MLKL inhibitors

文档序号:1712982 发布日期:2019-12-13 浏览:31次 中文

阅读说明:本技术 Mlkl抑制剂 (MLKL inhibitors ) 是由 张志远 王晓东 黄少强 闫博 刘磊 王华翌 姚郑林 韩建广 黄智� 曹伟业 于 2018-02-27 设计创作,主要内容包括:抑制细胞坏死性凋亡和/或人MLKL的嘌呤衍生物、其药物组合物,和使用有效量的化合物或组合物治疗MLKL介导的障碍的方法。所述MLKL介导的障碍为病理学相关的坏死性凋亡,包括缺血再灌注损伤、神经变性和炎性疾病,如急性胰腺炎、多发性硬化、炎性肠病和过敏性结肠炎。(Purine derivatives that inhibit necroptosis in cells and/or human MLKL, pharmaceutical compositions thereof, and methods of treating MLKL-mediated disorders using an effective amount of the compounds or compositions. The MLKL-mediated disorder is pathologically-associated necrotic apoptosis, including ischemia reperfusion injury, neurodegeneration, and inflammatory diseases, such as acute pancreatitis, multiple sclerosis, inflammatory bowel disease, and allergic colitis.)

1. A method of treating an MLKL-mediated disorder, disease, or condition, comprising treating a human in need thereof with an MLKL inhibitor compound of formula I:

Wherein:

Each of R1-R4 is independently H, or an optionally substituted, optionally heteroatom-containing, optionally cyclic C1-C18 hydrocarbyl group, or an optionally substituted heteroatom, and R1 and R2 are optionally linked to form a ring;

n is 0,1 or 2; and

Or a pharmaceutically acceptable salt, hydride, or stereoisomer of the compound.

2. The method of claim 1, wherein:

(a) One of R1-R4 is an alkyl carbocyclyl, such as methylcyclopropyl;

(b) one of R1-R4 includes fluoroalkyl, such as CF 3;

(c) R4 is alkylcyano or an alkyl CR, e.g. CH2CR, wherein R is H, or an optionally substituted, optionally heteroatom containing, optionally cyclic C1-C18 hydrocarbyl group; and/or

(d) r1 and R2 are linked to form a ring, e.g., R1/R2 is Me/Me or Me/Et linked to form a ring.

3. The method of any one of claims 1-2, wherein:

One, two or three of R1-R4 are Me;

n is 2;

One of R1-R4 is alkylcyano or an alkyl CR, such as CH2CR, wherein R is H or an optionally substituted, optionally heteroatom containing, optionally cyclic C1-C18 hydrocarbyl group; and/or.

4. The method of any one of claims 1-3, wherein:

Each of R1, R2 and R3 is Me;

n is 2; and

R4 is alkylcyano or an alkyl CR, e.g. CH2CR, wherein R is H, or an optionally substituted, optionally heteroatom-containing, optionally cyclic C1-C18 hydrocarbyl group.

5. The method of any one of claims 1-4, wherein R1 and R2 are linked to form a ring, such as R1/R2 is Me/Me or Me/Et linked to form a ring.

6. The method of any one of claims 1-5, wherein:

Optionally substituted, optionally heteroatom-containing, optionally cyclic C1-C18 hydrocarbyl is optionally substituted, optionally heteroatom-containing, optionally cyclic alkyl, alkenyl, or alkynyl, or optionally substituted, optionally heteroatom-containing aryl; and/or

Optionally substituted heteroatoms are halogen, optionally substituted hydroxy (alkoxy), optionally substituted acyl (formyl, alkanoyl, carbamoyl, carboxy, amido), optionally substituted amino (amino, alkylamino, dialkylamino, amido, sulfonamido), optionally substituted thiol (mercapto, alkylthio, arylthio), optionally substituted sulfinyl or sulfonyl (alkylsulfinyl, arylsulfinyl, alkylsulfonyl, arylsulfonyl), nitro or cyano.

7. The method of any one of claims 1-6, wherein the compound comprises a formula of Table 1 or 2.

8. The method of any of claims 1-7, wherein the MLKL-mediated disorder is a condition associated with cell death, such as ischemia reperfusion injury, neurodegeneration, and inflammation, including inflammatory diseases, such as acute pancreatitis, multiple sclerosis, inflammatory bowel disease, and allergic colitis.

9. The method of any one of claims 1-8, further comprising a prior step of diagnosing the disorder, disease, or condition, or a subsequent step of detecting an improvement in the resulting associated disorder, disease, or condition.

10. A composition comprising an agent, such as an anti-inflammatory agent, for treating a condition associated with necrotic apoptosis or cell death, such as ischemia reperfusion injury, neurodegeneration, and inflammation, and a second, different MLKL inhibitor compound of formula I:

Wherein:

Each of R1-R4 is independently H, or an optionally substituted, optionally heteroatom-containing, optionally cyclic C1-C18 hydrocarbyl group, or an optionally substituted heteroatom, and R1 and R2 are optionally linked to form a ring;

n is 0,1 or 2; and

Or a pharmaceutically acceptable salt, hydride, or stereoisomer of said compound.

11. An MLKL inhibitor compound of formula I:

Wherein:

each of R1-R4 is independently H, or an optionally substituted, optionally heteroatom-containing, optionally cyclic C1-C18 hydrocarbyl group, or an optionally substituted heteroatom, and R1 and R2 are optionally linked to form a ring; and

n is 0,1 or 2; and

(a) One of R1-R4 is an alkyl carbocyclyl, such as methylcyclopropyl;

(b) One of R1-R4 includes fluoroalkyl, such as CF 3;

(c) R4 is alkylcyano or an alkyl CR, e.g. CH2CR, wherein R is H, or an optionally substituted, optionally heteroatom containing, optionally cyclic C1-C18 hydrocarbyl group; and/or

(d) R1 and R2 are linked to form a ring, e.g., R1/R2 is Me/Me or Me/Et linked to form a ring;

Or a pharmaceutically acceptable salt, hydride, or stereoisomer of the compound.

12. The compound of claim 11, wherein:

One, two or three of R1-R4 are Me; and/or

n is 2.

13. The compound of claim 11 or 12, wherein:

Optionally substituted, optionally heteroatom-containing, optionally cyclic C1-C18 hydrocarbyl is optionally substituted, optionally heteroatom-containing, optionally cyclic alkyl, alkenyl, or alkynyl, or optionally substituted, optionally heteroatom-containing aryl; and/or

Optionally substituted heteroatoms are halogen, optionally substituted hydroxy (alkoxy), optionally substituted acyl (formyl, alkanoyl, carbamoyl, carboxy, amido), optionally substituted amino (amino, alkylamino, dialkylamino, amido, sulfonamido), optionally substituted thiol (mercapto, alkylthio, arylthio), optionally substituted sulfinyl or sulfonyl (alkylsulfinyl, arylsulfinyl, alkylsulfonyl, arylsulfonyl), nitro or cyano.

14. The compound of any one of claims 11-13, wherein the compound comprises the formula of a novel compound of table 1 or 2.

15. The compound or composition of any one of claims 10-14 in unit dosage form.

Summary of The Invention

The present invention provides methods and compositions for treating MLKL-mediated disorders, diseases, or conditions. In one aspect, the invention provides a method comprising treating a human in need thereof with an MLKL inhibitor compound of formula I:

Wherein:

Each of R1-R4 is independently H, or an optionally substituted, optionally heteroatom-containing, optionally cyclic C1-C18 hydrocarbyl group, or an optionally substituted heteroatom, and R1 and R2 are optionally linked to form a ring;

n is 0, 1 or 2; and

or a pharmaceutically acceptable salt, hydride, or stereoisomer of said compound.

In an embodiment:

(a) One of R1-R4 is an alkyl carbocyclyl, such as methylcyclopropyl;

(b) one of R1-R4 includes fluoroalkyl, such as CF 3;

(c) R4 is alkylcyano or an alkyl CR, e.g. CH2CR, wherein R is H, or an optionally substituted, optionally heteroatom containing, optionally cyclic C1-C18 hydrocarbyl group; and/or

(d) R1 and R2 are linked to form a ring, e.g., R1/R2 is Me/Me or Me/Et linked to form a ring.

In an embodiment:

One, two or three of R1-R4 are Me;

n is 2;

One of R1-R4 is alkylcyano or an alkyl CR, such as CH2CR, wherein R is H or an optionally substituted, optionally heteroatom containing, optionally cyclic C1-C18 hydrocarbyl group; and/or

In an embodiment:

Each of R1, R2 and R3 is Me;

n is 2; and

R4 is alkylcyano or an alkyl CR, e.g. CH2CR, wherein R is H or an optionally substituted, optionally heteroatom-containing, optionally cyclic C1-C18 hydrocarbyl group.

In an embodiment: r1 and R2 are linked to form a ring, e.g., R1/R2 is Me/Me or Me/Et linked to form a ring.

In an embodiment: optionally substituted, optionally heteroatom-containing, optionally cyclic C1-C18 hydrocarbyl is optionally substituted, optionally heteroatom-containing, optionally cyclic alkyl, alkenyl, or alkynyl, or optionally substituted, optionally heteroatom-containing aryl; and/or the optionally substituted heteroatom is halogen, optionally substituted hydroxy (e.g. alkoxy, aryloxy), optionally substituted acyl (e.g. formyl, alkanoyl, carbamoyl, carboxy, amido), optionally substituted amino such as (e.g. amino, alkylamino, dialkylamino, amido, sulfonamido), optionally substituted thiol (e.g. mercapto, alkylthio, arylthio), optionally substituted sulfinyl or sulfonyl (e.g. alkylsulfinyl, arylsulfinyl, alkylsulfonyl, arylsulfonyl), nitro or cyano.

In an embodiment: the compounds include the chemical formula of table 1.

In an embodiment: MLKL-mediated disorders are conditions associated with cell death, such as ischemia reperfusion injury, neurodegeneration, and inflammation, including inflammatory diseases, such as acute pancreatitis, multiple sclerosis, inflammatory bowel disease, and allergic colitis.

In embodiments, the method further comprises a prior step of diagnosing the disorder, disease, or condition, or a subsequent step of detecting an improvement in the resulting associated disorder, disease, or condition.

In another aspect, the present invention provides compositions comprising an agent, such as an anti-inflammatory agent, for treating a condition associated with necroptosis or cell death, such as ischemia reperfusion injury, neurodegeneration, and inflammation, and a second, different MLKL inhibitor compound of formula I:

Wherein:

Each of R1-R4 is independently H, or an optionally substituted, optionally heteroatom-containing, optionally cyclic C1-C18 hydrocarbyl group, or an optionally substituted heteroatom, and R1 and R2 are optionally linked to form a ring;

n is 0,1 or 2; and

Or a pharmaceutically acceptable salt, hydride, or stereoisomer of said compound.

In embodiments, the compositions share embodiments of the method.

In another aspect, the present invention provides MLKL inhibitor compounds of formula I:

Wherein:

Each of R1-R4 is independently H, or an optionally substituted, optionally heteroatom-containing, optionally cyclic C1-C18 hydrocarbyl group, or an optionally substituted heteroatom, and R1 and R2 are optionally linked to form a ring; and

n is 0,1 or 2; and

(a) One of R1-R4 is an alkyl carbocyclyl, such as methylcyclopropyl;

(b) One of R1-R4 includes fluoroalkyl, such as CF 3;

(c) R4 is alkylcyano or an alkyl CR, e.g. CH2CR, wherein R is H, or an optionally substituted, optionally heteroatom containing, optionally cyclic C1-C18 hydrocarbyl group; and/or

(d) R1 and R2 are linked to form a ring, e.g., R1/R2 is Me/Me or Me/Et linked to form a ring;

Or a pharmaceutically acceptable salt, hydride, or stereoisomer of said compound.

In an embodiment: one, two or three of R1-R4 are Me; and/or n is 2.

In an embodiment: the optionally substituted, optionally heteroatom-containing, optionally cyclic C1-C18 hydrocarbyl group is an optionally substituted, optionally heteroatom-containing, optionally cyclic alkyl, alkenyl, or alkynyl group, or an optionally substituted, optionally heteroatom-containing aryl group; and/or the optionally substituted heteroatom is halogen, optionally substituted hydroxy (e.g. alkoxy, aryloxy), optionally substituted acyl (e.g. formyl, alkanoyl, carbamoyl, carboxy, amido), optionally substituted amino (e.g. amino, alkylamino, dialkylamino, amido, sulfonamido), optionally substituted thiol (e.g. mercapto, alkylthio, arylthio), optionally substituted sulfinyl or sulfonyl (e.g. alkylsulfinyl, arylsulfinyl, alkylsulfonyl, arylsulfonyl), nitro or cyano.

In embodiments, the compounds include the chemical formula of the novel compounds of table 1.

In embodiments, the subject compound or composition is in unit dosage form.

The present invention includes all combinations of the specific embodiments recited herein.

Description of specific embodiments of the invention

The following description of specific embodiments and examples is provided by way of illustration and not by way of limitation. Those skilled in the art will readily recognize that various non-critical parameters may be changed or modified to produce substantially similar results. The present invention provides various embodiments.

Unless otherwise indicated or indicated to the contrary, in these descriptions and throughout the specification, the terms "a" and "an" mean one or more, the term "or" means and/or, and polynucleotide sequences are understood to include the opposite strands as well as the alternative backbones described herein. In addition, a generic expression is an abbreviation for the expression of all members of that generic expression; for example, the recitation of a (C1-C3) alkyl group is a shorthand for the recitation of all C1-C3 alkyl groups: methyl, ethyl and propyl, including isomers thereof.

the term "heteroatom" as used herein generally means any atom other than carbon or hydrogen. Preferred heteroatoms include oxygen (O), phosphorus (P), sulfur (S), nitrogen (N), and halogen, and preferred heteroatom functional groups are haloformyl, hydroxyl, aldehyde, amine, azo, carboxyl, cyano, thiocyano (thiocyanyl), carbonyl, halogen, hydroperoxy, imine, aldimine, isocyanate, nitrate, nitrile, nitrite, nitro, nitroso, phosphate, phosphonyl, sulfide, sulfonyl, sulfo, and mercapto.

Unless otherwise specified, the term "alkyl" by itself or as part of another substituent refers to a straight or branched chain or cyclic hydrocarbon group, or combinations thereof, that is fully saturated, having the indicated number of carbon atoms (i.e., C1-C8 means 1 to 8 carbons). Examples of alkyl groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, tert-butyl, isobutyl, sec-butyl, cyclohexyl, (cyclohexyl) methyl, cyclopropylmethyl, and equivalents and isomers such as n-pentyl, n-hexyl, n-heptyl, n-octyl. If cyclic, preferred alkyl groups are C1-C8, C3-C18.

The term "alkenyl" by itself or as part of another substituent means a straight or branched chain or cyclic hydrocarbon group, or combinations thereof, which may be mono-or polyunsaturated, having the indicated number of carbon atoms (i.e., C2-C8 means 2 to 8 carbons) and one or more double bonds. Examples of alkenyl groups include ethenyl, 2-propenyl, crotyl, 2-isopentenyl, 2- (butadienyl), 2, 4-pentadienyl, 3- (1, 4-pentadienyl), and higher homologs and isomers thereof. If cyclic, preferred alkenyl groups are C1-C8, C3-C18.

The term "alkynyl" by itself or as part of another substituent means a straight or branched chain hydrocarbon group or combination thereof, which may be mono-or polyunsaturated, having the indicated number of carbon atoms (i.e., C2-C8 means 2 to 8 carbons) and one or more triple bonds. Examples of alkynyl groups include ethynyl, 1-and 3-propynyl, 3-butynyl and higher homologs and isomers thereof. If cyclic, preferred alkynyl groups are C1-C8, C3-C18.

The term "alkylene" by itself or as part of another substituent means a divalent radical derived from an alkyl group, for example as-CH2-CH2-CH2-CH2examples are given by way of illustration. Typically, the alkyl (or alkylene) groups will have from 1 to 24 carbon atoms, with those groups having 10 or fewer carbon atoms being preferred in the present invention. "lower alkyl" or "lower alkylene" is a shorter chain alkyl or alkylene group, typically having 8 or fewer carbon atoms.

The terms "alkoxy", "alkylamino" and "alkylthio" (or thioalkoxy) are used in their conventional sense and refer to those alkyl groups attached to the remainder of the molecule through an oxygen atom, an amino group, or a sulfur atom, respectively.

Unless otherwise specified, the term "heteroalkyl," by itself or in combination with another term, means a stable straight or branched chain or cyclic hydrocarbon radical, or combinations thereof, consisting of the stated number of carbonsAtoms and one to three heteroatoms selected from O, N, P, Si and S, wherein the nitrogen, sulfur and phosphorus atoms may optionally be oxidized, and the nitrogen heteroatom may optionally be quaternized. The heteroatom (S) O, N, P and S can be placed at any internal position of the heteroalkyl group. The heteroatom Si may be placed anywhere in the heteroalkyl group, including where the alkyl group is attached to the remainder of the molecule. Examples include-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、-Si(CH3)3、-CH2-CH=N-OCH3and-CH-N (CH3) -CH3. Up to two heteroatoms may be consecutive, e.g. -CH2-NH-OCH3and-CH2-O-Si(CH3)3

Similarly, the term "heteroalkylene" by itself or as part of another substituent means a divalent radical derived from a heteroalkyl radical, for example, with a-CH2-CH2-S-CH2-CH2-and-CH2-S-CH2-CH2-NH-CH2Examples are given by way of illustration. For heteroalkylene groups, heteroatoms can also occupy either or both of the chain ends (e.g., alkyleneoxy, alkylenedioxy, alkyleneamino, alkylenediamino, and the like). Furthermore, for alkylene and heteroalkylene linking groups, no orientation of the linking group is implied.

Unless otherwise indicated, the terms "cycloalkyl" and "heterocycloalkyl", by themselves or in combination with other terms, represent cyclic forms of "alkyl" and "heteroalkyl", respectively. Thus, cycloalkyl groups have the indicated number of carbon atoms (i.e., C3-C8 means 3 to 8 carbons), and may also have one or two double bonds. The heterocycloalkyl group is comprised of the indicated number of carbon atoms and one to three heteroatoms selected from O, N, Si and S, and wherein the nitrogen and sulfur atoms may optionally be oxidized, and the nitrogen heteroatom may optionally be quaternized. In addition, for heterocycloalkyl, a heteroatom may occupy the position at which the heterocycle is attached to the remainder of the molecule. Examples of cycloalkyl groups include cyclopentyl, cyclohexyl, 1-cyclohexenyl, 3-cyclohexenyl, cycloheptyl, and the like. Examples of heterocycloalkyl include 1- (1,2,5, 6-tetrahydropyridinyl), 1-piperidinyl, 2-piperidinyl, 3-piperidinyl, 4-morpholinyl, 3-morpholinyl, tetrahydrofuran-2-yl, tetrahydrofuran-3-yl, tetrahydrothiophen-2-yl, tetrahydrothiophen-3-yl, 1-piperazinyl, 2-piperazinyl, and the like.

Unless otherwise specified, the terms "halo" and "halogen" by themselves or as part of another substituent means a fluorine, chlorine, bromine or iodine atom. In addition, terms such as "haloalkyl" are intended to include alkyl groups substituted with a number of the same or different halogen atoms in the range of 1 to (2m '+1), where m' is the total number of carbon atoms in the alkyl group. For example, the term "halo (C1-C4) alkyl" is intended to include trifluoromethyl, 2,2, 2-trifluoroethyl, 4-chlorobutyl, 3-bromopropyl, and the like. Thus, the term "haloalkyl" includes monohaloalkyl (alkyl substituted with one halogen atom) and polyhaloalkyl (alkyl substituted with a number of halogen atoms in the range of 2 to (2m '+1), where m' is the total number of carbon atoms in the alkyl group). Unless otherwise indicated, the term "perhaloalkyl" means an alkyl group substituted with (2m '+1) halogen atoms, where m' is the total number of carbon atoms in the alkyl group. For example, the term "perhalo (C1-C4) alkyl" is intended to include trifluoromethyl, pentachloroethyl, 1,1, 1-trifluoro-2-bromo-2-chloroethyl, and the like.

The term "acyl" refers to those groups derived from organic acids by removal of the hydroxyl portion of the acid. Thus, unsubstituted acyl is intended to include, for example, acetyl, propionyl, butyryl, decanoyl, pivaloyl, benzoyl and the like.

Unless otherwise indicated, the term "aryl" means a polyunsaturated, usually aromatic, hydrocarbon substituent which can be a single ring or multiple rings (up to three rings) which are fused together or linked covalently. Non-limiting examples of aryl groups include phenyl, 1-naphthyl, 2-naphthyl, 4-biphenyl, and 1,2,3, 4-tetrahydronaphthalene. Preferred aryl groups are C5-C18.

The term "heteroaryl" refers to an aryl (or ring) containing 0 to 4 heteroatoms selected from N, O and S, wherein the nitrogen and sulfur atoms are optionally oxidized, and the nitrogen heteroatom is optionally quaternized. The heteroaryl group may be attached to the remainder of the molecule through a heteroatom. Non-limiting examples of heteroaryl groups include 1-pyrrolyl, 2-pyrrolyl, 3-pyrazolyl, 2-imidazolyl, 4-imidazolyl, pyrazinyl, 2-oxazolyl, 4-oxazolyl, 2-phenyl-4-oxazolyl, 5-oxazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, 2-furyl, 3-furyl, 2-thienyl, 3-thienyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidyl, 4-pyrimidyl, 5-benzothiazolyl, purinyl, 2-benzimidazolyl, 5-indolyl, 1-isoquinolinyl, 5-isoquinolinyl, 2-quinoxalinyl, 5-quinoxalinyl, 3-quinolyl and 6-quinolyl.

For convenience, the term "aryl" when used in combination with other terms (e.g., aryloxy, arylsulfenoxy, arylalkyl) includes both aromatic and heteroaromatic rings as defined above. Thus, the term "arylalkyl" is intended to include those groups in which an aryl group is attached to an alkyl group (e.g., benzyl, phenethyl, pyridylmethyl, and the like), including those alkyl groups in which a carbon atom (e.g., a methylene group) has been replaced, for example, with an oxygen atom (e.g., phenoxymethyl, 2-pyridyloxymethyl, 3- (1-naphthyloxy) propyl, and the like).

Each of the above terms (e.g., "alkyl", "heteroalkyl", "aryl" and "heteroaryl") is intended to encompass both substituted and unsubstituted forms of the indicated group. Preferred substituents for each type of group are provided below.

Substituents for alkyl and heteroalkyl (and those groups referred to as alkylene, alkenyl, heteroalkylene, heteroalkenyl, alkynyl, cycloalkyl, heterocycloalkyl, cycloalkenyl, and heterocycloalkenyl) can be a variety of groups selected from: -OR ', - (O), (NR ', - (N-OR '), -NR ' R ", -SR ', halogen, -SiR ' R '", -oc (O) R ', -c (O) R ', -CO2R'、-CONR'R"、-OC(O)NR'R"、-NR"C(O)R'、-NR'-C(O)NR"R'"、-NR'-SO2NR'"、-NR"CO2R'、-NH-C(NH2)=NH、-NR'C(NH2)=NH、-NH-C(NH2)=NR'、-S(O)R'、-SO2R'、-SO2NR'R"、-NR"SO2R, -CN and-NO2The number ranges from 0 to 3, those having 0,1 or 2 substituents being particularly preferred. R ', R ", and R'" each independently refer to hydrogen, unsubstituted (C1-C8) alkyl and heteroalkyl groups, unsubstituted aryl groups, aryl groups substituted with one to three halogens, unsubstituted alkyl, alkoxy, or thioalkoxy groups, or aryl- (C1-C4) alkyl groups. When R' and R "are attached to the same nitrogen atom, they may combine with the nitrogen atom to form a 5-, 6-or 7-membered ring. For example, -NR' R "is intended to include 1-pyrrolidinyl and 4-morpholinyl. In general, alkyl or heteroalkyl groups have from 0 to 3 substituents, with those having two or fewer substituents being preferred in the present invention. More preferably, the alkyl or heteroalkyl group is unsubstituted or monosubstituted. Most preferably, the alkyl or heteroalkyl group is unsubstituted. From the above discussion of substituents, those skilled in the art will understand that the term "alkyl" is intended to include, for example, trihaloalkyl (e.g., -CF)3and-CH2CF3) A group of (1).

Preferred substituents for alkyl and heteroalkyl groups are selected from: -OR ', -O, -NR ' R ", -SR ', halogen, -SiR ' R" R ' ", -oc (O) R ', -c (O) R ', -CO2R'、-CONR'R"、-OC(O)NR'R"、-NR"C(O)R'、-NR"CO2R'、-NR'-SO2NR"R'"、-S(O)R'、-SO2R'、-SO2NR'R"、-NR"SO2R, -CN and-NO2Wherein R 'and R' are as defined above. Further preferred substituents are selected from: -OR ', -O, -NR ' R ", halogen, -oc (O) R ', -CO2R'、-CONR'R"、-OC(O)NR'R"、-NR"C(O)R'、-NR"CO2R'、-NR'-SO2NR"R'"、-SO2R'、-SO2NR'R"、-NR"SO2R, -CN and-NO2

Similarly, the substituents of the aryl and heteroaryl groups are different and are selected from: halogen, -OR ', -OC (O) R ', -NR ' R ", -SR ', -R ', -CN, -NO2、-CO2R'、-CONR'R"、-C(O)R'、-OC(O)NR'R"、-NR"C(O)R'、-NR"CO2R'、-NR'-C(O)NR"R'"、-NR'-SO2NR"R'"、-NH-C(NH2)=NH、-NR'C(NH2)=NH、-NH-C(NH2)=NR'、-S(O)R'、-SO2R'、-SO2NR'R"、-NR"SO2R、-N3、-CH(Ph)2Perfluoro (C1-C4) alkoxy and perfluoro (C1-C4) alkyl in an amount ranging from 0 to the total number of open valences on the aromatic ring system; and wherein R ', R ", and R'" are independently selected from the group consisting of hydrogen, (C1-C8) alkyl and heteroalkyl, unsubstituted aryl and heteroaryl, (unsubstituted aryl) - (C1-C4) alkyl, and (unsubstituted aryl) oxy- (C1-C4) alkyl. When the aryl group is 1,2,3, 4-tetrahydronaphthalene, it may be substituted with a substituted or unsubstituted (C3-C7) spirocycloalkyl group. The (C3-C7) spirocycloalkyl group may be substituted in the same manner as defined herein for "cycloalkyl". Typically, aryl or heteroaryl groups have from 0 to 3 substituents, with those groups having two or fewer substituents being preferred in the present invention. In one embodiment of the present invention, an aryl or heteroaryl group is unsubstituted or monosubstituted. In another embodiment, an aryl or heteroaryl group is unsubstituted.

Preferred substituents for the aryl and heteroaryl groups are selected from: halogen, -OR ', -OC (O) R ', -NR ' R ", -SR ', -R ', -CN, -NO2、-CO2R'、-CONR'R"、-C(O)R',-OC(O)NR'R"、-NR"C(O)R'、-S(O)R'、-SO2R'、-SO2NR'R"、-NR"SO2R、-N3、-CH(Ph)2Perfluoro (C1-C4) alkoxy and perfluoro (C1-C4) alkyl, wherein R 'and R' are as defined above. Further preferred substituents are selected from: halogen, -OR ', -OC (O) R', -NR 'R ", -R', -CN, -NO2、-CO2R'、-CONR'R"、-NR"C(O)R'、-SO2R'、-SO2NR'R"、-NR"SO2R, perfluoro (C1-C4) alkoxy, and perfluoro (C1-C4) alkyl.

substituent-CO as used herein2H includes its bioisosteric substitution; see, e.g., The Practice of Medicinal Chemistry; wermuth, c.g., ed; academic Press: New York, 1996; p.203.

Two substituents on adjacent atoms of the aromatic or heteroaromatic ring may optionally be replaced by a substituent of the formula-T-C (O) - (CH)2) q-U-substituent substitution, whereinT and U are independently-NH-, -O-, -CH2-or a single bond, and q is an integer from 0 to 2. Alternatively, two substituents on adjacent atoms of the aryl or heteroaryl ring may be optionally replaced by a substituent of the formula-A- (CH2) r-B-, wherein A and B are independently-CH2-、-O-、-NH-、-S-、-S(O)-、-S(O)2-、-S(O)2NR' -or a single bond, and r is an integer of 1 to 3. One of the single bonds of the new ring so formed may optionally be replaced by a double bond. Alternatively, two substituents on adjacent atoms of the aromatic or heteroaromatic ring may optionally be substituted by a group of formula- (CH)2)s-X-(CH2) t-wherein S and t are independently integers from 0 to 3, and X is-O-, -NR' -, -S (O)2-, or-S (O)2NR' -. -NR' -and-S (O)2The substituent R 'in NR' is selected from hydrogen or unsubstituted (C1-C6) alkyl.

In embodiments, the substituents, in particular those of R1-R4, are selected from:

In embodiments, R1 is-NR 5R6, wherein R5 and R6 are independently H, or an optionally substituted, optionally heteroatom-containing, optionally cyclic C1-C18 hydrocarbyl group, or an optionally substituted heteroatom. In an embodiment: the optionally substituted, optionally heteroatom-containing, optionally cyclic C1-C18 hydrocarbyl group is an optionally substituted, optionally heteroatom-containing, optionally cyclic alkyl, alkenyl, or alkynyl group, or an optionally substituted, optionally heteroatom-containing aryl group; and/or the optionally substituted heteroatom is halogen, optionally substituted hydroxy (e.g. alkoxy, aryloxy), optionally substituted acyl (e.g. formyl, alkanoyl, carbamoyl, carboxy, amido), optionally substituted amino (e.g. amino, alkylamino, dialkylamino, amido, sulfonamido), optionally substituted thiol (e.g. mercapto, alkylthio, arylthio), optionally substituted sulfinyl or sulfonyl (e.g. alkylsulfinyl, arylsulfinyl, alkylsulfonyl, arylsulfonyl), nitro or cyano.

in embodiments, R2, R3 and R4 are methyl, and/or R5 is H or methyl, and/or R6 is a substituent selected from:

In embodiments, R1 and R2 are linked to form a ring, such as R1/R2 is Me/Me or Me/Et linked to form a 5-or 6-membered ring, in particular wherein R3 is methyl, and/or R4 is a substituent selected from:

Preferred substituents are disclosed herein and exemplified in the tables, structures, examples and claims, and can be applied to different compounds of the invention, i.e., the substituents of any given compound can be used in combination with other compounds.

In particular embodiments, suitable substituents are independently substituted or unsubstituted heteroatoms, substituted or unsubstituted C1-C6 alkyl of 0-3 heteroatoms, substituted or unsubstituted C2-C6 alkenyl of 0-3 heteroatoms, substituted or unsubstituted C2-C6 alkynyl of 0-3 heteroatoms, or substituted or unsubstituted C6-C14 aryl of 0-3 heteroatoms, wherein each heteroatom is independently oxygen, phosphorus, sulfur, or nitrogen.

In more specific embodiments, suitable substituents are independently aldehyde groups, aldimine groups, alkanoyloxy groups, alkoxy groups, alkoxycarbonyl groups, alkoxy groups, alkyl groups, amine groups, azo groups, halogens, carbamoyl groups, carbonyl groups, carboxamide groups, carboxyl groups, cyano groups, ester groups, halogens, haloformyl groups, hydroperoxy groups, hydroxyl groups, imine groups, isocyanate groups, N-tert-butoxycarbonyl groups, nitrate groups, nitrile groups, nitrite groups, nitro groups, nitroso groups, phosphate groups, phosphono groups, sulfide groups, sulfonyl groups, sulfo groups, mercapto groups, thiol groups, thiocyano groups, trifluoromethyl groups, or trifluoromethyl ethers (OCF 3).

The term "pharmaceutically acceptable salts" is intended to include salts of the active compounds prepared with relatively nontoxic acids or bases, depending on the particular substituents present on the compounds described herein. When the compounds of the present invention contain relatively acidic functional groups, base addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired base, neat or in a suitable inert solvent. Examples of pharmaceutically acceptable base addition salts include sodium, potassium, calcium, ammonium, organic amine or magnesium salts, or similar salts. When the compounds of the present invention contain relatively basic functional groups, acid addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired acid, either neat or in a suitable inert solvent. Examples of pharmaceutically acceptable acid addition salts include those derived from inorganic acids such as hydrochloric, hydrobromic, nitric, carbonic, monohydrogencarbonic, phosphoric, monohydrogenphosphoric, dihydrogenphosphoric, sulfuric, monohydrogensulfuric, hydriodic, or phosphorous acids and the like, as well as those derived from relatively nontoxic organic acids such as acetic, propionic, isobutyric, oxalic, maleic, malonic, benzoic, succinic, suberic, fumaric, mandelic, phthalic, benzenesulfonic, p-toluenesulfonic, citric, tartaric, methanesulfonic, and the like. Also included are salts of amino acids, such as arginine and the like, and salts of organic acids, such as glucuronic or galacturonic acids and the like. Certain specific compounds of the invention contain both basic and acidic functionalities that allow the compounds to be converted into base or acid addition salts.

Neutral forms of the compounds may be regenerated by contacting the salt with a base or acid and isolating the parent compound in the conventional manner. The parent form of the compound differs from the various salt forms in certain physical properties, such as solubility in polar solvents, but otherwise for the purposes of this invention a salt is equivalent to the parent form of the compound.

In addition to salt forms, the present invention provides compounds in prodrug form. Prodrugs of the compounds described herein are those compounds that undergo chemical changes under physiological conditions to provide the compounds of the present invention. In addition, prodrugs can be converted to the compounds of the present invention by chemical or biochemical methods in an ex vivo environment. For example, prodrugs can be slowly converted to compounds of the present invention when co-disposed in a transdermal patch reservoir with a suitable enzyme or chemical agent. Prodrugs are often useful because, in some cases, they may be easier to administer than the parent drug. For example, they may be more orally bioavailable than the parent drug. Prodrugs may also have improved solubility in pharmaceutical compositions over the parent drug. A wide variety of prodrug derivatives are known in the art, such as those that rely on hydrolytic cleavage or oxidative activation of the prodrug. A non-limiting example of a prodrug is the administration of a compound of the invention as an ester ("prodrug"), but which is subsequently metabolically hydrolyzed to the carboxylic acid, the active entity. Additional examples include peptidyl derivatives of the compounds of the invention.

Certain compounds of the present invention may exist in unsolvated as well as solvated forms, including hydrated forms. In general, the solvated forms are equivalent to unsolvated forms and are intended to be encompassed within the scope of the present invention. Certain compounds of the present invention may exist in a variety of crystalline or amorphous forms. In general, all physical forms are equivalent for the uses contemplated by the present invention and are intended to be within the scope of the present invention.

Certain compounds of the present invention have asymmetric carbon atoms (optical centers) or double bonds; racemates, diastereomers, geometric isomers and individual isomers are intended to be included within the scope of the present invention.

The compounds of the invention may also contain unnatural proportions of atomic isotopes at one or more of the constituent atoms, such as deuterium, e.g., -CD3、CD2H or CDH2Instead of methyl. For example, the compounds may be treated with radioactive isotopes, such as tritium (A), (B), (C), (3H) Iodine-125 (125I) Or carbon-14 (14C) And (4) radioactive labeling. All isotopic variations of the compounds of the present invention, whether radioactive or not, are intended to be encompassed within the scope of the present invention.

The term "therapeutically effective amount" means that amount of the subject compound that will be sought by a researcher, veterinarian, medical doctor or other clinician, e.g., to elicit a biological or medical response in a tissue, system, animal or human, which is sufficient to prevent the development of, or alleviate to some extent, one or more symptoms of the condition or disorder being treated. The therapeutically effective amount will vary depending on the compound, the disease and its severity, and the age, weight, etc., of the mammal to be treated.

The invention also provides pharmaceutical compositions comprising a subject compound and a pharmaceutically acceptable excipient, particularly such compositions comprising a unit dose of a subject compound, particularly such compositions co-packaged with instructions describing the use of the composition for the treatment of an applicable disease or condition (herein).

Compositions for administration may take the form of bulk liquid solutions or suspensions or bulk powders. More commonly, however, the compositions are presented in unit dosage form to facilitate accurate administration. The term "unit dosage form" refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient. Typical unit dosage forms include pre-filled, pre-metered ampoules or syringes of liquid composition, or in the case of solid compositions, pills, tablets, capsules, lozenges and the like. In such compositions, the compound is generally a minor component (from about 0.1% to about 50% by weight, or preferably about 1% to 40% by weight), the remainder being various vehicles or carriers and processing aids that aid in forming the desired form of administration.

Suitable excipients or carriers and methods for preparing administrable compositions are known or apparent to those skilled in the art and are described in more detail in publications such as Remington's Pharmaceutical Science, Mack Publishing Co, NJ (1991). Furthermore, the compounds may be advantageously used in combination with other therapeutic agents, particularly other anti-necrosis agents, described herein or otherwise known in the art. Thus, the compositions may be administered alone, in combination, or in combination in a single dosage unit.

The amount administered will depend on the compound dosage form, route of administration, etc., and will generally be determined empirically in routine experimentation, and will necessarily vary depending on the target, host, and route of administration, etc. Generally, the amount of active compound in a unit dose formulation may be varied or adjusted from about 1,3, 10, or 30 to about 30, 100, 300, or 1000mg, depending on the particular application. In a particular embodiment, the unit dosage forms are packaged in a multi-pack suitable for sequential use, for example a blister pack comprising sheets of at least 6, 9 or 12 unit dosage forms. The actual dosage employed may vary depending upon the needs of the patient and the severity of the condition being treated. Determining the appropriate dosage for a particular situation is within the skill of the art. Typically, treatment is initiated with a smaller dose that is less than the optimal dose of the compound. Thereafter, the dose is increased by a small amount until the optimum effect under the circumstances is reached. For convenience, the total daily dose may be divided and administered in portions throughout the day, if desired.

The compounds may be administered by a variety of methods, including but not limited to parenteral, topical (topical), oral, or local (local) administration, for example, by aerosol or transdermal, for prophylactic and/or therapeutic treatment. In addition, the treatment regimen (e.g., the dose and number of administrations) can vary according to the observed effect of the administered therapeutic on the patient and according to the observed response of the disease to the administered therapeutic, according to the knowledge of the skilled clinician.

The therapeutic agents of the present invention may be administered in therapeutically effective doses and amounts during a therapeutically effective regimen for treating a patient. For more potent compounds, microgram (ug) amounts per kilogram of patient may be sufficient, for example in the range of about 1, 10 or 100ug/kg to about 0.01, 0.1, 1, 10 or 100mg/kg of patient body weight, although the optimal dose is compound specific and is usually empirically determined for each compound.

In general, routine experimentation in clinical trials will determine the specific range of optimal therapeutic effect, and for each therapeutic agent, each administration regimen and administration to a particular patient will also be adjusted to be within an effective and safe range depending on the patient's condition and response to the initial administration. However, the final administration regimen will be adjusted at the discretion of the attending clinician, taking into account factors such as age, condition and size of the patient, and efficacy of the compound, severity of the condition being treated, and the like. For example, a dosage regimen of the compound may be orally administered in two to four (preferably two) divided doses of 10mg to 2000 mg/day, preferably 10 to 1000 mg/day, more preferably 50 to 600 mg/day. Intermittent treatment (e.g., one of three weeks or three of four weeks) may also be used.

The compounds of the invention are particularly useful in the treatment of diseases/disorders which may be modulated at least in part by programmed necrosis, apoptosis or production of inflammatory cytokines, in particular inflammatory bowel disease (including crohn's disease and ulcerative colitis), psoriasis, retinal detachment, retinitis pigmentosa, macular degeneration, pancreatitis, atopic dermatitis, arthritis (including rheumatoid arthritis, spondyloarthritis, gout, systemic onset juvenile idiopathic arthritis (SoJIA), psoriatic arthritis), Systemic Lupus Erythematosus (SLE), sjogren's syndrome, systemic scleroderma, antiphospholipid syndrome (APS), vasculitis, osteoarthritis, liver injury/liver disease (non-alcoholic steatohepatitis, autoimmune hepatitis, autoimmune liver-gallbladder disease, Primary Sclerosing Cholangitis (PSC), Acetaminophen toxicity, hepatotoxicity), renal damage/injury (nephritis, kidney transplantation, surgery, administration of nephrotoxic drugs (e.g., cisplatin), Acute Kidney Injury (AKI)), celiac disease, autoimmune idiopathic thrombocytopenic purpura (autoimmune ITP), transplant rejection, solid organ ischemia reperfusion injury, sepsis, Systemic Inflammatory Response Syndrome (SIRS), cerebrovascular accident (CVA, stroke), Myocardial Infarction (MI), atherosclerosis, huntington's disease, alzheimer's disease, parkinson's disease, Amyotrophic Lateral Sclerosis (ALS), allergic diseases (including asthma and atopic dermatitis), multiple sclerosis, type I diabetes, wegener's granuloma, pulmonary sarcoidosis, Behcet's disease, interleukin 1 converting enzyme (ICE, also known as caspase-1) associated fever syndrome, chronic obstructive pulmonary disease, Chronic Obstructive Pulmonary Disease (COPD), tumor necrosis factor receptor-associated periodic syndrome (TRAPS), periodontitis, EMO deficiency syndrome (F- κ -B essential regulatory gene (also known as IKK γ or IKKG) deficiency syndrome), HOIL-1 deficiency ((also known as RBCK1) heme oxide IRP2 ubiquitin ligase-1 deficiency), linear ubiquitin chain assembly complex (LUBAC) deficiency syndrome, hematologic and solid organ malignancies, bacterial and viral infections (e.g., tuberculosis and influenza), and lysosomal storage diseases (especially gaucher disease) including GM2 gangliosidosis, α -mannosidosis, aspartylglucosaminuria, cholesteryl ester storage disease, chronic hexosaminidase a deficiency, cystinosis, nodaken disease (Danon disease), fabry disease (fabry disease), fabry disease (faner disease), Fucosidosis, galactosialidosis, GM1 gangliosidosis, mucolipidosis, infant free sialosis, juvenile hexosaminidase A deficiency, Krabbe Disease, lysosomal acid lipase deficiency, metachromatic leukodystrophy, mucopolysaccharidosis, multiple sulfatase deficiency, Niemann-Pick Disease, neuronal ceroid lipofuscinosis, Pompe Disease, compact osteogenesis imperfecta, Sandhoff Disease, Schindler Disease, sialosis, Tay-Sachs and Wolman Disease. Treatment of the above diseases/disorders may more particularly relate to ameliorating organ damage or damage sustained by the disease. For example, the compounds of the invention are particularly useful for ameliorating brain tissue damage or damage following ischemic or traumatic brain injury, or for ameliorating cardiac tissue damage or damage following myocardial infarction, or for ameliorating brain tissue damage or damage associated with huntington's disease, alzheimer's disease, or parkinson's disease, or for ameliorating liver tissue damage or damage associated with non-alcoholic steatohepatitis, autoimmune hepatitis, autoimmune liver gallbladder disease, or primary sclerosing cholangitis. In addition, treatment of a disease/disorder selected from the diseases/disorders described herein may more specifically relate to amelioration of tissue damage or damage associated with acetaminophen overdose, or kidney tissue damage or damage following kidney transplantation or administration of a nephrotoxic drug or substance (e.g., cisplatin).

The subject compounds are particularly useful in the treatment of inflammatory bowel disease (including crohn's disease and ulcerative colitis), psoriasis, retinal detachment, retinitis pigmentosa, arthritis (including rheumatoid arthritis, spondyloarthritis, gout, and systemic onset juvenile idiopathic arthritis (SoJIA)), transplant rejection, and/or solid organ ischemia reperfusion injury. The compound can also be used for treating burn.

The treatment of MLKL-mediated disease conditions, or more broadly, immune-mediated diseases, can be achieved using the compounds as monotherapy or in dual or multiple combination therapy, particularly for the treatment of refractory cases, such as in combination with other anti-inflammatory and/or anti-TNF agents that can be administered in therapeutically effective amounts, as is known in the art.

The subject compounds may be used alone or in combination with other therapeutic agents. Thus, combination therapy includes the administration of at least one pharmaceutically acceptable crystalline form of the compound and at least one other therapeutically active agent. The subject compound and other therapeutically active agent(s) may be administered together or separately in a single pharmaceutical composition, and when administered separately, this may occur simultaneously or sequentially in any order. The amounts of the subject compound and other therapeutically active agent(s), as well as the relative timing of administration, are selected to achieve the desired combination therapeutic effect. Thus, in another aspect, there is provided a combination comprising a pharmaceutically acceptable crystalline form of a compound and one or more other therapeutically active agents. Thus, in one aspect of the invention, a pharmaceutically acceptable crystalline form of a subject compound or a pharmaceutical composition comprising a pharmaceutically acceptable crystalline form of a subject compound may be used in combination with or include one or more other therapeutic agents, such as an anti-inflammatory agent and/or an anti-TNF agent. For example, the subject compounds may be administered in combination with other anti-inflammatory agents, including oral or topical corticosteroids (e.g., for any of the above indications)And budesonide), anti-TNF agents (including anti-TNF biologics), 5-aminosalicylic acid and mesalamine formulations, hydroxychloroquine, thiopurines (azathioprine, mercaptopurine), methotrexate, cyclophosphamideSporins, calcineurin inhibitors (cyclosporin, pimecrolimus, tacrolimus), mycophenolic acidmTOR inhibitors (temsirolimus, everolimus), JAK inhibitors (tofacitinib)) Syk inhibitor (fotatinib), anti-IL 6 biologic, anti-IL 1 (anakinra)) Kanamantimab (R)Linacloviranti-IL 12 and IL23 biologics (Ultecumab)) anti-IL 17 biologic (secukinumab), anti-CD 22 (epratuzumab), anti-integrin (natalizumab)) Vidolizumabanti-IFNa (sifamumab), anti-CD 20 or CD4 biological agents, and other cytokine inhibitors or biological agents of T-cell or B-cell receptors or interleukins.

Examples of suitable anti-inflammatory biological agents include(anti-IL 6R mAb), anti-CD 20 mAb (rituximab)Heofamu monochoria) Brazipu, AlbapulaAnakinraUltecal monoclonal antibodyAnd belimumabExamples of other suitable anti-inflammatory biological agents include canazumabLinaclovirSecukinumab, epratuzumab, sifacimumab and ustekumabExamples of suitable anti-TNF agent biologic agents include etanercept (etaneecept,) AdalimumabInfliximabCytuzumab ozogamicinAnd golimumab

a therapeutically "effective amount" is intended to mean an amount of a compound that is sufficient to effect a treatment as defined herein when administered to a patient in need of such treatment, e.g., an amount of a compound that is sufficient to modulate and/or inhibit the activity of MLKL when administered to a human in need thereof such that the disease condition mediated by MLKL is alleviated, or prevented.

"Treating" or "treatment" is intended to mean at least alleviating a disease or disorder in a patient. Therapeutic methods to alleviate a disease or disorder include the use of a compound in the present invention in any conventionally acceptable manner, e.g., for preventing, delaying, preventing, treating, or curing an MLKL-mediated disease or disorder as described above.

The compounds of the invention may be administered by any suitable route of administration, including systemic administration and topical administration. Systemic administration includes oral administration, parenteral administration, transdermal administration, rectal administration, and administration by inhalation. Parenteral administration refers to routes of administration other than enteral, transdermal or by inhalation, and is typically by injection or infusion. Parenteral administration includes intravenous, intramuscular, and subcutaneous injections or infusions. Inhalation refers to administration into the lungs of a patient by inhalation through the oral cavity or through the nasal passage. Topical administration includes application to the skin.

The compounds of the present invention may be administered in a single administration or according to a dosing regimen wherein multiple doses are administered at different time intervals over a given period of time. For example, administration may be once, twice, three times or four times daily. The administration may be carried out until the desired therapeutic effect is achieved or the desired therapeutic effect is maintained indefinitely. The dosage regimen for a compound of the invention depends on the pharmacokinetic properties of the compound, such as absorption, distribution and half-life, which can be determined by the skilled person. In addition, for the compounds of the present invention, suitable dosing regimens, including the duration of administration of such regimens, will depend upon the disease or disorder being treated, the severity of the disease or disorder being treated, the age and physical condition of the patient being treated, the medical history of the patient being treated, the nature of concurrent therapy, the desired therapeutic effect, and similar factors within the knowledge and expertise of the skilled artisan. Such a skilled artisan will further appreciate that appropriate dosage regimens may need to be adjusted in view of individual patient response to the administration regimen or changes in individual patient needs over time. The total daily dose is 1mg to 2000 mg.

For use in therapy, the compounds of the invention are typically, but not necessarily, formulated into pharmaceutical compositions or dosage units prior to administration to a patient. Thus, the present invention also relates to pharmaceutical compositions comprising a compound of the present invention and one or more pharmaceutically acceptable excipients. The invention also relates to a dosing unit comprising a compound of the invention and one or more pharmaceutically acceptable excipients.

The pharmaceutical compositions or administration units of the invention may be prepared and packaged in bulk form, wherein an effective amount of a compound of the invention may be extracted and then administered to a patient, e.g., with powders, syrups, and injections. Alternatively, the pharmaceutical compositions or administration units of the present invention may be prepared and packaged in unit dosage form. For oral use, for example, one or more tablets or capsules may be administered. One dose of the pharmaceutical composition comprises at least a therapeutically effective amount of a compound of the invention. When prepared in unit dosage form, a pharmaceutical composition or unit of administration may contain from 1mg to 1000mg of the subject compound.

As provided herein, a unit dosage form (pharmaceutical composition or administration unit) containing 1mg to 1000mg of the compound may be administered 1,2,3 or 4 times per day, preferably 1,2 or 3 times per day, more preferably once or twice per day, to achieve treatment of an MLKL-mediated disease or disorder.

As used herein, "pharmaceutically acceptable excipient" means a material, composition or vehicle involved in imparting shape or consistency to a composition. Each excipient, when mixed, must be compatible with the other ingredients of the pharmaceutical composition in order to avoid interactions that would substantially reduce the efficacy of the compounds of the invention when administered to a patient, as well as interactions that would result in a pharmaceutical composition that is not pharmaceutically acceptable. In addition, each excipient must, of course, be of sufficiently high purity to be pharmaceutically acceptable.

The compounds of the invention and the pharmaceutically acceptable excipient or excipients are generally formulated into a dosage form suitable for administration to a patient by the desired route of administration. Conventional dosage forms include: (1) dosage forms suitable for oral administration, such as tablets, capsules, caplets, pills, troches, powders, syrups, elixirs, suspensions, solutions, emulsions, sachets and cachets; (2) dosage forms suitable for parenteral administration, such as sterile solutions, suspensions, and powders for reconstitution and the like; (3) dosage forms suitable for transdermal administration, such as transdermal patches; (4) suitable for rectal administration, such as suppositories; (5) dosage forms suitable for inhalation, such as aerosols and solutions; and (6) dosage forms suitable for topical administration, such as creams, ointments, lotions, solutions, pastes, sprays, foams and gels.

Suitable pharmaceutically acceptable excipients will vary depending on the particular dosage form selected. In addition, suitable pharmaceutically acceptable excipients may be selected for the particular function they can serve in the composition. For example, certain pharmaceutically acceptable excipients may be selected because they facilitate the production of a homogeneous dosage form. Certain pharmaceutically acceptable excipients may be selected because they facilitate the production of stable dosage forms. Certain pharmaceutically acceptable excipients may be selected because they facilitate the carrying or transport of one or more compounds of the invention from one organ or part of the body to another organ or part of the body once administered to a patient. Certain pharmaceutically acceptable excipients may be selected because they can improve patient compliance.

Suitable pharmaceutically acceptable excipients include the following types of excipients: diluents, fillers, binders, disintegrants, lubricants, glidants, granulating agents, coating agents, wetting agents, solvents, co-solvents, suspending agents, emulsifiers, sweeteners, flavoring agents, taste masking agents, colorants, anti-caking agents, humectants, chelating agents, plasticizers, viscosity increasing agents, antioxidants, preservatives, stabilizers, surfactants, and buffers. One skilled in the art will appreciate that certain pharmaceutically acceptable excipients may serve more than one function and may serve alternative functions depending on the amount of excipient present in the formulation and the type of other ingredients present in the formulation. The skilled person has the knowledge and skill in the art to enable them to select suitable amounts of suitable pharmaceutically acceptable excipients for use in the present invention. In addition, one skilled in the art can obtain a number of sources describing pharmaceutically acceptable excipients, which can be used to select the appropriate pharmaceutically acceptable excipient. Examples include Remington's Pharmaceutical Sciences (Mack Publishing Company), The Handbook of Pharmaceutical Additives (The Handbook of Pharmaceutical Additives, Gower Publishing Limited) and The Handbook of Pharmaceutical Excipients (The Handbook of Pharmaceutical Excipients, The American Pharmaceutical Association and The Pharmaceutical Press).

The pharmaceutical compositions of the present invention are prepared using techniques and methods known to those skilled in the art. Some methods commonly used in the art are described in the pharmaceutical science of remington (supra). Accordingly, another embodiment of the present invention is a method of preparing a pharmaceutical composition or dosing unit comprising the step of admixing a pharmaceutically acceptable crystalline form of the subject compound with one or more pharmaceutically acceptable excipients.

In one aspect, the invention relates to a solid oral dosage form, such as a tablet or capsule, comprising an effective amount of a compound of the invention and a diluent or filler. Suitable diluents and fillers include lactose, sucrose, dextrose, mannitol, sorbitol, starches (e.g., corn starch, potato starch, and pregelatinized starch), cellulose and its derivatives (e.g., microcrystalline cellulose), calcium sulfate, and dibasic calcium phosphate. The oral solid dosage form may further comprise a binder. Suitable binders include starches (e.g., corn starch, potato starch, and pregelatinized starch), gelatin, acacia, sodium alginate, alginic acid, tragacanth gum, guar gum, povidone, and cellulose and its derivatives (e.g., microcrystalline cellulose). The oral solid dosage form may further comprise a disintegrant. Suitable disintegrants include crospovidone, sodium starch glycolate, croscarmellose sodium, alginic acid and carboxymethylcellulose sodium. The oral solid dosage form may further comprise a lubricant. Suitable lubricants include stearic acid, magnesium stearate, calcium stearate and talc.

It is understood that the examples and embodiments described herein are for illustrative purposes only and that various modifications or changes in light thereof will be suggested to persons skilled in the art and are to be included within the spirit and purview of this application and scope of the appended claims. All publications, patents, and patent applications cited herein, including citations therein, are hereby incorporated by reference in their entirety for all purposes.

an exemplary compound; in embodiments, the compound is selected from table 1.

Table 1. series 1: black, simple numbered compounds are new; the disclosed compositions and uses of bold red numbered compounds (1,2, 4, 5,7, 8, 10,18, 20, 21, 72, 73, 75, 87) are novel.

Synthesis of

Compound 1: 8-mercapto-1, 3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 8-chloro-1, 3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To 8-chloro-1, 3-dimethyl-1H-purine-2, 6(3H,7H) -dione (200.0mg,0.93mmol) and K2CO3To a solution of (154.8mg,1.12mmol) in anhydrous DMF (2mL) was added iodomethane (159.1mg,1.12mmol) and the mixture was stirred at room temperature under nitrogen for 4h. The reaction mixture was poured into water and extracted with dichloromethane (3x 5mL), the organic layer was separated and Na was added2SO4Dry, filter, concentrate and purify by silica gel chromatography (PE: EA ═ 2:1) to give a white solid (151.0mg, 70.4%).1H-NMR(400MHz,CDCl3):δ3.95(s,3H),3.55(s,3H),3.40(s,3H).MS(m/z):229.04[M+H]+.

(II) Synthesis of 8-mercapto-1, 3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 8-chloro-1, 3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione (151.0mg,0.66mmol) in anhydrous DMF (2mL) was added NaHS (111.3mg,1.98mmol) and heated to 105 ℃ for 7H. The mixture was then cooled to room temperature and acidified to PH 3. The mixture was poured into water and extracted with dichloromethane (3x 5mL), the organic layer was separated and Na was added2SO4Dry, filter, concentrate and purify by silica gel chromatography (PE: EA ═ 1:4) to give a white solid (128.0mg, 86.7%).1H-NMR(400MHz,DMSO-d6):δ13.68(s,1H),3.68(s,3H),3.37(s,3H),3.19(s,3H).MS(m/z):227.05[M+H]+.

Compound 2:1, 3, 7-trimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione

To compound 1(20.0mg,0.09mmol) and K2CO3To a solution of (14.7mg,0.11mmol) in anhydrous DMF (2mL) was added iodomethane (15.1mg,0.11mmol) and stirred at room temperature under nitrogen for 2 h. The reaction mixture was poured into water and extracted with dichloromethane (3 x 5mL), the organic layer was separated and Na was added2SO4Dry, filter, concentrate and purify by preparative TLC to give a white solid (19.2mg, 95.05%).1H-NMR(400MHz,CDCl3):δ3.84(s,3H),3.57(s,3H),3.39(s,3H),2.72(s,3H).MS(m/z):241.07[M+H]+.

Compound 3: 1,3, 7-trimethyl-8- (methylsulfinyl) -1H-purine-2, 6(3H,7H) -dione

Compound 4:1, 3, 7-trimethyl-8- (methylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

To a solution of Compound 2(18.0mg,0.07mmol) in MeOH (2mL) was added oxone (69.2mg,0.11mmol) in H2O (2mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 5mL), the organic layer separated and washed with Na2SO4Drying, filtration, concentration and purification by preparative TLC afforded compound 3 as a white solid (3.4mg, 20.5%).1H-NMR(400MHz,CDCl3):δ4.28(s,3H),3.58(s,3H),3.41(s,3H),3.18(s,3H).MS(m/z):257.06[M+H]+.

Compound 46.3mg (30.8%) was obtained as a white solid.1H-NMR(400MHz,CDCl3):δ4.32(s,3H),3.57(s,3H),3.43(s,3H),3.42(s,3H).MS(m/z):273.06[M+H]+.

Compound 5: 8- (ethylsulfanyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To Compounds 1(30mg) and K2CO3A solution of (21.98mg) in anhydrous DMF (2mL) was added iodoethane (24.8mg) and stirred at room temperature under nitrogen for 3 h. The reaction mixture was poured into water and extracted with ethyl acetate (3x 5mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by preparative TLC (PE: EA1:1) to give 25.2mg (74.8%) as a white solid.1H-NMR(400MHz,CDCl3):δ3.85(s,3H),3.57(s,3H),3.39(s,3H),3.28(q,J=7.6Hz,2H),1.43(t,J=7.6Hz,3H).MS(m/z):255.08[M+H]+.

Compound 6:8- (ethylsulfinyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

Compound 7: 8- (ethylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

to a solution of Compound 6(20mg) in MeOH (2mL) was added oxone (72.61mg) in H2O (2mL) solution. The mixture was then stirred at room temperature for 2 h. The solvent was then removed and extracted with dichloromethane (3x 5mL), the organic layer separated and washed with Na2SO4Drying, filtration, concentration and purification by preparative TLC yielded compound 65.7 mg (34.5%) as a white solid.1H-NMR(400MHz,CDCl3):δ4.28(s,3H),3.57(s,3H),3.42(s,3H),3.41(q,J=7.6Hz,2H),1.39(t,J=7.6Hz,3H).MS(m/z):271.08[M+H]+.

Compound 712.3mg (42.7%) was obtained as a white solid.1H-NMR(400MHz,CDCl3):δ4.33(s,3H),3.59(s,3H),3.55(q,J=7.6Hz,2H),3.42(s,3H),1.46(t,J=7.6Hz,3H).MS(m/z):287.07[M+H]+.

Compound 8: 1,3, 7-trimethyl-8- (propylsulfanyl) -1H-purine-2, 6(3H,7H) -dione

To Compounds 1(30mg) and K2CO3(21.98mg) in anhydrous DMF (2mL) was added 1-iodopropane (27.07mg) and stirred at room temperature under nitrogen for 3 h. The reaction mixture was poured into water and extracted with ethyl acetate (3x 5mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by preparative TLC (PE: EA1:1) to give 30.2mg (84.2%) as a white solid.1H-NMR(400MHz,CDCl3):δ3.85(s,3H),3.56(s,3H),3.39(s,3H),3.25(q,J=2.8Hz,2H),1.83-1.74(m,2H),1.05(t,J=7.2Hz,3H).MS(m/z):269.10[M+H]+.

Compound 9: 1,3, 7-trimethyl-8- (propylsulfinyl) -1H-purine-2, 6(3H,7H) -dione

Compound 10:1, 3, 7-trimethyl-8- (propylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

To a solution of Compound 8(25mg) in MeOH (2mL) was added oxone (86.01mg) in H2O (2mL) solution. The mixture was then stirred at room temperature for 3 h. The solvent was then removed and extracted with dichloromethane (3x 10mL), the organic layer separated and washed with Na2SO4Drying, filtration, concentration and purification by preparative TLC yielded 97.9mg (30.1%) of compound as a white solid.1H-NMR(400MHz,CDCl3):δ4.26(s,3H),3.56(s,3H),3.47(q,J=6.8Hz,2H),3.41(s,3H),1.89-1.67(m,2H),1.37(t,J=7.2Hz,3H).MS(m/z):285.09[M+H]+.

Compound 1015.2 mg (39.1%) was obtained as a white solid.1H-NMR(400MHz,CDCl3):δ4.32(s,3H),3.56(s,3H),3.49(q,J=5.6Hz,2H),3.41(s,3H),1.95-1.89(m,2H),1.10(t,J=7.6Hz,3H).MS(m/z):301.09[M+H]+.

Compound 11: 8- (Cyclopropylmethylthio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To Compounds 1(30mg) and K2CO3(bromomethyl) cyclopropane (21.34mg) was added to a solution of (21.98mg) in anhydrous DMF (2mL) and stirred at room temperature under nitrogen for 2.5 h. The reaction mixture was poured into water and extracted with ethyl acetate (3x 5mL), the organic layer was separated and Na was used2SO4Drying, filtration, concentration and purification by preparative TLC (PE: EA1:1) gave 36.5mg (97.3%) as a white solid.1H-NMR(400MHz,CDCl3):δ3.87(s,3H),3.56(s,3H),3.39(s,3H),3.21(d,J=7.6Hz,2H),1.21-1.17(m,1H),0.66-0.62(m,2H),0.35-0.33(m,2H).MS(m/z):281.10[M+H]+.

Compound 12: 8- (Cyclopropylmethylsulfinyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

Compound 13: 8- (Cyclopropylmethylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of Compound 11(23mg) in MeOH (2mL) was added oxone (75.7mg) in H2O (2mL) solution. The mixture was then stirred at room temperature for 2 h. The solvent was then removed and extracted with dichloromethane (3x 10mL), the organic layer separated and washed with Na2SO4Drying, filtration, concentration and purification by preparative TLC yielded 127.7mg (29.5%) of compound as a white solid.1H-NMR(400MHz,CDCl3):δ4.26(s,3H),3.56(s,3H),3.40(s,3H),3.33(d,J=7.6Hz,2H),1.15-1.05(m,1H),0.72-0.68(m,2H),0.42-0.36(m,2H).MS(m/z):297.09[M+H]+.

Compound 1313.4 mg (49.7%) was obtained as a white solid.1H-NMR(400MHz,CDCl3):δ4.36(s,3H),3.57(s,3H),3.43(s,3H),3.41(d,J=4.0Hz,2H),1.19-1.11(m,1H),0.71-0.66(m,2H),0.34-0.30(m,2H).MS(m/z):313.09[M+H]+.

Compound 14:1, 3, 7-trimethyl-8- (2,2, 2-trifluoroethylsulfinyl) -1H-purine-2, 6(3H,7H) -dione

Compound 15: 1,3, 7-trimethyl-8- (2,2, 2-trifluoroethylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 1,3, 7-trimethyl-8- (2,2, 2-trifluoroethylthio) -1H-purine-2, 6(3H,7H) -dione

To compound 1(30.0mg,0.13mmol) and K2CO3(21.4mg,0.15mmol) in anhydrous DMF (5mL) was added 2-bromo-1, 1, 1-trifluoroethane (25.9mg,0.15mmol) and stirred under nitrogen at room temperature overnight. The reaction mixture was then poured into water and extracted with EA (3x 10mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by preparative TLC to give a white solid (14.1 mg). MS (M/z) 309.06[ M + H]+.

(II) Synthesis of 1,3, 7-trimethyl-8- (2,2, 2-trifluoroethylsulfonyl) -1H-purine-2, 6(3H,7H) -dione and 1,3, 7-trimethyl-8- (2,2, 2-trifluoroethylsulfinyl) -1H-purine-2, 6(3H,7H) -dione

To a solution of 1,3, 7-trimethyl-8- (2,2, 2-trifluoroethylthio) -1H-purine-2, 6(3H,7H) -dione (10mg,0.03mmol) in DCM (2mL) was added m-CPBA (11.2mg,0.06 mmol). The mixture was then stirred at room temperature for 2 h. The solvent was then removed and extracted with dichloromethane (3x 5mL), the organic layer separated and washed with Na2SO4Drying, filtration, concentration and purification by preparative TLC afforded compound 14 as a white solid: (3.1mg,28.8%)。1H-NMR(400MHz,CDCl3):δ4.38(m,2H),4.35(s,3H),3.58(s,3H),3.42(s,3H).MS(m/z):341.05[M+H]+.

Compound 154.3 mg (30.4%) was obtained as a white solid.1H-NMR(400MHz,CDCl3):δ4.55-4.46(m,1H),4.28(s,3H),4.11-4.05(m,1H),3.58(s,3H),3.42(s,3H).MS(m/z):325.05[M+H]+.

Compound 16: 8- (2- (2- (2-hydroxyethoxy) ethoxy) ethylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 8- (2- (2- (2-hydroxyethoxy) ethoxy) ethylthio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To compound 1(100mg,0.44mmol) and Cs2CO3(172.9mg,0.53mmol) in dry DMF (4mL) was added 2- (2- (2-chloroethoxy) ethoxy) ethanol (89.4mg,0.53mmol) and reacted in the microwave at 130 ℃ for 1h on a Biotage Smith synthesizer. The mixture was then cooled to room temperature and acidified to PH3, extracted with dichloromethane (3 × 10mL), the organic layer separated, and Na was added2SO4Dried, filtered and concentrated to give a white solid (28.4 mg). MS (M/z) 359.13[ M + H]+.

(II) Synthesis of 8- (2- (2- (2-hydroxyethoxy) ethoxy) ethylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 8- (2- (2- (2-hydroxyethoxy) ethoxy) ethylthio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione (25.0mg,0.07mmol) in MeOH (2mL) was added a solution of potassium hydrogen persulfate (171.7mg,0.28mmol) in H2O (2 mL). The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 10mL), the organic layer was separated, dried over Na2SO4, filtered, concentrated and purified by preparative TLC to give a white solid (2.3mg, 8.43%). 1H-NMR (400MHz, CDCl3): delta 4.32(s,3H),4.00-3.92(M,2H),3.94-3.90(M,4H),3.77-3.66(M,2H),3.58(s,3H),3.57-3.42(M,4H),3.41(s,3H). MS (M/z):391.12[ M + H ] +.

Compound 17: 8- (but-3-ynylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 8- (but-3-ynylthio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To compound 1(50.0mg,0.22mmol) and K2CO3(36.6mg,0.26mmol) in anhydrous DMF (2mL) was added 4-bromobut-1-yne (35.3mg,0.26mmol) and stirred under nitrogen at room temperature overnight. The reaction mixture was then poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used2SO4Dried, filtered and concentrated to give a white solid (52.3 mg). MS (M/z) 279.08[ M + H]+.

(II) Synthesis of 8- (but-3-ynylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 8- (but-3-ynylthio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione (25.0mg,0.09mmol) in MeOH (2mL) was added potassium hydrogen persulfate (221.1mg,0.36mmol) in H2O (2mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 5mL), the organic layer separated and washed with Na2SO4dry, filter, concentrate and purify by prep TLC to give a white solid (20.1mg, 72.3%).1H-NMR(400MHz,CDCl3):δ4.32(s,3H),3.72(t,J=7.2Hz,2H),3.57(s,3H),3.41(s,3H),2.86-2.81(m,2H),1.98(t,J=2.8Hz,1H).MS(m/z):311.07[M+H]+.

Compound 18: 8- (isopropylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 8- (isopropylsulfanyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To compound 1(50.0mg,0.22mmol) and K2CO3(36.6mg,0.26mmol) in anhydrous DMF (5mL) was added 2-iodopropane (45.1mg,0.26mmol) under nitrogenStir at room temperature overnight. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by preparative TLC (PE: EA) to give a white solid (34.7mg, 58.6%).

(II) Synthesis of 8- (isopropylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 8- (isopropylsulfanyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione (30.0mg) in MeOH (2mL) was added oxone (275.3mg) in H2O (2mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 5mL), the organic layer separated and washed with Na2SO4Dried, filtered, concentrated and purified by preparative TLC (PE: EA1:1) to give 31.5mg (93.75%) as a white solid.1H-NMR(400MHz,CDCl3):δ4.34(s,3H),3.77-.372(m,1H),3.57(s,3H),3.42(s,3H),1.43(d,J=7.2Hz,6H).MS(m/z):301.09[M+H]+.

Compound 19: 8- (benzo [ d ] [1,3] dioxol-5-ylmethylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 5- (bromomethyl) benzo [ d ] [1,3] dioxoles

To a solution of benzo [ d ] [1,3] dioxol-5-ylcarbinol (1g) in DCM at 0 deg.C was slowly added tribromophosphine (2.65 g). The mixture was then stirred at room temperature overnight. The solvent was removed and extracted with dichloromethane (3x 15mL) and concentrated to give a yellow solid (1.2g, 84.51%).

(II) Synthesis of 8- ((benzo [ d ] [1,3] dioxol-5-ylmethyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To compound 1(50.0mg,0.22mmol) and K2CO3(36.6mg,0.26mmol) in anhydrous DMF (5mL) was added 5- (bromomethyl) benzo [ d][1,3]Dioxole (57.1mg,0.26mmol) and stirred under nitrogen at room temperature overnight. The reaction mixture is then poured into water and washed with dichloro-benzeneMethane (3x 10mL) extraction, separation of the organic layer, Na2SO4Dried, filtered, concentrated and purified by preparative TLC (PE: EA1:1) to give a white solid (23.4mg, 29.4%).

(III) Synthesis of 8- ((benzo [ d ] [1,3] dioxol-5-ylmethyl) sulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To 8- ((benzo [ d)][1,3]Dioxolen-5-ylmethyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione (50.0mg) in DCM (5mL) was added m-CPBA (47.9mg) and the mixture was stirred at room temperature for 2H. The solvent was then removed and extracted with dichloromethane (3 x 5mL), the organic layer separated and washed with Na2SO4Drying, filtration, concentration and purification by preparative TLC (1:1) gave 20.3mg (38.4%) as a white solid.1H-NMR(400MHz,CDCl3):δ6.74-6.71(m,2H),6.59(s,1H),5.98(s,2H),4.58(s,2H),3.91(s,3H),3.62(s,3H),3.40(s,3H).MS(m/z):393.08[M+H]+.

Compound 20:1, 3, 7-trimethyl-8- (phenylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 1,3, 7-trimethyl-8- (phenylthio) -1H-purine-2, 6(3H,7H) -dione

To 8-chloro-1, 3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione (100.0mg,0.44mmol) and Cs2CO3(354.5mg,1.09mmol) in dry DMF (5mL) was added thiophenol (57.9mg,0.53mmol) and stirred at room temperature under nitrogen overnight. The reaction mixture was then poured into water and extracted with EA (3x 10mL), the organic layer was separated and Na was used2SO4Dry, filter, concentrate and purify by prep TLC to give a white solid (54.7mg, 41.3%).1H-NMR(400MHz,CDCl3):δ7.45-7.31(m,5H),3.93(s,3H),3.57(s,3H),3.40(s,3H).MS(m/z):303.08[M+H]+.

(II) Synthesis of 1,3, 7-trimethyl-8- (phenylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

To 1,3, 7-trimethyl-8- (phenylthio) -1H-purine-2, 6(3H,7H) -dione (28.0mg,0.09mmol) in MeOH (5mL) was added potassium hydrogen persulfate (227.9mg,0.37mmol) in H2O (5mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 10mL), the organic layer separated and washed with Na2SO4Dry, filter, concentrate and purify by prep TLC to give a white solid (14.3mg, 46.2%).1H-NMR(400MHz,CDCl3):δ8.08-8.05(m,2H),7.73-7.70(m,1H),7.63-7.59(m,2H),4.32(s,3H),3.52(s,3H),3.38(s,3H).MS(m/z):335.07[M+H]+.

Compound 21: 8- (Benzylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 8- (benzylthio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To Compounds 1(70.0mg) and K2CO3(bromomethyl) benzene (63.5mg) was added to a solution of (52.1mg) in anhydrous DMF (5mL) and stirred under nitrogen at room temperature overnight. The reaction mixture was then poured into water and extracted with EA (3x 10mL), the organic layer was separated and Na was used2SO4Drying, filtering and concentrating to obtain white solid.

(II) Synthesis of 8- (benzylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 8- (benzylsulfanyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione (50.0mg) in MeOH (2mL) was added oxone (388.96mg) in H2O (2mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 5mL), the organic layer separated and washed with Na2SO4Dried, filtered, concentrated and purified by preparative TLC (PE: EA1:1) to give 28.4mg (51.6%) as a white solid.1H-NMR(400MHz,CDCl3):δ7.40-7.32(m,3H),7.17-7.15(m,2H),4.65(s,2H),3.70(s,3H),3.63(s,3H),3.40(s,3H).MS(m/z):349.09[M+H]+.

Compound 22: 1,3, 7-trimethyl-8- (phenethylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 1,3, 7-trimethyl-8- (phenethylthio) -1H-purine-2, 6(3H,7H) -dione

To Compounds 1(70.0mg) and K2CO3(2-bromoethyl) benzene (68.95mg) was added to a solution of (52.8mg) in anhydrous DMF (5mL) and stirred under nitrogen at room temperature overnight. The reaction mixture was then poured into water and extracted with EA (3x 10mL), the organic layer was separated and Na was used2SO4Dried, filtered and concentrated to give a yellow oil.

(II) Synthesis of 1,3, 7-trimethyl-8- (Phenylethylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

To a solution of 1,3, 7-trimethyl-8- (phenethylthio) -1H-purine-2, 6(3H,7H) -dione (20mg) in MeOH (1mL) was added oxone (149.1mg) in H2O (1mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 5mL), the organic layer separated and washed with Na2SO4Dried, filtered, concentrated and purified by preparative TLC (PE: EA1:1) to give 6.3mg (28.77%) as a white solid.1H-NMR(400MHz,CDCl3):δ7.29-7.17(m,5H),4.29(s,3H),3.83(t,J=8.0Hz,2H),3.54(s,3H),3.41(s,3H),3.21(t,J=8.0Hz,2H).MS(m/z):363.10[M+H]+.

Compound 23: 1,3, 7-trimethyl-8- (naphthalen-2-ylmethylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 1,3, 7-trimethyl-8- ((naphthalen-2-ylmethyl) thio) -1H-purine-2, 6(3H,7H) -dione

To Compounds 1(50mg) and K2CO3(36.6mg) in anhydrous DMF (5mL) was added 2- (bromomethyl) naphthalene (58.7mg) and stirred under nitrogen at room temperature overnight. The reaction mixture was then poured into water and extracted with EA (3 x 5mL), the organic layer was separated and Na was used2SO4Drying, filtration, concentration and purification by preparative TLC (PE: EA1:1) gave 20.4mg (25.2%) as a white solid.

(II) Synthesis of 1,3, 7-trimethyl-8- ((naphthalen-2-ylmethyl) sulfonyl) -1H-purine-2, 6(3H,7H) -dione

To a solution of 1,3, 7-trimethyl-8- ((naphthalen-2-ylmethyl) thio) -1H-purine-2, 6(3H,7H) -dione (10mg) in MeOH (1mL) was added oxone (67.2mg) in H2O (1mL) solution. The mixture was then stirred at room temperature overnight. The solvent was then removed and extracted with dichloromethane (3x 5mL), the organic layer separated and washed with Na2SO4Dried, filtered, concentrated and purified by preparative TLC (PE: EA1:2) to give 8.2mg (75.5%) as a white solid.1H-NMR(400MHz,CDCl3):δ7.85-7.75(m,4H),7.54-7.52(m,2H),7.24-7.20(m,1H),4.85(s,2H),3.72(s,3H),3.65(s,3H),3.38(s,3H),.MS(m/z):399.10[M+H]+.

Compound 24: 1,3, 7-trimethyl-8- (naphthalen-1-ylmethylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 1,3, 7-trimethyl-8- ((naphthalen-1-ylmethyl) thio) -1H-purine-2, 6(3H,7H) -dione

To Compounds 1(50mg) and K2CO3(36.6mg) in anhydrous DMF (5mL) was added 1- (bromomethyl) naphthalene (58.7mg) and stirred under nitrogen at room temperature overnight. The reaction mixture was then poured into water and extracted with EA (3x 5mL), the organic layer was separated and Na was used2SO4Drying, filtration, concentration and purification by preparative TLC (PE: EA1:1) gave 18.5mg (22.85%) as a white solid.

(II) Synthesis of 1,3, 7-trimethyl-8- ((naphthalen-1-ylmethyl) sulfonyl) -1H-purine-2, 6(3H,7H) -dione

To a solution of 1,3, 7-trimethyl-8- ((naphthalen-1-ylmethyl) thio) -1H-purine-2, 6(3H,7H) -dione (10mg) in MeOH (1mL) was added oxone (67.2mg) in H2O (1mL) solution. The mixture was then stirred at room temperature overnight. The solvent was then removed and extracted with dichloromethane (3x 5mL), the organic layer separated and washed with Na2SO4Drying, filtering, concentrating andPurification by preparative TLC (PE: EA1:2) gave 5.2mg (48.2%) as a white solid.1H-NMR(400MHz,CDCl3):δ7.95-7.85(m,3H),7.49-7.43(m,3H),7.41-7.35(m,1H),5.16(s,2H),3.64(s,3H),3.41(s,3H),3.36(s,3H).MS(m/z):399.10[M+H]+.

Compound 25: 5- ((4S) -2-oxohexahydro-1H-thieno [3,4-d ] imidazol-4-yl) -N- (2- (2- (2- (4- (2- (1,3, 7-trimethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-8-ylsulfonyl) ethyl) -1H-1,2, 3-triazol-1-yl) ethoxy) ethyl) pentanamide

To compound 17(10.0mg,0.03mmol), N- (2- (2- (2- (2-azidoethoxy) ethoxy) ethyl) -5- ((4S) -2-oxohexahydro-1H-thieno [3, 4-d)]Imidazol-4-yl) pentanamide (17.2mg,0.04mmol) and CuI (0.31mg,0.002mmol) in dry THF (1mL) TEA (6.6mg,0.06mmol) was added and stirred at room temperature under nitrogen for 4h. The reaction mixture was then poured into water and extracted with dichloromethane (3x 3mL), the organic layer was separated and Na was used2SO4Drying, filtration, concentration and purification by preparative HPLC gave a white solid (4.3mg, 14.7%).1H-NMR(400MHz,DMSO-d6):δ7.91(s,1H),7.81(t,J=5.6Hz,1H),6.43(br,1H),4.40(t,J=5.2Hz,2H),4.31-4.28(m,1H),4.18(s,3H),4.13-4.10(m,1H),3.98(t,J=7.6Hz,2H),3.73(t,J=5.2Hz,2H),3.49-3.47(m,6H),3.47(s,3H),3.37(t,J=6.0Hz,2H),3.25(s,3H),3.16-3.09(m,6H),2.83-2.79(m,1H),2.58-2.53(m,1H),2.04(t,J=7.2Hz,2H),1.60-1.57(m,1H),1.49-1.43(m,4H),1.23-1.13(m,3H).MS(m/z):755.29[M+H]+.

Compound 26:8- (2-Fluorobenzylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 8- ((2-fluorobenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To compound 1(50mg) andK2CO3(36.6mg) in anhydrous DMF (5mL) was added 1- (bromomethyl) -2-fluorobenzene (50.2mg) and the mixture was stirred under nitrogen at room temperature overnight. The reaction mixture was then poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by preparative TLC (PE: EA1:1) to give a white solid (24.8mg, 33.6%).

(II) Synthesis of 8- ((2-Fluorobenzyl) sulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 8- ((2-fluorobenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione (10.0mg) in MeOH (3mL) was added oxone (73.6mg) in H2O (3mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 5mL), the organic layer separated and washed with Na2SO4Drying, filtration, concentration and purification by preparative TLC yielded 6.3mg (57.5%) as a white solid.1H-NMR(400MHz,CDCl3):δ7.43-7.05(m,4H),4.75(s,2H),3.89(s,3H),3.60(s,3H),3.41(s,3H).MS(m/z):367.08[M+H]+.

Compound 27: 8- (3-Fluorobenzylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 8- ((3-fluorobenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To Compounds 1(50mg) and K2CO3(36.6mg) in anhydrous DMF (5mL) was added 1- (bromomethyl) -3-fluorobenzene (50.2mg) and the mixture was stirred under nitrogen at room temperature overnight. The reaction mixture was then poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used 2SO4Dried, filtered, concentrated and purified by preparative TLC (PE: EA1:1) to give a white solid (27.4mg, 37.1%).

(II) Synthesis of 8- ((3-fluorobenzyl) sulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To 8- ((3-fluorobenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6 (3)To a solution of H,7H) -dione (10.0mg) in MeOH (3mL) was added oxone (73.6mg) in H2O (3mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 5mL), the organic layer separated and washed with Na2SO4drying, filtration, concentration and purification by preparative TLC yielded 7.1mg (64.78%) as a white solid.1H-NMR(400MHz,CDCl3):δ7.35-6.97(m,4H),4.69(s,2H),3.91(s,3H),3.62(s,3H),3.40(s,3H).MS(m/z):367.08[M+H]+.

Compound 28: 8- (4-Fluorobenzylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 8- ((4-fluorobenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To Compounds 1(50mg) and K2CO3(36.6mg) in anhydrous DMF (5mL) was added 1- (bromomethyl) -4-fluorobenzene (50.2mg) and the mixture was stirred under nitrogen at room temperature overnight. The reaction mixture was then poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by preparative TLC (PE: EA1:1) to give a white solid (29.3mg, 39.65%).

(II) Synthesis of 8- ((4-fluorobenzyl) sulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 8- ((4-fluorobenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione (10.0mg) in MeOH (3mL) was added oxone (73.6mg) in H2O (3mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 5mL), the organic layer separated and washed with Na2SO4Drying, filtration, concentration and purification by preparative TLC yielded 8.4mg (76.64%) as a white solid.1H-NMR(400MHz,CDCl3):δ7.22(q,J=8.8Hz,2H),7.12(t,J=8.4Hz,2H),4.67(s,2H),3.89(s,3H),3.62(s,3H),3.41(s,3H).MS(m/z):367.08[M+H]+.

Compound 29: 8- (2-chlorobenzylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 8- (2-chlorobenzylthio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To compound 1(40.0mg,0.17mmol) and K2CO3(29.3mg,0.21mmol) in anhydrous DMF (2mL) was added 1- (bromomethyl) -2-chlorobenzene (43.6mg,0.21mmol) and stirred under nitrogen at room temperature overnight. The reaction mixture was then poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used2SO4Dry, filter, concentrate and purify by prep TLC to give a white solid (18.4mg, 30.2%).1H-NMR(400MHz,CDCl3):δ7.40-7.38(m,2H),7.23-7.19(m,1H),7.19-7.15(m,1H),4.56(s,2H),3.72(s,3H),3.61(s,3H),3.38(s,3H).MS(m/z):351.06[M+H]+.

(II) Synthesis of 8- (2-chlorobenzylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 8- (2-chlorobenzylthio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione (15.0mg,0.04mmol) in MeOH (3mL) was added potassium hydrogen persulfate (105.2mg,0.17mmol) in H2O (3mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 5mL), the organic layer separated and washed with Na2SO4Dry, filter, concentrate and purify by prep TLC to give a white solid (11.2mg, 68.3%).1H-NMR(400MHz,CDCl3):δ7.41-7.39(m,4H),4.88(s,2H),3.84(s,3H),3.58(s,3H),3.41(s,3H).MS(m/z):383.05[M+H]+.

Compound 30: 8- (3-chlorobenzylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

(I) synthesis of 8- ((3-chlorobenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To compound 1(40.0mg,0.17mmol) and K2CO3(293mg,0.21mmol) in anhydrous DMF (2mL) was added 1- (bromomethyl) -3-chlorobenzene (43.6mg,0.21mmol) and stirred under nitrogen at room temperature overnight. The reaction mixture was then poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by preparative TLC (PE: EA1:1) to give a white solid (29.3mg, 48.1%). MS (M/z) 351.06[ M + H]+.

(II) Synthesis of 8- ((3-chlorobenzyl) sulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 8- ((3-chlorobenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione (15.0mg,0.04mmol) in MeOH (5mL) was added potassium hydrogen persulfate (140.2mg) in H2O (5mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 5mL), the organic layer separated and washed with Na2SO4Drying, filtration, concentration and purification by preparative TLC yielded 15.4mg (70.65%) as a white solid.1H-NMR(400MHz,CDCl3):δ7.39-7.26(m,3H),7.12-7.10(m,1H),4.67(s,2H),3.91(s,3H),3.63(s,3H),3.40(s,3H).MS(m/z):383.05[M+H]+.

Compound 31: 8- (4-chlorobenzylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 8- ((4-chlorobenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To compound 1(40.0mg,0.17mmol) and K2CO3To a solution of (29.3mg,0.21mmol) in anhydrous DMF (2mL) was added 1- (bromomethyl) -4-chlorobenzene (43.6mg,0.21mmol) and stirred under nitrogen at room temperature overnight. The reaction mixture was then poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by preparative TLC (PE: EA1:1) to give a white solid (28.3mg, 46.3%). MS (M/z) 351.06[ M + H]+.

(II) Synthesis of 8- ((4-chlorobenzyl) sulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 8- ((4-chlorobenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione (20.0mg,0.04mmol) in MeOH (5mL) was added potassium hydrogen persulfate (140.2mg) in H2O (5mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 5mL), the organic layer separated and washed with Na2SO4Drying, filtration, concentration and purification by preparative TLC yielded 16.1mg (73.85%) as a white solid.1H-NMR(400MHz,CDCl3):δ7.35(d,J=1.6Hz,1H),7.33(d,J=1.6Hz,1H),7.19(d,J=2.0Hz,1H),7.17(d,J=2.0Hz,1H),4.67(s,2H),3.91(s,3H),3.62(s,3H),3.40(s,3H).MS(m/z):383.05[M+H]+.

Compound 32: 8- (2-Bromobenzylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 8- ((2-bromobenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

to Compounds 1(50mg) and K2CO3(36.6mg) in anhydrous DMF (5mL) was added 1-bromo-2- (bromomethyl) benzene (66.1mg) and stirred under nitrogen at room temperature overnight. The reaction mixture was then poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by preparative TLC (PE: EA1:1) to give a white solid (30.1mg, 34.4%). MS (M/z) 351.06[ M + H]+.

(II) Synthesis of 8- ((2-bromobenzyl) sulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 8- ((2-bromobenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione (10.0mg) in MeOH (3mL) was added oxone (62.2mg) in H2O (3mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3 x 5mL), the organic layer separated and washed with Na2SO4Drying, filtration, concentration and purification by preparative TLC yielded 8.3mg (76.85%) as a white solid.1H-NMR(400MHz,CDCl3):δ7.59-7.27(m,4H),4.91(s,2H),3.84(s,3H),3.58(s,3H),3.41(s,3H).MS(m/z):427.00[M+H]+.

Compound 33: 8- (3-Bromobenzylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 8- ((3-bromobenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To Compounds 1(50mg) and K2CO3(36.6mg) in anhydrous DMF (5mL) was added 1-bromo-3- (bromomethyl) benzene (66.1mg) and stirred under nitrogen at room temperature overnight. The reaction mixture was then poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by preparative TLC (PE: EA1:1) to give a white solid (34.5mg, 39.5%).

(II) Synthesis of 8- ((3-bromobenzyl) sulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 8- ((3-bromobenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione (10.0mg) in MeOH (3mL) was added oxone (62.2mg) in H2O (3mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 5mL), the organic layer separated and washed with Na2SO4Drying, filtration, concentration and purification by preparative TLC yielded 5.6mg (51.99%) as a white solid.1H-NMR(400MHz,CDCl3):δ7.53-7.51(m,1H),7.41(s,1H),7.23-7.16(m,2H),4.66(s,2H),3.91(s,3H),3.63(s,3H),3.41(s,3H).MS(m/z):427.00[M+H]+.

Compound 34: 8- (4-Bromobenzylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 8- ((4-bromobenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To Compounds 1(50mg) and K2CO3(36.6mg) in anhydrous DMF (5mL) was added 1-bromo-4- (bromomethyl) benzene (66.1mg) and stirred under nitrogen at room temperature overnight. The reaction mixture was then poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by preparative TLC (PE: EA1:1) to give a white solid (30.1mg, 34.4%).

(II) Synthesis of 8- ((4-bromobenzyl) sulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 8- ((4-bromobenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione (10.0mg) in MeOH (3mL) was added oxone (62.2mg) in H2O (3mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 5mL), the organic layer separated and washed with Na2SO4Dry, filter, concentrate and purify by preparative TLC to give 7.2mg (66.7%) as a white solid.1H-NMR(400MHz,CDCl3):δ7.49(d,J=8.4Hz,2H),7.12(d,J=8.4Hz,2H),4.66(s,2H),3.92(s,3H),3.62(s,3H),3.40(s,3H).MS(m/z):427.00[M+H]+.

Compound 35: 8- (2-methoxybenzylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 8- ((2-methoxybenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To Compounds 1(50mg) and K2CO3(36.6mg) in anhydrous DMF (5mL) was added 1- (chloromethyl) -2-methoxybenzene (41.6mg) and stirred under nitrogen at room temperature overnight. The reaction mixture was then poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by preparative TLC (PE: EA1:1) to give a white solid (30.1mg, 39.2%).

(II) Synthesis of 8- ((2-methoxybenzyl) sulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To 8- ((2-methoxybenzyl)Yl) thio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione (30mg) in MeOH (2mL) was added potassium hydrogen persulfate (212.6mg) in H2O (2mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 5mL), the organic layer separated and washed with Na2SO4Drying, filtration, concentration and purification by preparative TLC yielded 18.2mg (56.87%) as a white solid.1H-NMR(400MHz,CDCl3):δ7.36(t,J=7.6Hz,1H),7.16(d,J=7.2Hz,1H),6.94(t,J=7.2Hz,1H),6.82(d,J=8.0Hz,1H),4.74(s,2H),3.74(s,3H),3.61(s,6H),3.40(s,3H).MS(m/z):379.10[M+H]+.

Compound 36: 8- (3-methoxybenzylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 8- ((3-methoxybenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To Compounds 1(50mg) and K2CO3(36.6mg) in anhydrous DMF (5mL) was added 1- (bromomethyl) -2-methoxybenzene (53.4mg) and the mixture was stirred under nitrogen at room temperature overnight. The reaction mixture was then poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by preparative TLC (PE: EA1:1) to give a white solid (40.4mg, 52.6%).

(II) Synthesis of 8- ((3-methoxybenzyl) sulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 8- ((3-methoxybenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione (40mg) in MeOH (3mL) was added potassium hydrogen persulfate (283.5mg) in H2O (3mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 5mL), the organic layer separated and washed with Na2SO4Drying, filtration, concentration and purification by preparative TLC yielded 20.4mg (46.68%) as a white solid.1H-NMR(400MHz,CDCl3):δ7.22(d,J=8.0Hz,1H),6.94(q,J=8.4Hz,1H),6.76(s,1H),6.67(d,J=7.6Hz,1H),4.62(s,2H),3.78(s,3H),3.76(s,3H),3.63(s,3H),3.40(s,3H).MS(m/z):379.10[M+H]+.

Compound 37: 8- (4-methoxybenzylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 8- ((4-methoxybenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To Compounds 1(50mg) and K2CO3(36.6mg) in anhydrous DMF (5mL) was added 1- (chloromethyl) -4-methoxybenzene (41.6mg) and stirred under nitrogen at room temperature overnight. The reaction mixture was then poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by preparative TLC (PE: EA1:1) to give a white solid (23.4mg, 30.5%).

(II) Synthesis of 8- ((4-methoxybenzyl) sulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 8- ((4-methoxybenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione (20mg) in MeOH (2mL) was added potassium hydrogen persulfate (141.7mg) in H2O (2mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3 x 5mL), the organic layer separated and washed with Na2SO4Drying, filtration, concentration and purification by preparative TLC yielded 14.1mg (64.7%) as a white solid.1H-NMR(400MHz,CDCl3):δ7.08(d,J=7.6Hz,2H),6.84(d,J=7.2Hz,2H),4.59(s,2H),3.79(s,3H),3.77(s,3H),3.63(s,3H),3.40(s,3H).MS(m/z):379.10[M+H]+

Compound 38: 1,3, 7-trimethyl-8- (2-nitrobenzylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 1,3, 7-trimethyl-8- ((2-nitrobenzyl) thio) -1H-purine-2, 6(3H,7H) -dione

To Compounds 1(30mg) and K2CO3To a solution of (21.4mg) in anhydrous DMF (5mL) was added 1- (bromomethyl) -2-nitrobenzene (34.3mg) and the mixture was stirred under nitrogen at room temperature overnight. The reaction mixture was then poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by preparative TLC (PE: EA1:1) to give a white solid (16.8mg, 35%).

(II) Synthesis of 1,3, 7-trimethyl-8- ((2-nitrobenzyl) sulfonyl) -1H-purine-2, 6(3H,7H) -dione

To a solution of 1,3, 7-trimethyl-8- ((2-nitrobenzyl) thio) -1H-purine-2, 6(3H,7H) -dione (15mg) in MeOH (3mL) was added oxone (102.1mg) in H2O (3mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 5mL), the organic layer separated and washed with Na2SO4Dry, filter, concentrate and purify by preparative TLC to give 8.3mg (50.92%) as a white solid.1H-NMR(400MHz,CDCl3):δ8.03(t,J=6.8Hz,1H),7.68-7.58(m,3H),5.29(s,2H),4.08(s,3H),3.55(s,3H),3.41(s,3H).MS(m/z):394.07[M+H]+

Compound 39: 8- (4- (1H-pyrrol-1-yl) benzylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 1- (4- (bromomethyl) phenyl) -1H-pyrrole

To a solution of (4- (1H-pyrrol-1-yl) phenyl) methanol (100mg) in DCM at 0 deg.C was added tribromophosphine (232mg) slowly. The mixture was then stirred at room temperature overnight. The solvent was removed and extracted with dichloromethane (3x 5mL) and concentrated to give a yellow solid (63mg, 46.5%).

(II) Synthesis of 8- ((4- (1H-pyrrol-1-yl) benzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To compound 1(50.0mg,0.22mmol) and K2CO3To a solution of (36.6mg,0.26mmol) in anhydrous DMF (5mL) was added 1- (4- (bromomethyl) phenyl) -1H-pyrrole (62).6mg,0.26mmol) and stirred at room temperature under nitrogen overnight. The reaction mixture was then poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by preparative TLC (PE: EA1:1) to give a white solid (10.8mg, 12.81%).

(III) Synthesis of 8- ((4- (1H-pyrrol-1-yl) benzyl) sulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 8- ((4- (1H-pyrrol-1-yl) benzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione (10mg) in MeOH (3mL) was added potassium hydrogen persulfate (64.5mg) in H2O (3mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 5mL), the organic layer separated and washed with Na2SO4Dried, filtered, concentrated and purified by preparative TLC (PE: EA1:1) to give 3.8mg (32.5%) as a white solid.1H-NMR(400MHz,CDCl3):δ8.24(d,J=8.4Hz,2H),7.55(d,J=8.4Hz,2H),7.26-7.25(m,4H),4.87(s,2H),4.08(s,3H),3.62(s,3H),3.41(s,3H).MS(m/z):414.12[M+H]+

Compound 40:1, 3, 7-trimethyl-8- (3- (3- (trifluoromethyl) -3H-diazacyclopropen-3-yl) benzylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 1,3, 7-trimethyl-8- (3- (3- (trifluoromethyl) -3H-diazacyclopropen-3-yl) benzylthio) -1H-purine-2, 6(3H,7H) -dione

To compound 1(50.0mg,0.22mmol) and K2CO3(37.8mg,0.27mmol) in dry DMF (3mL) was added 3- (3- (bromomethyl) phenyl) -3- (trifluoromethyl) -3H-diazacyclopropene (73.8mg,0.27mmol) and stirred at room temperature under nitrogen overnight protected from light. The reaction mixture was then poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used2SO4Dry, filter, concentrate and purify by silica gel chromatography (PE: EA ═ 4:1 to DCM: MeOH ═ 10:1) to give a white solid (12.3mg, 13.1%).1H-NMR(400MHz,CDCl3):δ7.43(d,J=6.4Hz,1H),7.34(t,J=8.0Hz,1H),7.21(s,1H),7.11(d,J=8.0Hz,1H),4.43(s,2H),3.74(s,3H),3.61(s,3H),3.38(s,3H).MS(m/z):425.09[M+H]+.

(II) Synthesis of 1,3, 7-trimethyl-8- (3- (3- (trifluoromethyl) -3H-diazacyclopropen-3-yl) benzylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

To a solution of 1,3, 7-trimethyl-8- (3- (3- (trifluoromethyl) -3H-diazacyclopropen-3-yl) benzylthio) -1H-purine-2, 6(3H,7H) -dione (10mg,0.02mmol) in DCM (3mL) was added m-CPBA (6.2mg,0.03 mmol). The mixture was then stirred at room temperature for 2h in the dark. The solvent was then removed and extracted with dichloromethane (3x 10mL), the organic layer separated and washed with Na2SO4Dry, filter, concentrate and purify by prep TLC to give a white solid (5.7mg, 53.4%).1H-NMR(400MHz,CDCl3):δ7.45-7.39(m,2H),7.23(t,J=7.2Hz,1H),6.94(s,1H),4.70(s,2H),3.87(s,3H),3.64(s,3H),3.40(s,3H).MS(m/z):457.08[M+H]+.

Compound 41: 1,3, 7-trimethyl-8- (4- (3- (trifluoromethyl) -3H-diazacyclopropen-3-yl) benzylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

Compound was synthesized in a similar manner to compound 40 to give 8.4mg (77.7%) as a white solid.1H-NMR(400MHz,CDCl3):δ7.34(d,J=8.4Hz,2H),7.19(d,J=8.0Hz,2H),4.74(s,2H),3.97(s,3H),3.61(s,3H),3.41(s,3H).MS(m/z):457.08[M+H]+

Compound 42: 2- ((1,3, 7-trimethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-8-ylsulfonyl) methyl) benzonitrile

(I) Synthesis of 2- ((1,3, 7-trimethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-8-ylthio) methyl) benzonitrile

To compound 1(100.0mg,0.44mmol) and K2CO3(73.2mg,0.53mmol) of Anhydrous DTo the MF (5mL) solution was added 2- (bromomethyl) benzonitrile (103.1mg,0.53mmol) and stirred at room temperature under nitrogen overnight. The reaction mixture was then poured into water and extracted with EA (3x 10mL), the organic layer was separated and Na was used2SO4Dry, filter, concentrate and purify by preparative TLC to give a white solid (80.9mg, 53.62%).1H-NMR(400MHz,CDCl3):δ7.65-7.35(m,4H),4.62(s,2H),3.75(s,3H),3.57(s,3H),3.35(s,3H).MS(m/z):342.09[M+H]+.

(II) Synthesis of 2- ((1,3, 7-trimethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-8-ylsulfonyl) methyl) benzonitrile

To a solution of 2- ((1,3, 7-trimethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-8-ylthio) methyl) benzonitrile (15.0mg,0.04mmol) in MeOH (2mL) was added a solution of potassium hydrogen persulfate (108.2mg,0.17mmol) in H2O (5 mL). The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 5mL), the organic layer separated and washed with Na2SO4Dry, filter, concentrate and purify by prep TLC to give a white solid (7.2mg, 43.91%). 1H-NMR (400MHz, CDCl3): delta 7.71-7.53(M,4H),4.94(s,2H),4.01(s,3H),3.58(s,3H),3.41(s,3H). MS (M/z):374.08[ M + H]+.

Compound 43: 3- ((1,3, 7-trimethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-8-ylsulfonyl) methyl) benzonitrile

(I) Synthesis of 3- ((1,3, 7-trimethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-8-ylthio) methyl) benzonitrile

To compound 1(100.0mg,0.44mmol) and K2CO3(73.2mg,0.53mmol) in dry DMF (5mL) was added 3- (bromomethyl) benzonitrile (103.1mg,0.53mmol) and stirred under nitrogen at room temperature overnight. The reaction mixture was then poured into water and extracted with EA (3x 10mL), the organic layer was separated and Na was used2SO4Dry, filter, concentrate and purify by prep TLC to give a white solid (68.3mg, 45.3%).

(II) Synthesis of 3- ((1,3, 7-trimethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-8-ylsulfonyl) methyl) benzonitrile

To a solution of 3- ((1,3, 7-trimethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-8-ylthio) methyl) benzonitrile (15.0mg,0.04mmol) in MeOH (2mL) was added potassium hydrogen persulfate (108.2mg,0.17mmol) in H2O (5mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 5mL), the organic layer separated and washed with Na2SO4Drying, filtration, concentration and purification by preparative TLC yielded 10.7mg (65.3%) as a white solid.1H-NMR(400MHz,CDCl3):δ7.71-7.51(m,4H),4.79(s,2H),4.06(s,3H),3.62(s,3H),3.41(s,3H).MS(m/z):374.08[M+H]+.

Compound 44: 4- ((1,3, 7-trimethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-8-ylsulfonyl) methyl) benzonitrile

(I) synthesis of 4- ((1,3, 7-trimethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-8-ylthio) methyl) benzonitrile

To compound 1(50mg,0.22mmol) and K2CO3(37.8mg) in anhydrous DMF (2mL) was added 4- (bromomethyl) benzonitrile (51.2mg) and stirred under nitrogen at room temperature overnight. The reaction mixture was then poured into water and extracted with EA (3x 10mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:1) to give a white solid (23mg, 30.5%).

(II) Synthesis of 4- ((1,3, 7-trimethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-8-ylsulfonyl) methyl) benzonitrile

To a solution of 4- ((1,3, 7-trimethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-8-ylthio) methyl) benzonitrile (15.0mg,0.04mmol) in MeOH (2mL) was added a solution of potassium hydrogen persulfate (108.4mg,0.17mmol) in H2O (5 mL). The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 5mL), the organic layer separated and washed with Na2SO4Drying, filtering, concentrating and passing through to produce TLC purification (PE: EA1:2) gave 11.0mg (67.1%) as a white solid. 1H-NMR (400MHz, CDCl3): delta 7.68(d, J-8.0 Hz,2H),7.46(d, J-8.0 Hz,2H),4.81(s,2H),4.04(s,3H),3.61(s,3H),3.41(s, 3H): MS (M/z):374.08[ M + H ], (M + H): 374.08]+

Compound 45: 2- ((1,3, 7-trimethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-8-ylsulfonyl) methyl) benzamide

Compound 2- ((1,3, 7-trimethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-8-ylthio) methyl) benzonitrile is synthesized in the manner described for compound 42.

(I) Synthesis of 2- ((1,3, 7-trimethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-8-ylthio) methyl) benzamide

To a mixture of 2- ((1,3, 7-trimethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-8-ylthio) methyl) benzonitrile (15.0mg,0.04mmol) and K2CO3(0.85mg,0.006mmol) in DMSO (2mL) was added H slowly2O2(2.3mg,0.06mmol) and stirred at room temperature under nitrogen overnight. The reaction mixture was then poured into water and extracted with dichloromethane (3x 5mL), the organic layer was separated and Na was used2SO4Dried, filtered and concentrated to give a white solid (15.1 mg). MS (M/z) 360.11[ M + H]+.

(II) Synthesis of 2- ((1,3, 7-trimethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-8-ylsulfonyl) methyl) benzamide

To a solution of 2- ((1,3, 7-trimethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-8-ylthio) methyl) benzamide (10.0mg,0.03mmol) in MeOH (2mL) was added potassium hydrogen persulfate (68.5mg,0.11mmol) in H2O (2mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 5mL), the organic layer separated and washed with Na2SO4Dry, filter, concentrate and purify by prep TLC to give a white solid (8.7mg, 80.55%).1H-NMR(400MHz,DMSO-d6):δ7.77-7.37(m,4H),7.36(br,2H),5.38(s,2H),3.75(s,3H),3.38(s,3H),3.25(s,3H).MS(m/z):392.10[M+H]+.

Compound 46: 3- ((1,3, 7-trimethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-8-ylsulfonyl) methyl) benzamide

This compound was synthesized in a similar manner to compound 45 to give 7.3mg (67.6%) as a white solid.1H-NMR(400MHz,CDCl3):δ7.78-7.76(m,2H),7.46-7.45(m,2H),4.76(s,2H),3.91(s,3H),3.64(s,3H),3.40(s,3H).MS(m/z):392.10[M+H]+.

Compound 47: 4- ((1,3, 7-trimethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-8-ylsulfonyl) methyl) benzamide

This compound was synthesized in a similar manner to compound 45 to give 10.3mg (95.4%) as a white solid.1H-NMR(400MHz,CDCl3):δ7.80(d,J=8.0Hz,2H),7.37(d,J=8.4Hz,2H),4.77(s,2H),3.93(s,3H),3.62(s,3H),3.41(s,3H).MS(m/z):392.10[M+H]+.

Compound 48: 1,3, 7-trimethyl-8- (pyridin-2-ylmethylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

Compound 49: 2- ((1,3, 7-trimethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-8-ylsulfonyl) methyl) pyridine 1-oxide

(I) Synthesis of 1,3, 7-trimethyl-8- (pyridin-2-ylmethylthio) -1H-purine-2, 6(3H,7H) -dione

To compound 1(500.0mg,2.21mmol) and K2CO3(366.4mg,2.65mmol)To a solution of 2- (bromomethyl) pyridine (674.1mg,2.66mmol) in anhydrous DMF (25mL) was added and stirred under nitrogen at room temperature overnight. The reaction mixture was then poured into water and extracted with EA (3 × 50mL), the organic layer was separated and Na was used2SO4Drying, filtration, concentration and recrystallization in EA: PE ═ 1:5 gave a white solid (307.4 mg). MS (M/z) 318.09[ M + H]+.

(II) Synthesis of 1,3, 7-trimethyl-8- (pyridin-2-ylmethylsulfonyl) -1H-purine-2, 6(3H,7H) -dione and 2- ((1,3, 7-trimethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-8-ylsulfonyl) methyl) pyridine 1-oxide

To a solution of 1,3, 7-trimethyl-8- (pyridin-2-ylmethylthio) -1H-purine-2, 6(3H,7H) -dione (50.0mg,0.16mmol) in MeOH (3mL) was added oxone (387.9mg,0.64mmol) in H2O (3mL) solution. The mixture was then stirred at room temperature for 30 min. The solvent was then removed and extracted with dichloromethane (3x 5mL), the organic layer separated and washed with Na2SO4Drying, filtration, concentration and purification by preparative TLC afforded compound 48 as a white solid (14.2mg, 25.5%).1H-NMR(400MHz,CDCl3):δ8.50-8.48(m,1H),7.79-7.76(m,1H),7.51-7.33(m,2H),4.85(s,2H),3.88(s,3H),3.58(s,3H),3.41(s,3H).MS(m/z):350.08[M+H]+.

Compound 498.3 mg (75.4%) was obtained as a white solid.1H-NMR(400MHz,CDCl3):δ8.11-8.10(m,1H),7.61-7.60(m,1H),7.34-7.33(m,2H),5.09(s,2H),4.26(s,3H),3.47(s,3H),3.41(s,3H).MS(m/z):366.08[M+H]+.

Compound 50: 1,3, 7-trimethyl-8- (pyridin-3-ylmethylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

Compound 51: 3- ((1,3, 7-trimethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-8-ylsulfonyl) methyl) pyridine 1-oxide

(I) Synthesis of 1,3, 7-trimethyl-8- (pyridin-3-ylmethylthio) -1H-purine-2, 6(3H,7H) -dione

To Compounds 1(30mg) and K2CO3to a solution of (21.4mg) in anhydrous DMF (25mL) was added 3- (bromomethyl) pyridine (40.3mg) and the mixture was stirred under nitrogen at room temperature overnight. The reaction mixture was then poured into water and extracted with EA (3 × 50mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by preparative TLC (DCM: MeOH 10:1) to give a white solid (16.2mg, 38.6%). MS (M/z) 318.09[ M + H]+.

(II) Synthesis of 1,3, 7-trimethyl-8- (pyridin-3-ylmethylsulfonyl) -1H-purine-2, 6(3H,7H) -dione and 3- ((1,3, 7-trimethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-8-ylsulfonyl) methyl) pyridine 1-oxide

to a solution of 1,3, 7-trimethyl-8- (pyridin-3-ylmethylthio) -1H-purine-2, 6(3H,7H) -dione (10mg) in MeOH (2mL) was added oxone (77.6mg) in H2O (2mL) solution. The mixture was then stirred at room temperature for 30 min. The solvent was then removed and extracted with dichloromethane (3x 5mL), the organic layer separated and washed with Na2SO4Drying, filtration, concentration and purification by preparative TLC (DCM: MeOH 10:1) gave compound 504.3mg (37.3%) as a white solid.1H-NMR(400MHz,CDCl3):δ8.63(s,1H),8.46-8.45(m,1H),7.73-7.71(m,1H),7.34-7.33(m,1H),4.77(s,2H),3.97(s,3H),3.60(s,3H),3.39(s,3H).MS(m/z):350.08[M+H]+.

Compound 513.7 mg (35.7%) was obtained as a white solid.1H-NMR(400MHz,CDCl3):δ8.26(s,1H),8.21-8.20(m,1H),7.33-7.32(m,2H),4.75(s,2H),4.18(s,3H),3.60(s,3H),3.40(s,3H).MS(m/z):366.08[M+H]+.

Compound 52: 1,3, 7-trimethyl-8- (pyridin-4-ylmethylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

Compound 53: 4- ((1,3, 7-trimethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-8-ylsulfonyl) methyl) pyridine 1-oxide

(I) Synthesis of 1,3, 7-trimethyl-8- (pyridin-4-ylmethylthio) -1H-purine-2, 6(3H,7H) -dione

To Compounds 1(30mg) and K2CO3to a solution of (21.4mg) in anhydrous DMF (25mL) was added 3- (bromomethyl) pyridine (40.3mg) and the mixture was stirred under nitrogen at room temperature overnight. The reaction mixture was then poured into water and extracted with EA (3 × 50mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by preparative TLC (DCM: MeOH 10:1) to give a white solid (15mg, 35.7%). MS (M/z) 318.09[ M + H]+.

(II) Synthesis of 1,3, 7-trimethyl-8- (pyridin-4-ylmethylsulfonyl) -1H-purine-2, 6(3H,7H) -dione and 4- ((1,3, 7-trimethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-8-ylsulfonyl) methyl) pyridine 1-oxide

to a solution of 1,3, 7-trimethyl-8- (pyridin-3-ylmethylthio) -1H-purine-2, 6(3H,7H) -dione (10mg) in MeOH (2mL) was added oxone (77.6mg) in H2O (2mL) solution. The mixture was then stirred at room temperature for 30 min. The solvent was then removed and extracted with dichloromethane (3x 5mL), the organic layer separated and washed with Na2SO4Drying, filtration, concentration and purification by preparative TLC (DCM: MeOH 10:1) gave compound 524.2mg (35.2%) as a white solid. 1H-NMR (400MHz, CDCl3): delta 8.65(d, J ═ 4.8Hz,2H),7.27(d, J ═ 5.2Hz,2H),4.74(s,2H),4.02(s,3H),3.61(s,3H),3.40(s, 3H): MS (M/z):350.08[ M + H ]: 350.08]+.

533.2 mg (30.4%) of compound are obtained as a white solid. 1H-NMR (400MHz, CDCl3): delta 8.20(d, J ═ 6.8Hz,2H),7.31(d, J ═ 6.8Hz,2H),4.76(s,2H),4.19(s,3H),3.59(s,3H),3.41(s,3H). MS (M/z):366.08[ M + H ] +.

Compound 54: 8- (2, 3-dichlorobenzylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 8- (2, 3-dichlorobenzylthio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To compound 1(50.0mg,0.22mmol) and K2CO3(36.6mg,0.26mmol) in dry DMF (5mL) was added 1- (bromomethyl) -2, 3-dichlorobenzene (63.7mg,0.26mmol) and stirred at room temperature under nitrogen overnight. The reaction mixture was then poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by preparative TLC to give a white solid (14.3 mg). MS (M/z) 385.02[ M + H]+.

(II) Synthesis of 8- (2, 3-dichlorobenzylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 8- (2, 3-dichlorobenzylthio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione (10.0mg,0.03mmol) in MeOH (3mL) was added oxone (63.9mg,0.10mmol) in H2O (3mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 5mL), the organic layer separated and washed with Na2SO4Dry, filter, concentrate and purify by prep TLC to give a white solid (4.2mg, 38.9%).1H-NMR(400MHz,CDCl3):δ7.54-7.27(m,3H),4.97(s,2H),4.02(s,3H),3.57(s,3H),3.42(s,3H).MS(m/z):417.01[M+H]+.

Compound 55: 8- (2, 5-dichlorobenzylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 8- (2, 5-dichlorobenzylthio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To compound 1(50.0mg,0.22mmol) and K2CO3(36.6mg,0.26mmol) in dry DMF (5mL) was added 1- (bromomethyl) -2, 5-dichlorobenzene (63.7mg,0.26mmol) and stirred at room temperature under nitrogen overnight. The reaction mixture was then poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used2SO4Drying, filtering, concentrating and making into tabletPreparative TLC purification (PE: EA1:1) gave a white solid (20.1mg, 23.6%). MS (M/z) 385.02[ M + H]+.

(II) Synthesis of 8- (2, 5-dichlorobenzylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 8- (2, 5-dichlorobenzylthio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione (10.0mg,0.03mmol) in MeOH (3mL) was added oxone (63.9mg,0.10mmol) in H2o (3mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 5mL), the organic layer separated and washed with Na2SO4Drying, filtration, concentration and purification by preparative TLC (PE: EA1:1) gave 8.3mg (76.85%) as a white solid.1H-NMR(400MHz,CDCl3):δ7.48(s,1H),7.35-7.34(m,2H),4.88(s,2H),4.03(s,3H),3.58(s,3H),3.42(s,3H).MS(m/z):417.01[M+H]+.

Compound 56: 8- (3, 5-Dichlorobenzylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 8- (3, 5-dichlorobenzylthio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To compound 1(50.0mg,0.22mmol) and K2CO3To a solution of (36.6mg,0.26mmol) in anhydrous DMF (5mL) was added 1, 3-dichloro-5- (chloromethyl) benzene (51.8mg,0.26mmol) and stirred under nitrogen at room temperature overnight. The reaction mixture was then poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by preparative TLC (PE: EA1:1) to give a white solid (25.3mg, 29.7%). MS (M/z) 385.02[ M + H]+.

(II) Synthesis of 8- (3, 5-dichlorobenzylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of- (3, 5-dichlorobenzylthio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione (20mg) in MeOH (5mL) was added potassium hydrogen persulfate (127.7mg) in H2O (5mL) solution. The mixture was then stirred at room temperature for 5 h. Then removing the solvent andExtraction with dichloromethane (3x 5mL), separation of the organic layer, Na2SO4Drying, filtration, concentration and purification by preparative TLC (PE: EA1:1) gave 11.3mg (52.3%) as a white solid.1H-NMR(400MHz,CDCl3):δ7.39(s,1H),7.24(s,2H),4.68(s,2H),4.08(s,3H),3.62(s,3H),3.41(s,3H).MS(m/z):417.01[M+H]+.

Compound 57: 8- (2, 6-dichlorobenzylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 8- (2, 6-dichlorobenzylthio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To compound 1(50.0mg,0.22mmol) and K2CO3(36.6mg,0.26mmol) in dry DMF (5mL) was added 2- (bromomethyl) -1, 3-dichlorobenzene (63.7mg,0.26mmol) and stirred at room temperature under nitrogen overnight. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by preparative TLC (PE: EA1:1) to give a white solid (20.1mg, 23.6%). MS (M/z) 385.02[ M + H]+.

(II) Synthesis of 8- (2, 6-dichlorobenzylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

to a solution of 8- (2, 6-dichlorobenzylthio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione (10mg) in MeOH (3mL) was added potassium hydrogen persulfate (63.9mg) in H2O (3mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 5mL), the organic layer separated and washed with Na2SO4Dried, filtered, concentrated and purified by preparative TLC (PE: EA1:1) to give 7.9mg (73.1%) as a white solid.1H-NMR(400MHz,CDCl3):δ7.40-7.25(m,3H),5.16(s,2H),3.99(s,3H),3.57(s,3H),3.43(s,3H).MS(m/z):417.01[M+H]+.

Compound 58: 8- (2, 5-Dimethoxybenzylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 8- ((2, 5-dimethoxybenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To Compounds 1(50.0mg) and K2CO3(36.6mg) in dry DMF (5mL) was added 2- (bromomethyl) -1, 4-dimethoxybenzene (61.4mg) and stirred under nitrogen at room temperature overnight. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by preparative TLC (PE: EA1:1) to give a white solid (26.4mg, 31.8%).

(II) Synthesis of 8- ((2, 5-Dimethoxybenzyl) sulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 8- (2, 6-dichlorobenzylthio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione (10mg) in MeOH (5mL) was added potassium hydrogen persulfate (65.4mg) in H2O (5mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 5mL), the organic layer separated and washed with Na2SO4Drying, filtration, concentration and purification by preparative TLC (PE: EA1:1) gave 8.1mg (75%) as a white solid.1H-NMR(400MHz,CDCl3):δ6.87-6.75(m,3H),4.70(s,2H),3.82(s,3H),3.72(s,3H),3.59(s,3H),3.54(s,3H),3.39(s,3H).MS(m/z):409.11[M+H]+.

Compound 59: 8- (3, 5-Dimethoxybenzylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

This compound was synthesized in a similar manner to compound 58 to finally obtain 8.2mg (75.1%) as a white solid.1H-NMR(400MHz,CDCl3):δ6.45(s,1H),6.31(s,2H),4.58(s,2H),3.87(s,3H),3.72(s,6H),3.62(s,3H),3.40(s,3H).MS(m/z):409.11[M+H]+.

Compound 60: 8- (5-bromo-2-methoxybenzylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 4-bromo-1- (bromomethyl) -2-methoxybenzene

To a solution of (4-bromo-2-methoxyphenyl) methanol (100mg) in DME at 0 deg.C was added tribromophosphine (124.7mg) slowly. The mixture was then stirred at room temperature overnight. The solvent was removed and extracted with EA (3 × 10mL) and concentrated to give a white solid (136.3mg, 72%).

(II) Synthesis of 8- ((4-bromo-2-methoxybenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To compound 1(50.0mg,0.22mmol) and K2CO3To a solution of (36.6mg,0.26mmol) in anhydrous DMF (5mL) was added 4-bromo-1- (bromomethyl) -2-methoxybenzene (75mg,0.26mmol) and stirred under nitrogen at room temperature overnight. The reaction mixture was then poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by column chromatography (PE: EA1:1) to give a white solid (35mg, 37%).

(III) Synthesis of 8- ((4-bromo-2-methoxybenzyl) sulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 8- ((4-bromo-2-methoxybenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione (40mg) in MeOH (5mL) was added oxone (650mg) in H2O (5mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with EA (3x 10mL), the organic layer was separated and Na was added2SO4drying, filtration, concentration and purification by silica gel column chromatography gave 10mg (31%) as a white solid.1H-NMR(400MHz,CDCl3):δ7.107(d,J=2.0Hz,1H),7.09(s,1H),7.009(d,J=2.0Hz,1H),4.73(s,2H),3.95(s,3H),3.68(s,3H),3.60(s,3H),3.42(s,3H).MS(m/z):458.3[M+H]+.

Compound 61: 8- (2, 3-Dimethoxybenzylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 1- (bromomethyl) -2, 3-dimethoxybenzene

To a solution of (2, 3-dimethoxyphenyl) methanol (500mg) in DME at 0 deg.C was added tribromophosphine (805mg) slowly. The mixture was then stirred at room temperature overnight. The solvent was removed and extracted with EA (3 × 10mL) and concentrated to give a white solid (176.8mg, 26%).

(II) Synthesis of 8- ((2, 3-Dimethoxybenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To compound 1(50.0mg,0.22mmol) and K2CO3(62mg,0.44mmol) in dry DMF (5mL) was added 1- (bromomethyl) -2, 3-dimethoxybenzene (77mg,0.33mmol) and stirred under nitrogen at room temperature overnight. The reaction mixture was then poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by column chromatography (PE: EA1:1) to give a white solid (17mg, 20.5%).

(III) Synthesis of 8- ((2, 3-Dimethoxybenzyl) sulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 8- ((2, 3-dimethoxybenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione (17mg,0.045mmol) in MeOH (1mL) was added potassium hydrogen persulfate (280mg,0.45mmol) in H2O (1mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with EA (3x 10mL), the organic layer was separated and Na was added2SO4Dried, filtered, concentrated and purified by silica gel column chromatography (PE: EA1:1) to give 2mg (10.9%) as a white solid.1H-NMR(400MHz,CDCl3):δ7.73-7.71(m,1H),7.55-7.52(m,1H),6.69-6.67(m,1H),4.79(s,2H),3.88(s,3H),3.86(s,3H),3.85(s,3H),3.63(s,3H),3.43(s,3H).MS(m/z):409.4[M+H]+.

Compound 62: 8- (2, 6-Dimethoxybenzylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 2- (bromomethyl) -1, 3-dimethoxybenzene

To a solution of 1, 3-dimethoxy-2-methylbenzene (500mg,3.29mmol) in CCl4To the solution (10mL) was added NBS (585mg,3.29mmol) and AIBN (150mg) slowly. The mixture was then heated to 80 ℃ for 20 minutes until a white solid floated on the surface. The mixture was filtered and the filtrate was concentrated to give a residue. The residue was diluted and extracted with EA (3 × 10mL) and concentrated to give the crude product (purple solid).

(II) Synthesis of 8- ((2, 6-Dimethoxybenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To compound 1(50.0mg,0.22mmol) and K2CO3(62mg,0.44mmol) in dry DMF (5mL) was added 2- (bromomethyl) -1, 3-dimethoxybenzene (77mg,0.33mmol) and stirred under nitrogen at room temperature overnight. The reaction mixture was then poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by column chromatography (PE: EA1:1) to give a white solid (7mg, 8%).

(III) Synthesis of 8- ((2, 6-Dimethoxybenzyl) sulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 8- ((2, 6-dimethoxybenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione (7mg,0.019mmol) in MeOH (1mL) was added potassium hydrogen persulfate (120mg,0.2mmol) in H2O (1mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with EA (3x 10mL), the organic layer was separated and Na was added2SO4Dried, filtered, concentrated and purified by silica gel column chromatography (PE: EA1:1) to give 2mg (26.3%) as a white solid.1H-NMR(400MHz,CDCl3):δ7.308(t,J=8.4Hz,1H),6.514(d,J=8.4Hz,2H),4.82(s,2H),3.81(s,3H),3.64(s,6H),3.62(s,3H),3.42(s,3H).MS(m/z):409.4[M+H]+.

Compound 63: 8- (3-chloro-2-methoxybenzylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 1- (bromomethyl) -3-chloro-2-methoxybenzene

To a solution of 1-chloro-2-methoxy-3-methylbenzene (300mg,1.92mmol) in CCl4To the solution (10mL) was added NBS (350mg,1.92mmol) and AIBN (150mg) slowly. The mixture was then heated to 90 ℃ for 5 hours until a white solid floated on the surface. The mixture was filtered and the filtrate was concentrated to give a residue. The residue was diluted and extracted with EA (3 × 10mL) and concentrated to give the crude product (yellow solid).

(II) Synthesis of 8- ((3-chloro-2-methoxybenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To compound 1(50.0mg,0.22mmol) and K2CO3(62mg,0.44mmol) in dry DMF (5mL) was added 1- (bromomethyl) -3-chloro-2-methoxybenzene (80mg,0.33mmol) and stirred under nitrogen at room temperature overnight. The reaction mixture was then poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used2SO4dried, filtered, concentrated and purified by column chromatography (PE: EA1:1) to give a white solid (18mg, 21.39%).

(III) Synthesis of 8- ((3-chloro-2-methoxybenzyl) sulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 8- ((2, 6-dimethoxybenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione (18mg,0.048mmol) in MeOH (1mL) was added potassium hydrogen persulfate (300mg,0.48mmol) in H2O (1mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with EA (3x 10mL), the organic layer was separated and Na was added2SO4Dried, filtered, concentrated and purified by silica gel column chromatography (PE: EA1:1) to give 12mg (61.5%) as a white solid.1H-NMR(400MHz,CDCl3):δ7.432(dd,J=1.6Hz,J=8.0Hz,1H),7.147(dd,J=1.6Hz,J=8.0Hz,1H),7.06(t,J=8.0Hz,1H),4.81(s,2H),4.00(s,3H),3.90(s,3H),3.60(s,3H),3.43(s,3H).MS(m/z):413.8[M+H]+.

Compound 64: 8- (4-bromo-3-chlorobenzylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 1-bromo-4- (bromomethyl) -2-chlorobenzene

To CCl of 1-bromo-2-chloro-4-methylbenzene (500mg,2.43mmol)4To the solution (10mL) was added NBS (433mg,2.43mmol) and AIBN (200mg) slowly. The mixture was then heated to 90 ℃ for 5 hours until a white solid floated on the surface. The mixture was filtered and the filtrate was concentrated to give a residue. The residue was diluted and extracted with EA (3 × 10mL) and concentrated to give the crude product (yellow solid) 930 mg.

(II) Synthesis of 8- ((4-bromo-3-chlorobenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To compound 1(50.0mg,0.22mmol) and K2CO3(62mg,0.44mmol) in anhydrous DMF (5mL) was added 1-bromo-4- (bromomethyl) -2-chlorobenzene (95mg,0.33mmol) and stirred under nitrogen at room temperature overnight. The reaction mixture was then poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by column chromatography (PE: EA1:1) to give a white solid (20.5mg, 21.6%).

(III) Synthesis of 8- ((4-bromo-3-chlorobenzyl) sulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 8- ((4-bromo-3-chlorobenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione (20mg,0.048mmol) in MeOH (1mL) was added oxone (290mg,0.48mmol) in H2O (1mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with EA (3x 10mL), the organic layer was separated and Na was added2SO4Dried, filtered, concentrated and purified by silica gel column chromatography (PE: EA1:1) to give 10mg (46.5%) as a white solid.1H-NMR(400MHz,CDCl3):δ7.62(d,J=8.4Hz,1H),7.45(s,1H),7.07(d,J=8.4Hz,1H),4.68(s,2H),4.07(s,3H),3.62(s,3H),3.41(s,3H).MS(m/z):462.7[M+H]+.

Compound 65: 8- (2-ethoxy-5-methoxybenzylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 2-ethoxy-5-methoxybenzaldehyde

To 2-hydroxy-5-methoxybenzaldehyde (200mg,1.31mmol) and K2CO3To a solution of (220mg,1.6mmol) in anhydrous DMF (5mL) was added iodoethane (310mg,1.97mmol) and stirred under nitrogen at room temperature overnight. The reaction mixture was then poured into water and extracted with EA (3x 10mL), the organic layer was separated and Na was used2SO4Dried, filtered and concentrated to give a yellow solid (325.1 mg).

(II) Synthesis of 2-ethoxy-5-methoxyphenyl) methanol

At 0 ℃ under N2To a solution of 2-ethoxy-5-methoxybenzaldehyde (250mg,1.39mmol) in MeOH (5mL) under protection was added NaBH4. The mixture was then stirred at room temperature for 4h. The solvent was removed and extracted with EA (3 × 10mL) and concentrated to give a yellow solid (252.1 mg).

(III) Synthesis of 2- (bromomethyl) -1-ethoxy-4-methoxybenzene

To a solution of 2-ethoxy-5-methoxyphenyl) methanol (252mg,1.38mmol) in DME at 0 deg.C was added tribromophosphine (380mg,1.38mmol) slowly. The mixture was then stirred at room temperature overnight. The solvent was removed and extracted with EA (3 × 10mL), the organic layer was separated and Na was added2SO4Dried, filtered, concentrated and purified by column chromatography (PE: EA5:1) to give a white solid (336mg, 99%).

(IV) Synthesis of 8- ((2-ethoxy-5-methoxybenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To compound 1(30.0mg,0.133mmol) and K2CO3(36mg,0.265mmol) in anhydrous DMF (2mL) was added 2- (bromomethyl) -1-ethoxy-4-methoxybenzene (48mg,0.2mmol) and stirred under nitrogen at room temperature overnight. The reaction mixture was then poured into water and extracted with EA (3x 10mL), the organic layer was separated and Na was used2SO4drying, filtering, concentrating andPurification by column chromatography (PE: EA5:1) gave a white solid (5mg, 9.8%).

(V) Synthesis of 8- ((2-ethoxy-5-methoxybenzyl) sulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 8- ((2-ethoxy-5-methoxybenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione (5mg,0.013mmol) in MeOH (1mL) was added potassium hydrogen persulfate (80mg,0.13mmol) in H2O (1mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with EA (3 x 10mL), the organic layer was separated and Na was added2SO4Dried, filtered, concentrated and purified by silica gel column chromatography (PE: EA1:1) to give 2.3mg (42.6%) as a white solid.1H-NMR(400MHz,CDCl3):δ6.88(d,J=8.8Hz,1H),6.81(s,1H),6.75(d,J=8.8Hz,1H),4.72(s,2H),3.80-3.75(m,J=7.2Hz,2H),3.78(s,3H),3.74(s,3H),3.61(s,3H),3.41(s,3H),1.22(t,J=7.2Hz,3H).MS(m/z):423.4[M+H]+.

Compound 66: 8- (5-methoxy-2-propoxybenzylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

This compound was synthesized in a similar manner to compound 65 to give 2mg (22.7%) as a white solid.1H-NMR(400MHz,CDCl3):δ6.88(dd,J=3.2Hz,J=8.8Hz,1H),6.80(d,J=2.8Hz,1H),6.77(d,J=8.8Hz,1H),4.72(s,2H),3.78(s,3H),3.74(s,3H),3.68(t,J=6.4Hz,2H),3.61(s,3H),3.41(s,3H),1.63-1.58(m,J=6.4Hz,2H),0.94(t,J=7.2Hz,3H).MS(m/z):437.4[M+H]+.

Compound 67: 8- (5-ethoxy-2-methoxybenzylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 4-ethoxy-1-methoxy-2-methylbenzene

To 4-methoxy-3-methylphenol (200mg,1.45 mm)ol) and K2CO3(250mg,1.74mmol) in dry DMF (5mL) was added iodoethane (340mg,2.17mmol) and stirred at room temperature under nitrogen for 5 h. The reaction mixture was then poured into water and extracted with EA (3x 10mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by silica gel column chromatography (PE: EA5:1) to give a yellow solid (86mg, 35.8%).

(II) Synthesis of 2- (bromomethyl) -4-ethoxy-1-methoxybenzene

To CCl of 4-ethoxy-1-methoxy-2-methylbenzene (80mg,0.48mmol)4To the solution (2mL) was added NBS (86mg,0.48mmol) and AIBN (60mg) slowly. The mixture was then heated to 80 ℃ for 5 hours until a white solid floated on the surface. The mixture was filtered and the filtrate was concentrated to give a residue. The residue was diluted and extracted with EA (3 × 10mL) and concentrated to give 106mg of crude product (yellow solid).

(III) Synthesis of 8- ((5-ethoxy-2-methoxybenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To compound 1(30.0mg,0.133mmol) and K2CO3(36mg,0.265mmol) in anhydrous DMF (2mL) was added 2- (bromomethyl) -4-ethoxy-1-methoxybenzene (48mg,0.2mmol) and stirred under nitrogen at room temperature overnight. The reaction mixture was then poured into water and extracted with EA (3x 10mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by column chromatography (PE: EA5:1) to give a white solid (60 mg).

(IV) Synthesis of 8- ((5-ethoxy-2-methoxybenzyl) sulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 8- ((5-ethoxy-2-methoxybenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione (30mg,0.077mmol) in DCM (1mL) was added m-CPBA (15mg,0.077 mmol). The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with EA (3x 10mL), the organic layer was separated and Na was added2SO4Dried, filtered, concentrated and purified by silica gel column chromatography (PE: EA 1:1) to give 29mg (90.6%) as a white solid.1H-NMR(400MHz,CDCl3):δ6.88(dd,J=3.2Hz,J=8.8Hz,1H),6.79(d,J=2.8Hz,1H),6.74(d,J=8.8Hz,1H),4.70(s,2H),3.94(m,J=6.8Hz,2H),3.82(s,3H),3.62(s,3H),3.55(s,3H),3.41(s,3H),1.37(t,J=6.8Hz,3H).MS(m/z):423.4[M+H]+.

Compound 68: 8- (2-methoxy-5-propoxybenzylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

This compound was synthesized in a similar manner to compound 67 to give 23.1mg (72.2%) as a white solid.1H-NMR(400MHz,CDCl3):δ6.88(dd,J=3.2Hz,J=8.8Hz,1H),6.77(d,J=3.2Hz,1H),6.74(d,J=8.8Hz,1H),4.70(s,2H),3.83-3.80(m,2H),3.82(s,3H),3.62(s,3H),3.56(s,3H),3.41(s,3H),1.80-1.73(m,2H),1.00(t,J=7.2Hz,3H).MS(m/z):437.4[M+H]+.

Compound 69: 8- (3-bromo-2, 6-dimethoxybenzylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 1-bromo-3- (bromomethyl) -2, 4-dimethoxybenzene

To a solution of 1, 3-dimethoxy-2-methylbenzene (500mg,3.29mmol) in CCl4To the solution (10mL) was added NBS (585mg,3.29mmol) and AIBN (150mg) slowly. The mixture was then heated to 90 ℃ for 5 h. The mixture was filtered and the filtrate was concentrated to give a residue. The residue was diluted and extracted with EA (3 × 10mL), concentrated and purified by column chromatography (PE: EA5:1) to give a yellow solid (641mg, 56%).

(II) Synthesis of 8- ((3-bromo-2, 6-dimethoxybenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To compound 1(50.0mg,0.22mmol) and K2CO3(62mg,0.44mmol) in dry DMF (5mL) was added 1-bromo-3- (bromomethyl) -2, 4-dimethoxybenzene (80mg,0.33mmol) and stirred under nitrogen at room temperature overnight. The reaction mixture was then poured into water and washed with dichloromethaneAlkane (3x 10mL) extraction, separation of organic layer, Na2SO4Dried, filtered, concentrated and purified by column chromatography (PE: EA1:1) to give a white solid (67.5mg, 81%).

(III) Synthesis of 8- ((3-bromo-2, 6-dimethoxybenzyl) sulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 8- ((3-bromo-2, 6-dimethoxybenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione (30mg,0.08mmol) in MeOH (1mL) was added potassium hydrogen persulfate (655mg,0.8mmol) in H2O (1mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with EA (3x 10mL), the organic layer was separated and Na was added2SO4Dried, filtered, concentrated and purified by silica gel column chromatography (PE: EA1:1) to give 15mg (46.1%) as a white solid.1H-NMR(400MHz,CDCl3):δ7.53(d,J=9.2Hz,1H),6.58(d,J=8.8Hz,1H),4.86(s,2H),3.97(s,3H),3.86(s,3H),3.66(s,3H),3.60(s,3H),3.43(s,3H).MS(m/z):488.3[M+H]+.

Compound 70: 8- (4-chloro-2, 5-dimethoxybenzylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 4-chloro-2, 5-dimethoxybenzaldehyde

To 2-chloro-1, 4-dimethoxybenzene (2.5g,14.48mmol) and hexamethylenetetramine (2.05g,14.63mmol) was carefully added TFA (25 mL). The mixture was then heated to 95 ℃ overnight. The mixture was then poured into ice and washed with NaHCO3The pH was adjusted to 8. The solvent was then removed and extracted with dichloromethane (3x 20mL), the organic layer separated and washed with Na2SO4Dry, filter, concentrate and purify by silica gel chromatography (EA: PE ═ 1:10) to give a white solid (2.1g, 72.7%).1H-NMR(400MHz,CDCl3):δ10.39(s,1H),7.38(s,1H),7.06(s,1H),3.89(s,6H).MS(m/z):201.02[M+H]+.

(II) Synthesis of (4-chloro-2, 5-dimethoxyphenyl) methanol

To 4-chloro-2, 5-dimethoxybenzeneNaBH was carefully added to a solution of formaldehyde (1.5g,7.5mmol) in MeOH (20mL)4(0.85g,22.48 mmol). The mixture was then stirred at room temperature for 2 h. The solvent was then removed and extracted with dichloromethane (3x 10mL), the organic layer separated and washed with Na2SO4Dry, filter, concentrate and purify by silica gel chromatography (EA: PE ═ 1:4) to give a white solid (1.45g, 96%).1H-NMR(400MHz,CDCl3):δ6.95(s,1H),6.90(s,1H),4.65(s,1H),3.86(s,3H),3.81(s,3H),2.18(br,1H).MS(m/z):203.04[M+H]+.

(III) Synthesis of 1- (bromomethyl) -4-chloro-2, 5-dimethoxybenzene

To a solution of (4-chloro-2, 5-dimethoxyphenyl) methanol (500mg,2.48mmol) in DME (10mL) at 0 deg.C was added PBr carefully3(0.99g,3.69 mmol). The mixture was then stirred at room temperature overnight. The solvent was then removed and extracted with dichloromethane (3x 10mL), the organic layer separated and washed with Na2SO4Dried, filtered and concentrated to give a yellow solid (438.2 mg). MS (M/z) 264.96[ M + H]+.

(IV) Synthesis of 8- (4-chloro-2, 5-dimethoxybenzylthio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To compound 1(50.0mg,0.22mmol) and K2CO3To a solution of (36.6mg,0.26mmol) in anhydrous DMF (10mL) was added 1- (bromomethyl) -4-chloro-2, 5-dimethoxybenzene (71.2mg,0.26mmol) and stirred under nitrogen at room temperature overnight. The reaction mixture was then poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used2SO4Dry, filter, concentrate and purify by prep TLC to give a white solid (23.4mg, 25.8%).1H-NMR(400MHz,CDCl3):δ7.02(s,1H),6.91(s,1H),4.45(s,1H),3.82(s,3H),3.80(s,3H),3.76(s,3H),3.61(s,3H),3.39(s,3H).MS(m/z):411.08[M+H]+.

(V) Synthesis of 8- (4-chloro-2, 5-dimethoxybenzylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 8- (4-chloro-2, 5-dimethoxybenzylthio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione (10.0mg,0.02mmol) in DCM (2mL) was added m-CPBA (6.4mg,0.03 mmol). The mixture was then stirred at room temperature for 2 h. Then go toRemove solvent and extract with dichloromethane (3x 5mL), separate organic layer, use Na2SO4Dry, filter, concentrate and purify by prep TLC to give a white solid (8.2mg, 76.7%).1H-NMR(400MHz,CDCl3):δ6.93(s,1H),6.89(s,1H),4.73(s,1H),4.00(s,3H),3.84(s,3H),3.60(s,6H),3.42(s,3H).MS(m/z):443.07[M+H]+.

Compound 71: 8- (4-fluoro-2, 5-dimethoxybenzylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 4-fluoro-2, 5-dimethoxybenzaldehyde

A solution of 2-fluoro-1, 4-dimethoxybenzene (2.07g,13.26mmol) in DCM (15mL) was cooled to 5-6 deg.C in an ice bath. 6.75g (25.99mmol) of SnCl are added with vigorous stirring4Subsequently, dichloro (methoxy) methane (1.3g,11.41mmol) was added dropwise at a rate to maintain the internal temperature below 10 ℃. The mixture was then stirred at room temperature for 30min and poured into a mixture of 50g ice and 15mL HCl (concentrated). The reaction mixture was then extracted with dichloromethane (3x 20mL), the organic layer was separated, and Na was added2SO4Dry, filter, concentrate and purify by silica gel chromatography (EA: PE ═ 1:10) to give a green solid (1.9g, 77.86%).1H-NMR(400MHz,CDCl3):δ10.34(s,1H),7.43(d,J=9.6Hz,1H),6.77(d,J=12.4Hz,1H),3.87(s,3H),3.86(s,3H).MS(m/z):185.05[M+H]+.

(II) Synthesis of (4-fluoro-2, 5-dimethoxyphenyl) methanol

To a solution of 4-fluoro-2, 5-dimethoxybenzaldehyde (500.0mg,2.71mmol) in dry THF (10mL) was carefully added NaBH4(410.8mg,10.86 mmol). The mixture was then stirred at room temperature overnight. The solvent was then removed and extracted with dichloromethane (3x 15mL), the organic layer separated and washed with Na2SO4Dry, filter, concentrate and purify by prep TLC to give a white solid (238.1mg, 47.2%).1H-NMR(400MHz,CDCl3):δ6.96(d,J=9.6Hz,1H),6.68(d,J=12.8Hz,1H),4.62(s,2H),3.84(s,3H),3.79(s,3H).MS(m/z):187.07[M+H]+.

(III) Synthesis of 1- (bromomethyl) -4-fluoro-2, 5-dimethoxybenzene

To a solution of (4-fluoro-2, 5-dimethoxyphenyl) methanol (200mg,1.07mmol) in DME (5mL) at 0 deg.C was carefully added PBr3(431.2mg,1.67 mmol). The mixture was then stirred at room temperature overnight. The solvent was then removed and extracted with dichloromethane (3x 10mL), the organic layer separated and washed with Na2SO4Dried, filtered and concentrated to give a white solid (203.4 mg). MS (M/z) 248.98[ M + H]+.

(IV) Synthesis of 8- (4-fluoro-2, 5-dimethoxybenzylthio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To compound 1(50.0mg,0.22mmol) and K2CO3To a solution of (36.6mg,0.26mmol) in anhydrous DMF (10mL) was added 1- (bromomethyl) -4-fluoro-2, 5-dimethoxybenzene (65.8mg,0.26mmol) and stirred under nitrogen at room temperature overnight. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by preparative TLC to give a white solid (30.1 mg). MS (M/z) 395.11[ M + H]+.

(V) Synthesis of 8- (4-fluoro-2, 5-dimethoxybenzylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 8- (4-fluoro-2, 5-dimethoxybenzylthio) -1,3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione (20.0mg,0.05mmol) in DCM (5mL) was added m-CPBA (13.1mg,0.075 mmol). The mixture was then stirred at room temperature for 2 h. The solvent was then removed and extracted with dichloromethane (3x 5mL), the organic layer separated and washed with Na2SO4Dry, filter, concentrate and purify by prep TLC to give a white solid (13.4mg, 62.1%).1H-NMR(400MHz,CDCl3):δ6.96(d,J=9.2Hz,1H),6.66(d,J=12.4Hz,1H),4.71(s,1H),3.99(s,3H),3.82(s,3H),3.60(s,3H),3.58(s,3H),3.42(s,3H).MS(m/z):427.10[M+H]+.

Compound 72: 1, 3-dimethyl-8- (methylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

(I) synthesis of 1, 3-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione

To a solution of 8-chloro-1, 3-dimethyl-1H-purine-2, 6(3H,7H) -dione (100mg,0.46mmol) in anhydrous NMP (2mL) was added sodium methyl mercaptide (98.2mg,1.40mmol) and reacted in a microwave at 180 ℃ for 3H on a Biotage Smith synthesizer. The mixture was then cooled to room temperature and acidified to PH 3. The reaction mixture was then poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used2SO4Dried, filtered and concentrated to give a yellow oil (64.7 mg). MS (M/z) 227.05[ M + H]+.

(II) Synthesis of 1, 3-dimethyl-8- (methylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

To a solution of 1, 3-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione (30.0mg,0.13mmol) in MeOH (1mL) was added oxone (409.9mg,0.67mmol) in H2O (2mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 10mL), the organic layer separated and washed with Na2SO4Dry, filter, concentrate and purify by prep TLC to give a white solid (4.7mg, 10.9%).1H-NMR(400MHz,CDCl3):δ5.56(br,1H),3.68(s,3H),3.43(s,3H),3.19(s,3H).MS(m/z):259.04[M+H]+.

Compound 73: 7-Ethyl-1, 3-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 8-chloro-7-ethyl-1, 3-dimethyl-1H-purine-2, 6(3H,7H) -dione

To 8-chloro-1, 3-dimethyl-1H-purine-2, 6(3H,7H) -dione (200.0mg,0.93mmol) and K2CO3(154.8mg,1.12mmol) in anhydrous DMF (2mL) was added iodoethane (174.8mg,1.12mmol) and stirred at room temperature under nitrogen for 4h. The reaction mixture was then poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used2SO4Drying and filteringConcentrated and purified by silica gel chromatography (PE: EA ═ 2:1) to give a white solid (203.0 mg). MS (M/z) 243.06[ M + H]+.

(II) Synthesis of 7-Ethyl-1, 3-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione

To a solution of 8-chloro-7-ethyl-1, 3-dimethyl-1H-purine-2, 6(3H,7H) -dione (213mg,0.87mmol) in anhydrous DMF (2mL) was added sodium methyl mercaptide (1.5mL) and heated to 105 ℃ for 8H. The mixture was then cooled to room temperature and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was added2SO4Dry, filter, concentrate and purify by silica gel chromatography (PE: EA ═ 2:1) to give a white solid (31.2mg, 19.2%).1H-NMR(400MHz,CDCl3):δ4.29(q,J=7.2Hz,2H),3.57(s,3H),3.39(s,3H),2.72(s,3H),1.39(t,J=7.2Hz,3H).MS(m/z):255.08[M+H]+.

Compound 74: 7-Ethyl-1, 3-dimethyl-8- (methylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

To a solution of compound 74(24.7mg,0.09mmol) in MeOH (2mL) was added oxone (237.3mg,0.38mmol) in H2O (2mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 10mL), the organic layer separated and washed with Na2SO4Dry, filter, concentrate and purify by prep TLC to give a white solid (20.3mg, 73.1%).1H-NMR(400MHz,CDCl3):δ4.79(q,J=7.2Hz,2H),3.57(s,3H),3.45(s,3H),3.43(s,3H),1.54(t,J=7.2Hz,3H).MS(m/z):287.07[M+H]+.

Compound 75: 7-Ethyl-8- (ethylsulfanyl) -1, 3-dimethyl-1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 8-chloro-7-ethyl-1, 3-dimethyl-1H-purine-2, 6(3H,7H) -dione

To 8-chloro-1, 3-dimethyl-1H-purine-2, 6(3H,7H)) -diketone (200.0mg,0.93mmol) and K2CO3(154.8mg,1.12mmol) in anhydrous DMF (2mL) was added iodoethane (174.8mg,1.12mmol) and stirred at room temperature under nitrogen for 4h. The reaction mixture was then poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used2SO4Dry, filter, concentrate and purify by silica gel chromatography (PE: EA ═ 2:1) to give a white solid (203.0 mg). MS (M/z) 243.06[ M + H]+.

(II) Synthesis of 7-Ethyl-8-mercapto-1, 3-dimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 8-chloro-7-ethyl-1, 3-dimethyl-1H-purine-2, 6(3H,7H) -dione (140.0mg,0.57mmol) in anhydrous DMF (2mL) was added NaHS (97.2mg,1.73mmol) and heated to 105 ℃ for 7H. The mixture was then cooled to room temperature and acidified to PH 3. The reaction mixture was then poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used2SO4Dry, filter, concentrate and purify by silica gel chromatography (PE: EA ═ 1:2) to give a white solid (82.3mg, 59.3%).1H-NMR(400MHz,CDCl3):δ13.18(s,1H),4.79(q,J=7.2Hz,2H),3.58(s,3H),3.39(s,3H),1.40(t,J=7.2Hz,3H).MS(m/z):241.07[M+H]+.

(III) Synthesis of 7-Ethyl-8- (ethylsulfanyl) -1, 3-dimethyl-1H-purine-2, 6(3H,7H) -dione

To 7-ethyl-8-mercapto-1, 3-dimethyl-1H-purine-2, 6(3H,7H) -dione (30.0mg,0.12mmol) and K2CO3(20.7mg,0.15mmol) in anhydrous DMF (2mL) was added iodoethane (23.4mg,0.15mmol) and stirred at room temperature under nitrogen for 3 h. The reaction mixture was poured into water and extracted with dichloromethane (3 x 10mL), the organic layer was separated and Na was added2SO4Dry, filter, concentrate and purify by prep TLC to give a white solid (32.3mg, 99.38%).1H-NMR(400MHz,CDCl3):δ4.30(q,J=6.8Hz,2H),3.56(s,3H),3.39(s,3H),3.29(q,J=6.8Hz,2H),1.42(t,J=6.8Hz,3H),1.38(t,J=6.8Hz,3H)..MS(m/z):269.10[M+H]+.

Compound 76: 7-Ethyl-8- (ethylsulfinyl) -1, 3-dimethyl-1H-purine-2, 6(3H,7H) -dione

Compound 77: 7-Ethyl-8- (ethylsulfonyl) -1, 3-dimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of compound 75(25mg) in MeOH (2mL) was added oxone (86.3mg) in H2O (2mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 10mL), the organic layer separated and washed with Na2SO4Drying, filtration, concentration and purification by preparative TLC (PE: EA1:1) gave 763.8 mg (20.4%) of compound as a white solid.1H-NMR(400MHz,CDCl3):δ4.71(q,J=6.8Hz,2H),3.57(s,3H),3.44(s,3H),3.40(q,J=7.6Hz,2H),1.53(t,J=6.8Hz,3H),1.38(t,J=7.6Hz,3H).MS(m/z):285.09[M+H]+.

Compound 7718.2 mg (69.7%) was obtained as a white solid.1H-NMR(400MHz,CDCl3):δ4.74(q,J=7.2Hz,2H),3.53(q,J=7.2Hz,2H),3.52(s,3H),3.36(s,3H),1.47(t,J=7.2Hz,3H),1.39(t,J=7.2Hz,3H).MS(m/z):301.09[M+H]+.

Compound 78: 7-Ethyl-1, 3-dimethyl-8- (propylsulfanyl) -1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 8-chloro-7-ethyl-1, 3-dimethyl-1H-purine-2, 6(3H,7H) -dione

To 8-chloro-1, 3-dimethyl-1H-purine-2, 6(3H,7H) -dione (200.0mg,0.93mmol) and K2CO3(154.8mg,1.12mmol) in anhydrous DMF (2mL) was added iodoethane (174.8mg,1.12mmol) and stirred at room temperature under nitrogen for 4h. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by silica gel chromatography (PE: EA ═ 2:1) to give a white solid (203.0 mg). MS (M/z) 243.06[ M + H]+.

(II) Synthesis of 7-Ethyl-8-mercapto-1, 3-dimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 8-chloro-7-ethyl-1, 3-dimethyl-1H-purine-2, 6(3H,7H) -dione (140.0mg,0.57mmol) in anhydrous DMF (2mL) was added NaHS (97.2mg,1.73mmol) and heated to 105 ℃ for 7H. The mixture was then cooled to room temperature and acidified to PH 3. The reaction mixture was then poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used2SO4Dry, filter, concentrate and purify by silica gel chromatography (PE: EA ═ 1:2) to give a white solid (82.3mg, 59.3%). 1H-NMR (400MHz, CDCl3): delta 13.18(s,1H),4.79(q, J ═ 7.2Hz,2H),3.58(s,3H),3.39(s,3H),1.40(t, J ═ 7.2Hz,3H). MS (M/z):241.07[ M + H3: (M/z): d 13.18(s,1H),4.79(q, J ═ 7.2Hz,2H),3.58(s,3H)]+.

(III) Synthesis of 7-Ethyl-1, 3-dimethyl-8- (propylsulfanyl) -1H-purine-2, 6(3H,7H) -dione

To a solution of 7-ethyl-8-mercapto-1, 3-dimethyl-1H-purine-2, 6(3H,7H) -dione (30.0mg,0.12mmol) and K2CO3(20.7mg,0.15mmol) in anhydrous DMF (2mL) was added 1-iodopropane (25.5mg,0.15mmol) and stirred at room temperature under nitrogen for 3H. The reaction mixture was poured into water and extracted with EA (3 x 10mL), the organic layer was separated and Na was added2SO4Dry, filter, concentrate and purify by preparative TLC to give 31.5mg (89.3%) as a white solid.1H-NMR(400MHz,CDCl3):δ4.30(q,J=6.8Hz,2H),3.56(s,3H),3.39(s,3H),3.25(t,J=6.8Hz,2H),1.79(m,2H),1.38(t,J=6.8Hz,3H),1.04(t,J=7.6Hz,3H).MS(m/z):283.12[M+H]+.

Compound 79: 7-Ethyl-1, 3-dimethyl-8- (propylsulfinyl) -1H-purine-2, 6(3H,7H) -dione

Compound 80: 7-Ethyl-1, 3-dimethyl-8- (propylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

To compound 78(30mg) in MeOH (2mL) was added oxone (97.98mg) in H2O (2mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 10mL), the organic layer separated and washed with Na2SO4Drying, filtration, concentration and purification by preparative TLC gave 792.9mg (22.3%) of compound as a white solid.1H-NMR(400MHz,CDCl3):δ4.74-4.67(m,2H),3.57(s,3H),3.46(s,3H),3.51-3.25(m,2H),1.85(t,J=7.6Hz,2H),1.53(t,J=7.2Hz,3H),1.14(t,J=7.2Hz,3H).MS(m/z):299.11[M+H]+.

Compound 8019.2 mg (70.3%) was obtained as a white solid.1H-NMR(400MHz,CDCl3):δ4.81(q,J=7.2Hz,2H),3.54(s,3H),3.52(q,J=7.6Hz,2H),3.42(s,3H),1.94(t,J=7.6Hz,2H),1.53(t,J=7.2Hz,3H),1.11(t,J=7.6Hz,3H).MS(m/z):315.10[M+H]+.

Compound 81: 8- (Cyclopropylmethylthio) -7-ethyl-1, 3-dimethyl-1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 8-chloro-7-ethyl-1, 3-dimethyl-1H-purine-2, 6(3H,7H) -dione

To 8-chloro-1, 3-dimethyl-1H-purine-2, 6(3H,7H) -dione (200.0mg,0.93mmol) and K2CO3(154.8mg,1.12mmol) in anhydrous DMF (2mL) was added iodoethane (174.8mg,1.12mmol) and stirred at room temperature under nitrogen for 4h. The reaction mixture was then poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used2SO4Dry, filter, concentrate and purify by silica gel chromatography (PE: EA ═ 2:1) to give a white solid (203.0 mg). MS (M/z) 243.06[ M + H]+.

(II) Synthesis of 7-Ethyl-8-mercapto-1, 3-dimethyl-1H-purine-2, 6(3H,7H) -dione

to a solution of 8-chloro-7-ethyl-1, 3-dimethyl-1H-purine-2, 6(3H,7H) -dione (140.0mg,0.57mmol) in anhydrous DMF (2mL) was added NaHS (97.2mg,1.73mmol) and heatedHold to 105 ℃ for 7 h. The mixture was then cooled to room temperature and acidified to PH 3. The reaction mixture was then poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used2SO4Dry, filter, concentrate and purify by silica gel chromatography (PE: EA ═ 1:2) to give a white solid (82.3mg, 59.3%).1H-NMR(400MHz,CDCl3):δ13.18(s,1H),4.79(q,J=7.2Hz,2H),3.58(s,3H),3.39(s,3H),1.40(t,J=7.2Hz,3H).MS(m/z):241.07[M+H]+.

(III) Synthesis of 7-Ethyl-1, 3-dimethyl-8- (propylsulfanyl) -1H-purine-2, 6(3H,7H) -dione

to 7-ethyl-8-mercapto-1, 3-dimethyl-1H-purine-2, 6(3H,7H) -dione (30.0mg,0.12mmol) and K2CO3(20.7mg,0.15mmol) in anhydrous DMF (2mL) was added (bromomethyl) cyclopropane (20.25mg,0.15mmol) and stirred at room temperature under nitrogen for 3 h. The reaction mixture was poured into water and extracted with EA (3x 10mL), the organic layer was separated and Na was added2SO4Dried, filtered, concentrated and purified by preparative TLC (PE: EA1:1) to give 25.6mg (69.75%) as a white solid.1H-NMR(400MHz,CDCl3):δ4.33(q,J=7.2Hz,2H),3.57(s,3H),3.39(s,3H),3.21(d,J=7.6Hz,2H),1.40(q,J=7.2Hz,3H),1.24-1.14(m,1H),0.65-0.61(m,2H),0.35-0.33(m,2H).MS(m/z):295.12[M+H]+.

Compound 82: 8- (Cyclopropylmethylsulfonyl) -7-ethyl-1, 3-dimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of compound 81(23mg) in MeOH (1mL) was added oxone (240mg) in H2O (1mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 10mL), the organic layer separated and washed with Na2SO4Drying, filtration, concentration and purification by preparative TLC yielded 11.5mg (66.9%) as a white solid.1H-NMR(400MHz,CDCl3):δ4.84(q,J=6.8Hz,2H),3.56(s,3H),3.45(s,3H),3.42(d,J=5.2Hz,2H),1.54(t,J=6.8Hz,2H),1.20-1.15(m,1H),0.69-0.66(m,2H),0.36-0.32(m,2H).MS(m/z):327.10[M+H]+.

Compound 83: 1, 3-dimethyl-8- (methylsulfonyl) -7-propyl-1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 8-chloro-1, 3-dimethyl-7-propyl-1H-purine-2, 6(3H,7H) -dione

To 8-chloro-1, 3-dimethyl-1H-purine-2, 6(3H,7H) -dione (500.0mg,2.33mmol) and K2CO3(386.9mg,2.80mmol) in dry DMF (5mL) was added 1-iodopropane (476.5mg,2.80mmol) and stirred at room temperature under nitrogen for 4h. The reaction mixture was then poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by silica gel chromatography (PE: EA ═ 2:1) to give a white solid (467.3 mg). MS (M/z) 257.07[ M + H]+.

(II) Synthesis of 8-mercapto-1, 3-dimethyl-7-propyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 8-chloro-1, 3-dimethyl-7-propyl-1H-purine-2, 6(3H,7H) -dione (350.0mg,1.36mmol) in anhydrous DMF (5mL) was added NaHS (229.7mg,4.10mmol) and heated to 120 ℃ overnight. The mixture was then cooled to room temperature and acidified to PH 3. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10mL), the organic layer was separated and Na was used2SO4Dry, filter, concentrate and purify by silica gel chromatography (DCM: MeOH ═ 20:1) to give a white solid (235.2 mg). MS (M/z) 255.08[ M + H]+.

(III) Synthesis of 1, 3-dimethyl-8- (methylthio) -7-propyl-1H-purine-2, 6(3H,7H) -dione

To 8-mercapto-1, 3-dimethyl-7-propyl-1H-purine-2, 6(3H,7H) -dione (50.0mg,0.19mmol) and K2CO3To a solution of (32.5mg,0.23mmol) in anhydrous DMF (5mL) was added iodomethane (35.3mg,0.23mmol) and stirred at room temperature under nitrogen for 2 h. The reaction mixture was then poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used2SO4Drying, filtering, and concentratingAnd purified by preparative TLC to give a white solid (47.8mg, 90.62%).1H-NMR(400MHz,CDCl3):δ4.17(t,J=7.2Hz,2H),3.57(s,3H),3.39(s,3H),2.71(s,3H),1.87-1.78(m,2H),0.94(t,J=7.2Hz,3H).MS(m/z):269.10[M+H]+.

(IV) Synthesis of 1, 3-dimethyl-8- (methylsulfonyl) -7-propyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 1, 3-dimethyl-8- (methylthio) -7-propyl-1H-purine-2, 6(3H,7H) -dione (40.0mg,0.15mmol) in MeOH (5mL) was added oxone (366.9mg,0.59mmol) in H2O (5mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 10mL), the organic layer separated and washed with Na2SO4Dry, filter, concentrate and purify by prep TLC to give a white solid (41.9mg, 93.58%).1H-NMR(400MHz,CDCl3):δ4.67(t,J=6.0Hz,2H),3.57(s,3H),3.44(s,3H),3.42(s,3H),2.03-1.90(m,2H),1.00(t,J=7.6Hz,3H).MS(m/z):301.09[M+H]+.

Compound 84: 2- (1, 3-dimethyl-8- (methylsulfonyl) -2, 6-dioxo-2, 3-dihydro-1H-purin-7 (6H) -yl) acetonitrile

(I) Synthesis of 2- (8-chloro-1, 3-dimethyl-2, 6-dioxo-2, 3-dihydro-1H-purin-7 (6H) -yl) acetonitrile

To 8-chloro-1, 3-dimethyl-1H-purine-2, 6(3H,7H) -dione (3g) and K2CO32-Bromoacetonitrile (2g) was added (2.32g) to a solution of anhydrous DMF (30mL) and stirred at room temperature under nitrogen for 3 h. The reaction mixture was then poured into water and extracted with EA (3 × 50mL), the organic layer was separated and Na was used2SO4Dry, filter, concentrate and purify by silica gel chromatography (PE: EA ═ 1:1) to give a white solid (2.3g, 64.78%).

(II) Synthesis of 2- (8-mercapto-1, 3-dimethyl-2, 6-dioxo-2, 3-dihydro-1H-purin-7 (6H) -yl) acetonitrile

To 2- (8-chloro-1, 3-dimethyl-2, 6-dioxo-2, 3-dihydro-1H-purin-7 (6H) -yl) acetonitrile (100mg) in the absence of waterNaHS (44.3mg) was added to the DMF (3mL) solution and heated to 135 ℃ for 3 h. The mixture was then cooled to room temperature and acidified to PH 3. The reaction mixture was then poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used2SO4Dry, filter, concentrate and purify by silica gel chromatography (PE: EA ═ 1:1) to give a white solid (45.6mg, 45.96%).

(III) Synthesis of 2- (1, 3-dimethyl-8- (methylthio) -2, 6-dioxo-2, 3-dihydro-1H-purin-7 (6H) -yl) acetonitrile

to 2- (8-mercapto-1, 3-dimethyl-2, 6-dioxo-2, 3-dihydro-1H-purin-7 (6H) -yl) acetonitrile (20mg) and K2CO3To a solution of (13.2mg) in anhydrous DMF (5mL) was added iodomethane (13.5mg) and the mixture was stirred at room temperature under nitrogen for 2 h. The reaction mixture was then poured into water and extracted with EA (3x 10mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by preparative TLC (PE: EA1:1) to give a white solid.

(IV) Synthesis of 2- (1, 3-dimethyl-8- (methylsulfonyl) -2, 6-dioxo-2, 3-dihydro-1H-purin-7 (6H) -yl) acetonitrile

To a solution of 2- (1, 3-dimethyl-8- (methylthio) -2, 6-dioxo-2, 3-dihydro-1H-purin-7 (6H) -yl) acetonitrile (15mg) in MeOH (2mL) was added oxone (115.99mg) in H2O (2mL) solution. The mixture was then stirred at room temperature for 6 h. The solvent was then removed and extracted with dichloromethane (3x 5mL), the organic layer separated and washed with Na2SO4Dry, filter, concentrate and purify by preparative TLC to give 8.2mg (46.86%) as a white solid.1H-NMR(400MHz,CDCl3):δ5.81(s,2H),3.60(s,3H),3.45(s,3H),3.44(s,3H).MS(m/z):298.05[M+H]+.

Compound 85: 2- (1, 3-dimethyl-8- (methylthio) -2, 6-dioxo-2, 3-dihydro-1H-purin-7 (6H) -yl) acetamide

2- (1, 3-dimethyl-8- (methylthio) -2, 6-dioxo-2, 3-dihydro-1H-purin-7 (6H) -yl) acetonitrile was synthesized by the method described for compound 84.

To a solution of 2- (1, 3-dimethyl-8- (methylthio) -2, 6-dioxo-2, 3-dihydro-1H-purin-7 (6H) -yl) acetonitrile (100mg,0.39mmol) in EtOH (2mL) was added sodium methyl mercaptide (33.2mg,0.47mmol) and refluxed for 5H. The mixture was then cooled to room temperature and extracted with dichloromethane (3x 5mL), the organic layer was separated and Na was added2SO4Dry, filter, concentrate and purify by prep TLC to give a white solid (21.2mg, 19.1%).1H-NMR(400MHz,DMSO-d6):δ7.67(br,1H),7.33(br,1H),4.83(s,2H),3.44(s,3H),3.20(s,3H),2.65(s,3H).MS(m/z):284.07[M+H]+.

Compound 86: 2- (1, 3-dimethyl-8- (methylsulfonyl) -2, 6-dioxo-2, 3-dihydro-1H-purin-7 (6H) -yl) acetamide

To a solution of compound 85(20.0mg,0.07mmol) in MeOH (2mL) was added oxone (217.3mg,0.35mmol) in H2O (2mL) solution. The mixture was then stirred at room temperature for 8 h. The solvent was then removed and extracted with dichloromethane (3x 5mL), the organic layer separated and washed with Na2SO4Dry, filter, concentrate and purify by prep TLC to give a white solid (15.4mg, 73.65%).1H-NMR(400MHz,DMSO-d6):δ7.81(br,1H),7.42(br,1H),5.37(s,2H),3.45(s,3H),3.43(s,3H),3.23(s,3H).MS(m/z):316.06[M+H]+.

compound 87: 3, 7-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 8-chloro-3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione (4.0g,22.19mmol) in anhydrous THF (20mL) was added NCS (4.5g,33.71mmol) and stirred under nitrogen at room temperature overnight. The solvent was then removed and extracted with dichloromethane (3x 10mL), the organic layer separated and washed with Na2SO4Drying, filtration, concentration and recrystallization from EtOH: MeOH ═ 2:1 gave a white solid (2.5g, 52.3%).

(II) Synthesis of 3, 7-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione

To a solution of 8-chloro-3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione (150mg,0.70mmol) in anhydrous DMF (2mL) was added sodium methionate (195.9mg,2.79mmol) and reacted in a microwave at 130 ℃ for 1H on a Biotage Smith synthesizer. The mixture was then cooled to room temperature and acidified to PH 3. Then after stirring for 1h a solid precipitated out. The crude product was recrystallized from DCM: PE ═ 1:10 and filtered to give a white solid (89.5mg, 62.3%).1H-NMR(400MHz,DMSO-d6):δ11.08(br,1H),7.33(br,1H),3.72(s,3H),3.36(s,3H),2.67(s,3H).MS(m/z):227.05[M+H]+.

Compound 88: 3, 7-dimethyl-8- (methylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

To a solution of compound 88(20mg,0.08mmol) in MeOH (1mL) was added oxone (326.3mg,0.53mmol) in H2O (1mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 5mL), the organic layer separated and washed with Na2SO4Dry, filter, concentrate and purify by prep TLC to give a white solid (4.5mg, 39.4%).1H-NMR(400MHz,CDCl3):δ4.29(s,3H),3.52(s,3H),3.44(s,3H).MS(m/z):259.04[M+H]+.

Compound 89: ethyl-3, 7-dimethyl-8- (methylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 8-chloro-3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione (4.0g,22.19mmol) in anhydrous THF (20mL) was added NCS (4.5g,33.71mmol) and the mixture was taken under nitrogenStir overnight at room temperature under air. The solvent was then removed and extracted with dichloromethane (3x 10mL), the organic layer separated and washed with Na2SO4Drying, filtration, concentration and recrystallization in EtOH: MeOH ═ 2:1 gave a white solid (2.5 g). MS (M/z) 215.03[ M + H]+.

(II) Synthesis of 8-chloro-1-ethyl-3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione

to 8-chloro-3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione (500mg,2.33mmol) and K2CO3(460.3mg,2.79mmol) in dry DMF (5mL) was added iodoethane (519.6mg,2.79mmol) and stirred at room temperature under nitrogen for 4h. The reaction mixture was then poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used2SO4Dry, filter, concentrate and purify by silica gel chromatography (PE: EA ═ 2:1) to give a white solid (141.2mg, 16.3%).1H-NMR(400MHz,CDCl3):δ4.08(q,J=7.2Hz,2H),3.95(s,3H),3.54(s,3H),1.24(t,J=7.2Hz,3H).MS(m/z):243.06[M+H]+.

(III) Synthesis of 1-Ethyl-8-mercapto-3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 8-chloro-1-ethyl-3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione (40.0mg,0.16mmol) in anhydrous DMF (2mL) was added NaHS (26.9mg,0.48mmol) and heated to 100 ℃ for 3H. The mixture was then cooled to room temperature and acidified to PH 3. The reaction mixture was then poured into water and extracted with dichloromethane (3x 5mL), the organic layer was separated and Na was used2SO4Dried, filtered and concentrated to give a white solid (35.9 mg). MS (M/z) 241.07[ M + H]+.

(IV) Synthesis of 1-Ethyl-3, 7-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione

To 1-ethyl-8-mercapto-3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione (25.0mg,0.10mmol) and K2CO3(17.3mg,0.12mmol) in anhydrous DMF (1mL) was added iodomethane (17.8mg,0.12mmol) and stirred at room temperature under nitrogen for 2 h. The reaction mixture was then poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used2SO4Drying, filtering, concentrating and purifying by preparative TLC to obtainTo a white solid (12.0mg, 43.5%).1H-NMR(400MHz,CDCl3):δ4.07(t,J=6.8Hz,2H),3.84(s,3H),3.56(s,3H),2.71(s,3H),1.24(t,J=6.8Hz,3H).MS(m/z):255.08[M+1].

(V) Synthesis of 1-Ethyl-3, 7-dimethyl-8- (methylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

To a solution of 1-ethyl-3, 7-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione (10mg,0.03mmol) in MeOH (1mL) was added oxone (120.8mg,0.19mmol) in H2O (1mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 5mL), the organic layer separated and washed with Na2SO4Dry, filter, concentrate and purify by prep TLC to give a white solid (8.2mg, 77.4%).1H-NMR(400MHz,CDCl3):δ4.32(s,3H),4.08(q,J=6.0Hz,2H),3.56(s,3H),3.43(s,3H),1.25(t,J=6.0Hz,3H).MS(m/z):287.07[M+H]+.

Compound 90: 3, 7-dimethyl-8- (methylsulfonyl) -1-propyl-1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 8-chloro-3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione (4.0g,22.19mmol) in anhydrous THF (20mL) was added NCS (4.5g,33.71mmol) and stirred under nitrogen at room temperature overnight. The solvent was then removed and extracted with dichloromethane (3x 10mL), the organic layer separated and washed with Na2SO4Drying, filtration, concentration and recrystallization from EtOH: MeOH ═ 2:1 gave a white solid (2.5g, 52.3%).

(II) Synthesis of 3, 7-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione

A mixture of 8-chloro-3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione (100mg) and sodium thiomethoxide (130.5mg) in anhydrous DMF (4mL) was reacted in a microwave at 100 ℃ for 1H on a Biotage Smith synthesizer. The mixture was then cooled to room temperature and acidified to pH 5. The reaction mixture was poured into water and dichloromethane was usedAlkane (3x 10mL) extraction, separation of organic layer, Na2SO4dried, filtered and concentrated to give a white solid (67.8mg, 64.3%).

(III) Synthesis of 3, 7-dimethyl-8- (methylthio) -1-propyl-1H-purine-2, 6(3H,7H) -dione

To 3, 7-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione (30.0mg,0.13mmol) and K2CO3(21.9mg,0.15mmol) in anhydrous DMF (2mL) 1-iodopropane (27.1mg,0.15mmol) was added and stirred at room temperature under nitrogen for 2 h. The reaction mixture was then poured into water and extracted with dichloromethane (3x 5mL), the organic layer was separated and Na was used2SO4Dry, filter, concentrate and purify by prep TLC to give a white solid (18.7mg, 43.7%).1H-NMR(400MHz,CDCl3):δ3.94(t,J=7.6Hz,2H),3.83(s,3H),3.55(s,3H),2.70(s,3H),1.69-1.63(m,2H),0.95(t,J=7.6Hz,3H).MS(m/z):269.10[M+H]+.

(IV) Synthesis of 3, 7-dimethyl-8- (methylsulfonyl) -1-propyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 3, 7-dimethyl-8- (methylthio) -1-propyl-1H-purine-2, 6(3H,7H) -dione (15.0mg,0.05mmol) in MeOH (1mL) was added oxone (171.8mg,0.27mmol) in H2O (1mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 5mL), the organic layer separated and washed with Na2SO4Dry, filter, concentrate and purify by prep TLC to give a white solid (8.7mg, 61.4%).1H-NMR(400MHz,CDCl3):δ4.31(s,3H),3.96(t,J=5.6Hz,2H),3.55(s,3H),3.42(s,3H),1.69-1.63(m,2H),0.95(t,J=7.2Hz,3H).MS(m/z):301.09[M+H]+.

Compound 91: 1- (cyclopropylmethyl) -3, 7-dimethyl-8- (methylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 8-chloro-3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione

To 3, 7-dimethyl-1H-purine-2, 6(3H,7H) -A solution of the diketone (4.0g,22.19mmol) in dry THF (20mL) was added NCS (4.5g,33.71mmol) and stirred under nitrogen at room temperature overnight. The solvent was then removed and extracted with dichloromethane (3 x 10mL), the organic layer separated and washed with Na2SO4Drying, filtration, concentration and recrystallization in EtOH: MeOH ═ 2:1 afforded a white solid.

(II) Synthesis of 8-chloro-1- (cyclopropylmethyl) -3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione

To 8-chloro-3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione (500mg,2.33mmol) and K2CO3(385.8mg) in anhydrous DMF (5mL) was added (bromomethyl) cyclopropane (374.3mg) and stirred at room temperature under nitrogen for 4h. The reaction mixture was then poured into water and extracted with EA (3x 10mL), the organic layer was separated and Na was used2SO4Dry, filter, concentrate and purify by silica gel chromatography (PE: EA ═ 2:1) to give a white solid (325mg, 51.9%).

(III) Synthesis of 1- (cyclopropylmethyl) -8-mercapto-3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 8-chloro-1- (cyclopropylmethyl) -3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione (300mg) in anhydrous DMF (2mL) was added NaHS (187.6mg) and heated to 130 ℃ for 1.5H. The mixture was then cooled to room temperature and acidified to PH 3. The reaction mixture was then poured into water and extracted with dichloromethane (3x 5mL), the organic layer was separated and Na was used2SO4Drying, filtering and concentrating to obtain white solid.

(IV) Synthesis of 1- (cyclopropylmethyl) -3, 7-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione

To a mixture of 1- (cyclopropylmethyl) -8-mercapto-3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione (289.1mg) and K2CO3To a solution of (179.9mg) in anhydrous DMF (5mL) was added iodomethane (184.8mg) and the mixture was stirred at room temperature under nitrogen for 2 h. The reaction mixture was then poured into water and extracted with dichloromethane (3x 15mL), the organic layer was separated and Na was added2SO4Drying, filtration, concentration and purification on silica gel (PE: EA 4:1) gave a white solid (280mg, 92.1%).

(V) Synthesis of 1- (cyclopropylmethyl) -3, 7-dimethyl-8- (methylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

To a solution of 1- (cyclopropylmethyl) -3, 7-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione (250mg) in MeOH (5mL) was added oxone (2.74g) in H2O (5mL) solution. The mixture was then stirred at room temperature overnight. The solvent was then removed and extracted with dichloromethane (3x 15mL), the organic layer separated and washed with Na2SO4Drying, filtration, concentration and purification on silica gel (PE: EA1:1) gave 150mg (53.8%) as a white solid.1H-NMR(400MHz,CDCl3):δ4.31(s,3H),3.90(d,J=6.0Hz,2H),3.56(s,3H),3.43(s,3H),1.27-1.24(m,1H),0.48-0.41(m,4H).MS(m/z):313.09[M+H]+.

Compound 92: 3, 7-dimethyl-8- (methylsulfonyl) -1-phenyl-1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 8-chloro-3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione (4.0g,22.19mmol) in anhydrous THF (20mL) was added NCS (4.5g,33.71mmol) and stirred under nitrogen at room temperature overnight. The solvent was then removed and extracted with dichloromethane (3x 10mL), the organic layer separated and washed with Na2SO4Drying, filtration, concentration and recrystallization from EtOH: MeOH ═ 2:1 gave a white solid (2.5g, 52.3%).

(II) Synthesis of 3, 7-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione

A mixture of 8-chloro-3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione (100mg) and sodium thiomethoxide (130.5mg) in anhydrous DMF (4mL) was reacted in a microwave at 100 ℃ for 1H on a Biotage Smith synthesizer. The mixture was then cooled to room temperature and acidified to pH 5. The reaction mixture was poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was added2SO4Drying, filtering and concentrating to obtain white solid.

(III) Synthesis of 3, 7-dimethyl-8- (methylthio) -1-phenyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 3, 7-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione (50.0mg,0.22mmol) and phenylboronic acid (26.9mg,0.22mmol) in anhydrous DCM (2mL) were added TEA (90.5mg,0.88mmol) and Cu (OAc)2(60.1mg,0.33 mmol). The mixture was then stirred at room temperature overnight. The reaction mixture was then poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used2SO4Dry, filter, concentrate and purify by silica gel chromatography (PE: EA ═ 2:1) to give a white solid (15.0mg, 18.4%).1H-NMR(400MHz,CDCl3):δ7.51-7.22(m,5H),3.82(s,3H),3.60(s,3H),2.75(s,3H).MS(m/z):303.08[M+H]+.

(IV) Synthesis of 3, 7-dimethyl-8- (methylsulfonyl) -1-phenyl-1H-purine-2, 6(3H,7H) -dione

to a solution of 3, 7-dimethyl-8- (methylthio) -1-phenyl-1H-purine-2, 6(3H,7H) -dione (20.0mg,0.06mmol) in MeOH (2mL) was added oxone (162.8mg,0.26mmol) in H2O (2mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 5mL), the organic layer separated and washed with Na2SO4Dry, filter, concentrate and purify by prep TLC to give a white solid (14.3mg, 64.2%).1H-NMR(400MHz,CDCl3):δ7.54-7.21(m,5H),4.30(s,3H),3.60(s,3H),3.46(s,3H).MS(m/z):335.07[M+H]+.

Compound 93: 1-benzyl-3, 7-dimethyl-8- (methylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 8-chloro-3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione (4.0g,22.19mmol) in anhydrous THF (20mL) was added NCS (4.5g,33.71mmol) and stirred under nitrogen at room temperature overnight. The solvent was then removed and extracted with dichloromethane (3x 10mL), the organic layer separated and washed with Na2SO4Dried, filtered, concentrated and recrystallized in EtOH: MeOH 2:1,A white solid was obtained.

(II) Synthesis of 1-benzyl-8-chloro-3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione

To 8-chloro-3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione (500mg,2.33mmol) and K2CO3(385.8mg) in anhydrous DMF (5mL) was added (bromomethyl) benzene (478.6mg) and stirred at room temperature under nitrogen for 4h. The reaction mixture was then poured into water and extracted with EA (3x 10mL), the organic layer was separated and Na was used2SO4Dry, filter, concentrate and purify by silica gel chromatography (PE: EA ═ 1:1) to give a white solid (423.5mg, 58.5%).

(III) Synthesis of 1-benzyl-8-mercapto-3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 1-benzyl-8-chloro-3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione (150mg) in anhydrous DMF (5mL) was added NaHS (39.5mg) and heated to 130 ℃ for 1.5H. The mixture was then cooled to room temperature and acidified to PH 3. The reaction mixture was then poured into water and extracted with dichloromethane (3x 5mL), the organic layer was separated and Na was used2SO4Drying, filtering and concentrating to obtain white solid.

(IV) Synthesis of 1-benzyl-3, 7-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione

To 1-benzyl-8-mercapto-3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione (128mg) and K2CO3(66.99mg) in anhydrous DMF (5mL) was added iodomethane (68.89mg) and stirred at room temperature under nitrogen for 2 h. The reaction mixture was then poured into water and extracted with dichloromethane (3x 15mL), the organic layer was separated and Na was added2SO4Drying, filtration, concentration and purification on silica gel (PE: EA1:1) gave a white solid (108.3mg, 81.12%).

(V) Synthesis of 1-benzyl-3, 7-dimethyl-8- (methylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

To a solution of 1-benzyl-3, 7-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione (250mg) in MeOH (5mL) was added oxone (2.43g) in H2O (5mL) solution. The mixture was then stirred at room temperature overnight. The solvent was then removed and extracted with dichloromethane (3x 15mL),Separating the organic layer with Na2SO4Drying, filtration, concentration and purification on silica gel (PE: EA1:1) gave 138.2mg (50.1%) as a white solid.1H-NMR(400MHz,CDCl3):δ7.48-7.27(m,5H),5.19(s,2H),4.32(s,3H),3.54(s,3H),3.43(s,3H).MS(m/z):349.09[M+H]+.

Compound 94: 3, 7-dimethyl-8- (methylsulfonyl) -1-phenethyl-1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 8-chloro-3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione

to a solution of 3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione (4.0g,22.19mmol) in anhydrous THF (20mL) was added NCS (4.5g,33.71mmol) and stirred under nitrogen at room temperature overnight. The solvent was then removed and extracted with dichloromethane (3x 10mL), the organic layer separated and washed with Na2SO4Drying, filtration, concentration and recrystallization in EtOH: MeOH ═ 2:1 afforded a white solid.

(II) Synthesis of 8-chloro-3, 7-dimethyl-1-phenylethyl-1H-purine-2, 6(3H,7H) -dione

To 8-chloro-3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione (500mg,2.33mmol) and K2CO3(385.8mg) in anhydrous DMF (5mL) was added (2-bromoethyl) benzene (518.2mg) and stirred at room temperature under nitrogen for 4h. The reaction mixture was then poured into water and extracted with EA (3x 10mL), the organic layer was separated and Na was used2SO4dry, filter, concentrate and purify by silica gel chromatography (PE: EA ═ 1:1) to give a white solid (336.7mg, 47.6%).

(III) Synthesis of 8-mercapto-3, 7-dimethyl-1-phenylethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 8-chloro-3, 7-dimethyl-1-phenethyl-1H-purine-2, 6(3H,7H) -dione (150mg) in anhydrous DMF (5mL) was added NaHS (39.5mg) and heated to 130 ℃ for 1.5H. The mixture was then cooled to room temperature and acidified to PH 3. The reaction mixture was then poured into water and extracted with dichloromethane (3x 5mL), the organic layer was separated and Na was used2SO4Drying, filtering and concentrating to obtain white solid.

(IV) Synthesis of 3, 7-dimethyl-8- (methylthio) -1-phenethyl-1H-purine-2, 6(3H,7H) -dione

To 8-mercapto-3, 7-dimethyl-1-phenethyl-1H-purine-2, 6(3H,7H) -dione (128mg) and K2CO3(66.99mg) in anhydrous DMF (5mL) was added iodomethane (68.89mg) and stirred at room temperature under nitrogen for 2 h. The reaction mixture was then poured into water and extracted with dichloromethane (3x 15mL), the organic layer was separated and Na was added2SO4Drying, filtration, concentration and purification on silica gel (PE: EA1:1) gave a white solid.

(V) Synthesis of 3, 7-dimethyl-8- (methylsulfonyl) -1-phenethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 3, 7-dimethyl-8- (methylthio) -1-phenethyl-1H-purine-2, 6(3H,7H) -dione (100mg) in MeOH (5mL) was added oxone (931.5mg) in H2O (5mL) solution. The mixture was then stirred at room temperature overnight. The solvent was then removed and extracted with dichloromethane (3x 15mL), the organic layer separated and washed with Na2SO4Drying, filtration, concentration and purification on silica gel (PE: EA1:1) gave 48.9mg (45.7%) as a white solid.1H-NMR(400MHz,CDCl3):δ7.32-7.25(m,5H),4.32(s,3H),4.22(t,J=8.0Hz,2H),3.57(s,3H),3.44(s,3H),2.92(t,J=8.0Hz,2H).MS(m/z):363.10[M+H]+.

Compound 95: 3, 7-dimethyl-8- (methylsulfonyl) -1- (prop-2-ynyl) -1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 8-chloro-3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione (4.0g,22.19mmol) in anhydrous THF (20mL) was added NCS (4.5g,33.71mmol) and stirred under nitrogen at room temperature overnight. The solvent was then removed and extracted with dichloromethane (3x 10mL), the organic layer separated and washed with Na2SO4Drying, filtering, concentrating and dissolving in EtOH MeRecrystallization from OH ═ 2:1 gave a white solid (2.5g, 52.3%).

(II) Synthesis of 3, 7-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione

A mixture of 8-chloro-3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione (100mg) and sodium thiomethoxide (130.5mg) in anhydrous DMF (4mL) was reacted in a microwave at 100 ℃ for 1H on a Biotage Smith synthesizer. The mixture was then cooled to room temperature and acidified to pH 5. The reaction mixture was poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was added2SO4Dried, filtered and concentrated to give a white solid (67.8mg, 64.3%).

(III) Synthesis of 3, 7-dimethyl-8- (methylthio) -1- (prop-2-yn-1-yl) -1H-purine-2, 6(3H,7H) -dione

To 3, 7-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione (30.0mg,0.13mmol) and K2CO3(21.9mg,0.15mmol) in anhydrous DMF (2mL) was added 3-bromoprop-1-yne (18.9mg,0.15mmol) and stirred at room temperature under nitrogen for 2 h. The reaction mixture was then poured into water and extracted with dichloromethane (3x 5mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by preparative TLC (PE: EA1:1) to give a white solid (10.2mg, 34.7%).

(IV) Synthesis of 3, 7-dimethyl-8- (methylsulfonyl) -1- (prop-2-yn-1-yl) -1H-purine-2, 6(3H,7H) -dione

To a solution of 3, 7-dimethyl-8- (methylthio) -1- (prop-2-yn-1-yl) -1H-purine-2, 6(3H,7H) -dione (10mg) in MeOH (1mL) was added oxone (116.3mg) in H2O (1mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 5mL), the organic layer separated and washed with Na2SO4Dried, filtered, concentrated and purified by preparative TLC (PE: EA1:2) to give 4.8mg (45.7%) as a white solid.1H-NMR(400MHz,CDCl3):δ4.79(d,J=2.0Hz,3H),4.32(s,3H),3.59(s,3H),3.44(s,3H),2.19(t,J=2.0Hz,1H).MS(m/z):297.06[M+H]+.

Compound 96: 2- (3, 7-dimethyl-8- (methylsulfonyl) -2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-1-yl) -N, N-dimethylacetamide

(I) Synthesis of 8-chloro-3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione (4.0g,22.19mmol) in anhydrous THF (20mL) was added NCS (4.5g,33.71mmol) and stirred under nitrogen at room temperature overnight. The solvent was then removed and extracted with dichloromethane (3x 10mL), the organic layer separated and washed with Na2SO4Drying, filtration, concentration and recrystallization from EtOH: MeOH ═ 2:1 gave a white solid (2.5g, 52.3%).

(II) Synthesis of 3, 7-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione

A mixture of 8-chloro-3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione (100mg) and sodium thiomethoxide (130.5mg) in anhydrous DMF (4mL) was reacted in a microwave at 100 ℃ for 1H on a Biotage Smith synthesizer. The mixture was then cooled to room temperature and acidified to pH 5. The reaction mixture was poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was added2SO4Dried, filtered and concentrated to give a white solid (67.8mg, 64.3%).

(III) Synthesis of 2- (3, 7-dimethyl-8- (methylthio) -2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-1-yl) -N, N-dimethylacetamide

To 3, 7-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione (50.0mg) and K2CO3(36.6mg) in anhydrous DMF (2mL) was added 2-chloro-N, N-dimethylacetamide (32.1mg) and stirred at room temperature under nitrogen for 2 h. The reaction mixture was then poured into water and extracted with dichloromethane (3x 5mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by preparative TLC (PE: EA1:1) to give a white solid (43.4mg, 63.9%).

(IV) Synthesis of 2- (3, 7-dimethyl-8- (methylsulfonyl) -2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-1-yl) -N, N-dimethylacetamide

To 2- (3, 7-dimethyl-8-) (To a solution of methylthio) -2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-1-yl) -N, N-dimethylacetamide (10mg) in MeOH (1mL) was added oxone (78.9mg) in H2O (1mL) solution. The mixture was then stirred overnight. The solvent was then removed and extracted with dichloromethane (3x 5mL), the organic layer separated and washed with Na2SO4Dried, filtered, concentrated and purified by preparative TLC (PE: EA1:1) to give 6.8mg (63.1%) as a white solid.1H-NMR(400MHz,CDCl3):δ4.82(s,2H),4.28(s,3H),3.55(s,3H),3.41(s,3H),3.11(s,3H),2.97(s,3H).MS(m/z):344.10[M+H]+.

Compound 97: 4- (2- (3, 7-dimethyl-8- (methylsulfonyl) -2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-1-yl) ethyl) morpholine 4-oxide

(I) Synthesis of 8-chloro-3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione (4.0g,22.19mmol) in anhydrous THF (20mL) was added NCS (4.5g,33.71mmol) and stirred under nitrogen at room temperature overnight. The solvent was then removed and extracted with dichloromethane (3x 10mL), the organic layer separated and washed with Na2SO4Drying, filtration, concentration and recrystallization from EtOH: MeOH ═ 2:1 gave a white solid (2.5g, 52.3%).

(II) Synthesis of 3, 7-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione

A mixture of 8-chloro-3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione (100mg) and sodium thiomethoxide (130.5mg) in anhydrous DMF (4mL) was reacted in a microwave at 100 ℃ for 1H on a Biotage Smith synthesizer. The mixture was then cooled to room temperature and acidified to pH 5. The reaction mixture was poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was added2SO4Dried, filtered and concentrated to give a white solid (67.8mg, 64.3%).

(III) Synthesis of 3, 7-dimethyl-8- (methylthio) -1- (2-morpholinoethyl) -1H-purine-2, 6(3H,7H) -dione

To 3, 7-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione (15mg) and Cs2CO3(25.94mg) in anhydrous DMF (2mL) was added 4- (2-chloroethyl) morpholine hydrochloride (14.8mg) and reacted in a microwave at 110 ℃ for 1h on a BiotageSmith synthesizer. The reaction mixture was then cooled to room temperature and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used2SO4Dried, filtered and concentrated to give a yellow oil (20.1mg, 89.3%).

(IV) Synthesis of 4- (2- (3, 7-dimethyl-8- (methylsulfonyl) -2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-1-yl) ethyl) morpholine 4-oxide

To a solution of 3, 7-dimethyl-8- (methylthio) -1- (2-morpholinoethyl) -1H-purine-2, 6(3H,7H) -dione (10mg,0.03mmol) in DCM (2mL) was added m-CPBA (12.73mg,0.07 mmol). The mixture was then stirred at room temperature for 2 h. The solvent was then removed and extracted with dichloromethane (3x 5mL), the organic layer separated and washed with Na2SO4Drying, filtration, concentration and purification by preparative HPLC gave a white solid (4.1mg, 39.7%).1H-NMR(400MHz,CDCl3):δ4.60(m,2H),4.30-4.15(m,7H),4.15-3.97(m,2H),3.97-3.55(m,2H),3.41(s,3H)3.39-3.37(m,5H).MS(m/z):388.12[M+H]+.

Compound 98: 1- (furan-2-yl) -3, 7-dimethyl-8- (methylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 8-chloro-3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione (4.0g,22.19mmol) in anhydrous THF (20mL) was added NCS (4.5g,33.71mmol) and stirred under nitrogen at room temperature overnight. The solvent was then removed and extracted with dichloromethane (3x 10mL), the organic layer separated and washed with Na2SO4Drying, filtration, concentration and recrystallization from EtOH: MeOH ═ 2:1 gave a white solid (2.5g, 52.3%).

(II) Synthesis of 3, 7-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione

A mixture of 8-chloro-3, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione (100mg) and sodium thiomethoxide (130.5mg) in anhydrous DMF (4mL) was reacted in a microwave at 100 ℃ for 1H on a Biotage Smith synthesizer. The mixture was then cooled to room temperature and acidified to pH 5. The reaction mixture was poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was added2SO4Drying, filtering and concentrating to obtain white solid.

(III) Synthesis of 1- (Furan-2-yl) -3, 7-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione

a solution of CuCl (3.0mg,0.03mol), 2-furanboronic acid (59mg,0.53mmol) and 3A molecular sieve (100mg, freshly activated) in 1, 2-dichloroethane (4mL) was cooled to 0 ℃ under a nitrogen atmosphere. Anhydrous pyridine (0.3mL) was added followed by a solution of 3, 7-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione (60mg,0.265mmol) in 1, 2-dichloroethane (1 mL). The reaction mixture was allowed to warm to room temperature. Will be charged with dry O2Was attached to the flask and the mixture was stirred for 48 h. The reaction mixture was filtered and the filtrate was concentrated and purified by preparative TLC (PE/EA ═ 2:3) to give a white solid (34mg, 44%):1H NMR(400MHz,CDCl3)δ7.56-7.51(m,1H),7.54-7.52(m,1H),6.39-6.37(m,1H),3.80(s,3H),3.58(s,3H),2.74(s,3H);LC-MS 293.3[MH]+.

(IV) Synthesis of 1- (furan-2-yl) -3, 7-dimethyl-8- (methylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

To a solution of 1- (furan-2-yl) -3, 7-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione (34mg,0.12mmol) in MeOH (2.5mL) was added oxone (359mg,0.60mmol) in H2Mixture in O (2 mL). The reaction mixture was stirred at room temperature for 16 h. The reaction mixture was washed with DCM (30mL) and H2Partition between O (5 mL). Drying the organic layer (Na)2SO4) Concentrated and purified by preparative TLC (PE: EA ═ 2:3) to give a white solid (16mg, 42%):1H NMR(400MHz,CDCl3)δ7.48-7.46(m,1H),6.57-6.55(m,1H),6.42-6.40(m,1H),4.29(s,3H),3.58(s,3H),3.45(s,3H);LC-MS 325.2[MH]+.

Compound 99: 1, 7-dimethyl-8- (methylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 1, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 4-amino-1-methyl-1H-imidazole-5-carboxylic acid ethyl ester (2.0g,11.83mmol) and ethyl methylcarbamate (2.92g,28.32mmol) in dry THF (2mL) was reacted at 75 ℃ under nitrogen for 30 min. Potassium 2-methylpropane-2-oleate (1.98g,17.64mmol) was then added to the mixture and reacted at 75 ℃ overnight. The solvent was then removed and extracted with dichloromethane (3x 50mL), the organic layer separated and washed with Na2SO4Dry, filter, concentrate and purify by silica gel chromatography (DCM: MeOH ═ 20:1) to give a white solid (1.2g, 56.33%).1H-NMR(400MHz,DMSO-d6):δ11.82(s,1H),7.90(s,1H),3.84(s,3H),3.16(s,3H).MS(m/z):181.06[M+H]+.

(II) Synthesis of 8-chloro-1, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 1, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione (0.53g,2.94mmol) in anhydrous THF (10mL) was added NCS (0.59g,4.36mmol) and stirred under nitrogen at room temperature overnight. The solvent was then removed and extracted with dichloromethane (3x 20mL), the organic layer separated and washed with Na2SO4Dried, filtered and concentrated to give a white solid (0.45 g). MS (M/z) 215.03[ M + H]+.

(III) Synthesis of 1, 7-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione

To 8-chloro-1, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione (100mg,0.46mmol) in DMF (10mL) and H2To a solution of O (1mL) was added sodium methyl mercaptide (130.8mg,1.86mmol) and reacted in a microwave at 120 ℃ for 1h on a Biotage Smith synthesizer. The mixture was then cooled to room temperature and acidified to PH 3. The reaction mixture was poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was added2SO4Dried, filtered and concentrated to give a white solid (38.3 mg). MS (M/z) 227.05[ M + H]+.

(IV) Synthesis of 1, 7-dimethyl-8- (methylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

To a solution of 1, 7-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione (15.0mg,0.06mmol) in MeOH (3mL) was added oxone (163.2mg,0.26mmol) in H2O (5mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 10mL), the organic layer separated and washed with Na2SO4Dry, filter, concentrate and purify by prep TLC to give a white solid (12.8mg, 74.85%).1H-NMR(400MHz,DMSO-d6):δ12.18(s,1H),4.16(s,3H),3.48(s,3H),3.19(s,3H).MS(m/z):259.04[M+H]+.

Compound 100: 3-Ethyl-1, 7-dimethyl-8- (methylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 3-ethyl-1, 7-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione

Compound 1, 7-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione was synthesized from 4-amino-1-methyl-1H-imidazole-5-carboxylic acid ethyl ester in the manner described for compound 99.

To 1, 7-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione (15.0mg,0.06mmol) and K2CO3To a solution of (11.1mg,0.08mmol) in anhydrous DMF (2mL) was added iodoethane (12.4mg,0.08mmol) and stirred at room temperature under nitrogen for 2 h. The reaction mixture was then poured into water and extracted with dichloromethane (3x 5mL), the organic layer was separated and Na was used2SO4Drying, filtration, concentration and purification by preparative TLC yielded a white solid (11.0mg, 65.48%).1H-NMR(400MHz,CDCl3):δ4.15(q,J=6.0Hz,2H),3.83(s,3H),3.38(s,3H),2.70(s,3H),1.32(t,J=6.0Hz,3H).MS(m/z):255.08[M+H]+.

(II) Synthesis of 3-Ethyl-1, 7-dimethyl-8- (methylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

To a solution of 3-ethyl-1, 7-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione (10.0mg,0.04mmol) in MeOH (1mL) was addedInto potassium hydrogen persulfate (96.8mg) of H2O (1mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 10mL), the organic layer separated and washed with Na2SO4Dry, filter, concentrate and purify by prep TLC to give a white solid (10.5mg, 92.9%).1H-NMR(400MHz,CDCl3):δ4.31(s,3H),4.15(q,J=6.4Hz,2H),3.44(s,3H),3.41(s,3H),1.33(t,J=6.4Hz,3H).MS(m/z):287.07[M+H]+.

Compound 101: 1, 7-dimethyl-8- (methylsulfonyl) -3-propyl-1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 1, 7-dimethyl-8- (methylthio) -3-propyl-1H-purine-2, 6(3H,7H) -dione

Compound 1, 7-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione was synthesized from 4-amino-1-methyl-1H-imidazole-5-carboxylic acid ethyl ester in the manner described for compound 99.

To 1, 7-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione (30mg) and K2CO3(29.97mg) in anhydrous DMF (5mL) was added 1-iodopropane (27.1mg) and stirred at room temperature under nitrogen for 2 h. The reaction mixture was then poured into water and extracted with dichloromethane (3x 5mL), the organic layer was separated and Na was used2SO4dried, filtered, concentrated and purified by preparative TLC (PE: EA1:1) to give a white solid (31.1mg, 87.36%).

(II) Synthesis of 1, 7-dimethyl-8- (methylsulfonyl) -3-propyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 1, 7-dimethyl-8- (methylthio) -3-propyl-1H-purine-2, 6(3H,7H) -dione (30mg) in MeOH (2mL) was added oxone (275.3mg) in H2O (2mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 10mL), the organic layer separated and washed with Na2SO4Dried, filtered, concentrated and purified by preparative TLC (PE: EA1:1) to give 28.3mg (84.3%) as a white solid.1H-NMR(400MHz,CDCl3):δ4.31(s,3H),4.05(t,J=7.6Hz,2H),3.44(s,3H),3.41(s,3H),1.80-1.74(m,2H),0.96(t,J=7.6Hz,3H).MS(m/z):301.09[M+H]+.

Compound 102: 3- (cyclopropylmethyl) -1, 7-dimethyl-8- (methylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 3- (cyclopropylmethyl) -1, 7-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione

Compound 1, 7-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione was synthesized from 4-amino-1-methyl-1H-imidazole-5-carboxylic acid ethyl ester in the manner described for compound 99.

To 1, 7-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione (30mg) and K2CO3(bromomethyl) cyclopropane (21.3mg) was added to a solution of (21.97mg) in anhydrous DMF (5mL) and stirred at room temperature under nitrogen for 2 h. The reaction mixture was then poured into water and extracted with dichloromethane (3x 5mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by preparative TLC (PE: EA1:1) to give a white solid (31.9mg, 85.98%).

(II) Synthesis of 3- (cyclopropylmethyl) -1, 7-dimethyl-8- (methylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

To a solution of 3- (cyclopropylmethyl) -1, 7-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione (30mg) in MeOH (2mL) was added oxone (263.5mg) in H2O (2mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 10mL), the organic layer separated and washed with Na2SO4Drying, filtration, concentration and purification by preparative TLC (PE: EA1:1) gave 31.4mg (94.1%) as a white solid.1H-NMR(400MHz,CDCl3):δ4.32(s,3H),3.95(d,J=7.2Hz,2H),3.43(s,3H),3.42(s,3H),1.33-1.30(m,1H),0.52-0.49(m,2H),0.46-0.44(m,2H).MS(m/z):313.09[M+H]+.

Compound 103: 3-benzyl-1, 7-dimethyl-8- (methylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 3-benzyl-1, 7-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione

Compound 1, 7-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione was synthesized from 4-amino-1-methyl-1H-imidazole-5-carboxylic acid ethyl ester in the manner described for compound 99.

To 1, 7-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione (30mg) and K2CO3(bromomethyl) benzene (27.3mg) was added to a solution of (21.97mg) in anhydrous DMF (5mL) and stirred at room temperature under nitrogen for 2 h. The reaction mixture was then poured into water and extracted with dichloromethane (3x 5mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by preparative TLC (PE: EA1:1) to give a white solid (32.1mg, 76.6%).

(II) Synthesis of 3-benzyl-1, 7-dimethyl-8- (methylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

To a solution of 3-benzyl-1, 7-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione (30mg) in MeOH (2mL) was added oxone (233.1mg) in H2O (2mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 10mL), the organic layer separated and washed with Na2SO4Dried, filtered, concentrated and purified by preparative TLC (PE: EA1:1) to give 30.9mg (93.6%) as a white solid.1H-NMR(400MHz,CDCl3):δ7.47-7.28(m,5H),5.23(s,2H),4.30(s,3H),3.45(s,3H),3.40(s,3H).MS(m/z):349.09[M+H]+.

Compound 104: n- (3- (1, 7-dimethyl-8- (methylsulfonyl) -2, 6-dioxo-1H-purin-3 (2H,6H,7H) -yl) propyl) -5- ((4R) -2-oxohexahydro-1H-thieno [3,4-d ] imidazol-4-yl) pentanamide

(I) Synthesis of N- (3-chloropropyl) -5- ((4R) -2-oxohexahydro-1H-thieno [3,4-d ] imidazol-4-yl) pentanamide

To 5- ((4R) -2-oxohexahydro-1H-thieno [3, 4-d)]To a solution A of imidazol-4-yl) pentanoic acid (2.0g,8.18mmol) and CDI (2.0g,12.33mmol) in anhydrous DMF/THF (20mL/5mL) was added DIEA (1.6g,12.37 mmol). The mixture was stirred at 0 ℃ for 0.5 h. To an additional solution B of 3-chloroprop-1-amine hydrochloride (1.4g,10.76mmol) in anhydrous DMF (10mL) was added DIEA (1.6g,12.37mmol) and stirred at room temperature for 0.5 h. Solution B was then added to solution a and stirred at room temperature overnight. The solvent was removed and the pH was adjusted to 5. The mixture was then extracted with DCM (3 × 50mL) and NaHCO3The organic layer was washed (3 × 50mL) and recrystallized from DCM: PE ═ 7:3 to give a white solid (1.82g, 37.2%).1H-NMR(400MHz,DMSO-d6):δ7.87-7.83(m,1H),6.42(s,1H),6.35(s,1H),4.31-4.28(m,1H),4.14-4.10(m,1H),3.60(t,J=6.0Hz,2H),3.16-3.07(m,3H),2.84-2.79(m,1H),2.58-2.55(m,1H),2.05(t,J=7.6Hz,2H),1.86-1.79(m,2H),1.61-1.57(m,1H),1.51-1.43(m,3H),1.33-1.26(m,2H).MS(m/z):320.11[M+H]+.

(II) Synthesis of N- (3- (1, 7-dimethyl-8- (methylsulfonyl) -2, 6-dioxo-1H-purin-3 (2H,6H,7H) -yl) propyl) -5- ((4R) -2-oxohexahydro-1H-thieno [3,4-d ] imidazol-4-yl) pentanamide

To compound 99(50.0mg,0.19mmol) and K2CO3(32.1mg,0.23mmol) in anhydrous DMF (10mL) was added N- (3-chloropropyl) -5- ((4R) -2-oxohexahydro-1H-thieno [3, 4-d)]Imidazol-4-yl) pentanamide (74.2mg,0.23mmol) and stirring at room temperature under nitrogen for 2 h. The reaction mixture was then poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used2SO4Drying, filtration, concentration and purification by preparative HPLC gave a white solid (8.2mg, 7.8%).1H-NMR(400MHz,CDCl3):δ6.71-6.68(m,1H),6.28-6.23(m,1H),5.83-5.79(m,1H),4.59-4.56(m,1H),4.41-4.38(m,1H),4.33(s,3H),4.16(t,J=6.0Hz,2H),3.41(s,6H),3.22-3.18(m,5H),2.96-2.92(m,1H),2.78-2.74(m,1H),2.30-2.22(m,2H),1.97-1.94(m,2H),1.74-1.69(m,2H),1.51-1.47(m,2H).MS(m/z):542.18[M+H]+.

Compound 105: n- (2- (2- (2- (1, 7-dimethyl-8- (methylsulfonyl) -2, 6-dioxo-1H-purin-3 (2H,6H,7H) -yl) ethoxy) ethyl) -5- ((4R) -2-oxohexahydro-1H-thieno [3,4-d ] imidazol-4-yl) pentanamide

To compound 99(50.0mg,0.19mmol) and K2CO3(32.1mg,0.23mmol) in anhydrous DMF (10mL) was added 4-methylbenzenesulfonic acid 2- (2- (2- (5- ((4R) -2-oxohexahydro-1H-thieno [3, 4-d)]imidazol-4-yl) pentanamido) ethoxy) ethyl ester (123.1mg,0.23mmol) and stirred under nitrogen at room temperature overnight. The reaction mixture was then poured into water and extracted with dichloromethane (3x 5mL), the organic layer was separated and Na was used2SO4Drying, filtration, concentration and purification by preparative HPLC gave 15.2mg (12.77%) as a white solid.1H-NMR(400MHz,CDCl3):δ6.45-6.32(m,2H),5.81-5.77(m,1H),4.59-4.55(m,1H),4.40-4.35(m,1H),4.31-4.29(m,5H),3.87-3.82(m,2H),3.63-3.65(m,2H),3.56-3.50(m,4H),3.41-3.40(m,8H),3.21-3.11(m,1H),2.96-2.89(m,1H),2.77-2.73(m,1H),2.28-2.19(m,2H),1.75-1.64(m,4H),1.50-1.40(m,2H).MS(m/z):616.21[M+H]+.

Compound 106: 1, 7-dimethyl-8- (methylsulfonyl) -3- (prop-2-ynyl) -1H-purine-2, 6(3H,7H) -dione

To compounds 99(20mg) and K2CO3(12.8mg) in anhydrous DMF (5mL) was added 3-bromoprop-1-yne (10.96mg) and stirred at room temperature under nitrogen for 2 h. The reaction mixture was then poured into water and extracted with dichloromethane (3x 5mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by preparative TLC (PE: EA1:1) to give 10.3mg (44.89%) as a white solid.1H-NMR(400MHz,CDCl3):δ4.82(d,J=2.4Hz,2H),4.31(s,3H),3.46(s,3H),3.42(s,3H),2.25(t,J=2.4Hz,1H).MS(m/z):297.06[M+H]+.

Compound 107: 2- (1, 7-dimethyl-8- (methylsulfonyl) -2, 6-dioxo-1H-purin-3 (2H,6H,7H) -yl) acetonitrile

To compounds 99(10mg) and K2CO3(6.4mg) in anhydrous DMF (5mL) 2-bromoacetonitrile (5.6mg) was added and stirred at room temperature under nitrogen for 2 h. The reaction mixture was then poured into water and extracted with dichloromethane (3x 5mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by preparative TLC (PE: EA1:1) to give 4.7mg (40.9%) as a white solid.1H-NMR(400MHz,CDCl3):δ4.97(s,2H),4.33(s,3H),3.46(s,3H),3.43(s,3H).MS(m/z):298.05[M+H]+.

Compound 108: 1, 7-dimethyl-8- (methylsulfonyl) -3- (3-phenylprop-2-ynyl) -1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of (3-bromoprop-1-yn-1-yl) benzene

To a DME solution of 3-phenylprop-2-yn-1-ol (300mg) was slowly added tribromophosphine (912.7 mg). The mixture was then stirred at room temperature under nitrogen for 2 h. The mixture was then poured into ice and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used2SO4Drying, filtration and concentration gave 323.4mg (73.2%) as a red oil.

(II) Synthesis of 1, 7-dimethyl-8- (methylsulfonyl) -3- (3-phenylprop-2-yn-1-yl) -1H-purine-2, 6(3H,7H) -dione

To compounds 99(10mg) and K2CO3(6.4mg) in anhydrous DMF (5mL) was added (3-bromoprop-1-yn-1-yl) benzene (9.1mg) and stirred at room temperature under nitrogen for 2 h. The reaction mixture was then poured into water and extracted with EA (3x 5mL), the organic layer was separated and Na was used2SO4Drying, filtration, concentration and purification by preparative TLC (PE: EA1:1) gave 8.3mg (57.7%) as a white solid.1H-NMR(400MHz,CDCl3):δ7.35-7.26(m,5H),5.04(s,2H),4.30(s,3H),3.44(s,3H),3.41(s,3H).MS(m/z):373.09[M+H]+.

Compound 109: 3- (3- (3-hydroxyphenyl) prop-2-yn-1-yl) -1, 7-dimethyl-8- (methylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 3- (3- ((tetrahydro-2H-pyran-2-yl) oxy) prop-1-yn-1-yl) phenol

To a solution of 2- (prop-2-yn-1-yloxy) tetrahydro-2H-pyran (2.6g,18mmol) in DMF (10mL) was added 3-iodophenol (1g,5mmol), copper (I) iodide (catalytic amount), (Ph)3P)4Pd (catalytic amount) and TEA (1g,9 mmol). The mixture was then heated to 90 ℃ for 2 hours under nitrogen. The resulting reaction was cooled to room temperature, filtered and extracted with EA (3 × 5mL), the organic layer was separated, and Na was added2SO4Dried, filtered, concentrated and purified by column chromatography (PE: EA 10:1) to give a yellow oil (1.3g, 93%). MS (M/z):233.3[ M + H]+.

(II) Synthesis of 3- (3-hydroxypropan-1-yn-1-yl) phenol

To a solution of 3- (3- ((tetrahydro-2H-pyran-2-yl) oxy) prop-1-yn-1-yl) phenol (1.3g,5.6mmol) in MeOH (15mL) was added 4-methylbenzenesulfonic acid (193mg,1.12mmol) and stirred at room temperature for 2H. The reaction mixture was then concentrated and extracted with EA (3 x 5mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by column chromatography (PE: EA1:1) to give a yellow oil (600mg, 75%). MS (M/z) 149.2[ M + H]+.

(III) Synthesis of 3- (3-bromoprop-1-yn-1-yl) phenol

To a solution of 3- (3-hydroxyprop-1-yn-1-yl) phenol (430mg,2.8mmol) in DCM (8mL) at 0 deg.C was slowly added triphenylphosphine (1.29g,3.2mmol) and tetrabromomethane (2.15g,5.6 mmol). The mixture was then stirred at room temperature for 2 h. The reaction mixture was then poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used2SO4Drying, filtering, concentrating and purifying by column chromatographyPurification (PE: EA5:1) gave 600mg (95%) of a yellow oil as a yellow oil. MS (M/z) 212.06[ M + H]+.

(IV) Synthesis of 3- (3- (3-hydroxyphenyl) prop-2-yn-1-yl) -1, 7-dimethyl-8- (methylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

To compounds 99(75mg,0.3mmol) and K2CO3(70mg,0.48mmol) in dry DMF (3mL) was added 3- (3-bromoprop-1-yn-1-yl) phenol (60mg,0.3mmol) and stirred at room temperature under nitrogen for 0.5 h. The reaction mixture was then poured into water and extracted with EA (3x 5mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by column chromatography (PE: EA1:1) to yield 80mg (87%) as a white solid.1H-NMR(400MHz,DMSO):δ9.64(s,1H),7.14(s,1H),6.83-6.75(m,3H),4.98(s,2H),4.21(s,3H),3.54(s,3H),3.28(s,3H).MS(m/z):389.1[M+H]+.

Compound 110: 3- (3- (3-methoxyphenyl) prop-2-yn-1-yl) -1, 7-dimethyl-8- (methylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

To compound 109(30mg,0.08mmol) and K2CO3To a solution of (22mg,0.16mmol) in anhydrous DMF (2mL) was added iodomethane (17mg,0.12mmol) and stirred at room temperature for 0.5 h. The reaction mixture was then poured into water and extracted with EA (3x 5mL), the organic layer was separated and Na was used2SO4Drying, filtration, concentration and purification by column chromatography (PE: EA 1:1) gave 26mg (83.66%) as a white solid.1H-NMR(400MHz,CDCL3):δ7.18(t,J=8.0Hz,1H),7.01-6.98(m,1H),6.93-6.92(m,1H),6.88-6.85(m,1H),5.07(s,2H),4.33(s,3H),3.78(s,3H),3.48(s,3H),3.45(s,3H).MS(m/z):403.4[M+H]+.

Compound 111: 1, 7-dimethyl-8- (methylsulfonyl) -3- (3- (3-propoxyphenyl) prop-2-yn-1-yl) -1H-purine-2, 6(3H,7H) -dione

To a solution of compound 109(30mg,0.08mmol) and K2CO3(22mg,0.16mmol) in anhydrous DMF (2mL) was added 1-iodopropane (20mg,0.12mmol) and stirred at room temperature for 0.5 h. The reaction mixture was then poured into water and extracted with EA (3x 5mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by column chromatography (PE: EA5:1) to give 25mg (80.7%) as a white solid.1H-NMR(400MHz,CDCL3):δ7.16(t,J=8.0Hz,1H),6.98-6.96(m,1H),6.93-6.92(m,1H),6.87-6.84(m,1H),5.06(s,2H),4.33(s,3H),3.88(t,J=6.4Hz,2H),3.48(s,3H),3.45(s,3H),1.82-1.73(m,2H),1.01(t,J=7.2Hz,3H).MS(m/z):431.4[M+H]+.

Compound 112: 3- (3- (4-fluorophenyl) prop-2-yn-1-yl) -1, 7-dimethyl-8- (methylsulfonyl) -3, 7-dihydro-1H-purine-2, 6-dione

Compound 109 was synthesized in a similar manner to compound 109.1H NMR(400MHz,cdcl3)δ7.41–7.36(m,2H),7.01–6.94(m,2H),5.05(s,2H),4.33(s,3H),3.47(s,3H),3.45(s,3H).MS(m/z):431.4[M+H]+.

Compound 113: 3- (3- (3-aminophenyl) prop-2-yn-1-yl) -1, 7-dimethyl-8- (methylsulfonyl) -3, 7-dihydro-1H-purine-2, 6-dione

compound 113 was prepared (yellow solid, 8mg, yield 70%) from 1, 7-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione according to a similar procedure described for compound 109.1H NMR(400MHz,cdcl3)δ7.07(t,J=8.0Hz,1H),6.82(d,J=8.0Hz,1H),6.76(s,1H),6.67(d,J=8.0Hz,1H),5.04(s,2H),4.32(s,3H),3.47(s,3H),3.44(s,3H).13C NMR(101MHz,cdcl3)δ155.25(s),150.38(s),146.16(s),145.93(s),144.89(s),129.16(s),122.76(s),122.32(s),118.20(s),115.72(s),109.89(s),83.90(s),81.84(s),42.74(s),33.58(s),29.67(s),28.35(s).HRMS-ESI+:[M+H]+Calcd for C17H17N5O4S, 388.1074; found 388.1074.

Compound 114: 1, 7-dimethyl-3- (3- (3- (methylamino) phenyl) prop-2-yn-1-yl) -8- (methylsulfonyl) -3, 7-dihydro-1H-purine-2, 6-dione

Compound 114 (yellow solid, 10mg, yield 72%) was prepared from 1, 7-dimethyl-8- (methylthio) -1H-purine-2, 6(3H,7H) -dione according to a similar procedure described for compound 109.1H NMR(400MHz,dmso)δ7.04(t,J=8.0Hz,1H),6.55-6.49(m,3H),5.79(d,J=5.2Hz,1H),4.97(s,2H),4.21(s,3H),3.55(s,3H),3.28(s,3H),2.62(d,J=5.2Hz,3H).13C NMR(101MHz,dmso)δ155.29(s),150.42(s),150.20(s),145.68(s),144.85(s),129.51(s),122.30(s),119.08(s),114.19(s),113.09(s),110.06(s),84.09(s),83.03(s),43.65(s),34.50(s),33.63(s),29.88(s),28.44(s).HRMS-ESI+:[M+H]+Calcd for C18H19N5O4S, 402.1231; found 402.1231.

Compound 115: 8- ((cyclopropylmethyl) sulfonyl) -1, 7-dimethyl-3- (3-phenylprop-2-yn-1-yl) -3, 7-dihydro-1H-purine-2, 6-dione

Compound 115 (yellow solid, 6mg, yield 70%) was prepared from 8- ((cyclopropylmethyl) thio) -1, 7-dimethyl-3, 7-dihydro-1H-purine-2, 6-dione according to a similar procedure described for compound 109.1H-NMR(400MHz,CDCl3)δ7.38-7.36(m,2H),7.30-7.25(m,3H),5.08(s,2H),4.36(s,3H),3.46(d,J=4.0Hz,2H),3.45(s,3H),1.21-1.15(m,1H),0.62(q,J=4.0Hz,2H),0.31(q,J=4.0Hz,2H).MS(m/z):413.46[M+H]+.

Compound 116: 8- ((cyclopropylmethyl) sulfonyl) -3- (3- (3-hydroxyphenyl) prop-2-yn-1-yl) -1, 7-dimethyl-3, 7-dihydro-1H-purine-2, 6-dione

Compound 16 (yellow solid, 22mg, yield 80%) was prepared from 8- ((cyclopropylmethyl) thio) -1, 7-dimethyl-3, 7-dihydro-1H-purine-2, 6-dione according to a similar procedure described for compound 109.1H-NMR(400MHz,CDCl3)δ7.13(t,J=8.0Hz,1H),6.94(d,J=8.0Hz,1H),6.85(s,1H),6.79(d,J=8.0Hz,1H),5.06(s,2H),4.36(s,3H),3.45(d,J=4.0Hz,2H),3.44(s,3H),1.19-1.17(m,1H),0.63(q,J=4.0Hz,2H),0.31(q,J=4.0Hz,2H).MS(m/z):429.46[M+H]+.

Compound 117: 8- ((cyclopropylmethyl) sulfonyl) -3- (3- (3-methoxyphenyl) prop-2-yn-1-yl) -1, 7-dimethyl-3, 7-dihydro-1H-purine-2, 6-dione

Compound 117 (white solid, 8mg, yield 73%) was prepared from 8- ((cyclopropylmethyl) thio) -1, 7-dimethyl-3, 7-dihydro-1H-purine-2, 6-dione according to a similar procedure described for compound 109. MS (M/z) 443.49[ M + H ] +.

Compound 118: 8- ((cyclopropylmethyl) sulfonyl) -3- (3- (3-fluorophenyl) prop-2-yn-1-yl) -1, 7-dimethyl-3, 7-dihydro-1H-purine-2, 6-dione

Compound 118 (white solid, 10mg) was prepared from 8- ((cyclopropylmethyl) thio) -1, 7-dimethyl-3, 7-dihydro-1H-purine-2, 6-dione according to a similar procedure described for compound 109. MS (M/z) 431.45[ M + H ] +.

Compound 119: 3- (3- (3-chlorophenyl) prop-2-yn-1-yl) -8- ((cyclopropylmethyl) sulfonyl) -1, 7-dimethyl-3, 7-dihydro-1H-purine-2, 6-dione

Compound 119 (white solid, 16mg) was prepared from 8- ((cyclopropylmethyl) thio) -1, 7-dimethyl-3, 7-dihydro-1H-purine-2, 6-dione according to a similar procedure described for compound 109. MS (M/z) 447.91[ M + H ] +.

Compound 120: 8- ((cyclopropylmethyl) sulfonyl) -3- (3- (4-fluorophenyl) prop-2-yn-1-yl) -1, 7-dimethyl-3, 7-dihydro-1H-purine-2, 6-dione

Compound 120 (white solid, 22mg, yield 85%) was prepared from 8- ((cyclopropylmethyl) thio) -1, 7-dimethyl-3, 7-dihydro-1H-purine-2, 6-dione according to a similar procedure described for compound 109.1H-NMR(400MHz,CDCl3)δ7.39–7.32(m,2H),7.00–6.92(m,2H),5.06(s,2H),4.36(s,3H),3.45(d,J=6.4Hz,2H),3.44(s,3H),1.21-1.4(m,1H),0.62(q,J=6.4Hz,2H),0.30(q,J=6.4Hz,2H).MS(m/z):431.45[M+H]+.

Compound 121: 3- (3- (4-chlorophenyl) prop-2-yn-1-yl) -8- ((cyclopropylmethyl) sulfonyl) -1, 7-dimethyl-3, 7-dihydro-1H-purine-2, 6-dione

Compound 121 (white solid, 16mg) was prepared from 8- ((cyclopropylmethyl) thio) -1, 7-dimethyl-3, 7-dihydro-1H-purine-2, 6-dione according to a similar procedure described for compound 109.1H-NMR(400MHz,CDCl3)δ7.32–7.29(m,2H),7.25–7.23(m,2H),5.06(s,2H),4.36(s,3H),3.45(d,J=6.8Hz,2H),3.44(s,3H),1.21-1.14(m,1H),0.62(q,J=6.4Hz,2H),0.30(q,J=6.4Hz,2H).MS(m/z):447.91[M+H]+.

Compound 122: 3- (3- (4-bromophenyl) prop-2-yn-1-yl) -8- ((cyclopropylmethyl) sulfonyl) -1, 7-dimethyl-3, 7-dihydro-1H-purine-2, 6-dione

According to a similar method to that described for TC013172Compound 122 (white solid, 22mg) was prepared from 8- ((cyclopropylmethyl) thio) -1, 7-dimethyl-3, 7-dihydro-1H-purine-2, 6-dione.1H-NMR(400MHz,CDCl3)δ7.41–7.39(m,2H),7.24–7.22(m,2H),5.05(s,2H),4.35(s,3H),3.45(d,J=5.6Hz,2H),3.44(s,3H),1.21-1.13(m,1H),0.62(q,J=5.6Hz,2H),0.30(q,J=5.6Hz,2H).MS(m/z):492.36[M+H]+.

Compound 123: (3- (3- (8- ((cyclopropylmethyl) sulfonyl) -1, 7-dimethyl-2, 6-dioxo-1, 2,6, 7-tetrahydro-3H-purin-3-yl) prop-1-yn-1-yl) phenyl) carbamic acid tert-butyl ester

Compound 123 (yellow solid, 10mg) was prepared from 8- ((cyclopropylmethyl) thio) -1, 7-dimethyl-3, 7-dihydro-1H-purine-2, 6-dione according to a similar procedure described for compound 109. MS (M/z) 528.59[ M + H ] +.

Compound 124: 3- (3- (3-aminophenyl) prop-2-yn-1-yl) -8- ((cyclopropylmethyl) sulfonyl) -1, 7-dimethyl-3, 7-dihydro-1H-purine-2, 6-dione

Compound 124 (yellow solid, 13mg) was prepared from 8- ((cyclopropylmethyl) thio) -1, 7-dimethyl-3, 7-dihydro-1H-purine-2, 6-dione according to a similar procedure described for compound 109.1H-NMR(400MHz,CDCl3)δ7.72–7.26(m,1H),7.03(t,J=8.0Hz,1H),6.76–6.60(m,2H),5.05(s,2H),4.33(s,3H),3.45(d,J=8.4Hz,2H),3.44(s,3H),1.21-1.13(m,1H),0.62(q,J=5.6Hz,2H),0.30(q,J=5.6Hz,2H).MS(m/z):428.48[M+H]+.

Compound 125: 8- ((cyclopropylmethyl) sulfonyl) -1, 7-dimethyl-3- (3- (3-nitrophenyl) prop-2-yn-1-yl) -3, 7-dihydro-1H-purine-2, 6-dione

Compound 125 (yellow solid, 11mg) was prepared from 8- ((cyclopropylmethyl) thio) -1, 7-dimethyl-3, 7-dihydro-1H-purine-2, 6-dione according to a similar procedure described for compound 109.1H-NMR(400MHz,CDCl3)δ8.21(s,1H),8.16–8.14(m,1H),7.7-7.68(m,1H),7.47(t,J=8.0Hz,1H),5.10(s,2H),4.37(s,3H),3.46(d,J=8.0Hz,2H),3.45(s,3H),1.21-1.13(m,1H),0.62(q,J=5.6Hz,2H),0.30(q,J=5.6Hz,2H).MS(m/z):458.46[M+H]+.

Compound 126- ((cyclopropylmethyl) sulfonyl) -1, 7-dimethyl-3- (3- (2- (methylamino) pyridin-4-yl) prop-2-yn-1-yl) -3, 7-dihydro-1H-purine-2, 6-dione

Compound 126 (white solid, 12mg) was prepared from 8- ((cyclopropylmethyl) thio) -1, 7-dimethyl-3, 7-dihydro-1H-purine-2, 6-dione according to a similar procedure described for compound 109.1H-NMR(400MHz,CDCl3)δ7.91(d,J=5.2Hz,1H),6.53(d,J=5.2Hz,1H),6.43(s,1H),5.08(s,2H),4.37(s,3H),3.45(d,J=7.2Hz,2H),3.44(s,3H),2.88(d,J=3.6Hz,3H),1.20-1.15(m,1H),0.62(q,J=5.6Hz,2H),0.30(q,J=5.6Hz,2H).MS(m/z):443.49[M+H]+.

Compound 127: 8- ((2, 5-Dimethoxybenzyl) sulfonyl) -1, 7-dimethyl-3- (3- (2- (methylamino) pyridin-4-yl) prop-2-yn-1-yl) -3, 7-dihydro-1H-purine-2, 6-dione

Compound 127 (white solid, 5mg) was prepared from 8- ((cyclopropylmethyl) thio) -1, 7-dimethyl-3, 7-dihydro-1H-purine-2, 6-dione according to a similar procedure described for compound 109. MS (M/z) 539.58[ M + H ] +.

Compound 128: 8- ((2, 5-Dimethoxybenzyl) sulfinyl) -1, 7-dimethyl-3- (3- (2- (methylamino) pyridin-4-yl) prop-2-yn-1-yl) -3, 7-dihydro-1H-purine-2, 6-dione

Compound 128 (white solid, 3mg) was prepared from 8- ((cyclopropylmethyl) thio) -1, 7-dimethyl-3, 7-dihydro-1H-purine-2, 6-dione according to a similar procedure described for compound 109. MS (M/z) 523.58[ M + H ] +.

Compound 129: 3- (2, 5-Dimethoxybenzyl) -8- (2, 5-Dimethoxybenzylsulfonyl) -1, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 2- (bromomethyl) -1, 4-dimethoxybenzene

To a solution of 1, 4-dimethoxy-2-methylbenzene (500mg,3.29mmol) in CCl4To the solution (10mL) was added NBS (585mg,3.29mmol) and AIBN (150mg) slowly. The mixture was then heated to 80 ℃ for 1 h. The mixture was filtered and the filtrate was concentrated to give a residue. The residue was diluted, extracted with EA (3 × 10mL) and concentrated to give a yellow solid.

(II) Synthesis of 3- (2, 5-Dimethoxybenzyl) -8- ((2, 5-Dimethoxybenzyl) thio) -1, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione

To 8-mercapto-1, 3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione (30.0mg,0.14mmol) and K2CO3(16mg) in dry DMF (5mL) was added 2- (bromomethyl) -1, 4-dimethoxybenzene (40mg,0.18mmol) and stirred under nitrogen at room temperature overnight. The reaction mixture was then poured into water and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by column chromatography (PE: EA1:1) to give a white solid (83 mg).

(III) Synthesis of 3- (2, 5-Dimethoxybenzyl) -8- ((2, 5-Dimethoxybenzyl) sulfonyl) -1, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 3- (2, 5-dimethoxybenzyl) -8- ((2, 5-dimethoxybenzyl) thio) -1, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione (30mg,0.06mmol) in DCM (2mL) was added m-CPBA (30mg,0.18 mmol). Then in the chamberThe mixture was stirred at room temperature for 2 h. The solvent was then removed and extracted with EA (3x 10mL), the organic layer was separated and Na was added2SO4Dried, filtered, concentrated and purified by silica gel column chromatography (PE: EA5:1) to give 25mg (81%) as a white solid.1H-NMR(400MHz,CDCl3):δ6.96(d,J=8.8Hz,1H),6.94-6.91(m,1H),6.86(d,J=3.6Hz,2H),6.84-6.81(m,1H),6.42(d,J=3.2Hz,1H),5.06(s,2H),4.78(s,2H),3.80(d,J=2.0Hz,6H),3.66(s,3H),3.63(s,3H),3.41(s,3H),3.28(s,3H).MS(m/z):545.5[M+H]+.

Compound 130: 1, 7-dimethyl-8- (methylsulfonyl) -3- ((1-phenyl-1H-1, 2, 3-triazol-5-yl) methyl) -3, 7-dihydro-1H-purine-2, 6-dione

To a solution of 1, 7-dimethyl-8- (methylthio) -3- (prop-2-yn-1-yl) -3, 7-dihydro-1H-purine-2, 6-dione (20mg,0.08mmol) in DMF (1mL) was added azidobenzene (15mg), copper (I) iodide (catalytic amount) and TEA (catalytic amount) and the mixture was stirred at room temperature under nitrogen for 2 hours. The resulting reaction was extracted with DCM (3 × 5mL), the organic layer was separated and Na was added2SO4Dried, filtered, concentrated and purified by column chromatography (DCM: MeOH 10:1) to give a white solid. Intermediate (15mg) was dissolved in THF (1mL) and H2O (1mL) in a mixture with oxone (50 mg). The mixture was then stirred at room temperature overnight. The resulting reaction was extracted with DCM (3 × 5mL), the organic layer was separated and Na was added2SO4Dried, filtered, concentrated and purified by preparative TLC (DCM: MeOH 10:1) to give a white solid (5 mg).1H-NMR(400MHz,CDCl3)δ8.13(s,1H),7.70-7.68(m,2H),7.53-7.48(m,2H),7.44-7.42(m,1H),5.44(s,2H),4.29(s,3H),3.48(s,3H),3.40(s,3H).MS(m/z):416.43[M+H]+.

Compound 131: 1, 7-dimethyl-8- (methylsulfonyl) -3- (thien-2-ylmethyl) -1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 2- (bromomethyl) thiophene

to a DME solution of thien-2-ylmethanol (1g) was slowly added tribromophosphine (3.5g) and the mixture was then stirred at room temperature under nitrogen for 2 h. The mixture was then poured into ice and extracted with dichloromethane (3x 10mL), the organic layer was separated and Na was used2SO4Drying, filtration and concentration gave 1.04g (67.1%) as a red oil.

(II) Synthesis of 1, 7-dimethyl-8- (methylsulfonyl) -3- (thien-2-ylmethyl) -1H-purine-2, 6(3H,7H) -dione

To compounds 99(10mg) and K2CO3(6.5mg) in anhydrous DMF (5mL) was added 2- (bromomethyl) thiophene (8.3mg) and stirred at room temperature under nitrogen for 2 h. The reaction mixture was then poured into water and extracted with EA (3x 5mL), the organic layer was separated and Na was used2SO4Drying, filtration, concentration and purification by preparative TLC (PE: EA1:1) gave 5.4mg (39.5%) as a white solid.1H-NMR(400MHz,CDCl3):δ7.26-7.21(m,2H),6.95-6.92(m,1H),5.40(s,2H),4.29(s,3H),3.49(s,3H),3.40(s,3H).MS(m/z):355.05[M+H]+.

Compound 132: 4- (1, 7-dimethyl-8- (methylsulfonyl) -2, 6-dioxo-1H-purin-3 (2H,6H,7H) -yl) -N-ethylbut-2-ynylamide

To compounds 99(10mg) and K2CO3To a solution of (6.5mg) in anhydrous DMF (5mL) was added 4-bromo-N-ethylbut-2-ynylamide (8.8mg) and the mixture was stirred at room temperature under nitrogen for 2 h. The reaction mixture was then poured into water and extracted with EA (3x 5mL), the organic layer was separated and Na was used2SO4Drying, filtration, concentration and purification by preparative TLC (PE: EA1:1) gave 5.2mg (36.6%) as a white solid.1H-NMR(400MHz,CDCl3):δ5.83(s,1H),4.94(s,2H),4.32(s,3H),3.45(s,3H),3.42(s,3H),3.30(q,J=7.6Hz,2H),1.14(t,J=7.6Hz,3H).MS(m/z):368.10[M+H]+.

Compound 133: 8- ((cyclopropylmethyl) sulfonyl) -3- (2-fluorobenzyl) -1, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione

(I) Synthesis of 1, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione

A solution of 4-amino-1-methyl-1H-imidazole-5-carboxylic acid ethyl ester (2.0g,11.83mmol) and ethyl methylcarbamate (2.92g,28.32mmol) in dry THF (2mL) was reacted at 75 deg.C under nitrogen for 30 min. Potassium 2-methylpropane-2-oleate (1.98g,17.64mmol) was then added to the mixture and reacted at 75 ℃ overnight. The solvent was then removed and extracted with dichloromethane (3 x 50mL), the organic layer separated and washed with Na2SO4Dry, filter, concentrate and purify by silica gel chromatography (DCM: MeOH ═ 20:1) to give a white solid (1.2g, 56.33%).1H-NMR(400MHz,DMSO-d6):δ11.82(s,1H),7.90(s,1H),3.84(s,3H),3.16(s,3H).MS(m/z):181.06[M+H]+.

(II) Synthesis of 8-chloro-1, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 1, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione (0.53g,2.94mmol) in anhydrous THF (10mL) was added NCS (0.59g,4.36mmol) and stirred under nitrogen at room temperature overnight. The solvent was then removed and extracted with dichloromethane (3x 20mL), the organic layer separated and washed with Na2SO4Dried, filtered and concentrated to give a white solid (0.45 g). MS (M/z) 215.03[ M + H]+.

(III) Synthesis of 8-mercapto-1, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 8-chloro-1, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione (500mg,2.2mmol) in anhydrous DMF was added sodium hydrosulfide (370mg,6.5mmol) and heated to 100 ℃ for 12 hours. The reaction mixture was allowed to change to pH 5 and poured slowly into water (10mL) to give a yellow precipitate and filtered to give the crude product.1H-NMR(400MHz,DMSO-d6):δ3.68(s,3H),3.38(s,3H),3.20(s,3H).MS(m/z):227.2[M+H]+.

(IV) Synthesis of 8- ((cyclopropylmethyl) thio) -1, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione

To 8-mercapto-1, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione (1g,4.7mmol) and K2CO3(1.32g,9.4mmol) in dry DMF (15mL) was added (bromomethyl) cyclopropane (580mg,4.24mmol) and stirred at room temperature under nitrogen for 2 h. The reaction mixture was then poured into water and extracted with EA (3x 10mL), the organic layer was separated and Na was used2SO4Drying, filtration, concentration and purification by column chromatography (PE: EA1:1) gave 1.06g (88.8%) as a white solid. MS (M/z) 267.3[ M + H]+.

(V) Synthesis of 8- ((cyclopropylmethyl) sulfonyl) -1, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione

To a solution of 8- ((cyclopropylmethyl) thio) -1, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione (1g,3.76mmol) in DCM (10mL) was added m-CPBA (1.95g,11.3 mmol). The mixture was then stirred at room temperature for 2 h. The solvent was then removed and extracted with dichloromethane (3x 10mL), the organic layer separated and washed with Na2SO4Dried, filtered, concentrated and purified by column chromatography (PE: EA1:1) to give a white solid (800mg, 71.4%). MS (M/z) 299.32[ M + H]+.

(VI) Synthesis of 3- (2-chlorobenzyl) -8- ((cyclopropylmethyl) sulfonyl) -1, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione

To a mixture of 8- ((cyclopropylmethyl) sulfonyl) -1, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione (20mg,0.068mmol) and K2CO3(19mg,0.134mmol) in anhydrous DMF (2mL) was added 1- (bromomethyl) -2-fluorobenzene (21mg,0.101mmol) and stirred at room temperature under nitrogen for 0.5 h. The reaction mixture was then poured into water and extracted with EA (3x 5mL), the organic layer was separated and Na was used2SO4Dried, filtered, concentrated and purified by column chromatography (PE: EA5:1) to yield finally 16mg (60%) as a white solid.1H-NMR(400MHz,CDCl3):δ7.35-7.30(m,2H),7.08-7.00(m,2H),5.34(s,2H),4.34(s,3H),3.41(d,J=7.2Hz,5H),1.11-1.06(m,1H),0.64-0.59(m,2H),0.25-0.21(m,2H).MS(m/z):407.43[M+H]+.

Compound 134: 8- ((cyclopropylmethyl) sulfonyl) -3- (3-fluorobenzyl) -1, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione

This compound was synthesized in a similar manner to that described for compound 133 to give finally 18mg (66.7%) as a white solid.1H-NMR(400MHz,CDCl3):δ7.30-7.28(m,1H),7.25(s,1H),7.18-7.15(m,1H),6.71-6.93(m,1H),5.24(s,2H),4.34(s,3H),3.43-3.40(m,5H),1.15-1.10(m,1H),0.66-0.62(m,2H),0.25-0.21(m,2H).MS(m/z):407.43[M+H]+.

Compound 135: 8- ((cyclopropylmethyl) sulfonyl) -3- (4-fluorobenzyl) -1, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione

This compound was synthesized in a similar manner as described for compound 133 to give finally 16mg (59.3%) as a white solid.1H-NMR(400MHz,CDCl3):δ7.49-7.45(m,2H),7.00-6.96(m,2H),5.21(s,2H),4.34(s,3H),3.42(d,J=7.2Hz,5H),1.17-1.07(m,1H),0.67-0.62(m,2H),0.27-0.23(m,2H).MS(m/z):407.43[M+H]+.

Compound 136: 3- (2-chlorobenzyl) -8- ((cyclopropylmethyl) sulfonyl) -1, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione

This compound was synthesized in a similar manner as described for compound 133 to finally obtain 27mg (96.4%) as a white solid.1H-NMR(400MHz,CDCl3):δ7.38(dd,J=1.6Hz,J=7.6Hz,1H),7.23-7.14(m,2H),7.05(dd,J=1.6Hz,J=7.6Hz,1H),5.39(s,2H),4.35(s,3H),3.45(s,3H),3.37(d,J=7.2Hz,2H),1.11-1.03(m,1H),0.61-0.56(m,2H),0.22-0.19(m,2H).MS(m/z):423.89[M+H]+.

Compound 137: 3- (3-chlorobenzyl) -8- ((cyclopropylmethyl) sulfonyl) -1, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione

This compound was synthesized in a similar manner to that described for compound 133 to give 20mg (71.4%) as a white solid.1H-NMR(400MHz,CDCl3):δ7.46(s,1H),7.37-7.35(m,1H),7.25-7.23(m,2H),5.22(s,2H),4.34(s,3H),3.42(t,J=4.0Hz,J=3.2Hz,5H),1.16-1.08(m,1H),0.65-0.61(m,2H),0.25-0.21(m,2H).MS(m/z):423.89[M+H]+.

Compound 138: 3- (4-chlorobenzyl) -8- ((cyclopropylmethyl) sulfonyl) -1, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione

This compound was synthesized in a similar manner to that described for compound 133 to give 16mg (58%) as a white solid.1H-NMR(400MHz,CDCl3):δ7.42-7.39(m,2H),7.28-7.27(m,1H),7.25(d,J=2.4Hz,1H),5.21(s,2H),4.34(s,3H),3.41(s,3H),3.40(d,J=7.6Hz,2H)1.15-1.07(m,1H),0.67-0.62(m,2H),0.26-0.22(m,2H).MS(m/z):423.89[M+H]+.

compound 139: 3- (2-bromobenzyl) -8- ((cyclopropylmethyl) sulfonyl) -1, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione

This compound was synthesized in a similar manner to that described for compound 133 to give 25mg (80.6%) as a white solid.1H-NMR(400MHz,CDCl3):δ7.57(dd,J=1.2Hz,J=8.0Hz,1H),7.22-7.18(m,1H),7.15-7.10(m,1H),6.98(dd,J=1.6Hz,J=7.6Hz,1H),5.37(s,2H),4.35(s,3H),3.45(s,3H),3.38(d,J=7.2Hz,2H),1.10-1.03(m,1H),0.60-0.54(m,2H),0.22-0.12(m,2H).MS(m/z):468.34[M+H]+.

Compound 140: 3- (3-bromobenzyl) -8- ((cyclopropylmethyl) sulfonyl) -1, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione

this compound was synthesized in a similar manner to that described for compound 133 to give 22mg (71%) as a white solid.1H-NMR(400MHz,CDCl3):δ7.63(s,1H),7.42-7.39(m,2H),7.20-7.16(m,1H),5.21(s,2H),4.34(s,3H),3.42(d,J=6.0Hz,5H),1.17-1.07(m,1H),0.66-0.61(m,2H),0.26-0.22(m,2H).MS(m/z):468.34[M+H]+.

Compound 141: 3- (4-bromobenzyl) -8- ((cyclopropylmethyl) sulfonyl) -1, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione

This compound was synthesized in a similar manner to that described for compound 133 to give 25mg (80.6%) as a white solid.1H-NMR(400MHz,CDCl3):δ7.43-7.41(m,2H),7.35-7.33(m,2H),5.19(s,2H),4.34(s,3H),3.40(d,J=6.0Hz,5H),1.16-1.06(m,1H),0.67-0.62(m,2H),0.26-0.22(m,2H).MS(m/z):468.34[M+H]+.

Compound 142: 8- ((cyclopropylmethyl) sulfonyl) -1, 7-dimethyl-3- (pyridin-2-ylmethyl) -1H-purine-2, 6(3H,7H) -dione

This compound was synthesized in a similar manner to that described for compound 133 to give finally 16mg (61.5%) as a white solid.1H-NMR(400MHz,CDCl3):δ8.45(d,J=4.8Hz,1H),7.66-7.62(m,1H),7.26-7.24(m,1H),7.17-7.14(1H),5.40(s,2H),4.35(s,3H),3.43(s,3H),3.32(d,J=7.2Hz,2H),1.13-1.00(m,1H),0.59-0.54(m,2H),0.19-0.15(m,2H).MS(m/z):390.43[M+H]+.

Compound 143: 8- ((cyclopropylmethyl) sulfonyl) -1, 7-dimethyl-3- (pyridin-3-ylmethyl) -1H-purine-2, 6(3H,7H) -dione

This compound was synthesized in a similar manner to that described for compound 133 to give finally 16mg (61.5%) as a white solid.1H-NMR(400MHz,CDCl3):δ8.80-8.55(m,2H),7.86(d,J=8.0Hz,1H),7.29(s,1H),5.27(s,2H),4.34(s,3H),3.42(d,J=7.2Hz,2H),3.41(s,3H),1.18-1.07(m,1H),0.68-0.63(m,2H),0.28-0.24(m,2H).MS(m/z):390.43[M+H]+.

Compound 144: 8- ((cyclopropylmethyl) sulfonyl) -1, 7-dimethyl-3- (pyridin-4-ylmethyl) -1H-purine-2, 6(3H,7H) -dione

This compound was synthesized in a similar manner to that described for compound 133 to give finally 18mg (69.2%) as a white solid.1H-NMR(400MHz,CDCl3):δ8.57(brs,2H),7.30(d,J=4.8Hz,2H),5.25(s,2H),4.35(s,3H),3.43(s,3H),3.37(d,J=7.2Hz,2H),1.13-1.04(m,1H),0.66-0.61(m,2H),0.24-0.20(m,2H).MS(m/z):390.43[M+H]+.

Compound 145: 3,5, 7-trimethyl-2- (methylsulfonyl) -2H-pyrazolo [3,4-d ] pyrimidine-4, 6(5H,7H) -dione

(I) synthesis of 6-hydrazino-1, 3-dimethylpyrimidine-2, 4(1H,3H) -dione

To a solution of 6-chloro-1, 3-dimethylpyrimidine-2, 4(1H,3H) -dione (2.0g,11.45mmol) in isopropanol (6mL) was added hydrazine hydrate (6mL) and stirred at room temperature overnight. The precipitated white solid was then filtered off and washed with water (3 × 15mL) and dried thoroughly to give TM (1.67g, 85.6%) as a white solid.1H-NMR(400MHz,DMSO-d6):δ8.06(s,1H),5.09(s,1H),4.37(s,2H),3.21(s,3H),3.08(s,3H).MS(m/z):171.08[M+H]+.

(II) Synthesis of 3,5, 7-trimethyl-2H-pyrazolo [3,4-d ] pyrimidine-4, 6(5H,7H) -dione

6-hydrazino-1, 3-dimethylpyrimidine-2, 4(1H,3H) -dione (1.6g,9.40mmol) and acetic anhydride (1)0mL) was refluxed in anhydrous pyridine (12mL) for 3 h. The reaction was then cooled to 0 ℃ and acidified with 1N HCl (30 mL). The resulting solid was collected by filtration, washed with 1N HCl (2 x 5mL), water (2 x 10mL) and dried to give TM (1.1g, 60.5%) as a white solid.1H-NMR(400MHz,DMSO-d6):δ3.64(br,1H),3.19(s,3H),2.84(s,3H),2.65(s,3H).MS(m/z):195.08[M+H]+.

(III) Synthesis of 3,5, 7-trimethyl-2- (methylsulfonyl) -2H-pyrazolo [3,4-d ] pyrimidine-4, 6(5H,7H) -dione

To 3,5, 7-trimethyl-2H-pyrazolo [3,4-d]To an aqueous solution of pyrimidine-4, 6(5H,7H) -dione (50.0mg,0.26mmol) in NaOH (aq.) (10mL) was added methanesulfonyl chloride (44.2mg,0.38 mmol). The mixture was then stirred at room temperature overnight. A solid precipitated out. The crude product was purified by preparative TLC to give TM (31.2mg, 44.6%) as a white solid.1H-NMR(400MHz,CDCl3):δ3.51(s,3H),3.42(s,3H),3.37(s,3H),2.92(s,3H).MS(m/z):273.06[M+H]+.

Compound 146: 1,3, 9-trimethyl-8- (methylsulfonyl) -3, 9-dihydro-1H-purine-2, 6-dione

To a solution of 5, 6-diamino-1, 3-dimethylpyrimidine-2, 4(1H,3H) -dione (500mg,3mmol) in 95% ethanol (3mL) was added methyl isothiocyanate (215mg) and stirred at 100 ℃ for 6H. The precipitated white solid was then filtered off and washed with water (3x 15mL) and dried thoroughly to give 250mg of 1- (6-amino-1, 3-dimethyl-2, 4-dioxo-1, 2,3, 4-tetrahydropyrimidin-5-yl) -3-methylthiourea as a yellow solid.

The yellow solid (0.04mol) was dissolved in concentrated hydrochloric acid and refluxed vigorously for 6 hours. The precipitate was filtered from the hot mixture and washed with concentrated hydrochloric acid to give 80mg of 8-mercapto-1, 3, 9-trimethyl-3, 9-dihydro-1H-purine-2, 6-dione as a yellow solid.

The residue (20mg) was dissolved in anhydrous DMF (2mL) and K was added2CO3(50mg) and iodomethane (25mg) and stirred at room temperature under nitrogen for 2 h. The reaction mixture was poured into water and extracted with dichloromethane (3)5mL), the organic layer was separated and washed with Na2SO4Drying, filtration, concentration and purification by preparative TLC yielded 1,3, 9-trimethyl-8- (methylthio) -3, 9-dihydro-1H-purine-2, 6-dione as a yellow solid (15 mg).

To a solution of 1,3, 9-trimethyl-8- (methylthio) -3, 9-dihydro-1H-purine-2, 6-dione (12mg) in MeOH (3mL) was added oxone (92mg) in H2O (3mL) solution. The mixture was then stirred at room temperature for 5 h. The solvent was then removed and extracted with dichloromethane (3x 10mL), the organic layer separated and washed with Na2SO4Drying, filtration, concentration and purification by preparative TLC yielded 1,3, 9-trimethyl-8- (methylsulfonyl) -3, 9-dihydro-1H-purine-2, 6-dione as a white solid (6 mg). MS (M/z) 273.06[ M + H]+.

Compound 147 was prepared according to the procedure outlined in the scheme:

(a)K2CO3,DMF,rt,1h;(b)K2CO3,DMF,rt,1h;(c)Pd(PPh3)4CuI, TEA, anhydrous DMF,60 ℃,6 h; (d) potassium hydrogen persulfate, MeOH H2O 1:1, rt,4H.

Compound 148 was prepared according to the procedure outlined in the scheme:

(a) Refluxing sodium ethoxide and ethanol; (b) potassium tert-butoxide, anhydrous THF,75 ℃ overnight; (c) k2CO3, DMF, rt,1 h; (d) pd (PPh)3)4CuI, TEA, anhydrous DMF,60 ℃,6 h; (e) potassium hydrogen persulfate, MeOH H2O 1:1, rt,4H.

Preparation of compound 149 according to the procedure outlined in the scheme

(a)DMF,60℃,3h;(b)K2CO3,DMF,rt,1h;(c)Pd(PPh3)4CuI, TEA, anhydrous DMF,60 ℃,6 h; (d) potassium hydrogen persulfate, MeOH H2O 1:1, rt,4H.

Preparation of Compounds 150-155 according to the methods outlined in the schemes

(a)K2CO3,DMF,rt,1h;(b)K2CO3,DMF,rt,1h;(c)Pd(PPh3)4CuI, TEA, anhydrous DMF,60 ℃,6 h; (d) potassium hydrogen persulfate, MeOH H2O 1:1, rt,4H.

Preparation of Compounds 156-158 according to the methods outlined in the scheme

(a) Anhydrous DMF at 120 ℃ for 6 h; (b) NaOCl (aq), HCl, -25 ℃; (c) k2CO3,DMF,rt,1h.

Compounds 159-175 were prepared according to the procedure outlined in the scheme:

(a)K2CO3,DMF,rt,1h;(b)K2CO3,DMF,rt,1h;(c)Pd(PPh3)4CuI, TEA, anhydrous DMF,60 ℃,6 h; (d) potassium hydrogen persulfate, MeOH H2O 1:1,rt,4h.

Compound 176 was prepared according to the procedure outlined in the scheme

(a)Pd(PPh3)4CuI, TEA, anhydrous DMF,60 ℃,6 h; (b) k2CO3DMF, rt,1 h; (c) potassium hydrogen persulfate, MeOH H2O 1:1, rt,4H.

Necrotic apoptosis assay

The method comprises the following steps: HT-29 cells were cultured in McCoy's 5A medium (Invitrogen). On the first day, HT-29 cells were plated at a density of 2500-. The next day, necrosis was induced by addition of 20ng/ml TNF-a (T), 100nM Smac mimic (S) and 20mM z-VAD (Z). At the same time, 10mM of compound from a chemical library of about 200000 compounds was fed into each well. After 24 hours of treatment, Cell viability was determined by measuring ATP levels using the Cell Titer-Glo luminescent Cell viability assay kit. CellTiter-Glo assay (Promega) was performed according to the manufacturer's instructions. Luminescence was recorded with a PerkinElmer ensspire multimode plate reader. Surviving cells were normalized to those treated with DMSO. RIPA-56 was used as a positive control for screening necrotic apoptosis inhibitors. Data are presented as mean ± standard deviation of duplicate data.

Necrotic apoptotic activity of the series 1 compounds; 147-176 activity was extrapolated.

An exemplary compound; in embodiments, the compound is selected from table 2.

TABLE 2 series 2 Compounds

Series 2; compound 1:

8- ((2-hydroxyethyl) sulfonyl) -1,3, 7-trimethyl-3, 7-dihydro-1H-purine-2, 6-dione

And (1).

To a solution of 8-chloro-1, 3-dimethyl-3, 7-dihydro-1H-purine-2, 6-dione (428mg,2.0mmol) in DMF (6mL) was added MeI (188. mu.L, 3.0mmol) and K2CO3(414mg,3.0 mmol). The mixture was then stirred at room temperature for 3 hours. With EtOAc and H2The reaction mixture was extracted 3 times. The combined organic layers were washed with brine and Na2SO4Dry, concentrate and purify further by silica gel column chromatography (DCM/MeOH ═ 200/1) to give 409mg of 1-1 as a yellow solid (90%). LC-MS (ESI) M/z [ M + H ]]+calculated 229.04, found 229.06

And 2. step 2.

Will NaSCH3(170mg,3.0mmol) was added to a solution of 1-1(456mg,2.0mmol) in DMF (6 mL). The reaction mixture was stirred at 110 ℃ for 18 hours. The solvent was evaporated under reduced pressure and further purified by silica gel column chromatography (DCM/MeOH ═ 100/1) to give 93mg of 1-2 (88%). LC-MS (ESI) M/z [ M + H ]]+Calculated 227.05, found 227.03

And 3. step 3.

To 1-2(40mg,0.18mmol) and K2CO3(61mg,0.44mmol) in anhydrous DMF (2mL)2-Bromoethanol (23. mu.L, 0.33mmol) was added and stirred at room temperature under nitrogen for 3 hours. The reaction mixture was poured into water and extracted with ethyl acetate (3 × 5mL), the organic layer was separated and Na was added2SO4Drying, filtration, concentration and further purification by silica gel column chromatography (EA/PE ═ 30/70) gave 47mg of 1-3 (80%). LC-MS (ESI) M/z [ M + H ]]+Calculated 271.08, found 271.06

And 4. step 4.

To a solution of 1-3(47mg,0.18mmol) in MeOH (2mL) was added oxone (214mg,0.35mmol) in H2O (2mL) solution. The mixture was then stirred at room temperature for 3 hours. The solvent was then removed and extracted with dichloromethane (3X 5mL), the organic layer separated and Na2SO4Drying, filtration, concentration and purification by preparative TLC gave compound 131.7 mg (60%) as a white solid.1H NMR(400Hz,CDCl3)δ4.34(s,3H),4.20(t,J=5.6Hz,2H),3.77(t,J=5.6Hz,2H),3.56(s,3H),3.42(s,3H).LC-MS(ESI)m/z:[M+H]+Calculated 303.07, found 303.16

Compound 2:

8- ((2-methoxyethyl) sulfonyl) -1,3, 7-trimethyl-3, 7-dihydro-1H-purine-2, 6-dione

And (1).

2-1 was prepared according to the method of 1 from 1-2(40mg,0.175mmol) and 1-bromo-2-methoxyethane (31 μ L,0.33mmol) in 100% yield (50 mg). LC-MS (ESI) M/z [ M + H ]]+calculated 285.09, found 285.13

And 2. step 2.

Preparation of 2 from 2-1(50mg,0.18mmol) and oxone (214mg,0.35mmol) according to the method of 1 in 43% yield (24mg) as a yellow solid.1H NMR(400Hz,CDCl3)δ4.29(s,3H),3.87(t,J=5.6Hz,2H),3.73(t,J=5.6Hz,2H),3.57(s,3H),3.41(s,3H),3.25(s,3H).LC-MS(ESI)m/z:[M+H]+Calculated 317.08, found 317.20

Compound 3:

8- ((3-hydroxypropyl) sulfonyl) -1,3, 7-trimethyl-3, 7-dihydro-1H-purine-2, 6-dione

And (1).

3-1 was prepared according to the procedure for Compound 1 from 1-2(40mg,0.175mmol) and 3-bromopropan-1-ol (25 μ L,0.26mmol) in 100% yield (49 mg). LC-MS (ESI) M/z [ M + H ]]+Calculated 285.09, found 285.13

And 2. step 2.

Preparation of 3 from 3-1(49mg,0.18mmol) and oxone (214mg,0.35mmol) according to the method of 1 was in 32% yield (17.8mg) as a yellow solid.1H NMR(400Hz,CDCl3)δ4.32(s,3H),3.80(m,2H),9.71(m,3H),3.56(s,2H),3.41(s,3H),2.15(m,3H).LC-MS(ESI)m/z:[M+H]+Calculated 317.08, found 317.20

Compound 4:

8- ((cyclopropylmethyl) sulfonyl) -7-ethyl-1, 3-dimethyl-1H-purine-2, 6(3H,7H) -dione

And (1).

Iodothane (437mg,2.8mmol) and K were added to a solution of 8-chloro-1, 3-dimethyl-3, 7-dihydro-1H-purine-2, 6-dione (500mg,2.3mmol) in DMF (10mL) at room temperature2CO3(483mg,3.5mmol) and the mixture was then stirred at room temperature overnight. Completion of the reaction was monitored by UPLC-MS. The solvent was then evaporated to give crude 566mg 4-1 as a yellow solid without further purification. LC-MS (ESI) M/z [ M + H ]]+Calculated 243.06, found 243.12

And 2. step 2.

To a solution of 4-1(566mg,2.3mmol) in DMF (10mL) at room temperature was added NaSH (170mg,3.03 mmol). The mixture was then stirred at 110 ℃ for 2 hours. Completion of the reaction was monitored by UPLC-MS. The solvent was then evaporated to give crude 500mg 4-2 as a yellow solid without further purification. LC-MS (ESI) M/z [ M + H ]]+Calculated 241.07, found 241.01

And 3. step 3.

To a solution of 4-2(200mg,0.83mmol) in DMF (3mL) at room temperature was added (bromomethyl) cyclopropane (135mg,1.0mmol) and K2CO3(173mg,1.25mmol), and the mixture was then stirred at room temperature for 2 hours. Completion of the reaction was monitored by UPLC-MS. The reaction mixture was poured into water and extracted with DCM. The organic layer was separated and the aqueous layer was repeatedly extracted with DCM (2X 20 mL). With Na2SO4The combined extracts were dried and concentrated. The resulting residue was purified by silica gel column chromatography (PE: EA ═ 1:1) to give 234mg of 4-3 as a yellow solid in yield: 95 percent. LC-MS (ESI) M/z [ M + H ]]+Calculated 295.12, found 295.20

And 4. step 4.

To 4-3(100mg,0.34mmol) of methanol (3mL) and H2To a solution of O (1mL) was added oxone (626mg,1.2 mmol). The mixture was then stirred at room temperature for 20 hours. Completion of the reaction was monitored by UPLC-MS. The reaction mixture was poured into water and extracted with DCM. The organic layer was separated and the aqueous layer was repeatedly extracted with DCM (2X 20 mL). With Na2SO4The combined extracts were dried and concentrated. The resulting residue was purified by silica gel column chromatography (PE: EA ═ 5:1) to give 72mgd of 4 as a yellow solid in yield: and (4) 64.9%. 1H NMR (400MHz, CDCl)3)δ4.67(q,J=8Hz,2H),3.98-3.93(m,2H),3.41(s,3H),3.32-3.29(m,3H),1.38(t,J=8Hz,3H),1.04-0.96(m,1H),0.53(q,J=4Hz,2H),0.18(q,J=4Hz,2H).LC-MS(ESI)m/z:[M+H]+Calculated 327.10, found 327.37.

Compound 5:

7- (2-hydroxyethyl) -1, 3-dimethyl-8- (methylsulfonyl) -3, 7-dihydro-1H-purine-2, 6-dione

And (1).

To a solution of 8-chloro-1, 3-dimethyl-3, 7-dihydro-1H-purine-2, 6-dione (214mg,1.0mmol) in DMF (5mL) was added 2-bromoethanol (106. mu.L, 1.5mmol) and K2CO3(276mg,2.0 mmol). Heating the mixture to 11 by microwave radiationThe temperature was maintained at 0 ℃ for 1 hour. Evaporation of the solvent under reduced pressure gave the crude product 5-1, which was used in the next reaction step without further purification. For 5-1, LC-MS (ESI) M/z [ M + H ]]+Calculated 259.05, found 259.11.

And 2. step 2.

Will NaSCH3(105mg,1.5mmol) was added to a solution of 5-1 in DMF (4 mL). The reaction mixture was stirred at room temperature for 4 hours. Evaporation of the solvent under reduced pressure gave crude 5-2 and further purification by silica gel column chromatography (DCM/MeOH ═ 100/1) to give 5-2 as a white solid (34% over two steps). LC-MS (ESI) M/z [ M + H ]]+Calculated 271.08, found 271.01.

And 3. step 3.

Preparation of 5 from 5-2(40mg,0.15mmol) and oxone (272mg,0.44mmol) according to the method of 1 in 46% yield (21.0mg) as a white solid.1H NMR(400Hz,CDCl3)δ4.94(t,J=5.2Hz,2H),4.05(t,J=5.2Hz,2H),3.58(s,3H),3.45(s,3H),3.42(s,3H).LC-MS(ESI)m/z:[M+H]+Calculated 303.27, found 303.34.

Compound 6: 7- (2-methoxyethyl) -1, 3-dimethyl-8- (methylsulfonyl) -3, 7-dihydro-1H-purine-2, 6-dione and compound 7: 7- (2-methoxyethyl) -1, 3-dimethyl-8- (methylsulfinyl) -3, 7-dihydro-1H-purine-2, 6-dione

And (1).

6-1 was prepared from 8-chloro-1, 3-dimethyl-1H-purine-2, 6(3H,7H) -dione (214mg,1.0mmol) and 1-bromo-2-methoxyethane according to the procedure for Compound 1. 6-1 was used in the next reaction step without any further purification. LC-MS (ESI) M/z [ M + H ]]+Calculated 273.07, found 273.13

And 2. step 2.

The method according to 1, from 6-1(2724mg,1.0mmol) and NaSCH3(105mg,1.5mmol) preparation 6-2 in 36% yield (two steps). LC-MS (ESI) M/z [ M + H ]]+Calculated 285.09, found 285.13

And 3. step 3.

According to1 from 6-2(80mg,0.28mmol) and oxone (522mg,0.85mmol) to give 6 in 12% yield (10.0mg) as a white solid.1H NMR:(400Mz,CDCl3):δ4.49(t,J=5.2Hz,2H),3.74(t,J=5.2Hz,2H),3.54(s,3H),3.37(s,3H),3.32(s,3H)3.31(s,3H).LC-MS(ESI)m/z:[M+H]+Calculated 317.18, found 317.24.

Preparation of 7 from 6-2(80mg,0.28mmol) and oxone (522mg,0.85mmol) according to the method of 1 in 10% yield (9.2mg) as a white solid.1H NMR:(400Mz,CDCl3):δ4.93-4.88(m,1H),4.69-4.63(m,1H),3.71-3.65(m,2H),3.59(s,3H),3.39(s,3H),3.27(s,3H),3.21(s,3H).LC-MS(ESI)m/z:[M+H]+calculated 301.20, found 301.21.

Compound 8: 3- (3- (3-hydroxyphenyl) prop-2-yn-1-yl) -1, 7-dimethyl-8- (methylsulfinyl) -3, 7-dihydro-1H-purine-2, 6-dione

Prepared according to the procedure for compound 1 from 3- (3- (3-hydroxyphenyl) prop-2-yn-1-yl) -1, 7-dimethyl-8- (methylthio) -3, 7-dihydro-1H-purine-2, 6-dione (17mg,0.048mmol) and oxone (73mg,0.12mmol) in 55% yield (9.8mg) as a yellow solid.1H NMR(400Hz,CDCl3)δ7.08(m,1H),6.90(m,1H),8.84(m,1H),6.75(m,1H),5.04(s,2H),4.24(s,3H),3.43(s,3H),3.23(s,3H).LC-MS(ESI)m/z:[M+H]+Calculated 373.09, found 373.13

Compound 9- (3- (1H-indol-6-yl) prop-2-yn-1-yl) -1, 7-dimethyl-8 (methylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

And (1).

In N2To a solution of 1, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione (748mg,4.2mmol) in THF (20mL) was added NCS (832mg,6.3mmol) under atmosphere. The mixture was then stirred at room temperature overnight. Completion of the reaction was monitored by UPLC-MS. Pouring the reaction mixture intoWater and extracted with DCM. The organic layer was separated and the aqueous layer was repeatedly extracted with DCM (2X 20 mL). With Na2SO4The combined extracts were dried and concentrated. The resulting residue was purified by silica gel column chromatography (PE: EA ═ 5:1) to give 908mg of 9-1 as a yellow solid, yield: 100 percent. LC-MS (ESI) M/z [ M + H ]]+calculated 215.03, found 215.09

And 2. step 2.

To a solution of 9-1(1.33g,6.2mmol) in DMF (20mL) at room temperature was added 3-bromoprop-1-yne (1.47g,12.4mmol) and K2CO3(1.71g,12.4mmol), and then the mixture was stirred at room temperature for 2 hours. Completion of the reaction was monitored by UPLC-MS. The reaction mixture was poured into water and extracted with DCM. The organic layer was separated and the aqueous layer was repeatedly extracted with DCM (2X 20 mL). With Na2SO4The combined extracts were dried and concentrated. The resulting residue was purified by silica gel column chromatography (PE: EA ═ 3:1) to give 2.01g of 9-2 as a yellow solid, yield: 93.5 percent. LC-MS (ESI) M/z [ M + H ]]+Calculated 253.03, found 253.05.

And 3. step 3.

To a solution of 9-2(200mg,0.80mmol) in DMF (5mL) was added NaSMe (67mg,0.95mmol) at room temperature. The mixture was then stirred at room temperature for 2 hours. Completion of the reaction was monitored by UPLC-MS. The reaction mixture was poured into water and extracted with DCM. The organic layer was separated and the aqueous layer was repeatedly extracted with DCM (2X 20 mL). With Na2SO4The combined extracts were dried and concentrated. The resulting residue was purified by silica gel column chromatography (PE: EA ═ 3:1) to give 271mg of 9-3 as a yellow solid in yield: 90 percent. LC-MS (ESI) M/z [ M + H ]]+Calculated 264.07, found 264.12

And 4. step 4.

To 9-3(271mg,0.79mmol) of methanol (9mL) and H2To a solution of O (3mL) was added oxone (1.46g,2.38 mmol). The mixture was then stirred at room temperature for 20 hours. Completion of the reaction was monitored by UPLC-MS. The reaction mixture was poured into water and extracted with DCM. The organic layer was separated and the aqueous layer was repeatedly extracted with DCM (2X 20 mL). With Na2SO4The combined extracts were dried and concentrated. The obtained residue was purified by silica gel column chromatography (PE: EA: 5:1) to obtainTo 205mg of 9-4 as a yellow solid, yield: 87.2 percent. LC-MS (ESI) M/z [ M + H ]]+Calculated 297.06, found 297.12

And 5. step 5.

In N2To a solution of 9-4(40mg,0.14mmol) in DMF (2mL) under atmosphere was added 6-iodo-1H-indole (40mg,0.16mmol), TEA (33mg,0.32mmol), Pd (PPh)3)4(8mg,0.007mmol) and CuI (3mg,0.014 mmol). The mixture was then degassed for 15 min. The mixture was then stirred at 55 ℃ for 4 hours. Completion of the reaction was monitored by UPLC-MS. The reaction mixture was poured into water and extracted with DCM. The organic layer was separated and the aqueous layer was repeatedly extracted with DCM (2X 20 mL). With Na2SO4The combined extracts were dried and concentrated. The resulting residue was purified by silica gel column chromatography (PE: EA ═ 1:1) to give 20mg of 9 as a yellow solid in yield: 36.4 percent. 1H NMR (400MHz, DMSO). delta.11.23 (s,1H),7.50(d, J ═ 8Hz,1H),7.44-7.42(M,2H),6.99(d, J ═ 8Hz,1H),6.43(s,1H),5.01(s,2H),4.22(s,3H),3.56(s,3H),3.29(s,3H). LC-MS (ESI) M/z: [ M + H]+Calculated 412.40, found 412.43.

Compound 10: n- (3- (3- (1, 7-dimethyl-8- (methylsulfonyl) -2, 6-dioxo-1H-purin-3 (2H,6H,7H) -yl) prop-1-yn-1-yl) phenyl) acetamide

And (1).

Preparation of 10-1 from 9-4(50mg,0.17mmol) and 3-iodoaniline (45mg,0.20mmol) according to the procedure for compound 9, in 74% yield (48 mg). LC-MS (ESI) M/z [ M + H ]]+Calculated 388.10, found 388.19

And 2. step 2.

To a solution of 10-1(20mg,0.052mmol) in DCM (2mL) at 0 deg.C was added TEA (12mg,0.11mmol) and Ac2O (7mg,0.062 mmol). The mixture was then stirred at room temperature for 3 hours. Completion of the reaction was monitored by UPLC-MS. The reaction mixture was poured into water and extracted with DCM. The organic layer was separated and the aqueous layer was repeatedly extracted with DCM (2X 20 mL). With Na2SO4The combined extracts were dried and concentrated. The residue obtained is chromatographed on a column of silica gelSpectral purification (PE: EA ═ 1:1) afforded 14mg of 10 as a yellow solid in yield: and (3.6). 1H NMR (400MHz, DMSO) δ 10.00(s,1H),7.74(t, J ═ 4Hz,1H),7.48-7.45(M,1H),7.27(t, J ═ 8Hz,1H),7.08-7.05(M,1H),5.00(s,2H),4.21(s,3H),3.54(s,3H),3.28(s,3H),2.03(s,3H), LC-ms (esi) M/z: [ M + H,3H ], 3.5 (s,3H),3.28(s,3H), and [ M + H,3H ], [ LC-ms (esi) ]]+Calculated 430.41, found 430.45.

Compound 11: n- (3- (3- (1, 7-dimethyl-8- (methylsulfonyl) -2, 6-dioxo-1H-purin-3 (2H,6H,7H) -yl) prop-1-yn-1-yl) phenyl) benzamide

Prepared according to the procedure for compound 10 from 10-1(24mg,0.062mmol), TEA (14mg,0.14mmol) and benzoyl chloride (11mg,0.074mmol) as 11 in 55% yield (16 mg).1H NMR(400MHz,DMSO)δ10.30(s,1H),7.94(s,1H),7.93-7.92(m,2H),7.77-7.74(m,1H),7.60-7.58(m,1H),7.55-7.51(m,2H),7.35(t,J=8Hz,1H),7.16-7.14(m,1H),5.02(s,2H),4.21(s,3H),3.56(s,3H),3.29(s,3H).492.52.LC-MS(ESI)m/z:[M+H]+Calculated 492.13, found 492.20.

Compound 12: 3- (3- (3- ((2- (2-methoxyethoxy) ethyl) amino) phenyl) prop-2-yn-1-yl) -1, 7-dimethyl-8- (methylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

And (1).

To a solution of tert-butyl (3-iodophenyl) carbamate (1 eq) in THF at 0 deg.C was added NaH (1.2 eq) and stirred at room temperature for 30 min. 1-bromo-2- (2-methoxyethoxy) ethane (1.2 eq) was then added to the reaction mixture and stirred overnight. Completion of the reaction was monitored by UPLC-MS. The reaction mixture was poured into water and extracted with DCM. The organic layer was separated and the aqueous layer was repeatedly extracted with DCM (2X 20 mL). With Na2SO4the combined extracts were dried and concentrated. The resulting residue was purified by silica gel column chromatography (PE: EA ═ 5:1) to give 12-1 as a yellow solid.

And 2. step 2.

To a solution of 12-1(1 eq) in DCM at 0 deg.C was added TFA (5 eq). The mixture was then stirred at room temperature for 4 hours. Completion of the reaction was monitored by UPLC-MS. The reaction mixture was poured into water and extracted with DCM. The organic layer was separated and the aqueous layer was repeatedly extracted with DCM (2X 20 mL). With Na2SO4The combined extracts were dried and concentrated. The resulting residue was purified by silica gel column chromatography (PE: EA ═ 1:1) to give 12-2 as a yellow solid.

And 3. step 3.

In N2To a 12-2(1 eq) solution in DMF was added under an atmosphere 9-4(1.2 eq), TEA (2.4 eq), Pd (PPh)3)4(0.05 eq.) and CuI (0.1 eq.). The mixture was then stirred at 55 ℃ for 4 hours. Completion of the reaction was monitored by UPLC-MS. The reaction mixture was poured into water and extracted with DCM. The organic layer was separated and the aqueous layer was repeatedly extracted with DCM (2X 20 mL). With Na2SO4The combined extracts were dried and concentrated. The resulting residue was purified by silica gel column chromatography (PE: EA ═ 1:1) to give 12 as a yellow solid.1H NMR(400MHz,CDCl3)δ7.12(t,J=8Hz,1H),6.87(d,J=8Hz,1H),6.83(s,1H),6.74(d,J=8Hz,1H),5.05(s,2H),4.33(s,3H),3.72(t,J=4Hz,2H),3.66-3.64(m,2H),3.57-3.55(m,2H),3.48(s,3H),3.45(s,3H),3.40(s,3H),3.30(t,J=4Hz,2H).LC-MS(ESI)m/z:[M+H]+calculated 490.47, found 490.54.

Compound 13: 3- (3- (3- (benzylamino) phenyl) prop-2-yn-1-yl) -1, 7-dimethyl-8- (methylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

Preparation 13 was prepared according to method 12. 1H NMR (400MHz, DMSO) δ 7.31(s,2H),7.30(d, J ═ 4Hz,2H),7.00(t, J ═ 8Hz,1H),6.60-6.52(M,3H),6.70(t, J ═ 4Hz,1H),4.94(s,2H),4.23(d, J ═ 8Hz,2H),4.19(s,3H),3.52(s,3H),3.26(s,3H), LC-ms (esi) M/z: [ M + H ] M/z]+Calculated 478.46, found 478.54.

Compound 14,1, 7-dimethyl-8- (methylsulfonyl) -3- (3- (3- ((2-morpholinoethyl) amino) phenyl) prop-2-yn-1-yl) -1H-purine-2, 6(3H,7H) -dione

Preparation 14 was made according to method 12.1H NMR(400Hz,CDCl3)δ7.05(t,J=8.0Hz,1H),6.72(d,J=7.2Hz,1H),6.64(s,1H),6.56(d,J=8.0Hz,1H),5.04(s,2H),4.30(s,3H),3.73(m,4H),3.43(s,3H),3.46(s,3H),3.15(m,2H),2.66(m,2H),2.52(m,4H).LC-MS(ESI)m/z:[M+H]+Calculated 501.18, found 501.27

Compound 15: 3- (3- (3- ((2- (dimethylamino) ethyl) amino) phenyl) prop-2-yn-1-yl) -1, 7-dimethyl-8- (methylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

preparation 15 was prepared according to the method of 12.1H NMR(400MHz,CDCl3)δ7.07(t,J=8Hz,1H),6.75(d,J=8Hz,1H),6.66(s,1H),6.60(d,J=8Hz,1H),5.30(s,4H),5.05(s,2H),4.33(s,3H),3.48(s,3H),3.44(s,3H),3.20(s,3H),2.71(s,3H).LC-MS(ESI)m/z:[M+H]+Calculated 459.17, found 459.25

Compound 16: 1, 7-dimethyl-8- (methylsulfonyl) -3- (3- (3- (pentylamino) phenyl) prop-2-yn-1-yl) -1H-purine-2, 6(3H,7H) -dione

Preparation 16 was made according to method 12.1H NMR(400MHz,CDCl3)δ7.09(t,J=8Hz,1H),6.81(d,J=8Hz,1H),6.74(s,1H),6.66(s,1H),5.05(s,2H),4.33(s,3H),3.48(s,3H),3.45(s,3H),3.07(t,J=8Hz,2H),1.36-1.32(m,6H),0.90(t,J=4Hz,3H).LC-MS(ESI)m/z:[M+H]+Calculated 459.47, found 458.55.

Compound 17: n- (3- (3- (1, 7-dimethyl-8- (methylsulfonyl) -2, 6-dioxo-1H-purin-3 (2H,6H,7H) -yl) prop-1-yn-1-yl) phenyl) -2-morpholinylacetamide

And (1).

at room temperature under N2To a solution of 2-morpholinylacetic acid (100mg,0.69mmol) in DMF (2mL) under atmosphere was added 3-iodoaniline (151mg,0.69mmol), EDCI (199mg,1.03mmol) and DMAP (127mg,1.03 mmol). The mixture was then stirred at 55 ℃ for 4 hours. Completion of the reaction was monitored by UPLC-MS. The reaction mixture was poured into water and extracted with DCM. The organic layer was separated and the aqueous layer was repeatedly extracted with DCM (2X 20 mL). With Na2SO4The combined extracts were dried and concentrated. The resulting residue was purified by silica gel column chromatography (PE: EA ═ 1:1) to give 214mg 17-1 as a yellow solid in yield: 89.9 percent. LC-MS (ESI) M/z [ M + H ]]+Calculated 347.02, found 347.14.

Step 2 Sonogashira coupling

Preparation 17 according to the procedure of 12 in 31% yield (16 mg).1H NMR(400MHz,CDCl3)δ7.68(s,1H),7.55(d,J=8Hz,1H),7.24(s,1H),7.16(d,J=8Hz,1H),5.06(s,2H),4.34(s,3H),3.84(s,4H),3.49(s,3H),3.45(s,3H),2.10(s,2H),1.25(s,4H).LC-MS(ESI)m/z:[M+H]+Calculated 515.48, found 515.55.

Compound 18: 1, 7-dimethyl-8- (methylsulfonyl) -3- (3- (3- ((2- (piperidin-1-yl) ethyl) amino) phenyl) prop-2-yn-1-yl) -1H-purine-2, 6(3H,7H) -dione

Preparation of 18 from 3-iodo-N- (2- (piperidin-1-yl) ethyl) aniline (37mg,0.11mmol) according to method 12 in 17% yield (12mg) as a yellow solid.1H NMR(400Hz,CDCl3)δ7.70(s,1H),7.53(d,J=3.2Hz,1H),7.48(m,1H),7.28(m,1H),5.09(s,2H),4.29(s,3H),3.62(m,4H),3.51(s,3H),3.44(s,3H),2.62(m,4H),1.72(m,3H),1.42(m,3H).LC-MS(ESI)m/z:[M+H]+Calculated 499.38, found 499.42

Compound 19: 8- ((cyclopropylmethyl) sulfonyl) -1, 7-dimethyl-3- (3- (3- ((2-morpholinoethyl) amino) phenyl) prop-2-yn-1-yl) -3, 7-dihydro-1H-purine-2, 6-dione

Preparation of 19 from 3-iodo-N- (2-morpholinoethyl) aniline (30mg,0.09mmol) according to method 12, in 45% yield (18mg) as a yellow solid.1H NMR(400Hz,CDCl3)δ7.05(m,1H),6.72(d,J=7.2Hz,1H),6.64-6.58(m,2H),5.05(s,2H),4.32(s,3H),3.83-3.70(m,4H),3.43(s,3H),3.40(m,2H),3.19(m,1H),2.73(m,1H),2.59(m,2H),0.94(m,1H),0.62(m,2H),0.32(m,2H).LC-MS(ESI)m/z:[M+H]+Calculated 541.22, found 541.30

Compound 20: n- (3- (3- (1, 7-dimethyl-8- (methylsulfonyl) -2, 6-dioxo-1H-purin-3 (2H,6H,7H) -yl) prop-1-yn-1-yl) -5-fluorophenyl) -2-morpholinylacetamide

Preparation of 20 according to the method of 17 in 31% yield (16mg) as a yellow solid.1H NMR(400MHz,CDCl3)δ9.97(s,1H),7.60(d,J=12Hz,1H),7.55(s,1H),6.96(d,J=4Hz,1H),5.00(s,2H),4.20(s,3H),3.63-3.60(m,4H),3.54(s,3H),3.27(s,3H),3.11(s,2H),2.48-2.45(m,4H).LC-MS(ESI)m/z:[M+H]+533.45, found 533.54.

Compound 21: 3- (3- (3-methoxy-5- ((2-morpholinoethyl) amino) phenyl) prop-2-yn-1-yl) -1, 7-dimethyl-8- (methylsulfonyl) -1H-purine-2, 6(3H,7H) -dione

Prepared according to the method of 12 from 3-bromo-5-methoxy-N- (2-morpholinoethyl) aniline and 1, 7-dimethyl-8- (methylsulfonyl) -3- (prop-2-yn-1-yl) -1H-purine-2, 6(3H,7H) -dione in 9.4% (6mg) as a yellow solid. 1H NMR (400MHz, CDCl)3)δ6.44(s,1H)6.39(s,1H),6.28(s,1H),5.04(s,2H),4.34(s,3H),3.97(s,4H),3.74(s,3H),3.49(s,3H),3.44(s,3H),2.22(t,J=8Hz,2H)2.10(s,2H),1.25(s,4H).LC-MS(ESI)m/z:[M+H]+531.51, found 531.60.

Compound 22: n- (4-chloro-3- (3- (8- ((cyclopropylmethyl) sulfonyl) -1, 7-dimethyl-2, 6-dioxo-1H-purin-3 (2H,6H,7H) -yl) prop-1-yn-1-yl) phenyl) -2-morpholinylacetamide

Prepared according to the method of 12 from N- (4-chloro-3-iodophenyl) -2-morpholinylacetamide and 8- ((cyclopropylmethyl) sulfonyl) -1, 7-dimethyl-3- (prop-2-yn-1-yl) -1H-purine-2, 6(3H,7H) -dione in 16% yield (12mg) as a yellow solid.1H NMR(400MHz,DMSO)δ9.92(s,1H),7.88(d,J=4Hz,1H),7.63(dd,J=4,8Hz,1H),7.45(d,J=8Hz,1H),5.06(s,2H),4.34(s,3H),3.84(s,4H),3.49(s,3H),3.38(m,2H),2.10(s,2H),1.25(s,4H),1.04-0.96(m,1H),0.53(q,J=4Hz,2H),0.18(q,J=4Hz,2H).LC-MS(ESI)m/z:[M+H]+589.09, found 589.16.

Compound 23: 1, 7-dimethyl-8- (methylsulfonyl) -3- (3- (2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) prop-2-yn-1-yl) -3, 7-dihydro-1H-purine-2, 6-dione (244)

According to the method of 12, from 5-iodo-1, 3-dihydro-2H-benzo [ d]Imidazol-2-one (22mg,0.09mmol) was prepared 23 in 2% yield (7mg) as a yellow solid.1H NMR(400Hz,CDCl3)δ7.87(s,1H),7.71(m,1H),7.62(m,1H),5.34(m,2H),3.20(s,3H),3.76(s,6H).LC-MS(ESI)m/z:[M+H]+Calculated 429.09, found 429.17

Compound 24: 3- (3- (2-Aminopyrimidin-4-yl) prop-2-yn-1-yl) -1, 7-dimethyl-8- (methylsulfonyl) -3, 7-dihydro-1H-purine-2, 6-dione (245)

Prepared from 4-iodopyrimidin-2-amine 24 in 36% yield (16mg) as a yellow solid according to the procedure of 232.1H NMR(400Hz,DMSO)δ8.22(d,J=4.8Hz,1H),6.76(s,2H),6.1(d,J=4.2Hz,1H),5.01(s,2H),4.21(s,3H),3.54(s,3H),3.28(s,3H).LC-MS(ESI)m/z:[M+H]+Calculated 390.21, found 390.29

Compound 25:

N, N' - (5- (3- (1, 7-dimethyl-8- (methylsulfonyl) -2, 6-dioxo-1H-purin-3 (2H,6H,7H) -yl) prop-1-yn-1-yl) -1, 3-phenylene) diacetamide

And (1).

SnCl was added to a solution of 1-iodo-3, 5-dinitrobenzene (400mg,1.36mmol) in ethanol (10mL) at room temperature2.(H2O)2(1.84g,8.16 mmol). The mixture was then stirred at 70 ℃ for 2 hours. Completion of the reaction was monitored by UPLC-MS. The reaction mixture was poured into water and extracted with DCM. The organic layer was separated and the aqueous layer was repeatedly extracted with DCM (2X 20 mL). With Na2SO4The combined extracts were dried and concentrated. The resulting residue was purified by silica gel column chromatography (DCM: MeOH ═ 10:1) to give 160mg of 25-1 as a yellow solid, yield: 50.3 percent. LC-MS (ESI) M/z [ M + H ]]+calculated 234.97, found 235.02

And 2. step 2.

To a solution of 25-1(50mg,0.21mmol) in DCM (3mL) was added acetic anhydride (66mg,0.64mmol) and TEA (87mg,0.86mmol) at 0 deg.C. The mixture was then stirred at room temperature for 1 hour. Completion of the reaction was monitored by UPLC-MS. The reaction mixture was poured into water and extracted with DCM. The organic layer was separated and the aqueous layer was repeatedly extracted with DCM (2X 20 mL). With Na2SO4The combined extracts were dried and concentrated. The resulting residue was purified by silica gel column chromatography (DCM: MeOH ═ 40:1) to give 50mg of 25-2 as a yellow solid, yield: 73.5 percent. LC-MS (ESI) M/z [ M + H ]]+Calculated 318.99, found 319.03

And 3. step 3.

In N2To a solution of 1, 7-dimethyl-8- (methylsulfonyl) -3- (prop-2-yn-1-yl) -1H-purine-2, 6(3H,7H) -dione (25mg,0.084mmol) in DMF (2mL) under atmosphere was added 25-2(27mg,0.084mmol), TEA (21mg,0.202mmol), Pd (PPh)3)4(4.9mg,0.004mmol) and CuI (1.6mg,0.008 mmol). The mixture was then stirred at 60 ℃ for 4 hours. Completion of the reaction was monitored by UPLC-MS. The reaction mixture was poured into water and extracted with DCM. The organic layer was separated and the aqueous layer was repeatedly extracted with DCM (2X20 mL). With Na2SO4The combined extracts were dried and concentrated. The resulting residue was purified by silica gel column chromatography (PE: EA ═ 1:1) to give 8.5mg of 25 as a yellow solid in yield: 20.7 percent. 1H NMR (400MHz, DMSO). delta.10.00 (s,2H),7.76(s,1H),7.38(d, J ═ 4Hz,2H),4.98(s,2H),4.20(s,3H),3.53(s,3H),3.27(s,3H),2.01(s,6H). LC-MS (ESI) M/z: [ M + H: [ M ], [ M ] H]+Calculated 487.42, found 487.50

Compound 26: n, N' - (5- (3- (1, 7-dimethyl-8- (methylsulfonyl) -2, 6-dioxo-1H-purine-3 (2H,6H,7H) -yl) prop-1-yn-1-yl) -1, 3-phenylene) bis (2-morpholinylacetamide)

And (1).

At room temperature under N2To a solution of 2-morpholinoacetic acid (62mg,0.43mmol) in DMF (2mL) under an atmosphere were added 25-1(50mg,0.21mmol), EDCI (124mg,0.64mmol) and DMAP (79mg,0.64 mmol). The mixture was then stirred at 55 ℃ for 4 hours. Completion of the reaction was monitored by UPLC-MS. The reaction mixture was poured into water and extracted with DCM. The organic layer was separated and the aqueous layer was repeatedly extracted with DCM (2X 20 mL). With Na2SO4The combined extracts were dried and concentrated. The resulting residue was purified by silica gel column chromatography (PE: EA ═ 3:1) to give 21mg of 26-1 as a yellow solid in yield: 20.2 percent. LC-MS (ESI) M/z [ M + H ]]+Calculated 489.09, found 489.41

And 2. step 2.

In N21, 7-dimethyl-8- (methylsulfonyl) amine under atmosphereTo a solution of acyl) -3- (prop-2-yn-1-yl) -1H-purine-2, 6(3H,7H) -dione (16mg,0.053mmol) in DMF (2mL) was added 26-1(21mg,0.043mmol), TEA (11mg,0.10mmol), Pd (PPh)3)4(2.5mg,0.002mmol) and CuI (0.82mg,0.004 mmol). The mixture was then stirred at 55 ℃ for 4 hours. Completion of the reaction was monitored by UPLC-MS. The reaction mixture was poured into water and extracted with DCM. The organic layer was separated and the aqueous layer was repeatedly extracted with DCM (2X 20 mL). With Na2SO4The combined extracts were dried and concentrated. The resulting residue was purified by silica gel column chromatography (PE: EA ═ 1:1) to give 2.6mg of 26 as a yellow solid in yield: 9.2 percent. 1H NMR (400MHz, CDCl)3)δ9.07(s,2H),7.87(t,J=4Hz,1H),7.43(d,J=4Hz,2H),5.05(s,2H),4.34(s,3H),3.79(t,J=4Hz,8H),3.49(s,3H),3.45(s,3H),3.13(s,4H),2.61(m.8H).LC-MS(ESI)m/z:[M+H]+Calculated 657.61, found 657.71.

Compound 27: n, N' - (5- (3- (1, 7-dimethyl-8- (methylsulfonyl) -2, 6-dioxo-1, 2,6, 7-tetrahydro-3H-purin-3-yl) prop-1-yn-1-yl) -1, 3-phenylene) dicyclopropane-amide

And (1).

To a solution of 5-iodobenzene-1, 3-diamine (60mg,0.26mmol) in DCM (5mL) was added cyclopropanecarbonyl chloride (116. mu.L, 0.64mmol), Et3N (98. mu.L, 0.64mmol), and the mixture was then stirred at room temperature for 3 hours. Completion of the reaction was monitored by UPLC-MS. The solvent was evaporated under reduced pressure and further purified by silica gel column chromatography (PE/EA ═ 60/40) to give 40mg of 27-1 as a yellow solid (42%). LC-MS (ESI) M/z [ M + H ]]+Calculated 371.01, found 371.18.

Step 2 Sonogashira coupling

Prepared from 27-1(56mg,0.15mmol) according to the method of 12 in 18% yield (10mg) as a yellow solid.1H NMR(400Hz,CDCl3)δ7.91(s,1H),7.79(s,1H),7.29(s,1H),5.01(s,2H),4.33(s,3H),3.48(s,3H),3.41(s,3H),1.51(m,2H),1.03(m,4H),0.83(m,4H).LC-MS(ESI)m/z:[M+H]+Calculated 459.16, found 459.23.

Compound 28: n, N' - (5- (3- (8- ((cyclopropylmethyl) sulfonyl) -1, 7-dimethyl-2, 6-dioxo-1, 2,6, 7-tetrahydro-3H-purin-3-yl) prop-1-yn-1-yl) -1, 3-phenylene) bis (2-morpholinylacetamide)

Preparation 28 from N, N' - (5-iodo-1, 3-phenylene) bis (2-morpholinylacetamide) (48mg,0.11mmol) according to 22 in 10% yield (6.2mg) as a yellow solid.1H NMR(400Hz,CDCl3)δ7.89(s,1H),7.21(s,1H),5.06(s,2H),4.34(s,3H),3.81(m,8H),3.40(s,3H),3.21(s,4H),2.73(m,8H),3.41(m,2H),0.63(m,4H),0.35(m,4H).LC-MS(ESI)m/z:[M+H]+Calculated 697.27, found 697.35.

Compound 29: 3- (3- (3, 5-bis ((2-morpholinoethyl) amino) phenyl) prop-2-yn-1-yl) -1, 7-dimethyl-8- (methylsulfonyl) -3, 7-dihydro-1H-purine-2, 6-dione

Prepared from 5-iodo-N1, N3-bis (2-morpholinoethyl) benzene-1, 3-diamine (66mg,0.15mmol) according to method 12 in 10% yield (6.1mg) as a yellow solid.1H NMR(400Hz,CDCl3)δ6.25(s,2H),6.17(s,1H),5.03(s,2H),4.33(s,3H),3.63(s,3H),3.48(s,3H),3.45(s,3H),3.40(s,3H),3.70-3.67(m,4H),3.63-3.62(m,4H),3.56-3.54(m,8H).LC-MS(ESI)m/z:[M+H]+Calculated 607.25, found 607.34.

Compound 30: n, N' - (5- (3- (8- ((cyclopropylmethyl) sulfonyl) -1, 7-dimethyl-2, 6-dioxo-1, 2,6, 7-tetrahydro-3H-purin-3-yl) prop-1-yn-1-yl) -1, 3-phenylene) dicyclopropane-amide

Prepared 30 according to the procedure of 12 in 10% yield (5.0mg) as a yellow solid.1H NMR(400Hz,CDCl3)δ7.82(s,1H),7.61(s,1H),7.28(s,1H),5.03(s,2H),4.34(s,3H),3.41(s,3H),3.48-3.37(m,2H),1.21(m,2H),1.01(m,3H),0.82(m,2H),0.63(m,4H),0.34(m,4H).LC-MS(ESI)m/z:[M+H]+Calculated 579.19, found 579.24.

Compound 31: n- (4- (3- (8- ((cyclopropylmethyl) sulfonyl) -1, 7-dimethyl-2, 6-dioxo-1, 2,6, 7-tetrahydro-3H-purin-3-yl) prop-1-yn-1-yl) pyridin-2-yl) cyclopropanecarboxamide

Preparation of 31 according to the procedure of 12 in 27% yield (10mg) as a yellow solid.1H NMR(400Hz,CDCl3)δ8.25(s,1H),8.12(s,1H),7.0(d,d,J=4.8Hz,1H),5.08(s,2H),4.37(s,3H),3.45(s,3H),3.46-3.38(m,2H),1.7(m,1H),1.42(m,1H),1.08(m,2H),0.92(m,2H),0.65(m,2H),0.35(m,2H).LC-MS(ESI)m/z:[M+H]+Calculated 497.15, found 497.23.

Compound 32: n- (4- (3- (8- ((cyclopropylmethyl) sulfonyl) -1, 7-dimethyl-2, 6-dioxo-1, 2,6, 7-tetrahydro-3H-purin-3-yl) prop-1-yn-1-yl) pyridin-2-yl) -2-morpholinylacetamide

Preparation of 32 according to the procedure of 12 in 15% yield (6mg) as a yellow solid.1H NMR(400Hz,CDCl3)δ8.22(m,2H),7.02(d,J=4.8Hz,1H),5.09(s,2H),4.37(s,3H),3.83(m,4H),3.41(m,5H),3.38(m,2H)2.69(m,4H),0.83(m,1H),0.63(m,2H),0.34(m,2H).LC-MS(ESI)m/z:[M+H]+Calculated 556.19, found 556.26.

Compound 33: 8- ((cyclopropylmethyl) sulfonyl) -3- (3- (2- (ethylamino) pyridin-4-yl) prop-2-yn-1-yl) -1, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione

Stirring of 8- ((cyclopropylmethyl) sulfonyl) -1, 7-dimethyl at room temperature under an argon atmosphere3- (prop-2-yn-1-yl) -1H-purine-2, 6(3H,7H) -dione (25mg,75nmol), N-Ethyl-4-iodopyridin-2-amine (28mg,0.11mmol), PdCl2(PPh3)2(5.3mg,7.5nmol), copper (I) iodide (0.7mg,3.75nmol) in DMF (3mL) for 5 min. Triethylamine (0.25ml,0.75mmol) was added, the reaction was sealed and stirred overnight. The reaction mixture was filtered, the filtrate was washed with water and extracted with DCM (3 × 5 mL). The combined organic layers were washed with brine and dried over MgSO4, filtered and concentrated in vacuo. The residue was purified by silica gel column chromatography with 100:1DCM/MeOH to give 33 in 66% yield as a pale yellow solid.1H NMR:(400Mz,CDCl3):δ7.89(d,J=5.6Hz,1H),6.51(d,J=5.6Hz,1H),6.42(s,1H),5.08(s,2H),4.36(s,3H),3.45(d,J=7.2Hz,2H),3.45(s,3H),3.23(q,J=7.2Hz,2H),1.26(t,J=7.2Hz,3H),0.88(m,1H),0.65(m,2H),0.31(m,2H).LC-MS(ESI)m/z:[M+H]+Calculated 457.16, found 457.25.

Compound 34: 3- (3- (2- ((cyclopropylmethyl) amino) pyridin-4-yl) prop-2-yn-1-yl) -8- ((cyclopropylmethyl) sulfonyl) -1, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione

Prepared according to the method of 12 from 8- ((cyclopropylmethyl) sulfonyl) -1, 7-dimethyl-3- (prop-2-yn-1-yl) -1H-purine-2, 6(3H,7H) -dione (25mg,75nmol) and N- (cyclopropylmethyl) -4-iodopyridin-2-amine (30mg,0.11mmol) 34 in 63% yield (36mg) as a pale yellow solid.1H NMR:(400Mz,CDCl3):δ7.87(d,J=5.6Hz,1H),6.51(d,J=5.6Hz,1H),6.46(s,1H),5.08(s,2H),4.36(s,3H),3.45(d,J=7.2Hz,2H),3.45(s,3H),3.06(d,J=6.8Hz,2H),0.90(m,2H),0.64(m,2H),0.57(m,2H),0.30(m,2H),0.26(m,2H).LC-MS(ESI)m/z:[M+H]+Calculated 483.17, found 483.25.

Compound 35: 8- ((cyclopropylmethyl) sulfonyl) -3- (3- (2- (isopropylamino) pyridin-4-yl) prop-2-yn-1-yl) -1, 7-dimethyl-1H-purine-2, 6(3H,7H) -dione

Prepared according to the method of 12 from 8- ((cyclopropylmethyl) sulfonyl) -1, 7-dimethyl-3- (prop-2-yn-1-yl) -1H-purine-2, 6(3H,7H) -dione (25mg,75nmol) and 4-iodo-N-isopropylpyridin-2-amine (29mg,0.11mmol) as 35 in 30% yield (10.5mg) as a pale yellow solid.1H NMR:(400Mz,CDCl3):δ7.90(d,J=5.6Hz,1H),6.50(d,J=5.6Hz,1H),6.41(s,1H),5.08(s,2H),4.37(s,3H),3.84-3.67(m,1H),3.45(d,J=7.2Hz,2H),3.45(s,3H),1.24(s,3H),1.22(s,3H),0.87(m,1H),0.64(m,2H),0.31(m,2H).LC-MS(ESI)m/z:[M+H]+Calculated 471.17, found 471.24.

Compound 36: n- (3- (3- (1, 7-dimethyl-8- (methylsulfonyl) -2, 6-dioxo-1, 2,6, 7-tetrahydro-3H-purin-3-yl) prop-1-yn-1-yl) -5- (2-morpholinylacetamido) phenyl) cyclopropanecarboxamide

And (1).

At room temperature under N2to a solution of cyclopropanecarboxylic acid (85mg,1.0mmol) in DMF (8mL) was added 5-iodobenzene-1, 3-diamine (234mg,1.0mmol), EDCI (192mg,1.0mmol) and DMAP (122mg,1.0mmol) under atmosphere. The mixture was then stirred at room temperature for 5 hours. Completion of the reaction was monitored by UPLC-MS. The reaction mixture was poured into water and extracted with DCM. The organic layer was separated and the aqueous layer was repeatedly extracted with DCM (2 × 20 mL). With Na2SO4The combined extracts were dried and concentrated. The crude product 36-1 was used in the next reaction step without any further purification.

And 2. step 2.

36-2 was prepared according to the procedure of 36-1 in 11% yield (47mg in two steps) as a yellow solid.

And 3. step 3.

Preparation of 36 according to method 12 in 12% (6mg) as a yellow solid.1H NMR(400Hz,CDCl3)δ7.7(s,1H),7.40(s,1H),7.36(s,1H),5.03(s,2H),4.33(s,3H),3.81(s,3H),3.43(s,3H),3.41(s,2H),3.17(m,4H),2.6(m,4H),1.52(m,1H)1.03(m,2H),0.85(m,2H).LC-MS(ESI)m/z:[M+H]+CalculatingValue 598.20, found 598.27.

Compound 37: 3- (3- (3, 5-bis ((cyclopropylmethyl) amino) phenyl) prop-2-yn-1-yl) -1, 7-dimethyl-8- (methylsulfonyl) -3, 7-dihydro-1H-purine-2, 6-dione

Prepared 37 according to the procedure of 12 in 12% yield (6.2mg) as a yellow solid.1H NMR(400Hz,CDCl3)δ6.14(s,2H),5.94(s,1H),5.03(s,2H),4.32(s,3H),3.47(s,3H),3.43(s,3H),2.90(d,J=7.2Hz,4H),1.03(m,2H),0.52(m,4H),0.23(m,4H).LC-MS(ESI)m/z:[M+H]+Calculated 511.20, found 511.27.

Compound 38: n- (3- (3- (1, 7-dimethyl-8- (methylsulfonyl) -2, 6-dioxo-1, 2,6, 7-tetrahydro-3H-purin-3-yl) prop-1-yn-1-yl) -5- (2-methoxyacetamido) phenyl) cyclopropanecarboxamide

and (1).

38-1 was prepared from 36-1(80mg,0.27) in 38% yield (38mg) as a yellow solid according to the method of 36.

And 2. step 2.

Preparation of 38 according to the procedure of 12 in 30% yield (13mg) as a yellow solid.1H NMR(400Hz,CDCl3)δ7.83(s,1H),7.40(s,1H),7.37(s,1H),5.03(s,2H),4.33(s,3H),3.96(s,2H),3.49(s,3H),3.47(s,3H),3.41(s,3H),1.21(m,1H),1.04(m,2H),0.85(m,2H).LC-MS(ESI)m/z:[M+H]+Calculated 543.16, found 543.25.

Compound 39: n- (3- (3- (1, 7-dimethyl-8- (methylsulfonyl) -2, 6-dioxo-1, 2,6, 7-tetrahydro-3H-purin-3-yl) prop-1-yn-1-yl) -5- (2- (dimethylamino) acetamido) phenyl) cyclopropanecarboxamide

And (1).

39-1 was prepared from 36-1(80mg,0.27) in 39% yield (40mg) as a yellow solid according to the method of 36.

And 2. step 2.

Preparation of 39 according to the procedure of 12 in 9% yield (5mg) as a yellow solid.1H NMR(400Hz,CDCl3)δ7.83(s,1H),7.43(s,1H),7.37(s,1H),5.03(s,2H),4.34(s,3H),3.49(s,3H),3.47(s,3H),3.11(s,2H),2.38(s,6H).LC-MS(ESI)m/z:[M+H]+Calculated 556.19, found 556.28.

Compound 40: n- (3-amino-5- (3- (1, 7-dimethyl-8- (methylsulfonyl) -2, 6-dioxo-1H-purin-3 (2H,6H,7H) -yl) prop-1-yn-1-yl) phenyl) cyclopropanecarboxamide

Prepared according to the method of 12 from 1, 7-dimethyl-8- (methylsulfonyl) -3- (prop-2-yn-1-yl) -1H-purine-2, 6(3H,7H) -dione (39.5mg,0.13mmol) and N- (3-amino-5-iodophenyl) cyclopropanecarboxamide (60.4mg,0.2mmol) in 46% yield (28.6mg) as a white solid.1H NMR(400Mz,CDCl3):7.08(s,1H),6.79(s,1H),6.45(s,1H),5.03(s,2H),4.33(s,3H),3.48(s,3H),3.44(s,3H),1.04(m,1H),0.86(m,4H).LC-MS(ESI)m/z:[M+H]+calculated 471.14, found 471.23.

Compound 41: n- (3- (3- (1, 7-dimethyl-8- (methylsulfonyl) -2, 6-dioxo-1, 2,6, 7-tetrahydro-3H-purin-3-yl) prop-1-yn-1-yl) -5- (2-methoxyethoxy) phenyl) cyclopropanecarboxamide

Preparation of 41 according to the method of 12 gave a yield of 13% (5.8 mg).1H NMR(400Hz,CDCl3)δ7.20(s,1H),7.15(s,1H),6.64(s,1H),5.01(s,2H),4.31(s,3H),4.03(m,2H),3.67(m,1H),3.46(s,3H),3.42(s,3H),3.40(s,3H),1.51(m,1H),1.00(m,2H),0.79(m,2H).LC-MS(ESI)m/z:[M+H]+Calculated 530.16, found 530.22.

Compound 42: n- (3- (3- (1, 7-dimethyl-8- (methylsulfonyl) -2, 6-dioxo-1, 2,6, 7-tetrahydro-3H-purin-3-yl) prop-1-yn-1-yl) -5- (2-methoxyethoxy) phenyl) cyclopropanecarboxamide

42 was prepared according to the procedure of 12 in 12% yield (5.5 mg).1H NMR(400Hz,CDCl3)δ7.32(s,1H),7.23(s,1H),6.45(s,1H),5.30(s,2H),4.33(s,3H),4.31-4.28(m,2H),3.46(s,3H),3.45(s,3H),2.84(s,6H),1.1(m,1H),0.98(m,2H),0.81(m,2H).LC-MS(ESI)m/z:[M+H]+Calculated 543.19, found 543.25.

Compound 43: n- (4- (3- (1, 7-dimethyl-8- (methylsulfonyl) -2, 6-dioxo-1, 2,6, 7-tetrahydro-3H-purin-3-yl) prop-1-yn-1-yl) -6- (trifluoromethyl) pyridin-2-yl) cyclopropanecarboxamide

Prepared 43 according to the procedure of 12 in 40% yield (16 mg).1H NMR(400Hz,DMSO)δ8.68(s,1H),8.24(s,1H),5.09(s,2H),4.21(s,3H),3.52(s,3H),3.28(s,3H),1.41(m,1H),0.85(m,4H).LC-MS(ESI)m/z:[M+H]+Calculated 525.11, found 525.19.

Compound 44: n- (3- (3- (1, 7-dimethyl-8- (methylsulfonyl) -2, 6-dioxo-1, 2,6, 7-tetrahydro-3H-purin-3-yl) prop-1-yn-1-yl) -5- (trifluoromethyl) phenyl) cyclopropanecarboxamide

Preparation of 44 according to the procedure of 12 in 36% yield (15.9 mg).1H NMR(400Hz,DMSO)δ7.99(s,1H),7.89(s,1H),7.39(s,1H),5.03(s,2H),4.21(s,3H),3.32(s,6H),3.43(s,3H),3.23(s,3H),1.2(m,1H),0.82(m,4H).LC-MS(ESI)m/z:[M+H]+Calculated 524.11, found 524.19

Compound 45: n- (4- (3- (8- ((cyclopropylmethyl) sulfonyl) -1, 7-dimethyl-2, 6-dioxo-1, 2,6, 7-tetrahydro-3H-purin-3-yl) prop-1-yn-1-yl) -6- (trifluoromethyl) pyridin-2-yl) cyclopropanecarboxamide

Preparation of 45 according to the procedure of 12 in 32% yield (18 mg).1H NMR(400Hz,CDCl3)δ8.68(s,1H),8.23(s,1H),7.39(s,1H),5.09(s,2H),4.23(s,3H),3.55(d,J=7.2Hz,2H),3.28(s,3H),1.2(m,2H),0.83(m,4H),0.49(m,4H),LC-MS(ESI)m/z:[M+H]+Calculated 565.14, found 565.21

Compound 46: n- (3- (3- (8- ((cyclopropylmethyl) sulfonyl) -1, 7-dimethyl-2, 6-dioxo-1, 2,6, 7-tetrahydro-3H-purin-3-yl) prop-1-yn-1-yl) -5- (trifluoromethyl) phenyl) cyclopropanecarboxamide

Preparation of 46 according to the procedure of 12 in 22% yield (12.5 mg).1H NMR(400Hz,CDCl3)δ7.79(s,1H),7.76(s,1H),7.32(s,1H),5.06(s,2H),4.37(s,3H),3.46(s,3H),3.45(m,2H),1.27(m,2H),1.08(m,2H),0.88(m,2H),0.65(m,2H),0.33(m,2H).LC-MS(ESI)m/z:[M+H]+Calculated 564.14, found 564.23

compound 47: n, N' - (5- (3- (1, 7-dimethyl-8- (methylsulfinyl) -2, 6-dioxo-1, 2,6, 7-tetrahydro-3H-purin-3-yl) prop-1-yn-1-yl) -1, 3-phenylene) dicyclopropane-amide

47 was prepared according to the procedure of 12 in 11% yield (5.3 mg).1H NMR(400Hz,CDCl3)δ7.83(s,1H),7.37(s,2H),5.00(s,2H),4.12(s,3H),3.27(s,3H),3.14(s,3H),1.73(m,2H),0.78(m,8H).LC-MS(ESI)m/z:[M+H]+Calculated 523.17, found 523.22

Compound 48: n- (4- (3- (1, 7-dimethyl-8- (methylsulfinyl) -2, 6-dioxo-1, 2,6, 7-tetrahydro-3H-purin-3-yl) prop-1-yn-1-yl) -6- (trifluoromethyl) pyridin-2-yl) cyclopropanecarboxamide

Preparation of 48 according to method 12 in 34% (17mg) yield.1H NMR(400Hz,DMSO)δ8.68(s,1H),8.23(s,1H),5.11(s,2H),4.12(s,3H),3.26(s,3H),3.11(s,3H),1.23(m,1H),1.12(m,2H),0.85(m,2H).LC-MS(ESI)m/z:[M+H]+Calculated 509.11, found 509.20

Compound 49:

N- (3- (3- (1, 7-dimethyl-8- (methylsulfinyl) -2, 6-dioxo-1, 2,6, 7-tetrahydro-3H-purin-3-yl) prop-1-yn-1-yl) -5- (trifluoromethyl) phenyl) cyclopropanecarboxamide (272)

49 was prepared according to the procedure of 12 in 41% yield (21 mg).1H NMR(400Hz,CDCl3)δ7.99(s,1H),7.88(s,1H),7.38(s,1H),5.04(s,2H),4.12(s,3H),3.28(s,3H),3.10(s,3H),1.77(m,1H),1.17(m,2H),0.84(m,2H).LC-MS(ESI)m/z:[M+H]+Calculated 508.12, found 508.21

Compound 50 and compound 51:

N, N '- (5- (3- (8- (ethylsulfonyl) -1, 7-dimethyl-2, 6-dioxo-1H-purin-3 (2H,6H,7H) -yl) prop-1-yn-1-yl) -1, 3-phenylene) bicyclopropane amide and N, N' - (5- (3- (8- (ethylsulfinyl) -1, 7-dimethyl-2, 6-dioxo-1H-purin-3 (2H,6H,7H) -yl) prop-1-yn-1-yl) -1, 3-phenylene) bicyclopropane amide

And (1).

To a solution of 27 in anhydrous DMF (0.5mL) at 0 deg.C was added NaHS (6.7mg,0.12mmol) and the temperature was allowed to rise to room temperature. The mixture was stirred at room temperature for 3 hours. The mixture was then acidified to pH 3 and extracted with dichloromethane (3 × 10mL), the organic layer was separated, washed with brine, and washed with Na2SO4Dry, filter, concentrate and purify by silica gel chromatography (PE: EA ═ 1:4) to give 77mg of 50-1 as a white solid (67%). LC-MS (ESI) M/z [ M + H ]]+Calculated 493.16, found 493.23

and 2. step 2.

To the mixture of 50-1(20.0mg,0.04mmol) and K2CO3To a solution of (16.8mg,0.12mmol) in anhydrous DMF (1mL) was added iodoethane (0.01mL,0.12mmol) and stirred at room temperature under nitrogen for 2 h. The reaction mixture was then poured into water and extracted with dichloromethane (3x 5mL), the organic layer was separated, washed with brine, washed with Na2SO4Drying, filtration and concentration gave crude product 50-2. The crude product was used directly without further purification. LC-MS (ESI) M/z [ M + H ]]+Calculated 521.19, found 521.25

And 3. step 3.

To a solution of the product of 50-2 in DCM (1mL) at 0 deg.C was added m-CPBA (13.8mg,0.06 mmol). The mixture was then stirred at room temperature for 2 hours. The solvent was then removed and extracted with dichloromethane (3x 5mL), the organic layer separated and washed with Na2SO4Dry, filter, concentrate and purify by prep TLC to give 50(4.6mg) as a white solid (two steps 21%).1H NMR(400Mz,CDCl3):δ7.82(s,1H),7.33(s,2H),5.03(s,2H),4.35(s,3H),3.61(q,J=7.2Hz,2H),3.44(s,3H),1.47(t,J=7.2Hz,3H),1.04(m,2H),0.86(m,8H).LC-MS(ESI)m/z:[M+H]+Calculated 553.18, found 553.24

Compound 51 was obtained as a white solid (13% in two steps).1H NMR(400Mz,CDCl3):δ7.82(s,1H),7.33(s,2H),5.03(s,2H),4.30(s,3H),3.61(q,J=7.2Hz,2H),3.44(s,3H),1.47(t,J=7.2Hz,3H),1.04(m,2H),0.88(m,8H).LC-MS(ESI)m/z:[M+H]+Calculated 537.18, found 537.25

Compound 52: n, N' - (5- (3- (1, 7-dimethyl-2, 6-dioxo-8- (benzenesulfonyl) -1H-purin-3 (2H,6H,7H) -yl) prop-1-yn-1-yl) -1, 3-phenylene) bicyclopropane carboxamide

52 (white solid, 5.7mg, two step yield 23%) was prepared from N, N' - (5- (3- (8-mercapto-1, 7-dimethyl-2, 6-dioxo-1H-purin-3 (2H,6H,7H) -yl) prop-1-yn-1-yl) -1, 3-phenylene) dicyclopropane amide according to a similar procedure outlined for 50.1H NMR(400Mz,CDCl3):δ8.02(s,2H),7.81(s,1H),7.27-7.41(m,5H),5.03(s,2H),4.35(s,3H),3.44(s,3H),1.04(m,2H),0.86(m,8H).LC-MS(ESI)m/z:[M+H]+Calculated 601.18, found 601.25.

Compound 53: n, N' - (5- (3- (1, 7-dimethyl-2, 6-dioxo-8- (propylsulfonyl) -1H-purin-3 (2H,6H,7H) -yl) prop-1-yn-1-yl) -1, 3-phenylene) bicyclopropane carboxamide

Prepared 53 from 50-1 (white solid, 4.7mg, two step yield 21%) according to similar procedure outlined for 50.1H NMR(400Mz,CDCl3):δ7.84(s,1H),7.33(s,2H),5.04(s,2H),4.35(s,3H),3.55(m,2H),3.44(s,3H),1.64(m,2H),1.04(m,2H),0.86(m,11H).LC-MS(ESI)m/z:[M+H]+Calculated 567.19, found 567.25.

compound 54:

N- (3- (3- (1, 7-dimethyl-8- (methylsulfinyl) -2, 6-dioxo-1, 2,6, 7-tetrahydro-3H-purin-3-yl) prop-1-yn-1-yl) -5- (2-methoxyethoxy) phenyl) cyclopropanecarboxamide

54 was prepared according to the procedure of 12 in 10% yield (3.0 mg).1H NMR(400Hz,CDCl3)δ7.26(s,1H),7.21(s,1H),6.23(s,1H),5.01(s,2H),4.12(s,3H),4.03(t,J=4.8Hz,2H),3.60(t,J=4.8Hz,2H),3.30(s,3H),3.27(s,3H),3.14(s,3H),1.77(m,1H),1.15(m,2H),0.86(m,2H).LC-MS(ESI)m/z:[M+H]+Calculated 514.17, found 514.24.

Compound 55: n-benzyl-1, 3, 7-trimethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purine-8-sulfonamide

8-mercapto-1, 3, 7-trimethyl-1H-purine-2, 6(3H,7H) -dione (50mg,0.22mmol) was stirred in a mixture of 2mL DCM and 2mL 1M HCl for 10min at-10 to-5 ℃ in a flask. Cold (5 ℃) sodium hypochlorite (6% solution, 0.68M,1mL,0.72mmol) was added dropwise with very rapid stirring, maintaining the temperature at-10 to-5 ℃. After the addition was complete, the mixture was stirred at-10 to-5 ℃ for 15 min. The mixture was transferred to a separatory funnel (precooled with ice water) and CH was rapidly separated2Cl2Layers were collected in a clean flask cooled with a dry ice-acetone bath. Benzylamine (1.4mL,12.5mmol) followed by CH was added with stirring2Cl2The layer turned into a white suspension. The flask was transferred to an ice-water bath and the suspension was stirred at 0 ℃ for 30 min. The suspension was then washed with 1M phosphoric acid (all solids dissolved at once) and then with water and brine. Drying (Na)2SO4) And further purified by silica gel column chromatography (DCM/MeOH ═ 200/1) to give 7.9mg 55 as a yellow solid (10%).1H NMR(400Hz,DMSO-d6)δ7.26-7.17(m,5H),5.76(s,2H),4.05(s,3H),3.40(s,3H),3.23(s,3H).LC-MS(ESI)m/z:[M+H]+Calculated 364.10, found 364.19

Compound 56: n,1,3, 7-tetramethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purine-8-sulfonamide

Prepared 56 according to the method of 55 as a yellow solid.1H NMR(400Hz,CDCl3)δ4.26(s,3H),3.56(s,3H),3.41(s,3H),2.98(s,3H).LC-MS(ESI)m/z:[M+H]+Calculated 288.07, found 288.12

Compound 57:

N-ethyl-1, 3, 7-trimethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purine-8-sulfonamide

57 was prepared according to the method of 55 as a yellow solid.1H NMR(400Hz,CDCl3)δ4.25(s,3H),3.56(s,3H),3.40(s,3H),3.33(m,2H),1.27(m,3H).LC-MS(ESI)m/z:[M+H]+Calculated 302.07, found 302.14

Compound 58: 1,3, 7-trimethyl-2, 6-dioxo-N-propyl-2, 3,6, 7-tetrahydro-1H-purine-8-sulfonamide

Prepared 58 according to the method of 55 as a yellow solid.1H NMR(400Hz,CDCl3)δ4.21(s,3H),3.53(s,3H),3.31(s,3H),3.14(t,J=6.8Hz,2H),1.57(m,2H),0.94(t,J=6.4Hz,3H).LC-MS(ESI)m/z:[M+H]+Calculated 316.10, found 316.17

Compound 59: n, N,1,3, 7-pentamethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purine-8-sulfonamide

59 was prepared according to the method of 55 as a yellow solid.1H NMR(400Hz,CDCl3)δ4.24(s,3H),3.56(s,3H),3.42(s,3H),3.23(s,3H),2.92(s,3H).LC-MS(ESI)m/z:[M+H]+Calculated 302.08, found 302.14

Compound 60: 1,3, 7-trimethyl-8- (pyrrolidin-1-ylsulfonyl) -3, 7-dihydro-1H-purine-2, 6-dione

Prepared 60 according to the method of 55 as a yellow solid.1H NMR(400Hz,CDCl3)δ4.24(s,3H),3.59(m,2H),3.54(s,3H),3.42(s,3H),2.05(m,2H).LC-MS(ESI)m/z:[M+H]+Calculated 328.10, found 328.18

Compound 61:1,3, 7-trimethyl-2, 6-dioxo-N- (2,2, 2-trifluoroethyl) -2,3,6, 7-tetrahydro-1H-purine-8-sulfonamide

Prepared 61 according to the method of 55 as a yellow solid.1H NMR(400Hz,CDCl3)δ4.31(s,3H),3.49(s,3H),3.40(s,3H),3.3(m,2H).LC-MS(ESI)m/z:[M+H]+Calculated 356.06, found 356.12

Compound 62:

1, 3-dimethyl-6, 7-dihydrothiazolo [2,3-f ] purine-2, 4(1H,3H) -dione 8, 8-dioxide

And (1).

To a solution of 8-chloro-1, 3-dimethyl-3, 7-dihydro-1H-purine-2, 6-dione (456mg,2.0mmol) in DMF (6mL) was added NaSH (170mg,3.0 mmol). The mixture was then stirred by microwave irradiation at 110 ℃ for 2 hours. Completion of the reaction was monitored by UPLC-MS. The solvent was evaporated under reduced pressure and further purified by silica gel column chromatography (DCM/MeOH ═ 100/1) to give 404mg of 62-1 as a yellow solid (88%). LC-MS (ESI) M/z [ M + H ]]+Calculated 213.04, found 213.12

And 2. step 2.

To a solution of 62-1(80mg,0.38mmol) in DMF (2mL) was added 1, 2-dibromoethane (204. mu.L, 2.4mmol) and K2CO3(130mg,0.95 mmol). The mixture was heated to 110 ℃ by microwave irradiation for 1.5 h. The solvent was evaporated under reduced pressure and further purified by silica gel column chromatography (DCM/MeOH. RTM. 200/1) to give 53mg of 62-2 as a yellow solid (59%), LC-MS (ESI) M/z: [ M + H ]]+Calculated 239.05, found 239.14

And 3. step 3.

Preparation of 62 from 62-2(50mg,0.21mmol) and oxone (193mg,0.32mmol) according to the method of 1 in 18% yield (10.0mg) as a white solid.1H NMR:(400Hz,DMSO-d6)δ4.79(t,J=6.4Hz,2H),4.28(t,J=6.4Hz,2H),3.44(s,3H),3.24(s,3H).LC-MS(ESI)m/z:[M+H]+calculated 271.04, found 271.13

Compound 63: 1, 3-dimethyl-7, 8-dihydro-6H- [1,3] thiazino [2,3-f ] purine-2, 4(1H,3H) -dione 9, 9-dioxide

And (1).

Prepared from 8-mercapto-1, 3-dimethyl-1H-purine-2, 6(3H,7H) -dione (60mg,0.28mmol) and 1, 3-dibromopropane (142. mu.L, 1.4mmol) according to method 62 in 90% yield (10.0 mg). LC-MS (ESI) M/z [ M + H ]]+Calculated 253.07, found 253.13

And 2. step 2.

Prepared from 63-1(65mg,0.26mmol) and oxone (475mg,0.77mmol) according to the method of 1 to 63 in 8% yield (6.0mg) as a yellow solid.1H NMR:(400Hz,CDCl3)δ4.60(t,J=5.6Hz,2H),3.45(m,2H),3.61(s,3H),3.19(s,3H),2.81(m,2H).LC-MS(ESI)m/z:[M+H]+Calculated 285.06, found 285.13

Compound 64: 3-methyl-1- (prop-2-yn-1-yl) -6, 7-dihydrothiazolo [2,3-f ] purine-2, 4(1H,3H) -dione 8, 8-dioxide

And (1).

To a solution of cyanocarboiminodithiodimethyl ester (2.0g) in EtOH (60mL) was added 2-aminoethane-1-thiol (1.06 g). The mixture was then stirred at 90 ℃ for 2 hours. Completion of the reaction was monitored by UPLC-MS. The solvent was evaporated under reduced pressure and further purified by silica gel column chromatography (DCM/MeOH ═ 100/1) to give 1.7g of 64-1 as a yellow solid (100%). LC-MS (ESI) M/z [ M + H ]]+Calculated 128.02, found 128.11

And 2. step 2.

to a solution of 64-1(500mg) in DMF (8mL) was added ethyl 2-chloroacetate (517mg) and NaH. The mixture was stirred at room temperature for 5 hours. The solvent was evaporated under reduced pressure and further purified by silica gel column chromatography (DCM/MeOH. RTM. 200/1) to give 392mg of 64-2 as a yellow solid (50%), LC-MS (ESI) M/z: [ M + H ]]+Calculated 214.06, found 214.12

And 3. step 3.

EtONa was added to a solution of 64-2(200mg) in EtOH (6 mL). The mixture was then stirred at 90 ℃ for 2 hours. Completion of the reaction was monitored by UPLC-MS. The solvent was evaporated under reduced pressure and further purified by silica gel column chromatography (DCM/MeOH ═ 100/1) to give 108mg of 64-3 as a yellow solid (53%). LC-MS (ESI) M/z [ M + H ]]+Calculated 214.06, found 214.12

And 4. step 4.

To a solution of 64-3(1.49g) in THF (60mL) were added ethyl methylcarbamate (1.73g) and t-BuOK (2.35 g). The mixture was then stirred at 50 ℃ for 5 hours. Completion of the reaction was monitored by UPLC-MS. The solvent was evaporated under reduced pressure and further purified by silica gel column chromatography (DCM/MeOH ═ 100/1) to give 625mg 64-4 as a yellow solid (40%). LC-MS (ESI) M/z [ M + H ]]+Calculated 225.04, found 225.13

And 5. step 5.

To a solution of 64-4(500mg) in DMF (5mL) was added propargyl bromide (336mg) and K2CO3(615 mg). The mixture was stirred at room temperature for 3 h. The solvent was evaporated under reduced pressure and further purified by silica gel column chromatography (DCM/MeOH ═ 200/1) to give 3438mg 64-5 as a yellow solid (75%), LC-ms (esi) M/z: [ M + H ]]+Calculated 263.05, found 263.12

And 6. step 6.

preparation of 64 from 64-5(10mg,0.038mmol) and oxone (95mg,0.15mmol) according to the method of 1 gave 64 in a yield of 30% (3.3mg) as a yellow solid.1H NMR(400Hz,CDCl3)δ4.89(d,J=0.8Hz,2H),4.85(m,2H),4.03(m,2H),3.48(s,3H),2.28(t,J=1.6Hz,1H).LC-MS(ESI)m/z:[M+H]+Calculated 295.04, found 295.11

Compound 65: 1- (3- (3-hydroxyphenyl) prop-2-yn-1-yl) -3-methyl-6, 7-dihydrothiazolo [2,3-f ] purine-2, 4(1H,3H) -dione 8-oxide and compound 66: 1- (3- (3-hydroxyphenyl) prop-2-yn-1-yl) -3-methyl-6, 7-dihydrothiazolo [2,3-f ] purine-2, 4(1H,3H) -dione 8, 8-dioxide

Step 1 Sonogashira coupling

To 3-methyl-1- (prop-2-yn-1-yl) -6, 7-dihydrothiazolo [2,3-f]To a solution of purine-2, 4(1H,3H) -dione (75mg,0.29mmol) in DMF (5mL) was added 3-iodophenol (143mg,0.43mmol), Pd (PPh)3)2(20mg,0.028mmol)、CuI(5.3mg,0.028mmol)、Et3N (60. mu.L, 0.43 mmol). The mixture was then degassed for 15min and then stirred at 55 ℃ for 2.5 h. Completion of the reaction was monitored by UPLC-MS. The solvent was evaporated under reduced pressure and further purified by silica gel column chromatography (DCM/MeOH ═ 100/1) to give 42.6mg of 65-1 as a yellow solid (42%). LC-MS (ESI) M/z [ M + H ]]+Calculated 355.08, found 355.16

And 2. step 2.

Preparation of 65 from 65-1(8mg,0.023mmol) and oxone (41mg,0.068mmol) according to the method of 1 in 43% yield (3.6mg) as a yellow solid.1H NMR(400Hz,CDCl3)δ7.12(t,J=7.6Hz,1H),6.95(d,J=7.6Hz,1H),8.84(m,1H),6.87(t,J=2.4Hz,1H),6.78(m,1H),5.1(s,2H),4.97(m,1H),4.84(m,1H),3.84(m,2H),3.45(s,3H).LC-MS(ESI)m/z:[M+H]+Calculated 371.07, found 371.13

Prepared according to the method of 1 from 65-1(8mg,0.023mmol) and oxone (41mg,0.068mmol) as 66 in 36% yield (3.0mg) as a yellow solid.1H NMR(400Hz,DMSO)δ7.15(t,J=8.0Hz,1H),6.82-6.75(m,4H),5.0(s,2H),4.81(t,J=6.4Hz,2H),4.29(t,J=6.4Hz,2H),3.28(s,3H).LC-MS(ESI)m/z:[M+H]+Calculated 387.07, found 387.14

Compound 67: 3-methyl-1- (3- (3- ((2-morpholinoethyl) amino) phenyl) prop-2-yn-1-yl) -6, 7-dihydrothiazolo [2,3-f ] purine-2, 4(1H,3H) -dione 8, 8-dioxide

Preparation of 67 from 3-iodo-N- (2-morpholinoethyl) aniline (41mg,0.12mmol) according to method 12 in 24% yield (11mg) as a yellow solid.1H NMR(400Hz,CDCl3)δ7.05(m,1H),6.75(d,J=7.6Hz,1H),6.65(s,1H),6.55(m,1H),5.08(s,2H),4.84(t,J=6.0Hz,2H),4.01(t,J=6.0Hz,2H),3.75(t,J=7.6Hz,4H)3.43(s,3H),3.17(t,J=6.0Hz,2H),2.67(t,J=6.0Hz,2H),2.54(m,4H).LC-MS(ESI)m/z:[M+H]+Calculated 499.17, found 499.23

Compound 68:

N, N' - (5- (3- (1, 7-dimethyl-8- (methylthio) -2, 6-dioxo-1H-purin-3 (2H,6H,7H) -yl) prop-1-yn-1-yl) -1, 3-phenylene) bicyclopropane carboxamide

To a solution of 27(7.0mg,0.013mmol) in DMF (2mL) was added MeSNa (1.4mg,0.02 mmol). The mixture was then stirred at room temperature for 4 hours. The solvent was then removed and extracted with dichloromethane (3X 5mL), the organic layer separated and Na2SO4Dry, filter, concentrate and purify by preparative TLC to give 683.3 mg (50%) as a white solid. LC-MS (ESI) M/z [ M + H ]]+Calculated 507.17, found 507.23

Compound 69:

1, 7-dimethyl-8- (methylsulfonyl) -3- (prop-2-yn-1-yl) -6-thioxo-6, 7-dihydro-1H-purin-2 (3H) -one

To a solution of the compound 1, 7-dimethyl-8- (methylsulfonyl) -3- (prop-2-yn-1-yl) -1H-purine-2, 6(3H,7H) -dione (60mg,0.2mmol) in dioxane (3mL) was added P2S5(108mg,0.24 mmol). The mixture was then stirred at 80 ℃ for 4 hours. The solvent was then removed and extracted with dichloromethane (3X 5mL), the organic layer separated and Na2SO4Drying, filtration, concentration and purification by preparative TLC afforded 6919 mg (30%) of compound as a white solid. LC-MS (ESI) M/z [ M + H ]]+Calculated 313.04, found 313.13.

Compounds 70-72 were prepared according to the procedures outlined in the schemes

Preparation 73-74 according to the methods outlined in the schemes

75-77 prepared according to the methods outlined in the schemes

preparation 78 according to the procedure outlined in the scheme

Preparation of 79-80 according to the procedure outlined in the scheme

Preparation of 81-82 according to the procedure outlined in the scheme

83-85 prepared according to the methods outlined in the schemes

86-87 prepared according to the methods outlined in the schemes

Preparation of 88-90 according to the procedure outlined in the scheme

Preparation of 91-100 according to methods for Compounds 70-90

Preparation of 101-110 according to the methods outlined in the schemes

Preparation 111 according to the procedure outlined in the scheme

Preparation 112-113 according to the methods outlined in the schemes

114-117 prepared according to the methods outlined in the schemes

118-120 was prepared according to the method of compounds 110-117.

Necrotic apoptotic activity of compounds of series 2;

Experiment: discovery of a novel class of highly potent necrotic apoptosis inhibitors targeting mixed lineage kinase domain-like proteins

In previous work, we performed cell-based high throughput screening using a chemical library of approximately 200000 compounds to identify inhibitors of necrotic apoptosis and to identify a variety of compounds that protect human colorectal adenocarcinoma (HT-29) cells from TNF- α/Smac mimetics/Z-VAD-fmk (TSZ) -induced necrotic apoptosis[4]. Among these hit compounds, one of them was developed as the first oneMLKL inhibitor NSA revealing the biological function of MLKL in necrotizing apoptosis[4]. Another hit was identified as an inhibitor of RIP1 and was further developed as a highly potent and selective anti-inflammatory agent, RIPA-56[10]. We investigated another hit compound from this high throughput screen, compound 1 (compound 4, supra), which has good anti-necrotic apoptotic potency (EC)50=390±8nM)。

To determine whether compound 1 interacts with an unknown target or with one of three proteins (RIP1, RIP3, and MLKL) known to be directly involved in TSZ-induced necrotic apoptosis, we performed several experiments to determine the effect of compound 1 on the function of RIP1, RIP3, and MLKL. First, we tested whether compound 1 altered RIP1 and RIP3 kinase activity in the enzyme assay, as their kinase activity is essential for the progression of necrotic apoptosis. We found that compound 1 does not inhibit the kinase activity of RIP1 or RIP3 at concentrations up to 10 μ M, which rules out the possibility of RIPK1 or RIPK3 becoming targets. Compound 1 did not block phosphorylation of MLKL based on western blot analysis with anti-phosphorylated MLKL (pmlkl) antibody. However, compound 1 did block the binding of MLKL to NSA according to a binding competition experiment that tested compound 1 for its ability to compete for MLKL binding to NSA. This suggests that MLKL may be the target of compound 1.

To obtain direct evidence that compound 1 targets MLKL, we used a chemogenetic approach. Specifically, affinity probes were used to target compound 1. We have synthesized a series of derivatives based on compound 1 with the aim of improving their anti-necrotic apoptotic efficacy and obtaining high affinity probes for target recognition. We first performed SAR studies at position 8/1/7 of compound 1 by substituting the original methyl group with various functional groups. The results of the necroptosis analysis indicated that all tested substitutions at position 8/1/7 had a detrimental effect on efficacy, indicating that the methyl group was best in the tested structures at these positions. Then, we turned our efforts to the 3-position of compound 1. Related derivative compounds 7-12 and 14-18 were synthesized by the route shown in Table A.

TABLE A. (A) Synthesis of Compounds 7-12 and 14-18. (a) Potassium tert-butoxide, anhydrous THF,75 ℃ overnight; (b) NCS, THF, rt, overnight; (c) NaSMe, DMF, 100 ℃,3 h; (d) potassium hydrogen persulfate, MeOH H2O 1: rt, 4H; (e) k2CO3, DMF, rt,1 h; (f) 3-bromoprop-1-yne, K2CO3, DMF, rt,1 h; (g) pd (PPh3)4, CuI, TEA, anhydrous DMF,60 ℃,6 h; potassium hydrogen persulfate, MeOH H2O 1:1, rt,4H. (B) Structure of compound 19.

The imidazole derivative 2 is cyclized with ethyl methylcarbamate (3) to give 1, 7-dimethylxanthine (4). Subsequent substitution of the chlorine and the mercapto group produces purine derivative 6, which purine derivative 6 is oxidized by oxone to produce 7. Substitution of 7 with different bromo derivatives yields the target compounds 8-12. Compounds 14-18 were synthesized using intermediate 6 as the starting material. The intermediate 6 is substituted with 3-bromoprop-1-yne to give an intermediate 13; subsequent Suzuki coupling with different iodobenzene derivatives and oxidation with potassium hydrogen persulfate provided the target compounds 14-18.

The anti-necrotic apoptotic efficacy of compounds 7-12 and 14-18 was evaluated in the HT-29 cell line; EC (EC)50The values are shown in table B.

TABLE B efficacy of Compounds 7-12 and 14-18 in TSZ-induced necrotic apoptosis assay in HT-29 cells (EC)50)。

[a]Inducing necroptosis in HT-29 cells for 24 hours; EC given herein50Values are the average of at least three independent analyses.

Due to the lack of methyl groups, compound 7 decreased potency by more than 10-fold relative to compound 1. Reduced potency of ethyl analogue 8 compared to Compound 1Lower by a factor of 5. Further extension of the alkyl chain length restored some efficacy (see compounds 9 and 10). Interestingly, compound 12 with an acetylene group was 3 times more potent than compound 1 (methyl group); however, benzyl analog 11 decreased potency by 4-fold relative to compound 12, indicating the presence of a relatively narrow hydrophobic space in the target. To detect the putative binding pocket beyond the defined region of the propynyl group, we synthesized 14 by adding a phenyl group to the end of the propynyl group at 12. Surprisingly, compound 14 showed a 6-fold improvement in potency over compound 12, indicating the presence of "open space" in this region for additional interaction between the target and the inhibitor. To investigate the possibility of additional hydrogen bonding interactions around the phenyl group, we designed and synthesized compounds 15-18. Surprisingly, compounds 15(3-OH), 17 (3-NH)2) And 18(3-NHMe) had an inhibitory potency against necroptosis of cells of 2nM, 7nM and 7nM, respectively. In contrast, methylation of 3-OH of compound 15 (compound 16) resulted in a 15-fold decrease in potency. The enhanced potency of compounds 15, 17 and 18 indicates that hydrogen bonding is likely to occur between the hydrogen bond donor, i.e., the 3-OH of compound 15 (or the 3-NHR of compounds 17-18) and the acceptor residue on the target. The methoxy group of compound 16(3-OMe) lacks hydrogen bond donating properties, eliminating hydrogen bond formation and resulting in loss of potency. Our SAR study successfully improved the potency of the initial hit compound, compound 1, from EC50A value of 390nM decreased until EC50The value was 2nM (Compound 15). This important achievement suggests that MLKL is a pharmaceutically acceptable target and provides a useful chemical tool to study the function of MLKL.

Given that certain heteroaromatic sulfonyl groups can be replaced by various nucleophiles, we speculate that the methylsulfonyl group in compound 1 may act as a covalently bound leaving group in its interaction with MLKL[11]. To test this hypothesis, we synthesized compound 19, which is a close structural analog to compound 1, but in which the methylsulfonyl group had no leaving ability. Compound 19 showed no efficacy in the necroptosis assay, indicating that covalent binding appears to be critical for the efficacy of these inhibitors. We further confirmed the irreversible covalent binding behavior of compounds 12 and 15 by comparing their potency in a washed/unwashed cell assay (wash/no-wash cell assay). For both compounds, the EC recorded for the washed sample50Value and EC of unwashed sample50The values are comparable, indicating that these compounds are covalent inhibitors.

Similar to compound 1, compounds 12 and 14-16(10 μ M) did not inhibit the kinase activity of RIP1 or RIP3 in an in vitro assay. To determine whether the direct target of this series of compounds is MLKL, we performed activity-based protein profiling (ABPP). Compound 12 (an affinity probe) was incubated with MLKL-flag-HT-29 cell lysate for 2 hours, followed by a click reaction to conjugate the probe to a biotin tag. The biotin-labeled compound-protein complex was pulled down and analyzed by SDS-PAGE. We observed that compound 12 could indeed covalently bind to MLKL in cell lysates, and that compound 15 was more effective than 12 in binding MLKL.

The recently developed BTC-ABPP method in our laboratory greatly facilitates the identification of target protein probe modification sites in living cells[12]. Here, we used BTC-ABPP with HT-29 cells to identify binding sites for Compound 12 and MLKL. The b/y ion spectrum from MS/MS analysis showed that in the SNICR peptide of human MLKL, the covalent binding site of compound 12 was Cys-86; this is the same residue proposed as the modification site for NSA[4]. To further validate the relationship between probe binding at this site and the function of compound 12 in necrotic apoptosis, MLKL knockout RIP3-HeLa cells were transfected with either wild-type or C86S (cysteine 86 mutated to serine) mutant forms of MLKL. TSZ stimulation of both types of cells resulted in necrotic apoptosis, indicating that the C86S mutant form of MLKL normally functions in transducing necrotic apoptotic signals. Compound 12 protected cells transfected with wild-type MLKL, but not cells transfected with C86S-MLKL from necrotic apoptosis, further demonstrating that compound 12 inhibits MLKL by binding directly to Cys-86. Cys-86 in human MLKL corresponds in structure to the tryptophan residue in mouse and rat MLKL. Mouse MLHelix H4 containing Cys-86 at the N-terminal domain of the human MLKL structure is absent from KL[13]. In view of this, we speculate that compounds 12 and 15 may show poor inhibition of mouse or rat MLKL function in necrotic apoptosis. We performed necrotic apoptosis assays in mouse MEF and L929 cell lines and rat L6 cell line and found that neither compound 12 nor compound 15 showed inhibitory activity, even beyond their previously established EC50the same is true at the concentration of the values. These results demonstrate that Cys-86, which targets MLKL, is essential for the covalent binding and potency of this series of MLKL inhibitors.

Previous studies have shown that MLKL forms homooligomers when it is phosphorylated by RIP 3. This is a key step in the translocation of MLKL from the cytoplasm to the cell membrane, during which it mediates ion influx and/or causes structural disruption of the cell[5]. To evaluate the mechanism of action of this series of compounds on MLKL, we examined the state of MLKL (oligomeric and phosphorylated states) under TSZ stimulation in the presence of compounds 12 or 15. We first demonstrated that both compounds 12 and 15 can block the formation of MLKL homologous oligomers. Next, we evaluated the effect of MLKL inhibitors on the transfer of MLKL from the cytoplasm to the cell membrane. HT-29 cells were treated with TSZ in the presence of RIPA-56, NSA, Compound 12 or Compound 15 for 6 hours. Proteins from cell lysates were isolated and the soluble and membrane phases were analyzed with anti-MLKL and anti-pMLKL antibodies, respectively. As expected, compounds 12 and 15 did not disrupt phosphorylation of MLKL, but did reduce the level of MLKL in the membrane phase, indicating that these MLKL inhibitors block the transfer of MLKL to the cell membrane, thereby protecting the cells from necrotic apoptosis. We also performed immunofluorescent staining in HT-29 cells using monoclonal anti-pMLKL antibodies. After TSZ stimulation, large bright green fluorescent spots (pMLKL) appeared in the plasma membrane. RIPA-56 and GW806742X completely blocked phosphorylation of MLKL and no pMLKL spot was observed in cells treated with these compounds. In contrast, small pMLKL spots dispersed in the cytoplasm of cells treated with the MLKL inhibitor NSA and compounds 12 and 15, indicating that NSA and compounds 12 and 15 share a similar mode of action and block the transfer of MLKL to the cell membrane. These results were compared with earlier studiesTo make[5]

1.aL.Sun,X.Wang,Trends Biochem.Sci 2014,39,587-593;bM.Pasparakis,P.Vandenabeele,Nature 2015,517,311-320.

2.N.Holler,R.Zaru,O.Micheau,M.Thome,A.Attinger,S.Valitutti,J.-L.Bodmer,P.Schneider,B.Seed,J.Tschopp,Nat.Immunol.2000,1,489-495.

3.aS.He,L.Wang,L.Miao,T.Wang,F.Du,L.Zhao,X.Wang,Cell 2009,137,1100-1111;bD.-W.Zhang,J.Shao,J.Lin,N.Zhang,B.-J.Lu,S.-C.Lin,M.-Q.Dong,J.Han,Science 2009,325,332-336.

4, L.Sun, H.et al, Cell 2012,148, 213-.

5.aH.Wang,L.Sun,L.Su,J.Rizo,L.Liu,L.-F.Wang,F.-S.Wang,X.Wang,Mol.Cell 2014,54,133-146;bZ.Cai,S.Jitkaew,J.Zhao,H.-C.Chiang,S.Choksi,J.Liu,Y.Ward,L.-g.Wu,Z.-G.Liu,Nat.Cell Biol.2014,16,55-65.

aJ.Wu, et al, Cell Res.2013,23, 994-1006; bd. ofengeim, y.ito, a.najafov, y.zhang, b.shan, j.p.dewitt, j.ye, x.zhang, a.chang, h.vakifahmaloglu-Norberg, cellports 2015,10, 1836-; pierce, a, negroni, l, stronti, r, vitali, e, press, j, bertin, p, j, gough, m, aloi, s, cucchiara, Am j, gastroenterol, 2014,109,279-287.

J.M.Hildebrand, et al, Proc.Natl.Acad.Sci.U.S.A.2014,111,15072-15077.

8.D.Liao,L.Sun,W.Liu,S.He,X.Wang,X.Lei,Medchemcomm 2014,5,333-337.

Sammond, et al, bioorg, med, chem, lett, 2005,15, 3519-side 3523.

Y.ren, et al, J.Med.chem. (http:// dx.doi.org/10.1021/acs.jmedchem.6b01196, in Press)

11.P.Lan, et al Tetrahedron Lett.2008,49,1910-1914.

12.H.Sun,Y.Ren,W.Hou,L.Li,F.Zeng,S.Li,Y.Ma,X.Liu,S.Chen,Z.Zhang,Chem.Commun.2016,52,10225-10228.

13.L.Su,B.Quade,H.Wang,L.Sun,X.Wang,J.Rizo,Structure 2014,22,1489-1500.

144页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:制备吲哚啉并苯并二氮杂*衍生物的方法

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!