CN109851638B - Substituted diaminopyrimidine compounds - Google Patents

Substituted diaminopyrimidine compounds Download PDF

Info

Publication number
CN109851638B
CN109851638B CN201910066072.9A CN201910066072A CN109851638B CN 109851638 B CN109851638 B CN 109851638B CN 201910066072 A CN201910066072 A CN 201910066072A CN 109851638 B CN109851638 B CN 109851638B
Authority
CN
China
Prior art keywords
compound
added
cancer
stirred
alk
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
CN201910066072.9A
Other languages
Chinese (zh)
Other versions
CN109851638A (en
Inventor
王义汉
李焕银
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Shenzhen Targetrx Inc
Original Assignee
Shenzhen Targetrx Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Shenzhen Targetrx Inc filed Critical Shenzhen Targetrx Inc
Publication of CN109851638A publication Critical patent/CN109851638A/en
Application granted granted Critical
Publication of CN109851638B publication Critical patent/CN109851638B/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
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/32One oxygen, sulfur or nitrogen atom
    • C07D239/42One nitrogen atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • 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 Table
    • 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
    • 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 Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6561Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing systems of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring or ring system, with or without other non-condensed hetero rings

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Epidemiology (AREA)
  • Hematology (AREA)
  • Oncology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention provides a substituted diaminopyrimidine compound shown as a formula (I), or pharmaceutically acceptable salt, crystal form, prodrug, metabolite, hydrate, solvate, stereoisomer or isotope derivative thereof, and a pharmaceutical composition and application thereof. The compound can be used for treating ALK-mediated related diseases, such as non-small cell lung cancer, breast cancer, neural tumor, esophagus cancer, soft tissue cancer, lymphoma and leukemia.

Description

Substituted diaminopyrimidine compounds
Technical Field
The invention belongs to the technical field of medicines. In particular, the present invention relates to substituted diaminopyrimidine compounds having an inhibitory effect on protein tyrosine kinases, to pharmaceutical compositions comprising the compounds, to processes for preparing the compounds and to the use of the compounds in therapy. Wherein the compounds can be used as a new generation ALK inhibitor and can be used for treating ALK mediated cancer, and the compounds have more excellent pharmacokinetic properties.
Background
Anaplastic Lymphoma Kinase (ALK) is a receptor-type protein tyrosine kinase, belonging to the insulin receptor superfamily. It was discovered in 1994 from Anaplastic Large Cell Lymphoma (ALCL) by Morris and Shiota et al as a product of chromosomal rearrangement, and the most common way of fusion is the fusion of the NPM (nucleocapsid) gene on chromosome 5 with the ALK gene on chromosome 2. NPM-ALK fusion proteins were detected in nearly 75% of ALK-positive ALCL patients, and in several subsequent studies, different ALK fusion forms were found in a wide variety of cancers, including inflammatory myofibroblastic tumors and diffuse large B-cell lymphomas. Nevertheless, the importance of ALK kinases as a target for effective antitumor drugs is not fully recognized. Until 2007, Soda et al found that the occupancy probability of the EML4-ALK fusion protein in non-small cell lung cancer (NSCLC) was 5%, and the importance of ALK kinase as a target of an antitumor drug was not highlighted. This is because the number of cancer patients worldwide is very large, with lung cancer at the first, more than 8000 new cases of ALK-positive lung cancer annually in the united states, and more than 6.5 ten thousand new cases annually in china, and the global 5-year survival rate for lung cancer is only 15%.
It is attractive to the eye that patients positive for the EML4-ALK gene do not normally carry Epidermal Growth Factor Receptor (EGFR) or murine Kirsten sarcoma virus (KRAS) mutations, making the EML4-ALK fusion gene a unique molecular target for non-small cell lung cancer. In addition, amplification or point mutation of the ALK gene has been found in neuroblastoma (Neuroblastomas), anaplastic thyroid cancer (anaplastic thyroid cancer), and ovarian cancer (ovarian cancer).
The first small-molecule inhibitor Crizotinib (xalkonib, xalkorri) aiming at the ALK fusion gene is developed by pyroxene and belongs to the first generation of ALK inhibitors. However, despite crizotinib achieving an objective response rate of 60-74% and a good median progression-free survival (8-11 months) in ALK + NSCLC patients, most patients develop disease recurrence after 1 year of treatment, i.e. develop acquired resistance. Acquired resistance mechanisms of crizotinib have also been identified, including gain of ALK fusion genes, activation of signaling pathways, secondary mutations in the ALK kinase region, and other mechanisms. Approximately 40% of ALK-positive patients do not respond objectively as soon as they receive crizotinib treatment, and 1/3 crizotinib-resistant patients develop secondary mutations that induce secondary resistance.
There are several second-generation ALK inhibitors that are effective in overcoming the deficiencies in resistance to crizotinib treatment, such as Ceritinib (Zykadia, nova pharmaceuticals) and einitinib (Alectinib, alekensa, roche pharmaceuticals). However, while these second generation inhibitors are effective in overcoming most crizotinib resistance mutations, they are still ineffective against some mutations, such as ceritinib pair F1174C/V, alentinib pair I1171N/T/S and they are still not as effective against G1202R.
Furthermore, poor absorption, distribution, metabolism and/or excretion (ADME) properties are known to be the major cause of failure in many drug candidate clinical trials. Many drugs currently on the market also have limited their range of application due to poor ADME properties. The rapid metabolism of drugs can result in the difficulty of obtaining many drugs that are otherwise effective in treating disease due to their rapid metabolic clearance from the body. Although frequent or high dose administration may solve the problem of rapid clearance of the drug, this method may cause problems such as poor patient compliance, side effects caused by high dose administration, and increased treatment costs. In addition, rapidly metabolizing drugs may also expose patients to undesirable toxic or reactive metabolites.
There is therefore a great need to develop new ALK inhibitors that are more effective, safer and better in pharmacokinetic properties.
Summary of The Invention
In view of the above technical problems, the present invention discloses a novel diaminopyrimidine compound, a composition thereof, and a use thereof, which have better efficacy and safety, lower side effects, better pharmacodynamic/pharmacokinetic properties, can be used for treating ALK kinase-mediated cancer, and have high selectivity for a drug-resistant mutation L1196M.
In contrast, the invention adopts the following technical scheme:
in a first aspect of the invention, there is provided a compound of formula (I):
Figure BDA0001955701090000031
wherein:
R1and R2Independently selected from H, D, halogen, -CN, -OH, -OC1-6Alkyl, -NH2、-NHC1-6Alkyl, -N (C)1-6Alkyl radical)2、C1-6Alkyl radical, C1-6Haloalkyl, C2-6Alkenyl radical, C2-6Alkynyl, or R1And R2Form C with the atoms to which they are attached6-10Aryl or 5-10 membered heteroaryl, preferably pyrrolyl; wherein said group is optionally substituted with 1,2, 3,4,5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15D;
R3selected from H, D, halogen, -CN or C1-6An alkoxy group; wherein said C1-6Alkoxy is optionally substituted with 1,2, 3,4,5, 6, 7, 8, 9, 10, 11, 12, or 13D;
R4selected from H, D, halogen, -CN or C1-6An alkoxy group; wherein said C1-6Alkoxy is optionally substituted with 1,2, 3,4,5, 6, 7, 8, 9, 10, 11, 12, or 13D;
R5selected from:
Figure BDA0001955701090000032
optionally substituted with 1,2, 3,4,5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15D;
R6selected from the group consisting of:
Figure BDA0001955701090000033
which is optionally substituted with 1,2, 3,4,5 or 6D;
denotes a bond to the parent nucleus;
provided that when R is3Is C1-6Alkoxy or deuterated derivatives thereof, at least one of the following options holds:
(1)R1and R2Form C with the atoms to which they are attached6-10Aryl or 5-10 membered heteroaryl;
(2) except for R3In addition, the molecule also has at least one D atom;
(3)R5is a formula (b);
or a pharmaceutically acceptable salt, crystal form, prodrug, metabolite, hydrate, solvate, stereoisomer, or isotopic derivative thereof.
In another aspect, the invention provides a pharmaceutical composition comprising a compound of the invention and a pharmaceutically acceptable excipient. In a specific embodiment, the compounds of the present invention are provided in an effective amount in the pharmaceutical composition. In particular embodiments, the compounds of the present invention are provided in a therapeutically effective amount. In a particular embodiment, the compounds of the present invention are provided in a prophylactically effective amount.
In another aspect, the invention provides pharmaceutical compositions comprising a compound of the invention and a pharmaceutically acceptable excipient, which also contain an additional therapeutic agent.
In another aspect, the present invention provides a method for preparing the pharmaceutical composition as described above, comprising the steps of: pharmaceutically acceptable excipients are mixed with the compounds of the present invention to form pharmaceutical compositions.
In another aspect, the present invention provides a method of treating a cancer-related disorder caused by ALK mutation in a subject in need thereof, the method comprising administering to the subject an effective dose of a compound of the present invention. In specific embodiments, the cancer is selected from non-small cell lung cancer, breast cancer, neural tumors (such as glioblastoma and neuroblastoma); esophageal cancer, soft tissue cancer (such as rhabdomyosarcoma, etc.); various forms of lymphoma, such as non-hodgkin's lymphoma (NHL), known as Anaplastic Large Cell Lymphoma (ALCL); various forms of leukemia. In particular embodiments, the non-small cell lung cancer is ALK-positive non-small cell lung cancer. In particular embodiments, the compound is administered orally, subcutaneously, intravenously, or intramuscularly. In particular embodiments, the compound is administered chronically.
In another aspect, the present invention provides the use of a compound of the present invention in the manufacture of a medicament for the treatment of ALK-mediated cancer. In specific embodiments, the cancer is selected from the group consisting of non-small cell lung cancer, breast cancer, neural tumors, esophageal cancer, soft tissue cancer, lymphoma, and leukemia. In specific embodiments, the non-small cell lung cancer is ALK-positive non-small cell lung cancer; wherein the lymphoma is anaplastic large cell lymphoma.
Other objects and advantages of the present invention will be apparent to those skilled in the art from the following detailed description, examples and claims.
Detailed Description
Definition of
Chemical definition
The definitions of specific functional groups and chemical terms are described in more detail below.
When a range of values is recited, it is intended to include each value and every subrange within the range. E.g. "C1-C6Alkyl "includes C1、C2、C3、C4、C5、C6、C1-C6、C1-C5、C1-C4、C1-C3、C1-C2、C2-C6、C2-C5、C2-C4、C2-C3、C3-C6、C3-C5、C3-C4、C4-C6、C4-C5And C5-C6An alkyl group.
“C1-6Alkyl "refers to a straight or branched chain saturated hydrocarbon group having 1 to 6 carbon atoms. In some embodiments, C1-4Alkyl groups are preferred. C1-6Examples of alkyl groups include: methyl (C)1) Ethyl (C)2) N-propyl (C)3) Isopropyl (C)3) N-butyl (C)4) Tert-butyl (C)4) Sec-butyl (C)4) Isobutyl (C)4) N-pentyl group (C)5) 3-pentyl radical (C)5) Pentyl group (C)5) Neopentyl (C)5) 3-methyl-2-butyl (C)5) Tert-amyl (C)5) And n-hexyl (C)6). The term "C1-6Alkyl "also includes heteroalkyl wherein one or more (e.g., 1,2, 3, or 4) carbon atoms are replaced with a heteroatom (e.g., oxygen, sulfur, nitrogen, boron, silicon, phosphorus). The alkyl group may be optionally substituted with one or more substituents, for example, 1 to 5 substituents, 1 to 3 substituents, or 1 substituent. Conventional alkyl abbreviations include: me (-CH)3)、Et(-CH2CH3)、iPr(-CH(CH3)2)、nPr(-CH2CH2CH3)、n-Bu(-CH2CH2CH2CH3) Or i-Bu (-CH)2CH(CH3)2)。
“C2-6Alkenyl "refers to a straight or branched hydrocarbon group having 2 to 6 carbon atoms and at least one carbon-carbon double bond. In some embodiments, C2-4Alkenyl groups are preferred. C2-6Examples of alkenyl groups include: vinyl radical (C)2) 1-propenyl (C)3) 2-propenyl (C)3) 1-butenyl (C)4) 2-butenyl (C)4) Butadienyl radical (C)4) Pentenyl (C)5) Pentadienyl (C)5) Hexenyl (C)6) And so on. The term "C2-6Alkenyl "also includes heteroalkenyl groups in which one or more (e.g., 1,2, 3, or 4) carbon atoms are replaced with a heteroatom (e.g., oxygen, sulfur, nitrogen, boron, silicon, phosphorus). The alkenyl group may be optionally substituted with one or more substituents, for example, 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
“C2-6Alkynyl "refers to a straight or branched hydrocarbon group having 2 to 6 carbon atoms, at least one carbon-carbon triple bond, and optionally one or more carbon-carbon double bonds. In some embodiments, C2-4Alkynyl groups are preferred. C2-6Examples of alkynyl groups include, but are not limited to: ethynyl (C)2) 1-propynyl (C)3) 2-propynyl (C)3) 1-butynyl (C)4) 2-butynyl (C)4) Pentynyl group (C)5) Hexynyl (C)6) And so on. The term "C2-6Alkynyl also includes heteroalkynyl in which one or more (e.g., 1,2, 3, or 4) carbon atoms are replaced with a heteroatom (e.g., oxygen, sulfur, nitrogen, boron, silicon, phosphorus). An alkynyl group may be optionally substituted with one or more substituents, for example, 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
"halo" or "halogen" refers to fluoro (F), chloro (Cl), bromo (Br), and iodo (I). In some embodiments, the halogen group is F, -Cl, or Br. In some embodiments, the halogen group is F or Cl. In some embodiments, the halogen group is F.
“C1-6Haloalkyl "represents the above-mentioned" C1-6Alkyl "substituted with one or more halo groups. Examples include monohalogen substituted, dihalogen substituted and polyhaloalkyl including perhalo. A monohalogen substituent may have an iodine, bromine, chlorine or fluorine atom in the group; two halogen substituents and multiple halogen substituents may have two or more of the same halogen atom or a combination of different halogens. Examples of preferred haloalkyl groups include monofluoromethyl, difluoromethyl, trifluoromethyl, chloromethyl, dichloromethyl, trichloromethyl, pentafluoroethyl, heptafluoropropyl, difluorochloromethyl, dichlorofluoroMethyl, difluoroethyl, difluoropropyl, dichloroethyl and dichloropropyl. The haloalkyl group can be substituted at any available point of attachment, for example, 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
“C1-C6Alkoxy "refers to the group-OR, where R is substituted OR unsubstituted C1-C6An alkyl group. In some embodiments, C1-C4Alkoxy groups are particularly preferred. Specific said alkoxy groups include, but are not limited to: methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, tert-butoxy, sec-butoxy, n-pentoxy, n-hexoxy and 1, 2-dimethylbutoxy.
“C6-10Aryl "refers to a group having a monocyclic or polycyclic (e.g., bicyclic) 4n +2 aromatic ring system (e.g., having 6 or 10 pi electrons shared in a cyclic arrangement) of 6 to 10 ring carbon atoms and zero heteroatoms. In some embodiments, an aryl group has six ring carbon atoms ("C)6Aryl "; for example, phenyl). In some embodiments, an aryl group has ten ring carbon atoms ("C)10Aryl "; e.g., naphthyl, e.g., 1-naphthyl and 2-naphthyl). Aryl also includes ring systems in which the aforementioned aryl ring is fused to one or more cycloalkyl or heterocyclyl groups, and the point of attachment is on the aryl ring, in which case the number of carbon atoms continues to represent the number of carbon atoms in the aryl ring system. The aryl group may be optionally substituted with one or more substituents, for example, 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
"5-10 membered heteroaryl" refers to a group having a 5-10 membered monocyclic or bicyclic 4n +2 aromatic ring system (e.g., having 6 or 10 pi electrons shared in a cyclic arrangement) with ring carbon atoms and 1-4 ring heteroatoms, wherein each heteroatom is independently selected from nitrogen, oxygen, and sulfur. In heteroaryl groups containing one or more nitrogen atoms, the point of attachment may be a carbon or nitrogen atom, as valency permits. Heteroaryl bicyclic ring systems may include one or more heteroatoms in one or both rings. Heteroaryl also includes ring systems in which the aforementioned heteroaryl ring is fused to one or more cycloalkyl or heterocyclyl groups, and the point of attachment is on the heteroaryl ring, in which case the number of carbon atoms continues to represent the number of carbon atoms in the heteroaryl ring system. In some embodiments, 5-6 membered heteroaryl groups are particularly preferred, which are 5-6 membered monocyclic or bicyclic 4n +2 aromatic ring systems having ring carbon atoms and 1-4 ring heteroatoms. Exemplary 5-membered heteroaryl groups containing one heteroatom include, but are not limited to: pyrrolyl, furanyl and thienyl. Exemplary 5-membered heteroaryl groups containing two heteroatoms include, but are not limited to: imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl and isothiazolyl. Exemplary 5-membered heteroaryl groups containing three heteroatoms include, but are not limited to: triazolyl, oxadiazolyl (e.g., 1,2, 4-oxadiazolyl), and thiadiazolyl. Exemplary 5-membered heteroaryl groups containing four heteroatoms include, but are not limited to: a tetrazolyl group. Exemplary 6-membered heteroaryl groups containing one heteroatom include, but are not limited to: a pyridyl group. Exemplary 6-membered heteroaryl groups containing two heteroatoms include, but are not limited to: pyridazinyl, pyrimidinyl and pyrazinyl. Exemplary 6-membered heteroaryl groups containing three or four heteroatoms include, but are not limited to: triazinyl and tetrazinyl. Exemplary 7-membered heteroaryl groups containing one heteroatom include, but are not limited to: azepinyl, oxacycloheptyl, and thiacycloheptyl trienyl groups. Exemplary 5, 6-bicyclic heteroaryls include, but are not limited to: indolyl, isoindolyl, indazolyl, benzotriazolyl, benzothienyl, isobenzothienyl, benzofuranyl, benzisothiafuranyl, benzimidazolyl, benzoxazolyl, benzisoxazolyl, benzooxadiazolyl, benzothiazolyl, benzisothiazolyl, benzothiadiazolyl, indezinyl, and purinyl. Exemplary 6, 6-bicyclic heteroaryls include, but are not limited to: naphthyridinyl, pteridinyl, quinolinyl, isoquinolinyl, cinnolinyl, quinoxalinyl, phthalazinyl and quinazolinyl. The heteroaryl group may be optionally substituted with one or more substituents, for example, 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
Other definitions
The term "pharmaceutically acceptable salts" means, within the scope of sound medical judgment, suitable for use with humans and humansThose salts which contact the tissues of lower animals without undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, the pharmaceutically acceptable salts are described in detail by Berge et al in J.pharmaceutical Sciences (1977)66: 1-19. Pharmaceutically acceptable salts of the compounds of the present invention include salts derived from suitable inorganic and organic acids and bases. Examples of pharmaceutically acceptable non-toxic acid addition salts are salts of amino groups with inorganic acids, such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid, or with organic acids, such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid, or using methods used in the art, such as ion exchange methods. Other pharmaceutically acceptable salts include: adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cypionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, gluconate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, picrate, etc, Stearates, succinates, sulfates, tartrates, thiocyanates, p-toluenesulfonates, undecanoates, pentanoates, and the like. Pharmaceutically acceptable salts derived from suitable bases include alkali metals, alkaline earth metals, ammonium and N+(C1-4Alkyl radical)4And (3) salt. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium salts, and the like. Further pharmaceutically acceptable salts include, if appropriate, non-toxic ammonium salts, quaternary ammonium salts and amine cations formed using counterions such as halide, hydroxide, carboxylateAcid radical, sulfate radical, phosphate radical, nitrate radical, lower alkyl sulfonate radical and aryl sulfonate radical.
The "subject" to which the drug is administered includes, but is not limited to: a human (i.e., a male or female of any age group, e.g., a pediatric subject (e.g., an infant, a child, an adolescent) or an adult subject (e.g., a young adult, a middle-aged adult, or an older adult)) and/or a non-human animal, e.g., a mammal, e.g., a primate (e.g., a cynomolgus monkey, a rhesus monkey), a cow, a pig, a horse, a sheep, a goat, a rodent, a cat, and/or a dog. In some embodiments, the subject is a human. In some embodiments, the subject is a non-human animal.
"disease," "disorder," and "condition" are used interchangeably herein.
As used herein, unless otherwise specified, the term "treatment" includes the effect that occurs when a subject has a particular disease, disorder or condition, which reduces the severity of the disease, disorder or condition, or delays or slows the progression of the disease, disorder or condition ("therapeutic treatment"), and also includes the effect that occurs before the subject begins to have the particular disease, disorder or condition ("prophylactic treatment").
Generally, an "effective amount" of a compound is an amount sufficient to elicit a biological response of interest. As will be appreciated by those of ordinary skill in the art, the effective amount of a compound of the present invention may vary depending on the following factors: for example, biological goals, pharmacokinetics of the compound, the disease being treated, mode of administration, and the age, health, and condition of the subject. An effective amount includes both therapeutically and prophylactically therapeutically effective amounts.
As used herein, unless otherwise specified, a "therapeutically effective amount" of a compound is an amount sufficient to provide a therapeutic benefit in the treatment of a disease, disorder, or condition, or to delay or minimize one or more symptoms associated with a disease, disorder, or condition. A therapeutically effective amount of a compound refers to the amount of a therapeutic agent, alone or in combination with other therapies, that provides a therapeutic benefit in the treatment of a disease, disorder, or condition. The term "therapeutically effective amount" can include an amount that improves the overall treatment, reduces or avoids symptoms or causes of a disease or disorder, or enhances the therapeutic efficacy of other therapeutic agents.
As used herein, unless otherwise specified, a "prophylactically effective amount" of a compound is an amount sufficient to prevent a disease, disorder, or condition, or one or more symptoms associated with a disease, disorder, or condition, or to prevent recurrence of a disease, disorder, or condition. A prophylactically effective amount of a compound refers to the amount of a therapeutic agent, alone or in combination with other agents, that provides a prophylactic benefit in preventing a disease, disorder, or condition. The term "prophylactically effective amount" can include an amount that improves overall prophylaxis, or an amount that enhances the prophylactic efficacy of other prophylactic agents.
"combination" and related terms refer to the simultaneous or sequential administration of the therapeutic agents of the present invention. For example, a compound of the invention may be administered simultaneously or sequentially with another therapeutic agent in separate unit dosage forms, or simultaneously with another therapeutic agent in a single unit dosage form.
Herein, "deuterated", unless otherwise specified, means that one or more hydrogens of a compound or group are replaced with deuterium; deuterium can be mono-, di-, poly-, or fully substituted.
The invention also includes isotopically-labeled compounds, equivalent to those disclosed herein for the original compound. Examples of isotopes that can be listed as compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine and chlorine, respectively2H,3H,13C,14C,15N,17O,18O,31P,32P,35S,18F and36and (4) Cl. The compounds of the present invention, or enantiomers, diastereomers, isomers, or pharmaceutically acceptable salts or solvates thereof, wherein isotopes or other isotopic atoms containing such compounds are within the scope of the present invention. Certain isotopically-labelled compounds of the invention, e.g.3H and14among these, the radioactive isotope of C is useful in tissue distribution experiments of drugs and substrates. Tritium, i.e.3H and carbon 14, i.e.14C, their preparation and detection are relatively easy, and are the first choice among isotopes. Isotopically labeled compounds can be prepared by conventional methods by substituting readily available isotopically labeled reagents for non-isotopically labeled reagents using the protocols set forth in the examples.
The compounds of the invention may include one or more asymmetric centers, and thus may exist in a variety of "stereoisomeric" forms, e.g., enantiomeric and/or diastereomeric forms. For example, the compounds of the present invention may be individual enantiomers, diastereomers or geometric isomers (e.g., cis and trans isomers), or may be in the form of mixtures of stereoisomers, including racemic mixtures and mixtures enriched in one or more stereoisomers. Isomers may be separated from mixtures by methods known to those skilled in the art, including: chiral High Pressure Liquid Chromatography (HPLC) and the formation and crystallization of chiral salts; alternatively, preferred isomers may be prepared by asymmetric synthesis.
The compounds of the present invention may be in amorphous or crystalline form. Furthermore, the compounds of the present invention may exist in one or more crystalline forms. Accordingly, the present invention includes within its scope all amorphous or crystalline forms of the compounds of the present invention. The term "crystalline form" refers to the different arrangements of chemical drug molecules, typically expressed as the presence of the drug substance in the solid state. One drug can exist in a plurality of crystal form substances, and different crystal forms of the same drug can be dissolved and absorbed in vivo differently, so that the dissolution and release of the preparation can be influenced.
The term "solvate" refers to a complex of a compound of the present invention coordinated to solvent molecules in a specific ratio. "hydrate" refers to a complex formed by coordination of a compound of the present invention with water.
The term "prodrug" refers to a compound that is converted in vivo by hydrolysis, for example in the blood, into its active form with a medicinal effect. Pharmaceutically acceptable Prodrugs are described in t.higuchi and v.stella, Prodrugs as Novel Delivery Systems, vol.14 of a.c.s.symposium Series, Edward b.roche, ed., Bioreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon Press, 1987, and d.fleisher, s.ramon and h.bara "Improved oral Delivery: the solubility limits of the above overcom by the use of the drugs ", Advanced Drug Delivery Reviews (1996)19, (2)115-130, each of which is incorporated herein by reference.
A prodrug is any covalently bonded compound of the present invention that releases the parent compound in vivo when such prodrug is administered to a patient. Prodrugs are generally prepared by modifying functional groups in a manner such that the modification is effected by routine manipulation or in vivo cleavage to produce the parent compound. Prodrugs include, for example, compounds of the present invention wherein a hydroxy, amino, or sulfhydryl group is bonded to any group that, when administered to a patient, cleaves to form a hydroxy, amino, or sulfhydryl group. Thus, representative examples of prodrugs include, but are not limited to, acetate/amide, formate/amide, and benzoate/amide derivatives of hydroxy, mercapto, and amino functional groups of the compounds of formula (I). In addition, in the case of carboxylic acid (-COOH), esters such as methyl ester, ethyl ester, and the like may be used. The ester itself may be active and/or may hydrolyze under in vivo conditions in the human body. Suitable pharmaceutically acceptable in vivo hydrolysable ester groups include those which readily break down in the human body to release the parent acid or salt thereof.
Detailed Description
Compound (I)
In one embodiment, the present invention relates to compounds of formula (I):
Figure BDA0001955701090000101
wherein:
R1and R2Independently selected from H, D, halogen, -CN, -OH, -OC1-6Alkyl, -NH2、-NHC1-6Alkyl, -N (C)1-6Alkyl radical)2、C1-6Alkyl radical, C1-6Haloalkyl, C2-6Alkenyl radical, C2-6Alkynyl, or R1And R2Form C with the atoms to which they are attached6-10Aryl or 5-10 membered heteroaryl, preferably pyrrolyl; wherein said group is optionally substituted with 1,2, 3,4,5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15D;
R3selected from H, D, halogen, -CN or C1-6An alkoxy group; wherein said C1-6Alkoxy is optionally substituted with 1,2, 3,4,5, 6, 7, 8, 9, 10, 11, 12, or 13D;
R4selected from H, D, halogen, -CN or C1-6An alkoxy group; wherein said C1-6Alkoxy is optionally substituted with 1,2, 3,4,5, 6, 7, 8, 9, 10, 11, 12, or 13D;
R5selected from:
Figure BDA0001955701090000111
optionally substituted with 1,2, 3,4,5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15D;
R6selected from:
Figure BDA0001955701090000112
which is optionally substituted with 1,2, 3,4,5 or 6D;
denotes a bond to the parent nucleus;
provided that when R is3Is C1-6Alkoxy or deuterated derivatives thereof, at least one of the following options holds:
(1)R1and R2Form C with the atoms to which they are attached6-10Aryl or 5-10 membered heteroaryl;
(2) except for R3In addition, the molecule also has at least one D atom;
(3)R5is formula (b);
or a pharmaceutically acceptable salt, crystal form, prodrug, metabolite, hydrate, solvate, stereoisomer, or isotopic derivative thereof.
In another embodiment, the invention relates to compounds of formula (Ia):
Figure BDA0001955701090000113
wherein the content of the first and second substances,
R1’、R2’、R3’、R4’、R5’、R6’、R7’、R8’and R9’Each independently selected from hydrogen or deuterium;
X1、X2、X3、X4and X5Each independently selected from CH3、CD3、CHD2Or CH2D;
Provided that when X is1、X2、X3、X4And X5Are all CH3When R is1’、R2’、R3’、R4’、R5’、R6’、R7’、R8’And R9’At least one of which is deuterium;
or a pharmaceutically acceptable salt, crystal form, prodrug, metabolite, hydrate, solvate, stereoisomer, or isotopic derivative thereof.
In more specific embodiments, R1’、R2’、R3’、R4’Is deuterium.
In a more specific embodiment, X1And X2Each independently selected from CD3、CHD2Or CH2D; preferably, X1And X2Is a CD3
In more specific embodiments, R9’Is deuterium.
In a more specific embodiment, X4And X5Each independently selected from CD3、CHD2Or CH2D; preferably, X4And X5Is a CD3
As a specific embodiment of the present invention, the compound of formula (Ia) contains at least one deuterium atom, more preferably two deuterium atoms, more preferably three deuterium atoms, more preferably four deuterium atoms, more preferably five deuterium atoms, more preferably six deuterium atoms, more preferably seven deuterium atoms, more preferably eight deuterium atoms, more preferably nine deuterium atoms, more preferably ten deuterium atoms, more preferably eleven deuterium atoms, more preferably twelve deuterium atoms, more preferably thirteen deuterium atoms, more preferably fourteen deuterium atoms, more preferably fifteen deuterium atoms, more preferably sixteen deuterium atoms, more preferably seventeen deuterium atoms, more preferably eighteen deuterium atoms, more preferably nineteen deuterium atoms, more preferably twenty-one deuterium atoms, more preferably twenty-two deuterium atoms, more preferably twenty-three deuterium atoms, more preferably twenty-four deuterium atoms.
As a specific embodiment of the present invention, the deuterium isotope content of deuterium at the deuterium position is at least 0.015% greater than the natural deuterium isotope content, preferably greater than 30%, more preferably greater than 50%, more preferably greater than 75%, more preferably greater than 95%, more preferably greater than 99%.
Specifically, in the present invention R1’、R2’、R3’、R4’、R5’、R6’、R7’、R8’、R9’、X1、X2、X3、X4And X5The deuterium isotope content in each deuterated position is at least 5%, preferably greater than 10%, more preferably greater than 15%, more preferably greater than 20%, more preferably greater than 25%, more preferably greater than 30%, more preferably greater than 35%, more preferably greater than 40%, more preferably greater than 45%, more preferably greater than 50%, more preferably greater than 55%, more preferably greater than 60%, more preferably greater than 65%, more preferably greater than 70%, more preferably greater than 75%, more preferably greater than 80%, more preferably greater than 85%, more preferably greater than 90%, more preferably greater than 95%, more preferably greater than 99%.
In another embodiment, R of the compound of formula (Ia)1’、R2’、R3’、R4’、R5’、R6’、R7’、R8’、R9’、X1、X2、X3、X4And X5Preferably, at least one of the deuterium containing atoms, more preferably two deuterium containing atoms, more preferably three deuterium containing atoms, more preferably four deuterium containing atoms, more preferably five deuterium containing atoms, more preferably six deuterium containing atoms, more preferably seven deuterium containing atoms, more preferably eight deuterium containing atoms, more preferably nine deuterium containing atoms, more preferably ten deuterium containing atoms, more preferably eleven deuterium atoms, more preferably twelve deuterium atoms, more preferably thirteen deuterium atoms, more preferably fourteen deuterium atoms, more preferably fifteen deuterium atoms, more preferably sixteen deuterium atoms, more preferably seventeen deuterium atoms, more preferably eighteen deuterium atoms, more preferably nineteen deuterium atoms, more preferably twenty-one deuterium atoms, more preferably twenty-two deuterium atoms, more preferably twenty-three deuterium atoms, more preferably twenty-four deuterium atoms. In particular, the compound of formula (Ia) contains at least one, two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, fifteen, sixteen, seventeen, eighteen, nineteen, twenty-one, twenty-two, twenty-three, twenty-four deuterium atoms.
As a specific embodiment of the present invention, X1And X2Each independently selected from CH3、CD3、CHD2Or CH2D; in another embodiment, X1Is CH3(ii) a In another embodiment, X1Is a CD3(ii) a In another embodiment, X1Is CH2D; in another embodiment, X1Is CHD2(ii) a In another specific embodiment, X2Is CH3(ii) a In another embodiment, X2Is a CD3(ii) a In another embodiment, X2Is CH2D; in another embodiment, X2Is CHD2(ii) a In another embodiment, X1Is CH3,X2Is a CD3(ii) a In another embodiment, X1Is a CD3,X2Is CH3(ii) a In another embodiment, X1Is CH3,X2Is CH3(ii) a In another embodiment, X1Is a CD3,X2Is a CD3
As a specific embodiment of the present invention, X3Is selected from CH3、CD3、CHD2Or CH2D; in another embodiment, X3Is CH3(ii) a In another embodiment, X3Is a CD3(ii) a In another embodiment, X3Is CH2D; in another embodiment, X1Is CHD2
As a specific embodiment of the present invention, X4And X5Each independently selected from CH3、CD3、CHD2Or CH2D; in another specific embodiment, X4Is CH3(ii) a In another embodiment, X4Is a CD3(ii) a In another embodiment, X4Is CH2D; in another embodiment, X4Is CHD2(ii) a In another embodiment, X5Is CH3(ii) a In another embodiment, X5Is a CD3(ii) a In another embodiment, X5Is CH2D; in another embodiment, X5Is CHD2(ii) a In another embodiment, X4Is CH3,X5Is a CD3(ii) a In another embodiment, X4Is a CD3,X5Is CH3(ii) a In another embodiment, X4Is CH3,X5Is CH3(ii) a In another embodiment, X4Is a CD3,X5Is a CD3
As a particular embodiment of the invention, R1’、R2’、R3’、R4’、R5’、R6’、R7’、R8’And R9’Each independently selected from hydrogen or deuterium; in another embodiment, R1’Is hydrogen; in another embodiment, R1’Is deuterium; in another embodiment, R2’Is hydrogen; in another embodiment, R2’Is deuterium; in another embodiment, R3’Is hydrogen; in another embodiment, R3’Is deuterium; in another embodiment, R4’Is hydrogen; in another embodiment, R4’Is deuterium; in another embodiment, R5’Is hydrogen; in another embodiment, R5’Is deuterium; in another embodiment, R6’Is hydrogen; in another embodiment, R6’Is deuterium; in another embodiment, R7’Is hydrogen; in another embodiment, R7’Is deuterium; in another embodiment, R8’Is hydrogen; in another embodiment, R8’Is deuterium; in another embodiment, R9’Is hydrogen; in another embodiment, R9’Is deuterium; in another embodiment, R1’、R2’、R3’、R4’Are the same; in another embodiment, R6’、R7’、R8’And R9’Are the same; in another embodiment, R1’、R2’、R3’、R4’Are all deuterium; in another embodiment, R1’、R2’、R3’、R4’Are both hydrogen; in another embodiment, R1’、R2’、R3’、R4’And R5’Are all deuterium; in another embodiment, R1’、R2’、R3’、R4’And R5’Are both hydrogen; in another embodiment, R6’、R7’、R8’And R9’Are all deuterium; in another embodiment, R6’、R7’、R8’And R9’Are all hydrogen.
In another embodiment, the invention relates to compounds of formula (X):
Figure BDA0001955701090000141
wherein the content of the first and second substances,
R1、R2、R3、R4、R5、R6、R7、R8and R9Each independently selected from hydrogen or deuterium;
Y1、Y2、Y3、Y4、Y5、Y6、Y7and Y8Each independently selected from hydrogen or deuterium;
X1、X2、X3、X4and X5Each independently selected from CH3、CD3、CHD2Or CH2D;
Provided that if X is1、X2、X3、X4And X5Are all CH3Then R1、R2、R3、R4、R5、R6、R7、R8、R9、Y1、Y2、Y3、Y4、Y5、Y6、Y7And Y8At least one of which is deuterium; and if X1、X2、X4And X5Are all CH3,R1、R2、R3、R4、R5、R6、R7、R8、R9、Y1、Y2、Y3、Y4、Y5、Y6、Y7And Y8Are both hydrogen, then X3Selected from CHD2Or CH2D;
Or a pharmaceutically acceptable salt, crystal form, prodrug, metabolite, hydrate, solvate, stereoisomer, or isotopic derivative thereof.
In one embodiment, Y1、Y2、Y3、Y4、Y5、Y6、Y7And Y8Each independently selected from hydrogen or deuterium. In another specific embodiment, Y1Is hydrogen; in another specific embodiment, Y1Is deuterium; in another specific embodiment, Y2Is hydrogen; in another embodiment, Y2Is deuterium; in another embodiment, Y3Is hydrogen; in another embodiment, Y3Is deuterium; in another specific embodiment, Y4Is hydrogen; in another specific embodiment, Y4Is deuterium; in another embodiment, Y5Is hydrogen; in another embodiment, Y5Is deuterium; in another embodiment, Y6Is hydrogen; in another embodiment, Y6Is deuterium; in another embodiment, Y7Is hydrogen; in another specific embodiment, Y7Is deuterium; in another embodiment, Y8Is hydrogen; in another embodiment, Y8Is deuterium.
Furthermore, R1-R9And X1-X5As for R in the compounds of formula (Ia)1’-R9’And X1-X5As defined.
In another embodiment, the invention relates to compounds of formula (Ib):
Figure BDA0001955701090000151
wherein the content of the first and second substances,
R1and R2Independently selected from H, halogen, -CN, -OH, -OC1-6Alkyl, -NH2、-NHC1-6Alkyl, -N (C)1-6Alkyl radical)2、C1-6Alkyl radical, C1-6Haloalkyl, C2-6Alkenyl radical, C2-6Alkynyl, or R1And R2Form C with the atoms to which they are attached6-10Aryl or 5-10 membered heteroaryl, preferably pyrrolyl;
R3selected from H, halogen, -CN or C1-6An alkoxy group;
R4selected from H, halogen, -CN or C1-6An alkoxy group;
R5selected from the group consisting of:
Figure BDA0001955701090000161
with the proviso that when R3Is C1-6When alkoxy, at least one of the following options holds:
(1)R1and R2Form C with the atoms to which they are attached6-10Aryl or 5-10 membered heteroaryl;
(2)R5is formula (b);
preferably, the first and second electrodes are formed of a metal,
R1and R2Independently selected from H or halogen, or R1And R2Form a pyrrolyl group with the atoms to which they are attached;
R3is selected from H or C1-6An alkoxy group;
R4selected from H, halogen or C1-6An alkoxy group;
R5selected from:
Figure BDA0001955701090000162
provided that when R is1When it is halogen, R3Is other than C1-6An alkoxy group;
or a pharmaceutically acceptable salt, crystal form, prodrug, metabolite, hydrate, solvate, stereoisomer, or isotopic derivative thereof.
In one embodiment, R1And R2Independently selected from H, chlorine, or R1And R2The atoms to which they are attached form a pyrrole ring.
In one embodiment, R1Is chlorine, and R2Is H.
In one embodiment, R1And R2The atoms to which they are attached form a pyrrole ring.
In one embodiment, R3Selected from H, methoxy, ethoxy, isopropoxy or tert-butoxy.
In one embodiment, R3Is H.
In one embodiment, R3Is methoxy.
In one embodiment, R4Selected from H, fluoro, chloro, methoxy, ethoxy, isopropoxy or tert-butoxy.
In one embodiment, R4Is H.
In one embodiment, R4Is fluorine.
In one embodiment, R4Is methoxy.
In one embodiment, R1When it is fluorine or chlorine, R3Is not methoxy, ethoxy isopropoxy or tert-butoxy.
In one embodiment, R1When it is chlorine, R3Is not methoxy.
In another embodiment, the invention relates to compounds of formula (Ic):
Figure BDA0001955701090000171
wherein the content of the first and second substances,
R3is selected from H;
R4selected from H, halogen or C1-6An alkoxy group; preferably, R4Selected from methoxy or fluoro; preferably, R4Is methoxy;
or a pharmaceutically acceptable salt, crystal form, prodrug, metabolite, hydrate, solvate, stereoisomer, or isotopic derivative thereof.
In another embodiment, the invention relates to compounds of formula (Id):
Figure BDA0001955701090000172
wherein the content of the first and second substances,
R3selected from H, halogen, -CN or C1-6An alkoxy group; preferably, R3Is selected from H or C1-6An alkoxy group; preferably, R3Is H; preferably, R3Is C1-6An alkoxy group; preferably, R3Is methoxy;
R4selected from H, halogen, -CN or C1-6An alkoxy group; preferably, R4Selected from H, halogen or C1-6An alkoxy group; preferably, R4Selected from halogen or C1-6An alkoxy group; preferably, R4Selected from methoxy or fluoro; preferably, R4Is H;
or a pharmaceutically acceptable salt, crystal form, prodrug, metabolite, hydrate, solvate, stereoisomer, or isotopic derivative thereof.
In more specific embodiments, R3Is H, and R4Selected from H, halogen or C1-6An alkoxy group; preferably, R3Is H, and R4Selected from halogen or C1-6An alkoxy group; preferably, R3Is H, and R4Selected from methoxy or fluoro.
In more specific embodiments, R4Is H, and R3Is selected from H or C1-6An alkoxy group; preferably, R4Is H, and R3Is C1-6An alkoxy group; preferably, R4Is H, and R3Is methoxy.
In one embodiment, R3Selected from H, methoxy, ethoxy, isopropoxy or tert-butoxy.
In one embodiment, R3Is H.
In one embodiment, R3Is methoxy.
In one embodiment, R4Selected from H, fluoro, chloro, bromo, methoxy, ethoxy, isopropoxy or tert-butoxy.
In one embodiment, R4Is H.
In one embodiment, R4Is fluorine.
In one embodiment, R4Is methoxy.
In a more specific embodiment, the compounds of the present invention are selected from the following compounds, or pharmaceutically acceptable salts, prodrugs, hydrates or solvates, polymorphs, stereoisomers or isotopic variations thereof:
Figure BDA0001955701090000181
Figure BDA0001955701090000191
Figure BDA0001955701090000201
pharmaceutical compositions, formulations and kits
In another aspect, the present invention provides a pharmaceutical composition comprising a compound of the present invention (also referred to as "active ingredient") and a pharmaceutically acceptable excipient. In some embodiments, the pharmaceutical composition comprises an effective amount of an active ingredient. In some embodiments, the pharmaceutical composition comprises a therapeutically effective amount of an active ingredient. In some embodiments, the pharmaceutical composition comprises a prophylactically effective amount of an active ingredient.
Pharmaceutically acceptable excipients for use in the present invention refer to non-toxic carriers, adjuvants or vehicles that do not destroy the pharmacological activity of the compounds formulated therewith. Pharmaceutically acceptable carriers, adjuvants or vehicles that may be used in the compositions of the present invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins (such as human serum albumin), buffer substances (such as phosphates), glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes (such as protamine sulfate), disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, silica gel, magnesium trisilicate, polyvinylpyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and wool fat.
The invention also includes kits (e.g., pharmaceutical packages). The provided kits can include a compound of the invention, an additional therapeutic agent, and first and second containers (e.g., vials, ampoules, bottles, syringes, and/or dispensable packages or other suitable containers) containing the compound of the invention, the additional therapeutic agent. In some embodiments, provided kits may also optionally include a third container containing a pharmaceutically acceptable excipient for diluting or suspending a compound of the invention and/or other therapeutic agent. In some embodiments, the compound of the present invention and the additional therapeutic agent provided in the first container and the second container are combined to form one unit dosage form.
The following formulation examples illustrate representative pharmaceutical compositions that can be prepared according to the present invention. However, the present invention is not limited to the following pharmaceutical compositions.
Exemplary formulation 1-tablet: the compound of the invention in dry powder form may be mixed with the dry gel binder in a weight ratio of about 1: 2. A smaller amount of magnesium stearate was added as a lubricant. The mixture is shaped in a tablet press to 0.3-30mg tablets (each tablet containing 0.1-10mg of active compound).
Exemplary formulation 2-tablet: the compound of the invention in dry powder form may be mixed with the dry gel binder in a weight ratio of about 1: 2. A smaller amount of magnesium stearate was added as a lubricant. The mixture is shaped in a tablet press into 30-90mg tablets (each tablet containing 10-30mg of active compound).
Exemplary formulation 3-tablet: the compound of the invention in dry powder form may be mixed with the dry gel binder in a weight ratio of about 1: 2. A smaller amount of magnesium stearate was added as a lubricant. The mixture is shaped in a tablet press to 90-150mg tablets (each tablet containing 30-50mg of active compound).
Exemplary formulation 4-tablet: the compound of the invention in dry powder form may be mixed with the dry gel binder in a weight ratio of about 1: 2. A smaller amount of magnesium stearate was added as a lubricant. The mixture is shaped in a tablet press into 150-240mg tablets (each containing 50-80mg of active compound).
Exemplary formulation 5-tablet: the compound of the invention in dry powder form may be mixed with the dry gel binder in a weight ratio of about 1: 2. A smaller amount of magnesium stearate was added as a lubricant. The mixture is shaped in a tablet press to 240-270mg tablets (each containing 80-90mg of active compound).
Exemplary formulation 6-tablet: the compound of the invention in dry powder form may be mixed with the dry gel binder in a weight ratio of about 1: 2. A smaller amount of magnesium stearate was added as a lubricant. The mixture is shaped in a tablet press into 270-450mg tablets (each containing 90-150mg of active compound).
Exemplary formulation 7-tablet: the compound of the invention in dry powder form may be mixed with the dry gel binder in a weight ratio of about 1: 2. A smaller amount of magnesium stearate was added as a lubricant. The mixture was shaped into 450-900mg tablets (each tablet containing 150-300mg of active compound) in a tablet press.
Exemplary formulation 8-capsule: the compound of the invention in dry powder form may be mixed with a starch diluent in a weight ratio of about 1: 1. The mixture is filled into 250mg capsules (each containing 125mg of active compound).
Exemplary formulation 9-liquid: the compound of the present invention (125mg) may be mixed with sucrose (1.75g) and xanthan gum (4mg), and the resulting mixture may be blended, passed through a No.10 mesh U.S. sieve, and then mixed with a previously prepared aqueous solution of microcrystalline cellulose and sodium carboxymethylcellulose (11:89, 50 mg). Sodium benzoate (10mg), flavouring and colouring agents were diluted with water and added with stirring. Then, sufficient water may be added to give a total volume of 5 mL.
Exemplary formulation 10-injection: the compounds of the present invention may be dissolved or suspended in aqueous media, which may be injected in buffered sterile saline, to a concentration of about 5 mg/mL.
Administration of drugs
The pharmaceutical compositions provided by the present invention may be administered by a number of routes including, but not limited to: oral, parenteral, inhalation, topical, rectal, nasal, buccal, vaginal, by implant or other modes of administration. For example, parenteral administration as used herein includes subcutaneous administration, intradermal administration, intravenous administration, intramuscular administration, intraarticular administration, intraarterial administration, intrasynovial administration, intrasternal administration, intracerebrospinal administration, intralesional administration, and intracranial injection or infusion techniques.
Typically, an effective amount of a compound provided herein is administered. The amount of compound actually administered can be determined by a physician, as the case may be, including the condition to be treated, the chosen route of administration, the compound actually administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like.
When used to prevent a condition according to the invention, a subject at risk of developing the condition is administered a compound provided herein, typically based on physician's advice and under the supervision of a physician, at a dosage level as described above. Subjects at risk of developing a particular disorder, typically include subjects with a family history of the disorder, or those determined to be particularly susceptible to developing the disorder by genetic testing or screening.
The pharmaceutical compositions provided herein may also be administered chronically ("chronic administration"). By long-term administration is meant administration of the compound or pharmaceutical composition thereof over a long period of time, e.g., 3 months, 6 months, 1 year, 2 years, 3 years, 5 years, etc., or may continue for an indefinite period of time, e.g., for the remainder of the subject's life. In some embodiments, chronic administration is intended to provide a constant level of the compound in the blood over a prolonged period of time, e.g., within the therapeutic window.
Various methods of administration may be used to further deliver the pharmaceutical compositions of the present invention. For example, in some embodiments, the pharmaceutical composition may be administered as a bolus, e.g., in order to increase the concentration of the compound in the blood to an effective level. The bolus dose depends on the targeted systemic level of the active ingredient through the body, e.g., intramuscular or subcutaneous bolus doses result in slow release of the active ingredient, while a bolus delivered directly to the vein (e.g., by IV intravenous drip) can be delivered more rapidly, resulting in a rapid rise in the concentration of the active ingredient in the blood to an effective level. In other embodiments, the pharmaceutical composition may be administered as a continuous infusion, e.g., by IV intravenous drip, to provide a steady state concentration of the active ingredient in the body of the subject. Furthermore, in other embodiments, a bolus dose of the pharmaceutical composition may be administered first, followed by continuous infusion.
Oral compositions may take the form of bulk liquid solutions or suspensions or bulk powders. More generally, however, the compositions are provided in unit dosage form for convenient administration of the precise dosage. The term "unit dosage form" refers to physically discrete units suitable as unitary dosages for human patients and other mammals, each unit containing a predetermined quantity of active material suitable for the purpose of producing the desired therapeutic effect, in association with a suitable pharmaceutical excipient. Typical unit dosage forms include pre-filled, pre-measured ampoules or syringes of the liquid compositions, or pills, tablets, capsules and the like in the case of solid compositions. In such compositions, the compound is typically a minor component (about 0.1 to about 50% by weight, or preferably about 1 to about 40% by weight), with the remainder being various carriers or excipients and processing aids useful in forming the desired form of administration.
For oral doses, a representative regimen is one to five oral doses per day, in particular two to four oral doses, typically three oral doses. Using these dosing modes, each dose provides about 0.01 to about 20mg/kg of a compound of the invention, with preferred doses each providing about 0.1 to about 10mg/kg, especially about 1 to about 5 mg/kg.
In order to provide a blood level similar to, or lower than, the use of the injected dose, a transdermal dose is generally selected in an amount of from about 0.01 to about 20% by weight, preferably from about 0.1 to about 10% by weight, and more preferably from about 0.5 to about 15% by weight.
From about 1 to about 120 hours, especially 24 to 96 hours, the injection dosage level is in the range of about 0.1 mg/kg/hour to at least 10 mg/kg/hour. To obtain sufficient steady state levels, a preload bolus of about 0.1mg/kg to about 10mg/kg or more may also be administered. For human patients of 40 to 80kg, the maximum total dose cannot exceed about 2 g/day.
Liquid forms suitable for oral administration may include suitable aqueous or nonaqueous carriers, as well as buffers, suspending and dispersing agents, coloring and flavoring agents, and the like. Solid forms may include, for example, any of the following components, or compounds with similar properties: a binder, for example, microcrystalline cellulose, gum tragacanth or gelatin; excipients, for example, starch or lactose, disintegrants, for example, alginic acid, Primogel or corn starch; lubricants, for example, magnesium stearate; glidants, e.g., colloidal silicon dioxide; sweetening agents, for example, sucrose or saccharin; or a flavoring agent, for example, peppermint, methyl salicylate, or orange flavoring.
Injectable compositions are typically based on sterile saline or phosphate buffered saline for injection, or other injectable excipients known in the art. As previously mentioned, in such compositions, the active compound is typically a minor component, often about 0.05 to 10% by weight, with the remainder being injectable excipients and the like.
Transdermal compositions are typically formulated as topical ointments or creams containing the active ingredient. When formulated as an ointment, the active ingredient is typically combined with a paraffinic or water-miscible ointment base. Alternatively, the active ingredient may be formulated as a cream with a cream base, for example of the oil-in-water type. Such transdermal formulations are well known in the art and typically include other components for enhancing stable skin penetration of the active ingredient or formulation. All such known transdermal formulations and compositions are included within the scope of the present invention.
The compounds of the invention may also be administered by transdermal means. Thus, transdermal administration can be achieved using a reservoir (reservoir) or porous membrane type, or a patch of various solid matrices.
The above components of the compositions for oral, injection or topical administration are merely representative. Other materials and processing techniques are described in Remington's Pharmaceutical Sciences,17thedition,1985, Mack Publishing Company, Easton, Pennsylvania, section 8, which is incorporated herein by reference.
The compounds of the present invention may also be administered in sustained release form, or from a sustained release delivery system. A description of representative sustained release materials can be found in Remington's Pharmaceutical Sciences.
The invention also relates to pharmaceutically acceptable formulations of the compounds of the invention. In one embodiment, the formulation comprises water. In another embodiment, the formulation comprises a cyclodextrin derivative. The most common cyclodextrins are α -, β -and γ -cyclodextrins consisting of 6, 7 and 8 α -1, 4-linked glucose units, respectively, which optionally include one or more substituents on the linked sugar moiety, including but not limited to: methylated, hydroxyalkylated, acylated and sulfoalkyl ether substitution. In some embodiments, the cyclodextrin is sulfoalkyl ether β -cyclodextrin, e.g., sulfobutyl ether β -cyclodextrin, also known as Captisol. See, e.g., U.S.5,376,645. In some embodiments, the formulation includes hexapropyl- β -cyclodextrin (e.g., 10-50% in water).
Method of treatment
The present invention provides for the administration of a compound of the present invention, or a pharmaceutically acceptable salt, stereoisomer, solvate, hydrate, crystalline form, prodrug, or isotopic derivative thereof, to a subject in need thereof, or the administration of a pharmaceutical composition described herein for the treatment of cancer. In some embodiments, the cancer is an ALK-driven cancer. In some embodiments, the cancer is non-small cell lung cancer.
A "therapeutically effective amount" is an amount effective to detect killing or inhibiting the growth or spread of cancer cells; the size or number of weights; or other measure of the level, stage, progression or severity of the cancer. The precise amount required may vary from subject to subject, depending on the species, age, and general health of the subject, the severity of the disease, the particular anticancer agent, its mode of administration, combination therapy with other therapies, and the like.
Disclosed herein are compounds having biological properties that make them targets for the treatment or modulation of diseases that may involve kinases, symptoms of such diseases, or the effects of other physiological events mediated by kinases. For example, various compounds disclosed herein can inhibit the tyrosine kinase activity of ALK, fak and c-met, particularly tyrosine kinases believed to mediate the growth, development and/or metastasis of cancer. Various compounds as disclosed herein were also found to have potent in vitro activity against cancer cell lines. Thus, such compounds are useful for the target of treating cancer (including solid tumors as well as lymphomas and including cancers that are resistant to other therapies).
In some embodiments, the cancer is an ALK-driven cancer. In some embodiments, the cancer is non-small cell lung cancer (NSCLC). In some embodiments, the cancer is an ALK-positive NSCLC. In some embodiments, the cancer is locally advanced or metastatic ALK-positive NSCLC. In some embodiments, the cancer/patient has been previously treated with crizotinib or another tyrosine kinase inhibitor. In some embodiments, the cancer/patient has not been previously treated with an ALK inhibitor.
Such cancers include, but are not limited to, breast cancer, non-small cell lung cancer, neural tumors (such as glioblastoma and neuroblastoma); esophageal cancer, soft tissue cancer (such as rhabdomyosarcoma, etc.); various forms of lymphoma, such as non-hodgkin's lymphoma (NHL), known as Anaplastic Large Cell Lymphoma (ALCL); various forms of leukemia; and include cancers mediated by ALK or c-met.
Anaplastic Lymphoma Kinase (ALK) is a transmembrane receptor tyrosine kinase, which belongs to the insulin receptor subfamily. ALK Receptor Tyrosine Kinases (RTKs) were originally identified as being involved in a human non-hodgkin's lymphoma subtype known as Anaplastic Large Cell Lymphoma (ALCL). ALK generally has a restricted distribution in mammalian cells, and is found at significant levels only in the nervous system during embryonic development, suggesting a role for ALK in brain development.
In addition to its role in normal development, expression of full-length normal ALK has been detected in cell lines derived from a variety of tumors, such as neuroglioblastoma, neuroectodermal tumors and glioblastoma, and breast cancer and melanoma lines.
Like other RTKs, translocation affects the ALK gene, resulting in expression of a protocell fusion kinase, the most common of which is NPM-ALK. For example, approximately sixty percent of Anaplastic Large Cell Lymphomas (ALCLs) are associated with chromosomal mutations that produce fusion proteins consisting of the intracellular domain of nucleolar phosphoprotein (NMP) ALK. This mutant protein, NPM-ALK, has a constitutively active tyrosine kinase domain that is responsible for its oncogenic properties by activating downstream effectors. Experimental data have demonstrated that abnormal expression of constitutively active ALK is directly involved in the pathogenesis of ALCL and that such inhibition of ALK can significantly impede the growth of ALK-positive lymphoma cells. Constitutively activated chimeric ALK has been demonstrated to be present in about 60% of Inflammatory Myofibroblastic Tumors (IMT), a slow-growing sarcoma that mainly affects children and young adults. Furthermore, the current report has described the occurrence of the variant ALK fusion TPM4-ALK in the context of Squamous Cell Carcinoma (SCC) of the esophagus. ALK is therefore one of the few examples of RTKs involved in neoplasia in both nonhematopoietic and hematopoietic malignancies. Recently, inversion within chromosome 2p has been shown to result in the formation of a fusion gene comprising a portion of the 4(EML4) gene like the echinoderm microtubule-associated protein and the Anaplastic Lymphoma Kinase (ALK) gene in non-small cell lung cancer cells.
In some embodiments, ALK inhibitors may create a long lasting cure when used as a single therapeutic agent or in combination with current chemotherapy in ALCL, IMT, proliferative disorders, glioblastoma, and other possible solid tumors cited herein, or may be used as a single therapeutic agent in the maintenance of preventing relapse in patients in need of such treatment.
The compounds as disclosed herein may be administered as part of a treatment regimen in which the compound is the sole active agent, or in combination with one or more other therapeutic agents as part of a combination therapy. When administered as one component of a combination therapy, the therapeutic agents being administered can be formulated to be administered simultaneously or sequentially at different time points (e.g., within 72 hours, 48 hours, or 24 hours of each other) as separate compositions, or the therapeutic agents can be formulated together as a single pharmaceutical composition and administered simultaneously.
Thus, administration of the compounds of the invention may be combined with at least one additional treatment known to those skilled in the art to prevent or treat cancer (such as radiation therapy or cytostatics, cytotoxic agents, other anti-cancer agents and other drugs) to alleviate the symptoms of cancer or any drug side effects. Non-limiting examples of additional therapeutic agents include agents suitable for immunotherapy (such as, for example, PD-1 or PDL-1 inhibitors), anti-angiogenesis agents (such as, for example, bevacizumab), and/or chemotherapy agents.
If formulated as a fixed dose, such combination products employ a compound as disclosed herein within an acceptable dosage range. When a combined preparation is appropriate, a compound as disclosed herein may be administered sequentially with other anti-cancer or cytotoxic agents. The compounds as disclosed herein may be administered prior to, concurrently with, or after the administration of the other anti-cancer or cytotoxic agent.
Currently, standard treatment of primary tumors consists of surgical resection followed by radiation or chemotherapy, as appropriate, and is usually administered intravenously. The usual chemotherapeutic regimen consists of a DNA alkylating agent, a DNA intercalating agent, a CDK inhibitor or a microtubule poison. The chemotherapy dose used is just below the maximum tolerated dose, and thus dose-limiting toxicities typically include nausea, vomiting, diarrhea, hair loss, neutropenia, and the like.
There are a large number of antineoplastic agents available for commercial use, clinical evaluation and preclinical development, which can be selected for the treatment of cancer by combination drug chemotherapy. And there are several major classes of such antineoplastic agents, namely antibiotic-type agents, alkylating agents, antimetabolite agents, anti-hormone agents, immunological agents, interferon-type agents, and a class of miscellaneous agents.
Examples of other therapeutic agents include, but are not limited to, one or more of the following: anti-cancer alkylating or intercalating agents (e.g., nitrogen mustards, chlorambucil, cyclophosphamide, melphalan, and ifosfamide); antimetabolites (e.g., methotrexate); purine antituberculosis agents or pyrimidine antagonists (e.g., 5-fluorouracil, cytarabine and gemcitabine); spindle inhibitors (e.g., vinblastine, vincristine, vinorelbine paclitaxel); podophyllotoxins (e.g., etoposide, irinotecan, topotecan); antibiotics (such as doxorubicin, bleomycin and mitomycin); nitrosoureas (e.g., carmustine, lomustine); inorganic ions (such as cisplatin, carboplatin, oxaliplatin, or oxisplatin); enzymes (e.g., asparaginase); hormones (such as tamoxifen, leuprolide, flutamide, or megestrol); mTOR inhibitors (e.g., sirolimus (rapamycin), temsirolimus (CCI779), everolimus (RAD001), AP23573, or other compounds disclosed in U.S. patent 7091213); proteasome inhibitors (e.g., velcade, other proteasome inhibitors (e.g., Src, Bcr/Abl, kdr, flt3, aurora-2, glycogen synthase kinase 3(GSK-3), EGFR kinases (e.g., Iressa, Tarceva, etc.), VEGF-R kinases, PDGF-R kinases, etc.), antibodies, soluble receptors or other receptor antagonists against receptors or hormones involved in cancer (including receptors such as EGFR, ErbB2, VEGFR, PDGFR, and IGF-R; and drugs such as Herceptin, avastin, erbitux, etc.), etc. examples of other therapeutic agents include, but are not limited to, purine, alemtuzmab, hexamethamine, amifostine, nartrozole, antibodies against prostate specific membrane antigens (e.g., MLN-591, MLN591RL, and MLN2704), diarsenia, bexatin, milbevacycin, leucasabine, capecitabine, Waadeps, Wasabia, and so forth Chlorambucil, cisplatin-epinephrine gel, cladribine, cytarabine liposome, daunorubicin, rubicin, dexrazoxane, docetaxel, doxorubicin, Elliott's B solution, epirubicin, estramustine phosphate, etoposide phosphate, exemestane, fludarabine, 5-FU, fulvestrant, gemcitabine, gemumab-ozomicin, goserelin acetate, hydroxyurea, idarubicin, ifosfamide, imatinib mesylate, irinotecan (or other topoisomerase inhibitors including antibodies such as MLN576(XR11576)), letrozole, calcium folinate, levoimidazole, liposomal daunorubicin, melphalan, L-PAM, mesna, methotrexate, methoxsalen, mitomycin C, mitoxantrone, MLN518 or MLN (or other t-3 receptor tyrosine kinases, or other t-3 receptor tyrosine kinases, Inhibitors of PDFG-R or C-kit), itoxantrone, paclitaxel, pegase, pentastatin, rituximab, talc, tamoxifen, temozolamide, teniposide, VM-26, topotecan, toremifene, 2C4 (or other antibodies that interfere with HER 2-mediated signaling), tretinoin, ATRA, valrubicin, vinorelbine, or disodium aminodiphosphate, disodium zoledronate, or other diphosphates.
Examples
The invention will be further illustrated with reference to the following specific examples. It should be understood that these examples are for illustrative purposes only and are not intended to limit the scope of the present invention. The experimental procedures, in which specific conditions are not noted in the following examples, are generally carried out under conventional conditions or conditions recommended by the manufacturers. Parts and percentages are parts and percentages by weight unless otherwise indicated.
In general, in the preparative schemes, each reaction is usually carried out in an inert solvent at a temperature ranging from room temperature to reflux temperature (e.g., from 0 ℃ to 100 ℃, preferably from 0 ℃ to 80 ℃). The reaction time is usually 0.1 to 60 hours, preferably 0.5 to 24 hours.
3Example 1(2- ((2- ((4- (4- (di (methyl-d) amino) piperidin-1-yl) -2-methoxyphenyl) amino) - Preparation of 5-chloropyrimidin-4-yl) amino) phenyl) dimethylphosphine oxide (Compound T-1)
Figure BDA0001955701090000291
The synthesis was carried out using the following route:
Figure BDA0001955701090000292
step 1 Synthesis of Compound 3
To a 50mL single neck flask equipped with magnetic stirring was added compound 1(1.71g, 10mmol) and acetonitrile (30mL) in sequence, the mixture was stirred, compound 2(2.6g, 13mmol) and potassium carbonate (2.07g, 15mmol) were added, the reaction mixture was warmed to 70 ℃ under nitrogen and the reaction was stirred for 3 hours. After cooling to room temperature, the solvent was evaporated under reduced pressure, water (60mL) was added to precipitate a large amount of yellow solid, which was filtered, washed with water (10mL) and dried to obtain 2.5g of yellow solid with a yield of 71.2%. LC-MS (APCI) M/z 352.2(M +1)+.
Step 2 Synthesis of Compound 4
To a 250mL single neck flask equipped with magnetic stirring was added compound 3(2.5g, 7.12mmol) and ethyl acetate (10mL), the solution was stirred, a solution of hydrogen chloride in isopropanol (30mL, 5M) was added dropwise, and the reaction was stirred at room temperature under nitrogen for 2 hours. A large amount of white solid was precipitated, ethyl acetate (100mL) was added to dilute the reaction mixture, the mixture was filtered, ethyl acetate was washed (20mL), methylene chloride (100mL) was added to the cake, a methanol solution (7M) of ammonia was added with stirring to adjust the pH to 12, the mixture was stirred for 10 minutes, the generated ammonium chloride was filtered off, and the filtrate was concentrated to give 1.5g of a yellow solid with a yield of 83.9%. LC-MS (APCI): M/z 252.2(M +1)+.
Step 3 Synthesis of Compound 5
Add Compound 4(0.25g, 1.0mmol) and MeOD (5mL) sequentially to a 50mL single-neck flask equipped with magnetic stirring at room temperature, dissolve with stirring, cool in an ice-water bath, and add deuterated Formaldehyde in heavy water (0.48g, 3.0mmol, 20% w/w) and CH dropwise3COOD (1 drop), stirring for 10min, adding NaBD3CN (0.20g, 3.0mmol), the ice bath was removed and the reaction stirred at room temperature under nitrogen for 2 hours. The reaction was quenched by the addition of triethylamine (0.5mL), the solvent was removed by concentration under reduced pressure, and the residue was passed through a silica gel column to give 0.24g of a yellow solid in 84.2% yield. LC-MS (APCI): M/z 286.1(M +1)+.
Step 4 Synthesis of Compound 6
To a 50mL single-neck flask equipped with magnetic stirring was added compound 5(0.24g,0.84mmol) and methanol (5mL) in order, the mixture was stirred to dissolve, Pd/C (24mg, 10%) was added, vacuum was applied and hydrogen replacedAnd the reaction is stirred overnight under hydrogen atmosphere. The hydrogen balloon was removed, dichloromethane (20mL) was added for dilution, the catalyst was filtered off, and the filtrate was concentrated under reduced pressure to dryness to give 0.2g of a brown oil in 93.4% yield. LC-MS (APCI) M/z 256.1(M +1)+.
Step 5 Synthesis of Compound T-1
A25 mL single neck flask equipped with a magnetic stirring condenser was charged with Compound 6(200mg, 0.78mmol), Compound 7(247mg, 0.78mmol) and ethylene glycol monomethyl ether (5mL), stirred to dissolve, added dropwise with hydrogen chloride isopropanol solution (1.17mmol, 0.23mL, 5M), warmed to 120 ℃ under nitrogen and stirred overnight. After cooling to room temperature, water (10mL) and saturated sodium bicarbonate (5mL) were added, dichloromethane was extracted (15mLx3), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, concentrated, and the residue was passed through a silica gel column to give 240mg of a white solid with a yield of 44.9%. LC-MS (APCI): M/z 535.2(M +1)+.1H NMR(300MHz,CDCl3)δ10.84(s,1H),8.64-8.60(m,1H),8.15(d,J=9.0Hz,1H),8.10(s,1H),7.51(t,J=6.0Hz,1H),7.34-7.27(m,2H),7.17-7.12(m,1H),6.53(d,J=2.4Hz,1H),6.48(dd,J=9.0Hz,J=2.4Hz,1H),3.88(s,3H),3.72(d,J=12.0Hz,2H),3.13-3.10(m,1H),2.77(t,J=12.0Hz,2H),2.30-2.24(m,2H),2.00-1.91(m,2H),1.87(s,3H),1.83(s,3H).
4Example 2(2- ((5-chloro-2- ((4- (4- (dimethylamino) piperidin-1-yl-3, 3,5,5-d) -2-methoxy) Preparation of phenyl) amino) pyrimidin-4-yl) amino) phenyl) dimethylphosphine oxide (compound T-2)
Figure BDA0001955701090000311
The synthesis was carried out using the following route:
Figure BDA0001955701090000312
step 1 Synthesis of Compound 8
Sequentially stirring with magnetic forceA stirred 50mL one-neck flask was charged with Compound 1(1.71g, 10mmol) and acetonitrile (20mL), stirred to dissolve, 4-piperidone hydrochloride monohydrate (2.0g, 13mmol) and DIPEA (N, N-diisopropylethylamine, 3.90g, 30mmol) were added, the reaction mixture was warmed to 80 ℃ under nitrogen and stirred overnight. After cooling to room temperature, the solvent was evaporated under reduced pressure, water (60mL) was added to precipitate a large amount of yellow solid, which was filtered, washed with a large amount of water (100mL) and dried to obtain 2.1g of yellow solid in 84.0% yield. LC-MS (APCI) M/z 251.2(M +1)+.1H NMR(300MHz,DMSO-d6)δ7.93(d,J=9.6Hz,1H),6.62(dd,J=9.6Hz,J=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).
Step 2 Synthesis of Compound 9
To a 50mL three-necked flask equipped with magnetic stirring, compound 8(2.1g,8.4mmol) and deuterated chloroform (100mL) were added sequentially, the mixture was stirred to dissolve, 1,5, 7-triazabicyclo (4,4,0) dec-5-ene (100mg) was added, and the reaction was stirred at room temperature under nitrogen overnight. The reaction mixture was washed twice with dilute hydrochloric acid (10mL, 1M), water (20mL), saturated sodium bicarbonate (10mL), saturated brine (10mL), dried over anhydrous sodium sulfate, filtered, and concentrated to dryness to give 1.9g of a yellow solid with a yield of 90.5%. LC-MS (APCI) M/z 255.2(M +1)+.1H NMR(400MHz,CDCl3)δ8.03(d,J=9.6Hz,1H),6.45(dd,J=9.6Hz,J=2.4Hz,1H),6.35(d,J=2.4Hz,1H),3.97(s,3H),3.80(s,4H)。
Step 3 Synthesis of Compound 10
Compound 9(510mg, 2.0mmol) and deuterated chloroform (5mL) were added sequentially to a 50mL two-necked flask equipped with magnetic stirring at room temperature, the mixture was stirred to dissolve, a solution of dimethylamine in tetrahydrofuran (2mL, 4.0mmol, 2M) was added, tetraisopropyl titanate (2.82g, 10mmol) was added under nitrogen, and the reaction was stirred at room temperature overnight. In an ice-water bath, anhydrous ethanol (20mL) was added to dilute the reaction mixture, sodium cyanoborohydride (410mg, 5.0mmol) was added, and the reaction mixture was stirred at room temperature under nitrogen atmosphere for 3 hours. Adding water (50mL) and ethyl acetate (60mL), stirring for 10min, filtering off insoluble solids, separating the organic layer, extracting with ethyl acetate (40mLx2), combining the organic phases, drying over anhydrous sodium sulfate, filtering, concentrating and passing through a silica gel column400mg of yellow solid are obtained with a yield of 70.7%. LC-MS (APCI): M/z 284.2(M +1)+.1H NMR(500MHz,CDCl3)δ8.00(d,J=9.0Hz,1H),6.42(dd,J=9.0Hz,J=3.0Hz,1H),6.32(d,J=3.0Hz,1H),3.95(s,3H),3.93(d,J=13.0Hz,2H),2.97(d,J=13.0Hz,2H),2.43(s,1H),2.35(s,6H).
Step 4 Synthesis of Compound 11
To a 50mL single neck flask equipped with magnetic stirring was added compound 10(0.24g,0.84mmol) and methanol (5mL) in that order, stirred to dissolve, Pd/C (24mg, 10%) was added, evacuated and replaced with hydrogen three times, and the reaction was stirred under a hydrogen balloon atmosphere overnight. The hydrogen balloon was removed, dichloromethane (20mL) was added for dilution, the catalyst was filtered off, and the filtrate was concentrated under reduced pressure to dryness to give 0.2g of a brown oil in 93.4% yield. LC-MS (APCI) 254.1(M +1) M/z+.
Step 5 Synthesis of Compound T-2
A25 mL single neck flask equipped with a magnetic stirring condenser was charged with compound 11(200mg, 0.78mmol), compound 7(247mg, 0.78mmol) and ethylene glycol monomethyl ether (5mL), stirred to dissolve, added dropwise with hydrogen chloride isopropanol solution (1.17mmol, 0.23mL, 5M), warmed to 120 ℃ under nitrogen and stirred overnight. After cooling to room temperature, water (10mL) and saturated sodium bicarbonate (5mL) were added, dichloromethane was extracted (15mLx3), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, concentrated, and the residue was passed through a silica gel column to give 240mg of a white solid with a yield of 44.9%. LC-MS (APCI): M/z 533.2(M +1)+.1H NMR(300MHz,CDCl3)δ10.81(s,1H),8.65-8.61(m,1H),8.12-8.09(m,2H),7.53(t,J=6.0Hz,1H),7.33-7.25(m,1H),7.18-7.13(m,1H),6.56(d,J=2.4Hz,1H),6.52-6.48(m,1H),3.87(s,3H),3.64(d,J=12.6Hz,2H),2.68(d,J=12.6Hz,2H),2.41(s,7H),1.86(s,3H),1.82(s,3H).
3 5Example 3(2- ((2- ((4- (4- (di (methyl-d) amino) piperidin-1-yl-3, 3,4,5,5-d) -2-methoxy) Preparation of phenyl) amino) -5-chloropyrimidin-4-yl) amino) phenyl) dimethylphosphine oxide (Compound T-3)
Figure BDA0001955701090000331
The synthesis was carried out using the following route:
Figure BDA0001955701090000341
step 1 Synthesis of Compound 12
Add Compound 9(255mg, 1.0mmol) and deuterated chloroform (3mL) sequentially at room temperature to a 50mL two-necked flask equipped with magnetic stirring, stir to dissolve clear, add NH (CD)3)2To a solution of tetrahydrofuran (1mL, 2.0mmol, 2M), tetraisopropyl titanate (1.41g, 5mmol) was added under nitrogen, and the reaction was stirred at room temperature overnight. Under ice-water bath, absolute ethyl alcohol (10mL) is added to dilute the reaction solution, and NaBD is added3CN (205mg, 2.5mmol), after addition, the reaction was stirred at room temperature under nitrogen for 3 hours. Water (25mL) and ethyl acetate (30mL) were added, stirred for 10min, the insoluble solid was filtered off, the organic layer was separated, extracted with ethyl acetate (20mLx2), the organic phases combined, dried over anhydrous sodium sulfate, filtered, concentrated and passed through a silica gel column to give 200mg of a yellow solid in 70.7% yield. LC-MS (APCI): M/z 291.2(M +1)+.1H NMR(300MHz,CDCl3)δ8.05(d,J=9.0Hz,1H),6.45(dd,J=9.0Hz,J=3.5Hz,1H),6.34(d,J=3.5Hz,1H),3.98(s,3H),2.63(s,4H).
Step 2 Synthesis of Compound 13
To a 50mL single neck flask equipped with magnetic stirring was added compound 12(0.2g,0.71mmol) and methanol (5mL) in that order, the solution was stirred, Pd/C (20mg, 10%) was added, vacuum was applied and hydrogen replaced three times, and the reaction was stirred under hydrogen balloon overnight. The hydrogen balloon was removed, dichloromethane (20mL) was added for dilution, the catalyst was filtered off, and the filtrate was concentrated under reduced pressure to dryness to give 0.16g of a brown oil in 86.7% yield. LC-MS (APCI): M/z 261.1(M +1)+.
Step 3 Synthesis of Compound T-3
A25 mL single-neck flask equipped with a magnetic stirring condenser was charged with Compound 13(160mg, 0.61mmol), Compound 7(192mg, 0.61mmol) and ethylene glycol monomethyl ether (4mL), and the mixture was stirred to dissolveTo the mixture was added dropwise a solution of hydrogen chloride in isopropanol (0.91mmol, 0.18mL, 5M), warmed to 120 ℃ under nitrogen and stirred overnight. After cooling to room temperature, water (10mL) and saturated sodium bicarbonate (5mL) were added, dichloromethane was extracted (15mLx3), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, concentrated, and the residue was passed through a silica gel column to give 200mg of a white solid with a yield of 60.7%. LC-MS (APCI): M/z 540.2(M +1)+.1H NMR(300MHz,CDCl3)δ10.81(s,1H),8.65-8.61(m,1H),8.12-8.09(m,2H),7.53(t,J=6.0Hz,1H),7.33-7.25(m,1H),7.18-7.13(m,1H),6.56(d,J=2.4Hz,1H),6.52-6.48(m,1H),3.87(s,3H),3.64(d,J=12.6Hz,2H),2.68(d,J=12.6Hz,2H),1.86(s,3H),1.82(s,3H).
4Example 4(2- ((5-chloro-2- ((4- (4- (dimethylamino) piperidin-1-yl-2, 2,6,6-d) -2-methoxy) Preparation of phenyl) amino) pyrimidin-4-yl) amino) phenyl) dimethylphosphine oxide (compound T-4)
Figure BDA0001955701090000351
The synthesis was carried out using the following route:
Figure BDA0001955701090000361
step 1 Synthesis of Compound 16
To a 50mL single neck flask equipped with magnetic stirring was added benzylamine (compound 14, 1.07g, 10mmol) and DCOD in heavy water (4.0g, 25mmol, 20% w/w), and the reaction was stirred at room temperature under nitrogen for 1 hour. Then, compound 15(1.25g, 11mmol) was added dropwise thereto, and after completion of the addition, the temperature was raised to 40 ℃ and the reaction was stirred overnight with heat preservation. The reaction was quenched by addition of water (15mL), adjusted to pH 9 by addition of solid potassium carbonate, extracted with ethyl acetate (20mLx3), the organic phases combined, dried over anhydrous sodium sulfate, filtered, concentrated and passed through a silica gel column to give 1.2g of a colorless oil in 53.8% yield. LC-MS (APCI): M/z 224.1(M +1)+
Step 2 Synthesis of Compound 17
To a 50mL single neck flask equipped with magnetic stirring was added compound 16(1.2g, 5.38mmol) and methanol (10mL), Pd (OH) was added2C (300mg, 5%), evacuated and replaced with hydrogen 3 times, and the reaction was stirred under a hydrogen balloon at room temperature overnight. The hydrogen balloon was removed, the reaction was diluted with dichloromethane (30mL), the insoluble catalyst was filtered off, the filtrate was washed with dichloromethane (5mL), and the filtrate was concentrated to dryness to give 0.53g of a colorless oil in 92.9% yield. Was used directly in the next step.
Step 3 Synthesis of Compound 18
To a 50mL single neck flask equipped with magnetic stirring was added compound 17(0.53g, 5.0mmol) and acetonitrile (30mL) in sequence, the mixture was stirred, compound 1(0.85g, 5.0mmol) and potassium carbonate (1.1g, 7.5mmol) were added, the reaction mixture was warmed to 70 ℃ under nitrogen and the reaction was stirred for 3 hours with incubation. After cooling to room temperature, the solvent was evaporated under reduced pressure, water (60mL) was added to precipitate a large amount of yellow solid, which was filtered, washed with water (10mL) and dried to obtain 1.0g of yellow solid in 78.1% yield. LC-MS (APCI) M/z 257.2(M +1)+.
Step 4 Synthesis of Compound 19
Compound 18(1.0g, 3.92mmol) and dichloromethane (30mL) were added sequentially to a 50mL single neck flask equipped with magnetic stirring, the solution was stirred, molecular sieves (2.0g) were added, N-methyl-N-morpholine oxide (NMO, 700mg, 6.0mmol) and tetrapropyl ammonium homoruthenate (TPAP, 70mg, 0.2mmol) were added slowly dropwise in an ice water bath, after the addition was complete, the reaction was stirred for 20 minutes at nitrogen atmosphere with incubation, the ice bath was removed, and the reaction was stirred at room temperature for 2 hours. The organic solvent was evaporated under reduced pressure, concentrated and subjected to silica gel column chromatography to obtain 950mg of a white solid with a yield of 93.1%. LC-MS (APCI) M/z 255.1(M +1)+1H NMR(400MHz,CDCl3)δ8.03(d,J=9.6Hz,1H),6.45(dd,J=9.6Hz,J=2.4Hz,1H),6.35(d,J=2.4Hz,1H),3.97(s,3H),2.82(s,4H)。
Step 5 Synthesis of Compound 20
Add compound 19(510mg, 2.0mmol) and dichloromethane (5mL) sequentially to a 50mL two-necked flask equipped with magnetic stirring at room temperature, stir to dissolve, add dimethylamine in tetrahydrofuran (2mL, 4.0mmol, 2M), add tetraiso titanate under nitrogenPropyl ester (2.82g, 10mmol), the reaction was stirred at room temperature overnight. In an ice-water bath, anhydrous ethanol (20mL) was added to dilute the reaction solution, sodium cyanoborohydride (410mg, 5.0mmol) was added, and after completion of the addition, the reaction mixture was stirred at room temperature under a nitrogen atmosphere for 3 hours. Water (50mL) and ethyl acetate (60mL) were added, stirred for 10min, the insoluble solid was filtered off, the organic layer was separated, extracted with ethyl acetate (40mLx2), the organic phases combined, dried over anhydrous sodium sulfate, filtered, concentrated and passed through a silica gel column to give 400mg of a yellow solid in 70.7% yield. LC-MS (APCI): M/z 284.2(M +1)+.1H NMR(500MHz,CDCl3)δ8.00(d,J=9.0Hz,1H),6.42(dd,J=9.0Hz,J=3.0Hz,1H),6.32(d,J=3.0Hz,1H),3.95(s,3H),2.47-2.43(m,1H),2.35(s,6H),1.75-1.72(m,2H),1.43-1.38(m,2H).
Step 6 Synthesis of Compound 21
To a 50mL single neck flask equipped with magnetic stirring was added compound 20(0.24g,0.84mmol) and methanol (5mL) in that order, the solution was stirred, Pd/C (24mg, 10%) was added, vacuum was applied and hydrogen replaced three times, and the reaction was stirred under hydrogen balloon overnight. The hydrogen balloon was removed, dichloromethane (20mL) was added for dilution, the catalyst was filtered off, and the filtrate was concentrated under reduced pressure to dryness to give a brown oil 0.2g, 93.4% yield. LC-MS (APCI) 254.1(M +1) M/z+.
Step 7 Synthesis of Compound T-4
A25 mL single neck flask equipped with a magnetic stirring condenser was charged with compound 21(200mg, 0.78mmol), compound 7(247mg, 0.78mmol) and ethylene glycol monomethyl ether (5mL), stirred to dissolve, added dropwise with hydrogen chloride isopropanol solution (1.17mmol, 0.23mL, 5M), warmed to 120 ℃ under nitrogen and stirred overnight. After cooling to room temperature, water (10mL) and saturated sodium bicarbonate (5mL) were added, dichloromethane was extracted (15mLx3), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, concentrated, and the residue was passed through a silica gel column to give 240mg of a white solid in 44.9% yield. LC-MS (APCI): M/z 535.2(M +1)+.1H NMR(300MHz,CDCl3)δ10.84(s,1H),8.64-8.60(m,1H),8.15(d,J=9.0Hz,1H),8.10(s,1H),7.51(t,J=6.0Hz,1H),7.34-7.27(m,2H),7.17-7.12(m,1H),6.53(d,J=2.4Hz,1H),6.48(dd,J=9.0Hz,J=2.4Hz,1H),3.88(s,3H),3.13-3.10(m,1H),2.30-2.24(m,2H),2.00-1.91(m,2H),1.87(s,3H),1.83(s,3H).
Example 5(2- ((5-chloro-2- ((4- (4- (dimethylamino) piperidin-1-yl) -2-methoxyphenyl) amino) pyrimidine 3Preparation of pyridin-4-yl) amino) phenyl) bis (methyl-d) phosphine oxide (Compound T-5)
Figure BDA0001955701090000381
The synthesis was carried out using the following route:
Figure BDA0001955701090000382
Figure BDA0001955701090000391
step 1 Synthesis of Compound 23
Compound 1(887mg, 4.72mmol) and acetonitrile (20mL) are added sequentially to a 50mL single neck flask equipped with magnetic stirring, the supernatant is stirred, dihydrochloride of compound 22 (1.1g, 5.66mmol) and potassium carbonate (2.2g, 15.57mmol) are added, the reaction mixture is warmed to 60 ℃ under nitrogen and the reaction is stirred for 3 hours with warming. After cooling to room temperature, the solvent was evaporated under reduced pressure, water (60mL) was added to precipitate a large amount of yellow solid, which was filtered, washed with water (10mL) and dried to obtain 1.0g of yellow solid with a yield of 75.9%. LC-MS (APCI) 280.2(M +1) M/z+.
Step 2 Synthesis of Compound 24
To a 50mL single neck flask equipped with magnetic stirring was added compound 23(1.0g,3.58mmol) and methanol (20mL) in that order, the mixture was stirred to dissolve, Pd/C (100mg, 10%) was added, vacuum was applied and hydrogen replaced three times, and the reaction was stirred under a hydrogen balloon atmosphere overnight. The hydrogen balloon was removed, dichloromethane (50mL) was added for dilution, the catalyst was filtered off, and the filtrate was concentrated under reduced pressure to dryness to give 0.82g of a brown oil in 89.4% yield. LC-MS (APCI): M/z 250.1(M +1)+.
Step 3 Synthesis of Compound 27
A100 mL three-necked flask equipped with a magnetic stir and condenser was charged with magnesium powder (1.80g, 74.87mmol), evacuated and purged with nitrogen 3 times, and added with diethyl ether (30mL) and CD under nitrogen3I (10.0g, 68.96mmol) was added dropwise and the reaction was refluxed at elevated temperature for 2 hours. After cooling to 0 ℃ and slowly adding dropwise a solution of dimethyl phosphite (2.5g, 23mmol) in ether (10mL), the reaction was stirred at room temperature for 1 hour. Potassium carbonate cold water (9.6g, 10mL) was added to quench the reaction, the resulting solid was filtered off, the filter cake was washed with ethanol (20mL), concentrated under reduced pressure, and the precipitated solid was filtered off to give 840mg of an anhydrous oil in 13.3% yield.
Step 4 Synthesis of Compound 29
To a 50mL single neck flask equipped with magnetic stirring was added compound 27(840mg, 10.04mmol) and DMF (5mL), the mixture was stirred to dissolve, and compound 28(2.18g, 10mmol), Pd (OAc) were added2(56mg, 0.25mmol) and BINAP (1,1 '-binaphthyl-2, 2' -bisdiphenylphosphine, 311mg, 0.5mmol), evacuated and replaced with nitrogen three times, warmed to 150 ℃ under nitrogen atmosphere and stirred for 3 hours with heat preservation. Cooled to room temperature, concentrated under reduced pressure, and the residue was passed through a silica gel column to give 1.2g of a white solid in a yield of 68.5%. LC-MS (APCI): M/z 176.1(M +1)+.
Step 5 Synthesis of Compound 31
To a 50mL single neck flask equipped with magnetic stirring was added compound 29(1.2g, 6.90mmol) and acetonitrile (10mL), the solution was stirred, compound 30(1.49g, 8.23mmol) and potassium carbonate (1.13g, 8.23mmol) were added, the temperature was raised to 60 ℃ under nitrogen and the reaction was stirred for 4 hours. After cooling to room temperature, water (20mL) was added, extraction was performed with ethyl acetate (40mLx2), the organic phases were combined, washed with saturated brine (20mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure, and the residue was passed through a silica gel column to give 0.96g of a white solid with a yield of 43.4%. LC-MS (APCI): M/z 322.1(M +1)+.
Step 6 Synthesis of Compound T-5
To a 25mL single neck flask equipped with a magnetically stirred condenser was added compound 24(200mg, 0.78mmol), compound 31(250mg, 0.78mmol) and ethylene glycol monomethyl ether (5mL), the solution was stirred, and a solution of hydrogen chloride in isopropanol (1.17mmol, 0.23mL, 5M) was added dropwise under a nitrogen atmosphereThe reaction was warmed to 120 ℃ and stirred overnight with heat. After cooling to room temperature, water (10mL) and saturated sodium bicarbonate (5mL) were added, dichloromethane was extracted (15mLx3), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, concentrated, and the residue was passed through a silica gel column to give 240mg of a white solid with a yield of 44.9%. LC-MS (APCI): M/z 535.2(M +1)+.1H NMR(300MHz,CDCl3)δ(ppm):10.84(s,1H),8.64-8.60(m,1H),8.15(d,J=9.0Hz,1H),8.10(s,1H),7.51(t,J=6.0Hz,1H),7.34-7.27(m,2H),7.17-7.12(m,1H),6.53(d,J=2.4Hz,1H),6.48(dd,J=9.0Hz,J=2.4Hz,1H),3.88(s,3H),3.58(d,J=12.0Hz,2H),3.29-3.18(m,1H),2.78(s,6H),2.65(t,J=12.0Hz,2H),2.28(d,J=12.0Hz,2H),2.04-1.85(m,2H)。
3Example 6(2- ((2- ((4- (4- (di (methyl-d) amino) piperidin-1-yl) -2-methoxyphenyl) amino) - 3Preparation of 5-chloropyrimidin-4-yl) amino) phenyl) bis (methyl-d) phosphine oxide (Compound T-6)
Figure BDA0001955701090000411
The synthesis was carried out using the following route:
Figure BDA0001955701090000412
a25 mL single neck flask equipped with a magnetic stirring condenser was charged with Compound 6(200mg, 0.78mmol), Compound 31(250mg, 0.78mmol) and ethylene glycol monomethyl ether (5mL), stirred to dissolve, added dropwise with hydrogen chloride isopropanol solution (1.17mmol, 0.23mL, 5M), warmed to 120 ℃ under nitrogen and stirred overnight. After cooling to room temperature, water (10mL) and saturated sodium bicarbonate (5mL) were added, dichloromethane was extracted (15mLx3), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, concentrated, and the residue was passed through a silica gel column to give 240mg of a white solid with a yield of 44.9%. LC-MS (APCI) M/z 541.2(M +1)+.1H NMR(300MHz,CDCl3)δ10.84(s,1H),8.64-8.60(m,1H),8.15(d,J=9.0Hz,1H),8.10(s,1H),7.51(t,J=6.0Hz,1H),7.34-7.27(m,2H),7.17-7.12(m,1H),6.53(d,J=2.4Hz,1H),6.48(dd,J=9.0Hz,J=2.4Hz,1H),3.88(s,3H),3.72(d,J=12.0Hz,2H),3.13-3.10(m,1H),2.77(t,J=12.0Hz,2H),2.30-2.24(m,2H),2.00-1.91(m,2H).
Example 7(2- ((5-chloro-2- ((4- (4- (dimethylamino) piperidin-1-yl) -3-methoxyphenyl) amino) pyrimidine Preparation of pyridin-4-yl) amino) phenyl) dimethylphosphine oxide (Compound T-1a)
Figure BDA0001955701090000421
The synthesis was carried out using the following route:
Figure BDA0001955701090000422
step 1 Synthesis of Compound 3a
Compound 1a (887mg, 4.72mmol) and acetonitrile (20mL) are added sequentially to a 50mL single neck flask equipped with magnetic stirring, the supernatant is stirred, the dihydrochloride of compound 2a (1.1g, 5.66mmol) and potassium carbonate (2.2g, 15.57mmol) are added, the reaction mixture is warmed to 70 ℃ under nitrogen and the reaction is stirred for 3 hours with incubation. After cooling to room temperature, the solvent was evaporated under reduced pressure, water (60mL) was added to precipitate a large amount of yellow solid, which was filtered, washed with water (10mL) and dried to obtain 1.0g of yellow solid with a yield of 75.9%. LC-MS (APCI): M/z 280.2(M +1)+.
Step 2 Synthesis of Compound 4a
To a 50mL single neck flask equipped with magnetic stirring and a condenser was added compound 3a (1.0g,3.58mmol) and ethanol/water (30mL, 3/1), the solution was stirred, reduced iron powder (33.8mmol, 1.9g) and ammonium chloride (10.14mmol, 543mg) were added, and the reaction was allowed to warm to reflux under nitrogen and held for 1 hour. Cooling to room temperature, filtering, washing the filter cake with ethanol (5mL), concentrating to remove the organic solvent, extracting with dichloromethane (40mLx3), combining the organic phases, drying over anhydrous sodium sulfate, filtering, concentrating to obtain a brown solid 800mg, 89.4% yield。LC-MS(APCI):m/z=250.2(M+1)+.
Step 3 Synthesis of Compound T-1a
A25 mL single neck flask equipped with a magnetic stirring condenser was charged with compound 4a (800mg, 3.21mmol), compound 5a (1.01g, 3.21mmol) and ethylene glycol monomethyl ether (10mL), stirred to dissolve, added dropwise with hydrogen chloride isopropanol solution (4.82mmol, 0.96mL, 5M), warmed to 120 ℃ under nitrogen and stirred overnight with incubation. After cooling to room temperature, water (10mL) and saturated sodium bicarbonate (5mL) were added, the mixture was extracted with dichloromethane (15mLx3), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, concentrated, and the residue was passed through a silica gel column to give 600mg of a white solid with a yield of 35.3%. LC-MS (APCI): M/z 529.2(M +1)+.1H NMR(400MHz,CDCl3)δ(ppm):10.93(s,1H),8.62-8.59(m,1H),8.09(s,1H),7.34(t,J=8.0Hz,1H),7.31-7.25(m,1H),7.15(d,J=2.4Hz,1H),7.14-7.11(m,1H),7.05(dd,J=8.0Hz,J=2.4Hz,1H),6.98(s,1H),6.84(d,J=8.0Hz,1H),3.76(s,3H),3.58(d,J=12.0Hz,2H),3.29-3.18(m,1H),2.78(s,6H),2.65(t,J=12.0Hz,2H),2.28(d,J=12.0Hz,2H),2.04-1.85(m,2H),1.85(s,3H),1.82(s,3H).
Example 8(2- ((5-chloro-2- ((4- (4- (dimethylamino) piperidin-1-yl) -3-fluorophenyl) amino) pyrimidine- Preparation of 4-yl) amino) phenyl) dimethylphosphine oxide (Compound T-2a)
Figure BDA0001955701090000431
The synthesis was carried out using the following route:
Figure BDA0001955701090000441
step 1 Synthesis of Compound 7a
To a 50mL single neck flask equipped with magnetic stirring was added compound 6a (750mg, 4.72mmol) and acetonitrile (20mL) in sequence, the supernatant stirred, compound 2a dihydrochloride (1.1g, 5.66mmol) and potassium carbonate (2.2g, 15.66 mmol) added57mmol), the reaction mixture was warmed to 70 ℃ under nitrogen and the reaction was stirred for 3 hours with heat. After cooling to room temperature, the solvent was evaporated under reduced pressure, water (60mL) was added to precipitate a large amount of yellow solid, which was filtered, washed with water (10mL) and dried to obtain 955mg of yellow solid in a yield of 75.9%. LC-MS (APCI) M/z 268.2(M +1)+.
Step 2 Synthesis of Compound 8a
To a 50mL single neck flask equipped with magnetic stirring and a condenser was added compound 7a (955mg, 3.58mmol) and ethanol/water (30mL, 3/1), the solution was stirred, reduced iron powder (33.8mmol, 1.9g) and ammonium chloride (10.14mmol, 543mg) were added, and the reaction was warmed to reflux under nitrogen and incubated for 1 hour. Cool to room temperature, filter, wash the filter cake with ethanol (5mL), concentrate to remove organic solvent, extract with dichloromethane (40mLx3), combine the organic phases, dry over anhydrous sodium sulfate, filter, and concentrate to give a brown solid 760mg, 89.4% yield. LC-MS (APCI): M/z 238.2(M +1)+.
Step 3 Synthesis of Compound T-2a
A25 mL single neck flask equipped with a magnetic stirring condenser was charged with compound 8a (760mg, 3.21mmol), compound 5a (1.01g, 3.21mmol) and ethylene glycol monomethyl ether (10mL), stirred to dissolve, added dropwise with hydrogen chloride isopropanol solution (4.82mmol, 0.96mL, 5M), warmed to 120 ℃ under nitrogen and stirred overnight with incubation. After cooling to room temperature, water (10mL) and saturated sodium bicarbonate (5mL) were added, dichloromethane was extracted (15mLx3), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, concentrated, and the residue was passed through a silica gel column to give 600mg of a white solid with a yield of 36.1%. LC-MS (APCI): M/z 517.2(M +1)+.1H NMR(400MHz,CDCl3)δ(ppm):10.91(s,1H),8.57-8.53(m,1H),8.09(s,1H),7.63(dd,J=14.4Hz,J=2.4Hz,1H),7.52(t,J=8.0Hz,1H),7.33-7.29(m,1H),7.18-7.16(m,1H),7.08(s,1H),7.01(dd,J=8.8Hz,J=1.6Hz,1H),6.87(t,J=8.8Hz,1H),3.51(d,J=12.0Hz,2H),3.21-3.09(m,1H),2.77(s,6H),2.77-2.73(m,2H),2.28(d,J=12.0Hz,2H),2.00-1.95(m,2H),1.85(s,3H),1.82(s,3H).
Example 9(2- ((2- ((4- (4- (dimethylamino) piperidin-1-yl) -2-methoxyphenyl) amino) -7H-py-ridine Pyrrolo [2,3-d]Preparation of pyrimidin-4-yl) amino) phenyl) dimethylphosphine oxide (compound T-3a)
Figure BDA0001955701090000451
The synthesis was carried out using the following route:
Figure BDA0001955701090000452
step 1 Synthesis of Compound 10a
To a 250mL single neck flask equipped with magnetic stirring was added compound 9a (3.0g, 16.11mmol) and dichloromethane (80mL), the solution was stirred, p-toluenesulfonyl chloride (TsCl, 3.18g, 16.92mmol), triethylamine (3.24g, 32.22mmol) and DMAP (4-dimethylaminopyridine, 60mg, 0.48mmol) were added in that order, and after the addition, the reaction mixture was stirred at room temperature under nitrogen overnight. The reaction was quenched by the addition of water (50mL), the organic layer was separated, the aqueous layer was extracted with dichloromethane (40mLx2), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated, and the residue was passed through a silica gel column to give 4.8g of a white solid with a yield of 89.1%. LC-MS (APCI) M/z 342.1(M +1)+.1H NMR(300MHz,CDCl3)δ(ppm):8.12(d,J=5.1Hz,1H),8.03(d,J=8.4Hz,2H),7.50(d,J=8.4Hz,2H),6.98(d,J=4.2Hz,1H),2.38(s,3H).
Step 2 Synthesis of Compound 12a
To a 100mL single neck flask equipped with magnetic stirring was added compound 10a (3.4g, 10mmol) and DMF (20mL), the solution was stirred, compound 11a (1.7g, 10mmol) and DIPEA (1.3g, 10mmol) were added, the temperature was raised to 110 ℃ under nitrogen and the reaction was stirred overnight with incubation. Cooled to room temperature, water (60mL) was added, ethyl acetate extracted (50mLx3), the organic phases combined, washed with water (100mLx2), dried over anhydrous sodium sulfate, filtered, concentrated, and the residue was passed through a silica gel column to give 3.0g of a white solid in 63.2% yield. LC-MS (APCI): M/z 475.2(M +1)+.
Step 3 Synthesis of Compound 14a
Is equipped with magnetic stirringA20 mL stirred microwave tube was charged with compound 12a (304mg, 0.64mmol) and anhydrous t-butanol (10mL), stirred to dissolve, compound 13a (200mg, 0.8mmol) and anhydrous potassium carbonate (166mg, 1.2mmol) were added, nitrogen was bubbled for 3 minutes, and xphos (2-dicyclohexyl-2 ', 4 ', 6 ' -triisopropyl biphenyl, 30mg) and Pd were added2(dba)3(tris (dibenzylideneacetone) dipalladium, 30mg), the microwave tube was sealed in a nitrogen atmosphere, placed in a microwave reactor and heated to 160 ℃ and stirred for 1 hour with constant temperature. After cooling to room temperature, ethyl acetate (30mL) was added to dilute the solution, the insoluble solid was filtered off, the filtrate was concentrated, and the residue was passed through a silica gel column to give 307mg of a white solid with a yield of 55.8%. LC-MS (APCI) M/z 688.2(M +1)+.
Step 4 Synthesis of Compound T-3a
To a 50mL single neck flask equipped with magnetic stirring was added compound 14a (300mg, 0.44mmol) and isopropanol, the solution was stirred, aqueous sodium hydroxide (0.87mL, 1.76mmol, 2M) was added, the reaction mixture was warmed to 60 ℃ under nitrogen and the reaction was stirred for 3 hours. Cooled to room temperature, water (20mL) was added, dichloromethane extracted (30mLx3), the organic phases combined, dried over anhydrous sodium sulfate, filtered, concentrated, and the residue passed through a silica gel column to give 140mg of a white solid in 60.1% yield. LC-MS (APCI): M/z 534.2(M +1)+.1H NMR(400MHz,CDCl3)δ(ppm):11.18(s,1H),9.20(s,1H),8.96-8.93(m,1H),8.28(d,J=8.4Hz,1H),7.52(t,J=8.0Hz,1H),7.27-7.19(m,2H),7.09-7.05(m,1H),6.76(d,J=4.0Hz.1H),6.58-6.51(m,3H),3.89(s,3H),3.70(d,J=12.0Hz,2H),3.21-3.19(m,1H),2.78(s,6H),2.77-2.74(m,2H),2.28(d,J=12.0Hz,2H),2.00-1.94(m,2H),1.88(s,3H),1.85(s,3H).
Example 10(2- ((2- ((4- (4- (dimethylamino) piperidin-1-yl) -3-methoxyphenyl) amino) -7H-pir-o) Pyrrolo [2,3-d]Preparation of pyrimidin-4-yl) amino) phenyl) dimethylphosphine oxide (compound T-4a)
Figure BDA0001955701090000471
The synthesis was carried out using the following route:
Figure BDA0001955701090000472
step 1 Synthesis of Compound 15a
To a 20mL microwave tube equipped with magnetic stirring was added compound 12a (304mg, 0.64mmol) and dry t-butanol (10mL), the solution was stirred, compound 4a (200mg, 0.8mmol) and dry potassium carbonate (166mg, 1.2mmol) were added, nitrogen bubbled for 3min, xphos (30mg) and Pd were added2(dba)3(30mg), the microwave tube was sealed under nitrogen, placed in a microwave reactor and heated to 160 ℃ with stirring for 1 hour. After cooling to room temperature, ethyl acetate (30mL) was added to dilute the solution, the insoluble solid was filtered off, the filtrate was concentrated, and the residue was passed through a silica gel column to give 307mg of a white solid with a yield of 55.8%. LC-MS (APCI) M/z 688.2(M +1)+.
Step 2 Synthesis of Compound T-4a
To a 50mL single neck flask equipped with magnetic stirring was added compound 15a (300mg, 0.44mmol) and isopropanol, the solution was stirred, aqueous sodium hydroxide (0.87mL, 1.76mmol, 2M) was added, the reaction mixture was warmed to 60 ℃ under nitrogen and the reaction was stirred for 3 hours. Cooled to room temperature, water (20mL) was added, dichloromethane extracted (30mLx3), the organic phases combined, dried over anhydrous sodium sulfate, filtered, concentrated, and the residue passed through a silica gel column to give 140mg of a white solid in 60.1% yield. LC-MS (APCI): M/z 534.2(M +1)+.1H NMR(300MHz,DMSO-D6)δ(ppm):11.64(s,1H),11.28(s,1H),9.18-9.13(m,1H),8.83(s,1H),7.61-7.46(m,3H),7.36(dd,J=9.0Hz,J=2.4Hz,1H),7.07(t,J=4.5Hz,1H),6.95-6.93(m,1H),6.82(d,J=8.4Hz,1H),6.38-6.36(m,1H),3.79(s,3H),3.47-3.43(m,2H),2.68(s,6H),2.58-2.52(m,2H),2.08(d,J=11.4Hz,2H),1.86(s,3H),1.81(s,3H),1.80-1.72(m,2H).
Example 11(2- ((2- ((4- (4- (dimethylamino) piperidin-1-yl) -3-fluorophenyl) amino) -7H-pyrrolo [2,3-d]Preparation of pyrimidin-4-yl) amino) phenyl) dimethylphosphine oxide (compound T-5a)
Figure BDA0001955701090000481
The synthesis was carried out using the following route:
Figure BDA0001955701090000482
step 1 Synthesis of Compound 16a
To a 20mL microwave tube equipped with magnetic stirring was added compound 12a (304mg, 0.64mmol) and dry t-butanol (10mL), the solution was stirred, compound 8a (189mg, 0.8mmol) and dry potassium carbonate (166mg, 1.2mmol) were added, nitrogen bubbled for 3 minutes, xphos (30mg) and Pd were added2(dba)3(30mg), the microwave tube was sealed under nitrogen, placed in a microwave reactor and heated to 160 ℃ with stirring for 1 hour. After cooling to room temperature, ethyl acetate (30mL) was added to dilute the solution, the insoluble solid was filtered off, the filtrate was concentrated, and the residue was passed through a silica gel column to give 307mg of a white solid with a yield of 55.8%. LC-MS (APCI) M/z 688.2(M +1)+.
Step 2 Synthesis of Compound T-5a
To a 50mL single neck flask equipped with magnetic stirring was added compound 16a (300mg, 0.44mmol) and isopropanol, the solution was stirred, aqueous sodium hydroxide (0.87mL, 1.76mmol, 2M) was added, the reaction mixture was warmed to 60 ℃ under nitrogen and the reaction was stirred for 3 hours. Cooled to room temperature, water (20mL) was added, dichloromethane extracted (30mLx3), the organic phases combined, dried over anhydrous sodium sulfate, filtered, concentrated, and the residue passed through a silica gel column to give 140mg of a white solid in 60.1% yield. LC-MS (APCI): M/z 522.2(M +1)+.1H NMR(400MHz,CDCl3)δ(ppm):11.14(s,1H),8.94-8.90(m,2H),7.72(dd,J=14.8Hz,J=2.4Hz,1H),7.53(t,J=8.0Hz,1H),7.26-7.22(m,1H),7.10-7.05(m,2H),6.91(t,J=9.2Hz,1H),6.87(s,1H),6.78(d,J=3.6Hz,1H),6.58(d,J=3.6Hz,1H),3.45(d,J=12.0Hz,2H),2.67(t,J=12.0Hz,2H),2.48-2.40(m,1H),2.39(s,6H),1.97(d,J=12.0Hz,2H),1.87(s,3H),1.83(s,3H),1.82-1.77(m,2H).
Example 12(2- ((5)-chloro-2- ((4- ((dimethylamino) methyl) -2-methoxyphenyl) amino) pyrimidine-4- Preparation of the base) amino) phenyl) dimethylphosphine oxide (Compound T)
Figure BDA0001955701090000491
The synthesis was carried out using the following route:
Figure BDA0001955701090000501
step 1 Synthesis of Compound 3b
To a 50mL single neck flask equipped with magnetic stirring was added compound 1b (887mg, 4.72mmol) and acetonitrile (20mL) in sequence, the mixture stirred until clear, compound 2b (1.0g, 4.72mmol) and potassium carbonate (2.2g, 15.57mmol) were added, the reaction mixture was warmed to 70 ℃ under nitrogen and the reaction was stirred for 3 hours with incubation. After cooling to room temperature, the solvent was evaporated under reduced pressure, water (60mL) was added to precipitate a large amount of yellow solid, which was filtered, washed with water (10mL) and dried to obtain 1.4g of yellow solid with a yield of 81.7%. LC-MS (APCI): M/z 364.2(M +1)+.
Step 2 Synthesis of Compound 4b
Compound 3b (1.4g, 3.86mmol) and ethyl acetate (20mL) were added sequentially to a 100mL single neck flask equipped with magnetic stirring, the solution was stirred, a solution of hydrogen chloride in isopropanol (20mL, 5M) was added, and the reaction mixture was stirred at room temperature under nitrogen for 1 hour. The solvent was evaporated under reduced pressure, saturated aqueous sodium bicarbonate (20mL) and dichloromethane (30mL) were added, the organic layer was separated, the aqueous layer was extracted with dichloromethane (20mLx2), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the yellow solid was concentrated at 1.0g, 98.1% yield. LC-MS (APCI): M/z ═ 264.2(M +1)+.
Step 3 Synthesis of Compound 5b
To a 50mL single neck flask equipped with magnetic stirring was added compound 4b (1.0g, 3.78mmol) and methanol (20mL) in sequence, the mixture was stirred to a clear solution, aqueous formaldehyde (617mg, 7.6mmol) and glacial acetic acid (2 drops) were added, and the mixture was stirred under nitrogenStirring for 10 minutes, adding NaBH under cooling in ice water bath3CN (718mg,11.4mmol), after addition, the reaction was stirred at room temperature under nitrogen for 2 hours. The solvent was distilled off under reduced pressure, and the residue was passed through a silica gel column to give 935mg of a yellow solid in 89.3% yield, LC-MS (APCI): M/z 278.2(M +1)+.
Step 4 Synthesis of Compound 6b
To a 50mL single neck flask equipped with magnetic stirring and a condenser was added compound 5b (935mg, 3.38mmol) and ethanol/water (30mL, 3/1), the solution was stirred, reduced iron powder (33.8mmol, 1.9g) and ammonium chloride (10.14mmol, 543mg) were added, and the reaction was warmed to reflux under nitrogen and held for 1 hour. Cool to room temperature, filter, wash the filter cake with ethanol (5mL), concentrate to remove the organic solvent, extract with dichloromethane (40mLx3), combine the organic phases, dry over anhydrous sodium sulfate, filter, and concentrate to give 850mg of a brown solid in 99.6% yield. LC-MS (APCI) M/z 248.3(M +1)+.
Step 5 Synthesis of Compound T
A25 mL single neck flask equipped with a magnetic stirring condenser was charged with compound 6b (850mg, 3.44mmol), compound 7b (1.3g, 4.13mmol) and ethylene glycol monomethyl ether (10mL), stirred to dissolve, added dropwise with hydrogen chloride isopropanol solution (5.16mmol, 1.03mL, 5M), warmed to 120 ℃ under nitrogen and stirred overnight with incubation. After cooling to room temperature, water (10mL) and saturated sodium bicarbonate (5mL) were added, dichloromethane was extracted (15mLx3), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, concentrated, and the residue was passed through a silica gel column to give 600mg of a white solid with a yield of 33.1%. LC-MS (APCI): M/z 527.2(M +1)+.1H NMR(400MHz,CDCl3)δ(ppm):10.80(s,1H),8.68-8.65(m,1H),8.07(s,1H),7.98(d,J=8.8Hz,1H),7.49(t,J=8.0Hz,1H),7.30-7.25(m,1H),7.14-7.11(m,2H),6.29(s,1H),6.26-6.24(m,1H),3.86(s,3H),3.35(t,J=8.8Hz,2H),3.23-3.20(m,2H),2.99-2.97(m,2H),2.82(t,J=8.8Hz,2H),2.43-2.40(m,2H),1.85(s,3H),1.82(s,3H).
EXAMPLE 13 biological evaluation of Compounds
The compounds of the invention were evaluated in a number of assays to determine their biological activity. For example, compounds of the invention can be tested for their ability to inhibit a variety of protein kinases of interest. Some of the compounds tested showed potent inhibitory activity on ALK kinase.
(1) Evaluation of kinase inhibition
Compound preparation: test compounds were dissolved in DMSO to make 20mM stock. Compounds were diluted to 0.1mM (100-fold final dilution) in DMSO and diluted in 3-fold gradients, 11 concentrations, prior to use. When adding medicine, the medicine is diluted by buffer solution into 4 times of the dilution solution with final concentration.
And (3) kinase detection: after buffer preparation, the enzyme was mixed with the compounds of different concentrations prepared by dilution in advance, and left at room temperature for 30 minutes, each concentration being double-well. The corresponding substrate and ATP were added and the reaction was carried out at room temperature for 60 minutes (negative and positive controls were set). And (3) after the reaction is finished, adding an antibody for detection, incubating at room temperature for 60 minutes, then carrying out Evnvision detection, and collecting data. Data analysis and mapping were performed according to XLfit5 software. By this formula (IC50 ═ ABS onset [ (ABS test-ABS onset)/(ABS control-ABS onset)]x 100) computing IC50The value is obtained. Wherein A represents IC50Is 1-2.5nM, B denotes IC502.5-5nM, C represents IC50Is 5-10nM, D represents IC50Is 10-15nM, E represents IC5015-30 nM.
The compounds of the present invention and positive controls were tested in the above kinase inhibition experiments and found to have more potent activity on ALK and ALK [ L1196M ]. The results for representative example compounds are summarized in table 1 below.
Table 1:
Figure BDA0001955701090000521
Figure BDA0001955701090000531
(2) cytotoxicity test
The in vitro antiproliferative activity of the compounds of the invention on 3 strains of cells cultured in vitro was examined by the CellTiter-Glo method. The experimental result shows that the compound has strong inhibition effect on the in vitro proliferation of the EML4-ALK and EML4-ALK L1196M mutant cells cultured in vitro.
Cell line: BaF3 parent; BaF3[ EML4-ALK ] (from Yammincord) and BaF3[ EML4-ALK L1196M ] (from Yammincord); wherein BaF3parental is cultured in RPMI1640 medium containing 10ng/ml IL-3, 0% fetal calf serum, 100U/ml penicillin, and 100. mu.g/ml streptomycin, and BaF3[ EML4-ALK ] and BaF3[ EML4-ALK L1196M ] are cultured in RPMI1640 medium containing 10% fetal calf serum, 100U/ml penicillin, and 100. mu.g/ml streptomycin.
Reagents and consumables: RPMI-1640(GIBCO, Cat. No. A10491-01); fetal bovine serum (GIBCO, catalog No. 10099141); 0.25% trypsin-EDTA (GIBCO, cat No. 25200); penicillin-streptomycin, liquid (GIBCO, cat # 15140-122); DMSO (Sigma, cat # D2650); CellTiter-Glo test kit (Promega, cat. No. G7572), 96-well plate (Corning, cat. No. 3365).
The specific experimental method comprises the following steps:
1. the test compound was dissolved in DMSO to form a stock solution, and the stock solution was subjected to gradient dilution to obtain a 10-fold working concentration solution.
2. Cells in the logarithmic growth phase were diluted with a culture solution to be adjusted to a specific cell concentration, and 90. mu.l of a cell suspension was added to a 96-well plate so that the cell density reached a prescribed concentration. The cells were cultured overnight in a 5% carbon dioxide incubator at 37 ℃.
3. 10 μ l of the drug solution was added to each well of the 96-well plate in which the cells had been seeded. The highest concentration of the tested compound was 20 μ M, 10 concentrations, 3-fold gradient dilution, double wells.
4. After the cells were cultured for 72 hours, CellTiter-Glo examined the cell viability. Dose-response curves were generated and IC calculated using GraphPad Prism software50
The compounds of the present invention were tested in the above cytotoxicity experiments and found to be resistant to Ba/F3ALK and Ba/F3ALK [ L1196M ]]Has strong activity. The results of the inhibition of in vitro proliferation of cancer cells of representative examples are summarized in Table 2 below, wherein A represents IC50Less than or equal to 15nM, B denotes IC5015-50nM, C denotes IC50Is 50-500nM, D represents IC50500-1000nM, E represents IC50Is an IC50>1000nM。
TABLE 2
Figure BDA0001955701090000541
In addition, the compounds of the present invention are also selective for Ba/F3ALK and Ba/F3ALK [ L1196M ] as compared to Ba/F3 parent. For example, compound T-1 has a Ba/F3ALK/BaF3 partial selectivity greater than 400 and a Ba/F3ALK [ L1196M ]/Ba/F3 partial selectivity greater than 200.
(3) Metabolic stability evaluation
Microsome experiment: human liver microsomes: 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).
Preparing a stock solution: an amount of the compound of example was weighed out finely and dissolved in DMSO to 5mM each.
Preparation of phosphate buffer (100mM, pH 7.4): 150mL of 0.5M potassium dihydrogenphosphate and 700mL of 0.5M dipotassium hydrogenphosphate solution prepared in advance were mixed, the pH of the mixture was adjusted to 7.4 with the 0.5M dipotassium hydrogenphosphate solution, diluted 5-fold with ultrapure water before use, and magnesium chloride was added to obtain a phosphate buffer solution (100mM) containing 100mM potassium phosphate and 3.3mM magnesium chloride at a pH of 7.4.
NADPH regenerating system solution (containing 6.5mM NADP, 16.5mM G-6-P, 3U/mL G-6-P D, 3.3mM magnesium chloride) was prepared and placed on wet ice before use.
Preparing a stop solution: acetonitrile solution containing 50ng/mL propranolol hydrochloride and 200ng/mL tolbutamide (internal standard). 25057.5 mu L of phosphate buffer solution (pH7.4) is taken to a 50mL centrifuge tube, 812.5 mu L of human liver microsome is respectively added and mixed evenly, and liver microsome dilution liquid with the protein concentration of 0.625mg/mL is obtained. 25057.5 mu L of phosphate buffer (pH7.4) is taken to a 50mL centrifuge tube, 812.5 mu L of SD rat liver microsome is respectively added, and the mixture is mixed evenly to obtain liver microsome dilution with the protein concentration of 0.625 mg/mL.
Incubation of the samples: the stock solutions of the corresponding compounds were diluted to 0.25mM with 70% acetonitrile in water, respectively, and used as working solutions. 398. mu.L of human liver microsome or rat liver microsome dilutions were added to a 96-well plate (N2), 2. mu.L of 0.25mM working solution was added, and mixed well.
Determination of metabolic stability: 300. mu.L of pre-cooled stop solution was added to each well of a 96-well deep-well plate and placed on ice as a stop plate. The 96-well incubation plate and the NADPH regeneration system are placed in a 37 ℃ water bath box, shaken at 100 rpm and pre-incubated for 5 min. mu.L of the incubation solution was taken out from each well of the incubation plate, added to the stop plate, mixed well, and supplemented with 20. mu.L of NADPH regenerating system solution as a 0min sample. Then 80. mu.L of NADPH regenerating system solution was added to each well of the incubation plate, the reaction was started, and the timer was started. The reaction concentration of the corresponding compound was 1. mu.M, and the protein concentration was 0.5 mg/mL. When the reaction was carried out for 10min, 30 min and 90min, 100. mu.L of each reaction solution was added to the stop plate and vortexed for 3min to terminate the reaction. The stop plates were centrifuged at 5000 Xg for 10min at 4 ℃. And (3) taking 100 mu L of supernatant to a 96-well plate in which 100 mu L of distilled water is added in advance, mixing uniformly, and performing sample analysis by adopting LC-MS/MS.
And (3) data analysis: and detecting peak areas of the corresponding compound and the internal standard through an LC-MS/MS system, and calculating the peak area ratio of the compound to the internal standard. The slope is determined by plotting the natural logarithm of the percentage remaining of the compound against time and calculating t according to the formula1/2And CLintWhere V/M is equal to 1/protein concentration.
Figure BDA0001955701090000561
t1/2(min);CLint(μL/min/mg)。
The compounds of the invention were evaluated for their metabolic stability in human and rat liver microsomes. The half-life and intrinsic hepatic clearance as indicators of metabolic stability are shown in table 3 below. The compound of the invention can obviously improve the metabolic stability.
TABLE 3
Figure BDA0001955701090000562
(4) Pharmacokinetic experiment of rat
6 male Sprague-Dawley rats, 7-8 weeks old, weighing about 210g, were divided into 2 groups of 3 animals each, and a single dose of the compound (10 mg/kg per oral dose) was administered intravenously or orally to compare the pharmacokinetic differences.
Rats were fed with standard feed and given water. Fasting began 16 hours prior to the experiment. The drug was dissolved with PEG400 and dimethyl sulfoxide. Blood was collected from the orbit at 0.083 hr, 0.25 hr, 0.5 hr, 1 hr, 2 hr, 4 hr, 6 hr, 8 hr, 12 hr and 24 hr post-dose.
The rats were briefly anesthetized after ether inhalation and 300 μ L of blood was collected from the orbit into a test tube. There was 30 μ L of 1% heparin salt solution in the tube. Before use, the tubes were dried overnight at 60 ℃. After completion of blood collection at the last time point, rats were sacrificed after ether anesthesia.
Immediately after blood collection, the tubes were gently inverted at least 5 times to ensure mixing and then placed on ice. The blood samples were centrifuged at 5000rpm for 5 minutes at 4 ℃ to separate the plasma from the erythrocytes. Pipette 100 μ L of plasma into a clean plastic centrifuge tube, designating the name of the compound and the time point. Plasma was stored at-80 ℃ before analysis. The concentration of the compounds of the invention in plasma was determined by LC-MS/MS. Pharmacokinetic parameters were calculated based on the plasma concentration of each animal at different time points.
Experiments show that the compound has better pharmacokinetic properties in animals, so the compound has better pharmacodynamics and treatment effects.
The foregoing is a more detailed description of the invention in connection with specific preferred embodiments and it is not intended that the invention be limited to these specific details. For those skilled in the art to which the invention pertains, several simple deductions or substitutions can be made without departing from the spirit of the invention, and all shall be considered as belonging to the protection scope of the invention.

Claims (5)

1. A compound selected from the following compounds, or a pharmaceutically acceptable salt, crystal form, or stereoisomer thereof:
Figure FDA0003590877610000011
2. a pharmaceutical composition comprising a pharmaceutically acceptable excipient and a compound of claim 1, or a pharmaceutically acceptable salt, crystal, or stereoisomer thereof.
3. Use of a compound of claim 1, or a pharmaceutically acceptable salt, crystal form, or stereoisomer thereof, or a pharmaceutical composition of claim 2, in the manufacture of a medicament for treating an ALK-mediated cancer.
4. The use according to claim 3, wherein said cancer is selected from the group consisting of non-small cell lung cancer, breast cancer, neural tumors, esophageal cancer, soft tissue cancer, lymphoma and leukemia.
5. The use according to claim 4, wherein the non-small cell lung cancer is ALK-positive non-small cell lung cancer; wherein the lymphoma is anaplastic large cell lymphoma.
CN201910066072.9A 2018-02-07 2019-01-24 Substituted diaminopyrimidine compounds Active CN109851638B (en)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
CN2018101216410 2018-02-07
CN201810121641 2018-02-07
CN2018102041466 2018-03-13
CN2018102040285 2018-03-13
CN201810204028 2018-03-13
CN201810204146 2018-03-13

Publications (2)

Publication Number Publication Date
CN109851638A CN109851638A (en) 2019-06-07
CN109851638B true CN109851638B (en) 2022-05-31

Family

ID=66895834

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201910066072.9A Active CN109851638B (en) 2018-02-07 2019-01-24 Substituted diaminopyrimidine compounds

Country Status (2)

Country Link
CN (1) CN109851638B (en)
WO (1) WO2019154091A1 (en)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110467637B (en) * 2018-05-09 2022-02-18 北京赛特明强医药科技有限公司 Bisaminyl chloropyrimidine compound containing phosphine oxide substituted aniline, preparation method and application thereof
CN111836819A (en) * 2018-05-24 2020-10-27 北京赛特明强医药科技有限公司 Arylamine-substituted pyrrolopyrimidine compound, and preparation method and application thereof
CN110526915B (en) * 2018-05-25 2022-02-01 首药控股(北京)股份有限公司 Preparation method of anaplastic lymphoma kinase inhibitor
US20230026840A1 (en) * 2019-11-21 2023-01-26 Tyk Medicines, Inc. Compound used as egfr kinase inhibitor and use thereof
CN112239422B (en) * 2020-10-16 2022-07-22 河北化工医药职业技术学院 Bugatinib intermediate, salt thereof, preparation method thereof and preparation method of brigatinib

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102020522A (en) * 2009-09-21 2011-04-20 陈松源 Preparation method and application of deuterated drugs
CN103664726A (en) * 2012-08-31 2014-03-26 南京华狮化工有限公司 Deuterated bimatoprost or derivative thereof and preparation method and application of deuterated bimatoprost or derivative thereof
CN105061506A (en) * 2015-07-27 2015-11-18 苏州明锐医药科技有限公司 Preparation method for anti-tumor drug AP26113
CN106220608A (en) * 2016-07-25 2016-12-14 张柏 Diphenylamino pyrimidine and triaizine compounds, its Pharmaceutical composition and purposes
CN107698603A (en) * 2016-08-09 2018-02-16 厦门大学 Thienopyrimidines, its preparation method, Pharmaceutical composition and its application

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0321710D0 (en) * 2003-09-16 2003-10-15 Novartis Ag Organic compounds
ES2645689T3 (en) * 2008-05-21 2017-12-07 Ariad Pharmaceuticals, Inc. Phosphorus derivatives as kinase inhibitors
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
JP6887430B2 (en) * 2015-11-27 2021-06-16 チア タイ ティエンチン ファーマシューティカル グループ カンパニー リミテッドChia Tai Tianqing Pharmaceutical Group Co., Ltd. Deuterated Brigatinib derivatives, pharmaceutical compositions containing such derivatives, and their use.
CN113912648A (en) * 2015-12-02 2022-01-11 深圳市塔吉瑞生物医药有限公司 Diaminopyrimidine compound and composition containing same
EP3492462B1 (en) * 2016-07-26 2023-08-30 Shenzhen TargetRx, Inc. Amino pyrimidine compound for inhibiting protein tyrosine kinase activity

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102020522A (en) * 2009-09-21 2011-04-20 陈松源 Preparation method and application of deuterated drugs
CN103664726A (en) * 2012-08-31 2014-03-26 南京华狮化工有限公司 Deuterated bimatoprost or derivative thereof and preparation method and application of deuterated bimatoprost or derivative thereof
CN105061506A (en) * 2015-07-27 2015-11-18 苏州明锐医药科技有限公司 Preparation method for anti-tumor drug AP26113
CN106220608A (en) * 2016-07-25 2016-12-14 张柏 Diphenylamino pyrimidine and triaizine compounds, its Pharmaceutical composition and purposes
CN107698603A (en) * 2016-08-09 2018-02-16 厦门大学 Thienopyrimidines, its preparation method, Pharmaceutical composition and its application

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Deuterated Drug Molecules: Focus on FDA-Approved Deutetrabenazine;Bruce E. Maryanoff et al.;《Biochemistry》;20181231;第57卷;472-473 *
First deuterated drug approved;Charles Schmidt;《NATURE BIOTECHNOLOGY》;20171231;第35卷(第6期);493-494 *
氘代药物进展;刘洁;《医药化工》;20161231;第42卷(第4期);199-200 *

Also Published As

Publication number Publication date
CN109851638A (en) 2019-06-07
WO2019154091A1 (en) 2019-08-15

Similar Documents

Publication Publication Date Title
CN109851638B (en) Substituted diaminopyrimidine compounds
CN111285886B (en) Substituted pyrazolo [1,5-a ] pyridine compounds, compositions comprising the same and uses thereof
TW202200562A (en) Quinoxaline dione derivative as irreversible inhibitor of KRAS G12C mutant protein
CN109627263B (en) Diphenylaminopyrimidine compounds for inhibiting kinase activity
WO2021057877A1 (en) Substituted aromatic fused ring derivative and composition comprising same, and use thereof
WO2019201131A1 (en) Di(hetero)aryl macrocyclic compound for inhibiting protein kinase activity
EP3715350B1 (en) Arylphosphine oxides for inhibiting kinase activity
JP2023109924A (en) Dihydroimidazopyrazinone compound, composition including the compound, and use thereof
CN109593102B (en) Preparation method and crystal form of deuterated diphenylaminopyrimidine compound
CN108947974B (en) Aminopyrimidine compound, composition containing aminopyrimidine compound and application of aminopyrimidine compound
CN110066272B (en) Substituted benzo [ d ] imidazoles and pharmaceutical compositions thereof
CN108137614B (en) Fused pyrimidine compound, composition containing compound and application of compound
WO2022242712A1 (en) Substituted macrocyclic compound, composition comprising compound, and use thereof
WO2021057796A1 (en) Substituted fused tricyclic derivative, and composition and use thereof
EP3816162A1 (en) Diarylpyrazole compound, composition comprising same, and use thereof
CN114874189B (en) Substituted heteroaryl derivatives, compositions and uses thereof
CN113336760B (en) Substituted amide derivatives, compositions and uses thereof
CN113493439B (en) Substituted acrylamide derivative, composition and application thereof
JP6073480B2 (en) PI3K and / or mTOR inhibitors
WO2020168963A1 (en) Substituted fused aromatic ring derivative, composition and use thereof
WO2014154026A1 (en) PI3K AND/OR mTOR INHIBITOR PRODRUG

Legal Events

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