CN110573509B - MLKL inhibitors - Google Patents

MLKL inhibitors Download PDF

Info

Publication number
CN110573509B
CN110573509B CN201880027489.8A CN201880027489A CN110573509B CN 110573509 B CN110573509 B CN 110573509B CN 201880027489 A CN201880027489 A CN 201880027489A CN 110573509 B CN110573509 B CN 110573509B
Authority
CN
China
Prior art keywords
dione
purine
compound
mmol
solution
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN201880027489.8A
Other languages
Chinese (zh)
Other versions
CN110573509A (en
Inventor
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
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
National Institute of Biological Sciences Beijin
Original Assignee
National Institute of Biological Sciences Beijin
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by National Institute of Biological Sciences Beijin filed Critical National Institute of Biological Sciences Beijin
Publication of CN110573509A publication Critical patent/CN110573509A/en
Application granted granted Critical
Publication of CN110573509B publication Critical patent/CN110573509B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • A61K31/52Purines, e.g. adenine
    • A61K31/522Purines, e.g. adenine having oxo groups directly attached to the heterocyclic ring, e.g. hypoxanthine, guanine, acyclovir
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D473/00Heterocyclic compounds containing purine ring systems
    • C07D473/02Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6
    • C07D473/04Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6 two oxygen atoms
    • C07D473/06Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6 two oxygen atoms with radicals containing only hydrogen and carbon atoms, attached in position 1 or 3
    • C07D473/08Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6 two oxygen atoms with radicals containing only hydrogen and carbon atoms, attached in position 1 or 3 with methyl radicals in positions 1 and 3, e.g. theophylline
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D473/00Heterocyclic compounds containing purine ring systems
    • C07D473/02Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6
    • C07D473/04Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6 two oxygen atoms
    • C07D473/06Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6 two oxygen atoms with radicals containing only hydrogen and carbon atoms, attached in position 1 or 3
    • C07D473/10Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6 two oxygen atoms with radicals containing only hydrogen and carbon atoms, attached in position 1 or 3 with methyl radicals in positions 3 and 7, e.g. theobromine
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D473/00Heterocyclic compounds containing purine ring systems
    • C07D473/02Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6
    • C07D473/04Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6 two oxygen atoms
    • C07D473/06Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6 two oxygen atoms with radicals containing only hydrogen and carbon atoms, attached in position 1 or 3
    • C07D473/12Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6 two oxygen atoms with radicals containing only hydrogen and carbon atoms, attached in position 1 or 3 with methyl radicals in positions 1, 3, and 7, e.g. caffeine
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D513/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
    • C07D513/12Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00 in which the condensed system contains three hetero rings
    • C07D513/14Ortho-condensed systems

Abstract

Purine derivatives that inhibit necrotic apoptosis and/or human MLKL, pharmaceutical compositions thereof, and methods of treating MLKL-mediated disorders using an effective amount of a compound or composition. The MLKL-mediated disorders are pathology-related necrotizing apoptosis, including ischemia reperfusion injury, neurodegeneration and inflammatory diseases such as acute pancreatitis, multiple sclerosis, inflammatory bowel disease and allergic colitis.

Description

MLKL inhibitors
Introduction to the invention
We disclose herein the development of novel mixed lineage kinase domain like protein (MLKL) inhibitors with single nanomolar potency. Using a method of converting biochemistry to chemical activity-based protein profiling (BTC-ABPP), we were able to determine that inhibitors were covalently bound to cysteine 86 (Cys-86) of MLKL. This is the first example of the identification of the binding site of an MLKL inhibitor using LC-MS/MS.
Necrotic apoptosis is a type of programmed necrotic cell death that involves organ development, tissue homeostasis, inflammation, and disease pathogenesis [1] . Following induction of necrotic apoptosis, receptor interacting protein kinase 1 (RIP 1) [2] 、RIP3 [3] And MLKL [4] Are activated in sequence. MLKL as an effector of necrotic apoptosis can subsequently form oligomers and migrate to cell membranes where it mediates ion influx and/or causes destruction of cellular structures [5] . Recent studies have established that MLKL is involved in a variety of human inflammatory diseasesIncluding acute pancreatitis, multiple sclerosis, inflammatory bowel disease, and allergic colitis [6] . In this case, MLKL is considered as a potential therapeutic target for drug discovery.
There are only two known inhibitors of MLKL: necrosulfonamide (NSA) [4] And GW806742X [7] . The moderate potency and narrow structure-activity relationship (SAR) spectrum of NSA has limited its development as a drug [1a,8] . GW806742X, which targets the pseudo kinase domain of MLKL, has been found to have off-target activity against other kinases, including VEGFR2 [9] . We also disclose that GW806742X also has off-target activity against RIP1 and RIP3, so it is not possible to conclude that GW 806742X-mediated prevention of necrotic apoptosis is dependent only on the targeting of MLKL. In view of the deficiencies of the MLKL inhibitors reported so far, there is an urgent need to develop new, highly potent MLKL inhibitors.
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:
Figure BDA0002247439760000021
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 joined to form a ring;
n is 0, 1 or 2; and
or a pharmaceutically acceptable salt, hydride or stereoisomer of said compound.
In embodiments:
(a) One of R1-R4 is an alkyl carbocyclyl, such as methylcyclopropyl;
(b) One of R1-R4 includes a fluoroalkyl group, such as CF3;
(c) R4 is alkylcyano or alkylCR, e.gCH 2 CR, 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 embodiments:
one, two or three of R1-R4 are Me;
n is 2;
one of R1-R4 is alkylcyano or alkylCR, e.g. CH 2 CR, wherein R is H or an optionally substituted, optionally heteroatom-containing, optionally cyclic C1-C18 hydrocarbyl group; and/or
In embodiments:
each of R1, R2 and R3 is Me;
n is 2; and
r4 is alkylcyano or alkylCR, e.g. CH 2 CR, wherein R is H or an optionally substituted, optionally heteroatom-containing, optionally cyclic C1-C18 hydrocarbyl group.
In embodiments: 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 embodiments: an optionally substituted, optionally heteroatom-containing, optionally cyclic C1-C18 hydrocarbyl is an optionally substituted, optionally heteroatom-containing, optionally cyclic alkyl, alkenyl or alkynyl, or an optionally substituted, optionally heteroatom-containing aryl; and/or optionally substituted heteroatoms are 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, sulfonamide), 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 formulas of table 1.
In embodiments: 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 associated disorder, disease or condition.
In another aspect, the invention provides a composition comprising a medicament, 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:
Figure BDA0002247439760000031
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 joined 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 invention provides an MLKL inhibitor compound of formula I:
Figure BDA0002247439760000041
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 joined 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 a fluoroalkyl group, such as CF3;
(c) R4 is alkylcyano or alkylCR, e.g. CH 2 CR, 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 embodiments: one, two or three of R1-R4 are Me; and/or n is 2.
In embodiments: the 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 (e.g. amino, alkylamino, dialkylamino, amido, sulfonamide), 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 formulas of the novel compounds of table 1.
In embodiments, the subject compounds or compositions are in unit dosage form.
The present invention includes all combinations of the specific embodiments described herein.
Description of specific embodiments of the invention
The following description of specific implementations 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.
Throughout this description and throughout this specification, unless indicated or otherwise, the terms "a" and "an" mean one or more, the term "or" means and/or, and the polynucleotide sequence is understood to include the opposite strand as well as the alternative backbones described herein. Further, a generic expression is an abbreviation for the expression of all members of the generic class; for example, the recitation of (C1-C3) alkyl is shorthand for all recitations of C1-C3 alkyl: 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 functionalities are haloformyl, hydroxy, aldehyde, amine, azo, carboxyl, cyano, thiocyanyl, carbonyl, halogen, peroxy hydroxy, imine, aldimine, isocyanide, isocyanate, nitrate, nitrile, nitrite, nitro, nitroso, phosphate, phosphonyl, sulfide, sulfonyl, sulfo, and mercapto.
Unless otherwise indicated, the term "alkyl" by itself or as part of another substituent refers to a straight or branched or cyclic hydrocarbon group, or a combination 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, t-butyl, isobutyl, sec-butyl, cyclohexyl, (cyclohexyl) methyl, cyclopropylmethyl, and homologs and isomers such as n-pentyl, n-hexyl, n-heptyl, n-octyl, and the like. If cyclic, the preferred alkyl groups are C1-C8, C3-C18.
The term "alkenyl" by itself or as part of another substituent means a straight or branched or cyclic 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 double bonds. Examples of alkenyl groups include vinyl, 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 hydrocarbon radical 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, e.g., as-CH 2 -CH 2 -CH 2 -CH 2 -for example. Typically, alkyl (or alkylene) groups will have from 1 to 24 carbon atoms, with those 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 indicated, the term "heteroalkyl" by itself or in combination with another term means a stable straight or branched or cyclic hydrocarbon group or combination thereof, consisting of the recited number of carbon atoms 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 heteroatoms may optionally be quaternized. Heteroatoms (heteroatoms) O, N, P and S can be placed at any internal position of the heteroalkyl group. The heteroatom Si may be placed at any position of the heteroalkyl group, including where the alkyl group is attached to the remainder of the molecule. Examples include-CH 2 -CH 2 -O-CH 3 、-CH 2 -CH 2 -NH-CH 3 、-CH 2 -CH 2 -N(CH 3 )-CH 3 、-CH 2 -S-CH 2 -CH 3 、-CH 2 -CH 2 、-S(O)-CH 3 、-CH 2 -CH 2 -S(O) 2 -CH 3 、-CH=CH-O-CH 3 、-Si(CH 3 ) 3 、-CH 2 -CH=N-OCH 3 and-ch=ch-N (CH 3) -CH 3 . At most two heteroatoms may be consecutive, e.g. -CH 2 -NH-OCH 3 and-CH 2 -O-Si(CH 3 ) 3
Similarly, the term "heteroalkylene" by itself or as part of another substituent means a divalent group derived from a heteroalkyl group, e.g., in-CH 2 -CH 2 -S-CH 2 -CH 2 -and-CH 2 -S-CH 2 -CH 2 -NH-CH 2 -for example. For heteroalkylene groups, the heteroatom may also occupy either or both of the chain ends (e.g., alkyleneoxy, alkylenedioxy, alkyleneamino, alkylenediamino, and the like). Furthermore, no orientation of the linking groups is implied for the alkylene and heteroalkylene linking groups.
Unless otherwise indicated, the terms "cycloalkyl" and "heterocycloalkyl" by themselves or in combination with other terms, refer to the cyclic forms of "alkyl" and "heteroalkyl," respectively. Thus, a cycloalkyl group has 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 heteroaryl group consists 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 can optionally be oxidized and the nitrogen heteroatom can optionally be quaternized. In addition, for heterocycloalkyl, the heteroatom may occupy the position where 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 groups 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 indicated, 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 the same or different numbers of halogen atoms ranging from 1 to (2 m '+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-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 ranging from 2 to (2 m '+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 (2 m '+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-trifluoro-2-bromo-2-chloroethyl, and the like.
The term "acyl" refers to those groups derived from organic acids by removal of the hydroxyl moiety 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, typically aromatic, hydrocarbon substituent which may be a single ring or multiple rings (up to three rings) fused together or covalently linked. 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. Heteroaryl groups may be attached to the remainder of the molecule through heteroatoms. 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-pyrimidinyl, 4-pyrimidinyl, 5-benzothiazolyl, purinyl, 2-benzimidazolyl, 5-indolyl, 1-isoquinolyl, 5-isoquinolyl, 2-quinoxalinyl, 5-quinoxalinyl, 3-quinolinyl and 6-quinolinyl.
For simplicity, the term "aryl" when used in combination with other terms (e.g., aryloxy, arylthio, arylalkyl) includes 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, etc.), including those alkyl groups whose carbon atoms (e.g., methylene groups) have been replaced with, for example, oxygen atoms (e.g., phenoxymethyl, 2-pyridyloxymethyl, 3- (1-naphthyloxy) propyl, etc.).
Each of the above terms (e.g., "alkyl," "heteroalkyl," "aryl," and "heteroaryl") is intended to include both substituted and unsubstituted forms of the indicated group. Preferred substituents for each type of group are provided below.
Substituents for alkyl and heteroalkyl (those groups referred to as alkylene, alkenyl, heteroalkylene, heteroalkenyl, alkynyl, cycloalkyl, heterocycloalkyl, cycloalkenyl, and heterocycloalkenyl) can be various groups selected from the group consisting of: -OR ', =o, =nr', =n-OR ', -NR' R ', -SR', halogen, -SiR 'R', -OC (O) R ', -C (O) R', -CO 2 R'、-CONR'R"、-OC(O)NR'R"、-NR"C(O)R'、-NR'-C(O)NR"R'"、-NR'-SO 2 NR'"、-NR"CO 2 R'、-NH-C(NH 2 )=NH、-NR'C(NH 2 )=NH、-NH-C(NH 2 )=NR'、-S(O)R'、-SO 2 R'、-SO 2 NR'R"、-NR"SO 2 R, -CN and-NO 2 The 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, unsubstituted aryl, aryl 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. Typically, alkyl or heteroalkyl groups have from 0 to 3 substituents, with those groups 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 foregoing discussion of substituents, those skilled in the art will understand that the term "alkyl" is intended to include, for example, trihaloalkyl (e.g., -CF 3 and-CH 2 CF 3 ) Is a group of (2).
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', -CO 2 R'、-CONR'R"、-OC(O)NR'R"、-NR"C(O)R'、-NR"CO 2 R'、-NR'-SO 2 NR"R'"、-S(O)R'、-SO2R'、-SO 2 NR'R"、-NR"SO 2 R, -CN and-NO 2 Wherein R 'and R' are as defined above. Further preferred substituents are selected from: -OR ', =o, -NR ' R ", halogen, -OC (O) R ', -CO 2 R'、-CONR'R"、-OC(O)NR'R"、-NR"C(O)R'、-NR"CO 2 R'、-NR'-SO 2 NR"R'"、-SO 2 R'、-SO 2 NR'R"、-NR"SO 2 R, -CN and-NO 2
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, -NO 2 、-CO 2 R'、-CONR'R"、-C(O)R'、-OC(O)NR'R"、-NR"C(O)R'、-NR"CO2R'、-NR'-C(O)NR"R'"、-NR'-SO 2 NR"R'"、-NH-C(NH2)=NH、-NR'C(NH 2 )=NH、-NH-C(NH 2 )=NR'、-S(O)R'、-SO 2 R'、-SO 2 NR'R"、-NR"SO 2 R、-N 3 、-CH(Ph) 2 Perfluoro (C1-C4) alkoxy and perfluoro (C1-C4) alkyl in an amount ranging from 0 to the total number of open valencies 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 or unsubstituted (C3-C7) spirocycloalkylAnd (3) group substitution. The (C3-C7) spirocycloalkyl groups may be substituted in the same manner as defined herein for "cycloalkyl". Typically, aryl or heteroaryl groups have 0 to 3 substituents, those groups having two or fewer substituents are preferred in the present invention. In one embodiment of the invention, the aryl or heteroaryl group is unsubstituted or monosubstituted. In another embodiment, the aryl or heteroaryl group is unsubstituted.
Preferred substituents for aryl and heteroaryl groups are selected from: halogen, -OR ', -OC (O) R', -NR 'R', -SR ', -R', -CN, -NO 2 、-CO 2 R'、-CONR'R"、-C(O)R',-OC(O)NR'R"、-NR"C(O)R'、-S(O)R'、-SO 2 R'、-SO 2 NR'R"、-NR"SO 2 R、-N 3 、-CH(Ph) 2 Perfluoro (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', -CN, -NO 2 、-CO 2 R'、-CONR'R"、-NR"C(O)R'、-SO 2 R'、-SO 2 NR'R"、-NR"SO 2 R, perfluoro (C1-C4) alkoxy and perfluoro (C1-C4) alkyl.
substituent-CO as used herein 2 H includes bioisostere substitutions thereof; see, e.g., the Practice of Medicinal Chemistry; wermuth, c.g., ed.; academic Press New York,1996; p.203.
The two substituents on adjacent atoms of the aromatic or heteroaromatic ring may optionally be represented by the formula-T-C (O) - (CH) 2 ) Substituents of q-U-, wherein T and U are independently-NH-, -O-, -CH 2 -or a single bond, and q is an integer from 0 to 2. Alternatively, two substituents on adjacent atoms of an aromatic or heteroaromatic ring may be optionally replaced by substituents of the formula-A- (CH 2) r-B-, wherein A and B are independently-CH 2 -、-O-、-NH-、-S-、-S(O)-、-S(O) 2 -、-S(O) 2 NR' -or a single bond, and r is an integer of 1 to 3. One of the single bonds of the new ring thus formed may optionally be replaced by a double bond. Alternatively, two substituents on adjacent atoms of an aromatic or heteroaromatic ring may optionally be represented by formula- (CH) 2 )s-X-(CH 2 ) t-wherein s and t are independently substitutedThe ground is an integer of 0 to 3, and X is-O-, -NR' -, -S-, -S (O) 2 -, or-S (O) 2 NR' -. -NR' -and-S (O) 2 The substituent R 'in NR' -is selected from hydrogen or unsubstituted (C1-C6) alkyl.
In embodiments, the substituents, particularly the substituents of R1-R4, are selected from:
Figure BDA0002247439760000101
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 embodiments: the 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 (e.g. amino, alkylamino, dialkylamino, amido, sulfonamide), 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 the group consisting of:
Figure BDA0002247439760000111
in embodiments, R1 and R2 are linked to form a ring, e.g., R1/R2 is Me/Me or Me/Et linked to form a 5-or 6-membered ring, particularly wherein R3 is methyl, and/or R4 is a substituent selected from the group consisting of:
Figure BDA0002247439760000112
preferred substituents are disclosed herein and illustrated in the tables, structures, examples and claims, and may be applied to the various compounds of the present invention, i.e., substituents of any given compound may 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, aldimine, alkanoyloxy, alkoxy, alkoxycarbonyl, alkoxy, alkyl, amine, azo, halogen, carbamoyl, carbonyl, carboxamido, carboxyl, cyano, ester, halo, haloformyl, peroxy hydroxy, imine, isocyanide, isocyanate, N-t-butoxycarbonyl, nitrate, nitrile, nitrite, nitro, nitroso, phosphate, phosphonyl, sulfide, sulfonyl, sulfo, mercapto, thiol, thiocyano, trifluoromethyl or trifluoromethyl ether (OCF 3).
The term "pharmaceutically acceptable salts" is intended to include salts of the active compounds prepared with relatively non-toxic 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 (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, monohydrocarbonic, phosphoric, monohydrogenphosphoric, dihydrogenphosphoric, sulfuric, monohydrogensulfuric, hydroiodic or phosphorous acids and the like, and those derived from relatively non-toxic 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 acid or galacturonic acid and the like. Certain specific compounds of the invention contain both basic and acidic functionalities that allow the compounds to be converted to base or acid addition salts.
The neutral form of the compound may be regenerated by contacting the salt with a base or acid and isolating the parent compound in a 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 the present invention, the 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. Alternatively, prodrugs can be converted to the compounds of the present invention in an ex vivo environment by chemical or biochemical means. For example, prodrugs can be slowly converted to the compounds of the present invention when placed 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 have higher oral bioavailability 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 then metabolically hydrolyzed to the carboxylic acid, i.e., the active entity. Further examples include peptidyl derivatives of the compounds of the invention.
Certain compounds of the invention may exist in unsolvated forms as well as solvated forms, including hydrated forms. In general, 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 invention have asymmetric carbon atoms (optical centers) or double bonds; racemates, diastereomers, geometric isomers and individual isomers are all 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. -CD 3 、CD 2 H or CDH 2 Replacing methyl. For example, the compounds may be administered using a radioisotope, such as tritium @, for example 3 H) Iodine-125% 125 I) Or C-14% 14 C) And (3) radiolabeling. 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" refers to an amount of a subject compound that will elicit the biological or medical response of a tissue, system, animal or human that is being sought, for instance, by a researcher, veterinarian, medical doctor or other clinician, which is sufficient to prevent the development of or to alleviate, to a certain 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, as well as 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 the subject compound, particularly such compositions co-packaged with instructions describing use of the compositions for treating an applicable disease or disorder (herein).
The composition for administration may take the form of a bulk liquid solution or suspension or a bulk powder. More commonly, however, the composition is 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 compositions, or in the case of solid compositions, pills, tablets, capsules, lozenges and the like. In such compositions, the compound is typically a minor component (from about 0.1% to about 50% by weight, or preferably about 1% to 40% by weight), with the remainder being various vehicles or carriers and processing aids that assist in forming the desired administration form.
Suitable excipients or carriers and methods for preparing the administrable compositions are known or obvious 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 described herein or otherwise known in the art, in particular other anti-necrotic agents. Thus, the compositions may be administered in single dosage units, alone, in combination, or in combination.
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, route of administration, etc. Generally, the amount of active compound in a unit dosage formulation can vary or be adjusted from about 1, 3, 10, or 30 to about 30, 100, 300, or 1000mg, depending on the particular application. In a specific embodiment, the unit dosage forms are packaged in multiple packages suitable for sequential use, such as blister packages comprising at least 6, 9 or 12 sheets of unit dosage forms. The actual dosage used may vary depending on the needs of the patient and the severity of the condition being treated. It is within the skill of the art to determine the appropriate dosage for a particular situation. Generally, treatment is initiated with a smaller dose than the optimal dose of the compound. Thereafter, the dosage is increased by a small amount until the optimum effect in the case is reached. For convenience, the total daily dose may be divided and administered in portions within the day, if desired.
The compounds may be administered by a variety of methods including, but not limited to, parenteral, topical, oral, or local (local) administration, e.g., by aerosol or transdermal, for prophylactic and/or therapeutic treatment. Furthermore, the treatment regimen (e.g., the dosage and number of administrations) may vary depending on the observed effect of the administered therapeutic agent on the patient and on the observed response of the disease to the administered therapeutic agent, 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 efficacious compounds, microgram (ug) amounts per kilogram of patient may be sufficient, e.g., 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 typically 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 regimen of administration, as well as administration to a particular patient, will also be tailored 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 in accordance with the discretion of the attending clinician, taking into account factors such as age, condition and size of the patient, as well as the efficacy of the compound, severity of the disease being treated, and the like. For example, the dosage regimen of the compounds may be orally administered in two to four (preferably two) divided doses of 10 mg to 2000 mg/day, preferably 10 to 1000 mg/day, more preferably 50 to 600 mg/day. Intermittent therapy (e.g., one or three of four weeks) may also be used.
The compounds of the invention are particularly useful in the treatment of diseases/disorders that may be modulated at least in part by programmed necrosis, apoptosis or inflammatory cytokine production, particularly 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 (nonalcoholic steatohepatitis, alcoholic steatohepatitis, autoimmune hepatitis, autoimmune liver and gall disease, primary Sclerosing Cholangitis (PSC), acetaminophen toxicity, hepatotoxicity), kidney damage/injury (nephritis, kidney transplantation, surgery, administration of nephrotoxic drugs (e.g. cisplatin), acute Kidney Injury (AKI)), celiac disease, autoimmune idiopathic thrombocytopenic platelet (sira), systemic vascular inflammatory response (cvp), vascular graft (cvp), vascular reperfusion injury (septicemia), systemic vascular graft(s), vascular graft (septicemia), 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 mellitus, wegener's granulomatosis, pulmonary sarcoidosis, behcet's Disease, interleukin 1 converting enzyme (ICE, also known as caspase-1) associated fever syndrome, chronic Obstructive Pulmonary Disease (COPD), tumor necrosis factor receptor associated periodic syndrome (TRAPS), periodontitis, EMO deficiency syndrome (F-kappa-B essential regulator gene (also known as IKKgamma or IKKG) deficiency syndrome), HOIL-1 deficiency (also known as RBCK 1) heme oxidation IRP2 ubiquitin ligase-1 deficiency), linear ubiquitin chain assembly complex (LUBAC) deficiency syndrome, hematological and solid organ malignancies, bacterial and viral infections (such as tuberculosis and influenza) and lysosomal storage diseases (especially high snows (Gaucher Disease), including GM2 gangliosidosis, alpha-mannosidosis, aspartylglucosamine urosis, cholesterol ester storage Disease, chronic hexosaminidase A deficiency, cystinosis, danondisease, fabry Disease, farber Disease, fucosidosis, galactosialidosis, GM1 gangliosidosis, mucosis, infant free sialidosis, juvenile hexosaminidase A deficiency, crabbe Disease (Krabbe Disease), lysosomal acid lipase deficiency, metachromatic leukodystrophy, mucopolysaccharidosis, multiple sulfatase deficiency, niman-Pick Disease (Niemann-Pick Disease), neuronal ceroid lipofuscinosis, pompe disease, compact osteogenesis imperfecta (Sandhoff disease), schindler disease, sialidosis, tay-Sachs and Wolman disease. Treatment of the above-mentioned diseases/disorders may more particularly involve amelioration of organ damage or injury caused by the persistence of the disease. For example, the compounds of the invention may be particularly useful for ameliorating brain tissue damage or injury following ischemic or traumatic brain injury, or for ameliorating heart tissue damage or injury following myocardial infarction, or for ameliorating brain tissue damage or injury associated with huntington's disease, alzheimer's disease, or parkinson's disease, or for ameliorating liver tissue damage or injury associated with non-alcoholic steatohepatitis, autoimmune hepatitis, autoimmune hepatobiliary disease, or primary sclerosing cholangitis. In addition, the treatment of a disease/disorder selected from the group of diseases/disorders described herein may more particularly involve the amelioration of tissue damage or injury associated with excess acetaminophen, or the amelioration of kidney tissue damage or injury following kidney transplantation or administration of a nephrotoxic drug or substance (e.g., cisplatin).
The subject compounds are particularly useful for treating 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.
Treatment of MLKL-mediated disease conditions, or more broadly, immune-mediated diseases, may be achieved using the compounds as monotherapy or in dual or multiple combination therapies, particularly for refractory cases, as in combination with other anti-inflammatory and/or anti-TNF agents that may be administered in therapeutically effective amounts, as known in the art.
The subject compounds may be used alone or in combination with other therapeutic agents. Thus, combination therapy comprises administration of at least one pharmaceutically acceptable crystalline form of a compound and at least one other therapeutically active agent. The subject compound and the 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) and the relative timing of administration are selected to achieve the desired combination therapeutic effect. Thus, in another aspect, a combination comprising a pharmaceutically acceptable crystalline form of a compound and one or more other therapeutically active agents is provided. Thus, in one aspect of the invention, a pharmaceutically acceptable crystalline form of the subject compound or a pharmaceutical composition comprising a pharmaceutically acceptable crystalline form of the subject compound may be used in combination with or include one or more other therapeutic agents, such as anti-inflammatory agents and/or anti-TNF agents. For example, for any of the above indications, the subject compounds may be administered in combination with other anti-inflammatory agents, including oral or topical corticosteroids (e.g
Figure BDA0002247439760000161
And budesonide), anti-TNF agents (including anti-TNF biologic agents), 5-aminosalicylic acid and mesalamine formulations, hydroxychloroquine, thiopurines (azathioprine, mercaptopurine), methotrexate, cyclophosphamide, cyclosporine, calcineurin inhibitors (cyclosporine, pimecrolimus, tacrolimus), mycophenolic acid>
Figure BDA0002247439760000162
mTOR inhibitors (temsirolimus, everolimus), JAK inhibitors (tofacitinib +.>
Figure BDA0002247439760000163
) Syk inhibitor (fositinib), anti-IL 6 biologic, anti-IL 1 (afatinib)That is, emotion->
Figure BDA0002247439760000164
) Carbamab->
Figure BDA0002247439760000165
Li Naxi Pu->
Figure BDA0002247439760000166
anti-IL 12 and IL23 biological agent (Utexab->
Figure BDA0002247439760000167
) anti-IL 17 biologic (Stuzumab), anti-CD 22 (epratuzumab), anti-integrin (natalizumab) and +.>
Figure BDA0002247439760000168
) Windouzumab->
Figure BDA0002247439760000169
anti-IFNa (cetrimab), anti-CD 20 or CD4 biologic agents, and other cytokine inhibitors or biologic agents of T cell or B cell receptors or interleukins.
Examples of suitable anti-inflammatory biologic agents include
Figure BDA00022474397600001610
(anti-IL 6R mAb), anti-CD 20 mAb (rituximab ++>
Figure BDA00022474397600001611
And ofatuzumab->
Figure BDA00022474397600001612
) Arbazedox->
Figure BDA00022474397600001613
Anakinra->
Figure BDA00022474397600001614
Utex monoclonal antibody->
Figure BDA00022474397600001615
And belimumab->
Figure BDA00022474397600001616
Examples of other suitable anti-inflammatory biological agents include cinaman +.>
Figure BDA00022474397600001617
Li Naxi Pu->
Figure BDA00022474397600001618
Sukunmab, epaizumab, sibutrab and Utekunmab ++ >
Figure BDA00022474397600001619
Examples of suitable anti-TNF agent biologic agents include etanercept (etanercept,) and ∈>
Figure BDA0002247439760000174
) Adalimumab->
Figure BDA0002247439760000171
Infliximab->
Figure BDA0002247439760000172
Cetuximab->
Figure BDA0002247439760000173
And golimumab->
Figure BDA0002247439760000175
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 person in need thereof such that a disease condition mediated by MLKL is alleviated, ameliorated, or prevented.
"treatment" or "treatment" is intended to mean at least alleviating a disease or disorder in a patient. Methods of treatment for alleviating 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 local 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 inhalation through the mouth or through the nasal passages into the lungs of a patient. Topical administration includes application to the skin.
The compounds of the invention may be administered at once or according to a dosing regimen wherein multiple doses are administered at different time intervals over a given period of time. For example, the administration may be once, twice, three times or four times daily. The administration may be performed 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 on 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 the 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 dosing regimens may need to be adjusted in view of the individual patient's response to a dosing regimen or the change in the individual patient's needs over time. The total daily dose is 1mg to 2000mg.
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. The invention therefore also relates to pharmaceutical compositions comprising a compound of the invention and one or more pharmaceutically acceptable excipients. The invention also relates to a dosage unit comprising a compound of the invention and one or more pharmaceutically acceptable excipients.
The pharmaceutical compositions or dosage 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, for example, with powders, syrups and injections. Alternatively, the pharmaceutical compositions or dosage units of the present invention may be prepared and packaged in unit dosage forms. For oral use, for example, one or more tablets or capsules may be administered. A dose of a pharmaceutical composition comprises at least a therapeutically effective amount of a compound of the invention. When prepared in unit dosage form, the 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 a 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 effect treatment of an MLKL-mediated disease or disorder.
As used herein, "pharmaceutically acceptable excipient" means a material, composition, or vehicle that is involved in imparting a shape or consistency to the composition. Each excipient, when mixed, must be compatible with the other ingredients of the pharmaceutical composition, thereby avoiding interactions that would greatly 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 pharmaceutically unacceptable. In addition, each excipient must of course be of sufficiently high purity to be pharmaceutically acceptable.
The compounds of the invention and pharmaceutically acceptable excipient or excipients are typically 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, powders for reconstitution, and the like; (3) a dosage form suitable for transdermal administration, such as a transdermal patch; (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 upon the particular dosage form selected. In addition, suitable pharmaceutically acceptable excipients may be selected for the particular function they may perform in the composition. For example, certain pharmaceutically acceptable excipients may be selected as 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 as they facilitate carrying or transporting one or more compounds of the invention from one organ or body part to another organ or body part 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, tackifiers, antioxidants, preservatives, stabilizers, surfactants, and buffers. Those 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 artisan will have knowledge and skill in the art to enable them to select the appropriate amount of suitable pharmaceutically acceptable excipients for use in the present invention. In addition, many sources describing pharmaceutically acceptable excipients are available to those skilled in the art, and can be used to select the appropriate pharmaceutically acceptable excipient. Examples include the pharmaceutical science of Remington (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 sciences of ramington (supra). Thus, another embodiment of the invention is a method of preparing a pharmaceutical composition or dosage unit comprising the step of mixing 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, guar gum, povidone, and cellulose and derivatives thereof (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 sodium carboxymethylcellulose. 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 references therein, are incorporated by reference in their entirety for all purposes.
Exemplary compounds; in embodiments, the compound is selected from table 1.
Table 1. Series 1: the black, simple numbered compound is 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 new.
Figure BDA0002247439760000201
/>
Figure BDA0002247439760000211
/>
Figure BDA0002247439760000221
/>
Figure BDA0002247439760000231
/>
Figure BDA0002247439760000241
/>
Figure BDA0002247439760000251
/>
Figure BDA0002247439760000261
/>
Figure BDA0002247439760000271
/>
Figure BDA0002247439760000281
Synthesis
Compound 1: 8-mercapto-1, 3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione
Figure BDA0002247439760000282
(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 (200.0 mg,0.93 mmol) and K 2 CO 3 To a solution of (154.8 mg,1.12 mmol) in anhydrous DMF (2 mL) was added methyl iodide (159.1 mg,1.12 mmol) and stirred under nitrogen at room temperature for 4h. The reaction mixture was poured into water and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by silica gel chromatography (PE: ea=2:1) to give a white solid (151.0 mg, 70.4%). 1 H-NMR(400MHz,CDCl 3 ):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 (151.0 mg,0.66 mmol) in anhydrous DMF (2 mL) was added NaHS (111.3 mg,1.98 mmol) 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 (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by silica gel chromatography (PE: ea=1:4) to give a white solid (128.0 mg, 86.7%). 1 H-NMR(400MHz,DMSO-d 6 ):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
Figure BDA0002247439760000291
To compound 1 (20.0 mg,0.09 mmol) and K 2 CO 3 To a solution of (14.7 mg,0.11 mmol) in anhydrous DMF (2 mL) was added methyl iodide (15.1 mg,0.11 mmol) and stirred under nitrogen at room temperature for 2h. The reaction mixture was poured into water and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give a white solid (19.2 mg, 95.05%). 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000292
Compound 4:1,3, 7-trimethyl-8- (methylsulfonyl) -1H-purine-2, 6 (3H, 7H) -dione
Figure BDA0002247439760000301
To a solution of Compound 2 (18.0 mg,0.07 mmol) in MeOH (2 mL) was added potassium hydrogen persulfate (69.2 mg,0.11 mmol) in H 2 O (2 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Drying, filtration, concentration and purification by preparative TLC gave compound 3 as a white solid (3.4 mg, 20.5%). 1 H-NMR(400MHz,CDCl 3 ):δ4.28(s,3H),3.58(s,3H),3.41(s,3H),3.18(s,3H).MS(m/z):257.06[M+H]+.
46.3mg (30.8%) of compound were obtained as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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- (ethylthio) -1,3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione
Figure BDA0002247439760000302
To compound 1 (30 mg) and K 2 CO 3 To a solution of (21.98 mg) in anhydrous DMF (2 mL) was added ethyl iodide (24.8 mg) and stirred under nitrogen at room temperature for 3h. The reaction mixture was poured into water and extracted with ethyl acetate (3 x 5 ml), the organic layer was separated, and the mixture was taken up in Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:1) to give 25.2mg (74.8%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000303
Compound 7:8- (ethylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione
Figure BDA0002247439760000304
To a solution of Compound 6 (20 mg) in MeOH (2 mL) was added potassium hydrogen persulfate (72.61 mg) H 2 O (2 mL) solution. The mixture was then stirred at room temperature for 2h. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Drying, filtration, concentration and purification by preparative TLC gave compound 6.7 mg (34.5%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000311
To compound 1 (30 mg) and K 2 CO 3 To a solution of (21.98 mg) in anhydrous DMF (2 mL) was added 1-iodopropane (27.07 mg) and stirred under nitrogen at room temperature for 3h. The reaction mixture was poured into water and extracted with ethyl acetate (3 x 5 ml), the organic layer was separated, and the mixture was taken up in Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:1) to give 30.2mg (84.2%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000312
Compound 10:1,3, 7-trimethyl-8- (propylsulfonyl) -1H-purine-2, 6 (3H, 7H) -dione
Figure BDA0002247439760000313
To a solution of Compound 8 (25 mg) in MeOH (2 mL) was added potassium hydrogen persulfate (86.01 mg) H 2 O (2 mL) solution. The mixture was then stirred at room temperature for 3h. The solvent was then removed and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was extracted with Na 2 SO 4 Drying, filtration, concentration and purification by preparative TLC gave 97.9mg (30.1%) of compound as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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 10.2 mg (39.1%) was obtained as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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- (cyclopropylmethylsulfanyl) -1,3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione
Figure BDA0002247439760000321
To compound 1 (30 mg) and K 2 CO 3 To a solution of (21.98 mg) in anhydrous DMF (2 mL) was added (bromomethyl) cyclopropane (21.34 mg) and the mixture was stirred at room temperature under nitrogen for 2.5h. The reaction mixture was poured into water and extracted with ethyl acetate (3 x 5 ml), the organic layer was separated, and the mixture was taken up in Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:1) to give 36.5mg (97.3%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000322
Compound 13:8- (cyclopropylmethylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione
Figure BDA0002247439760000323
To a solution of Compound 11 (23 mg) in MeOH (2 mL) was added potassium hydrogen persulfate (75.7 mg) H 2 O (2 mL) solution. The mixture was then stirred at room temperature for 2h. The solvent was then removed and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was extracted with Na 2 SO 4 Drying, filtration, concentration and purification by preparative TLC gave 12.7 mg (29.5%) of compound as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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]+.
13.4mg (49.7%) of compound 13 are obtained as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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-trifluoroethylsulfinyl) -1H-purine-2, 6 (3H, 7H) -dione
Figure BDA0002247439760000331
Compound 15:1,3, 7-trimethyl-8- (2, 2-trifluoroethylsulfonyl) -1H-purine-2, 6 (3H, 7H) -dione
Figure BDA0002247439760000332
(I) Synthesis of 1,3, 7-trimethyl-8- (2, 2-trifluoroethylthio) -1H-purine-2, 6 (3H, 7H) -dione
To compound 1 (30.0 mg,0.13 mmol) and K 2 CO 3 To a solution of (21.4 mg,0.15 mmol) in anhydrous DMF (5 mL) was added 2-bromo-1, 1-trifluoroethane (25.9 mg,0.15 mmol) and stirred under nitrogen overnight at room temperature. The reaction mixture was then poured into water and extracted with EA (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, 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-trifluoroethylsulfonyl) -1H-purine-2, 6 (3H, 7H) -dione and 1,3, 7-trimethyl-8- (2, 2-trifluoroethylsulfinyl) -1H-purine-2, 6 (3H, 7H) -dione
To a solution of 1,3, 7-trimethyl-8- (2, 2-trifluoroethylthio) -1H-purine-2, 6 (3H, 7H) -dione (10 mg,0.03 mmol) in DCM (2 mL) was added m-CPBA (11.2 mg,0.06 mmol). The mixture was then stirred at room temperature for 2h. The solvent was then removed and dichloromethane was used (3×5 ml) extraction, separation of the organic layer, na 2 SO 4 Drying, filtration, concentration and purification by preparative TLC gave compound 14 as a white solid (3.1 mg, 28.8%). 1 H-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 15.3 mg (30.4%) is obtained as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000341
(I) Synthesis of 8- (2- (2- (2-hydroxyethoxy) ethoxy) ethylthio) -1,3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione
To compound 1 (100 mg,0.44 mmol) and Cs 2 CO 3 To a solution of (172.9 mg,0.53 mmol) in anhydrous DMF (4 mL) was added 2- (2- (2-chloroethoxy) ethoxy) ethanol (89.4 mg,0.53 mmol) and reacted in the microwave over a Biotage Smith synthesizer at 130℃for 1h. The mixture was then cooled to room temperature and acidified to ph=3, extracted with dichloromethane (3×10 ml), the organic layer was separated, and the organic layer was purified over Na 2 SO 4 Drying, filtration and concentration gave 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 (25.0 mg,0.07 mmol) in MeOH (2 mL) was added a solution of potassium hydrogen persulfate (171.7 mg,0.28 mmol) in H2O (2 mL). The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, dried over Na2SO4, filtered, concentrated and purified by preparative TLC to give a white solid (2.3 mg, 8.43%). 1H-NMR (400 MHz, CDCl 3): 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): 391.12[ M+H ] + ].
Compound 17:8- (but-3-ynylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione
Figure BDA0002247439760000342
(I) Synthesis of 8- (but-3-ynylthio) -1,3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione
To compound 1 (50.0 mg,0.22 mmol) and K 2 CO 3 To a solution of (36.6 mg,0.26 mmol) in anhydrous DMF (2 mL) was added 4-bromobut-1-yne (35.3 mg,0.26 mmol) and stirred at room temperature under nitrogen overnight. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Drying, filtration and concentration gave 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 (25.0 mg,0.09 mmol) in MeOH (2 mL) was added potassium hydrogen persulfate (221.1 mg,0.36 mmol) in H 2 O (2 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give a white solid (20.1 mg, 72.3%). 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000351
(I) Synthesis of 8- (isopropylsulfanyl) -1,3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione
To compound 1 (50.0 mg,0.22 mmol) and K 2 CO 3 To a solution of (36.6 mg,0.26 mmol) in anhydrous DMF (5 mL) was added 2-iodopropane (45.1 mg,0.26 mmol) and stirred under nitrogen overnight at room temperature. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA) to give a white solid (34.7 mg, 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 (30.0 mg) in MeOH (2 mL) was added potassium hydrogen persulfate (275.3 mg) H 2 O (2 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA1: 1) to give 31.5mg (93.75%) as a white solid. 1 H-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-ylmethyl sulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione
Figure BDA0002247439760000361
(I) Synthesis of 5- (bromomethyl) benzo [ d ] [1,3] dioxole
To a solution of benzo [ d ] [1,3] dioxol-5-yl-methanol (1 g) in DCM was slowly added tribromophosphine (2.65 g) at 0deg.C. The mixture was then stirred at room temperature overnight. The solvent was removed and extracted with dichloromethane (3 x 15 ml) and concentrated to give a yellow solid (1.2 g, 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 (50.0 mg)0.22 mmol) and K 2 CO 3 To a solution of (36.6 mg,0.26 mmol) in anhydrous DMF (5 mL) was added 5- (bromomethyl) benzo [ d ]][1,3]Dioxolane (57.1 mg,0.26 mmol) was stirred overnight at room temperature under nitrogen. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA1: 1) to give a white solid (23.4 mg, 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]To a solution of dioxol-5-ylmethyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione (50.0 mg) in DCM (5 mL) was added m-CPBA (47.9 mg) and the mixture was stirred at room temperature for 2H. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (1:1) to give 20.3mg (38.4%) as a white solid. 1 H-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- (benzenesulfonyl) -1H-purine-2, 6 (3H, 7H) -dione
Figure BDA0002247439760000362
(I) Synthesis of 1,3, 7-trimethyl-8- (phenylsulfanyl) -1H-purine-2, 6 (3H, 7H) -dione
To 8-chloro-1, 3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione (100.0 mg,0.44 mmol) and Cs 2 CO 3 To a solution of (354.5 mg,1.09 mmol) in anhydrous DMF (5 mL) was added thiophenol (57.9 mg,0.53 mmol) and stirred under nitrogen overnight at room temperature. The reaction mixture was then poured into water and extracted with EA (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give a white solid (54.7 mg, 41.3%). 1 H-NMR(400MHz,CDCl 3 ):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- (benzenesulfonyl) -1H-purine-2, 6 (3H, 7H) -dione
To a solution of 1,3, 7-trimethyl-8- (phenylsulfanyl) -1H-purine-2, 6 (3H, 7H) -dione (28.0 mg,0.09 mmol) in MeOH (5 mL) was added potassium hydrogen persulfate (227.9 mg,0.37 mmol) in H 2 O (5 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was extracted with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give a white solid (14.3 mg, 46.2%). 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000371
(I) Synthesis of 8- (benzylthio) -1,3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione
To compound 1 (70.0 mg) and K 2 CO 3 To a solution of (52.1 mg) in anhydrous DMF (5 mL) was added (bromomethyl) benzene (63.5 mg) and stirred under nitrogen overnight at room temperature. The reaction mixture was then poured into water and extracted with EA (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Drying, 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- (benzylthio) -1,3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione (50.0 mg) in MeOH (2 mL) was added potassium hydrogen persulfate (388.96 mg) H 2 O (2 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:1) to give 28.4mg (51.6%) as a white solid. 1 H-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
Figure BDA0002247439760000381
(I) Synthesis of 1,3, 7-trimethyl-8- (phenethylthio) -1H-purine-2, 6 (3H, 7H) -dione
To compound 1 (70.0 mg) and K 2 CO 3 To a solution of (52.8 mg) in anhydrous DMF (5 mL) was added (2-bromoethyl) benzene (68.95 mg) and stirred at room temperature under nitrogen overnight. The reaction mixture was then poured into water and extracted with EA (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Drying, filtering and concentrating to obtain yellow oily matter.
(II) Synthesis of 1,3, 7-trimethyl-8- (phenethylsulfonyl) -1H-purine-2, 6 (3H, 7H) -dione
To a solution of 1,3, 7-trimethyl-8- (phenylethylsulfanyl) -1H-purine-2, 6 (3H, 7H) -dione (20 mg) in MeOH (1 mL) was added potassium hydrogen persulfate (149.1 mg) H 2 O (1 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA1: 1) to give 6.3mg (28.77%) as a white solid. 1 H-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-ylmethyl sulfonyl) -1H-purine-2, 6 (3H, 7H) -dione
Figure BDA0002247439760000382
(I) Synthesis of 1,3, 7-trimethyl-8- ((naphthalen-2-ylmethyl) thio) -1H-purine-2, 6 (3H, 7H) -dione
Orientation methodCompound 1 (50 mg) and K 2 CO 3 To a solution of (36.6 mg) in anhydrous DMF (5 mL) was added 2- (bromomethyl) naphthalene (58.7 mg) and stirred at room temperature under nitrogen overnight. The reaction mixture was then poured into water and extracted with EA (3 x 5 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA1: 1) to give 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 (10 mg) in MeOH (1 mL) was added potassium hydrogen persulfate (67.2 mg) H 2 O (1 mL) solution. The mixture was then stirred at room temperature overnight. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:2) to give 8.2mg (75.5%) as a white solid. 1 H-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-ylmethyl sulfonyl) -1H-purine-2, 6 (3H, 7H) -dione
Figure BDA0002247439760000391
(I) Synthesis of 1,3, 7-trimethyl-8- ((naphthalen-1-ylmethyl) thio) -1H-purine-2, 6 (3H, 7H) -dione
To compound 1 (50 mg) and K 2 CO 3 To a solution of (36.6 mg) in anhydrous DMF (5 mL) was added 1- (bromomethyl) naphthalene (58.7 mg) and stirred at room temperature under nitrogen overnight. The reaction mixture was then poured into water and extracted with EA (3 x 5 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:1) to give 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 (10 mg) in MeOH (1 mL) was added potassium hydrogen persulfate (67.2 mg) H 2 O (1 mL) solution. The mixture was then stirred at room temperature overnight. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:2) to give 5.2mg (48.2%) as a white solid. 1 H-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
Figure BDA0002247439760000401
To compound 17 (10.0 mg,0.03 mmol), N- (2- (2- (2- (2-azidoethoxy) ethoxy) ethyl) -5- ((4S) -2-oxohexahydro-1H-thieno [3, 4-d)]To a solution of imidazol-4-yl) valeramide (17.2 mg,0.04 mmol) and CuI (0.31 mg,0.002 mmol) in dry THF (1 mL) was added TEA (6.6 mg,0.06 mmol) and stirred under nitrogen at room temperature for 4h. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 3 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative HPLC to give a white solid (4.3 mg, 14.7%). 1 H-NMR(400MHz,DMSO-d 6 ):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
Figure BDA0002247439760000402
(I) Synthesis of 8- ((2-fluorobenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione
To compound 1 (50 mg) and K 2 CO 3 To a solution of (36.6 mg) in anhydrous DMF (5 mL) was added 1- (bromomethyl) -2-fluorobenzene (50.2 mg) and stirred under nitrogen overnight at room temperature. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:1) to give a white solid (24.8 mg, 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 (10.0 mg) in MeOH (3 mL) was added potassium hydrogen persulfate (73.6 mg) H 2 O (3 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give 6.3mg (57.5%) as a white solid. 1 H-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
Figure BDA0002247439760000411
(I) Synthesis of 8- ((3-fluorobenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione
To compound 1 (50 mg) and K 2 CO 3 To a solution of (36.6 mg) in anhydrous DMF (5 mL) was added 1- (bromomethyl) -3-fluorobenzene (50.2 mg) and stirred under nitrogen overnight at room temperature. Then the reaction is carried outThe mixture was poured into water and extracted with dichloromethane (3 x 10 ml) and the organic layer was separated using Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:1) to give a white solid (27.4 mg, 37.1%).
(II) Synthesis of 8- ((3-fluorobenzyl) sulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione
To a solution of 8- ((3-fluorobenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione (10.0 mg) in MeOH (3 mL) was added potassium hydrogen persulfate (73.6 mg) H 2 O (3 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give 7.1mg (64.78%) as a white solid. 1 H-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
Figure BDA0002247439760000412
(I) Synthesis of 8- ((4-fluorobenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione
To compound 1 (50 mg) and K 2 CO 3 To a solution of (36.6 mg) in anhydrous DMF (5 mL) was added 1- (bromomethyl) -4-fluorobenzene (50.2 mg) and stirred under nitrogen overnight at room temperature. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:1) to give a white solid (29.3 mg, 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 (10.0 mg) in MeOH (3 mL) was added potassium hydrogen persulfate (73.6 mg) H 2 O (3 mL) solution. Then stirring and mixing at room temperatureThe reaction time was 5h. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give 8.4mg (76.64%) as a white solid. 1 H-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-chlorobenzyl sulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione
Figure BDA0002247439760000421
(I) Synthesis of 8- (2-chlorobenzylthio) -1,3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione
To compound 1 (40.0 mg,0.17 mmol) and K 2 CO 3 To a solution of (29.3 mg,0.21 mmol) in anhydrous DMF (2 mL) was added 1- (bromomethyl) -2-chlorobenzene (43.6 mg,0.21 mmol) and stirred under nitrogen overnight at room temperature. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give a white solid (18.4 mg, 30.2%). 1 H-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 (15.0 mg,0.04 mmol) in MeOH (3 mL) was added potassium hydrogen persulfate (105.2 mg,0.17 mmol) in H 2 O (3 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give a white solid (11.2 mg, 68.3%). 1 H-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-chlorobenzyl sulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione
Figure BDA0002247439760000431
(I) Synthesis of 8- ((3-chlorobenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione
To compound 1 (40.0 mg,0.17 mmol) and K 2 CO 3 To a solution of (29.3 mg,0.21 mmol) in anhydrous DMF (2 mL) was added 1- (bromomethyl) -3-chlorobenzene (43.6 mg,0.21 mmol) and stirred under nitrogen overnight at room temperature. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA1: 1) to give a white solid (29.3 mg, 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 (15.0 mg,0.04 mmol) in MeOH (5 mL) was added potassium hydrogen persulfate (140.2 mg) 2 O (5 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give 15.4mg (70.65%) as a white solid. 1 H-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-chlorobenzyl sulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione
Figure BDA0002247439760000432
(I) Synthesis of 8- ((4-chlorobenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione
To compound 1 (40.0 mg,0.17 mmol) and K 2 CO 3 To a solution of (29.3 mg,0.21 mmol) in anhydrous DMF (2 mL) was added 1- (bromomethyl) -4-chlorobenzene (43.6 mg,0.21 mmol) and stirred under nitrogen overnight at room temperature. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:1) to give a white solid (28.3 mg, 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 (20.0 mg,0.04 mmol) in MeOH (5 mL) was added potassium hydrogen persulfate (140.2 mg) 2 O (5 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give 16.1mg (73.85%) as a white solid. 1 H-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
Figure BDA0002247439760000441
(I) Synthesis of 8- ((2-bromobenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione
To compound 1 (50 mg) and K 2 CO 3 To a solution of (36.6 mg) in anhydrous DMF (5 mL) was added 1-bromo-2- (bromomethyl) benzene (66.1 mg) and the mixture was stirred at room temperature under nitrogen overnight. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:1) to give a white solid (30.1 mg, 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 (10.0 mg) in MeOH (3 mL) was added potassium hydrogen persulfate (62.2 mg) H 2 O (3 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give 8.3mg (76.85%) as a white solid. 1 H-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
Figure BDA0002247439760000451
(I) Synthesis of 8- ((3-bromobenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione
To compound 1 (50 mg) and K 2 CO 3 To a solution of (36.6 mg) in anhydrous DMF (5 mL) was added 1-bromo-3- (bromomethyl) benzene (66.1 mg) and the mixture was stirred at room temperature under nitrogen overnight. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA1: 1) to give a white solid (34.5 mg, 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 (10.0 mg) in MeOH (3 mL) was added potassium hydrogen persulfate (62.2 mg) H 2 O (3 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give 5.6mg (51.99%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000452
(I) Synthesis of 8- ((4-bromobenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione
To compound 1 (50 mg) and K 2 CO 3 To a solution of (36.6 mg) in anhydrous DMF (5 mL) was added 1-bromo-4- (bromomethyl) benzene (66.1 mg) and stirred under nitrogen overnight at room temperature. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:1) to give a white solid (30.1 mg, 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 (10.0 mg) in MeOH (3 mL) was added potassium hydrogen persulfate (62.2 mg) H 2 O (3 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give 7.2mg (66.7%) as a white solid. 1 H-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
Figure BDA0002247439760000461
(I) Synthesis of 8- ((2-methoxybenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione
To compound 1 (50 mg) and K 2 CO 3 To a solution of (36.6 mg) in anhydrous DMF (5 mL) was added 1- (chloromethyl) -2-methoxybenzene (41.6 mg) and stirred under nitrogen overnight at room temperature. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:1) to give a white solid (30.1 mg, 39.2%).
(II) Synthesis of 8- ((2-methoxybenzyl) sulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione
To a solution of 8- ((2-methoxybenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione (30 mg) in MeOH (2 mL) was added potassium hydrogen persulfate (212.6 mg) H 2 O (2 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give 18.2mg (56.87%) as a white solid. 1 H-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
Figure BDA0002247439760000471
(I) Synthesis of 8- ((3-methoxybenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione
To compound 1 (50 mg) and K 2 CO 3 To a solution of (36.6 mg) in anhydrous DMF (5 mL) was added 1- (bromomethyl) -2-methoxybenzene (53.4 mg) and stirred under nitrogen overnight at room temperature. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA1: 1) to give a white solid (40.4 mg, 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 (40 mg) in MeOH (3 mL) was added potassium hydrogen persulfate (283.5 mg) H 2 O (3 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give 20.4mg (46.68%) as a white solid. 1 H-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
Figure BDA0002247439760000472
(I) Synthesis of 8- ((4-methoxybenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione
To compound 1 (50 mg) and K 2 CO 3 To a solution of (36.6 mg) in anhydrous DMF (5 mL) was added 1- (chloromethyl) -4-methoxybenzene (41.6 mg) and stirred under nitrogen overnight at room temperature. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:1) to give a white solid (23.4 mg, 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 (20 mg) in MeOH (2 mL) was added potassium hydrogen persulfate (141.7 mg) H 2 O (2 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Drying, filtering and concentratingThe condensation was purified by preparative TLC to give 14.1mg (64.7%) as a white solid. 1 H-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-nitrobenzenesulfonyl) -1H-purine-2, 6 (3H, 7H) -dione
Figure BDA0002247439760000481
(I) Synthesis of 1,3, 7-trimethyl-8- ((2-nitrobenzyl) thio) -1H-purine-2, 6 (3H, 7H) -dione
To compound 1 (30 mg) and K 2 CO 3 To a solution of (21.4 mg) in anhydrous DMF (5 mL) was added 1- (bromomethyl) -2-nitrobenzene (34.3 mg) and stirred at room temperature under nitrogen overnight. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:1) to give a white solid (16.8 mg, 35%).
Synthesis of (II) 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 (15 mg) in MeOH (3 mL) was added potassium hydrogen persulfate (102.1 mg) H 2 O (3 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give 8.3mg (50.92%) as a white solid. 1 H-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
Figure BDA0002247439760000491
(I) Synthesis of 1- (4- (bromomethyl) phenyl) -1H-pyrrole
To a solution of (4- (1H-pyrrol-1-yl) phenyl) methanol (100 mg) in DCM was slowly added tribromophosphine (232 mg) at 0deg.C. The mixture was then stirred at room temperature overnight. The solvent was removed and extracted with dichloromethane (3×5 ml) and concentrated to give a yellow solid (63 mg, 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 (50.0 mg,0.22 mmol) and K 2 CO 3 To a solution of (36.6 mg,0.26 mmol) in anhydrous DMF (5 mL) was added 1- (4- (bromomethyl) phenyl) -1H-pyrrole (62.6 mg,0.26 mmol) and stirred under nitrogen overnight at room temperature. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:1) to give a white solid (10.8 mg, 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 (10 mg) in MeOH (3 mL) was added potassium hydrogen persulfate (64.5 mg) H 2 O (3 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:1) to give 3.8mg (32.5%) as a white solid. 1 H-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-diazepin-3-yl) benzylsulfonyl) -1H-purine-2, 6 (3H, 7H) -dione
Figure BDA0002247439760000492
(I) Synthesis of 1,3, 7-trimethyl-8- (3- (3- (trifluoromethyl) -3H-diazepin-3-yl) benzylthio) -1H-purine-2, 6 (3H, 7H) -dione
To compound 1 (50.0 mg,0.22 mmol) and K 2 CO 3 To a solution of (37.8 mg,0.27 mmol) in anhydrous DMF (3 mL) was added 3- (3- (bromomethyl) phenyl) -3- (trifluoromethyl) -3H-diazepine (73.8 mg,0.27 mmol) and stirred overnight at room temperature under nitrogen in the absence of light. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by silica gel chromatography (PE: ea=4:1 to DCM: meoh=10:1) to give a white solid (12.3 mg, 13.1%). 1 H-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-diazepin-3-yl) benzylsulfonyl) -1H-purine-2, 6 (3H, 7H) -dione
To a solution of 1,3, 7-trimethyl-8- (3- (3- (trifluoromethyl) -3H-diazepin-3-yl) benzylthio) -1H-purine-2, 6 (3H, 7H) -dione (10 mg,0.02 mmol) in DCM (3 mL) was added m-CPBA (6.2 mg,0.03 mmol). The mixture was then stirred at room temperature in the dark for 2h. The solvent was then removed and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was extracted with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give a white solid (5.7 mg, 53.4%). 1 H-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-diazepin-3-yl) benzylsulfonyl) -1H-purine-2, 6 (3H, 7H) -dione
Figure BDA0002247439760000501
The compound was synthesized in a similar manner to the compound 40 to give 8.4mg (77.7%) As a white solid. 1 H-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
Figure BDA0002247439760000502
(I) Synthesis of 2- ((1, 3, 7-trimethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-8-ylsulfanyl) methyl) benzonitrile
To compound 1 (100.0 mg,0.44 mmol) and K 2 CO 3 To a solution of (73.2 mg,0.53 mmol) in anhydrous DMF (5 mL) was added 2- (bromomethyl) benzonitrile (103.1 mg,0.53 mmol) and stirred under nitrogen overnight at room temperature. The reaction mixture was then poured into water and extracted with EA (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give a white solid (80.9 mg, 53.62%). 1 H-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-ylsulfanyl) methyl) benzonitrile (15.0 mg,0.04 mmol) in MeOH (2 mL) was added a solution of potassium hydrogen persulfate (108.2 mg,0.17 mmol) in H2O (5 mL). The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give a white solid (7.2 mg, 43.91%). 1H-NMR (400 MHz, CDCl 3): 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
Figure BDA0002247439760000511
(I) Synthesis of 3- ((1, 3, 7-trimethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-8-ylsulfanyl) methyl) benzonitrile
To compound 1 (100.0 mg,0.44 mmol) and K 2 CO 3 To a solution of (73.2 mg,0.53 mmol) in anhydrous DMF (5 mL) was added 3- (bromomethyl) benzonitrile (103.1 mg,0.53 mmol) and stirred under nitrogen overnight at room temperature. The reaction mixture was then poured into water and extracted with EA (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give a white solid (68.3 mg, 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-ylsulfanyl) methyl) benzonitrile (15.0 mg,0.04 mmol) in MeOH (2 mL) was added potassium hydrogen persulfate (108.2 mg,0.17 mmol) H 2 O (5 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give 10.7mg (65.3%) as a white solid. 1 H-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
Figure BDA0002247439760000521
(I) Synthesis of 4- ((1, 3, 7-trimethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-8-ylsulfanyl) methyl) benzonitrile
To compound 1 (50 mg,0.22 mmol) and K 2 CO 3 To a solution of (37.8 mg) of anhydrous DMF (2 mL) was added 4- (bromomethyl) benzonitrile [ (]51.2 mg) and stirred at room temperature under nitrogen overnight. The reaction mixture was then poured into water and extracted with EA (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:1) to give a white solid (23 mg, 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-ylsulfanyl) methyl) benzonitrile (15.0 mg,0.04 mmol) in MeOH (2 mL) was added a solution of potassium hydrogen persulfate (108.4 mg,0.17 mmol) in H2O (5 mL). The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:2) to give 11.0mg (67.1%) as a white solid. 1H-NMR (400 MHz, CDCl 3): 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): 374.08[ M+H ] ]+
Compound 45:2- ((1, 3, 7-trimethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-8-ylsulfonyl) methyl) benzamide
Figure BDA0002247439760000531
Compound 2- ((1, 3, 7-trimethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-8-ylsulfanyl) methyl) benzonitrile was synthesized in the manner described in compound 42.
(I) Synthesis of 2- ((1, 3, 7-trimethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-8-ylsulfanyl) methyl) benzamide
To 2- ((1, 3, 7-trimethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-8-ylsulfanyl) methyl) benzonitrile (15.0 mg,0.04 mmol) and K 2 CO 3 To a solution of (0.85 mg, 0.006mmol) in DMSO (2 mL) was slowly added H 2 O 2 (2.3 mg,0.06 mmol) and stirred at room temperature under nitrogen overnight. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Drying, filtration and concentration gave 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-ylsulfanyl) methyl) benzamide (10.0 mg,0.03 mmol) in MeOH (2 mL) was added potassium hydrogen persulfate (68.5 mg,0.11 mmol) H 2 O (2 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give a white solid (8.7 mg, 80.55%). 1 H-NMR(400MHz,DMSO-d 6 ):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
Figure BDA0002247439760000532
This compound was synthesized in a similar manner to compound 45, and finally obtained as 7.3mg (67.6%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000541
This compound was synthesized in a similar manner to compound 45, and finally obtained as 10.3mg (95.4%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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-ylmethyl sulfonyl) -1H-purine-2, 6 (3H, 7H) -dione
Figure BDA0002247439760000542
Compound 49:2- ((1, 3, 7-trimethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-8-ylsulfonyl) methyl) pyridine 1-oxide
Figure BDA0002247439760000543
(I) Synthesis of 1,3, 7-trimethyl-8- (pyridin-2-ylmethylsulfanyl) -1H-purine-2, 6 (3H, 7H) -dione
To compound 1 (500.0 mg,2.21 mmol) and K 2 CO 3 To a solution of (366.4 mg,2.65 mmol) in anhydrous DMF (25 mL) was added 2- (bromomethyl) pyridine (674.1 mg,2.66 mmol) and stirred under nitrogen overnight at room temperature. The reaction mixture was then poured into water and extracted with EA (3 x 50 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Drying, 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-ylmethylsulfanyl) -1H-purine-2, 6 (3H, 7H) -dione (50.0 mg,0.16 mmol) in MeOH (3 mL) was added potassium hydrogen persulfate (387.9 mg,0.64 mmol) H 2 O (3 mL) solution. The mixture was then stirred at room temperature for 30min. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Drying, filtration, concentration and purification by preparative TLC gave compound 48 as a white solid (14.2 mg, 25.5%). 1 H-NMR(400MHz,CDCl 3 ):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 49.3 mg (75.4%) as white solid was obtained. 1 H-NMR(400MHz,CDCl 3 ):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-ylmethyl sulfonyl) -1H-purine-2, 6 (3H, 7H) -dione
Figure BDA0002247439760000551
Compound 51:3- ((1, 3, 7-trimethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-8-ylsulfonyl) methyl) pyridine 1-oxide
Figure BDA0002247439760000552
(I) Synthesis of 1,3, 7-trimethyl-8- (pyridin-3-ylmethylsulfanyl) -1H-purine-2, 6 (3H, 7H) -dione
To compound 1 (30 mg) and K 2 CO 3 To a solution of (21.4 mg) in anhydrous DMF (25 mL) was added 3- (bromomethyl) pyridine (40.3 mg) and stirred at room temperature under nitrogen overnight. The reaction mixture was then poured into water and extracted with EA (3 x 50 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (DCM: meOH 10:1) to give a white solid (16.2 mg, 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-ylmethylsulfanyl) -1H-purine-2, 6 (3H, 7H) -dione (10 mg) in MeOH (2 mL) was added potassium hydrogen persulfate (77.6 mg) H 2 O (2 mL) solution. The mixture was then stirred at room temperature for 30min. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (DCM: meOH 10:1) to give 504.3mg (37.3%) of compound as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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]+.
3.7mg (35.7%) of compound 51 were obtained as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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-ylmethyl sulfonyl) -1H-purine-2, 6 (3H, 7H) -dione
Figure BDA0002247439760000561
Compound 53:4- ((1, 3, 7-trimethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-8-ylsulfonyl) methyl) pyridine 1-oxide
Figure BDA0002247439760000562
/>
(I) Synthesis of 1,3, 7-trimethyl-8- (pyridin-4-ylmethylsulfanyl) -1H-purine-2, 6 (3H, 7H) -dione
To compound 1 (30 mg) and K 2 CO 3 To a solution of (21.4 mg) in anhydrous DMF (25 mL) was added 3- (bromomethyl) pyridine (40.3 mg) and stirred at room temperature under nitrogen overnight. The reaction mixture was then poured into water and extracted with EA (3 x 50 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (DCM: meOH 10:1) to give a white solid (15 mg, 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 1,3, 7-trimethyl-8- (pyridin-3-ylmethylsulfanyl) -1H-purine-2, 6To a solution of (3H, 7H) -dione (10 mg) in MeOH (2 mL) was added potassium hydrogen persulfate (77.6 mg) in H 2 O (2 mL) solution. The mixture was then stirred at room temperature for 30min. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (DCM: meOH 10:1) to give 524.2mg (35.2%) of compound as a white solid. 1H-NMR (400 MHz, CDCl 3): delta 8.65 (d, J=4.8 Hz, 2H), 7.27 (d, J=5.2 Hz, 2H), 4.74 (s, 2H), 4.02 (s, 3H), 3.61 (s, 3H), 3.40 (s, 3H): 350.08[ M+H ] ]+.
Compound 53.2 mg (30.4%) as white solid was obtained. 1H-NMR (400 MHz, CDCl 3): delta 8.20 (d, J=6.8 Hz, 2H), 7.31 (d, J=6.8 Hz, 2H), 4.76 (s, 2H), 4.19 (s, 3H), 3.59 (s, 3H), 3.41 (s, 3H): 366.08[ M+H ] +.
Compound 54:8- (2, 3-dichlorobenzylsulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione
Figure BDA0002247439760000571
(I) Synthesis of 8- (2, 3-dichlorobenzylthio) -1,3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione
To compound 1 (50.0 mg,0.22 mmol) and K 2 CO 3 To a solution of (36.6 mg,0.26 mmol) in anhydrous DMF (5 mL) was added 1- (bromomethyl) -2, 3-dichlorobenzene (63.7 mg,0.26 mmol) and stirred under nitrogen overnight at room temperature. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, 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 (10.0 mg,0.03 mmol) in MeOH (3 mL) was added potassium hydrogen persulfate (63.9 mg,0.10 mmol) 2 O (3 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give a white solid (4.2 mg, 38.9%). 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000572
(I) Synthesis of 8- (2, 5-dichlorobenzylthio) -1,3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione
To compound 1 (50.0 mg,0.22 mmol) and K 2 CO 3 To a solution of (36.6 mg,0.26 mmol) in anhydrous DMF (5 mL) was added 1- (bromomethyl) -2, 5-dichlorobenzene (63.7 mg,0.26 mmol) and stirred under nitrogen overnight at room temperature. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:1) to give a white solid (20.1 mg, 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 (10.0 mg,0.03 mmol) in MeOH (3 mL) was added potassium hydrogen persulfate (63.9 mg,0.10 mmol) 2 O (3 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:1) to give 8.3mg (76.85%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000581
(I) Synthesis of 8- (3, 5-dichlorobenzylthio) -1,3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione
To compound 1 (50.0 mg,0.22 mmol) and K 2 CO 3 To a solution of (36.6 mg,0.26 mmol) in anhydrous DMF (5 mL) was added 1, 3-dichloro-5- (chloromethyl) benzene (51.8 mg,0.26 mmol) and stirred under nitrogen overnight at room temperature. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:1) to give a white solid (25.3 mg, 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 (20 mg) in MeOH (5 mL) was added potassium hydrogen persulfate (127.7 mg) 2 O (5 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:1) to give 11.3mg (52.3%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000591
(I) Synthesis of 8- (2, 6-dichlorobenzylthio) -1,3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione
To compound 1 (50.0 mg,0.22 mmol) and K 2 CO 3 To a solution of (36.6 mg,0.26 mmol) in anhydrous DMF (5 mL) was added 2- (bromomethyl) -1, 3-dichlorobenzene (63.7 mg,0.26 mmol) and stirred under nitrogen overnight at room temperature. ThenThe reaction mixture was poured into water and extracted with dichloromethane (3 x 10 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:1) to give a white solid (20.1 mg, 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 (10 mg) in MeOH (3 mL) was added potassium hydrogen persulfate (63.9 mg) H 2 O (3 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:1) to give 7.9mg (73.1%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000592
(I) Synthesis of 8- ((2, 5-dimethoxybenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione
To compound 1 (50.0 mg) and K 2 CO 3 To a solution of (36.6 mg) in anhydrous DMF (5 mL) was added 2- (bromomethyl) -1, 4-dimethoxybenzene (61.4 mg) and stirred under nitrogen overnight at room temperature. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:1) to give a white solid (26.4 mg, 31.8%).
(II) Synthesis of 8- ((2, 5-dimethoxybenzyl) sulfonyl) -1,3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione
To 8- (2, 6-dichlorobenzylthio) -1,3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione (10 mg) in MeOH (5 mL)H of potassium hydrogen persulfate (65.4 mg) was added to the solution 2 O (5 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:1) to give 8.1mg (75%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000601
This compound was synthesized in a similar manner to compound 58, and finally 8.2mg (75.1%) was obtained as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000602
(I) Synthesis of 4-bromo-1- (bromomethyl) -2-methoxybenzene
To a DME solution of (4-bromo-2-methoxyphenyl) methanol (100 mg) at 0deg.C was slowly added tribromophosphine (124.7 mg). The mixture was then stirred at room temperature overnight. The solvent was removed and extracted with EA (3 x 10 ml) and concentrated to give a white solid (136.3 mg, 72%).
(II) Synthesis of 8- ((4-bromo-2-methoxybenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione
To compound 1 (50.0 mg,0.22 mmol) and K 2 CO 3 To a solution of (36.6 mg,0.26 mmol) in anhydrous DMF (5 mL) was added 4-bromo-1- (bromomethyl) -2-methoxybenzene (75 mg,0.26 mmol) and under nitrogenStir at room temperature overnight. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by column chromatography (PE: EA 1:1) to give a white solid (35 mg, 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 (40 mg) in MeOH (5 mL) was added potassium hydrogen persulfate (650 mg) H 2 O (5 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with EA (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by silica gel column chromatography to give 10mg (31%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000611
/>
(I) Synthesis of 1- (bromomethyl) -2, 3-dimethoxybenzene
To a DME solution of (2, 3-dimethoxyphenyl) methanol (500 mg) was slowly added tribromophosphine (805 mg) at 0deg.C. The mixture was then stirred at room temperature overnight. The solvent was removed and extracted with EA (3 x 10 ml) and concentrated to give a white solid (176.8 mg, 26%).
(II) Synthesis of 8- ((2, 3-dimethoxybenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione
To compound 1 (50.0 mg,0.22 mmol) and K 2 CO 3 To a solution of (62 mg,0.44 mmol) in anhydrous DMF (5 mL) was added 1- (bromomethyl) -2, 3-dimethoxybenzene (77 mg,0.33 mmol) and stirred overnight at room temperature under nitrogen. The reaction mixture was then poured into water and taken up in dichloromethane3 x 10 ml) and separating the organic layer with Na 2 SO 4 Dried, filtered, concentrated and purified by column chromatography (PE: EA 1:1) to give a white solid (17 mg, 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 (17 mg,0.045 mmol) in MeOH (1 mL) was added potassium hydrogen persulfate (280 mg,0.45 mmol) 2 O (1 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with EA (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by silica gel column chromatography (PE: EA 1:1) to give 2mg (10.9%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000621
(I) Synthesis of 2- (bromomethyl) -1, 3-dimethoxybenzene
To CCl of 1, 3-dimethoxy-2-methylbenzene (500 mg,3.29 mmol) 4 To the solution (10 mL) was slowly added NBS (585 mg,3.29 mmol) and AIBN (150 mg). 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 x 10 ml) 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 (50.0 mg,0.22 mmol) and K 2 CO 3 To a solution of (62 mg,0.44 mmol) in anhydrous DMF (5 mL) was added 2- (bromomethyl) -1, 3-dimethoxybenzene (77 mg,0.33 mmol) and under nitrogenStir overnight at room temperature under air. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by column chromatography (PE: EA 1:1) to give a white solid (7 mg, 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 (7 mg,0.019 mmol) in MeOH (1 mL) was added potassium hydrogen persulfate (120 mg,0.2 mmol) in H 2 O (1 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with EA (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by silica gel column chromatography (PE: EA1: 1) to give 2mg (26.3%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000631
(I) Synthesis of 1- (bromomethyl) -3-chloro-2-methoxybenzene
To CCl of 1-chloro-2-methoxy-3-methylbenzene (300 mg,1.92 mmol) 4 To the solution (10 mL) was slowly added NBS (350 mg,1.92 mmol) and AIBN (150 mg). 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 x 10 ml) 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 (50.0 mg,0.22 mmol) and K 2 CO 3 (62 mg,0.44 mmol) in anhydrous DMF (5 mL)1- (bromomethyl) -3-chloro-2-methoxybenzene (80 mg,0.33 mmol) was added to the solution and stirred overnight at room temperature under nitrogen. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by column chromatography (PE: EA 1:1) to give a white solid (18 mg, 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 (18 mg,0.048 mmol) in MeOH (1 mL) was added potassium hydrogen persulfate (300 mg,0.48 mmol) 2 O (1 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with EA (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by silica gel column chromatography (PE: EA 1:1) to give 12mg (61.5%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000641
(I) Synthesis of 1-bromo-4- (bromomethyl) -2-chlorobenzene
To CCl of 1-bromo-2-chloro-4-methylbenzene (500 mg,2.43 mmol) 4 To the solution (10 mL) was slowly added NBS (433 mg,2.43 mmol) and AIBN (200 mg). 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 x 10 ml) and concentrated to give the crude product (yellow solid) 930mg.
(II) Synthesis of 8- ((4-bromo-3-chlorobenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione
To compound 1 (50.0 mg,0.22 mmol) and K 2 CO 3 To a solution of (62 mg,0.44 mmol) in anhydrous DMF (5 mL) was added 1-bromo-4- (bromomethyl) -2-chlorobenzene (95 mg,0.33 mmol) and stirred under nitrogen overnight at room temperature. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by column chromatography (PE: EA 1:1) to give a white solid (20.5 mg, 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 (20 mg,0.048 mmol) in MeOH (1 mL) was added potassium hydrogen persulfate (290 mg,0.48 mmol) 2 O (1 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with EA (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by silica gel column chromatography (PE: EA 1:1) to give 10mg (46.5%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000651
(I) Synthesis of 2-ethoxy-5-methoxybenzaldehyde
To 2-hydroxy-5-methoxybenzaldehyde (200 mg,1.31 mmol) and K 2 CO 3 To a solution of (220 mg,1.6 mmol) in anhydrous DMF (5 mL) was added ethyl iodide (310 mg,1.97 mmol) and stirred under nitrogen overnight at room temperature. The reaction mixture was then poured into water and extracted with EA (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Drying, filtration and concentration gave a yellow solid (325.1 mg).
(II) Synthesis of 2-ethoxy-5-methoxyphenyl) methanol
At 0℃under N 2 To a solution of 2-ethoxy-5-methoxybenzaldehyde (250 mg,1.39 mmol) in MeOH (5 mL) was added NaBH under protection 4 . The mixture was then stirred at room temperature for 4h. The solvent was removed and extracted with EA (3 x 10 ml) and concentrated to give a yellow solid (252.1 mg).
(III) Synthesis of 2- (bromomethyl) -1-ethoxy-4-methoxybenzene
To a DME solution of 2-ethoxy-5-methoxyphenyl) methanol (252 mg,1.38 mmol) was slowly added tribromophosphine (380 mg,1.38 mmol) at 0deg.C. The mixture was then stirred at room temperature overnight. The solvent was removed and extracted with EA (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by column chromatography (PE: EA 5:1) to give a white solid (336 mg, 99%).
(IV) Synthesis of 8- ((2-ethoxy-5-methoxybenzyl) thio) -1,3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione
To compound 1 (30.0 mg,0.133 mmol) and K 2 CO 3 To a solution of (36 mg,0.265 mmol) in anhydrous DMF (2 mL) was added 2- (bromomethyl) -1-ethoxy-4-methoxybenzene (48 mg,0.2 mmol) and stirred under nitrogen overnight at room temperature. The reaction mixture was then poured into water and extracted with EA (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by column chromatography (PE: EA 5:1) to give a white solid (5 mg, 9.8%).
Synthesis of (V) 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 (5 mg,0.013 mmol) in MeOH (1 mL) was added potassium hydrogen persulfate (80 mg,0.13 mmol) 2 O (1 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with EA (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by silica gel column chromatography (PE: EA1: 1) to give 2.3mg (42.6%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000661
This compound was synthesized in a similar manner to compound 65 to yield 2mg (22.7%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000662
(I) Synthesis of 4-ethoxy-1-methoxy-2-methylbenzene
To 4-methoxy-3-methylphenol (200 mg,1.45 mmol) and K 2 CO 3 To a solution of (250 mg,1.74 mmol) in anhydrous DMF (5 mL) was added ethyl iodide (340 mg,2.17 mmol) and stirred under nitrogen at room temperature for 5h. The reaction mixture was then poured into water and extracted with EA (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by silica gel column chromatography (PE: EA 5:1) to give a yellow solid (86 mg, 35.8%).
(II) Synthesis of 2- (bromomethyl) -4-ethoxy-1-methoxybenzene
To CCl of 4-ethoxy-1-methoxy-2-methylbenzene (80 mg,0.48 mmol) 4 To the (2 mL) solution were slowly added NBS (86 mg,0.48 mmol) and AIBN (60 mg). 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 residueAnd the remainder. The residue was diluted and extracted with EA (3 x 10 ml) 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 (30.0 mg,0.133 mmol) and K 2 CO 3 To a solution of (36 mg,0.265 mmol) in anhydrous DMF (2 mL) was added 2- (bromomethyl) -4-ethoxy-1-methoxybenzene (48 mg,0.2 mmol) and stirred under nitrogen overnight at room temperature. The reaction mixture was then poured into water and extracted with EA (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by column chromatography (PE: EA 5:1) 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 (30 mg,0.077 mmol) in DCM (1 mL) was added m-CPBA (15 mg,0.077 mmol). The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with EA (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by silica gel column chromatography (PE: EA 1:1) to give 29mg (90.6%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000671
This compound was synthesized in a similar manner to compound 67 to yield 23.1mg (72.2%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000681
(I) Synthesis of 1-bromo-3- (bromomethyl) -2, 4-dimethoxybenzene
To CCl of 1, 3-dimethoxy-2-methylbenzene (500 mg,3.29 mmol) 4 To the solution (10 mL) was slowly added NBS (585 mg,3.29 mmol) and AIBN (150 mg). The mixture was then heated to 90 ℃ for 5h. The mixture was filtered and the filtrate was concentrated to give a residue. The residue was diluted and extracted with EA (3 x 10 ml), concentrated and purified by column chromatography (PE: EA 5:1) to give a yellow solid (641 mg, 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 (50.0 mg,0.22 mmol) and K 2 CO 3 To a solution of (62 mg,0.44 mmol) in anhydrous DMF (5 mL) was added 1-bromo-3- (bromomethyl) -2, 4-dimethoxybenzene (80 mg,0.33 mmol) and stirred under nitrogen overnight at room temperature. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by column chromatography (PE: EA1: 1) to give a white solid (67.5 mg, 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 (30 mg,0.08 mmol) in MeOH (1 mL) was added potassium hydrogen persulfate (65 mg,0.8 mmol) H 2 O (1 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with EA (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Drying, filtering, concentrating and passing through silica gel columnChromatography (PE: EA 1:1) gave 15mg (46.1%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000691
(I) Synthesis of 4-chloro-2, 5-dimethoxybenzaldehyde
To 2-chloro-1, 4-dimethoxybenzene (2.5 g,14.48 mmol) and hexamethylenetetramine (2.05 g,14.63 mmol) were carefully added TFA (25 mL). The mixture was then heated to 95 ℃ and held overnight. The mixture was then poured into ice and extracted with NaHCO 3 Ph=8 was adjusted. The solvent was then removed and extracted with dichloromethane (3 x 20 ml), the organic layer was separated, and the solvent was removed using Na 2 SO 4 Dried, filtered, concentrated and purified by silica gel chromatography (EA: pe=1:10) to give a white solid (2.1 g, 72.7%). 1 H-NMR(400MHz,CDCl 3 ):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 a solution of 4-chloro-2, 5-dimethoxybenzaldehyde (1.5 g,7.5 mmol) in MeOH (20 mL) was carefully added NaBH 4 (0.85 g,22.48 mmol). The mixture was then stirred at room temperature for 2h. The solvent was then removed and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was extracted with Na 2 SO 4 Dried, filtered, concentrated and purified by silica gel chromatography (EA: pe=1:4) to give a white solid (1.45 g, 96%). 1 H-NMR(400MHz,CDCl 3 ):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 DME solution (10 mL) of (4-chloro-2, 5-dimethoxyphenyl) methanol (500 mg,2.48 mmol) at 0deg.C) Carefully add PBr 3 (0.99 g,3.69 mmol). The mixture was then stirred at room temperature overnight. The solvent was then removed and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was extracted with Na 2 SO 4 Drying, filtration and concentration gave 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 (50.0 mg,0.22 mmol) and K 2 CO 3 To a solution of (36.6 mg,0.26 mmol) in anhydrous DMF (10 mL) was added 1- (bromomethyl) -4-chloro-2, 5-dimethoxybenzene (71.2 mg,0.26 mmol) and stirred under nitrogen overnight at room temperature. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give a white solid (23.4 mg, 25.8%). 1 H-NMR(400MHz,CDCl 3 ):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]+.
Synthesis of (V) 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 (10.0 mg,0.02 mmol) in DCM (2 mL) was added m-CPBA (6.4 mg,0.03 mmol). The mixture was then stirred at room temperature for 2h. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give a white solid (8.2 mg, 76.7%). 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000701
(I) Synthesis of 4-fluoro-2, 5-dimethoxybenzaldehyde
A solution of 2-fluoro-1, 4-dimethoxybenzene (2.07 g,13.26 mmol) in DCM (15 mL) was cooled to 5-6deg.C in an ice bath. 6.75g (25.99 mmol) of SnCl are added with vigorous stirring 4 Dichloro (methoxy) methane (1.3 g,11.41 mmol) was then added dropwise at a rate that maintained 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 (thick). The reaction mixture was then extracted with dichloromethane (3 x 20 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by silica gel chromatography (EA: pe=1:10) to give a green solid (1.9 g, 77.86%). 1 H-NMR(400MHz,CDCl 3 ):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 (500.0 mg,2.71 mmol) in anhydrous THF (10 mL) was carefully added NaBH 4 (410.8 mg,10.86 mmol). The mixture was then stirred at room temperature overnight. The solvent was then removed and extracted with dichloromethane (3 x 15 ml), the organic layer was separated, and the solvent was removed using Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give a white solid (238.1 mg, 47.2%). 1 H-NMR(400MHz,CDCl 3 ):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 DME solution (5 mL) of (4-fluoro-2, 5-dimethoxyphenyl) methanol (200 mg,1.07 mmol) at 0deg.C was carefully added PBr 3 (431.2 mg,1.67 mmol). The mixture was then stirred at room temperature overnight. The solvent was then removed and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was extracted with Na 2 SO 4 Drying, filtration and concentration gave 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 (50.0 mg,0.22 mmol) and K 2 CO 3 To a solution of (36.6 mg,0.26 mmol) in anhydrous DMF (10 mL) was added 1- (bromomethyl) -4-fluoro-2, 5-dimethoxybenzene (65.8 mg,0.26 mmol) and stirred overnight at room temperature under nitrogen. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give a white solid (30.1 mg). MS (m/z): 395.11[ M+H ]]+.
Synthesis of (V) 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 (20.0 mg,0.05 mmol) in DCM (5 mL) was added m-CPBA (13.1 mg,0.075 mmol). The mixture was then stirred at room temperature for 2h. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give a white solid (13.4 mg, 62.1%). 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000711
(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 (100 mg,0.46 mmol) in anhydrous NMP (2 mL) was added sodium methyl mercaptide (98.2 mg,1.40 mmol) and reacted in microwaves on a Biotage Smith synthesizer at 180℃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 (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Drying, filtration and concentration gave 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 (30.0 mg,0.13 mmol) in MeOH (1 mL) was added potassium hydrogen persulfate (409.9 mg,0.67 mmol) H 2 O (2 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was extracted with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give a white solid (4.7 mg, 10.9%). 1 H-NMR(400MHz,CDCl 3 ):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- (methylsulfanyl) -1H-purine-2, 6 (3H, 7H) -dione
Figure BDA0002247439760000721
(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 (200.0 mg,0.93 mmol) and K 2 CO 3 To a solution of (154.8 mg,1.12 mmol) in anhydrous DMF (2 mL) was added iodoethane (174.8 mg,1.12 mmol) and stirred under nitrogen at room temperature for 4h. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, 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-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 (213 mg,0.87 mmol) in anhydrous DMF (2 mL) was added sodium methyl mercaptide (1.5 mL) and heated to 105℃for 8H. The mixture was then cooled to room temperature and extracted with dichloromethane (3 x 10 ml), the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by silica gel chromatography (PE: ea=2:1) to give a white solid(31.2mg,19.2%)。 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000722
To a solution of compound 74 (24.7 mg,0.09 mmol) in MeOH (2 mL) was added potassium hydrogen persulfate (237.3 mg,0.38 mmol) H 2 O (2 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was extracted with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give a white solid (20.3 mg, 73.1%). 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000731
(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 (200.0 mg,0.93 mmol) and K 2 CO 3 To a solution of (154.8 mg,1.12 mmol) in anhydrous DMF (2 mL) was added iodoethane (174.8 mg,1.12 mmol) and stirred under nitrogen at room temperature for 4h. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, 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 8-chloro-7-ethylTo a solution of 1, 3-dimethyl-1H-purine-2, 6 (3H, 7H) -dione (140.0 mg,0.57 mmol) in anhydrous DMF (2 mL) was added NaHS (97.2 mg,1.73 mmol) 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 (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by silica gel chromatography (PE: ea=1:2) to give a white solid (82.3 mg, 59.3%). 1 H-NMR(400MHz,CDCl 3 ):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]+.
Synthesis of (III) 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 (30.0 mg,0.12 mmol) and K 2 CO 3 To a solution of (20.7 mg,0.15 mmol) in anhydrous DMF (2 mL) was added iodoethane (23.4 mg,0.15 mmol) and stirred under nitrogen at room temperature for 3h. The reaction mixture was poured into water and extracted with dichloromethane (3 x 10 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give a white solid (32.3 mg, 99.38%). 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000741
Compound 77: 7-ethyl-8- (ethylsulfonyl) -1, 3-dimethyl-1H-purine-2, 6 (3H, 7H) -dione
Figure BDA0002247439760000742
To a solution of compound 75 (25 mg) in MeOH (2 mL) was added potassium hydrogen persulfate (86.3 mg) H 2 O (2 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was extracted with Na 2 SO 4 Drying, filtration, concentration and purification by preparative TLC (PE: EA 1:1) gave 76.8 mg (20.4%) of compound as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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 77.2 mg (69.7%) as white solid was obtained. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000743
(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 (200.0 mg,0.93 mmol) and K 2 CO 3 To a solution of (154.8 mg,1.12 mmol) in anhydrous DMF (2 mL) was added iodoethane (174.8 mg,1.12 mmol) and stirred under nitrogen at room temperature for 4h. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, 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 (140.0 mg,0.57 mmol) in anhydrous DMF (2 mL) was added NaHS (97.2 mg,1.73 mmol) 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 (3 x 10 ml), and the organics separatedLayer with Na 2 SO 4 Dried, filtered, concentrated and purified by silica gel chromatography (PE: ea=1:2) to give a white solid (82.3 mg, 59.3%). 1H-NMR (400 MHz, CDCl 3): delta 13.18 (s, 1H), 4.79 (q, J=7.2 Hz, 2H), 3.58 (s, 3H), 3.39 (s, 3H), 1.40 (t, J=7.2 Hz, 3H): MS (m/z): 241.07[ M+H)]+.
Synthesis of (III) 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 (30.0 mg,0.12 mmol) and K2CO3 (20.7 mg,0.15 mmol) in anhydrous DMF (2 mL) was added 1-iodopropane (25.5 mg,0.15 mmol) and stirred under nitrogen at room temperature for 3H. The reaction mixture was poured into water and extracted with EA (3 x 10 ml), the organic layer was separated, taken up in Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give 31.5mg (89.3%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000751
Compound 80: 7-ethyl-1, 3-dimethyl-8- (propylsulfonyl) -1H-purine-2, 6 (3H, 7H) -dione
Figure BDA0002247439760000752
To a solution of compound 78 (30 mg) in MeOH (2 mL) was added potassium hydrogen persulfate (97.98 mg) H 2 O (2 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was extracted with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give compound 792.9mg (22.3%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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 80.2 mg (70.3%) as white solid was obtained. 1 H-NMR(400MHz,CDCl 3 ):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- (cyclopropylmethylsulfanyl) -7-ethyl-1, 3-dimethyl-1H-purine-2, 6 (3H, 7H) -dione
Figure BDA0002247439760000761
(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 (200.0 mg,0.93 mmol) and K 2 CO 3 To a solution of (154.8 mg,1.12 mmol) in anhydrous DMF (2 mL) was added iodoethane (174.8 mg,1.12 mmol) and stirred under nitrogen at room temperature for 4h. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, 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 (140.0 mg,0.57 mmol) in anhydrous DMF (2 mL) was added NaHS (97.2 mg,1.73 mmol) 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 (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by silica gel chromatography (PE: ea=1:2) to give a white solid (82.3 mg, 59.3%). 1 H-NMR(400MHz,CDCl 3 ):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]+.
Synthesis of (III) 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 (30.0 mg,0.12 mmol) and K 2 CO 3 (20.7 mg,0.15 mmol) in anhydrous DMF (2 mL) was added (bromomethyl) cyclopropane (20.25 mg,0.15 mmol) and stirred under nitrogen at room temperature for 3h. The reaction mixture was poured into water and extracted with EA (3 x 10 ml), the organic layer was separated, taken up in Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:1) to give 25.6mg (69.75%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000771
To a solution of compound 81 (23 mg) in MeOH (1 mL) was added potassium hydrogen persulfate (240 mg) H 2 O (1 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was extracted with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give 11.5mg (66.9%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000772
(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 (500.0 mg,2.33 mmol) and K 2 CO 3 To a solution of (386.9 mg,2.80 mmol) in anhydrous DMF (5 mL) was added 1-iodopropane (476.5 mg,2.80 mmol) and stirred under nitrogen at room temperature for 4h. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, 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 (350.0 mg,1.36 mmol) in anhydrous DMF (5 mL) was added NaHS (229.7 mg,4.10 mmol) 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 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified 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 (50.0 mg,0.19 mmol) and K 2 CO 3 To a solution of (32.5 mg,0.23 mmol) in anhydrous DMF (5 mL) was added methyl iodide (35.3 mg,0.23 mmol) and stirred under nitrogen at room temperature for 2h. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give a white solid (47.8 mg, 90.62%). 1 H-NMR(400MHz,CDCl 3 ):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]+.
Synthesis of (IV) 1, 3-dimethyl-8- (methylsulfonyl) -7-propyl-1H-purine-2, 6 (3H, 7H) -dione
To 1, 3-dimethyl-8- (methyl)To a solution of thio) -7-propyl-1H-purine-2, 6 (3H, 7H) -dione (40.0 mg,0.15 mmol) in MeOH (5 mL) was added potassium hydrogen persulfate (366.9 mg,0.59 mmol) H 2 O (5 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was extracted with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give a white solid (41.9 mg, 93.58%). 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000781
(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 (3 g) and K 2 CO 3 To a solution of (2.32 g) in anhydrous DMF (30 mL) was added 2-bromoacetonitrile (2 g) and stirred under nitrogen at room temperature for 3h. The reaction mixture was then poured into water and extracted with EA (3 x 50 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by silica gel chromatography (PE: ea=1:1) to give a white solid (2.3 g, 64.78%).
(II) Synthesis of 2- (8-mercapto-1, 3-dimethyl-2, 6-dioxo-2, 3-dihydro-1H-purin-7 (6H) -yl) acetonitrile
To a solution of 2- (8-chloro-1, 3-dimethyl-2, 6-dioxo-2, 3-dihydro-1H-purin-7 (6H) -yl) acetonitrile (100 mg) in anhydrous DMF (3 mL) was added NaHS (44.3 mg) and heated to 135℃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 (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by silica gel chromatography (PE: ea=1:1) to give a white solid (45.6 mg, 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 (20 mg) and K 2 CO 3 To a solution of (13.2 mg) in anhydrous DMF (5 mL) was added methyl iodide (13.5 mg) and stirred under nitrogen at room temperature for 2h. The reaction mixture was then poured into water and extracted with EA (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:1) 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- (methylsulfanyl) -2, 6-dioxo-2, 3-dihydro-1H-purin-7 (6H) -yl) acetonitrile (15 mg) in MeOH (2 mL) was added potassium hydrogen persulfate (115.99 mg) 2 O (2 mL) solution. The mixture was then stirred at room temperature for 6h. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give 8.2mg (46.86%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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- (methylsulfanyl) -2, 6-dioxo-2, 3-dihydro-1H-purin-7 (6H) -yl) acetamide
Figure BDA0002247439760000801
2- (1, 3-dimethyl-8- (methylthio) -2, 6-dioxo-2, 3-dihydro-1H-purin-7 (6H) -yl) acetonitrile was synthesized as described in compound 84.
To a solution of 2- (1, 3-dimethyl-8- (methylsulfanyl) -2, 6-dioxo-2, 3-dihydro-1H-purin-7 (6H) -yl) acetonitrile (100 mg,0.39 mmol) in EtOH (2 mL) was added sodium methyl mercaptide (33.2 mg,0.47 mmol) and refluxed for 5H. The mixture was then cooled to room temperature and extracted with dichloromethane (3 x 5 ml), the organic layer was separated, and the organic layer was taken up in Na 2 SO 4 Drying and filteringConcentrated and purified by preparative TLC to give a white solid (21.2 mg, 19.1%). 1 H-NMR(400MHz,DMSO-d 6 ):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
Figure BDA0002247439760000802
To a solution of Compound 85 (20.0 mg,0.07 mmol) in MeOH (2 mL) was added potassium hydrogen persulfate (217.3 mg,0.35 mmol) in H 2 O (2 mL) solution. The mixture was then stirred at room temperature for 8h. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give a white solid (15.4 mg, 73.65%). 1 H-NMR(400MHz,DMSO-d 6 ):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
Figure BDA0002247439760000803
(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 (4.0 g,22.19 mmol) in anhydrous THF (20 mL) was added NCS (4.5 g,33.71 mmol) and stirred under nitrogen overnight at room temperature. The solvent was then removed and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was extracted with Na 2 SO 4 Drying, filtration, concentration and recrystallization from EtOH: meoh=2:1 afforded a white solid (2.5 g, 52.3%).
(II) Synthesis of 3, 7-dimethyl-8- (methylthio) -1H-purine-2, 6 (3H, 7H) -dione
To 8-chloro-3, 7-dimethyl-1H-purine-2, 6 (3H, 7H) -dione (150 mg,0.70 mmol)To a solution of anhydrous DMF (2 mL) was added sodium methyl mercaptide (195.9 mg,2.79 mmol) and reacted in the microwave on a Biotage Smith synthesizer at 130℃for 1h. 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 in DCM: pe=1:10 and filtered to give a white solid (89.5 mg, 62.3%). 1 H-NMR(400MHz,DMSO-d 6 ):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
Figure BDA0002247439760000811
To a solution of compound 88 (20 mg,0.08 mmol) in MeOH (1 mL) was added potassium hydrogen persulfate (326.3 mg,0.53 mmol) H 2 O (1 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give a white solid (4.5 mg, 39.4%). 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000812
(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 (4.0 g,22.19 mmol) in anhydrous THF (20 mL) was added NCS (4.5 g,33.71 mmol) and stirred under nitrogen overnight at room temperature. The solvent was then removed and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was extracted with Na 2 SO 4 Dried, filtered, concentrated and recrystallized from EtOH: meoh=2:1 to give 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 (500 mg,2.33 mmol) and K 2 CO 3 To a solution of (460.3 mg,2.79 mmol) in anhydrous DMF (5 mL) was added iodoethane (519.6 mg,2.79 mmol) and stirred under nitrogen at room temperature for 4h. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by silica gel chromatography (PE: ea=2:1) to give a white solid (141.2 mg, 16.3%). 1 H-NMR(400MHz,CDCl 3 ):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 (40.0 mg,0.16 mmol) in anhydrous DMF (2 mL) was added NaHS (26.9 mg,0.48 mmol) and heated to 100deg.C 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 (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Drying, filtration and concentration gave a white solid (35.9 mg). MS (m/z): 241.07[ M+H ]]+.
Synthesis of (IV) 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 (25.0 mg,0.10 mmol) and K 2 CO 3 To a solution of (17.3 mg,0.12 mmol) in anhydrous DMF (1 mL) was added methyl iodide (17.8 mg,0.12 mmol) and stirred under nitrogen at room temperature for 2h. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give a white solid (12.0 mg, 43.5%). 1 H-NMR(400MHz,CDCl 3 ):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].
Synthesis of (V) 1-ethyl-3, 7-dimethyl-8- (methylsulfonyl) -1H-purine-2, 6 (3H, 7H) -dione
To a solution of 1-ethyl-3, 7-dimethyl-8- (methylsulfanyl) -1H-purine-2, 6 (3H, 7H) -dione (10 mg,0.03 mmol) in MeOH (1 mL) was added potassium hydrogen persulfate (120.8 mg,0.19 mmol) in H 2 O (1 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give a white solid (8.2 mg, 77.4%). 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000831
(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 (4.0 g,22.19 mmol) in anhydrous THF (20 mL) was added NCS (4.5 g,33.71 mmol) and stirred under nitrogen overnight at room temperature. The solvent was then removed and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was extracted with Na 2 SO 4 Drying, filtration, concentration and recrystallization from EtOH: meoh=2:1 afforded a white solid (2.5 g, 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 (100 mg) and sodium methyl mercaptide (130.5 mg) in anhydrous DMF (4 mL) was reacted in microwaves for 1H on a Biotage Smith synthesizer at 100 ℃. The mixture was then cooled to room temperature and acidified to ph=5. The reaction mixture was poured into water and extracted with dichloromethane (3 x 10 ml) and the organic layer was separated with Na 2 SO 4 Drying, filtration and concentration gave a white solid (67.8 mg, 64.3%).
Synthesis of (III) 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 (30.0 mg,0.13 mmol) and K 2 CO 3 To a solution of (21.9 mg,0.15 mmol) in anhydrous DMF (2 mL) was added 1-iodopropane (27.1 mg,0.15 mmol) and stirred under nitrogen at room temperature for 2h. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give a white solid (18.7 mg, 43.7%). 1 H-NMR(400MHz,CDCl 3 ):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]+.
Synthesis of (IV) 3, 7-dimethyl-8- (methylsulfonyl) -1-propyl-1H-purine-2, 6 (3H, 7H) -dione
To a solution of 3, 7-dimethyl-8- (methylsulfanyl) -1-propyl-1H-purine-2, 6 (3H, 7H) -dione (15.0 mg,0.05 mmol) in MeOH (1 mL) was added potassium hydrogen persulfate (171.8 mg,0.27 mmol) H 2 O (1 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give a white solid (8.7 mg, 61.4%). 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000841
(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 (4.0 g,22.19 mmol) in anhydrous THF (20 mL) was added NCS (4.5 g,33.71 mmol) and stirred under nitrogen overnight at room temperature. The solvent was then removed and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was extracted with Na 2 SO 4 DryingFiltration, concentration and recrystallization from 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 (500 mg,2.33 mmol) and K 2 CO 3 To a solution of (385.8 mg) in anhydrous DMF (5 mL) was added (bromomethyl) cyclopropane (374.3 mg) and the mixture was stirred at room temperature under nitrogen for 4h. The reaction mixture was then poured into water and extracted with EA (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by silica gel chromatography (PE: ea=2:1) to give a white solid (325 mg, 51.9%).
Synthesis of (III) 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 (300 mg) in anhydrous DMF (2 mL) was added NaHS (187.6 mg) 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 (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Drying, filtering and concentrating to obtain white solid.
Synthesis of (IV) 1- (cyclopropylmethyl) -3, 7-dimethyl-8- (methylthio) -1H-purine-2, 6 (3H, 7H) -dione
To 1- (cyclopropylmethyl) -8-mercapto-3, 7-dimethyl-1H-purine-2, 6 (3H, 7H) -dione (289.1 mg) and K 2 CO 3 To a solution of (179.9 mg) in anhydrous DMF (5 mL) was added methyl iodide (184.8 mg) and stirred under nitrogen at room temperature for 2h. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 15 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified on silica gel (PE: EA 4:1) to give a white solid (280 mg, 92.1%).
Synthesis of (V) 1- (cyclopropylmethyl) -3, 7-dimethyl-8- (methylsulfonyl) -1H-purine-2, 6 (3H, 7H) -dione
To a solution of 1- (cyclopropylmethyl) -3, 7-dimethyl-8- (methylsulfanyl) -1H-purine-2, 6 (3H, 7H) -dione (250 mg) in MeOH (5 mL) was added hydrogen persulfateH of Potassium (2.74 g) 2 O (5 mL) solution. The mixture was then stirred at room temperature overnight. The solvent was then removed and extracted with dichloromethane (3 x 15 ml), the organic layer was separated, and the solvent was removed using Na 2 SO 4 Dried, filtered, concentrated and purified on silica gel (PE: EA 1:1) to give 150mg (53.8%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000851
(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 (4.0 g,22.19 mmol) in anhydrous THF (20 mL) was added NCS (4.5 g,33.71 mmol) and stirred under nitrogen overnight at room temperature. The solvent was then removed and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was extracted with Na 2 SO 4 Drying, filtration, concentration and recrystallization from EtOH: meoh=2:1 afforded a white solid (2.5 g, 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 (100 mg) and sodium methyl mercaptide (130.5 mg) in anhydrous DMF (4 mL) was reacted in microwaves for 1H on a Biotage Smith synthesizer at 100 ℃. The mixture was then cooled to room temperature and acidified to ph=5. The reaction mixture was poured into water and extracted with dichloromethane (3 x 10 ml) and the organic layer was separated with Na 2 SO 4 Drying, 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 (50.0 mg,0.22 mmol) and phenylboronic acid (26.9 mg,0.22 mmol) in anhydrous DCM (2 mL) was added T EA (90.5 mg,0.88 mmol) and Cu (OAc) 2 (60.1 mg,0.33 mmol). The mixture was then stirred at room temperature overnight. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by silica gel chromatography (PE: ea=2:1) to give a white solid (15.0 mg, 18.4%). 1 H-NMR(400MHz,CDCl 3 ):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]+.
Synthesis of (IV) 3, 7-dimethyl-8- (methylsulfonyl) -1-phenyl-1H-purine-2, 6 (3H, 7H) -dione
To a solution of 3, 7-dimethyl-8- (methylsulfanyl) -1-phenyl-1H-purine-2, 6 (3H, 7H) -dione (20.0 mg,0.06 mmol) in MeOH (2 mL) was added potassium hydrogen persulfate (162.8 mg,0.26 mmol) H 2 O (2 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give a white solid (14.3 mg, 64.2%). 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000861
(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 (4.0 g,22.19 mmol) in anhydrous THF (20 mL) was added NCS (4.5 g,33.71 mmol) and stirred under nitrogen overnight at room temperature. The solvent was then removed and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was extracted with Na 2 SO 4 Dried, filtered, concentrated and recrystallized from EtOH: meoh=2:1 to give a white solid.
(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 (500 mg,2.33 mmol) and K 2 CO 3 To a solution of (385.8 mg) in anhydrous DMF (5 mL) was added (bromomethyl) benzene (478.6 mg) and stirred under nitrogen at room temperature for 4h. The reaction mixture was then poured into water and extracted with EA (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by silica gel chromatography (PE: ea=1:1) to give a white solid (423.5 mg, 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 (150 mg) in anhydrous DMF (5 mL) was added NaHS (39.5 mg) 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 (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Drying, filtering and concentrating to obtain white solid.
Synthesis of (IV) 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 (128 mg) and K 2 CO 3 To a solution of (66.99 mg) in anhydrous DMF (5 mL) was added methyl iodide (68.89 mg) and stirred under nitrogen at room temperature for 2h. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 15 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified on silica gel (PE: EA 1:1) to give a white solid (108.3 mg, 81.12%).
Synthesis of (V) 1-benzyl-3, 7-dimethyl-8- (methylsulfonyl) -1H-purine-2, 6 (3H, 7H) -dione
To a solution of 1-benzyl-3, 7-dimethyl-8- (methylsulfanyl) -1H-purine-2, 6 (3H, 7H) -dione (250 mg) in MeOH (5 mL) was added H potassium hydrogen persulfate (2.43 g) 2 O (5 mL) solution. The mixture was then stirred at room temperature overnight. The solvent was then removed and extracted with dichloromethane (3 x 15 ml), the organic layer was separated, and the solvent was removed using Na 2 SO 4 Dried, filtered, concentrated and purified on silica gel (PE: EA 1:1) to give 138.2mg (50.1%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000871
(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 (4.0 g,22.19 mmol) in anhydrous THF (20 mL) was added NCS (4.5 g,33.71 mmol) and stirred under nitrogen overnight at room temperature. The solvent was then removed and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was extracted with Na 2 SO 4 Dried, filtered, concentrated and recrystallized from EtOH: meoh=2:1 to give a white solid.
(II) Synthesis of 8-chloro-3, 7-dimethyl-1-phenethyl-1H-purine-2, 6 (3H, 7H) -dione
To 8-chloro-3, 7-dimethyl-1H-purine-2, 6 (3H, 7H) -dione (500 mg,2.33 mmol) and K 2 CO 3 To a solution of (385.8 mg) in anhydrous DMF (5 mL) was added (2-bromoethyl) benzene (518.2 mg) and stirred under nitrogen at room temperature for 4h. The reaction mixture was then poured into water and extracted with EA (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by silica gel chromatography (PE: ea=1:1) to give a white solid (336.7 mg, 47.6%).
Synthesis of (III) 8-mercapto-3, 7-dimethyl-1-phenethyl-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 (150 mg) in anhydrous DMF (5 mL) was added NaHS (39.5 mg) 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 (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Drying, filtering and concentrating to obtain white solid.
Synthesis of (IV) 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 (128 mg) and K 2 CO 3 To a solution of (66.99 mg) in anhydrous DMF (5 mL) was added methyl iodide (68.89 mg) and stirred under nitrogen at room temperature for 2h. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 15 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified on silica gel (PE: EA 1:1) to give a white solid.
Synthesis of (V) 3, 7-dimethyl-8- (methylsulfonyl) -1-phenethyl-1H-purine-2, 6 (3H, 7H) -dione
To a solution of 3, 7-dimethyl-8- (methylsulfanyl) -1-phenethyl-1H-purine-2, 6 (3H, 7H) -dione (100 mg) in MeOH (5 mL) was added H potassium hydrogen persulfate (931.5 mg) 2 O (5 mL) solution. The mixture was then stirred at room temperature overnight. The solvent was then removed and extracted with dichloromethane (3 x 15 ml), the organic layer was separated, and the solvent was removed using Na 2 SO 4 Drying, filtration, concentration and purification on silica gel (PE: EA 1:1) gave 48.9mg (45.7%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000881
(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 (4.0 g,22.19 mmol) in anhydrous THF (20 mL) was added NCS (4.5 g,33.71 mmol) and stirred under nitrogen overnight at room temperature. The solvent was then removed and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was extracted with Na 2 SO 4 Drying, filtration, concentration and recrystallization from EtOH: meoh=2:1 afforded a white solid (2.5 g, 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 (100 mg) and sodium methyl mercaptide (130.5 mg) in anhydrous DMF (4 mL) was reacted in microwaves on a Biotage Smith synthesizer at 100deg.C for 1H. The mixture was then cooled to room temperature and acidified to ph=5. The reaction mixture was poured into water and extracted with dichloromethane (3 x 10 ml) and the organic layer was separated with Na 2 SO 4 Drying, filtration and concentration gave a white solid (67.8 mg, 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 (30.0 mg,0.13 mmol) and K 2 CO 3 To a solution of (21.9 mg,0.15 mmol) in anhydrous DMF (2 mL) was added 3-bromoprop-1-yne (18.9 mg,0.15 mmol) and stirred under nitrogen at room temperature for 2h. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA1: 1) to give a white solid (10.2 mg, 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- (methylsulfanyl) -1- (prop-2-yn-1-yl) -1H-purine-2, 6 (3H, 7H) -dione (10 mg) in MeOH (1 mL) was added potassium hydrogen persulfate (116.3 mg) H 2 O (1 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:2) to give 4.8mg (45.7%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000891
/>
(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 (4.0 g,22.19 mmol) in anhydrous THF (20 mL) was added NCS (4.5 g,33.71 mmol) and stirred under nitrogen overnight at room temperature. The solvent was then removed and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was extracted with Na 2 SO 4 Drying, filtration, concentration and recrystallization from EtOH: meoh=2:1 afforded a white solid (2.5 g, 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 (100 mg) and sodium methyl mercaptide (130.5 mg) in anhydrous DMF (4 mL) was reacted in microwaves on a Biotage Smith synthesizer at 100deg.C for 1H. The mixture was then cooled to room temperature and acidified to ph=5. The reaction mixture was poured into water and extracted with dichloromethane (3 x 10 ml) and the organic layer was separated with Na 2 SO 4 Drying, filtration and concentration gave a white solid (67.8 mg, 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 (50.0 mg) and K 2 CO 3 To a solution of (36.6 mg) in anhydrous DMF (2 mL) was added 2-chloro-N, N-dimethylacetamide (32.1 mg) and stirred under nitrogen at room temperature for 2h. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:1) to give a white solid (43.4 mg, 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 a solution of 2- (3, 7-dimethyl-8- (methylsulfanyl) -2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purin-1-yl) -N, N-dimethylacetamide (10 mg) in MeOH (1 mL) was addedH of Potassium hydrogen persulfate (78.9 mg) 2 O (1 mL) solution. The mixture was then stirred overnight. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:1) to give 6.8mg (63.1%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000901
(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 (4.0 g,22.19 mmol) in anhydrous THF (20 mL) was added NCS (4.5 g,33.71 mmol) and stirred under nitrogen overnight at room temperature. The solvent was then removed and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was extracted with Na 2 SO 4 Drying, filtration, concentration and recrystallization from EtOH: meoh=2:1 afforded a white solid (2.5 g, 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 (100 mg) and sodium methyl mercaptide (130.5 mg) in anhydrous DMF (4 mL) was reacted in microwaves for 1H on a Biotage Smith synthesizer at 100 ℃. The mixture was then cooled to room temperature and acidified to ph=5. The reaction mixture was poured into water and extracted with dichloromethane (3 x 10 ml) and the organic layer was separated with Na 2 SO 4 Drying, filtration and concentration gave a white solid (67.8 mg, 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 (15 mg) and Cs 2 CO 3 To a solution of (25.94 mg) in anhydrous DMF (2 mL) was added 4- (2-chloroethyl) morpholine hydrochloride (14.8 mg) and reacted in the microwave at 110℃for 1h on a Biotage Smith synthesizer. The reaction mixture was then cooled to room temperature and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Drying, filtration and concentration gave a yellow oil (20.1 mg, 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 (10 mg,0.03 mmol) in DCM (2 mL) was added m-CPBA (12.73 mg,0.07 mmol). The mixture was then stirred at room temperature for 2h. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative HPLC to give a white solid (4.1 mg, 39.7%). 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000911
(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 (4.0 g,22.19 mmol) in anhydrous THF (20 mL) was added NCS (4.5 g,33.71 mmol) and stirred under nitrogen overnight at room temperature. The solvent was then removed and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was extracted with Na 2 SO 4 Drying, filtration, concentration and recrystallization from EtOH: meoh=2:1 afforded a white solid (2.5 g, 52.3%).
(II) Synthesis of 3, 7-dimethyl-8- (methylthio) -1H-purine-2, 6 (3H, 7H) -dione
8-chloro-3, 7-dimethyl-1H-purine-2, 6 (3H, 7H) -dioneA mixture of (100 mg) and sodium methyl mercaptide (130.5 mg) in anhydrous DMF (4 mL) was reacted in a microwave over a Biotage Smith synthesizer at 100deg.C for 1h. The mixture was then cooled to room temperature and acidified to ph=5. The reaction mixture was poured into water and extracted with dichloromethane (3 x 10 ml) and the organic layer was separated with Na 2 SO 4 Drying, 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 (3.0 mg,0.03 mol), 2-furanboronic acid (59 mg,0.53 mmol) and 3A molecular sieve (100 mg, freshly activated) in 1, 2-dichloroethane (4 mL) was cooled to 0℃under a nitrogen atmosphere. Anhydrous pyridine (0.3 mL) was added followed by a solution of 3, 7-dimethyl-8- (methylthio) -1H-purine-2, 6 (3H, 7H) -dione (60 mg,0.265 mmol) in 1, 2-dichloroethane (1 mL). The reaction mixture was allowed to warm to room temperature. Will be filled with dry O 2 Is connected to the flask and the mixture is stirred for 48h. The reaction mixture was filtered and the filtrate was concentrated and purified by preparative TLC (PE/ea=2:3) to give a white solid (34 mg, 44%): 1 H NMR(400MHz,CDCl 3 )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- (methylsulfanyl) -1H-purine-2, 6 (3H, 7H) -dione (34 mg,0.12 mmol) in MeOH (2.5 mL) was added potassium hydrogen persulfate (319 mg,0.60 mmol) in H 2 Mixtures in O (2 mL). The reaction mixture was stirred at room temperature for 16h. The reaction mixture was taken up in DCM (30 mL) and H 2 O (5 mL) between partitions. The organic layer was dried (Na 2 SO 4 ) Concentrated and purified by preparative TLC (PE: ea=2:3) to give a white solid (16 mg, 42%): 1 H NMR(400MHz,CDCl 3 )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
Figure BDA0002247439760000921
(I) Synthesis of 1, 7-dimethyl-1H-purine-2, 6 (3H, 7H) -dione
To a solution of ethyl 4-amino-1-methyl-1H-imidazole-5-carboxylate (2.0 g,11.83 mmol) and ethyl methylcarbamate (2.92 g,28.32 mmol) in anhydrous THF (2 mL) was reacted at 75deg.C under nitrogen for 30min. Potassium 2-methylpropyl-2-oleate (1.98 g,17.64 mmol) was then added to the mixture and reacted overnight at 75 ℃. The solvent was then removed and extracted with dichloromethane (3 x 50 ml), the organic layer was separated, and the solvent was extracted with Na 2 SO 4 Dried, filtered, concentrated and purified by silica gel chromatography (DCM: meoh=20:1) to give a white solid (1.2 g, 56.33%). 1 H-NMR(400MHz,DMSO-d 6 ):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 (0.53 g,2.94 mmol) in anhydrous THF (10 mL) was added NCS (0.59 g,4.36 mmol) and stirred under nitrogen overnight at room temperature. The solvent was then removed and extracted with dichloromethane (3 x 20 ml), the organic layer was separated, and the solvent was removed using Na 2 SO 4 Drying, filtration and concentration gave 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 (100 mg,0.46 mmol) in DMF (10 mL) and H 2 To a solution of O (1 mL) was added sodium methyl mercaptide (130.8 mg,1.86 mmol) and reacted in a microwave over a Biotage Smith synthesizer at 120℃for 1h. The mixture was then cooled to room temperature and acidified to ph=3. The reaction mixture was poured into water and extracted with dichloromethane (3 x 10 ml) and the organic layer was separated with Na 2 SO 4 Drying, filtration and concentration gave 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 (15.0 mg,0.06 mmol) in MeOH (3 mL) was added potassium hydrogen persulfate (163.2 mg,0.26 mmol) H 2 O (5 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was extracted with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give a white solid (12.8 mg, 74.85%). 1 H-NMR(400MHz,DMSO-d 6 ):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
Figure BDA0002247439760000941
(I) Synthesis of 3-ethyl-1, 7-dimethyl-8- (methylthio) -1H-purine-2, 6 (3H, 7H) -dione
The compound 1, 7-dimethyl-8- (methylsulfanyl) -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 (15.0 mg,0.06 mmol) and K 2 CO 3 To a solution of (11.1 mg,0.08 mmol) in anhydrous DMF (2 mL) was added iodoethane (12.4 mg,0.08 mmol) and stirred under nitrogen at room temperature for 2h. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give a white solid (11.0 mg, 65.48%). 1 H-NMR(400MHz,CDCl 3 ):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- (methylsulfanyl) -1H-purine-2, 6 (3H, 7H) -dione (10.0 mg,0.04 mmol) in MeOH (1 mL) was added potassium hydrogen persulfate (96.8) mg) of H 2 O (1 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was extracted with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give a white solid (10.5 mg, 92.9%). 1 H-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
Figure BDA0002247439760000942
(I) Synthesis of 1, 7-dimethyl-8- (methylthio) -3-propyl-1H-purine-2, 6 (3H, 7H) -dione
The compound 1, 7-dimethyl-8- (methylsulfanyl) -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 (30 mg) and K 2 CO 3 To a solution of (29.97 mg) in anhydrous DMF (5 mL) was added 1-iodopropane (27.1 mg) and stirred under nitrogen at room temperature for 2h. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:1) to give a white solid (31.1 mg, 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- (methylsulfanyl) -3-propyl-1H-purine-2, 6 (3H, 7H) -dione (30 mg) in MeOH (2 mL) was added H potassium hydrogen persulfate (275.3 mg) 2 O (2 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was extracted with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:1) to give 28.3mg (84.3%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000951
(I) Synthesis of 3- (cyclopropylmethyl) -1, 7-dimethyl-8- (methylthio) -1H-purine-2, 6 (3H, 7H) -dione
The compound 1, 7-dimethyl-8- (methylsulfanyl) -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 (30 mg) and K 2 CO 3 To a solution of (21.97 mg) in anhydrous DMF (5 mL) was added (bromomethyl) cyclopropane (21.3 mg) and the mixture was stirred at room temperature under nitrogen for 2h. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:1) to give a white solid (31.9 mg, 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- (methylsulfanyl) -1H-purine-2, 6 (3H, 7H) -dione (30 mg) in MeOH (2 mL) was added potassium hydrogen persulfate (263.5 mg) H 2 O (2 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was extracted with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:1) to give 31.4mg (94.1%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000961
(I) Synthesis of 3-benzyl-1, 7-dimethyl-8- (methylthio) -1H-purine-2, 6 (3H, 7H) -dione
The compound 1, 7-dimethyl-8- (methylsulfanyl) -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 (30 mg) and K 2 CO 3 To a solution of (21.97 mg) in anhydrous DMF (5 mL) was added (bromomethyl) benzene (27.3 mg) and stirred under nitrogen at room temperature for 2h. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:1) to give a white solid (32.1 mg, 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- (methylsulfanyl) -1H-purine-2, 6 (3H, 7H) -dione (30 mg) in MeOH (2 mL) was added potassium hydrogen persulfate (233.1 mg) H 2 O (2 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was extracted with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA1: 1) to give 30.9mg (93.6%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000971
(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 (2.0 g,8.18 mmol) and CDI (2.0 g,12.33 mmol) in anhydrous DMF/THF (20 mL/5 mL) was added DIEA (1.6 g,12.37 mmol). The mixture was stirred at 0℃for 0.5h. To a solution B of additional 3-chloropropan-1-amine hydrochloride (1.4 g,10.76 mmol) in anhydrous DMF (10 mL) was added DIEA (1.6 g,12.37 mmol) and stirred at room temperature for 0.5h. Solution B was then added to solution a and stirred at room temperature overnight. The solvent was removed and ph=5 was adjusted. The mixture was then extracted with DCM (3×50 ml), with NaHCO 3 The organic layer was washed (3×50 ml) and recrystallized from DCM: pe=7:3 to give a white solid (1.82 g, 37.2%). 1 H-NMR(400MHz,DMSO-d 6 ):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 (50.0 mg,0.19 mmol) and K 2 CO 3 To a solution of (32.1 mg,0.23 mmol) in anhydrous DMF (10 mL) was added N- (3-chloropropyl) -5- ((4R) -2-oxohexahydro-1H-thieno [3, 4-d)]Imidazol-4-yl) pentanamide (74.2 mg,0.23 mmol) and stirred under nitrogen at room temperature for 2h. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative HPLC to give a white solid (8.2 mg, 7.8%). 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000981
To compound 99 (50.0 mg,0.19 mmol) and K 2 CO 3 To a solution of (32.1 mg,0.23 mmol) in anhydrous DMF (10 mL) was added 4-methylbenzenesulfonic acid 2- (2- (2- (5- ((4R) -2-oxohexahydro-1H-thieno [3, 4-d))]Imidazol-4-yl) pentanamido) ethoxy) ethyl ester (123.1 mg,0.23 mmol) and stirred at room temperature under nitrogen overnight. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative HPLC to give 15.2mg (12.77%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000982
To compound 99 (20 mg) and K 2 CO 3 To a solution of (12.8 mg) in anhydrous DMF (5 mL) was added 3-bromoprop-1-yne (10.96 mg) and stirred under nitrogen at room temperature for 2h. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:1) to give 10.3mg (44.89%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000991
To compound 99 (10 mg) and K 2 CO 3 To a solution of (6.4 mg) in anhydrous DMF (5 mL) was added 2-bromoacetonitrile (5.6 mg) and the mixture was stirred at room temperature under nitrogen for 2h. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:1) to give 4.7mg (40.9%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760000992
(I) Synthesis of (3-bromoprop-1-yn-1-yl) benzene
To a DME solution of 3-phenylpropan-2-yn-1-ol (300 mg) was slowly added tribromophosphine (912.7 mg). The mixture was then stirred under nitrogen at room temperature for 2h. The mixture was then poured into ice and extracted with dichloromethane (3 x 10 ml), the organic layer was separated with Na 2 SO 4 Drying, 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 compound 99 (10 mg) and K 2 CO 3 (3-Bromoprop-1-yn-1-yl) benzene (9.1 mg) was added to a solution of (6.4 mg) in anhydrous DMF (5 mL) and stirred at room temperature under nitrogen for 2h. The reaction mixture was then poured into water and extracted with EA (3 x 5 ml), separatedSeparating the organic layer with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:1) to give 8.3mg (57.7%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760001001
(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 (2.6 g,18 mmol) in DMF (10 mL) was added 3-iodophenol (1 g,5 mmol), copper (I) iodide (catalytic amount), (Ph 3 P) 4 Pd (catalytic amount) and TEA (1 g,9 mmol). The mixture was then heated to 90 ℃ under nitrogen for 2 hours. The resulting reaction was cooled to room temperature, filtered and extracted with EA (3 x 5 ml), the organic layer was separated, taken up in Na 2 SO 4 Dried, filtered, concentrated and purified by column chromatography (PE: EA 10:1) to give a yellow oil (1.3 g, 93%). MS (m/z): 233.3[ M+H ]]+.
(II) Synthesis of 3- (3-hydroxy-prop-1-yn-1-yl) phenol
To a solution of 3- (3- ((tetrahydro-2H-pyran-2-yl) oxy) prop-1-yn-1-yl) phenol (1.3 g,5.6 mmol) in MeOH (15 mL) was added 4-methylbenzenesulfonic acid (193 mg,1.12 mmol) and stirred at room temperature for 2H. The reaction mixture was then concentrated and extracted with EA (3 x 5 ml), the organic layer was separated, and the mixture was taken up in Na 2 SO 4 Dried, filtered, concentrated and purified by column chromatography (PE: EA 1:1) to give a yellow oil (600 mg, 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-hydroxypropyl-1-yn-1-yl) phenol (430 mg,2.8 mmol) in DCM (8 mL) at 0deg.C was slowly added triphenylphosphine (1.29 g,3.2 mmol) and tetrabromomethane (2.15 g,5.6 mmol). Then stirring and mixing at room temperatureAnd (3) carrying out object 2h. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by column chromatography (PE: EA 5:1) to give 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 compound 99 (75 mg,0.3 mmol) and K 2 CO 3 To a solution of (70 mg,0.48 mmol) in anhydrous DMF (3 mL) was added 3- (3-bromoprop-1-yn-1-yl) phenol (60 mg,0.3 mmol) and stirred under nitrogen at room temperature for 0.5h. The reaction mixture was then poured into water and extracted with EA (3 x 5 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by column chromatography (PE: EA 1:1) to give 80mg (87%) as a white solid. 1 H-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
Figure BDA0002247439760001011
To compound 109 (30 mg,0.08 mmol) and K 2 CO 3 To a solution of (22 mg,0.16 mmol) in anhydrous DMF (2 mL) was added methyl iodide (17 mg,0.12 mmol) and stirred at room temperature for 0.5h. The reaction mixture was then poured into water and extracted with EA (3 x 5 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by column chromatography (PE: EA 1:1) to give 26mg (83.66%) as a white solid. 1 H-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
Figure BDA0002247439760001012
To a solution of compound 109 (30 mg,0.08 mmol) and K2CO3 (22 mg,0.16 mmol) in anhydrous DMF (2 mL) was added 1-iodopropane (20 mg,0.12 mmol) and stirred at room temperature for 0.5h. The reaction mixture was then poured into water and extracted with EA (3 x 5 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by column chromatography (PE: EA 5:1) to give 25mg (80.7%) as a white solid. 1 H-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
Figure BDA0002247439760001021
Compound 109 was synthesized in a similar manner. 1 H NMR(400MHz,cdcl 3 )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
Figure BDA0002247439760001022
Compound 113 (yellow solid, 8mg, yield 70%) was prepared from 1, 7-dimethyl-8- (methylsulfanyl) -1H-purine-2, 6 (3H, 7H) -dione according to a similar method as described for compound 109. 1 H NMR(400MHz,cdcl 3 )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). 13 C NMR(101MHz,cdcl 3 )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] + Calculated 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
Figure BDA0002247439760001023
Compound 114 (yellow solid, 10mg, yield 72%) was prepared from 1, 7-dimethyl-8- (methylsulfanyl) -1H-purine-2, 6 (3H, 7H) -dione according to a similar method as described for compound 109. 1 H 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). 13 C 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] + Calculated 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
Figure BDA0002247439760001031
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 method as described for compound 109. 1 H-NMR(400MHz,CDCl 3 )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
Figure BDA0002247439760001032
Compound 16 (yellow solid, 22mg, 80% yield) was prepared from 8- ((cyclopropylmethyl) thio) -1, 7-dimethyl-3, 7-dihydro-1H-purine-2, 6-dione according to a similar procedure as described for compound 109. 1 H-NMR(400MHz,CDCl 3 )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
Figure BDA0002247439760001041
Compound 117 (white solid, 8mg, 73% yield) was prepared from 8- ((cyclopropylmethyl) thio) -1, 7-dimethyl-3, 7-dihydro-1H-purine-2, 6-dione according to a similar procedure as 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
Figure BDA0002247439760001042
Compound 118 (white solid, 10 mg) was prepared from 8- ((cyclopropylmethyl) thio) -1, 7-dimethyl-3, 7-dihydro-1H-purine-2, 6-dione according to a similar procedure as 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
Figure BDA0002247439760001043
Compound 119 (white solid, 16 mg) was prepared from 8- ((cyclopropylmethyl) thio) -1, 7-dimethyl-3, 7-dihydro-1H-purine-2, 6-dione according to a similar procedure as 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
Figure BDA0002247439760001051
Compound 120 (white solid, 22mg, 85% yield) was prepared from 8- ((cyclopropylmethyl) thio) -1, 7-dimethyl-3, 7-dihydro-1H-purine-2, 6-dione according to a similar procedure as described for compound 109. 1 H-NMR(400MHz,CDCl 3 )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
Figure BDA0002247439760001052
Compound 121 (white solid, 16 mg) was prepared from 8- ((cyclopropylmethyl) thio) -1, 7-dimethyl-3, 7-dihydro-1H-purine-2, 6-dione according to a similar procedure as described for compound 109. 1 H-NMR(400MHz,CDCl 3 )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
Figure BDA0002247439760001053
Compound 122 (white solid, 22 mg) was prepared from 8- ((cyclopropylmethyl) thio) -1, 7-dimethyl-3, 7-dihydro-1H-purine-2, 6-dione according to a similar procedure as described for TC 013172. 1 H-NMR(400MHz,CDCl 3 )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
Figure BDA0002247439760001061
Compound 123 (yellow solid, 10 mg) was prepared from 8- ((cyclopropylmethyl) thio) -1, 7-dimethyl-3, 7-dihydro-1H-purine-2, 6-dione according to a similar procedure as 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
Figure BDA0002247439760001062
Compound 124 (yellow solid, 13 mg) was prepared from 8- ((cyclopropylmethyl) thio) -1, 7-dimethyl-3, 7-dihydro-1H-purine-2, 6-dione according to a similar procedure as described for compound 109. 1 H-NMR(400MHz,CDCl 3 )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
Figure BDA0002247439760001071
Compound 125 (yellow solid, 11 mg) was prepared from 8- ((cyclopropylmethyl) thio) -1, 7-dimethyl-3, 7-dihydro-1H-purine-2, 6-dione according to a similar procedure as described for compound 109. 1 H-NMR(400MHz,CDCl 3 )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 126:8- ((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
Figure BDA0002247439760001072
Compound 126 (white solid, 12 mg) was prepared from 8- ((cyclopropylmethyl) thio) -1, 7-dimethyl-3, 7-dihydro-1H-purine-2, 6-dione according to a similar procedure as described for compound 109. 1 H-NMR(400MHz,CDCl 3 )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
Figure BDA0002247439760001081
Compound 127 (white solid, 5 mg) was prepared from 8- ((cyclopropylmethyl) thio) -1, 7-dimethyl-3, 7-dihydro-1H-purine-2, 6-dione according to a similar procedure as 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
Figure BDA0002247439760001082
Compound 128 (white solid, 3 mg) was prepared from 8- ((cyclopropylmethyl) thio) -1, 7-dimethyl-3, 7-dihydro-1H-purine-2, 6-dione according to a similar procedure as 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
Figure BDA0002247439760001083
(I) Synthesis of 2- (bromomethyl) -1, 4-dimethoxybenzene
To CCl of 1, 4-dimethoxy-2-methylbenzene (500 mg,3.29 mmol) 4 To the solution (10 mL) was slowly added NBS (585 mg,3.29 mmol) and AIBN (150 mg). The mixture was then heated to 80 ℃ for 1h. The mixture was filtered and the filtrate was concentrated to give a residue. The residue was diluted, extracted with EA (3 x 10 ml) 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 (30.0 mg,0.14 mmol) and K 2 CO 3 To a solution of (16 mg) in anhydrous DMF (5 mL) was added 2- (bromomethyl) -1, 4-dimethoxybenzene (40 mg,0.18 mmol) and under nitrogenStir at room temperature overnight. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by column chromatography (PE: EA 1:1) 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 (30 mg,0.06 mmol) in DCM (2 mL) was added m-CPBA (30 mg,0.18 mmol). The mixture was then stirred at room temperature for 2h. The solvent was then removed and extracted with EA (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by silica gel column chromatography (PE: EA 5:1) to give 25mg (81%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760001091
To a solution of 1, 7-dimethyl-8- (methylthio) -3- (prop-2-yn-1-yl) -3, 7-dihydro-1H-purine-2, 6-dione (20 mg,0.08 mmol) in DMF (1 mL) was added azidobenzene (15 mg), 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 (3X 5 mL) and the organic layer was separated using Na 2 SO 4 Dried, filtered, concentrated and purified by column chromatography (DCM: methanol 10:1) to give a white solid. Intermediate (15 mg) was dissolved in THF (1 mL) and H 2 O (1 mL) with potassium hydrogen persulfate (50 mg). The mixture was then stirred at room temperature overnight. The resulting reaction was extracted with DCM (3X 5 mL)The organic layer was separated from the residue using Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (DCM: methanol 10:1) to give a white solid (5 mg). 1 H-NMR(400MHz,CDCl 3 )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- (thiophen-2-ylmethyl) -1H-purine-2, 6 (3H, 7H) -dione
Figure BDA0002247439760001101
(I) Synthesis of 2- (bromomethyl) thiophene
To a DME solution of thiophen-2-yl-methanol (1 g) was slowly added tribromophosphine (3.5 g), and the mixture was then stirred under nitrogen at room temperature for 2h. The mixture was then poured into ice and extracted with dichloromethane (3 x 10 ml), the organic layer was separated with Na 2 SO 4 Drying, filtration and concentration gave 1.04g (67.1%) as a red oil.
(II) Synthesis of 1, 7-dimethyl-8- (methylsulfonyl) -3- (thiophen-2-ylmethyl) -1H-purine-2, 6 (3H, 7H) -dione
To compound 99 (10 mg) and K 2 CO 3 To a solution of (6.5 mg) in anhydrous DMF (5 mL) was added 2- (bromomethyl) thiophene (8.3 mg) and stirred under nitrogen at room temperature for 2h. The reaction mixture was then poured into water and extracted with EA (3 x 5 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:1) to give 5.4mg (39.5%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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-yninamide
Figure BDA0002247439760001102
To compound 99 (10 mg) and K 2 CO 3 To a solution of (6.5 mg) in anhydrous DMF (5 mL) was added 4-bromo-N-ethylbut-2-ynamide (8.8 mg) and stirred under nitrogen at room temperature for 2h. The reaction mixture was then poured into water and extracted with EA (3 x 5 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC (PE: EA 1:1) to give 5.2mg (36.6%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760001111
(I) Synthesis of 1, 7-dimethyl-1H-purine-2, 6 (3H, 7H) -dione
A solution of ethyl 4-amino-1-methyl-1H-imidazole-5-carboxylate (2.0 g,11.83 mmol) and ethyl methylcarbamate (2.92 g,28.32 mmol) in anhydrous THF (2 mL) was reacted at 75deg.C under nitrogen for 30min. Potassium 2-methylpropyl-2-oleate (1.98 g,17.64 mmol) was then added to the mixture and reacted overnight at 75 ℃. The solvent was then removed and extracted with dichloromethane (3 x 50 ml), the organic layer was separated, and the solvent was extracted with Na 2 SO 4 Dried, filtered, concentrated and purified by silica gel chromatography (DCM: meoh=20:1) to give a white solid (1.2 g, 56.33%). 1 H-NMR(400MHz,DMSO-d 6 ):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 (0.53 g,2.94 mmol) in anhydrous THF (10 mL) was added NCS (0.59 g,4.36 mmol) and stirred under nitrogen overnight at room temperature. The solvent was then removed and extracted with dichloromethane (3 x 20 ml), the organic layer was separated, and the solvent was removed using Na 2 SO 4 Drying, filtering, concentrating to obtainWhite solid (0.45 g). MS (m/z): 215.03[ M+H ]]+.
Synthesis of (III) 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 (500 mg,2.2 mmol) in anhydrous DMF was added sodium bisulfide (370 mg,6.5 mmol) and heated to 100deg.C for 12 hours. The reaction mixture was changed to pH 5 and slowly poured into water (10 mL) to give a yellow precipitate and filtered to finally give the crude product. 1 H-NMR(400MHz,DMSO-d 6 ):δ3.68(s,3H),3.38(s,3H),3.20(s,3H).MS(m/z):227.2[M+H]+.
Synthesis of (IV) 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 (1 g,4.7 mmol) and K 2 CO 3 To a solution of (1.32 g,9.4 mmol) in anhydrous DMF (15 mL) was added (bromomethyl) cyclopropane (580 mg,4.24 mmol) and stirred under nitrogen at room temperature for 2h. The reaction mixture was then poured into water and extracted with EA (3 x 10 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Dried, filtered, concentrated and purified by column chromatography (PE: EA 1:1) to give 1.06g (88.8%) as a white solid. MS (m/z): 267.3[ M+H ]]+.
Synthesis of (V) 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 (1 g,3.76 mmol) in DCM (10 mL) was added m-CPBA (1.95 g,11.3 mmol). The mixture was then stirred at room temperature for 2h. The solvent was then removed and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was extracted with Na 2 SO 4 Dried, filtered, concentrated and purified by column chromatography (PE: EA 1:1) to give a white solid (800 mg, 71.4%). MS (m/z): 299.32[ M+H ]]+.
Synthesis of (VI) 3- (2-chlorobenzyl) -8- ((cyclopropylmethyl) sulfonyl) -1, 7-dimethyl-1H-purine-2, 6 (3H, 7H) -dione
To 8- ((cyclopropylmethyl) sulfonyl) -1, 7-dimethyl-1H-purine-2, 6 (3H, 7H) -dione (20 mg,0.068 mmol) and K 2 CO 3 To a solution of (19 mg,0.134 mmol) in anhydrous DMF (2 mL) was added1- (bromomethyl) -2-fluorobenzene (21 mg,0.101 mmol) was added and stirred under nitrogen at room temperature for 0.5h. The reaction mixture was then poured into water and extracted with EA (3 x 5 ml), the organic layer was separated, and the solvent was removed with Na 2 SO 4 Drying, filtration, concentration and purification by column chromatography (PE: EA 5:1) gave finally 16mg (60%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760001121
The compound was synthesized in a similar manner as described for compound 133, resulting in 18mg (66.7%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760001131
The compound was synthesized in a similar manner as described for compound 133, resulting in 16mg (59.3%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760001132
The compound was synthesized in a similar manner as described for compound 133, eventually obtaining 27mg (96.4%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760001141
The compound was synthesized in a similar manner as described for compound 133, resulting in 20mg (71.4%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760001142
The compound was synthesized in a similar manner as described for compound 133, resulting in 16mg (58%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760001143
The compound was synthesized in a similar manner as described for compound 133, resulting in 25mg (80.6%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760001151
The compound was synthesized in a similar manner as described for compound 133, resulting in 22mg (71%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760001152
The compound was synthesized in a similar manner as described for compound 133, resulting in 25mg (80.6%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760001153
The compound was synthesized in a similar manner as described for compound 133, resulting in 16mg (61.5%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760001161
The compound was synthesized in a similar manner as described for compound 133, resulting in 16mg (61.5%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760001162
The compound was synthesized in a similar manner as described for compound 133, eventually yielding 18mg (69.2%) as a white solid. 1 H-NMR(400MHz,CDCl 3 ):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
Figure BDA0002247439760001163
(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 (2.0 g,11.45 mmol) in isopropanol (6 mL) was added hydrazine hydrate (6 mL) and stirred at room temperature overnight. The precipitated white solid was then filtered off and washed with water (3 x 15 ml) and dried well to give TM (1.67 g, 85.6%) as a white solid. 1 H-NMR(400MHz,DMSO-d 6 ):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
A mixture of 6-hydrazino-1, 3-dimethylpyrimidine-2, 4 (1H, 3H) -dione (1.6 g,9.40 mmol) and acetic anhydride (10 mL) was refluxed in anhydrous pyridine (12 mL) for 3h. The reaction was then cooled to 0deg.C and acidified with 1N HCl (30 mL). The solid obtained was collected by filtration, washed with 1N HCl (2 x 5 ml), water (2 x 10 ml) and dried to give TM (1.1 g, 60.5%) as a white solid. 1 H-NMR(400MHz,DMSO-d 6 ):δ3.64(br,1H),3.19(s,3H),2.84(s,3H),2.65(s,3H).MS(m/z):195.08[M+H]+.
Synthesis of (III) 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 (50.0 mg,0.26 mmol) in NaOH (aqueous solution) (10 mL) was added methanesulfonyl chloride (44.2 mg,0.38 mmol). The mixture was then stirred at room temperature overnight. The solid precipitated out. The crude product was purified by preparative TLC to give TM (31.2 mg, 44.6%) as a white solid. 1 H-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
Figure BDA0002247439760001171
To a solution of 5, 6-diamino-1, 3-dimethylpyrimidine-2, 4 (1H, 3H) -dione (500 mg,3 mmol) in 95% ethanol (3 mL) was added methyl isothiocyanate (215 mg) and stirred at 100℃for 6h. The precipitated white solid was then filtered off and washed with water (3×15 ml), 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 (0.04 mol) 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 (20 mg) was dissolved in anhydrous DMF (2 mL) and K was added 2 CO 3 (50 mg) and methyl iodide (25 mg) and stirred at room temperature under nitrogen for 2h. The reaction mixture was poured into water and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give 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- (methylsulfanyl) -3, 9-dihydro-1H-purine-2, 6-dione (12 mg) in MeOH (3 mL) was added potassium hydrogen persulfate (92 mg) H 2 O (3 mL) solution. The mixture was then stirred at room temperature for 5h. The solvent was then removed and extracted with dichloromethane (3 x 10 ml), the organic layer was separated, and the solvent was extracted with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give 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 schemes:
Figure BDA0002247439760001181
(a)K 2 CO 3 ,DMF,rt,1h;(b)K 2 CO 3 ,DMF,rt,1h;(c)Pd(PPh 3 ) 4 CuI, TEA, anhydrous DMF,60℃for 6h; (d) Potassium hydrogen persulfate, meOH, H2O 1:1, rt,4H.
Compound 148 was prepared according to the procedure outlined in the schemes:
Figure BDA0002247439760001182
(a) Sodium ethoxide, ethanol, reflux; (b) potassium tert-butoxide, anhydrous THF,75 ℃, overnight; (c) K2CO3, DMF, rt,1h; (d) Pd (PPh) 3 ) 4 CuI, TEA, anhydrous DMF,60℃for 6h; (e) Potassium hydrogen persulfate, meOH, H2O 1:1, rt,4H.
Compound 149 was prepared according to the procedure outlined in the schemes
Figure BDA0002247439760001191
(a)DMF,60℃,3h;(b)K 2 CO 3 ,DMF,rt,1h;(c)Pd(PPh 3 ) 4 CuI, TEA, anhydrous DMF,60℃for 6h; (d) Potassium hydrogen persulfate, meOH, H2O 1:1, rt,4H.
Compounds 150-155 were prepared according to the methods outlined in the schemes
Figure BDA0002247439760001192
(a)K 2 CO 3 ,DMF,rt,1h;(b)K 2 CO 3 ,DMF,rt,1h;(c)Pd(PPh 3 ) 4 CuI, TEA, anhydrous DMF,60℃for 6h; (d) Potassium hydrogen persulfate, meOH, H2O 1:1, rt,4H.
Compounds 156-158 were prepared according to the methods outlined in the schemes
Figure BDA0002247439760001193
(a) Anhydrous DMF at 120℃for 6h; (b) NaOCl (aqueous solution), HCl, -25 ℃; (c) K (K) 2 CO 3 ,DMF,rt,1h.
Compounds 159-175 were prepared according to the procedure outlined in the schemes:
Figure BDA0002247439760001194
(a)K 2 CO 3 ,DMF,rt,1h;(b)K 2 CO 3 ,DMF,rt,1h;(c)Pd(PPh 3 ) 4 CuI, TEA, anhydrous DMF,60℃for 6h; (d) Potassium hydrogen persulfate, meOH H 2 O 1:1,rt,4h.
Compound 176 was prepared according to the procedure outlined in the schemes
Figure BDA0002247439760001201
(a)Pd(PPh 3 ) 4 CuI, TEA, anhydrous DMF,60℃for 6h; (b) K (K) 2 CO 3 DMF, rt,1h; (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 in 96-well assay plates at a density of 2500-3500 cells per well. The next day necrosis was induced by the addition of 20ng/ml TNF-a (T), 100nM Smac mimetic (S) and 20mM Z-VAD (Z). Simultaneously, 10mM of a compound from a chemical library of about 200000 compounds was delivered 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 analysis (Promega) was performed according to the manufacturer's instructions. Luminescence was recorded with a PerkinElmer EnSpire multimode plate reader. Surviving cells were normalized to those treated with DMSO. RIPA-56 was used as a positive control for screening for inhibitors of necrotic apoptosis. Data are expressed as mean ± standard deviation of duplicate data.
Series 1 compound necrotic apoptotic activity; the activity of 147-176 was extrapolated.
Figure BDA0002247439760001202
/>
Figure BDA0002247439760001211
Exemplary compounds; in an embodiment, the compound is selected from table 2.
TABLE 2 series 2 Compounds
Figure BDA0002247439760001221
/>
Figure BDA0002247439760001231
/>
Figure BDA0002247439760001241
/>
Figure BDA0002247439760001251
/>
Figure BDA0002247439760001261
/>
Figure BDA0002247439760001271
/>
Figure BDA0002247439760001281
Figure BDA0002247439760001291
/>
Series 2; compound 1:
8- ((2-hydroxyethyl) sulfonyl) -1,3, 7-trimethyl-3, 7-dihydro-1H-purine-2, 6-dione
Figure BDA0002247439760001292
Step 1.
To a solution of 8-chloro-1, 3-dimethyl-3, 7-dihydro-1H-purine-2, 6-dione (428 mg,2.0 mmol) in DMF (6 mL) was added MeI (188. Mu.L, 3.0 mmol) and K 2 CO 3 (414 mg,3.0 mmol). The mixture was then stirred at room temperature for 3 hours. With EtOAc and H 2 The reaction mixture was extracted 3 times. The organic layers were combined, washed with brine, and dried over Na 2 SO 4 Dried, concentrated and further purified by silica gel column chromatography (DCM/meoh=200/1) to give 409mg 1-1 as a yellow solid (90%). LC-MS (ESI) M/z: [ M+H ]] + Calculated 229.04, measured 229.06
And 2, step 2.
Will NaSCH 3 (170 mg,3.0 mmol) was added to a solution of 1-1 (455 mg,2.0 mmol) 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 1-2 (88%). LC-MS (ESI) M/z: [ M+H ]] + Calculated 227.05, measured 227.03
And 3, step 3.
To 1-2 (40 mg,0.18 mmol) and K 2 CO 3 To a solution of (61 mg,0.44 mmol) in anhydrous DMF (2 mL) was added 2-bromoethanol (23. Mu.L, 0.33 mmol) and stirred under nitrogen at room temperature for 3 hours. The reaction mixture was poured into water and extracted with ethyl acetate (3×5 mL), the organic layer was separated, taken up in Na 2 SO 4 Drying, 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, measured 271.06
And 4, step 4.
To a solution of 1-3 (47 mg,0.18 mmol) in MeOH (2 mL) was added potassium hydrogen persulfate (214 mg,0.35 mmol) H 2 O (2 mL) solution. The mixture was then stirred at room temperature for 3 hours. The solvent was then removed and extracted with dichloromethane (3×5 mL), the organic layer was separated, and the solvent was removed using Na 2 SO 4 Drying, filtration, concentration and purification by preparative TLC gave compound 1.7mg (60%) as a white solid. 1 H 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, measured 303.16
Compound 2:
8- ((2-methoxyethyl) sulfonyl) -1,3, 7-trimethyl-3, 7-dihydro-1H-purine-2, 6-dione
Figure BDA0002247439760001301
Step 1.
2-1 was prepared according to the method of 1 from 1-2 (40 mg,0.175 mmol) and 1-bromo-2-methoxyethane (31. Mu.L, 0.33 mmol) in 100% yield (50 mg). LC-MS (ESI) M/z: [ M+H ]] + Calculated 285.09, measured 285.13
And 2, step 2.
Preparation of 2 from 2-1 (50 mg,0.18 mmol) and potassium hydrogen persulfate (214 mg,0.35 mmol) according to the method of 1 gave a yield of 43% (24 mg) as a yellow solid. 1 H 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, measured 317.20
Compound 3:
8- ((3-hydroxypropyl) sulfonyl) -1,3, 7-trimethyl-3, 7-dihydro-1H-purine-2, 6-dione
Figure BDA0002247439760001302
Step 1.
3-1 was prepared according to the method of compound 1 from 1-2 (40 mg,0.175 mmol) and 3-bromopropan-1-ol (25 μl,0.26 mmol) in 100% yield (49 mg). LC-MS (ESI) M/z: [ M+H ]] + Calculated 285.09, measured 285.13
And 2, step 2.
According to the method of 1, from 3-1 (49 mg,0.18 mmol) and potassium hydrogen persulfate (214 mg,0.35 mmol) preparation 3 in 32% yield (17.8 mg) as yellow solid. 1 H 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, measured 317.20
Compound 4:
8- ((cyclopropylmethyl) sulfonyl) -7-ethyl-1, 3-dimethyl-1H-purine-2, 6 (3H, 7H) -dione
Figure BDA0002247439760001311
Step 1.
To a solution of 8-chloro-1, 3-dimethyl-3, 7-dihydro-1H-purine-2, 6-dione (500 mg,2.3 mmol) in DMF (10 mL) was added iodoethane (437 mg,2.8 mmol) and K at room temperature 2 CO 3 (483 mg,3.5 mmol) and then the mixture was stirred at room temperature overnight. UPLC-MS monitors the completion of the reaction. The solvent was then evaporated to give crude 566mg of 4-1 as a yellow solid without further purification. LC-MS (ESI) M/z: [ M+H ]] + Calculated 243.06, measured 243.12
And 2, step 2.
To a solution of 4-1 (566 mg,2.3 mmol) in DMF (10 mL) was added NaSH (170 mg,3.03 mmol) at room temperature. The mixture was then stirred at 110℃for 2 hours. UPLC-MS monitors the completion of the reaction. The solvent was then evaporated to give crude 500mg of 4-2 as a yellow solid without further purification. LC-MS (ESI) M/z: [ M+H ] ] + Calculated 241.07, measured 241.01
And 3, step 3.
To a solution of 4-2 (200 mg,0.83 mmol) in DMF (3 mL) at room temperature was added (bromomethyl) cyclopropane (135 mg,1.0 mmol) and K 2 CO 3 (173 mg,1.25 mmol) and then the mixture was stirred at room temperature for 2 hours. UPLC-MS monitors the completion of the reaction. 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 Na 2 SO 4 The 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, yield: 95%. LC-MS (ESI) M/z: [ M+H ]] + Calculated 295.12, measured 295.20
And 4, step 4.
To 4-3 (100 mg,0.34 mmol) of methanol (3 mL) and H 2 To a solution of O (1 mL) was added potassium hydrogen persulfate (626 mg,1.2 mmol). The mixture was then stirred at room temperature for 20 hours. UPLC-MS monitors the completion of the reaction. 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 Na 2 SO 4 The combined extracts were dried and concentrated. The resulting residue was purified by silica gel column chromatography (PE: ea=5:1) to give 4 of 72mgd as a yellow solid, yield: 64.9%.1H NMR (400 MHz, 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, measured 327.37.
Compound 5:
7- (2-hydroxyethyl) -1, 3-dimethyl-8- (methylsulfonyl) -3, 7-dihydro-1H-purine-2, 6-dione
Figure BDA0002247439760001321
Step 1.
To a solution of 8-chloro-1, 3-dimethyl-3, 7-dihydro-1H-purine-2, 6-dione (214 mg,1.0 mmol) in DMF (5 mL) was added 2-bromoethanol (106. Mu.L, 1.5 mmol) and K 2 CO 3 (276 mg,2.0 mmol). The mixture was heated to 110 ℃ for 1 hour by microwave radiation. Evaporation of the solvent under reduced pressure gave 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, measured 259.11.
And 2, step 2.
Will NaSCH 3 (105 mg,1.5 mmol) 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, which was further purified by silica gel column chromatography (DCM/MeOH=100/1) to give 5-2 as a white solid (34% in two steps). LC-MS (ESI) M/z: [ M+H ]] + Calculated 271.08, measured values271.01.
And 3, step 3.
Prepared 5 from 5-2 (40 mg,0.15 mmol) and potassium hydrogen persulfate (272 mg,0.44 mmol) according to the method of 1 in 46% yield (21.0 mg) as a white solid. 1 H 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, measured 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
Figure BDA0002247439760001331
Step 1.
6-1 was prepared according to the procedure for compound 1 from 8-chloro-1, 3-dimethyl-1H-purine-2, 6 (3H, 7H) -dione (214 mg,1.0 mmol) and 1-bromo-2-methoxyethane. 6-1 was used in the next reaction step without any further purification. LC-MS (ESI) M/z: [ M+H ]] + Calculated 273.07, measured 273.13
And 2, step 2.
According to method 1, from 6-1 (2724 mg,1.0 mmol) and NaSCH 3 (105 mg,1.5 mmol) preparation 6-2 in 36% yield (two steps). LC-MS (ESI) M/z: [ M+H ]] + Calculated 285.09, measured 285.13
And 3, step 3.
According to the method of 1, 6 was prepared from 6-2 (80 mg,0.28 mmol) and potassium hydrogen persulfate (522 mg,0.85 mmol) in 12% yield (10.0 mg) as a white solid. 1 H 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, measured 317.24.
Preparation 7 was carried out according to the method of 1 from 6-2 (80 mg,0.28 mmol) and potassium hydrogen persulfate (522 mg,0.85 mmol) in 10% yield (9.2 mg) as a white solid. 1 H 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
Figure BDA0002247439760001341
According to the method of compound 1, 8 was prepared from 3- (3- (3-hydroxyphenyl) prop-2-yn-1-yl) -1, 7-dimethyl-8- (methylsulfanyl) -3, 7-dihydro-1H-purine-2, 6-dione (17 mg,0.048 mmol) and potassium hydrogen persulfate (73 mg,0.12 mmol) in 55% (9.8 mg) as a yellow solid. 1 H 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, measured 373.13
Compound 9:3- (3- (1H-indol-6-yl) prop-2-yn-1-yl) -1, 7-dimethyl-8 (methylsulfonyl) -1H-purine-2, 6 (3H, 7H) -dione
Figure BDA0002247439760001342
Step 1.
At N 2 To a solution of 1, 7-dimethyl-1H-purine-2, 6 (3H, 7H) -dione (748 mg,4.2 mmol) in THF (20 mL) was added NCS (832 mg,6.3 mmol) under an atmosphere. The mixture was then stirred at room temperature overnight. UPLC-MS monitors the completion of the reaction. 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 Na 2 SO 4 The 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%. LC-MS (ESI) M/z: [ M+H ]] + Calculated 215.03, measured 215.09
And 2, step 2.
To a solution of 9-1 (1.33 g,6.2 mmol) in DMF (20 mL) was added 3-bromoprop-1-yne (1.47 g,12.4 mmol) and K at room temperature 2 CO 3 (1.71 g,12.4 mmol) and then the mixture was stirred at room temperature for 2 hours. UPLC-MS monitors the completion of the reaction. 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 Na 2 SO 4 The 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%. LC-MS (ESI) M/z: [ M+H ] ] + Calculated 253.03, measured 253.05.
And 3, step 3.
To a solution of 9-2 (200 mg,0.80 mmol) in DMF (5 mL) was added NaSMe (67 mg,0.95 mmol) at room temperature. The mixture was then stirred at room temperature for 2 hours. UPLC-MS monitors the completion of the reaction. 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 Na 2 SO 4 The 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, yield: 90%. LC-MS (ESI) M/z: [ M+H ]] + Calculated 264.07, measured 264.12
And 4, step 4.
To 9-3 (271mg, 0.79 mmol) of methanol (9 mL) and H 2 To a solution of O (3 mL) was added potassium hydrogen persulfate (1.46 g,2.38 mmol). The mixture was then stirred at room temperature for 20 hours. UPLC-MS monitors the completion of the reaction. 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 Na 2 SO 4 The combined extracts were dried and concentrated. The resulting residue was purified by silica gel column chromatography (PE: ea=5:1) to give 205mg of 9-4 as a yellow solid, yield: 87.2%. LC-MS (ESI) M/z: [ M+H ]] + Calculated 297.06, measured 297.12
And 5, step 5.
At N 2 To a solution of 9-4 (40 mg,0.14 mmol) in DMF (2 mL) was added 6-iodo-1H-indole (40 mg,0.16 mmol), TEA (33 mg,0.32 mmol), pd (PPh) under an atmosphere 3 ) 4 (8 mg, 0.0071 mmol) and CuI (3 mg,0.014 mmol). The mixture was then degassed for 15min. The mixture was then stirred at 55℃for 4 hours. UPLC-MS monitors the completion of the reaction. The reaction is carried outThe 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 Na 2 SO 4 The 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, yield: 36.4%.1H NMR (400 MHz, DMSO). Delta.11.23 (s, 1H), 7.50 (d, J=8 Hz, 1H), 7.44-7.42 (M, 2H), 6.99 (d, J=8 Hz, 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, measured 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
Figure BDA0002247439760001361
Step 1.
10-1 was prepared according to the method of compound 9 from 9-4 (50 mg,0.17 mmol) and 3-iodoaniline (45 mg,0.20 mmol) in 74% yield (48 mg). LC-MS (ESI) M/z: [ M+H ]] + Calculated 388.10, measured 388.19
And 2, step 2.
To a solution of 10-1 (20 mg,0.052 mmol) in DCM (2 mL) at 0deg.C was added TEA (12 mg,0.11 mmol) and Ac 2 O (7 mg,0.062 mmol). The mixture was then stirred at room temperature for 3 hours. UPLC-MS monitors the completion of the reaction. 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 Na 2 SO 4 The combined extracts were dried and concentrated. The resulting residue was purified by silica gel column chromatography (PE: ea=1:1) to give 14mg of 10 as a yellow solid, yield: 63.6%.1H NMR (400 MHz, DMSO). Delta.10.00 (s, 1H), 7.74 (t, J=4 Hz, 1H), 7.48-7.45 (M, 1H), 7.27 (t, J=8 Hz, 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)] + Calculated 430.41, measured 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
Figure BDA0002247439760001362
According to the method for compound 10, 11 was prepared from 10-1 (24 mg,0.062 mmol), TEA (14 mg,0.14 mmol) and benzoyl chloride (11 mg,0.074 mmol) in 55% yield (16 mg). 1 H 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, measured 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
Figure BDA0002247439760001371
Step 1.
To a solution of tert-butyl (3-iodophenyl) carbamate (1 eq.) in THF at 0 ℃ NaH (1.2 eq.) was added and stirred at room temperature for 30min. 1-bromo-2- (2-methoxyethoxy) ethane (1.2 eq) was then added to the reaction mixture and stirred overnight. UPLC-MS monitors the completion of the reaction. 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 Na 2 SO 4 The 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 0deg.C was added TFA (5 eq.). The mixture was then stirred at room temperature for 4 hours. UPLC-MS monitors the completion of the reaction. 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 Na 2 SO 4 The 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 yellowColor solids.
And 3, step 3.
At N 2 9-4 (1.2 eq), TEA (2.4 eq), pd (PPh) were added to a solution of 12-2 (1 eq) in DMF under an atmosphere 3 ) 4 (0.05 equivalent) and CuI (0.1 equivalent). The mixture was then stirred at 55℃for 4 hours. UPLC-MS monitors the completion of the reaction. 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 Na 2 SO 4 The 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. 1 H NMR(400MHz,CDCl 3 )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, measured 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
Figure BDA0002247439760001381
13 is prepared according to the method of 12. 1H NMR (400 MHz, DMSO). Delta.7.31 (s, 2H), 7.30 (d, J=4 Hz, 2H), 7.00 (t, J=8 Hz, 1H), 6.60-6.52 (M, 3H), 6.70 (t, J=4 Hz, 1H), 4.94 (s, 2H), 4.23 (d, J=8 Hz, 2H), 4.19 (s, 3H), 3.52 (s, 3H), 3.26 (s, 3H). LC-MS (ESI) M/z: LC-MS (ESI) M/z: [ M+H)] + 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
Figure BDA0002247439760001382
Preparation 14 according to the method of 12. 1 H 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, measured 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
Figure BDA0002247439760001391
Preparation 15 was according to the method of 12. 1 H NMR(400MHz,CDCl 3 )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, measured 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
Figure BDA0002247439760001392
Preparation 16 was according to the method of 12. 1 H NMR(400MHz,CDCl 3 )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, measured 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
Figure BDA0002247439760001393
Step 1.
At room temperature under N 2 2-morpholinoacetic acid (100 mg, To a solution of 0.69 mmol) in DMF (2 mL) was added 3-iodoaniline (151 mg,0.69 mmol), EDCI (199mg, 1.03 mmol) and DMAP (127 mg,1.03 mmol). The mixture was then stirred at 55℃for 4 hours. UPLC-MS monitors the completion of the reaction. 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 Na 2 SO 4 The 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, yield: 89.9%. LC-MS (ESI) M/z: [ M+H ]] + Calculated 347.02, measured 347.14.
Step 2 Sonogashira coupling
Preparation 17 was prepared according to the method of 12 in 31% yield (16 mg). 1 H NMR(400MHz,CDCl 3 )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
Figure BDA0002247439760001401
/>
Preparation 18 from 3-iodo-N- (2- (piperidin-1-yl) ethyl) aniline (37 mg,0.11 mmol) according to method 12, yield 17% (12 mg) as yellow solid. 1 H 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, measured 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
Figure BDA0002247439760001402
According to the method of 12, 19 was prepared from 3-iodo-N- (2-morpholinoethyl) aniline (30 mg,0.09 mmol) in 45% yield (18 mg) as yellow solid. 1 H 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, measured 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
Figure BDA0002247439760001411
Preparation 20 was prepared according to the method of 17 in 31% yield (16 mg) as yellow solid. 1 H NMR(400MHz,CDCl 3 )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
Figure BDA0002247439760001412
/>
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 was prepared 21 in 9.4% (6 mg) as a yellow solid. 1H NMR (400 MHz, 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
Figure BDA0002247439760001421
Prepared according to the method of 12 from N- (4-chloro-3-iodophenyl) -2-morpholinoacetamide and 8- ((cyclopropylmethyl) sulfonyl) -1, 7-dimethyl-3- (prop-2-yn-1-yl) -1H-purine-2, 6 (3H, 7H) -dione in 16% (12 mg) yield as yellow solid. 1 H 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)
Figure BDA0002247439760001422
According to the method of 12, from 5-iodo-1, 3-dihydro-2H-benzo [ d ]]Imidazol-2-one (22 mg,0.09 mmol) was prepared 23 in 2% yield (7 mg) as a yellow solid. 1 H 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, measured 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)
Figure BDA0002247439760001423
Preparation of 24 from 4-iodopyrimidin-2-amine according to method 232The rate was 36% (16 mg) as a yellow solid. 1 H 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, measured 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) diacet-amide
Figure BDA0002247439760001431
Step 1.
To a solution of 1-iodo-3, 5-dinitrobenzene (400 mg,1.36 mmol) in ethanol (10 mL) was added SnCl at room temperature 2 .(H 2 O) 2 (1.84 g,8.16 mmol). The mixture was then stirred at 70℃for 2 hours. UPLC-MS monitors the completion of the reaction. 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 Na 2 SO 4 The 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%. LC-MS (ESI) M/z: [ M+H ]] + Calculated 234.97, measured 235.02
And 2, step 2.
To a solution of 25-1 (50 mg,0.21 mmol) in DCM (3 mL) at 0deg.C was added acetic anhydride (66 mg,0.64 mmol) and TEA (87 mg,0.86 mmol). The mixture was then stirred at room temperature for 1 hour. UPLC-MS monitors the completion of the reaction. 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 Na 2 SO 4 The 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%. LC-MS (ESI) M/z: [ M+H ]] + Calculated 318.99, measured 319.03
And 3, step 3.
At N 2 To 1, 7-dimethyl-8- (methylsulfonyl) -3 under an atmosphereTo a solution of- (prop-2-yn-1-yl) -1H-purine-2, 6 (3H, 7H) -dione (25 mg,0.084 mmol) in DMF (2 mL) was added 25-2 (27 mg,0.084 mmol), TEA (21 mg,0.202 mmol), pd (PPh) 3 ) 4 (4.9 mg,0.004 mmol) and CuI (1.6 mg,0.008 mmol). The mixture was then stirred at 60℃for 4 hours. UPLC-MS monitors the completion of the reaction. 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 Na 2 SO 4 The 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, yield: 20.7%.1H NMR (400 MHz, DMSO). Delta.10.00 (s, 2H), 7.76 (s, 1H), 7.38 (d, J=4 Hz, 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)] + Calculated 487.42, measured 487.50
Compound 26: 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) bis (2-morpholinylacetamide
Figure BDA0002247439760001441
Step 1.
At room temperature under N 2 To a solution of 2-morpholinoacetic acid (62 mg,0.43 mmol) in DMF (2 mL) was added 25-1 (50 mg,0.21 mmol), EDCI (124 mg,0.64 mmol) and DMAP (79 mg,0.64 mmol) under an atmosphere. The mixture was then stirred at 55℃for 4 hours. UPLC-MS monitors the completion of the reaction. 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 Na 2 SO 4 The combined extracts were dried and concentrated. The resulting residue was purified by silica gel column chromatography (PE: ea=3:1) to give 21mg 26-1 as a yellow solid, yield: 20.2%. LC-MS (ESI) M/z: [ M+H ]] + Calculated 489.09, measured 489.41
And 2, step 2.
At N 2 To a solution of 1, 7-dimethyl-8- (methylsulfonyl) -3- (prop-2-yn-1-yl) -1H-purine-2, 6 (3H, 7H) -dione (16 mg,0.053 mmol) in DMF (2 mL) was added 26-1 (21 mg,0.043 mmol) under an atmosphere,TEA(11mg,0.10mmol)、Pd(PPh 3 ) 4 (2.5 mg,0.002 mmol) and CuI (0.82 mg,0.04 mmol). The mixture was then stirred at 55℃for 4 hours. UPLC-MS monitors the completion of the reaction. 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 Na 2 SO 4 The 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, yield: 9.2%.1H NMR (400 MHz, 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, measured 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) bicyclo propane carboxamide
Figure BDA0002247439760001451
Step 1.
To a solution of 5-iodobenzene-1, 3-diamine (60 mg,0.26 mmol) in DCM (5 mL) was added cyclopropanecarbonyl chloride (116. Mu.L, 0.64 mmol), et 3 N (98. Mu.L, 0.64 mmol) and then the mixture was stirred at room temperature for 3 hours. UPLC-MS monitors the completion of the reaction. The solvent was evaporated under reduced pressure and further purified by silica gel column chromatography (PE/ea=60/40) to give 40mg 27-1 as a yellow solid (42%). LC-MS (ESI) M/z: [ M+H ] ] + Calculated 371.01, measured 371.18.
Step 2 Sonogashira coupling
Preparation 27 from 27-1 (56 mg,0.15 mmol) was prepared according to the method of 12 in 18% yield (10 mg) as yellow solid. 1 H 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, measured 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
Figure BDA0002247439760001461
Prepared 28 from N, N' - (5-iodo-1, 3-phenylene) bis (2-morpholinylacetamide) (48 mg,0.11 mmol) according to method 22 in 10% yield (6.2 mg) as yellow solid. 1 H 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, measured 697.35.
Compound 29:3- (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
Figure BDA0002247439760001462
Preparation 29 from 5-iodo-N1, N3-bis (2-morpholinoethyl) benzene-1, 3-diamine (66 mg,0.15 mmol) according to method 12 gave 10% yield (6.1 mg) as yellow solid. 1 H 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, measured 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) dicyclohexyl propane carboxamide
Figure BDA0002247439760001471
Preparation 30 according to the method of 12 in 10% yield (5.0 mg) as yellow solid. 1 H 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, measured 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
Figure BDA0002247439760001472
Preparation 31 was prepared according to the method of 12 in 27% yield (10 mg) as yellow solid. 1 H 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, measured 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
Figure BDA0002247439760001473
Preparation 32 according to the method of 12 in 15% yield (6 mg) as yellow solid. 1 H 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, measured 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
Figure BDA0002247439760001481
Stirring at room temperature under argon atmosphere8- ((cyclopropylmethyl) sulfonyl) -1, 7-dimethyl-3- (prop-2-yn-1-yl) -1H-purine-2, 6 (3H, 7H) -dione (25 mg,75 nmol), N-ethyl-4-iodopyridin-2-amine (28 mg,0.11 mmol), pdCl 2 (PPh 3 ) 2 (5.3 mg,7.5 nmol), copper (I) iodide (0.7 mg,3.75 nmol) in DMF (3 mL) for 5min. Triethylamine (0.25 ml,0.75 mmol) 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 as a pale yellow solid in 66% yield. 1 H 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, measured 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
Figure BDA0002247439760001482
According to the method of 12, 34 was prepared from 8- ((cyclopropylmethyl) sulfonyl) -1, 7-dimethyl-3- (prop-2-yn-1-yl) -1H-purine-2, 6 (3H, 7H) -dione (25 mg,75 nmol) and N- (cyclopropylmethyl) -4-iodopyridin-2-amine (30 mg,0.11 mmol) in 63% (36 mg) as a pale yellow solid. 1 H 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, measured 483.25.
Compound 35:8- ((cyclopropylmethyl) sulfonyl) -3- (3- (2- (isopropylamino) pyridin-4-yl) prop-2-yn-1-yl) -1, 7-dimethyl-1H-purin-2, 6 (3H, 7H) -dione
Figure BDA0002247439760001491
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 (25 mg,75 nmol) and 4-iodo-N-isopropylpyridin-2-amine (29 mg,0.11 mmol) in 30% (10.5 mg) yield as a pale yellow solid. 1 H 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, measured 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
Figure BDA0002247439760001492
Step 1.
At room temperature under N 2 To a solution of cyclopropanecarboxylic acid (85 mg,1.0 mmol) in DMF (8 mL) was added 5-iodobenzene-1, 3-diamine (234 mg,1.0 mmol), EDCI (192 mg,1.0 mmol) and DMAP (122 mg,1.0 mmol) under an atmosphere. The mixture was then stirred at room temperature for 5 hours. UPLC-MS monitors the completion of the reaction. 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 Na 2 SO 4 The 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 (47 mg in two steps) as a yellow solid.
And 3, step 3.
Preparation 36 was prepared according to the method of 12 in 12% yield (6 mg) as yellow solid. 1 H 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] + Calculated 598.20, measured 598.27.
Compound 37:3- (3, 5-bis ((cyclopropylmethyl) amino) phenyl) prop-2-yn-1-yl) -1, 7-dimethyl-8- (methylsulfonyl) -3, 7-dihydro-1H-purine-2, 6-dione
Figure BDA0002247439760001501
Preparation 37 according to the method of 12 gave a yield of 12% (6.2 mg) as a yellow solid. 1 H 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, measured 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
Figure BDA0002247439760001502
Step 1.
38-1 was prepared from 36-1 (80 mg, 0.27) according to the method of 36 in 38% (38 mg) yield as a yellow solid.
And 2, step 2.
Preparation 38 was prepared according to the method of 12 in 30% yield (13 mg) as yellow solid. 1 H 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, measured 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
Figure BDA0002247439760001511
Step 1.
39-1 was prepared from 36-1 (80 mg, 0.27) according to the method of 36 in 39% (40 mg) yield as a yellow solid.
And 2, step 2.
Preparation 39 was prepared according to the method of 12 in 9% yield (5 mg) as yellow solid. 1 H 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, measured 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
Figure BDA0002247439760001521
According to the method of 12, 40 was prepared from 1, 7-dimethyl-8- (methylsulfonyl) -3- (prop-2-yn-1-yl) -1H-purine-2, 6 (3H, 7H) -dione (39.5 mg,0.13 mmol) and N- (3-amino-5-iodophenyl) cyclopropanecarboxamide (60.4 mg,0.2 mmol) in 46% yield (28.6 mg) as a white solid. 1 H 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, measured 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
Figure BDA0002247439760001522
Preparation 41 was prepared according to the method of 12 in 13% yield (5.8 mg). 1 H 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, measured 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
Figure BDA0002247439760001523
Preparation 42 was prepared according to the method of 12 in 12% (5.5 mg) yield. 1 H 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, measured 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
Figure BDA0002247439760001531
Preparation 43 according to the method of 12 in 40% yield (16 mg). 1 H 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, measured 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
Figure BDA0002247439760001532
Preparation 44 according to the method of 12 gave a yield of 36% (15.9 mg). 1 H 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, measured 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
Figure BDA0002247439760001541
Preparation 45 according to the method of 12 gave a yield of 32% (18 mg). 1 H 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, measured 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
Figure BDA0002247439760001542
Preparation 46 was prepared according to the method of 12 in 22% yield (12.5 mg). 1 H 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, measured 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) bicyclo propane carboxamide
Figure BDA0002247439760001543
Preparation 47 was prepared according to the procedure 12 in 11% yield (5.3mg)。 1 H 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, measured 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
Figure BDA0002247439760001551
Preparation 48 according to the method of 12 in 34% yield (17 mg). 1 H 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, measured 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)
Figure BDA0002247439760001552
Preparation 49 according to the method of 12 gave a yield of 41% (21 mg). 1 H 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, measured 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) dicyclohexyl-propanecarboxamide 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) dicyclohexyl-propanecarboxamide
Figure BDA0002247439760001561
Step 1.
To a solution of 27 in anhydrous DMF (0.5 mL) was added NaHS (6.7 mg,0.12 mmol) at 0deg.C and the temperature was allowed to warm 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 x 10 ml), the organic layer was separated, washed with brine, and dried over Na 2 SO 4 Dried, filtered, concentrated and purified 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, measured 493.23
And 2, step 2.
To 50-1 (20.0 mg,0.04 mmol) and K 2 CO 3 To a solution of (16.8 mg,0.12 mmol) in anhydrous DMF (1 mL) was added iodoethane (0.01 mL,0.12 mmol) and stirred under nitrogen at room temperature for 2 hours. The reaction mixture was then poured into water and extracted with dichloromethane (3 x 5 ml), the organic layer was separated, washed with brine, and dried over Na 2 SO 4 Drying, filtration and concentration gave crude 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 50-2 of the product in DCM (1 mL) at 0deg.C was added m-CPBA (13.8 mg,0.06 mmol). The mixture was then stirred at room temperature for 2 hours. The solvent was then removed and extracted with dichloromethane (3 x 5 ml) and the organic layer was separated with Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give 50 (4.6 mg) as a white solid (21% in two steps). 1 H 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, measured 553.24
Compound 51 was obtained as a white solid (13% in two steps). 1 H 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, measured 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) dicyclohexyl propane carboxamide
Figure BDA0002247439760001571
According to a similar procedure outlined for 50, 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) dicyclohexyl-propanecarboxamide. 1 H 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, measured 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) dicyclohexyl propane carboxamide
Figure BDA0002247439760001572
According to a similar procedure outlined for 50, prepare 53 from 50-1 (white solid, 4.7mg, two-step yield 21%). 1 H 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, measured 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
Figure BDA0002247439760001581
Preparation 54 was prepared according to the method of 12 in 10% yield (3.0 mg). 1 H 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, measured 514.24.
Compound 55: n-benzyl-1, 3, 7-trimethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purine-8-sulphonamide
Figure BDA0002247439760001582
8-mercapto-1, 3, 7-trimethyl-1H-purine-2, 6 (3H, 7H) -dione (50 mg,0.22 mmol) was stirred in a mixture of 2mL DCM and 2mL 1M HCl in a flask at-10 to-5℃for 10min. Cold (5 ℃) sodium hypochlorite (6% solution, 0.68M,1mL,0.72 mmol) 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 15min. The mixture was transferred to a separatory funnel (pre-cooled with ice water) and the CH was separated rapidly 2 Cl 2 Layers were collected in a clean flask cooled with a dry ice-acetone bath. Benzylamine (1.4 mL,12.5 mmol) was added with stirring followed by CH 2 Cl 2 The layer turned into a white suspension. The flask was moved to an ice-water bath and the suspension was stirred at 0 ℃ for 30min. The suspension was then washed with 1M phosphoric acid (all solids dissolved at once), then with water and brine. Drying (Na) 2 SO 4 ) And further purified by silica gel column chromatography (DCM/meoh=200/1) to give 7.9mg 55 as a yellow solid (10%). 1 H NMR(400Hz,DMSO-d 6 )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, measured 364.19
Compound 56: n,1,3, 7-tetramethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purine-8-sulphonamide
Figure BDA0002247439760001591
Preparation 56 according to method 55 is a yellow solid. 1 H 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, measured 288.12
Compound 57:
n-ethyl-1, 3, 7-trimethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purine-8-sulphonamide
Figure BDA0002247439760001592
Preparation 57 was prepared as a yellow solid according to procedure 55. 1 H 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, measured 302.14
Compound 58:1,3, 7-trimethyl-2, 6-dioxo-N-propyl-2, 3,6, 7-tetrahydro-1H-purine-8-sulfonamide
Figure BDA0002247439760001593
Preparation 58 was prepared as a yellow solid according to method 55. 1 H 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, measured 316.17
Compound 59: n, N,1,3, 7-pentamethyl-2, 6-dioxo-2, 3,6, 7-tetrahydro-1H-purine-8-sulfonamide
Figure BDA0002247439760001594
Preparation 59 was prepared as a yellow solid according to the method of 55. 1 H 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, measured 302.14
Compound 60:1,3, 7-trimethyl-8- (pyrrolidin-1-ylsulfonyl) -3, 7-dihydro-1H-purine-2, 6-dione
Figure BDA0002247439760001601
Preparation 60 was prepared as a yellow solid according to procedure 55. 1 H 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, measured 328.18
Compound 61:1,3, 7-trimethyl-2, 6-dioxo-N- (2, 2-trifluoroethyl) -2,3,6, 7-tetrahydro-1H-purine-8-sulphonamide
Figure BDA0002247439760001602
Preparation 61 was a yellow solid according to procedure 55. 1 H 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, measured 356.12
Compound 62:
1, 3-dimethyl-6, 7-dihydrothiazolo [2,3-f ] purine-2, 4 (1H, 3H) -dione 8, 8-dioxide
Figure BDA0002247439760001603
Step 1.
To a solution of 8-chloro-1, 3-dimethyl-3, 7-dihydro-1H-purine-2, 6-dione (4576 mg,2.0 mmol) in DMF (6 mL) was added NaSH (170 mg,3.0 mmol). The mixture was then stirred by microwave radiation at 110 ℃ for 2 hours. UPLC-MS monitors the completion of the reaction. The solvent was evaporated under reduced pressure and further purified by silica gel column chromatography (DCM/meoh=100/1) to give 404mg 62-1 as a yellow solid (88%). LC-MS (ESI) M/z: [ M+H ]] + Calculated 213.04, measured 213.12
And 2, step 2.
To a solution of 62-1 (80 mg,0.38 mmol) in DMF (2 mL) was added 1, 2-dibromoethane (204. Mu.L, 2.4 mmol) and K 2 CO 3 (130 mg,0.95 mmol). The mixture is subjected to microwave irradiationHeated to 110℃for 1.5h. The solvent was evaporated under reduced pressure and further purified by silica gel column chromatography (DCM/meoh=200/1) to give 53mg 62-2 as a yellow solid (59%), LC-MS (ESI) M/z: [ m+h] + Calculated 239.05, measured 239.14
And 3, step 3.
According to the method of 1, 62 was prepared from 62-2 (50 mg,0.21 mmol) and potassium hydrogen persulfate (193 mg,0.32 mmol) in 18% yield (10.0 mg) as a white solid. 1 H NMR:(400Hz,DMSO-d 6 )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, measured 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
Figure BDA0002247439760001611
Step 1.
63-1 was prepared from 8-mercapto-1, 3-dimethyl-1H-purine-2, 6 (3H, 7H) -dione (60 mg,0.28 mmol) and 1, 3-dibromopropane (142. Mu.L, 1.4 mmol) in 90% yield (10.0 mg) according to the method of 62. LC-MS (ESI) M/z: [ M+H ]] + Calculated 253.07, measured 253.13
And 2, step 2.
Preparation 63 was prepared from 63-1 (65 mg,0.26 mmol) and potassium hydrogen persulfate (470 mg,0.77 mmol) according to the method of 1 in 8% yield (6.0 mg) as a yellow solid. 1 H 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, measured 285.13
Compound 64: 3-methyl-1- (prop-2-yn-1-yl) -6, 7-dihydrothiazolo [2,3-f ] purin-2, 4 (1H, 3H) -dione 8, 8-dioxide
Figure BDA0002247439760001621
Step 1.
To cyanocarbaiminodithiodimethyl ester2.0 g) of EtOH (60 mL) was added 2-aminoethane-1-thiol (1.06 g). The mixture was then stirred at 90℃for 2 hours. UPLC-MS monitors the completion of the reaction. The solvent was evaporated under reduced pressure and further purified by silica gel column chromatography (DCM/meoh=100/1) to give 1.7g 64-1 as a yellow solid (100%). LC-MS (ESI) M/z: [ M+H ]] + Calculated 128.02, measured 128.11
And 2, step 2.
To a solution of 64-1 (500 mg) in DMF (8 mL) was added ethyl 2-chloroacetate (517 mg) 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=200/1) to give 390 mg 64-2 as a yellow solid (50%), LC-MS (ESI) M/z: [ m+h ] + Calculated 214.06, measured 214.12
And 3, step 3.
To a solution of 64-2 (200 mg) in EtOH (6 mL) was added EtONa. The mixture was then stirred at 90℃for 2 hours. UPLC-MS monitors the completion of the reaction. The solvent was evaporated under reduced pressure and further purified by silica gel column chromatography (DCM/meoh=100/1) to give 108mg 64-3 as a yellow solid (53%). LC-MS (ESI) M/z: [ M+H ]] + Calculated 214.06, measured 214.12
And 4, step 4.
To a solution of 64-3 (1.49 g) in THF (60 mL) were added methyl-urethane (1.73 g) and t-BuOK (2.35 g). The mixture was then stirred at 50℃for 5 hours. UPLC-MS monitors the completion of the reaction. 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, measured 225.13
And 5, step 5.
To a solution of 64-4 (500 mg) in DMF (5 mL) was added propargyl bromide (336 mg) and K 2 CO 3 (615 mg). The mixture was stirred at room temperature for 3h. 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, measured 263.12
And 6, step 6.
According to the method of 1, a mixture of 64-5 (10 mg,0.038 mmol) and potassium hydrogen persulfate (95 mg,0.15 mmol) of preparation 64 was 30% yield (3.3 mg) as yellow solid. 1 H 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, measured 295.11
Compound 65:1- (3- (3-hydroxyphenyl) prop-2-yn-1-yl) -3-methyl-6, 7-dihydrothiazolo [2,3-f ] purine-2, 4 (1 h,3 h) -dione 8-oxide and compound 66:1- (3- (3-hydroxyphenyl) prop-2-yn-1-yl) -3-methyl-6, 7-dihydrothiazolo [2,3-f ] purin-2, 4 (1H, 3H) -dione 8, 8-dioxide
Figure BDA0002247439760001631
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 (75 mg,0.29 mmol) in DMF (5 mL) was added 3-iodophenol (143 mg,0.43 mmol), pd (PPh) 3 ) 2 (20mg,0.028mmol)、CuI(5.3mg,0.028mmol)、Et 3 N (60. Mu.L, 0.43 mmol). The mixture was then degassed for 15min and then stirred at 55 ℃ for 2.5 hours. UPLC-MS monitors the completion of the reaction. The solvent was evaporated under reduced pressure and further purified by silica gel column chromatography (DCM/meoh=100/1) to give 42.6mg 65-1 as a yellow solid (42%). LC-MS (ESI) M/z: [ M+H ]] + Calculated 355.08, measured 355.16
And 2, step 2.
65 was prepared according to the method of 1 from 65-1 (8 mg,0.023 mmol) and potassium hydrogen persulfate (41 mg,0.068 mmol) in 43% yield (3.6 mg) as a yellow solid. 1 H 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, measured 371.13
Preparation 66 according to the method of 1 from 65-1 (8 mg,0.023 mmol) and potassium hydrogen persulfate (41 mg,0.068 mmol) was found to be a yellow solid in 36% yield (3.0 mg). 1 H 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, measured 387.14
Compound 67: 3-methyl-1- (3- (3- ((2-morpholinoethyl) amino) phenyl) prop-2-yn-1-yl) -6, 7-dihydrothiazolo [2,3-f ] purin-2, 4 (1H, 3H) -dione 8, 8-dioxide
Figure BDA0002247439760001641
67 was prepared according to the method of 12 from 3-iodo-N- (2-morpholinoethyl) aniline (41 mg,0.12 mmol) in 24% yield (11 mg) as yellow solid. 1 H 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, measured 499.23
Compound 68:
n, N' - (5- (3- (1, 7-dimethyl-8- (methylsulfanyl) -2, 6-dioxo-1H-purin-3 (2H, 6H, 7H) -yl) prop-1-yn-1-yl) -1, 3-phenylene) bicyclo propane carboxamide
Figure BDA0002247439760001642
To a solution of 27 (7.0 mg,0.013 mmol) in DMF (2 mL) was added MeSNa (1.4 mg,0.02 mmol). The mixture was then stirred at room temperature for 4 hours. The solvent was then removed and extracted with dichloromethane (3×5 mL), the organic layer was separated, and the solvent was removed using Na 2 SO 4 Dried, filtered, concentrated and purified by preparative TLC to give 68.3 mg (50%) as a white solid. LC-MS (ESI) M/z: [ M+H ]] + Calculated 507.17, measured 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
Figure BDA0002247439760001651
To a solution of the compound 1, 7-dimethyl-8- (methylsulfonyl) -3- (prop-2-yn-1-yl) -1H-purine-2, 6 (3H, 7H) -dione (60 mg,0.2 mmol) in dioxane (3 mL) was added P 2 S 5 (108 mg,0.24 mmol). The mixture was then stirred at 80℃for 4 hours. The solvent was then removed and extracted with dichloromethane (3×5 mL), the organic layer was separated, and the solvent was removed using Na 2 SO 4 Drying, filtration, concentration and purification by preparative TLC gave 69 mg (30%) of compound as a white solid. LC-MS (ESI) M/z: [ M+H ]] + Calculated 313.04, measured 313.13.
Compounds 70-72 were prepared according to the methods outlined in the schemes
Figure BDA0002247439760001652
Preparation 73-74 according to the procedure outlined in the schemes
Figure BDA0002247439760001653
Preparation of 75-77 according to the procedure outlined in the schemes
Figure BDA0002247439760001661
Preparation 78 according to the procedure outlined in the scheme
Figure BDA0002247439760001662
Preparation 79-80 according to the procedure outlined in the schemes
Figure BDA0002247439760001663
Preparation 81-82 according to the procedure outlined in the schemes
Figure BDA0002247439760001664
Preparation 83-85 according to the procedure outlined in the schemes
Figure BDA0002247439760001671
Preparation 86-87 according to the procedure outlined in the schemes
Figure BDA0002247439760001672
Preparation 88-90 according to the procedure outlined in the schemes
Figure BDA0002247439760001673
Preparation of 91-100 according to the method of Compounds 70-90
Preparation 101-110 according to the procedure outlined in the schemes
Figure BDA0002247439760001674
Preparation 111 according to the procedure outlined in the schemes
Figure BDA0002247439760001681
Preparation 112-113 according to the procedure outlined in the schemes
Figure BDA0002247439760001682
Preparation 114-117 according to the procedure outlined in the schemes
Figure BDA0002247439760001683
118-120 are prepared according to the method of compounds 110-117.
Series 2 compound necrotic apoptotic activity;
Figure BDA0002247439760001684
/>
Figure BDA0002247439760001691
experiment: discovery of novel high-efficiency necrotic apoptosis inhibitors targeting mixed lineage kinase domain-like proteins
In previous work, we performed a cell-based high throughput screen using a chemical library of about 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 mimetic/Z-VAD-fmk (TSZ) -induced necrotic apoptosis [4] . Among these hit compounds, one was developed as the first MLKL inhibitor NSA, which reveals the biological function of MLKL in necrotic apoptosis [4] . Another hit compound was identified as a RIP1 inhibitor and further developed as a highly potent and selective anti-inflammatory agent RIPA-56 [10] . We studied another hit compound from this high throughput screen, compound 1 (Compound 4, supra), which has good anti-necrotic apoptotic efficacy (EC 50 =390±8nM)。
To determine whether compound 1 interacted with an unknown target or with one of three proteins known to directly participate in TSZ-induced necrotic apoptosis (RIP 1, RIP3, and MLKL), 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 alters RIP1 and RIP3 kinase activity in an enzymatic assay, as their kinase activity is essential for the progression of necroptosis. We found that compound 1 does not inhibit kinase activity of RIP1 or RIP3 at concentrations up to 10 μm, which precludes the possibility of RIPK1 or RIPK3 being the target. Based on western blot analysis with anti-phosphorylated MLKL (pMLKL) antibodies, compound 1 did not block the phosphorylation of MLKL. However, compound 1 did block MLKL binding to NSA according to a binding competition assay that tested compound 1's ability to compete with NSA for MLKL binding. This suggests that MLKL may be a target for compound 1.
To obtain direct evidence for compound 1 targeting MLKL, we used chemical genetic methods. Specifically, the target of compound 1 was found using an affinity probe. We synthesized a series of derivatives based on compound 1 with the aim of increasing its anti-necrotic apoptotic efficacy and obtaining high affinity probes for target recognition. We first performed SAR studies on the 8/1/7 position of compound 1 by substituting the original methyl group with various functional groups. The necrotic apoptosis assay results indicated that all tested substitutions at positions 8/1/7 had a detrimental effect on potency, indicating that methyl groups were the best of the tested structures at these positions. Then, we turn our effort to the 3-position of compound 1. Related derivative compounds 7-12 and 14-18 were synthesized by the routes shown in Table A.
Figure BDA0002247439760001711
Synthesis of compounds 7-12 and 14-18 of (a). (a) potassium tert-butoxide, anhydrous THF,75 ℃, overnight; (b) NCS, THF, rt, overnight; (c) NaSMe, DMF,100 ℃,3h; (d) potassium hydrogen persulfate, meOH: H2O 1:1, rt,4H; (e) K2CO3, DMF, rt,1h; (f) 3-bromoprop-1-yne, K2CO3, DMF, rt,1h; (g) Pd (PPh 3) 4, cuI, TEA, anhydrous DMF,60 ℃,6h; potassium hydrogen persulfate, meOH, H2O 1:1, rt,4H. (B) Structure of Compound 19.
The imidazole derivative 2 was cyclized with methyl ethyl carbamate (3) to give 1, 7-dimethylxanthine (4). Subsequent substitution with chlorine and sulfhydryl groups yields the purine derivative 6, which purine derivative 6 is oxidized by potassium hydrogen persulfate to yield 7. Substitution of 7 with a different bromo derivative yields the target compounds 8-12. Compounds 14-18 were synthesized using intermediate 6 as starting material. Intermediate 6 is substituted with 3-bromoprop-1-yne to afford intermediate 13; followed by a Suzuki coupling with a different iodo benzene derivative and oxidation with potassium hydrogen persulfate to give the target compounds 14-18.
The anti-necrotic efficacy of compounds 7-12 and 14-18 was evaluated in the HT-29 cell line; EC (EC) 50 The values are shown in table B.
TABLE B efficacy of Compounds 7-12 and 14-18 in analysis of TSZ-induced necrotic apoptosis in HT-29 cells (EC 50 )。
Figure BDA0002247439760001712
Figure BDA0002247439760001721
[a]Inducing necrotic apoptosis in HT-29 cells for 24 hours; EC given herein 50 The values are the average of at least three independent analyses.
Compound 7 was more than 10-fold less potent than compound 1 due to the lack of methyl groups. The potency of ethyl analog 8 was reduced 5-fold compared to compound 1. Further extension of the alkyl chain length restored some efficacy (see compounds 9 and 10). Interestingly, compound 12 with propynyl group was 3 times more potent than compound 1 (methyl group); however, benzyl analog 11 was reduced 4-fold in potency relative to compound 12, indicating the presence of a relatively narrow hydrophobic space in the target. To detect putative binding pockets beyond the region defined by propynyl groups, we synthesised 14 by adding a phenyl group at the end of the propynyl group of 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 interactions between the target and the inhibitor. To investigate the possibility of creating 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) have inhibition potency 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 suggests that hydrogen bonding may be formed 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 studies successfully improved the efficacy of the initial hit compound, compound 1, from EC 50 A value of 390nM until EC is reduced 50 The 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 can act as a covalently bound leaving group in its interaction with MLKL [11] . To verify this hypothesis, we synthesized compound 19, which is a close structural analog of compound 1, but in which the methylsulfonyl group has no dissociative ability. Compound 19 showed no efficacy in the necrotic apoptosis assay, indicating that covalent binding appears to be critical for the efficacy of these inhibitors. By comparing the efficacy of compounds 12 and 15 in washed/unwashed cell assays (wash/no-wash cell assays), we further confirmed the irreversible covalent binding behavior of compounds 12 and 15. For both compounds, EC of the wash samples were recorded 50 Value and EC of unwashed samples 50 The values are comparable, indicating that these compounds are covalent inhibitors.
Like compound 1, compounds 12 and 14-16 (10 μm) did not inhibit kinase activity of RIP1 or RIP3 in vitro assays. To determine if 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 the biotin tag. The biotin-labeled compound-protein complex was pulled down and analyzed by SDS-PAGE. We observed that compound 12 did bind covalently to MLKL in the cell lysate and that compound 15 was effectively over 12 in binding to MLKL.
The recently developed BTC-ABPP method in our laboratory greatly facilitates the identification of modification sites for target protein probes in living cells [12] . Here we used BTC-ABPP together with HT-29 cells to identify the binding sites of Compound 12 and MLKL. B/y ion spectra from MS/MS analysis indicated 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 site of NSA modification [4] . To further verify the relationship between probe binding at this site and the function of compound 12 in necrotic apoptosis, MLKL knockdown RIP3-HeLa cells were transfected with wild-type or C86S (cysteine 86 mutated to serine) mutant forms of MLKL. TSZ stimulation of both cell types resulted in necrotic apoptosis, suggesting that the C86S mutant form of MLKL plays a normal role in transducing necrotic apoptosis signals. Compound 12 protected cells transfected with wild-type MLKL, but not cells transfected with C86S-MLKL from necrotic apoptosis, further confirming that compound 12 inhibited MLKL by direct binding to Cys-86. Cys-86 in human MLKL corresponds structurally to tryptophan residues in mouse and rat MLKL. Spiral H4 comprising Cys-86 at the N-terminal domain of the human MLKL structure is absent in mouse MLKL [13] . In view of this, we speculate that compounds 12 and 15 may show poor inhibition of the function of mouse or rat MLKL in necrotic apoptosis. We performed necrotic apoptosis assays in the mouse MEF and L929 cell lines and the rat L6 cell line and found that neither compound 12 nor compound 15 showed inhibitory activity, even beyond their previously established ECs 50 The same is true for the concentration of values. These results confirm that Cys-86 of the target MLKL is necessary for the covalent binding and potency of the series of MLKL inhibitors.
Previous studies have shown that MLKL forms a homooligomer when it is phosphorylated by RIP 3. This is a key step in the transfer of MLKL from the cytoplasm to the cell membrane, in the process of which it mediates ion influx and/or causes destruction of cellular structures [5] . To evaluate the mechanism of action of this series of compounds on MLKL, we detected the status of MLKL (oligomeric and phosphorylated status) in the presence of compound 12 or 15 under TSZ stimulation. We first demonstrate that both compounds 12 and 15 can block the formation of MLKL homo-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 the 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 spots (pMLKL) appear in the plasma membrane. RIPA-56 and GW806742X completely blocked the phosphorylation of MLKL, and no pMLKL spot was observed in cells treated with these compounds. In contrast, small pMLKL spots were 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 are consistent with earlier studies [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.
Sun, H, et al Cell 2012,148,213-227.
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.Vakifahmetoglu-Norberg, cell reports 2015,10,1836-1849; cM.Pierdomenico, A.Negroni, L.Stronati, R.Vitali, E.Prete, J.Bertin, P.J.Gough, M.Aloi, S.Cucchiara, am J. Gastroentol.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.
D.m. sammond, et al, biorg.med.chem.lett.2005, 15,3519-3523.
Y.ren, et al, J.Med.chem. (http:// dx.doi.org/10.1021/acs.jmed chem.6b01128, in Press)
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.

Claims (7)

1. An MLKL inhibitor compound of formula I:
Figure FDA0004121449860000011
wherein:
R 1 in the presence of a hydrogen atom, which is H,
Figure FDA0004121449860000012
Figure FDA0004121449860000013
Figure FDA0004121449860000014
wherein->
Figure FDA0004121449860000015
R in (2) is F, cl, br,>
Figure FDA0004121449860000016
R 2 and R is 3 Is methyl;
R 4 is that
Figure FDA0004121449860000017
Figure FDA0004121449860000018
Wherein (1)>
Figure FDA0004121449860000019
R in the formula is F, cl, br, me, CF 3 ,/>
Figure FDA00041214498600000110
Figure FDA00041214498600000111
Figure FDA0004121449860000021
And
n is 1 or 2;
or a pharmaceutically acceptable salt of said compound.
2. The compound of claim 1, wherein:
R 1 is methyl or cyclopropylmethyl.
3. The compound of claim 1 or 2, wherein one or more hydrogens are in the form of deuterium.
4. The compound of claim 1 or 2, wherein the compound is a peptide of formula (I) 3 、CD 2 H or CDH 2 Replacement armorA base.
5. A MLKL inhibitor compound having a structure selected from the group consisting of series 1:
Figure FDA0004121449860000022
/>
Figure FDA0004121449860000031
/>
Figure FDA0004121449860000041
6. a compound having the structure of series 2 selected from the group consisting of:
Figure FDA0004121449860000042
/>
Figure FDA0004121449860000051
/>
Figure FDA0004121449860000061
/>
Figure FDA0004121449860000071
/>
Figure FDA0004121449860000081
7. a pharmaceutical composition comprising a compound of any one of claims 1-6 and a pharmaceutically acceptable excipient, which is a predetermined unit dosage form selected from a pill, tablet or capsule.
CN201880027489.8A 2017-02-28 2018-02-27 MLKL inhibitors Active CN110573509B (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
CNPCT/CN2017/075248 2017-02-28
CN2017075248 2017-02-28
PCT/CN2018/077464 WO2018157800A1 (en) 2017-02-28 2018-02-27 Mlkl inhibitors

Publications (2)

Publication Number Publication Date
CN110573509A CN110573509A (en) 2019-12-13
CN110573509B true CN110573509B (en) 2023-05-26

Family

ID=63369796

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201880027489.8A Active CN110573509B (en) 2017-02-28 2018-02-27 MLKL inhibitors

Country Status (2)

Country Link
CN (1) CN110573509B (en)
WO (1) WO2018157800A1 (en)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111053907A (en) * 2019-12-31 2020-04-24 厦门大学 Application of Setdb1 gene as target in treating inflammatory bowel disease
CN113150009B (en) * 2020-01-07 2023-05-23 爱科诺生物医药(香港)有限公司 Heterocyclic compound with activity of inhibiting programmed cell necrosis pathway and application thereof
CN113135940B (en) * 2020-01-20 2022-05-03 成都大学 Benzothiazole pyrimidine ring compound and preparation method and application thereof
WO2024050825A1 (en) * 2022-09-09 2024-03-14 National Institute Of Biological Sciences, Beijing Compounds as mlkl inhibitors

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IT1248528B (en) * 1991-06-21 1995-01-19 Pierrel Spa AROMATIC ETHER AND THIOETHER (HETER) DERIVATIVES HAVING ANTI-HYPERLIPIDEMIC ACTIVITY, PROCEDURE FOR THEIR PREPARATION AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM.
US7732447B2 (en) * 2006-06-22 2010-06-08 Cephalon, Inc. Fused [d]pyridazin-7-ones
CN102516384A (en) * 2011-11-03 2012-06-27 北京生命科学研究所 MLKL protein and application of MLKL protein as target point of cell necrosis inhibitor
TW201444844A (en) * 2013-03-14 2014-12-01 Hydra Biosciences Inc Substituted xanthines and methods of use thereof
HUE033528T4 (en) * 2013-03-15 2018-05-02 Hydra Biosciences Inc Substituted xanthines and methods of use thereof
SG11201609350XA (en) * 2014-05-15 2016-12-29 Catalyst Therapeutics Pty Ltd Methods for inhibiting necroptosis

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Discovery of a new class of highly potent necroptosis inhibitors targeting the mixed lineage kinase domain-like protein;Bo Yan等;《Chemical Communications (Cambridge, United Kingdom)》;20170307;第53卷(第26期);第3638页图3,表1,Supporting Information表S1,第3638页右栏最后一行、第3639页左栏第一段 *

Also Published As

Publication number Publication date
CN110573509A (en) 2019-12-13
WO2018157800A1 (en) 2018-09-07

Similar Documents

Publication Publication Date Title
AU2020203967B2 (en) Amide-substituted heterocyclic compounds useful as modulators of il-12, il-23 and/or ifn alpha responses
CN110573509B (en) MLKL inhibitors
EP3066078B1 (en) Alkyl-amide-substituted pyridyl compounds useful as modulators of il-12, il-23 and/or ifnalpha responses
EP3057586B1 (en) Bromodomain inhibitors
AU2009232017B2 (en) 1-heterocyclyl-1,5-dihydro-pyrazolo[3,4-d] pyrimidin-4-one derivatives and their use as PDE9A modulators
NL2000480C2 (en) NEW PHARMACEUTICAL FUNDS.
JP6867417B2 (en) N- (substituted phenyl) -sulfonamide derivative as a kinase inhibitor
JP2022506887A (en) Nitrogen-containing condensed heterocyclic SHP2 inhibitor compound, production method and use
EP3585780A1 (en) Aryl hydrocarbon receptor (ahr) modulator compounds
WO2006080450A1 (en) Igf-1r inhibitor
WO2014074660A1 (en) ALKYL-AMIDE-SUBSTITUTED PYRIDYL COMPOUNDS USEFUL AS MODULATORS OF IL-12, IL-23 AND/OR IFNα RESPONSES
JPH11209350A (en) Nitrogen-containing heterocyclic derivative and medicine containing the same
JPWO2005095419A1 (en) Thiazolopyrimidine derivatives
WO2003048132A1 (en) Imidazopyridines, pyrimidines and triazines for enhancing cognition as gaba-a alpha 5 receptor subtype ligands
AU2006328934A1 (en) Pyrrolo [1,2-a]quinoxaline derivatives as adenosine A3 receptor modulators and uses thereof
AU2018289864B2 (en) Coumarin-like cyclic compound as MEK inhibitor and use thereof
KR20200089219A (en) Aryl or heteroaryl derivatives, and pharmaceutical composition thereof for use in preventing or treating kinase-related disease
EP1867648A1 (en) Substituted aminopyrazolopyridines and salts thereof, their preparations and pharmaceutical compositions comprising them.
CN117083268A (en) Amino heteroaryl compounds and compositions
JP6511692B2 (en) Hydroxypurine compound and its application
US11666577B2 (en) MLKL inhibitors
JP2012211085A (en) Hedgehog signal inhibitor
JP2005526817A (en) Novel 4- (pyrrolopyrimidin-6-yl) benzenesulfonamide derivative
CA3158731A1 (en) Adenosine receptor antagonist compounds
EA007226B1 (en) Imidazoquinoline derivatives

Legal Events

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