[go: up one dir, main page]
More Web Proxy on the site http://driver.im/

CN111377906B - Substituted pyrazine compounds, their preparation and use - Google Patents

Substituted pyrazine compounds, their preparation and use Download PDF

Info

Publication number
CN111377906B
CN111377906B CN201811616735.1A CN201811616735A CN111377906B CN 111377906 B CN111377906 B CN 111377906B CN 201811616735 A CN201811616735 A CN 201811616735A CN 111377906 B CN111377906 B CN 111377906B
Authority
CN
China
Prior art keywords
compound
pharmaceutically acceptable
acceptable salt
hydrogen
independently
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
CN201811616735.1A
Other languages
Chinese (zh)
Other versions
CN111377906A (en
Inventor
刘金明
何婷
蔡家强
康熙伟
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
Priority to CN201811616735.1A priority Critical patent/CN111377906B/en
Publication of CN111377906A publication Critical patent/CN111377906A/en
Application granted granted Critical
Publication of CN111377906B publication Critical patent/CN111377906B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings

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

The present invention discloses a class of substituted pyrazine compounds as adenosine receptor antagonists, methods of their preparation, intermediates, pharmaceutical compositions containing them and their therapeutic use.

Description

Substituted pyrazine compounds, their preparation and use
Technical Field
The present invention relates to a class of substituted pyrazine compounds as adenosine receptor antagonists, methods of preparation thereof, intermediates, pharmaceutical compositions containing them and therapeutic uses thereof.
Background
Adenosine is a signaling molecule that inhibits inflammation and immune responses in vivo, and extracellular adenosine is mainly of 2 origins, namely, intracellular adenosine transport and extracellular adenosine hydrolysis. Adenosine can be produced in many types of tumor tissue and maintained at high levels in the tumor microenvironment. The main pathway for adenosine production in the tumor microenvironment is that CD73 enzyme, which is highly expressed on the surface of tumor cells, can catalyze 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) and the family of receptors comprises four major receptors, the a1, A2a, A2b and A3 receptors. Among them, 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.
The adenosine A2a receptor is 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 adenosine A2a receptor on the surface of tumor tissue infiltration immunocyte, so that cAMP amount in the immunocyte is increased, the capability of the immunocyte for attacking the tumor is inhibited, the organism is enabled to generate immune tolerance, and the tumor cell can escape immune monitoring of the organism, which is mainly shown in 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 evade surveillance and attack by the immune system, increasing their survival rate. The A2a receptor gene knockout mouse can strengthen the anti-tumor immunity 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 high level, and the activation of the adenosine A2a receptor can promote the organism to generate immune tolerance, promote the formation of 'immune escape' or 'immune suppression' of tumor cells, and create favorable conditions for the generation and development of tumors. The adenosine A2a receptor antagonist directly targets the adenosine A2a receptor on the surface of an immune cell, inhibits the activation of the receptor, further inhibits the generation of cAMP in the immune cell, eliminates the T cell immune function inhibition mediated by the activation of the adenosine A2a receptor, and achieves the effect of treating tumors. Therefore, the adenosine A2a receptor antagonist has good application prospect in the pharmaceutical industry as a tumor treatment drug. 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.
With respect to compounds having an antagonistic effect on the adenosine A2a receptor, the company Corvus CPI-444 is currently in phase I clinical studies, with the indication being a tumor. Heretofore, CPI-444 has been used in clinical trials for the treatment of central nervous system disorders. On the other hand, WO0162233 and WO2002014282 disclose that aminopyridine compounds have an antagonistic action against adenosine A2a receptor and disclose that they are useful as therapeutic agents for parkinson's disease or senile dementia. WO0162233, WO2003035639, WO2004016605 and WO2005079801 disclose that aminopyrimidine compounds have an antagonistic action on adenosine A2a receptor and are useful as therapeutic agents for diseases such as parkinson's disease or neuralgia. WO2011095625 discloses aminotriazine compounds having antagonistic action against adenosine A2a receptors and as therapeutic agents for dyskinesias, stroke, or parkinson's disease.
Therefore, adenosine A2a receptor antagonist has good application prospect in the pharmaceutical industry as a medicament, but there is little research on the anti-tumor application of the adenosine A2a receptor antagonist. There is a need in the art to develop adenosine A2a receptor antagonists for the treatment of tumors.
Disclosure of Invention
The present invention provides a class of substituted pyrazine compounds which possess adenosine receptor antagonistic activity, particularly high selectivity for the A2a receptor relative to the a1 receptor. The compounds of the present invention, which are adenosine receptor antagonists, particularly A2a receptor selective antagonists, have good antitumor activity. The compounds of the invention also have a variety of advantageous properties, such as good physicochemical properties (e.g., solubility, physical and/or chemical stability); good pharmacokinetic properties (e.g. good bioavailability, suitable blood concentration, half-life and duration of action); good safety (lower toxicity and/or fewer side effects, wider therapeutic window).
One aspect of the present invention provides a compound, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug thereof, wherein said compound has the structure of formula (I):
Figure BDA0001925977890000021
wherein:
x is N or CR 7
R 1 And R 2 Each independently selected from hydrogen and C 1-6 Alkyl, -C (O) -R a 、-C(O)OR a 、-C(O)-NR a R b 、-S(O) m OR a and-S (O) m -NR a R b
R 3 Selected from hydrogen, halogen, CN, OH, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 3-6 Cycloalkyl, COOH, -C (O) O-C 1-6 Alkyl, -C (O) -NR c R d 、S(O) m OH、-S(O) m O-C 1-6 Alkyl and-S (O) m -NR c R d
R 4 、R 5 And R 6 Each occurrence is independently selected from hydrogen, halogen, CN, OH, C 1-6 Alkyl radical, C 1-6 Haloalkyl and C 1-6 An alkoxy group;
R 7 selected from hydrogen, halogen, CN, OH, C 1-6 Alkyl radical, C 1-6 Haloalkyl and C 1 -6 alkoxy;
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 C substituted by 5-or 6-membered heteroaryl 1-6 An alkyl group; or R a And R b Together with the nitrogen atom to which they are commonly attached form a 3-to 6-membered azaheterocyclyl;
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 C substituted by 5-or 6-membered heteroaryl 1-6 An alkyl group; or R c And R d Together with the nitrogen atom to which they are commonly attached form a 3-to 6-membered azaheterocyclyl;
m is 1 or 2;
n is 0, 1 or 2;
p is 0, 1 or 2; and is
q is 0, 1 or 2.
Another aspect of the invention provides a pharmaceutical composition comprising 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 invention provides the use of 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, in the manufacture of a medicament for the treatment of an inhibition of T cell immune function in a subject.
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 treatment of an inhibition of T cell immune function in a subject.
Another aspect of the present invention provides a method of treating a subject for T cell immune function suppression, said method 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.
Another aspect of the invention provides the use of 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, in the manufacture of a medicament for the treatment of a disease associated with the adenosine receptor.
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 treatment of a disease associated with the adenosine receptor.
Another aspect of the present invention provides a method of treating a disease associated with an adenosine receptor, 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.
Another aspect of the invention provides a process for preparing a compound of the invention.
Definition of
Unless defined otherwise below, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art. The techniques used herein refer to those commonly understood in the art, including those variations or equivalents thereof that would be appreciated by those skilled in the art. While the following terms are believed to be understood by those skilled in the art, the following definitions are set forth to better explain the present invention.
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.
The term "alkyl" as used herein is defined as a straight or branched chain saturated aliphatic hydrocarbon. In some embodiments, the alkyl group has 1 to 12, e.g., 1 to 6, carbon atoms. For example, as used herein, the term "C 1-6 Alkyl "refers to a straight chain of 1 to 6 carbon atoms orA branched group (e.g. methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl or n-hexyl) which is optionally substituted with 1 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.). The term "C 1-4 Alkyl "refers to a straight or branched aliphatic hydrocarbon chain of 1 to 4 carbon atoms (i.e., methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, or tert-butyl).
As used herein, the term "alkoxy" refers to a group having the structure "alkyl-O-" (wherein alkyl is as defined above) which is optionally substituted with 1 or more (such as 1 to 3) suitable substituents, for example, halogen (in which case the group is referred to as "haloalkoxy"). As used herein, the term "C 1-6 Alkoxy "means having" C 1-6 alkyl-O- "structure (wherein C 1-6 Alkyl is as defined above), e.g. C 1-4 Alkoxy radical, C 1-2 Alkoxy radical, C 1 Alkoxy radical, C 2 Alkoxy radical, C 3 Alkoxy radical, C 4 Alkoxy radical, C 5 Alkoxy or C 6 An alkoxy group. Specific examples include, but are not limited to, methoxy, ethoxy, propoxy, isopropoxy, n-butoxy, 2-butoxy, isopropoxy, sec-butoxy, tert-butoxy, n-pentoxy, isopentoxy, n-hexoxy, and the like.
As used herein, the term "cycloalkyl" refers to a saturated or 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 ring, including spiro, fused or bridged systems (such as bicyclo [ 1.1.1)]Pentyl, bicyclo [2.2.1]Heptyl, bicyclo [3.2.1]Octyl or bicyclo [5.2.0]Nonyl, decalinyl, etc.), optionally substituted with 1 or more (such as 1 to 3) suitable substituents. The cycloalkyl group has 3 to 15 carbon atoms. For example, the term "C 3-6 Cycloalkyl "refers to a saturated or unsaturated, non-aromatic, monocyclic or polycyclic (such as bicyclic) hydrocarbon ring of 3 to 6 ring-forming carbon atoms (e.g., cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl) optionally substituted with 1 or more (such as 1 to 3) suitable substituents, for example, methyl-substituted cyclopropyl.
As used herein, the term "aryl" refers to an all-carbon monocyclic or fused ring polycyclic aromatic group having a conjugated pi-electron system. For example, as used herein, the term "C 6-14 Aryl "means an aromatic group containing 6 to 14 carbon atoms, such as phenyl or naphthyl. 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" refers to a monovalent monocyclic, bicyclic or tricyclic aromatic ring system having 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 ring atoms, in particular 1 or 2 or 3 or 4 or 5 or 6 or 9 or 10 carbon atoms, and which contains at least one heteroatom (which may be the same or different, for example oxygen, nitrogen or sulfur) and which, in addition, in some cases, may be benzo-fused. In particular, heteroaryl is selected from thienyl, furyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl and the like, and benzo derivatives thereof; or pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, and the like, and benzo derivatives thereof.
As used herein, the term "halo" or "halogen" group is defined to include F, Cl, Br, or I.
As used herein, the term "heterocyclyl" refers to a saturated or partially unsaturated monocyclic or polycyclic group having 2, 3, 4,5, 6, 7, 8, 9 carbon atoms in the ring and one or more (e.g., 1, 2, 3, or 4) selected from N, NR (R represents a hydrogen atom or a substituent such as, but not limited to, alkyl or cycloalkyl), O, C (═ O), S, S (═ O), S (═ O) 2 A group of (1). In particular, 3-10 membered heterocyclic groups are those having 3-10 carbon atoms in the ring and heteroRadicals of atoms, for example, having 4 to 10, 4 to 7, 4 to 6, 5 to 7, and 5 to 6 carbon and heteroatoms (referred to as 4 to 10, 4 to 7, 4 to 6, 5 to 7, and 5 to 6 membered heterocyclic groups, respectively), such as, but not limited to, oxiranyl, aziridinyl, azetidinyl, oxetanyl, tetrahydrofuryl, pyrrolidinyl, pyrrolidinonyl, imidazolidinyl, pyrazolidinyl, tetrahydropyranyl, piperidinyl, morpholinyl, dithianyl (dithianyl), thiomorpholinyl, piperazinyl, trithianyl (trithianyl), and the like; and bicyclic derivatives thereof, such as, but not limited to, pyrrolidinyl-cyclopropyl, cyclopent-aziridinyl, pyrrolidinyl-cyclobutyl, pyrrolidinyl-pyrrolidinyl, pyrrolidinyl-piperidinyl, pyrrolidinyl-piperazinyl, pyrrolidinyl-morpholinyl; or a spiro derivative; or benzo derivatives or heteroaryl and derivatives, such as, but not limited to
Figure BDA0001925977890000041
And the like.
As used herein, the term "azaheterocyclyl" refers to a heterocyclyl group as described above, wherein at least one ring member is a nitrogen atom. As used herein, the term "3 to 6 membered azaheterocyclyl" refers to a saturated or partially unsaturated monocyclic heterocyclic group having 3, 4,5 or 6 ring members, wherein at least one ring member is a nitrogen atom, which may optionally further comprise one or more (e.g. 1, 2 or 3) groups selected from N, NR (R represents a hydrogen atom or a substituent such as, but not limited to, alkyl or cycloalkyl), O, C ═ O, S, S ═ O and S (═ O) 2 The ring member of (1). In some embodiments, the azaheterocyclyl is attached to the remainder of the molecule through a C atom; in some embodiments, the azaheterocyclyl, e.g., as described herein with respect to R a And R b And R c And R d The "nitrogen heterocyclyl" as defined is attached to the remainder of the molecule through a nitrogen atom. In particular, examples of 3-membered azaheterocyclyl groups include, but are not limited to, aziridinyl; examples of 4-membered azaheterocyclyl groups include, but are not limited to, azetidinyl and 1, 3-oxaazetidinyl; examples of the 5-membered azaheterocyclyl group includeBut are not limited to, for example, pyrrolyl, tetrahydropyrrolyl, pyrrolinyl, pyrrolidinonyl, imidazolyl, imidazolidinyl, imidazolinyl, pyrazolyl, pyrazolinyl; examples of 6 membered azaheterocyclyl include, but are not limited to, piperidinyl, morpholinyl, thiomorpholinyl, piperazinyl.
The term "substituted" means that one or more (e.g., 1, 2, 3, or 4) hydrogens on the designated atom is replaced with a selected group, provided that the designated atom's normal valency at the present instance is not exceeded, and that the substitution results in a stable compound. The number of substituent groups selected is permissible only if such combination forms a stable compound.
If a substituent is described as "optionally substituted," the substituent may be (1) unsubstituted or (2) substituted. If a carbon of a substituent is described as optionally substituted with one or more of the list of substituents, then one or more hydrogens on the carbon 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 may each be replaced with an independently selected optional substituent.
If a substituent is described as "independently selected", each substituent may be the same as 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.
Unless indicated, as used herein, the point of attachment of a substituent may be from any suitable position of the substituent.
When a bond of a substituent is shown to pass through a bond in a ring without specifying a position, then such substituent may be bonded to any appropriate 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 a group having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number prevailing in natureAtomic substitution of mass number. Examples of isotopes suitable for inclusion in compounds of the invention include, but are not limited to, isotopes of hydrogen (e.g. deuterium (D, 2 H) tritium (T, 3 H) ); isotopes of carbon (e.g. of 11 C、 13 C and 14 C) (ii) a Isotopes of chlorine (e.g. of chlorine) 36 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); 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 are useful in drug and/or substrate tissue distribution studies (e.g., assays). 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 result in 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 a mixture of two or more structurally distinct forms in rapid equilibrium (commonly referred to as tautomers). Representative examples of tautomers include keto-enol tautomers, phenol-keto tautomers, nitroso-oxime tautomers, imine-enamine tautomers, and the like. For example, a dihydropyrimidine radical may exist in solution in equilibrium with the following tautomeric forms:
Figure BDA0001925977890000042
it is understood that the scope of this application encompasses all such isomers or mixtures thereof in any proportion (e.g., 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%).
Solid (-), solid wedge shapes may be used herein
Figure BDA0001925977890000043
Or virtual wedge shape
Figure BDA0001925977890000044
Carbon-carbon bonds of the compounds of the invention are depicted. The solid line is used to depict the representation of the bond to the asymmetric carbon atom, including all possible stereoisomers at that carbon atom (e.g., particular enantiomers, racemic mixtures, etc.). The use of solid or dashed wedges to depict bonds to asymmetric carbon atoms indicates 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 may exist in the form of stereoisomers (which include 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 present 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 the 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, esters, solvates, metabolites, isotopically labeled compounds or prodrugs, 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, including suitable inorganic and organic acids. Examples include aspartate, benzoate, bicarbonate/carbonate, bisulfate/sulfate, fumarate, glucoheptonate, gluconate, glucuronate, hexafluorophosphate, hydrobromide/bromide, hydroiodide, maleate, malonate, methylsulfate, naphthoate, nicotinate, nitrate, orotate, oxalate, palmitate and other similar salts.
Suitable base addition salts are formed from bases which form pharmaceutically acceptable salts and include suitable inorganic and organic bases. Examples include aluminum, arginine, choline, diethylamine, lysine, magnesium, meglumine, potassium and other similar salts.
For a review of suitable Salts, see 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.
As used herein, the term "ester" means an ester derived from a compound of the respective general formula in the present application, including physiologically hydrolysable esters, which can be hydrolysed under physiological conditions to release the compound of the invention in the form of the free acid or alcohol. The compounds of the invention may themselves also be esters.
The compounds of the invention may be present in the form of solvates (e.g. 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 can form N-oxides because nitrogen requires an available lone pair for oxidation to an 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-; a.r.katitzky and a.j.boulton, eds., Academic Press; and G.W.H.Cheeseman and E.S.G.Werstuk, Advances in Heterocyclic Chemistry, vol.22, pp 390-.
Also included within the scope of the present invention are metabolites of the compounds of the present invention, i.e., substances formed in vivo upon administration of the compounds of the present 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 or may not themselves be pharmacologically active, which may be converted to the compounds of the present invention having the desired activity by, for example, hydrolytic cleavage when administered into or onto the body. 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 substituting certain moieties known to those skilled in the art as "pro-moieties" (e.g., "Design of Prodrugs", described in h. bundgaard (Elsevier, 1985)) for appropriate functional groups present in compounds of the invention.
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.
As used herein, the term "about" means within ± 10% of the stated numerical value, such as within ± 5% or within ± 2%.
Detailed Description
Compound and preparation method thereof
One aspect of the present invention provides a compound, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug thereof, wherein said compound has the structure of formula (I):
Figure BDA0001925977890000061
wherein:
x is N or CR 7
R 1 And R 2 Each independently selected from hydrogen, C 1-6 Alkyl, -C (O) -R a 、-C(O)OR a 、-C(O)-NR a R b 、-S(O) m OR a and-S (O) m -NR a R b
R 3 Selected from hydrogen, halogen, CN, OH, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 3-6 Cycloalkyl, COOH, -C (O) O-C 1-6 Alkyl, -C (O) -NR c R d 、S(O) m OH、-S(O) m O-C 1-6 Alkyl and-S (O) m -NR c R d
R 4 、R 5 And R 6 Each occurrence is independently selected from hydrogen, halogen, CN, OH, C 1-6 Alkyl radical, C 1-6 Haloalkyl and C 1-6 An alkoxy group;
R 7 selected from hydrogen, halogen, CN, OH, C 1-6 Alkyl radical, C 1-6 Haloalkyl and C 1-6 An alkoxy group;
R a and R b Each independently selected from hydrogen, C 1-6 Alkyl radical, C 3-6 Cycloalkyl, hydroxy C 1-6 Alkyl radical, C 1-6 alkoxy-C 1-6 Alkyl-and C substituted by 5-or 6-membered heteroaryl 1-6 An alkyl group; or R a And R b Together with the nitrogen atom to which they are commonly attached form a 3-to 6-membered azaheterocyclyl;
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 C substituted by 5-or 6-membered heteroaryl 1-6 An alkyl group; or R c And R d Together with the nitrogen atom to which they are commonly attached form a 3-to 6-membered azaheterocyclyl;
m is selected from 1 or 2;
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 is 1 Selected from hydrogen, C 1-4 Alkyl, -C (O) -R a 、-C(O)OR a 、-C(O)-NR a R b 、-S(O) m OR a and-S (O) m -NR a R b
In a preferred embodiment, R 1 Selected from hydrogen and C 1-4 An alkyl group. Preferably, R 1 Selected from hydrogen, methyl, ethyl and isopropyl. More preferably, R 1 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 is a And R b Each independently selected from hydrogen and C 1-4 Alkyl radical, C 3-6 Cycloalkyl, hydroxy C 1-4 Alkyl radical, C 1-4 alkoxy-C 1-4 Alkyl-and C substituted by 5-or 6-membered heteroaryl 1-4 An alkyl group.
In a preferred embodiment, R a And R b Each independently selected from C 1-4 Alkyl and C substituted by 5-or 6-membered heteroaryl 1-4 An alkyl group;
preferably, R a And R b Each independently selected from C 1-4 Alkyl and C substituted by 6-membered heteroaryl 1-4 An alkyl group;
preferably, R a And R b Each independently selected from C 1-4 Alkyl and C substituted by pyridyl 1-4 An alkyl group;
preferably, R a And R b Each independently selected from methyl, ethyl, isopropyl and methyl, ethyl and isopropyl substituted with pyridyl;
preferably, R a And R b One selected from methyl, ethyl, isopropyl and the other selected from methyl, ethyl and isopropyl substituted with pyridyl;
preferably, R a And R b One of which is methyl and the other is methyl substituted by pyridyl;
more preferably, R a And R b One of which is methyl and the other is
Figure BDA0001925977890000071
In other embodiments, R a And R b Together with the nitrogen atom to which they are commonly attached, form a 4-to 6-membered azaheterocyclyl.
In a preferred embodiment, R a And R b Together with the nitrogen atom to which they are commonly attached form a 5 or 6 membered azaheterocyclyl;
preferably, R a And R b Together with the nitrogen atom to which they are commonly attached form a 6-membered azaheterocyclyl;
more preferably, R a And R b Together with the nitrogen atom to which they are commonly attached
Figure BDA0001925977890000072
More preferably, R a And R b Together with the nitrogen atom to which they are jointly attached
Figure BDA0001925977890000073
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 is 2 Selected from hydrogen, C 1-4 Alkyl, -C (O) -R a 、-C(O)OR a 、-C(O)-NR a R b 、-S(O) m OR a and-S (O) m -NR a R b
In a preferred embodiment, R 2 Selected from hydrogen, C 1-4 Alkyl, -C (O) -NR a R b and-S (O) m -NR a R b
Preferably, R 2 Selected from hydrogen, methyl, ethyl, isopropyl, -C (O) -NR a R b and-S (O) m -NR a R b
Preferably, R 2 Selected from hydrogen, -C (O) -NR a R b and-S (O) m -NR a R b
More preferably, R 2 Selected from:
hydrogen, hydrogen,
Figure BDA0001925977890000074
Figure BDA0001925977890000075
More preferably, R 2 Selected from:
hydrogen, hydrogen,
Figure BDA0001925977890000076
More preferably, R 2 Selected from:
hydrogen, hydrogen,
Figure BDA0001925977890000077
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 is c And R d Each independently selected from hydrogen and C 1-4 Alkyl radical, C 3-6 Cycloalkyl, hydroxy C 1-4 Alkyl radical, C 1-4 alkoxy-C 1-4 Alkyl-and C substituted by 5-or 6-membered heteroaryl 1-4 An alkyl group; or R c And R d Together with the nitrogen atom to which they are commonly attached, form a 4-to 6-membered azaheterocyclyl group, preferably a 5-or 6-membered azaheterocyclyl group.
In a preferred embodiment, R c And R d Each independently selected from hydrogen and C 1-4 An alkyl group; preferably, R c And R d Each independently selected from hydrogen, methyl, ethyl and isopropyl;
more preferably, R c And R d Each independently selected from hydrogen and methyl.
In some embodiments, the present invention provides a compound as described above, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N, thereof-an oxide, an isotopically-labelled compound, metabolite or prodrug, wherein R 3 Selected from hydrogen, halogen, CN, OH, C 1-4 Alkyl radical, C 1-4 Haloalkyl, C 3-6 Cycloalkyl, COOH, -C (O) O-C 1-4 Alkyl, -C (O) -NR c R d 、S(O) m OH、-S(O) m O-C 1-4 Alkyl and-S (O) m -NR c R d
In a preferred embodiment, R 3 Selected from hydrogen, halogen, CN, OH, C 1-4 Alkyl radical, C 1-4 Haloalkyl, COOH, -C (O) O-C 1-4 Alkyl, -C (O) -NR c R d 、S(O) m OH、-S(O) m O-C 1-4 Alkyl and-S (O) m -NR c R d
Preferably, R 3 Selected from hydrogen, F, Cl, Br, I, CN, OH, methyl, ethyl, isopropyl, CH 2 F、CHF 2 、CF 3 、COOH、-C(O)O-CH 3 、-C(O)O-Et、-C(O)O-iPr、-C(O)O-tBu、-C(O)-NR c R d 、S(O) m OH、-S(O) m O-CH 3 、-S(O) m O-Et、-S(O) m O-iPr、-S(O) m O-tBu and-S (O) m -NR c R d
Preferably, R 3 Selected from hydrogen, CN, COOH, -C (O) O-CH 3 、-C(O)O-Et、-C(O)O-iPr、-C(O)O-tBu、-C(O)-NR c R d 、S(O) m OH、-S(O) m O-CH 3 、-S(O) m O-Et、-S(O) m O-iPr、-S(O) m O-tBu and-S (O) m -NR c R d
Preferably, R 3 Selected from hydrogen, CN, COOH, -C (O) -NR c R d 、S(O) m OH and-S (O) m -NR c R d
Preferably, R 3 Selected from hydrogen, CN, COOH, -C (O) -NH 2 、-C(O)-NH(C 1-4 Alkyl), S (O) m OH、-S(O) m -NH 2 、-S(O) m -NH(C 1-4 Alkyl groups);
more preferably, R 3 Selected from hydrogen, CN, COOH, -C (O) -NH 2 、-C(O)-NHCH 3 、-C(O)-NH-Et、-C(O)-NH-iPr、S(O) m OH、-S(O) m -NH 2 、-S(O) m -NHCH 3 、-S(O) m -NH-Et、-S(O) m -NH-iPr;
More preferably, R 3 Selected from hydrogen, CN, COOH, -C (O) -NH 2 and-C (O) -NHCH 3
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, -C (O) -NR a R b and-S (O) m -NR a R b
Preferably selected from: hydrogen, hydrogen,
Figure BDA0001925977890000081
Figure BDA0001925977890000082
Preferably selected from: hydrogen, hydrogen,
Figure BDA0001925977890000083
Or
More preferably from hydrogen,
Figure BDA0001925977890000084
And is
R 3 Is hydrogen;
in 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, wherein:
R 2 is hydrogen, and
R 3 selected from hydrogen, halogen, CN, OH, COOH, -C (O) O-C 1-4 Alkyl, -C (O) -NR c R d 、S(O) m OH、-S(O) m O-C 1-4 Alkyl and-S (O) m -NR c R d
Preferably selected from hydrogen, CN, COOH, -C (O) -NH 2 、-C(O)-NH(C 1-4 Alkyl), S (O) m OH、-S(O) m -NH 2 、-S(O) m -NH(C 1-4 Alkyl groups);
more preferably from hydrogen, CN, COOH, -C (O) -NH 2 、-C(O)-NHCH 3 、-C(O)-NH-Et、-C(O)-NH-iPr、S(O) m OH、-S(O) m -NH 2 、-S(O) m -NHCH 3 、-S(O) m -NH-Et、-S(O) m -NH-iPr; or
More preferably from hydrogen, CN, COOH, -C (O) -NH 2 and-C (O) -NHCH 3
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 is 4 Independently at each occurrence, selected from hydrogen, halogen, CN, OH, C 1-4 Alkyl radical, C 1-4 Haloalkyl and C 1-4 An alkoxy group.
In a preferred embodiment, R 4 Independently at each occurrence is selected from hydrogen, F, Cl, Br, I, CN, OH, methyl, ethyl, isopropyl, CH 2 F、CHF 2 、CF 3 Methoxy, ethoxy and isopropoxy;
preferably, R 4 Independently at each occurrence, selected from hydrogen, F, Cl, Br, I, CN and OH;
preferably, R 4 Independently at each occurrence, selected from F, Cl, Br and I;
more preferably, R 4 Independently at each occurrence is F.
In some embodiments, the present invention provides a compound as described above, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide thereofA compound, isotopically-labelled compound, metabolite or prodrug, wherein R 5 Independently at each occurrence, selected from hydrogen, halogen, CN, OH, C 1-4 Alkyl radical, C 1-4 Haloalkyl and C 1-4 An alkoxy group.
In a preferred embodiment, R 5 Independently at each occurrence is selected from hydrogen, F, Cl, Br, I, CN, OH, methyl, ethyl, isopropyl, CH 2 F、CHF 2 、CF 3 Methoxy, ethoxy and isopropoxy;
preferably, R 5 Independently at each occurrence is selected from hydrogen, F, Cl, Br, I, CN, OH, methyl, ethyl, isopropyl, CH 2 F、CHF 2 、CF 3 Methoxy, ethoxy and isopropoxy;
more preferably, R 5 Independently at each occurrence 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 is 6 Independently at each occurrence, selected from hydrogen, halogen, CN, OH, C 1-4 Alkyl radical, C 1-4 Haloalkyl and C 1-4 An alkoxy group.
In a preferred embodiment, R 6 Independently at each occurrence is selected from hydrogen, F, Cl, Br, I, CN, OH, methyl, ethyl, isopropyl, CH 2 F、CHF 2 、CF 3 Methoxy, ethoxy and isopropoxy;
preferably, R 6 Independently at each occurrence, selected from methyl, ethyl, isopropyl, CH 2 F、CHF 2 And CF 3
Preferably, R 6 Independently at each occurrence, selected from the group consisting of methyl, ethyl and isopropyl;
more preferably, R 6 Independently at each occurrence is methyl.
In some embodiments, the present invention provides a compound 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, 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 CR 7
In a preferred embodiment, R 7 Selected from hydrogen, halogen, CN, OH, C 1-4 Alkyl radical, C 1-4 Haloalkyl and C 1-4 An alkoxy group;
preferably, R 7 Selected from hydrogen, F, Cl, Br, I, CN, OH, methyl, ethyl, isopropyl, CH 2 F、CHF 2 、CF 3 Methoxy, ethoxy and isopropoxy;
more preferably, R 7 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 N is 1 or 2, preferably 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 0 or 1, preferably 0.
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 1 or 2, preferably 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 (II):
Figure BDA0001925977890000091
wherein R is 1 、R 2 、R 3 、R 4 、R 5 、R 6 N and p are as defined above;
preferably, the compound has the structure of formula (III):
Figure BDA0001925977890000101
wherein R is 2 、R 3 、R 4 、R 5 、R 6 N and p are as defined above;
more preferably, the compound has the structure of formula (IV):
Figure BDA0001925977890000102
wherein R is 2 、R 3 、R 4 And R 6 As defined above;
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 BDA0001925977890000103
in another aspect, the present invention provides a method of making a compound of the present invention.
The reaction schemes described below illustrate general synthetic methods for the compounds of the present invention and are not limiting.
Route 1
Figure BDA0001925977890000104
Wherein R is 5 、R 6 P and q are as defined above; preferably, R 5 Is H; preferably, R 6 Is C 1 -4 alkyl, preferably methyl; preferably, p is 0; preferably, q is 1; LG (Ligno-lead-acid) 3 Is a leaving group such as halogen or sulfonyl ester, e.g. Cl, Br, I, tosylate, triflate, perfluorobutylsulfonate, preferably Br; m 2 is-B (OH) 2 、-Sn(C 1 -4 alkyl group) 3 -ZnCl, -ZnBr, -ZnI, preferably
Figure BDA0001925977890000111
The first step is as follows: subjecting compound 1 to a cyclization reaction to produce compound 2;
for example, the reaction may be carried out in the presence of a suitable ammonium salt (e.g. ammonium acetate) in an activated formic acid derivative (e.g. triethyl orthoformate) at a temperature of, for example, 60 ℃ to 140 ℃.
The second step is that: converting compound 2 to compound 3;
for example, the conversion reaction may be carried out under nitrogen protection in an organic solvent (e.g., toluene, xylene, tetrahydrofuran, ethylene glycol dimethyl ether, 1, 4-dioxane, N-dimethylformamide, dimethyl sulfoxide, or N-methylpyrrolidone) in an organic or inorganic base (e.g., sodium tert-butoxide, potassium tert-butoxide, cesium carbonate, potassium carbonate, cesium carbonate, sodium bicarbonate, potassium phosphate, potassium acetate) and a catalyst (e.g., a palladium catalyst such as tris (dibenzylideneacetone) dipalladium, tetrakis (triphenylphosphine) palladium, [1,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride, [1,1' -bis (diphenylphosphino) ferrocene]By reacting compound 2 with a suitable reagent (e.g. boron) in the presence of palladium dichloride dichloromethane complex, bis (triphenylphosphine) palladium dichloride, palladium acetate, at a temperature of e.g. 60 ℃ to 140 ℃Acid esters, e.g. pinacol ester of diboronic acid
Figure BDA0001925977890000112
) The reaction is completed.
Route 2
Figure BDA0001925977890000113
Wherein R is 1 、R 2 、R 3 、R 4 And n is as defined above; preferably, R 1 And R 3 Each is H; preferably, R 4 Is halogen, preferably F; preferably, n is 1; LG (Ligno-lead-acid) 1 And LG 2 Each is a leaving group such as halogen or sulfonyl ester, e.g. Cl, Br, I, tosylate, triflate, perfluorobutylsulfonate, preferably Cl, Br or I; m 1 is-B (OH) 2 、-Sn(C 1 -4 alkyl group) 3 -ZnCl, -ZnBr, -ZnI, or
Figure BDA0001925977890000114
preferably-B (OH) 2
The first step is as follows: carrying out nucleophilic substitution or coupling reaction on the compound 4 and the compound 5 to generate a compound 6;
for example, the reaction can be carried out under nitrogen protection in an organic solvent (e.g., toluene, xylene, tetrahydrofuran, ethylene glycol dimethyl ether, 1, 4-dioxane, N-dimethylformamide, dimethyl sulfoxide, or N-methylpyrrolidone) or a mixed system thereof with water, in the presence of an organic or inorganic base (e.g., sodium tert-butoxide, potassium tert-butoxide, cesium carbonate, potassium carbonate, cesium carbonate, sodium hydrogen carbonate, potassium phosphate, potassium acetate) and a catalyst (e.g., a palladium catalyst such as tris (dibenzylideneacetone) dipalladium, tetrakis (triphenylphosphine) palladium, [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride dichloromethane complex, bis (triphenylphosphine) palladium dichloride, palladium acetate), at a temperature of, for example, 60 ℃ to 140 ℃.
The second step is that: subjecting compound 6 to a substitution reaction to produce compound 7;
for example, the reaction may be carried out in an organic solvent (e.g., N-dimethylformamide, N-methylpyrrolidone, methanol, ethanol, tetrahydrofuran, 1, 4-dioxane) at, for example, room temperature with a suitable reagent such as N-bromosuccinimide.
The third step: carrying out coupling reaction on the compound 7 and the compound 3 to generate a compound 8;
for example, the reaction can be carried out under nitrogen protection in an organic solvent (e.g., toluene, xylene, tetrahydrofuran, ethylene glycol dimethyl ether, 1, 4-dioxane, N-dimethylformamide, dimethyl sulfoxide, or N-methylpyrrolidone) or a mixed system thereof with water, in the presence of an organic or inorganic base (e.g., sodium tert-butoxide, potassium tert-butoxide, cesium carbonate, potassium carbonate, cesium carbonate, sodium hydrogen carbonate, potassium phosphate, potassium acetate) and a catalyst (e.g., a palladium catalyst such as tris (dibenzylideneacetone) dipalladium, tetrakis (triphenylphosphine) palladium, [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride dichloromethane complex, bis (triphenylphosphine) palladium dichloride, palladium acetate), at a temperature of, for example, 60 ℃ to 140 ℃.
The fourth step: nucleophilic substitution of compound 8 with compound 9 produces a compound of formula (II).
For example, the reaction can be carried out in an organic solvent (e.g., dichloromethane, tetrahydrofuran, 1, 4-dioxane, diethyl ether, acetonitrile) in the presence of an organic base (e.g., N-diisopropylethylamine, triethylamine, pyridine, and 4-dimethylaminopyridine) and a suitable reagent (such as phenyl chloroformate), for example, at room temperature.
Route 3
Figure BDA0001925977890000121
Wherein R is 1 、R 2 、R 4 、R c 、R d And n is asAs defined above; preferably, R 1 And R 2 Each is H; preferably, R 4 Is halogen, preferably F; preferably, n is 1; LG (Ligno-lead-acid) 1 And LG 2 Each is a leaving group such as halogen or sulfonyl ester, e.g. Cl, Br, I, tosylate, triflate, perfluorobutylsulfonate, preferably Cl, Br or I; m 1 is-B (OH) 2 、-Sn(C 14 Alkyl radical) 3 -ZnCl, -ZnBr, -ZnI, or
Figure BDA0001925977890000122
preferably-B (OH) 2 (ii) a Hal is halogen, for example Cl, Br or I, preferably Br.
The first step is as follows: carrying out nucleophilic substitution or coupling reaction on the compound 10 and the compound 5 to generate a compound 11;
for example, the reaction can be carried out under nitrogen protection in an organic solvent (e.g., toluene, xylene, tetrahydrofuran, ethylene glycol dimethyl ether, 1, 4-dioxane, N-dimethylformamide, dimethyl sulfoxide, or N-methylpyrrolidone) or a mixed system thereof with water, in the presence of an organic or inorganic base (e.g., sodium tert-butoxide, potassium tert-butoxide, cesium carbonate, potassium carbonate, cesium carbonate, sodium hydrogen carbonate, potassium phosphate, potassium acetate) and a catalyst (e.g., a palladium catalyst such as tris (dibenzylideneacetone) dipalladium, tetrakis (triphenylphosphine) palladium, [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride dichloromethane complex, bis (triphenylphosphine) palladium dichloride, palladium acetate), at a temperature of, for example, 60 ℃ to 140 ℃.
The second step is that: subjecting compound 11 to a substitution reaction to produce compound 12;
for example, the reaction may be carried out in an organic solvent (e.g., N-dimethylformamide, N-methylpyrrolidone, methanol, ethanol, tetrahydrofuran, 1, 4-dioxane) at, for example, room temperature with a suitable reagent such as N-bromosuccinimide.
The third step: carrying out coupling reaction on the compound 12 and the compound 3 to generate a compound 13;
for example, the reaction can be carried out under nitrogen protection in an organic solvent (e.g., toluene, xylene, tetrahydrofuran, ethylene glycol dimethyl ether, 1, 4-dioxane, N-dimethylformamide, dimethyl sulfoxide, or N-methylpyrrolidone) or a mixed system thereof with water, in the presence of an organic or inorganic base (e.g., sodium tert-butoxide, potassium tert-butoxide, cesium carbonate, potassium carbonate, cesium carbonate, sodium hydrogen carbonate, potassium phosphate, potassium acetate) and a catalyst (e.g., a palladium catalyst such as tris (dibenzylideneacetone) dipalladium, tetrakis (triphenylphosphine) palladium, [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride dichloromethane complex, bis (triphenylphosphine) palladium dichloride, palladium acetate), at a temperature of, for example, 60 ℃ to 140 ℃.
The fourth step: subjecting compound 13 to a substitution reaction to produce compound 14;
for example, the reaction can be carried out in an organic solvent (e.g., N-dimethylformamide, N-methylpyrrolidone, methanol, ethanol, tetrahydrofuran, 1, 4-dioxane) at a temperature, e.g., room temperature, with a suitable reagent, such as N-bromosuccinimide.
The fifth step: subjecting compound 14 and compound 15 to a coupling reaction to produce compound 16;
for example, the reaction can be carried out under nitrogen protection in an organic solvent (e.g., 1, 4-dioxane, N-dimethylformamide, methanol, ethanol, toluene) in the presence of a catalyst (e.g., a palladium catalyst such as tris (dibenzylideneacetone) dipalladium, tetrakis (triphenylphosphine) palladium, [1,1' -bis (diphenylphosphino) ferrocene ] dichloropalladium dichloromethane complex, bis (triphenylphosphine) dichloropalladium, palladium acetate) under heating (e.g., microwave) at a temperature of, for example, 90 ℃ to 140 ℃.
And a sixth step: subjecting compound 16 to a hydrolysis reaction to produce compound 17;
for example, the reaction may be carried out in the presence of an inorganic base (e.g., sodium hydroxide, potassium hydroxide) in an organic solvent (e.g., methanol, ethanol, isopropanol, ethylene glycol, 1, 4-dioxane, dimethyl sulfoxide) or a mixed system thereof with water at a temperature of, for example, 60 ℃ to 140 ℃.
The seventh step: the compound 17 and the compound 18 are subjected to condensation reaction to produce the compound of the formula (II).
For example, the reaction can be carried out in an organic solvent (e.g., tetrahydrofuran, dichloromethane, 1, 4-dioxane, N-dimethylformamide) in the presence of a condensing agent (e.g., N ' -carbonyldiimidazole, 2- (7-azobenzotriazol) -N, N ' -tetramethyluronium hexafluorophosphate, benzotriazol-1-yloxytris (dimethylamino) phosphonium hexafluorophosphate, 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride, preferably N, N ' -carbonyldiimidazole) at a temperature such as room temperature.
In obtaining the compound of formula (II), in particular the compound of formula (IV) as described above, preferably wherein R 1 、R 2 And R 3 Each is H; r 4 Is halogen, preferably F; and R is 6 Is C 1-4 After the compound of formula (IV) with alkyl, preferably methyl, further compounds of the invention can be synthesized therefrom, for example, in which R is 1 、R 2 、R 3 、R 6 Compounds of formula (IV) having other meanings as defined above.
Pharmaceutical compositions and methods of treatment
As noted above, adenosine A2a receptor activation can mediate T cell immune function suppression, as noted above, and is associated with the development of tumors. The inventors of the present invention have found that the compounds of the present invention have adenosine receptor antagonistic activity, in particular, high selectivity for the A2a receptor relative to the a1 receptor. While not wishing to be bound by any theory, the inventors believe that by virtue of such activity, the compounds of the invention are capable of treating T cell immune function suppression (particularly mediated by adenosine A2a receptor activation) and diseases involving adenosine receptors (particularly adenosine A2a receptor, more particularly adenosine A2a receptor activation), including tumors.
Accordingly, another aspect of the present invention provides a pharmaceutical composition or 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, and one or more pharmaceutically acceptable carriers. The pharmaceutical composition is used for treating T cell immune function inhibition or diseases related to adenosine receptors.
In some embodiments, the pharmaceutical composition or pharmaceutical formulation may further comprise one or more other therapeutic agents, for example, other therapeutic agents for the treatment of T cell immune function suppression or diseases associated with adenosine receptors.
Another aspect of the invention provides a method of making a pharmaceutical composition or pharmaceutical formulation 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, and one or more pharmaceutically acceptable carriers.
Another aspect of the invention provides the use of a compound of the invention, or a pharmaceutically acceptable salt, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug, a pharmaceutical composition of the invention, or a pharmaceutical formulation of the invention, in the manufacture of a medicament for the treatment of an inhibition of T cell immune function in a subject.
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, a pharmaceutical composition of the invention, or a pharmaceutical formulation of the invention, for use in the treatment of an inhibition of T cell immune function in a subject.
Another aspect of the present invention provides a method of treating T cell immune function suppression in a subject, said method 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, a pharmaceutical composition of the present invention, or a pharmaceutical formulation of the present invention.
In preferred embodiments of these aspects, said suppression of T cell immune function is mediated by activation of the adenosine A2a receptor.
Another aspect of 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, a pharmaceutical composition of the present invention, or a pharmaceutical formulation of the present invention, in the manufacture of a medicament for the treatment of a disease associated with the adenosine receptor.
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, a pharmaceutical composition of the present invention or a pharmaceutical formulation of the present invention, for use in the treatment of a disease associated with the adenosine receptor.
Another aspect of the present invention provides a method of treating a disease associated with an adenosine receptor, 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, a pharmaceutical composition of the present invention, or a pharmaceutical formulation of the present invention.
In a preferred embodiment of these aspects, the adenosine receptor is the adenosine A2a receptor. In a preferred embodiment, the disease is associated with adenosine A2a receptor activation. Preferably, the disease is selected from the group consisting of tumors, including benign tumors and malignant tumors, preferably malignant tumors.
As described above, many studies have been conducted on adenosine A2a receptor antagonists for the treatment of central nervous system diseases. For example, CPI-444 has been used in clinical trials for the treatment of central nervous system disorders. WO0162233 and WO2002014282 disclose that aminopyridine compounds having adenosine A2a receptor antagonistic action can be used as therapeutic agents for parkinson's disease or senile dementia. WO0162233, WO2003035639, WO2004016605 and WO2005079801 disclose that aminopyrimidine compounds having adenosine A2a receptor antagonistic action can be used as therapeutic agents for diseases such as Parkinson's disease or neuralgia. WO2011095625 discloses aminotriazine compounds having adenosine A2a receptor antagonistic action as therapeutic agents for dyskinesias, stroke, or parkinson's disease. These studies indicate, on the one hand, that adenosine A2a receptor antagonists of the prior art are able to cross the blood-brain barrier; on the other hand, there is little research in the art regarding the anti-tumor use of adenosine A2a receptor antagonists. Moreover, if these active compounds disclosed in the prior art are used to treat peripheral tumors in an individual, they are likely to cause damage to the central nervous system of the individual, resulting in serious toxic side effects, due to their ability to cross the blood-brain barrier. In contrast, some compounds of the present invention are peripherally selective and do not cross the blood-brain barrier into the brain, and thus have low or no central nervous system toxic side effects in exerting anti-tumor efficacy against peripheral tumors. These compounds are good candidates for therapeutic agents for peripheral tumors.
Thus, in a preferred embodiment, the tumors described above are peripheral benign tumors and peripheral malignant tumors, preferably peripheral malignant tumors.
As used herein, the term "peripheral tumor" refers to tumors present in body sites other than the central nervous system, including benign tumors and malignant tumors, including head and neck, extremities, skin, eyes, ears, mouth, nose, tongue, lips, larynx, pharynx, esophagus, internal organs (e.g., bronchi and lungs, kidney, liver, gall bladder, pancreas, stomach, small intestine, large intestine, mesentery, peritoneum, breast, ovary, uterus, fallopian tube, bladder, ureter, prostate, adrenal gland, etc.) and connective tissues (including bones, blood, lymph), and the like.
As used herein, the term "pharmaceutically acceptable carrier" refers to a diluent, adjuvant, excipient, or vehicle with which a therapeutic agent is administered, 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 used in the pharmaceutical compositions or formulations 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. Examples of suitable pharmaceutically acceptable carriers are described in Remington's Pharmaceutical Sciences (1990).
The pharmaceutical composition or pharmaceutical formulation of the invention may act systemically and/or locally. For this purpose, they can be administered by a suitable route. For these routes of administration, administration may be in a suitable dosage form.
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.
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 administration may be given, several divided doses may be administered over time, or the dose may be proportionally reduced or increased depending on the therapeutic situation. It is noted that dosage values may vary with the type and severity of the condition, 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's needs.
The amount of a compound of the invention administered will depend on the severity of the individual, disorder or condition being treated, the frequency of administration, the disposition of the compound and the judgment of the prescribing physician. Generally, the effective dose is from about 0.0001 to about 50mg per kg body weight per day. In some cases, dosage levels not 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 to be administered throughout the day.
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 1000 mg.
As used herein, unless otherwise indicated, the term "treating" means reversing, alleviating, inhibiting, or eliminating the progression of, or preventing, a disease or disorder or one or more symptoms of such a disease or disorder to which such term applies.
As used herein, the term "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.).
Detailed Description
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 H NMR was measured using a JEOL Eclipse400 NMR spectrometer using deuterated methanol (CD) as the solvent 3 OD), deuterated chloroform (CDCl) 3 ) Or hexadeutero dimethyl sulfoxide (DMSO-d) 6 ) Internal standard is Tetramethylsilane (TMS) with chemical shift (delta) of 10 -6 (ppm) is given as a unit.
MS was measured with an Agilent (ESI) mass spectrometer, model Agilent 6120B, manufactured by Agilent.
The preparation method of the high performance liquid chromatograph comprises the following steps:
the instrument model is as follows: agilent 1260; a chromatographic column: waters Xbridge Prep C18OBD (19 mm. times.150 mm. times.5.0 μm); temperature of the chromatographic column: 25 ℃; flow rate: 20.0 mL/min; detection wavelength: 214 nm; elution gradient: (0 min: 10% A, 90% B; 16.0 min: 90% A, 10% B); a mobile phase A: acetonitrile; mobile phase B: 0.05% aqueous ammonium bicarbonate solution.
Thin layer chromatography silica gel plate (TLC) an aluminum plate (20X 20cm) from Merck was used, and GF 254(1mm) from Nicotiana Xinno chemical Co., Ltd was used for separation and purification by thin layer chromatography.
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 used a Biotage Initiator + (400W, RT-300 ℃) microwave reactor.
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 examples, the reaction temperature is room temperature (20 ℃ to 35 ℃);
the reagents used in the present invention were purchased from Acros Organics, Aldrich Chemical Company, Texas Chemical, and the like.
The abbreviations in this application have the following meanings:
abbreviations Means of
DMF N, N-dimethylformamide
DIPEA N, N-diisopropylethylamine
TLC Thin layer chromatography
Pd(dppf)Cl 2 [1,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride
Et Ethyl radical
iPr Isopropyl group
tBu Tert-butyl radical
Preparation of an intermediate:
intermediate preparation example 1: preparation of 4-methyl-6- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) quinazoline
Figure BDA0001925977890000151
The first step is as follows: preparation of 6-bromo-4-methyl quinazoline
1- (2-amino-5-bromophenyl) ethanone (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 the reaction was completed, it was cooled to room temperature, the reaction solution was concentrated, and the residue was diluted with water and extracted with ethyl acetate. The combined organic phases were dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated, and the residue was purified by silica gel column chromatography (eluent: ethyl acetate/petroleum ether: 1/3) to give the title compound of this step (2.6g, 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
To a solution of 6-bromo-4-methyl quinazoline (2g,8.9mmol) in 1, 4-dioxane (50mL) was added, under a nitrogen atmosphere, in order, pinacol diboron diboride (3.4g,13.5mmol), potassium acetate (1.76g,16.9mmol) and Pd (dppf) Cl 2 (0.65g,0.9mmol) and reacted 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 diluted with water and extracted with ethyl acetate. The combined organic phases were dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated, and the residue was purified by silica gel column chromatography (eluent: ethyl acetate/petroleum ether: 1/2) to give the title compound of the present step (2.1g, yield: 87%).
MS m/z(ESI):271.2[M+H] +
Preparation of the Compounds of the invention
Example 1: preparation of 6- (4-fluorophenyl) -5- (4-methylquinazolin-6-yl) pyrazin-2-amine
Figure BDA0001925977890000161
The first step is as follows: preparation of 6- (4-fluorophenyl) pyrazine-2-amine
6-chloropyrazin-2-amine (1g,7.72mmol), 4-fluorophenyl) boronic acid (1.40g,10.03mmol) and cesium carbonate (5.03g,15.44mmol) were added successively to a mixture of 1, 4-dioxane (30mL) and water (3mL), replaced three times with nitrogen, and Pd (dppf) Cl was added 2 (284.1mg,0.39mmol) and reacted at 95 ℃ for 12 hours. After the reaction was completed, the reaction solution was allowed to stand and filtered. The filtrate was concentrated, and the residue was purified by silica gel column chromatography (eluent: ethyl acetate/petroleum ether: 1/2) to give the title compound of this step (1.2g, yield: 82.2%).
MS m/z(ESI):190.1[M+H] +
The second step is that: preparation of 5-bromo-6- (4-fluorophenyl) pyrazin-2-amine
6- (4-fluorophenyl) pyrazin-2-amine (1g,5.29mmol) was dissolved in DMF (25mL), N-bromosuccinimide (940.8mg,5.29mmol) was added, and the reaction was allowed to proceed at 25 ℃ for 2 hours. The reaction solution was poured into water, stirred for 10 minutes, and then extracted three times with ethyl acetate. The organic phases were combined and washed three times with saturated brine, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated, and the residue was purified by silica gel column chromatography (eluent: ethyl acetate/petroleum ether: 1/2) to give the title compound of this step (700mg, yield: 49.4%).
MS m/z(ESI):268.0[M+H] +
The third step: preparation of 6- (4-fluorophenyl) -5- (4-methylquinazolin-6-yl) pyrazin-2-amine
5-bromo-6- (4-fluorophenyl) pyrazin-2-amine (3.0g,11.2mmol) is added to a mixture of 1, 4-dioxane (100mL) and water (20mL), followed by the sequential addition of 4-methyl-6- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) quinazoline (3.6g,13.4mmol), potassium carbonate (3.1g,22.4mmol) and Pd (dppf) Cl 2 (0.4g,0.6mmol), and the reaction mixture was purged with nitrogen 3 times and then reacted at 100 ℃ for 4 hours. After the reaction, the reaction mixture was cooled to room temperature, poured into water, and extracted with ethyl acetate. The organic phases were combined and dried over anhydrous sodium sulfate, then filtered. The filtrate was concentrated, and the residue was purified by silica gel column chromatography (eluent: ethyl acetate/petroleum ether: 3/1) to give the title compound (3.0g, yield: 81%).
MS m/z(ESI):332.1[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ:9.06(s,1H),8.06(d,J=9.2Hz,2H),7.84(d,J=2.0Hz,2H),7.41-7.37(m,2H),7.15(t,J=8.8Hz,2H),6.85(s,2H),2.69(s,3H)。
Example 2: preparation of 3- (6- (4-fluorophenyl) -5- (4-methylquinazolin-6-yl) pyrazin-2-yl) -1-methyl-1- (pyridin-2-ylmethyl) urea
Figure BDA0001925977890000162
6- (4-fluorophenyl) -5- (4-methylquinazolin-6-yl) pyrazin-2-amine (100.0mg,0.3mmol) was dissolved in tetrahydrofuran (15mL), DIPEA (0.6mL,3.2mmol) and phenyl chloroformate (89.0mg,0.3mmol) were added at-40 ℃ and reacted at-40 ℃ for 2 hours. Then, N-methyl-1- (pyridin-2-yl) methylamine (70.4mg,0.6mmol) was added and reacted at 0 ℃ for 2 hours. After the completion of the reaction, the reaction solution was concentrated, and the residue was purified by silica gel column chromatography (eluent: ethyl acetate) to obtain the title compound (55.0mg, yield: 37%).
MS m/z(ESI):480.2[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ:9.20(s,1H),9.10(s,1H),8.58(d,J=4.0Hz,1H),8.23(d,J=1.2Hz,1H),8.00-7.72(m,3H),7.58-7.42(m,3H),7.43-7.26(m,2H),7.18(t,J=9.2Hz,2H),4.74(s,2H),3.08(s,3H),2.74(s,3H)。
Example 3: preparation of N- (6- (4-fluorophenyl) -5- (4-methylquinazolin-6-yl) pyrazin-2-yl) morpholine-4-carboxamide
Figure BDA0001925977890000171
The title compound of example 3 (110.0mg, yield: 82%) was synthesized in a similar manner to the procedure described in example 2, substituting morpholine for N-methyl-1- (pyridin-2-yl) methylamine in example 2.
MS m/z(ESI):445.2[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ:9.85(s,1H),9.16(s,1H),9.10(s,1H),8.22(d,J=1.6Hz,1H),7.92-7.87(m,2H),7.61-7.40(m,2H),7.20-7.17(m,2H),3.76-3.58(m,4H),3.59-3.45(m,4H),2.73(s,3H)。
Example 4: preparation of 3-amino-5- (4-fluorophenyl) -6- (4-methylquinazolin-6-yl) pyrazine-2-carbonitrile
Figure BDA0001925977890000172
The first step is as follows: preparation of 3-bromo-6- (4-fluorophenyl) -5- (4-methylquinazolin-6-yl) pyrazin-2-amine
6- (4-fluorophenyl) -5- (4-methylquinazolin-6-yl) pyrazin-2-amine (200.0mg,0.6mmol) was dissolved in DMF (15mL), N-bromosuccinimide (107.4mg,0.6mmol) was added and the reaction was allowed to proceed at 25 ℃ for 2 h. The reaction solution was poured into water and filtered. The filter cake was washed with water and then dried to give the title compound of this step (220.0mg, yield: 84%).
MS m/z(ESI):410.0[M+H] +
The second step is that: preparation of 3-amino-5- (4-fluorophenyl) -6- (4-methylquinazolin-6-yl) pyrazine-2-carbonitrile
3-bromo-6- (4-fluorophenyl) -5- (4-methylquinazolin-6-yl) pyrazin-2-amine (0.9g, 2.2mmol) and cuprous cyanide (0.4g, 4.4mmol) were added to DMF (8mL) and reacted in a microwave at 120 ℃ for 2 hours. After completion of the reaction, extraction was performed with water and ethyl acetate. The organic phases were combined and washed with saturated brine, then the organic phases were concentrated, and the obtained residue was purified by high performance liquid chromatography to obtain the title compound (590.0mg, yield: 71%).
MS m/z(ESI):357.1[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ:9.08(s,1H),8.12(d,J=1.2Hz,1H),7.84-7.78(m,2H),7.69(s,2H),7.43(dd,J=8.8,5.6Hz,2H),7.19(dd,J=16.0,5.6Hz,2H),2.72(s,3H)。
Example 5: preparation of 3-amino-5- (4-fluorophenyl) -6- (4-methylquinazolin-6-yl) pyrazine-2-carboxylic acid
Figure BDA0001925977890000173
Sodium hydroxide (0.3g,7.50mmol) was added to a mixture of ethanol (2mL) and water (5mL), followed by 3-amino-5- (4-fluorophenyl) -6- (4-methylquinazolin-6-yl) pyrazine-2-carbonitrile (0.1g,0.3mmol) and reacted at 100 ℃ for 2 hours. The reaction solution was extracted with ethyl acetate, and the pH of the aqueous phase was adjusted to 2 with 1N hydrochloric acid, followed by concentration. Dichloromethane was added for dilution and then filtered. The filtrate was concentrated, and the residue was purified by silica gel thin layer chromatography preparation plate (developing solvent: dichloromethane/methanol-10/1) to give the title compound (20.0mg, yield: 18%).
MS m/z(ESI):376.1[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ:9.08(s,1H),8.15(d,J=1.2Hz,2H),7.81-7.76(m,2H),7.69(s,2H),7.43(dd,J=8.8,5.6Hz,2H),7.22-7.18(m,2H),2.89(s,3H)。
Example 6: preparation of 3-amino-5- (4-fluorophenyl) -6- (4-methylquinazolin-6-yl) pyrazine-2-carboxamide
Figure BDA0001925977890000181
3-amino-5- (4-fluorophenyl) -6- (4-methylquinazolin-6-yl) pyrazine-2-carboxylic acid (100.0mg, 0.27mmol) and DIPEA (172.2mg, 1.33mmol) were added to tetrahydrofuran (30mL), followed by ammonium chloride (71.3mg, 1.33mmol) and benzotriazol-1-yloxytris (dimethylamino) phosphonium hexafluorophosphate (108.8mg, 0.53mmol), and reacted at 25 ℃ for 4 hours. Then, the reaction solution was concentrated, and the residue was purified by high performance liquid chromatography to give the title compound (35mg, yield: 33%).
MS m/z(ESI):375.1[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ:9.07(s,1H),8.32(s,1H),8.23(d,J=1.6Hz,1H),8.00(dd,J=8.8,2.0Hz,2H),7.89-7.64(m,3H),7.46(dd,J=8.8,5.6Hz,2H),7.19(t,J=8.8Hz,2H),2.75(s,3H)。
Example 7: preparation of 3-amino-5- (4-fluorophenyl) -N-methyl-6- (4-methylquinazolin-6-yl) pyrazine-2-carboxamide
Figure BDA0001925977890000182
The title compound of example 7 (18.0mg, yield: 11%) was synthesized in a similar manner to the procedure described in example 6, using 2M methylamine tetrahydrofuran solution in place of ammonium chloride in example 6.
MS m/z(ESI):389.1[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ:9.08(s,1H),8.81(d,J=4.8Hz,1H),8.16(d,J=1.6Hz,1H),8.06(dd,J=8.8,2.0Hz,1H),7.86(d,J=8.8Hz,3H),7.55-7.32(m,2H),7.30-7.03(m,2H),2.87(d,J=4.8Hz,3H),2.72(s,3H)。
Biological assay
Experimental example 1 determination of competitive inhibition constants (Ki) for adenosine A1 and A2a receptors
Reagent:
[ 3 H]-DPCPX:PerkinElmer,NET974250UC
[ 3 H]-CGS-21680:PerkinElmer,NET1021250UC
a1 receptor cell membrane (human): perkinelmer, ES-010-M400UA
A2a receptor cell membrane (human): PerkinElmer, RBHA2AM400UA
Microscint 20cocktail scintillation fluid: PerkinElmer, 6013329
PEI(Poly ethyleneimine):Sigma,P3143
CGS15943:Sigma,C199
Instruments and consumables:
MicroBeta2Reader,PerkinElmer
unifilter-96GF/C filter plate: perkin Elmer, 6005174
96-well plate: agilent, 5042-1385
Buffer solution:
a1 assay buffer: 25mM HEPES, 5mM MgCl 2 ,1mM CaCl 2 100mM NaCl, pH 7.4. The buffer solution is used for diluting A1 receptor cell membrane (human source) and 3 H]-DPCPX。
a1 wash buffer: 25mM HEPES, 5mM MgCl 2 ,1mM CaCl 2 ,100mM NaCl,pH 7.4。
A2a assay buffer: 50mM Tris-HCl, 10mM MgCl 2 1mM EDTA, pH 7.4. The buffer solution is used for diluting A2a receptor cell membrane (human source) and 3 H]-CGS-21680。
a2a wash buffer: 50mM Tris-HCl, 154mM NaCl, pH 7.4.
The experimental method comprises the following steps:
diluting the A1 receptor cell membrane to 0.025 mu g/mu l by adopting an A1 experiment buffer solution to obtain an A1 receptor cell membrane diluent; a2a receptor cell membranes were diluted to 0.05. mu.g/. mu.l with A2a assay buffer to give A2a receptor cell membrane dilutions.
Test compounds and CGS15943 were diluted in DMSO gradient. Mu.l each of the test compound, high control (0.5% DMSO), and low control (1000nM CGS15943) was added to a 96-well plate, and then 100. mu. l A1 diluted cell membrane of receptor (containing 2.5. mu.g of cell membrane) was added to each well to obtain an A1 assay plate. Mu.l each of the test compound, high control (0.5% DMSO), and low control (1000nM CGS15943) was added to a 96-well plate, and then 100. mu. l A2a diluted cell membrane of receptor (containing 5.0. mu.g of cell membrane) was added to each well to obtain an A2a assay plate.
To an A1 detection plate, 100. mu.l of a radioisotope labeled ligand [ alpha ], [ beta ] -peptide 3 H]DPCPX (diluted with A1 assay buffer, working concentration 1.0 nM). To a A2a detection plate was added 100. mu.l of a radioisotope labeled ligand [ alpha ], [ solution ] 3 H]CGS-21680 (diluted with A2a assay buffer, working concentration 6.0nM), blocked with tape A1 and A2a assay plates and incubated at room temperature for 1h and 2h, respectively.
A Unifilter-96GF/C filter plate was prepared by adding 50. mu.l of 0.3% PEI to each well of the Unifilter-96GF/C filter plate and incubating at room temperature for not less than 0.5 h.
And respectively transferring the reaction liquid in the incubated A1 detection plate and A2a detection plate to two Unifilter-96GF/C filter plates, respectively washing by using pre-cooled corresponding washing buffer solutions, and then drying the filter plates. After sealing the bottom of the filter plate, 50. mu.l Microscint 20cocktail scintillation fluid was added, then 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 was 100 — (experimental well signal value-low vs. well signal average)/(high vs. well signal average-low vs. well signal average) × 100.
Fitting IC Using XLfit 50 。Ki=IC 50 /(1+ isotopic concentration/Kd); wherein Kd is the dissociation constant of the isotopically labeled ligand.
The results are detailed in table 1:
TABLE 1 competitive inhibition constants (Ki) for the compounds of the invention for adenosine A2a, A1 receptors
Example numbering A1,Ki(nM) A2a,Ki(nM) A1/A2a, Ki ratio
1 202.4 3.5 58.6
2 1933.7 3.3 581.2
3 326.0 5.2 62.9
4 132.8 1.7 78.3
6 891.1 4.8 183.8
7 1036.5 8.8 117.7
The data in table 1 show that the compounds of the invention have good affinity for the adenosine A2a receptor and weaker affinity for the adenosine a1 receptor.
Experimental example 2: rat Pharmacokinetic (PK) study
The compounds of the invention were administered to male SD rats by Intravenous (IV) and intragastric (PO) administration, respectively, to investigate pharmacokinetic characteristics. The dosages of IV and PO were 1mg/kg and 5mg/kg, respectively, with IV vehicle being 5% DMSO: 5% Solutol: 90% physiological saline, and PO vehicle being 0.5% MC (sodium methyl cellulose). Blood was collected at time points before (0h) IV administration and 0.083, 0.25, 0.5, 1, 2, 4, 6, 8 and 24h after administration, blood was collected at time points before (0h) PO administration and 0.25, 0.5, 1, 2, 4, 6, 8 and 24h after administration, blood was 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 tables 2 and 3.
TABLE 2 pharmacokinetic parameters of IV administered Compounds in rats
Figure BDA0001925977890000191
As shown in Table 2, the in vivo exposure (AUC) in rats by the compound of example 6 administered IV at a dose of 1mg/kg last ) 1688h ng/mL, corresponding to maximum blood concentration (C) max ) 951ng/mL indicates that the compound of example 6 has excellent drug exposure in rats by IV administration.
TABLE 3 pharmacokinetic parameters of PO dosed Compounds in rats
Figure BDA0001925977890000201
As shown in Table 3, AUC in rat blood of the compound of example 6 administered by PO at a dose of 5mg/kg last 2802h ng/mL C in rat blood max 525ng/mL, indicating that the compound of example 6 has excellent drug exposure in the rat blood system by PO administration.
The bioavailability of compound 6 orally administered to rats was calculated to be 32.4% compared to intravenous administration, indicating that the compound of the present invention (e.g., the compound of example 6) has an excellent oral absorption effect in rats.
Various modifications of the invention in addition to those described herein will be apparent to those skilled in the art from the foregoing description. Such modifications are also intended to fall within the scope of the appended claims. Each reference (including all patents, patent applications, journal articles, books, and any other publications) cited in this application is hereby incorporated by reference in its entirety.

Claims (62)

1. A compound or a pharmaceutically acceptable salt thereof, wherein the compound has the structure of formula (I):
Figure FDA0003705527180000011
wherein:
x is N;
R 1 and R 2 Each independently selected from hydrogen and-C (O) -NR a R b
R 3 Selected from hydrogen, CN, COOH, and-C (O) -NR c R d
R 4 、R 5 And R 6 Each independently at each occurrence is selected from the group consisting of hydrogen, halogen, and C 1-6 An alkyl group;
R a and R b Each independently selected from hydrogen and C 1-6 Alkyl and C substituted by 5-or 6-membered heteroaryl 1-6 An alkyl group; or R a And R b Together with the nitrogen atom to which they are commonly attached form a 3-to 6-membered azaheterocyclyl;
R c and R d Each independently selected from hydrogen and C 1-6 An alkyl group;
n is 0, 1 or 2;
p is 0, 1 or 2; and is
q is 0, 1 or 2;
and when R is 1 、R 2 、R 4 、R 5 And R 6 When both are hydrogen, R 3 Not hydrogen or CN.
2. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein R 1 Is hydrogen.
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 1-4 Alkyl and C substituted by 5-or 6-membered heteroaryl 1-4 An alkyl group; or
R a And R b Together with the nitrogen atom to which they are commonly attached, form a 4-to 6-membered azaheterocyclyl.
4. The compound of claim 3, or a pharmaceutically acceptable salt thereof, wherein R a And R b Each independently selected from C 1-4 Alkyl and C substituted by 5-or 6-membered heteroaryl 1-4 An alkyl group.
5. The compound of claim 4, or a pharmaceutically acceptable salt thereof, wherein R a And R b Each independently selected from C 1-4 Alkyl and C substituted by 6-membered heteroaryl 1-4 An alkyl group.
6. The compound of claim 5, or a pharmaceutically acceptable salt thereof, wherein R a And R b Each independently selected from C 1-4 Alkyl and C substituted by pyridyl 1-4 An alkyl group.
7. The compound of claim 6, or a pharmaceutically acceptable salt thereof, wherein R a And R b Each independently selected from methyl, ethyl, isopropyl and methyl, ethyl and isopropyl substituted with pyridyl.
8. The compound of claim 7, or a pharmaceutically acceptable salt thereof, wherein R a And R b One selected from methyl, ethyl, isopropyl and the other selected from methyl, ethyl and isopropyl substituted with pyridyl.
9. The compound of claim 8, or a pharmaceutically acceptable salt thereof, wherein R a And R b One of which is methyl and the other is methyl substituted by pyridyl.
10. The compound of claim 9, or a pharmaceutically acceptable salt thereof, wherein R a And R b One of which is methyl and the other is
Figure FDA0003705527180000021
11. The compound of claim 3, or a pharmaceutically acceptable salt thereof, wherein R a And R b Together with the nitrogen atom to which they are commonly attached, form a 5 or 6 membered azaheterocyclyl.
12. The compound of claim 11, or a pharmaceutically acceptable salt thereof, wherein R a And R b Together with the nitrogen atom to which they are commonly attached, form a 6-membered azaheterocyclyl.
13. The compound of claim 12, or a pharmaceutically acceptable salt thereof, wherein R a And R b Together with the nitrogen atom to which they are jointly attached
Figure FDA0003705527180000022
14. The compound of claim 13, or a pharmaceutically acceptable salt thereof, wherein R a And R b Together with the nitrogen atom to which they are jointly attached
Figure FDA0003705527180000023
15. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein R 2 Selected from:
hydrogen, hydrogen,
Figure FDA0003705527180000024
16. The compound of claim 15, or a pharmaceutically acceptable salt thereof, wherein R 2 Selected from:
hydrogen, hydrogen,
Figure FDA0003705527180000025
17. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein R c And R d Each independently selected from hydrogen and C 1-4 An alkyl group.
18. The compound of claim 17, or a pharmaceutically acceptable salt thereof, wherein R c And R d Each independently selected from hydrogen, methyl, ethyl and isopropyl.
19. The compound of claim 18, or a pharmaceutically acceptable salt thereof, wherein R c And R d Each independently selected from hydrogen and methyl.
20. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein R 3 Selected from hydrogen, CN, COOH, -C (O) -NH 2 and-C (O) -NH (C) 1-4 Alkyl groups).
21. The compound of claim 20, or a pharmaceutically acceptable salt thereof, wherein R 3 Selected from hydrogen, CN, COOH, -C (O) -NH 2 、-C(O)-NHCH 3 C (O) -NH-Et and C (O) -NH-iPr.
22. The compound of claim 21, or a pharmaceutically acceptable salt thereof, wherein R 3 Selected from hydrogen, CN, COOH, -C (O) -NH 2 and-C (O) -NHCH 3
23. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein:
R 2 selected from: hydrogen and-C (O) -NR a R b (ii) a And is provided with
R 3 Is hydrogen;
or
R 2 Is hydrogen, and
R 3 selected from hydrogen, CN, COOH, -C (O) -NR c R d
24. The compound of claim 23, or a pharmaceutically acceptable salt thereof, wherein:
R 2 selected from: hydrogen, hydrogen,
Figure FDA0003705527180000031
25. The compound of claim 24, or a pharmaceutically acceptable salt thereof, wherein:
R 2 selected from hydrogen,
Figure FDA0003705527180000032
26. The compound of claim 23, or a pharmaceutically acceptable salt thereof, wherein:
R 3 selected from hydrogen, CN, COOH, -C (O) -NH 2 、-C(O)-NH(C 1-4 Alkyl groups).
27. The compound of claim 26, or a pharmaceutically acceptable salt thereof, wherein:
R 3 selected from hydrogen, CN, COOH, -C (O) -NH 2 、-C(O)-NHCH 3 、-C(O)-NH-Et、-C(O)-NH-iPr。
28. The compound of claim 27, or a pharmaceutically acceptable salt thereof, wherein:
R 3 selected from hydrogen, CN, COOH, -C (O) -NH 2 and-C (O) -NHCH 3
29. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein R 4 Independently at each occurrence, selected from hydrogen, halogen, C 1-4 An alkyl group.
30. The compound of claim 29, or a pharmaceutically acceptable salt thereof, wherein R 4 Independently at each occurrence is selected from hydrogen, F, Cl, Br, I, methyl, ethyl, isopropyl.
31. The compound of claim 30, or a pharmaceutically acceptable salt thereof, wherein R 4 Independently at each occurrence is selected from hydrogen, F, Cl, Br, I.
32. The compound of claim 31, or a pharmaceutically acceptable salt thereof, wherein R 4 Independently at each occurrence is selected from F, Cl, Br and I.
33. The compound of claim 32, or a pharmaceutically acceptable salt thereof, wherein R 4 Independently at each occurrence is F.
34. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein R 5 Independently at each occurrence, selected from hydrogen, halogen, C 1-4 An alkyl group.
35. The compound of claim 34, or a pharmaceutically acceptable salt thereof, wherein R 5 Independently at each occurrence is selected from hydrogen, F, Cl, Br, I, methyl, ethyl, isopropyl.
36. The compound of claim 35, or a pharmaceutically acceptable salt thereof, wherein R 5 Independently at each occurrence is hydrogen.
37. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein R 6 Independently at each occurrence, selected from hydrogen, halogen, C 1-4 An alkyl group.
38. The compound of claim 37, or a pharmaceutically acceptable salt thereof, wherein R 6 Independently at each occurrence is selected from hydrogen, F, Cl, Br, I, methyl, ethyl, isopropyl.
39. The compound of claim 38, or a pharmaceutically acceptable salt thereof, wherein R 6 Independently at each occurrence, is selected from methyl, ethyl, isopropyl.
40. The compound of claim 39, or a pharmaceutically acceptable salt thereof, wherein R 6 Independently at each occurrence is methyl.
41. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein n is 1 or 2.
42. The compound of claim 41, or a pharmaceutically acceptable salt thereof, wherein n is 1.
43. The compound of claim 42, or a pharmaceutically acceptable salt thereof, wherein p is 0 or 1.
44. The compound of claim 43, or a pharmaceutically acceptable salt thereof, wherein p is 0.
45. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein q is 1 or 2.
46. The compound of claim 45, or a pharmaceutically acceptable salt thereof, wherein q is 1.
47. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein the compound has the structure of formula (II):
Figure FDA0003705527180000041
wherein R is 1 、R 2 、R 3 、R 4 、R 5 、R 6 N and p are as defined in any one of claims 1 to 46.
48. The compound of claim 47, or a pharmaceutically acceptable salt thereof, wherein the compound has the structure of formula (III):
Figure FDA0003705527180000042
wherein R is 2 、R 3 、R 4 、R 5 、R 6 N and p are as defined in any one of claims 1 to 46.
49. The compound of claim 48, or a pharmaceutically acceptable salt thereof, wherein the compound has the structure of formula (IV):
Figure FDA0003705527180000051
wherein R is 2 、R 3 、R 4 And R 6 As defined in any one of claims 1 to 46.
50. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein the compound is selected from the group consisting of:
Figure FDA0003705527180000052
51. a pharmaceutical composition comprising a compound of any one of claims 1 to 50, or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable carriers.
52. A method of making a pharmaceutical composition comprising combining a compound of any one of claims 1 to 50, or a pharmaceutically acceptable salt thereof, with one or more pharmaceutically acceptable carriers.
53. Use of a compound of any one of claims 1 to 50, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of claim 51, in the manufacture of a medicament for treating suppression of T cell immune function in a subject.
54. The use of claim 53, wherein said suppression of T cell immune function is mediated by activation of the adenosine A2a receptor.
55. Use of a compound according to any one of claims 1 to 50, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 51, in the manufacture of a medicament for the treatment of a disease associated with the adenosine receptor.
56. The use of claim 55, wherein the adenosine receptor is the adenosine A2a receptor.
57. The use according to claim 56, wherein the disease is associated with adenosine A2a receptor activation.
58. The use of claim 57, wherein the disease is selected from the group consisting of tumors, including benign tumors and malignant tumors.
59. The use of claim 58, wherein the disease is a malignant tumor.
60. The use of claim 58, wherein the tumor is a peripheral benign tumor or a peripheral malignant tumor.
61. The use of claim 60, wherein the tumor is a peripheral malignancy.
62. The use of any one of claims 53 to 61, wherein the medicament is a medicament administered by oral, intravenous, intra-arterial, subcutaneous, intraperitoneal, intramuscular, or transdermal routes.
CN201811616735.1A 2018-12-28 2018-12-28 Substituted pyrazine compounds, their preparation and use Active CN111377906B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201811616735.1A CN111377906B (en) 2018-12-28 2018-12-28 Substituted pyrazine compounds, their preparation and use

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201811616735.1A CN111377906B (en) 2018-12-28 2018-12-28 Substituted pyrazine compounds, their preparation and use

Publications (2)

Publication Number Publication Date
CN111377906A CN111377906A (en) 2020-07-07
CN111377906B true CN111377906B (en) 2022-09-02

Family

ID=71220832

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201811616735.1A Active CN111377906B (en) 2018-12-28 2018-12-28 Substituted pyrazine compounds, their preparation and use

Country Status (1)

Country Link
CN (1) CN111377906B (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007017096A1 (en) * 2005-07-29 2007-02-15 Laboratorios Almirall, S.A. Pyrazine derivatives useful as adenosine receptor antagonists
CN101468965A (en) * 2003-03-24 2009-07-01 默克公司 Biaryl substituted 6-membered heterocyles as sodium channel blockers
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
CN111163780A (en) * 2017-07-18 2020-05-15 诺维逊生物股份有限公司 Heterocyclic compounds as adenosine antagonists

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BRPI0415863A (en) * 2003-10-27 2007-01-09 Astellas Pharma Inc pyrazine derivatives and their pharmaceutical uses

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101468965A (en) * 2003-03-24 2009-07-01 默克公司 Biaryl substituted 6-membered heterocyles as sodium channel blockers
WO2007017096A1 (en) * 2005-07-29 2007-02-15 Laboratorios Almirall, S.A. Pyrazine derivatives useful as adenosine receptor antagonists
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
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
CN111163780A (en) * 2017-07-18 2020-05-15 诺维逊生物股份有限公司 Heterocyclic compounds as adenosine antagonists

Also Published As

Publication number Publication date
CN111377906A (en) 2020-07-07

Similar Documents

Publication Publication Date Title
EP3558989B1 (en) Triazolo[1,5-a]pyridine derivatives as immunomodulators
EP3558973B1 (en) Pyridine derivatives as immunomodulators
EP3390361B1 (en) N-phenyl-pyridine-2-carboxamide derivatives and their use as pd-1/pd-l1 protein/protein interaction modulators
EP3472167B1 (en) Heterocyclic compounds as immunomodulators
EP3394033B1 (en) Heterocyclic compounds as immunomodulators
JP2023126907A (en) Rho-related protein kinase inhibitor, pharmaceutical composition containing the same, and method of preparation and use thereof
WO2021249057A1 (en) Heterocyclic compound and use thereof
AU2017376629A1 (en) Benzimidazole compounds as c-Kit inhibitors
TW201833102A (en) Amine-substituted heterocyclic compounds as ehmt2 inhibitors and methods of use thereof
CN105524048A (en) Indazole compound as FGFR kinase inhibitor and its preparation method and use
CN112920180A (en) Glutaminase inhibitors
CN110582491A (en) Rho-related protein kinase inhibitor, pharmaceutical composition containing same, and preparation method and application thereof
JP2024505732A (en) Pyridopyrimidinone derivatives and their production methods and uses
CN112457308B (en) Novel tricyclic aromatic heterocyclic compound, preparation method, pharmaceutical composition and application thereof
CN116568681A (en) SOS1 inhibitor, pharmaceutical composition containing same and application thereof
TW201917129A (en) Triacyclic derivatives containing pyrazole, their preparation methods and applications thereof
WO2021041976A1 (en) Perk inhibiting indolinyl compounds
CA3130245A1 (en) Heterocyclic compound, pharmaceutical composition comprising same, preparation method therefor, and use thereof
CN113166119A (en) Substituted aryl compound and preparation method and application thereof
TW202140446A (en) Quinolyl phosphine oxide compound, and composition and application thereof
CN111377906B (en) Substituted pyrazine compounds, their preparation and use
CN111377873B (en) Aminopyrimidine compounds, their preparation and use
CN113166109B (en) Aminopyridine compound and preparation method and application thereof
AU2018260390A1 (en) Methods of use for trisubstituted benzotriazole derivatives as dihydroorotate oxygenase inhibitors
WO2021233454A1 (en) Novel tricyclic aromatic heterocyclic compound and preparation method therefor, pharmaceutical composition and use thereof

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