethyl] 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-aminopropanoyl] piperazin-1-yl, the other R5 is methoxy or methyl,
R1, R1’a nd R2 are hydrogen, and R3 is H, F, Cl, CH3 or CN,
Q is not 4, 5, 6, 7-tetrahydropyrazolo [1, 5-a] pyridine-3-yl, pyrazolo [1, 5-a] pyridin-3-yl or 1H-indol-3-yl.
5. At least one compound of any one of 1 to 4 and/or at least one pharmaceutically acceptable salt thereof, wherein the formula is
Figure PCTCN2015081332-appb-000005
6. At least one compound of any one of 1 to 5 and/or at least one pharmaceutically acceptable salt thereof, wherein R1’ is hydrogen.
7. At least one compound of any one of 1 to 5 and/or at least one pharmaceutically acceptable salt thereof, wherein R1’ is selected from halogen, preferably R1’ is fluorine.
8. At least one compound of any one of 1 to 7 and/or at least one pharmaceutically acceptable salt thereof, wherein R1 is hydrogen.
9. At least one compound of any one of 1 to 8 and/or at least one pharmaceutically acceptable salt thereof, wherein R2 is hydrogen.
10. At least one compound of any one of 1 to 9 and/or at least one pharmaceutically acceptable salt thereof, wherein R3 is selected from hydrogen, halogen, and C1-10 alkyl.
11. At least one compound of 10 and/or at least one pharmaceutically acceptable salt thereof, wherein R3 is hydrogen.
12. At least one compound of any one of 1 to11 and/or at least one pharmaceutically acceptable salt thereof, wherein Q is selected from heteroaryl.
13. At least one compound of 12 and/or at least one pharmaceutically acceptable salt thereof, wherein Q is selected from
Figure PCTCN2015081332-appb-000006
14. At least one compound of any one of 1 to 13 and/or at least one pharmaceutically acceptable salt thereof, wherein R4 is selected from hydrogen, C1-10 alkyl, halogen and CN, wherein C1-10 alkyl are unsubstituted or substituted with at least one substituent, such as one, two, three, or four substituents, independently selected from R6a.
15. At least one compound of 14 and/or at least one pharmaceutically acceptable salt thereof, wherein R4 is selected from hydrogen, methyl, halogen and CN, wherein methyl are unsubstituted or substituted with at least one substituent independently selected from R6a, preferably R6a is fluorine.
16. At least one compound of any one of 1 to 15 and/or at least one pharmaceutically acceptable salt thereof, wherein R5 is independently selected from OR8, heterocyclyl, NR7R8, wherein heterocyclyl is unsubstituted or substituted with at least one substituent, such as one, two, three, or four substituents, independently selected from R6a.
17. At least one compound of 16 and/or at least one pharmaceutically acceptable salt thereof, wherein R5 is independently 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-methylacetamido) ethyl) amino) .
18. At least one compound, 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- (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-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-methylpiperazin-1 -yl) pyridin-3-yl) acrylamide,
N- (6-ethoxy-5- ( (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- (piperazin-1-yl) pyridin -3-yl) acrylamide,
N- (6-ethoxy-5- ( (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- (4-methylpiperazin-1-yl) pyridin-3-yl) acrylamide,
N- (6-isopropoxy-5- ( (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- (piperazin-1-yl) pyridin-3-yl) acrylamide,
N- (6-isopropoxy-5- ( (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- (4-methylpiperazin-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-methylindolizin-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide,
N- (2- ( (2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5- ( (4- (1-methylindolizin-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-methylindolizin-1-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide,
N- (2- ( (2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5- ( (4- (3-methylindolizin-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] pyridin-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] pyridin-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-methylimidazo [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-methylimidazo [1, 5-a] pyridin-3-yl) pyrimidin-2-yl) amino) phenyl) acrylamide,
N- (2- ( (2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ( (4- (1-methylimidazo [1, 5-a] pyridin-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-methylimidazo [1, 5-a] pyridin-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] pyridin-1-yl) pyrimidin-2-yl) amino) pyridin-3-yl) -2-fluoroacrylamide,
and pharmaceutically acceptable salts thereof.
In another of its aspects, there is provided a pharmaceutical composition comprising at least one compound of any one of 1-18 and pharmaceutically acceptable salts thereof, wherein the composition is adapted for administration by a route selected from the group consisting of orally, parenterally, intraperitoneally, intravenously, intraarterially, transdermally, sublingually, intramuscularly, rectally, transbuccally, intranasally, liposomally, via inhalation, vaginally, intraoccularly, via local delivery (for example by catheter or stent) , subcutaneously, intraadiposally, intraarticularly, and intrathecally.
In yet another of its aspects, there is provided a kit comprising at least one compound of any one of 1-18 and pharmaceutically acceptable salts thereof, and instructions which comprise one or more forms of information selected from the group consisting of indicating a disease state for which the composition is to be administered, storage information for the composition, dosing information and instructions regarding how to administer the composition. In one particular variation, the kit comprises the compound in a multiple dose form.
In still another of its aspects, there is provided an article of manufacture comprising at least one compound of any one of 1-18 and pharmaceutically acceptable salts thereof, and packaging materials. In one variation, the packaging material comprises a container for housing the compound. In one particular variation, the container comprises a label indicating one or more members of the group consisting of a disease state for which the compound is to be administered, storage information, dosing information and/or instructions regarding how to administer the compound. In another variation, the article of manufacture comprises the compound in a multiple dose form.
In a further of its aspects, there is provided a therapeutic method comprising administering to a subject at least one compound of any one of 1-18 and pharmaceutically acceptable salts thereof.
In another of its aspects, there is provided a method of inhibiting a mutant EGFR kinase comprising contacting the mutant EGFR with at least one compound of any one of 1-18 and pharmaceutically acceptable salts thereof.
In yet another of its aspects, there is provided a method of inhibiting a mutant EGFR comprising causing at least one compound of any one of 1-18 and pharmaceutically acceptable salts thereof to be present in a subject in order to inhibit the mutant EGFR in vivo.
In another of its aspects, there is provided a method of treating a disease state for which a mutant EGFR possesses activity that contributes to the pathology and/or symptomology of the disease state, the method comprising causing at least one compound of any one of 1-18 and pharmaceutically acceptable salts thereof to be present in a subject in a therapeutically effective amount for the disease state.
In one variation of each of the above methods the disease state is selected from the group consisting of cancerous hyperproliferative disorders (e.g., brain, lung, squamous cell, bladder, gastric, pancreatic, breast, head, neck, renal, kidney, ovarian, prostate, colorectal, epidermoid, esophageal, testicular, gynecological or thyroid cancer) ; non-cancerous hyperproliferative disorders (e.g., benign hyperplasia of the skin (e.g., psoriasis) , restenosis, and benign prostatic hypertrophy (BPH) ) ; pancreatitis; kidney disease; pain; preventing blastocyte implantation; treating diseases related to vasculogenesis or angiogenesis (e.g., tumor angiogenesis, acute and chronic inflammatory disease such as rheumatoid arthritis, atherosclerosis, inflammatory bowel disease, skin diseases such as psoriasis, exzema, and scleroderma, diabetes, diabetic retinopathy, retinopathy of prematurity, age-related macular degeneration, hemangioma, glioma, melanoma, Kaposi's sarcoma and ovarian, breast, lung, pancreatic, prostate, colon and epidermoid cancer) ; asthma; neutrophil chemotaxis (e.g., reperfusion injury in myocardial infarction and stroke and inflammatory arthritis) ; septic shock; T-cell mediated diseases where immune suppression would be of value (e.g., the prevention of organ transplant rejection, graft versus host disease, lupus erythematosus, multiple sclerosis, and rheumatoid arthritis) ; atherosclerosis; inhibition of keratinocyte responses to growth factor cocktails; chronic obstructive pulmonary disease (COPD) and other diseases.
In another of its aspects, there is provided a method of treating a disease state for which a mutation in the mutant EGFR gene contributes to the pathology and/or symptomology of the disease state including, for example, melanomas, lung cancer, colon cancer and other tumor types.
In still another of its aspects, the present invention relates to the use of at least one compound of any one of 1-18 and pharmaceutically acceptable salts thereof as a medicament. In yet another of its aspects, the present invention relates to the use of at least one compound of any one of 1-18 and pharmaceutically acceptable salts thereof in the manufacture of a medicament for inhibiting a mutant EGFR.
In a further of its aspects, the present invention relates to the use of at least one compound of any one of 1-18 and pharmaceutically acceptable salts thereof in the manufacture of a medicament for treating a disease state for which a mutant EGFR possesses activity that contributes to the pathology and/or symptomology of the disease state.
Administration and Pharmaceutical Compositions
In general, compounds of the disclosure will be administered in therapeutically effective amounts via any of the usual and acceptable modes known in the art, either singly or in combination with one or more therapeutic agents. A therapeutically effective amount may vary widely depending on the severity of the disease, the age and relative health of the subject, the potency of the compound used and other factors known to those of ordinary skill in the art. For example, for the treatment of neoplastic diseases and immune system disorders, the required dosage will also vary depending on the mode of administration, the particular condition to be treated and the effect desired.
In general, satisfactory results are indicated to be obtained systemically at daily dosages of from about 0.001 to about 100 mg/kg per body weight, or particularly, from about 0.03 to 2.5 mg/kg per body weight. An indicated daily dosage in the larger mammal, e. g. humans, may be in the range from about 0.5 mg to about 2000 mg, or more particularly, from about 0.5 mg to about 1000 mg,  conveniently administered, for example, in divided doses up to four times a day or in retard form. Suitable unit dosage forms for oral administration comprise from ca. 1 to 50 mg active ingredient.
Compounds of the disclosure may be administered as pharmaceutical compositions by any conventional route; for example, 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 lotions, gels, ointments or creams, or in a nasal or suppository form.
Pharmaceutical compositions comprising a compound of the present disclosure in free form or in a pharmaceutically acceptable salt form in association with at least one pharmaceutically acceptable carrier or diluent may be manufactured in a conventional manner by mixing, granulating, coating, dissolving or lyophilizing processes. For example, pharmaceutical compositions comprising a compound of the disclosure in association with at least one pharmaceutical acceptable carrier or diluent may be manufactured in conventional manner by mixing with a pharmaceutically acceptable carrier or diluent. Unit dosage forms for oral administration contain, for example, from about 0.1 mg to about 500 mg of active substance.
In one embodiment, the pharmaceutical compositions are solutions of the active ingredient, including suspensions or dispersions, such as isotonic aqueous solutions. In the case of lyophilized compositions comprising the active ingredient alone or together with a carrier such as mannitol, dispersions or suspensions can be made up before 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, or microbicides, such as sorbic acid or benzoic acid. The solutions or suspensions may further comprise viscosity-increasing agents, including but not limited to, sodium carboxymethylcellulose, carboxymethylcellulose, dextran, polyvinylpyrrolidone, gelatins, or solubilizers, e.g. Tween 80 (polyoxyethylene (20) sorbitan mono-oleate) .
Suspensions in oil may comprise as the oil component the vegetable, synthetic, or semi-synthetic oils customary for injection purposes. Examples include liquid fatty acid esters that  contain as the acid component a long-chained 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, for example oleic acid, elaidic acid, erucic acid, brassidic acid and linoleic acid, and if desired, may contain antioxidants, for example vitamin E, 3-carotene or 3, 5-di-tert-butyl-hydroxytoluene. The alcohol component of these fatty acid esters may have six carbon atoms and may be monovalent or polyvalent, for example a mono-, di-or trivalent, alcohol. Suitable alcohol components include but are not limited to methanol, ethanol, propanol, butanol or pentanol or isomers thereof; glycol and glycerol.
Other suitable fatty acid esters include but are not limited ethyl-oleate, isopropyl myristate, isopropyl palmitate,
Figure PCTCN2015081332-appb-000007
M 2375, (polyoxyethylene glycerol) ,
Figure PCTCN2015081332-appb-000008
M 1944 CS (unsaturated polyglycolized glycerides prepared by alcoholysis of apricot kernel oil and comprising glycerides and polyethylene glycol ester) , LABRASOLTM (saturated polyglycolized glycerides prepared by alcoholysis of TCM and comprising glycerides and polyethylene glycol ester; all available from GaKefosse, France) , and/or
Figure PCTCN2015081332-appb-000009
812 (triglyceride of saturated fatty acids of chain length C8 to C12 from Hüls AG, Germany) , and vegetable oils such as cottonseed oil, almond oil, olive oil, castor oil, sesame oil, soybean oil, or groundnut oil.
Pharmaceutical compositions for oral administration may be obtained, for example, by combining the active ingredient with one or more solid carriers, and if desired, granulating a resulting mixture, and processing the mixture or granules by the inclusion of additional excipients, to form tablets or tablet cores.
Suitable carriers include but are not limited to fillers, such as sugars, for example lactose, saccharose, mannitol or sorbitol, cellulose preparations, and/or calcium phosphates, for example tricalcium phosphate or calcium hydrogen phosphate, and also binders, such as starches, for example corn, wheat, rice or potato starch, methylcellulose, hydroxypropyl methylcellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone, and/or, if desired, disintegrators, such as the above-mentioned starches, carboxymethyl starch, crosslinked polyvinylpyrrolidone, alginic acid or a  salt thereof, such as sodium alginate. Additional excipients include flow conditioners 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.
Tablet cores may be provided with suitable, optionally enteric, coatings through the use of, inter alia, concentrated sugar solutions which may comprise gum arable, talc, polyvinylpyrrolidone, polyethylene glycol and/or titanium dioxide, or coating solutions in suitable organic solvents or solvent mixtures, or, for the preparation of enteric coatings, solutions of suitable cellulose preparations, such as 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 active ingredient.
Pharmaceutical compositions for oral administration may also include hard capsules comprising gelatin or soft-sealed capsules comprising gelatin and a plasticizer, such as glycerol or sorbitol. The hard capsules may contain the active ingredient 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 oil or liquid polyethylene glycols or fatty acid esters of ethylene or propylene glycol, to which stabilizers and detergents, for example of the polyoxyethylene sorbitan fatty acid ester type, may also be added.
Pharmaceutical compositions suitable for rectal administration are, for example, suppositories comprising 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 aqueous solutions of an active ingredient in water-soluble form, for example of a water-soluble salt, or aqueous injection suspensions that contain viscosity-increasing substances, for example sodium carboxymethylcellulose, sorbitol and/or dextran, and, if desired, stabilizers. The active ingredient, optionally together with excipients, can also be in the form of a lyophilizate and can be made into a  solution before parenteral administration by the addition of suitable solvents. Solutions such as are used, for example, for parenteral administration can also be employed as infusion solutions. The manufacture of injectable preparations is usually carried out under sterile conditions, as is the filling, for example, into ampoules or vials, and the sealing of the containers.
The compounds of the disclosure may be administered as the sole active ingredient, or together with other drugs useful against neoplastic diseases or useful in immunomodulating regimens. For example, the compounds of the disclosure may be used in accordance with the disclosure in combination with pharmaceutical compositions effective in various diseases as described above, e.g. with cyclophosphamide, 5-fluorouracil, fludarabine, gemcitabine, cisplatinum, carboplatin, vincristine, vinblastine, etoposide, irinotecan, paclitaxel, docetaxel, rituxan, doxorubicine, gefitinib, or imatinib; or also with cyclosporins, rapamycins, ascomycins or their immunosuppressive analogs, e.g. cyclosporin A, cyclosporin G, FK-506, sirolimus or everolimus, corticosteroids, e.g. prednisone, cyclophosphamide, azathioprene, methotrexate, gold salts, sulfasalazine, antimalarials, brequinar, leflunomide, mizoribine, mycophenolic acid, mycophenolate, mofetil, 15-deoxyspergualine, immuno-suppressive monoclonal antibodies, e.g. monoclonal antibodies to leukocyte receptors, e.g. 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, e.g. CTLA41g.
The disclosure also provides for a pharmaceutical combinations, e.g. a kit, comprising a) a first agent which is a compound of the disclosure as disclosed herein, in free form or in pharmaceutically acceptable salt form, and b) at least one co-agent. The kit can comprise instructions for its administration.
EXAMPLES
Various methods may be developed for synthesizing the at least one compound of formula (I) and/or at least one pharmaceutically acceptable salt thereof. Representative methods for  synthesizing the at least one compound of formula (I) and/or at least one pharmaceutically acceptable salt thereof are provided in the Examples. It is noted, however, that the at least one compound of formula (I) and/or at least one pharmaceutically acceptable salt thereof may also be synthesized by other synthetic routes that others may devise.
It will be readily recognized that certain compounds of formula (I) have atoms with linkages to other atoms that confer a particular stereochemistry to the compound (e.g., chiral centers) . It is recognized that synthesis of the at least one compound of formula (I) and/or at least one pharmaceutically acceptable salt thereof may result in the creation of mixtures of different stereoisomers (enantiomers, diastereomers) . Unless a particular stereochemistry is specified, recitation of a compound is intended to encompass all of the different possible stereoisomers.
The at least one compound of formula (I) can also be prepared as a pharmaceutically acceptable acid addition salt by, for example, reacting the free base form of the at least one compound with a pharmaceutically acceptable inorganic or organic acid. Alternatively, a pharmaceutically acceptable base addition salt of the at least one compound of formula (I) can be prepared by, for example, reacting the free acid form of the at least one compound with a pharmaceutically acceptable inorganic or organic base. Inorganic and organic acids and bases suitable for the preparation of the pharmaceutically acceptable salts of compounds of formula (I) are set forth in the definitions section of this Application. Alternatively, the salt forms of the compounds of formula (I) can be prepared using salts of the starting materials or intermediates.
The free acid or free base forms of the compounds of formula (I) can be prepared from the corresponding base addition salt or acid addition salt form. For example, a compound of formula (I) in an acid addition salt form can be converted to the corresponding free base thereof by treating with a suitable base (e.g., ammonium hydroxide solution, sodium hydroxide, and the like) . A compound of formula (I) in a base addition salt form can be converted to the corresponding free acid thereof by, for example, treating with a suitable acid (e.g., hydrochloric acid, etc) .
The N-oxides of the at least one compound of formula (I) and/or at least one pharmaceutically acceptable salt thereof can be prepared by methods known to those of ordinary  skill in the art. For example, N-oxides can be prepared by treating an unoxidized form of the compound of formula (I) with an oxidizing agent (e.g., trifluoroperacetic acid, permaleic acid, perbenzoic acid, peracetic acid, meta-chloroperoxybenzoic acid, or the like) in a suitable inert organic solvent (e.g., a halogenated hydrocarbon such as dichloromethane) at approximately 0 to 80 ℃.Alternatively, the N-oxides of the compounds of formula (I) can be prepared from the N-oxideof an appropriate starting material.
Compounds of formula (I) in an unoxidized form can be prepared from N-oxides of compounds of formula (I) by, for example, treating with a reducing agent (e.g., sulfur, sulfur dioxide, triphenyl phosphine, lithium borohydride, sodium borohydride, phosphorus trichloride, tribromide, and the like) in an suitable inert organic solvent (e.g., acetonitrile, ethanol, aqueous dioxane, and the like) at 0 to 80 ℃.
Protected derivatives of the compounds of formula (I) can be made by methods known to those of ordinary skill in the art. A detailed description of the techniques applicable to the creation of protecting groups and their removal can be found in T.W. Greene, Protecting Groups in Organic Synthesis, 3rd edition, John Wiley &Sons, Inc. 1999.
As used herein the symbols and conventions used in these processes, schemes and examples are consistent with those used in the contemporary scientific literature, for example, the Journal of the American Chemical Society or the Journal of Biological Chemistry. Standard single-letter or three-letter abbreviations are generally used to designate amino acid residues, which are assumed to be in the L-configuration unless otherwise noted. Unless otherwise noted, all starting materials were obtained from commercial suppliers and used without further purification. For example, the following abbreviations may be used in the examples and throughout the specification: g (grams) ; mg (milligrams) ; L (liters) ; mL (milliliters) ; μL (microliters) ; psi (pounds per square inch) ; M (molar) ; mM (millimolar) ; i.v. (intravenous) ; Hz (Hertz) ; MHz (megahertz) ; mol (moles) ; mmol (millimoles) ; RT (room temperature) ; min (minutes) ; h (hours) ; mp (melting point) ; TLC (thin layer chromatography) ; Rt (retention time) ; RP (reverse 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-dimethoxyethane) ; DCM (dichloromethane) ; DCE (dichloroethane) ; DMF (N, N-dimethylformamide) ; DMPU (N, N'-dimethylpropyleneurea) ; 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 (atmosphere) ; TMSE (2- (trimethylsilyl) ethyl) ; TMS (trimethylsilyl) ; TIPS (triisopropylsilyl) ; TBS (t-butyldimethylsilyl) ; DMAP (4-dimethylaminopyridine) ; Me (methyl) ; OMe (methoxy) ; Et (ethyl) ; tBu (tert-butyl) ; HPLC (high pressure liquid chomatography) ; BOP (bis (2-oxo-3-oxazolidinyl) phosphinic chloride) ; TBAF (tetra-n-butylammonium fluoride) ; m-CPBA (meta-chloroperbenzoic acid) .
References to ether or Et2O are to diethyl ether; brine refers to a saturated aqueous solution of NaCl. Unless otherwise indicated, all temperatures are expressed in ℃ (degrees Centigrade) . All reactions were conducted under an inert atmosphere at RT unless otherwise noted.
1H NMR spectra were recorded on a Varian Mercury Plus 400. Chemical shifts are expressed in parts per million (ppm) . Coupling constants are in units of hertz (Hz) . Splitting patterns describe apparent multiplicities and are designated as s (singlet) , d (doublet) , t (triplet) , q (quartet) , m (multiplet) , and br (broad) .
Low-resolution mass spectra (MS) and compound purity data were acquired on a Shimadzu LC/MS single quadrapole system equipped with electrospray ionization (ESI) source, UV detector (220 and 254 nm) , and evaporative light scattering detector (ELSD) . Thin-layer chromatography was performed on 0.25 mm E. Merck silica gel plates (60F-254) , visualized with UV light, 5%ethanolic phosphomolybdic acid, ninhydrin, or p-anisaldehyde solution. Flash column chromatography was performed on silica gel (230-400 mesh, Merck) .
Synthetic Schemes
At least one compound of formula I and/or at least one pharmaceutically acceptable salt thereof may be synthesized according to a variety of reaction schemes. Some illustrative schemes are provided below and in the examples. Other reaction schemes could be readily devised by those skilled in the art in view of the present disclosure.
In the reactions described hereinafter it may be necessary to protect reactive functional groups, for example hydroxy, amino, imino, thio or carboxy groups, where these are desired in the final product, to avoid their unwanted participation in the reactions. Conventional protecting groups may be used in accordance with standard practice, for examples see T.W. Greene and P.G.M. Wuts in "Protective Groups in Organic Chemistry" John Wiley and Sons, 1991.
Synthetic methods for preparing the compounds of the present disclosure are illustrated in the following Schemes and Examples. Starting materials are commercially available or may be made according to procedures known in the art or as illustrated herein.
The intermediates shown in the following schemes are either known in the literature or may be prepared by a variety of methods familiar to those skilled in the art.
The compound of formula I of the present disclosure can be prepared as shown in Scheme 1. Coupling of intermediates such as those of formula III with amino-arene such as IV using such coupling conditions as Buchwald amination reaction or amination conditions known in the literature provides the intermediate VII or VI. Reduction of the nitro group in intermediate VII using conditions such as Fe/NH4Cl in a solvent or mixture of solvents such as EtOH and H2O provides amine VIII. Alternatively intermediate VIII can be prepared by cleavage of the protecting group on the amine of intermediate VI. intermediate VI can be obtained by the coupling between intermediate III and amino-arene V. Coupling of amine VIII with acid IX or acyl chloride X provides compound I. Compound of formula I can also be synthesized via a two-step sequence by reacting amine VIII with acyl chloride XI followed by elimination reaction in the presence of a base such as NaOH in a solvent such as H2O.
Figure PCTCN2015081332-appb-000010
An alternative approach to the key intermediate amine VIII is shown in Scheme 2. Coupling of amine XIII with halide XIV or XV under similar conditions as described in Scheme 1 or known in the literature provides nitro VII or intermediate VI, which can be further converted to amine VIII under conditions as described in Scheme 1.
Figure PCTCN2015081332-appb-000011
In some cases, compound of formula I can be prepared by coupling of intermediate III with intermediate VIa under the conditions as described in Scheme 1 or Scheme 2 as shown in Scheme 3.
Figure PCTCN2015081332-appb-000012
One preparation of VIaa is illustrated in Scheme 4. Starting from the commercially available dichloropyridine 1, intermediate VIaa can be prepared via a sequence of region-selective displacement of chloride, bromination, amination, deprotection, acylation and reduction.
Figure PCTCN2015081332-appb-000013
As an illustration of the preparation of intermediate XIV, an approach to XIVa is described in Scheme 5. Sequential reaction of commercial available dichloropyridine with amine and sodium alkoxide followed by halogenation provides intermediate XIVa.
Figure PCTCN2015081332-appb-000014
In some cases the order of carrying out the foregoing reaction schemes may be varied to facilitate the reaction or to avoid unwanted reaction products. The following examples are  provided so that the invention might be more fully understood. These examples are illustrative only and should not 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-indol-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 (5.79 g, 30.0 mmol) in dry THF (30 mL) at 0℃ was added methanol (1.15 mL, 28.5 mmol) followed NaH (60%, 1.20 g, 30.0 mmol) in 4 portions. The mixture was stirred at 0℃ for 1 h and then at r.t. for 1 h. The mixture was diluted with water (50 mL) and extracted with ethyl acetate (2 × 30 mL) . The extracts were washed with brine and dried over MgSO4. Solvent was evaporated, and the residue was crystallized from hexanes to give 6-Chloro-2-methoxy-3-nitropyridine (1a) as pale yellow solid (4.04 g, 71%) .
N1- (6-Methoxy-5-nitropyridin-2-yl) -N1, N2, N2-trimethylethane-1, 2-diamine (1b)
The solution of 6-Chloro-2-methoxy-3-nitropyridine (1a) (3.77 g, 20.0 mmol) and N1, N1, N2-trimethylethane-1, 2-diamine (2.24 g, 22.0 mmol) in ethanol (45 mL) was heated at reflux for 3 h. The solvent was evaporated under reduced pressure. The residue was purified by column chromatography on silica gel eluting with DCM/methanol/ammonia (92:6:2) to provide N1- (6-Methoxy-5-nitropyridin-2-yl) -N1, N2, N2-trimethylethane-1, 2-diamine (1b) (2.40 g, 47%) . MS-ESI (m/z) : 255 [M + 1] +.
N1- (3-Bromo-6-methoxy-5-nitropyridin-2-yl) -N1, N2, N2-trimethylethane-1, 2-diamine (1c)
To a suspension of N1- (6-Methoxy-5-nitropyridin-2-yl) -N1, N2, N2-trimethylethane-1, 2-diamine (1b) (1.33 g, 5.24 mmol) in acetonitrile (30 mL) at 0℃ was added NBS (1.40 g, 7.82 mmol) . The mixture was stirred at 0℃ for 1 h. Solvent was evaporated under reduced pressure. The residue was purified by chromatography on silica gel eluting with DCM/methanol/ammonia (92:6:2)  to give N1- (3-Bromo-6-methoxy-5-nitropyridin-2-yl) -N1, N2, N2-trimethylethane-1, 2-diamine (1c) as yellow solid (1.47 g, 84%) . MS-ESI (m/z) : 333 and 335 [1:1, M + 1] +.
N2- (2- (dimethylamino) ethyl) -N3- (diphenylmethylene) -6-methoxy-N2-methyl-5-nitropyri  dine-2, 3-diamine (1d)
The mixture of N1- (3-Bromo-6-methoxy-5-nitropyridin-2-yl) -N1, N2, N2-trimethylethane -1, 2-diamine (1c) (666 mg, 2.00 mmol) , Pd2 (dba) 3 (55 mg, 0.060 mmol) , (+/-) -BINAP (75 mg, 0.120 mmol) , benzophenone imine (471 mg, 2.60 mmol) , and t-BuONa (250 mg, 2.60 mmol) in toluene (8 mL) was heated under nitrogen at 110C for 2 h, and cooled to room temperature. The solid was removed by filtration through celite and washed with ethyl acetate. Solvents were evaporated under reduced pressure. The residue was purified by column chromatography on silica gel eluting with 20-40%ethyl acetate in hexanes to give N2- (2- (dimethylamino) ethyl) -N3-(diphenylmethylene) -6-methoxy-N2-methyl-5-nitropyridine-2, 3-diamine (1d) as pale yellow solid (227 mg, 26%) . MS-ESI (m/z) : 434 [M + 1] +.
N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-5-nitropyridine-2, 3-diamine (1e) 
To a solution of N2- (2- (dimethylamino) ethyl) -N3- (diphenylmethylene) -6-methoxy-N2-methyl-5-nitropyridine-2, 3-diamine (1d) (194 mg, 0.45 mmol) in methanol (5 mL) at r.t. was added concentrated hydroloric acid (1 mL) . The mixture was stirred at room temperature for 1 h. The mixture was diluted with NaHCO3 (aq. ) and extracted with ethyl acetate (3 ×) . The extracts were washed with water and brine, and dried over MgSO4. Solvents were evaporated under reduced pressure. The residue was purified by column chromatography on silica gel eluting with 3-10%methanol in DCM to give N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-5-nitropyridine -2, 3-diamine (1e) as yellow solid (12.4 mg, 10%) . MS-ESI (m/z) : 270 [M + 1] +.
N- (2- ( (2- (Dimethylamino) ethyl) (methyl) amino) -6-methoxy-5-nitropyridin-3-yl) acrylam ide (1f)
To a mixture of N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-5-nitropyridine -2, 3-diamine (1e) (12.4 mg, 0.046 mmol) in THF-H2O (10:1, 1 mL) at 0℃ was added  3-chloropropanoyl chloride (14 mg, 0.11 mmol) . The mixture was stirred at 0℃ for 0.5 h. NaOH (20 mg, 0.50 mmol) was added. The mixture was heated at 60 ℃ for 3 h and cooled to r.t.. The mixture was diluted with water and extracted with ethyl acetate (2 ×) . Solvents were evaporated under reduced pressure. The residue was purified by column chromatography on silica gel eluting with 5%methanol in DCM to give N- (2- ( (2- (Dimethylamino) ethyl) (methyl) amino) -6-methoxy-5-nitropyridin-3-yl) acrylamide (1f) (6.7 mg, 45%) . MS-ESI (m/z) : 324.2 [M + 1] +.
N- (5-Amino-2- ( (2- (dimethylamino) ethyl) (methyl) amino) -6-methoxypyridin-3-yl) acryla  mide (1g)
The mixture of N- (2- ( (2- (Dimethylamino) ethyl) (methyl) amino) -6-methoxy-5-nitropyridin-3-yl) acrylamide (1f) (6.7 mg, 0.021 mmol) , Fe (7.0 mg, 0.12 mmol) , and NH4Cl (1.1 mg, 0.021 mmol) in ethanol-H2O (3:1, 2 mL) was heated at reflux for 6 h. Solvents were evaporated under reduced pressure. The residue was purified by column chromatography on silica gel eluting with DCM/methanol/ammonia (92:6:2) to give N- (5-Amino-2- ( (2- (dimethylamino) ethyl) (methyl) amino) -6-methoxypyridin-3-yl) acrylamide (1g) (3.6 mg, 59%) . MS-ESI (m/z) : 294 [M + 1] +.
N- (2- ( (2- (Dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- (4- (1-methyl-1H-indol-3- yl) pyrimidin-2-ylamino) pyridin-3-yl) acrylamide (1)
The mixture of N- (5-Amino-2- ( (2- (dimethylamino) ethyl) (methyl) amino) -6-methoxypyridin-3-yl) acrylamide (1g) (3.6 mg, 0.012 mmol) , 3- (2-chloropyrimidin-4-yl) -1-methyl-1H-indole (1h) (6.0 mg, 0.025 mmol) (prepared by following the literature, WO2013/14448) , and p-TsOH·H2O (4.7 mg, 0.025 mmol) in 2-pentanol (0.5 mL) was heated at 105℃ for 2 h. Solvent was evaporated under reduced pressure. The residue was purified by column chromatography on silica gel eluting with DCM/methanol (95:5) to give N- (2- ( (2- (Dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-ylamino) pyridin-3-yl) acrylamide (1) (5.0 mg, 82%) . MS-ESI (m/z) : 500 [M + 1] +.
Example 2
N- (6-Methoxy-2- (methyl (2- (methylamino) ethyl) amino) -5- ( (4- (1-methyl-1H-indol-3-yl) p  yrimidin-2-yl) amino) pyridin-3-yl) acrylamide (2)
tert-Butyl 2- ( (6-methoxy-5-nitropyridin-2-yl) (methyl) amino) ethyl (methyl) carbamate  (2b)
The solution of 1a (4.00 g, 21.3 mmol) and tert-butyl methyl (2- (methylamino) ethyl) carbamate (2a, 4.38 g, 22.3 mmol) (prepared by following the literature, J. Med. Chem., 1992, 35, 565) in ethanol (47 mL) was heated at reflux for 3 h. The solvent was evaporated under reduced pressure. The residue was purified by column chromatography on silica gel eluting with 15-30%ethyl acetate in hexanes to give tert-Butyl 2- ( (6-methoxy-5-nitropyridin-2-yl) (methyl) amino) ethyl (methyl) carbamate (2b) (4.65 g, 63%) as yellow solid. MS-ESI (m/z) : 341 [M + 1] +.
tert-Butyl 2- ( (3-bromo-6-methoxy-5-nitropyridin-2-yl) (methyl) amino) ethyl (methyl) carbamate (2c)
To a suspension of tert-Butyl 2- ( (6-methoxy-5-nitropyridin-2-yl) (methyl) amino) ethyl (methyl) carbamate (2b) (4.32 g, 12.7 mmol) in acetonitrile (50 mL) at 0C was added NBS (3.40 g, 19.1 mmol) . The mixture was stirred at 0℃ for 1 h. Solvent was evaporated under reduced pressure. The residue was purified by column chromatography on silica gel eluting with 20-30%ethyl acetate hexanes to give tert-Butyl 2- ( (3-bromo-6-methoxy-5-nitropyridin-2-yl) (methyl) amino) ethyl (methyl) carbamate (2c) as yellow solid (5.00 g, 93%) . MS-ESI (m/z) : 419 and 421 [1:1, M +1] +.
tert-Butyl 2- ( (3- (diphenylmethyleneamino) -6-methoxy-5-nitropyridin-2-yl)  (methyl) amino) ethyl (methyl) carbamate (2d)
The mixture of tert-Butyl 2- ( (3-bromo-6-methoxy-5-nitropyridin-2-yl) (methyl) amino) ethyl (methyl) carbamate (2c) (2.095 g, 5.00 mmol) , Pd2 (dba) 3 (137 mg, 0.150 mmol) , (+/-) -BINAP (187 mg, 0.300 mmol) , benzophenone imine (1.177 g, 6.50 mmol) , and t-BuONa (624 mg, 6.50 mmol) in toluene (20 mL) was heated under nitrogen at 110C for 3 h, and cooled to room temperature. The solid was removed by filtration through celite and washed with ethyl acetate.  Solvents were evaporated under reduced pressure. The residue was purified by column chromatography on silica gel eluting with 20-40%ethyl acetate in hexanes to give tert-Butyl 2- ( (3- (diphenylmethyleneamino) -6-methoxy-5-nitropyridin-2-yl) (methyl) amino) ethyl (methyl) carbamate (2d) as pale yellow solid (600 mg, 23%) . MS-ESI (m/z) : 520 [M + 1] +.
tert-Butyl 2- ( (3-amino-6-methoxy-5-nitropyridin-2-yl) (methyl) amino) ethyl (methyl) carbamate (2e)
To a solution of tert-Butyl 2- ( (3- (diphenylmethyleneamino) -6-methoxy-5-nitropyridin -2-yl) (methyl) amino) ethyl (methyl) carbamate (2d) (580 mg, 1.12 mmol) in ethanol (20 mL) at r.t. was added hydroloric acid (1 N, 0.5 mL) . The mixture was heated at 50C for 7 h. The mixture was diluted with NaHCO3 (aq. ) and extracted with ethyl acetate (3 ×) . The extracts were washed with water and brine, and dried over MgSO4. Solvents were evaporated under reduced pressure. The residue was purified by column chromatography on silica gel eluting with 20-40%ethyl acetate in hexanes to give tert-Butyl 2- ( (3-amino-6-methoxy-5-nitropyridin-2-yl) (methyl) amino) ethyl (methyl) carbamate (2e) as orange solid (280 mg, 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 mixture of tert-Butyl 2- ( (3-amino-6-methoxy-5-nitropyridin-2-yl) (methyl) amino) ethyl (methyl) carbamate (2e) (280 mg, 0.789 mmol) in THF-H2O (10:1, 5.5 mL) at 0℃ was added 3-chloropropanoyl chloride (120 mg, 0.946 mmol) . The mixture was stirred at 0℃ for 0.5 h. NaOH (126 mg, 3.76 mmol) was added. The mixture was heated at 65℃ for 1 h and cooled to r.t.. The mixture was diluted with water and extracted with ethyl acetate (2 ×) . Solvents were evaporated under reduced pressure. The residue was purified by column chromatography on silica gel eluting with 30-50%ethyl acetate in hexanes to give tert-Butyl 2- ( (3-acrylamido-6-methoxy-5-nitropyridin -2-yl) (methyl) amino) ethyl (methyl) carbamate (2f) (185 mg, 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)
The mixture of tert-Butyl 2- ( (3-acrylamido-6-methoxy-5-nitropyridin-2-yl) (methyl) amino) ethyl (methyl) carbamate (2f) (185 mg, 0.452 mmol) , iron powder (152 mg, 2.71 mmol) , and NH4Cl (24.2 mg, 0.452 mmol) in ethanol-H2O (3:1, 15 mL) was heated at reflux for 1 h. The solid was removed by filtration through celite and washed with ethyl acetate. The filtrates were diluted with ethyl acetate and washed with water and brine. Solvents were evaporated under reduced pressure to give crude tert-Butyl 2- ( (3-acrylamido-5-amino-6-methoxypyridin-2-yl) (methyl) amino) ethyl (methyl) carbamate (2g) (149 mg, 87%) as 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) p yrimidin-2-yl) amino) pyridin-3-yl) acrylamide (2)
The mixture of tert-Butyl 2- ( (3-acrylamido-5-amino-6-methoxypyridin-2-yl) (methyl) amino) ethyl (methyl) carbamate (2g) (94.0 mg, 0.248 mmol) , 3- (2-chloropyrimidin-4-yl) -1 -methyl-1H-indole (1h, 72.5 mg, 0.298 mmol) , and p-TsOH·H2O (56.6 mg, 0.298 mmol) in 2-pentanol (1.5 mL) was heated at 105C for 2 h. Solvent was evaporated under reduced pressure. The residue was purified by column chromatography on silica gel eluting with DCM/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) (100.0 mg, 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-ind ol-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (3)
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) (15.4 mg, 0.0317 mmol) in 1, 2-dichloroethane (1 mL) at room temperature was added acetaldehyde (40%in water, 9.0 mg, 0.082 mmol) followed by NaBH (OAc) 3 (8.7 mg, 0.041 mmol) . The mixture was stirred at room temperature for 1 h. The mixture was diluted with saturated aqueous NaHCO3 (5 mL) and extracted with DCM (2 × 5 mL) . The extracts were dried over Na2SO4. Solvents were evaporated under  reduced pressure. The residue was purified by column chromatography on silica gel eluting with DCM/methanol/ammonia (95:4:1) to give 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) as brown solid (12.5 mg, 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-indol- 3-yl) pyrimidin-2-ylamino) 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-ylamino) pyridin-3-yl) acrylamide (2) (28.3 mg, 0.058 mmol) in DCM (1 mL) was added DIPEA (15.2 μL, 0.087 mmol) , and cooled to 0℃. A solution of Ac2O (5.9 mg in 0.20 mL DCM) was added dropwise. The mixture was stirred at room temperature for 1 h. The mixture was diluted with saturated aqueous NaHCO3 (5 mL) and extracted with DCM (2 × 5 mL) . The extracts were dried over Na2SO4. Solvents were evaporated under reduced pressure. The residue was purified by column chromatography on silica gel eluting with 2-3%methanol in DCM to give N- (6-Methoxy-2- (methyl (2- (N-methylacetamido) ethyl) amino) -5- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-ylamino) pyridin-3-yl) acrylamide (4) as yellow solid (15.1 mg, 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) pyr imidin-2-ylamino) pyridin-3-yl) acrylamide (5)
6-chloro-2-ethoxy-3-nitropyridine (5a)
6-chloro-2-ethoxy-3-nitropyridine (5a) was prepared according to the synthetic method of 1a by replacing methanol with ethanol. MS-ESI (m/z) : 203 [M + 1] +.
N- (6-Ethoxy-2- (methyl (2- (methylamino) ethyl) amino) -5- (4- (1-methyl-1H-indol-3-yl) pyr imidin-2-ylamino) pyridin-3-yl) acrylamide (5)
The title compound N- (6-Ethoxy-2- (methyl (2- (methylamino) ethyl) amino) -5-(4- (1-methyl-1H-indol-3-yl) pyrimidin-2-ylamino) pyridin-3-yl) acrylamide (5) (36.1 mg) was prepared according to the synthetic method of Example 2 by replacing 6-Chloro-2-methoxy-3-nitropyridine (1a) with 6-chloro-2-ethoxy-3-nitropyridine (5a) . MS-ESI (m/z) : 501 [M + 1] +.
Example 6
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)
To a solution of N- (6-Ethoxy-2- (methyl (2- (methylamino) ethyl) amino) -5- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-ylamino) pyridin-3-yl) acrylamide (5) (10.1 mg, 0.020 mmol) in 1, 2-dichloroethane (0.6 mL) at room temperature was added formaldehyde (37%in water, 17 mg) followed by NaBH (OAc) 3 (6.4 mg, 0.030 mmol) . The mixture was stirred at room temperature for 1 h.The mixture was diluted with saturated aqueous NaHCO3 (2 mL) and extracted with DCM (2 × 2 mL) . The extracts were dried over Na2SO4. Solvents were evaporated under reduced pressure. The residue was purified by column chromatography on silica gel eluting with 5-10%methanol in DCM to give 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) as grey solid (8.5 mg, 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-ylamino) pyridin-3-yl) acrylamide (7)
6-chloro-2-isopropoxy-3-nitropyridine (7a)
The title compound 6-chloro-2-isopropoxy-3-nitropyridine (7a) was prepared according to the synthetic method of 1a by replacing methanol with isopropanol. 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-ylamino) pyridin-3-yl) acrylamide (7)
The title compound 7 (36.1 mg) was prepared according to the synthetic method of Example 2 by replacing 6-Chloro-2-methoxy-3-nitropyridine (1a) with 6-chloro-2-isopropoxy-3-nitropyridine (7a) . MS-ESI (m/z) : 515 [M + 1] +.
Example 8
N- (2- ( (2- (Dimethylamino) ethyl) (methyl) amino) -6-isopropoxy-5- (4- (1-methyl-1H-indol- 3-yl) pyrimidin-2-ylamino) pyridin-3-yl) acrylamide (8)
The title compound 8 (6.3 mg, 75%) was prepared according to the synthetic method of Example 6 by replacing N- (6-Ethoxy-2- (methyl (2- (methylamino) ethyl) amino) -5- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-ylamino) 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-ylamino) pyridin-3-yl) acr ylamide (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) p yridin-3-yl) acrylamide (9)
The title compound 9 (25 mg) was prepared according to the synthetic method of Example 2 by replacing tert-butyl methyl (2- (methylamino) ethyl) carbamate (2a) with tert-butyl piperazine-1-carboxylate. 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-methylpiperaz in-1-yl) pyridin-3-yl) acrylamide (10)
The title compound 10 (5.2 mg) was prepared according to the synthetic method of Example 6 by replacing N- (6-Ethoxy-2- (methyl (2- (methylamino) ethyl) amino) -5- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-ylamino) pyridin-3-yl) acrylamide (5) 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) . 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) py ridin-3-yl) acrylamide (11)
The title compound 11 (55 mg) was prepared according to the synthetic method of Example 2 by replacing methyl (2- (methylamino) ethyl) carbamate (2a) with tert-butyl piperazine-1-carboxylate and replacing 6-Chloro-2-methoxy-3-nitropyridine (1a) 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-methylpiperazi n-1-yl) pyridin-3-yl) acrylamide (12)
The title compound 12 (7.2 mg) was prepared according to the synthetic method of Example 6 by replacing N- (6-Ethoxy-2- (methyl (2- (methylamino) ethyl) amino) -5- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-ylamino) pyridin-3-yl) acrylamide (5) 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) . 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-y l) pyridin-3-yl) acrylamide (13)
The title compound 13 (75 mg) was prepared according to the synthetic method of Example 2 by replacing tert-butyl methyl (2- (methylamino) ethyl) carbamate (2a) with tert-butyl piperazine-1-carboxylate and replacing 6-Chloro-2-methoxy-3-nitropyridine (1a) with 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-methylpipe razin-1-yl) pyridin-3-yl) acrylamide (14)
The title compound 14 (7.4 mg) was prepared according to the synthetic method of Example 6 by replacing N- (6-Ethoxy-2- (methyl (2- (methylamino) ethyl) amino) -5- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-ylamino) pyridin-3-yl) acrylamide (5) 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) . 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)
N1- (6-chloro-3-nitropyridin-2-yl) -N1, N2, N2-trimethylethane-1, 2-diamine (15a)
To a solution of 2, 6-dichloro-3-nitropyridine (19.3 g, 100.0 mmol) and triethylamine (20.2 g, 200.0 mmol) in dry dichloromethane (200 mL) was added a solution of N1, N1, N2-trimethylethane-1, 2-diamine (10.2 g, 100.0 mmol) in dry dichloromethane (20 ml) dropwise at 0℃for over 0.5 h . Then the reaction mixture was stirred for 1.5 h at 0℃. The mixture was diluted with water (200 mL) and extracted with dichloromethane (100 mL × 3) . The organic layers were combined, washed with brine (200 mL) , and dried over Na2SO4. The solids were filtered and the filtrate was concentrated in vacuum to give N1- (6-chloro-3-nitropyridin-2-yl) -N1, N2, N2-trimethylethane-1, 2-diamine (15a) as yellow oil (23.8 g, 91%) . MS-ESI (m/z) : 259 [M + 1] +.
N1- (6-methoxy-3-nitropyridin-2-yl) -N1, N2, N2-trimethylethane-1, 2-diamine (15b)
To a solution of N1- (6-chloro-3-nitropyridin-2-yl) -N1, N2, N2-trimethylethane-1, 2-diamine (15a) (23.8 g, 91.9 mmol) in methanol (200 mL) was added sodium methoxide (10.8 g, 183.8 mmol) at 0-5℃. The mixture was heated to reflux for 2 h. Then the mixture was cooled to r.t. and methanol was evaporated, diluted with water (200 mL) and extracted with dichloromethane (200 mL × 2) . The organic layers were combined and washed with water (200 mL × 2) and brine (200 mL) , and dried over Na2SO4. The solids were filtered out and the filtrate was concentrated in  vacuum. The crude product was purified by column chromatography on silica gel eluting with ethyl acetate/petroleum ether (1:1) to give N1- (6-methoxy-3-nitropyridin-2-yl) -N1, N2, N2-trimethylethane-1, 2-diamine (15b) as yellow oil (18.0 g, 77%) . MS-ESI (m/z) : 255 [M + 1] +.
N1- (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N1, N2, N2-trimethylethane-1, 2-diamine (15c)
To a solution of N1- (6-methoxy-3-nitropyridin-2-yl) -N1, N2, N2-trimethylethane-1, 2-diamine (15b) (11.0 g, 43.1 mmol) in glacial acetic acid (120 mL) was added N-iodosuccinimide (11.6 g, 51.8 mmol) for 3 batches at 0-5℃, and the mixture was stirred at room temperature for 4.5 h. Then solvent was evaporated under reduced pressure, diluted with water (300 mL) and extracted with ethyl acetate (200 mL × 3) . The organic layers were combined, washed with water (200 mL × 2) and brine (200 mL) , and dried over Na2SO4. The solids were filtered out and the filtrate was concentrated in vacuum. The crude product was purified by column chromatography on silica gel eluting with ethyl acetate/petroleum ether (1:1) to give N1- (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N1, N2, N2-trimethylethane-1, 2-diamine (15c) as yellow solid (14.0 g, 88%) . MS-ESI (m/z) : 381 [M + 1] +.
N1- (5- ( (diphenylmethylene) amino) -6-methoxy-3-nitropyridin-2-yl) -N1, N2, N2-trimethylet hane-1, 2-diamine (15d)
A mixture of N1- (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N1, N2, N2-trimethylethane -1, 2-diamine (15c) (10.0 g, 26.2 mmol) , tris (dibenzylideneacetone) dipalladium (0) (1.2 g, 1.3 mmol) , (+/-) -2, 2'-bis (diphenylphosphino) -1, 1'-binaphthyl (1.6 g, 2.6 mmol) , benzophenone imine (7.1 g, 39.4 mmol) , and sodium tert-butoxide (3.8 g, 39.4 mmol) in toluene (150 mL) was stirred at 110℃under an inert atmosphere of nitrogen for 16 h. The resulting mixture was cooled to room temperature, the solids were removed by filtration through celite and washed with ethyl acetate. Solvents were evaporated under reduced pressure. The residue was purified by column chromatography on silica gel eluting with ethyl acetate to give N1- (5- ( (diphenylmethylene) amino) -6-methoxy-3-nitropyridin-2-yl) -N1, N2, N2-trimethylethane-1, 2-diamine (15d) as red oil (2.2 g, 19%) . MS-ESI (m/z) : 434 [M + 1] +.
N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-3-nitropyridine-2, 5-diamine (15e)
To a solution of N1- (5- ( (diphenylmethylene) amino) -6-methoxy-3-nitropyridin-2-yl) -N1, N2, N2-trimethylethane-1, 2-diamine (15d) (2.2 g, 5.1 mmol) in tetrahydrofuran (150 mL) was added 0.5 N hydrochloric acid (24 mL) at room temperature. The resulting solution was stirred at room temperature for 1.5 h. Tetrahydrofuran was evaporated under reduced pressure. The pH value of the mixture was adjusted to 9-10 with potassium carbonate. The resulting mixture was extracted with dichloromethane (30 mL × 4) . The organic layers were combined, washed with brine (20 mL) , and dried over Na2SO4. The solids were filtered out and the filtrate was concentrated under vacuum. The residue was purified by column chromatography on silica gel eluting with dichloromethane/methanol (20:1~10:1) to give N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-3-nitropyridine-2, 5-diamine (15e) as red oil (0.7 g, 51%) . MS-ESI (m/z) : 270 [M + 1] +.
7-fluoro-1-methyl-1H-indole (15f)
To a solution of 7-fluoro-1H-indole (1.0 g, 7.4 mmol) in N, N-dimethylformamide (10 mL) was added sodium hydride (60%dispersion in mineral oil, 0.44g, 11.1 mmol) at 0-5℃. The resulting suspension was stirred at room temperature for 20 min. Iodomethane (0.7 mL, 11.1 mmol) was added at room temperature. Then the resulting mixture was stirred at room temperature for 1 h. The reaction was quenched by water (40 mL) , and the mixture was extracted with ethyl acetate (30 mL × 3) . The organic layers were combined, washed with water (30 mL × 3) and brine (30 mL) , and dried over Na2SO4. The solids were filtered out and the filtrate was concentrated in vacuum. The residue was purified by column chromatography on silica gel eluting with ethyl acetate/petroleum ether (1:20) to give 7-fluoro-1-methyl-1H-indole (15f) as light yellow liquid (1.0 g, 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) (0.60 g, 4.0 mmol) , 2, 4-dichloropyrimidine (0.60 g, 4.0 mmol) , anhydrous Iron (III) chloride (0.65 g, 4.0 mmol) in 1, 2-dimethoxyethane (12 mL) was stirred at 60℃ for 7 h. The reaction was quenched by water (40  mL) , the resulting mixture was extracted with ethyl acetate (30 mL × 3) , the organic layers were combined and washed with water (30 mL × 3) and brine (30 mL) , dried over anhydrous sodium sulfate. The solids were filtered out and the filtrate was concentrated in vacuum. The residue was purified by column chromatography on silica gel 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 brown solid (0.50 g, 48%) . MS-ESI (m/z) : 262 [M + 1] +.
N2- (2- (dimethylamino) ethyl) -N5- (4- (7-fluoro-1-methyl-1H-indol-3-yl) pyrimidin-2-yl) -6  -methoxy-N2-methyl-3-nitropyridine-2, 5-diamine (15h)
A mixture of 3- (2-chloropyrimidin-4-yl) -7-fluoro-1-methyl-1H-indole (15g) (30.0 mg, 0.11 mmol) , N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-3-nitropyridine-2, 5-diamine (15e) (31.0 mg, 0.11 mmol) , cesium carbonate (112.0 mg, 0.33 mmol) , tris (dibenzylideneacetone) dipalladium (0) (21.0 mg, 0.022 mmol) , (+/-) -2, 2'-bis (diphenylphosphino) -1, 1'-binaphthyl (29.0 mg, 0.044 mmol) in dioxane (3 mL) was stirred at 100℃ for 7 h, and cooled to r.t.. The reaction was quenched by water (30 mL) , the resulting mixture was extracted with ethyl acetate (30 mL × 3) , the organic layers were combined and washed with water (30 mL × 3) and brine (30 mL) , dried over Na2SO4. The solids were filtered out and the filtrate was concentrated in vacuum. The residue was purified by column chromatography on silica gel eluting with dichloromethane/methanol/aqueous ammonia (100:5:0.5) to give N2- (2- (dimethylamino) ethyl) -N5- (4- (7-fluoro-1-methyl-1H-indol-3-yl) pyrimidin-2-yl) -6-methoxy-N2-methyl-3-nitropyridine-2, 5-diamine (15h) as yellow solid (34.0 mg, 60%) . MS-ESI (m/z) : 495 [M + 1] +.
N2- (2- (dimethylamino) ethyl) -N5- (4- (7-fluoro-1-methyl-1H-indol-3-yl) pyrimidin-2-yl) -6  -methoxy-N2-methylpyridine-2, 3, 5-triamine (15i)
A mixture of N2- (2- (dimethylamino) ethyl) -N5- (4- (7-fluoro-1-methyl-1H-indol-3-yl) pyrimidin-2-yl) -6-methoxy-N2-methyl-3-nitropyridine-2, 5-diamine (15h) (34.0 mg, 0.07 mmol) , iron powder (41.0 mg, 0.73 mmol) , ammonium chloride (13.0 mg, 0.24 mmol) in ethanol (12 mL) and water (3 mL) was stirred at 80℃ for 1.5 h. The solids were filtered out, washed with ethanol (5 mL × 3) , and the filtrate was concentrated in vacuum. The residue was purified by column  chromatography on silica gel eluting with dichloromethane/methanol/aqueous ammonia (100:5:0.5~100:10:0.5) to give N2- (2- (dimethylamino) ethyl) -N5- (4- (7-fluoro-1-methyl-1H-indol-3-yl) pyrimidin-2-yl) -6-methoxy-N2-methylpyridine-2, 3, 5-triamine (15i) as yellow oil (28.0 mg, 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) acrylamide (15)
To a solution of N2- (2- (dimethylamino) ethyl) -N5- (4- (7-fluoro-1-methyl-1H-indol-3-yl) pyrimidin-2-yl) -6-methoxy-N2-methylpyridine-2, 3, 5-triamine (15i) (28.0 mg, 0.06 mmol) in anhydrous dichloromethane (1 mL) was added acryloyl chloride (5.5 mg, 0.06 mmol) at 0℃. The resulting solution was stirred at 0℃ for 1 h. The reaction was quenched by saturated aqueous sodium bicarbonate (20 mL) , and the resulting mixture was extracted with dichloromethane (20 mL × 3) . The organic layers were combined, washed with water (20 mL × 3) and brine (20 mL) , and dried over Na2SO4. The solids were filtered out and the filtrate was concentrated in vacuum. The residue was purified by column chromatography on silica gel eluting with dichloromethane/methanol/aqueous 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 yellow solid (20.0 mg, 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)
The title compound 16 (20.0 mg) was prepared according to the synthetic method of Example 15 by replacing 7-fluoro-1H-indole with 6-fluoro-1H-indole. 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)
The title compound 17 (25.0 mg) was prepared according to the synthetic method of Example 15 by replacing 7-fluoro-1H-indole with 5-fluoro-1H-indole. 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 (23.0 mg) was prepared according to the synthetic method of Example 15 by replacing 7-fluoro-1H-indole with 4-fluoro-1H-indole. 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 (0.50 g, 3.8 mmol) , 2, 4-dichloro-5-fluoropyrimidine (0.64 g, 3.8 mmol) , anhydrous Iron (III) chloride (0.62 g, 3.8 mmol) in 1, 2-dimethoxyethane (7.0 mL) was stirred at 60℃ for 7 h. The reaction was quenched by water (40 mL) , and the resulting mixture was extracted with ethyl acetate (30 mL × 3) . The organic layers were combined, washed with water (30 mL × 3) and brine (30 mL) , and dried over Na2SO4. The solids were filtered out and the filtrate was concentrated in vacuum. The residue was purified by column chromatography on silica gel 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 brown solid (0.54 g, 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)
The title compound 19 (23.0 mg) was prepared according to the synthetic method of Example 15 by replacing 3- (2-chloropyrimidin-4-yl) -7-fluoro-1-methyl-1H-indole (15g) with 3- (2-chloro-5-fluoropyrimidin-4-yl) -1-methyl-1H-indole (19a) . 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- (dimethylamin o) ethyl) (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 (0.50 g, 3.8 mmol) , 2, 4, 5-trichloropyrimidine (0.70 g, 3.8 mmol) , anhydrous Iron (III) chloride (0.62 g, 3.8 mmol) in 1, 2-dimethoxyethane (7.0 mL) was stirred at 60℃ for 7 h. The reaction was quenched by water (40 mL) , and the resulting mixture was extracted with ethyl acetate (30 mL × 3) . The organic layers were combined, washed with water (30 mL × 3) and brine (30 mL) , and dried over Na2SO4. The solids were filtered out and the filtrate was concentrated in vacuum. The residue was purified by column chromatography on silica gel eluting with ethyl acetate/petroleum ether (1:5~1:2) to give 3- (2, 5-dichloropyrimidin-4-yl) -1-methyl-1H-indole (20a) as brown solid (0.56 g, 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- (dimethylamin o) ethyl) (methyl) amino) -6-methoxypyridin-3-yl) acrylamide (20)
The title compound 20 (20.0 mg) was prepared according to the synthetic method of Example 15 by replacing 3- (2-chloropyrimidin-4-yl) -7-fluoro-1-methyl-1H-indole (15g) with 3- (2, 5-dichloropyrimidin-4-yl) -1-methyl-1H-indole (20a) . 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 (0.50 g, 3.8 mmol) , 2, 4-dichloro-5-methylpyrimidine (0.62 g, 3.8 mmol) , anhydrous Iron (III) chloride (0.62 g, 3.8 mmol) in 1, 2-dimethoxyethane (7.0 mL) was stirred at 60℃ for 7 h. The reaction was quenched by water (40 mL) , the resulting mixture was extracted with ethyl acetate (30 mL × 3) , the organic layers were combined and washed with water  (30 mL × 3) and brine (30 mL) , dried over Na2SO4. The solids were filtered out and the filtrate was concentrated in vacuum. The residue was purified by column chromatography on silica gel 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 brown solid (0.50 g, 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)
The title compound 21 (26.0 mg) was prepared according to the synthetic method of Example 15 by replacing 3- (2-chloropyrimidin-4-yl) -7-fluoro-1-methyl-1H-indole (15g) with 21a. MS-ESI (m/z) : 515 [M + 1] +.
Example 22
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)
5-bromo-3-methoxy-2-nitropyridine (22a)
5-bromo-3-methoxy-2-nitropyridine (22a) was prepared according to the synthetic method described in US2006/84665.
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) (2.8 g, 12.02 mmol) , tert-butyl methyl (2- (methylamino) ethyl) carbamate (2.67 g, 13.22 mmol) , Pd (OAc) 2 (0.27 g, 1.20 mmol) , Xantphos (0.69 g, 1.20 mmol) and Cs2CO3 (11.7 g, 35.91 mmol) in toluene (56 mL) was stirred at 100℃ under nitrogen for 2.5 h. The mixture was cooled to r.t., quenched with water (200 mL) and extracted with EA (3×50 mL) . The extracts were washed with brine, dried over Na2SO4, concentrated and purified by column chromatography on silica gel eluting with 50%ethyl acetate in  hexanes to give tert-butyl (2- ( (5-methoxy-6-nitropyridin-3-yl) (methyl) amino) ethyl) (methyl) carbamate (22b) as yellow solid (3.0 g, 73%) . MS-ESI (m/z) : 341 [M + 1] +.
tert-Butyl (2- ( (2-bromo-5-methoxy-6-nitropyridin-3-yl) (methyl) amino) ethyl) (methyl)  carbamate (22c)
To a solution of tert-butyl (2- ( (5-methoxy-6-nitropyridin-3-yl) (methyl) amino) ethyl) (methyl) carbamate (22b) (1.70 g, 5.00 mmol) in acetic acid (30 mL) at 0℃ was added a solution of Br2 (0.88 g, 5.50 mmol) in acetic acid (5 mL) . The mixture was stirred at 0℃ for 0.5 h and then at room temperature for 1 h. The mixture was diluted with water (150 mL) and extracted with ethyl acetate (3 ×) . The extracts were washed with saturated aqueous NaHCO3 and brine and dried over Na2SO4. Solvent was evaporated under reduced pressure. The residue was purified by column chromatography on silica gel eluting with 20-40%ethyl acetate in hexanes to give tert-Butyl (2- ( (2-bromo-5-methoxy-6-nitropyridin-3-yl) (methyl) amino) ethyl) (methyl) carbamate (22c) as yellow solid (1.53 g, 73%) . MS-ESI (m/z) : 419.2 and 421.2 [1:1, M + 1] +.
tert-Butyl (2- ( (2- ( (diphenylmethylene) amino) -5-methoxy-6-nitropyridin-3-yl) (methyl)  amino) ethyl) (methyl) carbamate (22d)
The mixture of tert-Butyl (2- ( (2-bromo-5-methoxy-6-nitropyridin-3-yl) (methyl) amino) ethyl) (methyl) carbamate (22c) (419 mg, 1.00 mmol) , Pd2 (dba) 3 (27.5 mg, 0.030 mmol) , (+/-) -BINAP (37.4 mg, 0.060 mmol) , benzophenone imine (199 mg, 1.10 mmol) , and t-BuONa (192 mg, 2.00 mmol) in toluene (5 mL) was heated under nitrogen at 80C for 4 h. The mixture was cooled to room temperature, the solvent was evaporated under reduced pressure. THF (14 mL) and 0.5 N HCl (14 mL) were added successively. The mixture was stirred at r.t. for 1.5 h. The mixture was diluted with NaHCO3 (50 mL) , extracted with ethyl acetate (2×) . The extracts were washed with brine and dried over Na2SO4. Solvent was evaporated under reduced pressure. The residue was purified by column chromatography on silica gel eluting with 30-50%ethyl acetate in hexanes to give tert-Butyl (2- ( (2- ( (diphenylmethylene) amino) -5-methoxy-6-nitropyridin-3-yl) (methyl) amino) ethyl) (methyl) carbamate (22d) as yellow solid (76 mg, 21%) . MS-ESI (m/z) : 356.2 [M + 1] +.
tert-Butyl (2- ( (2-acrylamido-5-methoxy-6-nitropyridin-3-yl) (methyl) amino) ethyl)  (methyl) carbamate (22e)
To a mixture of tert-Butyl (2- ( (2- ( (diphenylmethylene) amino) -5-methoxy-6-nitropyridin-3-yl) (methyl) amino) ethyl) (methyl) carbamate (22d) (75 mg, 0.211 mmol) in acetonitrile (3 mL) at 0℃ was added 3-chloropropanoyl chloride (53.6 mg, 0.422 mmol) followed by DMAP (51.5 mg, 0.422 mmol) and DIPEA (81.7 mg, 0.633 mmol) . The mixture was stirred at 0℃ for 2.5 h. NaOH (10 N, 0.8 mL) was added. The mixture was stirred at r.t. for 1.5 h. The mixture was diluted with water and extracted with ethyl acetate (2 ×) . Solvents were evaporated under reduced pressure. The residue was purified by column chromatography on silica gel eluting with 5%methanol in DCM to give tert-Butyl (2- ( (2-acrylamido-5-methoxy-6-nitropyridin-3-yl) (methyl) amino) ethyl) (methyl) carbamate (22e) (28.5 mg, 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)
The mixture of tert-Butyl (2- ( (2-acrylamido-5-methoxy-6-nitropyridin-3-yl) (methyl) amino) ethyl) (methyl) carbamate (22e) (28.5 mg, 0.070 mmol) , Fe (19.5 mg, 0.348 mmol) , and NH4Cl (3.7 mg, 0.070 mmol) in ethanol-H2O (3:1, 1.5 mL) was heated at reflux for 2 h. Solvents were evaporated under reduced pressure. The residue was purified by silica gel column eluting with 92:6:2 DCM-methanol-ammonia to give tert-Butyl (2- ( (2-acrylamido-6-amino-5-methoxypyridin-3-yl) (methyl) amino) ethyl) (methyl) carbamate (22f) (9.0 mg, 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)
The mixture of tert-Butyl (2- ( (2-acrylamido-6-amino-5-methoxypyridin-3-yl) (methyl) amino) ethyl) (methyl) carbamate (22f) (8.6 mg, 0.023 mmol) , 3- (2-chloropyrimidin-4-yl) -1-methyl-1H-indole (1h, 6.6 mg, 0.027 mmol) , Pd2 (dba) 3 (3.4 mg, 0.0036 mmol) , BINAP (2.2 mg,  0.0036 mmol) , and Cs2CO3 (11 mg, 0.034 mmol) in dioxane (0.5 mL) was heated at 100℃ for 2.5 h. The mixture was diluted with water and extracted with dichloromethane. Solvent was evaporated under reduced pressure. The residue was purified by silica gel column eluting with 95:5 DCM-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) (3.3 mg, 25%) . MS-ESI (m/z) : 586.3 [M + 1] +.
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)
To a solution of 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) (3.3 mg) in DCM (0.5 mL) at r.t. was added TFA (0.5 mL) . The mixture was stirred at r.t. for 40 min. Solvents were evaporated. To this was added NaBH (OAc) 3 (3.5 mg) , 1, 2-dichloroethane (0.3 mL) and CH2O (37%aqueous, 15 mg) . The mixture was stirred at r.t. for 1 h. The mixture was diluted with NaHCO3, extracted with DCM. The crude product was purified by silica gel column, eluted with 95:4:1 DCM-MeOH-NH4OH 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) (2.5 mg, 89%) . MS-ESP: 501.4 [M + 1] +.
Example 23
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)
N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-N5- (4- (1-methyl-1H-indol-3-yl) pyri midin-2-yl) -3-nitropyridine-2, 5-diamine (23a)
The title compound 23a was prepared according to the synthetic method of Example 2 by replacing N1- (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N1, N2, N2-trimethylethane-1, 2-diamine (2g) with N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-3-nitropyridine-2, 5-diamine (15e) . MS-ESI (m/z) : 477 [M + 1] +.
N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-N5- (4- (1-methyl-1H-indol-3-yl) pyri midin-2-yl) pyridine-2, 3, 5-triamine (23b)
The title compound 23b was prepared according to the synthetic method of 15i by replacing N1- (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N1, N2, N2-trimethylethane-1, 2-diamine (15h) with N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-N5- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) -3-nitropy9ridine-2, 5-diamine (23a) . MS-ESI (m/z) : 447 [M + 1] +.
N- (2- ( (2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ( (4- (1-methylimidazo [1, 5- a] pyridin-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) -2-fluoroacrylamide (23)
A mixture of N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-N5- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) pyridine-2, 3, 5-triamine (23b) (20 mg, 0.045 mmol) , HOBT (18 mg, 0.135 mmol) , EDCI (26 mg, 0.135) , TEA (14 mg, 0.135 mmol) , in DMF (2 ml) was stirred at 35℃for 3 h. The reaction was cooled to room temperature and quenched by water (5 mL) . The mixture was adjusted to pH 9~10 using sodium carbonate. The resulting mixture was extracted with EA (8 mL × 3) . The organic layers were combined and washed with brine (20 mL) , dried over Na2SO4. The solids were filtered out and the filtrate was concentrated in vacuum. The residue was purified by column chromatography on silica gel eluting with dichloromethane/methanol (10:1) as the eluant to give N- (2- ( (2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ( (4- (1-methylimidazo [1, 5-a] pyridin-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-indol-3- yl) pyrimidin-2-yl) amino) phenyl) -2-fluoroacrylamide (24)
N1- (2- (dimethylamino) ethyl) -5-methoxy-N1-methyl-2-nitrobenzene-1, 4-diamine (24a)
A mixture of 4-fluoro-2-methoxy-5-nitroaniline (3 g, 16.0 mmol) (prepared by following the literature WO2013/14448) , N1, N1, N2-trimethylethane-1, 2-diamine (2 g, 19.7 mmol)  and DIPEA (2.7 g, 20.8 mmol) in DMA was stirred at 80 ℃ for 5 h and then cooled to r.t.. DMA was evaporated, and the mixture was diluted with water (50 mL) and extracted with dichloromethane (50 mL × 3) . The organic layers were combined and washed with water (100 mL ) and brine (100 mL) , dried over Na2SO4. The solids were filtered out and the filtrate was concentrated in vacuum. The residue was purified by column chromatography on silica gel eluting with dichloromethane/methanol (20:1~10:1) to give N1- (2- (dimethylamino) ethyl) -5-methoxy-N1-methyl-2-nitrobenzene-1, 4-diamine (24a) . MS-ESI (m/z) : 269 [M + 1] +.
N- (2- ( (2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5- ( (4- (1-methylimidazo [1, 5- a] pyridin-3-yl) pyrimidin-2-yl) amino) phenyl) -2-fluoroacrylamide (24)
The title compound 24 was prepared according to the synthetic method of Example 23 by replacing N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-3-nitropyridine-2, 5-diamine (15e) with N1- (2- (dimethylamino) ethyl) -5-methoxy-N1-methyl-2-nitrobenzene-1, 4-diamine (24a) . MS-ESI (m/z) : 517 [M + 1] +.
Example 25
N- (2- ( (2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ( (4- (1-methylindolizin-3-yl ) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (25)
1- (2-ethoxy-2-oxoethyl) -2-ethylpyridin-1-ium bromide (25a)
To a mixture of 2-ethylpyridine (10 g, 0.09 mol) in EtOH (200 mL) was added ethyl 2-bromoacetate (23 g, 0.14 mmol) . The mixture was heated to reflux for 16 h, then cooled to ambient temperature, and concentrated in vacuum. The crude product 1- (2-ethoxy-2-oxoethyl) -2-ethylpyridin-1-ium bromide (25a) was used for next step without further purification. MS-ESI (m/z) : 194 [M] +.
ethyl 1-methylindolizine-3-carboxylate (25b)
To a solution of 1- (2-ethoxy-2-oxoethyl) -2-ethylpyridin-1-ium bromide (25a) (10 g, 0.036 mol) and K2CO3 (15 g, 0.10 mol) in EtOH (100 mL) was added (E) -ethyl 2-cyano-3-ethoxyacrylate (25 g, 0.14 mol) . The mixture was heated to reflux for 15 h, then cooled to ambient temperature, and concentrated in vacuum. The residue was extracted with EtOAc (50 mL × 3) . The combined organic layer was washed with brine, dried over Na2SO4, filtered and concentrated in vacuum. The residue was purified by flash column chromatography eluting with hexane/ethyl acetate (50:1~20:1) to give ethyl 1-methylindolizine-3-carboxylate (25b) . MS-ESI (m/z) : 275 [M + 1] +.
N-methoxy-N, 1-dimethylindolizine-3-carboxamide (25c)
To a solution of ethyl 1-methylindolizine-3-carboxylate (25b) (3.0 g, 0.014 mol) and N, O-Dimethylhydroxylamine hydrochloride (3.6 g, 0.037 mol) in THF (50 mL) was added i-PrMgCl (2 M in Et2O, 35 mL , 0.07 mol) at 0℃. The mixture was stirred for 1 h then quenched with H2O. The mixture was extracted with EtOAc (50 mL × 3) . The combined organic layer was washed with brine, dried over Na2SO4, filtered and concentrated in vacuum. The residue was purified by flash column chromatography eluting with hexane/ethyl acetate (20:1~10:1) to give N-methoxy-N, 1-dimethylindolizine-3-carboxamide (25c) . MS-ESI (m/z) : 219 [M + 1] +.
1- (1-methylindolizin-3-yl) ethanone (25d)
To a solution of N-methoxy-N, 1-dimethylindolizine-3-carboxamide (25c) (2.6 g, 0.012 mol) in THF (50 mL) was added MeMgBr (3 M in Et2O, 8.0 mL , 0.024 mol) at 0℃. The mixture was stirred for 1 h, then quenched with H2O and extracted with EtOAc (50 mL × 3) . The combined organic layer was washed with brine, dried over Na2SO4, filtered and concentrated in vacuum. This crude product 1- (1-methylindolizin-3-yl) ethanone (25d) was used for next step without further purification. MS-ESI (m/z) : 174 [M + 1] +.
(E) -1- (1-methylindolizin-3-yl) -3- (pyrrolidin-1-yl) prop-2-en-1-one (25e)
To a solution of 1- (1-methylindolizin-3-yl) ethanone (25d) (2.0 g, 0.012 mol) in DMF-DMA (25 mL) was added pyrrolidine (10 mL, 0.12 mol) at 25℃. The mixture was stirred for  1 h at 90℃, and then extracted with EtOAc (50 mL × 3) . The organic layer was washed with brine, dried over Na2SO4, filtered and concentrated in vacuum. The residue was purified by column chromatography on silica gel eluting with hexane/ethyl acetate (10:1~2:1) to give (E) -1- (1-methylindolizin-3-yl) -3- (pyrrolidin-1-yl) prop-2-en-1-one (25e) . MS-ESI (m/z) : 255 [M + 1] +.
4- (1-methylindolizin-3-yl) pyrimidin-2-amine (25f)
To a solution of (E) -1- (1-methylindolizin-3-yl) -3- (pyrrolidin-1-yl) prop-2-en-1-one (25e) (1.0 g, 0.0039 mol) and K2CO3 (2.7 g, 0.012 mol) in i-PrOH (10 mL) was added guanidine hydrochloride (1.1 g , 0.19 mol) at 25℃. The mixture was stirred for 16 h at 130℃, then cooled to ambient temperature, extracted with EtOAc (50 mL x 3) . The organic layer was washed with brine, dried over Na2SO4, filtered and concentrated in vacuum. The residue was purified by flash column chromatography eluting with DCM/MeOH (30:1) to give 4- (1-methylindolizin-3-yl) pyrimidin-2-amine (25f) . MS-ESI (m/z) : 225 [M + 1] +.
N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-N5- (4- (1-methylindolizin-3-yl) pyrim idin-2-yl) -3-nitropyridine-2, 5-diamine (25g)
To a solution of 4- (1-methylindolizin-3-yl) pyrimidin-2-amine (25f) (150 mg, 0.67 mmol) , N1- (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N1, N2, N2-trimethylethane-1, 2-diamine (15c) (382 mg, 0.33 mmol) , and Cs2CO3 (0.87 g, 2.67 mmol) in toluene (5 mL) and 1, 4-dioxane (2.5 mL) was added Xantphos (193 mg , 0.33 mmol) and Pd (OAc) 2 (75 mg, 0.33 mmol) at 25℃. The mixture was stirred for 3 h at 110℃, then cooled to ambient temperature, and extracted with EtOAc (50 mL × 3) . The organic layer was washed with brine, dried over Na2SO4, filtered and concentrated in vacuum. The residue was purified by flash column chromatography eluting with DCM/MeOH (40:1~20:1) to give N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-N5- (4- (1-methylindolizin-3-yl) pyrimidin-2-yl) -3-nitropyridine-2, 5-diamine (25g) . MS-ESI (m/z) : 477 [M + 1] +.
N- (2- ( (2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- (4- (1-methylindolizin-3-yl)  pyrimidin-2-ylamino) pyridin-3-yl) acrylamide (25)
The title compound 25 was prepared according to the synthetic method of Example 15 by replacing N1- (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N1, N2, N2-trimethylethane-1, 2-diamine (15h) with N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-N5- (4- (1-methylindolizin-3-yl) pyrimidin-2-yl) -3-nitropyridine-2, 5-diamine (25g) . MS-ESI (m/z) : 501 [M + 1] +.
Example 26
N- (2- ( (2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5- ( (4- (1-methylindolizin-3-yl  ) pyrimidin-2-yl) amino) phenyl) acrylamide (26)
N1- (4-iodo-5-methoxy-2-nitrophenyl) -N1, N2, N2-trimethylethane-1, 2-diamine (26a)
To a solution of N1- (2- (dimethylamino) ethyl) -5-methoxy-N1-methyl-2-nitrobenzene-1, 4-diamine (24a) (3.2 g, 11.9 mmol) in 1 N H2SO4 (50 mL) was added NaNO2 (1.28 g, 17.9 mmol) at 0℃. The mixture was stirred at 0℃ for 15 min, then KI (4.0 g, 23.8 mmol) was added. The mixture was stirred at 90℃ for 1 h. The reaction was cooled to room temperature. The mixture was adjusted to pH 9~10 using sodium carbonate. The resulting mixture was extracted with EA (20 mL × 3) . The organic layers were combined, washed with brine (20 mL) , and dried over Na2SO4. The solids were filtered out and the filtrate was concentrated in vacuum. 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 (26a) . MS-ESI (m/z) : 380 [M + 1] +.
N- (2- ( (2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5- ( (4- (1-methylindolizin-3-yl  ) pyrimidin-2-yl) amino) phenyl) acrylamide (26)
The title compound 26 was prepared according to the synthetic method of Example 25 by replacing N1- (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N1, N2, N2-trimethylethane-1, 2-diamine (15c) with N1- (4-iodo-5-methoxy-2-nitrophenyl) -N1, N2, N2-trimethylethane-1, 2-diamine (26a) . MS-ESI (m/z) : 500 [M + 1] +.
Example 27
N- (5- ( (4- (1-cyanoindolizin-3-yl) pyrimidin-2-yl) amino) -2- ( (2- (dimethylamino) ethyl) (me thyl) amino) -6-methoxypyridin-3-yl) acrylamide (27)
1- (2-ethoxy-2-oxoethyl) pyridin-1-ium bromide (27a)
To a mixture of pyridine (10 g, 0.09 mol) in EtOH (200 mL) was added ethyl 2-bromoacetate (23 g, 0.14 mmol) . The mixture was heated to reflux for 16 h, then cooled to ambient temperature, and concentrated in vacuum. The crude product 1- (2-ethoxy-2-oxoethyl) pyridin-1-ium bromide (27a) was used for next step without further purification. MS-ESI (m/z) : 167 [M] +.
ethyl 1-cyanoindolizine-3-carboxylate (27b)
To a solution of 1- (2-ethoxy-2-oxoethyl) pyridin-1-ium bromide (27a) (10 g, 0.04 mol) and TEA (29 mL, 0.2 mol) in EtOH (250 mL) was added (E) -3-methoxyacrylonitrile (10 g, 0.12 mol) . The mixture was heated to reflux for 15 h, then cooled to ambient temperature, and concentrated in vacuum. The residue was extracted with EtOAc (50 mL x 3) . The organic layers were washed with brine, dried over Na2SO4, and concentrated in vacuum. The residue was purified by flash column chromatography eluting with hexane/ethyl acetate (50:1~20:1) to give ethyl 1-cyanoindolizine-3-carboxylate (27b) . MS-ESI (m/z) : 215 [M + 1] +.
N- (5- ( (4- (1-cyanoindolizin-3-yl) pyrimidin-2-yl) amino) -2- ( (2- (dimethylamino) ethyl) (me thyl) amino) -6-methoxypyridin-3-yl) acrylamide (27)
The title compound 27 was prepared according to the synthetic method of Example 25 by replacing ethyl 1-methylindolizine-3-carboxylate (25b) with ethyl 1-cyanoindolizine-3-carboxylate (27b) . MS-ESI (m/z) : 501 [M + 1] +.
Example 28
N- (5- ( (4- (1-cyanoindolizin-3-yl) pyrimidin-2-yl) amino) -2- ( (2- (dimethylamino) ethyl) (me thyl) amino) -4-methoxyphenyl) acrylamide (28)
The title compound 28 was prepared according to the synthetic method of Example 27 by replacing N1- (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N1, N2, N2-trimethylethane-1, 2-diamine (15c) with N1- (4-iodo-5-methoxy-2-nitrophenyl) -N1, N2, N2-trimethylethane-1, 2-diamine (26a) . MS-ESI (m/z) : 511 [M + 1] +.
Example 29
N- (2- ( (2- (dimethylamino) ethyl) (methyl) amino) -5- ( (4- (indolizin-3-yl) pyrimidin-2-yl) ami no) -6-methoxypyridin-3-yl) acrylamide (29)
The title compound 29 was prepared according to the synthetic method of Example 25 by replacing 2-ethylpyridine with 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) ami no) -4-methoxyphenyl) acrylamide (30)
The title compound 30 was prepared according to the synthetic method of Example 29 by replacing N1- (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N1, N2, N2-trimethylethane-1, 2-diamine (15c) with N1- (4-iodo-5-methoxy-2-nitrophenyl) -N1, N2, N2-trimethylethane-1, 2-diamine (26a) . MS-ESI (m/z) : 486 [M + 1] +.
Example 31
N- (5- ( (4- (1-chloroindolizin-3-yl) pyrimidin-2-yl) amino) -2- ( (2- (dimethylamino) ethyl) (m ethyl) amino) -6-methoxypyridin-3-yl) acrylamide (31)
ethylindolizine-3-carboxylate (31a)
The title compound ethyl indolizine-3-carboxylate (31a) was prepared according to the synthetic method of compound 25b by replacing 2-ethylpyridine with 2-methylpyridine. MS-ESI (m/z) : 190 [M + 1] +.
ethyl 1-chloroindolizine-3-carboxylate (31b)
To a solution of ethyl indolizine-3-carboxylate (31a) (0.42 g, 2.2 mmol) in DMF (5 mL) was added NCS (0.3 g, 2.2 mmol) at 0℃. The mixture was stirred at 25℃ for 1 h, and extracted with EtOAc (50 mL × 3) . The organic layers were washed with brine, dried over Na2SO4, and concentrated in vacuum. The residue was purified by flash column chromatography eluting with hexane/ethyl acetate (50:1) to give ethyl 1-chloroindolizine-3-carboxylate (31b) . MS-ESI (m/z) : 224 [M + 1] +.
N- (5- ( (4- (1-chloroindolizin-3-yl) pyrimidin-2-yl) amino) -2- ( (2- (dimethylamino) ethyl) (m ethyl) amino) -6-methoxypyridin-3-yl) acrylamide (31)
The title compound 31 was prepared according to the synthetic method of Example 25 by replacing compound ethyl 1-methylindolizine-3-carboxylate (25b) with ethyl 1-chloroindolizine-3-carboxylate (31b) . MS-ESI (m/z) : 521 [M + 1] +.
Example 32
N- (5- ( (4- (1-chloroindolizin-3-yl) pyrimidin-2-yl) amino) -2- ( (2- (dimethylamino) ethyl) (m ethyl) amino) -4-methoxyphenyl) acrylamide (32)
The title compound 32 was prepared according to the synthetic method of Example 31 by replacing N1- (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N1, N2, N2-trimethylethane-1, 2-diamine (15c) with N1- (4-iodo-5-methoxy-2-nitrophenyl) -N1, N2, N2-trimethylethane-1, 2-diamine (26a) . MS-ESI (m/z) : 520 [M + 1] +.
Example 33
N- (2- ( (2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ( (4- (3-methylindolizin-1-yl  ) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (33)
3-methylindolizine-1-carbonitrile (33a)
A mixture of 2-bromopropanal (7.26 g, 53.4 mmol) and 2-pyridylacetonitrile (6.3 g, 53.4 mmol) in THF (70 mL) was heated to 70℃ for 24 h. The mixture was concentrated in vacuum. The residue was diluted with EtOAc and hydrochloride acid (20 mL, 2 M) . The organic layer was  washed with sat. aq. NaHCO3 solution, brine, dried over Na2SO4, and concentrated in vacuum. The residue was purified by flash column chromatography eluting with petroleum ether/ethyl acetate (10:1~5:1) to give 3-methylindolizine-1-carbonitrile (33a) . MS-ESI (m/z) : 157 [M + 1] +.
1- (3-methylindolizin-1-yl) ethan-1-imine (33b)
To a solution of 3-methylindolizine-1-carbonitrile (33a) (1.6 g, 10.3 mmol) in THF was added 3 M CH3MgBr (18 mL) at ice-water bath, and the reaction mixture was stirred at ambient temperature for 15 h. The mixture was quenched with sat. aq. NH4Cl solution, and extracted with dichloromethane. The organic layer was washed with brine, dried over Na2SO4 and concentrated in vacuum to give 1- (3-methylindolizin-1-yl) ethan-1-imine (33b) . MS-ESI (m/z) : 173 [M + 1] +.
1- (3-methylindolizin-1-yl) ethan-1-one (33c)
To a solution of 1- (3-methylindolizin-1-yl) ethan-1-imine (33b) (1.0 g, 5.8 mmol) in THF was added dropwise 6 N H2SO4 (60 mL) at ambient temperature, heated to 55℃ for 24 h, then cooled to ambient temperature. The reaction mixture was diluted with water and DCM. The organic layer was separated, washed with sat. aq. NaHCO3 solution, brine, dried over Na2SO4 and concentrated in vacuum. The residue was purified by flash column chromatography to give 1- (3-methylindolizin-1-yl) ethan-1-one (33c) . MS-ESI (m/z) : 174 [M + 1] +.
1- (3-methylindolizin-1-yl) -3- (pyrrolidin-1-yl) prop-2-en-1-one (33d)
To a solution of 1- (3-methylindolizin-1-yl) ethan-1-one (33c) (110mg, 0.7mmol) in DMF was added DMF-DMA (170mg, 1.4 mmol) and pyrrolidine (50mg, 0.7mmol) . The mixture was heated to 90℃ for 1.5 h. After cooled to ambient temperature, the reaction mixture was diluted with water and extracted with EtOAc. The organic layers were washed with brine, dried over Na2SO4 and concentrated to give crude product 1- (3-methylindolizin-1-yl) -3- (pyrrolidin-1-yl) prop-2-en-1-one (33d) . MS-ESI (m/z) : 255 [M + 1] +.
4- (3-methylindolizin-1-yl) pyrimidin-2-amine (33e)
To a solution of 1- (3-methylindolizin-1-yl) -3- (pyrrolidin-1-yl) prop-2-en-1-one (33d) (300 mg, 1.2 mmol) in propanol was added guanidine hydrochloride (337 mg, 3.5 mmol) and K2CO3 (662 mg, 4.8 mmol) . The mixture was heated to 120℃ for 20 h in a sealed tube. After cooled to ambient temperature, the reaction mixture was diluted with water and extracted with EtOAc. The organic layers were washed with brine, dried over Na2SO4 and concentrated to give crude product 4- (3-methylindolizin-1-yl) pyrimidin-2-amine (33e) . MS-ESI (m/z) : 225 [M + 1] +.
N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-N5- (4- (3-methylindolizin-1-yl) pyrim idin-2-yl) -3-nitropyridine-2, 5-diamine (33f)
A mixture of 4- (3-methylindolizin-1-yl) pyrimidin-2-amine (33e) (30.0 mg, 0.13 mmol) , 25c (76.0 mg, 0.20 mmol) , Pd (OAc) 2 (15.0 mg, 0.07 mmol) , Xantphos (38.0 mg, 0.07 mmol) , and Cs2CO3 (170 mg, 0.52 mmol) in toluene (1mL) and dioxane (0.5 mL) was heated under nitrogen at 110 ℃ for 3 h. After cooled to room temperature, the reaction mixture was diluted with water and extracted with EtOAc. The organic layers were washed with brine, dried over Na2SO4 and concentrated. 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-methylindolizin-1-yl) pyrimidin-2-yl) -3-nitropyridine-2, 5-diamine (33f) . MS-ESI (m/z) : 477 [M + 1] +.
N- (2- ( (2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ( (4- (3-methylindolizin-1-yl  ) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (33)
The title compound 33 was prepared according to the synthetic method of Example 15 by replacing N1- (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N1, N2, N2-trimethylethane-1, 2-diamine (15h) with compound N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-N5- (4- (3-methylindolizin-1-yl) pyrimidin-2-yl) -3-nitropyridine-2, 5-diamine (33f) . MS-ESI (m/z) : 501 [M + 1] +.
Example 34
N- (2- ( (2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5- ( (4- (3-methylindolizin-1-yl  ) pyrimidin-2-yl) amino) phenyl) acrylamide (34)
The title compound 34 was prepared according to the synthetic method of Example 33 by replacing N1- (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N1, N2, N2-trimethylethane-1, 2-diamine (15c) with N1- (4-iodo-5-methoxy-2-nitrophenyl) -N1, N2, N2-trimethylethane-1, 2-diamine (26a) . MS-ESI (m/z) : 500 [M + 1] +.
Example 35
N- (5- ( (4- (3-cyanoindolizin-1-yl) pyrimidin-2-yl) amino) -2- ( (2- (dimethylamino) ethyl) (me thyl) amino) -6-methoxypyridin-3-yl) acrylamide (35)
1-acetylindolizine-3-carbonitrile (35a)
A mixture of 1- (cyanomethyl) pyridin-1-ium bromide (10 g, 50.3 mmol) , 4-methoxybut-3-en-2-one (5.1 g, 50.3 mmol) and triethylamine (10 g, 100.6 mmol) in EtOH (100 mL) was heated to 80℃ for 7 h. The reaction mixture was concentrated in vacuum. The residue was diluted with DCM and water. The organic layer was separated, washed with brine, dried over Na2SO4 and concentrated. The residue was stirred for 0.5 h in iso-propanol. Yellow solid was collected by filtration and dried in vacuum to give 1-acetylindolizine-3-carbonitrile (35a) . MS-ESI (m/z) : 185 [M + 1] +.
1- (3- (pyrrolidin-1-yl) acryloyl) indolizine-3-carbonitrile (35b)
To a solution of 1-acetylindolizine-3-carbonitrile (35a) (400 mg, 2.17 mmol) in DMF was added DMF-DMA (516 mg, 4.34 mmol) and pyrrolidine (308 mg, 4.34 mmol) . The reaction mixture was heated to 90℃ for 1 h. After cooled to ambient temperature, the reaction mixture was diluted with water and extracted with EtOAc. The organic layers were washed with brine, dried over Na2SO4 and concentrated to give the crude product 1- (3- (pyrrolidin-1-yl) acryloyl) indolizine-3-carbonitrile (35b) . 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) (637 mg, 2.4 mmol) in 1-butanol was added guanidine hydrochloride (690 mg, 7.2 mmol) and K2CO3 (1.3 g,  9.6 mmol) . The mixture was heated to 115℃ for 20 h. After cooled to ambient temperature, the reaction mixture was diluted with water and extracted with EtOAc. The organic layers were washed with brine, dried over Na2SO4 and concentrated to give the crude product 1- (2-aminopyrimidin-4-yl) indolizine-3-carbonitrile (35c) . MS-ESI (m/z) : 236 [M + 1] +.
N- (5- ( (4- (3-cyanoindolizin-1-yl) pyrimidin-2-yl) amino) -2- ( (2- (dimethylamino) ethyl) (me thyl) amino) -6-methoxypyridin-3-yl) acrylamide (35)
The title compound 35 was prepared according to the synthetic method of Example 33 by replacing 4- (3-methylindolizin-1-yl) pyrimidin-2-amine (33e) with 1- (2-aminopyrimidin-4-yl) indolizine-3-carbonitrile (35c) . MS-ESI (m/z) : 512 [M + 1] + .
Example 36
N- (5- ( (4- (3-cyanoindolizin-1-yl) pyrimidin-2-yl) amino) -2- ( (2- (dimethylamino) ethyl) (me thyl) amino) -4-methoxyphenyl) acrylamide (36)
The title compound 36 was prepared according to the synthetic method of Example 35 by replacing N1- (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N1, N2, N2-trimethylethane-1, 2-diamine (15c) with N1- (4-iodo-5-methoxy-2-nitrophenyl) -N1, N2, N2-trimethylethane-1, 2-diamine (26a) . MS-ESI (m/z) : 511 [M + 1] +.
Example 37
N- (2- ( (2- (dimethylamino) ethyl) (methyl) amino) -5- ( (4- (indolizin-1-yl) pyrimidin-2-yl) ami no) -6-methoxypyridin-3-yl) acrylamide (37)
1- (2-ethoxy-2-oxoethyl) pyridin-1-ium bromide (37a)
A mixture of pyridine (2.00 g, 25.30 mmol) and ethyl 2-bromoacetate (6.3 g, 38.00 mmol) in EtOH (20 mL) was heated under nitrogen at 70℃ for 1 h. The solvent was evaporated under reduced pressure. The residue was filtered through celite pad and the pad was washed with PE, gave 6.9 g 1- (2-ethoxy-2-oxoethyl) pyridin-1-ium bromide (37a) . MS-ESI (m/z) : 167 [M + 1] +.
ethyl 1-acetylindolizine-3-carboxylate (37b)
To a mixture of 1- (2-ethoxy-2-oxoethyl) pyridin-1-ium bromide (37a) (5.50 g, 15.10 mmol) and 4-methoxybut-3-en-2-one (2.3 g, 22.3 mmol) in EtOH (65 mL) at 20℃, was added TEA (4.50 g, 44.60 mmol) . The mixture was heated at 80℃ overnight. The mixture was cooled to room temperature and diluted with water. The solution was extracted with EA (3 × 50mL) . The organic layer was removed 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) (2.60 g, 11.3 mmol) in MeOH (30 mL) was added 10%NaOH (2.0 g, 50.6m mol) at 20℃. The mixture was heated at 65℃for 1 h. After the solvent was evaporated under reduced pressure, PPA (10.0g) and MeOH (40 ml) was add to the residue and the mixture was heated at 65℃ for 40 min. The mixture was cooled to room temperature and diluted with water. The solution was extracted with DCM (3 × 50mL) . The extracts were washed with NaHCO3, brine, dried over Na2SO4 and concentrated to give 1- (indolizin-1-yl) ethan-1-one (37c) (1.23 g) . MS-ESI (m/z) : 160 [M + 1] +.
N- (5- (4- (indolizin-1-yl) pyrimidin-2-ylamino) -2- (N- (2- (dimethylamino) ethyl) -N-methyla mino) -6-methoxypyridin-3-yl) acrylamide (37)
The title compound 37 was prepared according to the synthetic method of Example 35 by replacing 1-acetylindolizine-3-carbonitrile (35a) with1- (indolizin-1-yl) ethan-1-one (37c) . MS-ESI (m/z) : 487 [M + 1] +.
Example 38
N- (2- ( (2- (dimethylamino) ethyl) (methyl) amino) -5- ( (4- (indolizin-1-yl) pyrimidin-2-yl) ami no) -4-methoxyphenyl) acrylamide (38)
The title compound 38 was prepared according to synthetic the method of Example 37 by replacing N1- (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N1, N2, N2-trimethylethane-1, 2-diamine (15c) with N1- (4-iodo-5-methoxy-2-nitrophenyl) -N1, N2, N2-trimethylethane-1, 2-diamine (26a) . MS-ESI (m/z) : 486 [M + 1] +.
Example 39
N- (5- ( (4- (3-chloroindolizin-1-yl) pyrimidin-2-yl) amino) -2- ( (2- (dimethylamino) ethyl) (m ethyl) amino) -6-methoxypyridin-3-yl) acrylamide (39)
1- (3-chloroindolizin-1-yl) ethan-1-one (39a)
A mixture of 1- (indolizin-1-yl) ethan-1-one (37c) (600 mg, 3.8 mmol) , CuCl (962 mg, 5.7 mmol) in CH3CN (6 mL) was stirred at r.t. for 4 h. The mixture was diluted with water (10 mL) and extracted with DCM (3 × 10 mL) . The extracts were dried over Na2SO4. Solvents were evaporated under reduced pressure. The residue was purified through flash column chromatography eluting with PE/EA (8:1~6:1) to give 1- (3-chloroindolizin-1-yl) ethan-1-one (39a) . MS-ESI (m/z) : 194 [M + 1] +.
N- (5- ( (4- (3-chloroindolizin-1-yl) pyrimidin-2-yl) amino) -2- ( (2- (dimethylamino) ethyl) (m ethyl) amino) -6-methoxypyridin-3-yl) acrylamide (39)
The title compound 39 was prepared according to the synthetic method of Example 35 by replacing 1-acetylindolizine-3-carbonitrile (35a) with1- (3-chloroindolizin-1-yl) ethan-1-one (39a) . MS-ESI (m/z) : 521 [M + 1] +.
Example 40
N- (5- ( (4- (3-chloroindolizin-1-yl) pyrimidin-2-yl) amino) -2- ( (2- (dimethylamino) ethyl) (m ethyl) amino) -4-methoxyphenyl) acrylamide (40)
The title compound 40 was prepared according to the synthetic method of Example39by replacing N1- (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N1, N2, N2-trimethylethane-1, 2-diamine (15c) withN1- (4-iodo-5-methoxy-2-nitrophenyl) -N1, N2, N2-trimethylethane-1, 2-diamine (26a) . MS-ESI (m/z) : 520 [M + 1] +.
Example 41
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 (41)
1- (2-chloropyrimidin-4-yl) -3-methyl-1H-indole (41a)
To a solution of 2, 4-dichloropyrimidine (2.89 g, 20.0 mmol) and 3-methyl-1H-indole (1.31 g, 10.0 mmol) in DMA (50 mL) at room temperature was added HOBT (306 mg, 2.0 mmol) followed K2CO3 (1.93 g, 14.0 mmol) . The mixture was stirred at 70℃ for 24 h. The mixture was diluted with water (50 mL) and extracted with ethyl acetate (2 × 30 mL) . The extracts were washed with brine and dried over Na2SO4. Solvent was evaporated. The residue was purified by column chromatography on silica gel eluting with PE/EtOAc (10:1~5:1) to give 1- (2-chloropyrimidin-4-yl) -3-methyl-1H-indole (41a) as yellow solid (1 g, 40%) .
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 (41)
The title compound 41 (17.3 mg) was prepared by using the same procedure as described for 1 by replacing 3- (2-chloropyrimidin-4-yl) -1-methyl-1H-indole (1h) with 1- (2-chloropyrimidin-4-yl) -3-methyl-1H-indole (41a) . MS-ESI (m/z) : 501 [M + 1] +
Example 42
N- (2- ( (2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5- ( (4- (3-methyl-1H-indol-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)
The mixture of 1- (2-chloropyrimidin-4-yl) -3-methyl-1H-indole (41a) (278 mg, 1.14 mmol) , 4-fluoro-2-methoxy-5-nitroaniline (213 mg, 1.14 mmol) (prepared by following the literature, WO2013/14448) , and p-TsOH·H2O (260 mg, 1.37 mmol) in 2-pentanol (4 mL) was heated at 105℃ for 2 h. The mixture was filtrated, and the filtration was evaporated under reduced pressure to give N- (4-fluoro-2-methoxy-5-nitrophenyl) -4- (3-methyl-1H-indol-1-yl) pyrimidin-2-amine (42a) (320 mg, 72%) . MS-ESI (m/z) : 394 [M + 1] +.
N1- (2- (dimethylamino) ethyl) -5-methoxy-N1-methyl-N4- (4- (3-methyl-1H-indol-1-yl) pyri midin-2-yl) -2-nitrobenzene-1, 4-diamine (42b)
The solution of N- (4-fluoro-2-methoxy-5-nitrophenyl) -4- (3-methyl-1H-indol-1-yl) pyrimidin-2-amine (42a) (160 mg, 0.407 mmol) , N1, N1, N2-trimethylethane-1, 2-diamine (0.064 mL , 0.489 mmol) and DIPEA (0.088 mL, 0.529 mmol) in DMA (1.6 mL) was heated at 85℃ for 3 h. The solvent was evaporated under reduced pressure and the residue was purified by column chromatography on silica gel eluting with DCM/methanol/ammonia (92:6:2) to provide 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) (139 mg, 73%) . MS-ESI (m/z) : 476 [M + 1] +.
N1- (2- (dimethylamino) ethyl) -5-methoxy-N1-methyl-N4- (4- (3-methyl-1H-indol-1-yl) pyri midin-2-yl) -2-nitrobenzene-1, 4-diamine (42c)
The mixture of 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) (139 mg, 0.293 mmol) , iron powder (98 mg, 1.76 mmol) , and NH4Cl (22.0 mg, 0.293 mmol) in ethanol-H2O (3:1, 15 mL) was heated at reflux for 1 h. The solid was removed by filtration through celite and washed with ethyl acetate. The filtrates were diluted with ethyl acetate and washed with water and brine. The solvent was evaporated under reduced pressure and the residue was column chromatography on silica gel eluting with DCM-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-nitrobenzene-1, 4-diamine (42c) (120 mg, 92%) . MS-ESI (m/z) : 446 [M + 1] +.
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)
To a mixture of 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) (120 mg, 0.27 mmol) in THF-H2O (10:1, 3 mL) at 0℃ was added 3-chloropropanoyl chloride (0.031 mL, 0.32 mmol) . The mixture was stirred at 0℃ for 2 h. Ammonium Hydroxidethe (0.5mL) was added. The mixture was diluted  with water and extracted with ethyl acetate. Solvents were evaporated under reduced pressure. The residue was purified by column chromatography on silica gel eluting with 2-5%methanol in DCM 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) (100 mg, 69%) . MS-ESI (m/z) : 536 [M + 1] +.
N- (2- ( (2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5- ( (4- (3-methyl-1H-indol-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) propanamide (42d) (100 mg, 0.187 mmol) in THF-H2O (10:1, 2 mL) at room temperature was added NaOH (60 mg, 1.5 mmol) . The mixture was stirred for 2 h. The mixture was diluted with water and extracted with ethyl acetate. Solvents were evaporated under reduced pressure. The residue was purified by column chromatography on silica gel eluting with 1-2%methanol in DCM to give N- (2- ( (2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5- ( (4- (3-methyl-1H-indol-1-yl) pyrimidin-2-yl) amino) phenyl) acryl amide (42) (60 mg, 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-methylpiperaz in-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) p henyl) acrylamide (43a)
The title compound 43a (10 mg) was prepared according to the synthetic method of Example 41 by replacing N1, N1, N2-trimethylethane-1, 2-diamine with tert-butyl piperazine-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-methylpiperazin-1-yl) pyridin-3-yl) acrylamide (43)
The title compound 43 (4.9 mg) was prepared according to the synthetic method of Example 6 by replacing N- (6-Ethoxy-2- (methyl (2- (methylamino) ethyl) amino) -5- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-ylamino) pyridin-3-yl) acrylamide (5) with N- (4-methoxy-5- ( (4- (3-methyl-1H-indol-1-yl) pyrimidin-2-yl) amino) -2- (piperazin-1-yl) phenyl) acrylamide (43a) . 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-methylpiperaz in-1-yl) phenyl) acrylamide (44)
The title compound 44 was prepared according to the synthetic method of Example 42 by replacing N1, N1, N2-trimethylethane-1, 2-diamine with 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) (m ethyl) amino) -6-methoxypyridin-3-yl) acrylamide (45)
1- (2-chloropyrimidin-4-yl) -1H-indole-3-carbonitrile (45b)
The title compound 1- (2-chloropyrimidin-4-yl) -1H-indole-3-carbonitrile (45b) (220 mg) was prepared according to the synthetic method of 41a by replacing 3-methyl-1H-indole with 1H-indole-3-carbonitrile (45a) (This reagent was prepared according to the synthetic method described in literature: 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) (m ethyl) amino) -6-methoxypyridin-3-yl) acrylamide (45)
The title compound 45 (7.0 mg) was prepared according to the synthetic method of Example 15 by replacing 3- (2-chloropyrimidin-4-yl) -7-fluoro-1-methyl-1H-indole (15g) with 1- (2-chloropyrimidin-4-yl) -1H-indole-3-carbonitrile (45b) . 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) (m ethyl) amino) -4-methoxyphenyl) acrylamide (46)
The title compound 46 (5 mg) was prepared according to the synthetic method of Example 45 by replacing N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-3-nitropyridine-2, 5-diamine (15e) with N1- (2- (dimethylamino) ethyl) -5-methoxy-N1-methyl-2-nitrobenzene-1, 4-diamine (24a) . MS-ESI (m/z) : 511 [M + 1] +.
Example 47
N- (5- ( (4- (1-chloroimidazo [1, 5-a] pyridin-3-yl) pyrimidin-2-yl) amino) -2- ( (2- (dimethyla mino) ethyl) (methyl) amino) -6-methoxypyridin-3-yl) acrylamide (47)
2- (methylthio) -N- (pyridin-2-yl) pyrimidine-4-carboxamide (47a)
A mixture of pyridin-2-ylmethanamine (900 mg, 8.3 mmol) , 2- (methylthio) pyrimidine -4-carboxylic acid (1 g, 5.9 mmol) (prepared by following the literature WO2006/117368) , HOBT (2.4 g, 17.7 mmol) , EDCI (3.4 g, 17.7 mmol) , TEA (1.78 g, 17.7 mmol) in DMF (10 ml) was stirred at r.t. for 2 h. The reaction was cooled to room temperature and quenched by water (10 mL) . The mixture was adjusted to pH 9~10 using sodium carbonate. The resulting mixture was extracted with EA (15 mL × 3) . The organic layers were combined and washed with brine (20 mL) , dried over Na2SO4. The solids were filtered out and the filtrate was concentrated in vacuum. The residue was purified by column chromatography on silica gel eluting with PE/EA (2:1~1:1) to give 2- (methylthio) -N- (pyridin-2-yl) pyrimidine-4-carboxamide (47a) . MS-ESI (m/z) : 247 [M + 1] +.
3- (2- (methylthio) pyrimidin-4-yl) imidazo [1, 5-a] pyridine (47b)
To a solution of 2- (methylthio) -N- (pyridin-2-yl) pyrimidine-4-carboxamide (47a) (370 mg, 1.5 mmol) in CH3CN (2 mL) was added POCl3 (2.5 mL) , the mixture was stirred at 85 ℃ for 20 h. The reaction was cooled to room temperature and quenched by water (10 mL) . The mixture was adjusted to pH 8-9 using NaOH. The resulting mixture was extracted with DCM (8 mL × 3) . The organic layers were combined and washed with brine (20 mL) , dried over Na2SO4 and concentrated  in vacuum to give crude product 3- (2- (methylthio) pyrimidin-4-yl) imidazo [1, 5-a] pyridine (47b) MS-ESI (m/z) : 243 [M + 1] +
1-chloro-3- (2- (methylthio) pyrimidin-4-yl) imidazo [1, 5-a] pyridine (47c)
To a solution of 3- (2- (methylthio) pyrimidin-4-yl) imidazo [1, 5-a] pyridine (200 mg, 0.83 mmol) in DMF (2 mL) was add NCS (144 mg, 1.08 mmol) at r.t.. The mixture was stirred at r.t. for 2 h. The reaction was quenched by water (5 mL) . The resulting mixture was extracted with DCM (5 mL × 2) . The organic layers were combined and washed with brine (10 mL) , dried over Na2SO4 and concentrated in vacuum to give crude product 1-chloro-3- (2- (methylthio) pyrimidin-4-yl) imidazo [1, 5-a] pyridine (47c) . MS-ESI (m/z) : 277 [M + 1] +
4- (1-chloroimidazo [1, 5-a] pyridin-3-yl) pyrimidin-2-ol (47d)
To a solution of 1-chloro-3- (2- (methylthio) pyrimidin-4-yl) imidazo [1, 5-a] pyridine (47c) (150 mg, 0.54 mmol) in NMP (5mL) was add 10%NaOH (2 mL) . The mixture was stirred at 100 ℃for 24 h. The reaction was cooled to room temperature. The mixture was adjusted to pH 5-6 using 2 N HCl. The resulting mixture was extracted with DCM (10 mL × 3) . The organic layers were combined and washed with brine (20 mL) , dried over Na2SO4 and concentrated in vacuum to give crude product 4- (1-chloroimidazo [1, 5-a] pyridin-3-yl) pyrimidin-2-ol (47d) . MS-ESI (m/z) : 247 [M + 1] +
1-chloro-3- (2-chloropyrimidin-4-yl) imidazo [1, 5-a] pyridine (47e)
To a solution of 4- (1-chloroimidazo [1, 5-a] pyridin-3-yl) pyrimidin-2-ol (47d) (140 mg, 0.57 mmol) in CH3CN (1 mL) was added POCl3 (1 mL) . The mixture was stirred at 80℃ for 1.5 h. The reaction was cooled to room temperature and quenched by water (5 mL) . The mixture was adjusted to pH 8-9 using Na2CO3. The resulting mixture was extracted with DCM (8 mL × 3) . The organic layers were combined and washed with brine (20 mL) , dried over Na2SO4 and concentrated in vacuum to give crude product 1-chloro-3- (2-chloropyrimidin-4-yl) imidazo [1, 5-a] pyridine (47e) . MS-ESI (m/z) : 265 [M + 1] +
N1- (4- (1-chloroimidazo [1, 5-a] pyridin-3-yl) pyrimidin-2-yl) -N4- (2- (dimethylamino) ethyl  ) -2-methoxy-N4-methyl-5-nitrobenzene-1, 4-diamine (47f)
A mixture of 1-chloro-3- (2-chloropyrimidin-4-yl) imidazo [1, 5-a] pyridine (47e) (54 mg, 0.20 mmol) , N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-3-nitropyridine-2, 5-diamine (15e) (61 mg, 0.22 mmol) and MsOH (27 mg, 0.24 mmol) in PrOH (2.5 mL) was stirred at 90℃ for 3 h. The reaction was cooled to room temperature and quenched by water (10 mL) . The mixture was adjusted to pH 8-9 using Na2CO3. The resulting mixture was extracted with ethyl acetate (30 mL ×3) .The organic layers were combined and washed with water (30 mL) and brine (30 mL) , dried over Na2SO4. The solids were filtered out and the filtrate was concentrated in vacuum. The residue was purified by chromatography on silica gel column eluting with dichloromethane/methanol (10:1) give N1- (4- (1-chloroimidazo [1, 5-a] pyridin-3-yl) pyrimidin-2-yl) -N4- (2- (dimethylamino) ethyl) -2-methoxy -N4-methyl-5-nitrobenzene-1, 4-diamine (47f) . MS-ESI (m/z) : 498 [M + 1] +..
N- (5- ( (4- (1-chloroimidazo [1, 5-a] pyridin-3-yl) pyrimidin-2-yl) amino) -2- ( (2- (dimethyla mino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide (47)
The title compound (47) was prepared according to the synthetic method of Example 15 by replacing N2- (2- (dimethylamino) ethyl) -N5- (4- (7-fluoro-1-methyl-1H-indol-3-yl) pyrimidin-2-yl) -6-methoxy-N2-methyl-3-nitropyridine-2, 5-diamine (15h) with N1- (4- (1-chloroimidazo [1, 5-a] pyridin-3-yl) pyrimidin-2-yl) -N4- (2- (dimethylamino) ethyl) -2-methoxy -N4-methyl-5-nitrobenzene-1, 4-diamine (47f) . MS-ESI (m/z) : 468 [M + 1] +.
Example 48
N- (5- ( (4- (1-chloroimidazo [1, 5-a] pyridin-3-yl) pyrimidin-2-yl) amino) -2- ( (2- (dimethyla  mino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide (48)
The title compound (48) was prepared according to the synthetic method of Example 47 by replacing N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-3-nitropyridine-2, 5-diamine (15e) with N1- (4-iodo-5-methoxy-2-nitrophenyl) -N1, N2, N2-trimethylethane-1, 2-diamine (24a) . MS-ESI (m/z) : 521 [M + 1] +.
Example 49
N- (2- ( (2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ( (4- (1- (trifluoromethyl) imi dazo [1, 5-a] pyridin-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (49)
1-iodo-3- (2- (methylthio) pyrimidin-4-yl) imidazo [1, 5-a] pyridine (49a)
To a solution of 3- (2- (methylthio) pyrimidin-4-yl) imidazo [1, 5-a] pyridine (47b) (300 mg, 1.24 mmol) in DMF (5 mL) was add NIS (362 mg, 1.61 mmol) at r.t.. The mixture was stirred at r.t. for 2 h. The reaction was quenched by water (5 mL) . The resulting mixture was extracted with DCM (8 mL × 2) . The organic layers were combined and washed with brine (10 mL) , dried over Na2SO4 and concentrated in vacuum to give crude product 1-iodo-3- (2- (methylthio) pyrimidin-4-yl) imidazo [1, 5-a] pyridine (49a) . MS-ESI (m/z) : 369 [M + 1] +
3- (2- (methylthio) pyrimidin-4-yl) -1- (trifluoromethyl) imidazo [1, 5-a] pyridine (49b)
A mixture of 1-iodo-3- (2- (methylthio) pyrimidin-4-yl) imidazo [1, 5-a] pyridine (49a) (200 mg, 0.54 mmol) , (Trifluoromethyl) trimethylsilane (153 mg, 1.08 mmol) , Phen (194 mg, 1.08 mmol) , Trimethyl borate (112 mg, 1.08 mmol) , Potassium fluoride (63 mg, 1.08 mmol) and copper (I) iodide (154 mg, 0.81 mmol) in DMSO (5 mL) was stirred at 60℃ overnight. The reaction was cooled to room temperature and quenched by water (10 mL) . The resulting mixture was extracted with ethyl acetate (10 mL × 3) , the organic layers were combined and washed with water (20 mL) and brine (20 mL) , dried over Na2SO4. The solids were filtered out and the filtrate was concentrated in vacuum. The residue was purified by column chromatography on silica gel eluting with acetate/petroleum ether (1:2) to give 3- (2- (methylthio) pyrimidin-4-yl) -1- (trifluoromethyl) imidazo [1, 5-a] pyridine (49b) . MS-ESI (m/z) : 311 [M + 1] +..
N- (2- ( (2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ( (4- (1- (trifluoromethyl) imi dazo [1, 5-a] pyridin-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (49)
The title compound 49 was prepared according to the synthetic method of Example 47 by replacing 1-chloro-3- (2- (methylthio) pyrimidin-4-yl) imidazo [1, 5-a] pyridine (47c) with  3- (2- (methylthio) pyrimidin-4-yl) -1- (trifluoromethyl) imidazo [1, 5-a] pyridine (49b) . MS-ESI (m/z) : 556 [M + 1] +
Example 50
N- (2- ( (2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5- ( (4- (1- (trifluoromethyl) imi dazo [1, 5-a] pyridin-3-yl) pyrimidin-2-yl) amino) phenyl) acrylamide (50)
The title compound 50 was prepared according to the synthetic method of Example 47 by replacing N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-3-nitropyridine-2, 5-diamine (15e) with N1- (2- (dimethylamino) ethyl) -5-methoxy-N1-methyl-2-nitrobenzene-1, 4-diamine (24a) . MS-ESI (m/z) : 555 [M + 1] +.
Example 51
N- (2- ( (2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ( (4- (1-methylimidazo [1, 5- a] pyridin-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (51)
1-methyl-3- (2- (methylthio) pyrimidin-4-yl) imidazo [1, 5-a] pyridine (51a)
The title compound 51a was prepared according to the synthetic method of 47b by replacing pyridin-2-ylmethanamine with 1- (pyridin-2-yl) ethan-1-amine. MS-ESI (m/z) : 257 [M + 1] +.
3- (2-chloropyrimidin-4-yl) -1-methylimidazo [1, 5-a] pyridine (51b)
The title compound 51b was prepared according to the synthetic method of 47e by replacing 1-chloro-3- (2- (methylthio) pyrimidin-4-yl) imidazo [1, 5-a] pyridine (47c) with 1-methyl-3- (2- (methylthio) pyrimidin-4-yl) imidazo [1, 5-a] pyridine (51a) . MS-ESI (m/z) : 245 [M + 1] +.
N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-N5- (4- (1-methylimidazo [1, 5-a] pyrid  in-3-yl) pyrimidin-2-yl) pyridine-2, 3, 5-triamine (51c)
The title compound (51c) was prepared according to the synthetic method of 15i by replacing 3- (2-chloropyrimidin-4-yl) -7-fluoro-1-methyl-1H-indole (15g) with 3- (2-chloropyrimidin-4-yl) -1-methylimidazo [1, 5-a] pyridine (51b) . MS-ESI (m/z) : 448 [M + 1] +.
N- (2- ( (2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ( (4- (1-methylimidazo [1, 5- a] pyridin-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (51)
The title compound 51 was prepared according to the synthetic method of Example 15 by replacing N2- (2- (dimethylamino) ethyl) -N5- (4- (7-fluoro-1-methyl-1H-indol-3-yl) pyrimidin-2-yl) -6-methoxy-N2-methylpyridine-2, 3, 5-triamine (15i) with N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-N5- (4- (1-methylimidazo [1, 5-a] pyridin-3-yl) pyrimidin-2-yl) pyridine-2, 3, 5-tri amine (51c) . MS-ESI (m/z) : 502 [M + 1] +.
Example 52
N- (2- ( (2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5- ( (4- (1-methylimidazo [1, 5- a] pyridin-3-yl) pyrimidin-2-yl) amino) phenyl) acrylamide (52)
The title compound 52 was prepared according to the synthetic method of Example 51 by replacing N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-3-nitropyridine-2, 5-diamine (15e) with N1- (2- (dimethylamino) ethyl) -5-methoxy-N1-methyl-2-nitrobenzene-1, 4-diamine (24a) . MS-ESI (m/z) : 501 [M + 1] +.
Example 53
N- (2- ( (2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ( (4- (1-methylimidazo [1, 5- a] pyridin-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) -2-fluoroacrylamide (53)
The title compound (53) was prepared according to the method of Example 23 by replacing N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-N5- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) pyridine-2, 3, 5-triamine (23b) with N2- (2- (dimethylamino) ethyl) -6-methoxy -N2-methyl-N5- (4- (1-methylimidazo [1, 5-a] pyridin-3-yl) pyrimidin-2-yl) pyridine-2, 3, 5-triamine (51c) . MS-ESI (m/z) : 520 [M + 1] +.
Example 54
N- (5- ( (4- (1, 2-dihydropyrrolo [3, 2, 1-hi] indol-5-yl) pyrimidin-2-yl) amino) -2- ( (2- (dimethy lamino) ethyl) (methyl) amino) -6-methoxypyridin-3-yl) acrylamide (54)
2- (2-nitrophenyl) ethanol (54a)
A mixture of 1-methyl-2-nitrobenzene (48 g, 0.35 mol) , Formaldehyde (10.4 g, 0.35 mol) , 20%NaOH (3.6 ml) and DMSO was heated at 50℃ for 2 h. Then the mixture was cooled to r.t., diluted with water (200 mL) and extracted with Ethyl acetate (300 mL × 3) . The organic layers were combined and washed with water (200 mL × 2) and brine (200 mL) , dried over Na2SO4. The solids were filtered out and the filtrate was concentrated in vacuum. The crude product was purified by column chromatography on silica gel eluting with ethyl acetate/petroleum ether (1:1) to give 2- (2-nitrophenyl) ethanol (54a) as brown oil (14.4 g, 25%) .
tert-butyldimethyl (2-nitrophenethoxy) silane (54b)
To a solution of 2- (2-nitrophenyl) ethanol (54a) (1.7 g, 0.01 mol) in dry DCM (15 ml) was added the imidazole (0.88 g, 0.013 mol) followed by tert-Butyldimethylsilyl chloride (2.1 g, 0.014 mol) at r.t for 2 h. Then the mixture was filtered, diluted with DCM (30 mL) and washed with 1 N HCl (30 mL) , water and aq NaHCO3. The organic layers were combined and dried over Na2SO4. The solids were filtered out and the filtrate was concentrated in vacuum. The crude product was concentrated to give tert-butyldimethyl (2-nitrophenethoxy) silane (54b) as brown oil (2.8 g, 97%)
7- (2- ( (tert-butyldimethylsilyl) oxy) ethyl) -1H-indole (54c)
To a solution of tert-butyldimethyl (2-nitrophenethoxy) silane (54b) (4.5 g, 16 mmol) in dry THF (50 ml) was added dropwise the vinylmagnesium bromide (56 ml, 56 mmol) under N2 at -70℃. The mixture was slowly warm to r.t. The mixture was poured into aq NH4Cl and extracted with ethyl acetate (50 mL) . The organic layers were combined and dried over Na2SO4. The solids were filtered out and the filtrate was concentrated in vacuum. The crude product was purified by column chromatography on silica gel eluting with ethyl acetate/petroleum ether (1:100) to give  7- (2- ( (tert-butyldimethylsilyl) oxy) ethyl) -1H-indole (54c) as yellow oil (2.6 g, 59%) . MS-ESI (m/z) : 276 [M + 1] +
2- (3- (2-chloropyrimidin-4-yl) -1H-indol-7-yl) ethanol (54d)
The title compound 54d (0.33 g, 30%, brown solid) was prepared by using the same procedure as described for 15g by replacing 7-fluoro-1-methyl-1H-indole (15f) with 7- (2- ( (tert-butyldimethylsilyl) oxy) ethyl) -1H-indole (54c) . MS-ESI (m/z) : 274 [M + 1] +
7- (2-chloroethyl) -3- (2-chloropyrimidin-4-yl) -1H-indole (54e)
To the suspension of 2- (3- (2-chloropyrimidin-4-yl) -1H-indol-7-yl) ethanol (54d) (50 mg, 0.18 mmol) in dry CAN (5 ml) was added dropwise thionyl chloride (0.44 mg, 0.36 mmol) at 0℃.And then the resulting mixture was stirred for overnight at r.t. The mixture was cooled to r.t, quenched with aq NaHCO3 (20 ml) and extracted with ethyl acetate (20 mL) . The organic layers were combined and washed with water (20 mL × 2) and brine (200 mL) , dried over anhydrous sodium sulfate. The solids were filtered out and the filtrate was concentrated under vacuum. The crude product was purified by column chromatography on silica gel eluting with ethyl acetate/petroleum ether (1:3) to give 7- (2-chloroethyl) -3- (2-chloropyrimidin-4-yl) -1H-indole (54e) as brown solid (0.30 g, 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) (33 mg, 0.11 mmol) in DMF (3 ml) was added Cs2CO3 (78 mg, 0.22 mmol) at r.t, and then was heated to 60℃ for 10 h. The mixture was cooled to r.t, quenched with water (20 ml) and extracted with ethyl acetate (20 mL) . The organic layers were combined and washed with water (20 mL × 2) and brine (20 mL) , dried over Na2SO4. The solids were filtered out and the filtrate was concentrated in vacuum to give 5- (2-chloropyrimidin-4-yl) -1, 2-dihydropyrrolo [3, 2, 1-hi] indole (54f) as brown solid (0.30 g, 100%) . MS-ESI (m/z) : 256 [M + 1] +
N- (5- ( (4- (1, 2-dihydropyrrolo [3, 2, 1-hi] indol-5-yl) pyrimidin-2-yl) amino) -2- ( (2- (dimethy lamino) ethyl) (methyl) amino) -6-methoxypyridin-3-yl) acrylamide (54)
The title compound 54 (5.0 mg) was prepared by using the same procedure as described for 15 by replacing 3- (2-chloropyrimidin-4-yl) -7-fluoro-1-methyl-1H-indole (15g) with 5- (2-chloropyrimidin-4-yl) -1, 2-dihydropyrrolo [3, 2, 1-hi] indole (54f) . MS-ESI (m/z) : 513 [M + 1] +.
Example 55
N- (5- ( (4- (1, 2-dihydropyrrolo [3, 2, 1-hi] indol-5-yl) pyrimidin-2-yl) amino) -2- ( (2- (dimethy lamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide (55)
The title compound (55) was prepared according to the synthetic method of Example 54 by replacing N1- (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N1, N2, N2-trimethylethane-1, 2-diamine (15c) withN1- (4-iodo-5-methoxy-2-nitrophenyl) -N1, N2, N2-trimethylethane-1, 2-diamine (24a) . 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 (20 g, 0.14 mol) in ethane-1, 2-diol (70 ml) was added hydrazine hydrate (4.7 ml, 0.15 mol) at r.t while maintaining this temperature for 2 h , and then was heated to 150℃ for overnight. The mixture was cooled to r.t, quenched with water (200 ml) and extrated with DCM (200 mL) . The organic layers were combined and washed with water (200 mL × 2) and brine (200 mL) , dried over anhydrous sodium sulfate. The solids were filtered out and the filtrate was concentrated under vacuum. The crude product was purified by column chromatography on silica gel eluting with ethyl acetate/petroleum ether (1:20) to give 3-methyl-1H-indazole (56a) as brown solid (7 g, 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) (1.1 g, 8.3 mmol) in dry DMF was added NaH at 0℃ while maintaining this temperature for 2 h, and then was added 2, 4-dichloropyrimidine  (1.24 g, 8.3 mmol) at 0℃. The resulting mixture was stirred at r.t for overnight. The mixture was quenched with aq NH4Cl (20 ml) and extracted with ethyl acetate (50 mL) . The organic layers were combined and washed with water (50 mL × 2) and brine (50 mL) , dried over Na2SO4. The solids were filtered out and the filtrate was concentrated in vacuum. The crude product was purified by column chromatography on silica gel eluting with ethyl acetate/petroleum ether (1:20) to give 1- (2-chloropyrimidin-4-yl) -3-methyl-1H-indazole (56b) as white solid (0.85 g, 43%) . MS-ESI (m/z) : 245 [M + 1] +
N- (5- ( (4- (4, 5-dihydropyrrolo [3, 2, 1-hi] indol-1-yl) pyrimidin-2-yl) amino) -2- ( (2- (dimethy lamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide (56)
The title compound 56 (8.0 mg) was prepared by using the same procedure as described for 15 by replacing 3- (2-chloropyrimidin-4-yl) -7-fluoro-1-methyl-1H-indole (15g) with 1- (2-chloropyrimidin-4-yl) -3-methyl-1H-indazole (56b) . 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)
The title compound 57 (5.0 mg) was prepared according to the synthetic method of Example 23 by replacing 3- (2-chloropyrimidin-4-yl) -1-methyl-1H-indole (1h) with 1- (2-chloropyrimidin-4-yl) -3-methyl-1H-indazole (56b) . 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 synthetic method described in literature: J. Heterocyclic 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)
The title compound 58 was prepared according to the synthetic method of Example 25 by replacing 1- (1-methylindolizin-3-yl) ethanone (25d) with 1- (1-methyl-1H-indazol-3-yl) ethanone (58a) . 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)
N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-N5- (4- (1-methyl-1H-indazol-3-yl) py rimidin-2-yl) -3-nitropyridine-2, 5-diamine (59a)
The compound 59a was prepared according to the synthetic method of Example 25 by replacing 1- (1-methylindolizin-3-yl) ethanone (25d) with 1- (1-methyl-1H-indazol-3-yl) ethanone (58a) . MS-ESI (m/z) : 448 [M + 1] +.
N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-N5- (4- (1-methyl-1H-indazol-3-yl) py rimidin-2-yl) pyridine-2, 3, 5-triamine (59b)
The title compound 59b was prepared according to the synthetic method of 15i by replacing N1- (5-iodo-6-methoxy-3-nitropyridin-2-yl) -N1, N2, N2-trimethylethane-1, 2-diamine (15h) with N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-N5- (4- (1-methyl-1H-indazol-3-yl) pyrimidin-2-yl) -3-nitropyridine-2, 5-diamine (59a) . 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)
The title compound (59) was prepared according to the synthetic method of Example 23 by replacing N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-N5- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) pyridine-2, 3, 5-triamine (23b) with N2- (2- (dimethylamino) ethyl) -6-methoxy  -N2-methyl-N5- (4- (1-methyl-1H-indazol-3-yl) pyrimidin-2-yl) pyridine-2, 3, 5-triamine (59b) . MS-ESI (m/z) : 520 [M + 1] +.
Example 60
N- (2- ( (2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ( (4- (3-methylimidazo [1, 5- a] pyridin-1-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (60)
1- (3-methylimidazo [1, 5-a] pyridin-1-yl) ethan-1-one (60a)
The title compound 60a was prepared according to the synthetic method described in literature: Journal of Chemical society., 1955, 2834.
N- (2- ( (2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ( (4- (3-methylimidazo [1, 5- a] pyridin-1-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide (60)
The title compound 60 was prepared according to the synthetic method of Example 25 by replacing 1- (1-methylindolizin-3-yl) ethanone (25d) with 1- (3-methylimidazo [1, 5-a] pyridin-1-yl) ethan-1-one (60a) . MS-ESI (m/z) : 502 [M + 1] +.
Example 61
N- (2- ( (2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ( (4- (3-methylimidazo [1, 5- a] pyridin-1-yl) pyrimidin-2-yl) amino) pyridin-3-yl) -2-fluoroacrylamide (61)
N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-N5- (4- (3-methylimidazo [1, 5-a] pyrid in-1-yl) pyrimidin-2-yl) pyridine-2, 3, 5-triamine (61a)
The compound 61a was prepared according to the synthetic method of Example 25 by replacing 1- (1-methylindolizin-3-yl) ethanone (25d) with 1- (3-methylimidazo [1, 5-a] pyridin-1-yl) ethan-1-one (60a) . MS-ESI (m/z) : 448 [M + 1] +.
N- (2- ( (2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ( (4- (3-methylimidazo [1, 5- a] pyridin-1-yl) pyrimidin-2-yl) amino) pyridin-3-yl) -2-fluoroacrylamide (61)
The title compound 61 was prepared according to the synthetic method of Example 23 by replacing N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-N5- (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) pyridine-2, 3, 5-triamine (23b) with N2- (2- (dimethylamino) ethyl) -6-methoxy -N2-methyl-N5- (4- (3-methylimidazo [1, 5-a] pyridin-1-yl) pyrimidin-2-yl) pyridine-2, 3, 5-triamine (61a) . MS-ESI (m/z) : 520 [M + 1] +.
Cell Proliferation Assays
To investigate whether a compound is able to inhibit the activity of L858R/T790M EGFR in cells, a mechanism-based assay using H1975 cell was developed. In this assay, inhibition of L858R/T790M EGFR was detected by the inhibition of H1975 cells proliferation. H1975 cells were cultured in culture flasks to 40-80%confluence in RPMI-1640 plus 10%fetal bovine serum. Cells were collected and plated onto 96-well plates at desired cell density (3000 cells/well) . Plates were incubated overnight at 37℃, with 5%CO2 to adhere. Compounds were added to the plates, the final compound concentrations were 10000, 3333.3, 1111.1, 270.4, 123.5, 41.2, 13.7, 4.6 and 1.5 nM. Place plates at 37 ℃, with 5%CO2 for 48 h. After removing the medium, 20 μl MTS /100 μl medium mixture solution were added to each well and incubate the plates for exactly 1.5 hours. Stop the reaction by adding 25 μl 10%SDS per well. Measure absorbance at 490 nm and 650 nm (reference wavelength) . IC50 was calculated using GraphPad Prism 5.0.
To investigate whether a compound is able to inhibit the activity of WT EGFR in cells, a mechanism-based assay using A431 cell was developed. In this assay, inhibition of WT EGFR was detected by the inhibition of A431 cells proliferation. A431 cells were cultured in culture flasks to 40-80%confluence in DMEM plus 10%fetal bovine serum. Cells were collected and plated onto 96-well plates at desired cell density (3000 cells/well) . Plates were incubated overnight at 37℃, with 5%CO2 to adhere. Compounds were added to the plates, the final compound concentrations were 10000, 3333.3, 1111.1, 270.4, 123.5, 41.2, 13.7, 4.6 and 1.5 nM. Place plates at 37 ℃, with 5%CO2 for 48 h. After removing the medium, 20 μl MTS /100 μl medium mixture solution were added to each well and incubate the plates for exactly 1.5 hours. Stop the reaction by adding 25 μl  10%SDS per well. Measure absorbance at 490 nm and 650 nm (reference wavelength) . IC50 was calculated using GraphPad Prism 5.0.
Select compounds prepared as described above were assayed according to the biological procedures described herein. The results are given in the table 1.
Table 1
Example 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 (24)

  1. At least one compound of formula (I) :
    Figure PCTCN2015081332-appb-100001
    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;
    R1 is selected from hydrogen, C1-10alkyl, C3-10cycloalkyl, C3-10cycloalkyl-C1-4alkyl, heterocyclyl, and heterocyclyl-C1-4alkyl, wherein alkyl, cycloalkyl, and heterocyclyl are each unsubstituted or substituted with at least one substituent, such as one, two, three, or four substituents, independently selected from R6a
    R1’ is selected from hydrogen and halogen;
    R2 and R3 are independently selected from hydrogen, halogen, hydroxyl, CN, C1-10alkyl, C2-10alkenyl, C2-10alkynyl, C3-10cycloalkyl, C3-10cycloalkyl-C1-4alkyl, heterocyclyl, heterocyclyl-C1-4alkyl, aryl, aryl-C1-4alkyl, heteroaryl, and heteroaryl-C1-4alkyl, wherein alkyl, alkenyl, alkynyl, cycloalkyl, and heterocyclyl are each unsubstituted or substituted with at least one  substituent, such as one, two, three, or four substituents independently selected from R6a, and each aryl and heteroaryl is unsubstituted or substituted with at least one substituent, such as one, two, three, or four substituents, independently selected from R6b; or R2 and R3 together with the carbon atoms 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, and optionally substituted with 1or 2 R6a groups;
    each R4 is independently selected from hydrogen, C1-10alkyl, C2-10alkenyl, 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 (CR9R10tNR7R8, -C (O) R7, -CO2R8, -CO2 (CR9R10tCONR7R8, -OC (O) R7, -CN, -C (O) NR7R8, -NR7C (O) R8, -OC (O) NR7R8, -NR7C (O) OR8, -NR7C (O) NR7R8, and -CR7 (N-OR8) , wherein C1-10alkyl, C2-10alkenyl, C2-10alkynyl, and C3-10cycloalkyl are each unsubstituted or substituted with at least one substituent, such as one, two, three, or four substituents, independently selected from R6a
    each R5 is independently selected from hydrogen, C1-10alkyl, C2-10alkenyl, C2-10alkynyl, C3-10cycloalkyl, C3-10cycloalkyl-C1-4alkyl, heterocyclyl, heterocyclyl-C1-4alkyl, aryl, aryl-C1-4alkyl, heteroaryl, and heteroaryl-C1-4alkyl, -OR8, -NR7S (O) rR8, -NO2, halogen, -S (O) rR7, -SR8, -S (O) 2OR7, -OS (O) 2R8, -S (O) rNR7R8, -NR7R8, -O (CR9R10tNR7R8, -C (O) R7, -CO2R8, -CO2 (CR9R10tCONR7R8, -OC (O) R7, -CN, -C (O) NR7R8, -NR7C (O) R8, -OC (O) NR7R8, -NR7C (O) OR8, -NR7C (O) NR7R8, and -CR7 (N-OR8) , wherein C1-10alkyl, C2-10alkenyl, C2-10alkynyl, C3-10cycloalkyl and heterocyclyl are each unsubstituted or substituted with at least one substituent, such as one, two, three, or four substituents independently selected from R6a, and each aryl and heteroaryl is unsubstituted or substituted with at least one substituent, such as one, two, three, or four substituents, independently selected from R6b
    each R6a is independently selected from C1-10alkyl, C2-10alkenyl, C2-10alkynyl, C3-10cycloalkyl, -OR8, -NR7S (O) rR8, -NO2, halogen, -S (O) rR7, -SR8, -S (O) 2OR7, -OS (O) 2R8, -S (O) rNR7R8, -NR7R8, - (CR9R10tOR8, - (CR9R10tNR7R8, - (CR9R10tSR8, - (CR9R10tS (O) rR8, - (CR9R10tCO2R8, - (CR9R10tCONR7R8, - (CR9R10tNR7CO2R8, - (CR9R10tOCONR7R8, - (CR9R10tNR7CONR7R8,  - (CR9R10tNR7SO2NR7R8, -O (CR9R10tNR7R8, -C (O) R7, -C (O) (CR9R10tOR8, -C (O) (CR9R10tNR7R8, -C (O) (CR9R10tSR8, -C (O) (CR9R10tS (O) rR8, -CO2R8, -CO2 (CR9R10tCONR7R8, -OC (O) R7, -CN, -C (O) NR7R8, -NR7C (O) R8, -OC (O) NR7R8, -NR7C (O) OR8, -NR7C (O) NR7R8, and -CR7 (N-OR8) ;
    each R6b is independently selected from R6a, aryl, aryl-C1-4alkyl, heteroaryl, and heteroaryl-C1-4alkyl;
    each R7 and each R8 are independently selected from hydrogen, C1-10alkyl, C2-10alkenyl, C2-10alkynyl, cycloalkyl, cycloalkyl-C1-4alkyl, heterocyclyl, heterocyclyl-C1-4alkyl, aryl, aryl-C1-4alkyl, heteroaryl, and heteroaryl-C1-4alkyl, wherein alkyl, alkenyl, alkynyl, cycloalkyl, and heterocyclyl are each unsubstituted or substituted with at least one substituent, such as one, two, three, or four substituents independently selected from R6a, and aryl and heteroaryl are each unsubstituted or substituted with at least one substituent, such as one, two, three, or four substituents independently selected from R6b; or R7 and R8 together with the atom (s) to which they are attached form a heterocyclic ring of 4 to 12 members containing 0, 1, or 2 additional heteroatoms independently selected from oxygen, sulfur and nitrogen, and optionally substituted with 1or 2 R6b groups;
    each R9 and each R10 are independently selected from hydrogen, C1-10alkyl, C2-10alkenyl, C2-10alkynyl, cycloalkyl, cycloalkyl-C1-4alkyl, heterocyclyl, heterocyclyl-C1-4alkyl, aryl, aryl-C1-4alkyl, heteroaryl, and heteroaryl-C1-4alkyl; or R9 and R10 together with the carbon atom (s) to which they are attached form a ring of 3 to 7 members containing 0, 1, or 2 heteroatoms independently selected from oxygen, sulfur and nitrogen, and optionally substituted with 1or 2 R6a groups;
    m is independently selected from 0, 1, 2, and 3;
    n is independently selected from 0, 1, 2, and 3;
    each r is independently selected from 1 and 2;
    each t is independently selected from 1, 2, and 3.
  2. At least one compound of claim 1 and/or at least one pharmaceutically acceptable salt thereof, wherein X is selected from N and C, and Y is C.
  3. At least one compound of any one of claims 1 to 2 and/or at least one pharmaceutically acceptable salt thereof, wherein X is N, and Y is C.
  4. At least one compound of any one of claims 1 to 2 and/or at least one pharmaceutically acceptable salt thereof, wherein when both X and Y are C, and,
    n = 2, one R5 is (3R) -3- (dimethylamino) pyrrolidin-1-yl, (3S) -3- (dimethylamino) pyrrolidin-1-yl, 3- (dimethylamino) azetidin-1-yl, [2- (dimethylamino) ethyl] - (methyl) amino,
    [2- (methylamino) ethyl] (methyl) amino, 5-methyl-2, 5-diazaspiro [3.4] oct-2-yl,
    (3aR, 6aR) -5-methylhexa-hydro-pyrrolo [3, 4-b] pyrrol-1 (2H) -yl,
    1-methyl-1, 2, 3, 6-tetrahydropyridin-4-yl, 4-methylpiperizin-1-yl,
    4- [2- (dimethylamino) -2-oxoethyl] 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-aminopropanoyl] piperazin-1-yl, the other R5 is methoxy or methyl, and,
    R1, R1’ and R2 are hydrogen, and R3 is H, F, Cl, CH3 or CN,
    Q is not 4, 5, 6, 7-tetrahydropyrazolo [1, 5-a] pyridine-3-yl, pyrazolo [1, 5-a] pyridin-3-yl or 1H-indol-3-yl.
  5. At least one compound of any one of claims 1 to 4 and/or at least one pharmaceutically acceptable salt thereof, wherein the formula is
    Figure PCTCN2015081332-appb-100002
  6. At least one compound of any one of claims 1 to 5 and/or at least one pharmaceutically acceptable salt thereof, wherein R1’ is hydrogen.
  7. At least one compound of any one of claims 1 to 5 and/or at least one pharmaceutically acceptable salt thereof, wherein R1’ is selected from halogen, preferably R1’ is fluorine.
  8. At least one compound of any one of claims 1 to 7 and/or at least one pharmaceutically acceptable salt thereof, wherein R1 is hydrogen.
  9. At least one compound of any one of claims 1 to 8 and/or at least one pharmaceutically acceptable salt thereof, wherein R2 is hydrogen.
  10. At least one compound of any one of claims 1 to 9 and/or at least one pharmaceutically acceptable salt thereof, wherein R3 is selected from hydrogen, halogen, and C1-10 alkyl.
  11. At least one compound of claim 10 and/or at least one pharmaceutically acceptable salt thereof, wherein R3 is hydrogen.
  12. At least one compound of any one of claims 1 to 11 and/or at least one pharmaceutically acceptable salt thereof, wherein Q is selected from heteroaryl.
  13. At least one compound of claim 12 and/or at least one pharmaceutically acceptable salt thereof, wherein Q is selected from
    Figure PCTCN2015081332-appb-100003
  14. At least one compound of any one of claims 1 to 13 and/or at least one pharmaceutically acceptable salt thereof, wherein R4 is selected from hydrogen, C1-10alkyl, halogen and CN, wherein C1-10alkyl are unsubstituted or substituted with at least one substituent, such as one, two, three, or four substituents, independently selected from R6a.
  15. At least one compound of claim 14 and/or at least one pharmaceutically acceptable salt thereof, wherein R4 is selected from hydrogen, methyl, halogen and CN, wherein methyl are unsubstituted or substituted with at least one substituent independently selected from R6a, preferably R6a is fluorine.
  16. At least one compound of any one of claims 1 to 15 and/or at least one pharmaceutically acceptable salt thereof, wherein R5 is independently selected from OR8, heterocyclyl, NR7R8, wherein heterocyclyl is unsubstituted or substituted with at least one substituent, such as one, two, three, or four substituents, independently selected from R6a.
  17. At least one compound of claim 16 and/or at least one pharmaceutically acceptable salt thereof, wherein R5 is independently 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-methylacetamido) ethyl) amino) .
  18. At least one compound, 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- (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-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-methylpiperazin-1-yl) pyridin-3-yl) acrylamide,
    N- (6-ethoxy-5- ( (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- (piperazin-1-yl) pyridin-3-yl) acrylamide,
    N- (6-ethoxy-5- ( (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- (4-methylpiperazin-1-yl) pyridin-3-yl) acrylamide,
    N- (6-isopropoxy-5- ( (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- (piperazin-1-yl) pyridin-3-yl) acrylamide,
    N- (6-isopropoxy-5- ( (4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- (4-methylpiperazin-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-in dol-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-methylindolizin-3-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide,
    N- (2- ( (2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5- ( (4- (1-methylindolizin-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-methylindolizin-1-yl) pyrimidin-2-yl) amino) pyridin-3-yl) acrylamide,
    N- (2- ( (2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5- ( (4- (3-methylindolizin-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] pyridin-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] pyridin-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) imidaz o [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) imidaz o [1, 5-a] pyridin-3-yl) pyrimidin-2-yl) amino) phenyl) acrylamide,
    N- (2- ( (2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ( (4- (1-methylimidazo [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-methylimidazo [1, 5-a] pyridin-3-yl) pyrimidin-2-yl) amino) phenyl) acrylamide,
    N- (2- ( (2- (dimethylamino) ethyl) (methyl) amino) -6-methoxy-5- ( (4- (1-methylimidazo [1, 5-a] pyridin-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-methylimidazo [1, 5-a] pyridin-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] pyridin-1-yl) pyrimidin-2-yl) amino) pyridin-3-yl) -2-fluoroacrylamide,
    and pharmaceutically acceptable salts thereof.
  19. A pharmaceutical composition, comprising at least one compound of claims 1 to 18, and/or at least one pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier.
  20. A method for modulating EGFR mutant, comprising administering to a subject in need thereof a therapeutically effective amount of at least one compound of claims 1 to 18, and/or at least one pharmaceutically acceptable salt thereof , or at least one pharmaceutical composition thereof.
  21. A method to treat, ameliorate or prevent a condition, which responds to inhibition of EGFR mutant, comprising administering to a subject in need of such treatment an effective amount of at least one compound of claims 1 to 18, and/or at least one pharmaceutically acceptable salt thereof, or of at least one pharmaceutical composition thereof, and optionally in combination with a second therapeutic agent.
  22. A method for making a medicament for treating a condition, which responds to inhibition of EGFR mutant, comprising including to the medicament at least one compound of claims 1 to 18, and/or at least one pharmaceutically acceptable salt thereof.
  23. A method for treating a cell proliferative disorder, comprising administering to a subject in need of such treatment an effective amount of at least one compound of claims 1 to 18, and/or at least one pharmaceutically acceptable salt thereof or of at least one pharmaceutical composition thereof, and optionally in combination with a second therapeutic agent.
  24. A method for making a medicament for treating a cell-proliferative disorder, comprising including in the medicament at least one compound of claims 1 to 18, and/or at least one pharmaceutically acceptable salt thereof.
PCT/CN2015/081332 2014-06-12 2015-06-12 Certain protein kinase inhibitors WO2015188777A1 (en)

Priority Applications (3)

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

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201462011109P 2014-06-12 2014-06-12
US62/011,109 2014-06-12
US201462090360P 2014-12-11 2014-12-11
US62/090,360 2014-12-11

Publications (1)

Publication Number Publication Date
WO2015188777A1 true WO2015188777A1 (en) 2015-12-17

Family

ID=54832920

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2015/081332 WO2015188777A1 (en) 2014-06-12 2015-06-12 Certain protein kinase inhibitors

Country Status (2)

Country Link
CN (3) CN111875585B (en)
WO (1) WO2015188777A1 (en)

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016105525A3 (en) * 2014-12-23 2016-09-29 Dana-Farber Cancer Institute, Inc. Novel pyrimidines as egfr inhibitors and methods of treating disorders
CN106117185A (en) * 2015-08-31 2016-11-16 广州科擎新药开发有限公司 2,4 2 nitrogen-containing group substituted pyrimidines compounds and its preparation method and application
CN106279160A (en) * 2016-03-18 2017-01-04 海南越康生物医药有限公司 N phenyl 2 amino-metadiazine compound preparation method and purposes
WO2017111076A1 (en) 2015-12-24 2017-06-29 協和発酵キリン株式会社 α, β UNSATURATED AMIDE COMPOUND
CN106928150A (en) * 2015-12-31 2017-07-07 恩瑞生物医药科技(上海)有限公司 Acrylamide anil and its application pharmaceutically
CN106957304A (en) * 2017-04-25 2017-07-18 耿岳 A kind of graphene-supported FeCl3The preparation method of catalyst and its purposes in preparation Anti-cancer medicament intermediate
EP3207035A4 (en) * 2014-10-13 2018-03-14 Yuhan Corporation Compounds and compositions for modulating egfr mutant kinase activities
CN107827875A (en) * 2017-09-25 2018-03-23 文韬创新药物研究(北京)有限责任公司 A kind of application of benzimidazoles derivative as the inhibitor of Cyclin dependent kinase 4/6
WO2018235926A1 (en) 2017-06-23 2018-12-27 協和発酵キリン株式会社 α, β-UNSATURATED AMIDE COMPOUND
US10179784B2 (en) 2014-11-05 2019-01-15 Inventisbio Shanghai Ltd. Pyrimidine or pyridine compounds, preparation method therefor and pharmaceutical uses thereof
US20190023689A1 (en) * 2016-01-07 2019-01-24 Cs Pharmatech Limited Selective inhibitors of clinically important mutants of the egfr tyrosine kinase
US10428081B2 (en) * 2014-10-11 2019-10-01 Shanghai Hansoh Biomedical Co., Ltd. EGFR inhibitor, preparation method and use thereof
US10513509B2 (en) 2016-05-26 2019-12-24 Recurium Ip Holdings, Llc EGFR inhibitor compounds
US10738067B2 (en) 2018-11-01 2020-08-11 Syros Pharmaceuticals, Inc. Inhibitors of cyclin-dependent kinase 7 (CDK7)
WO2020230091A1 (en) * 2019-05-14 2020-11-19 Janssen Biotech, Inc. Combination therapies with bispecific anti-egfr/c-met antibodies and third generation egfr tyrosine kinase inhibitors
US10906901B2 (en) 2017-07-19 2021-02-02 Hainan Yuekang Biomedicines Co., Ltd. Crystal form and salt form of N-phenyl-2-aminopyrimidine compound, and preparation method therefor
USRE48687E1 (en) 2014-07-29 2021-08-17 Shanghai Allist Pharmaceuticals Co., Ltd. Pyridinylaminopyrimidine derivatives, preparation process and use thereof
WO2021177343A1 (en) * 2020-03-04 2021-09-10 国立研究開発法人理化学研究所 Method for releasing compound
AU2020255100A8 (en) * 2019-03-29 2021-12-02 Shenzhen Forward Pharmaceuticals Co., Ltd. N-heteroaromatic amide derivatives for treatment of cancer
US11311542B2 (en) 2018-01-16 2022-04-26 Syros Pharmaceuticals, Inc. Inhibitors of cyclin dependent kinase 7 (CDK7)
US11459391B2 (en) 2019-02-26 2022-10-04 Janssen Biotech, Inc. Combination therapies and patient stratification with bispecific anti-EGFR/c-Met antibodies
WO2022271861A1 (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
US11708362B2 (en) 2017-07-28 2023-07-25 Yuhan Corporation Process for preparing aminopyrimidine derivatives
US11918592B2 (en) 2018-01-16 2024-03-05 Syros Pharmaceuticals, Inc. Inhibitors of cyclin dependent kinase 7 (CDK7)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
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
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
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 (2)

* 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

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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

Patent Citations (2)

* 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

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
RICHARD A. WARD ET AL.: "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., vol. 56, no. 17, 9 August 2013 (2013-08-09), pages 7025 - 7048, XP002734712, ISSN: 0022-2623 *

Cited By (58)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
USRE48687E1 (en) 2014-07-29 2021-08-17 Shanghai Allist Pharmaceuticals Co., Ltd. Pyridinylaminopyrimidine derivatives, preparation process and use thereof
USRE49851E1 (en) 2014-07-29 2024-02-27 Shanghai Allist Pharmaceuticals Co., Ltd. Pyridinylaminopyrimidine derivatives, preparation process and use thereof
US10428081B2 (en) * 2014-10-11 2019-10-01 Shanghai Hansoh Biomedical Co., Ltd. EGFR inhibitor, preparation method and use thereof
KR102487451B1 (en) 2014-10-13 2023-01-11 주식회사유한양행 Compounds and compositions for modulating egfr mutant kinase activities
EP3929190A1 (en) * 2014-10-13 2021-12-29 Yuhan Corporation Compounds and compositions for modulating egfr mutant kinase activities
EP3604294A1 (en) * 2014-10-13 2020-02-05 Yuhan Corporation Compounds and compositions for modulating egfr mutant kinase activities
KR20210011068A (en) * 2014-10-13 2021-01-29 주식회사유한양행 Compounds and compositions for modulating egfr mutant kinase activities
JP2019163277A (en) * 2014-10-13 2019-09-26 ユハン コーポレーションYuhan Corporation Compounds and compositions for modulating egfr mutant kinase activities
EP3207035A4 (en) * 2014-10-13 2018-03-14 Yuhan Corporation Compounds and compositions for modulating egfr mutant kinase activities
US11203589B2 (en) 2014-11-05 2021-12-21 InventisBio Co., Ltd. Pyrimidine or pyridine compounds, preparation method therefor and pharmaceutical uses thereof
US11498921B1 (en) 2014-11-05 2022-11-15 InventisBio Co., Ltd. Pyrimidine or pyridine compounds, preparation method therefor and pharmaceutical uses thereof
US10179784B2 (en) 2014-11-05 2019-01-15 Inventisbio Shanghai Ltd. Pyrimidine or pyridine compounds, preparation method therefor and pharmaceutical uses thereof
US10266517B2 (en) 2014-12-23 2019-04-23 Dana-Farber Cancer Institute, Inc. Pyrimidines as EGFR inhibitors and methods of treating disorders
US11673882B2 (en) 2014-12-23 2023-06-13 Dana-Farber Cancer Institute, Inc. Pyrimidines as EGFR inhibitors and methods of treating disorders
US10870636B2 (en) 2014-12-23 2020-12-22 Dana-Farber Cancer Institute, Inc. Pyrimidines as EGFR inhibitors and methods of treating disorders
WO2016105525A3 (en) * 2014-12-23 2016-09-29 Dana-Farber Cancer Institute, Inc. Novel pyrimidines as egfr inhibitors and methods of treating disorders
US11267802B2 (en) 2015-08-31 2022-03-08 Bebetter Med Inc. 2,4-bis(nitrogen-containing group)-substituted pyrimidine compound, preparation method and use thereof
EP3345900A4 (en) * 2015-08-31 2019-04-10 Guangzhou Bebetter Medicine Technology Co., Ltd. 2, 4-di-(nitrogen containing group) substituted pyrimidine compound and preparation method and use thereof
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
CN106117185A (en) * 2015-08-31 2016-11-16 广州科擎新药开发有限公司 2,4 2 nitrogen-containing group substituted pyrimidines compounds and its preparation method and application
US11332455B2 (en) 2015-12-24 2022-05-17 Kyowa Kirin Co., Ltd. α,β-unsaturated amide compound
WO2017111076A1 (en) 2015-12-24 2017-06-29 協和発酵キリン株式会社 α, β UNSATURATED AMIDE COMPOUND
EP3401309A4 (en) * 2015-12-24 2019-11-27 Kyowa Kirin Co., Ltd. a, ß UNSATURATED AMIDE COMPOUND
US10787428B2 (en) 2015-12-24 2020-09-29 Kyowa Kirin Co., Ltd. α,β-unsaturated amide compound
AU2021204307B2 (en) * 2015-12-24 2023-06-01 Kyowa Kirin Co., Ltd. a, ß unsaturated amide compound
AU2016382515B2 (en) * 2015-12-31 2020-12-17 Ancureall Pharmaceutical (Shanghai) Co., Ltd. Acrylanilide derivative, preparation method therefor, and applications thereof in pharmacy
JP2019506449A (en) * 2015-12-31 2019-03-07 恩瑞生物医▲薬▼科技(上▲海▼)有限公司Ancureall Pharmaceutical(Shanghai)Co.,Ltd. Acrylic anilide derivative, its production method and its pharmaceutical application
CN106928150A (en) * 2015-12-31 2017-07-07 恩瑞生物医药科技(上海)有限公司 Acrylamide anil and its application pharmaceutically
RU2742372C2 (en) * 2015-12-31 2021-02-05 Энкьюриэлл Фармасьютикал (Шанхай) Ко., Лтд. Acrylanilide derivative, a method for production thereof and use thereof in pharmacology
EP3398939A4 (en) * 2015-12-31 2019-02-13 Ancureall Pharmaceutical (Shanghai) Co., Ltd. Acrylanilide derivative, preparation method therefor, and applications thereof in pharmacy
US20190023689A1 (en) * 2016-01-07 2019-01-24 Cs Pharmatech Limited Selective inhibitors of clinically important mutants of the egfr tyrosine kinase
US10435388B2 (en) 2016-01-07 2019-10-08 Cs Pharmatech Limited Selective inhibitors of clinically important mutants of the EGFR tyrosine kinase
CN106279160A (en) * 2016-03-18 2017-01-04 海南越康生物医药有限公司 N phenyl 2 amino-metadiazine compound preparation method and purposes
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
US10513509B2 (en) 2016-05-26 2019-12-24 Recurium Ip Holdings, Llc EGFR inhibitor compounds
US11098030B2 (en) 2016-05-26 2021-08-24 Recurium Ip Holdings, Llc EGFR inhibitor compounds
CN106957304A (en) * 2017-04-25 2017-07-18 耿岳 A kind of graphene-supported FeCl3The preparation method of catalyst and its purposes in preparation Anti-cancer medicament intermediate
JP7221202B2 (en) 2017-06-23 2023-02-13 協和キリン株式会社 α, β unsaturated amide compounds
JPWO2018235926A1 (en) * 2017-06-23 2020-05-21 協和キリン株式会社 α, β unsaturated amide compounds
KR20200019979A (en) 2017-06-23 2020-02-25 쿄와 기린 가부시키가이샤 α, β unsaturated amide compounds
US11447471B2 (en) 2017-06-23 2022-09-20 Kyowa Kirin Co., Ltd. α,β-unsaturated amide compound
AU2018289759B2 (en) * 2017-06-23 2022-03-10 Kyowa Kirin Co., Ltd. Alpha, beta-unsaturated amide compound
WO2018235926A1 (en) 2017-06-23 2018-12-27 協和発酵キリン株式会社 α, β-UNSATURATED AMIDE COMPOUND
US10906901B2 (en) 2017-07-19 2021-02-02 Hainan Yuekang Biomedicines Co., Ltd. Crystal form and salt form of N-phenyl-2-aminopyrimidine compound, and preparation method therefor
US11708362B2 (en) 2017-07-28 2023-07-25 Yuhan Corporation Process for preparing aminopyrimidine derivatives
CN107827875A (en) * 2017-09-25 2018-03-23 文韬创新药物研究(北京)有限责任公司 A kind of application of benzimidazoles derivative as the inhibitor of Cyclin dependent kinase 4/6
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
US11311542B2 (en) 2018-01-16 2022-04-26 Syros Pharmaceuticals, Inc. Inhibitors of cyclin dependent kinase 7 (CDK7)
US11918592B2 (en) 2018-01-16 2024-03-05 Syros Pharmaceuticals, Inc. Inhibitors of cyclin dependent kinase 7 (CDK7)
US10738067B2 (en) 2018-11-01 2020-08-11 Syros Pharmaceuticals, Inc. Inhibitors of cyclin-dependent kinase 7 (CDK7)
US11459391B2 (en) 2019-02-26 2022-10-04 Janssen Biotech, Inc. Combination therapies and patient stratification with bispecific anti-EGFR/c-Met antibodies
AU2020255100B2 (en) * 2019-03-29 2022-11-24 Shenzhen Forward Pharmaceuticals Co., Ltd. N-heteroaromatic amide derivatives for treatment of cancer
AU2020255100C1 (en) * 2019-03-29 2023-02-23 Shenzhen Forward Pharmaceuticals Co., Ltd. N-heteroaromatic amide derivatives for treatment of cancer
AU2020255100A8 (en) * 2019-03-29 2021-12-02 Shenzhen Forward Pharmaceuticals Co., Ltd. N-heteroaromatic amide derivatives for 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
WO2020230091A1 (en) * 2019-05-14 2020-11-19 Janssen Biotech, Inc. Combination therapies with bispecific anti-egfr/c-met antibodies and third generation egfr tyrosine kinase inhibitors
WO2021177343A1 (en) * 2020-03-04 2021-09-10 国立研究開発法人理化学研究所 Method for releasing compound
WO2022271861A1 (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

Also Published As

Publication number Publication date
CN111892579A (en) 2020-11-06
CN111875585A (en) 2020-11-03
CN111892579B (en) 2023-07-25
CN111875585B (en) 2023-06-23
CN106660993A (en) 2017-05-10
CN106660993B (en) 2020-09-11

Similar Documents

Publication Publication Date Title
WO2015188777A1 (en) Certain protein kinase inhibitors
JP6570001B2 (en) ALK kinase inhibitor
EP3149008B1 (en) Certain protein kinase inhibitors
US10022374B2 (en) Certain protein kinase inhibitors
EP3224248B1 (en) Certain protein kinase inhibitors
US20160145244A1 (en) Certain protein kinase inhibitors

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 15806251

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 15806251

Country of ref document: EP

Kind code of ref document: A1