USRE48687E1 - Pyridinylaminopyrimidine derivatives, preparation process and use thereof - Google Patents

Pyridinylaminopyrimidine derivatives, preparation process and use thereof Download PDF

Info

Publication number
USRE48687E1
USRE48687E1 US16/517,790 US201516517790A USRE48687E US RE48687 E1 USRE48687 E1 US RE48687E1 US 201516517790 A US201516517790 A US 201516517790A US RE48687 E USRE48687 E US RE48687E
Authority
US
United States
Prior art keywords
methyl
amino
alkyl
ethyl
pyridin
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
US16/517,790
Other languages
English (en)
Inventor
Huibing Luo
Huayong Zhou
Shuhui Wang
Yong Wu
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Shanghai Allist Pharmaceuticals Inc
Original Assignee
Shanghai Allist Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Shanghai Allist Pharmaceuticals Inc filed Critical Shanghai Allist Pharmaceuticals Inc
Priority to US16/517,790 priority Critical patent/USRE48687E1/en
Assigned to SHANGHAI ALLIST PHARMACEUTICALS CO., LTD. reassignment SHANGHAI ALLIST PHARMACEUTICALS CO., LTD. CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: SHANGHAI ALLIST PHARMACEUTICALS, INC.
Application granted granted Critical
Publication of USRE48687E1 publication Critical patent/USRE48687E1/en
Assigned to SHANGHAI ALLIST PHARMACEUTICALS CO., LTD. reassignment SHANGHAI ALLIST PHARMACEUTICALS CO., LTD. CHANGE OF ADDRESS Assignors: SHANGHAI ALLIST PHARMACEUTICALS CO., LTD.
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • 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
    • 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
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/444Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring heteroatom, e.g. amrinone
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings

Definitions

  • the present invention relates to pyridinylaminopyrimidine derivatives, which selectively inhibit the activity of mutation-type epidermal growth factor receptor (EGFR), a pharmaceutically acceptable salt thereof, a process for preparing the same, a pharmaceutical composition containing said derivative and a pharmaceutically acceptable salt thereof, uses of said derivative and a pharmaceutically acceptable salt thereof in treating some mutation-type EGFR mediated diseases and in manufacture of a medicament for treating some mutation-type EGFR mediated diseases.
  • EGFR epidermal growth factor receptor
  • Cancer has been considered as a disease of the intracellular signal transconducing system or signal transduction mechanism.
  • the most common cause of cancer is a series of defects, either in proteins, when they are mutated, or in the regulation of the quantities of the proteins in the cells such that they are over or under produced. Mutations to the cell surface receptors, which usually transduce the signals into the cells by means of tyrosine kinases, can lead to activation of the kinase in the absence of ligand, and passing of a signal which does not really exist. Alternatively, many receptor tyrosine kinases can be overexpressed on the cell surface leading to an inappropriately strong response to a weak signal.
  • Epidermal cell growth factors receptors are identified as one significant driving factor in the process for cellular growth and proliferation.
  • the epidermal cell growth factors receptors family is composed of EGFR (Erb-B1), Erb-B2 (HER-2/neu), Erb-B3 and Erb-B4.
  • the epidermal cell growth factors receptors are concerned in the process for most cancers, such as lung cancer, colon cancer and breast cancer.
  • the overexpression and mutation of EGFR have been proved to be the leading risk factor for a breast cancer with poor prognosis.
  • each of the above four members of the receptors family can aggregate with another member into a heterodimer, and form a signal transduction complex. Overexpression of one or more member(s) of this family in a malignant tumor will result in a synergistic signal transduction.
  • EGFR belongs to the protein tyrosine kinase (PTK) family.
  • the protein tyrosine kinase is an group of enzymes which catalyze the transportation of phosphate groups from adenosine triphosphate (ATP) to the tyrosine residue located in a protein substrate.
  • ATP adenosine triphosphate
  • Protein tyrosine kinases function in normal cell growth.
  • the overexpression and mutation of EGFR may cause the activation of receptors without ligands and the phosphorylation of some proteins, and then the signal for cell division is produced. As a result, EGFR may magnify the weak signal excessively by its own tyrosine-kinase action, and render the overproliferation of cells.
  • PTK inhibitors as a potential anti-cancer therapeutic drug are of wide concern.
  • Typical representatives of currently market available EGFR reversible inhibitors include Gefitinib, Erlotinib and Lapatinib, and inhibit the EGFR wild-type and activating mutations (e.g. Exon 19 deletion activating mutation, or L858R activating mutation).
  • Their structures are as follows, and are respectively useful for treating non-small cell lung cancer (NSCLC) and breast cancer.
  • NSCLC non-small cell lung cancer
  • Clinical study proves gefitinib and erlotinib have a favorable therapeutic effect on NSCLC patients with EGFR exon 19 deletion or L858R mutation.
  • NSCLC non-small cell lung cancer
  • erlotinib have a favorable therapeutic effect on NSCLC patients with EGFR exon 19 deletion or L858R mutation.
  • their limitations are that patients develop drug resistance after treatment, so that inhibitors of this type are limited in their further clinical applications.
  • T790M is located at the entrance of the ATP binding pocket of EGFR, and the size of its side chain directly affects the ability of EGFR binding to ATP.
  • the T790M mutation spatially inhibits the interaction of the EGFR inhibitor and the ATP binding site, increases the affinity of EGFR to ATP, and makes the cells resistant to the EGFR inhibitors.
  • irreversible EGFR inhibitors Compared to reversible EGFR inhibitors, irreversible EGFR inhibitors have very prominent advantages. Irreversible EGFR inhibitors can inhibit EGFR for a long time and are only limited by the normal rate of receptor re-binding (also called reversion). It is found that the irreversible EGFR inhibitor can covalently bind to the cysteine residue (Cys797) of the EGFR by Michael addition reaction and expand the binding sites of irreversible EGFR inhibitors and the ATP, so that the resistance caused by the T790M mutation can be overcame to some extent (Li D et al., Oncogene, 27:4702-4711, 2008).
  • the above-mentioned reversible or irreversible EGFR inhibitors being currently marketed or under development, are mainly quinazoline compounds.
  • the currently reported quinazoline EGFR inhibitors are the ATP competitive inhibitors of wild-type EGFR, leading to the occurrence of some side-reaction.
  • a group of pyrimidine-based irreversible EGFR inhibitors which are specific to the EGFR T790M was reported by the researchers, and the structures are shown below.
  • these pyrimidine-based compounds Compared to the existing aniline quinazoline EGFR inhibitors, these pyrimidine-based compounds have a 30-100 fold higher inhibition activity for the EGFR T790M, and a 100 fold lower inhibition activity for the wild-type EGFR (WenjunZhou et al., Nature, 462:1070-1074, 2009). However, these pyrimidine-based compounds did not enter the clinical study later.
  • the present invention provides a compound represented by the following general formula (I), or a pharmaceutically acceptable salt thereof:
  • Ring A is aryl or heteroaryl
  • R 1 is selected from a group consisting of hydrogen, halogen, C 1 -C 4 alkyl, haloC 1 -C 4 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl or —CN
  • R 2 is selected from a group consisting of C 1 -C 4 alkyl, haloC 1 -C 4 alkyl, C 2 -C 6 alkenyl, —(CH 2 ) q OR 7 , —(CH 2 ) q NR 7 R 7 ′ or —(CH 2 ) q C(O)R 7
  • R 4 is
  • Each R 5 is dependently independently halogen, C 1 -C 4 alkyl, haloC 1 -C 4 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, —OR 6 , —C(O)R 7 , —C(O)NR 7 R 7 ′, —OR 7 , —NR 7 R 7 ′, —CN or —NO 2 ;
  • R 3 is selected from a group consisting of halogen, —CN, —NO 2 , C 1 -C 4 alkyl, haloC 1 -C 4 alkyl, —C(O)R 6 , —C(O)R 7 , —C(O)NR 7 R 7 ′, —OR 7 , —OR 6 , —NHR 7 , —NR 7 —(C 1 -C 4 alkyl), —NR 7 -(haloC 1 -C 4 alkyl), —NR 7 (
  • the present invention provides a compound represented by the general formula (I), which can inhibit one or more EGFR activating or resistant mutations, such as L858R activating mutation, Exon 19 deletion activating mutation, and T790M resistant mutation.
  • the present compound can be useful in treating the cancer patient who has been resistant to the existing therapy based on the EGFR inhibitor.
  • the present invention provides a compound represented by the general formula (I), which shows a higher inhibition to the activating or resistant mutation-type EGFR than the wild-type EGFR. Due to the reduced toxicity associated with the inhibition of the wild-type EGFR, it is therefore expected that the compound of the present invention is more useful as a therapeutic agent, in particular for treating the cancer.
  • the present invention also provides a process for preparing the compound represented by the general formula (I) of the present invention.
  • the present invention also provides a pharmaceutical composition, comprising the compound represented by the general formula (I) of the present invention or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, excipient or diluent.
  • the present invention also provides use of the compound represented by the general formula (I) of the present invention or a pharmaceutically acceptable salt thereof for treating an EGFR activating or resistant mutation-mediated disease, in particular cancer, in mammals, in particular human.
  • the present invention also provides use of the compound represented by the general formula (I) of the present invention or a pharmaceutically acceptable salt thereof in manufacture of a medicament for treating an EGFR activating or resistant mutation-mediated disease, in particular cancer, in mammals, in particular human.
  • the present invention also provides a method for treating an EGFR activating or resistant mutation-mediated disease, in particular cancer, in mammals, in particular human, said method comprises administrating to a patient the compound represented by the general formula (I) or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition comprising a therapeutically effective amount of the compound represented by the general formula (I) and a pharmaceutically acceptable carrier, excipient or diluent.
  • the present invention also provides a method of selectively inhibiting the EGFR activating or resistant mutation over the wild-type EGFR (WT EGFR), said method comprises contacting a biological sample with or administrating to a patient the compound represented by the general formula (I) or a pharmaceutically acceptable salt thereof or a pharmaceutical composition comprising the same.
  • the cancer as mentioned in the present invention can be selected from a group consisting of lung cancer, ovarian cancer, cervical cancer, breast cancer, stomach cancer, colorectal cancer, pancreatic cancer, glioma, glioblastoma, melanoma, prostate cancer, leukemia, lymphoma, non-Hodgkin's lymphoma, hepatocytes cancer, gastrointestinal stromal tumor (GIST), thyroid cancer, cholangiocarcinoma, endometrial cancer, renal cancer, anaplastic large cell lymphoma, acute myeloid leukemia (AML), multiple myeloma, and mesothelioma.
  • lung cancer ovarian cancer, cervical cancer, breast cancer, stomach cancer, colorectal cancer, pancreatic cancer, glioma, glioblastoma, melanoma, prostate cancer, leukemia, lymphoma, non-Hodgkin's lymphoma, hepatocytes cancer, gastrointestinal stromal
  • Ring A is heteroaryl
  • Ring A is indolyl, indazolyl, pyrro[2,3-c]pyridinyl pyrrolo[2,3-c]pyridinyl, pyrro[3,2-c]pyridinyl pyrrolo[3,2-c]pyridinyl, pyrro[2,3-b]pyridinyl pyrrolo[2,3-b]pyridinyl, pyrro[3,2-b]pyridinyl pyrrolo[3,2-b]pyridinyl, pyrro[2,3-b]pyrazinyl pyrrolo[2,3-b]pyrazinyl, indolin-2-onyl, pyridinyl, pyrazolyl or pyrimidinyl.
  • R 1 is hydrogen, halogen or haloC 1 -C 4 alkyl.
  • R 1 is hydrogen, chloro, fluoro or trifluoromethyl.
  • R 2 is C 1 -C 4 alkyl or haloC 1 -C 4 alkyl, preferably C 2 -C 4 alkyl or haloC 2 -C 4 alkyl, more preferably isopropyl or trifluoroethyl.
  • R 4 is
  • R 7 and R 7 ′ are each independently hydrogen or C 1 -C 4 alkyl.
  • R 4 is
  • R 7 is hydrogen
  • R 3 is selected from a group consisting of
  • halogen —CN, —NO 2 , C 1 -C 4 alkyl, haloC 1 -C 4 alkyl, —C(O)R 7 , —C(O)NR 7 R 7 ′, —OR 7 , —NHR 7 , —NR 7 —(C 1 -C 4 alkyl), —NR 7 (CH 2 ) n C(O)R 6 or —NR 6 R 7 ,
  • heterocycloalkyl that is unsubstituted or substituted with 1-3 substituents selected from halogen, C 1 -C 4 alkyl, haloC 1 -C 4 alkyl, —(CH 2 ) n OH, —NR 7 R 7 ′, —OR 7 or —C(O)R 7 ;
  • R 6 is —(CH 2 ) q OR 7 , —(CH 2 ) q NR 7 R 7 ′, —(CH 2 ) q C(O)R 7 or —(CH 2 ) q C(O)NR 7 R 7 ′;
  • R 7 and R 7 ′ are each independently hydrogen, C 1 -C 4 alkyl or haloC 1 -C 4 alkyl, or R 7 , R 7 ′ and the nitrogen atom attached thereto are cyclized together to form a heterocycloalkyl;
  • n 0, 1, 2, 3 or 4;
  • q 0, 1, 2, 3 or 4.
  • R 3 is —NR 6 R 7 , wherein R 6 is —(CH 2 ) q NR 7 R 7 ′, R 7 and R 7 ′ are each independently hydrogen or C 1 -C 4 alkyl, q is 2.
  • R 3 is a heterocycloalkyl substituted by one substituent selected from halogen, C 1 -C 4 alkyl, haloC 1 -C 4 alkyl or —NR 7 R 7 ′, R 7 and R 7 ′ are each independently hydrogen or C 1 -C 4 alkyl; more preferably, the heterocycloalkyl is pyrrolidinyl.
  • each R 5 is dependently independently halogen, C 1 -C 4 alkyl, haloC 1 -C 4 alkyl, —OR 7 , —NR 7 R 7 ′, —CN or —NO 2 , R 7 and R 7 ′ are each independently hydrogen or C 1 -C 4 alkyl, m is 1, 2 or 3.
  • each R 5 is dependently independently halogen, C 1 -C 4 alkyl, —OR 7 or —NR 7 R 7 ′, R 7 and R 7 ′ are each independently hydrogen or C 1 -C 4 alkyl, m is 1, 2 or 3.
  • the specifically preferable compound represented by the general formula (I) or a pharmaceutically acceptable salt thereof according to the present invention includes:
  • the present invention also provides a process for preparing the compound represented by the general formula (I), which comprises the steps of:
  • L represents a leaving group, including hydrogen, halogen or
  • compounds (a) and (b) are used as starting material, and subjected to substitution under a catalyst to produce an Intermediate 2; the Intermediate 2 and an Intermediate 1 are subjected to substitution or coupling reaction to produce a compound (c), the nitro group of the compound (c) is reduced to produce a compound (d), the compound (d) is acylated to produce a compound (I); or the Intermediate 2 and an Intermediate 1′ are subjected to substitution or coupling reaction to directly produce a compound (I).
  • the catalyst for carrying out the substitution reaction of the compounds (a) and (b) includes a Lewis acid such as AlCl 3 or a transition metal catalyst such as bis(pinacolato)diboron/PdCl 2 (dppf), PdCl 2 (dppf); the substitution or coupling reaction of Intermediate 2 and Intermediate 1 can also be carried out under the catalysis of a transition metal catalyst including but not limited to Pd2(dba)3/xantphos; conventional reducing agents well known in the art are used in the reduction of the nitro group, includes but is not limited to iron powders, zinc powders, sodium sulfide, H 2 /PtO 2 ; the acylation of the compound (d) is carried out with the corresponding acyl halide such as acyl chloride.
  • a Lewis acid such as AlCl 3
  • a transition metal catalyst such as bis(pinacolato)diboron/PdCl 2 (dppf), PdCl 2 (dppf)
  • the preparation process for Intermediate 1 and Intermediate 1′ comprises the steps of,
  • R 2 , R 3 and R 4 are defined as in the above general formula (I); 2,6-dichloro-3-nitropyridine is used as starting material, and subjected to etherification to produce a compound (e), the nitro group of the compound (e) is reduced to produce a compound (f), the compound (f) is then subjected to a reaction to produce a compound (g), the compound (g) is subjected to nitration to produce a compound (h), the compound (h) and R 3 H are subjected to substitution to produce a compound (i), and the compound (i) is then deprotected to produce the Intermediate 1; the compound (i) is subjected to Boc-protection to produce a compound (j), the compound (j) is then subjected to acetyl-deprotection to produce a compound (k), the nitro group of the compound (k) is reduced to produce a compound (l), the compound (l) is subjected to acylation to produce a compound (m),
  • the etherification reaction is carried out in presence of a strong base which includes, but is not limited to sodium hydride, potassium hydride, sodium hydroxide, potassium hydroxide, sodium ethoxide, and sodium methoxide; conventional reducing agents well known in the art are used in the reduction of the nitro group, includes but is not limited to iron powders, zinc powders, sodium sulfide, H 2 /PtO 2 ; protection and deprotection are the conventional method well known in the art, and are carried out in an appropriate acidic or basic condition.
  • a strong base which includes, but is not limited to sodium hydride, potassium hydride, sodium hydroxide, potassium hydroxide, sodium ethoxide, and sodium methoxide
  • conventional reducing agents well known in the art are used in the reduction of the nitro group, includes but is not limited to iron powders, zinc powders, sodium sulfide, H 2 /PtO 2
  • protection and deprotection are the conventional method well known in the art, and
  • halogen means fluoro, chloro, bromo iodo and the like, preferably fluoro, chloro and bromo, and more preferably chloro.
  • C 1 -C 4 alkyl means methyl, ethyl, propyl, isopropyl, butyl, iso-butyl, sec-butyl or tert-butyl
  • C 2 -C 4 alkyl means ethyl, propyl, isopropyl, butyl, iso-butyl, sec-butyl or tert-butyl, preferably ethyl, propyl, isopropyl or butyl, more preferably isopropyl.
  • haloC 1 -C 4 alkyl means the C 1 -C 4 alkyl, as defined herein, which is substituted with one or more halogen atoms, preferably 1-5 halogen atoms, including but not limited to, trifluoromethyl, trifluoroethyl, difluoromethyl, 1-chloro-2-fluoroethyl and the like.
  • haloC 2 -C 4 alkyl includes but is not limited to trifluoroethyl, difluoromethyl, 1-chloro-2-fluoroethyl and the like, preferably trifluoroethyl.
  • alkenyl means a mono-valent group derived from a hydrocarbon group
  • C 2 -C 6 alkenyl means an alkenyl group containing 2 to 6 carbon atoms and at least containing one C—C double bond, including but not limited to, ethenyl, propenyl, butenyl, 2-methyl-2-butenyl, 2-methyl-2-pentenyl and the like.
  • alkynyl means a mono-valent group derived from a hydrocarbon group
  • C 2 -C 6 alkynyl means an alkynyl group containing 2 to 6 carbon atoms and at least containing one C—C triple bond, including but not limited to, ethynyl, propynyl, 1-butynyl, 2-butynyl and the like.
  • cycloalkyl means a mono-valent group derived from monocyclic or polycyclic, saturated or partially unsaturated aliphatic carbocyclic compounds
  • C 3 -C 8 -cycloalkyl includes but is not limited to cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclooctenyl, cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl, cycloheptenyl
  • C 9 -C 12 - includes but is not limited to bicyclo[2.2.1]heptyl, bicyclo[2.2.1]octyl and the like.
  • heterocycloalkyl means a monovalent monocyclic group, which is saturated or partially unsaturated (but not aromatic) and contains 3-8 ring members, preferably 4-7 ring members, or a monovalent fused bicyclic group, which is saturated or partially unsaturated (but not aromatic) and contains 5-12 ring members, preferably 7-10 ring members, wherein 1-4 ring heteroatom(s) is/are selected from a group consisting of O, S and N, and the remaining ring atoms are carbon.
  • Said heterocycloalkyl includes but is not limited to azetidinyl, oxetanyl, pyrrolidinyl, piperidyl, morpholinyl, piperazinyl, tetrahydropyranyl, pyrazolidinyl, pyrazolinyl, imidazolinyl, imidazolidinyl, [1,3]dioxolane (dioxolane), dihydropyridinyl, tetrahydropyridinyl, hexahydropyridinyl, oxazolinyl, oxazolidinyl, iso-oxazolidinyl, thiazolinyl, thiazolidinyl, tetrahydrothiazolyl, iso-tetrahydrothiazolyl, octahydroindolyl, octahydroisoindolyl, tetrahydrofuryl
  • aryl means an aromatic cyclic hydrocarbyl, which is a fused or non-fused carbonaceous ring system containing one or more aromatic rings, and includes but is not limited to phenyl, naphthyl, tetrahydronaphthyl, indanyl, indenyl and the like, preferably an aryl containing 6-14 carbon atoms, more preferably an aryl containing 6-10 carbon atoms, such as phenyl and naphthyl, more preferably phenyl.
  • heteroaryl means 5-6 membered monocyclic heteroaryl containing 1-4 heteroatoms selected from N, S or O, or bicyclic heteroaryl formed by fusing said 5-6 membered monocyclic heteroaryl with a benzene ring, pyridine ring or pyrrole ring, said heteroaryl can be partially saturated.
  • Said heteroaryl includes but is not limited to furyl, thienyl, pyrrolyl, imidazolyl, pyrazolyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, triazolyl, tetrazolyl, thiadiazolyl, pyridinyl, pyrimidinyl, pyridazinyl, pyrazinyl, benzofuranyl, benzothienyl, benzothiadiazolyl, benzothiazolyl, benzimidazolyl, indolyl, isoindolyl, indazolyl, quinolyl, isoquinolyl, quinazolinyl, 1,2,3,4-tetrahydroisoquinolyl, pyrro[2,3-c]pyridinyl pyrrolo[2,3-c]pyridinyl, pyrro[3,2-c]pyridinyl pyrrol
  • the present invention also includes the pharmaceutically acceptable salt of the compound represented by formula (I).
  • pharmaceutically acceptable salt means relatively nontoxic acid addition salts or base addition salts of the compound of the present invention.
  • Said acid addition salts are the salts formed between the compound represented by formula (I) of the present invention and suitable inorganic acids or organic acids. Said salts may be prepared during the final separation and purification processes of the compounds, or may be prepared through the reaction of purified compound represented by formula (I) in the form of free base thereof and suitable organic acids or inorganic acids.
  • Representative acid addition salts includes hydrobromic acid salt, hydrochloric acid salt, sulfate, bisulfate, sulfite, acetate, oxalate, valerate, oleate, palmate, stearate, laurate, borate, benzoate, lactate, phosphate, hydrogen phosphate, carbonate, bicarbonate, toluate, citrate, maleate, fumarate, succinate, tartrate, benzoate, mesylate, p-tosylate, glyconate, lactobionate and laurylsulfonate and the like.
  • Said base addition salts are the salts formed between the compound represented by formula (I) and suitable inorganic bases or organic bases, including such as the salts formed with alkali metals, alkaline earth metals, quaternary ammonium cations, such as sodium salts, lithium salts, potassium salts, calcium salts, magnesium salts, tetramethylammonium salts, tetraethylammonium salt and the like; amine salts, including the salts formed with ammonia (NH 3 ), primary amines, secondary amines or tertiary amines, such as: methylamine salts, dimethylamine salts, trimethylamine salts, triethylamine salts, ethylamine salts and the like.
  • suitable inorganic bases or organic bases including such as the salts formed with alkali metals, alkaline earth metals, quaternary ammonium cations, such as sodium salts, lithium salts, potassium salts, calcium salts, magnesium salts
  • the compound of the present invention or a pharmaceutically acceptable salt thereof can be administered to mammals, such as human, and administrated orally, rectally, parenterally (intravenously, intramuscularly or subcutaneously), topically (such as in the form of powders, ointments or drops), or intratumorally.
  • the administration dosage of the compound of the present invention can be about 0.05-50 mg/kg body weight/day, e.g. 0.1-45 mg/kg body weight/day, 0.5-35 mg/kg body weight/day.
  • the compound of the present invention or a pharmaceutically acceptable salt thereof can be formulated into the solid dosage forms for oral administration, which includes but is not limited to capsules, tablets, pills, powders and granules and the like.
  • the compounds represented by formula (I) of the present invention as active ingredients are admixed with at least one conventional inert excipients (or carriers), such as sodium citrate or dicalcium phosphate, or admixed with the following ingredients: (1) fillers or extenders, such as, starch, lactose, sucrose, glucose, mannitol and silicic acid and the like; (2) adhesives, such as, hydroxymethylcellulose, alginate, gelatin, polyvinyl pyrrolidine, sucrose and acacia and the like; (3) humectants, such as, glycerol and the like; (4) disintegrating agents, such as, agar, calcium carbonate, potato starch or tapioca, alginic acid, certain composite silicate and sodium carbon
  • Said solid dosage forms such as tablets, sugar pills, capsules, pills and granules can also by coated or microencapsulated by coatings and shell materials such as enteric coatings and other materials well known in the art. They may comprise opacifying agents, and the release of active ingredients in these compositions may be carried out in a certain portion of digestive tube in a retarded manner.
  • coatings and shell materials such as enteric coatings and other materials well known in the art. They may comprise opacifying agents, and the release of active ingredients in these compositions may be carried out in a certain portion of digestive tube in a retarded manner.
  • the examples for embedding components that may be adopted are polymers and waxes. If necessary, active ingredients can also be formulated into the form of microcapsules with one or more of the above excipients.
  • the compound of the present invention or a pharmaceutically acceptable salt thereof can be formulated into liquid dosage forms for oral administration, including but not limited to pharmaceutically acceptable emulsions, solutions, suspensions, syrups and tinctures and the like.
  • the liquid dosage forms may comprise inert diluents customarily used in the art, such as water and other solvents, solubilizers and emulsifiers, such as, ethanol, isopropyl alcohol, ethyl carbonate, ethyl acetate, propylene glycol, 1,3-butanediol, dimethyl formamide, and oils, especially cottonseed oil, peanut oil, corn germ oil, olive oil, castor oil and sesame oil and the like or mixtures of these materials and the like.
  • the liquid dosage forms of the present invention may also comprise conventional auxiliaries, such as moistening agents, emulsifiers
  • Said suspending agents includes, such as, ethoxylated isostearyl alcohol, polyoxyethylene sorbitol and sorbitan ester, microcrystalline cellulose, aluminium methoxide and agar and the like or mixtures of these materials.
  • the compound of the present invention or a pharmaceutically acceptable salt thereof can be formulated into dosage forms for parenteral injection, including but not limited to physiologically acceptable sterile aqueous or anhydrous solutions, dispersions, suspensions or emulsions, and sterile powder for re-dissolving into sterile injectable solutions or dispersions.
  • Suitable carriers, diluents, solvents or excipients include water, ethanol, polyhydric alcohol and suitable mixtures thereof.
  • the compound of the present invention or a pharmaceutically acceptable salt thereof can also be formulated into dosage forms for topical administration, including but not limited to ointments, powders, suppositories, drops, propellants and inhalants and the like.
  • the compounds represented by formula (I) of the present invention or a pharmaceutically acceptable salt thereof as active ingredients are admixed together with physiologically acceptable carriers and optional preservatives, buffers, or if necessary, propellants, under sterile condition.
  • the present invention also provides a pharmaceutical composition containing the compound represented by formula (I) of the present invention or a pharmaceutically acceptable salt thereof as active ingredients, and pharmaceutically acceptable carriers, excipients or diluents.
  • the compound represented by formula (I) of the present invention or a pharmaceutically acceptable salt thereof is generally admixed with pharmaceutically acceptable carriers, excipients or diluents.
  • the content of the compound of the general formula (I) or a pharmaceutically acceptable salt thereof can be 0.01-1000 mg, for example 0.05-800 mg, 0.1-500 mg, 0.01-300 mg, 0.01-200 mg, 0.05-150 mg, 0.05-50 mg and the like.
  • composition of the present invention may be formulated into conventional pharmaceutical preparations, such as tablets, pills, capsules, powder, granules, emulsions, suspensions, dispersions, solutions, syrups, elixirs, ointments, drops, suppositories, inhalants, propellants and the like.
  • the compound of the present invention or a pharmaceutically acceptable salt thereof may be administered alone or in combination with other pharmaceutically acceptable therapeutic agents, especially with other anti-tumor drugs.
  • the therapeutic agents include but are not limited to anti-tumor drugs which exert an influence on the chemical structure of DNA, such as Cisplatin, anti-tumor drugs which affect the synthesis of nucleic acid, such as Methotrexate (MTX), 5-Fluorouracil (5FU) and the like, anti-tumor drugs which affect the transcription of nucleic acid, such as Adriamycin, Epirubicin, Aclacinomycin, Mitramycin and the like, anti-tumor drugs which exert an influence on synthesis of tubulin, such as Paclitaxel, Vinorelbine and the like, aromatase inhibitors such as Aminoglutethimide, Lentaron, Letrozole, Anastrozole and the like, inhibitors of the cell signal pathway such as epidermal growth factor receptor inhibitors Imatinib, Gefitinib
  • Each therapeutic agent to be combined can be administered simultaneously or sequentially, and can be administered either in a unitary formulation or in separate formulations.
  • Such combination includes not only the combination of the compound of the present invention with another active ingredient but also the combination of the compound of the present invention with two or more other active ingredients.
  • the compound of the present invention has a good proliferation inhibition effect on the activating mutation or resistant mutation tumor cells and a weak proliferation inhibition effect on the wild-type EGFR cancer cells, and has a good selectivity.
  • the compound of the present invention can be used as the medicament for treating a disease or condition mediated by the activity of EGFR activating or resistant mutation, in particular tumor, e.g. cancer.
  • Said cancer includes but is not limited to, e.g.
  • lung cancer ovarian cancer, cervical cancer, breast cancer, stomach cancer, colorectal cancer, pancreatic cancer, glioma, glioblastoma, melanoma, prostate cancer, leukemia, lymphoma, non-Hodgkin's lymphoma, hepatocytes cancer, gastrointestinal stromal tumor (GIST), thyroid cancer, cholangiocarcinoma, endometrial cancer, renal cancer, anaplastic large cell lymphoma, acute myeloid leukemia (AML), multiple myeloma, mesothelioma, in particular a type of tumor wherein threonine at position 790 of the epidermal growth factor receptor is mutated into methionine (EGFR T790M).
  • GIST gastrointestinal stromal tumor
  • AML acute myeloid leukemia
  • mesothelioma in particular a type of tumor wherein threonine at position 790 of the epidermal growth factor receptor is mutated into me
  • the compound of the present invention can be used as medicament for treating the non-small cell cancer (EGFR T790M). It can be used to overcome the resistency problem caused by EGFR T790M after Gefitinib and Erlotinib are clinically used. Due to the reduced toxicity associated with the inhibition of the wild-type EGFR, it is therefore expected that the compound of the present invention will produce a relatively small toxic and side-effect upon being applied to the cancer treatment.
  • EGFR T790M non-small cell cancer
  • the pharmacodynamic action of the compound of the present invention in terms of inhibiting the proliferation of cancer cells may be assayed by conventional methods.
  • One preferable evaluation method of which is Sulforhodamine B (SRB) protein staining method, which calculates the inhibition ratio of a drug against the proliferation of cancer cells by measuring the change in optical absorption value generated after the drug has acted on the cancer cells.
  • Inhibition ratio (%) [(blank control OD ⁇ inhibitor OD)/blank control OD] ⁇ 100%
  • Blank control OD the OD value of the well of normally growed cells without the action of a drug.
  • Inhibitor OD the OD value of the well of cells with the action of the added compounds to be screened.
  • the median inhibitory concentration (IC 50 ) value is obtained by the software GraphPad Prism 5.0 by the 4-parameter logistic curve fit calculation. Each experiment is repeated three times, and the average IC 50 value for three experiments is used as the final index for the inhibitory ability.
  • the pharmacodynamic action of the compound of the present invention in terms of inhibiting the growth of transplanted tumors in animal may be assayed by conventional methods.
  • One preferable evaluation method of which is the inhibitory effect on the growth of subcutaneously transplanted tumors of human lung cancer H1975-bearing nude mice.
  • the experimental method is as follows: human lung cancer H1975 cell strain (5 ⁇ 10 6 /each mouse) is inoculated to nude mice subcutaneously at the right side of the back thereof. After the tumors grow to 100-150 mm 3 on average, the animals are divided into groups randomly according to the tumor size and the animal weight.
  • test compounds are administered by intragastric administration in a certain dosages, and solvent control groups are administered with equal amount of solvent by intragastric administration, wherein the administration is performed once per day for a continuous period of 12 days.
  • the animal weight and the tumor size are measured twice per week, so as to observe whether or not the toxic reaction occurs.
  • FIG. 1 is the tumor volume curve for subcutaneously transplanted tumors of human lung cancer H1975-bearing nude mice at the administration dosage of 25 mg/kg of the compound of Example 3 and AZD9291.
  • FIG. 2 is the body weight curve for human lung cancer H1975-bearing nude mice at the administration dosage of 25 mg/kg of the compound of Example 3 and AZD9291.
  • 6-chloro-2-methoxypyridin-3-amine 1.6 g, 10.1 mmol
  • diisopropylethylamine 2.6 ml, 15.1 mmol
  • dichloromethane 100 ml dichloromethane.
  • the mixture was cooled to 5° C. in an ice bath.
  • Acetyl chloride (0.86 ml, 12.1 mmol) was added. The reaction continued for 1.25 hours.
  • Step 5 Synthesis of N- ⁇ 6- ⁇ [2-(dimethylamino)ethyl](methyl)amino ⁇ -2-methoxy-5-nitropyridin-3-yl ⁇ acetamide hydrochloride
  • N-(6-chloro-2-methoxy-5-nitropyridin-3-yl)acetamide 1.0 g, 4.1 mmol
  • 30 ml acetonitrile 30 ml acetonitrile
  • N,N,N′-trimethylethylenediamine 0.6 g, 6.1 mmol
  • the reaction mixture was reacted at 80° C. for 3 hours.
  • the reaction mixture was concentrated under a reduced pressure to about 1 ⁇ 3 of the original volume.
  • 50 ml ethyl acetate was added.
  • the mixture was stirred for several minutes, a solid precipitated and was filtered to produce 1.1 g of an beige solid with a yield of 87%.
  • Step 6 Synthesis of N 2 -methyl-N 2 -[2-(dimethylamino)ethyl]-6-methoxy-3-nitropyridin-2,5-diamine hydrochloride
  • Step 5 Synthesis of N- ⁇ 6- ⁇ [2-(dimethylamino)ethyl](methyl)amino ⁇ -2-isopropyloxy-5-nitropyridin-3-yl ⁇ acetamide
  • N-(6-chloro-2-isopropyloxy-5-nitropyridin-3-yl)acetamide 15 g, 54.8 mmol
  • 150 ml acetonitrile 150 ml acetonitrile
  • N,N,N′-trimethylethylenediamine 7.28 g, 71.3 mmol
  • potassium carbonate 15.15 g, 110 mmol
  • Step 6 Synthesis of N 2 -methyl-N 2 -[2-(dimethylamino)ethyl]-6-isopropyloxy-3-nitropyridin-2,5-diamine
  • Step 5 Synthesis of N- ⁇ 6- ⁇ [2-(dimethylamino)ethyl](methyl)amino ⁇ -2-(2,2,2-trifluoroethoxyl)-5-nitropyridin-3-yl ⁇ acetamide
  • N-[6-chloro-2-(2,2,2-trifluoroethoxyl)]-5-nitropyridin-3-yl)acetamide (626 mg, 2 mmol)
  • 10 ml acetonitrile 10 ml acetonitrile
  • N,N,N′-trimethylethylenediamine 224 mg, 2.2 mmol
  • potassium carbonate 138 mg, 4 mmol
  • Step 6 Synthesis of N 2 -methyl-N 2 -[2-(dimethylamino)ethyl]-6-(2,2,2-trifluoroethoxyl)-3-nitropyridin-2,5-diamine
  • Step 1 Synthesis of N-tert-butoxycarbonyl-N- ⁇ 6- ⁇ [2-(dimethylamino)ethyl](methyl)amino ⁇ -2-isopropyloxy-5-nitropyridin-3-yl ⁇ acetamide
  • Step 2 Synthesis of tert-butyl ⁇ 6-( ⁇ [2-(dimethylamino)ethyl](methyl)amino ⁇ -2-isopropyloxy-5-nitropyridin-3-yl ⁇ carbamate
  • N-tert-butoxycarbonyl-N- ⁇ 6- ⁇ [2-(dimethylamino)ethyl](methyl)amino ⁇ -2-isopropyloxy-5-nitropyridin-3-yl ⁇ acetamide 24 g, 54.6 mmol
  • 240 ml methanol 240 ml methanol.
  • the mixture was cooled to 0° C.
  • Sodium methoxide (2.95 g, 54.6 mmol) was added.
  • the mixture was slowly warmed up to room temperature and reacted overnight.
  • the reaction mixture was concentrated under a reduced pressure.
  • the residue was dissolved in 300 ml ethyl acetate, and washed with 100 ml water.
  • the organic phase was dried with anhydrous sodium sulfate, filtered, and evaporated to dryness under a reduced pressure to produce 18 g of a product with a yield of 83%.
  • Step 3 Synthesis of tert-butyl ⁇ 5-amino-6- ⁇ [2-(dimethylamino)ethyl](methyl)amino ⁇ -2-isopropyloxypyridin-3-yl ⁇ carbamate
  • Step 4 Synthesis of tert-butyl ⁇ 5-acrylamide-6- ⁇ [2-(dimethylamino)ethyl](methyl)amino ⁇ -2-isopropyloxypyridin-3-yl ⁇ carbamate
  • Step 2 Synthesis of tert-butyl ⁇ 6- ⁇ [2-(dimethylamino)ethyl](methyl)amino ⁇ -2-(2,2,2-trifluoroethoxyl)-5-nitropyridin-3-yl ⁇ carbamate
  • Step 3 Synthesis of tert-butyl ⁇ 5-amino-6- ⁇ [2-(dimethylamino)ethyl](methyl)amino ⁇ -2-(2,2,2-trifluoroethoxyl)pyridin-3-yl ⁇ carbamate
  • Step 4 Synthesis of tert-butyl ⁇ 5-acrylamide-6- ⁇ [2-(dimethylamino)ethyl](methyl)amino ⁇ -2-(2,2,2-trifluoroethoxyl)pyridin-3-yl ⁇ carbamate
  • Step 1 Synthesis of 3-bromo-1-p-tosyl-1H-pyrro[2,3-b]pyridine 3-bromo-1-p-tosyl-1H-pyrrolo[2,3-b]pyridine
  • Step 2 Synthesis of 3-(2,5-dichloropyrimidin-4-yl)-1-p-tosyl-1H-pyrro[2,3-b]pyridine 3-(2,5-dichloropyrimidin-4-yl)-1-p-tosyl-1H-pyrrolo[2,3-b]pyridine
  • Step 3 Synthesis of 3-(2,5-dichloropyrimidin-4-yl)-1H-pyrro[2,3-b]pyridine 3-(2,5-dichloropyrimidin-4-yl)-1H-pyrrolo[2,3-b]pyridine
  • the reaction mixture was washed with 100 ml water.
  • the organic phase was dried with anhydrous sodium sulfate, and filtered.
  • the filtrate was evaporated to dryness under a reduced pressure.
  • the slurry was filtered by suction to produce 500 mg of an off-white solid with a yield of 83%.
  • Step 4 Synthesis of 3-(2,5-dichloropyrimidin-4-yl)-1-methyl-1H-pyrro[2,3-b]pyridine 3-(2,5-dichloropyrimidin-4-yl)-1-methyl-1H-pyrrolo[2,3-b]pyridine
  • Step 1 Synthesis of N 2 -methyl-N 2 -[2-(dimethylamino)ethyl]-6-isopropyloxy-N 5 [5-chloro-4-(1-methyl-1H-indol-3-yl)pyrimidin-2-yl]-3-nitropyridin-2,5-diamine
  • N 2 -methyl-N 2 -[2-(dimethylamino)ethyl]-6-isopropyloxy-3-nitropyridin-2,5-diamine (490 mg, 1.65 mmol)
  • 3-(2,5-dichloropyrimidin-4-yl)-1-methyl-1H-indole 550 mg, 1.98 mmol
  • tris(dibenzylideneacetone)dipalladium (226 mg, 0.2475 mmol
  • 4,5-Bis(diphenylphosphino)-9,9-dimethylxanthene (286 mg, 0.495 mmol)
  • potassium phosphate (874 mg, 4.125 mmol) and 15 ml dioxane.
  • Step 2 Synthesis of N 2 -methyl-N 2 -[2-(dimethylamino)ethyl]-6-isopropyloxy-N 5 -[5-chloro-4-(1-methyl-1H-indol-3-yl)pyrimidin-2-yl]pyridin-2,3,5-triamine
  • a reduced iron powders 240 mg, 4.26 mmol. The mixture was stirred at 80° C. for 1 hour.
  • Step 3 Synthesis of N- ⁇ 2- ⁇ [2-(dimethylamino)ethyl](methyl)amino ⁇ -6-isopropyloxy-5- ⁇ 5-chloro-[4-(1-methyl-1H-indol-3-yl)pyrimidin-2-yl]amino ⁇ pyridin-3-yl ⁇ acrylamide
  • N 2 -methyl-N 2 -[2-(dimethylamino)ethyl]-6-isopropyloxy-N 5 -[5-chloro-4-(1-methyl-1H-indol-3-yl)pyrimidin-2-yl]pyridin-2,3,5-triamine 196 mg, 0.385 mmol
  • the reaction mixture was cooled in an ice-water bath.
  • 0.5 N of a solution of acryloyl chloride in dichloromethane (0.8 ml, 0.4 mmol) and triethylamine (0.15 ml, 1.08 mmol) were added.
  • Step 1 Synthesis of N 2 -methyl-N 2 -[2-(dimethylamino)ethyl]-6-isopropyloxy-N 5 -[4-(1-methyl-1H-indol-3-yl)pyrimidin-2-yl]-3-nitropyridin-2,5-diamine
  • the compound was synthesized in the same manner as those in Step 1 of Example 1 with a yield of 100%.
  • Step 2 Synthesis of N 2 -methyl-N 2 -[2-(dimethylamino)ethyl]-6-isopropyloxy-N 5 -[4-(1-methyl-1H-indol-3-yl)pyrimidin-2-yl]pyridin-2,3,5-triamine
  • N 2 -methyl-N 2 -[2-(dimethylamino)ethyl]-6-isopropyloxy-N 5 -[4-(1-methyl-1H-indol-3-yl)pyrimidin-2-yl]-3-nitropyridin-2,5-diamine 200 mg, 0.397 mmol was dissolved in 12 ml methanol. 35 mg platinum dioxide was added and hydrogen was introduced. The resulting mixture was stirred at room temperature for 1.5 hour, and filtered.
  • Step 3 Synthesis of N- ⁇ 2- ⁇ [2-(dimethylamino)ethyl](methyl)amino ⁇ -6-isopropyloxy-5- ⁇ [4-(1-methyl-1H-indol-3-yl)pyrimidin-2-yl]amino ⁇ pyridin-3-yl ⁇ acrylamide
  • Step 1 Synthesis of N 2 -methyl-N 2 -[2-(dimethylamino)ethyl]-6-(2,2,2-trifluoroethoxyl)-N 5 -[4-(1-methyl-1H-indol-3-yl)pyrimidin-2-yl]-3-nitropyridin-2,5-diamine
  • Step 2 Synthesis of N 2 -methyl-N 2 -[2-(dimethylamino)ethyl]-6-(2,2,2-trifluoroethoxyl)-N 5 -[4-(1-methyl-1H-indol-3-yl)pyrimidin-2-yl]pyridin-2,3,5-triamine
  • Step 3 Synthesis of N- ⁇ 2- ⁇ [2-(dimethylamino)ethyl](methyl)amino ⁇ -6-(2,2,2-trifluoroethoxyl)-5- ⁇ [4-(1-methyl-1H-indol-3-yl)pyrimidin-2-yl]amino ⁇ pyridin-3-yl ⁇ acrylamide
  • Step 1 Synthesis of N 2 -methyl-N 2 -[2-(dimethylamino)ethyl]-6-(2,2,2-trifluoroethoxyl)-N 5 -[5-chloro-4-(1-methyl-1H-indol-3-yl)pyrimidin-2-yl]-3-nitropyridin-2,5-diamine
  • the compound was synthesized in the same manner as those in Step 1 of Example 1 with a yield of 86%.
  • Step 2 Synthesis of N 2 -methyl-N 2 -[2-(dimethylamino)ethyl]-6-(2,2,2-trifluoroethoxyl)-N 5 -[5-chloro-4-(1-methyl-1H-indol-3-yl)pyrimidin-2-yl]pyridin-2,3,5-triamine
  • Step 3 Synthesis of N- ⁇ 2- ⁇ [2-(dimethylamino)ethyl](methyl)amino ⁇ -6-(2,2,2-trifluoroethoxyl)-5- ⁇ 5-chloro-[4-(1-methyl-1H-indol-3-yl)pyrimidin-2-yl]amino ⁇ pyridin-3-yl ⁇ acrylamide
  • Step 1 Synthesis of N 2 -methyl-N 2 -[2-(dimethylamino)ethyl]-6-isopropyloxy-N 5 -[4-(1-methyl-5-fluoro-1H-indol-3-yl)pyrimidin-2-yl]-3-nitropyridin-2,5-diamine
  • Step 2 Synthesis of N 2 -methyl-N 2 -[2-(dimethylamino)ethyl]-6-isopropyloxy-N 5 -[4-(1-methyl-5-fluoro-1H-indol-3-yl)pyrimidin-2-yl]pyridin-2,3,5-triamine
  • Step 3 Synthesis of N- ⁇ 2- ⁇ [2-(dimethylamino)ethyl](methyl)amino ⁇ -6-isopropyloxy-5- ⁇ [4-(1-methyl-5-fluoro-1H-indol-3-yl)pyrimidin-2-yl]amino ⁇ pyridin-3-yl ⁇ acrylamide
  • Step 1 Synthesis of N 2 -methyl-N 2 -[2-(dimethylamino)ethyl]-6-isopropyloxy-N 5 -[4-(1-methyl-5,6-difluoro-1H-indol-3-yl)pyrimidin-2-yl]-3-nitropyridin-2,5-diamine
  • Step 2 Synthesis of N 2 -methyl-N 2 -[2-(dimethylamino)ethyl]-6-isopropyloxy-N 5 -[4-(1-methyl-5,6-difluoro-1H-indol-3-yl)pyrimidin-2-yl]pyridin-2,3,5-triamine
  • Step 3 Synthesis of N- ⁇ 2- ⁇ [2-(dimethylamino)ethyl](methyl)amino ⁇ -6-isopropyloxy-5- ⁇ [4-(1-methyl-5,6-difluoro-1H-indol-3-yl)pyrimidin-2-yl]amino ⁇ pyridin-3-yl ⁇ acrylamide
  • Example 8 N- ⁇ 2- ⁇ [2-(dimethylamino)ethyl](methyl)amino ⁇ -6-isopropyloxy-5- ⁇ 5-chloro-[4-(1-methyl-5,6-difluoro-1H-indol-3-yl)pyrimidin-2-yl]amino ⁇ pyridin-3-yl ⁇ acrylamide
  • Example 17 N- ⁇ 2- ⁇ [2-(dimethylamino)ethyl](methyl)amino ⁇ -6-isopropyloxy-5- ⁇ 5-chloro-[4-(1-methyl-5,6-difluoro-1H-indol-3-yl)pyrimidin-2-yl]amino ⁇ pyridin-3-yl ⁇ acrylamide methanesulfonate
  • the compound was synthesized in the substantially same manner as those in Example 16. Ethyl acetate was added to the final crude product. The mixture was ultrasonically treated and filtered to produce a product with a yield of 43%.
  • the compound was synthesized in the substantially same manner as those in Example 16. Ethyl acetate was added to the final crude product. The mixture was ultrasonically treated and filtered to produce a product with a yield of 96%.
  • Example 19 N- ⁇ 2- ⁇ [2-(dimethylamino)ethyl](methyl)amino ⁇ -6-isopropyloxy-5- ⁇ [5-chloro-4-(1-methyl-1H-pyrro[2,3-b]pyridin-3-yl)pyrimidin-2-yl]amino ⁇ pyridin-3-yl ⁇ acrylamide N- ⁇ 2- ⁇ [2-(dimethylamino)ethyl](methyl)amino ⁇ -6-isopropyloxy-5- ⁇ [5-chloro-4-(1-methyl-1H-pyrrolo[2,3-b]pyridin-3-yl)pyrimidin-2-yl]amino ⁇ pyridin-3-yl ⁇ acrylamide
  • Step 1 Synthesis of N 2 -methyl-N 2 -[2-(dimethylamino)ethyl]-6-isopropyloxy-N 5 -[5-chloro-4-(1-methyl-1H-pyrro[2,3-b]pyridin-3-yl)pyrimidin-2-yl]-3-nitropyridin-2,5-diamine N 2 -methyl-N 2 -[2-(dimethylamino)ethyl]-6-isopropyloxy-N 5 -[5-chloro-4-(1-methyl-1H-pyrrolo[2,3-b]pyridin-3-yl)pyrimidin-2-yl]-3-nitropyridin-2,5-diamine
  • Step 2 Synthesis of N 2 -methyl-N 2 -[2-(dimethylamino)ethyl]-6-isopropyloxy-N 5 -[5-chloro-4-(1-methyl-1H-pyrro[2,3-b]pyridin-3-yl)pyrimidin-2-yl]pyridin-2,3,5-triamine N 2 -methyl-N 2 -[2-(dimethylamino)ethyl]-6-isopropyloxy-N 5 -[5-chloro-4-(1-methyl-1H-pyrrolo[2,3-b]pyridin-3-yl)pyrimidin-2-yl]pyridin-2,3,5-triamine
  • Step 3 Synthesis of N- ⁇ 2- ⁇ [2-(dimethylamino)ethyl](methyl)amino ⁇ -6-isopropyloxy-5- ⁇ [5-chloro-4-(1-methyl-1H-pyrro[2,3-b]pyridin-3-yl)pyrimidin-2-yl]amino ⁇ pyridin-3-yl ⁇ acrylamide N- ⁇ 2- ⁇ [2-(dimethylamino)ethyl](methyl)amino ⁇ -6-isopropyloxy-5- ⁇ [5-chloro-4-(1-methyl-1H-pyrrolo[2,3-b]pyridin-3-yl)pyrimidin-2-yl]amino ⁇ pyridin-3-yl ⁇ acrylamide
  • Example 20 N- ⁇ 2- ⁇ [2-(dimethylamino)ethyl](methyl)amino ⁇ -6-isopropyloxy-5- ⁇ [5-chloro-4-(1-methyl-1H-pyrro[2,3-b]pyridin-5-yl)pyrimidin-2-yl]amino ⁇ pyridin-3-yl ⁇ acrylamide N- ⁇ 2- ⁇ [2-(dimethylamino)ethyl](methyl)amino ⁇ -6-isopropyloxy-5- ⁇ [5-chloro-4-(1-methyl-1H-pyrrolo[2,3-b]pyridin-5-yl)pyrimidin-2-yl]amino ⁇ pyridin-3-yl ⁇ acrylamide
  • Cells in the logarithmic phase were inoculated to 96-well culture plates (cell density: 5000/well, cell suspension: 180 ⁇ l/well), and cultured at 37° C. under 5% CO 2 for 24 hours. After the culturing, the cells adhered to the well walls.
  • Each of compounds was dissolved in DMSO in advance to formulate a 10 nM stock solution. Upon testing, the stock solution was diluted with complete medium to 10 times the target concentration in another 96-cell plate. And then the compound was added at 20 ⁇ l/cell in the 96-well plate in which the cells were inoculated, i.e. the target concentration could be reached. The well for each concentration was triplicated, and the blank control was established.
  • Cells continued to be cultured at 37° C. under 5% CO 2 for 72 hours. After the termination of culturing, 50 ⁇ l pre-cooled (4° C.) 50% trichloroacetic acid, i.e., TCA was added to each of wells (final concentration 10%), and was placed at 4° C. for 1 hour to fix the cells. The resulting matter was washed with purified water for at least 5 times, and dried naturally in air or at 60° C. in an oven. 4 mg/ml Sulforhodamine B (SRB) solution prepared by 1% glacial acetic acid/purified water was added at 100 ⁇ l/well to each well so as to stain for 1 hour at room temperature. The supernatant was discarded.
  • SRB Sulforhodamine B
  • the test results showed that the compounds of the present invention had a strong proliferation inhibition effect on human lung cancer cell (HCC827, EGFR Exon 19 deletion activating mutation) and human lung cancer cell (H1975, EGFR L858R/T790M resistant mutation), a relatively weak proliferation inhibition effect on human skin cancer cell (A431, wild-type EGFR), that is to say, the compounds of the present invention had a good selectivity.
  • H1975 was placed in a RPMI-1640 medium containing 10% FBS, and cultivated in a temperature-constant incubator containing 5% CO 2 at 37° C. The cells in exponential growth phase were collected and counted for inoculation.
  • Test animals 15 BALB/c nude mices, 15 males and 0 female, 6 weeks old, 18-20 g, commercially available from Shanghai Lab. Animal Research Center
  • test groups were established: 0.5% sodium carboxymethylcellucose solvent control group, the groups of the compound of Example 3 at 25 mg/kg and the groups of AZD9291 at 25 mg/kg, respectively.
  • mice human lung cancer H1975 cell strain (5 ⁇ 10 6 /each mouse) was inoculated to nude mice subcutaneously at the right side of the back thereof. Each mouse was inoculated with 0.1 ml, and the tumor growth was observed regularly. After the tumors grew to 100-150 mm 3 on average, the mice were divided into groups randomly according to the tumor size and the mouse weight.
  • the compound of Example 3 and AZD9291 were administered by intragastric administration in the dosage of 25 mg/kg, and solvent control groups were administered with equal amount of solvent by intragastric administration, wherein the administration was performed once per day for a continuous period of 12 days. During the entire experimental process, the mouse weight and the tumor size were measured twice per week, so as to observe whether or not the toxic reaction occurs.
  • the tumor growth curves of three experimental groups are shown in FIG. 1
  • the mice's weight growth curves are shown in FIG. 2 .
  • the results show that the compounds of the present invention have a good inhibition effect on the growth of subcutaneously transplanted tumors of human lung cancer H1975-bearing nude mice, while having little effect on the weights of nude mice, and showing a good safety.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Hematology (AREA)
  • Oncology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
US16/517,790 2014-07-29 2015-07-29 Pyridinylaminopyrimidine derivatives, preparation process and use thereof Active USRE48687E1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/517,790 USRE48687E1 (en) 2014-07-29 2015-07-29 Pyridinylaminopyrimidine derivatives, preparation process and use thereof

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
CN201410365911.4A CN105315259B (zh) 2014-07-29 2014-07-29 吡啶胺基嘧啶衍生物、其制备方法及应用
CN201410365911.4 2014-07-29
PCT/CN2015/000540 WO2016015453A1 (zh) 2014-07-29 2015-07-29 吡啶胺基嘧啶衍生物、其制备方法及应用
US16/517,790 USRE48687E1 (en) 2014-07-29 2015-07-29 Pyridinylaminopyrimidine derivatives, preparation process and use thereof
US15/329,044 US10072002B2 (en) 2014-07-29 2015-07-29 Pyridinylaminopyrimidine derivatives, preparation process and use thereof

Publications (1)

Publication Number Publication Date
USRE48687E1 true USRE48687E1 (en) 2021-08-17

Family

ID=55216719

Family Applications (3)

Application Number Title Priority Date Filing Date
US17/376,987 Active USRE49851E1 (en) 2014-07-29 2015-07-29 Pyridinylaminopyrimidine derivatives, preparation process and use thereof
US16/517,790 Active USRE48687E1 (en) 2014-07-29 2015-07-29 Pyridinylaminopyrimidine derivatives, preparation process and use thereof
US15/329,044 Ceased US10072002B2 (en) 2014-07-29 2015-07-29 Pyridinylaminopyrimidine derivatives, preparation process and use thereof

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US17/376,987 Active USRE49851E1 (en) 2014-07-29 2015-07-29 Pyridinylaminopyrimidine derivatives, preparation process and use thereof

Family Applications After (1)

Application Number Title Priority Date Filing Date
US15/329,044 Ceased US10072002B2 (en) 2014-07-29 2015-07-29 Pyridinylaminopyrimidine derivatives, preparation process and use thereof

Country Status (9)

Country Link
US (3) USRE49851E1 (ja)
EP (1) EP3181560B1 (ja)
JP (1) JP6431593B6 (ja)
KR (1) KR101883125B1 (ja)
CN (1) CN105315259B (ja)
CA (1) CA2956628C (ja)
ES (1) ES2855050T3 (ja)
PL (1) PL3181560T3 (ja)
WO (1) WO2016015453A1 (ja)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
USRE49851E1 (en) * 2014-07-29 2024-02-27 Shanghai Allist Pharmaceuticals Co., Ltd. Pyridinylaminopyrimidine derivatives, preparation process and use thereof

Families Citing this family (41)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108430972B (zh) 2015-12-24 2022-12-27 协和麒麟株式会社 α,β不饱和酰胺化合物
EP3399968B8 (en) 2016-01-07 2021-12-01 Xuanzhu Biopharmaceutical Co., Ltd. Selective inhibitors of clinically important mutants of the egfr tyrosine kinase
CN107163027B (zh) 2016-03-07 2019-11-05 上海艾力斯医药科技有限公司 吡啶胺基嘧啶衍生物甲磺酸盐的结晶形式及其制备和应用
CN107163026B (zh) * 2016-03-07 2019-07-02 上海艾力斯医药科技有限公司 吡啶胺基嘧啶衍生物的盐及其制备方法和应用
WO2018019204A1 (zh) * 2016-07-26 2018-02-01 深圳市塔吉瑞生物医药有限公司 用于抑制蛋白酪氨酸激酶活性的氨基嘧啶类化合物
AU2018289759B2 (en) * 2017-06-23 2022-03-10 Kyowa Kirin Co., Ltd. Alpha, beta-unsaturated amide compound
DE102017123894B3 (de) 2017-10-13 2019-02-07 Carl Zeiss Meditec Ag Scheibe für ein HMD und HMD mit wenigstens einer Scheibe
WO2019164945A1 (en) * 2018-02-20 2019-08-29 Dana-Farber Cancer Institute, Inc. Pharmaceutical combinations of egfr inhibitors and methods of use thereof
CN110606842B (zh) * 2018-06-15 2021-06-01 上海艾力斯医药科技股份有限公司 吡啶胺基嘧啶衍生物的制备方法及其中间体
CN110606841A (zh) * 2018-06-15 2019-12-24 上海艾力斯医药科技有限公司 吡啶胺基嘧啶衍生物的晶型及其制备方法
CN110790749B (zh) * 2018-08-03 2023-07-14 北京普祺医药科技股份有限公司 一种含氮杂环化合物、药物组合物以及其用途
CN110857292A (zh) * 2018-08-22 2020-03-03 上海艾力斯医药科技有限公司 一种egfr激酶抑制剂及其制备方法和应用
US11066404B2 (en) 2018-10-11 2021-07-20 Incyte Corporation Dihydropyrido[2,3-d]pyrimidinone compounds as CDK2 inhibitors
US20220048891A1 (en) * 2018-12-21 2022-02-17 Shenzhen Targetrx, Inc. Aminopyrimidine compound used for inhibiting activity of protein kinase
US11384083B2 (en) 2019-02-15 2022-07-12 Incyte Corporation Substituted spiro[cyclopropane-1,5′-pyrrolo[2,3-d]pyrimidin]-6′(7′h)-ones as CDK2 inhibitors
TW202100520A (zh) 2019-03-05 2021-01-01 美商英塞特公司 作為cdk2 抑制劑之吡唑基嘧啶基胺化合物
US11919904B2 (en) 2019-03-29 2024-03-05 Incyte Corporation Sulfonylamide compounds as CDK2 inhibitors
CN111747950B (zh) * 2019-03-29 2024-01-23 深圳福沃药业有限公司 用于治疗癌症的嘧啶衍生物
US11440914B2 (en) 2019-05-01 2022-09-13 Incyte Corporation Tricyclic amine compounds as CDK2 inhibitors
WO2020223558A1 (en) 2019-05-01 2020-11-05 Incyte Corporation Tricyclic amine compounds as cdk2 inhibitors
JP2022534063A (ja) * 2019-05-22 2022-07-27 上▲海▼翰森生物医▲薬▼科技有限公司 インドール誘導体含有阻害剤、そのための調製方法及びその用途
CR20220066A (es) 2019-08-14 2022-11-28 Incyte Corp Compuestos de imidazolil pirimidinilamina como inhibidores de cdk2
PE20221905A1 (es) 2019-10-11 2022-12-23 Incyte Corp Aminas biciclicas como inhibidoras de la cdk2
CN110777391B (zh) * 2019-10-24 2021-01-22 浙江大学 一种吉非替尼中间体的电还原制备方法
WO2021094379A1 (en) 2019-11-12 2021-05-20 Astrazeneca Ab Epidermal growth factor receptor tyrosine kinase inhibitors for the treatment of cancer
KR20220130190A (ko) 2020-01-20 2022-09-26 아스트라제네카 아베 암 치료를 위한 표피성장인자 수용체 티로신 키나제 억제제
US20210369709A1 (en) 2020-05-27 2021-12-02 Astrazeneca Ab EGFR TKIs FOR USE IN THE TREATMENT OF NON-SMALL CELL LUNG CANCER
WO2022166916A1 (zh) * 2021-02-05 2022-08-11 深圳市塔吉瑞生物医药有限公司 取代的杂芳基衍生物及其组合物及用途
US11981671B2 (en) 2021-06-21 2024-05-14 Incyte Corporation Bicyclic pyrazolyl amines as CDK2 inhibitors
WO2023035223A1 (zh) * 2021-09-10 2023-03-16 上海艾力斯医药科技股份有限公司 药物组合物及其用途
CN113896716A (zh) * 2021-10-27 2022-01-07 浙江爱索拓科技有限公司 一种放射性同位素碳-14双标记甲磺酸伏美替尼合成方法
CN116159062A (zh) * 2021-11-24 2023-05-26 上海艾力斯医药科技股份有限公司 药物组合物及其用途
US11976073B2 (en) 2021-12-10 2024-05-07 Incyte Corporation Bicyclic amines as CDK2 inhibitors
WO2023187037A1 (en) 2022-03-31 2023-10-05 Astrazeneca Ab Epidermal growth factor receptor (egfr) tyrosine kinase inhibitors in combination with an akt inhibitor for the treatment of cancer
WO2023209086A1 (en) 2022-04-28 2023-11-02 Astrazeneca Ab Bicyclic heteroaromatic compounds for treating cancer
WO2023209088A1 (en) 2022-04-28 2023-11-02 Astrazeneca Ab Bicyclic heteroaromatic compounds and their use in the treatment of cancer
WO2023209090A1 (en) 2022-04-28 2023-11-02 Astrazeneca Ab Bicyclic heteroaromatic compounds and their application in the treatment of cancer
WO2023209084A1 (en) 2022-04-28 2023-11-02 Astrazeneca Ab Condensed bicyclic heteroaromatic compounds and their use in the treatment of cancer
WO2024002938A1 (en) 2022-06-27 2024-01-04 Astrazeneca Ab Combinations involving epidermal growth factor receptor tyrosine kinase inhibitors for the treatment of cancer
WO2024008929A1 (en) 2022-07-08 2024-01-11 Astrazeneca Ab Epidermal growth factor receptor tyrosine kinase inhibitors in combination with hgf-receptor inhibitors for the treatment of cancer
WO2024059962A1 (en) * 2022-09-19 2024-03-28 Shanghai Allist Pharmaceuticals Co., Ltd. Pharmaceutical composition and use thereof

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012061299A1 (en) 2010-11-01 2012-05-10 Avila Therapeutics, Inc. Heterocyclic compounds and uses thereof
CN102482277A (zh) 2009-05-05 2012-05-30 达纳-法伯癌症研究所有限公司 表皮生长因子受体抑制剂及治疗障碍的方法
WO2013014448A1 (en) 2011-07-27 2013-01-31 Astrazeneca Ab 2 - (2, 4, 5 - substituted -anilino) pyrimidine derivatives as egfr modulators useful for treating cancer
CN104761544A (zh) 2014-01-03 2015-07-08 南京波尔泰药业科技有限公司 Egfr酪氨酸激酶的临床重要突变体的选择性抑制剂
WO2015188777A1 (en) 2014-06-12 2015-12-17 Shanghai Fochon Pharmaceutical Co Ltd Certain protein kinase inhibitors
US10472349B2 (en) * 2016-03-07 2019-11-12 Shanghai Allist Pharmaceutical And Medical Tech Co Salt of pyridinyl amino pyrimidine derivative, preparation method therefor, and application thereof
US10550101B2 (en) * 2016-03-07 2020-02-04 Shanghai Allist Pharmaceutical And Medical Tech Co Crystalline forms of mesylate salt of pyridinyl amino pyrimidine derivative, preparation methods therefor, and applications thereof

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EA201391626A1 (ru) * 2011-05-04 2014-03-31 Ариад Фармасьютикалз, Инк. Соединения для ингибирования клеточной пролиферации в egfr-стимулированных типах рака
CN105315259B (zh) * 2014-07-29 2018-03-09 上海艾力斯医药科技有限公司 吡啶胺基嘧啶衍生物、其制备方法及应用

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102482277A (zh) 2009-05-05 2012-05-30 达纳-法伯癌症研究所有限公司 表皮生长因子受体抑制剂及治疗障碍的方法
WO2012061299A1 (en) 2010-11-01 2012-05-10 Avila Therapeutics, Inc. Heterocyclic compounds and uses thereof
WO2013014448A1 (en) 2011-07-27 2013-01-31 Astrazeneca Ab 2 - (2, 4, 5 - substituted -anilino) pyrimidine derivatives as egfr modulators useful for treating cancer
US20130053409A1 (en) 2011-07-27 2013-02-28 Astrazeneca Ab 2-(2,4,5-substituted-anilino) pyrimidine compounds
CN103702990A (zh) 2011-07-27 2014-04-02 阿斯利康(瑞典)有限公司 2-(2,4,5-取代苯胺)嘧啶衍生物作为egfr调谐子用于治疗癌症
US8946235B2 (en) * 2011-07-27 2015-02-03 Astrazeneca Ab 2-(2,4,5-substituted-anilino) pyrimidine compounds
CN104761544A (zh) 2014-01-03 2015-07-08 南京波尔泰药业科技有限公司 Egfr酪氨酸激酶的临床重要突变体的选择性抑制剂
WO2015188777A1 (en) 2014-06-12 2015-12-17 Shanghai Fochon Pharmaceutical Co Ltd Certain protein kinase inhibitors
US10472349B2 (en) * 2016-03-07 2019-11-12 Shanghai Allist Pharmaceutical And Medical Tech Co Salt of pyridinyl amino pyrimidine derivative, preparation method therefor, and application thereof
US10550101B2 (en) * 2016-03-07 2020-02-04 Shanghai Allist Pharmaceutical And Medical Tech Co Crystalline forms of mesylate salt of pyridinyl amino pyrimidine derivative, preparation methods therefor, and applications thereof

Non-Patent Citations (15)

* Cited by examiner, † Cited by third party
Title
Besse et al. "Neratinib (HKI-272), an irreversible pan-ErbB receptor tyrosine kinase inhibitor: preliminary results of a phase 2 trial in patients with advanced non-small cell lung cancer", Poster Session—Phase II 203:64 (2008).
Chinese Office Action corresponding to Chinese Application No. 201410365911.4 dated Jul. 25, 2017.
Extended European Search Report corresponding to European Application No. 15828199.8 dated Nov. 29, 2017.
International Search Report corresponding to International Application No. PCT/CN2015/000540 dated Oct. 28, 2015.
Jänne et al. "Multicenter, Randomized, Phase II Trial of CI-1033, an Irreversible Pan-ERBB Inhibitor, for Previously Treated Advanced Non-Small-Cell Lung Cancer", J. Clin. Oncol. 25(25):3936-3944 (2007).
Jänne et al. "Phase I Dose-Escalation Study of the Pan-HER Inhibitor, PF299804, in Patients with Advanced Malignant Solid Tumors", Clin. Cancer Res. 17:1131-1139 (2011).
Katakami et al. "LUX-Lung 4: A Phase II Trial of Afatinib in Patients with Advanced Non-Small-Cell Lung Cancer who Progressed During Prior Treatment with Erlotinib, Gefitinib, or Both", J. Clin. Oncol. 31:3335-3341 (2013).
Landi et al. "Irreversible EGFR-TKIs: dreaming perfection", Transl Lung Cancer Res. 2(1);40-49 (2013).
Li et al. "BIBW2992, an irreversible EGFR/HER2 inhibitor highly effective in preclinical lung cancer models", Oncogene 27:4702-4711 (2008).
Office Action corresponding to Canadian Application No. 2,956,628 dated Apr. 25, 2018.
Office Action corresponding to Japanese Patent Application No. 2017-504644 dated Mar. 13, 2018.
Pao et al. "Acquired Resistance of Lung Adenocarcinomas to Gefitinib or Erlotinib is Associated with a Second Mutation in the EGFR Kinase Domain", PLoS Medicine 2(3):0225-0235 (2005).
Walter et al. "Discovery of a mutant-selective covalent inhibitor of EGFR that overcomes T790M-mediated resistance in NSCLC", Cancer Discov. 3(12):1404-1415 (2013).
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)", Journal of Medicinal Chemistry, American Chemical Society 56(17):7025-7048 (2013).
Zhou et al. "Novel mutant-selective EGFR kinase inhibitors against EGFR T790M", Nature 462(24):1070-1074 (2009).

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
USRE49851E1 (en) * 2014-07-29 2024-02-27 Shanghai Allist Pharmaceuticals Co., Ltd. Pyridinylaminopyrimidine derivatives, preparation process and use thereof

Also Published As

Publication number Publication date
JP2017525685A (ja) 2017-09-07
CN105315259B (zh) 2018-03-09
US10072002B2 (en) 2018-09-11
JP6431593B6 (ja) 2019-01-09
US20170210739A1 (en) 2017-07-27
ES2855050T3 (es) 2021-09-23
EP3181560B1 (en) 2020-11-25
KR20170031778A (ko) 2017-03-21
WO2016015453A1 (zh) 2016-02-04
CA2956628A1 (en) 2016-02-04
EP3181560A4 (en) 2017-12-27
CN105315259A (zh) 2016-02-10
EP3181560A1 (en) 2017-06-21
USRE49851E1 (en) 2024-02-27
KR101883125B1 (ko) 2018-07-27
JP6431593B2 (ja) 2018-11-28
PL3181560T3 (pl) 2021-06-14
CA2956628C (en) 2018-10-23

Similar Documents

Publication Publication Date Title
USRE48687E1 (en) Pyridinylaminopyrimidine derivatives, preparation process and use thereof
JP7181558B2 (ja) Egfrモジュレーターとしての置換2-アミノピリミジン誘導体
US10239864B2 (en) 2-H-indazole derivatives as cyclin-dependent kinase (CDK) inhibitors and therapeutic uses thereof
US10912779B2 (en) Quinazolines and azaquinazolines as dual inhibitors of RAS/RAF/MEK/ERK and PI3K/AKT/PTEN/mTOR pathways
US11352341B2 (en) 2H-indazole derivatives as CDK4 and CDK6 inhibitors and therapeutic uses thereof
EP0802914B1 (en) Aryl- and heteroaryl- purine and pyridopyrimidine derivatives
JP6139782B2 (ja) 置換ピラゾロピリミジン化合物、及びその薬学的に許容される塩、並びにこれらの溶媒和物、立体異性体、及び互変異性体、並びにこれらを含む医薬組成物
US10377747B2 (en) 2-arylamino pyridine, pyrimidine or triazine derivatives, preparation method and use thereof
US20190367520A1 (en) New compound having fgfr inhibitory activity and preparation and application thereof
US9550738B2 (en) Bicyclic compounds as kinases inhibitors
US20220259235A1 (en) EGFR Inhibitor, Composition, and Preparation Method Therefor
US20170101414A1 (en) Novel quinoline-substituted compound
WO2013170770A1 (zh) 具有抗肿瘤活性的乙炔衍生物
US20150152088A1 (en) Alkynyl heteroaromatic compound and use thereof
WO2013170671A1 (zh) 蝶啶酮衍生物及其作为egfr、blk、flt3抑制剂的应用
CN112638897B (zh) 一种egfr激酶抑制剂及其制备方法和应用
EP0853616B1 (en) Bicyclic 4-aralkylaminopyrimidine derivatives as tyrosine kinase inhibitors
WO2019177374A1 (ko) 2, 4, 5-치환된 피리미딘 유도체, 이의 제조방법 및 이를 유효성분으로 포함하는 암 또는 염증질환의 예방 또는 치료용 약학적 조성물
WO2020125513A1 (zh) 作为cdk抑制剂的大环化合物、其制备方法及其在医药上的应用
WO2017211216A1 (zh) 稠合嘧啶哌啶环衍生物及其制备方法和应用
CN115109036B (zh) 一种喹唑啉类化合物、组合物及其应用
WO2022171139A1 (zh) 一种巨环化合物、药物组合物以及其用途
CN113527215A (zh) 一种喹唑啉类化合物、制备方法及其应用

Legal Events

Date Code Title Description
FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO UNDISCOUNTED (ORIGINAL EVENT CODE: BIG.); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO SMALL (ORIGINAL EVENT CODE: SMAL); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

AS Assignment

Owner name: SHANGHAI ALLIST PHARMACEUTICALS CO., LTD., CHINA

Free format text: CHANGE OF NAME;ASSIGNOR:SHANGHAI ALLIST PHARMACEUTICALS, INC.;REEL/FRAME:052365/0074

Effective date: 20191223

FEPP Fee payment procedure

Free format text: PETITION RELATED TO MAINTENANCE FEES GRANTED (ORIGINAL EVENT CODE: PTGR); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

CC Certificate of correction
FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO UNDISCOUNTED (ORIGINAL EVENT CODE: BIG.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

CC Certificate of correction
MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 4TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1551); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 4

AS Assignment

Owner name: SHANGHAI ALLIST PHARMACEUTICALS CO., LTD., CHINA

Free format text: CHANGE OF ADDRESS;ASSIGNOR:SHANGHAI ALLIST PHARMACEUTICALS CO., LTD.;REEL/FRAME:063663/0430

Effective date: 20230516