WO2016184313A1 - 羟基嘌呤类化合物及其应用 - Google Patents

羟基嘌呤类化合物及其应用 Download PDF

Info

Publication number
WO2016184313A1
WO2016184313A1 PCT/CN2016/081103 CN2016081103W WO2016184313A1 WO 2016184313 A1 WO2016184313 A1 WO 2016184313A1 CN 2016081103 W CN2016081103 W CN 2016081103W WO 2016184313 A1 WO2016184313 A1 WO 2016184313A1
Authority
WO
WIPO (PCT)
Prior art keywords
mmol
methyl
dione
pyrimidine
group
Prior art date
Application number
PCT/CN2016/081103
Other languages
English (en)
French (fr)
Inventor
吴凌云
张鹏
张丽
陈曙辉
Original Assignee
南京明德新药研发股份有限公司
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 南京明德新药研发股份有限公司 filed Critical 南京明德新药研发股份有限公司
Priority to US15/575,315 priority Critical patent/US10618898B2/en
Priority to CN201680029214.9A priority patent/CN107614501B/zh
Publication of WO2016184313A1 publication Critical patent/WO2016184313A1/zh

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D473/00Heterocyclic compounds containing purine ring systems
    • C07D473/02Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6
    • C07D473/04Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6 two oxygen atoms
    • C07D473/06Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6 two oxygen atoms with radicals containing only hydrogen and carbon atoms, attached in position 1 or 3
    • C07D473/10Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6 two oxygen atoms with radicals containing only hydrogen and carbon atoms, attached in position 1 or 3 with methyl radicals in positions 3 and 7, e.g. theobromine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D473/00Heterocyclic compounds containing purine ring systems
    • C07D473/02Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6
    • C07D473/04Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6 two oxygen atoms
    • C07D473/06Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6 two oxygen atoms with radicals containing only hydrogen and carbon atoms, attached in position 1 or 3
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D475/00Heterocyclic compounds containing pteridine ring systems
    • C07D475/02Heterocyclic compounds containing pteridine ring systems with an oxygen atom directly attached in position 4
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Definitions

  • the present invention relates to a series of hydroxy steroids and their use as PDE2 or TNF ⁇ inhibitors, in particular to compounds of the formula ( ⁇ ), tautomers thereof or pharmaceutically acceptable salts thereof.
  • Phosphodiesterase catalyzes the hydrolysis of cyclized nucleotides cGMP and cAMP, and regulates various physiological responses by controlling the intramolecular concentrations of these two important secondary signaling factors. Cyclization of nucleotides cGMP and cAMP intramolecular Abnormal regulation is the cause of many diseases. Several drugs have been used to improve and treat diseases by inhibiting PDE activity, such as PDE5 inhibitors for pulmonary hypertension and PDE4 inhibitors for arthritis caused by psoriasis. There are eleven major classes of phosphodiesterase genes, each of which can express several subtypes. There are more than 100 PDE subtypes in total. Different subtypes have different structures, different tissue distributions, and circularized nuclei. The activities of cGMP and cAMP are also very different, and the physiological functions of regulation are also very different.
  • PDE2 phosphodiesterase can catalyze the hydrolysis of cyclized nucleotides cGMP and cAMP, while cAMP activity is regulated by cGMP, which plays a key role in the balance of cGMP and cAMP functions in cells.
  • PDE2 is widely expressed in human tissues and is mainly distributed in the heart, central nervous system, liver, adrenal gland, endothelial cells, and platelets. PDE2 is involved in the regulation of various physiological activities, such as central learning, memory and cognition, maintaining the basic rhythm of the heart, smooth muscle and endothelial cells, permeability of endothelial cells, and regulating inflammation.
  • PDE2 knockout mice directly cause embryonic death. It can be used in a variety of central, cardiovascular, and inflammatory responses by inhibiting PDE2 activity.
  • Non-selective PDE inhibitory activities of various natural and synthetic terpenoids have long been discovered, such as caffeine, theophylline, pentoxifylline and the like.
  • Pentoxifylline PDE2 activity
  • PDE2 activity is clinically approved for lower limb paralysis caused by peripheral vascular occlusion. The main role is to reduce blood viscosity, increase red blood cell deformation, and inhibit platelet aggregation.
  • Novel highly selective PDE2 inhibitors have also been reported to control endothelial cell division and angiogenesis, and to improve central cognitive impairment.
  • the development and application of the new novel selective PDE2 inhibitors are still very limited, and the discovery and application of new PDE2 inhibitors have broad prospects.
  • TNF- ⁇ Tumor necrosis factor alpha
  • IL-1 interleukon-1
  • IL-6 proinflammatory cytokines
  • IMID immune-mediated inflammatory diseases
  • RA rheumatoid arthritis
  • psoriatic arthritis psoriatic arthritis
  • JCA juvenile chronic arthritis
  • TNF- ⁇ inhibitors TNF- ⁇ inhibitors
  • PDE2 can regulate the expression of TNF- ⁇ in a mechanism, so that the level of TNF- ⁇ can be controlled by regulating PDE2 activity, thereby controlling inflammatory response.
  • the present invention provides a compound of the formula (I), a tautomer thereof or a pharmaceutically acceptable salt thereof,
  • Ring B is a 4- to 6-membered cyclic ether or alkoxy-cycloalkyl group optionally substituted by 1 to 3 R;
  • L is a C 1-3 alkyl group optionally substituted by 1 to 2 R;
  • Ring A is selected from a 5- to 6-membered aryl or heteroaryl group optionally substituted with 1 or 2 R 1 ;
  • R 1 is selected from the group consisting of halogen, OH, NH 2 , optionally substituted by 1 to 3 R 2 : C 1-6 alkyl or heteroalkyl, 3 to 6-membered cycloalkyl or heterocycloalkyl, 3 - a 6-membered cycloalkyl or heterocycloalkyl substituted C 1-6 alkyl or heteroalkyl;
  • R 2 is selected from the group consisting of halogen, OH, NH 2 , Me, CF 3 , OMe, OCF 3 ;
  • the hetero atom represents a hetero atom selected from O, S, N, and the number of heteroatoms on each heteroalkyl or heterocycloalkyl group is independently selected from 1, 2 or 3;
  • R is selected from halogen, N(R')(R'), C 1-3 alkyl or heteroalkyl optionally substituted by 1 to 3 R';
  • R' is selected from the group consisting of H, halogen, NH 2 , Me, CF 3 , OMe, OCF 3 .
  • R 1 is selected from the group consisting of halogen, OH, NH 2 , optionally substituted by 1 to 3 R 2 : C 1-4 alkyl or heteroalkyl, 3 to 5 membered cycloalkyl Or a heterocycloalkyl substituted C 1-3 alkyl or heteroalkyl group.
  • R 1 is selected from the group consisting of Me, CF 3 , Et, CH 2 CF 3 ,
  • R is selected from the group consisting of F, Cl, Br, I, Me,
  • the ring B is selected from the group consisting of optionally substituted by 1 to 3 R:
  • the above B is selected from the group consisting of optionally substituted by 1 to 3 R:
  • the above B is selected from the group consisting of:
  • the L is selected from the group consisting of: 1 to 2 R substituted: methylene,
  • the above L is selected from the group consisting of: methylene,
  • the ring A is selected from the group consisting of optionally substituted with 1 or 2 R 1 : imidazolyl, pyrazolyl, pyridyl, pyrazinyl, pyridazinyl, pyrimidinyl, phenyl.
  • the above ring A is optionally substituted by 1 or 2 R 1
  • the ring A is selected from the group consisting of:
  • the structural unit From:
  • the above compound is selected from the group consisting of
  • the invention also provides the use of the above compounds, their tautomers or their chemically acceptable salts in the preparation of PDE2 inhibitors and TNFa inhibitors.
  • C 1-12 is selected from C 1 , C 2 , C 3 , C 4 , C 5 , C 6 , C 7 , C 8 , C 9 , C 10 , C 11 and C 12 ;
  • C 3-12 is selected from C 3 , C 4 , C 5 , C 6 , C 7 , C 8 , C 9 , C 10 , C 11 and C 12 .
  • pharmaceutically acceptable as used herein is intended to mean that those compounds, materials, compositions and/or dosage forms are within the scope of sound medical judgment and are suitable for use in contact with human and animal tissues. Without excessive toxicity, irritation, allergic reactions or other problems or complications, commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically acceptable salt refers to a salt of a compound of the invention prepared from a compound having a particular substituent found in the present invention and a relatively non-toxic acid or base.
  • a base addition salt can be obtained by contacting a neutral amount of such a compound with a sufficient amount of a base in a neat solution or a suitable inert solvent.
  • Pharmaceutically acceptable base addition salts include sodium, potassium, calcium, ammonium, organic ammonia or magnesium salts or similar salts.
  • an acid addition salt can be obtained by contacting a neutral form of such a compound with a sufficient amount of an acid in a neat solution or a suitable inert solvent.
  • pharmaceutically acceptable acid addition salts include inorganic acid salts including, for example, hydrochloric acid, hydrobromic acid, nitric acid, carbonic acid, hydrogencarbonate, phosphoric acid, monohydrogen phosphate, dihydrogen phosphate, sulfuric acid, Hydrogen sulfate, hydroiodic acid, phosphorous acid, etc.; and an organic acid salt, such as acetic acid, propionic acid, isobutyric acid, maleic acid, malonic acid, benzoic acid, succinic acid, suberic acid, Similar acids such as fumaric acid, lactic acid, mandelic acid, phthalic acid, benzenesulfonic acid, p-toluenesulfonic acid, citric acid, tartaric acid, and me
  • the salt is contacted with a base or acid in a conventional manner, and the parent compound is separated, thereby regenerating the neutral form of the compound.
  • the parent form of the compound differs from the form of its various salts by certain physical properties, such as differences in solubility in polar solvents.
  • a "pharmaceutically acceptable salt” is a derivative of a compound of the invention wherein the parent compound is modified by salt formation with an acid or with a base.
  • pharmaceutically acceptable salts include, but are not limited to, inorganic or organic acid salts of bases such as amines, alkali metal or organic salts of acid groups such as carboxylic acids, and the like.
  • Pharmaceutically acceptable salts include the conventional non-toxic salts or quaternary ammonium salts of the parent compound, for example salts formed from non-toxic inorganic or organic acids.
  • non-toxic salts include, but are not limited to, those derived from inorganic acids and organic acids selected from the group consisting of 2-acetoxybenzoic acid, 2-hydroxyethanesulfonic acid, acetic acid, ascorbic acid, Benzenesulfonic acid, benzoic acid, hydrogencarbonate, carbonic acid, citric acid, edetic acid, ethane disulfonic acid, ethanesulfonic acid, fumaric acid, glucoheptose, gluconic acid, glutamic acid, glycolic acid, Hydrobromic acid, hydrochloric acid, hydroiodide, hydroxyl, hydroxynaphthalene, isethionethane, lactic acid, lactose, dodecylsulfonic acid, maleic acid, malic acid, mandelic acid, methanesulfonic acid, nitric acid, oxalic acid, Pamoic acid, pantothenic acid, phenylacetic acid, phen
  • the pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound containing an acid group or a base by conventional chemical methods.
  • such salts are prepared by reacting these compounds in water or an organic solvent or a mixture of the two via a free acid or base form with a stoichiometric amount of a suitable base or acid.
  • a nonaqueous medium such as ether, ethyl acetate, ethanol, isopropanol or acetonitrile is preferred.
  • the compounds provided herein also exist in the form of prodrugs.
  • Prodrugs of the compounds described herein are readily chemically altered under physiological conditions to convert to the compounds of the invention.
  • prodrugs can be converted to the compounds of the invention by chemical or biochemical methods in an in vivo setting.
  • Certain compounds of the invention may exist in unsolvated or solvated forms, including hydrated forms.
  • the solvated forms are equivalent to the unsolvated forms and are included within the scope of the invention.
  • Certain compounds of the invention may have asymmetric carbon atoms (optical centers) or double bonds. Racemates, diastereomers, geometric isomers and individual isomers are included within the scope of the invention.
  • the compounds of the invention may exist in specific geometric or stereoisomeric forms.
  • the present invention contemplates all such compounds, including the cis and trans isomers, the (-)- and (+)-p-enantiomers, the (R)- and (S)-enantiomers, and the diastereomeric a conformation, a (D)-isomer, a (L)-isomer, and a racemic mixture thereof, and other mixtures, such as enantiomerically or diastereomeric enriched mixtures, all of which belong to It is within the scope of the invention.
  • Additional asymmetric carbon atoms may be present in the substituents such as alkyl groups. All such isomers, as well as mixtures thereof, are included within the scope of the invention.
  • optically active (R)- and (S)-isomers as well as the D and L isomers can be prepared by chiral synthesis or chiral reagents or other conventional techniques. If an enantiomer of a compound of the invention is desired, it can be prepared by asymmetric synthesis or by derivatization with a chiral auxiliary wherein the resulting mixture of diastereomers is separated and the auxiliary group cleaved to provide pure The desired enantiomer.
  • a diastereomeric salt is formed with a suitable optically active acid or base, and then resolved by art.
  • a suitable optically active acid or base e.g., an amino group
  • an acidic functional group e.g., a carboxyl group
  • the diastereomer is resolved and the pure enantiomer is recovered.
  • the separation of enantiomers and diastereomers is generally accomplished by the use of chromatography using a chiral stationary phase, optionally in combination with chemical derivatization (eg, formation of an amino group from an amine). Formate).
  • the compounds of the present invention may contain unnatural proportions of atomic isotopes on one or more of the atoms that make up the compound.
  • radiolabeled compounds can be used, such as tritium (3 H), iodine -125 (125 I) or C-14 (14 C). Alterations of all isotopic compositions of the compounds of the invention, whether radioactive or not, are included within the scope of the invention.
  • pharmaceutically acceptable carrier refers to any formulation or carrier medium that is capable of delivering an effective amount of an active substance of the present invention, does not interfere with the biological activity of the active substance, and has no toxic side effects to the host or patient, including water, oil, Vegetables and minerals, cream bases, lotion bases, ointment bases, etc. These bases include suspending agents, tackifiers, transdermal enhancers and the like. Their formulations are well known to those skilled in the cosmetic or topical pharmaceutical arts. For additional information on vectors, reference is made to Remington: The Science and Practice of Pharmacy, 21st Ed., Lippincott, Williams & Wilkins (2005), the contents of which are hereby incorporated by reference.
  • excipient generally refers to the carrier, diluent and/or vehicle required to formulate an effective pharmaceutical composition.
  • an "effective amount” or “therapeutically effective amount” with respect to a pharmaceutical or pharmacologically active agent refers to a sufficient amount of a drug or agent that is non-toxic but that achieves the desired effect.
  • an "effective amount” of an active substance in a composition refers to the amount required to achieve the desired effect when used in combination with another active substance in the composition.
  • the determination of the effective amount varies from person to person, depending on the age of the recipient and The general case also depends on the particular active substance, and a suitable effective amount in a case can be determined by a person skilled in the art according to routine experimentation.
  • active ingredient refers to a chemical entity that is effective in treating a target disorder, disease or condition.
  • substituted means that any one or more hydrogen atoms on a particular atom are replaced by a substituent, including variants of heavy hydrogen and hydrogen, as long as the valence of the particular atom is normal and the substituted compound is stable.
  • it means that two hydrogen atoms are substituted.
  • Ketone substitution does not occur on the aryl group.
  • optionally substituted means that it may or may not be substituted, and unless otherwise specified, the kind and number of substituents may be arbitrary on the basis of chemically achievable.
  • any variable eg, R
  • its definition in each case is independent.
  • the group may optionally be substituted with at most two R, and each case has an independent option.
  • combinations of substituents and/or variants thereof are permissible only if such combinations result in stable compounds.
  • one of the variables When one of the variables is selected from a single bond, it means that the two groups to which it is attached are directly linked. For example, when L represents a single bond in A-L-Z, the structure is actually A-Z.
  • substituents When a bond of a substituent can be cross-linked to two atoms on a ring, the substituent can be bonded to any atom on the ring.
  • substituents do not indicate which atom is attached to a compound included in the chemical structural formula including but not specifically mentioned, such a substituent may be bonded through any atomic phase thereof.
  • Combinations of substituents and/or variants thereof are permissible only if such combinations result in stable compounds. For example, a structural unit It is indicated that it can be substituted at any position on the cyclohexyl or cyclodiene.
  • R', R", R"', R"" and R""' are each independently preferred Hydrogen, substituted or unsubstituted heteroalkyl, substituted or unsubstituted aryl (eg substituted by 1 to 3 halogen aryl), substituted or unsubstituted alkyl, alkoxy, sulphur Alkoxy group or aralkyl group.
  • each R group is independently selected as if present Each of these groups of more than one R', R", R"', R"" and R""' groups.
  • R' and R" When R' and R" are attached to the same nitrogen atom, they may be bonded to the nitrogen The atoms combine to form a 5-, 6- or 7-membered ring.
  • -NR'R is intended to include, but is not limited to, 1-pyrrolidinyl and 4-morpholinyl.
  • alkyl is intended to include carbon.
  • a group bonded to a non-hydrogen group such as a haloalkyl group (e.g., -CF 3 , -CH 2 CF 3 ) and an acyl group (e.g., -C(O)CH 3 , -C(O)CF 3 ,- C(O)CH 2 OCH 3 , etc.).
  • a non-hydrogen group such as a haloalkyl group (e.g., -CF 3 , -CH 2 CF 3 ) and an acyl group (e.g., -C(O)CH 3 , -C(O)CF 3 ,- C(O)CH 2 OCH 3 , etc.).
  • Two substituents on adjacent atoms of the aryl or heteroaryl ring may be optionally substituted with a substituent of the formula -TC(O)-(CRR')qU-, wherein T and U are independently selected From -NR-, -O-, CRR'- or a single bond, q is an integer from 0 to 3.
  • two substituents on adjacent atoms of the aryl or heteroaryl ring may be optionally substituted with a substituent of the formula -A(CH2)r B-, wherein A and B are independently selected From -CRR'-, -O-, -NR-, -S-, -S(O)-, S(O) 2 -, -S(O) 2 NR'- or a single bond, r is 1 to 4 The integer.
  • a single bond on the new ring thus formed can be replaced with a double bond.
  • two substituents on adjacent atoms of the aryl or heteroaryl ring may be optionally substituted with a substituent of the formula -A(CH2)r B-, wherein s and d are each independently An integer selected from 0 to 3, X is -O-, -NR', -S-, -S(O)-, -S(O) 2 - or -S(O) 2 NR'-.
  • the substituents R, R', R" and R"' are each independently preferably selected from hydrogen and substituted or unsubstituted (C 1 -C 6 )alkyl.
  • halo or halogen
  • haloalkyl is intended to include both monohaloalkyl and polyhaloalkyl.
  • halo(C 1 -C 4 )alkyl is intended to include, but is not limited to, trifluoromethyl, 2,2,2-trifluoroethyl, 4-chlorobutyl, 3-bromopropyl, and the like. Wait.
  • haloalkyl groups include, but are not limited to, trifluoromethyl, trichloromethyl, pentafluoroethyl, and pentachloroethyl.
  • Alkoxy represents the above alkyl group having a specified number of carbon atoms attached through an oxygen bridge.
  • the C 1-6 alkoxy group includes a C 1 , C 2 , C 3 , C 4 , C 5 and C 6 alkoxy groups.
  • alkoxy groups include, but are not limited to, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, sec-butoxy, tert-butoxy, n-pentyloxy and S- Pentyloxy.
  • Cycloalkyl includes saturated cyclic groups such as cyclopropyl, cyclobutyl or cyclopentyl.
  • the 3-7 cycloalkyl group includes C 3 , C 4 , C 5 , C 6 and C 7 cycloalkyl groups.
  • Alkenyl includes hydrocarbon chains in a straight or branched configuration wherein one or more carbon-carbon double bonds, such as vinyl and propylene groups, are present at any stable site on the chain.
  • halo or halogen refers to fluoro, chloro, bromo and iodo.
  • hetero denotes a hetero atom or a hetero atomic group (ie, a radical containing a hetero atom), including atoms other than carbon (C) and hydrogen (H), and radicals containing such heteroatoms, including, for example, oxygen (O).
  • N nitrogen
  • S sulfur
  • Si silicon
  • Ge germanium
  • Al aluminum
  • ring means substituted or unsubstituted cycloalkyl, heterocycloalkyl, cycloalkenyl, heterocycloalkenyl, cycloalkynyl, heterocycloalkynyl, aryl or heteroaryl. So-called rings include single rings, interlocking rings, spiral rings, parallel rings or bridge rings. The number of atoms on the ring is usually defined as the number of elements of the ring. For example, "5 to 7-membered ring” means 5 to 7 atoms arranged in a circle. Unless otherwise specified, the ring optionally contains from 1 to 3 heteroatoms.
  • 5- to 7-membered ring includes, for example, phenyl, pyridine, and piperidinyl; on the other hand, the term “5- to 7-membered heterocycloalkyl ring” includes pyridyl and piperidinyl, but does not include phenyl.
  • ring also includes ring systems containing at least one ring, each of which "ring” independently conforms to the above definition.
  • heterocycle or “heterocyclyl” means a stable monocyclic, bicyclic or tricyclic ring containing a hetero atom or a heteroatom group which may be saturated, partially unsaturated or unsaturated ( Aromatic) which comprise a carbon atom and 1, 2, 3 or 4 ring heteroatoms independently selected from N, O and S, wherein any of the above heterocycles may be fused to a phenyl ring to form a bicyclic ring.
  • Nitrogen and sulfur heteroatoms can be optionally oxidized (ie NO and S(O)p).
  • the nitrogen atom can be substituted or unsubstituted (i.e., N or NR, wherein R is H or other substituents as already defined herein).
  • the heterocyclic ring can be attached to the side groups of any hetero atom or carbon atom to form a stable structure. If the resulting compound is stable, the heterocycles described herein can undergo substitutions at the carbon or nitrogen sites.
  • the nitrogen atom in the heterocycle is optionally quaternized.
  • a preferred embodiment is that when the total number of S and O atoms in the heterocycle exceeds 1, these heteroatoms are not adjacent to each other. Another preferred embodiment is that the total number of S and O atoms in the heterocycle does not exceed one.
  • aromatic heterocyclic group or "heteroaryl” as used herein means a stable 5, 6, or 7 membered monocyclic or bicyclic or aromatic ring of a 7, 8, 9 or 10 membered bicyclic heterocyclic group, It contains carbon atoms and 1, 2, 3 or 4 ring heteroatoms independently selected from N, O and S.
  • the nitrogen atom can be substituted or unsubstituted (i.e., N or NR, wherein R is H or other substituents as already defined herein).
  • the nitrogen and sulfur heteroatoms can be optionally oxidized (i.e., NO and S(O)p). It is worth noting that the total number of S and O atoms on the aromatic heterocycle does not exceed one.
  • Bridged rings are also included in the definition of heterocycles.
  • a bridged ring is formed when one or more atoms (ie, C, O, N, or S) join two non-adjacent carbon or nitrogen atoms.
  • Preferred bridged rings include, but are not limited to, one carbon atom, two carbon atoms, one nitrogen atom, two nitrogen atoms, and one carbon-nitrogen group. It is worth noting that a bridge always converts a single ring into a three ring. In the bridged ring, a substituent on the ring can also be present on the bridge.
  • heterocyclic compounds include, but are not limited to, acridinyl, octanoyl, benzimidazolyl, benzofuranyl, benzofuranylfuranyl, benzindenylphenyl, benzoxazolyl, benzimidin Oxazolinyl, benzothiazolyl, benzotriazolyl, benzotetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazolyl, oxazolyl, 4aH-carbazolyl, Porphyrin, chroman, chromene, porphyrin-decahydroquinolinyl, 2H, 6H-1,5,2-dithiazinyl, dihydrofuro[2,3-b] Tetrahydrofuranyl, furyl, furfuryl, imidazolidinyl, imidazolinyl, imidazolyl, 1H-carbazolyl, nonenyl,
  • hydrocarbyl or its subordinate concept (such as alkyl, alkenyl, alkynyl, phenyl, etc.) by itself or as part of another substituent means straight-chain, branched or cyclic
  • the hydrocarbon radical or a combination thereof may be fully saturated, mono- or polyunsaturated, may be monosubstituted, disubstituted or polysubstituted, and may be monovalent (such as methyl), divalent (such as methylene) or polyvalent (methine), may include a divalent or polyvalent radical having the specified number of carbon atoms (e.g., C 1 -C 10 represents 1 to 10 carbons).
  • Hydrocarbyl includes, but is not limited to, aliphatic hydrocarbyl groups including chain and cyclic, including but not limited to alkyl, alkenyl, alkynyl groups including, but not limited to, 6-12 members.
  • An aromatic hydrocarbon group such as benzene, naphthalene or the like.
  • hydrocarbyl means a straight or branched chain radical or a combination thereof, which may be fully saturated, unitary or polyunsaturated, and may include divalent and multivalent radicals.
  • saturated hydrocarbon radicals include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, isobutyl, sec-butyl, isobutyl, cyclohexyl, (cyclohexyl).
  • a homolog or isomer of a methyl group, a cyclopropylmethyl group, and an atomic group such as n-pentyl, n-hexyl, n-heptyl, n-octyl.
  • the unsaturated alkyl group has one or more double or triple bonds, and examples thereof include, but are not limited to, a vinyl group, a 2-propenyl group, a butenyl group, a crotyl group, a 2-isopentenyl group, and a 2-(butadienyl group). ), 2,4-pentadienyl, 3-(1,4-pentadienyl), ethynyl, 1- and 3-propynyl, 3-butynyl, and higher homologs and Structure.
  • heterohydrocarbyl or its subordinate concept (such as heteroalkyl, heteroalkenyl, heteroalkynyl, heteroaryl, etc.), by itself or in combination with another term, means a stable straight chain, branched chain. Or a cyclic hydrocarbon radical or a combination thereof having a number of carbon atoms and at least one heteroatom.
  • heteroalkyl by itself or in conjunction with another term refers to a stable straight chain, branched hydrocarbon radical or combination thereof, having a number of carbon atoms and at least one heteroatom.
  • the heteroatoms are selected from the group consisting of B, O, N, and S, wherein the nitrogen and sulfur atoms are optionally oxidized, and the nitrogen heteroatoms are optionally quaternized.
  • the heteroatom or heteroatom group can be located at any internal position of the heterohydrocarbyl group (including where the hydrocarbyl group is attached to the rest of the molecule).
  • Up to two heteroatoms may be consecutive, for example, -CH 2 -NH-OCH 3.
  • alkoxy alkylamino and “alkylthio” (or thioalkoxy) are customary expressions and refer to those alkane which are attached to the remainder of the molecule through an oxygen atom, an amino group or a sulfur atom, respectively.
  • Base group alkoxy
  • cycloalkyl refers to any heterocyclic alkynyl group, etc., by itself or in combination with other terms, denotes a cyclized “hydrocarbyl group” or “heterohydrocarbyl group”, respectively.
  • a hetero atom may occupy a position at which the hetero ring is attached to the rest of the molecule.
  • cycloalkyl groups include, but are not limited to, cyclopentyl, cyclohexyl, 1-cyclohexenyl, 3-cyclohexenyl, cycloheptyl, and the like.
  • heterocyclic groups include 1-(1,2,5,6-tetrahydropyridyl), 1-piperidinyl, 2-piperidinyl, 3-piperidinyl, 4-morpholinyl, 3-morpholinyl, tetrahydrofuran-2-yl, tetrahydrofuran-3-yl, tetrahydrothiophen-2-yl, tetrahydrothiophen-3-yl, 1-piperazinyl and 2-piperazinyl.
  • aryl denotes a polyunsaturated, aromatic hydrocarbon substituent which may be monosubstituted, disubstituted or polysubstituted, may be monovalent, divalent or polyvalent, it may be monocyclic or Polycyclic (such as 1 to 3 rings; at least one of which is aromatic), which are fused together or covalently linked.
  • heteroaryl refers to an aryl (or ring) containing one to four heteroatoms. In an illustrative example, the heteroatoms are selected from the group consisting of B, N, O, and S, wherein the nitrogen and sulfur atoms are optionally oxidized, and the nitrogen atom is optionally quaternized.
  • a heteroaryl group can be attached to the remainder of the molecule through a heteroatom.
  • aryl or heteroaryl groups include phenyl, 1-naphthyl, 2-naphthyl, 4-biphenyl, 1-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 3-pyridyl Azyl, 2-imidazolyl, 4-imidazolyl, pyrazinyl, 2-oxazolyl, 4-oxazolyl, 2-phenyl-4-oxazolyl, 5-oxazolyl, 3-isoxan Azyl, 4-isoxazolyl, 5-isoxazolyl, 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, 2-furyl, 3-furyl, 2-thienyl, 3-thiophene , 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidinyl, 4-pyrimidinyl, 5-benzothiazolyl, 5-
  • aryl groups when used in conjunction with other terms (eg, aryloxy, arylthio, aralkyl), include aryl and heteroaryl rings as defined above.
  • aralkyl is intended to include those radicals to which an aryl group is attached to an alkyl group (eg, benzyl, phenethyl, pyridylmethyl, and the like), including wherein the carbon atom (eg, methylene) has been, for example, oxygen.
  • alkyl groups substituted by an atom such as phenoxymethyl, 2-pyridyloxymethyl 3-(1-naphthyloxy)propyl and the like.
  • leaving group refers to a functional group or atom which may be substituted by another functional group or atom by a substitution reaction (for example, an affinity substitution reaction).
  • substituent groups include triflate; chlorine, bromine, iodine; sulfonate groups such as mesylate, tosylate, p-bromobenzenesulfonate, p-toluenesulfonic acid Esters and the like; acyloxy groups such as acetoxy, trifluoroacetoxy and the like.
  • protecting group includes, but is not limited to, "amino protecting group", “hydroxy protecting group” or “thiol protecting group”.
  • amino protecting group refers to a protecting group suitable for preventing side reactions at the amino nitrogen position.
  • Representative amino protecting groups include, but are not limited to, formyl; acyl, such as alkanoyl (e.g., acetyl, trichloroacetyl or trifluoroacetyl); alkoxycarbonyl, e.g., tert-butoxycarbonyl (Boc) Arylmethoxycarbonyl, such as benzyloxycarbonyl (Cbz) and 9-fluorenylmethoxycarbonyl (Fmoc); arylmethyl, such as benzyl (Bn), trityl (Tr), 1, 1-di -(4'-methoxyphenyl)methyl; silyl groups such as trimethylsilyl (TMS) and tert-
  • hydroxy protecting group refers to a protecting group suitable for use in preventing hydroxy side reactions.
  • Representative hydroxy protecting groups include, but are not limited to, alkyl groups such as methyl, ethyl and t-butyl groups; acyl groups such as alkanoyl groups (e.g., acetyl); arylmethyl groups such as benzyl (Bn), Oxybenzyl (PMB), 9-fluorenylmethyl (Fm) and diphenylmethyl (diphenylmethyl, DPM); silyl groups such as trimethylsilyl (TMS) and tert-butyl Dimethylsilyl (TBS) and the like.
  • alkyl groups such as methyl, ethyl and t-butyl groups
  • acyl groups such as alkanoyl groups (e.g., acetyl)
  • arylmethyl groups such as benzyl (Bn), Oxybenzyl (PMB), 9-fluoreny
  • the compounds of the present invention can be prepared by a variety of synthetic methods well known to those skilled in the art, including the specific embodiments set forth below, combinations thereof with other chemical synthetic methods, and those well known to those skilled in the art. Equivalent alternatives, preferred embodiments include, but are not limited to, embodiments of the invention.
  • Oxetane-3-ylmethyl methanesulfonate 200 mg, 1.20 mmol
  • 3,7-dimethyl-1H-indole-2,6(3H,7H)-dione 216 mg, 1.20
  • Methyl acetate 20.0 mg, 0.120 mmol
  • potassium carbonate 250 mg, 1.81 mmol
  • the reaction solution was warmed to 120 ° C and stirred for 3 hours.
  • Methyl 2-(2-(benzyloxy)ethyl)-3-oxopentanoate (2.50 g, 9.47 mmol) was dissolved in methanol (30 mL). (mmol), the reaction solution was stirred at 25 ° C for 1 hour. The reaction was quenched with water (10 mL)EtOAc. The organic phase was washed with a saturated sodium chloride solution (20 mL ⁇ 3), dried over anhydrous sodium sulfate, filtered and evaporated. Methyl 2-(2-(benzyloxy)ethyl)-3-hydroxypentanoate (2.01 g, mp.
  • Ethyl ethyl-2-ethoxyphosphoric acid ethyl ester (4.03 g, 17.9 mmol) was dissolved in tetrahydrofuran (20 mL), sodium hydrogen (719 mg, 17.9 mmol) was added at 0 ° C.
  • Heterocyclobutane-3-carbaldehyde (900 mg, 8.99 mmol) was reacted at room temperature for 2 hours. The reaction was quenched by the addition of saturated aqueous ammonium chloride (10 mL). It was extracted with ethyl acetate (20 mL ⁇ 3), dried over anhydrous sodium sulfate, filtered, evaporated, evaporated, evaporated.
  • Ethyl (3-methyloxetan-3-yl)prop-2-enoate (800 mg, yellow oil), yield: 52%.
  • Tetrahydropyran-4-ylmethanol 500 mg, 4.30 mmol
  • triethylamine 870 mg, 8.60 mmol
  • dichloromethane 10 mL
  • methanesulfonyl chloride 985 mg, 8.60 mmol
  • the reaction was carried out at 25 ° C for 1 hour.
  • the reaction was quenched by the addition of water (10 mL). Extracted with dichloromethane (10 mL x 3), dried over anhydrous sodium sulfate, filtered and evaporated. , Yield: 84%.
  • Methyl tetrahydro-2H-pyran-4-carboxylate (2.50 g, 17.3 mmol) was dissolved in anhydrous tetrahydrofuran (50 mL), and a solution of lithium diisopropylamide was slowly added dropwise at -78 ° C under nitrogen atmosphere. (2M n-hexane solution, 10.4 mL, 20.8 mmol), and the mixture was stirred at -78 ° C for one hour. Methyl iodide (4.92 g, 34.7 mmol) was added slowly and stirring was continued for 1 hour. The reaction was quenched by the addition of water (20 mL). The reaction mixture was extracted with EtOAc (EtOAc (EtOAc)EtOAc. 0.4) Methyl 4-methyltetrahydro-2H-pyran-4-carboxylate (1.20 g, yellow oil).
  • Methyl 4-methyltetrahydro-2H-pyran-4-carboxylate (1.20 g, 7.59 mmol) was dissolved in anhydrous tetrahydrofuran (10 mL), and lithium tetrahydroaluminum (576 mg, 15.2 mmol) was added at 0 °C. .
  • the reaction solution was warmed to 25 ° C and stirred for 1 hour. Quenched with water (20 mL), EtOAc (EtOAc m. 0.2), (4-methyltetrahydro-2H-pyran-4-yl)methanol (700 mg,yield of yellow oil).
  • Methyl tetrahydro-2H-pyran-4-carboxylate (2.50 g, 17.3 mmol) was dissolved in anhydrous tetrahydrofuran (50 mL), and a solution of lithium diisopropylamide was slowly added dropwise at -78 ° C under nitrogen atmosphere. (2M n-hexane solution, 10.4 mL, 20.8 mmol), and the mixture was stirred at -78 ° C for one hour. Iodoethane (5.41 g, 34.7 mmol) was slowly added and stirring was continued for 1 hour. The reaction was quenched by the addition of water (20 mL).
  • Methyllithium (1.6 M in diethyl ether, 20.9 mL, 33.4 mmol) was dissolved in anhydrous diethyl ether (30 mL) EtOAc. The reaction solution was reacted at 0 ° C for 0.5 hour. A solution of 2-methyl-2H-pyran-4(3H)-one (1.25 g, 11.2 mmol) in diethyl ether (5 mL) was slowly added. The reaction solution was warmed to 20 ° C and stirred for 3 hours.
  • Ethyl triphenylphosphine (3.26 g, 9.37 mmol) was dissolved in anhydrous toluene (10 mL) and ((2S,6S)-2,6-dimethyl-tetrahydro-pyran-4-one was added. (400 mg, 3.13 mmol). The reaction solution was heated to 110 ° C for 72 hours.
  • Lithium tetrahydroaluminum (92.8 mg, 2.45 mmol) was slowly added to tetrahydrofuran (7 mL) dissolved in methyl-4-methoxycyclohexanecarboxylic acid (280 mg, 1.63 mmol). The reaction mixture was stirred at room temperature for 4 hours, and the reaction solution was cooled to 0 ° C with ice water, and water (0.1 mL), 15% sodium hydroxide (0.1 mL) and water (0.3 mL) were added slowly.
  • Methyl 4-nitro-pyrazole-5-carboxylate (25.0 g, 146 mmol) was dissolved in N,N-dimethylformamide (350 mL). The reaction was stirred at 0 ° C for 1 hour, and 2,2,2-trifluoroethyl trifluoromethanesulfonate (33.9 g, 146 mmol) was added dropwise. The reaction solution was stirred at 25 ° C for 18 hours. Water (1.2 L) was added to the reaction mixture, which was extracted with ethyl acetate (300 mL).
  • Methyl 4-nitro-1-(2,2,2-trifluoroethyl)-pyrazole-5-carboxylate (7.50 g, 29.6 mmol) was dissolved in methanol (100 mL) and dry palladium carbon (palladium 10) %, water 1%, 750 mg), and the reaction solution was reacted under a hydrogen pressure of 40 psi for 3 hours at room temperature.
  • the reaction mixture was filtered, and the filtrate was evaporated toluiserjjjjjjjjjjjj .
  • reaction solution was heated to 120 ° C and stirred for 3 hours.
  • the reaction solution was cooled to room temperature, poured into water (30 mL) The combined organic layers were dried with anhydrous sodium
  • the crude product was isolated and purified by preparative high performance liquid chromatography to give 6-((3-ethyloxetan-3-yl)methyl)-4-methyl-1-(2,2,2-trifluoroethyl )-pyrazolo[4,3-d]pyrimidine-5,7-dione (10 mg), yield: 24%.
  • Tetrahydrofuran-3-ylmethyl methanesulfonate (30.0 mg, 0.166 mmol), 4-methyl-1-(2,2,2-trifluoroethyl)pyrazolo[4,3-d]pyrimidine -5,7-dione (41.3 mg, 0.166 mmol), potassium carbonate (46.0 mg, 0.333 mmol) and potassium iodide (5.5 mg, 0.033 mmol) were dissolved in N,N-dimethylformamide (5 mL). The reaction solution was heated to 120 ° C and stirred for 3 hours.
  • Tetrahydropyran-4-ylmethanol (1.00 g, 8.61 mmol) and triethylamine (1.74 g, 17.2 mmol) were dissolved in anhydrous dichloromethane (20 mL). (1.28 g, 11.2 mmol). The reaction solution was stirred at 0 ° C for 1 hour. The reaction was quenched with saturated aqueous sodium hydrogen sulfate (50 mL). The combined organic phases were washed with EtOAc EtOAc (EtOAc m. Yellow oil), Yield: 78%. MS-ESI calcd for [M+H] + 195.
  • 2-Tetrahydropyran-4-ylethyl methanesulfonate (50.0 mg, 0.240 mmol), 4-methyl-1-(2,2,2-trifluoroethyl)pyrazolo[4, 3-d]pyrimidine-5,7-dione (59.6 mg, 0.240 mmol), potassium carbonate (66.4 mg, 0.480 mmol) and potassium iodide (7.9 mg, 0.048 mmol) dissolved in N,N-dimethylformamide ( 5 mL).
  • the reaction solution was heated to 120 ° C and stirred for 3 hours.
  • Methyl 4-nitro-pyrazole-5-carboxylate (22.0 g, 129 mmol) was dissolved in N,N-dimethylformamide (350 mL). The reaction was stirred at 0 ° C for 1 hour, sodium iodide (21.2 g, 141 mmol) was added, and bromomethylcyclopropane (19.1 g, 141 mmol) was added dropwise. The reaction solution was stirred at 25 ° C for 18 hours. Water (1.2 L) was added to the reaction mixture, which was extracted with ethyl acetate (300 mL).
  • Methyl 1-(cyclopropylmethyl)-4-nitro-pyrazole-5-carboxylate (5.00 g, 22.2 mmol) was dissolved in methanol (70 mL), dry palladium carbon (palladium 10%, water 1%) , 500 mg), the reaction was carried out under a hydrogen pressure of 40 psi for 3 hours at room temperature.
  • Methyl 1-(cyclopropylmethyl)-4-(2,2,2-trifluoroacetamido)-pyrazole-5-carboxylate (6.20 g, 21.3 mmol) was dissolved in N,N-dimethyl Formamide (50 mL) was added potassium carbonate (4.41 g, 31.9 mmol). The reaction solution was heated to 80 ° C for 1 hour. Cool to room temperature and add methyl iodide (4.53 g, 31.9 mmol). The reaction solution was stirred at room temperature for 18 hours. Water (300 mL) was added to the reaction mixture, and ethyl acetate (100 mL x 3).
  • Methyl 1-(cyclopropylmethyl)-4-(2,2,2-trifluoro-N-methylacetamido)-pyrazole-5-carboxylate (6.40 g, 21.0 mmol) was dissolved in tetrahydrofuran ( 30 mL) and water (30 mL) were added lithium hydroxide monohydrate (4.40 g, 105 mmol), and the mixture was stirred at room temperature for 18 hours. Di-tert-butyl dicarbonate (9.15 g, 41.9 mmol) was added, and the reaction solution was continued at room temperature for 16 hours.
  • Tetrahydrofuran-3-ylmethyl methanesulfonate (30.0 mg, 0.166 mmol), 1-methylpyrido[2,3-d]pyrimidine-2,4-dione (29.5 mg, 0.166 mmol), carbonic acid Potassium (46.0 mg, 0.333 mmol) and potassium iodide (5.5 mg, 0.033 mmol) were dissolved in N,N-dimethylformamide (5 mL). The reaction solution was heated to 120 ° C and stirred for 3 hours.
  • 2-Tetrahydropyran-4-ylethyl methanesulfonate (50.0 mg, 0.240 mmol), 1-methylpyrido[2,3-d]pyrimidine-2,4-dione (42.5 mg, 0.240 mmol), potassium carbonate (66.4 mg, 0.480 mmol) and potassium iodide (7.9 mg, 0.048 mmol) were dissolved in N,N-dimethylformamide (5 mL). The reaction solution was heated to 120 ° C and stirred for 3 hours.
  • 3-Fluoro-isonicotinic acid (3.00 g, 21.3 mmol) was dissolved in dioxane (6 mL) and 30% aqueous methylamine (22.0 g, 213 mmol). The reaction solution was warmed to 140 ° C and stirred for 14 hours. Concentrated hydrochloric acid (12 N, 3 mL) was added, the pH was adjusted to 3, filtered, and then filtered to give 3-(methylamino)isoniconic acid (3.00 g, yellow solid).
  • N-(2-Chloro-3-pyridyl)carbamic acid tert-butyl ester (11.0 g, 48.1 mmol) was dissolved in anhydrous tetrahydrofuran (150 mL). 57.7 mmol), the reaction solution was stirred at 0 ° C for half an hour. Methyl iodide (10.2 g, 72.2 mmol) was slowly added and stirred at room temperature for 12 hours. The reaction was quenched by the addition of water (50 mL). The reaction mixture was extracted with EtOAc (EtOAc (EtOAc)EtOAc. Methyl)carbamic acid tert-butyl ester (11.0 g, colorless oil), yield: 94%.
  • 2-Chloro-N-methylpyridin-3-amine (5.50 g, 38.6 mmol) was dissolved in methanol (100 mL), and the reaction solution was added to 1,1'-bis(diphenylphosphine) at 25 °C. Ferrocene palladium chloride (2.82 g, 3.86 mmol). The reaction solution was reacted in a carbon monoxide atmosphere (50 psi) at 50 ° C for 56 hours.
  • Methyl 3-(methylamino)pyridine-2-carboxylate (6.00 g, 36.1 mmol) was dissolved in methanol (100 mL) and aqueous ammonia (1.27 g, 36.1 mmol). The reaction solution was stirred at 40 ° C for 18 hours.
  • 6-Amino-1-methylpyrimidine-2,4-dione (10.0 g, 70.1 mmol) was dissolved in water (100 mL), and hydrochloric acid (7 mL, 84.0 mmol, 12 N) was added dropwise with stirring at 0 °C.
  • Sodium nitrite (5.80 g, 84.2 mmol) was dissolved in water (50 mL) and added dropwise to a mixture. The reaction solution was stirred at 25 ° C for 2 hours, filtered and washed with cold water to give a purple solid.
  • 6-Amino-1-methylpyrimidine-2,4-dione (3.50 g, 24.8 mmol) was added to formamide (5.00 g, 111 mmol), and the reaction mixture was heated to 180 ° C, stirred for 3 hours, and lowered to room temperature. Filtration, water (10 mL) was added to the filtrate and stirred, and filtered again to give 1-methylpyrimido[4,5-d]pyrimidine-2,4-dione (1.60 g, pale yellow solid), yield 36% .
  • EXPERIMENTAL OBJECTIVE To detect the AMP/GMP concentration produced in the reaction system by detecting the AlexaFluor 633 fluorescent dye substituted on the AMP/GMP antibody by fluorescence polarization analysis, and calculate the IC 50 value of the PDE2 phosphodiesterase inhibition of the test compound.
  • buffer solution 10 mM Tris-HCl, pH 7.5, 5 mM MgCl 2 , 0.01% Brij 35, 1 mM DTT, and 1% DMSO.
  • Enzyme Expression of recombinant full-length human PDE2A protein in insect Sf9 cells using baculovirus using N-terminal GST tag
  • the freshly prepared buffer solution was placed in the reaction solution, and then added to the reaction well, and the DMSO solution of the test compound was added through an Echo 550 non-contact nano-sound sonic pipetting system, and then pre-incubated for 10 minutes at room temperature, and the substrate was added (1 ⁇ M).
  • the reaction was initiated by cGMP) and allowed to react at room temperature for one hour. Then join the detection system ( AMP 2 /GMP 2 antibody, AMP2/GMP2 AlexaFluor 633 fluorescent dye), reacted at room temperature for 90 minutes, and then detected fluorescence polarization using Ex/Em 620/688.
  • Fluorescence polarization intensity was converted to nM concentration by AMP/GMP standard curve, then relative enzyme activity inhibition relative to DMSO blank was calculated, and IC50 values and curves were calculated using the Prism software package (GraphPad Software, San Diego California, USA).
  • Test article (compound prepared in each example) PDE2 phosphodiesterase inhibitory activity
  • Example 1 Example 2 -- Example 3 + Example 4 + Example 5 + Example 6 --
  • the compounds of the invention have significant and unexpected PDE2A protease inhibitory activity.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Rheumatology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pain & Pain Management (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

本发明公开了一系列羟基嘌呤类化合物及其作为PDE2或TNFα抑制剂的应用,具体公开了式(Ⅰ)所示化合物、其互变异构体或其药学上可接受的盐。

Description

羟基嘌呤类化合物及其应用 技术领域
本发明涉及一系列羟基嘌呤类化合物及其作为PDE2或TNFα抑制剂的应用,具体涉及式(Ι)所示化合物、其互变异构体或其药学上可接受的盐。
技术背景
磷酸二酯酶(PDE)催化水解环化核苷酸cGMP和cAMP,通过控制这两个重要的二级信号因子的分子内浓度调控各种生理反应.环化核苷酸cGMP和cAMP分子内的调控异常是导致许多疾病的原因,现在已经有多个药物通过抑制PDE活性来改善和治疗疾病,如PDE5抑制剂用于肺动脉高压,PDE4抑制剂用于银屑病引起的关节炎.目前已知的磷酸二酯酶基因共有十一个大类,每一类又可以表达若干亚型,总共有超过100种PDE亚型.不同的亚型具有不同的结构,不同的组织分布,对环化核苷酸cGMP和cAMP的活性也有很大的不同,调控的生理功能也是千差万别.
PDE2磷酸二酯酶可以催化水解环化核苷酸cGMP和cAMP,同时cAMP活性受cGMP调控,对于细胞内的cGMP和cAMP功能平衡起关键作用。PDE2在人体组织中广泛表达,分布主要是心脏,中枢神经系统,肝脏,肾上腺,内皮细胞,和血小板等。PDE2参与调节各项生理活性,如中枢的学习、记忆和认知等过程,维持心脏,平滑肌和内皮细胞的基本节律,内皮细胞的通透性,调节炎症反应。PDE2基因敲初小鼠直接导致胚胎死亡。通过抑制PDE2活性可能用于各种中枢,心血管疾病,和控制炎症反应。
多种天然和合成的嘌呤类化合物的非选择性PDE抑制活性很早就被发现,如咖啡因,茶碱,己酮可可碱等。己酮可可碱(PDE2活性)临床上批准用于周边血管诸塞造成的下肢跛行,主要作用是降低血液粘度,提高红细胞变形,抑制血小板凝聚等。新型的高选择性PDE2抑制剂也有报道用于控制内皮细胞的分裂和血管再生,和改善中枢认知障碍。但总体新型的选择性PDE2抑制剂的开发和应用还非常有限,发现和应用新型PDE2抑制剂具有广阔的前景。
肿瘤坏死因子α(tumor necrosis factor alpha,TNF-α)是一种具有多种生物学活性的细胞因子,对多种疾病的发生、发展及转归具有重要影响。TNF-α主要由单核细胞和巨噬细胞系产生,参与机体的免疫调节和细胞因子网络协调。正常情况下,TNF-α对免疫防御和免疫监督起着重要作用,但在某些情况下却有不良作用。研究显示,TNF-α过量表达可诱导促炎细胞因子如白介素1(interleukon-1,IL-1)、IL-6等的表达、增加内皮细胞通透性、上调粘附分子表达、激活中性白细胞和嗜酸细胞,并且诱导骨滑膜细胞和软骨细胞分泌急性期物质和组织降解酶等促进炎症的发生。这些病理反应在许多免疫介导的炎症性疾病(Immune-mediated inflammatory diseases,IMID)的发生发展中起着非常重要的作用,如风湿性关节炎(rheumatoid arthritis,RA)、牛皮癣关节炎(psoriatic arthritis,PsA)、强直性脊椎炎(ankylosing spondylitis,AS)、炎症性肠炎(inflammatory bowel disease,IBD)、幼年型慢性关节炎(juvenile chronic arthritis,JCA)以及脉管炎(vasculitis)等。研究表明,TNF-α是以上多种IMID的理想靶标,使用TNF-α拮抗药物(TNF-αinhibitors)来中和过量的TNF-α,是有效防治因TNF-α过度表达所致慢性炎症性疾病的理想途径。PDE2从机理上可以调控TNF-α的表达,因此可以通过调节PDE2活性了控制TNF-α的水平,从而可以实现控制炎症反应。
发明内容
本发明提供式(Ⅰ)所示化合物、其互变异构体或其药学上可接受的盐,
Figure PCTCN2016081103-appb-000001
其中,
环B为任选被1~3个R取代的4~6元环醚或烷氧基-环烷基;
L为任选被1~2个R取代的C1-3烷基;
环A选自任选被1或2个R1取代的5~6元芳基或杂芳基;
R1选自卤素、OH、NH2、任选被1~3个R2取代的:C1-6烷基或杂烷基、3~6元环烷基或杂环烷基、被3~6元环烷基或杂环烷基取代的C1-6烷基或杂烷基;
R2选自卤素、OH、NH2、Me、CF3、OMe、OCF3
所述杂代表杂原子,选自O、S、N,每个杂烷基或杂环烷基上的杂原子数目分别独立地选自1、2或3;
R选自卤素、N(R’)(R’)、任选被1~3个R’取代的C1-3烷基或杂烷基;
R’选自H、卤素、NH2、Me、CF3、OMe、OCF3
本发明的一个方案中,上述R1选自卤素、OH、NH2、任选被1~3个R2取代的:C1-4烷基或杂烷基、被3~5元环烷基或杂环烷基取代的C1-3烷基或杂烷基。
本发明的一个方案中,上述R1选自:Me、CF3、Et、CH2CF3
Figure PCTCN2016081103-appb-000002
Figure PCTCN2016081103-appb-000003
本发明的一个方案中,上述,R选自:F、Cl、Br、I、Me、
Figure PCTCN2016081103-appb-000004
本发明的一个方案中,上述环B选自任选被1~3个R取代的:
Figure PCTCN2016081103-appb-000005
Figure PCTCN2016081103-appb-000006
本发明的一个方案中,上述B选自任选被1~3个R取代的:
Figure PCTCN2016081103-appb-000007
Figure PCTCN2016081103-appb-000008
本发明的一个方案中,上述B选自:
Figure PCTCN2016081103-appb-000009
Figure PCTCN2016081103-appb-000010
本发明的一个方案中,上述L选自任选被1~2个R取代的:亚甲基、
Figure PCTCN2016081103-appb-000011
本发明的一个方案中,上述L选自:亚甲基、
Figure PCTCN2016081103-appb-000012
本发明的一个方案中,上述环A选自任选被1或2个R1取代的:咪唑基、吡唑基、吡啶基、吡嗪基、哒嗪基、嘧啶基、苯基。
本发明的一个方案中,上述环A自任选被1或2个R1取代的
Figure PCTCN2016081103-appb-000013
Figure PCTCN2016081103-appb-000014
本发明的一个方案中,上述环A选自:
Figure PCTCN2016081103-appb-000015
Figure PCTCN2016081103-appb-000016
本发明的一个方案中,上述结构单元
Figure PCTCN2016081103-appb-000017
选自:
Figure PCTCN2016081103-appb-000018
Figure PCTCN2016081103-appb-000019
Figure PCTCN2016081103-appb-000020
本发明的一个方案中,上述化合物选自:
Figure PCTCN2016081103-appb-000021
Figure PCTCN2016081103-appb-000022
Figure PCTCN2016081103-appb-000023
Figure PCTCN2016081103-appb-000024
本发明还提供了上述化合物、其互变异构体或其化学上可接受的盐在制备PDE2抑制剂和TNFa抑制剂中的应用。
相关定义
除非另有说明,本文所用的下列术语和短语旨在具有下列含义。一个特定的术语或短语在没有特别定义的情况下不应该被认为是不确定的或不清楚的,而应该按照普通的含义去理解。当本文中出现商品名时,意在指代其对应的商品或其活性成分。
C1-12选自C1、C2、C3、C4、C5、C6、C7、C8、C9、C10、C11和C12;C3-12选自C3、C4、C5、C6、C7、C8、C9、C10、C11和C12
这里所采用的术语“药学上可接受的”,是针对那些化合物、材料、组合物和/或剂型而言,它们在可靠的医学判断的范围之内,适用于与人类和动物的组织接触使用,而没有过多的毒性、刺激性、过敏性反应或其它问题或并发症,与合理的利益/风险比相称。
术语“药学上可接受的盐”是指本发明化合物的盐,由本发明发现的具有特定取代基的化合物与相对无毒的酸或碱制备。当本发明的化合物中含有相对酸性的功能团时,可以通过在纯的溶液或合适的惰性溶剂中用足够量的碱与这类化合物的中性形式接触的方式获得碱加成盐。药学上可接受的碱加成盐包括钠、钾、钙、铵、有机氨或镁盐或类似的盐。当本发明的化合物中含有相对碱性的官能团时,可以通过在纯的溶液或合适的惰性溶剂中用足够量的酸与这类化合物的中性形式接触的方式获得酸加成盐。药学上可接受的酸加成盐的实例包括无机酸盐,所述无机酸包括例如盐酸、氢溴酸、硝酸、碳酸,碳酸氢根,磷酸、磷酸一氢根、磷酸二氢根、硫酸、硫酸氢根、氢碘酸、亚磷酸等;以及有机酸盐,所述有机酸包括如乙酸、丙酸、异丁酸、马来酸、丙二酸、苯甲酸、琥珀酸、辛二酸、反丁烯二酸、乳酸、扁桃酸、邻苯二甲酸、苯磺酸、对甲苯磺酸、柠檬酸、酒石酸和甲磺酸等类似的酸;还包括氨基酸(如精氨酸等)的盐,以及如葡糖醛酸等有机酸的盐(参见Berge et al.,"Pharmaceutical Salts",Journal of Pharmaceutical Science 66:1-19(1977))。本发明的某些特定的化合物含有碱性和酸性的官能团,从而可以被转换成任一碱或酸加成盐。
优选地,以常规方式使盐与碱或酸接触,再分离母体化合物,由此再生化合物的中性形式。化合物的母体形式与其各种盐的形式的不同之处在于某些物理性质,例如在极性溶剂中的溶解度不同。
本文所用的“药学上可接受的盐”属于本发明化合物的衍生物,其中,通过与酸成盐或与碱成盐的方式修饰所述母体化合物。药学上可接受的盐的实例包括但不限于:碱基比如胺的无机酸或有机酸盐、酸根比如羧酸的碱金属或有机盐等等。药学上可接受的盐包括常规的无毒性的盐或母体化合物的季铵盐,例如无毒的无机酸或有机酸所形成的盐。常规的无毒性的盐包括但不限于那些衍生自无机酸和有机酸的盐,所述的无机酸或有机酸选自2-乙酰氧基苯甲酸、2-羟基乙磺酸、乙酸、抗坏血酸、苯磺酸、苯甲酸、碳酸氢根、碳酸、柠檬酸、依地酸、乙烷二磺酸、乙烷磺酸、富马酸、葡庚糖、葡糖酸、谷氨酸、乙醇酸、氢溴酸、盐酸、氢碘酸盐、羟基、羟萘、羟乙磺酸、乳酸、乳糖、十二烷基磺酸、马来酸、苹果酸、扁桃酸、甲烷磺酸、硝酸、草酸、双羟萘酸、泛酸、苯乙酸、磷酸、多聚半乳糖醛、丙酸、水杨酸、硬脂酸、亚乙酸、琥珀酸、氨基磺酸、对氨基苯磺酸、硫酸、单宁、酒石酸和对甲苯磺酸。
本发明的药学上可接受的盐可由含有酸根或碱基的母体化合物通过常规化学方法合成。一般情况下,这样的盐的制备方法是:在水或有机溶剂或两者的混合物中,经由游离酸或碱形式的这些化合物与化学计量的适当的碱或酸反应来制备。一般地,优选醚、乙酸乙酯、乙醇、异丙醇或乙腈等非水介质。
除了盐的形式,本发明所提供的化合物还存在前药形式。本文所描述的化合物的前药容易地在生理条件下发生化学变化从而转化成本发明的化合物。此外,前体药物可以在体内环境中通过化学或生化方法被转换到本发明的化合物。
本发明的某些化合物可以以非溶剂化形式或者溶剂化形式存在,包括水合物形式。一般而言,溶剂化形式与非溶剂化的形式相当,都包含在本发明的范围之内。
本发明的某些化合物可以具有不对称碳原子(光学中心)或双键。外消旋体、非对映异构体、几何异构体和单个的异构体都包括在本发明的范围之内。
本文中消旋体、ambiscalemic and scalemic或者对映体纯的化合物的图示法来自Maehr,J.Chem.Ed.1985,62:114-120。1985年,62:114-120。除非另有说明,用楔形键和虚线键表示一个立体中心的绝对构型。当本文所述化合物含有烯属双键或其它几何不对称中心,除非另有规定,它们包括E、Z几何异构体。同样地,所有的互变异构形式均包括在本发明的范围之内。
本发明的化合物可以存在特定的几何或立体异构体形式。本发明设想所有的这类化合物,包括顺式和反式异构体、(-)-和(+)-对对映体、(R)-和(S)-对映体、非对映异构体、(D)-异构体、(L)-异构体,及其外消旋混合物和其他混合物,例如对映异构体或非对映体富集的混合物,所有这些混合物都属于本发明的范围之内。烷基等取代基中可存在另外的不对称碳原子。所有这些异构体以及它们的混合物,均包括在本发明的范围之内。
可以通过的手性合成或手性试剂或者其他常规技术制备光学活性的(R)-和(S)-异构体以及D和L异构体。如果想得到本发明某化合物的一种对映体,可以通过不对称合成或者具有手性助剂的衍生作用来制备,其中将所得非对映体混合物分离,并且辅助基团裂开以提供纯的所需对映异构体。或者,当分子中含有碱性官能团(如氨基)或酸性官能团(如羧基)时,与适当的光学活性的酸或碱形成非对映异构体的盐,然后通过本领域所公知的拆分方法进行非对映异构体拆分,然后回收得到纯的对映体。此外,对映异构体和非对映异构体的分离通常是通过使用色谱法完成的,所述色谱法采用手性固定相,并任选地与化学衍生法相结合(例如由胺生成氨基甲酸盐)。
本发明的化合物可以在一个或多个构成该化合物的原子上包含非天然比例的原子同位素。例如,可用放射性同位素标记化合物,比如氚(3H),碘-125(125I)或C-14(14C)。本发明的化合物的所有同位素组成的变换,无论放射性与否,都包括在本发明的范围之内。
术语“药学上可接受的载体”是指能够递送本发明有效量活性物质、不干扰活性物质的生物活性并且对宿主或者患者无毒副作用的任何制剂或载体介质代表性的载体包括水、油、蔬菜和矿物质、膏基、洗剂基质、软膏基质等。这些基质包括悬浮剂、增粘剂、透皮促进剂等。它们的制剂为化妆品领域或局部药物领域的技术人员所周知。关于载体的其他信息,可以参考Remington:The Science and Practice of Pharmacy,21st Ed.,Lippincott,Williams&Wilkins(2005),该文献的内容通过引用的方式并入本文。
术语“赋形剂”通常是指配制有效的药物组合物所需要载体、稀释剂和/或介质。
针对药物或药理学活性剂而言,术语“有效量”或“治疗有效量”是指无毒的但能达到预期效果的药物或药剂的足够用量。对于本发明中的口服剂型,组合物中一种活性物质的“有效量”是指与该组合物中另一种活性物质联用时为了达到预期效果所需要的用量。有效量的确定因人而异,取决于受体的年龄和一 般情况,也取决于具体的活性物质,个案中合适的有效量可以由本领域技术人员根据常规试验确定。
术语“活性成分”、“治疗剂”,“活性物质”或“活性剂”是指一种化学实体,它可以有效地治疗目标紊乱、疾病或病症。
术语“被取代的”是指特定原子上的任意一个或多个氢原子被取代基取代,包括重氢和氢的变体,只要特定原子的价态是正常的并且取代后的化合物是稳定的。当取代基为酮基(即=O)时,意味着两个氢原子被取代。酮取代不会发生在芳香基上。术语“任选被取代的”是指可以被取代,也可以不被取代,除非另有规定,取代基的种类和数目在化学上可以实现的基础上可以是任意的。
当任何变量(例如R)在化合物的组成或结构中出现一次以上时,其在每一种情况下的定义都是独立的。因此,例如,如果一个基团被0-2个R所取代,则所述基团可以任选地至多被两个R所取代,并且每种情况下的R都有独立的选项。此外,取代基和/或其变体的组合只有在这样的组合会产生稳定的化合物的情况下才是被允许的。
当其中一个变量选自单键时,表示其连接的两个基团直接相连,比如A-L-Z中L代表单键时表示该结构实际上是A-Z。
当一个取代基的键可以交叉连接到一个环上的两个原子时,这种取代基可以与这个环上的任意原子相键合。当所列举的取代基中没有指明其通过哪一个原子连接到化学结构通式中包括但未具体提及的化合物时,这种取代基可以通过其任何原子相键合。取代基和/或其变体的组合只有在这样的组合会产生稳定的化合物的情况下才是被允许的。例如,结构单元
Figure PCTCN2016081103-appb-000025
表示其可在环己基或者环基二烯上的任意一个位置发生取代。
烷基和杂烷基原子团的取代基一般被称为“烷基取代基”,它们可以选自但不限于下列基团中的一个或多个:-R’、-OR’、=O、=NR’、=N-OR’、-NR’R”、-SR’、卤素、-SiR’R”R”’、OC(O)R’、-C(O)R’、-CO2R’、-CONR’R”、-OC(O)NR’R”、-NR”C(O)R’、NR’C(O)NR”R”’、-NR”C(O)2R’、-NR””’-C(NR’R”R’”)=NR””、NR””C(NR’R”)=NR’”、-S(O)R’、-S(O)2R’、-S(O)2NR’R”、NR”SO2R’、-CN、–NO2、-N3、-CH(Ph)2和氟代(C1-C4)烷基,取代基的数目为0~(2m’+1),其中m’是这类原子团中碳原子的总数。R'、R”、R”'、R””和R””’各自独立地优选氢、被取代或未被取代的杂烷基、被取代或未被取代的芳基(例如被1~3个卤素取代芳基)、被取代或未被取代的烷基、烷氧基、硫代烷氧基基团或芳烷基。当本发明的化合物包括一个以上的R基团时,例如,每一个R基团是独立地加以选择的,如同当存在一个以上的R'、R”、R”'、R””和R””’基团时的每个这些基团。当R'和R”附着于同一个氮原子时,它们可与该氮原子结合形成5-,6-或7-元环。例如,-NR'R“意在包括但不仅限于1-吡咯烷基和4-吗啉基。根据上述关于取代基的讨论中,本领域技术人员可以理解,术语“烷基”意在包括碳原子键合于非氢基团所构成的基团,如卤代烷基(例如-CF3、-CH2CF3)和酰基(例如-C(O)CH3、-C(O)CF3、-C(O)CH2OCH3等)。
与烷基原子团所述取代基相似,芳基和杂芳基取代基一般统称为“芳基取代基”,选自例如-R’、-OR’、-NR’R”、-SR’、-卤素,-SiR’R”R”’、OC(O)R’、-C(O)R’、-CO2R’、-CONR’R”、-OC(O)NR’R”、-NR”C(O)R’、NR’C(O)NR”R”’、-NR”C(O)2R’、-NR””’-C(NR’R”R’”)=NR””、NR””C(NR’R”)=NR’”、-S(O)R’、-S(O)2R’、-S(O)2NR’R”、NR”SO2R’、-CN、–NO2、-N3、-CH(Ph)2、氟(C1-C4)烷氧基和氟(C1-C4)烷基等,取代基 的数量为0到芳香环上开放化合价的总数之间;其中R’、R”、R”’、R””和R””’独立地优选自氢、被取代或未被取代的烷基、被取代或未被取代的杂烷基、被取代或未被取代的芳基和被取代或未被取代的杂芳基。当本发明的化合物包括一个以上的R基团时,例如,每个R基团是独立地加以选择的,如同当存在一个以上R’、R”、R”’、R””和R””’基团时的每个这些基团。
芳基或杂芳基环的相邻原子上的两个取代基可以任选地被通式为–T-C(O)-(CRR’)q-U-的取代基所取代,其中T和U独立地选自-NR-、-O-、CRR'-或单键,q是0到3的整数。作为替代选择,芳基或杂芳基环的相邻原子上的两个取代基可以任选地被通式为–A(CH2)r B-的取代基所取代,其中A和B独立的选自–CRR’-、-O-、-NR-、-S-、-S(O)-、S(O)2-、-S(O)2NR’-或单键,r是1~4的整数。任选地,由此形成的新环上的一个单键可以替换为双键。作为替代选择,芳基或杂芳基环的相邻原子上的两个取代基可以任选地被通式为–A(CH2)r B-的取代基所取代,其中s和d分别独立的选自0~3的整数,X是–O-、-NR’、-S-、-S(O)-、-S(O)2-或–S(O)2NR’-。取代基R、R’、R”和R”’分别独立地优选自氢和被取代或未被取代的(C1-C6)烷基。
除非另有规定,术语“卤代素”或“卤素”本身或作为另一取代基的一部分表示氟、氯、溴或碘原子。此外,术语“卤代烷基”意在包括单卤代烷基和多卤代烷基。例如,术语“卤代(C1-C4)烷基”意在包括但不仅限于三氟甲基、2,2,2-三氟乙基、4-氯丁基和3-溴丙基等等。
卤代烷基的实例包括但不仅限于:三氟甲基、三氯甲基、五氟乙基,和五氯乙基。“烷氧基”代表通过氧桥连接的具有特定数目碳原子的上述烷基。C1-6烷氧基包括C1、C2、C3、C4、C5和C6的烷氧基。烷氧基的例子包括但不限于:甲氧基、乙氧基、正丙氧基、异丙氧基、正丁氧基、仲丁氧基、叔丁氧基、正戊氧基和S-戊氧基。“环烷基”包括饱和环基,如环丙基、环丁基或环戊基。3-7环烷基包括C3、C4、C5、C6和C7环烷基。“链烯基”包括直链或支链构型的烃链,其中该链上任何的稳定位点上存在一个或多个碳-碳双键,例如乙烯基和丙烯基。
术语“卤”或“卤素”是指氟、氯、溴和碘。
除非另有规定,术语“杂”表示杂原子或杂原子团(即含有杂原子的原子团),包括碳(C)和氢(H)以外的原子以及含有这些杂原子的原子团,例如包括氧(O)、氮(N)、硫(S)、硅(Si)、锗(Ge)、铝(Al)、硼(B)、-O-、-S-、=O、=S、-C(=O)O-、-C(=O)-、-C(=S)-、-S(=O)、-S(=O)2-,以及任选被被取代的-C(=O)N(H)-、-N(H)-、-C(=NH)-、-S(=O)2N(H)-或-S(=O)N(H)-。
除非另有规定,“环”表示被取代或未被取代的环烷基、杂环烷基、环烯基、杂环烯基、环炔基、杂环炔基、芳基或杂芳基。所谓的环包括单环、联环、螺环、并环或桥环。环上原子的数目通常被定义为环的元数,例如,“5~7元环”是指环绕排列5~7个原子。除非另有规定,该环任选地包含1~3个杂原子。因此,“5~7元环”包括例如苯基、吡啶和哌啶基;另一方面,术语“5~7元杂环烷基环”包括吡啶基和哌啶基,但不包括苯基。术语“环”还包括含有至少一个环的环系,其中的每一个“环”均独立地符合上述定义。
除非另有规定,术语“杂环”或“杂环基”意指稳定的含杂原子或杂原子团的单环、双环或三环,它们可以是饱和的、部分不饱和的或不饱和的(芳族的),它们包含碳原子和1、2、3或4个独立地选自N、O和S的环杂原子,其中上述任意杂环可以稠合到一个苯环上形成双环。氮和硫杂原子可任选被氧化(即 NO和S(O)p)。氮原子可以是被取代的或未取代的(即N或NR,其中R是H或本文已经定义过的其他取代基)。该杂环可以附着到任何杂原子或碳原子的侧基上从而形成稳定的结构。如果产生的化合物是稳定的,本文所述的杂环可以发生碳位或氮位上的取代。杂环中的氮原子任选地被季铵化。一个优选方案是,当杂环中S及O原子的总数超过1时,这些杂原子彼此不相邻。另一个优选方案是,杂环中S及O原子的总数不超过1。如本文所用,术语“芳族杂环基团”或“杂芳基”意指稳定的5、6、7元单环或双环或7、8、9或10元双环杂环基的芳香环,它包含碳原子和1、2、3或4个独立地选自N、O和S的环杂原子。氮原子可以是被取代的或未取代的(即N或NR,其中R是H或本文已经定义过的其他取代基)。氮和硫杂原子可任选被氧化(即NO和S(O)p)。值得注意的是,芳香杂环上S和O原子的总数不超过1。桥环也包含在杂环的定义中。当一个或多个原子(即C、O、N或S)连接两个不相邻的碳原子或氮原子时形成桥环。优选的桥环包括但不限于:一个碳原子、两个碳原子、一个氮原子、两个氮原子和一个碳-氮基。值得注意的是,一个桥总是将单环转换成三环。桥环中,环上的取代基也可以出现在桥上。
杂环化合物的实例包括但不限于:吖啶基、吖辛因基、苯并咪唑基、苯并呋喃基、苯并巯基呋喃基、苯并巯基苯基、苯并恶唑基、苯并恶唑啉基、苯并噻唑基、苯并三唑基、苯并四唑基、苯并异恶唑基、苯并异噻唑基、苯并咪唑啉基、咔唑基、4aH-咔唑基、咔啉基、苯并二氢吡喃基、色烯、噌啉基十氢喹啉基、2H,6H-1,5,2-二噻嗪基、二氢呋喃并[2,3-b]四氢呋喃基、呋喃基、呋咱基、咪唑烷基、咪唑啉基、咪唑基、1H-吲唑基、吲哚烯基、二氢吲哚基、中氮茚基、吲哚基、3H-吲哚基、isatino基、异苯并呋喃基、吡喃、异吲哚基、异二氢吲哚基、异吲哚基、吲哚基、异喹啉基、异噻唑基、异恶唑基、亚甲二氧基苯基、吗啉基、萘啶基,八氢异喹啉基、恶二唑基、1,2,3-恶二唑基、1,2,4-恶二唑基、1,2,5-恶二唑基、1,3,4-恶二唑基、恶唑烷基、恶唑基、异恶唑基、羟吲哚基、嘧啶基、菲啶基、菲咯啉基、吩嗪、吩噻嗪、苯并黄嘌呤基、酚恶嗪基、酞嗪基、哌嗪基、哌啶基、哌啶酮基、4-哌啶酮基、胡椒基、蝶啶基、嘌呤基、吡喃基、吡嗪基、吡唑烷基、吡唑啉基、吡唑基、哒嗪基、吡啶并恶唑、吡啶并咪唑、吡啶并噻唑、吡啶基、嘧啶基、吡咯烷基、吡咯啉基、2H-吡咯基、吡咯基、吡唑基、喹唑啉基、喹啉基、4H-喹嗪基、喹喔啉基、奎宁环基、四氢呋喃基、四氢异喹啉基、四氢喹啉基、四唑基,6H-1,2,5-噻二嗪基、1,2,3-噻二唑基、1,2,4-噻二唑基、1,2,5-噻二唑基、1,3,4-噻二唑基、噻蒽基、噻唑基、异噻唑基噻吩基、噻吩基、噻吩并恶唑基、噻吩并噻唑基、噻吩并咪唑基、噻吩基、三嗪基、1,2,3-三唑基、1,2,4-三唑基、1,2,5-三唑基、1,3,4-三唑基和呫吨基。还包括稠环和螺环化合物。
除非另有规定,术语“烃基”或者其下位概念(比如烷基、烯基、炔基、苯基等等)本身或者作为另一取代基的一部分表示直链的、支链的或环状的烃原子团或其组合,可以是完全饱和的、单元或多元不饱和的,可以是单取代、二取代或多取代的,可以是一价(如甲基)、二价(如亚甲基)或者多价(如次甲基),可以包括二价或多价原子团,具有指定数量的碳原子(如C1-C10表示1至10个碳)。“烃基”包括但不限于脂肪烃基和芳香烃基,所述脂肪烃基包括链状和环状,具体包括但不限于烷基、烯基、炔基,所述芳香烃基包括但不限于6-12元的芳香烃基,例如苯、萘等。在一些实施例中,术语“烃基”表示直链的或支链的原子团或它们的组合,可以是完全饱和的、单元或多元不饱和的,可以包括二价和多价原子团。饱和烃原子团的实例包括但不限于甲基、乙基、正丙基、异丙基、正丁基、叔丁基、异丁基、 仲丁基、异丁基、环己基、(环己基)甲基、环丙基甲基,以及正戊基、正己基、正庚基、正辛基等原子团的同系物或异构体。不饱和烷基具有一个或多个双键或三键,其实例包括但不限于乙烯基、2-丙烯基、丁烯基、巴豆基、2-异戊烯基、2-(丁二烯基)、2,4-戊二烯基、3-(1,4-戊二烯基)、乙炔基、1-和3-丙炔基,3-丁炔基,以及更高级的同系物和异构体。
除非另有规定,术语“杂烃基”或者其下位概念(比如杂烷基、杂烯基、杂炔基、杂芳基等等)本身或者与另一术语联合表示稳定的直链的、支链的或环状的烃原子团或其组合,有一定数目的碳原子和至少一个杂原子组成。在一些实施例中,术语“杂烷基”本身或者与另一术语联合表示稳定的直链的、支链的烃原子团或其组合物,有一定数目的碳原子和至少一个杂原子组成。在一个典型实施例中,杂原子选自B、O、N和S,其中氮和硫原子任选地被氧化,氮杂原子任选地被季铵化。杂原子或杂原子团可以位于杂烃基的任何内部位置(包括该烃基附着于分子其余部分的位置)。实例包括但不限于-CH2-CH2-O-CH3、-CH2-CH2-NH-CH3、-CH2-CH2-N(CH3)-CH3、-CH2-S-CH2-CH3、-CH2-CH2、-S(O)-CH3、-CH2-CH2-S(O)2-CH3、-CH=CH-O-CH3、-CH2-CH=N-OCH3和–CH=CH-N(CH3)-CH3。至多两个杂原子可以是连续的,例如-CH2-NH-OCH3
术语“烷氧基”、“烷氨基”和“烷硫基”(或硫代烷氧基)属于惯用表达,是指分别通过一个氧原子、氨基或硫原子连接到分子的其余部分的那些烷基基团。
除非另有规定,术语“环烃基”、“杂环烃基”或者其下位概念(比如芳基、杂芳基、环烷基、杂环烷基、环烯基、杂环烯基、环炔基、杂环炔基等等)本身或与其他术语联合分别表示环化的“烃基”、“杂烃基”。此外,就杂烃基或杂环烃基(比如杂烷基、杂环烷基)而言,杂原子可以占据该杂环附着于分子其余部分的位置。环烷基的实例包括但不限于环戊基、环己基、1-环己烯基、3-环己烯基、环庚基等。杂环基的非限制性实例包括1-(1,2,5,6-四氢吡啶基)、1-哌啶基、2-哌啶基,3-哌啶基、4-吗啉基、3-吗啉基、四氢呋喃-2-基、四氢呋喃吲哚-3-基、四氢噻吩-2-基、四氢噻吩-3-基,1-哌嗪基和2-哌嗪基。
除非另有规定,术语“芳基”表示多不饱和的芳族烃取代基,可以是单取代、二取代或多取代的,可以是一价、二价或者多价,它可以是单环或多环(比如1至3个环;其中至少一个环是芳族的),它们稠合在一起或共价连接。术语“杂芳基”是指含有一至四个杂原子的芳基(或环)。在一个示范性实例中,杂原子选自B、N、O和S,其中氮和硫原子任选地被氧化,氮原子任选地被季铵化。杂芳基可通过杂原子连接到分子的其余部分。芳基或杂芳基的非限制性实施例包括苯基、1-萘基、2-萘基、4-联苯基、1-吡咯基、2-吡咯基、3-吡咯基、3-吡唑基、2-咪唑基、4-咪唑基、吡嗪基、2-恶唑基、4-恶唑基、2-苯基-4-恶唑基、5-恶唑基、3-异恶唑基、4-异恶唑基、5-异恶唑基、2-噻唑基、4-噻唑基、5-噻唑基、2-呋喃基、3-呋喃基、2-噻吩基、3-噻吩基、2-吡啶基、3-吡啶基、4-吡啶基、2-嘧啶基、4-嘧啶基、5-苯并噻唑基、嘌呤基、2-苯并咪唑基、5-吲哚基、1-异喹啉基、5-异喹啉基、2-喹喔啉基、5-喹喔啉基、3-喹啉基和6-喹啉基。上述任意一个芳基和杂芳基环系的取代基选自下文所述的可接受的取代基。
为简便起见,芳基在与其他术语联合使用时(例如芳氧基、芳硫基、芳烷基)包括如上定义的芳基和杂芳基环。因此,术语“芳烷基”意在包括芳基附着于烷基的那些原子团(例如苄基、苯乙基、吡啶基甲基等),包括其中碳原子(如亚甲基)已经被例如氧原子代替的那些烷基,例如苯氧基甲基、2-吡啶氧甲基3-(1-萘氧基)丙基等。
术语“离去基团”是指可以被另一种官能团或原子通过取代反应(例如亲和取代反应)所取代的官能团或原子。例如,代表性的离去基团包括三氟甲磺酸酯;氯、溴、碘;磺酸酯基,如甲磺酸酯、甲苯磺酸酯、对溴苯磺酸酯、对甲苯磺酸酯等;酰氧基,如乙酰氧基、三氟乙酰氧基等等。
术语“保护基”包括但不限于“氨基保护基”、“羟基保护基”或“巯基保护基”。术语“氨基保护基”是指适合用于阻止氨基氮位上副反应的保护基团。代表性的氨基保护基包括但不限于:甲酰基;酰基,例如链烷酰基(如乙酰基、三氯乙酰基或三氟乙酰基);烷氧基羰基,如叔丁氧基羰基(Boc);芳基甲氧羰基,如苄氧羰基(Cbz)和9-芴甲氧羰基(Fmoc);芳基甲基,如苄基(Bn)、三苯甲基(Tr)、1,1-二-(4'-甲氧基苯基)甲基;甲硅烷基,如三甲基甲硅烷基(TMS)和叔丁基二甲基甲硅烷基(TBS)等等。术语“羟基保护基”是指适合用于阻止羟基副反应的保护基。代表性羟基保护基包括但不限于:烷基,如甲基、乙基和叔丁基;酰基,例如链烷酰基(如乙酰基);芳基甲基,如苄基(Bn),对甲氧基苄基(PMB)、9-芴基甲基(Fm)和二苯基甲基(二苯甲基,DPM);甲硅烷基,如三甲基甲硅烷基(TMS)和叔丁基二甲基甲硅烷基(TBS)等等。
本发明的化合物可以通过本领域技术人员所熟知的多种合成方法来制备,包括下面列举的具体实施方式、其与其他化学合成方法的结合所形成的实施方式以及本领域技术上人员所熟知的等同替换方式,优选的实施方式包括但不限于本发明的实施例。
本发明所使用的所有溶剂是市售的,无需进一步纯化即可使用。本发明采用下述缩略词:aq代表水;HATU代表O-(7-氮杂苯并三唑-1-基)-N,N,N',N'-四甲基脲六氟磷酸盐;EDC代表N-(3-二甲基氨基丙基)-N'-乙基碳二亚胺盐酸盐;m-CPBA代表3-氯过氧苯甲酸;eq代表当量、等量;CDI代表羰基二咪唑;DCM代表二氯甲烷;PE代表石油醚;DIAD代表偶氮二羧酸二异丙酯;DMF代表N,N-二甲基甲酰胺;DMSO代表二甲亚砜;EtOAc代表乙酸乙酯;EtOH代表乙醇;MeOH代表甲醇;CBz代表苄氧羰基,是一种胺保护基团;BOC代表叔丁基羰基是一种胺保护基团;HOAc代表乙酸;NaCNBH3代表氰基硼氢化钠;r.t.代表室温;O/N代表过夜;THF代表四氢呋喃;Boc2O代表二-叔丁基二碳酸酯;TFA代表三氟乙酸;DIPEA代表二异丙基乙基胺;SOCl2代表氯化亚砜;CS2代表二硫化碳;TsOH代表对甲苯磺酸;NFSI代表N-氟-N-(苯磺酰基)苯磺酰胺;NCS代表1-氯吡咯烷-2,5-二酮;n-Bu4NF代表氟化四丁基铵;iPrOH代表2-丙醇;mp代表熔点;LDA代表二异丙基胺基锂;TMSCF3代表三氟甲基三甲基硅烷;Ti(Oi-Pr)4代表钛酸四异丙酯;MSCl代表甲烷磺酰氯;DMAP代表N,N-二甲基-4-氨基吡啶;TEA代表三乙胺;BnBr代表苄溴;DIEA代表二异丙基乙胺;BH3DMS代表硼烷二甲硫醚;DMP代表戴斯马丁过碘烷;TBAF代表四丁基氟化胺;HOBT代表1-羟基苯并三唑;AIBN代表偶氮二异丁腈;NBS代表N-溴代丁二酰亚胺。
化合物经手工或者
Figure PCTCN2016081103-appb-000026
软件命名,市售化合物采用供应商目录名称。
具体实施方式
下面通过实施例对本发明进行详细描述,但并不意味着对本发明任何不利限制。
实施例1
3,7-二甲基-1-(氧杂环丁烷-3-基甲基)嘌呤-2,6-二酮
Figure PCTCN2016081103-appb-000027
第一步
氧杂环丁烷-3-基甲基甲磺酸酯
将氧杂环丁烷-3-基-甲醇(150mg,1.70mmol)和三乙胺(344mg,3.40mmol)溶于二氯甲烷(5mL)中,0℃下加入甲烷磺酰氯(390mg,3.40mmol)。反应液缓慢升至室温,搅拌2小时。加入饱和碳酸氢钠水溶液(10mL)淬灭反应。用二氯甲烷萃取(10mL x 3)。合并有机相,用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到氧杂环丁烷-3-基甲基甲磺酸酯(200mg,黄色油状),产率:70%。MS-ESI计算值[M+H]+167,实测值167。
第二步
3,7-二甲基-1-(氧杂环丁烷-3-基甲基)嘌呤-2,6-二酮
将氧杂环丁烷-3-基甲基甲磺酸酯(200mg,1.20mmol),3,7-二甲基-1H-嘌呤-2,6(3H,7H)-二酮(216mg,1.20mmol),碘化钾(20.0mg,0.120mmol)和碳酸钾(250mg,1.81mmol)溶于N,N-二甲基甲酰胺(10mL)中。反应液升温至120℃,搅拌3小时。冷却至室温,过滤,滤液减压浓缩用制备型高效液相色谱纯化,得到3,7-二甲基-1-(氧杂环丁烷-3-基甲基)嘌呤-2,6-二酮(100mg),产率:33%。
1H NMR:(400MHz,Methonal-d4)δ7.87(s,1H),4.74(t,J=6.8Hz,2H),4.61(t,J=6.8Hz,2H),4.30(d,J=6.8Hz,2H),3.96(s,3H),3.51(s,3H),3.42-3.35(m,1H)。MS-ESI计算值[M+H]+251,实测值251。
实施例2
1-(3-氟-氧杂环丁烷-3-基甲基)-3,7-二甲基-3,7-二氢-嘌呤-2,6-二酮
Figure PCTCN2016081103-appb-000028
Figure PCTCN2016081103-appb-000029
第一步
2-((苄氧基)甲基)丙-2-烯基-1-醇
在0℃条件下,向氢化钠(3.63g,90.8mmol)的四氢呋喃(180mL)溶液中滴加2-甲烯基丙烷-1,3-二醇(8.00g,90.8mmol)的四氢呋喃(20mL)溶液。反应液在0℃搅拌1小时。加入四丁基碘化铵(33.5g,90.8mmol)和卞溴(15.5g,90.8mmol)。反应混合物在室温反应3小时。反应液冷却至0℃,加水(200mL)淬灭。用乙酸乙酯萃取(100mL x 3)。合并有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱层析法分离纯化(3:1石油醚/乙酸乙酯),得到2-((苄氧基)甲基)丙-2-烯基-1-醇(7.00,无色液体),产率:43%。
1H NMR:(400MHz,CDCl3)δ7.38-7.30(m,5H),5.22(s,1H),5.16(s,1H),4.53(s,2H),4.20(s,2H),4.10(s,2H)。
第二步
3-(苄氧基)-2-(溴甲基)-2-氟丙烷-1-醇
在0℃条件下,向2-((苄氧基)甲基)丙-2-烯基-1-醇(2.00g,11.2mmol)的二氯甲烷(30mL)溶液加入氢氟酸三乙胺盐(4.50g,27.9mmol)和溴代丁二酰亚胺(3.00g,16.8mmol)。反应液在室温搅拌3小时。加水(30mL)淬灭反应。用二氯甲烷(30mL x 2)萃取。合并有机相用依次用饱和碳酸氢钠水溶液和饱和食盐水洗涤。用无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱层析法分离纯化(5:1石油醚/乙酸乙酯)得到3-(苄氧基)-2-(溴甲基)-2-氟丙烷-1-醇(1.40g,无色油状物),产率:45%。
1H NMR:(400MHz,CDCl3)δ7.38-7.31(m,5H),4.60(s,2H),3.89(d,J=16.0Hz,2H),3.77(d,J=16.0Hz,2H),3.70-3.65(m,2H)。
第三步
3-((苄氧基)甲基)-3-氟环氧丙烷
在0℃条件下,向3-(苄氧基)-2-(溴甲基)-2-氟丙烷-1-醇(200mg,0.721mmol)的无水四氢呋喃(2mL)溶液中加入氢化钠(75.0mg,1.88mmol)。反应液在室温条件搅拌过夜。加入冰水(20mL)淬灭反应,用乙酸乙酯(10mL x 3)萃取,合并有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱层析法分离纯化(3:1石油醚/乙酸乙酯)得到3-((苄氧基)甲基)-3-氟环氧丙烷(42.0mg, 无色液体),产率:30%。
1H NMR:(400MHz,CDCl3)δ7.40-7.32(m,5H),4.79(dd,J=20.0,8.0Hz,2H),4.64(s,2H),4.60(dd,J=20.0,8.0Hz,2H),3.82(d,J=20.0Hz,2H).
第四步
(3-氟-氧杂环丁烷-3-基)甲醇
将3-苄氧基甲基-3-氟-氧杂环丁烷(190mg,0.968mmol)溶于四氢呋喃(10mL),加入醋酸(0.25mL),干钯碳(钯10%,水1%,20mg),室温下,反应液于氢气(30psi)下反应5小时。反应液过滤,得到(3-氟-氧杂环丁烷-3-基)甲醇四氢呋喃溶液,直接用于下一步反应。
第五步
甲磺酸3-氟-氧杂环丁烷-3-基甲酯
氮气保护下,向上步得到的(3-氟-氧杂环丁烷-3-基)甲醇(104mg,0.980mmol)四氢呋喃溶液中加入三乙胺(298mg,2.94mmol)和甲烷磺酰氯(225mg,1.96mmol),反应液室温反应3小时。加入二氯甲烷(30mL),依次用0.5N盐酸水溶液(15mL)和饱和碳酸氢钠溶液(15mL)洗涤反应液,用饱和食盐水有机相(10mL)洗涤有机相,无水硫酸钠干燥,减压浓缩,得到甲磺酸3-氟-氧杂环丁烷-3-基甲酯(102mg,黄色油状物),产率:57%。
1H NMR:(400MHz,CDCl3)δ4.84(dd,J=18.4,8.4Hz,2H),4.67-4.51(m,4H),3.09(s,3H).M MS-ESI计算值[M+H]+185,实测值185.
第六步
1-(3-氟-氧杂环丁烷-3-基甲基)-3,7-二甲基-3,7-二氢-嘌呤-2,6-二酮
将甲磺酸3-氟-氧杂环丁烷-3-基甲酯(100mg,0.543mmol)溶于N,N-二甲基甲酰胺(2mL),加入3,7-二甲基-1H-嘌呤-2,6(3H,7H)-二酮(97.8mg,0.543mmol),碳酸钾(150mg,1.09mmol)和碘化钾(9.0mg,0.054mmol)。反应液加热到120℃,搅拌3小时。减压浓缩,剩余物用高效液相色谱法纯化,得1-(3-氟-氧杂环丁烷-3-基甲基)-3,7-二甲基-3,7-二氢-嘌呤-2,6-二酮(28.0mg),产率:19%。
1H NMR:(400MHz,CDCl3)δ7.54(s,1H),4.92(d,J=9.2Hz,1H),4.86(d,J=9.2Hz,1H),4.80(d,J=8.8Hz,1H),4.74(d,J=8.8Hz,1H),4.54(d,J=10.0Hz,2H),3.99(s,3H),3.59(s,3H).MS-ESI计算值[M+H]+269,实测值269。
实施例3
3,7-二甲基-1-((3-甲基氧杂环丁烷-3-基)甲基)-1H-嘌呤-2,6(3H,7H)-二酮
Figure PCTCN2016081103-appb-000030
将混合物3,7-二甲基-1H-嘌呤-2,6(3H,7H)-二酮(200mg,1.10mmol),3-(氯甲基)-3-甲基氧杂环丁烷(198mg,1.65mmol),碳酸钾(304mg,2.20mmol)和碘甲烷(18.0mg,0.109mmol)溶于N,N-二甲基甲酰胺(2mL)中,反应液在微波反应器中在130℃反应15分钟。反应液减压浓缩用制备高效液相色谱法分离纯化得到3,7-二甲基-1-((3-甲基氧杂环丁烷-3-基)甲基)-1H-嘌呤-2,6(3H,7H)-二酮。
1H NMR:(400MHz,Methonal-d4)δ7.91(s,1H),4.78(d,J=6.4Hz,2H),4.23(d,J=6.4Hz,2H),4.13(s,2H),3.99(s,3H),3.54(s,3H),1.36(s,3H)。MS-ESI计算值[M+H]+265,实测值265。
实施例4
Figure PCTCN2016081103-appb-000031
第一步
(3-乙基氧杂环丁烷-3-基)甲基甲磺酸酯
将(3-乙基氧杂环丁烷-3-基)甲醇(64.0mg,0.552mmol)和三乙胺(111mg,1.10mmol)溶于二氯甲烷(20mL)中,0℃条件下加入甲烷磺酰氯(94.9mg,0.829mmol)。反应液于室温搅拌2小时后,加入二氯甲烷(20mL)稀释,用饱和碳酸氢钠溶液(20mL x 2)洗涤,有机相无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱层析法分离纯化(4:1石油醚/乙酸乙酯,Rf=0.5)得到(3-乙基氧杂环丁烷-3-基)甲基甲磺酸酯(100mg,无色油状物),产率:93%。
MS-ESI计算值[M+H]+195,实测值195。
第二步
1-((3-乙基氧杂环丁烷-3-基)甲基)3,7-二甲基-1H-嘌呤-2,6(3H,7H)-二酮
将(3-乙基氧杂环丁烷-3-基)甲基甲磺酸酯(100mg,0.520mmol)溶于N,N-二甲基甲酰胺(20mL)中,反应液于室温条件下加入3,7-二甲基-1H-嘌呤-2,6(3H,7H)-二酮(92.7mg,0.520mmol),碳酸钾(107mg,0.780mmol)和碘化钾(86.3mg,0.520mmol)。反应液加热至100℃,反应2小时,加入乙酸乙酯(20mL)稀释,有机相用饱和碳酸氢钠溶液(20mL x 2)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,用高效液相色谱纯化得1-((3-乙基氧杂环丁烷-3-基)甲基)3,7-二甲基-1H-嘌呤-2,6(3H,7H)-二酮(80.0mg),产率:56%。
1H NMR:(400MHz,CDCl3)δ7.53(s,1H),4.62(d,J=6.4Hz,2H),4.28(d,J=6.4Hz,2H),4.06(s,2H),3.98(s,3H),3.58(s,3H),1.82(q,J=7.2Hz,2H),1.06(t,J=7.2Hz,3H)。MS-ESI计算值[M+H]+279,实测值279。
实施例5
1-(2-(2-乙基氧杂环丁烷-3-基)乙基)-3,7-二甲基-1H-嘌呤-2,6(3H,7H)-二酮
Figure PCTCN2016081103-appb-000032
第一步
2-(2-(苄氧基)乙基)-3-氧代戊酸甲酯
0℃下,将氢化钠(842mg,35.1mmol)加入到3-氧代戊酸甲酯(3.04g,23.4mmol)的四氢呋喃(100mL)溶液中。反应液在0℃搅拌1个小时。将((2-溴乙氧基)-甲基)-苯(10.0g,46.7mmol)的四氢呋喃(10mL)溶液于0℃下滴加到反应液中。反应液在70℃下搅拌24小时后降温到0℃,加入饱和氯化铵溶液(20mL)淬灭反应,用乙酸乙酯萃取(20mL x 3)。有机相用饱和食盐水洗涤(100mL x3),无水硫酸钠干燥后减压浓缩。用硅胶柱色谱法分离纯化(20:1石油醚/乙酸乙酯,Rf=0.3)得到2-(2-(苄氧基)乙基)-3-氧代戊酸甲酯(3.01g,无色油状),产率:49%。
第二步
2-(2-(苄氧基)乙基)-3-羟基戊酸甲酯
将2-(2-(苄氧基)乙基)-3-氧代戊酸甲酯(2.50g,9.47mmol)溶于甲醇(30mL)中,0℃条件下加入硼氢化锂(208mg,9.47mmol),反应液在25℃下搅拌1小时。加入水(10mL)淬灭反应,用乙酸乙酯(30mL x 3)萃取。有机相用饱和氯化钠溶液(20mL x 3)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱层析法分离纯化(3:1石油醚/乙酸乙酯,Rf=0.5)得到2-(2-(苄氧基)乙基)-3-羟基戊酸甲酯(2.01g,无色油状物),产率:80%。
1H NMR:(400MHz,Methonal-d4)δ7.35-7.33(m,5H),4.47(s,2H),3.63-3.61(m,3H),3.52-3.49(m,1H),2.62-2.45(m,2H),2.15-1.85(m,2H),1.57-1.25(m,3H),1.00-0.98(m,3H)。
第三步
2-(2-(苄氧基)乙基)戊烷-1,3-二醇
在氮气保护,0℃将氢化铝锂(323mg,8.27mmol)缓慢加入2-(2-(苄氧基)乙基)-3-羟基戊酸甲酯(2.00g,7.52mmol)的四氢呋喃(100mL)溶液中。反应液在0℃搅拌1小时。反应液冷却至0℃,依次缓慢加入水(0.33mL),15%氢氧化钠溶液(0.33mL)及水(0.99mL)。过滤,滤液减压浓缩得到产物2-(2-(苄氧基)乙基)戊烷-1,3-二醇(1.50g,黄色油状),产率:76%。
1H NMR:(400MHz,Methonal-d4)δ7.36-7.28(m,5H),4.52(s,2H),3.66-3.57(m,5H),1.73–1.69(m,5H),1.00-0.95(m,3H)。
第四步
2-(2-(苄氧基)乙基)-3-羟基戊基甲磺酸酯
将2-(2-(苄氧基)乙基)戊烷-1,3-二醇(1.50g,6.30mmol)及三乙胺(1.91g,18.9mmol)溶于二氯甲烷(20mL)中,在0℃下缓慢加入甲烷磺酰氯(846mg,7.56mmol)。反应液缓慢升至25℃,搅拌0.5小时。加入水(20mL)淬灭反应,用二氯甲烷萃取(30mL x 3)。合并有机相,有机相用饱和氯化钠溶液(20mL x 3)洗涤,用无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法纯化(10:1石油醚/乙酸乙酯,Rf=0.4)得到产物2-(2-(苄氧基)乙基)-3-羟基戊基甲磺酸酯(1.50g,黄色油状物),产率:74%。
1H NMR:(400MHz,Methonal-d4)δ7.34-7.27(m,5H),4.50(s,2H),4.32-4.26(m,2H),3.60-3.50(m,3H),3.00(s,3H),1.96-1.70(m,5H),0.98-0.94(m,3H)。
第五步
3-(2-(苄氧基)乙基)-2-乙基氧杂环丁烷
0℃下,将氢化钠(228mg,9.49mmol)加入到2-(2-(苄氧基)乙基)-3-羟基戊基甲磺酸酯(1.50g,4.75mmol)的四氢呋喃(10mL)溶液中。反应液缓慢升至25℃,搅拌12个小时。加入饱和氯化铵溶液(5mL)淬灭反应,用乙酸乙酯萃取(20mL x 3)。有机相用饱和食盐水洗涤(20mL x 3),无水硫酸钠干燥后减压浓缩。用硅胶柱色谱法分离纯化(15:1石油醚/乙酸乙酯,Rf=0.5)得到3-(2-(苄氧基)乙基)-2-乙基氧杂环丁烷(800mg,无色油状),产率:77%。
1H NMR:(400MHz,Methonal-d4)δ7.37-7.28(m,5H),4.60-4.58(m,1H),4.57-4.56(m,1H),4.48(s,2H),4.44-4.25(m,1H),3.49-3.44(m,2H),2.77-2.75(m,1H),1.98-1.94(m,2H),1.74-1.63(m,2H),0.92-0.88(m,3H)。
第六步
2-(2-乙基氧杂环丁烷-3-基)乙醇
将3-(2-(苄氧基)乙基)-2-乙基氧杂环丁烷(800mg,3.64mmol)溶解在二氯甲烷(10mL)溶液中,加入三氯化铁(1.17g,7.27mmol)。反应液在25℃,氮气保护下,搅拌0.5小时。反应液过滤,滤液减压浓缩,硅胶柱色谱法分离纯化(3:1石油醚/乙酸乙酯,Rf=0.5)得到2-(2-乙基氧杂环丁烷-3-基)乙醇(60.0mg,无色油状物),产率:13%。
1H NMR:(400MHz,Methonal-d4)δ3.85-3.82(m,3H),3.77-3.75(m,2H),2.08-2.03(m,2H),1.61-1.52(m,3H),1.05-0.96(m,3H)。
第七步
2-(2-乙基氧杂环丁烷-3-基)乙基甲磺酸酯
将2-(2-乙基氧杂环丁烷-3-基)乙醇(60.0mg,0.463mmol)及三乙胺(93.0mg,0.923mmol)溶于二氯甲烷(2mL)中,在0℃下缓慢加入甲烷磺酰氯(62.2mg,0.556mmol)。反应液缓慢升至25℃,搅拌0.5小时。加入水(5mL)淬灭反应,用二氯甲烷萃取(10mL x 3)。合并有机相,有机相用饱和氯化钠溶液(10mL x 3)洗涤,用无水硫酸钠干燥,过滤,滤液减压浓缩,得到产物2-(2-乙基氧杂环丁烷-3-基)乙基甲磺酸酯(40.0mg,黄色油状物),产率:42%。
第八步
1-(2-(2-乙基氧杂环丁烷-3-基)乙基)-3,7-二甲基-1H-嘌呤-2,6(3H,7H)-二酮
将2-(2-乙基氧杂环丁烷-3-基)乙基甲磺酸酯(40.0mg,0.192mmol),3,7-二甲基-1H-嘌呤-2,6(3H,7H)-二酮(34.6mg,0.192mmol),碘化钾(3.5mg,0.019mmol)和碳酸钾(53.0mg,0.384mmol)溶于无水N,N-二甲基甲酰胺(2mL)中。反应液加热至120℃,反应3小时。反应液冷却至20℃,过滤,用制备高效液相色谱纯化,得到1-(2-(2-乙基氧杂环丁烷-3-基)乙基)-3,7-二甲基-1H-嘌呤-2,6(3H,7H)-二酮(3.0mg),产率:5%。
1H NMR:(400MHz,Methonal-d4)δ7.52(s,1H),4.17-4.14(m,1H),4.00(s,3H),3.98-3.96(m,1H),3.86-3.85(m,2H),3.59(s,3H),3.58-3.57(m,1H),2.41-2.39(m,1H),1.95-1.94(m,1H),1.85-1.81(m,1H),1.60-1.55(m,2H),1.00-0.96(m,3H)。
MS-ESI计算值[M+H]+293,实测值293。
实施例6
3,7-二甲基-1-[3-(3-甲基氧杂环丁烷-3-基)丙基]嘌呤-2,6-二酮
Figure PCTCN2016081103-appb-000033
第一步
3-(3-甲基氧杂环丁烷-3-基)丙-2-烯酸乙酯
将乙基-2-乙氧基磷酸乙酯(4.03g,17.9mmol)溶于四氢呋喃(20mL)中,0℃下加入钠氢(719mg,17.9mmol),反应0.5小时后加入3-甲基氧杂环丁烷-3-甲醛(900mg,8.99mmol),室温反应2小时。加入饱和氯化铵溶液(10mL)淬灭反应。用乙酸乙酯萃取(20mL x 3),无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法分离纯化(10:1石油醚/乙酸乙酯,Rf值=0.2)得到3-(3-甲基氧杂环丁烷-3-基)丙-2-烯酸乙酯(800mg,黄色油状),产率:52%。
1H NMR:(400MHz,Methonal-d4)δ7.27(d,J=16.0Hz,1H),5.93(d,J=16.0Hz,1H),4.67(d,J=6.0Hz,2H),4.48(d,J=6.0Hz,2H),4.20(q,J=7.2Hz,2H),1.53(s,3H),1.29(t,J=7.2Hz,3H)。MS-ESI计算值[M+H]+171,实测值171。
第二步
乙基3-(3-甲基氧杂环丁烷-3-基)丙酸乙酯
将3-(3-甲基氧杂环丁烷-3-基)丙-2-烯酸乙酯(550mg,3.23mmol),湿钯碳(100mg,3.23mmol)混合溶于四氢呋喃(30mL)中,氢气(15psi)氛围下反应12小时。反应液过滤,滤液减压浓缩得到乙基3-(3-甲基氧杂环丁烷-3-基)丙酸乙酯(500mg,黄色油状),产率:90%。
1H NMR:(400MHz,Methonal-d4)δ4.45(d,J=6.0Hz,2H),4.33(d,J=6.0Hz,2H),4.14(q,J=7.2Hz,2H),2.35(t,J=4.8Hz,2H),1.98(t,J=4.8Hz,2H),1.31(s,3H),1.25(t,J=7.2Hz,3H)。MS-ESI计算值[M+H]+173,实测值173。
第三步
3-(3-甲基氧杂环丁烷-3-基)丙-1-醇
将乙基3-(3-甲基氧杂环丁烷-3-基)丙酸乙酯(550mg,3.19mmol)溶于四氢呋喃(10mL)中,0℃下,加入四氢锂铝(242mg,6.38mmol),反应1小时。加入水(10mL)淬灭反应。用乙酸乙酯萃取(10mL x 3),无水硫酸钠干燥,过滤,滤液减压浓缩得到3-(3-甲基氧杂环丁烷-3-基)丙-1-醇(250mg,无色油状),产率:60%。
MS-ESI计算值[M+H]+131,实测值131。
第四步
3-(3-甲基氧杂环丁烷-3-基)丙基甲磺酸酯
将3-(3-甲基氧杂环丁烷-3-基)丙-1-醇(250mg,1.92mmol)和三乙胺(583mg,5.76mmol)溶于二氯甲烷(5mL)中,0℃下加入甲烷磺酰氯(659mg,5.76mmol)。反应液缓慢升至室温,搅拌2小时。加入碳酸氢钠水溶液(10mL)淬灭反应。用二氯甲烷萃取(10mL x 3)。合并有机相,用饱和食盐水洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到3-(3-甲基氧杂环丁烷-3-基)丙基甲磺酸酯(200mg,黄色油状),产率:50%。
1H NMR:(400MHz,Methonal-d4)δ4.47(d,J=6.0Hz,2H),4.37(d,J=6.0Hz,2H),4.28(t,J=6.4Hz,2H),3.09(s,3H),1.71-1.50(m,4H),1.31(s,3H)。MS-ESI计算值[M+H]+209,实测值209。
第五步
3,7-二甲基-1-[3-(3-甲基氧杂环丁烷-3-基)丙基]嘌呤-2,6-二酮
将3-(3-甲基氧杂环丁烷-3-基)丙基甲磺酸酯(200mg,0.960mmol),3,7-二甲基-1H-嘌呤-2,6(3H,7H)-二酮(173mg,0.960mmol),碘化钾(15.9mg,0.0960mmol)和碳酸钾(265mg,1.92mmol)溶于N,N-二甲基甲酰胺(10mL)中。反应液升温至120℃,搅拌3小时。冷却至室温,过滤,滤液减压浓缩用制备型高效液相色谱纯化,得到3,7-二甲基-1-[3-(3-甲基氧杂环丁烷-3-基)丙基]嘌呤-2,6-二酮(50.0mg,),产率:18%。
1H NMR:(400MHz,Methonal-d4)δ7.89(s,1H),4.46(d,J=6.0Hz,2H),4.36(d,J=6.0Hz,2H),4.03-3.95(m,5H),3.55(s,3H),1.74-1.61(m,4H),1.31(s,3H)。MS-ESI计算值[M+H]+293,实测值293。
实施例7
3,7-二甲基-1-((四氢呋喃-2-基)甲基)-1H-嘌呤-2,6(3H,7H)-二酮
Figure PCTCN2016081103-appb-000034
将混合物3,7-二甲基-1H-嘌呤-2,6(3H,7H)-二酮(538mg,2.99mmol),2-(氯甲基)四氢呋喃(300mg,2.49mmol),碳酸钾(688mg,4.98mmol)和碘甲烷(41.0mg,0.25mmol)溶于N,N-二甲基甲酰胺(3mL)中,反应液加热至130℃反应3小时。反应液减压浓缩用制备高效液相色谱法分离纯化得到3,7-二甲基-1-((四氢呋喃-2-基)甲基)-1H-嘌呤-2,6(3H,7H)-二酮(50.0mg),产率:8%。
1H NMR:(400MHz,Methonal-d4)δ7.87(s,1H),4.36-4.28(m,1H),4.23-4.16(m,1H),3.97(s,3H),3.93-3.83(m,2H),3.76-3.69(m,1H),3.53(s,3H),2.07-1.86(m,3H),1.79-1.69(m,1H)。MS-ESI计算值[M+H]+265,实测值265。
实施例8
Figure PCTCN2016081103-appb-000035
第一步
(四氢呋喃-3-基)甲基甲磺酸酯
将(四氢呋喃-3-基)甲醇(56.0mg,0.552mmol)和三乙胺(111mg,1.10mmol)溶于二氯甲烷(20mL)中,0℃条件下加入甲烷磺酰氯(94.9mg,0.829mmol)。反应液于室温搅拌2小时后,加入二氯甲烷(20mL)稀释,用饱和碳酸氢钠溶液(20mL x 2)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱层析法分离纯化(4:1石油醚/乙酸乙酯,Rf=0.5)得到(四氢呋喃-3-基)甲基甲磺酸酯(90.0mg,无色油状物),产率:90%。
MS-ESI计算值[M+H]+181,实测值181。
第二步
3,7-二甲基-1-((四氢呋喃-3-基)甲基)-1H-嘌呤-2,6(3H,7H)-二酮
将(四氢呋喃-3-基)甲基甲磺酸酯(90.0mg,0.510mmol)溶于N,N-二甲基甲酰胺(20mL)中,反应液于室温条件下加入3,7-二甲基-1H-嘌呤-2,6(3H,7H)-二酮(92.7mg,0.520mmol),碳酸钾(107mg,0.780mmol)和碘化钾(86.3mg,0.520mmol)。反应液加热至100℃,反应2小时,加入乙酸乙酯(20mL)稀释,有机相用饱和碳酸氢钠(20mL x 2)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,用高效液相色谱纯化得3,7-二甲基-1-((四氢呋喃-3-基)甲基)-1H-嘌呤-2,6(3H,7H)-二酮(80.0mg),产率:60%。
1H NMR:(400MHz,CDCl3)δ7.49(s,1H),3.95-3.93(m,1H),3.88(s,3H),3.86(m,2H),3.66-3.60(m,2H),3.46-3.45(m,1H),3.42(s,3H),2.62-2.57(m,1H),1.86-1.82(m,1H),1.63-1.58(m,1H)。MS-ESI计算值[M+H]+265,实测值265。
实施例9
3,7-二甲基-1-[2-(2-甲基四氢呋喃-3-基)乙基]嘌呤-2,6-二酮
Figure PCTCN2016081103-appb-000036
第一步
2-(2-甲基-3-亚基)乙酸乙酯
将乙基-2-乙氧基磷酸乙酯(2.24g,9.98mmol)溶于四氢呋喃(20mL)中,0℃下加入钠氢(399mg, 9.98mmol),反应0.5小时后加入2-甲基四氢呋喃-3-酮(500mg,4.99mmol),室温反应2小时。加入饱和氯化铵溶液(10mL)淬灭反应。用乙酸乙酯萃取(30mL x 3),无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法分离纯化(10:1石油醚/乙酸乙酯,Rf值=0.6)得到2-(2-甲基-3-亚基)乙酸乙酯(400mg,黄色油状),产率:47%。
1H NMR:(400MHz,Methonal-d4)δ5.76(s,1H),4.45-4.42(m,1H),4.19(q,J=7.2Hz,2H),4.12-4.09(m,1H),3.80-3.76(m,1H),3.17–3.11(m,1H),2.98-2.92(m,1H),1.34(d,J=6.0Hz,3H),1.28(t,J=7.2Hz,3H)。MS-ESI计算值[M+H]+171,实测值171。
第二步
2-(2-甲基四氢呋喃-3-基)乙酸乙酯
将2-(2-甲基-3-亚基)乙酸乙酯(1.70g,9.99mmol),湿钯碳(100mg,3.23mmol)混合溶于四氢呋喃(30mL)中,氢气(15psi)氛围下反应12小时。反应液过滤,滤液减压浓缩得到乙基2-(2-甲基四氢呋喃-3-基)乙酸乙酯(1.50g,黄色油状)。产率:87%。
1H NMR:(400MHz,Methonal-d4)δ4.17(q,J=7.2Hz,2H),3.92-3.55(m,3H),2.62-2.10(m,4H),1.72-1.64(m,1H),1.32-1.22(m,6H)。MS-ESI计算值[M+H]+173,实测值173。
第三步
2-(2-甲基四氢呋喃-3-基)乙醇
将2-(2-甲基四氢呋喃-3-基)乙酸乙酯(250mg,1.45mmol)溶于四氢呋喃(10mL)中,0℃下,加入四氢锂铝(61.0mg,1.61mmol),反应1小时。加入水(10mL)淬灭反应。用乙酸乙酯萃取(10mL x 3),无水硫酸钠干燥,过滤,滤液减压浓缩得到2-(2-甲基四氢呋喃-3-基)乙醇(180mg,无色油状),产率:95%。
1H NMR:(400MHz,Methonal-d4)δ3.96-3.51(m,5H),2.26-2.08(m,2H),1.77-1.62(m,2H),1.48-1.35(m,1H),1.31-1.13(m,3H)。MS-ESI计算值[M+H]+131,实测值131。
第四步
2-(2-甲基四氢呋喃-3-基)乙基甲磺酸酯
将2-(2-甲基四氢呋喃-3-基)乙醇(180mg,1.38mmol)和三乙胺(280mg,2.77mmol)溶于二氯甲烷(5mL)中,0℃下加入甲烷磺酰氯(310mg,2.71mmol)。反应液缓慢升至室温,搅拌2小时。加入碳酸氢钠水溶液(10mL)淬灭反应。用二氯甲烷萃取(10mL x 3)。合并有机相,用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到2-(2-甲基四氢呋喃-3-基)乙基甲磺酸酯(200mg,黄色油状),产率:69%。
MS-ESI计算值[M+H]+209,实测值209。
第五步
3,7-二甲基-1-[2-(2-甲基四氢呋喃-3-基)乙基]嘌呤-2,6-二酮
将2-(2-甲基四氢呋喃-3-基)乙基甲磺酸酯(200mg,0.960mmol),3,7-二甲基-1H-嘌呤-2,6(3H,7H)-二酮(173mg,0.960mmol),碘化钾(16.0mg,0.0960mmol)和碳酸钾(200mg,1.45mmol)溶于N,N-二甲基甲酰胺(10mL)中。反应液升温至120℃,搅拌3小时。冷却至室温,过滤,滤液 减压浓缩,用制备型高效液相色谱纯化,得到3,7-二甲基-1-[2-(2-甲基四氢呋喃-3-基)乙基]嘌呤-2,6-二酮(20.0mg),产率:7%。
1H NMR:(400MHz,Methonal-d4)δ7.86(s,1H),4.13-4.03(m,3H),3.99(s,3H),3.95-3.44(m,2H),3.52(s,3H),2.22-1.65(m,4H),1.60-1.53(m,1H),1.25-1.02(m,3H)。MS-ESI计算值[M+H]+293,实测值293。
实施例10
3,7-二甲基-1-((四氢-2H-吡喃-4-基)甲基)-1H-嘌呤-2,6-(3H,7H)-二酮
Figure PCTCN2016081103-appb-000037
第一步
(四氢-2H-吡喃-4-基)甲基甲磺酸酯
将四氢吡喃-4-基甲醇(500mg,4.30mmol)和三乙胺(870mg,8.60mmol)溶于二氯甲烷(10mL)中,0℃下加入甲烷磺酰氯(985mg,8.60mmol)。25℃反应1小时。加入水(10mL)淬灭反应。用二氯甲烷萃取(10mL x 3),无水硫酸钠干燥,过滤,滤液减压浓缩,得到(四氢-2H-吡喃-4-基)甲基甲磺酸酯(700mg,黄色油状),产率:84%。
1H NMR:(400MHz,Methonal-d4)δ4.10(d,J=6.4Hz,2H),3.99-3.96(m,2H),3.48-3.42(m,2H),3.09(s,3H),2.05-2.03(m,1H),1.72-1.67(m,2H),1.43-1.39(m,2H)。MS-ESI计算值[M+H]+195,实测值195。
第二步
3,7-二甲基-1-((四氢-2H-吡喃-4-基)甲基)-1H-嘌呤-2,6-(3H,7H)-二酮
将(四氢-2H-吡喃-4-基)甲基甲磺酸酯(700mg,3.60mmol),3,7-二甲基-1H-嘌呤-2,6-(3H,7H)-二酮(649mg,3.60mmol),碘化钾(119mg,0.720mmol)和碳酸钾(995mg,7.20mmol)溶于N,N-二甲基甲酰胺(20mL)中。反应液升温至120℃,搅拌3小时。冷却至室温,过滤,滤液减压浓缩。残余物用甲醇(10mL)洗涤得到3,7-二甲基-1-((四氢-2H-吡喃-4-基)甲基)-1H-嘌呤-2,6-(3H,7H)-二酮(200mg),产率:20%。
1H NMR:(400MHz,Methonal-d4)δ7.88(s,1H),3.99(s,3H),3.96-3.91(m,4H),3.54(s,3H),3.40-3.36(m,2H),2.10-2.07(m,1H),1.60-1.56(m,2H),1.46-1.40(m,2H)。MS-ESI计算值[M+H]+279,实测值279。
实施例11
3,7-二甲基-1-((4-甲基四氢-2H-吡喃-4-基)甲基)-1H-嘌呤-2,6-(3H,7H)-二酮
3,7-二甲基-1-(2-(3-甲基四氢呋喃-3-基)乙基)-1H-嘌呤-2,6-(3H,7H)-二酮
Figure PCTCN2016081103-appb-000038
第一步
4-甲基四氢-2H-吡喃-4-羧酸甲酯
将四氢-2H-吡喃-4-羧酸甲酯(2.50g,17.3mmol)溶于无水四氢呋喃(50mL)中,在氮气保护,-78℃时缓慢滴加二异丙基氨基锂溶液(2M正己烷溶液,10.4mL,20.8mmol),反应液在-78℃搅拌1小时。缓慢加入碘甲烷(4.92g,34.7mmol),继续搅拌1小时。加入水(20mL)淬灭反应。反应液用乙酸乙酯(50mL x 3)萃取,合并有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法纯化(10:1石油醚/乙酸乙酯,Rf=0.4)得到4-甲基四氢-2H-吡喃-4-羧酸甲酯(1.20g,黄色油状物),产率:44%。
1H NMR:(400MHz,Methonal-d4)δ3.79-3.75(m,2H),3.71(s,3H),3.48-3.42(m,2H),2.06-2.02(m,2H),1.51-1.44(m,2H),1.20(s,3H)。
第二步
(4-甲基四氢-2H-吡喃-4-基)甲醇
将4-甲基四氢-2H-吡喃-4-羧酸甲酯(1.20g,7.59mmol)溶于无水四氢呋喃(10mL)中,0℃下加入四氢铝锂(576mg,15.2mmol)。反应液升温至25℃,搅拌1小时。加水(20mL)淬灭,用乙酸乙酯(50mL x 3)萃取,无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法纯化(1:1石油醚/乙酸乙酯,Rf=0.2),得到(4-甲基四氢-2H-吡喃-4-基)甲醇(700mg,黄色油状物),产率:71%。
1H NMR:(400MHz,Methonal-d4)δ3.75-3.71(m,2H),3.68-3.66(m,2H),3.33(s,2H),1.62-1.55(m,2H),1.31-1.26(m,2H),1.03(s,3H)。
第三步
(4-甲基四氢-2H-吡喃-4-基)甲基甲磺酸酯
将(4-甲基四氢-2H-吡喃-4-基)甲醇(700mg,5.38mmol)溶解在二氯甲烷(10mL)中,在0℃下加入三乙胺(1.09g,10.8mmol)和甲烷磺酰氯(739mg,6.46mmol)。反应液在0℃下反应2小时。加入碳酸氢钠饱和水溶液(10mL)淬灭,用二氯甲烷(50mL x 3)萃取,合并有机相,用饱和氯化钠溶 液(50mL x 3)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得到(4-甲基四氢-2H-吡喃-4-基)甲基甲磺酸酯(700mg,黄色油状物),产率:63%。
1H NMR:(400MHz,Methonal-d4)δ4.08(s,2H),3.73-3.71(m,2H),3.68-3.65(m,2H),3.08(s,3H),1.66-1.59(m,2H),1.39-1.35(m,2H),1.12(s,3H)。
第四步
3,7-二甲基-1-((4-甲基四氢-2H-吡喃-4-基)甲基)-1H-嘌呤-2,6-(3H,7H)-二酮
3,7-二甲基-1-(2-(3-甲基四氢呋喃-3-基)乙基)-1H-嘌呤-2,6-(3H,7H)-二酮
(4-甲基四氢-2H-吡喃-4-基)甲基甲磺酸酯(300mg,1.44mmol),3,7-二甲基-1H-嘌呤-2,6-(3H,7H)-二酮(259mg,1.44mmol),碘化钾(23.9mg,0.144mmol)和碳酸钾(239mg,1.73mmol)溶于无水N,N-二甲基甲酰胺(10mL)中。反应液加热至130℃,微波反应2小时。反应液冷却至20℃,过滤,用制备高效液相色谱纯化,得到3,7-二甲基-1-((4-甲基四氢-2H-吡喃-4-基)甲基)-1H-嘌呤-2,6-(3H,7H)-二酮(异构体1)(80.0mg),产率:19%,和3,7-二甲基-1-(2-(3-甲基四氢呋喃-3-基)乙基)-1H-嘌呤-2,6-(3H,7H)-二酮(异构体2)(90.0mg),产率:24%。
3,7-二甲基-1-((4-甲基四氢-2H-吡喃-4-基)甲基)-1H-嘌呤-2,6-(3H,7H)-二酮。1H NMR:(400MHz,Methonal-d4)δ7.87(s,1H),3.97(s,3H),3.96(s,2H),3.78-3.74(m,2H),3.66-3.64(m,2H),3.33(s,3H),1.68-1.62(m,2H),1.35-1.31(m,2H),1.02(s,3H)。MS-ESI计算值[M+H]+293,实测值293。
3,7-二甲基-1-(2-(3-甲基四氢呋喃-3-基)乙基)-1H-嘌呤-2,6-(3H,7H)-二酮。1H NMR:(400MHz,Methonal-d4)δ7.87(s,1H),4.04-3.99(m,2H),3.92(s,3H),3.90-3.89(m,2H),3.61-3.33(m,5H),1.97-1.93(m,1H),1.77-1.70(m,3H),1.02(s,3H)。MS-ESI计算值[M+H]+293,实测值293。
实施例12
1-((4-乙基四氢-2H-吡喃-4-基)甲基)-3,7-二甲基-1H-嘌呤-2,6-(3H,7H)-二酮
1-(2-(3-乙基四氢-呋喃-3-基)乙基)-3,7-二甲基-1H-嘌呤-2,6-(3H,7H)-二酮
Figure PCTCN2016081103-appb-000039
第一步
4-乙基四氢-2H-吡喃-4-羧酸甲酯
将四氢-2H-吡喃-4-羧酸甲酯(2.50g,17.3mmol)溶于无水四氢呋喃(50mL)中,在氮气保护,-78℃时缓慢滴加二异丙基氨基锂溶液(2M正己烷溶液,10.4mL,20.8mmol),反应液在-78℃搅拌1小时。缓慢加入碘乙烷(5.41g,34.7mmol),继续搅拌1小时。加入水(20mL)淬灭反应。反应液用乙酸乙酯(50mL x 3)萃取,合并有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法纯化(10:1石油醚/乙酸乙酯,Rf=0.4)得到4-乙基四氢-2H-吡喃-4-羧酸甲酯(1.00g,黄色油状),产率:33%。
1H NMR:(400MHz,Methonal-d4)δ3.84-3.81(m,2H),3.73(s,3H),3.47-3.40(m,2H),2.10-2.06(m,2H),1.53-1.28(m,2H),0.85-0.81(m,2H),1.20(t,J=7.2Hz,3H)。
第二步
(4-乙基四氢-2H-吡喃-4-基)甲醇
将4-乙基四氢-2H-吡喃-4-羧酸甲酯(1.00g,5.81mmol)溶于无水四氢呋喃(10mL)中,0℃下加入四氢铝锂(220mg,5.81mmol)。反应液升温至25℃,搅拌1小时。加水(20mL)淬灭,用乙酸乙酯(50mL x 3)萃取,无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法纯化(1:1石油醚/乙酸乙酯,Rf=0.2),得到(4-乙基四氢-2H-吡喃-4-基)甲醇(600mg,黄色油状物),产率:72%。
1H NMR:(400MHz,Methonal-d4)δ3.75-3.66(m,4H),3.45(s,2H),1.54-1.50(m,4H),1.42-1.38(m,2H),0.88-0.84(m,3H)。
第三步
(4-乙基四氢-2H-吡喃-4-基)甲基甲磺酸酯
将(4-乙基四氢-2H-吡喃-4-基)甲醇(600mg,4.16mmol)溶解在二氯甲烷(10mL)中,在0℃下加入三乙胺(843mg,8.32mmol)和甲烷磺酰氯(572mg,4.99mmol)。反应液在0℃下反应2小时。加入碳酸氢钠饱和水溶液(10mL)淬灭,用二氯甲烷(50mL x 3)萃取,合并有机相,用饱和氯化钠溶液(50mL x 3)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得到(4-乙基四氢-2H-吡喃-4-基)甲基甲磺酸酯(600mg,黄色油状物),产率:65%。
1H NMR:(400MHz,Methonal-d4)δ4.12(s,2H),3.71-3.69(m,4H),3.11(s,3H),1.61-1.52(m,4H),1.28-1.24(m,2H),0.93-0.89(m,3H)。
第四步
1-((4-乙基四氢-2H-吡喃-4-基)甲基)-3,7-二甲基-1H-嘌呤-2,6-(3H,7H)-二酮
1-(2-(3-乙基四氢-呋喃-3-基)乙基)-3,7-二甲基-1H-嘌呤-2,6-(3H,7H)-二酮
(4-乙基四氢-2H-吡喃-4-基)甲基甲磺酸酯(300mg,1.35mmol),3,7-二甲基-1H-嘌呤-2,6-(3H,7H)-二酮(243mg,1.35mmol),碘化钾(22.4mg,0.135mmol)和碳酸钾(224mg,1.62mmol)溶于无水N,N-二甲基甲酰胺(10mL)中。反应液加热至130℃,微波反应2小时。反应液冷却至20℃,过滤,用制备高效液相色谱纯化,得到1-((4-乙基四氢-2H-吡喃-4-基)甲基)-3,7-二甲基-1H-嘌呤-2,6-(3H,7H)-二酮(异构体1)(120mg),产率:29%,和1-(2-(3-乙基四氢-呋喃-3-基)乙基)-3, 7-二甲基-1H-嘌呤-2,6-(3H,7H)-二酮(异构体2)(80.0mg),产率:19%。
1-((4-乙基四氢-2H-吡喃-4-基)甲基)-3,7-二甲基-1H-嘌呤-2,6-(3H,7H)-二酮。1H NMR:(400MHz,Methonal-d4)δ7.88(s,1H),4.03(s,2H),3.98(s,3H),3.73-3.72(m,2H),3.65-3.63(m,2H),3.53(s,3H),1.62-1.52(m,4H),1.52-1.43(m,2H),0.98-0.94(m,3H)。MS-ESI计算值[M+H]+307,实测值307。
1-(2-(3-乙基四氢-呋喃-3-基)乙基)-3,7-二甲基-1H-嘌呤-2,6-(3H,7H)-二酮。1H NMR:(400MHz,Methonal-d4)δ7.87(s,1H),4.00-3.98(m,5H),3.87-3.86(m,2H),3.63-3.61(m,2H),3.56(s,3H),1.90-1.80(m,1H),1.75-1.72(m,3H),1.58-1.54(m,2H),1.05-1.01(m,3H)。MS-ESI计算值[M+H]+307,实测值307。
实施例13
3,7-二甲基-1-(2-(四氢-2H-吡喃-4-基)乙基)-1H-嘌呤-2,6(3H,7H)-二酮
Figure PCTCN2016081103-appb-000040
将4-(2-溴乙基)四氢-2H-吡喃(200mg,1.00mmol),3,7-二甲基-1H-嘌呤-2,6(3H,7H)-二酮(186mg,1.00mmol)碘化钾(17.0mg,0.100mmol)及碳酸钾(414mg,3.00mmol)溶于N,N-二甲基甲酰胺(4mL)中,反应液加热至130℃反应3小时。反应液冷却至25℃,加入饱和食盐水淬灭反应,用乙酸乙酯萃取(50mL x 3),有机相用无水硫酸钠干燥,过滤,滤液减压浓缩,用高效制备板纯化(乙酸乙酯,Rf=0.5)得到产物3,7-二甲基-1-(2-(四氢-2H-吡喃-4-基)乙基)-1H-嘌呤-2,6(3H,7H)-二酮(224mg),产率:77%。
1H NMR:(400MHz,Methonal-d4)δ7.86(s,1H),4.06-4.01(m,2H),3.97(s,3H),3.92(dd,J=12,3.2Hz,2H),3.53(s,3H),3.44-3.38(m,2H),1.73(d,J=12.8Hz,2H),1.61-1.55(m,3H),1.38-1.24(m,2H)。MS-ESI计算值[M+H]+293,实测值293。
实施例14
1-[2-((2S,6S)-2,6-二甲基四氢-2H-吡喃-4-基)-乙基]-3,7-二甲基-1H-2,6(3H,7H)-二酮
Figure PCTCN2016081103-appb-000041
Figure PCTCN2016081103-appb-000042
第一步
2-甲基-2H-吡喃-4(3H)-酮
(E)-((4-甲氧基-1,3-二烯-2-基)氧基)三甲基硅烷(5.00g,29.0mmol)和乙醛(55.0g,58.0mmol)溶于无水乙醚(50mL)中,在-78℃下加入三氟化硼乙醚(4.33g,30.5mmol),反应在-78℃搅拌2.5小时。加入饱和溶液氯化铵(40mL)淬灭,用乙酸乙酯(20mL x 3)萃取,无水硫酸钠干燥,过滤,滤液减压浓缩。用硅胶柱色谱法分离纯化(10:1的石油醚/乙酸乙酯,Rf=0.6),得到2-甲基-2H-吡喃-4(3H)-酮(1.25g,黄色油状物),产率:38%。
1H NMR:(400MHz,Methonal-d4)δ7.75(d,J=6.4Hz,1H),5.65(d,J=6.4Hz,1H),4.63-4.58(m,1H),2.59-2.45(m,2H),1.46(d,J=3.2Hz,3H)。
第二步
(2S,6S)-2,6-二甲基-四氢-吡喃-4-酮
甲基锂(1.6M乙醚溶液,20.9mL,33.4mmol)在0℃下溶于无水乙醚(30mL)溶液中,氮气保护下加入碘化亚铜(4.25g,22.3mmol)。反应液在0℃下反应0.5小时。缓慢加入2-甲基-2H-吡喃-4(3H)-酮(1.25g,11.2mmol)的乙醚(5mL)溶液。反应液升温至20℃,搅拌3小时。加入饱和氯化铵溶液(20mL)淬灭,用乙酸乙酯(20mL x 3)萃取,无水硫酸钠干燥,过滤,滤液减压浓缩,通过硅胶柱色谱法分离纯化(10:1石油醚/乙酸乙酯,Rf=0.3),得到(2S,6S)-2,6-二甲基-四氢-吡喃-4-酮(400mg,黄色固体),产率:29%。
1H NMR:(400MHz,Methonal-d4)δ4.36-4.31(m,2H),2.58-2.53(m,2H),2.28-2.25(m,2H),1.25(d,J=6.0Hz,6H)。
第三步
(2,6-二甲基-四氢-吡喃-4-亚基)-乙酸乙酯
将三苯基膦乙酸乙酯(3.26g,9.37mmol)溶于无水甲苯(10mL)中,加入((2S,6S)-2,6-二甲基-四氢-吡喃-4-酮(400mg,3.13mmol)。反应液加热至110℃,反应72小时。冷却至20℃,加水(50 mL)淬灭,用乙酸乙酯(20mL x 3)萃取,无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法(10:1的石油醚/乙酸乙酯,Rf=0.7)纯化,得到(2,6-二甲基-四氢-吡喃-4-亚基)-乙酸乙酯(200mg,黄色油状物),产率:32%。
1H NMR:(400MHz,Methonal-d4)δ5.77(s,1H),4.14-4.07(m,4H),2.98-2.97(m,1H),2.84-2.82(m,1H),2.44-2.40(m,1H),2.11-2.09(m,1H),1.28-1.25(m,3H),1.18-1.16(m,6H)。
第四步
(2,6-二甲基-四氢-吡喃-4-基)-乙酸乙酯
将(2,6-二甲基-四氢-吡喃-4-亚基)-乙酸乙酯(200mg,1.01mmol)溶解在乙酸乙酯(20mL)溶液中,加入湿钯碳(10%,20.0mg)。反应液在25℃,氢气气球压力下,搅拌2小时。反应液过滤,滤液减压浓缩得到(2,6-二甲基-四氢-吡喃-4-基)-乙酸乙酯(150mg,黄色油状物),产率:75%。
1H NMR:(400MHz,Methonal-d4)δ4.20-4.10(m,3H),3.83-3.82(m,1H),2.28-2.21(m,3H),1.72-1.69(m,1H),1.58-1.55(m,1H),1.31-1.30(m,1H),1.28-1.26(m,6H),1.12-1.11(m,1H),1.02-1.01(m,3H)。
第五步
2-(2,6-二甲基-四氢-吡喃-4-基)-乙醇
将(2,6-二甲基-四氢-吡喃-4-基)-乙酸乙酯(150mg,0.750mmol)溶于无水四氢呋喃(2mL)中,0℃下加入四氢铝锂(57.0mg,1.50mmol)。反应液升温至25℃,搅拌1小时。加水(10mL)淬灭,用乙酸乙酯(10mL x 3)萃取,无水硫酸钠干燥,过滤,滤液减压浓缩,用制备TLC板分离纯化(3:1石油醚/乙酸乙酯,Rf=0.5),得到2-(2,6-二甲基-四氢-吡喃-4-基)-乙醇(70.0mg,黄色油状物),产率:86%。
1H NMR:(400MHz,Methonal-d4)δ4.20-4.18(m,1H),3.83-3.81(m,1H),3.64-3.61(m,2H),1.99-1.98(m,1H),1.70-1.68(m,1H),1.55-1.53(m,1H),1.47-1.42(m,3H),1.31-1.29(m,3H),1.13-1.11(m,3H),0.85-0.82(m,1H)。
第六步
2-((2S,6S)-2,6-二甲基四氢-2H-吡喃-4-基)乙基甲磺酸酯
将2-(2,6-二甲基-四氢-吡喃-4-基)-乙醇(150mg,0.949mmol)溶解在二氯甲烷(5mL)中,在0℃下加入三乙胺(287mg,2.85mmol)和甲烷磺酰氯(213mg,1.90mmol)。反应液在25℃下反应2小时。加入碳酸氢钠饱和水溶液(10mL)淬灭,用二氯甲烷(10mL x 3)萃取,合并有机相,用饱和氯化钠溶液洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得到2-((2S,6S)-2,6-二甲基四氢-2H-吡喃-4-基)乙基甲磺酸酯(200mg,黄色油状物),产率:90%。
1H NMR:(400MHz,Methonal-d4)δ4.33-4.30(m,2H),4.23-4.20(m,1H),3.85-3.82(m,1H),3.08(s,3H),2.03-2.14(m,1H),1.68-1.67(m,1H),1.65-1.63(m,2H),1.58-1.56(m,1H),1.41-1.39(m,1H),1.31-1.29(m,3H),1.14-1.12(m,3H),0.89-0.86(m,1H)。
第七步
1-[2-((2S,6S)-2,6-二甲基四氢-2H-吡喃-4-基)-乙基]-3,7-二甲基-1H-2,6(3H,7H)-二酮
将2-((2S,6S)-2,6-二甲基四氢-2H-吡喃-4-基)乙基甲磺酸酯(100mg,0.424mmol),3,7-二甲基-3,7-二氢-嘌呤-2,6-二酮(83.9mg,0.466mmol),碘化钾(7.4mg,0.047mmol)和碳酸钾(109mg,0.790mmol)溶解在N,N-二甲基甲酰胺(3mL)中,反应液加热至120℃,反应3小时。反应冷却至20℃,过滤,滤液用制备高效液相色谱分离纯化,得到1-[2-((2S,6S)-2,6-二甲基四氢-2H-吡喃-4-基)-乙基]-3,7-二甲基-1H-2,6(3H,7H)-二酮(40.0mg),收率:30%。
1H NMR:(400MHz,Methonal-d4)δ8.14(s,1H),4.21-4.18(m,1H),4.07-4.05(m,2H),4.01(s,3H),3.82-3.81(m,1H),3.54(s,3H),1.85-1.66(m,1H),1.55-1.51(m,2H),1.40-1.39(m,2H),1.29-1.28(m,1H),1.27-1.26(m,3H),1.12-1.11(m,3H),0.89-0.86(m,1H)。MS-ESI计算值[M+H]+321,实测值321。
实施例15
1-(2-(3-乙基四氢-2H-吡喃-4-基)乙基)-3,7-二甲基-1H-嘌呤-2,6-(3H,7H)-二酮
Figure PCTCN2016081103-appb-000043
第一步
3-乙基二氢-2H-吡喃-4-(3H)-酮
将二氢-2H-吡喃-4-(3H)-酮(5.00g,50.0mmol)和六甲基磷酰胺(8.95g,50.0mmol)溶于四氢呋喃(50mL)中,-78℃条件下缓慢滴加二异丙基胺基锂(50mL,2.0M四氢呋喃溶液,100mmol),在-78℃条件下搅拌30分钟。反应液于0℃条件下加入碘乙烷(16.2g,75.0mmol)搅拌2小时。反应液中加入水(15mL),乙酸乙酯(40mL x 3)萃取,合并有机相并用饱和氯化钠(20mL x 2)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱层析法分离纯化(10:1石油醚/乙酸乙酯,Rf=0.5)得到3-乙基二氢-2H-吡喃-4-(3H)-酮(1.20g,无色油状物),产率:19%。
1H NMR:(400MHz,CDCl3)δ4.15-4.11(m,2H),3.77-3.76(m,1H),3.47-3.42(m,1H),2.45-2.40(m,3H),1.80-1.78(m,2H),0.91(t,J=7.2Hz,3H)。MS-ESI计算值[M+H]+129,实测值129。
第二步
乙基2-(3-乙基二氢-2H-吡喃-4(3H)-亚基)乙酸乙酯
将3-乙基二氢-2H-吡喃-4(3H)-酮(600mg,4.68mmol)和乙基2-(二乙氧基磷酰基)乙酸乙酯(1.15 g,5.15mmol)溶于四氢呋喃(30mL),反应液于0℃条件下加入氢化钠(224mg,9.36mmol)。室温搅拌30分钟后,反应液在0℃条件下加入水(10mL)。反应液中加入乙酸乙酯(30mL)稀释,有机相用水(20mL x 2)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱层析法分离纯化(10:1石油醚/乙酸乙酯,Rf=0.7)得到乙基2-(3-乙基二氢-2H-吡喃-4(3H)-亚基)乙酸乙酯(800mg,黄色油状物),产率:86%。MS-ESI计算值[M+H]+199,实测值199。
第三步
2-(3-乙基四氢-2H-吡喃-4-基)乙酸乙酯
将乙基2-(3-乙基二氢-2H-吡喃-4(3H)-亚基)乙酸乙酯(800mg,4.04mmol)溶于甲醇(40mL)中,室温条件下加入湿钯碳(10%,0.02g)。反应体系加氢气球置换3次,然后室温反应2小时,反应液过滤浓缩得到2-(3-乙基四氢-2H-吡喃-4-基)乙酸乙酯(600mg,黄色油状物),产率:74%。MS-ESI计算值[M+H]+201,实测值201。
第四步
2-(3-乙基四氢-2H-吡喃-4-基)乙醇
将2-(3-乙基四氢-2H-吡喃-4-基)乙酸乙酯(600mg,3.00mmol,)溶于四氢呋喃(30mL)中,在0℃条件下加入氢化锂铝(170mg,4.50mmol),室温搅拌2小时后,反应液中分别加入水(0.2mL),15%氢氧化钠(0.2mL)和水(0.6mL),搅拌20分钟,反应液中加入乙酸乙酯(30mL)稀释,有机相用水(20mL x 2)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到2-(3-乙基四氢-2H-吡喃-4-基)乙醇(400mg,黄色油状物),产率:84%。
MS-ESI计算值[M+H]+159,实测值159。
第五步
2-(3-乙基四氢-2H-吡喃-4-基)乙基甲磺酸酯
将2-(3-乙基四氢-2H-吡喃-4-基)乙醇(474mg,3.00mmol)和三乙胺(455mg,4.50mmol)溶于二氯甲烷(30mL)中,反应液于0℃条件下加入甲磺酰氯(412mg,3.60mmol)。0℃条件下搅拌2小时,反应液中加入二氯甲烷(30mL)稀释,有机相用水(20mL x 2)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到2-(3-乙基四氢-2H-吡喃-4-基)乙基甲磺酸酯(600mg,黄色油状物)产率:85%。MS-ESI计算值[M+H]+237,实测值237。
第六步
1-(2-(3-乙基四氢-2H-吡喃-4-基)乙基)-3,7-二甲基-1H-嘌呤-2,6-(3H,7H)-二酮
将2-(3-乙基四氢-2H-吡喃-4-基)乙基甲磺酸酯(400mg,1.69mmol)和3,7-二甲基-1H-嘌呤-2,6-(3H,7H)-二酮(305mg,1.69mmol)溶于N,N-二甲基甲酰胺(20mL)中,室温条件下加入碳酸钾(467mg,3.38mmol)和碘化钾(28.0mg,0.169mmol)。反应液于100℃条件下搅拌2小时,反应液冷却至室温浓缩,加入乙酸乙酯(30mL)稀释,有机相用水(20mL x 2)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,用高效液相色谱纯化得1-(2-(3-乙基四氢-2H-吡喃-4-基)乙基)-3,7-二甲基-1H-嘌呤-2,6-(3H,7H)-二酮(200mg),产率:37%。
1H NMR:(400MHz,CDCl3)δ7.49(s,1H),3.97-3.95(m,5H),3.83-3.77(m,2H),3.43(s,3H), 3.42-3.35(m,2H),1.79-1.41(m,8H),0.91-0.82(m,3H)。MS-ESI计算值[M+H]+321,实测值321。
实施例16
1-(2-(2-甲基四氢-2H-吡喃-4-基)乙基)-3,7-二甲基-1H-嘌呤-2,6(3H,7H)-二酮
Figure PCTCN2016081103-appb-000044
第一步
2-(2-甲基四氢-2H-吡喃-4-基)乙基甲磺酸酯
将2-(2-甲基四氢-2H-吡喃-4-基)乙醇(100mg,0.630mmol)和N,N-二异丙基乙胺(122mg,0.940mmol)溶于二氯甲烷(20mL)中,0℃下加入甲烷磺酰氯(86.9mg,0.750mmol)。反应液于25℃搅拌0.5小时后,加入二氯甲烷稀释(20mL),用饱和碳酸氢钠洗涤(30mL x 2),有机相用无水硫酸钠干燥,浓缩得2-(2-甲基四氢-2H-吡喃-4-基)乙基甲磺酸酯(180mg,黄色油状物),收率:100%。
第二步
1-(2-(2-甲基四氢-2H-吡喃-4-基)乙基)-3,7-二甲基-1H-嘌呤-2,6(3H,7H)-二酮
将2-(2-甲基四氢-2H-吡喃-4-基)乙基甲磺酸酯(180mg,0.760mmol)溶于N,N-二甲基甲酰胺(10mL)中,反应液于25℃条件下加入3,7-二甲基-1H-嘌呤-2,6(3H,7H)-二酮(146mg,0.800mmol),碘化钾(13.0mg,0.0800mmol)和碳酸钾(211mg,1.60mmol)。反应液加热至120℃反应2小时,加入乙酸乙酯(30mL)稀释,用饱和碳酸氢钠洗涤(20mL x 2),有机相用无水硫酸钠干燥,浓缩得黄色油状物,经高效液相色谱法分离纯化得到1-(2-(2-甲基四氢-2H-吡喃-4-基)乙基)-3,7-二甲基-1H-嘌呤-2,6(3H,7H)-二酮(50.0mg),收率:20%。
1H NMR:(400MHz,Methanol-d4)δ7.87(s,1H),4.05-3.96(m,6H),3.54-3.19(m,5H),1.80-1.45(m,8H),0.98-0.92(m,4H)。MS-ESI计算值[M+H]+321,实测值321。
实施例17
3,7-二甲基-1-(2-吗啡啉乙基)-嘌呤-2,6-二酮
Figure PCTCN2016081103-appb-000045
Figure PCTCN2016081103-appb-000046
在室温下,向混合物1-(3-氯丙基)-3,7-二甲基-嘌呤-2,6(3H,7H)-二酮(71.9mg,0.826mmol)和吗啡啉(50.0mg,0.207mmol)的乙腈(2mL)溶液中加入碳酸钾(138mg,1.03mmol)和碘化钾(86.3mg,0.517mmol)。反应液在90℃搅拌4个小时。加入水(5mL)淬灭反应,用乙酸乙酯萃取(5mL x 3)。有机相用饱和食盐水洗涤,无水硫酸钠干燥后减压浓缩。用制备高效液相色谱分离纯化得到3,7-二甲基-1-(2-吗啡啉乙基)-嘌呤-2,6-二酮(15.0mg),产率:25%。1H NMR:(400MHz,Methonal-d4)δ7.86(s,1H),4.16(t,J=6.4Hz,2H),3.97(s,3H),3.68-3.65(m,4H),3.52(s,3H),2.65-2.57(m,6H)。MS-ESI计算值[M+H]+294,实测值294。
实施例18
((-4-甲氧基苯基)甲基)-3,7-二甲基-1H-嘌呤-2,6-(3H,7H)-二酮
Figure PCTCN2016081103-appb-000047
第一步
4-甲氧基环己烷羧酸甲酯
将4-甲氧基环己烷甲酸(300mg,1.90mmol)溶于甲醇(7mL)中,在0℃下缓慢加入二氯亚砜(1.13g,9.50mmol),反应液室温搅拌12小时。反应液减压浓缩,得到粗产品4-甲氧基环己烷羧酸甲酯(283mg,黄色油状),产率:86%。
1H NMR:(400MHz,Methonal-d4)δ3.65(s,3H),3.41-3.35(m,1H),3.30(s,3H),2.44-2.38(m,1H),1.84-1.75(m,4H),1.67-1.54(m,4H)
第二步
(4-甲氧基环己基)甲醇
0℃时,在氮气保护下将四氢铝锂(92.8mg,2.45mmol)缓慢加入溶有甲基-4-甲氧基环己烷羧酸(280mg,1.63mmol)的四氢呋喃(7mL)中,反应室温搅拌4小时,反应液用冰水浴中冷却至0℃,依次缓慢加入水(0.1mL),15%氢氧化钠(0.1mL)及水(0.3mL)。反应液升温至室温后搅拌半小时,过滤,滤饼用四氢呋喃(8mL x 2)洗涤,滤液减压浓缩得(4-甲氧基环己基)甲醇(213mg,黄色油 状),产率:91%。1H NMR:(400MHz,Methonal-d4)δ3.49-3.45(m,1H),3.39-3.37(m,2H),3.32(s,3H),1.95-1.80(m,3H),1.56-1.48(m,4H),1.36-1.27(m,2H)。
第三步
(4-甲氧基环己基)甲基甲磺酸酯
将(4-甲氧基环己基)甲醇(210mg,1.46mmol),三乙胺(443mg,4.38mmol)溶于二氯甲烷(7mL)中,在0℃下缓慢加入甲烷磺酰氯(250mg,2.19mmol)。反应液室温搅拌过夜,加入水,用二氯甲烷(50mL x 3)萃取,有机相用饱和食盐水(25mL x 2)洗涤并用无水硫酸钠干燥,过滤,滤液减压浓缩,得到的产品用硅胶柱色谱法纯化(5:1石油醚/乙酸乙酯,Rf=0.4)得到产物(4-甲氧基环己基)甲基甲磺酸酯(240mg,黄色油状),产率:74%。1H NMR:(400MHz,Methonal-d4)δ4.07(d,J=6.4Hz,2H),3.51-3.47(m,1H),3.32(s,3H),3.07(s,3H),1.98-1.92(m,2H),1.85-1.78(m,1H),1.63-1.36(m,6H)。
第四步
1((-4-甲氧基环己基)甲基)-3,7-二甲基-1H-嘌呤-2,6-(3H,7H)-二酮
将(4-甲氧基环己基)甲基甲磺酸酯(240mg,1.08mmol),3,7-二甲基-1H-嘌呤-2,6-(3H,7H)-二酮(233mg,1.30mmol)及碘化钾(17.9mg,0.108mmol)溶于N,N-二甲基甲酰胺(5mL)中,加入碳酸钾(298mg,2.16mmol),反应130℃加热回流4小时。反应液冷却至室温,过滤,滤液减压浓缩,得到的产品用制备高效液相色谱纯化得到产物1((-4-甲氧基环己基)甲基)-3,7-二甲基-1H-嘌呤-2,6-(3H,7H)-二酮(67.0mg),产率:20%。1H NMR:(400MHz,Methonal-d4)δ7.88(s,1H),3.99(s,3H),3.89(d,J=7.2Hz,2H),3.54(s,3H),3.47-3.42(m,1H),3.32(s,3H),1.94-1.87(m,3H),1.47-1.39(m,6H)。MS-ESI计算值[M+H]+307,实测值307。
实施例19
3-甲基-1-((3-甲基氧杂环丁烷-3-基)甲基)-7-(2,2,2-三氟乙基)-1H-嘌呤-2,6(3H,7H)-二酮
Figure PCTCN2016081103-appb-000048
第一步
3-甲基-1-((3-甲基氧杂环丁烷-3-基)甲基)-7-(2,2,2-三氟乙基)-1H-嘌呤-2,6(3H,7H)-二酮将3-甲基-7-(2,2,2-三氟乙基)-1H-嘌呤-2,6(3H,7H)-二酮(200mg,0.806mmol),3-(氯甲基)-3-甲基氧杂环丁烷(97.2mg,0.806mmol)及碘化钾(13.4mg,0.0806mmol)溶于N,N-二甲基甲酰胺(5mL)中,加入碳酸钾(223mg,1.61mmol),反应130℃加热回流2.5小时。反应液冷却至25℃,过滤,滤液减压浓缩,得到的产品用制备高效液相色谱纯化得到产物3-甲基-1-((3-甲基氧杂环丁烷-3-基)甲基)-7-(2,2,2-三氟乙基)-1H-嘌呤-2,6(3H,7H)-二酮(48.0mg),产率:18%。 1H NMR:(400M Hz,Methonal-d4)δ8.09(s,1H),5.25-5.18(m,2H),4.75(d,J=6.4Hz,2H),4.23(d,J=6.4Hz,2H),4.15(s,2H),3.56(s,3H),1.35(s,3H)。MS-ESI计算值[M+H]+333,实测值333。
实施例20
1-(2-((2R,6R)-2,6-二甲基四氢-2H-吡喃-4-基)乙基)-3-甲基-7-(2,2,2-三氟乙基)-1H-嘌呤2,6(3H,7H)-二酮
Figure PCTCN2016081103-appb-000049
将3-甲基-7-(2,2,2-三氟乙基)-1H-嘌呤-2,6(3H,7H)-二酮(100mg,0.4mmol),碘化钾(7.0mg,0.040mmol)及碳酸钾(165mg,1.20mmol)溶于N,N-二甲基甲酰胺(3mL)中,反应加热至130℃反应1小时,加入2-((2S,6S)-2,6-二甲基四氢-2H-吡喃-4-基)乙基甲磺酸酯(50.0mg,0.200mmol),在130℃继续反应2.5小时。反应液直接过滤,滤液减压浓缩,得到粗产品用制备高效液相色谱纯化得产物1-(2-((2R,6R)-2,6-二甲基四氢-2H-吡喃-4-基)乙基)-3-甲基-7-(2,2,2-三氟乙基)-1H-嘌呤2,6(3H,7H)-二酮(68.0mg),产率:87%。1H NMR:(400MHz,Methonal-d4)δ8.08(s,1H),5.27-5.21(m,2H),4.22-4.15(m,1H),4.10-4.03(m,2H),3.80-3.75(m,1H),3.57(s,3H),1.95-1.30(m,7H)1.28(d,J=6.8Hz,3H),1.12(d,J=6.8Hz,3H)。MS-ESI计算值[M+H]+389,实测值389。
实施例21
1-(2-((2R,6R)-2,6-二甲基四氢-2H-吡喃-4-基)乙基)基-7-异丙基-3-甲基-1H-嘌呤-2,6(3H,7H)-二酮
Figure PCTCN2016081103-appb-000050
将2-((2S,6S)-2,6-二甲基四氢-2H-吡喃-4-基)乙基甲磺酸酯(100mg,0.424mmol),7-异丙基-3- 甲基-1H-嘌呤-2,6(3H,7H)-二酮(100mg,0.466mmol),碘化钾(7.4mg,0.047mmol)和碳酸钾(109mg,0.790mmol)溶于无水N,N-二甲基甲酰胺(3mL)中,反应液加热至120℃,搅拌3小时。反应冷却至20℃,过滤,滤液用制备高效液相色谱分离纯化,得到1-(2-((2R,6R)-2,6-二甲基四氢-2H-吡喃-4-基)乙基)基-7-异丙基-3-甲基-1H-嘌呤-2,6(3H,7H)-二酮(70.0mg),收率:46%。
1H NMR:(400MHz,Methonal-d4)δ8.92(s,1H),5.21-5.15(m,1H),4.21-4.09(m,1H),4.08-4.06(m,2H),4.05-4.04(m,1H),3.58(s,3H),1.86-1.80(m,1H),1.68-1.66(m,2H),1.65-1.64(m,6H),1.57-1.55(m,2H),1.54-1.52(m,1H),1.30-1.28(m,3H),1.13-1.12(m,3H),0.90-0.87(m,1H)。MS-ESI计算值[M+H]+349,实测值349。
实施例22
7-(环丙基甲基)-3-甲基-1-((3-甲基氧杂环丁烷-3-基)甲基)-1H-嘌呤-2,6(3H,7H)-二酮
Figure PCTCN2016081103-appb-000051
第一步
7-(环丙基甲基)-3-甲基-1-((3-甲基氧杂环丁烷-3-基)甲基)-1H-嘌呤-2,6(3H,7H)-二酮
将7-(环丙基甲基)-3-甲基-1H-嘌呤-2,6(3H,7H)-二酮(250mg,1.14mmol),3-(氯甲基)-3-甲基氧杂环丁烷(137mg,1.14mmol)及碘化钾(18.9mg,0.114mmol)溶于N,N-二甲基甲酰胺(5mL)中,加入碳酸钾(315mg,2.28mmol),反应130℃加热回流2.5小时。反应液冷却至25℃,过滤,滤液减压浓缩,得到的产品用制备高效液相色谱纯化得到产物7-(环丙基甲基)-3-甲基-1-((3-甲基氧杂环丁烷-3-基)甲基)-1H-嘌呤-2,6(3H,7H)-二酮(55.0mg),产率:16%。1H NMR:(400M Hz,Methonal-d4)δ8.01(s,1H),4.76(d,J=6.4Hz,2H),4.24-4.18(m,4H),4.14(s,2H),3.55(s,3H),1.39-1.33(m,4H),0.62-0.56(m,2H),0.48-0.43(m,2H)。MS-ESI计算值[M+H]+305,实测值305。
实施例23
7-(环丙基甲基-1-(2-((2R,6R)-2,6-二甲基四氢-2H-吡喃-4-基)乙基-3-甲基-1H-嘌呤-2,6(3H,7H)-二酮
Figure PCTCN2016081103-appb-000052
Figure PCTCN2016081103-appb-000053
将((2S,6S)-2,6-二甲基四氢-2H-吡喃-4-基)乙基甲磺酸酯(100mg,0.424mmol)和7-(环丙基甲基)-3-甲基-1H-嘌呤-2,6(3H,7H)-二酮(102mg,0.466mmol)溶于在N,N-二甲基甲酰胺(3mL),氮气保护下加入碳酸钾(109mg,0.790mmol)和碘化钾(7.4mg,0.047mmol)。反应液加热至120℃,搅拌3小时。冷却至20℃后,将混合物过滤,滤液通过制备高效液相色谱分离纯化,得到7-(环丙基甲基)-1-(2-((2R,6R)-2,6-二甲基四氢-2H-吡喃-4-基)乙基)-3-甲基-1H-嘌呤-2,6(3H,7H)-二酮(70.0mg),收率:46%。
1H NMR:(400MHz,Methonal-d4)δ8.02(s,1H),4.25-4.21(m,3H),4.20-4.19(m,2H),4.08-4.02(m,1H),3.55(s,3H),1.87-1.80(m,1H),1.66-1.56(m,2H),1.54-1.50(m,2H),1.42-1.40(m,2H),1.29-1.27(m,3H),1.13-1.12(m,3H),0.89-0.86(m,1H),0.63-0.60(m,2H),0.49-0.47(m,2H)。MS-ESI计算值[M+H]+361,实测值361。
实施例24
7-(环丙基甲基)-1-((4-甲氧基环己基)甲基)-3-甲基-嘌呤-2,6-二酮
Figure PCTCN2016081103-appb-000054
第一步
7-(环丙基甲基)-1-((4-甲氧基环己基)甲基)-3-甲基-嘌呤-2,6-二酮
将(4-甲氧基环己基)甲基甲磺酸酯(30.0mg,0.135mmol),7-(环丙基甲基)-3-甲基-嘌呤-2,6-二酮(26.8mg,0.121mmol),碘化钾(2.2mg,0.014mmol)和碳酸钾(37.3mg,0.269mmol)溶于N,N-二甲基甲酰胺(5mL)中。反应液升温至120℃,搅拌3小时。冷却至室温,过滤,滤液减压浓缩,剩余物用制备型高效液相色谱纯化得到7-(环丙基甲基)-1-((4-甲氧基环己基)甲基)-3-甲基-嘌呤-2,6-二酮(20.0mg),产率:43%。1H NMR:(400MHz,Methonal-d4)δ8.01(s,1H),4.20(d,J=7.2Hz,2H),3.88(d,J=16.0Hz,2H),3.56(s,3H),3.34(s,3H),3.17-3.14(m,1H),2.09-2.06(m,2H),1.75-1.73(m,3H),1.41-1.39(m,1H),1.34-1.10(m,4H),0.63-0.61(m,2H),0.48-0.47(m,2H)。MS-ESI计算值[M+H]+347,实测值347。
实施例25
7-异丁基-3-甲基-1-((3-甲基氧杂环丁烷-3-基)甲基)-1H-嘌呤-2,6(3H,7H)-二酮
Figure PCTCN2016081103-appb-000055
第一步
7-异丁基-3-甲基-1-((3-甲基氧杂环丁烷-3-基)甲基)-1H-嘌呤-2,6(3H,7H)-二酮
将7-异丁基-3-甲基-1H-嘌呤-2,6(3H,7H)-二酮(200mg,0.900mmol),3-(氯甲基)-3-甲基氧杂环丁烷(109mg,0.900mmol)及碘化钾(14.9mg,0.0900mmol)溶于N,N-二甲基甲酰胺(5mL)中,加入碳酸钾(249mg,1.80mmol),反应130℃加热回流2.5小时。反应液冷却至25℃,过滤,滤液减压浓缩,得到的产品用制备高效液相色谱纯化得到产物7-异丁基-3-甲基-1-((3-甲基氧杂环丁烷-3-基)甲基)-1H-嘌呤-2,6(3H,7H)-二酮(34.0mg),产率:12%。1H NMR:(400M Hz,Methonal-d4)δ7.95(s,1H),4.75(d,J=1.2Hz,2H),4.22(d,J=6.4Hz,2H),4.15-4.12(m,4H),3.55(s,3H),2.23-2.12(m,1H),1.35(s,3H),0.92(d,J=6.8Hz,6H)。MS-ESI计算值[M+H]+307,实测值307。
实施例26
1-(2-((2R,6R)-2,6-二甲基四氢-2H-吡喃-4-基)乙基)-7-异丁基-3-甲基-1H-嘌呤-2,6(3H,7H)-二酮
Figure PCTCN2016081103-appb-000056
将7-异丁基-3-甲基-1H-嘌呤-2,6(3H,7H)-二酮(90.0mg,0.400mmol),碘化钾(7.0mg,0.040mmol)及碳酸钾(165mg,1.20mmol)溶于N,N-二甲基甲酰胺(3mL)中,反应加热至130℃反应1小时,加入2-((2S,6S)-2,6-二甲基四氢-2H-吡喃-4-基)乙基甲磺酸酯(50.0mg,0.200mmol),在130℃继续反应2.5小时。反应液直接过滤,滤液减压浓缩,得到粗产品用制备高效液相色谱纯化得产物1-(2-((2R,6R)-2,6-二甲基四氢-2H-吡喃-4-基)乙基)-7-异丁基-3-甲基-1H-嘌呤-2,6(3H,7H)-二酮(71.0mg),产率:97%。
1H NMR:(400MHz,Methonal-d4)δ7.93(s,1H),4.24-3.98(m,5H),3.86-3.78(m,1H),3.55(s,3H),2.26-2.16(m,1H),1.79-1.77(m,1H),1.72-1.65(m,1H),1.60-1.47(m,2H),1.45-1.38(m,1H),1.28(d,J=6.8Hz,3H),1.12(d,J=6.8Hz,3H),0.94-0.88(m,8H)。MS-ESI计算值[M+H]+363,实测值363。
实施例27
4-甲基-6-((3-甲基氧杂环丁烷-3-基)甲基)-1-(2,2,2-三氟乙基)-吡唑并[4,3-d]嘧啶-5,7-二酮
Figure PCTCN2016081103-appb-000057
第一步
4-硝基-吡唑-5-甲酸甲酯
将4-硝基-吡唑-5-甲酸(45.0g,286mmol)溶于甲醇(700mL),0℃下滴加氯化亚砜(102g,859mmol)。反应液在25℃下搅拌反应18小时。反应液减压浓缩,得4-硝基-吡唑-5-甲酸甲酯(49.0g,白色固体),产率:100%。1H NMR:(400MHz,CDCl3)δ8.53(s,1H),4.06(s,3H)。MS-ESI计算值[M+H]+172,实测值172。
第二步
4-硝基-1-(2,2,2-三氟乙基)-吡唑-5-甲酸甲酯
将4-硝基-吡唑-5-甲酸甲酯(25.0g,146mmol)溶于N,N-二甲基甲酰胺(350mL),0℃下分批加入 氢化钠(6.43g,161mmol)。0℃下搅拌反应1小时,滴加2,2,2-三氟乙基三氟甲烷磺酸酯(33.9g,146mmol)。反应液在25℃下搅拌反应18小时。向反应液中加入水(1.2L),用乙酸乙酯萃取(300mL x 2)。合并有机相,饱和食盐水洗涤(500mL),无水硫酸钠干燥,过滤,减压浓缩,剩余物用硅胶柱色谱法分离纯化(5:1石油醚/乙酸乙酯,Rf=0.3),得到4-硝基-1-(2,2,2-三氟乙基)-吡唑-5-甲酸甲酯(8.00g,无色油状物),产率:22%。1H NMR:(400MHz,CDCl3)δ8.13(s,1H),5.06(q,J=8.0Hz,2H),4.04(s,3H)。MS-ESI计算值[M+H]+254,实测值254。
第三步
4-氨基-1-(2,2,2-三氟乙基)-吡唑-5-甲酸甲酯
将4-硝基-1-(2,2,2-三氟乙基)-吡唑-5-甲酸甲酯(7.50g,29.6mmol)溶于甲醇(100mL),加入干钯碳(钯10%,水1%,750mg),室温下,反应液于40psi氢气压力下反应3小时。反应液过滤,滤液减压浓缩得到4-氨基-1-(2,2,2-三氟乙基)-吡唑-5-甲酸甲酯(6.30g,类白色固体),产率:95%。
1H NMR:(400MHz,CDCl3)δ7.25(s,1H),5.10(q,J=8.4Hz,2H),4.21(s,2H),3.94(s,3H)。MS-ESI计算值[M+H]+224,实测值224。
第四步
4-(2,2,2-三氟乙酰胺)-1-(2,2,2-三氟乙基)-吡唑-5-甲酸甲酯
将4-氨基-1-(2,2,2-三氟乙基)-吡唑-5-甲酸甲酯(6.30g,28.2mmol)溶于二氯甲烷(100mL),氮气保护下滴加三氟乙酸酐(8.89g,42.4mmol),反应液室温下搅拌2小时,用饱和碳酸氢钠溶液(100mL)淬灭反应,二氯甲烷(100mL)萃取,饱和食盐水(50mL)洗涤有机相,无水硫酸钠干燥,减压浓缩,得4-(2,2,2-三氟乙酰胺)-1-(2,2,2-三氟乙基)-吡唑-5-甲酸甲酯(9.20g粗产品,黄色油状物)。1H NMR:(400MHz,CDCl3)δ9.66(s,1H),8.45(s,1H),5.18(q,J=8.0Hz,2H),4.06(s,3H)。MS-ESI计算值[M+H]+320,实测值320。
第五步
4-(2,2,2-三氟-N-甲基乙酰胺)-1-(2,2,2-三氟乙基)-吡唑-5-甲酸甲酯
将4-(2,2,2-三氟乙酰胺)-1-(2,2,2-三氟乙基)-吡唑-5-甲酸甲酯(9.20g,28.8mmol)溶于N,N-二甲基甲酰胺(50mL),加入碳酸钾(5.98g,43.3mmol)。反应液加热至80℃反应1小时。冷却至室温,加入碘甲烷(6.14g,43.2mmol)。反应液室温搅拌18小时,然后向反应液中加入水(300mL),用乙酸乙酯萃取(100mL x 3)。合并有机相,饱和食盐水洗涤(100mL),无水硫酸钠干燥,过滤,减压浓缩,得4-(2,2,2-三氟-N-甲基乙酰胺)-1-(2,2,2-三氟乙基)-吡唑-5-甲酸甲酯(9.80g粗产品,黄色油状物)。1H NMR:(400MHz,CDCl3)δ7.65(s,1H),5.45-5.15(m,2H),3.93(s,3H),3.29(s,3H)。MS-ESI计算值[M+H]+334,实测值334。
第六步
4-[(叔丁氧基羰基)(甲基)氨基]-1-(2,2,2-三氟乙基)-吡唑-5-甲酸
将4-(2,2,2-三氟-N-甲基乙酰胺)-1-(2,2,2-三氟乙基)-吡唑-5-甲酸甲酯(9.90g,29.7mmol)溶于四氢呋喃(40mL)和水(40mL),加入一水合氢氧化锂(6.23g,0.149mol),室温搅拌反应18小时。加入二碳酸二叔丁酯(13.0g,59.4mmol),反应液室温下继续反应6小时,反应液减压浓缩, 用2N盐酸水溶液调节pH=4,过滤,干燥滤饼,得到4-[(叔丁氧基羰基)(甲基)氨基]-1-(2,2,2-三氟乙基)-吡唑-5-甲酸(8.00g,白色固体),产率:83%。1H NMR:(400MHz,CDCl3)δ7.58(s,1H),5.25(q,J=8.0Hz,2H),3.27(s,3H),1.42(s,9H)。MS-ESI计算值[M+H]+324,实测值324。
第七步
4-[(叔丁氧基羰基)(甲基)氨基]-1-(2,2,2-三氟乙基)-吡唑-5-甲酰胺
将4-[叔丁氧基羰基(甲基)氨基]-2-(2,2,2-三氟乙基)吡唑-5-羧酸,2-(7-偶氮苯并三氮唑)-四甲基脲六氟磷酸酯(13.8g,36.2mmol)和氯化铵(2.98g,55.7mmol)溶于二氯甲烷(120mL),室温下滴加三乙胺(4.23g,41.8mmol)。反应液室温搅拌18小时,向反应液中加入水(100mL),用二氯甲烷(100mL x 2)萃取,合并有机相,依次用饱和碳酸氢钠(50mL)和饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩,剩余物用乙醇(20mL),得到4-[(叔丁氧基羰基)(甲基)氨基]-1-(2,2,2-三氟乙基)-吡唑-5-甲酰胺(6.00g,白色固体),产率:67%。1H NMR:(400MHz,CDCl3)δ7.54(s,1H),5.25(q,J=8.0Hz,2H),3.22(s,3H),1.48(s,9H)。MS-ESI计算值[M+H]+323,实测值323。
第八步
4-(甲基氨基)-2-(2,2,2-三氟乙基)吡唑-5-甲酰胺
将4-[(叔丁氧基羰基)(甲基)氨基]-1-(2,2,2-三氟乙基)-吡唑-5-甲酰胺(5.00g,15.51mmol)溶于盐酸乙酸乙酯(50mL)。室温下搅拌反应18小时,减压浓缩。剩余物用甲醇(50mL)溶解,加入碳酸钾(5.36g,38.8mmol),室温搅拌2小时。减压浓缩,剩余物用二氯甲烷(100mL)萃取,过滤,滤液旋干,得到4-(甲基氨基)-2-(2,2,2-三氟乙基)吡唑-5-甲酰胺(2.90g,白色固体),产率:84%。1H NMR:(400MHz,Methanol-d4)δ7.93(s,1H),5.26(q,J=8.4Hz,2H),3.13(s,3H)。MS-ESI计算值[M+H]+223,实测值223。
第九步
4-甲基-1-(2,2,2-三氟乙基)-吡唑并[4,3-d]嘧啶-5,7-二酮
将4-(甲基氨基)-2-(2,2,2-三氟乙基)吡唑-5-甲酰胺(2.70g,12.2mmol)和1,1-羰基二咪唑(3.94g,24.3mmol)溶于N,N-二甲基甲酰胺(20mL),反应液加热至140℃反应1小时。冷却至室温,向反应液中加入水(100mL),固体析出,过滤收集,烘干滤饼,得到4-甲基-1-(2,2,2-三氟乙基)-吡唑并[4,3-d]嘧啶-5,7-二酮(1.80g,白色固体),产率:60%。1H NMR:(400MHz,DMSO-d6)δ11.56(s,1H),7.95(s,1H),5.35(q,J=8.8Hz,2H),3.33(s,3H)。MS-ESI计算值[M+H]+249,实测值249。
第十步
4-甲基-6-((3-甲基氧杂环丁烷-3-基)甲基)-1-(2,2,2-三氟乙基)-吡唑并[4,3-d]嘧啶-5,7-二酮将4-甲基-1-(2,2,2-三氟乙基)吡唑并[4,3-d]嘧啶-5,7-二酮(70.0mg,0.282mmol)溶于N,N-二甲基甲酰胺(2mL),加入3-(氯甲基)-3-甲基-氧杂环丁烷(40.8mg,0.338mmol),碳酸钾(78.0mg,0.564mmol)和碘化钾(56.2mg,0.338mmol)。反应液加热到120℃,搅拌1小时。反应液冷至 室温,过滤。滤液减压浓缩,剩余物用高效液相色谱法纯化,得4-甲基-6-[(3-甲基氧杂环丁烷-3-基)甲基]-1-(2,2,2-三氟乙基)吡唑并[4,3-d]嘧啶-5,7-二酮(30.0mg),产率:32%。1H NMR:(400MHz,CDCl3)δ7.57(s,1H),5.21(q,J=8.0Hz,2H),4.72(d,J=6.0Hz,2H),4.26(d,J=6.0Hz,2H),4.16(s,2H),3.52(s,3H),1.40(s,3H)。MS-ESI计算值[M+H]+333,实测值333。
实施例28
6-((3-乙基氧杂环丁烷-3-基)甲基)-4-甲基-1-(2,2,2-三氟乙基)-吡唑并[4,3-d]嘧啶-5,7-二酮
Figure PCTCN2016081103-appb-000058
第一步
(3-乙基氧杂环丁烷-3-基)甲基甲磺酸酯
将(3-乙基氧杂环丁烷-3-基)甲醇(1.00g,8.61mmol)和三乙胺(1.74g,17.2mmol)溶于无水二氯甲烷(15mL)中,氮气保护,0℃缓慢加入甲烷磺酰氯(1.28g,11.2mmol)。反应液在0℃,搅拌1小时。加入饱和碳酸氢钠溶液(50mL)淬灭反应,用二氯甲烷(20mL x 2)萃取。合并有机相,饱和氯化钠溶液(50mL x 2)洗涤,用无水硫酸镁干燥,过滤,滤液减压浓缩得到(3-乙基氧杂环丁烷-3-基)甲基甲磺酸酯(1.30g,黄色油状),产率:78%。MS-ESI计算值[M+H]+195,实测值195。
第二步
6-((3-乙基氧杂环丁烷-3-基)甲基)-4-甲基-1-(2,2,2-三氟乙基)-吡唑并[4,3-d]嘧啶-5,7-二酮将4-甲基-1-(2,2,2-三氟乙基)吡唑并[4,3-d]嘧啶-5,7-二酮(30.0mg,0.121mmol),(3-乙基氧杂环丁烷-3-基)甲基甲磺酸酯(35.2mg,0.181mmol),碳酸钾(33.2mg,0.242mmol)和碘化钾(4.0mg,0.024mmol)溶解于N,N-二甲基甲酰胺(10mL)中。反应液加热到120℃,搅拌3小时。反应液冷至室温,倒入水(30mL)中,用乙酸乙酯萃取(20mL x 3)。合并有机相,用无水硫酸钠干燥,过滤,滤液浓缩。粗品用制备高效液相色谱分离纯化得到6-((3-乙基氧杂环丁烷-3-基)甲基)-4-甲基-1-(2,2,2-三氟乙基)-吡唑并[4,3-d]嘧啶-5,7-二酮(10mg),产率:24%。1H NMR:(400MHz,Methonal-d4)δ7.86(s,1H),5.37-5.31(m,2H),4.64(d,J=6.4Hz,2H),4.30(d,J=6.4Hz,2H),4.14(s,2H),3.54(s,3H),1.84-1.79(m,2H),1.07(t,J=7.2Hz,3H)。MS-ESI计算值[M+H]+347,实测值347。
实施例29
4-甲基-6-((四氢呋喃-3-基)甲基)-1-(2,2,2-三氟乙基)-吡唑并[4,3-d]嘧啶-5,7-二酮
Figure PCTCN2016081103-appb-000059
Figure PCTCN2016081103-appb-000060
第一步
4-甲基-6-((四氢呋喃-3-基)甲基)-1-(2,2,2-三氟乙基)-吡唑并[4,3-d]嘧啶-5,7-二酮
将四氢呋喃-3-基甲基甲磺酸酯(30.0mg,0.166mmol),4-甲基-1-(2,2,2-三氟乙基)吡唑并[4,3-d]嘧啶-5,7-二酮(41.3mg,0.166mmol),碳酸钾(46.0mg,0.333mmol)和碘化钾(5.5mg,0.033mmol)溶解于N,N-二甲基甲酰胺(5mL)中。反应液加热到120℃,搅拌3小时。反应液冷至室温,倒入水(20mL)中,用乙酸乙酯萃取(20mL x 3)。合并有机相,用无水硫酸钠干燥,过滤,滤液浓缩,用制备高效液相色谱分离纯化得到4-甲基-6-((四氢呋喃-3-基)甲基)-1-(2,2,2-三氟乙基)-吡唑并[4,3-d]嘧啶-5,7-二酮(10.0mg),产率:18%。1H NMR:(400MHz,Methonal-d4)δ7.83(s,1H),5.38-5.30(m,2H),4.10-4.04(m,2H),3.92-3.87(m,1H),3.78-3.75(m,2H),3.64-3.62(m,1H),3.52(s,3H),2.76-2.72(m,1H),2.04-2.01(m,1H),1.79-1.72(m,1H)。MS-ESI计算值[M+H]+333,实测值333。
实施例30
6-((3-乙基氧杂环丁烷-3-基)甲基)-4-甲基-1-(2,2,2-三氟乙基)-吡唑并[4,3-d]嘧啶-5,7-二酮
Figure PCTCN2016081103-appb-000061
第一步
(四氢吡喃-4-基)甲基甲磺酸酯
将四氢吡喃-4-基甲醇(1.00g,8.61mmol)和三乙胺(1.74g,17.2mmol)溶于无水二氯甲烷(20mL)中,氮气保护,0℃缓慢加入甲烷磺酰氯(1.28g,11.2mmol)。反应液在0℃,搅拌1小时。加入饱和碳酸氢钠溶液(50mL)淬灭反应,用二氯甲烷(20mL x 2)萃取。合并有机相,饱和氯化钠溶液(50mL x 2)洗涤,用无水硫酸镁干燥,过滤,滤液减压浓缩得到(四氢吡喃-4-基)甲基甲磺酸酯(1.30g,黄色油状),产率:78%。MS-ESI计算值[M+H]+195,实测值195。
第二步
4-甲基-6-((四氢吡喃-4-基)甲基)-1-(2,2,2-三氟乙基)-吡唑并[4,3-d]嘧啶-5,7-二酮
将4-甲基-1-(2,2,2-三氟乙基)吡唑并[4,3-d]嘧啶-5,7-二酮(30.0mg,0.121mmol),(四氢吡喃-4-基)甲基甲磺酸酯(35.2mg,0.181mmol),碳酸钾(33.2mg,0.242mmol)和碘化钾(4.0mg,0.024 mmol)溶解于N,N-二甲基甲酰胺(10mL)中。反应液加热到120℃,搅拌3小时。反应液冷至室温,倒入水(30mL)中,用乙酸乙酯萃取(20mL x 3)。合并有机相,用无水硫酸钠干燥,过滤,滤液浓缩。用制备高效液相色谱分离纯化得到4-甲基-6-((四氢吡喃-4-基)甲基)-1-(2,2,2-三氟乙基)-吡唑并[4,3-d]嘧啶-5,7-二酮(10.0mg),产率:24%。1H NMR:(400MHz,Methonal-d4)δ7.80(s,1H),5.33-5.27(m,2H),3.93-3.89(m,4H),3.49(s,3H),3.36-3.34(m,2H),2.08-2.05(m,1H),1.57-1.53(m,2H),1.44-1.37(m,2H)。MS-ESI计算值[M+H]+347,实测值347。
实施例31
4-甲基-6-(2-(四氢吡喃-4-基)乙基)-1-(2,2,2-三氟乙基)-吡唑并[4,3-d]嘧啶-5,7-二酮
Figure PCTCN2016081103-appb-000062
第一步
4-甲基-6-(2-(四氢吡喃-4-基)乙基)-1-(2,2,2-三氟乙基)-吡唑并[4,3-d]嘧啶-5,7-二酮
将2-四氢吡喃-4-基乙基甲磺酸酯(50.0mg,0.240mmol),4-甲基-1-(2,2,2-三氟乙基)吡唑并[4,3-d]嘧啶-5,7-二酮(59.6mg,0.240mmol),碳酸钾(66.4mg,0.480mmol)和碘化钾(7.9mg,0.048mmol)溶解于N,N-二甲基甲酰胺(5mL)中。反应液加热到120℃,搅拌3小时。反应液冷至室温,倒入水(20mL)中,用乙酸乙酯萃取(20mL x 3)。合并有机相,用无水硫酸钠干燥,过滤,滤液浓缩。粗品用制备高效液相色谱分离纯化得到4-甲基-6-(2-(四氢吡喃-4-基)乙基)-1-(2,2,2-三氟乙基)-吡唑并[4,3-d]嘧啶-5,7-二酮(20.0mg),产率:23%。1H NMR:(400MHz,Methonal-d4)δ7.82(s,1H),5.36-5.27(m,2H),4.08-4.05(m,2H),3.95-3.91(m,2H),3.51(s,3H),3.44-3.41(m,2H),1.76-1.73(m,2H),1.60-1.58(m,3H),1.37-1.30(m,2H)。MS-ESI计算值[M+H]+361,实测值361。
实施例32
6-((4-甲氧基环己基)甲基)-4-甲基-1-(2,2,2-三氟乙基)-吡唑并[4,3-d]嘧啶-5,7-二酮
Figure PCTCN2016081103-appb-000063
第一步
6-((4-甲氧基环己基)甲基)-4-甲基-1-(2,2,2-三氟乙基)-吡唑并[4,3-d]嘧啶-5,7-二酮
将4-甲基-1-(2,2,2-三氟乙基)吡唑并[4,3-d]嘧啶-5,7-二酮(50.0mg,0.201mmol),((4-甲氧基环己基)甲基甲磺酸酯(44.8mg,0.201mmol),碳酸钾(55.7mg,0.403mmol)和碘化钾(6.7mg,0.040mmol,)溶解于N,N-二甲基甲酰胺(5mL)中。反应液加热到120℃,搅拌3小时。反应液冷至室温,倒入水(20mL)中,用乙酸乙酯萃取(20mL x 3)。合并有机相,用无水硫酸钠干燥,过滤,滤液浓缩,用制备高效液相色谱分离纯化得到6-((4-甲氧基环己基)甲基)-4-甲基-1-(2,2,2-三氟乙基)-吡唑并[4,3-d]嘧啶-5,7-二酮(20.0mg),产率:26%。1H NMR:(400MHz,Methonal-d4)δ7.82(s,1H),5.36-5.30(m,2H),3.89(d,J=6.8Hz,2H),3.51(s,3H),3.39(s,3H),3.21-3.15(m,1H),2.09-2.08(m,2H),1.81-1.73(m,3H),1.16-1.10(m,4H)。MS-ESI计算值[M+H]+375,实测值375。
实施例33
1-(环丙基甲基)-4-甲基-6-((3-甲基氧杂环丁烷-3-基)甲基)-吡唑并[4,3-d]嘧啶-5,7-二酮
Figure PCTCN2016081103-appb-000064
第一步
1-(环丙基甲基)-4-硝基-吡唑-5-甲酸甲酯
将4-硝基-吡唑-5-甲酸甲酯(22.0g,129mmol)溶于N,N-二甲基甲酰胺(350mL),0℃下分批加入氢化钠(5.66g,141mmol)。反应在0℃下搅拌1小时,加入碘化钠(21.2g,141mmol),滴加溴甲基环丙烷(19.1g,141mmol)。反应液在25℃下搅拌18小时。向反应液中加入水(1.2L),用乙酸乙酯萃取(300mL x 2)。合并有机相,饱和食盐水洗涤(500mL),用无水硫酸钠干燥,过滤,减压浓缩,剩余物用硅胶柱色谱法分离纯化(5:1石油醚/乙酸乙酯,Rf=0.3),得到1-(环丙基甲基)-4- 硝基-吡唑-5-甲酸甲酯(5.00g,无色油状物),产率:17%。1H NMR:(400MHz,CDCl3)δ8.04(s,1H),4.14(d,J=7.6Hz,2H),4.03(s,3H),1.40-1.23(m,1H),0.75-0.55(m,2H),0.47-0.34(m,2H)。MS-ESI计算值[M+H]+226,实测值226。
第二步
4-氨基-1-(环丙基甲基)-吡唑-5-甲酸甲酯
将1-(环丙基甲基)-4-硝基-吡唑-5-甲酸甲酯(5.00g,22.2mmol)溶于甲醇(70mL),加入干钯碳(钯10%,水1%,500mg),室温下,反应在40psi氢气压力下反应3小时。反应液过滤,滤液减压浓缩得到4-氨基-1-(环丙基甲基)-吡唑-5-甲酸甲酯(4.30g,类白色固体),产率:99%。1H NMR:(400MHz,CDCl3)δ7.11(s,1H),4.27(d,J=7.6Hz,2H),4.11(s,2H),3.91(s,3H),1.46-1.21(m,1H),0.53-0.43(m,2H),0.41-0.32(m,2H)。MS-ESI计算值[M+H]+196,实测值196。
第三步
1-(环丙基甲基)-4-(2,2,2-三氟乙酰氨基)-吡唑-5-甲酸甲酯
将4-氨基-1-(环丙基甲基)-吡唑-5-甲酸甲酯(4.30g,22.0mmol)溶于二氯甲烷(40mL),氮气保护下滴加三氟乙酸酐(6.94g,33.1mmol),反应液室温下搅拌2小时,用饱和碳酸氢钠溶液(50mL)淬灭反应,二氯甲烷(40mL)萃取,饱和食盐水(50mL)洗涤有机相,无水硫酸钠干燥,减压浓缩,得1-(环丙基甲基)-4-(2,2,2-三氟乙酰氨基)-吡唑-5-甲酸甲酯(6.30g,无色油状物),产率:98%。
1H NMR:(400MHz,CDCl3)δ9.72(s,1H),8.28(s,1H),4.37(d,J=7.2Hz,2H),4.09(s,3H),1.39-1.23(m,1H),0.60-0.48(m,2H),0.45-0.37(m,2H)。MS-ESI计算值[M+H]+292,实测值292。
第四步
1-(环丙基甲基)-4-(2,2,2-三氟-N-甲基乙酰氨基)-吡唑-5-甲酸甲酯
将1-(环丙基甲基)-4-(2,2,2-三氟乙酰氨基)-吡唑-5-甲酸甲酯(6.20g,21.3mmol)溶于N,N-二甲基甲酰胺(50mL),加入碳酸钾(4.41g,31.9mmol)。反应液加热至80℃反应1小时。冷却至室温,加入碘甲烷(4.53g,31.9mmol)。反应液室温搅拌18小时。向反应液中加入水(300mL),用乙酸乙酯萃取(100mL x 3)。合并有机相,饱和食盐水洗涤(100mL),无水硫酸钠干燥,过滤,减压浓缩,得1-(环丙基甲基)-4-(2,2,2-三氟-N-甲基乙酰氨基)-吡唑-5-甲酸甲酯(6.44g,黄色油状物),产率:99%。1H NMR:(400MHz,CDCl3)δ7.50(s,1H),4.43(d,J=7.2Hz,2H),3.90(s,3H),3.28(s,3H),1.43-1.27(m,1H),0.60-0.47(m,2H),0.45-0.33(m,2H)。MS-ESI计算值[M+H]+306,实测值306。
第五步
4-[叔丁氧基羰基)(甲基)氨基]1-(环丙基甲基)-吡唑-5-甲酸
将1-(环丙基甲基)-4-(2,2,2-三氟-N-甲基乙酰氨基)-吡唑-5-甲酸甲酯(6.40g,21.0mmol)溶于四氢呋喃(30mL)和水(30mL),加入一水合氢氧化锂(4.40g,105mmol),室温搅拌反应18小时。加入二碳酸二叔丁酯(9.15g,41.9mmol),反应液室温下继续反应16小时,反应液减压浓缩,用2N盐酸水溶液调节pH=4,过滤,干燥滤饼,得到4-((叔丁氧基羰基)(甲基)氨基)-1-(环丙基甲基) -吡唑-5-甲酸(4.50g,白色固体),产率:73%。1H NMR:(400MHz,CDCl3)δ7.46(s,1H),4.38(d,J=6.8Hz,2H),3.21(s,3H),1.58-1.25(m,10H),0.60-0.47(m,2H),0.45-0.37(m,2H)。MS-ESI计算值[M+H]+296,实测值296。
第六步
(5-氨基甲酰基-1-(环丙基甲基)-吡唑-4-基)(甲基)氨基甲酸叔丁酯
将4-[叔丁氧基羰基)(甲基)氨基]1-(环丙基甲基)-吡唑-5-甲酸(3.40g,11.5mmol),2-(7-偶氮苯并三氮唑)-四甲基脲六氟磷酸酯(5.69g,15.0mmol)和氯化铵(1.23g,23.0mmol)溶于二氯甲烷(120mL),室温下滴加三乙胺(1.75g,17.3mmol)。反应液室温搅拌18小时,向反应液中加入水(50mL),用二氯甲烷(500mL x 2)萃取,合并有机相,依次用饱和碳酸氢钠(50mL)和饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩,剩余物用乙醇(20mL),得到(5-氨基甲酰基-1-(环丙基甲基)-吡唑-4-基)(甲基)氨基甲酸叔丁酯(3.00g,粗产品,黄色油状物)。MS-ESI计算值[M+H]+295,实测值295。
第七步
1-(环丙基甲基)-4-(甲基氨基)-吡唑-5-甲酰胺
将(5-氨基甲酰基-1-(环丙基甲基)-吡唑-4-基)(甲基)氨基甲酸叔丁酯(3.30g,11.2mmol)溶于盐酸乙酸乙酯(25mL)。室温下搅拌反应18小时,减压浓缩,剩余物用甲醇(40mL)溶解,加入碳酸钾(3.10g,22.4mmol),室温搅拌2小时。反应液过滤,减压浓缩,剩余物用二氯甲烷(60mL)萃取,过滤,滤液旋干,剩余物用二氯甲烷(15mL)打浆,过滤,得到1-(环丙基甲基)-4-(甲基氨基)-吡唑-5-甲酰胺(1.45g,白色固体),产率:67%。1H NMR:(400MHz,CDCl3)δ7.34(s,2H),7.17(s,1H),4.62-4.47(m,1H),4.21(d,J=6.8Hz,2H),2.65(d,J=5.6Hz,3H),1.22-1.10(m,1H),0.43-0.34(m,2H),0.31-0.23(m,2H)。MS-ESI计算值[M+H]+195,实测值195。
第八步
1-(环丙基甲基)-4-甲基-吡唑并[4,3-d]嘧啶-5,7-二酮
将1-(环丙基甲基)-4-(甲基氨基)-吡唑-5-甲酰胺(1.45g,7.47mmol)溶于N,N-二甲基甲酰胺(10mL),0℃下,分批加入氢化钠(627mg,15.7mmol)。氮气保护下,反应液于0℃搅拌1小时。加入1,1-羰基二咪唑(1.82g,11.2mmol),反应液加热至75℃反应2小时。反应液冷至室温,加入水(80mL)淬灭反应,过滤,烘干滤饼,得到1-(环丙基甲基)-4-甲基-吡唑并[4,3-d]嘧啶-5,7-二酮(1.60g,白色固体),产率:97%。1H NMR:(400MHz,DMSO-d6)δ11.35(s,1H),7.72(s,1H),4.29(d,J=6.8Hz,2H),3.32(s,3H),1.17-1.07(m,1H),0.54-0.32(m,4H)。MS-ESI计算值[M+H]+221,实测值221。
第九步
1-(环丙基甲基)-4-甲基-6-((3-甲基氧杂环丁烷-3-基)甲基)-吡唑并[4,3-d]嘧啶-5,7-二酮
将1-(环丙基甲基)-4-甲基-吡唑并[4,3-d]嘧啶-5,7-二酮(62.1mg,0.282mmol)溶于N,N-二甲基甲酰胺(2mL),加入3-(氯甲基)-3-甲基-氧杂环丁烷(40.8mg,0.338mmol),碳酸钾(78.0mg,0.564mmol)和碘化钾(56.2mg,0.338mmol)。反应液加热到120℃,搅拌1小时。反应液冷至室温, 过滤。滤液减压浓缩,剩余物用高效液相色谱法纯化,得1-(环丙基甲基)-4-甲基-6-((3-甲基氧杂环丁烷-3-基)甲基)-吡唑并[4,3-d]嘧啶-5,7-二酮(46.0mg),产率:54%。1H NMR:(400MHz,CDCl3)δ7.42(s,1H),4.73(d,J=6.4Hz,2H),4.43(d,J=6.8Hz,2H),4.26(d,J=6.4Hz,2H),4.14(s,2H),3.50(s,3H),1.41(s,3H),1.40-1.28(m,1H),0.60-0.52(m,2H),0.51-0.38(m,2H)。MS-ESI计算值[M+H]+305,实测值305。
实施例34
1-(环丙基甲基)-6-((3-乙基氧杂环丁烷-3-基)甲基)-4-甲基-吡唑并[4,3-d]嘧啶-5,7-二酮
Figure PCTCN2016081103-appb-000065
第一步
1-(环丙基甲基)-6-((3-乙基氧杂环丁烷-3-基)甲基)-4-甲基-吡唑并[4,3-d]嘧啶-5,7-二酮
将(3-乙基氧杂环丁烷-3-基)甲基甲磺酸酯(34.4mg,0.177mmol),1-(环丙基甲基)-4-甲基-吡唑并[4,3-d]嘧啶-5,7-二酮(30.0mg,0.136mmol),碘化钾(2.3mg,0.014mmol)和碳酸钾(56.5mg,0.408mmol)溶于N,N-二甲基甲酰胺(3mL)中。反应液升温至120℃,搅拌3小时。冷却至室温,过滤,滤液减压浓缩,剩余物用制备型高效液相色谱纯化得到1-(环丙基甲基)-6-((3-乙基氧杂环丁烷-3-基)甲基)-4-甲基-吡唑并[4,3-d]嘧啶-5,7-二酮(21.0mg),产率:48%。1H NMR:(400MHz,CDCl3)δ7.44(s,1H),4.62(d,J=6.4Hz,2H),4.44(d,J=7.6Hz,2H),4.30(d,J=6.8Hz,2H),4.10(s,2H),3.52(s,3H),1.88-1.83(m,2H),1.37-1.36(m,1H),1.11-1.07(m,3H),0.56-0.46(m,4H)。MS-ESI计算值[M+H]+319,实测值319。
实施例35
1-(环丙基甲基)-4-甲基-6-((四氢呋喃-3-基)甲基)-吡唑并[4,3-d]嘧啶-5,7-二酮
Figure PCTCN2016081103-appb-000066
第一步
1-(环丙基甲基)-4-甲基-6-((四氢呋喃-3-基)甲基)-吡唑并[4,3-d]嘧啶-5,7-二酮
将(四氢呋喃-3-基)甲基甲磺酸酯(53.1mg,0.295mmol),1-(环丙基甲基)-4-甲基-吡唑并[4,3-d]嘧啶-5,7-二酮(50.0mg,0.227mmol)及碳酸钾(94.1mg,0.681mmol)溶于N,N-二甲基甲酰胺(5mL)中,加入碘化钾(3.8mg,0.023mmol),反应液在120℃搅拌3小时。反应液直接过滤,滤液减压浓缩,粗产品用制备高效液相色谱纯化得产物1-(环丙基甲基)-4-甲基-6-((四氢呋喃-3-基)甲基)-吡唑并[4,3-d]嘧啶-5,7-二酮(36.0mg),产率:52%。1H NMR:(400MHz,Methonal-d4)δ7.63(s,1H),4.41(d,J=7.2Hz,2H),3.95-3.90(m,1H),3.87-3.82(m,1H),3.77-3.72(m,1H),3.61-3.54(m,2H),3.45-3.42(m,1H),3.14(s,3H),2.59-2.52(m,1H),1.87-1.79(m,1H),1.62-1.53(m,1H),1.22-1.12(m,1H),0.36-0.25(m,4H)。MS-ESI计算值[M+H]+305,实测值305。
实施例36
1-(环丙基甲基)-4-甲基-6-((四氢吡喃-4-基)甲基)-吡唑并[4,3-d]嘧啶-5,7-二酮
Figure PCTCN2016081103-appb-000067
第一步
1-(环丙基甲基)-4-甲基-6-((四氢吡喃-4-基)甲基)-吡唑并[4,3-d]嘧啶-5,7-二酮
将(四氢吡喃-4-基)甲基甲磺酸酯(34.4mg,0.177mmol),1-(环丙基甲基)-4-甲基-吡唑并[4,3-d]嘧啶-5,7-二酮(30.0mg,0.136mmol),碘化钾(2.3mg,0.014mmol)和碳酸钾(56.5mg,0.408mmol)溶于N,N-二甲基甲酰胺(3mL)中。反应液升温至120℃,搅拌3小时。冷却至室温,过滤,滤液减压浓缩,剩余物用制备型高效液相色谱纯化得到1-(环丙基甲基)-4-甲基-6-((四氢吡喃-4-基)甲基)-吡唑并[4,3-d]嘧啶-5,7-二酮(30.0mg),产率:69%。1H NMR:(400MHz,CDCl3)δ7.42(s,1H),4.45(d,J=7.6Hz,2H),4.00-3.95(m,4H),3.49(s,3H),3.39-3.33(m,2H),2.10-2.06(m,1H),1.48-1.40(m,5H),0.57-0.48(m,4H)。MS-ESI计算值[M+H]+319,实测值319。
实施例37
1-(环丙基甲基)-4-甲基-6-(2-(四氢吡喃-4-基)乙基)-吡唑并[4,3-d]嘧啶-5,7-二酮
Figure PCTCN2016081103-appb-000068
Figure PCTCN2016081103-appb-000069
第一步
1-(环丙基甲基)-4-甲基-6-(2-(四氢吡喃-4-基)乙基)-吡唑并[4,3-d]嘧啶-5,7-二酮
将2-(四氢吡喃-4-基)乙基甲磺酸酯(50.0mg,0.227mmol),1-(环丙基甲基)-4-甲基-吡唑并[4,3-d]嘧啶-5,7-二酮(47.3mg,0.227mmol),碘化钾(3.8mg,0.023mmol)和碳酸钾(62.3mg,0.454mmol)溶于N,N-二甲基甲酰胺(3mL)中。反应液升温至120℃,搅拌3小时。冷却至室温,过滤,滤液减压浓缩,剩余物用制备型高效液相色谱纯化得到1-(环丙基甲基)-4-甲基-6-(2-(四氢吡喃-4-基)乙基)-吡唑并[4,3-d]嘧啶-5,7-二酮(20.0mg),产率:27%。1H NMR:(400MHz,CDCl3)δ7.40(s,1H),4.43(d,J=7.2Hz,2H),4.08-4.05(m,2H),3.98-3.94(m,2H),3.49(s,3H),3.38(t,J=1.6Hz,2H),1.73-1.69(m,2H),1.62-1.59(m,4H),1.39-1.36(m,2H),0.55-0.46(m,4H)。MS-ESI计算值[M+H]+333,实测值333。
实施例38
1-(环丙基甲基)-6-((4-甲氧基环己基)甲基)-4-甲基-吡唑并[4,3-d]嘧啶-5,7-二酮
Figure PCTCN2016081103-appb-000070
第一步
1-(环丙基甲基)-6-((4-甲氧基环己基)甲基)-4-甲基-吡唑并[4,3-d]嘧啶-5,7-二酮
将(4-甲氧基环己基)甲基甲磺酸酯(30.0mg,0.135mmol),1-(环丙基甲基)-4-甲基-吡唑并[4,3-d]嘧啶-5,7-二酮(26.8mg,0.121mmol),碘化钾(2.2mg,0.014mmol)和碳酸钾(37.3mg,0.269mmol)溶于N,N-二甲基甲酰胺(5mL)中。反应液升温至120℃,搅拌3小时。冷却至室温,过滤,滤液减压浓缩,剩余物用制备型高效液相色谱纯化得到1-(环丙基甲基)-6-((4-甲氧基环己基)甲基)-4-甲基-吡唑并[4,3-d]嘧啶-5,7-二酮(16.0mg),产率:34%。1H NMR:(400MHz,Methonal-d4)δ7.64(s,1H),4.42(d,J=6.8Hz,2H),3.89(d,J=6.8Hz,2H),3.50(s,3H),3.35(s,3H),3.20-3.15(m,1H),2.10-2.06(m,2H),1.76-1.73(m,3H),1.38-1.36(m,1H),1.16-1.12(m,4H),0.55-0.46(m,4H)。MS-ESI计算值[M+H]+347,实测值347。
实施例39
1-甲基-3-((3-甲基氧杂环丁烷-3-基)甲基)吡啶并[2,3-d]嘧啶-2,4-二酮
Figure PCTCN2016081103-appb-000071
第一步
2-(甲基氨基)烟腈
将2-氯-3-氰基吡啶(30.0g,216mmol)加入40%甲胺水溶液(300mL)中,加热至80℃搅拌2小时.反应液减压蒸馏浓缩,过滤所得到的固体用水(30mL x 3)洗涤,干燥得2-(甲基氨基)烟腈(22.3g,淡黄色固体),产率:76%。1H NMR:(400MHz,Methonal-d4)δ8.25-8.22(m,1H),7.79-7.74(m,1H),6.65-6.59(m,1H),2.96(s,3H)。
第二步
2-(甲基氨基)吡啶-3-甲酰胺
将2-(甲基氨基)烟腈(600mg,4.51mmol),碳酸钾(1.87mg,0.130mmol),双氧水(0.1mL)溶于二甲基亚砜(10mL)中,室温反应1小时,加入水(10mL)淬灭反应。用乙酸乙酯萃取(10mL x3),无水硫酸钠干燥,过滤,滤液减压浓缩。制备硅胶板板纯化(1:1石油醚/乙酸乙酯,Rf值=0.2),得到2-(甲基氨基)吡啶-3-甲酰胺(500mg,白色固体),产率:73%。1H NMR:(400MHz,DMSO-d6)δ8.45-8.40(br,1H),8.28(d,J=2.0Hz,1H),7.95-7.93(m,2H),7.35-7.30(br,1H),6.53(dd,J=7.6,2.0Hz,1H),3.03(d,J=4.8Hz,3H)。MS-ESI计算值[M+H]+152,实测值152。
第三步
1-甲基吡啶并[2,3-d]嘧啶-2,4(1H,3H)-二酮
将2-(甲基氨基)吡啶-3-甲酰胺(100mg,0.661mmol)和苯基异氰酸酯(157mg,1.32mmol)溶于甲苯(10mL)中,110℃搅拌12小时。加入水(10mL)淬灭反应。过滤得到1-甲基吡啶并[2,3-d]嘧啶-2,4(1H,3H)-二酮(20.0mg,黄色固体),产率:17%。1H NMR:(400MHz,DMSO-d6)δ11.72(s,1H),8.72(d,J=2.0Hz,1H),8.31(d,J=7.6Hz,1H),7.29(dd,J=7.6,2.0Hz,1H),3.48(s,3H)。MS-ESI计算值[M+H]+178,实测值178。
第四步
将1-甲基吡啶并[2,3-d]嘧啶-2,4-二酮(70.0mg,0.395mmol)溶于N,N-二甲基甲酰胺(2mL),加入3-(氯甲基)-3-甲基-氧杂环丁烷(52.4mg,0.435mmol),碳酸钾(109mg,0.790mmol)和碘化 钾(78.7mg,0.474mmol)。反应液加热到120℃,搅拌1小时。反应液冷至室温,过滤。滤液减压浓缩,剩余物用高效液相色谱法纯化,得1-甲基-3-((3-甲基氧杂环丁烷-3-基)甲基)吡啶并[2,3-d]嘧啶-2,4-二酮(44.0mg),产率:43%。1H NMR:(400MHz,CDCl3)δ8.69(d,J=2.0Hz,1H),8.46(d,J=5.6Hz,1H),7.27-7.18(m,1H),4.76(d,J=6.8Hz,2H),4.28(d,J=6.8Hz,2H),4.23(s,2H),3.73(s,3H),1.41(s,3H)。MS-ESI计算值[M+H]+262,实测值262。
实施例40
3-((3-乙基氧杂环丁烷-3-基)甲基)-1-甲基吡啶并[2,3-d]嘧啶-2,4-二酮
Figure PCTCN2016081103-appb-000072
第一步
3-((3-乙基氧杂环丁烷-3-基)甲基)-1-甲基吡啶并[2,3-d]嘧啶-2,4-二酮
将1-甲基吡啶并[2,3-d]嘧啶-2,4-二酮(30.0mg,0.169mmol),(3-乙基氧杂环丁烷-3-基)甲基甲磺酸酯(49.3mg,0.254mmol),碳酸钾(46.8mg,0.338mmol)和碘化钾(5.6mg,0.034mmol)溶解于N,N-二甲基甲酰胺(10mL)中。反应液加热到120℃,搅拌3小时。反应液冷至室温,倒入水(30mL)中,用乙酸乙酯萃取(20mL x 3)。合并有机相,用无水硫酸钠干燥,过滤,滤液浓缩。用制备高效液相色谱分离纯化得到3-((3-乙基氧杂环丁烷-3-基)甲基)-1-甲基吡啶并[2,3-d]嘧啶-2,4-二酮(10.0mg),产率:21%。1H NMR:(400MHz,Methonal-d4)δ8.74-8.73(m,1H),8.48-8.46(m,1H),7.35-7.32(m,1H),4.67(d,J=6.8Hz,2H),4.32(d,J=6.8Hz,2H),4.20(s,2H),3.72(s,3H),1.84-1.79(m,2H),1.08(t,J=7.2Hz,3H)。MS-ESI计算值[M+H]+276,实测值276。
实施例41
1-甲基-3-(四氢呋喃-3-基甲基)吡啶并[2,3-d]嘧啶-2,4-二酮
Figure PCTCN2016081103-appb-000073
第一步
1-甲基-3-(四氢呋喃-3-基甲基)吡啶并[2,3-d]嘧啶-2,4-二酮
将四氢呋喃-3-基甲基甲磺酸酯(30.0mg,0.166mmol),1-甲基吡啶并[2,3-d]嘧啶-2,4-二酮(29.5mg,0.166mmol),碳酸钾(46.0mg,0.333mmol)和碘化钾(5.5mg,0.033mmol,)溶解于N,N-二甲基甲酰胺(5mL)中。反应液加热到120℃,搅拌3小时。反应液冷至室温,倒入水(20mL)中,用乙酸乙酯萃取(20mL x 3)。合并有机相,无水硫酸钠干燥,过滤,滤液浓缩,用制备高效液相色谱分离纯化得到1-甲基-3-(四氢呋喃-3-基甲基)吡啶并[2,3-d]嘧啶-2,4-二酮(10.0mg),产率:23%。1H NMR:(400MHz,Methonal-d4)δ8.65-8.63(m,1H),8.46-8.41(m,1H),7.33-7.30(m,1H),4.14-4.12(m,2H),4.09-4.07(m,1H),3.79-3.75(m,2H),3.69(s,3H),3.65-3.62(m,1H),2.79-2.75(m,1H),2.06-2.01(m,1H),1.78-1.75(m,1H)。MS-ESI计算值[M+H]+262,实测值262。
实施例42
1-甲基-3-((四氢吡喃-4-基)甲基)吡啶并[2,3-d]嘧啶-2,4-二酮
Figure PCTCN2016081103-appb-000074
第一步
1-甲基-3-((四氢吡喃-4-基)甲基)吡啶并[2,3-d]嘧啶-2,4-二酮
将1-甲基吡啶并[2,3-d]嘧啶-2,4-二酮(30.0mg,0.169mmol),四氢吡喃-4-基甲基甲磺酸酯(49.4mg,0.254mmol),碳酸钾(46.8mg,0.338mmol)和碘化钾(5.6mg,0.034mmol)溶解于N,N-二甲基甲酰胺(10mL)中。反应液加热到120℃,搅拌3小时。反应液冷至室温,倒入水(30mL)中,用乙酸乙酯萃取(20mL x 3)。合并有机相,用无水硫酸钠干燥,过滤,滤液浓缩。用制备高效液相色谱分离纯化得到1-甲基-3-((四氢吡喃-4-基)甲基)吡啶并[2,3-d]嘧啶-2,4-二酮(10.0mg,黄色油状),产率:21%。1H NMR:(400MHz,Methonal-d4)δ8.71-8.69(m,1H),8.48-8.40(m,1H),7.33-7.30(m,1H),3.99-3.91(m,4H),3.69(s,3H),3.39-3.33(m,2H),2.13-2.10(m,1H),1.62-1.58(m,2H),1.44-1.40(m,2H)。MS-ESI计算值[M+H]+276,实测值276。
实施例43
1-甲基-3-(2-(四氢吡喃-4-基)乙基)吡啶并[2,3-d]嘧啶-2,4-二酮
Figure PCTCN2016081103-appb-000075
Figure PCTCN2016081103-appb-000076
第一步
1-甲基-3-(2-(四氢吡喃-4-基)乙基)吡啶并[2,3-d]嘧啶-2,4-二酮
将2-四氢吡喃-4-基乙基甲磺酸酯(50.0mg,0.240mmol),1-甲基吡啶并[2,3-d]嘧啶-2,4-二酮(42.5mg,0.240mmol),碳酸钾(66.4mg,0.480mmol)和碘化钾(7.9mg,0.048mmol)溶解于N,N-二甲基甲酰胺(5mL)中。反应液加热到120℃,搅拌3小时。反应液冷至室温,倒入水(20mL)中,用乙酸乙酯萃取(20mL x 3)。合并有机相,用无水硫酸钠干燥,过滤,滤液浓缩,用制备高效液相色谱分离纯化得到1-甲基-3-(2-(四氢吡喃-4-基)乙基)吡啶并[2,3-d]嘧啶-2,4-二酮(20.0mg),产率:29%。1H NMR:(400MHz,Methonal-d4)δ8.72-8.71(m,1H),8.47-8.45(m,1H),7.34-7.31(m,1H),4.14-4.10(m,2H),3.96-3.93(m,2H),3.70(s,3H),3.43-3.40(m,2H),1.78-1.75(m,2H),1.66-1.62(m,3H),1.38-1.32(m,2H)。MS-ESI计算值[M+H]+290,实测值290。
实施例44
3-((4-甲氧基环己基)甲基)-1-甲基吡啶并[2,3-d]嘧啶-2,4-二酮
Figure PCTCN2016081103-appb-000077
第一步
3-((4-甲氧基环己基)甲基)-1-甲基吡啶并[2,3-d]嘧啶-2,4-二酮
将1-甲基吡啶并[2,3-d]嘧啶-2,4-二酮(50.0mg,0.282mmol),(4-甲氧基环己基)甲基甲磺酸酯(62.7mg,0.282mmol),碳酸钾(78.0mg,0.564mmol)和碘化钾(9.4mg,0.056mmol)溶解于N,N-二甲基甲酰胺(5mL)中。反应液加热到120℃,搅拌3小时。反应液冷至室温,倒入水(20mL)中,用乙酸乙酯萃取(20mL x 3)。合并有机相,用无水硫酸钠干燥,过滤,滤液浓缩,用制备高效液相色谱分离纯化得到3-((4-甲氧基环己基)甲基)-1-甲基吡啶并[2,3-d]嘧啶-2,4-二酮(20.0mg),产率:23%。1H NMR:(400MHz,Methonal-d4)δ8.73-8.71(m,1H),8.47-8.45(m,1H),7.34-7.31(m,1H),3.95(d,J=6.8Hz,2H),3.71(s,3H),3.36(s,3H),3.20-3.16(m,1H),2.10-2.08(m,2H),1.84-1.76(m,3H),1.21-1.08(m,4H)。MS-ESI计算值[M+H]+304,实测值304。
实施例45
1-甲基-3-((3-环氧丙烷-3-基)甲基)吡啶并[3,4-d]嘧啶-2,4-二酮
Figure PCTCN2016081103-appb-000078
第一步
3-(甲基氨基)异烟酸
将3-氟异烟酸(3.00g,21.3mmol)溶于二氧六环(6mL),加30%甲胺水溶液(22.0g,213mmol)。反应液升温至140℃,搅拌14小时。加入浓盐酸(12N,3mL),调节pH值至3,过滤,滤饼干燥后得到3-(甲基氨基)异烟酸(3.00g,黄色固体),产率:93%。1H NMR:(400MHz,DMSO-d6)δ8.46(s,1H),7.89(s,1H),7.69(d,J=4.8Hz,1H),7.50(d,J=4.8Hz,1H),2.80(s,3H)。
第二步
3-(甲基氨基)异烟酰胺
将3-(甲基氨基)异烟酸(4.00g,26.3mmol),1-羟基苯并三唑(10.7g,78.9mmol),1-(3-二甲氨基丙基)-3-乙基碳二亚胺(15.1g,78.9mmol)和氯化铵(5.63g,105mmol)溶于N,N-二甲基甲酰胺(5mL)中。反应液25℃下,搅拌24小时。加入水(100mL)淬灭反应。混合物用异丙醇/氯仿(1:3)(50mL x 2)萃取。合并有机相,减压浓缩。剩余物加入二氯甲烷/甲醇(10:1,30mL),搅拌10分钟,过滤,滤饼干燥后得到3-(甲基氨基)异烟酰胺(3.50g,黄色固体),产率:88%。1H NMR:(400MHz,DMSO-d6)δ8.09(s,2H),7.80(d,J=5.2Hz,1H),7.62-7.61(m,1H),7.52-7.48(m,1H),7.43(d,J=5.2Hz,1H),2.84(d,J=5.2Hz,3H)。
第三步
1-甲基吡啶并[3,4-d]嘧啶-2,4-二酮
在0℃条件下,向3-(甲基氨基)异烟酰胺(3.40g,22.5mmol)的N,N-二甲基甲酰胺(50mL)溶液中加入氢化钠(1.80g,45.0mmol)。反应液在0℃搅拌1小时。加入羰基二咪唑(5.47g,33.7mmol)。反应混合物在室温反应1小时。反应液冷却至0℃,加水(20mL)淬灭。有白色固体析出,过滤,滤饼干燥后得得到1-甲基吡啶并[3,4-d]嘧啶-2,4-二酮(3.50g,黄色固体),产率:95%。1H NMR:(400MHz,DMSO-d6)δ11.83(s,1H),8.86(s,1H),8.48(d,J=4.8Hz,1H),7.82(d,J=4.8Hz,1H),3.49(s,3H)。
第四步
1-甲基-3-((3-环氧丙烷-3-基)甲基)吡啶并[3,4-d]嘧啶-2,4-二酮
将3-(氯甲基)-3-甲基-氧杂环丁烷(22.5mg,0.186mmol),1-甲基吡啶并[3,4-d]嘧啶-2,4-二酮(30.0mg,0.169mmol),碘化钾(2.8mg,0.017mmol)和碳酸钾(46.8mg,0.338mmol)溶于N,N-二甲基甲酰胺(5mL)中。反应液升温至120℃,搅拌3小时。冷却至室温,过滤,滤液减压浓缩,剩余物用制备型高效液相色谱纯化得到1-甲基-3-((3-环氧丙烷-3-基)甲基)吡啶并[3,4-d]嘧啶-2,4-二酮(10.0mg),产率:19%。1H NMR:(400MHz,Methonal-d4)δ8.88(s,1H),8.53(d,J=5.2Hz,1H),8.03(d,J=5.2Hz,1H),4.78(d,J=6.4Hz,2H),4.28-4.26(m,4H),3.69(s,3H),1.38(s,3H)。MS-ESI计算值[M+H]+262,实测值262。
实施例46
3-((3-乙基氧杂环丁烷-3-基)甲基)-1-甲基吡啶并[3,4-d]嘧啶-2,4-二酮
Figure PCTCN2016081103-appb-000079
第一步
3-((3-乙基氧杂环丁烷-3-基)甲基)-1-甲基吡啶并[3,4-d]嘧啶-2,4-二酮
将(3-乙基氧杂环丁烷-3-基)甲基甲磺酸酯(36.2mg,0.186mmol),1-甲基吡啶并[3,4-d]嘧啶-2,4-二酮(30.0mg,0.169mmol),碘化钾(2.8mg,0.017mmol)和碳酸钾(46.8mg,0.338mmol)溶于N,N-二甲基甲酰胺(5mL)中。反应液升温至120℃,搅拌3小时。冷却至室温,过滤,滤液减压浓缩,剩余物用制备型高效液相色谱纯化得到3-((3-乙基氧杂环丁烷-3-基)甲基)-1-甲基吡啶并[3,4-d]嘧啶-2,4-二酮(11.0mg),产率:22%。1H NMR:(400MHz,CDCl3)δ8.79(s,1H),8.60(d,J=4.8Hz,1H),8.01(d,J=4.8Hz,1H),4.62(d,J=6.8Hz,2H),4.32(d,J=6.4Hz,2H),4.20(s,2H),3.71(s,3H),1.85-1.79(m,2H),1.10-1.07(m,3H)。MS-ESI计算值[M+H]+276,实测值276。
实施例47
1-甲基-3-((四氢呋喃-3-基)甲基)吡啶并[3,4-d]嘧啶-2,4-二酮
Figure PCTCN2016081103-appb-000080
第一步
1-甲基-3-((四氢呋喃-3-基)甲基)吡啶并[3,4-d]嘧啶-2,4-二酮
将3-(四氢呋喃-3-基)甲基甲磺酸酯(33.6mg,0.186mmol),1-甲基吡啶并[3,4-d]嘧啶-2,4-二酮(30.0mg,0.169mmol),碘化钾(2.8mg,0.017mmol)和碳酸钾(46.8mg,0.338mmol)溶于N,N-二甲基甲酰胺(5mL)中。反应液升温至120℃,搅拌3小时。冷却至室温,过滤,滤液减压浓缩,剩余物用制备型高效液相色谱纯化得到1-甲基-3-((四氢呋喃-3-基)甲基)吡啶并[3,4-d]嘧啶-2,4-二酮(21.0mg),产率:45%。1H NMR:(400MHz,CDCl3)δ8.78(s,1H),8.60(d,J=4.8Hz,1H),8.03(d,J=4.8Hz,1H),4.23-4.21(m,2H),3.85-3.81(m,3H),3.71(s,3H),3.66-3.65(m,1H),2.81-2.77(m,1H),2.05-2.00(m,1H),1.81-1.77(m,1H)。MS-ESI计算值[M+H]+262,实测值262。
实施例48
1-甲基-3-((四氢吡喃-4-基)甲基)吡啶并[3,4-d]嘧啶-2,4-二酮
Figure PCTCN2016081103-appb-000081
第一步
1-甲基-3-((四氢吡喃-4-基)甲基)吡啶并[3,4-d]嘧啶-2,4-二酮
将(四氢吡喃-4-基)甲基甲磺酸酯(36.2mg,0.186mmol),1-甲基吡啶并[3,4-d]嘧啶-2,4-二酮(30.0mg,0.169mmol),碘化钾(2.8mg,0.017mmol)和碳酸钾(46.8mg,0.338mmol)溶于N,N-二甲基甲酰胺(5mL)中。反应液升温至120℃,搅拌3小时。冷却至室温,过滤,滤液减压浓缩,剩余物用制备型高效液相色谱纯化得到1-甲基-3-((四氢吡喃-4-基)甲基)吡啶并[3,4-d]嘧啶-2,4-二酮(10.0mg),产率:21%。1H NMR:(400MHz,CDCl3)δ8.76(s,1H),8.58(d,J=4.8Hz,1H),8.01(d,J=4.8Hz,1H),4.03(d,J=7.2Hz,2H),3.99-3.95(m,2H),3.70(s,3H),3.37-3.18(m,2H),2.13-2.09(m,1H),1.57-1.48(m,4H)。MS-ESI计算值[M+H]+276,实测值276。
实施例49
1-甲基-3-(2-(四氢吡喃-4-基)乙基)吡啶并[3,4-d]嘧啶-2,4-二酮
Figure PCTCN2016081103-appb-000082
Figure PCTCN2016081103-appb-000083
第一步
1-甲基-3-(2-(四氢吡喃-4-基)乙基)吡啶并[3,4-d]嘧啶-2,4-二酮
将2-(四氢吡喃-4-基)乙基甲磺酸脂(38.8mg,0.186mmol),1-甲基吡啶并[3,4-d]嘧啶-2,4-二酮(30.0mg,0.169mmol),碘化钾(2.8mg,0.017mmol)和碳酸钾(46.8mg,0.338mmol)溶于N,N-二甲基甲酰胺(5mL)中。反应液升温至120℃,搅拌3小时。冷却至室温,过滤,滤液减压浓缩,剩余物用制备型高效液相色谱纯化得到1-甲基-3-(2-(四氢吡喃-4-基)乙基)吡啶并[3,4-d]嘧啶-2,4-二酮(10.0mg),产率:20%。1H NMR:(400MHz,CDCl3)δ9.17(s,1H),8.62-8.60(m,1H),8.43-8.41(m,1H),4.16-4.13(m,2H),3.99-3.95(m,2H),3.77(s,3H),3.42-3.36(m,2H),1.72-1.64(m,5H),1.43-1.34(m,2H)。MS-ESI计算值[M+H]+290,实测值290。
实施例50
3-((4-甲氧基环己基)甲基)-1-甲基吡啶并[3,4-d]嘧啶-2,4-二酮
Figure PCTCN2016081103-appb-000084
第一步
3-((4-甲氧基环己基)甲基)-1-甲基吡啶并[3,4-d]嘧啶-2,4-二酮
将(4-甲氧基环己基)甲基甲磺酸酯(41.4mg,0.186mmol),1-甲基吡啶并[3,4-d]嘧啶-2,4-二酮(30.0mg,0.169mmol),碘化钾(2.8mg,0.017mmol)和碳酸钾(46.8mg,0.338mmol)溶于N,N-二甲基甲酰胺(5mL)中。反应液升温至120℃,搅拌3小时。冷却至室温,过滤,滤液减压浓缩,剩余物用制备型高效液相色谱纯化得到3-((4-甲氧基环己基)甲基)-1-甲基吡啶并[3,4-d]嘧啶-2,4-二酮(10.0mg),产率:19%。1H NMR:(400MHz,CDCl3)δ8.76(s,1H),8.58(d,J=4.8Hz,1H),8.01(d,J=4.8Hz,1H),3.99(d,J=7.2Hz,2H),3.69(s,3H),3.35(s,3H),3.15-3.11(m,1H),2.08-1.97(m,2H),1.86-1.82(m,3H),1.23-1.10(m,4H)。MS-ESI计算值[M+H]+304,实测值304。
实施例51
1-甲基-3-((3-甲基吡啶-3-基)甲基)吡啶并[4,3-d]嘧啶-2,4-二酮
Figure PCTCN2016081103-appb-000085
第一步
4-(甲基氨基)烟酸
将4-氯烟酸(7.00g,44.3mmol)溶于二氧六环(14mL),加30%甲胺水溶液(55.2g,444mmol)。反应液在微波中升温至100℃,搅拌50分钟。加入盐酸水溶液(4N,5mL),调节PH值至3,过滤,滤饼干燥后得到4-(甲基氨基)烟酸(5.00g,白色固体),产率:74%。1H NMR:(400MHz,DMSO-d6)δ8.52(s,1H),8.13(d,J=6.8Hz,1H),6.78(d,J=6.8Hz,1H),2.95(d,J=4.4Hz,3H)。
第二步
4-(甲基氨基)烟酰胺
将4-(甲基氨基)烟酸(5.20g,34.2mmol),1-羟基苯并三唑(27.7g,205mmol),1-乙基-(3-二甲基氨基丙基)碳酰二亚胺盐酸盐(39.3g,205mmol)和氯化铵(14.6g,273mmol)溶于N,N-二甲基甲酰胺(50mL)中。反应液25℃下,搅拌8小时。加入水(100mL)淬灭反应。混合物用异丙醇/氯仿(1:3)(30mL x 5)萃取。合并有机相,减压浓缩。剩余物加入二氯甲烷/甲醇(10:1,50mL),搅拌10分钟,过滤,滤饼干燥后得到4-(甲基氨基)烟酰胺(4.70g,白色固体),产率:91%。1H NMR:(400MHz,DMSO-d6)δ9.67(d,J=7.6Hz,1H),8.77(s,1H),8.52(s,1H),8.30-8.28(m,1H),7.87(s,1H),7.01(d,J=7.6Hz,1H),3.01(s,3H)。
第三步
1-甲基吡啶并[4,3-d]嘧啶-2,4-二酮
在0℃条件下,向4-(甲基氨基)烟酰胺(4.80g,31.8mmol)的N,N-二甲基甲酰胺(50mL)溶液中加入氢化钠(1.52g,63.5mmol)。反应液在0℃搅拌1小时。加入羰基二咪唑(7.72g,47.6mmol)。反应混合物在75℃反应2小时。反应液冷却至室温,加水(50mL)淬灭。加入盐酸水溶液(12N,5mL),调节PH值至3,有白色固体析出,过滤,滤饼干燥后得得到1-甲基吡啶并[4,3-d]嘧啶-2,4-二酮(3.50g,黄色固体),产率:95%。1H NMR:(400MHz,DMSO-d6)δ11.76(s,1H),8.97(s,1H),8.69(d,J=6.0Hz,1H),7.38(d,J=6.0Hz,1H),3.39(s,3H)。
第四步
1-甲基-3-((3-甲基吡啶-3-基)甲基)吡啶并[4,3-d]嘧啶-2,4-二酮
将3-(氯甲基)-3-甲基氧杂环丁烷(26.5mg,0.220mmol),1-甲基吡啶并[4,3-d]嘧啶-2,4-二酮(30.0mg,0.169mmol)及碳酸钾(70.2mg,0.508mmol)溶于N,N-二甲基甲酰胺(3mL)中,加入碘化钾(2.8mg,0.017mmol),反应液在120℃搅拌3小时。反应液直接过滤,滤液减压浓缩,粗产品用制备高效液相色谱纯化得产物1-甲基-3-((3-甲基吡啶-3-基)甲基)吡啶并[4,3-d]嘧啶-2,4-二酮(22.0mg),产率:48%。1H NMR:(400MHz,Methonal-d4)δ9.14(s,1H),8.71(d,J=6.4Hz,1H),7.44(d,J=6.4Hz,1H),4.79(d,J=6.0Hz,2H),4.27(d,J=6.0Hz,2H),4.25(s,2H),3.61(s,3H),1.38(s,3H)。MS-ESI计算值[M+H]+262,实测值262。
实施例52
3-((3-乙基氧杂环丁烷-3-基)甲基)-1-甲基吡啶并[4,3-d]嘧啶-2,4-二酮
Figure PCTCN2016081103-appb-000086
第一步
3-((3-乙基氧杂环丁烷-3-基)甲基)-1-甲基吡啶并[4,3-d]嘧啶-2,4-二酮
将(3-乙基氧杂环丁烷-3-基)甲基甲磺酸酯(42.7mg,0.220mmol),1-甲基吡啶并[4,3-d]嘧啶-2,4-二酮(30.0mg,0.169mmol)及碳酸钾(70.2mg,0.508mmol)溶于N,N-二甲基甲酰胺(3mL)中,加入碘化钾(2.8mg,0.017mmol),反应液在120℃搅拌3小时。反应液直接过滤,滤液减压浓缩,粗产品用制备高效液相色谱纯化得产物3-((3-乙基氧杂环丁烷-3-基)甲基)-1-甲基吡啶并[4,3-d]嘧啶-2,4-二酮(20.0mg),产率:42%。1H NMR:(400MHz,CDCl3)δ9.29(s,1H),8.75(d,J=6.0Hz,1H),7.09(d,J=6.0Hz,1H),4.61(d,J=6.4Hz,2H),4.31(d,J=6.4Hz,2H),4.17(s,2H),3.59(s,3H),1.83-1.78(q,J=7.6Hz,2H),1.07(t,J=7.6Hz,3H)。MS-ESI计算值[M+H]+276,实测值276。
实施例53
1-甲基-3-((四氢呋喃-3-基)甲基)吡啶并[4,3-d]嘧啶-2,4-二酮
Figure PCTCN2016081103-appb-000087
Figure PCTCN2016081103-appb-000088
第一步
1-甲基-3-((四氢呋喃-3-基)甲基)吡啶并[4,3-d]嘧啶-2,4-二酮
将(四氢呋喃-3-基)甲基甲磺酸酯(39.6mg,0.220mmol),1-甲基吡啶并[4,3-d]嘧啶-2,4-二酮(30.0mg,0.169mmol)及碳酸钾(70.2mg,0.508mmol)溶于N,N-二甲基甲酰胺(3mL)中,加入碘化钾(2.8mg,0.017mmol),反应液在120℃搅拌3小时。反应液直接过滤,滤液减压浓缩,粗产品用制备高效液相色谱纯化得产物1-甲基-3-((四氢呋喃-3-基)甲基)吡啶并[4,3-d]嘧啶-2,4-二酮(20.0mg),产率:43%。1H NMR:(400MHz,Methonal-d4)δ9.11(s,1H),8.68(d,J=6.0Hz,1H),7.42(d,J=6.0Hz,1H),4.17-4.04(m,2H),3.96-3.91(m,1H),3.81-3.73(m,2H),3.65-3.61(m,1H),3.60(s,3H),2.81-2.71(m,1H),2.08-2.00(m,1H),1.81-1.73(m,1H)。MS-ESI计算值[M+H]+262,实测值262。
实施例54
1-甲基-3-((四氢吡喃-4-基)甲基)吡啶并[4,3-d]嘧啶-2,4-二酮
Figure PCTCN2016081103-appb-000089
第一步
1-甲基-3-((四氢吡喃-4-基)甲基)吡啶并[4,3-d]嘧啶-2,4-二酮
将(四氢吡喃-4-基)甲基甲磺酸酯(42.7mg,0.220mmol),1-甲基吡啶并[4,3-d]嘧啶-2,4-二酮(30.0mg,0.169mmol)及碳酸钾(70.2mg,0.508mmol)溶于N,N-二甲基甲酰胺(3mL)中,加入碘化钾(2.8mg,0.017mmol),反应液在120℃搅拌3小时。反应液直接过滤,滤液减压浓缩,粗产品用制备高效液相色谱纯化得产物1-甲基-3-((四氢吡喃-4-基)甲基)吡啶并[4,3-d]嘧啶-2,4-二酮(19.0mg),产率:40%。1H NMR:(400MHz,Methonal-d4)δ9.12(s,1H),8.69(d,J=6.0Hz,1H),7.42(d,J=6.0Hz,1H),3.99(d,J=7.2Hz,2H),3.96-3.92(m,2H),3.60(s,3H),3.37-3.33(m,2H),2.13-2.11(m,1H),1.63-1.59(m,2H),1.47-1.37(m,2H)。MS-ESI计算值[M+H]+276,实测值276。
实施例55
1-甲基-3-(2-(四氢吡喃-4-基)乙基)吡啶并[4,3-d]嘧啶-2,4-二酮
Figure PCTCN2016081103-appb-000090
第一步
1-甲基-3-(2-(四氢吡喃-4-基)乙基)吡啶并[4,3-d]嘧啶-2,4-二酮
将2-(四氢吡喃-4-基)乙基甲磺酸酯(35.2mg,0.169mmol),1-甲基吡啶并[4,3-d]嘧啶-2,4-二酮(30.0mg,0.169mmol)及碳酸钾(70.2mg,0.508mmol)溶于N,N-二甲基甲酰胺(3mL)中,加入碘化钾(2.8mg,0.017mmol),反应液在120℃搅拌3小时。反应液直接过滤,滤液减压浓缩,粗产品用制备高效液相色谱纯化得产物1-甲基-3-(2-(四氢吡喃-4-基)乙基)吡啶并[4,3-d]嘧啶-2,4-二酮(25.0mg),产率:51%。1H NMR:(400MHz,Methonal-d4)δ9.12(s,1H),8.69(d,J=6.4Hz,1H),7.43(d,J=6.4Hz,1H),4.10(t,J=7.4Hz,2H),3.96-3.92(m,2H),3.60(s,3H),3.46-3.39(m,2H),1.75(d,J=12.8Hz,2H),1.66-1.61(m,3H),1.37-1.31(m,2H)。MS-ESI计算值[M+H]+290,实测值290。
实施例56
3-((4-甲氧基环己基)甲基)-1-甲基吡啶并[4,3-d]嘧啶-2,4-二酮
Figure PCTCN2016081103-appb-000091
第一步
3-((4-甲氧基环己基)甲基)-1-甲基吡啶并[4,3-d]嘧啶-2,4-二酮
将(4-甲氧基环己基)甲基甲磺酸酯(37.6mg,0.169mmol),1-甲基吡啶并[4,3-d]嘧啶-2,4-二酮(30.0mg,0.169mmol)及碳酸钾(70.2mg,0.508mmol)溶于N,N-二甲基甲酰胺(3mL)中,加入碘化钾(2.8mg,0.017mmol),反应液在120℃搅拌3小时。反应液直接过滤,滤液减压浓缩,粗产品用制备高效液相色谱纯化得产物3-((4-甲氧基环己基)甲基)-1-甲基吡啶并[4,3-d]嘧啶-2,4-二酮(20.0mg),产率:37%。1H NMR:(400MHz,Methonal-d4)δ9.14(s,1H),8.69(d,J=6.0Hz,1H),7.43(d,J=6.0Hz,1H),3.95(d,J=6.8Hz,2H),3.64(s,3H),3.35(s,3H),3.22-3.16(m,1H),2.09(d,J=8.8Hz,2H),1.85-1.77(m,3H),1.18-1.12(m,4H)。MS-ESI计算值[M+H]+304, 实测值304。
实施例57
1-甲基-3-[(3-甲基吡啶-3-基)甲基]吡啶并[3,2-d]嘧啶-2,4-二酮
Figure PCTCN2016081103-appb-000092
第一步
N-(2-氯-3-吡啶基)氨基甲酸叔丁脂
将2-氯吡啶-3-胺(30.0g,233mmol)溶于二氯甲烷(250mL)中并加入三乙胺(47.2g,467mmol)。0℃下滴加二碳酸二叔丁酯(102g,467mmol)。反应液在25℃下搅拌18小时。加入水(100mL)淬灭反应。反应液用乙酸乙酯(100mL x 3)萃取。合并有机相,用无水硫酸钠干燥,过滤,减压浓缩,剩余物用硅胶柱色谱法分离纯化(15:1石油醚/乙酸乙酯,Rf=0.6),得到N-(2-氯-3-吡啶基)氨基甲酸叔丁脂(11.0g,白色固体),产率:21%。1H NMR:(400MHz,DMSO-d6)δ8.89(s,1H),8.17-8.16(m,1H),8.03-8.01(m,1H),7.43-7.39(m,1H),1.47(s,9H)。
第二步
(2-氯吡啶-3-基)(甲基)氨基甲酸叔丁脂
将N-(2-氯-3-吡啶基)氨基甲酸叔丁脂(11.0g,48.1mmol)溶于无水四氢呋喃(150mL)中,在氮气保护,0℃时缓慢加入钠氢(1.39g,57.7mmol),反应液在0℃搅拌半小时。缓慢加入碘甲烷(10.2g,72.2mmol),室温搅拌12小时。加入水(50mL)淬灭反应。反应液用乙酸乙酯(80mL x 3)萃取,合并有机相,饱和食盐水(150mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得到(2-氯吡啶-3-基)(甲基)氨基甲酸叔丁脂(11.0g,无色油状物),产率:94%。1H NMR:(400Hz,DMSO-d6)δ8.33(d,J=4.8Hz,1H),7.92-7.90(m,1H),7.48(d,J=8.0Hz,1H),3.06(s,3H),1.45-1.14(m,9H)。MS-ESI计算值[M+H]+243,实测值243。
第三步
2-氯-N-甲基吡啶-3-胺
将(2-氯吡啶-3-基)(甲基)氨基甲酸叔丁脂(11.0g,45.3mmol)溶于乙酸乙酯(50mL),0℃下滴加4M盐酸乙酸乙酯(150mL)后于25℃搅拌15小时。反应液减压浓缩。用硅胶柱色谱法纯化(10:1石油醚/乙酸乙酯,Rf=0.3)得到2-氯-N-甲基吡啶-3-胺(5.50g,无色油状物),产率:85%。1H NMR:(400Hz,DMSO-d6)δ7.56(d,J=4.8Hz,1H),7.20-7.17(m,1H),6.95(d,J=8.0Hz,1H),5.76-5.73(m,1H),2.73(d,J=4.8Hz,3H)。MS-ESI计算值[M+H]+143,实测值143。
第四步
3-(甲基氨基)吡啶甲酸甲酯
将2-氯-N-甲基吡啶-3-胺(5.50g,38.6mmol)溶于甲醇(100mL)中,反应液于25℃条件下加入1,1'-双(二苯基磷)二茂铁氯化钯(2.82g,3.86mmol)。在50℃下,反应液于一氧化碳氛围中(50psi),反应56小时,反应液冷却至25℃,减压浓缩,用硅胶柱层析法分离纯化(5:1石油醚/乙酸乙酯,Rf=0.5)得到3-(甲基氨基)吡啶甲酸甲酯(6.00g,无色油状物),产率:94%。1H NMR:(400MHz,Methonal-d4)δ7.84(d,J=4.8Hz,1H),7.45-7.42(m,1H),7.29(d,J=8.0Hz,1H),3.93(s,3H),2.94(s,3H)。MS-ESI计算值[M+H]+167,实测值167。
第五步
3-(甲基氨基)吡啶-2-甲酰胺
将3-(甲基氨基)吡啶-2-羧酸甲酯(6.00g,36.1mmol)溶于甲醇(100mL),并加入氨水(1.27g,36.1mmol)。反应液在40℃下搅拌18小时。反应液加入水(200mL),用乙酸乙酯萃取(100mL x 2)。合并有机相,用无水硫酸钠干燥,过滤,减压浓缩得3-(甲基氨基)吡啶-2-甲酰胺(3.50g,黄色固体),产率:64%。1H NMR:(400MHz,DMSO-d6)δ8.27-8.23(br,1H),7.80-7.95(br,1H),7.76(d,J=4.0Hz,1H),7.37-7.32(m,2H),7.11(d,J=8.0Hz,1H),2.79(d,J=4.8Hz,3H)。
第六步
N-[(2-甲酰氨基-3-吡啶基)-N-甲基]-氨基甲酸乙酯
将3-(甲基氨基)吡啶-2-甲酰胺(1.70g,10.9mmol)溶于氯甲酸乙酯(35.3g,326mmol),反应液在90℃下搅拌1小时。反应液用饱和碳酸氢钠水溶液(20mL)淬灭,乙酸乙酯萃取(20mL x 2)。有机相减压浓缩,剩余物用硅胶柱色谱法分离纯化(1:1石油醚/乙酸乙酯,Rf=0.2),得N-[(2-甲酰氨基-3-吡啶基)-N-甲基]-氨基甲酸乙酯(2.00g,白色固体),产率:83%。1H NMR:(400MHz,DMSO-d6)δ8.48(d,J=4.0Hz,1H),7.90(br,1H),7.82(d,J=8.0Hz,1H),7.60-7.56(m,1H),7.50(br,1H),3.88(q,J=7.2Hz,2H),3.12(s,3H),1.00(t,J=7.2Hz,3H)。
第七步
1-甲基吡啶并[3,2-d]嘧啶-2,4-二酮
将N-[(2-甲酰氨基-3-吡啶基)-N-甲基]-氨基甲酸乙酯(2.00g,8.96mmol)和氢氧化钠(717mg,17.9mmol)溶于甲苯(25mL),反应液在110℃下搅拌0.5h。反应液加入水(15mL)稀释,用1N盐酸水溶液调节pH=7。过滤,滤饼用甲醇(15mL)稀释,减压浓缩得1-甲基吡啶并[3,2-d]嘧啶-2,4-二酮(1.09g,白色固体),产率:69%。1H NMR:(400MHz,DMSO-d6)δ11.72(s,1H),8.50(d, J=4.8Hz,1H),7.89(dd,J=8.0,4.8Hz,1H),7.74-7.71(m,1H),3.41(s,3H)。
第八步
1-甲基-3-[(3-甲基吡啶-3-基)甲基]吡啶并[3,2-d]嘧啶-2,4-二酮
将1-甲基吡啶并[3,2-d]嘧啶-2,4-二酮(30.0mg,0.169mmol),3-(氯甲基)-3-甲基氧杂环丁烷(26.5mg,0.220mmol)及碳酸钾(58.5mg,0.424mmol)溶于N,N-二甲基甲酰胺(4mL)中,加入碘化钾(2.8mg,0.017mmmol),反应120℃加热回流3小时。反应液直接过滤,滤液减压浓缩,粗产品用制备高效液相色谱纯化得产物1-甲基-3-[(3-甲基吡啶-3-基)甲基]吡啶并[3,2-d]嘧啶-2,4-二酮(12.0mg),产率:27%。1H NMR:(400MHz,Methonal-d4)δ8.57(d,J=4.8Hz,1H),8.00(d,J=8.0Hz,1H),7.82-7.77(m,1H),4.80(d,J=6.0Hz,2H),4.31-4.25(m,4H),3.64(s,3H),1.40(s,3H)。MS-ESI计算值[M+H]+262,实测值262。
实施例58
3-[(3-乙基氧杂环丁烷-3-基)甲基]-1-甲基吡啶并[3,2-d]嘧啶-2,4-二酮
Figure PCTCN2016081103-appb-000093
第一步
3-[(3-乙基氧杂环丁烷-3-基)甲基]-1-甲基吡啶并[3,2-d]嘧啶-2,4-二酮
将1-甲基吡啶并[3,2-d]嘧啶-2,4-二酮(30.0mg,169mmol),(3-乙基氧杂环丁烷-3-基)甲基甲磺酸酯(42.8mg,220mmol)及碳酸钾(70.2mg,508mmol)溶于N,N-二甲基甲酰胺(4mL)中,加入碘化钾(2.80mg,0.017mmol),反应120℃加热回流3小时。反应液直接过滤,滤液减压浓缩,粗产品用制备高效液相色谱纯化得产物3-[(3-乙基氧杂环丁烷-3-基)甲基]-1-甲基吡啶并[3,2-d]嘧啶-2,4-二酮(16.0mg),产率:34%。1H NMR:(400MHz,Methonal-d4)δ8.57(d,J=4.4Hz,1H),8.00(d,J=4.4Hz,1H),7.83-7.79(m,1H),4.69(d,J=6.8Hz,2H),4.33(d,J=6.8Hz,2H),4.25(s,2H),3.65(s,3H),1.86-1.80(m,2H),1.09(t,J=7.4Hz,3H)。MS-ESI计算值[M+H]+276,实测值276。
实施例59
1-甲基-3-((四氢呋喃-3-基)甲基)吡啶并[3,2-d]嘧啶-2,4-二酮
Figure PCTCN2016081103-appb-000094
Figure PCTCN2016081103-appb-000095
第一步
1-甲基-3-((四氢呋喃-3-基)甲基)吡啶并[3,2-d]嘧啶-2,4-二酮
将(四氢呋喃-3-基)甲基甲磺酸酯(30.5mg,0.169mmol),1-甲基吡啶并[3,2-d]嘧啶-2,4-二酮(25.0mg,0.141mmol),碘化钾(2.3mg,0.014mmol)和碳酸钾(39.0mg,0.282mmol)溶于N,N-二甲基甲酰胺(3mL)中。反应液升温至120℃,搅拌3小时。冷却至室温,过滤,滤液减压浓缩,剩余物用制备型高效液相色谱纯化得到1-甲基-3-((四氢呋喃-3-基)甲基)吡啶并[3,2-d]嘧啶-2,4-二酮(20.0mg),产率:54%。1H NMR:(400MHz,Methonal-d4)δ8.56(d,J=4.4Hz,1H),7.98(d,J=8.0Hz,1H),7.81-7.77(m,1H),4.21-4.14(m,2H),3.96-3.90(m,1H),3.81-3.78(m,2H),3.67-3.63(m,4H),2.82-2.79(m,1H),2.06-2.03(m,1H),1.83-1.79(m,1H)。MS-ESI计算值[M+H]+262,实测值262。
实施例60
1-甲基-3-((四氢吡喃-4-基)甲基)吡啶并[3,2-d]嘧啶-2,4-二酮
Figure PCTCN2016081103-appb-000096
第一步
1-甲基-3-((四氢吡喃-4-基)甲基)吡啶并[3,2-d]嘧啶-2,4-二酮
将(四氢吡喃-4-基)甲基甲磺酸酯(39.5mg,0.203mmol),1-甲基吡啶并[3,2-d]嘧啶-2,4-二酮(30.0mg,0.169mmol),碘化钾(2.8mg,0.017mmol)和碳酸钾(46.8mg,0.339mmol)溶于N,N-二甲基甲酰胺(3mL)中。反应液升温至120℃,搅拌3小时。冷却至室温,过滤,滤液减压浓缩,剩余物用制备型高效液相色谱纯化得到1-甲基-3-((四氢吡喃-4-基)甲基)吡啶并[3,2-d]嘧啶-2,4-二酮(20.0mg),产率:43%。1H NMR:(400MHz,Methonal-d4)δ8.56(d,J=3.6Hz,1H),7.98(d,J=8.0Hz,1H),7.81-7.77(m,1H),4.06-4.04(m,2H),3.96-3.93(m,2H),3.63(s,3H),3.38-3.33(m,2H),2.17-2.14(m,1H),1.64-1.61(m,2H),1.49-1.44(m,2H)。MS-ESI计算值[M+H]+276,实测值276。
实施例61
1-甲基-3-(2-(四氢吡喃-4-基)乙基)吡啶并[3,2-d]嘧啶-2,4-二酮
Figure PCTCN2016081103-appb-000097
第一步
1-甲基-3-(2-(四氢吡喃-4-基)乙基)吡啶并[3,2-d]嘧啶-2,4-二酮
将2-(四氢吡喃-4-基)乙基甲磺酸酯(35.3mg,0.169mmol),1-甲基吡啶并[3,2-d]嘧啶-2,4-二酮(25.0mg,0.141mmol),碘化钾(2.3mg,0.014mmol)和碳酸钾(39.0mg,0.282mmol)溶于N,N-二甲基甲酰胺(3mL)中。反应液升温至120℃,搅拌3小时。冷却至室温,过滤,滤液减压浓缩,剩余物用制备型高效液相色谱纯化得到1-甲基-3-(2-(四氢吡喃-4-基)乙基)吡啶并[3,2-d]嘧啶-2,4-二酮(20.0mg),产率:49%。1H NMR:(400MHz,Methonal-d4)δ8.55(d,J=4.0Hz,1H),7.99(d,J=8.0Hz,1H),7.78(d,J=8.0,4.0Hz,1H),4.19-4.15(m,2H),3.95-3.93(m,2H),3.63(s,3H),3.47-3.40(m,2H),1.79-1.76(m,2H),1.68-1.64(m,3H),1.37-1.33(m,2H)。MS-ESI计算值[M+H]+290,实测值290。
实施例62
3-((4-甲氧基环己基)甲基)-1-甲基吡啶并[3,2-d]嘧啶-2,4-二酮
Figure PCTCN2016081103-appb-000098
第一步
3-((4-甲氧基环己基)甲基)-1-甲基吡啶并[3,2-d]嘧啶-2,4-二酮
将(4-甲氧基环己基)甲基甲磺酸酯(37.7mg,0.169mmol),1-甲基吡啶并[3,2-d]嘧啶-2,4-二酮(25.0mg,0.141mmol),碘化钾(2.3mg,0.014mmol)和碳酸钾(39.0mg,0.282mmol)溶于N,N-二甲基甲酰胺(3mL)中。反应液升温至120℃,搅拌3小时。冷却至室温,过滤,滤液减压浓缩,剩余物用制备型高效液相色谱纯化得到3-((4-甲氧基环己基)甲基)-1-甲基吡啶并[3,2-d]嘧啶-2,4-二酮(20.0mg),产率:47%。1H NMR:(400MHz,Methonal-d4)δ8.55(d,J=4.0Hz,1H),7.97(d,J=8.4Hz,1H),7.80-7.77(m,1H),3.99-3.97(m,2H),3.62(s,3H),3.33(s,3H),3.18-3.15(m,1H),2.09-2.06(m,2H),1.86-1.76(m,3H),1.19-1.10(m,4H)。MS-ESI计 算值[M+H]+304,实测值304。
实施例63
1-甲基-3-((3-甲基氧杂环丁烷-3-基)甲基)蝶啶-2,4-二酮
Figure PCTCN2016081103-appb-000099
第一步
6-氨基-1-甲基嘧啶-2,4-二酮
在25℃下,将金属钠(7.80g,340mmol)分批加入到搅拌的无水乙醇(180mL)中,加热到80℃回流0.5小时。然后分批加入甲基脲(12.6g,170mmol),继续回流0.5小时。反应液滴加氰乙酸乙酯(19.0g,170mmol),有大量沉淀产生。继续回流3小时,减压回收乙醇。剩余物加水(50mL)溶解,用稀盐酸(1N)调节pH=7,过滤得到产物6-氨基-1-甲基嘧啶-2,4-二酮(7.60g,白色固体),产率:32%。1H NMR:(400MHz,DMSO-d6)δ10.39(s,1H),6.79(s,2H),4.54(s,1H),3.14(s,3H)。MS-ESI计算值[M+H]+142,实测值142。
第二步
5,6-二氨基-1-甲基嘧啶-2,4-二酮
将6-氨基-1-甲基嘧啶-2,4-二酮(10.0g,70.1mmol)溶于水(100mL)中,在0℃搅拌下滴加盐酸(7mL,84.0mmol,12N)。再将亚硝酸钠(5.80g,84.2mmol)溶于水(50mL)滴加到反应物中,出现紫色沉淀。反应液在25℃搅拌2小时,过滤,冷水洗涤,得紫色固体。将固体溶于水(100mL)中,在搅拌中分批加入低亚硫酸钠(18.7g,118mmol),加热到60℃搅拌0.5小时,降温至25℃搅拌16小时。过滤,分别用冷水(50mL)、乙醇(50mL)、丙酮(50mL)洗涤。干燥得产物5,6-二氨基-1-甲基嘧啶-2,4-二酮(8.60g,淡黄色固体),产率93%。1H NMR(400MHz,DMSO-d6)δ10.49(s,1H),6.15(s,2H),3.25(s,3H),2.95(s,2H)。MS-ESI计算值[M+H]+157,实测值157。
第三步
1-甲基蝶啶-2,4-二酮
将5,6-二氨基-1-甲基嘧啶-2,4-二酮(4.00g,25.6mmol)溶于水(150mL)中,在25℃下,一次性加入乙二醛(5.58g,38.4mmol,40%水溶液)。加热到60℃搅拌16小时。过滤,所得固体用水洗(50mL)得到产物1-甲基蝶啶-2,4-二酮(3.60g,黄色固体),产率:79%。
第四步
1-甲基-3-((3-甲基氧杂环丁烷-3-基)甲基)蝶啶-2,4-二酮
将1-甲基蝶啶-2,4-二酮(299mg,1.68mmol)溶于N,N-二甲基甲酰胺(8mL)中,在25℃加入3-(氯甲基)-3-甲基-氧杂环丁烷(222mg,1.85mmol),碘化钾(334mg,2.02mmol)和碳酸钾(464mg,3.36mmol),反应液加热到120℃,搅拌17小时。反应液冷至室温,过滤。滤液减压浓缩,剩余物用高效液相色谱法纯化,得1-甲基-3-((3-甲基氧杂环丁烷-3-基)甲基)蝶啶-2,4-二酮(40.0mg),产率:9%。1H NMR:(400MHz,CDCl3)δ8.68(d,J=4.0Hz,1H),8.62(d,J=4.0Hz,1H),4.78(d,J=8.0Hz,2H),4.32-4.29(m,4H),3.73(s,3H),1.42(s,3H)。计算值[M+H]+263,实测值263.
实施例64
3-((3-乙基氧杂环丁烷-3-基)甲基)-1-甲基蝶啶-2,4-二酮
Figure PCTCN2016081103-appb-000100
第一步
3-((3-乙基氧杂环丁烷-3-基)甲基)-1-甲基蝶啶-2,4-二酮
将1-甲基蝶啶-2,4-二酮(100mg,0.560mmol)溶于N,N-二甲基甲酰胺(4mL)中,在25℃加入(3-乙基氧杂环丁烷-3-基)甲基甲磺酸酯(119mg,0.620mmol),碘化钾(19.0mg,0.110mmol)和碳酸钾(155mg,1.12mmol),反应液加热到120℃,搅拌16小时。反应液冷至室温,过滤。滤液减压浓缩,剩余物用制备高效液相色谱纯化得产物3-((3-乙基氧杂环丁烷-3-基)甲基)-1-甲基蝶啶-2,4-二酮(18.0mg),产率:12%。1H NMR:(400MHz,CDCl3)δ8.68(d,J=2.0Hz,1H),8.62(d,J=2.0Hz,1H),4.66(d,J=6.4Hz,2H),4.35(d,J=6.8Hz,2H),4.26(s,2H),3.73(s,3H),1.86-1.80(m,2H),1.09(t,J=7.2Hz,3H)。MS-ESI计算值[M+H]+277,实测值277。
实施例65
1-甲基-3-((四氢呋喃-3-基)甲基)蝶啶-2,4-二酮
Figure PCTCN2016081103-appb-000101
Figure PCTCN2016081103-appb-000102
第一步
1-甲基-3-((四氢呋喃-3-基)甲基)蝶啶-2,4-二酮
将1-甲基蝶啶-2,4-二酮(60.0mg,0.337mmol)溶于N,N-二甲基甲酰胺(4mL)中,在25℃加入(四氢呋喃-3-基)甲基甲磺酸酯(61.0mg,0.337mmol),碘化钾(11.0mg,0.0670mmol)和碳酸钾(93.0mg,0.674mmol),反应液加热到120℃,搅拌16小时。反应液冷至室温,过滤。滤液减压浓缩,剩余物用制备高效液相色谱纯化得产物1-甲基-3-((四氢呋喃-3-基)甲基)蝶啶-2,4-二酮(6.0mg),产率:7%。1H NMR:(400MHz,CDCl3)δ8.66(d,J=2.4Hz,1H),8.61(d,J=2.4Hz,1H),4.31-4.25(m,1H),4.18-4.12(m,1H),3.98-3.94(m,1H),3.87-3.78(m,2H),3.73(s,3H),3.67-3.63(m,1H),2.84-2.78(m,1H),2.04-1.98(m,1H),1.83-1.75(m,1H)。MS-ESI计算值[M+H]+263,实测值263。
实施例66
1-甲基-3-((四氢吡喃-4-基)甲基)蝶啶-2,4-二酮
Figure PCTCN2016081103-appb-000103
第一步
1-甲基-3-((四氢吡喃-4-基)甲基)蝶啶-2,4-二酮
将1-甲基蝶啶-2,4-二酮(100mg,0.560mmol)溶于N,N-二甲基甲酰胺(4mL)中,在25℃加入(四氢吡喃-4-基)甲基甲磺酸酯(119mg,0.622mmol),碘化钾(19.0mg,0.112mmol)和碳酸钾(155mg,1.12mmol),反应液加热到120℃,搅拌16小时。反应液冷至室温,过滤。滤液减压浓缩,剩余物用制备高效液相色谱纯化得产物1-甲基-3-((四氢吡喃-4-基)甲基)蝶啶-2,4-二酮(14.0mg),产率:9%。1H NMR:(400MHz,CDCl3)δ8.65(d,J=2.0Hz,1H),8.60(d,J=2.0Hz,1H),4.08(d,J=7.2Hz,2H),3.98-3.95(m,2H),3.72(s,3H),3.37-3.31(m,2H),2.19-2.09(m,1H),1.61-1.49(m,4H)。MS-ESI计算值[M+H]+277,实测值277。
实施例67
1-甲基-3-(2-(四氢吡喃-4-基)乙基)蝶啶-2,4-二酮
Figure PCTCN2016081103-appb-000104
第一步
1-甲基-3-(2-(四氢吡喃-4-基)乙基)蝶啶-2,4-二酮
将2-(四氢吡喃-4-基)乙基甲磺酸酯(117mg,0.561mmol),1-甲基蝶啶-2,4-二酮(100mg,0.561mmol)及碳酸钾(233mg,1.68mmol)溶于N,N-二甲基甲酰胺(5mL)中,加入碘化钾(9.3mg,0.0561mmol),反应液在120℃搅拌3小时。反应液冷至室温,过滤。滤液减压浓缩,粗产品用制备高效液相色谱纯化得产物1-甲基-3-(2-(四氢吡喃-4-基)乙基)蝶啶-2,4-二酮(12.0mg),产率:7.4%。1H NMR:(400MHz,CDCl3)δ8.65(d,J=2.4Hz,1H),8.60(d,J=2.4Hz,1H),4.18(t,J=7.2Hz,2H),3.98-3.95(m,2H),3.72(s,3H),3.42-3.37(m,2H),1.73(d,J=12.8Hz,2H),1.67(t,J=7.2Hz,2H),1.58(s,1H),1.43-1.33(m,2H)。MS-ESI计算值[M+H]+291,实测值291。
实施例68
3-((4-甲氧基环己基)甲基)-1-甲基蝶啶-2,4-二酮
Figure PCTCN2016081103-appb-000105
第一步
3-((4-甲氧基环己基)甲基)-1-甲基蝶啶-2,4-二酮
将(4-甲氧基环己基)甲基甲磺酸酯(162mg,0.729mmol),1-甲基蝶啶-2,4-二酮(100mg,0.561mmol)及碳酸钾(233mg,1.68mmol)溶于N,N-二甲基甲酰胺(5mL)中,加入碘化钾(9.3mg,0.056mmol),反应液在120℃搅拌3小时。反应液冷至室温,过滤。滤液减压浓缩,粗产品用制备高效液相色谱纯化得产物3-((4-甲氧基环己基)甲基)-1-甲基蝶啶-2,4-二酮(19.0mg),产率:11%。1H NMR:(400MHz,CDCl3)δ8.64(d,J=2.0Hz,1H),8.59(d,J=2.0Hz,1H),4.03(d,J=7.2Hz,2H),3.72(s,3H),3.34(s,3H),3.15-3.09(m,1H),2.08-2.06(m,2H),1.90-1.84(m,1H),1.77(d,J=10.0Hz,2H),1.20-1.13(m,4H)。MS-ESI计算值[M+H]+305,实测值305。
实施例69
1-甲基-3-((3-甲基吡啶-3-基)甲基)嘧啶并[4,5-d]嘧啶-2,4-二酮
Figure PCTCN2016081103-appb-000106
第一步
1-甲基嘧啶并[4,5-d]嘧啶-2,4-二酮
将6-氨基-1-甲基嘧啶-2,4-二酮(3.50g,24.8mmol)加入到甲酰胺(5.00g,111mmol)中,反应液加热到180℃,搅拌3小时,降到室温过滤,将水(10mL)加入到滤液中搅拌,再次过滤得到1-甲基嘧啶并[4,5-d]嘧啶-2,4-二酮(1.60g,淡黄色固体),产率36%。1H NMR:(400MHz,DMSO-d6)δ9.16(s,1H),9.08(s,1H),3.43(s,3H)。
第二步
1-甲基-3-((3-甲基吡啶-3-基)甲基)嘧啶并[4,5-d]嘧啶-2,4-二酮
将1-甲基嘧啶并[4,5-d]嘧啶-2,4-二酮(300mg,1.68mmol)溶于N,N-二甲基甲酰胺(8mL)中,在25℃加入3-(氯甲基)-3-甲基氧杂环丁烷(403mg,1.85mmol),碳酸钾(465mg,3.37mmol)和碘化钾(335mg,2.02mmol)。反应液加热到120℃,搅拌16小时,反应液冷至室温,过滤。滤液减压浓缩,剩余物用高效液相色谱法纯化,得到1-甲基-3-((3-甲基吡啶-3-基)甲基)嘧啶并[4,5-d]嘧啶-2,4-二酮(135mg),产率:31%。1H NMR:(400MHz,CDCl3)δ9.29(s,1H),9.16(s,1H),4.73(d,J=6.4Hz,2H),4.29(d,J=6.4Hz,2H),4.22(s,2H),3.69(s,3H),1.39(s,3H)。MS-ESI计算值[M+H]+263,实测值263。
实施例70
3-((3-乙基氧杂环丁烷-3-基)甲基)-1-甲基嘧啶并[4,5-d]嘧啶-2,4-二酮
Figure PCTCN2016081103-appb-000107
第一步
3-((3-乙基氧杂环丁烷-3-基)甲基)-1-甲基嘧啶并[4,5-d]嘧啶-2,4-二酮
将1-甲基嘧啶并[4,5-d]嘧啶-2,4-二酮(60mg,0.337mmol)溶于N,N-二甲基甲酰胺(4mL)中,在25℃加入(3-乙基氧杂环丁烷-3-基)甲基甲磺酸酯(71.0mg,0.370mmol),碘化钾(11.0mg,0.0674 mmol)和碳酸钾(93.0mg,0.673mmol),反应液加热到120℃,搅拌16小时。反应液冷至室温,过滤。滤液减压浓缩,剩余物用制备高效液相色谱纯化得产物3-((3-乙基氧杂环丁烷-3-基)甲基)-1-甲基嘧啶并[4,5-d]嘧啶-2,4-二酮(43.0mg),产率:46%。1H NMR:(400MHz,CDCl3)δ9.31(s,1H),9.17(s,1H),4.61(d,J=6.4Hz,2H),4.34(d,J=6.4Hz,2H),4.18(s,2H),3.71(s,3H),1.81(q,J=7.2Hz,2H),1.08(t,J=7.2Hz,3H)。MS-ESI计算值[M+H]+277,实测值277。
实施例71
1-甲基-3-((四氢呋喃-3-基)甲基)嘧啶并[4,5-d]嘧啶-2,4-二酮
Figure PCTCN2016081103-appb-000108
第一步
1-甲基-3-((四氢呋喃-3-基)甲基)嘧啶并[4,5-d]嘧啶-2,4-二酮
将1-甲基嘧啶并[4,5-d]嘧啶-2,4-二酮(50.0mg,0.281mmol)溶于N,N-二甲基甲酰胺(4mL)中,在25℃加入(四氢呋喃-3-基)甲基甲磺酸酯(56.0mg,0.308mmol),碘化钾(9.0mg,0.056mmol)和碳酸钾(78.0mg,0.561mmol),反应液加热到120℃,搅拌16小时。反应液冷至室温,过滤。滤液减压浓缩,剩余物用制备高效液相色谱纯化得产物1-甲基-3-((四氢呋喃-3-基)甲基)嘧啶并[4,5-d]嘧啶-2,4-二酮(45.0mg),产率:61%。1H NMR:(400MHz,CDCl3)δ9.30(s,1H),9.15(s,1H),4.23-4.14(m,1H),4.11-4.03(m,1H),3.99-3.92(m,1H),3.85-3.75(m,2H),3.69(s,3H),3.64-3.59(m,1H),2.82-2.70(m,1H),2.06-1.96(m,1H),1.80-1.64(m,1H)。MS-ESI计算值[M+H]+263,实测值263。
实施例72
1-甲基-3-((四氢吡喃-4-基)甲基)嘧啶并[4,5-d]嘧啶-2,4-二酮
Figure PCTCN2016081103-appb-000109
第一步
1-甲基-3-((四氢吡喃-4-基)甲基)嘧啶并[4,5-d]嘧啶-2,4-二酮
将1-甲基蝶啶-2,4-二酮(60.0mg,0.337mmol)溶于N,N-二甲基甲酰胺(4mL)中,在25℃加入(四氢吡喃-4-基)甲基甲磺酸酯(72.0mg,0.370mmol),碘化钾(11.0mg,0.674mmol)和碳酸钾(93.0mg,0.674mmol),反应液加热到120℃,搅拌16小时。反应液冷至室温,过滤。滤液减压浓缩。剩余物用制备高效液相色谱纯化得产物1-甲基-3-((四氢吡喃-4-基)甲基)嘧啶并[4,5-d]嘧啶-2,4-二酮(43.0mg),产率:46%。1H NMR:(400MHz,CDCl3)δ9.30(s,1H),9.15(s,1H),4.02-3.94(m,4H),3.69(s,3H),3.38-3.30(m,2H),2.15-2.03(m,1H),1.67-1.58(m,2H),1.54-1.44(m,2H)。MS-ESI计算值[M+H]+277,实测值277。
实施例73
1-甲基-3-(2-(四氢吡喃-4-基)乙基)嘧啶并[4,5-d]嘧啶-2,4-二酮
Figure PCTCN2016081103-appb-000110
第一步
1-甲基-3-(2-(四氢吡喃-4-基)乙基)嘧啶并[4,5-d]嘧啶-2,4-二酮
将2-(四氢吡喃-4-基)乙基甲磺酸酯(117mg,0.561mmol),1-甲基嘧啶并[4,5-d]嘧啶-2,4-二酮(100.0mg,0.561mmol)及碳酸钾(233mg,1.68mmol)溶于N,N-二甲基甲酰胺(5mL)中,加入碘化钾(4.7mg,0.028mmol),反应液在120℃搅拌3小时。反应液冷至室温,过滤。滤液减压浓缩,粗产品用制备高效液相色谱纯化得产物1-甲基-3-(2-(四氢吡喃-4-基)乙基)嘧啶并[4,5-d]嘧啶-2,4-二酮(49.0mg),产率:30%。1H NMR:(400MHz,CDCl3)δ9.32(s,1H),9.17(s,1H),4.13(t,J=7.2Hz,2H),4.01-3.97(m,2H),3.71(s,3H),3.41(t,J=10.8Hz,2H),1.75-1.69(m,2H),1.67-1.63(m,2H),1.60(s,1H),1.42-1.36(m,2H)。MS-ESI计算值[M+H]+291,实测值291。
实施例74
3-((4-甲氧基环己基)甲基)-1-甲基嘧啶并[4,5-d]嘧啶-2,4-二酮
Figure PCTCN2016081103-appb-000111
第一步
3-((4-甲氧基环己基)甲7基)-1-甲基嘧啶并[4,5-d]嘧啶-2,4-二酮
将(4-甲氧基环己基)甲基甲磺酸酯(81.1mg,0.364mmol),1-甲基嘧啶并[4,5-d]嘧啶-2,4-二酮(50.0mg,0.280mmol)及碳酸钾(38.7mg,0.280mmol)溶于N,N-二甲基甲酰胺(5mL)中,加入碘化钾(46.5mg,0.280mmol),反应液在120℃搅拌3小时。反应液冷至室温,过滤。滤液减压浓缩,粗产品用制备高效液相色谱纯化得产物3-((4-甲氧基环己基)甲基)-1-甲基嘧啶并[4,5-d]嘧啶-2,4-二酮(53.0mg),产率:62%。1H NMR:(400MHz,Methonal-d4)δ9.20(s,1H),9.13(s,1H),3.93(d,J=7.2Hz,2H),3.67(s,3H),3.35(s,3H),3.22-3.16(m,1H),2.10-2.07(m,2H),1.84-1.78(m,3H),1.21-1.08(m,4H)。MS-ESI计算值[M+H]+305,实测值305。
实验例1:体外评价PDE2磷酸二酯酶抑制活性
实验目的:通过荧光偏振分析法检测AMP/GMP抗体上取代的AlexaFluor 633荧光染料来检测反应体系中产生的AMP/GMP浓度,计算待测化合物的PDE2磷酸二酯酶抑制IC50值。
实验材料:
测定緩沖溶液:10mM Tris-HCl,pH 7.5,5mM MgCl2,0.01%Brij 35,1mM DTT,and 1%DMSO.
酶:使用N端GST标签用杆状病毒在昆虫Sf9细胞中表达重组全长人PDE2A蛋白
底物:1μM cGMP
检测方法:
Figure PCTCN2016081103-appb-000112
AMP2/GMP2抗体,AMP2/GMP2AlexaFluor 633荧光染料
实验操作:
将新鲜制备的缓冲溶液配置酶溶液,然后加入到反应孔穴中,通过Echo550非接触式纳升级声波移液系统加入待测化合物的DMSO溶液,然后室温下预温育10分钟,加入底物(1μM cGMP)引发反应,室温反应一小时。然后加入检测系统(
Figure PCTCN2016081103-appb-000113
AMP2/GMP2抗体,AMP2/GMP2AlexaFluor 633荧光染料),室温下反应90分钟,然后使用Ex/Em 620/688检测荧光偏振。
荧光偏振强度通过AMP/GMP标准曲线换算成nM浓度,然后计算相对DMSO空白的相对酶活性抑制,利用Prism软件包(GraphPad Software,San Diego California,USA)计算IC50值和曲线
实验结果:
表1 PDE2磷酸二酯酶抑制活性测试结果
供试品(各实施例所制得的化合物) PDE2磷酸二酯酶抑制活性
实施例1 --
实施例2 --
实施例3 +
实施例4 +
实施例5 +
实施例6 --
实施例7 --
实施例8 --
实施例9 --
实施例10 --
实施例11异构体1 +
实施例11异构体2 --
实施例12异构体1 +
实施例12异构体2 +
实施例13 --
实施例14 +
实施例15 +
实施例16 +
实施例18 +
实施例19 --
实施例20 +
实施例21 +
实施例22 --
实施例23 +
实施例25 --
实施例26 +
实施例33 +
实施例39 --
注:10μM<+≤50μM;1μM<++≤10μM;+++≤1μM;--N/A
结论:本发明化合物具有显著甚至意料不到的PDE2A蛋白酶抑制活性。
实验例2:体外评价化合物对LPS诱导大鼠血液中TNF-α的影响
实验目的:在体外检测化合物对LPS诱导大鼠血液中TNF-α的影响,评估化合物对大鼠血液中LPS诱导TNF-α的抑制作用。
实验材料:
Sprague Dawley大鼠(雄性,210~260g,8~10周龄,上海斯莱克)
Rat TNF-alpha Quantikine ELISA Kit(R&D,#SRTA00)
实验操作:
配制浓度为1mM的待测化合物溶液,分别在48孔细胞培养板中加入40ul(化合物终浓度为100uM)。大鼠用异氟烷麻醉后,于心脏采血(肝素抗凝)。将血液加入已经加好待测化合物的48孔板中,每孔320ul。将48孔板放置于细胞培养箱中,孵育30分钟后取出,加入40ul LPS溶液(100ug/ml),混匀后放置于培养 箱中继续孵育。5小时后取出48孔板,血样转移至1.5ml离心管中,置于离心机中离心(4,500rpm,4℃,5minutes),分离上层得血浆,分装后速冻,保存在-80度冰箱。第二天按照试剂盒说明书操作用R&D ELISA试剂盒进行血浆样品中TNF-α水平检测。
实验结果:
表2 TNFα抑制活性测试结果
供试品(各实施例所制得的化合物) TNF-α抑制比率
实施例3 +
实施例17 --
注:80%>+≥60%;++≥80%;--N/A
结论:本发明化合物具有显著甚至意料不到的TNFα抑制活性。

Claims (11)

  1. 式(Ⅰ)所示化合物、其互变异构体或其药学上可接受的盐,
    Figure PCTCN2016081103-appb-100001
    其中,
    环B为任选被1~3个R取代的4~6元环醚或烷氧基-环烷基;
    L为任选被1~2个R取代的C1-3烷基;
    环A选自任选被1或2个R1取代的5~6元芳基或杂芳基;
    R1选自卤素、OH、NH2、任选被1~3个R2取代的:C1-6烷基或杂烷基、3~6元环烷基或杂环烷基、被3~6元环烷基或杂环烷基取代的C1-6烷基或杂烷基;
    R2选自卤素、OH、NH2、Me、CF3、OMe、OCF3
    所述杂代表杂原子,选自O、S、N,每个杂烷基或杂环烷基上的杂原子数目分别独立地选自1、2或3;
    R选自卤素、N(R’)(R’)、任选被1~3个R’取代的C1-3烷基或杂烷基;
    R’选自H、卤素、NH2、Me、CF3、OMe、OCF3
  2. 根据权利要求1所述的化合物,其中,所述R1选自卤素、OH、NH2、任选被1~3个R2取代的:C1-4烷基或杂烷基、被3~5元环烷基或杂环烷基取代的C1-3烷基或杂烷基;
    具体地,R1选自:Me、CF3、Et、CH2CF3
    Figure PCTCN2016081103-appb-100002
  3. 根据权利要求1所述化合物、其互变异构体或其药学上可接受的盐,其中,R选自:F、Cl、Br、I、Me、
    Figure PCTCN2016081103-appb-100003
  4. 根据权利要求1所述的化合物、其互变异构体或其药学上可接受的盐,其中所述环B选自任选被1~3个R取代的:
    Figure PCTCN2016081103-appb-100004
  5. 根据权利要求4所述的化合物、其互变异构体或其药学上可接受的盐,其中,所述B选自任选被1~3个R取代的:
    Figure PCTCN2016081103-appb-100005
    具体地,所述B选自:
    Figure PCTCN2016081103-appb-100006
    Figure PCTCN2016081103-appb-100007
  6. 根据权利要求1所述的化合物、其互变异构体或其药学上可接受的盐,其中,所述L选自任选被1~2个R取代的:亚甲基、
    Figure PCTCN2016081103-appb-100008
    具体地,L选自:亚甲基、
    Figure PCTCN2016081103-appb-100009
  7. 根据权利要求1所述的化合物、其互变异构体或其药学上可接受的盐,环A选自任选被1或2个R1取代的:咪唑基、吡唑基、吡啶基、吡嗪基、哒嗪基、嘧啶基、苯基。
  8. 根据权利要求7所述的化合物、其互变异构体或其药学上可接受的盐,环A自任选被1或2个R1取代的
    Figure PCTCN2016081103-appb-100010
    具体地,环A选自:
    Figure PCTCN2016081103-appb-100011
    Figure PCTCN2016081103-appb-100012
  9. 根据权利要求7所述的化合物、其互变异构体或其药学上可接受的盐,结构单元
    Figure PCTCN2016081103-appb-100013
    选自:
    Figure PCTCN2016081103-appb-100014
    Figure PCTCN2016081103-appb-100015
  10. 根据权利要求1所述化合物,其选自:
    Figure PCTCN2016081103-appb-100016
    Figure PCTCN2016081103-appb-100017
    Figure PCTCN2016081103-appb-100018
    Figure PCTCN2016081103-appb-100019
  11. 根据权利要求1~10任意一项所述的化合物、其互变异构体或其化学上可接受的盐在制备PDE2抑制剂和TNFα抑制剂中的应用。
PCT/CN2016/081103 2015-05-20 2016-05-05 羟基嘌呤类化合物及其应用 WO2016184313A1 (zh)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US15/575,315 US10618898B2 (en) 2015-05-20 2016-05-05 Hydroxyl purine compounds and use thereof
CN201680029214.9A CN107614501B (zh) 2015-05-20 2016-05-05 羟基嘌呤类化合物及其应用

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201510260884.9 2015-05-20
CN201510260884 2015-05-20

Publications (1)

Publication Number Publication Date
WO2016184313A1 true WO2016184313A1 (zh) 2016-11-24

Family

ID=57319487

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2016/081103 WO2016184313A1 (zh) 2015-05-20 2016-05-05 羟基嘌呤类化合物及其应用

Country Status (3)

Country Link
US (1) US10618898B2 (zh)
CN (1) CN107614501B (zh)
WO (1) WO2016184313A1 (zh)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023000636A1 (zh) * 2021-07-20 2023-01-26 韶远科技(上海)有限公司 一种4-甲基苯磺酸[(3-氟氧杂环丁烷-3-基)甲基]酯的合成方法

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104428302A (zh) * 2012-07-31 2015-03-18 勃林格殷格翰国际有限公司 4-甲基-2,3,5,9,9b-五氮杂-环戊二烯并[a]萘
CN105566324A (zh) * 2014-10-09 2016-05-11 四川好医生药业集团有限公司 羟基嘌呤类化合物及其应用

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU6702894A (en) * 1993-04-09 1994-11-08 Cell Therapeutics, Inc. Ring-substituted cell signaling inhibitors
US6323201B1 (en) 1994-12-29 2001-11-27 The Regents Of The University Of California Compounds for inhibition of ceramide-mediated signal transduction
PA8539301A1 (es) 2001-02-14 2002-09-30 Warner Lambert Co Inhibidores de la metaloproteinasa de la matriz
EA007485B1 (ru) 2001-02-24 2006-10-27 Берингер Ингельхайм Фарма Гмбх Унд Ко. Кг Производные ксантина, способ их получения, фармацевтическая композиция на их основе и способ ее получения
MXPA05000754A (es) 2002-07-17 2005-04-19 Warner Lambert Co Combinacion de un inhibidor carboxilico alosterico de la metaloproteinasa de matriz-13 con celecoxib o valdecoxib.
DE10235225B3 (de) 2002-08-01 2004-01-22 Albert-Ludwigs-Universität Freiburg Verfahren zur Durchführung chemischer Reaktionen unter Beteiligung von an fluorierten Trägermaterialien über Fluor-Fluor-Wechselwirkungen adsorbierten Verbindungen, fluoriertes Trägermaterial sowie die Verwendung des Trägermaterials
JP2008546752A (ja) 2005-06-21 2008-12-25 ザ ユニヴァーシティー コート オブ ザ ユニヴァーシティー オブ ダンディー インヒビター化合物
TW201623257A (zh) 2014-05-09 2016-07-01 奧利安公司 藥理活性之喹唑啉二酮衍生物
US10221177B2 (en) 2014-09-19 2019-03-05 Hydra Biosciences, Inc. Inhibiting the transient receptor potential A1 ion channel
EP3205652B1 (en) 2014-10-09 2022-03-09 Guangdong Raynovent Biotech Co., Ltd. Hydroxyl purine compounds and applications thereof
WO2016184312A1 (zh) * 2015-05-20 2016-11-24 南京明德新药研发股份有限公司 羟基嘌呤类化合物及其应用

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104428302A (zh) * 2012-07-31 2015-03-18 勃林格殷格翰国际有限公司 4-甲基-2,3,5,9,9b-五氮杂-环戊二烯并[a]萘
CN105566324A (zh) * 2014-10-09 2016-05-11 四川好医生药业集团有限公司 羟基嘌呤类化合物及其应用

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
AARON, T.L. ET AL.: "Predictable Stereoselective and Chemoselective Hydroxylations and Epoxidations with P450 3A4", J. AM. CHEM. SOC., no. 133, 29 April 2011 (2011-04-29), pages 7853 - 7858, XP055330076 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023000636A1 (zh) * 2021-07-20 2023-01-26 韶远科技(上海)有限公司 一种4-甲基苯磺酸[(3-氟氧杂环丁烷-3-基)甲基]酯的合成方法

Also Published As

Publication number Publication date
US10618898B2 (en) 2020-04-14
US20180148451A1 (en) 2018-05-31
CN107614501A (zh) 2018-01-19
CN107614501B (zh) 2020-01-14

Similar Documents

Publication Publication Date Title
ES2957233T3 (es) Inhibidores de quinasa tipo receptor de activina
CN111606908B (zh) Jak抑制剂化合物及其用途
JP6628930B1 (ja) Rsウイルス(rsv)複製の阻害活性を有するピペリジン置換ピラゾロ[1,5−a]ピリミジン誘導体
RU2553681C2 (ru) N-содержащие гетероарильные производные в качестве ингибиторов jak3 киназы
TWI694998B (zh) Jak抑制劑
TW201713665A (zh) 適用於治療與ntrk相關之病症的化合物及組合物
JP7459094B2 (ja) ピラゾリル-アミノ-ピリミジニル誘導体のベンズアミド、およびその組成物ならびに方法
KR20090101281A (ko) 퓨린 유도체
WO2015192760A1 (zh) 作为pi3k抑制剂的吡啶并[1,2-a]嘧啶酮类似物
AU2019353144B2 (en) Compounds and compositions for treating conditions associated with APJ receptor activity
EP3632903A1 (en) Compound used as autophagy regulator, and preparation method therefor and uses thereof
CN111032630B (zh) 一种化合物,其药物组合物及其用途及应用
KR20200140849A (ko) A2a 수용체 길항제로서의 피라졸로트리아졸로피리미딘 유도체
WO2016054971A1 (zh) 羟基嘌呤类化合物及其应用
CN107709323B (zh) 羟基嘌呤类化合物及其应用
CN111655689B (zh) 吡唑并吡啶酮化合物
CN114075200A (zh) 用于治疗重症肺炎的jak抑制剂化合物
CN107614501B (zh) 羟基嘌呤类化合物及其应用
KR102412045B1 (ko) 혈액응고인자 Xa 억제제로서의 히드라지드 화합물
TWI690529B (zh) 羥基嘌呤類化合物及其應用
TWI690526B (zh) 羥基嘌呤類化合物及其應用
WO2023194964A1 (en) Fused pyridazine derivatives as nlrp3 inhibitors
CN118139864A (zh) 新化合物
CN116082307A (zh) Hpk1抑制剂及其在医药上的应用
EA040746B1 (ru) ПРОИЗВОДНЫЕ ИМИДАЗО[1,5-а]ПИРАЗИНА В КАЧЕСТВЕ ИНГИБИТОРОВ PI3Kδ

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 16795800

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 15575315

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 16795800

Country of ref document: EP

Kind code of ref document: A1