CN106188138B - A kind of diaminopyrimidine compounds and the composition comprising the compound - Google Patents

A kind of diaminopyrimidine compounds and the composition comprising the compound Download PDF

Info

Publication number
CN106188138B
CN106188138B CN201610587472.0A CN201610587472A CN106188138B CN 106188138 B CN106188138 B CN 106188138B CN 201610587472 A CN201610587472 A CN 201610587472A CN 106188138 B CN106188138 B CN 106188138B
Authority
CN
China
Prior art keywords
compound
deuterium
diaminopyrimidine compounds
pharmaceutically acceptable
piperidin
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
CN201610587472.0A
Other languages
Chinese (zh)
Other versions
CN106188138A (en
Inventor
王义汉
李焕银
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Shenzhen Rui Rui Rui Biological Medicine Co Ltd
Original Assignee
Shenzhen Rui Rui Rui Biological Medicine 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
Application filed by Shenzhen Rui Rui Rui Biological Medicine Co Ltd filed Critical Shenzhen Rui Rui Rui Biological Medicine Co Ltd
Priority to CN201810922557.9A priority Critical patent/CN108948082A/en
Priority to CN202111097366.1A priority patent/CN113912648A/en
Priority to PCT/CN2016/096320 priority patent/WO2017092413A1/en
Publication of CN106188138A publication Critical patent/CN106188138A/en
Application granted granted Critical
Publication of CN106188138B publication Critical patent/CN106188138B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6558Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system
    • C07F9/65583Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system each of the hetero rings containing nitrogen as ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B59/00Introduction of isotopes of elements into organic compounds ; Labelled organic compounds per se
    • C07B59/004Acyclic, carbocyclic or heterocyclic compounds containing elements other than carbon, hydrogen, halogen, oxygen, nitrogen, sulfur, selenium or tellurium
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/05Isotopically modified compounds, e.g. labelled

Abstract

The present invention provides a kind of diaminopyrimidine compounds and comprising the composition of the compound, the invention discloses the diaminopyrimidine compounds as shown in formula (I) and the pharmaceutical compositions containing the compound or its crystal form, pharmaceutically acceptable salt, hydrate or solvate, stereoisomer, prodrug or isotopic variations.Diaminopyrimidine compounds disclosed by the invention and composition comprising the compound have excellent inhibition to protein kinase, there is better pharmacokinetic parameter characteristic simultaneously, the drug concentration of compound in animal body can be improved, to improve curative effect of medication and safety.

Description

A kind of diaminopyrimidine compounds and the composition comprising the compound
Technical field
The invention belongs to pharmaceutical technology field more particularly to a kind of diaminopyrimidine compounds and include the group of the compound Close object.
Background technology
In Past 30 Years, lung cancer mortality rises 465%, and incidence increases by 26.9% every year, it has also become China is first Position Death Cause for Malignant Tumors.Wherein non-small cell lung cancer (non-small cell lung cancer, NSCLC) accounts for all lungs 80% or more of cancer, only the NSCLC patient of one third there are the chance of operative treatment, about 70% patient when medical Belong to Locally Advanced or DISTANT METASTASES IN occur, loses the chance of operation, in this case, drug therapy is particularly important. Anaplastic lymphoma kinase (anaplasticlymphoma kinase, ALK) Gene Fusion in the recent period have become one it is important Biomarker, provide help for patient's selection of specific NSCLC subgroups, to carry out treatment using corresponding inhibitor.Lung Cancer research international association (IASLC) recommends to instruct patient screening using ALK fusion detections, and late selection can in adenocarcinoma patients The patient of ALK inhibitor for treating is adopted, no matter its sex, race, smoking history or other clinical risk factors.Using double labels point Patient of fluorescence in situ hybridization (FISH) detection for selecting acceptable ALK-TKI treatments from probe, this diagnostic method obtain U.S. FDA approval is obtained, is used in gram azoles treats the research that ALK resets tumour for Buddhist nun.Gram azoles is three phosphorus of oral type for Buddhist nun Adenosine monophosphate (ATP) competitive inhibitor, can inhibit ALK and MET tyrosine kinase, additionally it is possible to inhibit the work of ROS1 and RON kinases Property.
But gram azoles will appear following side effect for Buddhist nun:3-4 occurs for dysopia, gastrointestinal side effect, 16% case Grade liver transaminase levels increase.In addition, ALK positive patients are inevitable after gram azoles of incipient stage treats sensitive periods for Buddhist nun There is acquired resistance in ground.Therefore, for need to develop to it is with ALK kinase inhibiting activities and/or with more preferable pharmacodynamics/ The compound of pharmacokinetics performance.
Invention content
For the above technical problem, the invention discloses a kind of diaminopyrimidine compounds and include the combination of the compound Object, with better ALK kinase inhibiting activities and/or with more preferable pharmacodynamics/pharmacokinetics performance.
In this regard, the technical solution adopted by the present invention is:
The object of the present invention is to provide it is a kind of novel with ALK kinase inhibiting activities and/or with more preferable pharmacodynamics/ The compound of pharmacokinetics performance.
In the first aspect of the present invention, diaminopyrimidine compounds or its crystal form, medicine shown in a kind of formula (I) are provided Acceptable salt, hydrate or solvated compounds on.
In formula:
Wherein:R1a、R1b、R1c、R2a、R2b、R3a、R3b、R4a、R4b、R5a、R5b、R6、R7a、R7b、R8a、R8b、R9a、R9b、R10a、 R10b、R11、R12、R13、R14a、R14b、R14c、R15、R16、R17a、R17b、R17c、R18a、R18b、R18c、R19、R20、R21And R22Respectively solely It is on the spot hydrogen, deuterium, halogen or trifluoromethyl;
R16For hydrogen, deuterium, halogen, cyano, not deuterated C1-C6Alkyl or C1-C6Alkoxy, it is one or many deuterated or Complete deuterated C1-C6Alkyl or C1-C6Alkoxy, or C that one or more halogens replace or that perhalogeno element replaces1-C6Alkyl Or C1-C6Alkoxy;
Additional conditions are R1a、R1b、R1c、R2a、R2b、R3a、R3b、R4a、R4b、R5a、R5b、R6、R7a、R7b、R8a、R8b、R9a、R9b、 R10a、R10b、R11、R12、R13、R14a、R14b、R14c、R15、R16、R17a、R17b、R17c、R18a、R18b、R18c、R19、R20、R21And R22In At least one is deuterated or deuterium.
Shape and volume of the deuterium in drug molecule are substantially the same with hydrogen, if hydrogen is selectively replaced in drug molecule For deuterium, deuterated drug generally can also retain original bioactivity and selectivity.Inventor passes through it is experimentally confirmed that carbon deuterium key simultaneously Combination it is more more stable than the combination of C-H bond, the attributes such as absorption, distribution, metabolism and the excretion of some drugs can be directly affected, from And the effect of improving drug, safety and tolerance.
In another preferred example, deuterium isotopic content of the deuterium in each deuterated position is at least more than natural deuterium isotopic content (0.015%), it is preferably greater than 30%, even more preferably greater than 50%, even more preferably greater than 75%, even more preferably greater than 95%, more preferably More than 99%.
Specifically, R in the present invention1a、R1b、R1c、R2a、R2b、R3a、R3b、R4a、R4b、R5a、R5b、R6、R7a、R7b、R8a、 R8b、R9a、R9b、R10a、R10b、R11、R12、R13、R14a、R14b、R14c、R15、R16、R17a、R17b、R17c、R18a、R18b、R18c、R19、R20、 R21And R22Deuterium isotopic content is at least 5% in each deuterated position, is preferably greater than 10%, even more preferably greater than 15%, more preferably More than 20%, even more preferably greater than 25%, even more preferably greater than 30%, even more preferably greater than 35%, even more preferably greater than 40%, more preferably greatly In 45%, even more preferably greater than 50%, even more preferably greater than 55%, even more preferably greater than 60%, even more preferably greater than 65%, even more preferably greater than 70%, even more preferably greater than 75%, even more preferably greater than 80%, even more preferably greater than 85%, even more preferably greater than 90%, even more preferably greater than 95%, even more preferably greater than 99%.
In another preferred example, compound at least contains there are one D-atom in formula (I), and the number containing D-atom can be Any one in 1 to 38.
In another preferred example, compound at least contains there are one D-atom in formula (I), and the number containing D-atom can be Any one in 1 to 38.
In another preferred example, in formula (I) compound R1a、R1b、R1c、R2a、R2b、R3a、R3b、R4a、R4b、R5a、R5b、R6、 R7a、R7b、R8a、R8b、R9a、R9b、R10a、R10b、R11、R12、R13、R14a、R14b、R14c、R15、R16、R17a、R17b、R17c、R18a、R18b、 R18c、R19、R20、R21And R22, at least one of which R contains deuterium, and more preferably two R contain deuterium, and more preferably three R contain deuterium, and more preferably four A R contains deuterium, and more preferably five R contain deuterium, and more preferably six R contain deuterium, and more preferably seven R contain deuterium, and more preferably eight R contain deuterium, more preferably Nine, ground R contains deuterium, and more preferably ten R contain deuterium, and more preferably 11 R contain deuterium, and more preferably 12 R contain deuterium, more preferably 13 R Containing deuterium, more preferably 14 R contain deuterium, and more preferably 15 R contain deuterium, and more preferably 16 R contain deuterium, and more preferably 17 R contain deuterium, More preferably 18 R contain deuterium, and more preferably 19 R contain deuterium, and more preferably 20 R contain deuterium, and more preferably 21 R contain deuterium, more Good 22 R in ground contain deuterium, and more preferably 23 R contain deuterium, and more preferably 24 R contain deuterium, and more preferably 25 R contain Deuterium, more preferably 26 R contain deuterium, and more preferably 27 R contain deuterium, and more preferably 28 R contain deuterium, more preferably 29 R contains deuterium, and more preferably 30 R contain deuterium, and more preferably 31 R contain deuterium, and more preferably 32 R contain deuterium, and more preferably 33 A R contains deuterium, and more preferably 34 R contain deuterium, and more preferably 35 R contain deuterium, and more preferably 36 R contain deuterium, and more preferably three 17 R contain deuterium, and more preferably 38 R contain deuterium.
In another preferred example, R1a、R1bAnd R1cIt is each independently deuterium or hydrogen.
In another preferred example, R2a、R2b、R3a、R3b、R4a、R4b、R5aAnd R5bIt is each independently deuterium or hydrogen.
In another preferred example, R6For deuterium or hydrogen.
In another preferred example, R7a、R7b、R8a、R8b、R9a、R9b、R10aAnd R10bIt is each independently deuterium or hydrogen.
In another preferred example, R11、R12And R13It is each independently deuterium or hydrogen.
In another preferred example, R14a、R14bAnd R14cIt is each independently deuterium or hydrogen.
In another preferred example, R17a、R17bAnd R17cIt is each independently deuterium or hydrogen.
In another preferred example, R18a、R18bAnd R18cIt is each independently deuterium or hydrogen.
In another preferred example, R19、R20、R21And R22It is each independently deuterium or hydrogen.
In another preferred example, R16Separately select halogen, trifluoromethyl, cyano, one or many deuterated alkane Base and alkoxy.
In another preferred example, which is characterized in that R16It is chlorine.
In another preferred example, which is characterized in that R1a、R1b、R1cIt is deuterium.
In another preferred example, which is characterized in that R2a、R2b、R5a、R5bIt is deuterium.
In another preferred example, which is characterized in that R3a、R3b、R4a、R4bIt is deuterium.
In another preferred example, R2a、R2b、R3a、R3b、R4a、R4b、R5a、R5bIt is deuterium.
In another preferred example, R6It is deuterium.
In another preferred example, R7a、R7b、R10a、R10bIt is deuterium.
In another preferred example, R8a、R8b、R9a、R9bIt is deuterium.
In another preferred example, R7a、R7b、R8a、R8b、R9a、R9b、R10a、R10bIt is deuterium.
In another preferred example, R11、R13It is deuterium.
In another preferred example, R11、R12、R13It is deuterium.
In another preferred example, R14a、R14b、R14cIt is deuterium.
In another preferred example, R17a、R17b、R17c、R18a、R18b、R18cIt is deuterium.
In another preferred example, R20、R22It is deuterium.
In another preferred example, R19、R20、R21、R22It is deuterium.
In another preferred example, the compound is selected from the group compound or its pharmaceutically acceptable salt:
The chloro- N of 5-4[2- (solutions of dimethyl phosphoryl base) phenyl]-N2{ 2-d3- methoxyl groups -4- [4- (4- methylpiperazine-1-yls) Piperidin-1-yl] phenyl } pyrimidine -2,4- diamines, shown in structural formula such as formula (2);
The chloro- N of 5-4[2- (solutions of dimethyl phosphoryl base) phenyl]-N2{ 2- methoxyl groups -4- [4- (4-d3- methylpiperazine-1-yls) Piperidin-1-yl] phenyl } pyrimidine -2,4- diamines, shown in structural formula such as formula (3);
The chloro- N of 5-4[2- (solutions of dimethyl phosphoryl base) phenyl]-N2{ 2- methoxyl groups -4- [4- (4- thyl-piperazin -1- bases) -4- D- piperidin-1-yls] phenyl } pyrimidine -2,4- diamines, shown in structural formula such as formula (4);
The chloro- N of 5-4[2- (solutions of dimethyl phosphoryl base) phenyl]-N2{ 2- methoxyl groups -4- [4- (4- methyl -3,3,5,5-d4- piperazines Piperazine -1- bases) piperidin-1-yl] phenyl } pyrimidine -2,4- diamines, shown in structural formula such as formula (5);
In another preferred example, the compound is selected from the group compound or its pharmaceutically acceptable salt:
In another preferred example, the compound does not include non-deuterated compound.
In another preferred example, the non-deuterated compound is the chloro- N4- of 5- (2- (dimethyl oxygen phosphino-) phenyl)-N2- (2- methoxyl groups -4- (4- (4- methylpiperazine-1-yls)-piperidin-1-yl) phenyl) pyrimidine -2,4- diamines.
In the second aspect of the present invention, a kind of method preparing pharmaceutical composition, including step are provided:It will pharmaceutically Compound or its crystal form, pharmaceutically acceptable salt, hydrate described in acceptable carrier and first aspect present invention or Solvate is mixed, to form pharmaceutical composition.
In the third aspect of the present invention, provide a kind of pharmaceutical composition, it contain pharmaceutically acceptable carrier and Compound described in first aspect present invention or its crystal form, pharmaceutically acceptable salt, hydrate or solvate.
In another preferred example, the pharmaceutical composition is injection, wafer, tablet, pill, powder or granule.
In another preferred example, the pharmaceutical composition also contains other medicine, the other treatment Drug is cancer, angiocardiopathy, inflammation, infection, immunity disease, cell proliferation disorders, viral disease, metabolic disease Disease or the drug of organ transplant.
In the fourth aspect of the present invention, the compound or its crystal form, pharmacy described in first aspect present invention are provided The purposes of upper acceptable salt, prodrug, stereoisomer, isotopic variations, hydrate or solvate, they be used to prepare The pharmaceutical composition of protease inhibition.
In another preferred example, the pharmaceutical composition is for treating and preventing following disease:Cancer, cell proliferative Disease, inflammation, infection, immunity disease, organ transplant, viral disease, angiocardiopathy or metabolic disease.
In another preferred example, the cancer includes but is not limited to:Lung cancer, head and neck cancer, breast cancer, prostate cancer, Cancer of the esophagus, the carcinoma of the rectum, colon cancer, nasopharyngeal carcinoma, uterine cancer, cancer of pancreas, lymthoma, leukemia, osteosarcoma, melanoma, kidney, stomach Cancer, liver cancer, carcinoma of urinary bladder, thyroid cancer or colorectal cancer.
In another preferred example, the immunity disease or inflammation include but is not limited to:Rheumatoid arthritis, bone close Save inflammation, poker back, gout, asthma, bronchitis, rhinitis, chronic obstructive pulmonary disease, cystic fibrosis.
In another preferred example, the cell proliferation disorders refer to lung cancer, head and neck cancer, breast cancer, prostate cancer, food Road cancer, the carcinoma of the rectum, colon cancer, nasopharyngeal carcinoma, uterine cancer, cancer of pancreas, lymthoma, leukemia, osteosarcoma, melanoma, kidney, stomach Cancer, liver cancer, carcinoma of urinary bladder, thyroid cancer or colorectal cancer.
In another preferred example, the cancer is non-small cell lung cancer.
In the fifth aspect of the present invention, a kind of method inhibiting protein kinase (such as ALK kinases) or a kind of disease are provided Sick (such as cancer, cell proliferation disorders, inflammation, infection, immunity disease, organ transplant, viral disease, angiocardiopathy Or metabolic disease) therapy, it includes step:It is applied described in first aspect present invention to object in need for the treatment of Compound or its crystal form, pharmaceutically acceptable salt, hydrate or solvate, or described in application third aspect present invention Pharmaceutical composition.
It should be understood that within the scope of the present invention, above-mentioned each technical characteristic of the invention and have in below (eg embodiment) It can be combined with each other between each technical characteristic of body description, to form a new or preferred technical solution.As space is limited, exist This no longer tires out one by one states.
The invention also includes the compounds of isotope labelling, are equal to original chemical and are disclosed.This hair can be classified as The example of bright compound isotope includes hydrogen, carbon, nitrogen, oxygen, phosphorus, sulphur, fluorine and chlorine isotope, respectively such as2H,3H,13C,14C,15N,17O,18O,31P,32P,35S,18F and36Cl.Compound in the present invention or enantiomer, diastereomer, isomers or medicine Acceptable salt or solvate on, wherein containing the isotope of above compound or other other isotope atoms all at this Within the scope of invention.Certain compound isotopically labelleds in the present invention, such as3H and14The radioactive isotope of C also wherein, It is useful in the experiment of the Tissue distribution of drug and substrate.Tritium, i.e.,3H and carbon-14, i.e.,14C, their preparation and detection are compared It is easy, is the first choice in isotope.In addition, higher isotope substitution such as deuterium, i.e.,2H, since its good metabolic stability is at certain It is advantageous in a little therapies, such as in vivo therefore increase half-life period or reduction dosage can be paid the utmost attention in some cases. The compound of isotope labelling can use general method, non isotopic by being replaced with the isotope labeling reagent being easy to get Reagent can be prepared with the scheme in example.
Herein, unless otherwise instructed, " halogen " refers to F, Cl, Br and I.More preferably, halogen atom is selected from F, Cl and Br.
Herein, unless otherwise instructed, " C1-C6Alkyl " refers to the alkyl for the linear chain or branched chain for including 1-6 carbon atom, Such as methyl, ethyl, propyl, isopropyl, butyl, isobutyl group, tertiary butyl or similar group.
Herein, unless otherwise instructed, " deuterated " refers to one or more of compound or group hydrogen and is replaced by deuterium;Deuterium Generation can be a substitution, two substitutions, polysubstituted or full substitution.Term " one or more deuterated " and " one or many deuterated " It is used interchangeably.
Herein, unless otherwise instructed, " non-deuterated compound " refers to that ratio containing D-atom is not higher than the same position of natural deuterium The compound of cellulose content (0.015%).
In the present invention, pharmaceutically acceptable salt includes inorganic salts and organic salt.A kind of preferred salt is chemical combination of the present invention The salt that object is formed with acid.The acid for suitably forming salt includes but is not limited to:Hydrochloric acid, hydrobromic acid, hydrofluoric acid, sulfuric acid, nitric acid, phosphoric acid Equal inorganic acids;Formic acid, acetic acid, trifluoroacetic acid, propionic acid, oxalic acid, malonic acid, succinic acid, fumaric acid, maleic acid, lactic acid, apple Acid, tartaric acid, citric acid, picric acid, benzoic acid, methanesulfonic acid, ethanesulfonic acid, p-methyl benzenesulfonic acid, benzene sulfonic acid, naphthalene sulfonic acids etc. are organic Acid;And the amino acid such as proline, phenylalanine, aspartic acid, glutamic acid.Another kind of preferred salt be the compounds of this invention with The salt that alkali is formed, such as alkali metal salt (such as sodium salt or sylvite), alkali salt (such as magnesium salts or calcium salt), ammonium salt are (such as low Grade alkanol ammonium salt and other pharmaceutically acceptable amine salt), such as methylamine salt, ethylamine salt, propylamine salt, dimethyl amine salt, Trismethylamine salt, diethyl amine salt, triethyl amine salt, tert-butylamine salt, ethylenediamine salt, oxyethylamine salt, dihydroxy ethylamine salt, three hydroxyls Ethylamine salt, and the amine salt that is formed respectively by morpholine, piperazine, lysine.
Term " solvate " refers to the compounds of this invention and is coordinated the complex to form special ratios with solvent molecule." hydration Object " refers to the complex that the compounds of this invention carries out coordination formation with water.
Compared with prior art, beneficial effects of the present invention are:
(1) the compounds of this invention has excellent inhibition to protein kinase (kinase) (such as ALK kinases).
(2) by deuterate, this technology changes metabolism of the compound in organism, and compound is made to have better medicine generation Kinetic parameter characteristic.In such a case, it is possible to change dosage and form durative action preparation, improve applicability.
(3) use the hydrogen atom in deuterium substituted compound that can improve compound in animal body due to its deuterium isotope effect Interior drug concentration, to improve curative effect of medication.
(4) hydrogen atom in deuterium substituted compound is used, since certain metabolites are suppressed, the peace of compound may be improved Quan Xing.
Specific implementation mode
The preparation method of formula (I) structural compounds of the present invention is described more particularly below, but these specific methods are not to this Invention constitutes any restrictions.The compounds of this invention can also optionally will be describing or known in the art various in the present specification Synthetic method combines and is easily made, such combination can by those skilled in the art in the invention easily into Row.
The preparation method of the salt for the not deuterated diaminopyrimidine compounds and its physical compatibility that the present invention uses is Know.The preparation of corresponding deuterated diaminopyrimidine compounds can be raw material with corresponding deuterated initial compounds, with same Route synthesis.For example, formula (I) compound of the present invention can be prepared by the preparation method described in WO2012061299, difference It is to replace non-deuterated raw material with deuterated raw material in the reaction.
In general, in preparation flow, each reaction is usually in atent solvent, in room temperature to reflux temperature (such as 0 DEG C~100 DEG C, preferably 0 DEG C~80 DEG C) under carry out.Reaction time is usually -60 hours 0.1 hour, preferably 0.5-24 hours.
Following general preparation route can be used for synthesizing the compound of formula (I) structure of the present invention.The following institute of synthetic route Show:
The synthesis of piperazine substituted piperidine amine is as follows:
It is described in detail with reference to embodiment.
Embodiment 1
The chloro- N of 5- are prepared according to following synthetic route4[2- (solutions of dimethyl phosphoryl base) phenyl]-N2{ 2-d3- methoxyl groups -4- [4- (4- methylpiperazine-1-yls)-piperidin-1-yl] phenyl } pyrimidine -2,4- diamines (compound 9 in following synthetic routes):
It is prepared using following steps:
(1) prepare compound 2:
In 100mL single port bottles be added 30mL acetone, sequentially added under stirring the fluoro- 2- nitrophenols of 5- (2.0g, 12.7mmol), Anhydrous potassium carbonate (3.5g, 25.4mmol), deuterated iodomethane (2.4g, 16.5mmol), are warming up to 60 DEG C and protect Temperature stirring 2h.It is cooled to room temperature, rotary evaporation falls acetone, and water 20mL is added in residue, and ethyl acetate extracts (30mLx3), merges Organic layer, anhydrous sodium sulfate drying, filtering, filtrate are concentrated to give white solid 2.0g, yield 90%.
1H NMR(300MHz,CDCl3) (δ/ppm) 8.00-7.95 (m, 1H), 6.83-6.71 (m, 2H), LC-MS (APCI):M/z=175.2 (M+1)+, 95%.
(2) prepare compound 4:
N,N-Dimethylformamide (4mL) is added in 25mL single-necked flasks, the fluoro- 2-d3- first of 4- is sequentially added under stirring Oxygroup nitrobenzene (0.4g, 2.3mmol), 1- methyl -4- (piperidin-4-yl) piperazine hydrochloride (0.7g, 3.2mmol), anhydrous carbon Sour potassium (0.95g, 6.9mmol), reaction mixture are warming up to 80 DEG C, N2It is reacted overnight under atmosphere.It is cooled to room temperature, pours into ice water In (80mL), a large amount of yellow solids are precipitated, filter, and be dissolved in DCM (40mL), anhydrous sodium sulfate drying, filtering, filtrate concentration Obtain yellow solid 0.55g, yield 70.9%.
LC-MS(APCI):M/z=338.2 (M+1)+1H NMR(300MHz,CDCl3) (δ/ppm) 8.01 (d, J= 9.3Hz, 1H), 6.43 (dd, J=9.6Hz, J=2.7Hz, 1H), 6.31 (d, J=2.4Hz, 1H), 3.98-3.94 (m, 2H), 3.03-2.94(m,2H),2.65-2.62(m,4H),2.54-2.46(m,5H),2.32(s,3H),2.01-1.96(m,2H), 1.65-1.60(m,2H)。
(3) prepare compound 5:
Ethyl alcohol 6mL and water 2mL is added in 25mL single port bottles, 1- (1- (3-d3- methoxyl group -4- nitre is sequentially added under stirring Base phenyl) piperidin-4-yl) -4- methyl piperazines (0.2g, 0.59mmol), reduced iron powder (0.20g, 3.55mmol), ammonium chloride (16mg, 0.30mmol), reaction mixture N285 DEG C are warming up under atmosphere, and insulated and stirred reacts 1h.It is cooled to room temperature, is filtered solid Body substance, filtrate concentrate, and saturated sodium bicarbonate (5mL) is added in residue, and dichloromethane extracts (15mLx2), merges organic Phase, anhydrous sodium sulfate drying, filtering are concentrated to give off-white powder 0.15g, and yield 79.6% is directly thrown in next step.
(4) prepare compound 8:
In 25mL single port bottles be added DMF (3mL), sequentially added under stirring 2,5,6- trichloropyrimidines (0.72g, 3.9mmol), 2- (dimethyl phosphino-) aniline (0.5g, 3mmol), Anhydrous potassium carbonate (0.62g, 4.5mmol), are warming up to 60 DEG C and keep the temperature Stir 4h.It is cooled to room temperature, sequentially adds ethyl acetate (30mL), water (30mL), concussion layering, aqueous layer with ethyl acetate extraction (30mLx2) merges organic phase, washes (60mLx2), and the drying of organic layer anhydrous sodium sulfate is filtered, and concentration, residue crosses silica gel Column obtains faint yellow solid 0.8g, yield 84.6%.
LC-MS(APCI):M/z=175.2 (M+1)+1H NMR(CDCl3,300MHz)(δ/ppm)8.00-7.95(m, 1H),6.83-6.71(m,2H)。
(5) prepare compound 9:
Glycol monoethyl ether 2mL is added in 25mL single port bottles, 2,5-, bis- chloro- N- (2- (dimethyl is sequentially added under stirring Phosphoroso-) phenyl) pyrimidine -4- amine (60mg, 0.19mmol), 1- (1- (3-d3- methoxyl group -4- aminocarbonyl phenyls) piperidin-4-yl) - 4- methyl piperazines (60mg, 0.2mmol), concentrated hydrochloric acid (two drops), reaction mixture N2100 DEG C, and insulated and stirred are warming up under atmosphere Reaction is overnight.It is cooled to room temperature, saturated sodium bicarbonate water liquid (10mL) is added, dichloromethane extracts (15mLx3), merges organic Phase, anhydrous sodium sulfate drying, is filtered, concentration, is crossed silicagel column and is obtained white solid 60mg, yield 50.1%;
LC-MS(APCI):M/z=587.2 (M+1)+1H NMR(300MHz,DMSO-d6)(δ/ppm)11.18(s,1H), 8.51-8.47(m,1H),0.08-8.07(m,2H),7.57-7.50(m,1H),7.40-7.32(m,2H),7.12-7.07(m, 1H), 6.62 (d, J=2.4Hz, 1H), 6.47 (dd, J=9.0Hz, 2.4Hz, 1H), 3.77-3.72 (m, 2H), 2.82-2.61 (m,11H),2.48-2.34(m,3H),1.91-1.85(m,2H),1.79(s,3H),1.74(s,3H),1.59-1.52(m, 2H)。
Embodiment 2
The chloro- N of 5- are prepared according to following synthetic route4[2- (solutions of dimethyl phosphoryl base) phenyl]-N2{ 2- methoxyl group -4- [4- (4-d3- methylpiperazine-1-yls)-piperidin-1-yl] phenyl } pyrimidine -2,4- diamines (compound 15 in following synthetic routes):
Include the following steps:
(1) prepare compound 10:
N,N-Dimethylformamide (10mL) is added in 100mL single-necked flasks, the fluoro- 2- methoxyl groups of 4- are sequentially added under stirring Nitrobenzene (2g, 11.8mmol), piperidin-4-one hydrochloride (2.23g, 16.5mmol), Anhydrous potassium carbonate (4.88g, 35.4mmol), reaction mixture is warming up to 80 DEG C, N2It is reacted overnight under atmosphere.It is cooled to room temperature, is poured into ice water (80mL), is analysed Go out a large amount of yellow solids, filter, and be dissolved in DCM (100mL), anhydrous sodium sulfate drying is filtered, and filtrate is concentrated to give yellow solid 2.2g, yield 74.6%.
LC-MS(APCI):M/z=251.2 (M+1)+1H NMR (300MHz, DMSO-d6) (δ/ppm) 7.93 (d, J= 9.6Hz, 1H), 6.62 (dd, J=9.6Hz, 2.4Hz, 1H), 6.53 (d, J=2.4Hz, 1H), 3.94 (s, 3H), 3.84 (t, J =6.3Hz, 4H), 2.50 (t, J=6.3Hz, 4H).
(2) prepare compound 11:
Toluene 20mL is added in 100mL single-necked flasks, 1- (3- methoxyl group -4- nitrobenzophenones) piperazine is sequentially added under stirring Pyridine -4- ketone (0.53g, 2.1mmol), triethylamine (0.8mL), N-Boc piperazines (0.85g, 4.5mmol), N2It is stirred to react under atmosphere 30min is added at one time acetic acid sodium borohydride (0.4g, 1.92mmol), stirs 30min, adds acetic acid boron hydrogen again in three times Change sodium 1.2g, the reaction was complete.Saturated sodium bicarbonate water liquid (30mL) is added, separates organic layer, the extraction of water layer ethyl acetate (30mLx2) merges organic phase, and anhydrous sodium sulfate drying is filtered, and concentration, residue crosses silicagel column and obtains faint yellow solid 0.58g, Yield 65.7%.
LC-MS(APCI):M/z=421.2 (M+1)+1H NMR(300MHz,CDCl3) (δ/ppm) 8.01 (d, J= 9.3Hz, 1H), 6.43 (dd, J=9.6Hz, 2.7Hz, 1H), 6.31 (d, J=2.4Hz, 1H), 3.94 (s, 3H), 3.03-2.94 (m,2H),2.65-2.62(m,4H),2.54-2.46(m,5H),2.32(s,3H),2.01-1.96(m,2H),1.65-1.60 (m, 2H), 1.51 (s, 9H).
(3) prepare compound 12:
Dichloromethane 20mL is added in 100mL single-necked flasks, 4- (1- (3- methoxyl group -4- nitros are sequentially added under stirring Phenyl) piperidin-4-yl) piperazinyl -1- tert-butyl esters (0.58g, 1.4mmol), trifluoracetic acid (2mL), N2Stirring at normal temperature is anti-under atmosphere It answers 1h, reaction solution to be concentrated to dryness, saturated sodium bicarbonate water liquid (10mL) is added, mixture dichloromethane extracts (20mLx3), nothing Aqueous sodium persulfate is dried, and filtering is concentrated to give yellow solid 0.45g, yield 100%, LC-MS (APCI):M/z=321.2 (M+1)+
(4) prepare compound 13:
Acetonitrile 5mL is added in 25mL single-necked flasks, 4- (1- (3- methoxyl group -4- nitrobenzophenones) are sequentially added under stirring Piperidin-4-yl) piperazine (0.32g, 1mmol), be added triethylamine (0.12g, 1.2mmol), ice-water bath cooling, be slowly added dropwise into deuterium For iodomethane (0.16g, 1.1mmol), be stirred to react 30min under ice-water bath, be concentrated to dryness, residue cross silicagel column obtain it is yellow Color solid 0.15g, yield 44.5%.
LC-MS(APCI):M/z=338.2 (M+1)+1H NMR(300MHz,CDCl3) (δ/ppm) 8.01 (d, J= 9.3Hz, 1H), 6.43 (dd, J=9.6Hz, 2.7Hz, 1H), 6.31 (d, J=2.4Hz, 1H), 3.98-3.94 (m, 2H), 3.92 (s,3H),3.03-2.94(m,2H),2.65-2.62(m,4H),2.54-2.46(m,5H),2.01-1.96(m,2H),1.65- 1.60(m,2H)。
(5) prepare compound 14, preparation method is consistent with the preparation method of compound 5, the difference is that with 1- (1- (3- methoxyl group -4- nitrobenzophenones) piperidin-4-yl) -4-d3- methyl piperazines replacement 1- (1- (3-d3- methoxyl group -4- nitrobenzenes Base) piperidin-4-yl) -4- methyl piperazines.
(6) the chloro- N of 5- are prepared4[2- (dimethyl oxygen phosphino-) phenyl]-N2{ 2- methoxyl groups -4- [4- (4-d3- methyl piperazines Piperazine -1- bases)-piperidin-1-yl] phenyl } pyrimidine -2,4- diamines (compound 14), the preparation method of preparation method and compound 9 Unanimously, the difference is that substituting 1- using 1- (1- (3- methoxyl group -4- aminocarbonyl phenyls) piperidin-4-yl) -4-d3- methyl piperazines (1- (3-d3- methoxyl group -4- aminocarbonyl phenyls) piperidin-4-yl) -4- methyl piperazines.
LC-MS(APCI):M/z=587.2 (M+1)+1H NMR(300MHz,DMSO-d6)(δ/ppm)11.18(s,1H), 8.51-8.47(m,1H),0.08-8.07(m,2H),7.57-7.50(m,1H),7.40-7.32(m,2H),7.12-7.07(m, 1H), 6.62 (d, J=2.4Hz, 1H), 6.47 (dd, J=9.0Hz, 2.4Hz, 1H), 3.76-3.71 (m, 6H), 2.82-2.61 (m,11H),2.48-2.34(m,3H),1.91-1.85(m,2H),1.79(s,3H),1.74(s,3H),1.59-1.52(m, 2H)。
Embodiment 3
The chloro- N of 5- are prepared according to following synthetic route4[2- (solutions of dimethyl phosphoryl base) phenyl]-N2{ 2- methoxyl group -4- [4- (4- methylpiperazine-1-yls) -4-d- piperidin-1-yls] phenyl } pyrimidine -2,4- diamines (compound 20 in following synthetic routes):
Include the following steps:
(1) prepare compound 16:
Deuterated methanol 10mL is added in 50mL single-necked flasks, ice-water bath is added with stirring 1- (3- methoxyl group -4- nitrobenzenes Base) piperidin-4-one (0.25g, 1mmol), deuterated sodium borohydride (42mg, 1mmol), ice-water bath N are slowly added to after complete dissolved clarification2 It is stirred to react 5min under atmosphere, heavy water (2mL) is added, reaction, and stirring at normal temperature 30min is quenched, sequentially add water (30mL) and acetic acid Ethyl ester (30mL) separates organic layer, and water layer ethyl acetate extracts (30mLx2), and concentration, residue is again dissolved in ethyl acetate (50mL), saturated common salt water washing (20mLx1), the drying of organic phase anhydrous sodium sulfate, filtering are concentrated to give yellow solid 0.25g, Yield 96%.
LC-MS(ESI):M/z=254.2 (M+1)+1H NMR(300MHz,DMSO-d6) (δ/ppm) 7.88 (d, J= 9.3Hz, 1H), 6.58 (dd, J=9.3Hz, 2.4Hz, 1H), 6.49 (d, J=2.4Hz, 1H), 4.75 (s, 1H), 3.90 (s, 3H),3.83-3.77(m,2H),3.23-3.14(s,2H),1.84-1.76(m,2H),1.45-1.37(m,2H)。
(2) prepare compound 17:
Dichloromethane (15mL) is added in 50mL single-necked flasks, 1- (3- methyl -4- nitrobenzophenones)-is added under ice-water bath 4-d- piperidines -4- alcohol (0.25g, 1mmol) is added with stirring triethylamine (0.18g, 1.8mmol), is slowly added dropwise into sulfonyloxy methyl Chlorine (0.17g, 1.5mmol), room temperature N2It is stirred to react 1h under atmosphere.Water (20mL) is added, concussion separates organic layer, water layer dichloromethane Alkane extracts (20mLx2), merges organic layer, uses 0.5M HCl/waters liquid (20mLx1), saturated sodium bicarbonate water liquid successively (15mLx1), saturated salt solution (15mLx1), anhydrous sodium sulfate drying, filtering are concentrated to dryness to obtain yellow solid 0.3g, yield 90.9%, it is directly used in next step.
(3) prepare compound 18:
DMF (3mL) is added in 25mL single-necked flasks, 1- (3- methyl -4- nitrobenzophenones) -4-d- is sequentially added under stirring Piperidines -4- methanesulfonate esters (0.3g, 0.9mmol), 1- methyl piperazines (0.36g, 3.6mmol), Anhydrous potassium carbonate (0.62g, 4.5mmol), mixture is warming up to 100 DEG C, N2Insulated and stirred reaction is stayed overnight under atmosphere.It is cooled to room temperature, water (30mL) and second is added Acetoacetic ester (30mL) separates organic layer, and water layer ethyl acetate extracts (20mLx2), merges organic phase, washes (40mLx3), organic Layer anhydrous sodium sulfate drying, is filtered, concentration, residue crosses silicagel column and obtains yellow solid 100mg, yield 33.1%.
LC-MS(APCI):M/z=339.2 (M+1)+1H NMR(300MHz,CDCl3) (δ/ppm) 8.01 (d, J= 9.3Hz, 1H), 6.43 (dd, J=9.6Hz, 2.7Hz, 1H), 6.31 (d, J=2.4Hz, 1H), 3.98-3.94 (m, 2H), 3.03-2.94(m,2H),2.65-2.62(m,4H),2.54-2.46(m,5H),2.32(s,3H),2.01-1.96(m,2H), 1.65-1.60(m,2H)。
(4) 1- [1- (3- methoxyl group -4- aminocarbonyl phenyls) -4-d- piperidin-4-yls] -4- methyl piperazine (compounds are prepared 19), preparation method is consistent with the preparation method of compound 5, the difference is that with 1- [1- (3- methoxyl group -4- nitrobenzenes Base) -4-d- piperidin-4-yls] -4- methyl piperazines replacement 1- [1- (3-d3- methoxyl group -4- nitrobenzophenones) piperidin-4-yl] -4- first Base piperazine.
(5) the chloro- N of 5- are prepared4[2- (solutions of dimethyl phosphoryl base) phenyl]-N2{ 2- methoxyl groups -4- [4- (4- methyl piperazines -1- Base) -4-d- piperidin-1-yls] phenyl } pyrimidine -2,4- diamines (compound 20), the preparation method of preparation method and compound 9 Unanimously, the difference is that being substituted using 1- [1- (3- methoxyl group -4- aminocarbonyl phenyls) -4-d- piperidin-4-yls] -4- methyl piperazines 1- [1- (3-d3- methoxyl group -4- aminocarbonyl phenyls) piperidin-4-yl] -4- methyl piperazines.
LC-MS(APCI):M/z=587.2 (M+1)+1H NMR(300MHz,DMSO-d6)δ(ppm):11.18(s,1H), 8.51-8.47(m,1H),0.08-8.07(m,2H),7.57-7.50(m,1H),7.40-7.32(m,2H),7.12-7.07(m, 1H), 6.62 (d, J=2.4Hz, 1H), 6.47 (dd, J=9.0Hz, 2.4Hz, 1H), 3.76-3.71 (m, 6H), 2.82-2.61 (m,10H),2.48-2.34(m,3H),1.91-1.85(m,2H),1.79(s,3H),1.74(s,3H),1.59-1.52(m, 2H)。
Embodiment 4
Prepare the chloro- N of 5-4[2- (solutions of dimethyl phosphoryl base) phenyl]-N2{ 2- methoxyl groups -4- [4- (4- methyl -3,3,5,5- D4- piperazine -1- bases)-piperidin-1-yl] phenyl } pyrimidine -2,4- diamines (compound 21), structural formula is as follows:
With similar method described in embodiment 2, difference is that 4- methyl -3,3,5,5-d4- piperazines is used to replace N- methyl Piperazine, to which target compound be made.
Embodiment 5
Prepare the chloro- N of 5-4[2- (solutions of dimethyl phosphoryl base) phenyl]-N22- methoxyl groups -4- [4- (4- methyl 2,2,3,3,5, 5,6,6-d8- piperazine -1- bases) piperidin-1-yl] phenyl } pyrimidine -2,4- diamines (compound 22), structural formula is as follows:
With similar method described in embodiment 2, difference is to use 4- methyl -2,2,3,3,5,5,6,6-d8- piperazine generations For N methyl piperazine, to which target compound be made.
LC-MS(APCI):M/z=592.4 (M+1)+1H NMR(400MHz,CD3OD) (δ/ppm) 8.35 (dd, J= 8.4Hz, 4.4Hz, 1H), 8.04 (s, 1H), 7.69 (d, J=8.8Hz, 1H), 7.65-7.59 (m, 1H), 7.53 (t, J=8Hz, 1H), 7.29-7.25 (m, 1H), 6.67 (d, J=2.4Hz, 1H), 6.46 (dd, J=8.8Hz, 2.4Hz, 1H), 3.86 (s, 3H), 3.71 (d, J=12.8Hz, 2H), 2.76-2.70 (m, 2H), 2.61-2.56 (m, 4H), 2.04 (d, J=12.8Hz, 2H),1.87(s,3H),1.83(s,3H),1.76-1.65(m,2H)。
Embodiment 6
The chloro- N of 5- are prepared according to following synthetic route4[2- (solutions of dimethyl phosphoryl base) phenyl]-N2{ 2-d3- methoxyl groups -4- [4- (4-d3- methylpiperazine-1-yls) piperidin-1-yl] phenyl } pyrimidine -2,4- diamines (compounds in following synthetic routes 25):
(1) prepare compound 23:
Acetonitrile 5mL is added in 25mL single-necked flasks, 4- (1- (3- methoxyl group -4- nitrobenzophenones) are sequentially added under stirring Piperidin-4-yl) piperazine (0.32g, 1mmol), be added triethylamine (0.12g, 1.2mmol), ice-water bath cooling, be slowly added dropwise into deuterium For iodomethane (0.16g, 1.1mmol), be stirred to react 30min under ice-water bath, be concentrated to dryness, residue cross silicagel column obtain it is yellow Color solid 0.15g, yield 44.5%.
LC-MS(APCI):M/z=340.2 (M+1)+
(2) prepare compound 24, preparation method is consistent with the preparation method of compound 5, the difference is that with 1- [1- (3-d3- methoxyl group -4- nitrobenzophenones) piperidin-4-yl] -4-d3- methyl piperazines replacement 1- [1- (3-d3- methoxyl group -4- nitros Phenyl) piperidin-4-yl] -4- methyl piperazines.
(3) prepare compound 25, preparation method is consistent with the preparation method of compound 9, the difference is that using 1- [1- (3-d3- methoxyl group -4- aminocarbonyl phenyls) piperidin-4-yl] -4-d3- methyl piperazines substitute 1- [1- (3-d3- methoxyl group -4- amine Base phenyl) piperidin-4-yl] -4- methyl piperazines.
LC-MS(APCI):M/z=587.2 (M+1)+1H NMR(300MHz,DMSO-d6)(δ/ppm)11.18(s,1H), 8.51-8.47(m,1H),0.08-8.07(m,2H),7.57-7.50(m,1H),7.40-7.32(m,2H),7.12-7.07(m, 1H), 6.62 (d, J=2.4Hz, 1H), 6.47 (dd, J=9.0Hz, 2.4Hz, 1H), 3.76-3.71 (m, 6H), 2.82-2.61 (m,11H),2.48-2.34(m,3H),1.91-1.85(m,2H),1.79(s,3H),1.74(s,3H),1.59-1.52(m, 2H)。
Embodiment 7
The chloro- N of 5- are prepared according to following synthetic route4[2- (solutions of dimethyl phosphoryl base) phenyl]-N2{ 2- methoxyl group -4- [4- (4-d3- methyl -2,2,3,3,5,5,6,6-d8- piperazine -1- bases) piperidin-1-yl] phenyl } pyrimidine -2,4- diamines (following synthesis Compound 29 in route):
(1) prepare compound 27:
By compound 26 (214mg, 652 μm of ol), deuterated formaldehyde heavy aqueous solution (313mg, 1.95mmol, 20%/ D2) and CH O3COOD (1 drop) is stirred at room temperature 10 minutes, and deuterated sodium cyanoborohydride (129mg, 1.95mmol) is added, after After continuous stirring 30 minutes, triethylamine is added and neutralizes, yellow solid 175mg, yield are obtained by column chromatography separating purification after concentration It is 77.8%.
LC-MS(APCI):M/z=346.4 (M+1)+
(2) prepare compound 28, preparation method is consistent with the preparation method of compound 5, the difference is that with 1- [1- (3- methoxyl group -4- nitrobenzophenones) piperidin-4-yl] -4-d3- methyl -2,2,3,3,5,5,6,6-d8- piperazines replacement 1- [1- (3- D3- methoxyl group -4- nitrobenzophenones) piperidin-4-yl] -4- methyl piperazines.
(3) prepare compound 29, preparation method is consistent with the preparation method of compound 9, the difference is that using 1- [1- (methoxyl group -4- aminocarbonyl phenyls) piperidin-4-yl] -4-d3- methyl -2,2,3,3,5,5,6,6-d8- piperazines substitute 1- [1- (3-d3- methoxyl group -4- aminocarbonyl phenyls) piperidin-4-yl] -4- methyl piperazines.
LC-MS(APCI):M/z=595.4 (M+1)+1H NMR(300MHz,CDCl3)(δ/ppm)10.80(s,1H), 8.63 (dd, J=4.8Hz, 2.4Hz, 1H), 8.09-8.07 (m, 2H), 7.50 (t, J=4.5Hz, 1H), 7.30-7.25 (m, 2H), 7.14-7.10 (m, 1H), 6.55 (d, J=1.5Hz, 1H), 6.49 (dd, J=5.4Hz, 1.5Hz, 1H), 3.87 (s, 3H), 3.66 (d, J=7.5Hz, 2H), 2.73-2.68 (m, 2H), 2.40-2.36 (m, 1H), 1.95 (d, J=7.5Hz, 2H), 1.85(s,3H),1.82(s,3H),1.76-1.68(m,2H)。
Embodiment 8
The chloro- N of 5- are prepared according to following synthetic route4[2- (solutions of dimethyl phosphoryl base) phenyl]-N2{ 2-d3- methoxyl groups -4- [4- (4- methyl -2,2,3,3,5,5,6,6-d8- piperazine -1- bases) piperidin-1-yl] phenyl } pyrimidine -2,4- diamines (following synthesis Compound 32 in route):
(1) prepare compound 30, preparation method is consistent with the preparation method of compound 10, the difference is that using d3- The fluoro- 2- Nitroanisoles of 5- substitute the fluoro- 2- Nitroanisoles of 5-.
LC-MS(APCI):M/z=254.5 (M+1)+
(2) prepare compound 31:
By tetraisopropyl titanate (Ti (Oi-Pr)4, 5mL) and it is added to compound 30 (900mg, 3.6mmol) and 2,2,3,3, It in the solution of 5,5,6,6-d8- piperazines (474mg, 5.03mmol), is stirred overnight at room temperature, 10mL ethyl alcohol is added, continuously adds Itrile group sodium borohydride (678mg, 10.79mmol) after this mixed liquor is stirred at room temperature 3 hours, is poured into dissolved with 5g celite (celite) water (10mL), continues stirring 30 minutes, and obtaining yellow solid by column chromatography separating purification after removal solvent produces Object 300mg, yield 25.4%.
LC-MS(APCI):M/z=332.5 (M+1)+
(3) prepare compound 32, preparation method is consistent with the preparation method of compound 29, the difference is that using chemical combination 31 alternative compounds 26 of object, formaldehyde substitute deuterated formaldehyde, and itrile group sodium borohydride substitutes deuterated sodium borohydride.Finally obtain target production Object is white solid, total 40mg, yield 53.0%.
LC-MS(APCI):M/z=595.5 (M+1)+1H NMR(300MHz,CDCl3)(δ/ppm)10.80(s,1H), 8.62 (dd, J=8.1Hz, 4.5Hz, 1H), 8.11-8.08 (m, 2H), 7.50 (t, J=7.8Hz, 1H), 7.32-7.25 (m, 2H), 7.16-7.11 (m, 1H), 6.54 (d, J=1.6Hz, 1H), 6.48 (dd, J=8.4Hz, 2.4Hz, 1H), 3.66 (d, J= 12Hz, 2H), 2.74-2.67 (m, 2H), 2.61-2.55 (m, 1H), 2.48 (s, 3H), 2.02 (d, J=12.3Hz, 2H), 1.84 (s,3H),1.81(s,3H),1.79-1.73(m,2H)。
Embodiment 9
Prepare the chloro- N of 5-4[2- (solutions of dimethyl phosphoryl base) phenyl]-N22-d3- methoxyl groups -4- [4- (methyl -2 4-d2-, 2,3,3,5,5,6,6-d8- piperazine -1- bases) piperidin-1-yl] phenyl } pyrimidine -2,4- diamines (compound 33), structural formula is as follows It is shown:
It is similar to 7 the method for embodiment, the difference is that with 31 alternative compounds 26 of compound, itrile group sodium borohydride Substitute deuterated sodium borohydride.It is yellow solid, total 70mg, yield 27.2% to finally obtain target product.
LC-MS(APCI):M/z=597.4 (M+1)+1H NMR(300MHz,CDCl3)(δ/ppm)10.82(s,1H), 8.62 (dd, J=8.4Hz, 4.5Hz, 1H), 8.13-8.09 (m, 2H), 7.50 (t, J=7.5Hz, 1H), 7.33-7.26 (m, 2H), 7.16-7.10 (m, 1H), 6.54 (d, J=2.1Hz, 1H), 6.48 (dd, J=9.0Hz, 2.4Hz, 1H), 3.66 (d, J= 12.6Hz,2H),2.76-2.59(m,3H),2.56(s,1H),2.08-2.00(m,2H),1.86(s,3H),1.81(s,3H), 1.79-1.72(m,2H)。
Embodiment 10
Prepare the chloro- N of 5-4[2- (solutions of dimethyl phosphoryl base) phenyl]-N22-d3- methoxyl groups -4- [4- (methyl -2 4-d3-, 2,3,3,5,5,6,6-d8- piperazine -1- bases) piperidin-1-yl] phenyl } pyrimidine -2,4- diamines (compound 34), structural formula is as follows It is shown:
It is similar to 7 the method for embodiment, the difference is that with 31 alternative compounds 26 of compound.Finally obtain target Product is white solid, total 110mg, yield 36.7%.
LC-MS(APCI):M/z=598.4 (M+1)+1H NMR(300MHz,CDCl3) (δ/ppm) 8.35 (dd, J= 8.4Hz, 4.4Hz, 1H), 8.04 (s, 1H), 7.68 (d, J=8.4Hz, 1H), 7.65-7.59 (m, 1H), 7.52 (t, J=8Hz, 1H), 7.29-7.25 (m, 1H), 6.67 (d, J=6.8Hz, 1H), 6.46 (dd, J=8.8Hz, 2.4Hz, 1H), 3.71 (d, J= 12.4Hz, 2H), 2.76-2.70 (m, 2H), 2.64-2.58 (m, 1H), 2.04 (d, J=12.4Hz, 2H), 1.87 (s, 3H), 1.83(s,3H),1.76-1.66(m,2H)。
Embodiment 11
The biological assessment of compound
The compound of the present invention is evaluated in multiple tests to determine their biological activity.For example, can survey Examination the compounds of this invention inhibits the ability of a variety of concern of albumen kinases.The compound of some tests shows by force ALK kinases The inhibitory activity of effect.
(1) kinase inhibitory activity is evaluated
Compound is prepared:Test-compound is dissolved in DMSO and is made into 20mM mother liquors.Compound is diluted in DMSO using preceding At 0.1mM (dilution of 100 times of final concentrations), and 3 times of gradient dilutions are done, 11 concentration.When dosing 4 times are diluted to buffer solution The dilution of final concentration.
Kinase assay:After preparing buffer solution, enzyme is mixed with the various concentration compound that beforehand dilution is prepared, is placed at room temperature for 30 minutes, each concentration duplicate hole.Corresponding substrate and ATP, room temperature reaction 60 minutes (being provided with yin and yang attribute control) is added.Instead Addition antibody test should be finished, Evnvision is detected after sixty minutes for incubation at room temperature, gathered data.It is carried out according to XLfit5 softwares Data analysis and quasi- figure.And Buddhist nun's product as a contrast are replaced using gram azoles.
IC50=[(ABS tests-ABS starts)/(ABS controls-ABS starts)] x100
The results are shown in Table 1 for kinase inhibitory activity in embodiment.
1 Examples 1 to 10 of table replaces the kinase inhibitory activity contrast table of Buddhist nun with reference substance gram azoles
Embodiment is numbered ALK WT IC50(nM) ALK L1196M IC50(nM)
Embodiment 1 <20 <20
Embodiment 2 <20 <20
Embodiment 3 <20 <20
Embodiment 4 <20 <20
Embodiment 5 <20 <20
Embodiment 6 <20 <20
Embodiment 7 <20 <20
Embodiment 8 <20 <20
Embodiment 9 <20 <20
Embodiment 10 <20 <20
Reference substance gram azoles replaces Buddhist nun <20 >75
As shown in table 1, compared with ALK inhibitor gram azoles compare for Buddhist nun, the compounds of this invention to ALK L1196M be mutated Body surface reveals excellent inhibitory activity (IC50Less than 20), illustrate the compounds of this invention can pair between modification lymphom kinase (ALK) With very strong rejection ability.
(2) cytotoxicity experiment
Inhibiting effect to the compound on tumor cell of Examples 1 to 10 is had detected using tetrazolium salts (MTS) method, and with Gram azoles is reference substance for Buddhist nun.Experimental result is as shown in table 2.
2 Examples 1 to 10 of table replaces the cytotoxicity experiment contrast table of Buddhist nun with reference substance gram azoles
Embodiment is numbered ALK WT IC50(nM) ALK L1196M IC50(nM)
Embodiment 1 <20 <50
Embodiment 2 <20 <50
Embodiment 3 <20 <50
Embodiment 4 <20 <50
Embodiment 5 <20 <20
Embodiment 6 <20 <20
Embodiment 7 <20 <20
Embodiment 8 <20 <20
Embodiment 9 <20 <20
Embodiment 10 <20 <20
Reference substance gram azoles replaces Buddhist nun >70 >600
As shown in table 2, compared with ALK inhibitor gram azoles compare for Buddhist nun, the compounds of this invention all show inhibit expression The excellent antitumor activity of ALK mutant L1196M growth of cancer cells.
(3) metabolic stability is evaluated
Microsomal assay:People's hepatomicrosome:0.5mg/mL, Xenotech;Rat liver microsomes:0.5mg/mL, Xenotech;Coenzyme (NADPH/NADH):1mM, Sigma Life Science;Magnesium chloride:5mM, 100mM phosphate buffer (pH 7.4).
The preparation of storing solution:Precision weighs a certain amount of embodiment compound, and DMSO is used in combination to be dissolved to 5mM respectively.
The preparation of phosphate buffer (100mM, pH7.4):Take the 0.5M potassium dihydrogen phosphates 150mL for preparing in advance and The 0.5M dipotassium hydrogen phosphate solutions of 700mL mix, then adjust mixed liquor pH value to 7.4 with 0.5M dipotassium hydrogen phosphate solutions, use It is preceding to dilute 5 times with ultra-pure water, magnesium chloride is added, phosphate buffer (100mM) is obtained, wherein potassium phosphate containing 100mM, 3.3mM Magnesium chloride, pH 7.4.
It prepares NADPH regenerative systems solution and (contains 6.5mM NADP, 16.5mM G-6-P, 3U/mL G-6-P D, 3.3mM Magnesium chloride), using it is preposition in it is wet on ice.
Prepare terminate liquid:Acetonitrile containing 50ng/mL Propranolol Hydrochlorides and 200ng/mL orinases (internal standard) is molten Liquid.It takes in 25057.5 μ L phosphate buffers (pH7.4) to 50mL centrifuge tubes, is separately added into 812.5 μ L people's hepatomicrosomes, mix It is even, obtain the hepatomicrosome dilution of a concentration of 0.625mg/mL of albumen.Take 25057.5 μ L phosphate buffers (pH7.4) extremely In 50mL centrifuge tubes, 812.5 μ L SD rat liver microsomes are separately added into, mixing obtains the liver of a concentration of 0.625mg/mL of albumen Microsome dilution.
The incubation of sample:The storing solution of respective compound is diluted to 0.25mM respectively with the aqueous solution containing 70% acetonitrile, It is spare as working solution.It takes people's hepatomicrosome of 398 μ L or rat liver microsomes dilution that 96 holes are added respectively to be incubated in plate (N=2), it is separately added into the working solution of 2 μ L 0.25mM, mixing.
The measurement of metabolic stability:The terminate liquid of 300 μ L precoolings is added in every hole of 96 hole deep-well plates, is placed in ice On, as termination plate.96 holes are incubated plate and NADPH regenerative systems are placed in 37 DEG C of water baths, 100 revs/min of concussions are incubated in advance 5min.80 μ L Incubating Solutions addition termination plate is taken out per hole from plate is incubated, mixing supplements 20 μ LNADPH regenerative system solution, as 0min samples.Again to the NADPH regenerative system solution for being incubated plate 80 μ L of addition per hole, start reaction, starts timing.Corresponding chemical combination The reaction density of object is 1 μM, a concentration of 0.5mg/mL of albumen.When reaction 10,30,90min, 100 μ L reaction solutions are respectively taken, It is added in termination plate, vortex 3min terminates reaction.Termination plate is centrifuged into 10min under the conditions of 5000 × g, 4 DEG C.It takes on 100 μ L For clear liquid to being previously added in 96 orifice plates of 100 μ L distilled water, mixing carries out sample analysis using LC-MS/MS.
Data analysis:By LC-MS/MS system detectios respective compound and interior target peak area, calculate compound with it is interior Mark peak area ratio.Slope is measured with time mapping by the natural logrithm of the percentage of compound surplus, and according to following Formula calculates t1/2And CLint, wherein V/M is i.e. equal to 1/ albumen concentration.
To the compounds of this invention and its not deuterated compound tests compare simultaneously, and it is micro- with rat liver in people to evaluate it The metabolic stability of plastochondria.The half-life period of index as metabolic stability and liver clearance rate (Clint) are as shown in table 3. Using without deuterated compound AP26113 samples as a contrast in table 3;The AP26113 is the third generation for being the prior art ALK inhibitor is tried for treating the metastatic ALK positive non-small cell lung cancers for replacing Buddhist nun's tolerance to gram azoles in I/II phase clinic In testing, to the Patients with Non-small-cell Lung of the ALK positives, including brain metastes patient, AP26113 has duration antitumor activity.
As shown in table 3, by being compareed with without deuterated compound AP26113, the compounds of this invention can significantly improve Metabolic stability, and then be more suitable for preparing for treating the metastatic ALK positive non-small cell lung cancers for replacing Buddhist nun to be resistant to gram azoles Drug.
The metabolic stability contrast table of 3 Examples 1 to 10 of table and AP26113 control samples
(4) Pharmacokinetic Evaluation in rat
6 male Sprague-Dawley rats, 7-8 week old, weight about 210g are divided into 2 groups, every group 3, through vein or The compound (through vein 3mg/kg, taking orally 10mg/kg) of oral single dosage, compares its pharmacokinetic difference.
Rat is raised using standard feed, gives water.Experiment is fasted for first 16 hours.Drug is sub- with PEG400 and diformazan Sulfone dissolves.Eye socket is taken a blood sample, and time point of blood sampling is 0.083 hour after administration, 0.25 hour, 0.5 hour, 1 hour, 2 hours, 4 Hour, 6 hours, 8 hours, 12 hours and 24 hours.
Rat sucks of short duration anesthesia after ether, and eye socket acquires 300 μ L sample of blood in test tube.There are 30 μ L1% heparinates in test tube Solution.Before use, test tube is stayed overnight in 60 DEG C of drying.After being completed with the latter time point blood specimen collection, rat etherization After put to death.
After blood specimen collection, test tube is leniently overturned immediately at least 5 times, be positioned on ice after ensureing mixing fully.Blood sample is 4 DEG C 5000rpm is centrifuged 5 minutes, and blood plasma is detached with red blood cell.100 μ L blood plasma are sucked out to clean plastic centrifuge tube with pipettor In, show title and the time point of compound.Blood plasma is stored in -80 DEG C before being analyzed.It is measured in blood plasma with LC-MS/MS The concentration of the compounds of this invention.Pharmacokinetic parameter is based on every animal blood concentration in different time points into calculating.
Experimental result is as shown in table 4 below, relative to control compound AP26113, the compound 15 of embodiment 2 in the present invention Oral availability (F) it is suitable with control compound AP26113, but its Increased Plasma Half-life, metabolic stability are obviously improved;Implement The Oral availability of the compound 9 of example 1 increases substantially and (improves 20%), and illustrating it in animal body has better drug dynamic Mechanics.
4 pharmacokinetics in rats of table is tested
It should be understood that these examples are only for illustrating the present invention and are not intended to limit the scope of the present invention, in embodiment not The experimental method of actual conditions is indicated, usually according to normal condition, or according to the normal condition proposed by manufacturer.Unless in addition saying Bright, otherwise parts and percentages are parts by weight and weight percent.
The above content is a further detailed description of the present invention in conjunction with specific preferred embodiments, and it cannot be said that The specific implementation of the present invention is confined to these explanations.For those of ordinary skill in the art to which the present invention belongs, exist Under the premise of not departing from present inventive concept, a number of simple deductions or replacements can also be made, all shall be regarded as belonging to the present invention's Protection domain.

Claims (9)

1. a kind of diaminopyrimidine compounds, it is characterised in that:Diaminopyrimidine compounds or its pharmacy as shown in formula (I) Upper acceptable salt,
Wherein, R1a、R1b、R1c、R2a、R2b、R3a、R3b、R4a、R4b、R5a、R5b、R6、R7a、R7b、R8a、R8b、R9a、R9b、R10a、R10b、 R14a、R14b、R14cIt is each independently hydrogen or deuterium;
R16For Cl;
R11、R12、R13、R15、R17a、R17b、R17c、R18a、R18b、R18c、R19、R20、R21And R22For hydrogen;
Additional conditions are R1a、R1b、R1c、R2a、R2b、R3a、R3b、R4a、R4b、R5a、R5b、R6、R7a、R7b、R8a、R8b、R9a、R9b、R10a、 R10b、R14a、R14b、R14cIn it is at least one be deuterated or deuterium.
2. diaminopyrimidine compounds according to claim 1, it is characterised in that:R1a、R1bAnd R1cIt is deuterium.
3. diaminopyrimidine compounds according to claim 1, it is characterised in that:R7a、R7b、R8a、R8b、R9a、R9b、R10a And R10bIt is each independently deuterium or hydrogen.
4. diaminopyrimidine compounds according to claim 1, it is characterised in that:R14a、R14bAnd R14cIt is deuterium.
5. diaminopyrimidine compounds according to claim 1, it is characterised in that:The compound is selected from the group compound Or its pharmaceutically acceptable salt:
6. a kind of pharmaceutical composition preparation method, it is characterised in that:Pharmaceutically acceptable carrier and Claims 1 to 5 are appointed Diaminopyrimidine compounds or its pharmaceutically acceptable salt described in meaning one are mixed, to form pharmaceutical composition.
7. a kind of pharmaceutical composition, it is characterised in that:It contains pharmaceutically acceptable carrier and as Claims 1 to 5 is arbitrary The pharmaceutical composition of diaminopyrimidine compounds or its pharmaceutically acceptable salt described in one.
8. pharmaceutical composition according to claim 7, it is characterised in that:It also includes other drugs, and the drug is to control Treat cancer, angiocardiopathy, inflammation, infection, immunity disease, cell proliferation disorders, viral disease, metabolic disease, Or the drug of organ transplant.
9. a kind of diaminopyrimidine compounds as claimed in any one of claims 1 to 5, wherein or its pharmaceutically acceptable salt Purposes, it is characterised in that:It is used to prepare the pharmaceutical composition for inhibiting anaplastic lymphoma kinase.
CN201610587472.0A 2015-12-02 2016-07-22 A kind of diaminopyrimidine compounds and the composition comprising the compound Active CN106188138B (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
CN201810922557.9A CN108948082A (en) 2015-12-02 2016-07-22 A kind of diaminopyrimidine compounds and the composition comprising the compound
CN202111097366.1A CN113912648A (en) 2015-12-02 2016-07-22 Diaminopyrimidine compound and composition containing same
PCT/CN2016/096320 WO2017092413A1 (en) 2015-12-02 2016-08-23 Diaminopyrimidine compounds and composition comprising same

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN2015108755061 2015-12-02
CN201510875506 2015-12-02

Related Child Applications (2)

Application Number Title Priority Date Filing Date
CN201810922557.9A Division CN108948082A (en) 2015-12-02 2016-07-22 A kind of diaminopyrimidine compounds and the composition comprising the compound
CN202111097366.1A Division CN113912648A (en) 2015-12-02 2016-07-22 Diaminopyrimidine compound and composition containing same

Publications (2)

Publication Number Publication Date
CN106188138A CN106188138A (en) 2016-12-07
CN106188138B true CN106188138B (en) 2018-07-24

Family

ID=57491836

Family Applications (3)

Application Number Title Priority Date Filing Date
CN201810922557.9A Pending CN108948082A (en) 2015-12-02 2016-07-22 A kind of diaminopyrimidine compounds and the composition comprising the compound
CN201610587472.0A Active CN106188138B (en) 2015-12-02 2016-07-22 A kind of diaminopyrimidine compounds and the composition comprising the compound
CN202111097366.1A Pending CN113912648A (en) 2015-12-02 2016-07-22 Diaminopyrimidine compound and composition containing same

Family Applications Before (1)

Application Number Title Priority Date Filing Date
CN201810922557.9A Pending CN108948082A (en) 2015-12-02 2016-07-22 A kind of diaminopyrimidine compounds and the composition comprising the compound

Family Applications After (1)

Application Number Title Priority Date Filing Date
CN202111097366.1A Pending CN113912648A (en) 2015-12-02 2016-07-22 Diaminopyrimidine compound and composition containing same

Country Status (2)

Country Link
CN (3) CN108948082A (en)
WO (1) WO2017092413A1 (en)

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3381925B1 (en) * 2015-11-27 2020-12-16 Chia Tai Tianqing Pharmaceutical Group Co., Ltd. Deuterium-modified brigatinib derivatives, pharmaceutical compositions comprising same, and use thereof
CN106220608B (en) 2016-07-25 2018-11-27 安润医药科技(苏州)有限公司 Diphenylamino pyrimidine and triaizine compounds, its Pharmaceutical composition and purposes
WO2019007293A1 (en) * 2017-07-01 2019-01-10 浙江同源康医药股份有限公司 Compound used as alk kinase inhibitor and use thereof
WO2019134573A1 (en) * 2018-01-04 2019-07-11 深圳市塔吉瑞生物医药有限公司 Method for preparing deuterated diphenylaminopyrimidine compound and crystal form thereof
WO2019154091A1 (en) * 2018-02-07 2019-08-15 深圳市塔吉瑞生物医药有限公司 Substituted diaminopyrimidine compound
WO2021098883A1 (en) * 2019-11-21 2021-05-27 浙江同源康医药股份有限公司 Compound used as egfr kinase inhibitor and use thereof
WO2021150613A1 (en) 2020-01-20 2021-07-29 Incyte Corporation Spiro compounds as inhibitors of kras
US11739102B2 (en) 2020-05-13 2023-08-29 Incyte Corporation Fused pyrimidine compounds as KRAS inhibitors
WO2022072783A1 (en) 2020-10-02 2022-04-07 Incyte Corporation Bicyclic dione compounds as inhibitors of kras
CA3235146A1 (en) 2021-10-14 2023-04-20 Incyte Corporation Quinoline compounds as inhibitors of kras

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009045476A1 (en) * 2007-10-02 2009-04-09 Concert Pharmaceuticals, Inc. Pyrimidinedione derivatives
CA2723961C (en) * 2008-05-21 2017-03-21 Ariad Pharmaceuticals, Inc. Phosphorous derivatives as kinase inhibitors
US20150299166A1 (en) * 2012-12-20 2015-10-22 Concert Pharmaceuticals, Inc. Deuterated alk inhibitors
CN103965114B (en) * 2013-01-28 2016-01-06 苏州泽璟生物制药有限公司 Deuterated phenyl amino pyrimidine compounds and comprise the pharmaceutical composition of this compound
CN104109149B (en) * 2013-04-22 2018-09-28 苏州泽璟生物制药有限公司 Deuterated diaminopyrimidine compounds and the pharmaceutical composition comprising the compound
CN104327053A (en) * 2014-08-06 2015-02-04 北京凯悦宁医药科技有限公司 Deuterated crizotinib and derivative thereof, preparation method and application
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
CN105061506B (en) * 2015-07-27 2017-08-29 苏州明锐医药科技有限公司 Antineoplastic AP26113 preparation method
EP3381925B1 (en) * 2015-11-27 2020-12-16 Chia Tai Tianqing Pharmaceutical Group Co., Ltd. Deuterium-modified brigatinib derivatives, pharmaceutical compositions comprising same, and use thereof

Also Published As

Publication number Publication date
WO2017092413A1 (en) 2017-06-08
CN106188138A (en) 2016-12-07
CN108948082A (en) 2018-12-07
CN113912648A (en) 2022-01-11

Similar Documents

Publication Publication Date Title
CN106188138B (en) A kind of diaminopyrimidine compounds and the composition comprising the compound
EP3630761B1 (en) Kras g12c inhibitors and methods of using the same
CN108699081B (en) A kind of macrocyclic compound and the composition comprising the compound
WO2021072232A1 (en) Bicyclic amines as cdk2 inhibitors
CN105189478B (en) Uracil deoxyriboside triphosphatase inhibitor
CN107973791A (en) Condensed ring or tricyclic Arylpyrimidines compound are used as kinase inhibitor
CN109843888A (en) It can be used for the 6 for the treatment of cancer, 7,8,9- tetrahydro -3H- pyrazolo [4,3-f] isoquinilone derivatives
JP2019537610A (en) FGFR4 inhibitor, its production method and pharmaceutical application
EP3312180B1 (en) Use of pteridinone derivative serving as egfr inhibitor
BR112015002709B1 (en) COMPOUNDS, PHARMACEUTICAL COMPOSITION, USE THEREOF AND USE OF A COMPOUND COMBINATION OR PHARMACEUTICAL COMPOSITION AND A SECOND PROPHYLATIC OR THERAPEUTIC AGENT
US20220227729A1 (en) Identification and use of kras inhibitors
CN103717602A (en) Kinase inhibitors
BRPI0619272A2 (en) 3- (substituted amino) -pyrazolo [3,4-d] pyrimidines as kinase inhibitors ephb and vegfr2
BRPI0619517A2 (en) pyrazolo [1,5-a] pyridine-3-carboxylic acids as inhibitors of ephb and vegfr2 kinase
CN105315285A (en) 2,4-disubstituted 7H-pyrrolo[2,3-d]pyrimidine derivative, preparation method and medical uses thereof
TW201319067A (en) Triazolopyridine compounds
BR112016001333B1 (en) INDOL AND PYRROL DERIVATIVES AND THEIR USE, PROCESSES FOR THE PRODUCTION THEREOF, PHARMACEUTICAL COMPOSITIONS AND THEIR USE AND COMBINATIONS AND THEIR USE
CN108290899A (en) A kind of substituted Pyrrolopyrimidine compounds and its application
CN110214140A (en) Estrogenic agents
CN108026046B (en) Substituted quinazoline compounds and their use as inhibitors of G12C mutant KRAS, HRAS and/or NRAS proteins
CN105884695B (en) Heterocyclic derivatives species tyrosine kinase inhibitor
CN108368059A (en) A kind of substituted phthalazinone compounds and its pharmaceutical composition
JP2023533349A (en) Compounds as BTK inhibitors, and methods for producing and applying the same
CN104557913B (en) Pyridopyrimidine compounds as well as preparation method and application thereof
JP2021504332A (en) Pyrazolopyridinone compound

Legal Events

Date Code Title Description
C06 Publication
PB01 Publication
C10 Entry into substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant