[go: up one dir, main page]

CN111377873B - Aminopyrimidine compounds, their preparation and use - Google Patents

Aminopyrimidine compounds, their preparation and use Download PDF

Info

Publication number
CN111377873B
CN111377873B CN201911313568.8A CN201911313568A CN111377873B CN 111377873 B CN111377873 B CN 111377873B CN 201911313568 A CN201911313568 A CN 201911313568A CN 111377873 B CN111377873 B CN 111377873B
Authority
CN
China
Prior art keywords
compound
alkyl
pharmaceutically acceptable
hydrogen
acceptable salt
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
CN201911313568.8A
Other languages
Chinese (zh)
Other versions
CN111377873A (en
Inventor
刘金明
何婷
蔡家强
李晓勇
田强
宋宏梅
薛彤彤
王利春
王晶翼
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.)
Sichuan Kelun Biotech Biopharmaceutical Co Ltd
Original Assignee
Sichuan Kelun Biotech Biopharmaceutical Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Sichuan Kelun Biotech Biopharmaceutical Co Ltd filed Critical Sichuan Kelun Biotech Biopharmaceutical Co Ltd
Publication of CN111377873A publication Critical patent/CN111377873A/en
Application granted granted Critical
Publication of CN111377873B publication Critical patent/CN111377873B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/70Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings condensed with carbocyclic rings or ring systems
    • C07D239/72Quinazolines; Hydrogenated quinazolines
    • C07D239/74Quinazolines; Hydrogenated quinazolines with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, attached to ring carbon atoms of the hetero ring
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Landscapes

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

Abstract

Description

Aminopyrimidine compounds, their preparation and use
Technical Field
The invention belongs to the field of medicine, and particularly relates to an aminopyrimidine compound or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug thereof. The invention also relates to processes for the preparation of said compounds, intermediates, pharmaceutical compositions containing said compounds and their therapeutic use.
Background
Adenosine is a signaling molecule that inhibits inflammation and immune responses in vivo, extracellular adenosine is mainly of 2 origins, transport of intracellular adenosine and hydrolysis of extracellular adenosine. Adenosine can be produced in many types of tumor tissue and maintained at high levels in the tumor microenvironment. The main pathway of adenosine production in the tumor microenvironment is in tumor cells the highly surface-expressed CD73 enzyme catalyzes AMP to produce extracellular adenosine, which in turn leads to the maintenance of high concentrations of adenosine in the tumor microenvironment. Adenosine receptors are a class of G protein-coupled receptors (GPCRs) that include the four A1, A2a, A2b and A3 receptors. Among these, the A2a and A2b receptors are coupled to Gs proteins that activate adenylate cyclase, stimulating the production of intracellular cyclic adenosine monophosphate (cAMP) signaling molecules.
Adenosine A2a receptors are expressed on the surface of several cells in the immune system, such as T cells, NK cells, macrophages and dendritic cells. Adenosine generated by the tumor can interact with an adenosine A2a receptor on the surface of an immune cell infiltrated by a tumor tissue, so that the cAMP amount in the immune cell is increased, the capability of the immune cell to attack the tumor is inhibited, the organism generates immune tolerance, and the tumor cell can escape from the immune monitoring of the organism, and the adenosine generating method mainly comprises the following two aspects: (1) Blocking the activation and the function of immune cells which can kill tumor cells; (2) Increasing the number of regulatory T-cells (T-regs) that suppress the immune cell response to the tumor cell. Tumor cells use these mechanisms to escape surveillance and attack by the immune system, the survival rate of the user is improved. The A2A receptor gene knockout mouse can strengthen the anti-tumor immune function of CD8+ T cells, obviously inhibit the proliferation of tumors, transplant melanoma or lymphoma cells into a wild type mouse to grow more easily than transplant into an adenosine A2A receptor gene knockout mouse, and the adenosine A2A receptor gene knockout mouse has better response to tumor vaccines.
The adenosine A2a receptor is expressed on immune cells at a high level, and the activation of the adenosine A2a receptor can promote the organism to generate immune tolerance and promote the formation of 'immune escape' or 'immune suppression' of tumor cells, thereby creating favorable conditions for the generation and development of tumors. The adenosine A2a receptor antagonist directly targets the adenosine A2a receptor on the surface of immune cells, inhibits the activation of the receptor and further inhibits the generation of cAMP in the immune cells, restoring the T cell immune function inhibition mediated by the activation of adenosine A2a receptor, and achieving the effect of treating tumors. Therefore, the temperature of the molten metal is controlled, the adenosine A2a receptor antagonist has good application prospect in the pharmaceutical industry as a tumor treatment medicament. In addition to tumors, adenosine A2a receptors have been implicated in diseases such as parkinson's disease, alzheimer's disease, AIDS encephalopathy, multiple sclerosis, amyotrophic lateral sclerosis, huntington's disease, multiple system atrophy, cerebral ischemia, attention deficit hyperactivity disorder, sleep disorders, anxiety disorders, mood disorders, epilepsy, neuralgia, migraine.
CPI-444 from Corvus is a compound antagonistic to adenosine A2a receptors and is currently in phase I clinical studies for indications of tumors, before which CPI-444 was used in clinical trials for the treatment of central nervous system disorders. On the other hand, WO0162233A2 and WO2002014282A1 disclose that aminopyridine compounds have an antagonistic effect on adenosine A2a receptors, also discloses a therapeutic agent of Parkinson's disease or senile dementia. WO2003035639A1, WO2004016605A1 and WO2005079801A1 disclose aminopyrimidine compounds having antagonistic action on adenosine A2a receptors and are useful as therapeutic agents for diseases such as Parkinson's disease or neuralgia. WO2011095625A1 discloses an aminotriazine compound having an antagonistic effect on adenosine A2a receptors and is disclosed as a therapeutic agent for dyskinesia, stroke, or parkinson's disease.
Therefore, the adenosine A2a receptor antagonist has good application prospect in the pharmaceutical industry as a medicament. In order to achieve better tumor treatment effect and better meet the market demand, the development of a new adenosine A2a receptor antagonist with high efficiency and low toxicity is urgently needed.
Disclosure of Invention
The invention provides aminopyrimidine compounds which have good inhibitory action on adenosine A2a receptors and weak inhibitory action on adenosine A1 receptors, thus having good antitumor activity. The compounds of the invention also have good pharmacokinetic properties (e.g. good drug exposure and good oral absorption).
Some aspects of the invention provide a compound of formula (I) or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite, or prodrug thereof, wherein:
Figure BDA0002325201380000021
x is selected from N and CH;
R 1 selected from hydrogen, halogen, cyano, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 3-6 Cycloalkyl, carboxy, C 1-6 alkyl-OC (O) -, R a R b N-C (O) -, 5-6 membered heterocyclyl and 5-6 membered heteroaryl, wherein the heterocyclyl and heteroaryl are optionally substituted with substituents independently selected from the group consisting of: halogen, halogen C 1-6 An alkyl group C 1-6 Haloalkyl, C 3-6 Cycloalkyl radical, C 1-6 Alkoxy radical, R a R b N-、R a R b N-C (O) -, hydroxy-C 1-6 Alkyl-, C 1-6 alkoxy-C 1-6 Alkyl-, R a R b N-C 1-6 Alkyl-and R a R b N-C(O)-C 1-6 Alkyl-;
R 2 selected from hydrogen, halogen, cyano, C 1-6 Alkyl radical, C 1-6 Haloalkyl and C 1-6 An alkoxy group;
R 3 selected from hydrogen, halogen, C 1-6 Alkyl and C 1-6 A haloalkyl group;
R 4 selected from hydrogen, halogen, C 1-6 Alkyl and C 1-6 A haloalkyl group;
R a and R b Each independently selected from hydrogen and C 1-6 Alkyl radical, C 3-6 Cycloalkyl, hydroxy-C 1-6 Alkyl-, C 1-6 alkoxy-C 1-6 Alkyl-, R c R d N-C(O)-C 1-6 Alkyl-and 5-6 membered heterocyclyl-C 1-6 Alkyl-;
R c and R d Each independently selected from hydrogen and C 1-6 Alkyl radical, C 3-6 Cycloalkyl, hydroxy C 1-6 Alkyl radical, C 1-6 alkoxy-C 1-6 Alkyl-and 5-6 membered heterocyclyl-C 1-6 Alkyl-;
n is selected from 0, 1 or 2;
p is selected from 0, 1 or 2;
q is selected from 0, 1 or 2.
Another aspect of the invention provides a pharmaceutical composition comprising a prophylactically or therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug thereof, and one or more pharmaceutically acceptable carriers.
Another aspect of the invention provides a method of making a pharmaceutical composition, the method comprising combining a compound of the invention, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug thereof, with one or more pharmaceutically acceptable carriers.
Another aspect of the present invention provides a use of a compound of the present invention or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug thereof, or a pharmaceutical composition of the present invention for the preparation of a medicament for the prevention or treatment of an adenosine A2a receptor-related disease.
Another aspect of the invention provides a compound of the invention, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug thereof, or a pharmaceutical composition of the invention, for use in the inhibition of adenosine A2a receptor activity.
Another aspect of the present invention provides a compound of the present invention or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug thereof, or a pharmaceutical composition of the present invention, for use in the prevention or treatment of an adenosine A2a receptor-associated disease.
Another aspect of the present invention provides a method for preventing or treating an adenosine A2a receptor-related disease, which comprises administering to a subject in need thereof an effective amount of a compound of the present invention or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug thereof, or a pharmaceutical composition of the present invention.
Yet another aspect of the present invention aspects provide for the preparation of methods for preparing the compounds of the invention.
Definition of
Unless defined otherwise below, all technical and scientific terms used herein are intended to have the same meaning as commonly understood by one of ordinary skill in the art. Reference to the techniques used herein is intended to refer to techniques commonly understood in the art, including those variations of or alternatives to those techniques that would be apparent to one of ordinary skill in the art. While the following terms are believed to be well understood by those skilled in the art, the following definitions are set forth to better explain the present invention.
As used herein, the terms "comprising," "including," "having," "containing," or "involving," and other variations thereof herein, are inclusive or open-ended and do not exclude additional unrecited elements or method steps, although not necessarily present (i.e., these terms also encompass the terms "consisting essentially of … …" and "consisting of … …").
The term "alkyl" as used herein is defined as a straight or branched chain saturated aliphatic hydrocarbon group. In some embodiments, the alkyl group has 1 to 12, such as 1 to 6, carbon atoms, for example 1 to 3 carbon atoms. For example, as used herein, the term "C 1-6 Alkyl "refers to a straight or branched chain group having 1 to 6 carbon atoms (e.g., methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, or n-hexyl), which is optionally substituted with one or more (such as 1 to 3) suitable substituents such as halo (when the group is referred to as" haloalkyl ", e.g., CF) 3 、C 2 F 5 、CHF 2 、CH 2 F、CH 2 CF 3 、CH 2 Cl or-CH 2 CH 2 CF 3 Etc.).
As used herein, the term "cycloalkyl" refers to a saturated or partially unsaturated, non-aromatic, monocyclic or polycyclic (such as bicyclic) hydrocarbon ring (e.g., monocyclic, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, or bicyclic, including spiro, fused or bridged systems, such as bicyclo [1.1.1]Pentyl, bicyclo [2.2.1 ] s]Heptyl, bicyclo [3.2.1]Octyl or bicyclo [5.2.0]Nonyl, decalinyl, etc.), optionally substituted with one or more (such as 1 to 3) suitable substituents. The cycloalkyl group has 3 to 15, for example 3 to 6, carbon atoms. For example, the term "C 3-6 Cycloalkyl "refers to a saturated or partially unsaturated non-aromatic monocyclic or polycyclic (such as bicyclic) hydrocarbon ring having 3 to 6 ring-forming carbon atoms (e.g., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and the like) cyclobutyl, cyclopentyl or cyclohexyl), which is optionally substituted with one or more (such as 1 to 3) suitable substituents, for example methyl-substituted cyclopropyl.
As used herein, the term "alkoxy" means a group having an oxygen atom inserted at any reasonable position in the alkyl group (as defined above), e.g., C 1-8 Alkoxy radical, C 1-6 Alkoxy radical, C 1-4 Alkoxy, or C 1-3 An alkoxy group. C 1-6 Representative examples of alkoxy groups include, but are not limited toLimited to methoxy, ethoxy, propoxy, isopropoxy, n-butoxy, isobutoxy, tert-butoxy, pentoxy, hexoxy, -CH 2 -OCH 3 Etc., which may be optionally substituted with one or more (such as 1 to 3) same or different substituents.
As used herein, the term "halo" or "halogen" group is defined to include fluorine, chlorine, bromine, or iodine.
As used herein, the term "haloalkyl" refers to an alkyl group substituted with one or more (such as 1 to 3) identical or different halogen atoms, the term "C 1-8 Haloalkyl "," C 1-6 Halogenated alkyl radical "and" C 1-3 Haloalkyl "refers to haloalkyl groups having 1 to 8 carbon atoms, 1 to 6 carbon atoms, and 1-3 carbon atoms, respectively, e.g., -CF 3 、-C 2 F 5 、-CHF 2 、-CH 2 F、-CH 2 CF 3 、-CH 2 Cl or-CH 2 CH 2 CF 3 And the like.
The term "heterocyclyl" as used herein, refers to a saturated or partially unsaturated monocyclic or polycyclic group, e.g. having 2, 3, 4,5, 6, 7, 8 or 9 carbon atoms in the ring and one or more (e.g. 1, 2, 3 or 4) selected from nitrogen, oxygen or S (O) m (where m is an integer from 0 to 2) heteroatoms such as, but not limited to, oxiranyl, aziridinyl, azetidinyl, oxetanyl, tetrahydrofuryl, pyrrolidinyl, pyrrolidinonyl, imidazolidinyl, pyrazolidinyl, tetrahydropyranyl, piperidinyl, morpholinyl, dithianyl, thiomorpholinyl, piperazinyl, trithianyl, and the like. Correspondingly, the term "fused heterocyclyl" refers to polycyclic heterocyclyl groups in which each ring shares an adjacent pair of atoms with the other rings in the system, one or more of which rings may contain one or more double bonds, but none of which rings has a fully conjugated pi-electron system, in which one or more of the ring atoms is selected from nitrogen, oxygen or S (O) m (wherein m is an integer of 0 to 2) and the remaining ring atoms are carbon. Can be divided into bicyclic and tricyclic according to the number of constituent ringsA ring, a tetracyclic or polycyclic fused heterocyclic group, for example, including but not limited to a 5-membered/5-membered or 5-membered/6-membered bicyclic fused heterocyclic group.
As used herein, the term "spiroheterocyclyl" means a ring system in the form of a double ring having 7-12 ring atoms, wherein the two rings share 1 carbon atom (referred to as a "spiro atom").
As used herein, the term "aryl" or "aromatic ring" refers to an all-carbon monocyclic or fused ring polycyclic aromatic group having a conjugated pi-electron system. For example, the term "C 6-14 Aryl "or" C 6-14 Aromatic ring "refers to an aromatic group containing 6 to 14 carbon atoms, the term" C 6-10 Aryl "or" C 6-10 Aromatic ring "means containing 6 to 10 an aromatic group of carbon atoms, a group of carbon atoms, such as phenyl (ring) or naphthyl (ring). Aryl is optionally substituted with 1 or more (such as 1 to 3) suitable substituents (e.g. halogen, -OH, -CN, -NO) 2 、C 1-6 Alkyl, etc.).
As used herein, the term "heteroaryl" or "heteroaromatic ring" refers to a monocyclic, bicyclic, or tricyclic aromatic ring system containing at least one heteroatom selected from N, O and S, which for example have 5, 6, 8, 9, 10, 11, 12, 13 or 14 ring atoms and contain 1 or 2 or 3 or 4 or 5 or 6 or 9 or 10 carbon atoms and, in addition, may be benzo-fused in each case. For example, the heteroaryl or heteroaromatic ring may be selected from thienyl (ring), furyl (ring), pyrrolyl (ring), oxazolyl (ring), thiazolyl (ring), imidazolyl (ring), pyrazolyl (ring), isoxazolyl (ring), isothiazolyl (ring), oxadiazolyl (ring), triazolyl (ring), thiadiazolyl (ring), and the like, and benzo derivatives thereof; or pyridyl (ring), pyridazinyl (ring), pyrimidinyl (ring), pyrazinyl (ring), triazinyl (ring), etc., and benzo derivatives thereof.
The term "substituted" means that one or more (e.g., 1, 2, 3, or 4) hydrogens on the designated atom is replaced with a selection from the indicated group, provided that the designated atom's normal valency at the present time is not exceeded and the substitution results in a stable compound. Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.
If a substituent is described as "optionally substituted with …," the substituent may be (1) unsubstituted or (2) substituted. If a carbon of a substituent is described as being optionally substituted with one or more of a list of substituents, then one or more hydrogens on the carbon (to the extent of any hydrogens present) may be replaced individually and/or together with an independently selected optional substituent. If the nitrogen of a substituent is described as being optionally substituted with one or more of the list of substituents, then one or more hydrogens on the nitrogen (to the extent any hydrogen is present) may each be replaced with an independently selected optional substituent.
If a substituent is described as "independently selected from" a group of groups, each substituent is selected independently of the other. Thus, each substituent may be the same or different from another (other) substituent.
As used herein, the term "one or more" means 1 or more than 1, such as 2, 3, 4,5 or 10, under reasonable conditions.
As used herein, unless otherwise indicated, the point of attachment of a substituent may be from any suitable position of the substituent.
When a bond to a substituent is shown across the bond connecting two atoms in a ring, then such substituent may be bonded to any ring-forming atom in the substitutable ring.
The invention also includes all pharmaceutically acceptable isotopically-labeled compounds, which are identical to those of the present invention, except that one or more atoms are replaced by an atom having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number prevailing in nature. Examples of isotopes suitable for inclusion in compounds of the invention include, but are not limited to, isotopes of hydrogen (e.g. hydrogen) 2 H、 3 H. Deuterium D, tritium T); isotopes of carbon (e.g. of 11 C、 13 C and 14 c) Zxfoom Isotopes of chlorine (e.g. of chlorine) 37 Cl); isotopes of fluorine (e.g. of fluorine) 18 F) (ii) a Isotopes of iodine (e.g. of iodine) 123 I and 125 i) (ii) a Isotopes of nitrogen (e.g. of 13 N and 15 n); isotopes of oxygen (e.g. of 15 O、 17 O and 18 o is) (ii) a; isotopes of phosphorus (e.g. of phosphorus) 32 P); and isotopes of sulfur (e.g. of 35 S). Certain isotopically-labeled compounds of the present invention (e.g., those into which a radioisotope is incorporated) are useful in drug and/or substrate tissue distribution studies (e.g., assays). Radioisotope tritium (i.e. tritium 3 H) And carbon-14 (i.e. 14 C) Are particularly useful for this purpose because of their ease of incorporation and ease of detection. Using positron-emitting isotopes (e.g. of the type 11 C、 18 F、 15 O and 13 n) can be used to examine substrate receptor occupancy in Positron Emission Tomography (PET) studies. Isotopically labeled compounds of the present invention can be prepared by processes analogous to those described in the accompanying schemes and/or in the examples and preparations by using an appropriate isotopically labeled reagent in place of the non-labeled reagent previously employed. Pharmaceutically acceptable solvates of the invention include those in which the crystallization solvent may be isotopically substituted, e.g., D 2 O, acetone-d 6 Or DMSO-d 6
The term "stereoisomer" refers to an isomer formed as a result of at least one asymmetric center. In compounds having one or more (e.g., 1, 2, 3, or 4) asymmetric centers, they can give rise to racemic mixtures, single enantiomers, diastereomeric mixtures and individual diastereomers. Certain individual molecules may also exist as geometric isomers (cis/trans). Similarly, the compounds of the invention may exist as mixtures of two or more structurally different forms (commonly referred to as tautomers) in rapid equilibrium. Representative examples of tautomers include keto-enol tautomers, phenol-keto tautomers, nitroso-oxime tautomers, imine-enamine tautomers, and the like. It is understood that the scope of this application encompasses all such isomers or mixtures thereof in any ratio (e.g., 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%).
Solid lines may be used herein
Figure BDA0002325201380000041
Real wedge shaped->
Figure BDA0002325201380000042
Or a virtual wedge>
Figure BDA0002325201380000043
Chemical bonds of the compounds of the present invention are depicted. The use of a solid line to depict a bond to an asymmetric carbon atom is intended to indicate that all possible stereoisomers (e.g., particular enantiomers, racemic mixtures, etc.) at that carbon atom are included. The use of solid or dashed wedges to depict bonds to asymmetric carbon atoms is intended to indicate that the stereoisomers shown are present. When present in a racemic mixture, solid and dotted wedges are used to define the relative stereochemistry, not the absolute stereochemistry. Unless otherwise indicated, the compounds of the present invention are intended to exist in the form of stereoisomers, including cis and trans isomers, optical isomers (e.g., R and S enantiomers), diastereomers, geometric isomers, rotamers, conformers, atropisomers, and mixtures thereof. The compounds of the invention may exhibit more than one type of isomerization and consist of mixtures thereof (e.g., racemic mixtures and diastereomeric pairs).
The present invention encompasses all possible crystalline forms or polymorphs of the compounds of the present invention, which may be single polymorphs or mixtures of more than one polymorph in any ratio.
It will also be appreciated that certain compounds of the invention may be present in free form for use in therapy or, where appropriate, in the form of a pharmaceutically acceptable derivative thereof. In the present invention, pharmaceutically acceptable derivatives include, but are not limited to: pharmaceutically acceptable salts, solvates, metabolites or prodrugs thereof, which upon administration to a patient in need thereof are capable of providing, directly or indirectly, a compound of the present invention or a metabolite or residue thereof. Thus, when reference is made herein to "a compound of the invention," it is also intended to encompass the various derivative forms of the compounds described above.
Pharmaceutically acceptable salts of the compounds of the present invention include acid addition salts and base addition salts thereof.
Suitable acid addition salts are formed from acids which form pharmaceutically acceptable salts. Suitable base addition salts are formed from bases which form pharmaceutically acceptable salts.
A review of suitable Salts is given in Stahl and Wermuth, handbook of Pharmaceutical Salts: properties, selection, and Use (Wiley-VCH, 2002). Methods for preparing pharmaceutically acceptable salts of the compounds of the present invention are known to those skilled in the art.
The compounds of the invention may be present in the form of solvates, preferably hydrates, wherein the compounds of the invention comprise as structural element of the crystal lattice of the compound a polar solvent, such as in particular water, methanol or ethanol. The amount of polar solvent, particularly water, may be present in stoichiometric or non-stoichiometric proportions.
Those skilled in the art will appreciate that not all nitrogen-containing heterocycles are capable of forming N-oxides, since the available lone pair is required for oxidation of the nitrogen to the oxide; one skilled in the art will recognize nitrogen-containing heterocycles that are capable of forming N-oxides. Those skilled in the art will also recognize that tertiary amines are capable of forming N-oxides. Synthetic methods for preparing N-oxides of heterocycles and tertiary amines are well known to those skilled in the art and include oxidation of heterocycles and tertiary amines with peroxy acids such as peracetic and m-chloroperbenzoic acid (MCPBA), hydrogen peroxide, alkyl hydroperoxides such as t-butyl hydroperoxide, sodium perborate, and dioxiranes (dioxiranes) such as dimethyldioxirane. These methods for preparing N-oxides have been widely described and reviewed in the literature, see for example: T.L.Gilchrist, comprehensive Organic Synthesis, vol.7, pp 748-750; a.r.katitzky and a.j.boulton, eds., academic Press; and g.w.h.cheeseman and e.s.g.werstink, advances in Heterocyclic Chemistry, vol.22, pp 390-392, a.r.kattritzky and a.j.boulton, eds., academic Press.
Also included within the scope of the present invention are metabolites of the compounds of the invention, i.e., substances formed in vivo upon administration of the compounds of the invention. Such products may result, for example, from oxidation, reduction, hydrolysis, amidation, deamidation, esterification, enzymatic hydrolysis, etc., of the administered compound. Accordingly, the present invention includes metabolites of the compounds of the present invention, including compounds made by the process of contacting the compounds of the present invention with a mammal for a time sufficient to produce a metabolite thereof.
The present invention further includes within its scope prodrugs of the compounds of the present invention which are certain derivatives of the compounds of the present invention which may themselves have little or no pharmacological activity which, when administered into or onto the body, may be converted to the compounds of the present invention having the desired activity by, for example, hydrolytic cleavage. Typically such prodrugs will be functional derivatives of the compounds which are readily convertible in vivo into the desired therapeutically active compound. Further information on the use of prodrugs can be found in "Pro-drugs as Novel Delivery Systems", volume 14, ACS Symposium Series (T.Higuchi and V.Stella) and "Bioreversible Carriers in Drug Design," Pergamon Press,1987 (E.B.Roche editions, american Pharmaceutical Association). Prodrugs of the invention may be prepared, for example, by replacing appropriate functional groups present in a compound of the invention with certain moieties known to those skilled in the art as "pro-moieties", for example as described in "Design of Prodrugs", h.
The invention also encompasses compounds of the invention containing a protecting group. In any process for preparing the compounds of the present invention, it may be necessary and/or desirable to protect sensitive or reactive groups on any of the molecules concerned, thereby forming a chemically protected form of the compounds of the present invention. This can be achieved by conventional protecting Groups, for example, as described in Protective Groups in Organic Chemistry, ed.j.f.w.mcomie, plenum Press,1973; and T.W.Greene & P.G.M.Wuts, protective Groups in Organic Synthesis, john Wiley & Sons,1991, which are incorporated herein by reference. The protecting group may be removed at a suitable subsequent stage using methods known in the art.
The term "about" means within. + -. 10%, preferably within. + -. 5%, more preferably within. + -. 2% of the stated value.
Detailed Description
Compound (I)
In one aspect, the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite, or prodrug thereof:
Figure BDA0002325201380000051
wherein:
x is selected from N and CH;
R 1 selected from hydrogen, halogen, cyano, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 3-6 Cycloalkyl, carboxyl, C 1-6 alkyl-OC (O) -, R a R b N-C (O) -, 5-6 membered heterocyclyl and 5-6 membered heteroaryl, wherein the heterocyclyl and heteroaryl are optionally substituted with substituents independently selected from the group consisting of: halogen, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 3-6 Cycloalkyl radical, C 1-6 Alkoxy radical, R a R b N-、R a R b N-C (O) -, hydroxy-C 1-6 Alkyl-, C 1-6 alkoxy-C 1-6 Alkyl-, R a R b N-C 1-6 Alkyl-and R a R b N-C(O)-C 1-6 Alkyl-;
R 2 selected from hydrogen, halogen, cyano, C 1-6 Alkyl radical, C 1-6 Haloalkyl and C 1-6 An alkoxy group;
R 3 selected from hydrogen, halogen, C 1-6 Alkyl and C 1-6 A haloalkyl group;
R 4 selected from hydrogen, halogen, C 1-6 Alkyl and C 1-6 A haloalkyl group;
R a and R b Each independently selected from hydrogen and C 1-6 Alkyl radical, C 3-6 Cycloalkyl, hydroxy-C 1-6 Alkyl-, C 1-6 alkoxy-C 1-6 Alkyl-, R c R d N-C(O)-C 1-6 Alkyl-and 5-6 membered heterocyclyl-C 1-6 Alkyl-;
R c and R d Each independently selected from hydrogen and C 1-6 Alkyl radical, C 3-6 Cycloalkyl, hydroxy C 1-6 Alkyl radical, C 1-6 alkoxy-C 1-6 Alkyl-and 5-6 membered heterocyclyl-C 1-6 Alkyl-;
n is selected from 0, 1 or 2;
p is selected from 0, 1 or 2;
q is selected from 0, 1 or 2.
In some embodiments, the present invention provides a compound as described above, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite, or prodrug thereof, wherein:
R a and R b Each independently selected from hydrogen and C 1-6 Alkyl radical, C 3-6 Cycloalkyl, hydroxy C 1-6 Alkyl radical, C 1-6 alkoxy-C 1-6 Alkyl-and 5-6 membered heterocyclyl-C 1-6 An alkyl group-.
In some embodiments, the present invention provides a compound as described above, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite, or prodrug thereof, wherein:
R 1 selected from hydrogen, halogen, cyano, C 1-4 Alkyl radical, C 1-4 Haloalkyl, C 3-6 Cycloalkyl, carboxyl, C 1-4 alkyl-OC (O) -, R a R b N-C (O) -, 5-6 membered heterocyclyl and 5-6 membered heteroaryl, wherein the heterocyclyl and heteroaryl are optionally substituted with substituents independently selected from the group consisting of: halogen, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 3-6 Cycloalkyl, C 1-6 Alkoxy radical, R a R b N-、R a R b N-C (O) -, hydroxy-C 1-6 Alkyl-, C 1-6 alkoxy-C 1-6 Alkyl-, R a R b N-C 1-6 Alkyl-and R a R b N-C(O)-C 1-6 Alkyl-;
preferably, R 1 Selected from hydrogen, halogen, cyano, C 1-4 Alkyl radical, C 1-4 Haloalkyl, C 3-6 Cycloalkyl, carboxy, C 1-4 alkyl-OC (O) -, R a R b N-C (O) -, 5-6 membered heterocyclyl and 5-6 membered heteroaryl, wherein the heterocyclyl and heteroaryl are optionally substituted with substituents independently selected from the group consisting of: halogen, C 1-4 Alkyl radical, C 1-4 Haloalkyl, C 3-6 Cycloalkyl radical, C 1-4 Alkoxy radical, R a R b N-、R a R b N-C (O) -, hydroxy-C 1-4 Alkyl-, C 1-4 alkoxy-C 1-4 Alkyl-, R a R b N-C 1-4 Alkyl-and R a R b N-C(O)-C 1-4 Alkyl-;
preferably, R 1 Selected from hydrogen, halogen, cyano, C 1-4 Alkyl radical, C 3-6 Cycloalkyl, carboxy, C 1-4 alkyl-OC (O) -, R a R b N-C(O)-。
In some embodiments, the present invention provides a compound as described above, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite, or prodrug thereof, wherein: r 1 Selected from cyano and R a R b N-C(O)-。
In some embodiments, the present invention provides a compound as described above, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite, or prodrug thereof, wherein:
R a and R b Each independently selected from hydrogen and C 1-3 Alkyl radical, C 3-6 Cycloalkyl, hydroxy-C 1-3 Alkyl-, C 1-3 alkoxy-C 1-3 Alkyl-, R c R d N-C(O)-C 1-3 Alkyl-and 5-6 membered heterocyclyl-C 1-3 Alkyl-; preferably, R a And R b Each independently selected from hydrogen and C 1-3 Alkyl and R c R d N-C(O)-C 1-3 Alkyl-;
preferably, R a And R b Each independently selected from hydrogen, methyl, ethyl, propyl, R c R d N-C(O)-CH 2 -、R c R d N-C(O)-(CH 2 ) 2 -and R c R d N-C(O)-(CH 2 ) 3 -;
Preferably, R a And R b Each independently selected from hydrogen, methyl and R c R d N-C(O)-CH 2 -。
In some embodiments, the present invention provides a compound as described above, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite, or prodrug thereof, wherein: r a Is H, and R b Is hydrogen, methyl or R c R d N-C(O)-CH 2 -。
In some embodiments, the present invention provides a compound as described above, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite, or prodrug thereof, wherein:
R a and R b Each independently selected from hydrogen and C 1-3 Alkyl radical, C 3-6 Cycloalkyl, hydroxy-C 1-3 Alkyl-, C 1-3 alkoxy-C 1-3 Alkyl-and 5-6 membered heterocyclyl-C 1-3 Alkyl-;
preferably, R a And R b Each independently selected from hydrogen and C 1-3 An alkyl group;
preferably, R a And R b Each independently selected from hydrogen, methyl, ethyl and propyl;
preferably, R a And R b Each independently hydrogen.
In some embodiments, the present invention provides a compound as described above, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite, or prodrug thereof, wherein:
R c and R d Each independently selected from hydrogen and C 1-3 Alkyl radical, C 3-6 Cycloalkyl, hydroxy-C 1-3 Alkyl-, C 1-3 alkoxy-C 1-3 Alkyl-and 5-6 membered heterocyclyl-C 1-3 Alkyl-;
preferably, R c And R d Each independently selected from hydrogen and C 1-3 Alkyl radical (ii) a;
preferably, R c And R d Each independently selected from hydrogen, methyl, ethyl and propyl;
preferably, R c And R d Each independently hydrogen.
In some embodiments, the present invention provides a compound as described above, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite, or prodrug thereof, wherein: r 1 Is a cyano group.
In some embodiments, the present invention provides a compound as described above, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite, or prodrug thereof, wherein: r 1 Is H 2 N-C(O)-、NH(CH 3 ) -C (O) -or H 2 N-C(O)-CH 2 -NH-C(O)-。
In some embodiments, the present invention provides a compound as described above, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite, or prodrug thereof, wherein: r 1 Is H 2 N-C(O)-。
In some embodiments, the present invention provides a compound as described above, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite, or prodrug thereof, wherein:
R 2 selected from hydrogen, halogen, cyano, C 1-3 An alkyl group C 1-3 Haloalkyl and C 1-3 An alkoxy group;
preferably, R 2 Selected from the group consisting of hydrogen, fluoro, chloro, bromo, iodo, cyano, methyl, ethyl and propyl;
preferably, the first and second liquid crystal display panels are, R is 2 Is composed of fluorine.
In some embodiments, the present invention provides a compound as described above, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite, or prodrug thereof, wherein:
R 3 selected from hydrogen, halogen, C 1-3 Alkyl and C 1-3 A haloalkyl group;
preferably, R 3 Selected from hydrogen, fluoro, chloro, bromo, iodo, cyano, methyl, ethyl, and propyl;
preferably, the first and second electrodes are formed of a metal, R 3 Is hydrogen.
In some embodiments, the present invention provides a compound as described above, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite, or prodrug thereof, wherein:
R 4 selected from hydrogen, halogen, C 1-3 Alkyl radical and C 1-3 A haloalkyl group;
preferably, R 4 Selected from the group consisting of hydrogen, fluoro, chloro, bromo, iodo, cyano, methyl, ethyl and propyl;
preferably, R 4 Is methyl.
In some embodiments, the present invention provides a compound as described above, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite, or prodrug thereof, wherein: n is selected from 0 or 1; preferably, n is 1.
In some embodiments, the present invention provides a compound as described above, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite, or prodrug thereof, wherein: p is selected from 0 or 1; preferably, p is 1.
In some embodiments, the present invention provides a compound as described above, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite, or prodrug thereof, wherein: q is selected from 0 or 1; preferably, q is 1.
In some embodiments, the present invention provides a compound as described above, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite, or prodrug thereof, wherein:
the compound has the structure of formula (Ia):
Figure BDA0002325201380000071
wherein R is a 、R b 、R 2 、R 3 、R 4 X, n, p, q are as defined above for general formula (I);
preferably, the compound has the structure of formula (Ia-1), formula (Ia-2), or formula (Ia-3):
Figure BDA0002325201380000081
wherein R is 2 、R 3 、R 4 X, n, p, q are as defined above for formula (I).
In some embodiments, the present invention provides a compound as described above, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite, or prodrug thereof, wherein:
the compound has the structure of formula (I-5):
Figure BDA0002325201380000082
wherein R is 2 、R 3 、R 4 X, n, p, q are as defined above for formula (I).
In some embodiments, the present invention provides a compound as described above, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite, or prodrug thereof, wherein X is N; in other embodiments, X is CH.
In some embodiments, the present invention provides a compound as described above, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite, or prodrug thereof, wherein:
R 2 selected from hydrogen, halogen, cyano, C 1-3 Alkyl radical, C 1-3 Haloalkyl and C 1-3 Alkoxy, preferably halogen, more preferably F;
R 3 selected from hydrogen, halogen, C 1-3 Alkyl and C 1-3 Haloalkyl, preferably hydrogen;
R 4 selected from hydrogen, halogen, C 1-3 Alkyl and C 1-3 Haloalkyl, preferably C 1-3 Alkyl, more preferably methyl; and is
n, p and q are each 1.
In still other embodiments, the present invention provides a compound as described above, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite, or prodrug thereof, having the structure of formula (II):
Figure BDA0002325201380000083
wherein R is 1 、R 2 、R 3 、R 4 X, p are as defined above for formula (I).
In some such embodiments, X is N; in other embodiments, X is CH.
In some embodiments, the present invention provides a compound of formula (I) or formula (II) as described above, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite, or prodrug thereof, wherein:
x is N;
R 1 selected from hydrogen, halogen, cyano, C 1-4 Alkyl radical, C 1-4 Haloalkyl, C 3-6 Cycloalkyl, carboxy, C 1-4 alkyl-OC (O) -, R a R b N-C (O) -, 5-6 membered heterocyclyl and 5-6 membered heteroaryl, wherein the heterocyclyl and heteroaryl are optionally substituted with substituents independently selected from the group consisting of: halogen, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 3-6 Cycloalkyl radical, C 1-6 Alkoxy radical, R a R b N-、R a R b N-C (O) -, hydroxy-C 1-6 Alkyl-, C 1-6 alkoxy-C 1-6 Alkyl-, R a R b N-C 1-6 Alkyl-and R a R b N-C(O)-C 1-6 Alkyl-;
R 2 selected from hydrogen, halogen, cyano, C 1-3 Alkyl radical, C 1-3 Haloalkyl and C 1-3 An alkoxy group;
R 3 selected from hydrogen, halogen, C 1-3 Alkyl and C 1-3 A haloalkyl group;
R 4 selected from hydrogen, halogen, C 1-3 Alkyl and C 1-3 A haloalkyl group;
R a and R b Each independently selected from hydrogen and C 1-3 Alkyl radical, C 3-6 Cycloalkyl, hydroxy-C 1-3 Alkyl-, C 1-3 alkoxy-C 1-3 Alkyl-, R c R d N-C(O)-C 1-6 Alkyl-and 5-6 membered heterocyclyl-C 1-3 Alkyl-;
R c and R d Each independently selected from hydrogen, C 1-3 Alkyl radical, C 3-6 Cycloalkyl, hydroxy-C 1-3 Alkyl-, C 1-3 alkoxy-C 1-3 Alkyl-and 5-6 membered heterocyclyl-C 1-3 Alkyl-;
n is selected from 0 or 1;
p is selected from 0 or 1;
q is selected from 0 or 1.
In some embodiments, the present invention provides a compound of formula (I) or formula (II) as described above, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite, or prodrug thereof, wherein:
R a and R b Each independently selected from hydrogen, C 1-3 Alkyl radical, C 3-6 Cycloalkyl, hydroxy-C 1-3 Alkyl-, C 1-3 alkoxy-C 1-3 Alkyl-and 5-6 membered heterocyclyl-C 1-3 An alkyl group-.
In some such embodiments, the present invention provides a compound of formula (I) or formula (II) as described above, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically-labeled compound, metabolite or prodrug thereof, wherein:
x is N;
R 1 selected from cyano, H 2 N-C(O)-、NH(CH 3 ) -C (O) -and H 2 N-C(O)-CH 2 -NH-C(O)-;
R 2 Selected from halogen and cyano, preferably from fluorine, chlorine, bromine, iodine and cyano, preferably F;
R 3 is hydrogen;
R 4 is selected from C 1-3 Alkyl, preferably methyl;
n, p and q are each 1.
In other such embodiments, the present invention provides a compound of formula (I) or formula (II) as described above, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically-labeled compound, metabolite or prodrug thereof, wherein:
x is N;
R 1 selected from cyanoAnd H 2 N-C(O)-;
R 2 Is halogen; preferably, R 2 Is fluorine;
R 3 is hydrogen;
R 4 is methyl;
R a and R b Each independently is hydrogen;
n is 1; p is 0; q is 1.
In some embodiments, the present invention provides a compound as described above, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite, or prodrug thereof, wherein the compound is selected from the group consisting of:
Figure BDA0002325201380000091
Figure BDA0002325201380000101
preparation method
In another aspect, the present application provides methods of making the compounds of the present invention.
In some embodiments, the present invention provides a method of preparing a compound of formula (Ia-1):
reaction scheme 1
Figure BDA0002325201380000102
Wherein: r is 2 、R 3 、R 4 X, n, p, q are as defined above for formula (I), Y is a boronic acid or boronic ester group, preferably-B (OH) 2 Or
Figure BDA0002325201380000103
The method comprises the following steps:
(1) Reacting the compound I-1 with a compound IN-a to obtain a compound I-2;
the compound I-1 and the compound IN-a are subjected to coupling reaction to obtain a compound I-2. The coupling reaction is preferably carried out in the presence of a metal catalyst and a base. The metal catalyst is a palladium metal catalyst such as tetrakis (triphenylphosphine) palladium, [1,1 '-bis (diphenylphosphino) ferrocene ] palladium dichloride, [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride dichloromethane complex, bis (triphenylphosphine) palladium dichloride, palladium acetate, preferably tetrakis (triphenylphosphine) palladium. The base is an inorganic base such as potassium carbonate, cesium carbonate, sodium bicarbonate, potassium bicarbonate, preferably potassium carbonate. The coupling reaction is carried out in a suitable organic solvent or a mixed solvent of an organic solvent and water, the organic solvent may be selected from 1,4-dioxane, N-dimethylformamide, methanol, ethanol and toluene, preferably a mixed solvent of 1,4-dioxane and water. Preferably, the coupling reaction is carried out under a suitable protective atmosphere (e.g. nitrogen atmosphere). The coupling reaction is carried out at a suitable temperature, which may be 70 to 100 ℃, preferably 80 ℃. Preferably, the coupling reaction is carried out for a suitable time, which may be 1 to 24 hours, for example 6 hours.
(2) Reacting the compound I-2 with a halogenating agent to obtain a compound I-3;
the compound I-2 reacts with a halogenating agent in a solvent to obtain a compound I-3. The halogenating agent used may be iodine monochloride or N-iodosuccinimide, preferably iodine monochloride. The solvent is selected from N, N-dimethylformamide, N-methylpyrrolidone, glacial acetic acid, methanol, ethanol, tetrahydrofuran, 1,4-dioxane and the like, preferably glacial acetic acid. The reaction temperature is usually from-20 ℃ to room temperature, preferably room temperature. The reaction time is usually 10 to 18 hours, preferably 12 hours.
(3) Reacting the compound I-3 with the compound IN-b to obtain a compound I-4;
the compound I-3 and the compound IN-b are subjected to coupling reaction to obtain a compound I-4. The coupling reaction is preferably carried out in the presence of a metal catalyst and a base. The metal catalyst is a palladium metal catalyst such as tetrakis (triphenylphosphine) palladium, [1,1 '-bis (diphenylphosphino) ferrocene ] palladium dichloride, [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride dichloromethane complex, bis (triphenylphosphine) palladium dichloride, palladium acetate, preferably tetrakis (triphenylphosphine) palladium. The base is an inorganic base such as potassium carbonate, cesium carbonate, sodium bicarbonate, potassium bicarbonate, preferably potassium carbonate. The coupling reaction is carried out in a suitable organic solvent or a mixed solvent of an organic solvent and water, wherein the organic solvent can be 1,4-dioxane, N-dimethylformamide, methanol, ethanol and toluene, and preferably is a mixed solvent of 1,4-dioxane and water. Preferably, the coupling reaction is carried out under a suitable protective atmosphere (e.g. nitrogen atmosphere). The coupling reaction is carried out at a suitable temperature, which may be 70-100 ℃, preferably 80 ℃. Preferably, the coupling reaction is carried out for a suitable time, which may be 1 to 24 hours, for example 6 hours.
(4) Reacting the compound I-4 with IN-c to obtain a compound I-5
The compound I-4 and the compound IN-c are subjected to coupling reaction to obtain a compound I-5. The coupling reaction is preferably carried out in the presence of a metal catalyst. The metal catalyst may be selected from tris (dibenzylideneacetone) dipalladium, tetrakis (triphenylphosphine) palladium, [1,1 '-bis (diphenylphosphino) ferrocene ] palladium dichloride, [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride dichloromethane complex, bis (triphenylphosphine) palladium dichloride, palladium acetate, preferably tris (dibenzylideneacetone) dipalladium. The coupling reaction may be carried out with the addition of a suitable ligand, for example 1,1 '-bis (diphenylphosphino) ferrocene, 1.1' -binaphthyl-2.2 '-diphenylphosphine, preferably 1,1' -bis (diphenylphosphino) ferrocene. The coupling reaction is carried out in a suitable organic solvent which may be selected from 1,4-dioxane, N-dimethylformamide, methanol, ethanol, toluene, preferably N, N-dimethylformamide. Preferably, the coupling reaction is carried out under a suitable protective atmosphere (e.g. nitrogen atmosphere). The reaction temperature is generally from 90 to 140 ℃ and preferably 100 ℃. The reaction time is usually 10 to 18 hours, preferably 12 hours.
(5) Compound I-5 is hydrolyzed to obtain A compound of formula (I-A)
Reacting the compound I-5 with a base in a solvent to obtain the compound of the formula (I-A). The base may be selected from potassium carbonate, sodium carbonate, cesium carbonate, sodium hydroxide, potassium hydroxide, preferably potassium carbonate. Preferably, hydrogen peroxide can be added in the reaction. The reaction is carried out in a suitable organic solvent which may be selected from toluene, 1,4-dioxane, dimethyl sulfoxide, water, preferably dimethyl sulfoxide. The reaction temperature is preferably room temperature (25-30 ℃). The reaction time is usually 2 to 10 hours, preferably 2 hours.
In some embodiments, the present invention provides a method of preparing a compound of formula (Ia):
reaction scheme 2
Figure BDA0002325201380000111
Wherein R is a 、R b 、R 2 、R 3 、R 4 X, n, p, q are as described above for general formula (I);
the method comprises the following steps:
(1) The compound I-4 is subjected to carbonyl insertion reaction to obtain a compound I-6;
the compound I-4 is subjected to carbonyl insertion reaction to obtain a compound I-6. The carbonylation reaction is preferably carried out in the presence of a metal catalyst and a base. The metal catalyst is a palladium metal catalyst, such as bis (acetonitrile) palladium (II) chloride, [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride, [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride dichloromethane complex or palladium acetate, preferably [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride. The base may be selected from triethylamine and N, N-diisopropylethylamine, preferably triethylamine. The carbonylation reaction is carried out in a suitable organic solvent which may be selected from N, N-dimethylformamide, methanol and mixtures thereof, preferably a mixed solvent of N, N-dimethylformamide and methanol. The carbonyl insertion reaction is carried out under a carbon monoxide atmosphere. The carbonylation reaction is carried out at a suitable temperature, which may be 60 to 100 c, preferably 90 c. Preferably, the carbonylation reaction is carried out for a suitable time, which may be from 1 to 24 hours, for example 24 hours.
(2) Hydrolyzing the compound I-6 to obtain a compound I-7;
reacting the compound I-6 with alkali in a solvent to obtain a compound I-7. The base may be selected from sodium hydroxide, potassium hydroxide, lithium hydroxide, preferably sodium hydroxide. The reaction is carried out in a suitable solvent mixture of an organic solvent and water, which may be selected from the group consisting of tetrahydrofuran, 1,4-dioxane, methanol, ethanol, acetonitrile, and mixtures of two or more thereof, preferably methanol. The reaction temperature is preferably room temperature (25-30 ℃). The reaction time is usually 6 to 12 hours, preferably 12 hours.
(3) Carrying out condensation reaction on the compound I-7 and IN-d to obtain a compound shown IN a formula (Ia);
the compound I-7 and IN-d are subjected to condensation reaction to obtain the compound shown IN the formula (Ia). The condensation reaction is preferably carried out in the presence of a condensing agent. The condensing agent can be selected from 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethylurea hexafluorophosphate, 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride and carbonyldiimidazole, preferably carbonyldiimidazole. The reaction is carried out in a suitable organic solvent which may be selected from tetrahydrofuran, dichloromethane, N-dimethylformamide and acetonitrile, preferably N, N-dimethylformamide. The reaction temperature is preferably room temperature (25-30 ℃). Preferably, the condensation reaction is carried out for a suitable time, which may be 1 to 24 hours, for example 12 hours.
Pharmaceutical composition and pharmaceutical preparation
In some embodiments, the present invention provides pharmaceutical compositions comprising a prophylactically or therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite, or prodrug thereof, and one or more pharmaceutically acceptable carriers.
By "pharmaceutically acceptable carrier" in the context of the present invention is meant a diluent, adjuvant, excipient, or vehicle that is administered together with a therapeutic agent and which is, within the scope of sound medical judgment, suitable for contact with the tissues of humans and/or other animals without excessive toxicity, irritation, allergic response, or other problem or complication commensurate with a reasonable benefit/risk ratio.
Pharmaceutically acceptable carriers that may be employed in the pharmaceutical compositions of the present invention include, but are not limited to, sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water is an exemplary carrier when the pharmaceutical composition is administered intravenously. Physiological saline and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions. Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, maltose, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene glycol, water, ethanol and the like. The composition may also optionally contain minor amounts of wetting agents, emulsifying agents, or pH buffering agents. Oral formulations may contain standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate and the like. Examples of suitable pharmaceutically acceptable carriers are described in Remington's Pharmaceutical Sciences (1990).
In some embodiments, the present invention provides a pharmaceutical formulation comprising a compound of the present invention, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug thereof, or a mixture thereof, or a pharmaceutical composition of the present invention. The pharmaceutical composition or pharmaceutical formulation of the invention may act systemically and/or locally. For this purpose, they may be administered by a suitable route, for example by injection (e.g. intravenous, intra-arterial, subcutaneous, intraperitoneal, intramuscular injection, including instillation) or transdermally; or by oral, buccal, nasal, transmucosal, topical, in the form of ophthalmic preparations or by inhalation.
For these routes of administration, the pharmaceutical compositions of the present invention may be administered in suitable dosage forms. Such dosage forms include, but are not limited to, tablets, capsules, lozenges, hard candies, powders, sprays, creams, ointments, suppositories, gels, pastes, lotions, ointments, aqueous suspensions, injectable solutions, elixirs, syrups, and the like.
The compound of the invention may be present in the pharmaceutical composition or pharmaceutical formulation in an amount or amount of about 0.01mg to about 1000mg.
In some embodiments, the present invention provides a method of making a pharmaceutical composition of the present invention, the method comprising combining a compound of the present invention, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite, or prodrug thereof, with one or more pharmaceutically acceptable carriers.
Methods of treatment and uses
In some embodiments, the present invention provides the use of a compound of the present invention, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite, or prodrug thereof, or a pharmaceutical composition of the present invention, in the manufacture of a medicament for the prevention or treatment of an adenosine A2a receptor associated disease.
In some embodiments, the present invention provides a compound of the present invention, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug thereof, or a pharmaceutical composition of the present invention, for use in inhibiting adenosine A2a receptor activity.
In some embodiments, the present invention provides a compound of the present invention or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug thereof, or a pharmaceutical composition of the present invention, for use in the prevention or treatment of an adenosine A2a receptor associated disease.
In some embodiments, the present invention provides a method of preventing or treating an adenosine A2a receptor associated disease, comprising administering to a subject in need thereof an effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug thereof, or a pharmaceutical composition of the present invention.
In some embodiments, the adenosine A2a receptor associated disease is a tumor, preferably, the disease is cancer.
The term "effective amount" as used herein refers to an amount of a compound that, when administered, will alleviate one or more symptoms of the condition being treated to some extent.
The dosing regimen may be adjusted to provide the best desired response. For example, a single bolus may be administered, several divided doses may be administered over time, or the dose may be proportionally reduced or increased as the therapeutic situation dictates. It is noted that dosage values may vary with the type and severity of the condition being alleviated, and may include single or multiple doses. It is further understood that for any particular individual, the specific dosage regimen will be adjusted over time according to the individual need and the professional judgment of the person administering the composition or supervising the administration of the composition.
The amount of a compound of the invention administered will depend on the subject being treated, the severity of the disorder or condition, the rate of administration, the disposition of the compound, and the judgment of the prescribing physician. Generally, an effective dose is from about 0.0001 to about 50mg per kg body weight per day, e.g., from about 0.01 to about 10 mg/kg/day (single or divided administration). For a 70kg human, this may amount to about 0.007 mg/day to about 3500 mg/day, for example about 0.7 mg/day to about 700 mg/day. In some cases, dosage levels no higher than the lower limit of the aforesaid range may be sufficient, while in other cases still larger doses may be employed without causing any harmful side effects, provided that the larger dose is first divided into several smaller doses for administration throughout the day.
Unless otherwise indicated, the term "treating," as used herein, means reversing, alleviating, inhibiting the progression of, or one or more symptoms of, the disorder or condition to which such term applies.
As used herein, "individual" includes a human or non-human animal. Exemplary human individuals include human individuals (referred to as patients) having a disease (e.g., a disease described herein) or normal individuals. "non-human animals" in the context of the present invention include all vertebrates, such as non-mammals (e.g., birds, amphibians, reptiles) and mammals, such as non-human primates, livestock and/or domesticated animals (e.g., sheep, dogs, cats, cows, pigs, etc.).
Examples
Embodiments of the present invention will be described in detail below with reference to examples, but it will be understood by those skilled in the art that the following examples are only illustrative of the present invention and should not be construed as limiting the scope of the present invention. The examples, in which specific conditions are not specified, were conducted under conventional conditions or conditions recommended by the manufacturer. The reagents or instruments used are not indicated by the manufacturer, and are all conventional products commercially available.
The structure of the compound is determined by nuclear magnetic resonance 1 H NMR) or Mass Spectrometry (MS). 1 The H NMR was measured using a JEOL Eclipse 400 NMR spectrometer with a solvent selected from deuterated methanol (CD) 3 OD), deuterated chloroform (CDCl) 3 ) And hexadeuterio dimethyl sulfoxide (DMSO-d) 6 ) Internal standard is Tetramethylsilane (TMS), and chemical shift (delta) is 10 -6 (ppm) is given as a unit.
MS was determined using an Agilent (ESI) mass spectrometer, manufacturer: agilent, model: agilent 6120B.
When a preparative high performance liquid chromatograph is adopted for purification, the adopted instrument model is Agilent 1260, and a chromatographic column: waters SunAire Prep C18 OBD (19 mm. Times.150 mm. Times.5.0 μm); temperature of the chromatographic column: 25 ℃; flow rate: 20.0mL/min; detection wavelength: 214nm; elution gradient: (0min; mobile phase A:100% acetonitrile; mobile phase B:0.05% aqueous formic acid.
Thin layer chromatography silica gel plate (TLC) an aluminum plate (20X 20 cm) from Merck was used, and the specification for separation and purification by thin layer chromatography was GF 254 (1 mm) from Nicotiana.
The reaction was monitored by Thin Layer Chromatography (TLC) or LC-MS using a developing system of: dichloromethane and methanol system, n-hexane and ethyl acetate system, petroleum ether and ethyl acetate system, and volume ratio of solvent is regulated according to different polarities of the compounds or by adding triethylamine and the like.
The microwave reaction was carried out using a Biotage Initiator + (400W, RT-300 ℃ C.) microwave reactor.
Silica gel column chromatography: the column chromatography generally uses 200-300 mesh silica gel as a carrier. The system of eluents comprises: the volume ratio of the solvent is adjusted according to different polarities of the compounds in a dichloromethane and methanol system and a petroleum ether and ethyl acetate system, and a small amount of triethylamine can also be added for adjustment.
In the following examples, the reaction temperature is room temperature (20 ℃ C. To 35 ℃ C.), unless otherwise specified.
The reagents used in the present invention were purchased from Acros Organics, aldrich Chemical Company, texas Chemical, and the like.
In the conventional synthesis methods and examples of the compounds of the present invention, and intermediate synthesis examples, each abbreviation has the following meaning:
abbreviations Means of
DMF N, N-dimethylformamide
Pd 2 (dba) 3 Tris (dibenzylideneacetone) dipalladium
dppf 1,1' -bis (diphenylphosphino) ferrocene
TLC Thin layer chromatography
Pd(dppf)Cl 2 [1,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride
Preparation of intermediates
Intermediate preparation example 1: preparation of 4-methyl-6- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl) quinazoline (In-1)
Figure BDA0002325201380000141
The first step is as follows: preparation of 6-bromo-4-methyl quinazoline (In-1-b)
1- (2-amino-5-bromophenyl) ethanone (In-1-a) (3g, 14mmol), triethyl orthoformate (3.1g, 21mmol) and ammonium acetate (1.62g, 21mmol) were added to a reaction flask and reacted at 100 ℃ overnight. After completion of the reaction, it was cooled to room temperature, the reaction solution was concentrated, the residue was diluted with water and extracted with ethyl acetate, the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated, and the residue was purified by silica gel column chromatography (eluent: ethyl acetate/petroleum ether = 1/3) to obtain the title compound of this step (2.6 g, yield: 83%).
MS m/z(ESI):223.0[M+H] +
The second step is that: preparation of 4-methyl-6- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl) quinazoline (In-1)
To a solution of 6-bromo-4-methyl quinazoline (2g, 8.9 mmol) in 1,4-dioxane (50 mL) under a nitrogen atmosphere was added sequentially pinacoldiboron diboron (3.4 g,13.5 mmol), potassium acetate (1.76g, 16.9 mmol) and Pd (dppf) Cl 2 (0.65g, 0.9mmol), the reaction was carried out at 80 ℃ for 6 hours. After completion of the reaction, it was cooled to room temperature, the reaction solution was concentrated, the residue was diluted with water and extracted with ethyl acetate, the organic phases were combined, dried over anhydrous sodium sulfate, filtered, concentrated, and the residue was purified by silica gel column chromatography (eluent: ethyl acetate/petroleum ether = 1/2) to obtain the title compound (2.1 g, collectedRate: 87%).
MS m/z(ESI):271.2[M+H] +
Preparation of the compounds of the invention:
example 1: preparation of 2-amino-6- (4-fluorophenyl) -5- (4-methylquinazolin-6-yl) pyrimidine-4-carbonitrile (1)
Figure BDA0002325201380000142
The first step is as follows: preparation of 4-chloro-6- (4-fluorophenyl) pyrimidin-2-amine
4,6-dichloro-2-aminopyrimidine (5g, 30.6 mmol), 4-fluorophenylboronic acid (4.5g, 32.1mmol) and potassium carbonate (8.5g, 61.2mmol) were added to 1,4-dioxane (20 mL) and water (4 mL), nitrogen was replaced three times, tetrakis (triphenylphosphine) palladium (1.7 g, 1.5mmol) was added, and the reaction was carried out at 80 ℃ for 6 hours. After completion of the reaction, it was cooled to room temperature, the reaction solution was concentrated, and the residue was purified by silica gel column chromatography (eluent: ethyl acetate/petroleum ether = 1/5) to obtain the title compound of the present step (6.5 g, yield: 95%).
MS m/z(ESI):224.0[M+H] +
The second step is that: preparation of 4-chloro-6- (4-fluorophenyl) -5-iodo-2-aminopyrimidine
4-chloro-6- (4-fluorophenyl) pyrimidin-2-amine (5g, 22.4 mmol) was dissolved in acetic acid (50 mL), and a solution of iodine monochloride (10.9g, 67.2 mmol) in acetic acid (10 mL) was added dropwise at room temperature, and the reaction was continued at room temperature for 12 hours. The reaction solution was diluted with water, extracted with ethyl acetate, and the organic phases were combined and washed successively with aqueous sodium bisulfite and brine. The organic phase was dried, filtered, and the filtrate was concentrated to obtain the title compound of this step (7 g, yield: 89.7%).
MS m/z(ESI):349.9[M+H] +
The third step: preparation of 4-chloro-6- (4-fluorophenyl) -5- (4-methylquinazolin-6-yl) pyrimidin-2-amine
4-chloro-6- (4-fluorophenyl) -5-iodo-2-aminopyrimidine (3g, 8.6 mmol), 4-methyl-6- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl) quinazoline (2.5g, 9.5 mmol) and potassium carbonate (2.4g, 17.2 mmol) were added to 1,4-dioxane (40 mL) and water (10 mL), nitrogen was replaced three times, tetrakis (triphenylphosphine) palladium (0.55g, 0.5 mmol) was added, and the reaction was carried out at 80 ℃ for 6 hours. After completion of the reaction, it was cooled to room temperature, the reaction solution was concentrated, and the residue was purified by silica gel column chromatography (eluent: ethyl acetate/petroleum ether = 1/1) to obtain the title compound of this step (1 g, yield: 75%).
MS m/z(ESI):366.1[M+H] +
The fourth step: preparation of 2-amino-6- (4-fluorophenyl) -5- (4-methylquinazolin-6-yl) pyrimidine-4-carbonitrile (1)
4-chloro-6- (4-fluorophenyl) -5- (4-methylquinazolin-6-yl) pyrimidin-2-amine (0.4 g, 1.1mmol), zinc cyanide (0.13g, 1.1mmol) and dppf (60mg, 0.1mmol) were added to DMF (15 mL) with nitrogen replaced three times and Pd added 2 (dba) 3 (0.1g, 0.1mmol) and reacted at 100 ℃ for 12 hours. After the reaction is finished, cooling to room temperature, adding water for dilution, and extracting by ethyl acetate. The organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated, and the residue was purified by silica gel column chromatography (eluent: ethyl acetate/petroleum ether = 1/1) to obtain the title compound (0.22 g, yield: 53.6%).
MS m/z(ESI):357.1[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ:9.15(s,1H),8.42(d,J=2.0Hz,1H),7.91(d,J=8.8Hz,1H),7.71(dd,J=8.8,2.0Hz,1H),7.61(br,2H),7.37-7.31(m,2H),7.10-7.07(m,2H),2.84(s,3H)。
Example 2: preparation of 2-amino-6- (4-fluorophenyl) -5 (4-methylquinazolin-6-yl) pyrimidine-4-carboxamide (2)
Figure BDA0002325201380000151
To a solution of 2-amino-6- (4-fluorophenyl) -5- (4-methylquinazolin-6-yl) pyrimidine-4-carbonitrile (1) (100mg, 0.28mmol) in dimethyl sulfoxide (2 mL), potassium carbonate (80mg, 0.56mmol) and hydrogen peroxide (0.16g, 1.4 mmol) were added in this order, and the reaction was carried out at 25 ℃ for 2 hours. The reaction solution was poured into water, extracted with ethyl acetate, the organic phases were combined, washed three times with a saturated aqueous sodium sulfite solution, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated, and the residue was purified by preparative high performance liquid chromatography to give the title compound (30 mg, yield: 28.5%).
MS m/z(ESI):375.1[M+H] +
1 H NMR(400MHz,CD 3 OD)δ:9.02(s,1H),8.07(d,J=1.6Hz,1H),7.81(d,J=8.8Hz,1H),7.66(dd,J=8.8,2.0Hz,1H),7.33-7.30(m,2H),6.95-6.90(m,2H),2.84(s,3H)。
Example 3: preparation of 2-amino-6- (4-fluorophenyl) -N-methyl-5- (4-methylquinazolin-6-yl) pyrimidine-4-carboxamide (3)
Figure BDA0002325201380000152
The first step is as follows: preparation of methyl 2-amino-6- (4-fluorophenyl) -5- (4-methylquinazolin-6-yl) pyrimidine-4-carboxylate (3-2)
To a 50mL autoclave, 4-chloro-6- (4-fluorophenyl) -5- (4-methylquinazolin-6-yl) pyrimidin-2-amine (3-1) (0.65g, 1.8mmol), triethylamine (0.364g, 3.6mmol), methanol (5 mL) and DMF (0.5 mL) were added in this order, nitrogen was purged for three minutes, and then Pd (dppf) Cl was added thereto 2 (0.17g, 0.18mmol). The reaction mixture was replaced with a nitrogen atmosphere, evacuated, charged with carbon monoxide gas (pressure 8 atm), reacted at 90 ℃ (autoclave pressure 9 atm) for 24 hours, and then cooled to room temperature. The reaction solution was concentrated, and water was added to the obtained residue, followed by stirring for 5 minutes. The reaction mixture was allowed to stand, and the resulting solid was suction-filtered and dried to obtain the title compound (596 mg, yield: 85.1%) of this step.
MS m/z(ESI):390.1[M+H] +
The second step: preparation of 2-amino-6- (4-fluorophenyl) -5- (4-methylquinazolin-6-yl) pyrimidine-4-carboxylic acid (3-3)
To a 50mL round bottom flask, methyl 2-amino-6- (4-fluorophenyl) -5- (4-methylquinazolin-6-yl) pyrimidine-4-carboxylate (3-2) (0.5g, 1.3mmol) dissolved in methanol (5 mL) and water (1 mL) was added, and sodium hydroxide (0.52g, 13mmol) was added and reacted at 25 ℃ for 12 hours. The reaction solution was concentrated, then diluted with water and adjusted to a pH between 3 and 4 with 1N dilute hydrochloric acid. The reaction solution was left to stand, and the resulting solid was filtered with suction and washed with a small amount of water, followed by drying to obtain the title compound of this step (0.35 g, yield: 71.7%).
MS m/z(ESI):376.1[M+H] +
The third step: preparation of 2-amino-6- (4-fluorophenyl) -N-methyl-5- (4-methylquinazolin-6-yl) pyrimidine-4-carboxamide (3)
To a 10mL round bottom flask, 2-amino-6- (4-fluorophenyl) -5- (4-methylquinazolin-6-yl) pyrimidine-4-carboxylic acid (3-3) (50mg, 0.13mmol), carbonyldiimidazole (42mg, 0.26mmol), and DMF (1 mL) were added in this order, reacted at 25 ℃ for 4 hours, then methylamine hydrochloride (44mg, 0.65mmol) and triethylamine (66mg, 0.65mmol) were added, reacted at 25 ℃ for 12 hours, and then the reaction was quenched by adding methanol. The resulting mixture was purified by preparative high performance liquid chromatography to give the title compound (12 mg, yield: 23.5%).
MS m/z(ESI):376.1[M+H] +
1 H NMR(400MHz,CD 3 OD)δ:9.02(s,1H),8.00(d,J=1.6Hz,1H),7.82(d,J=8.8Hz,1H),7.66(dd,J=8.8,1.6Hz,1H),7.32-7.29(m,2H),6.92(t,J=8.8Hz,2H),2.82(s,3H),2.68(s,3H)。
Example 4: preparation of 2-amino-N- (2-amino-2-oxoethyl) -6- (4-fluorophenyl) -5- (4-methylquinazolin-6-yl) pyrimidine-4-carboxamide (4)
Figure BDA0002325201380000161
The title compound (8 mg, yield: 18.9%) was synthesized in a similar manner to the procedure described in the third step of example 3, using glycylamine hydrochloride instead of methylamine hydrochloride in the third step of example 3.
MS m/z(ESI):432.2[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ:9.06(s,1H),8.70(t,J=5.6Hz,1H),8.06(d,J=1.2Hz,1H),7.73(d,J=8.8Hz,1H),7.56(dd,J=8.8,1.6Hz,1H),7.2-7.01(m,5H),4.10-4.04(m,1H),3.63(d,J=5.2Hz,2H),3.30(s,1H),3.17(d,J=5.2Hz,1H),2.78(s,3H)。
Biological assay
Experimental example 1 determination of competitive inhibition constant (Ki) for adenosine A1, A2a receptors
Reagents for experiments:
CGS-15943:Sigma,C199
[ 3 H]-DPCPX:PerkinElmer,NET974250UC
[ 3 H]-CGS-21680:PerkinElmer,NET1021250UC
a1 receptor cell membrane stock (human): perkinelmer, ES-010-M400UA
A2A receptor cell membrane stock (human): perkinElmer, RBHA2AM400UA
Microscint20cocktail scintillation fluid: perkinElmer,6013329
PEI(Poly ethyleneimine):Sigma,P3143
Instruments and consumables:
MicroBeta 2 Reader,PerkinElmer
unifilter-96GF/C filter plate, perkin Elmer,6005174
96-well plate, agilent,5042-1385
A1 assay buffer: 25mM HEPES,5mM MgCl 2 ,1mM CaCl 2 100mM NaCl, pH7.4. For diluting A1 receptor cell membrane (of human origin) with [3H ]]-DPCPX
A1 wash buffer: 25mM HEPES,5mM MgCl 2 ,1mM CaCl 2 ,100mM NaCl,pH7.4
A2a assay buffer: 50mM Tris-HCl,10mM MgCl 2 1mM EDTA, pH7.4. For diluting A2a receptor cell membrane (of human origin) with [3H ]]-CGS-21680
A2a wash buffer: 50mM Tris-HCl,154mM NaCl, pH7.4
The experimental method comprises the following steps:
a1 receptor cell membranes are diluted to 0.025 mu g/mu l by adopting an A1 experiment buffer solution, and A2a receptor cell membranes are diluted to 0.05 mu g/mu l by adopting an A2a experiment buffer solution, so that an A1 receptor cell membrane diluent and an A2a receptor cell membrane diluent are respectively obtained.
Diluting test compound and CGS15943 with DMSO gradient, adding 1 μ l test compound, high control (0.5% DMSO), and low control (1000nM CGS15943) to 96-well plate, and adding 100 μ lA1 receptor cell membrane diluent (containing 2.5 μ g cell membrane) to each well to obtain A1 assay plate; mu.l of test compound, high control (0.5% DMSO), and low control (1000nM CGS15943) were added to a 96-well plate, and 100. Mu. lA2a receptor cell membrane dilution (containing 5.0. Mu.g cell membrane) was added to each well to obtain A2a assay plate;
mu.l of a radioisotope labeled ligand [2 ] was added to an A1 detection plate 3 H]DPCPX (diluted with A1 assay buffer, working concentration 1.0 nM); add 100. Mu.l to A2a assay plate A radioisotope labeled ligand [2 ] 3 H]CGS-21680 (diluted with A2a assay buffer, working concentration 6.0 nM); the A1 and A2a detection plates were blocked with tape and incubated for 1h and 2h, respectively, at room temperature.
The Unifilter-96GF/C filter plate was prepared, 50. Mu.l of 0.3% PEI was added to each well of the Unifilter-96GF/C filter plate, and the mixture was incubated at room temperature for not less than 0.5h.
After the incubation is finished, the reaction liquid in the A1 detection plate and the A2a detection plate is transferred to two Unifilter-96GF/C filter plates respectively, and the filter plates are washed by using corresponding precooled washing buffer solutions respectively and then dried. After sealing the bottom of the filter plate, 50. Mu.l of Microscint20cocktail scintillation fluid was added, the top of the filter plate was sealed, and the plate was read using a counter Microbeta2 Reader.
And (3) data analysis:
the inhibition rate was calculated using the following formula: inhibition rate% =100- (experimental well signal value-low value control signal average value)/(high value control signal average value-low value control signal average value) = 100; IC fitting Using EXCEL XLFit 50 (ii) a The competitive inhibition constant (Ki) is calculated as: ki = IC 50 /(1 + isotopically labeled ligand concentration/Kd); wherein K is d Is the dissociation constant of the isotopically labeled ligand.
The results of the competitive inhibition constant (Ki) assay for adenosine A2a, A1 receptors for the compounds of the invention are detailed in Table 1:
table 1: competitive inhibition constants (Ki) of the compounds of the invention for adenosine A2a, A1 receptors
Figure BDA0002325201380000171
The data in table 1 show that the compounds of the invention (e.g. compounds 1, 2, 3 and 4) have good affinity for the adenosine A2a receptor, weaker affinity for the adenosine A1 receptor and good selectivity for the adenosine A2a receptor.
Experimental example 2: rat Pharmacokinetic (PK) study
Male SD rats were administered the compounds of the invention by gavage (PO) and pharmacokinetic profiles were examined. The dose administered is 5mg/kg, the vehicle 0.5%. Blood was collected at time points of pre-dose (0 h) and 0.25, 0.5, 1, 2, 4,6, 8, 24, 32 and 48h post-dose using edta.k 2 Anticoagulated and centrifuged to obtain a plasma sample, and the plasma sample is stored at-80 ℃. Plasma samples were processed for precipitated protein and analyzed by LC-MS/MS. Pharmacokinetic parameters were calculated using WinNonlin 6.3 software using a non-compartmental model, and the results are shown in table 2.
Table 2: pharmacokinetic parameters in rats of PO administration of Compounds of the invention
Figure BDA0002325201380000172
As shown in Table 2, the C in rat blood of Compound 1 of the present invention administered by PO at a dose of 5mg/kg max Is 1350ng/mL, AUC last 43822h ng/mL, indicating that the compound of the present invention (e.g. compound 1) has excellent drug exposure and oral absorption in rat blood system by PO administration.

Claims (25)

1. A compound or a pharmaceutically acceptable salt thereof, wherein the compound has the structure of formula (I):
Figure FDA0004019287320000011
x is N or CH;
R 1 selected from cyano and R a R b N-C(O)-;
R 2 Selected from hydrogen, halogen, C 1-6 Alkyl and C 1-6 A haloalkyl group;
R 3 selected from hydrogen, halogen, C 1-6 Alkyl and C 1-6 A haloalkyl group;
R 4 selected from hydrogen, halogen, C 1-6 Alkyl and C 1-6 A haloalkyl group;
R a and R b Each independently selected from hydrogen and C 1-6 Alkyl and R c R d N-C(O)-C 1-6 Alkyl-;
R c and R d Each independently selected from hydrogen and C 1-6 An alkyl group;
n is selected from 0, 1 or 2;
p is selected from 0, 1 or 2;
q is selected from 0, 1 or 2.
2. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein:
R a and R b Each independently selected from hydrogen and C 1-6 An alkyl group.
3. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein:
R a and R b Each independently selected from hydrogen and C 1-3 Alkyl and R c R d N-C(O)-C 1-3 An alkyl radical.
4. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein: r a And R b Each independently selected from hydrogen, methyl, ethyl, propyl, R c R d N-C(O)-CH 2 -、R c R d N-C(O)-(CH 2 ) 2 -and R c R d N-C(O)-(CH 2 ) 3 -。
5. The compound of claim 1 or a pharmaceutically acceptable salt thereofA salt of acceptance, wherein: r a And R b Each independently selected from hydrogen, methyl and R c R d N-C(O)-CH 2 -。
6. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein: r a Is H, and R b Is hydrogen, methyl or R c R d N-C(O)-CH 2 -。
7. The compound of any one of claims 1 to 6, or a pharmaceutically acceptable salt thereof, wherein: r c And R d Each independently selected from hydrogen and C 1-3 An alkyl group.
8. The compound of claim 7, or a pharmaceutically acceptable salt thereof, wherein: r c And R d Each independently selected from hydrogen, methyl, ethyl and propyl.
9. The compound of claim 8, or a pharmaceutically acceptable salt thereof, wherein: r c And R d Each independently hydrogen.
10. The compound of any one of claims 1-6 and 8-9, or a pharmaceutically acceptable salt thereof, wherein: r 2 Selected from hydrogen, halogen, C 1-3 Alkyl and C 1-3 A haloalkyl group.
11. The compound of claim 10, or a pharmaceutically acceptable salt thereof, wherein:
R 2 selected from the group consisting of hydrogen, fluoro, chloro, bromo, iodo, methyl, ethyl and propyl.
12. The compound of claim 11, or a pharmaceutically acceptable salt thereof, wherein: r 2 Is fluorine.
13. The compound of any one of claims 1-6, 8-9, or 11-12, or a pharmaceutically acceptable salt thereof, wherein: r 3 Selected from hydrogen, halogen, C 1-3 Alkyl and C 1-3 A haloalkyl group.
14. The compound of claim 13, or a pharmaceutically acceptable salt thereof, wherein: r 3 Selected from the group consisting of hydrogen, fluoro, chloro, bromo, iodo, cyano, methyl, ethyl and propyl.
15. The compound of claim 13, or a pharmaceutically acceptable salt thereof, wherein: r 3 Selected from hydrogen.
16. The compound of any one of claims 1-6, 8-9, 11-12, or 14-15, or a pharmaceutically acceptable salt thereof, wherein: r is 4 Selected from hydrogen, halogen, C 1-3 Alkyl and C 1-3 A haloalkyl group.
17. The compound of claim 16, or a pharmaceutically acceptable salt thereof, wherein: r 4 Selected from the group consisting of hydrogen, fluoro, chloro, bromo, iodo, cyano, methyl, ethyl, and propyl.
18. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein: the compound has the structure of formula (Ia):
Figure FDA0004019287320000021
wherein R is a 、R b 、R 2 、R 3 、R 4 X, n, p, q are as defined in any one of claims 1 to 6, 8 to 9 or 11 to 12.
19. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein: the compound has the structure of formula (Ia-1), formula (Ia-2) or formula (Ia-3):
Figure FDA0004019287320000022
wherein R is 2 、R 3 、R 4 X, n, p, q are as defined in any one of claims 1 to 6, 8 to 9 or 11 to 12.
20. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein: the compound has the structure of formula (I-5):
Figure FDA0004019287320000031
wherein R is 2 、R 3 、R 4 X, n, p, q are as defined in any one of claims 1, 11-12, 14-15 and 17.
21. The compound of any one of claims 1 to 6, or a pharmaceutically acceptable salt thereof, wherein: x is N;
R 1 selected from cyano and R a R b N-C(O)-;
R 2 Selected from hydrogen, halogen, cyano, C 1-3 Alkyl and C 1-3 A haloalkyl group;
R 3 selected from hydrogen, halogen, C 1-3 Alkyl and C 1-3 A haloalkyl group;
R 4 selected from hydrogen, halogen, C 1-3 Alkyl and C 1-3 A haloalkyl group;
R a and R b Each independently selected from hydrogen, C 1-3 Alkyl and R c R d N-C(O)-C 1-6 Alkyl-;
R c and R d Each independently selected from hydrogen and C 1-3 An alkyl group;
n is selected from 0 or 1;
p is selected from 0 or 1;
q is selected from 0 or 1.
22. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein the compound is selected from the group consisting of:
Figure FDA0004019287320000032
23. a pharmaceutical composition comprising a prophylactically or therapeutically effective amount of a compound according to any one of claims 1-22, or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable carriers.
24. Use of a compound according to any one of claims 1 to 22 or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 23, in the manufacture of a medicament for the prevention or treatment of an adenosine A2a receptor-related disease.
25. The use of claim 24, wherein the adenosine A2a receptor associated disease is a tumor.
CN201911313568.8A 2018-12-28 2019-12-19 Aminopyrimidine compounds, their preparation and use Active CN111377873B (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN2018116169412 2018-12-28
CN201811616941 2018-12-28

Publications (2)

Publication Number Publication Date
CN111377873A CN111377873A (en) 2020-07-07
CN111377873B true CN111377873B (en) 2023-03-28

Family

ID=71213424

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201911313568.8A Active CN111377873B (en) 2018-12-28 2019-12-19 Aminopyrimidine compounds, their preparation and use

Country Status (1)

Country Link
CN (1) CN111377873B (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022042331A1 (en) * 2020-08-25 2022-03-03 四川科伦博泰生物医药股份有限公司 Heterocyclic compound, and preparation method therefor and use thereof
KR20230117416A (en) * 2021-01-18 2023-08-08 하이노바 파마슈티컬스 인코포레이티드 Methods for synthesizing aminopyrimidine-based FAK inhibitor compounds
CN116425683A (en) * 2023-04-04 2023-07-14 上药康丽(常州)药业有限公司 Minoxidil synthesis and refining method

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1438890A (en) * 2000-02-25 2003-08-27 弗·哈夫曼-拉罗切有限公司 Adenosine receptor modulators
CN1507436A (en) * 2001-03-02 2004-06-23 Aryl and heteroaryl urea CHK1 inhibitors for use as radiosensitizers and chamosensitizers
CN1575290A (en) * 2001-10-22 2005-02-02 卫材株式会社 Pyrimidine derivatives and pharmaceutical compositions containing that compounds
CN1809354A (en) * 2003-04-24 2006-07-26 麦克公司 Inhibitors of Akt activity
CN101268064A (en) * 2005-07-29 2008-09-17 奥米罗实验室有限公司 Pyrazine derivatives useful as adenosine receptor antagonists
CN102822150A (en) * 2010-02-05 2012-12-12 赫普泰雅治疗有限公司 1,2,4-Triazin-4-amine derivatives
WO2018130184A1 (en) * 2017-01-13 2018-07-19 江苏恒瑞医药股份有限公司 1,2,4-triazine-3-amine derivative, preparation method therefor, and use thereof in medicine
WO2019062803A1 (en) * 2017-09-28 2019-04-04 基石药业 Fused ring derivative as a2a receptor inhibitor
WO2019158070A1 (en) * 2018-02-15 2019-08-22 杭州阿诺生物医药科技有限公司 A2a and/or a2b receptor antagonist

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI301834B (en) * 2001-10-22 2008-10-11 Eisai R&D Man Co Ltd Pyrimidone compound and pharmaceutical composition including the same

Patent Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1438890A (en) * 2000-02-25 2003-08-27 弗·哈夫曼-拉罗切有限公司 Adenosine receptor modulators
CN1507436A (en) * 2001-03-02 2004-06-23 Aryl and heteroaryl urea CHK1 inhibitors for use as radiosensitizers and chamosensitizers
CN1575290A (en) * 2001-10-22 2005-02-02 卫材株式会社 Pyrimidine derivatives and pharmaceutical compositions containing that compounds
CN1809354A (en) * 2003-04-24 2006-07-26 麦克公司 Inhibitors of Akt activity
CN101268064A (en) * 2005-07-29 2008-09-17 奥米罗实验室有限公司 Pyrazine derivatives useful as adenosine receptor antagonists
CN102822150A (en) * 2010-02-05 2012-12-12 赫普泰雅治疗有限公司 1,2,4-Triazin-4-amine derivatives
WO2018130184A1 (en) * 2017-01-13 2018-07-19 江苏恒瑞医药股份有限公司 1,2,4-triazine-3-amine derivative, preparation method therefor, and use thereof in medicine
CN108884061A (en) * 2017-01-13 2018-11-23 江苏恒瑞医药股份有限公司 1,2,4- triazine -3- amine derivant, preparation method and its application in medicine
WO2019062803A1 (en) * 2017-09-28 2019-04-04 基石药业 Fused ring derivative as a2a receptor inhibitor
CN111542520A (en) * 2017-09-28 2020-08-14 基石药业(苏州)有限公司 Pycyclic derivatives as A2A receptor inhibitors
WO2019158070A1 (en) * 2018-02-15 2019-08-22 杭州阿诺生物医药科技有限公司 A2a and/or a2b receptor antagonist

Also Published As

Publication number Publication date
CN111377873A (en) 2020-07-07

Similar Documents

Publication Publication Date Title
JP7672451B2 (en) RHO-associated protein kinase inhibitors, pharmaceutical compositions containing same, and methods for preparing and using same - Patents.com
JP6948659B1 (en) Pyridadinyl thiaazole carboxamide compound
JP5820921B2 (en) 1,2-disubstituted heterocyclic compounds
JP2020525523A (en) RHO-related protein kinase inhibitor, pharmaceutical composition containing RHO-related protein kinase inhibitor, preparation method and use of the pharmaceutical composition
JP2020525525A (en) RHO-related protein kinase inhibitor, pharmaceutical composition containing RHO-related protein kinase inhibitor, preparation method and use of the pharmaceutical composition
EP3741752A1 (en) Tam family kinase /and csf1r kinase inhibitor and use thereof
KR20150092279A (en) Novel bi-ring phenyl-pyridines/pyrazines for the treatment of cancer
EP4289843A1 (en) Pyridopyrimidinone derivative, preparation method therefor, and use thereof
CN111377873B (en) Aminopyrimidine compounds, their preparation and use
WO2022028506A1 (en) Sos1 inhibitor, pharmaceutical composition containing same, and use therefor
KR20230004612A (en) Substituted pyridines for the treatment of inflammatory diseases
CN112969694A (en) Rho-related protein kinase inhibitor, pharmaceutical composition containing same and application thereof
JP2019524678A (en) Chiral heterocyclic compound having hedgehog pathway antagonist activity, method for producing the same, and application thereof
US20240279224A1 (en) Quinolines and azaquinolines as inhibitors of cd38
CA3130245A1 (en) Heterocyclic compound, pharmaceutical composition comprising same, preparation method therefor, and use thereof
JP2022515309A (en) Substituted aryl compounds, their production methods and uses
CN113166109B (en) Aminopyridine compound and preparation method and application thereof
JP2022544700A (en) Molecules that Bind TDP-43 for the Treatment of Amyotrophic Lateral Sclerosis and Related Disorders
JP7710756B2 (en) CTLA-4 small molecule degrader and uses thereof
CN111377906B (en) Substituted pyrazine compounds, their preparation and use
TWI894448B (en) Ctla-4 small molecule inhibitor and its application
WO2019141096A1 (en) Substituted urea compound and preparation method and use thereof
CN111808080B (en) Substituted pyridine or pyrimidine compound, preparation method and medical application thereof
KR20250025695A (en) CSF-1R inhibitors and uses thereof
CN118414335A (en) Compounds as Src inhibitors

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