WO2009006089A2 - A2 adenosine receptor agonists - Google Patents

A2 adenosine receptor agonists Download PDF

Info

Publication number
WO2009006089A2
WO2009006089A2 PCT/US2008/067978 US2008067978W WO2009006089A2 WO 2009006089 A2 WO2009006089 A2 WO 2009006089A2 US 2008067978 W US2008067978 W US 2008067978W WO 2009006089 A2 WO2009006089 A2 WO 2009006089A2
Authority
WO
WIPO (PCT)
Prior art keywords
compound
pharmaceutically acceptable
acceptable salt
group
nmr
Prior art date
Application number
PCT/US2008/067978
Other languages
French (fr)
Other versions
WO2009006089A3 (en
Inventor
Kenneth A. Jacobson
Zhan-Guo Gao
Hayamitsu Adachi
James M. Downey
Yanping Liu
Original Assignee
Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services
South Alabama Medical Science Foundation
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 Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services, South Alabama Medical Science Foundation filed Critical Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services
Publication of WO2009006089A2 publication Critical patent/WO2009006089A2/en
Publication of WO2009006089A3 publication Critical patent/WO2009006089A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/16Purine radicals
    • C07H19/167Purine radicals with ribosyl as the saccharide radical
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis

Definitions

  • a 2A , A 2B , and A 3 adenosine receptors
  • Activation or blocking one or more of these ARs leads to useful therapeutic applications.
  • activation of the A 2 ⁇ AR can induce angiogenesis, reduce vascular permeabilization, increase production of the anti- inflammatory cytokine IL-10, increase chloride secretion in epithelial cells, or increase release of inflammatory mediators from human and canine mast cells.
  • a 2 ⁇ AR agonists for use in therapeutic applications.
  • the invention provides A 2B adenosine receptor (AR) agonists of formula I.
  • exemplary compounds of formula I have enhanced potency at the A 2 ⁇ AR and reduced potency at other AR subtypes, wherein R 1 -R 4 , Z, and n are described below in detail.
  • compositions comprising at least one compound of formula I and methods of use thereof, for example, in a method of treating a disease based on the modulation of the adenosine system, e.g., septic shock, cystic fibrosis, restenosis, erectile dysfunction, inflammation, myocardial ischemia, and reperfusion injury.
  • adenosine system e.g., septic shock, cystic fibrosis, restenosis, erectile dysfunction, inflammation, myocardial ischemia, and reperfusion injury.
  • Figure 1 depicts chemical structures of exemplary compounds of formula I (8 and
  • Figure 2 depicts a reaction scheme to prepare intermediates for the 2-ether component of compounds of formula I in accordance with an embodiment of the invention.
  • Figure 3 depicts another reaction scheme to prepare intermediates for the 2-ether component of compounds of formula I in accordance with an embodiment of the invention.
  • Step (a) (i): TsCl, pyridine, CH 2 Cl 2 ; (ii): 3-butyn-l-ol, Pd(PPh 3 ) 2 Cl 2 , CuI, Et 3 N, DMF, 70 0 C; step (b): I 2 , PPh 3 , imidazole, Et 2 O-MeCN, room temperature; and step (c): LAH, THF, 0 0 C - room temperature.
  • Figure 4 depicts a reaction scheme to prepare compounds 8, 17-36, and 119-127 in accordance with an embodiment of the invention.
  • Figure 5 depicts a reaction scheme to prepare compound 39 in accordance with an embodiment of the invention.
  • step (b) (i): EtNH 2 HCl, DIPEA, DMF, 140 0 C;
  • Figure 6 depicts a reaction scheme to prepare compound 40 in accordance with an embodiment of the invention.
  • FIG. 7A depicts activation curves (log [Agonist] versus percent agonist efficacy) for 28 ( ⁇ ) in comparison to 2 ( ⁇ ) at the Ai adenosine receptors.
  • Figure 7B depicts activation curves (log [Agonist] versus percent agonist efficacy) for 28 ( ⁇ ) in comparison to 2 ( ⁇ ) at the A 2A adenosine receptors.
  • Figure 7C depicts activation curves (log [Agonist] versus percent agonist efficacy) for 28 ( A ) in comparison to 2 ( ⁇ ) at the A 2B adenosine receptors.
  • Figure 7D depicts activation curves (log [Agonist] versus percent agonist efficacy) for 28 ( ⁇ ) in comparison to 2 ( ⁇ ) at the A 3 adenosine receptors.
  • Figure 8 depicts the percent of risk zone infracted in an isolated rabbit heart for a control group and a group that had been administered 200 nM of 28 in accordance with an embodiment of the invention.
  • Figure 9 depicts the infarct size (cm ) as a function of risk size (cm ) in accordance with an embodiment of the invention.
  • the symbol • represents the control group, whereas O represents 28.
  • the present invention provides a compound of formula I or pharmaceutically acceptable salt thereof:
  • R 1 is selected from the group consisting of aryl, cycloalkylaryl, heterocycloalkylaryl, heteroaryl, heterocycloalkyl, and arylheterocycloalkyl, each of which is optionally substituted with 1 to 3 substituents selected from the group consisting of C M2 alkyl, hydroxyl, Ci -I2 alkoxy, halo, tosyl (Ts), CN, -C(O)OH, aminocarbonyl, amino, C 1 - J2 dialkylamino, and Ci -J2 alkylamino;
  • Z is a bond or -CH 2 -;
  • n is an integer of 1 to 4.
  • R 2 is selected from the group consisting Of-CH 2 OH, aminocarbonyl, Cj -J2 alkylaminocarbonyl, and di-Cj.j ⁇ alkylaminocarbonyl;
  • R 3 and R 4 are the same or different and each is selected from the group consisting of hydrogen, C J -CJ 2 alkyl, C 3 -C 8 cycloalkyl, C 6 -C 30 aryl, and imidamido;
  • R 2 is -CH 2 OH, and R 3 and R 4 are hydrogen, then -0-(CH 2 ) H -Z-R 1 is not benzyloxy, phenylalkoxy or a substituted phenylalkoxy, thiophenylalkoxy or a substituted thiophenylalkoxy, pyridylalkoxy, indolylalkoxy, naphthylalkoxy, biphenylalkoxy, or indolylalkoxy.
  • R 1 is selected from the group consisting Of C 6-3O aryl, heteroaryl, heterocycloalkyl, C 5-7 cycloalkyl-C 6-3 o aryl, heterocycloalkyl-Ce ⁇ o aryl, C 6-30 aryl- heteroaryl, and C 6-30 aryl-heterocycloalkyl.
  • R 1 is selected from the group consisting of
  • C 6-3O aryl e.g., phenyl, biphenyl, naphthyl, anthracenyl, pyrenyl
  • a heteroaryl consisting of one or two oxygen or sulfur atoms and/or from one to four nitrogen atoms provided that the total number of heteroatoms in each ring is four or less and each ring has at least one carbon atom (e.g., pyridinyl, pyridazinyl, pyrimidyl, pyrazinyl, triazinyl, pyrrolyl, pyrazolyl, imidazolyl, (1,2,3,)- and (l,2,4)-triazolyl, pyrazinyl, pyrimidinyl, tetrazolyl, furyl, thiophenyl, isothiazolyl, thiazolyl, isoxazolyl, oxadiazolyl, oxazolyl, pyrrolo[2,3-c]
  • R 5 and R 5 are the same or different and each is hydrogen or 1 to 3 substituents individually selected from the group consisting of Ci -I2 alkyl, hydroxyl, Ci -I2 alkoxy, halo, tosyl (Ts), CN, -C(O)OH, aminocarbonyl, amino, Ci -I2 dialkylamino, and Ci -I2 alkylamino; and
  • R 6 is selected from the group consisting of hydrogen, Ci -I2 alkyl, C 3-8 cycloalkyl, and C 6-30 aryl.
  • R 1 is selected from the group consisting of
  • R 1 is selected from the group consisting of
  • R 5 and R 5 are the same or different and each can be hydrogen, Ci -I2 alkyl, hydroxyl, C 1-I2 alkoxy, halo, or tosyl.
  • R 1 is selected from the group consisting of
  • n is 1, 2, or 3, particularly n is 2 or 3. In especially preferred compounds, n is 2.
  • R 2 is -CH 2 OH or C M2 alkylaminocarbonyl (e.g., N-methylcarboxaminocarbonyl, N-ethylcarboxaminocarbonyl, N- propylcarboxaminocarbonyl) .
  • C M2 alkylaminocarbonyl e.g., N-methylcarboxaminocarbonyl, N-ethylcarboxaminocarbonyl, N- propylcarboxaminocarbonyl
  • R 3 and R 4 are the same or different and each is selected from the group consisting of hydrogen, Ci-Ci 2 alkyl, and imidamido.
  • alkyl implies a straight or branched alkyl moiety containing from, for example, 1 to 12 carbon atoms, preferably from 1 to 8 carbon atoms, more preferably from 1 to 6 carbon atoms.
  • moieties include methyl, ethyl, propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, pentyl, isoamyl, hexyl, octyl, dodecanyl, and the like.
  • cycloalkyl means a cyclic moiety containing from, for example, 1-3 rings (i.e., monocyclic, bicyclic, tricyclic, or spiro), 3 to 8 carbon atoms per ring, preferably from 5 to 8 carbon atoms, more preferably from 5 to 6 carbon atoms.
  • moieties include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like.
  • aryl refers to an unsubstituted or substituted aromatic carbocyclic moiety, as commonly understood in the art, and includes monocyclic and polycyclic aromatics such as, for example, phenyl, biphenyl, naphthyl, anthracenyl, pyrenyl, and the like.
  • heterocycloalkyl means a cycloalkyl moiety having one or more heteroatoms selected from nitrogen, sulfur, and/or oxygen.
  • a heterocycloalkyl is a 5 or 6-membered monocyclic ring and contains one, two, or three heteroatoms selected from nitrogen, oxygen, and/or sulfur.
  • the heterocycloalkyl can be attached to the parent structure through a carbon atom or through any heteroatom of the heterocycloalkyl that results in a stable structure. Examples of such heterocyclic rings are pyrrolinyl, pyranyl, piperidyl, tetrahydrofuranyl, tetrahydrothiopheneyl, and morpholinyl.
  • arylheterocycloalkyl refers to a heterocycloalkyl, as defined herein, that is substituted with an aryl group, as defined herein, as a fused ring, e.g., benzo. Examples include 2,3-dihydrobenzo[b][l,4]dioxinyl, 2-indolinyl, and 3-indolinyl.
  • cycloalkylaryl and “heterocycloalkylaryl” refer to an aryl, as defined herein, that is substituted with a cycloalkyl group or a heterocycloalkyl group, respectively, as defined herein, or as a fused ring, e.g., benzo.
  • cycloalkylaryl groups include 5-, 6-, 7-, or 8-1,2,3,4-tetrahydronaphthalenyl and 4-, 5-, 6-, or 7-2,3-dihydro-lH-indenyl.
  • heterocycloalkylaryl examples include 5-, 6-, 7-, or 8-1,2,3,4-tetrahydroquinolinyl, 5-, 6-, 7-, or 8-1,2,3,4-tetrahydroquinoxalinyl, and 5-, 6-, 7-, or 8-2,3-dihydrobenzo[b][l,4]dioxinyl.
  • heteroaryl refers to aromatic 4, 5, or 6 membered monocyclic groups, 9 or 10 membered bicyclic groups, and 11 to 14 membered tricyclic aryl groups having one or more heteroatoms (O, S, or N).
  • Each ring of the heteroaryl group containing a heteroatom can contain one or two oxygen or sulfur atoms and/or from one to four nitrogen atoms provided that the total number of heteroatoms in each ring is four or less and each ring has at least one carbon atom.
  • the nitrogen and sulfur atoms may optionally be oxidized, and the nitrogen atoms may optionally be quaternized.
  • the heteroaryl group can be attached at any available nitrogen or carbon atom of any ring.
  • Illustrative examples of heteroaryl groups are pyridinyl, pyridazinyl, pyrimidyl, pyrazinyl, triazinyl, pyrrolyl, pyrazolyl, imidazolyl,
  • arylheteroaryl refers to heteroaryl, as defined herein, that is substituted with an aryl group, as defined herein, as a fused ring, e.g., benzo.
  • arylheteroaryl groups include benzimidazolyl, indolyl, indazolyl, benzo-l,2,3-triazolyl, 2-benzo[d] oxazolyl,
  • alkoxy embraces linear or branched alkyl groups that are attached to a an ether oxygen.
  • the alkyl group is the same as described herein. Examples of such substituents include methoxy, ethoxy, t-butoxy, and the like.
  • alkylamino refers to a group with one hydrogen and one alkyl group directly attached to a trivalent nitrogen atom.
  • dialkylamino refers to a group with two of the same or different alkyl groups directly attached to a trivalent nitrogen atom.
  • aminocarbonyl refers to the group -C(O)NH 2 .
  • alkylaminocarbonyl and “dialkylaminocarbonly” refer to the group -C(O)NRR', in which
  • R is hydrogen or an alkyl group and R' is the same or different alkyl group as described herein.
  • imidamido also known as guanidino, refers to the group
  • halo as used herein, means a substituent selected from Group VIIA, such as, for example, fluorine, bromine, chlorine, and iodine.
  • fused includes a polycyclic compound in which one ring contains one or more atoms preferably one, two, or three atoms in common with one or more other rings.
  • Ci -8 , Ci -6 , or CM alkyl, alkylamino, etc. any sub-range or individual number of carbon atoms falling within the indicated range also can be used.
  • a range of 1-8 carbon atoms e.g., Cj-Cg
  • 1-6 carbon atoms e.g., C I -C O
  • 1-4 carbon atoms e.g., Ci-C 4
  • 1-3 carbon atoms e.g., C1-C3
  • 2-8 carbon atoms e.g., C 2 -C 8
  • any chemical group e.g., alkyl, alkylamino, etc.
  • any sub-range thereof e.g., 1-2 carbon atoms, 1-3 carbon atoms, 1-4 carbon
  • the compounds of formula I can be prepared by any suitable method.
  • 2', 3', 5'-triacetyl-6-chloroguanosine can be synthetically modified to prepare 2- ether-substituted 2', 3', 5'-triacetyl-6-chloroadenosine derivatives.
  • a suitable amine e.g., ammonia, ethylamine hydrochloride, guanidine hydrochloride
  • a suitable amine e.g., ammonia, ethylamine hydrochloride, guanidine hydrochloride
  • the adenosine derivative is treated with a suitable strong acid or base (e.g., KOH) to form the compounds of formula I.
  • a suitable strong acid or base e.g., KOH
  • Isolation and purification of the compounds and intermediates described herein can be effected, if desired, by any suitable separation or purification procedure such as, for example, filtration, extraction, crystallization, column chromatography, thin-layer chromatography, thick-layer chromatography, preparative low or high-pressure liquid chromatography, or a combination of these procedures. Specific illustrations of suitable separation and isolation procedures can be had by reference to the Examples described herein. However, other equivalent separation or isolation procedures can also be used.
  • the present invention further provides a pharmaceutical composition comprising at least one compound of formula I or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier.
  • the pharmaceutically acceptable carrier can be any of those conventionally used and is limited only by chemico-physical considerations, such as solubility and lack of reactivity with the compound, and by the route of administration. It will be appreciated by one of skill in the art that, in addition to the following described pharmaceutical compositions; the compounds of the present invention can be formulated as inclusion complexes, such as cyclodextrin inclusion complexes or liposomes.
  • the pharmaceutically acceptable carriers described herein, for example, vehicles, adjuvants, excipients, or diluents, are well known to those who are skilled in the art and are readily available to the public. It is preferred that the pharmaceutically acceptable carrier be one which is chemically inert to the active compounds and one which has no detrimental side effects or toxicity under the conditions of use.
  • compositions of the present invention are merely exemplary and are in no way limiting.
  • Formulations suitable for oral administration can consist of (a) liquid solutions, such as an effective amount of the compound dissolved in diluents, such as water, saline, or orange juice; (b) capsules, sachets, tablets, lozenges, and troches, each containing a predetermined amount of the active ingredient, as solids or granules; (c) powders; (d) suspensions in an appropriate liquid; and (e) suitable emulsions.
  • Liquid formulations may include diluents, such as water and alcohols, for example, ethanol, benzyl alcohol, and the polyethylene alcohols, either with or without the addition of a pharmaceutically acceptable surfactant, suspending agent, or emulsifying agent.
  • Capsule forms can be of the ordinary hard- or soft-shelled gelatin type containing, for example, surfactants, lubricants, and inert fillers, such as lactose, sucrose, calcium phosphate, and cornstarch.
  • Tablet forms can include one or more of lactose, sucrose, mannitol, corn starch, potato starch, alginic acid, microcrystalline cellulose, acacia, gelatin, guar gum, colloidal silicon dioxide, croscarmellose sodium, talc, magnesium stearate, calcium stearate, zinc stearate, stearic acid, and other excipients, colorants, diluents, buffering agents, disintegrating agents, moistening agents, preservatives, flavoring agents, and pharmacologically compatible carriers.
  • Lozenge forms can comprise the active ingredient in a flavor, usually sucrose and acacia or tragacanth, as well as pastilles comprising the active ingredient in an inert base, such as gelatin and glycerin, or sucrose and acacia, emulsions, gels, and the like containing, in addition to the active ingredient, such carriers as are known in the art.
  • a flavor usually sucrose and acacia or tragacanth
  • pastilles comprising the active ingredient in an inert base, such as gelatin and glycerin, or sucrose and acacia, emulsions, gels, and the like containing, in addition to the active ingredient, such carriers as are known in the art.
  • the compounds of the present invention can be made into aerosol formulations to be administered via inhalation.
  • aerosol formulations can be placed into pressurized acceptable propellants, such as dichlorodifluoromethane, propane, nitrogen, and the like. They also may be formulated as pharmaceuticals for non-pressured preparations, such as in a nebulizer or an atomizer.
  • Formulations suitable for parenteral administration include aqueous and nonaqueous, isotonic sterile injection solutions, which can contain anti-oxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the intended recipient, and aqueous and non-aqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizers, and preservatives.
  • the compound can be administered in a physiologically acceptable diluent in a pharmaceutical carrier, such as a sterile liquid or mixture of liquids, including water, saline, aqueous dextrose and related sugar solutions, an alcohol, such as ethanol, isopropanol, or hexadecyl alcohol, glycols, such as propylene glycol or polyethylene glycol, glycerol ketals, such as 2,2-dimethyl-l,3-dioxolane- 4-methanol, ethers, such as poly(ethyleneglycol) 400, an oil, a fatty acid, a fatty acid ester or glyceride, or an acetylated fatty acid glyceride with or without the addition of a pharmaceutically acceptable surfactant, such as a soap or a detergent, suspending agent, such as pectin, carbomers, methylcellulose, hydroxypropylmethylcellulose, or carboxymethylcellulose, or emulsifying agents and other pharmaceutical
  • Oils which can be used in parenteral formulations include petroleum, animal, vegetable, or synthetic oils. Specific examples of oils include peanut, soybean, sesame, cottonseed, corn, olive, petrolatum, and mineral. Suitable fatty acids for use in parenteral formulations include oleic acid, stearic acid, and isostearic acid. Ethyl oleate and isopropyl myristate are examples of suitable fatty acid esters.
  • Suitable soaps for use in parenteral formulations include fatty alkali metal, ammonium, and triethanolamine salts
  • suitable detergents include (a) cationic detergents such as, for example, dimethyl dialkyl ammonium halides, and alkyl pyridinium halides, (b) anionic detergents such as, for example, alkyl, aryl, and olefin sulfonates, alkyl, olefin, ether, and monoglyceride sulfates, and sulfosuccinates, (c) nonionic detergents such as, for example, fatty amine oxides, fatty acid alkanolamides, and polyoxyethylene-polypropylene copolymers, (d) amphoteric detergents such as, for example, alkyl-beta-aminopropionates, and 2-alkyl-imidazoline quaternary ammonium salts, and (3) mixtures thereof.
  • the parenteral formulations will typically contain from about 0.5 to about 25% by weight of the active ingredient in solution. Suitable preservatives and buffers can be used in such formulations. In order to minimize or eliminate irritation at the site of injection, such compositions may contain one or more nonionic surfactants having a hydrophile-Iipophile balance (HLB) of from about 12 to about 17. The quantity of surfactant in such formulations ranges from about 5 to about 15% by weight. Suitable surfactants include polyethylene sorbitan fatty acid esters, such as sorbitan monooleate and the high molecular weight adducts of ethylene oxide with a hydrophobic base, formed by the condensation of propylene oxide with propylene glycol.
  • HLB hydrophile-Iipophile balance
  • parenteral formulations can be presented in unit-dose or multi- dose sealed containers, such as ampoules and vials, and can be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example, water, for injections, immediately prior to use.
  • sterile liquid carrier for example, water
  • Extemporaneous injection solutions and suspensions can be prepared from sterile powders, granules, and tablets of the kind previously described.
  • the compounds of the present invention may be made into injectable formulations.
  • the requirements for effective pharmaceutical carriers for injectable compositions are well known to those of ordinary skill in the art. See Pharmaceutics and Pharmacy Practice, J. B. Lippincott Co., Philadelphia, Pa., Banker and Chalmers, eds., pages 238-250 (1982), and ASHP Handbook on Injectable Drugs, Toissel, 4th ed., pages 622-630 (1986).
  • the compounds of the present invention may be made into suppositories by mixing with a variety of bases, such as emulsifying bases or water-soluble bases.
  • bases such as emulsifying bases or water-soluble bases.
  • Formulations suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams, or spray formulas containing, in addition to the active ingredient, such carriers as are known in the art to be appropriate.
  • Suitable carriers and their formulations are further described in A.R. Gennaro, ed., Remington: The Science and Practice of Pharmacy (19th ed.), Mack Publishing Company, Easton, PA (1995).
  • the compound of the invention or a composition thereof can potentially be administered as a pharmaceutically acceptable acid-addition, base neutralized or addition salt, formed by reaction with inorganic acids, such as hydrochloric acid, hydrobromic acid, perchloric acid, nitric acid, thiocyanic acid, sulfuric acid, and phosphoric acid, and organic acids such as formic acid, acetic acid, propionic acid, glycolic acid, lactic acid, pyruvic acid, oxalic acid, malonic acid, succinic acid, maleic acid, and fumaric acid, or by reaction with an inorganic base, such as sodium hydroxide, ammonium hydroxide, potassium hydroxide, and organic bases, such as mono-, di-, trialkyl, and aryl amines and substituted ethanolamines.
  • inorganic acids such as hydrochloric acid, hydrobromic acid, perchloric acid, nitric acid, thiocyanic acid, sulfuric acid, and phosphoric acid
  • organic acids
  • the conversion to a salt is accomplished by treatment of the base compound with at least a stoichiometric amount of an appropriate acid.
  • the free base is dissolved in an inert organic solvent such as diethyl ether, ethyl acetate, chloroform, ethanol, methanol, and the like, and the acid is added in a similar solvent.
  • the mixture is maintained at a suitable temperature (e.g., between 0 0 C and 50 0 C).
  • the resulting salt precipitates spontaneously or can be brought out of solution with a less polar solvent.
  • the neutral forms of the compounds can be regenerated by contacting the salt with a base or acid and isolating the parent compound in the conventional manner.
  • the parent form of the compound differs from the various salt forms in certain physical properties, such as solubility in polar solvents, but otherwise the salts are equivalent to the parent form of the compound for the purposes of the present invention.
  • the present invention provides a method for activating an A 2B adenosine receptor in a mammal comprising administering to the mammal an effective amount of at least one compound of formula I or a pharmaceutically acceptable salt thereof.
  • the method can further comprise activating an A 2A adenosine receptor upon administration of the compound of formula I.
  • the effective amount can be a therapeutically effective amount or a prophylactically effective amount.
  • the compound can be administered acutely or chronically.
  • the present invention also provides a method for activating an A 2B adenosine receptor in a cell comprising contacting the cell with a compound or pharmaceutically acceptable salt of formula I.
  • the method can further comprise activating an A 2A adenosine receptor upon contacting the cell with a compound of formula I.
  • the method includes contacting the cell and the compound or salt of formula I is carried out in vitro, in vivo, or ex vivo.
  • the term "in vitro” means that the cell is not in a living organism.
  • the term “in vivo” means that the cell is a part of a living organism or is the living organism.
  • ex vivo refers to the administration of a compound to a cell or a population of cells in vitro, followed by administration of the cell or population of cells to a host.
  • the cell is in or from a host.
  • Hosts include, for example, bacteria, yeast, fungi, plants, and mammals.
  • the host is a mammal.
  • mammals include, but are not limited to, the order Rodentia, such as mice, and the order Logomorpha, such as rabbits. It is preferred that the mammals are from the order Carnivora, including Felines (cats) and Canines (dogs).
  • the mammals are from the order Artiodactyla, including Bovines (cows) and Swines (pigs) or of the order Perssodactyla, including Equines (horses). It is most preferred that the mammals are of the order Primates, Ceboids, or Simioids (monkeys) or of the order Anthropoids (humans and apes). An especially preferred mammal is the human.
  • the host can be the unborn offspring of any of the forgoing hosts, especially mammals (e.g., humans), in which case any screening of the host or cells of the host, or administration of compounds to the host or cells of the host, can be performed in utero.
  • the amount or dose of a compound of formula I, a salt thereof, or a composition thereof should be sufficient to affect a therapeutic or prophylactic response in the host over a reasonable time frame.
  • the appropriate dose will depend upon the nature and severity of the disease or affliction to be treated or prevented, as well as by other factors. For instance, the dose also will be determined by the existence, nature and extent of any adverse side effects that might accompany the administration of a particular compound or salt.
  • the attending physician will decide the dosage of the compound of the present invention with which to treat each individual patient, taking into consideration a variety of factors, such as age, body weight, general health, diet, sex, compound or salt to be administered, route of administration, and the severity of the condition being treated. Typical doses might be, for example, 0.1 mg to 1 g daily, such as 5 mg to 500 mg daily.
  • the inventive compounds are contemplated to be useful for treating disorders in which therapeutic treatment is effected by activating the A 2B adenosine receptor.
  • the method can further comprise treating disorders in which therapeutic treatment is effected by activating both the A 2A and A 2B adenosine receptors.
  • the present invention contemplates a method of treating or preventing a disorder in a mammal comprising administering a therapeutically effective amount of at least one compound of formula I or a pharmaceutically acceptable salt thereof to the mammal, whereupon the disorder is treated or prevented.
  • the disorder is any disorder in which activation of the A 2B or both the A 2B and A 2A adenosine receptors is beneficial in its treatment or prevention.
  • disorders include, for example, septic shock, cystic fibrosis, restenosis, erectile dysfunction, inflammation, and cardiac ischemia.
  • the compounds or pharmaceutically acceptable salts of the invention find use in the preparation of a medicament for activating A 2B adenosine receptors in a mammal.
  • the present invention also provides a method of treating or preventing (a) myocardial ischemia or (b) myocardial ischemia/reperfusion injury in a mammal comprising administering a therapeutically effective amount of at least one compound of formula I or a pharmaceutically acceptable salt thereof to the mammal, whereupon the (a) myocardial ischemia or (b) myocardial ischemia/reperfusion injury is treated or prevented.
  • a period of myocardial ischemia followed by reperfusion produces damage to the myocardium.
  • the compound of formula I or a salt thereof is administered for at least the first 24 hrs following reperfusion.
  • the compound of formula I or a salt thereof can limit infarct size when administered at the time of reperfusion. If desired, the compound of formula I or a salt thereof can be administered prior to reperfusion. In a particularly preferred embodiment of this method, the compound administered is 2-(3"-(6"-bromo-indolyl)ethyloxy)adenosine (i.e., compound 28) or a pharmaceutically acceptable salt thereof.
  • the compounds or pharmaceutically acceptable salts of the invention find use in the preparation of a medicament for treating or preventing myocardial ischemia or myocardial ischemia/reperfusion injury.
  • Ischemic preconditioning protects a reperfused heart by inhibiting the formation of permeability transition pores that normally form in many of the heart's mitochondria in the first minutes of reperfusion. These large conductance pores span both the inner and outer membrane and depolarize the matrix which uncouples the mitochondria and halts the oxidative phosphorylation of ADP to ATP. Energy production is thus inhibited at a time when the ischemic cells need it the most. If enough of the cell's mitochondria experience pore formation, then the cell will swell uncontrollably because it lacks the energy required to run the membrane ion pumps that maintain volume control. Those cells suffer lethal membrane rupture and die.
  • Ischemic preconditioning causes activation of ERK and PB -kinase at reperfusion, which ultimately act to inhibit transition pore formation at reperfusion.
  • the signal transduction pathway responsible for the kinase activation in the preconditioned heart includes the A 2B receptor (Kuno et al., J. MoI. Cell. Cardiol, in press (2007)).
  • the untreated heart is reperfused, there are 3 populations of cells present. The first population is killed by the ischemia itself and represents that volume of necrotic tissue seen in the drug- treated groups in Figure 8.
  • the second population includes those cells that have experienced a sub-lethal injury and will recover spontaneously with no treatment. That population represents the surviving tissue seen in the control hearts in Figure 8.
  • the third population represents cells that are still viable at reperfusion but will soon be killed by transition pore formation. It is that population that is salvaged by a drug of formula I (necrotic tissue in the control group minus that in the drug-treated groups in Figure 8).
  • Compounds of formula I that are agonists for both A 2A and A 2B should protect against myocardial ischemia/reperfusion injury.
  • the mammal is a patient prone to reperfusion injury, for example, a patient with coronary artery diseases in general or a patient about to have occluded arteries opened (reperfused) by one or more various interventions (e.g., coronary artery bypass grafts, angioplasty, or thrombolytic therapy).
  • various interventions e.g., coronary artery bypass grafts, angioplasty, or thrombolytic therapy.
  • System A linear gradient solvent system: CH 3 CN/H 2 O from 20/80 to 40/60 in 20 min; the flow rate is 1 mL/min.
  • System B linear gradient solvent system: CH 3 CNZH 2 O from 20/80 to 60/40 in 20 min; the flow rate is 1 mL/min.
  • System C linear gradient solvent system: CH 3 CN/5mM TBAP from 20/80 to 60/40 in 20 min.; the flow rate is 1 mL/min.
  • System D linear gradient solvent system: CH 3 CN/5mM TBAP from 5/95 to 80/20 in 20 min.; the flow rate is 1 mL/min. Peaks are detected by UV absorption with a diode array detector. All derivatives tested for biological activity show >98% purity in the HPLC systems.
  • TLC analysis is carried out on aluminum sheets pre-coated with silica gel F 254 (0.2 mm) from Aldrich.
  • Low-resolution mass spectrometry is performed with a JEOL SXl 02 spectrometer with 6-kV Xe atoms following desorption from a glycerol matrix or on an Agilent LC/MS 1100 MSD, with a Waters (Milford, MA) Atlantis Cl 8 column.
  • High resolution mass spectroscopic (HRMS) measurements are performed on a proteomics optimized Q-TOF-2 (Micromass- Waters) using external calibration using polyalanine. Observed mass accuracies are those expected based on known performance of the instrument as well as trends in masses of standard compounds observed at intervals during the series of measurements. Reported masses are observed masses uncorrected for this time-dependent drift in mass accuracy.
  • binding and functional parameters are calculated using Prism 4.0 software (GraphPAD, San Diego, CA, USA). IC 50 values obtained from competition curves are converted to K; values using the Cheng-Prusoff equation (Cheng et al., Biochem. Pharmacol. 1973, 22, 3099-3108). Data are expressed as mean + standard error.
  • This example demonstrates a general tosylation procedure for the synthesis of 3- iodoethylindole derivatives, 44-46, 56-58, 71-73 and 84 used in the preparation of compounds in accordance with an embodiment of the invention. See Figure 2.
  • THF tetrahydrofuran
  • sodium hydride 60 %, 3 eq
  • Tosyl chloride (3 eq) is added to the suspension at 0 °C, and the reaction mixture is stirred at room temperature overnight.
  • reaction mixture is diluted with ethyl acetate and washed with water, dried over MgSO 4 , and filtered. The filtrate is evaporated to give a crude oil, which is subjected to column chromatography on silica gel. Elution with a mixture of toluene and acetone (40: 1) gives the desired tosylated derivative.
  • Compound 53 is identical to the known compound reported by Mewshaw et al. (J.
  • Compound 54 is identical to the commercially available compound.
  • This example demonstrates a general synthetic procedure for 3- hydroxyethylindole derivatives (67-70) via Fischer indole ring preparation used in the preparation of compounds in accordance with an embodiment of the invention. See Figure 2.
  • a solution of substituted phenylhydrazine hydrochloride and ethoxytetrahydrofuran (1.5 eq) in 95 % ethanol is refluxed overnight.
  • the reaction mixture is filtered through celite.
  • the filtrate is evaporated to give a crude solid.
  • the solid is dissolved in ethyl acetate, and the solution is washed with water, dried over MgSO 4 , and filtered.
  • the filtrate is evaporated to give a crude oil, which is subjected to column chromatography on silica gel. Elution with a mixture of toluene and acetone (2:1) gives the alcohol.
  • 6-Chloro-tryptophol (67), 6-Bromo-tryptophol (68), and 5-ChIoro-tryptophol (69) [0096] These compounds are identical to the known compounds. 67: WO 2001/049679;
  • reaction mixture is stirred overnight at room temperature.
  • the reaction is diluted with ethyl acetate, washed with water, dried over MgSO 4 , and filtered.
  • the filtrate is evaporated to give an oil, which is subjected to preparative TLC developed with a mixture of toluene and acetone (1:1) to give 128 (56 mg, 50 %).
  • This example demonstrates a synthesis of 6-guanidino-2-(3"- indolylethyloxy)adenosine (37) and 6-guanidino-2-(3"-(l"-(p- toluenesulfonyl)indolyl)ethyloxy)adenosine (38) in an embodiment of the invention. See Figure 5.
  • CHO Cho et al., Biochem. Pharmacol. 2004, 68, 1985-1993
  • DMEM fetal bovine serum
  • penicillin 100 ⁇ g/mL streptomycin
  • 2 ⁇ mol/mL glutamine 2 ⁇ mol/mL glutamine
  • 800 ⁇ g/mL geneticin 800 ⁇ g/mL geneticin.
  • cells are centrifuged at 500 g for 10 min, and the pellet is re- suspended in 50 mM Tris-HCl buffer (pH 8.0) containing 10 mM MgCl 2 , 1 mM EDTA, and 0.1 mg/mL CHAPS.
  • the suspension is homogenized with an electric homogenizer for 10 sec, and is then re-centrifuged at 20,000 g for 20 min at 4 0 C.
  • the resultant pellets are re- suspended in buffer in the presence of 3 Units/ml adenosine deaminase, and the suspension is stored at -80 0 C until the binding experiments.
  • the protein concentration is measured using the Bradford assay (Bradford et al., Anal. Biochem. 1976, 72, 248-254).
  • [0208] For binding to human A 1 receptors [ 3 H]CCPA (1 nM) is incubated with membranes (40 ⁇ g/tube) from CHO cells stably expressing human Aj receptors at 25 0 C for 60 min in 50 mM Tris-HCl buffer (pH 7.4; MgCl 2 , 10 mM) in a total assay volume of 200 ⁇ L. Nonspecific binding is determined using 10 ⁇ M of CPA.
  • membranes (20 ⁇ g/tube) from HEK-293 cells stably expressing human A 2A receptors are incubated with 15 nM [ 3 H]CGS21680 at 25 0 C for 60 min in 200 ⁇ l 50 mM Tris-HCl, pH 7.4, containing 10 mM MgCl 2 .
  • NECA (10 ⁇ M) is used to define nonspecific binding. The reaction is terminated by filtration with GF/B filters.
  • each tube contains 100 ⁇ L membrane suspension (20 ⁇ g protein), 50 ⁇ L of [ 125 I]I-AB-MECA (0.5 nM), and 50 ⁇ L of increasing concentrations of the nucleoside derivative in Tris-HCl buffer (50 mM, pH 7.4) containing 10 mM MgCl 2 , 1 mM EDTA.
  • Nonspecific binding is determined using 10 ⁇ M of Cl-IB-MECA in the buffer.
  • the mixtures are incubated at 25 0 C for 60 min. Binding reactions are terminated by filtration through Whatman GF/B filters under reduced pressure using a MT-24 cell harvester (Brandell, Gaithersburgh, MD, USA).
  • R 4 C(NH)NH 2
  • R 4 C(NH)NH 2
  • R 2 CONHEt aPotency of various adenosine derivatives in activation of the human A 28 AR expressed in CHO cells, with values expressed either the EC 50 (nM) or the percent stimulation at 10 ⁇ M (in parentheses).
  • binding affinities of the adenosine derivatives at human Ai , A 2A , and A 3 ARs expressed in CHO cells expressed as Kj value or percent displacement at 10 ⁇ M
  • maximal agonist effects at 10 ⁇ M at the A 3 AR Values for compounds 5 - 7 and 9 - 16 are from Gao et al., Biochem. Pharmacol.
  • NECA S'-N-ethylcarboxaminocarbonyladenosine 2 was considered to be the most potent known agonist at the A 2B AR, with an EC 50 of 140 nM (Hide et al., MoI. Pharmacol. 1992, 41, 352-359; Gao et al., Biochem. Pharmacol. 2004, 68, 1985- 1993; and Klotz et al., Naunyn-Schmiedeberg's Arch. Pharmacol. 1998, 357, 1-9).
  • Compound 28 is a full agonist at the A 2A and A 2B adenosine receptors.
  • the EC 50 values for 2 are 21.9 nM (A 2 A, Figure 7B) and 110 nM (A 2B , Figure 7C).
  • the EC 50 values of compound 28 are 39.7 nM (A 2A , Figure 7B) and 109 nM (A 2B , Figure 7C) are obtained.
  • the relative maximal efficacy of 28 at the Ai and A 3 adenosine receptors is 31.8% and 20.2 ⁇ 1.0% of the full agonist 2, respectively.
  • Compound 28 is more selective to A 2B adeonosine receptors than compound 2.
  • This example illustrates an anti-infarct effect of compound 28 in an isolated rabbit heart model of ischemia/reperfusion injury in accordance with an embodiment of the invention.
  • New Zealand White rabbits are anesthetized with pentobarbital sodium (30 mg/kg i.v.) and ventilated with 100% oxygen. A suture is passed around a coronary arterial branch.
  • the excised heart is perfused on a Langendorff apparatus with Krebs-Henseleit bicarbonate buffer bubbled with 95% O 2 /5% CO 2 to a pH of 7.35 - 7.45 at 38 0 C.
  • a fluid-filled latex balloon measures pressure in the left ventricle as the heart spontaneously beats in an isometric fashion on the balloon.
  • a prominent branch of the left coronary artery is occluded (regional ischemia) for
  • the percentage of the ischemic region that infarcts in a rabbit heart is a function of the absolute size of the risk zone. Therefore, infarct size is examined by plotting it against the risk zone to make sure that the apparent salvage seen in the drug is not an artifact resulting from smaller risk zones in the treatment group. As shown in Figure 9, a line is fit to the control data with an intercept at about 0.35 cm 3 . This result means that no infarction would occur in an untreated heart with a 30-minute insult if the risk zone is less than 0.35 cm 3 . Figure 9 shows that all of the drug-treated hearts fell well below the line. Thus for any given risk zone volume, the infarcts are clearly smaller in the treated groups.
  • a single high dose (e.g., 160 ⁇ g/kg) bolus plus a continued administration (e.g., 1920 ⁇ g/kg/hr) of compound 28 provides a reduction in the size of the infarcted area.
  • a single high dose (e.g., 160 ⁇ g/kg) bolus plus a continued administration (e.g., 1920 ⁇ g/kg/hr) of compound 28 provides a reduction in the size of the infarcted area.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Vascular Medicine (AREA)
  • General Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Biotechnology (AREA)
  • Urology & Nephrology (AREA)
  • Biochemistry (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Molecular Biology (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Saccharide Compounds (AREA)

Abstract

Disclosed are AZB adenosine receptor (AR) agonists of formula (I), in which R1, R2, R3, R4, Z, and n are defined herein. The invention also provides compositions comprising at least one compound of formula I and methods of use thereof, for example, in the treatment of septic shock, cystic fibrosis, restenosis, erectile dysfunction, inflammation, myocardial ischemia, and reperfusion injury.

Description

A2 ADENOSINE RECEPTOR AGONISTS
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims the benefit of United States Provisional Patent Application Nos. 60/947,066, filed June 29, 2007 and 60/950,250, filed July 17, 2007, the disclosures of which are incorporated by reference.
BACKGROUND OF THE INVENTION
[0002] There are four subtypes of adenosine receptors (ARs): A1, A2A, A2B, and A3 (Fredholm et al. Pharmacol. Rev. 2001, 53, 527-552). Activation or blocking one or more of these ARs leads to useful therapeutic applications. For example, activation of the A2βAR can induce angiogenesis, reduce vascular permeabilization, increase production of the anti- inflammatory cytokine IL-10, increase chloride secretion in epithelial cells, or increase release of inflammatory mediators from human and canine mast cells. Accordingly, there is a desire for A2βAR agonists for use in therapeutic applications.
BRIEF SUMMARY OF THE INVENTION
[0003] The invention provides A2B adenosine receptor (AR) agonists of formula I. Exemplary compounds of formula I have enhanced potency at the A2βAR and reduced potency at other AR subtypes, wherein R1 -R4, Z, and n are described below in detail.
Figure imgf000002_0001
(I)
It is contemplated that some compounds of formula I are agonists for both A2A and A2B adenosine receptors.
[0004] The invention also provides compositions comprising at least one compound of formula I and methods of use thereof, for example, in a method of treating a disease based on the modulation of the adenosine system, e.g., septic shock, cystic fibrosis, restenosis, erectile dysfunction, inflammation, myocardial ischemia, and reperfusion injury.
BRIEF DESCRIPTION OF THE SEVERAL VIEWS OF THE DRAWINGS
[0005] Figure 1 depicts chemical structures of exemplary compounds of formula I (8 and
17-40), in accordance with an embodiment of the invention.
[0006] Figure 2 depicts a reaction scheme to prepare intermediates for the 2-ether component of compounds of formula I in accordance with an embodiment of the invention.
Step (a): TsCl, NaH, THF, 0 0C - room temperature; step (b): NaI, DMF, 60 0C; step (c) (i):
TsOH-H2O, MeOH, 60 0C; (ii): LAH, THF, 4 0C - room temperature; step (d): EtOH, reflux; step (e): oxalyl chloride, Et2O; step (f): LAH, THF, reflux; and step (g): I2, PPh3, imidazole, benzene.
[0007] Figure 3 depicts another reaction scheme to prepare intermediates for the 2-ether component of compounds of formula I in accordance with an embodiment of the invention.
Step (a) (i): TsCl, pyridine, CH2Cl2; (ii): 3-butyn-l-ol, Pd(PPh3)2Cl2, CuI, Et3N, DMF, 70 0C; step (b): I2, PPh3, imidazole, Et2O-MeCN, room temperature; and step (c): LAH, THF, 0 0C - room temperature.
[0008] Figure 4 depicts a reaction scheme to prepare compounds 8, 17-36, and 119-127 in accordance with an embodiment of the invention. Step (a): R1CH2CH2-I (47 - 49, 59-61,
74-77, 85, 86, l-iodo-3-phenylpropane, 89, 94, 95 and 99, respectively), Cs2CO3, DMF, room temperature; step (b): saturated NH3 in EtOH, 120 0C; and step (c): deprotection of tosyl group of 18, 119-121, 26, 32, 122-126, 21 and 127; KOH, MeOH 70 - 90 0C.
[0009] Figure 5 depicts a reaction scheme to prepare compound 39 in accordance with an embodiment of the invention. Step (a): guanidine solution (Jacobson et al., Chem. Biol. 2005,
12, 237-247), DABCO, EtOH, 110 0C; and step (b) (i): EtNH2 HCl, DIPEA, DMF, 140 0C;
(ii): KOH, MeOH, 80 0C.
[0010] Figure 6 depicts a reaction scheme to prepare compound 40 in accordance with an embodiment of the invention. Step (a): 2,2-dimethoxypropane, p-TsOH-H2O, DMF; step
(b): KMnO4, KOH, H2O; step (c): PyBop, DIPEA, EtNH2 HCl, DMF; step (d): f-butyl nitrite,
2-PrOH/H2O (1:1); step (e): iodide 47, Cs2CO3, DMF, room temperature; step (f): saturated
NH3, EtOH, 120 0C; step (g): 80% AcOH, 80 0C; and step (h): KOH, MeOH, 70 0C. [0011] Figure 7A depicts activation curves (log [Agonist] versus percent agonist efficacy) for 28 (^) in comparison to 2 (■) at the Ai adenosine receptors.
[0012] Figure 7B depicts activation curves (log [Agonist] versus percent agonist efficacy) for 28 (^) in comparison to 2 (■) at the A2A adenosine receptors.
[0013] Figure 7C depicts activation curves (log [Agonist] versus percent agonist efficacy) for 28 (A) in comparison to 2 (■) at the A2B adenosine receptors.
[0014] Figure 7D depicts activation curves (log [Agonist] versus percent agonist efficacy) for 28 (^) in comparison to 2 (■) at the A3 adenosine receptors.
[0015] Figure 8 depicts the percent of risk zone infracted in an isolated rabbit heart for a control group and a group that had been administered 200 nM of 28 in accordance with an embodiment of the invention.
[0016] Figure 9 depicts the infarct size (cm ) as a function of risk size (cm ) in accordance with an embodiment of the invention. The symbol • represents the control group, whereas O represents 28.
DETAILED DESCRIPTION OF THE INVENTION
[0017] The present invention provides a compound of formula I or pharmaceutically acceptable salt thereof:
Figure imgf000004_0001
(I) wherein
R1 is selected from the group consisting of aryl, cycloalkylaryl, heterocycloalkylaryl, heteroaryl, heterocycloalkyl, and arylheterocycloalkyl, each of which is optionally substituted with 1 to 3 substituents selected from the group consisting of CM2 alkyl, hydroxyl, Ci-I2 alkoxy, halo, tosyl (Ts), CN, -C(O)OH, aminocarbonyl, amino, C1-J2 dialkylamino, and Ci-J2 alkylamino;
Z is a bond or -CH2-;
n is an integer of 1 to 4;
R2 is selected from the group consisting Of-CH2OH, aminocarbonyl, Cj-J2 alkylaminocarbonyl, and di-Cj.j alkylaminocarbonyl; and
R3 and R4 are the same or different and each is selected from the group consisting of hydrogen, CJ-CJ2 alkyl, C3-C8 cycloalkyl, C6-C30 aryl, and imidamido;
provided that when R2 is -CH2OH, and R3 and R4 are hydrogen, then -0-(CH2)H-Z-R1 is not benzyloxy, phenylalkoxy or a substituted phenylalkoxy, thiophenylalkoxy or a substituted thiophenylalkoxy, pyridylalkoxy, indolylalkoxy, naphthylalkoxy, biphenylalkoxy, or indolylalkoxy.
[0018] In preferred embodiments, R1 is selected from the group consisting Of C6-3O aryl, heteroaryl, heterocycloalkyl, C5-7 cycloalkyl-C6-3o aryl, heterocycloalkyl-Ce^o aryl, C6-30 aryl- heteroaryl, and C6-30 aryl-heterocycloalkyl. For example, R1 is selected from the group consisting of
C6-3O aryl (e.g., phenyl, biphenyl, naphthyl, anthracenyl, pyrenyl), a heteroaryl consisting of one or two oxygen or sulfur atoms and/or from one to four nitrogen atoms provided that the total number of heteroatoms in each ring is four or less and each ring has at least one carbon atom (e.g., pyridinyl, pyridazinyl, pyrimidyl, pyrazinyl, triazinyl, pyrrolyl, pyrazolyl, imidazolyl, (1,2,3,)- and (l,2,4)-triazolyl, pyrazinyl, pyrimidinyl, tetrazolyl, furyl, thiophenyl, isothiazolyl, thiazolyl, isoxazolyl, oxadiazolyl, oxazolyl, pyrrolo[2,3-c]pyridinyl, pyrrolo[3,2-c]pyridinyl, pyrrolo[2,3-b]pyridinyl, pyrrolo[3,2-b]pyridinyl, pyrrolo[3,2-d]pyrimidinyl, and pyrrolo[2,3- d]pyrimidinyl), a heterocycloalkyl consisting of a 5 or 6-membered monocyclic ring that contains one, two, or three heteroatoms selected from nitrogen, oxygen, and/or sulfur (e.g., pyrrolinyl, pyranyl, piperidyl, tetrahydrofuranyl, tetrahydrothiopheneyl, and moφholinyl), C5-7 cycloalkyl-fused phenyl (e.g., 5-, 6-, 7-, or 8-1,2,3,4-tetrahydronaphthalenyl and 4-, 5-, 6-, or 7-2,3-dihydro-lH-indenyl), heterocycloalkyl-fused phenyl, wherein the heterocycloalkyl consists of a 5 or 6- membered monocyclic ring that contains one, two, or three heteroatoms selected from nitrogen, oxygen, and/or sulfur (e.g., 5-, 6-, 7-, or 8-1,2,3,4- tetrahydroquinolinyl, 5-, 6-, 7-, or 8-1,2,3,4-tetrahydroquinoxalinyl, and 5-, 6-, 7-, or 8-2,3-dihydrobenzo[b][l,4]dioxinyl), a benzo-fused heteroaryl, wherein the heteroaryl consists of one or two oxygen or sulfur atoms and/or from one to four nitrogen atoms provided that the total number of heteroatoms in each ring is four or less and each ring has at least one carbon atom (e.g., benzimidazolyl, indolyl, indazolyl, benzo-1,2,3- triazolyl, 2-benzo[d]oxazolyl, 2-benzo[d]thiazolyl, 1,- 3-,or 4-isoquinolinyl, 2-, 3-, or 4-quinolinyl, and 2- or 3-quinoxalinyl), and a benzo-fused heterocycloalkyl, wherein the heterocycloalkyl consists of a 5 or 6- membered monocyclic ring that contains one, two, or three heteroatoms selected from N, O, and/or S (e.g., 2,3-dihydrobenzo[b][l,4]dioxinyl, 2- indolinyl, and 3-indolinyl). [0019] In particularly preferred embodiments, R1 is selected from the group consisting of
Figure imgf000006_0001
Figure imgf000007_0001
wherein
R5 and R5 are the same or different and each is hydrogen or 1 to 3 substituents individually selected from the group consisting of Ci-I2 alkyl, hydroxyl, Ci-I2 alkoxy, halo, tosyl (Ts), CN, -C(O)OH, aminocarbonyl, amino, Ci-I2 dialkylamino, and Ci-I2 alkylamino; and
R6 is selected from the group consisting of hydrogen, Ci-I2 alkyl, C3-8 cycloalkyl, and C6-30 aryl.
[0020] For example, R1 is selected from the group consisting of
Figure imgf000007_0002
Specific embodiments of these compounds include wherein R1 is selected from the group consisting of
Figure imgf000007_0004
and
Figure imgf000007_0003
[0021] In any of the foregoing embodiments, R5 and R5 are the same or different and each can be hydrogen, Ci-I2 alkyl, hydroxyl, C1-I2 alkoxy, halo, or tosyl. For instance, in preferred compounds, R1 is selected from the group consisting of
Figure imgf000008_0001
[0022] In an embodiment of the invention, in the compound of formula I, n is 1, 2, or 3, particularly n is 2 or 3. In especially preferred compounds, n is 2.
[0023] In another embodiment of the invention, R2 is -CH2OH or CM2 alkylaminocarbonyl (e.g., N-methylcarboxaminocarbonyl, N-ethylcarboxaminocarbonyl, N- propylcarboxaminocarbonyl) .
[0024] In preferred embodiments of the invention, R3 and R4 are the same or different and each is selected from the group consisting of hydrogen, Ci-Ci2 alkyl, and imidamido.
Compounds in which R3 is hydrogen and R4 is hydrogen, Ci-I2 alkyl, or imidamido are especially preferred. Figure 1 depicts the chemical structures of exemplary compounds of formula I.
[0025] Referring now to terminology used generically herein, the term "alkyl" implies a straight or branched alkyl moiety containing from, for example, 1 to 12 carbon atoms, preferably from 1 to 8 carbon atoms, more preferably from 1 to 6 carbon atoms. Examples of such moieties include methyl, ethyl, propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, pentyl, isoamyl, hexyl, octyl, dodecanyl, and the like.
[0026] The term "cycloalkyl," as used herein, means a cyclic moiety containing from, for example, 1-3 rings (i.e., monocyclic, bicyclic, tricyclic, or spiro), 3 to 8 carbon atoms per ring, preferably from 5 to 8 carbon atoms, more preferably from 5 to 6 carbon atoms. Examples of such moieties include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like.
[0027] The term "aryl" refers to an unsubstituted or substituted aromatic carbocyclic moiety, as commonly understood in the art, and includes monocyclic and polycyclic aromatics such as, for example, phenyl, biphenyl, naphthyl, anthracenyl, pyrenyl, and the like. An aryl moiety generally contains from, for example, 6 to 30 carbon atoms, preferably from 6 to 18 carbon atoms, more preferably from 6 to 14 carbon atoms and most preferably from 6 to 10 carbon atoms. It is understood that the term aryl includes carbocyclic moieties that are planar and comprise 4n+2 π electrons, according to Hϋckel's Rule, wherein n = 1, 2, or 3.
[0028] The term "heterocycloalkyl" means a cycloalkyl moiety having one or more heteroatoms selected from nitrogen, sulfur, and/or oxygen. Preferably, a heterocycloalkyl is a 5 or 6-membered monocyclic ring and contains one, two, or three heteroatoms selected from nitrogen, oxygen, and/or sulfur. The heterocycloalkyl can be attached to the parent structure through a carbon atom or through any heteroatom of the heterocycloalkyl that results in a stable structure. Examples of such heterocyclic rings are pyrrolinyl, pyranyl, piperidyl, tetrahydrofuranyl, tetrahydrothiopheneyl, and morpholinyl.
[0029] The term "arylheterocycloalkyl" refers to a heterocycloalkyl, as defined herein, that is substituted with an aryl group, as defined herein, as a fused ring, e.g., benzo. Examples include 2,3-dihydrobenzo[b][l,4]dioxinyl, 2-indolinyl, and 3-indolinyl. [0030] The terms "cycloalkylaryl" and "heterocycloalkylaryl" refer to an aryl, as defined herein, that is substituted with a cycloalkyl group or a heterocycloalkyl group, respectively, as defined herein, or as a fused ring, e.g., benzo. Examples of cycloalkylaryl groups include 5-, 6-, 7-, or 8-1,2,3,4-tetrahydronaphthalenyl and 4-, 5-, 6-, or 7-2,3-dihydro-lH-indenyl. Examples of heterocycloalkylaryl include 5-, 6-, 7-, or 8-1,2,3,4-tetrahydroquinolinyl, 5-, 6-, 7-, or 8-1,2,3,4-tetrahydroquinoxalinyl, and 5-, 6-, 7-, or 8-2,3-dihydrobenzo[b][l,4]dioxinyl. [0031] The term "heteroaryl" refers to aromatic 4, 5, or 6 membered monocyclic groups, 9 or 10 membered bicyclic groups, and 11 to 14 membered tricyclic aryl groups having one or more heteroatoms (O, S, or N). Each ring of the heteroaryl group containing a heteroatom can contain one or two oxygen or sulfur atoms and/or from one to four nitrogen atoms provided that the total number of heteroatoms in each ring is four or less and each ring has at least one carbon atom. The nitrogen and sulfur atoms may optionally be oxidized, and the nitrogen atoms may optionally be quaternized. The heteroaryl group can be attached at any available nitrogen or carbon atom of any ring. Illustrative examples of heteroaryl groups are pyridinyl, pyridazinyl, pyrimidyl, pyrazinyl, triazinyl, pyrrolyl, pyrazolyl, imidazolyl,
(1,2,3,)- and (l,2,4)-triazolyl, pyrimidinyl, tetrazolyl, furyl, thiophenyl, isothiazolyl, thiazolyl, isoxazolyl, oxadiazolyl, oxazolyl, pyrrolo[2,3-c]pyridinyl, pyrrolo[3,2-c]pyridinyl, pyrrolo[2,3-b]pyridinyl, pyrrolo[3,2-b]pyridinyl, pyrrolo[3,2-d]pyrimidinyl, and pyrrolo[2,3- d]pyrimidinyl.
[0032] The term "arylheteroaryl" refers to heteroaryl, as defined herein, that is substituted with an aryl group, as defined herein, as a fused ring, e.g., benzo. Examples of arylheteroaryl groups include benzimidazolyl, indolyl, indazolyl, benzo-l,2,3-triazolyl, 2-benzo[d] oxazolyl,
2-benzo[d]thiazolyl, 1,- 3-,or 4-isoquinolinyl, 2-, 3-, or 4-quinolinyl, and 2- or 3- quinoxalinyl.
[0033] The term "alkoxy" embraces linear or branched alkyl groups that are attached to a an ether oxygen. The alkyl group is the same as described herein. Examples of such substituents include methoxy, ethoxy, t-butoxy, and the like.
[0034] The term "alkylamino" refers to a group with one hydrogen and one alkyl group directly attached to a trivalent nitrogen atom. The term "dialkylamino" refers to a group with two of the same or different alkyl groups directly attached to a trivalent nitrogen atom.
[0035] The term "aminocarbonyl" refers to the group -C(O)NH2. The terms
"alkylaminocarbonyl" and "dialkylaminocarbonly" refer to the group -C(O)NRR', in which
R is hydrogen or an alkyl group and R' is the same or different alkyl group as described herein.
[0036] The term "imidamido," also known as guanidino, refers to the group
-C(=NH)NH2.
[0037] The term "halo" as used herein, means a substituent selected from Group VIIA, such as, for example, fluorine, bromine, chlorine, and iodine.
[0038] For the purpose of the present invention, the term "fused" includes a polycyclic compound in which one ring contains one or more atoms preferably one, two, or three atoms in common with one or more other rings.
[0039] Whenever a range of the number of atoms in a structure is indicated (e.g., a Ci-I2,
Ci-8, Ci-6, or CM alkyl, alkylamino, etc.), it is specifically contemplated that any sub-range or individual number of carbon atoms falling within the indicated range also can be used. Thus, for instance, the recitation of a range of 1-8 carbon atoms (e.g., Cj-Cg), 1-6 carbon atoms (e.g., CI-CO), 1-4 carbon atoms (e.g., Ci-C4), 1-3 carbon atoms (e.g., C1-C3), or 2-8 carbon atoms (e.g., C2-C8) as used with respect to any chemical group (e.g., alkyl, alkylamino, etc.) referenced herein encompasses and specifically describes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, and/or 12 carbon atoms, as appropriate, as well as any sub-range thereof (e.g., 1-2 carbon atoms, 1-3 carbon atoms, 1-4 carbon atoms, 1-5 carbon atoms, 1-6 carbon atoms, 1-7 carbon atoms, 1-8 carbon atoms, 1-9 carbon atoms, 1-10 carbon atoms, 1-11 carbon atoms, 1-12 carbon atoms, 2-3 carbon atoms, 2-4 carbon atoms, 2-5 carbon atoms, 2-6 carbon atoms, 2-7 carbon atoms, 2-8 carbon atoms, 2-9 carbon atoms, 2-10 carbon atoms, 2-11 carbon atoms, 2- 12 carbon atoms, 3-4 carbon atoms, 3-5 carbon atoms, 3-6 carbon atoms, 3-7 carbon atoms, 3-8 carbon atoms, 3-9 carbon atoms, 3-10 carbon atoms, 3-11 carbon atoms, 3-12 carbon atoms, 4-5 carbon atoms, 4-6 carbon atoms, 4-7 carbon atoms, 4-8 carbon atoms, 4-9 carbon atoms, 4-10 carbon atoms, 4-11 carbon atoms, and/or 4-12 carbon atoms, etc., as appropriate).
[0040] The compounds of formula I can be prepared by any suitable method. For example, 2', 3', 5'-triacetyl-6-chloroguanosine can be synthetically modified to prepare 2- ether-substituted 2', 3', 5'-triacetyl-6-chloroadenosine derivatives. To make a nucleophilic substitution of the 6-chloro group, a suitable amine (e.g., ammonia, ethylamine hydrochloride, guanidine hydrochloride) can be added. In case of deprotection of a protecting group (e.g., tosyl), the adenosine derivative is treated with a suitable strong acid or base (e.g., KOH) to form the compounds of formula I. Specific embodiments of the preparation of compounds of formula I and intermediates thereof are described in the Examples and in Figures 2-6.
[0041] Isolation and purification of the compounds and intermediates described herein can be effected, if desired, by any suitable separation or purification procedure such as, for example, filtration, extraction, crystallization, column chromatography, thin-layer chromatography, thick-layer chromatography, preparative low or high-pressure liquid chromatography, or a combination of these procedures. Specific illustrations of suitable separation and isolation procedures can be had by reference to the Examples described herein. However, other equivalent separation or isolation procedures can also be used. [0042] The present invention further provides a pharmaceutical composition comprising at least one compound of formula I or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier. The pharmaceutically acceptable carrier can be any of those conventionally used and is limited only by chemico-physical considerations, such as solubility and lack of reactivity with the compound, and by the route of administration. It will be appreciated by one of skill in the art that, in addition to the following described pharmaceutical compositions; the compounds of the present invention can be formulated as inclusion complexes, such as cyclodextrin inclusion complexes or liposomes. [0043] The pharmaceutically acceptable carriers described herein, for example, vehicles, adjuvants, excipients, or diluents, are well known to those who are skilled in the art and are readily available to the public. It is preferred that the pharmaceutically acceptable carrier be one which is chemically inert to the active compounds and one which has no detrimental side effects or toxicity under the conditions of use.
[0044] The choice of carrier will be determined in part by the particular active agent, as well as by the particular method used to administer the composition. Accordingly, there is a wide variety of suitable formulations of the pharmaceutical composition of the present invention. The following formulations for oral, aerosol, parenteral, subcutaneous, intravenous, intraarterial, intramuscular, interperitoneal, intrathecal, rectal, and vaginal administration are merely exemplary and are in no way limiting.
[0045] Formulations suitable for oral administration can consist of (a) liquid solutions, such as an effective amount of the compound dissolved in diluents, such as water, saline, or orange juice; (b) capsules, sachets, tablets, lozenges, and troches, each containing a predetermined amount of the active ingredient, as solids or granules; (c) powders; (d) suspensions in an appropriate liquid; and (e) suitable emulsions. Liquid formulations may include diluents, such as water and alcohols, for example, ethanol, benzyl alcohol, and the polyethylene alcohols, either with or without the addition of a pharmaceutically acceptable surfactant, suspending agent, or emulsifying agent. Capsule forms can be of the ordinary hard- or soft-shelled gelatin type containing, for example, surfactants, lubricants, and inert fillers, such as lactose, sucrose, calcium phosphate, and cornstarch. Tablet forms can include one or more of lactose, sucrose, mannitol, corn starch, potato starch, alginic acid, microcrystalline cellulose, acacia, gelatin, guar gum, colloidal silicon dioxide, croscarmellose sodium, talc, magnesium stearate, calcium stearate, zinc stearate, stearic acid, and other excipients, colorants, diluents, buffering agents, disintegrating agents, moistening agents, preservatives, flavoring agents, and pharmacologically compatible carriers. Lozenge forms can comprise the active ingredient in a flavor, usually sucrose and acacia or tragacanth, as well as pastilles comprising the active ingredient in an inert base, such as gelatin and glycerin, or sucrose and acacia, emulsions, gels, and the like containing, in addition to the active ingredient, such carriers as are known in the art.
[0046] The compounds of the present invention, alone or in combination with other suitable components, can be made into aerosol formulations to be administered via inhalation. These aerosol formulations can be placed into pressurized acceptable propellants, such as dichlorodifluoromethane, propane, nitrogen, and the like. They also may be formulated as pharmaceuticals for non-pressured preparations, such as in a nebulizer or an atomizer. [0047] Formulations suitable for parenteral administration include aqueous and nonaqueous, isotonic sterile injection solutions, which can contain anti-oxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the intended recipient, and aqueous and non-aqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizers, and preservatives. The compound can be administered in a physiologically acceptable diluent in a pharmaceutical carrier, such as a sterile liquid or mixture of liquids, including water, saline, aqueous dextrose and related sugar solutions, an alcohol, such as ethanol, isopropanol, or hexadecyl alcohol, glycols, such as propylene glycol or polyethylene glycol, glycerol ketals, such as 2,2-dimethyl-l,3-dioxolane- 4-methanol, ethers, such as poly(ethyleneglycol) 400, an oil, a fatty acid, a fatty acid ester or glyceride, or an acetylated fatty acid glyceride with or without the addition of a pharmaceutically acceptable surfactant, such as a soap or a detergent, suspending agent, such as pectin, carbomers, methylcellulose, hydroxypropylmethylcellulose, or carboxymethylcellulose, or emulsifying agents and other pharmaceutical adjuvants. [0048] Oils, which can be used in parenteral formulations include petroleum, animal, vegetable, or synthetic oils. Specific examples of oils include peanut, soybean, sesame, cottonseed, corn, olive, petrolatum, and mineral. Suitable fatty acids for use in parenteral formulations include oleic acid, stearic acid, and isostearic acid. Ethyl oleate and isopropyl myristate are examples of suitable fatty acid esters. Suitable soaps for use in parenteral formulations include fatty alkali metal, ammonium, and triethanolamine salts, and suitable detergents include (a) cationic detergents such as, for example, dimethyl dialkyl ammonium halides, and alkyl pyridinium halides, (b) anionic detergents such as, for example, alkyl, aryl, and olefin sulfonates, alkyl, olefin, ether, and monoglyceride sulfates, and sulfosuccinates, (c) nonionic detergents such as, for example, fatty amine oxides, fatty acid alkanolamides, and polyoxyethylene-polypropylene copolymers, (d) amphoteric detergents such as, for example, alkyl-beta-aminopropionates, and 2-alkyl-imidazoline quaternary ammonium salts, and (3) mixtures thereof.
[0049] The parenteral formulations will typically contain from about 0.5 to about 25% by weight of the active ingredient in solution. Suitable preservatives and buffers can be used in such formulations. In order to minimize or eliminate irritation at the site of injection, such compositions may contain one or more nonionic surfactants having a hydrophile-Iipophile balance (HLB) of from about 12 to about 17. The quantity of surfactant in such formulations ranges from about 5 to about 15% by weight. Suitable surfactants include polyethylene sorbitan fatty acid esters, such as sorbitan monooleate and the high molecular weight adducts of ethylene oxide with a hydrophobic base, formed by the condensation of propylene oxide with propylene glycol. The parenteral formulations can be presented in unit-dose or multi- dose sealed containers, such as ampoules and vials, and can be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example, water, for injections, immediately prior to use. Extemporaneous injection solutions and suspensions can be prepared from sterile powders, granules, and tablets of the kind previously described.
[0050] The compounds of the present invention may be made into injectable formulations. The requirements for effective pharmaceutical carriers for injectable compositions are well known to those of ordinary skill in the art. See Pharmaceutics and Pharmacy Practice, J. B. Lippincott Co., Philadelphia, Pa., Banker and Chalmers, eds., pages 238-250 (1982), and ASHP Handbook on Injectable Drugs, Toissel, 4th ed., pages 622-630 (1986).
[0051] Additionally, the compounds of the present invention may be made into suppositories by mixing with a variety of bases, such as emulsifying bases or water-soluble bases. Formulations suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams, or spray formulas containing, in addition to the active ingredient, such carriers as are known in the art to be appropriate.
[0052] Suitable carriers and their formulations are further described in A.R. Gennaro, ed., Remington: The Science and Practice of Pharmacy (19th ed.), Mack Publishing Company, Easton, PA (1995).
[0053] The compound of the invention or a composition thereof can potentially be administered as a pharmaceutically acceptable acid-addition, base neutralized or addition salt, formed by reaction with inorganic acids, such as hydrochloric acid, hydrobromic acid, perchloric acid, nitric acid, thiocyanic acid, sulfuric acid, and phosphoric acid, and organic acids such as formic acid, acetic acid, propionic acid, glycolic acid, lactic acid, pyruvic acid, oxalic acid, malonic acid, succinic acid, maleic acid, and fumaric acid, or by reaction with an inorganic base, such as sodium hydroxide, ammonium hydroxide, potassium hydroxide, and organic bases, such as mono-, di-, trialkyl, and aryl amines and substituted ethanolamines. The conversion to a salt is accomplished by treatment of the base compound with at least a stoichiometric amount of an appropriate acid. Typically, the free base is dissolved in an inert organic solvent such as diethyl ether, ethyl acetate, chloroform, ethanol, methanol, and the like, and the acid is added in a similar solvent. The mixture is maintained at a suitable temperature (e.g., between 0 0C and 50 0C). The resulting salt precipitates spontaneously or can be brought out of solution with a less polar solvent.
[0054] The neutral forms of the compounds can be regenerated by contacting the salt with a base or acid and isolating the parent compound in the conventional manner. The parent form of the compound differs from the various salt forms in certain physical properties, such as solubility in polar solvents, but otherwise the salts are equivalent to the parent form of the compound for the purposes of the present invention. [0055] The present invention provides a method for activating an A2B adenosine receptor in a mammal comprising administering to the mammal an effective amount of at least one compound of formula I or a pharmaceutically acceptable salt thereof. The method can further comprise activating an A2A adenosine receptor upon administration of the compound of formula I. The effective amount can be a therapeutically effective amount or a prophylactically effective amount. In addition, the compound can be administered acutely or chronically.
[0056] The present invention also provides a method for activating an A2B adenosine receptor in a cell comprising contacting the cell with a compound or pharmaceutically acceptable salt of formula I. The method can further comprise activating an A2A adenosine receptor upon contacting the cell with a compound of formula I. The method includes contacting the cell and the compound or salt of formula I is carried out in vitro, in vivo, or ex vivo. As used herein, the term "in vitro" means that the cell is not in a living organism. As used herein, the term "in vivo" means that the cell is a part of a living organism or is the living organism. The term "ex vivo" as used herein refers to the administration of a compound to a cell or a population of cells in vitro, followed by administration of the cell or population of cells to a host. [0057] Preferably the cell is in or from a host. Hosts include, for example, bacteria, yeast, fungi, plants, and mammals. Preferably, the host is a mammal. For purposes of the present invention, mammals include, but are not limited to, the order Rodentia, such as mice, and the order Logomorpha, such as rabbits. It is preferred that the mammals are from the order Carnivora, including Felines (cats) and Canines (dogs). It is more preferred that the mammals are from the order Artiodactyla, including Bovines (cows) and Swines (pigs) or of the order Perssodactyla, including Equines (horses). It is most preferred that the mammals are of the order Primates, Ceboids, or Simioids (monkeys) or of the order Anthropoids (humans and apes). An especially preferred mammal is the human. Furthermore, the host can be the unborn offspring of any of the forgoing hosts, especially mammals (e.g., humans), in which case any screening of the host or cells of the host, or administration of compounds to the host or cells of the host, can be performed in utero.
[0058] The amount or dose of a compound of formula I, a salt thereof, or a composition thereof should be sufficient to affect a therapeutic or prophylactic response in the host over a reasonable time frame. The appropriate dose will depend upon the nature and severity of the disease or affliction to be treated or prevented, as well as by other factors. For instance, the dose also will be determined by the existence, nature and extent of any adverse side effects that might accompany the administration of a particular compound or salt. Ultimately, the attending physician will decide the dosage of the compound of the present invention with which to treat each individual patient, taking into consideration a variety of factors, such as age, body weight, general health, diet, sex, compound or salt to be administered, route of administration, and the severity of the condition being treated. Typical doses might be, for example, 0.1 mg to 1 g daily, such as 5 mg to 500 mg daily.
[0059] Moreover, since the compound of formula I or pharmaceutically salt thereof acts as an agonist of an A2B adenosine receptor, the inventive compounds are contemplated to be useful for treating disorders in which therapeutic treatment is effected by activating the A2B adenosine receptor. The method can further comprise treating disorders in which therapeutic treatment is effected by activating both the A2A and A2B adenosine receptors. More specifically, the present invention contemplates a method of treating or preventing a disorder in a mammal comprising administering a therapeutically effective amount of at least one compound of formula I or a pharmaceutically acceptable salt thereof to the mammal, whereupon the disorder is treated or prevented. The disorder is any disorder in which activation of the A2B or both the A2B and A2A adenosine receptors is beneficial in its treatment or prevention. Such disorders include, for example, septic shock, cystic fibrosis, restenosis, erectile dysfunction, inflammation, and cardiac ischemia. The compounds or pharmaceutically acceptable salts of the invention find use in the preparation of a medicament for activating A2B adenosine receptors in a mammal. [0060] The present invention also provides a method of treating or preventing (a) myocardial ischemia or (b) myocardial ischemia/reperfusion injury in a mammal comprising administering a therapeutically effective amount of at least one compound of formula I or a pharmaceutically acceptable salt thereof to the mammal, whereupon the (a) myocardial ischemia or (b) myocardial ischemia/reperfusion injury is treated or prevented. A period of myocardial ischemia followed by reperfusion produces damage to the myocardium. In a particularly preferred embodiment, the compound of formula I or a salt thereof is administered for at least the first 24 hrs following reperfusion. In this method, the compound of formula I or a salt thereof can limit infarct size when administered at the time of reperfusion. If desired, the compound of formula I or a salt thereof can be administered prior to reperfusion. In a particularly preferred embodiment of this method, the compound administered is 2-(3"-(6"-bromo-indolyl)ethyloxy)adenosine (i.e., compound 28) or a pharmaceutically acceptable salt thereof. The compounds or pharmaceutically acceptable salts of the invention find use in the preparation of a medicament for treating or preventing myocardial ischemia or myocardial ischemia/reperfusion injury.
[0061] Ischemic preconditioning protects a reperfused heart by inhibiting the formation of permeability transition pores that normally form in many of the heart's mitochondria in the first minutes of reperfusion. These large conductance pores span both the inner and outer membrane and depolarize the matrix which uncouples the mitochondria and halts the oxidative phosphorylation of ADP to ATP. Energy production is thus inhibited at a time when the ischemic cells need it the most. If enough of the cell's mitochondria experience pore formation, then the cell will swell uncontrollably because it lacks the energy required to run the membrane ion pumps that maintain volume control. Those cells suffer lethal membrane rupture and die.
[0062] Ischemic preconditioning causes activation of ERK and PB -kinase at reperfusion, which ultimately act to inhibit transition pore formation at reperfusion. The signal transduction pathway responsible for the kinase activation in the preconditioned heart includes the A2B receptor (Kuno et al., J. MoI. Cell. Cardiol, in press (2007)). When the untreated heart is reperfused, there are 3 populations of cells present. The first population is killed by the ischemia itself and represents that volume of necrotic tissue seen in the drug- treated groups in Figure 8. The second population includes those cells that have experienced a sub-lethal injury and will recover spontaneously with no treatment. That population represents the surviving tissue seen in the control hearts in Figure 8. The third population represents cells that are still viable at reperfusion but will soon be killed by transition pore formation. It is that population that is salvaged by a drug of formula I (necrotic tissue in the control group minus that in the drug-treated groups in Figure 8).
[0063] There is a second wave of killing that is not detected by either of the models used in this study. In the blood-perfused heart, ischemically injured tissue that is still alive can become pro-inflammatory. Leukocytes and particularly lymphocytes can attack these cells and kill some of them over the first 24 hr of reperfusion (Yang et al., Circulation, 111: 2190- 2197 (2005)). A2A-selective agonists attenuate this process, and thus induce salvage by a mechanism unrelated to that from A2B agonists (Glover et al., Am. J. Physiol., 288: H1851- H1858 (2005)). Thus, some of those cells that survive the first several hours of reperfusion will be targeted 6-8 hr later by a second wave of cell death due to inflammation. Compounds of formula I that are agonists for both A2A and A2B (e.g., compound 28) should protect against myocardial ischemia/reperfusion injury.
[0064] The mammal is a patient prone to reperfusion injury, for example, a patient with coronary artery diseases in general or a patient about to have occluded arteries opened (reperfused) by one or more various interventions (e.g., coronary artery bypass grafts, angioplasty, or thrombolytic therapy).
[0065] The following examples further illustrate the invention but, of course, should not be construed as in any way limiting its scope.
Materials and instrumentation
[0066] 2-Amino-6-chloropurine-9-riboside, tryptophol, l-iodo-3-phenylpropane, 5- bromoindole-3 -acetic acid, 5-methoxyindole-3-acetic acid and other reagents and solvents are purchased from Sigma-Aldrich (St. Louis, MO), and 5-fluoroindole-3-acetic acid is purchased from Wako Chemicals USA, Inc. (Richmond, VA). Compound 69 is prepared as reported (Chou et al., Heterocycles 2003, 60, 1095-1110).
[0067] 1H NMR spectra are obtained with a Varian Gemini 300 spectrometer using
CDCl3, CD3OD as solvents. Chemical shifts are expressed in δ values (ppm) with tetramethylsilane (50.00) for CDCl3 and (53.30) for CD3OD. [0068] Purity of the nucleosides submitted for biological testing is checked using a Hewlett-Packard 1100 HPLC equipped with a Luna 5μ RP-C 18(2) analytical column (250 X 4.6 mm; Agilent Technologies, Santa Clara, CA). System A: linear gradient solvent system: CH3CN/H2O from 20/80 to 40/60 in 20 min; the flow rate is 1 mL/min., System B: linear gradient solvent system: CH3CNZH2O from 20/80 to 60/40 in 20 min; the flow rate is 1 mL/min., System C: linear gradient solvent system: CH3CN/5mM TBAP from 20/80 to 60/40 in 20 min.; the flow rate is 1 mL/min., System D: linear gradient solvent system: CH3CN/5mM TBAP from 5/95 to 80/20 in 20 min.; the flow rate is 1 mL/min. Peaks are detected by UV absorption with a diode array detector. All derivatives tested for biological activity show >98% purity in the HPLC systems.
[0069] TLC analysis is carried out on aluminum sheets pre-coated with silica gel F254 (0.2 mm) from Aldrich. Low-resolution mass spectrometry is performed with a JEOL SXl 02 spectrometer with 6-kV Xe atoms following desorption from a glycerol matrix or on an Agilent LC/MS 1100 MSD, with a Waters (Milford, MA) Atlantis Cl 8 column. High resolution mass spectroscopic (HRMS) measurements are performed on a proteomics optimized Q-TOF-2 (Micromass- Waters) using external calibration using polyalanine. Observed mass accuracies are those expected based on known performance of the instrument as well as trends in masses of standard compounds observed at intervals during the series of measurements. Reported masses are observed masses uncorrected for this time-dependent drift in mass accuracy.
[0070] In the pharmacological methods, [125I]N5-(4-amino-3-iodobenzyl)adenosine-5'-N- methyluronamide (I-AB-MECA; 2000 Ci/mmol), [3H]CCPA (2-chloro-N6- cyclopentyladenosine, 42.6 Ci/mmol), [3H]CGS21680 (2-[p-(2- carboxyethy^phenylethylaminol-S'-N-ethylcarboxaminocarbonyl-adenosine, 47 Ci/mmol), and [3H]cyclic AMP (40 Ci/mmol) are from Amersham Pharmacia Biotech (Buckinghamshire, UK).
[0071] For the statistical analysis, binding and functional parameters are calculated using Prism 4.0 software (GraphPAD, San Diego, CA, USA). IC50 values obtained from competition curves are converted to K; values using the Cheng-Prusoff equation (Cheng et al., Biochem. Pharmacol. 1973, 22, 3099-3108). Data are expressed as mean + standard error. EXAMPLE 1
[0072] This example demonstrates a general tosylation procedure for the synthesis of 3- iodoethylindole derivatives, 44-46, 56-58, 71-73 and 84 used in the preparation of compounds in accordance with an embodiment of the invention. See Figure 2. [0073] To a solution of the alcohol in THF (tetrahydrofuran) is added sodium hydride (60 %, 3 eq) at 0 0C, and the reaction mixture is stirred at 0 0C for 1 h. Tosyl chloride (3 eq) is added to the suspension at 0 °C, and the reaction mixture is stirred at room temperature overnight. The reaction mixture is diluted with ethyl acetate and washed with water, dried over MgSO4, and filtered. The filtrate is evaporated to give a crude oil, which is subjected to column chromatography on silica gel. Elution with a mixture of toluene and acetone (40: 1) gives the desired tosylated derivative.
EXAMPLE 2
[0074] This example demonstrates a general iodination procedure for the synthesis of compounds 47-49, 59-61, 74-77, and 85 used in the preparation of compounds in accordance with an embodiment of the invention. See Figure 2.
[0075] A solution of the tosylate and sodium iodide (3.5 eq) in N, N-dimethylformamide is stirred overnight at 60 0C. The reaction mixture is diluted with ethyl acetate and washed with water, dried over MgSO4, and filtered. The filtrate is evaporated to give a crude oil, which is subjected to column chromatography on silica gel. Elution with a mixture of hexanes and ethyl acetate (4:1) gives the iodide.
3-(p-Toluenesulfonyloxyethyl)-l-(p-toluenesulfonyl)indole (44)
[0076] The yield is 62%: 1H ΝMR (CDCl3) δ 7.93 (IH, d with small coupling, J = 8.0
Hz), 7.74 (2H, d, J= 8.2 Hz), 7.56 (2H, d, J= 8.5 Hz), 7.12 - 7.34 (7H, m), 4.24 (2H, t, J= 6.6 Hz), 3.01 (2H, t, J= 6.6 Hz), 2.38 (3H, s), 2.32 (3H, s); HRMS (ESI-MS m/z) calcd for C24H23NO5S2Na (M+Na)+ 492.0915, found 492.0914.
5-Methoxy-3-(p-toluenesulfonyloxyethyl)-l-(p-toluenesulfonyl)indole (45)
[0077] The yield is 56 %. 1H NMR (CDCl3) δ 7.82 (IH, d, J= 9.1 Hz), 7.71 (2H, d with small coupling, J= 8.5 Hz), 7.53 (2H, d with small coupling, J= 8.2 Hz), 7.27 (IH, s), 7.21 (2H, dd, J= 0.6 and 8.8 Hz), 7.15 (2H, dd, J= 0.6 and 8.5 Hz), 6.89 (IH, dd, J= 2.5 and 9.1 Hz), 6.71 (IH, d, J = 2.2 Hz), 4.23 (2H, t, J= 6.6 Hz), 3.77 (3H, s), 2.97 (2H, dt, J= 0.8 and 6.6 Hz), 2.39 (3H, s), 2.32 (3H, s); HRMS (ESI-MS m/z) calcd for C25H26NO6S2 500.1202 (M+H)+, found 500.1207.
l-(p-Toluenesulfonyl)-3-(p-toluenesulfonyloxyethyl)-5-(p-toluenesulfonyloxy) indole (46) [0078] The yield is 57%: 1H NMR (CDCl3) δ 7.80 (IH, d, J= 9.1 Hz), 7.71 (2H, d with small coupling, J= 8.5 Hz), 7.82 (2H, d with small coupling, J= 8.5 Hz), 7.58 (2H, d, with small coupling, J= 8.2 Hz), 7.36 (IH, s), 7.31 (2H, dd, J= 0.6 and 8.5 Hz), 7.25 (2H, d, J= 8.4 Hz), 7.19 (2H, d, J= 8.2 Hz), 6.97 (IH, d, J= 2.2 Hz), 6.84 (IH, dd, J= 2.3 and 8.9 Hz), 4.18 (2H, t, J= 6.5 Hz), 2.90 (2H, t, J= 6.5 Hz), 2.46 (3H, s), 2.40 (3H, s), 2.35 (3H, s); HRMS (ESI-MS m/z) calcd for C3iH30NO8S3 (M+H)+ 640.1134 found 640.1099.
3-Iodoethyl-l-(p-toluenesufonyl)mdole (47)
[0079] The yield is 70%: 1H NMR (CDCl3) δ 7.98 (IH, d with small coupling, J = 8.2
Hz), 7.76 (2H, dt, J= 1.9 and 8.5 Hz), 7.45 (2H, m), 7.32 (IH, ddd, J= 1.3, 7.1, and 8.4 Hz), 7.23 - 7.28 (2H, m), 7.20 (2H, d with small coupling, J= 8.0 Hz), 3.41 (2H, t with small coupling, J= 7.1 Hz), 3.24 (2H, t with small coupling, J= 7.3 Hz), 2.33 (3H, s); HRMS (ESI- MS m/z) calcd for Ci7Hi7NO2SI (M+H)+ 426.0025, found 426.0016.
3-Iodoethyl-5-methoxy-l-(p-toluenesulfonyl)indole (48)
[0080] The yield is 74 %. 1H NMR (CDCl3) δ 7.74 (IH, d with small coupling, J= 9.1
Hz), 7.73 (2H, d with small coupling, J= 8.2 Hz), 7.40 (IH, s), 7.20 (2H, dd, J= 0.7 and 8.7 Hz), 6.92 (IH, dd, J= 2.5 and 9.2 Hz), 6.85 (IH, d, J= 2.5 Hz), 3.82 (3H, s), 3.40 (2H, dt, J = 0.7 and 7.6 Hz), 3.20 (2H, t, J= 7.3 Hz), 2.33 (3H, s); HRMS (ESI-MS m/z) calcd for Ci8Hi9NO3SI (M+H)+ 456.0130, found 456.0135.
3-Iodoethyl-l-(p-toluenesulfonyl)-5-(p-toluenesulfonyloxy) indole (49)
[0081] The yield is 83%. 1H NMR (CDCl3) δ 7.85 (IH, d, J= 9.1 Hz), 7.73 (2H, d with small coupling, J= 8.5 Hz), 7.68 (2H, d with small coupling, J= 8.2 Hz), 7.47 (IH, s), 7.30 (2H, d, J= 8.3 Hz), 7.23 (2H, d, J= 8.3 Hz), 7.02 (IH, d, J= 2.5 Hz), 6.91 (IH, dd, J= 2.3 and 8.9 Hz), 3.26 (2H, t with small coupling, J =7.4 Hz), 3.12 (2H, t with small coupling, J= 7.1 Hz), 2.46 (3H, s), 2.36 (3H, s); APCI-MS (m/z) 596.0 (M+H)+. EXAMPLE 3
[0082] This example demonstrates a general procedure for the synthesis of 3- hydroxyethylindole derivatives 53-55 used in the preparation of compounds in accordance with an embodiment of the invention. See Figure 2.
[0083] To a solution of a 2- and/or 5-substituted-indole-3-acetic acid in methanol is added />-toluenesulfonic acid monohydrate (3 eq), and the reaction mixture is stirred at 600C overnight. After neutralization with IN aqueous NaOH, the solvent is evaporated leaving an oily residue, which is dissolved in ethyl acetate. The solution is washed with water, dried over MgSO4, and filtered. The filtrate is evaporated leaving an oily residue, which is subjected to column flush chromatography on silica gel. Elution with a mixture of toluene and acetone (5:1) gives the corresponding ester.
[0084] To a solution of the ester in THF is added lithium aluminum hydride (2.8 eq) at 0 0C, and the reaction mixture is stirred at 0 0C for 1 h and at room temperature for 1 h. After addition of ethyl acetate, the reaction mixture is stirred at room temperature for 30 min. The reaction mixture is diluted with ethyl acetate and washed with water, dried over MgSO4, and filtered. The filtrate is evaporated leaving an oily residue, which is subjected to column chromatography on silica gel. Elution with a mixture of toluene and acetone (3:1) gives the pure alcohol.
5-Fluoro-tryptopho] (53)
[0085] Compound 53 is identical to the known compound reported by Mewshaw et al. (J.
Med. Chem. 2004, 47, 3823-3842).
5-Bromo-tryptophol (54)
[0086] Compound 54 is identical to the commercially available compound.
3-Hydroxyethyl-5-methoxy-2-methylindole (55)
[0087] The yield is 81%. 1H NMR (CDCl3) δ 7.27 (IH, br s), 7.16 (IH, d, J= 8.8 Hz),
6.97 (IH, d, J= 2.2 Hz), 6.78 (IH, dd, J= 2.5 and 8.5 Hz), 3.78 - 3.88 (2H, m overlapped with OCH3), 3.85 (3H, s), 2.94 (2H, t, J= 6.5 Hz), 2.39 (3H, s); HRMS (ESI-MS m/z) calcd for Ci2Hi6NO2206.1181 , found 206.1190. S-Fluoro-l-Cp-toluenesulfonylJ-S-Cp-toluenesulfonyloxyethy^indoIe CSδ)
[0088] The yield is 58 %. 1H NMR (CDCl3) δ 7.87 (IH, dd, J= 4.1 and 9.1 Hz), 7.72
(2H, d with small couplings, J= 8.5 Hz), 7.55 (2H, d with small coupling, J= 8.2 Hz), 7.36 (IH, S), 7.24 (2H, d with small coupling, J= 8.0 Hz), 7.16 (2H, dd, J= 0.7 and 8.7 Hz), 7.00 (IH, dt, J= 2.4 and 9.0 Hz), 6.89 (IH, dd, J= 2.2 and 8.5 Hz), 4.22 (3H, t, J= 6.5 Hz), 2.95 (3H, t, J= 6.5 Hz), 2.39 (3H, s), 2.34 (3H, s); HRMS (ESI-MS m/z) calcd for C24H23NO5S2F (M+H)+ 488.1002, found 488.0995.
5-Bromo-3-(p-toluenesulfonyIoxyethyI)-l-(p-toIuenesulfonyl)indoIe (57)
[0089] The yield is 63%. 1H NMR (CDCl3) δ 7.80 (IH, d with small coupling, J= 9.6
Hz), 7.73 (2H, d with small coupling, J= 8.2 Hz), 7.52 (2H, d with small coupling, J= 8.5 Hz), 7.32-7.39 (3H, m), 7.25 (2H, d, J= 8.0 Hz), 7.13 (2H, d, J= 8.0 Hz), 4.23 (2H, t, J= 6.3 Hz), 2.95 (2H, dt, J= 0.8 and 6.5 Hz), 2.39 (3H, s), 2.34 (3H, s); HRMS (ESI-MS m/z) calcd for C24H22NO5S2BrLi (M+Li)+ 554.0283, found 554.0292.
5-Methoxy-2-methyl-3-(p-toluenesulfonyIoxyethyl)-l-(p-toluenesulfonyI)indole (58) [0090] The yield was 30%. 1H NMR (CDCl3) δ 8.02 (IH, d, J = 9.1 Hz), 7.58 (2H, d with small coupling, J= 8.2Hz), 7.48 (2H, d with small coupling, J= 8.2 Hz), 7.18 (2H, dd, J = 0.7 and 8.7 Hz), 7.13 (2H, dd, J= 0.6 and 8.5 Hz), 6.84 (IH, dd, J= 2.8 and 9.1 Hz), 6.64 (IH, d, J= 2.5 Hz), 4.11 (2H, t, J= 6.7 Hz), 3.79 (3H, s), 2.90 (2H, t, J= 6.7 Hz), 2.43 (3H, s), 2.39 (3H, s), 2.33 (3H, s); HRMS (ESI-MS m/z) calcd for C26H27NO6S2Na (M+Na)+ 536.1178, found 536.1186.
3-Iodoethyl-5-fluoro-l-(p-toluenesulfonyl)indole (59)
[0091] Yield 70 %. 1H NMR (CDCl3) δ 7.92 (IH, ddd, J= 0.6, 4.3 and 9.1 Hz), 7.74
(2H, dt, J= 1.9 and 8.5 Hz), 7.48 (IH, s), 7.22 (2H, d, J= 8.0 Hz), 7.09 (IH, dd, J= 2.5 and 8.5 Hz), 7.04 (IH, dt, J= 2.5 and 9.1 Hz), 3.39 (2H, dt, J= 0.7 and 6.8 Hz), 3.19 (2H, t, J= 7.3 Hz), 2.35 (3H, s); HRMS (ESI-MS m/z) calcd for Ci7Hi5NO2FS 316.0808, found 316.0810.
5-Bromo-3-Iodoethyl-l-(p-toluenesufonyl)indole (60)
[0092] The yield is 65 %. 1H NMR (CDCl3) δ 7.85 (IH, d, J= 8.8 Hz), 7.74 (2H, d with small coupling, J= 8.2 Hz), 7.57 (IH, d, J= 1.9 Hz), 7.45 (IH, s), 7.41 (IH, dd, J= 1.9 and 8.8 Hz), 7.22 (2H, d, J= 8.2 Hz), 3.88 (2H, t, J= 7.0 Hz), 3.19 (2H, t, J= 7.3 Hz), 2.35 (3H, s); HRMS (ESI-MS m/z) calcd for CnHi5NO2SBrI (M+H)+ 502.9025, found 502.9036.
3-Iodoethyl-5-methoxy-2-methyl-l-(p-toluenesufonyl)indole (61)
[0093] The yield is 85%. 1H NMR (CDCl3) δ 8.07 (IH, d, J= 9.1 Hz), 7.58 (2H, dt, J =
1.8 and 8.5 Hz), 7.17 (2H, d, J= 8.2 Hz), 6.87 (IH, dd, J= 2.5 and 9.1 Hz), 6.79 (IH, d, J = 2.5 Hz), 3.84 (3H, s), 3.26 (2H, m), 3.14 (2H, m), 2.52 (3H, s), 2.33 (3H, s); HRMS (ESI-MS m/z) calcd for Ci9H2INO3IS (M+H)+ 470.0287, found 470.0294.
EXAMPLE 4
[0094] This example demonstrates a general synthetic procedure for 3- hydroxyethylindole derivatives (67-70) via Fischer indole ring preparation used in the preparation of compounds in accordance with an embodiment of the invention. See Figure 2. [0095] A solution of substituted phenylhydrazine hydrochloride and ethoxytetrahydrofuran (1.5 eq) in 95 % ethanol is refluxed overnight. The reaction mixture is filtered through celite. The filtrate is evaporated to give a crude solid. The solid is dissolved in ethyl acetate, and the solution is washed with water, dried over MgSO4, and filtered. The filtrate is evaporated to give a crude oil, which is subjected to column chromatography on silica gel. Elution with a mixture of toluene and acetone (2:1) gives the alcohol.
6-Chloro-tryptophol (67), 6-Bromo-tryptophol (68), and 5-ChIoro-tryptophol (69) [0096] These compounds are identical to the known compounds. 67: WO 2001/049679;
68: Fuchs et al. (J Am. Chem. Soc. 2004, 126, 5068-5069); and 69 Chou {Heterocycles 2003, 60, 1095-1110).
5-Iodo-tryptophol (70)
[0097] The yield is 32 %. 1H NMR (CDCl3) δ 8.07 (IH, br s), 7.95 (IH, m), 7.45 (IH, dd, J= 1.7 and 8.5 Hz), 7.16 (IH, d, J= 8.5 Hz), 7.06 (IH, d, J = 2.2 Hz), 3.89 (2H, br s), 2.98 (2H, dt, J= 0.8 and 6.3 Hz), 1.45 (IH, br s); APCI-MS (m/z) 288.0 (M+H)+.
6-ChIoro-3-(p-toluenesulfonyloxyethyl)-l-(p-toluenesulfonyI)indole (71)
[0098] The yield is 32%. 1H NMR (CDCl3) δ 7.94 (IH, d, J= 1.7 Hz), 7.74 (2H, d with small coupling, J= 8.5 Hz), 7.51 (2H, d with small coupling, J= 8.2 Hz), 7.29 (2H, d, J= 8.5 Hz), 7.25 (IH, s), 7.19 (IH, d, J= 8.2 Hz), 7.10-7.16 (3H, m), 4.27 (2H, t, J= 6.3 Hz), 2.97 (2H, t, J= 6.3 Hz), 2.40 (3H, s), 2.35 (3H, s); APCI-MS (m/z) 504.1 (M+H)+.
6-Bromo-3-(p-toluenesulfonyloxyethyl)-l-(p-toluenesulfonyl)indoIe (72)
[0099] The yield is 30 %. 1H NMR (CDCl3) δ 8.10 (IH, d, J= 1.4Hz), 7.74 (2H, d with small coupling, J= 8.2 Hz), 7.51 (2H, d with small coupling, J= 8.2 Hz), 7.23-7.30 (5H, m), 7.13 (2H, dd, J= 1.7 and 8.2 Hz), 4.23 (2H, t, J= 6.3 Hz), 2.97 (2H, t, J= 6.3 Hz), 2.40 (3H, s), 2.35 (3H, s); APCI-MS (m/z) found 548.0 (M+H)+.
5-Chloro-3-(p-toIuenesulfonyloxyethyl)-l-(p-toluenesulfonyl)indole (73)
[00100] The yield is 30 %. 1H NMR (CDCl3) δ 7.85 (IH, dd, J= 0.6 and 8.8 Hz), 7.73
(2H, dt, J= 2.1 and 8.7 Hz), 7.53 (2H, dt, J= 1.8 and 8.5 Hz), 7.36 (IH, s), 7.21 - 7.26 (3H, m), 7.19 (IH, dd, J= 0.6 and 1.9 Hz), 7.14 (2H, dd, J= 0.5 and 8.5 Hz), 4.23 (2H, t, J= 6.5 Hz), 2.95 (2H, dt, J= 0.8 and 6.5 Hz), 2.39 (3H, s), 2.34 (3H, s); HRMS (ESI-MS m/z) calcd for C24H22NO5NaS2Cl 526.0526, found 526.0535.
6-Chloro-3-iodoethyl-l-(p-toluenesulfonyl)indole (74)
[0100] The yield is 67 %. 1H NMR (CDCl3) d 8.00 (IH, d, J = 1.7 Hz), 7.76 (2H, d with small coupling, J= 8.5 Hz), 7.43 (IH, s), 7.36 (IH, d, J= 8.5 Hz), 7.25 ~ 7.28 (2H overlapped with CHCl3), 7.22 (IH, dd, J= 1.8 and 8.4 Hz), 3.39 (2H, t with small coupling, J = 7.3 Hz), 3.21 (2H, t, Jwith small coupling, J= 7.3 Hz), APCI-MS (m/z) 460.0 (M+H)+.
6-Bromo-3-iodoethyl-l-(p-toluenesulfonyl)indole (75)
[0101] The yield is 67 %. 1H NMR (CDCl3) δ 8.16 (IH, d, J= 1.7 Hz), 7.76 (2H, dd, J =
1.9 and 8.5 Hz), 7.42 (IH, br s), 7.36 (IH, dd, J= 1.7 and 8.5 Hz), 7.30 (IH, d, J= 8.2 Hz), 7.25 (2H, d, J= 8.5 Hz), 3.38 (2H, t with small coupling, J= 7.4 Hz), 3.21 (2H, t with small coupling, J= 7.3 Hz), 2.36 (3H, s); HRMS (ESI-MS m/z) calcd for CnHi5BrINO2S (M)+ 502.9052, found 502.9066.
5-Chloro-3-iodoethyl-l-(p-toluenesulfonyl)indole (76)
[0102] The yield is 60 %. 1H NMR (CDCl3) δ 7.90 (IH, dd, J= 0.6 and 8.8 Hz), 7.74
(2H, d with small coupling, J= 8.2 Hz), 7.47 (IH, s), 7.41 (IH, dd, J= 0.6 and 1.9 Hz), 7.24- 7.29 (IH overlaped with CHCl3), 7.22 (2H, d, J= 8.3 Hz), 3.39 (2H, dt, J= 0.8 and 7.3 Hz), 3.20 (2H, t, J= 7.3 Hz), 2.35 (3H, s); HRMS (ESI-MS m/z) calcd for CI7HI6NO2ICIS (M+H)+ 459.9635, found 459.9625.
5-Iodo-3-iodoethyl-l-(p-toluenesuIfonyl)indole (77)
[0103] The yield is 68 %. 1H NMR (CDCl3) δ 7.77 (IH, d, J= 1.6 Hz), 7.74 (IH, d, J =
8.8 Hz), 7.73 (2H, d with small coupling, J= 8.5 Hz), 7.58 (IH, dd, J= 1.7 and 8.5 Hz), 7.41 (IH, s), 7.22 (2H, d, J= 8.2 Hz), 3.83 (2H, t, J= 7.2 Hz), 3.19 (2H, t, J= 7.3 Hz), 2.35 (3H, s); APCI-MS (m/z) 551.9 (M+H)+.
EXAMPLE 5
[0104] This example demonstrates a synthesis of ethyl 4-bromo-3-indolylglyoxylate (80) used in the preparation of compounds in accordance with an embodiment of the invention. See Figure 2.
[0105] To a solution of 78 (2.94 g, 15.0 mmol) in diethyl ether (60 mL) is added oxalyl chloride (3.01 ml, 34.5 mmol) at 0 0C, and the reaction mixture is stirred at room temperature for 1O h. After evaporation, ethanol (30 mL) is added to the solids, and the solution is stirred at room temperarure overnight. The solvent is evaporated to give a solid. This residue is dissolved in ethyl acetate, washed with water, dried over MgSO4, and filtered. The filtrate is evaporated to give a crude solid, which is subjected to column chromatography on silica gel. Elution with a mixture of hexanes and ethyl acetate (1:1) gives 80 (2.3g, 52 %). 1H NMR (CDCl3) δ 9.55 (IH, br s), 8.24 (IH, d, J= 3.3 Hz), 7.50 (IH, dd, J= 0.8 and 7.7 Hz), 7.42 (IH, dd, J= 0.8 and 8.3 Hz), 7.13 (IH, t, J= 8.0 Hz), 4.41 (2H, q, J= 7.1 Hz), 1.40 (3H, t, J = 7.1 Hz); APCI-MS (m/z) 296.0 (M+H)+.
EXAMPLE 6
[0106] This example demonstrates a synthesis of ethyl 7-bromo-3-indolylglyoxylate (81) in an embodiment of the invention. See Figure 2.
[0107] Compound 81 is obtained from 79 by the similar procedure for the preparation of 80 (yield 70 %). 1H NMR (CDCl3) δ 8.96 (IH, br s), 8.55 (IH, d, J= 3.3 Hz), 8.39 (IH, dd, J = 0.6 and 8.0 Hz), 7.48 (IH, dd, J= 0.8 and 8.0 Hz), 7.23 (IH, t, J= 7.8 Hz), 4.43 (2H, q, J= 7.1 Hz), 1.44 (3H, t, J= 7.1 Hz), APCI-MS (m/z) 296.0 (M+H)+. EXAMPLE 7
[0108] This example demonstrates a synthesis of 4-bromo-tryptophol (82) used in the preparation of compounds in accordance with an embodiment of the invention. See Figure 2. [0109] To a solution of 80 (20 mg, 0.0675 mmol) in THF (1.4 mL) is added lithium aluminum hydride (17.4 mg, 0.459 mmol), and the reaction mixture is refluxed for 2 h. The mixture is diluted with ethyl acetate and washed with water, dried over MgSO4, and filtered. The filtrate is evaporated to give a crude oil, which is subjected to preparative TLC developed with a mixture of hexanes and ethyl acetate (1 : 1) to give 82 (10 mg, 63 % yield). 1H NMR (CDCl3) δ 8.12 (IH, br s), 7.32 (IH, dd, J= 0.8 and 7.7 Hz), 7.28 (IH, dd, J= 0.8 and 7.7 Hz), 7.14 (IH, d, J= 2.5 Hz), 7.02 (IH, t, J= 7.8 Hz), 3.97 (2H, q, J= 6.1 Hz), 3.29 (2H, dt, J= 0.6 and 6.5 Hz), 1.46 (IH, t, J= 5.6 Hz); HRMS (ESI-MS m/z) calcd for Ci0Hi1BrNO (M+H)+ 240.0024, found 240.0028.
EXAMPLE 8
[0110] This example demonstrates a synthesis of 83-86 used in the preparation of compounds in accordance with an embodiment of the invention. See Figure 2. [0111] Compounds 83-86 are obtained from 81 by the similar procedure for the preparation of 82.
7-Bromo-tryptophol (83)
[0112] The yield is 52 %. IH NMR (CDCl3) δ 8.82 (IH, br s), 7.57 (IH, d, J = 8.0 Hz),
7.36 (IH, d, J = 8.0 Hz), 7.16 (IH, d, J = 2.2 Hz), 7.02 (IH, t, J = 7.8 Hz), 3.91 (2H, q, J = 6.2 Hz), 3.02 (2H, dt, J = 0.5 and 6.3 Hz), 1.46 (IH, t, J = 6.0 Hz); HRMS (ESI-MS m/z) calcd for Ci0HnNOBr (M+H)+ 240.0024, found 240.0031.
4-Bromo-3-(p-toluenesulfonyloxyethyl)-l-(p-toluenesulfonyl)indole (84)
[0113] The yield is 41 %. IH NMR (CDCl3) δ 7.91 (IH, dd, J = 0.8 and 8.2 Hz), 7.75
(2H, d with small coupling, J = 8.5 Hz), 7.55 (2H, d with small coupling, J = 8.2 Hz), 7.41 (IH, s), 7.28 (IH, dd, J = 1.0 and 7.8 Hz), 7.25 (2H, d, J = 8.2 Hz), 7.11 (2H, d, J = 8.0 Hz), 7.09 (IH, t, J = 8.0 Hz), 4.32 (2H, t, J = 6.5 Hz), 3.25 (2H, t, J = 6.3 Hz), 2.34 (6H, s); APCI- MS (m/z) 548.0 (M+H)+.
4-Bromo-3-iodoethyI-l-(p-toluenesulfonyl)-indole (85) [0114] The yield is 67 %. 1H NMR (CDCl3) δ 7.96 (IH, dd, J= 0.8 and 8.2 Hz), 7.76 (2H, d with small coupling, J= 8.5 Hz), 7.52 (IH, s), 7.38 (IH, dd, J= 0.8 and 8.0 Hz), 7.23 (2H, dd, J= 0.6 and 8.5 Hz), 7.13 (IH, t, J= 8.1 Hz), 3.45 (4H, s), 2.35 (2H, s); HRMS (ESI- MS m/z) calcd for Ci7Hi6NO2IBrS (M+H)+ 509.9130, found 509.9114.
7-Bromo-3-iodoethylindole (86)
[0115] The yield is 88 %. IH NMR (CDCl3) δ 8.21 (IH, br s), 7.52 (IH, dd, J = 0.8 and
8.0 Hz), 7.36 (IH, d, J = 7.7 Hz), 7.16 (IH, d, J = 2.2 Hz), 7.02 (IH, t, J = 7.7 Hz), 3.39-3.46 (2H, m), 3.29-3.37 (2H, m); HRMS (ESI-MS m/z) calcd for Ci0Hi0NBrI (M+H)+ 349.9041, found 349.9036.
EXAMPLE 9
[0116] This example demonstrates a synthesis of 2-hydroxyethyl-l-(p-toluenesulfonyl)- indole (88) used in the preparation of compounds in accordance with an embodiment of the invention. See Figure 3.
[0117] To a solution of N-tosyl-2-iodoanilide (1.172g. 3.14 mmol) in DMF are added 3- butyn-1-ol (1.42 ml, 18.8 mmol), copper iodide (119 mg, 0.628 mmol), triethyl amine (13.56 mL, 97.3 mmol) and Pd(PPh3)2Cl2 (220.3 mg, 0.314 mmol). The reaction mixture is stirred at 70 0C overnight. The reaction mixture is diluted with ethyl acetate. The solution is washed with water, dried over MgSO4, and filtered. The filtrate is evaporated to give an oil, which is subjected to column chromatography on silica gel. Elution with a mixture of hexanes and ethyl acetate (1:1) gives 88 (820 mg, 83%). 1H ΝMR (CDCl3) δ 8.16 (IH, d, J = 8.2 Hz), 7.61 (2H, d with small coupling, J= 8.2 Hz), 7.42 (IH, dd, J= 1.7 and 7.4 Hz), 7.28 (IH, dt, J= 2.2 and 7.6 Hz), 7.23 (IH overlapped with Ph), 7.18 (2H, d, J= 8.5 Hz), 6.50 (IH, s), 4.01 (2H, t, J= 6.1 Hz), 3.29 (2H, t, J= 6.2 Hz), 2.33 (3H, s); APCI-MS (m/z) 316.0 (M+H)+.
EXAMPLE 10
[0118] This example demonstrates a synthesis of 2-iodoethyl-l-(p-toluenesulfonyl)- indole (89) used in the preparation of compounds in an embodiment of the invention. See Figure 3.
[0119] To a solution of 88 (638 mg, 2.02 mmol) in a mixture of diethylether (24 mL) and acetnitrile (8 mL) are added triphenylphosphine (1.589 g, 6.06 mmol), imidazole (439 mg, 6.46 mmol), and iodine (1.639 g, 6.46 mmol). The reaction mixture is stirred at 0 0C for 1 h. The reaction mixture is diluted with ethyl acetate. The solution is washed with water, dried over MgSO4, and filtered. The filtrate is evaporated to give an oil, which is subjected to column chromatography on silica gel. Elution with a mixture of hexanes and ethyl acetate (4: 1) gives 89 (847 mg, 98 %). 1H NMR (CDCl3) δ 8.14 (IH, d with small coupling, J= 8.2 Hz), 7.60 (2H, dt, J= 2.0 and 8.6 Hz), 7.45 (IH, dd, J= 1.0 and 6.7 Hz), 7.30 (IH, dt, J= 1.5 and 8.0 Hz), 7.23 (IH, dd, J= 1.2 and 7.6 Hz), 7.18 (2H, d with small coupling, J= 8.0 Hz), 6.50 (IH, s), 3.46 - 3.60 (4H, m), 2.33 (3H, s); HRMS (ESI-MS m/z) calcd for Ci7Hi7NO2IS (M+H)+ 426,0025 found 426.0035.
EXAMPLE I l
[0120] This example demonstrates a synthesis of benzoimidazol-1-yl-ethanol (92) used in the preparation of compounds in an embodiment of the invention. See Figure 3. [0121] To a solution of benzoimidazol-1-yl-acetic acid (893 mg, 5.06 mmol) in THF (20 mL) is added lithium aluminum hydride (672 mg, 17.7 mmol) at 0 0C, and the reaction mixture is stirred at room temperature for 5 h. After dilution with ethyl acetate the solution is washed with water, dried over MgSO4, and filtered. The filtrate is evaporated to give a crude oil, which is subjected to column chromatography on silica gel. Elution with a mixture of chloroform and methanol (8: 1) gives 92 (620 mg, 76 %). 1H NMR (CDCl3) δ 7.63 (IH, s), 7.38 (IH, dt, J= 1.0 and 8.2 Hz), 7.31 (IH, dt, J= 1.0 and 8.0 Hz), 7.17 (IH, ddd, J= 1.0, 7.1, and 8.1 Hz), 7.06 (IH, ddd, J= 1.1, 7.1, and 8.1 Hz), 4.22 (2H, t, J = 4.9 Hz), 3.99 (2H, t, J= 4.8 Hz); HRMS (ESI-MS m/z) calcd for C9HnN2O (M+H)+ 163.0871, found 163.0880.
EXAMPLE 12
[0122] This example demonstrates a synthesis of benzotriazol-1-yl-ethanol (93) used in the preparation of compounds in an embodiment of the invention. See Figure 3. [0123] Procedure used for the preparation of 93 from 91 is similar to those used for the preparation of 92 from 90; amorphous solid, the yield is 59 %. 1H NMR (CDCl3) δ (IH, d with small coupling, J= 8.5 Hz), 7.61 (IH, d with small coupling, J= 8.2 Hz), 7.50 (IH, ddd, J= 1.1, 7.0 and 8.1 Hz), 7.36 (IH, ddd, J= 1.2, 6.9 and 8.1 Hz), 4.75 (2H, t, J= 5.1 Hz), 4.25 (2H, dd, J= 5.9 and 10.3 Hz), 2.55 (IH, t, J= 6.0 Hz); HRMS (ESI-MS m/z) calcd for C8Hi0N3O 164.0824 found 164.0812. EXAMPLE 13
[0124] This example demonstrates a synthesis of benzoimidazol-1-yl-ethyliodide (94) used in the preparation of compounds in an embodiment of the invention. See Figure 3. [0125] To a solution of 92 (22 mg, 0.134 mmol) in a mixture of acetonitrile (0.3 mL) and diethylether (0.9 mL) are added triphenylphosphine (105 mg, 0.402 mmol), imidazole (29 mg, 0.428 mmol), and iodine (108 mg, 0.428 mmol) at 0 0C. The reaction mixture is stirred for 2 h. The mixture is diluted with ethyl acetate, washed with water, dried over MgSO4, and filtered. The filtrate is evaporated to give a crude oil, which is subjected to preparative TLC developed with a mixture of toluene and acetone (2: 1) to give 94 (32.3 mg, 88 %). 1H NMR (CDCl3) δ 7.97 (IH, s), 7.80-7.87 (IH, m), 7.28-7.42 (3H, m), 4.58 (2H, t, J= 7.0 Hz), 3.51 (2H, t, J= 7.1 Hz); APCI-MS (m/z) 273.0 (M+H)+.
EXAMPLE 14
[0126] This example demonstrates a synthesis of benzotriazol-1-yl-ethyliodide (95) used in the preparation of compounds in an embodiment of the invention. See Figure 3. [0127] The procedure used for the preparation of 95 from 93 is similar to that used for the preparation of 94 from 92; amorphous solid, the yield is 88 %. 1H NMR (CDCl3) δ 8.09 (IH, dt, J= 1.0 and 8.3 Hz), 7.50 - 7.60 (2H, m), 7.40 (IH, ddd, J= 1.8, 6.3, and 8.1 Hz), 5.02 (2H, t, J= 7.3 Hz), 3.67 (2H, t, J= 7.3 Hz); HRMS (ESI- MS m/z) calcd for C8H9N3I (M+H)+ 273.9841, found 273.9833.
EXAMPLE 15
[0128] This example demonstrates a synthesis of 3-hydroxyethyl- 1 -(p- toluenesulfonyl)pyrrole (98) used in the preparation of compounds in an embodiment of the invention. See Figure 3.
[0129] To a solution of 97 (1.24 g, 4.21 mmol) in THF (20 mL) is added lithium aluminum hydride (340 mg, 6.32 mmol) at 0 0C. The reaction mixture is stirred for 2 h. After addition of ethyl acetate, the mixture is stirred for 30 min. The mixture is diluted with ethyl acetate, washed with water, dried over MgSO4, and filtered. The filtrate is evaporated to give a crude oil, which is subjected to column chromatography on silica gel. Elution with a mixture of chloroform and methanol (20:1) gives 98 (692 mg, 62 %). 1H NMR δ 7.74 (2H, d, J= 8.4 Hz), 7.28 (2H, d, J= 8.7 Hz), 7.10 (IH, t, J= 2.7 Hz), 6.99 (IH, m), 6.19 (IH, dd, J = 1.5 and 3.3 Hz), 3.74 (2H, t, J= 6.5 Hz), 2.64 (2H, t, J= 6.5 Hz), 2.40 (3H, s); HRMS (ESI-MS m/z) calcd for Ci3Hi6NO3S (M+H)+ 266.0851, found 266.0837.
EXAMPLE 16
[0130] This example demonstrates a synthesis of 3-iodoethyl-l-(p- toluenesulfonyl)pyrrole (99) used in the preparation of compounds in an embodiment of the invention. See Figure 3.
[0131] The procedure used for preparation of 99 from 98 is similar to that used for the preparation of 89 from 88; the yield was 62 %. 1H NMR (CDCl3) δ 7.73 (2H, d, J= 8.2 Hz), 7.29 (2H, d, J= 8.5 Hz), 7.08 (IH, t, J= 2.8 Hz), 6.99 (IH, m), 6.16 (IH, dd, J= 1.6 and 3.3 Hz), 3.24 (2H, t, J= 7.3 Hz), 2.96 (2H, t, J=7.6 Hz), 2.40 (3H, s); HRMS (ESI-MS m/z) calcd for Ci3Hi5NO2SI (M+H)+ 375.9868, found 375.9857.
EXAMPLE 17
[0132] This example demonstrates a general synthetic procedure for 2-substituted adenosine derivatives in an embodiment of the invention. See Figure 4. [0133] To a solution of 2', 3', 5'-triacetyl-6-chloroguanosine in N,N-dimethylformamide are added iodide (1.8 eq) and Cs2CO3 (2.7 eq) at room temperature, and the reaction mixture is stirred overnight. After dilution with ethyl acetate, the solution is washed with water twice, dried over MgSO4, and filtered. The filtrate is evaporated to give a crude oil, which is purified by column chromatography or preparative TLC on silica gel. Elution or developing with a mixture of toluene and acetone (4:1) gives the 2-substituted 2', 3', 5'-triacetyl-6- chloroadenosine derivative.
[0134] A solution of 2-substituted 2', 3', 5'-triacetyl-6-chloroadenosine derivative in saturated ammonia ethanol solution is stirred in sealed tube overnight at 110 - 120 0C. The solvent is evaporated to give an oil, which is subjected to preparative TLC developed with a mixture of chloroform and methanol (8:1) to give the 2-substituted adenosine derivative. [0135] In case of deprotection of the tosyl group as a 2-substituent, tosylated adenosine derivative is treated with KOH (20 eq) in methanol overnight at 70 0C in sealed tube. The reaction mixture is concentrated to a small amount of solution, which is subjected to preparative TLC developed with a mixture of chloroform and methanol (5:1) to give the final product. 6-Chloro-2-(3"-(l "-(p-toluenesuIfonyl)indolyl)ethyloxy)-3', 4', 5'-triacetyladenosine
(102)
[0136] The yield is 86 %. 1H NMR (CDCl3) δ 8.09 (IH, s), 7.98 (IH, d with small coupling, J= 6.6 Hz), 7.76 (2H, d with small coupling, J= 8.2 Hz), 7.61 (IH, d with small coupling, J= 7.1 Hz), 7.53 (IH, s), 7.14 - 7.36 (6H, m), 6.13 (IH, d, J= 4.7 Hz), 5.93 (IH, t, J= 5.1 Hz), 5.65 (IH, t, J = 5.5 Hz), 4.71 (2H, m), 4.39 - 4.49 (2H, m), 4.32 (IH, dd, J= 4.7 and 12.6 Hz), 3.24 (2H, t, J= 7.0 Hz), 2.32 (3H, s), 2.14 (3H, s), 2.09 (3H, s), 2.05 (3H, s); HRMS (ESI-MS m/z) calcd for C33H32N5Oi0ClSNa (M+Na)+ 748.1456, found 748.1455.
6-Chloro-2-(3"-(5"-methoxy-l"-(p-toluenesulfonyl)indolyl)ethyloxy)- 3', 4', 5'- triacetyladenosine (103)
[0137] The yield is 72 %. 1H NMR (CDCl3) δ 8.09 (IH, s), 7.86 (IH, dd, J= 9.1 Hz),
7.73 (2H, d, J= 8.5 Hz), 7.48 (IH, s), 7.20 (2H, d, J= 8.2 Hz), 7.03 (IH, d, J= 2.5 Hz), 6.92 (IH, dd, J= 2.3 and 8.9 Hz), 6.12 (IH, d, J= 4.7 Hz), 5.93 (IH, t, J= 5.1 Hz), 5.66 (IH, t, J = 5.6 Hz), 4.69 (2H, ddd, J= 3.8, 7.4, and 14.3 Hz), 4.40 - 4.49 (2H, m), 4.31 (IH, dd, J = 4.4 and 12.6 Hz), 3.85 (3H, s), 3.19 (2H, t, J= 6.7 Hz), 2.32 (3H, s), 2.14 (3H, s), 2.09 (3H, s), 2.05 (3H, s); HRMS (ESI-MS m/z) calcd for C34H35N5OnSCl (M+H)+ 756.1742, found 756.1735.
6-Chloro-2-(3"-(5"-(p-toluenesulfonyIoxy) -l"-(p-toluenesulfonyl)indolyl)ethyloxy)- 3',
4', 5'-triacetyladenosme (104)
[0138] The yield is 51 %. 1H NMR (CDCl3) δ 8.09 (IH, s), 7.83 (IH, d, J= 8.5 Hz), 7.72
(2H, d with small coupling, J= 8.5 Hz), 7.68 (2H, d with small coupling, J= 8.2 Hz), 7.57 (IH, s), 7.25-7.31 (3H, m), 7.22 (2H, d, J = 8.0 Hz), 6.83 (IH, dd, J= 2.3 and 8.9 Hz), 6.12 (IH, d, J= 4.4 Hz), 5.94 (IH, dd, J= 4.7 and 5.5 Hz), 5.67 (IH, t, J= 5.4 Hz), 4.64 (2H, m), 4.40-4.50 (2H, m), 4.31 (IH, dd, J= 5.1 and 12.8 Hz), 3.13 (2H, t, J= 6.7 Hz), 2.42 (3H, s), 2.34 (3H, s), 2.15 (3H, s), 2.08 (3H, s), 2.03 (3H, s); HRMS (ESI-MS m/z) calcd for C40H39ClN5Oi3S2 (M+H)+ 896.1674 found 896.1638.
6-Chloro-2-(3"-(5"-fluoro-l"-(p-toluenesulfonyl)indolyl)ethyloxy)-3', 4', 5'- triacetyladenosine (105)
[0139] The yield is 59 %. 1H NMR (CDCl3) δ 8.09 (IH, s), 7.91 (IH, dd, J= 4.4 and 9.1
Hz), 7.74 (2H, d with small coupling, J = 8.5 Hz), 7.57 (IH, s), 7.25-7.31 (IH overlapped with CHCl3), 7.22 (2H, d, J= 8.0 Hz), 7.04 (IH, dt, J = 2.6 and 9.0 Hz), 6.11 (IH, d, J= 4.7 Hz), 5.94 (IH, t, J= 4.9 Hz), 5.67 (IH, t, J= 5.5 Hz), 4.69 (2H, m), 4.40-4.50 (2H, m), 4.31 (IH, dd, J = 4.9 and 12.9 Hz), 3.18 (2H, t, J= 6.9 Hz), 2.33 (3H, s), 2.15 (3H, s), 2.09 (3H, s), 2.04 (3H, s); HRMS (ESI-MS m/z) calcd for C32H23NSO ioSFCl (M+H)+ 744.1542, found 744.1522.
2-(3"-(5"-Bromo-l"-(p-toluenesulfonyl)indolyl)ethyloxy)-6-chloro-3', 4', 5'- triacetyladenosine (106)
[0140] The yield is 47 %. 1H NMR (CDCl3) δ 8.09 (IH, s), 7.84 (IH, d, J= 8.8 Hz), 7.75
(IH, s), 7.73 (2H, d with small coupling, J= 6.6 Hz), 7.54 (IH, s), 7.40 (IH, dd, J= 1.8 and 8.9 Hz), 7.22 (2H, d, J= 8.5 Hz), 6.13 (IH, d, J = 4.7 Hz), 5.94 (IH, t, J = 4.9 Hz), 5.67 (IH, t, J = 5.4 Hz), 4.69 (2H, t, J= 6.6 Hz), 4.40-4.50 (2H, m), 4.31 (IH, dd, J= 5.2 Hz), 3.19 (2H, t, J= 6.7 Hz), 2.33 (3H,s), 2.15 (3H, s), 2.09 (3H, s), 2.03 (3H, s); HRMS (ESI-MS m/z) calcd for C33H32N5Oi0BSClBr (M+H)+ 804.0742, found 804.0752.
6-ChIoro-2-(3"-(5"-methoxy-2"-methyl-l"-0>-toluenesulfonyl)indolyl)ethyloxy)- 3',4',5'- triacetyladenosine (107)
[0141] The yield is 72 %. 1H NMR (CDCl3) δ 8.08 (IH, s), 8.07 (IH, d, J= 9.6 Hz), 7.60
(2H, d with small coupling, J= 8.5 Hz), 7.17 (2H, d, J= 8.0 Hz), 6.98 (IH, d, J= 2.5 Hz), 6.87 (IH, dd, J= 2.8 and 9.1 Hz), 6.12 (IH, d, J= 5.0 Hz), 5.83 (IH, t, J= 5.2 Hz), 4.36 - 4.54 (5H, m), 4.31 (IH, dd, J= 4.1 and 12.4 Hz), 3.86 (3H, s), 3.13 (2H, t, J= 7.8 Hz), 2.61 (3H, s), 2.32 (3H, s), 2.13 (3H,s), 2.06 (3H,s), 2.05 (3H, s); HRMS (ESI-MS m/z) calcd for C35H37N5OnSCl (M+H)+ 770.1899, found 770.1895.
6-Chloro-2-(3"-(6"-chIoro-l"-(p-toIuenesuIfonyI)indoIyI)ethyIoxy)-3', 4', 5'- triacetyladenosine (108)
[0142] The yield is 70 %. 1H NMR (CDCl3) d 8.09 (IH, s), 7.99 (IH, d, J= 1.9 Hz), 7.76
(2H, d with small coupling, J= 8.5 Hz), 7.53 (IH, d, J= 8.5 Hz), 7.52 (IH, s), 7.20 - 7.28 (3H, m), 6.11 (IH, d, J = 4.7 Hz), 5.95 (IH, t, J= 4.9 Hz), 5.66 (IH, t, J = 5.5 Hz), 4.68 (2H, m), 4.38-4.50 (2H, m), 4.31 (IH, dd, J= 4.5 and 12.2 Hz), 3.20 (2H, t, J= 6.9 Hz), 2.35 (3H, s), 2.14 (3H, s), 2.09 (3H, s), 2.06 (3H, s); APCI-MS (m/z) 760.1 (M+H)+.
2-(3"-(6"-Bromo-l "-(p-toluenesulfonyl)indolyl)ethyloxy)-6-chloro-3f, 4', 5'- triacetyladenosine (109)
[0143] The yield is 45 %. 1HNMR (CDCl3) δ 8.15 (IH, d, J= 1.6 Hz), 8.09 (IH, s), 7.76
(2H, d with small coupling, J= 8.2 Hz), 7.50 (IH, s), 7.49 (IH, d, J= 9.1 Hz), 7.38 (IH, dd, J = 1.7 and 8.5 Hz), 7.25 (2H, d, J= 8.2 Hz), 6.11 (IH, d, J= 4.7 Hz), 5.95 (IH, t, J= 4.9 Hz), 5.66 (IH, t, J= 5.5 Hz), 4.68 (2H, m), 4.18-4.50 (2H, m), 4.31 (IH, dd, J= 4.5 and 12.5 Hz), 3.20 (2H, t, J= 7.0 Hz), 2.35 (3H, s), 2.14 (3H, s), 2.09 (3H, s), 2.06 (3H, s); HRMS (ESI- MS m/z) calcd for C33H32N5Oi0SCl Br(M+H)+ 804.0742, found 804.0760.
6-Chloro-2-(3"-(5"-cMoro-l"-(p-toluenesulfonyl)indolyl)ethyloxy)- 3', 4', 5'- triacetyladenosine (110)
[0144] The yield is 46 %. 1H NMR (CDCl3) δ 8.09 (IH, s), 7.89 (IH, d, J= 9.1 Hz), 7.73
(2H, d with small coupling, J= 8.2 Hz), 7.59 (IH, d, J= 2.2 Hz), 7.24-7.30 (IH, m), 7.22 (2H, d, J= 8.0 Hz), 6.12 (IH, d, J= 4.4 Hz), 5.94 (IH, t, J= 5.1 Hz), 5.67 (IH, t, J= 5.4 Hz), 4.69 (2H, m), 4.40-4.49 (2H, m), 4.31 (IH, dd, J= 4.9 and 13.2 Hz), 3.18 (2H, t, J= 6.7 Hz), 2.33 (3H, s), 2.15 (3H, s), 2.09 (3H, s), 2.04 (3H, s); HRMS (ESI-MS m/z) calcd for C33H3IN5Oi0SCl2Na (M+Na)+ 782.1066, found 782.1071.
6-Chloro-2-(3"-(5"-iodo-l"-(p-tolueiiesuIfoiiyI)indoIyl)ethyloxy)- 3', 4', 5'- triacetyladenosine (111)
[0145] The yield is 45 %. 1H NMR (CDCl3) δ 8.09 (IH, s), 7.93 (IH, d, J= 1.7 Hz), 7.73
(3H, m), 7.57 (IH, dd, J= 1.8 and 8.7 Hz), 7.50 (IH, s), 7.22 (2H, d, J = 8.0 Hz), 6.13 (IH, d, J= 4.7 Hz), 5.94 (IH, t, J= 5.1 Hz), 5.67 (IH, t, J= 5.4 Hz), 4.68 (2H, t, J= 7.0 Hz), 4.40- 4.48 (2H, m), 4.31 (IH, dd, J= 5.1 and 13.3 Hz), 3.18 (2H, t, J= 6.9 Hz), 2.33 (3H, s), 2.15 (3H, s), 2.09 (3H, s), 2.04 (3H, s); HRMS (ESI-MS m/z) calcd for C33H32N5Oi0SClI (M+H)+ 852.0603, found 852.0566.
6-ChIoro-2-(3"-(4"-broino-l"-(p-toIuenesulfonyl)indolyl)ethyloxy)- 3', 4', 5'- triacetyladenosine (112)
[0146] The yield is 40 %. 1H NMR (CDCl3) δ 8.09 (IH, s), 7.95 (IH, dd, J= 0.8 and 8.2
Hz), 7.75 (2H, d with small coupling, J= 8.5 Hz), 7.59 (IH, s), 7.39 (IH, dd, J= 0.8 and 8.0 Hz), 7.23 (2H, d, J= 8.5 Hz), 7.12 (IH, t, J= 8.1 Hz), 6.13 (IH, d, J= 5.0 Hz), 5.92 (IH, t, J = 5.1 Hz), 5.64 (IH, t, J= 5.4 Hz), 4.75 (2H, m), 4.38-4.50 (2H, m), 4.33 (IH, dd, J= 4.7 and 12.9 Hz), 3.53 (2H, t, J= 7.0 Hz), 2.34 (3H, s), 2.13 (3H, s), 2.09 (3H, s), 2.05 (3H, s); APCI- MS (m/z) 806.1 (M+H)+. 6-Chloro-2-(3"-(7"-bromoindolyl)ethyloxy)- 3', 4', 5'-triacetyIadenosine (113)
[0147] The yield is 10 %. 1H NMR (CDCl3) δ 8.06 (IH, s), 7.66 (IH, d, J= 8.0 Hz), 7.35 (IH, d, J= 7.7 Hz), 7.27 (IH overlapped with CHC13), 7.03 (IH, t, J= 7.7 Hz), 6.11 (IH, d, J = 5.0 Hz), 5.91 (IH, t, J= 5.2 Hz), 5.64 (IH, t, J= 5.2 Hz), 4.70 (2H, m), 4.37-4.46 (2H, m), 4.32 (IH, dd, J= 5.4 and 13.1 Hz), 3.30 (2H, t,J= 7.1 Hz), 2.13 (3H, s), 2.07 (3H, s), 2.07 (3H, s); APCI-MS (m/z) 672.1 (M+Na)+.
6-Chloro-2-phenypropoxy-3', 4', 5'-triacetyladenosine (114)
[0148] The yield is 63 %. 1H NMR (CDCl3) δ 8.08 (IH, s), 7.16-7.34 (5H, m), 6.14 (IH, d, J= 4.9 Hz), 5.91 (IH, t, J= 5.4 Hz), 5.63 (IH, t, J= 5.2 Hz), 4.38 - 4.52 (5H, m), 4.31 (IH, dd, J= 3.8 and 11.8 Hz), 2.85 (2H, dd, J= 7.4 and 8.0 Hz), 2.12 - 2.24 (2H, m), 2.15 (3H, s), 2.10 (3H, s), 2.09 (3H, s); HRMS (ESI-MS m/z) calcd for C25H27N4O8ClLi (M+Li)+ 553.1677, found 553.1661.
6-Chloro-2-(2"-(l"-(p-toluenesulfonyl)indolyl)ethyloxy)- 3', 4', 5'-triacetyladenosine
(115)
[0149] The yield is 40 %. 1H NMR (CDCl3) δ 8.16 (IH, d, J= 8.2 Hz), 8.09 (IH, s), 7.63
(2H, d, J= 8.5 Hz), 7.42 (2H, d, J= 7.1 Hz), 7.15 - 7.32 (5H, m), 6.59 (IH, s), 6.16 (IH, d, J = 5.0 Hz), 5.87 (IH, t, J= 5.2 Hz), 5.61 (IH, t, J= 5.2 Hz), 4.83 (2H, m), 4.40 - 4.48 (2H, m), 4.34 (IH, dd, J= 4.9 and 13.2 Hz), 3.58 (2H, t, J= 6.6 Hz), 2.33 (3H, s), 2.12 (3H, s), 2.08 (3H,s), 2.07 (3H,s); HRMS (ESI-MS m/z) calcd for C33H33N5Oi0ClS (M+H)+ 726.1637, found 726.1640.
6-Chloro-2-(3"-(benzoimidazoI-l"-yl)ethyloxy)- 3', 4', 5'-triacetyladenosine (116)
[0150] The yield is 51 %. 1H NMR (CD3OD) δ 8.09 (2H, d, J= 8.0 Hz), 7.79 (IH, dd, J =
1.4 and 7.1 Hz), 7.55 (IH, dd, J= 1.1 and 7.1 Hz), 7.34 (IH, dt, J= 1.4 and 7.4 Hz), 7.28 (IH, dt, J = 1.4 and 7.5 Hz), 6.05 (IH, d, J= 4.7 Hz), 5.92 (IH, t, J= 4.9 Hz), 5.67 (IH, t, J = 5.4 Hz), 4.82 (2H, m), 4.66 (2H, t, J= 5.4 Hz), 4.39 - 4.48 (2H, m), 4.27 (IH, dd, J= 5.2 and 13.2 Hz), 2.16 (3H, s), 2.09 (3H, s), 2.02 (3H, s); HRMS (ESI-MS m/z) calcd for C25H26N6O8Cl (M+H)+ 573.1501, found 573.1503.
6-Chloro-2-(3"-(benzotriazol-l"-yl)ethoxy)- 3', 4', 5'-triacetyladenosine (117) [0151] The yield is 53 %. 1H NMR (CDCl3) δ 8.08 (IH, s), 8.03 (IH, dt, J= 0.8 and 8.5 Hz), 7.72 (IH, dt, J= 0.8 and 8.5 Hz), 7.52 (IH, ddd, J= 1.0, 7.1 and 8.1 Hz), 7.36 (IH, ddd, J= 1.1, 7.1 and 8.2 Hz), 6.07 (IH, d, J= 4.4 Hz), 5.89 (IH, dd, J= 4.7 and 5.5 Hz), 5.62 (IH, t, J= 5.4 Hz), 5.11 (2H, m), 5.00 (2H, m), 4.40-4.48 (2H, m), 4.26-4.34 (IH, m), 2.15, 2.10 and 2.05 (each 3H, s); HRMS (ESI-MS m/z) calcd for C24H25N7O8Cl (M+H)+ 574.1453, found 574.1456.
6-Chloro-2-(3"-(l"-/»-toluenesulfonyl)pyrrolyl)ethyloxy)- 3', 4', 5'-triacetyladenosine
(118)
[0152] The yield is 61%. 1H NMR (CDCl3) δ 8.08 (IH, s), 7.73 (IH, d, J= 8.5 Hz), 7.27
(2H, d overlapped with CHCl3), 7.08 (2H, m), 6.28 (IH, dd, J= 1.9 and 3.0 Hz), 6.13 (IH, d, J= 5.0 Hz), 5.91 (IH, t, J= 5.1 Hz), 5.63 (IH, t, J= 5.4 Hz), 4.57 (IH, dd, J= 2.8 and 7.4 Hz), 4.53 (IH, dd, J= 3.0 and 7.7 Hz), 4.38 - 4.45 (2H, m), 4.32 (IH, dd, J= 4.3 and 12.2 Hz), 2.95 (2H, t, J= 7.0 Hz), 2.40(3H, s), 2.15(3H, s), 2.09 (3H, s), 2.07 (3H, s); HRMS (ESI-MS m/z) calcd for C29H31N5O10SCI (M+H)+ 676.1480, found 676.1450.
2-(3"-(5"-Methoxy-l"-(p-toluenesulfonyl)indolyI)ethyloxy)adenosine (119)
[0153] The yield is 56 %. 1H NMR (CD3OD) δ 8.14 (IH, s), 7.81 (IH, d, J= 9.3 Hz),
7.67 (2H, dt, J =1.9 and 8.5 Hz), 7.52 (IH, s), 7.15 (2H, dd, J= 0.6 and 8.5 Hz), 7.02 (IH, d, J=2.5 Hz), 6.89 (IH, dd, J= 2.8 and 8.8 Hz), 5.89 (IH, d, J= 6.0 Hz), 4.72 (IH, t, J= 5.6 Hz), 4.55 (2H, dt, J= 1.1 and 6.3 Hz), 4.33 (IH, dd, J= 3.3 and 5.0 Hz), 4.12 (IH, q, J= 3.0 Hz), 3.88 (IH, dd, J= 2.7 and 12.4 Hz), 3.78 (3H, s), 3.74 (IH, dd, J=3.2 and 12.5 Hz), 3.11 (2H, t, J= 6.3 Hz), 2.25 (3H, s); HRMS (ESI-MS m/z) calcd for C28H3 iN6O8S (M+H)+ 611.1924, found 611.1899.
2-(3"-(5"-(p-toluenesulfonyloxy) -l"-(p-toluenesulfonyl)indolyl)ethyloxy)adenosine (120) [0154] The yield is 63 %. 1H NMR (CD3OD) δ 8.15 (IH, s), 7.86 (IH, d, J= 8.8 Hz),
7.71 (2H, d with small coupling, J= 8.5 Hz), 7.64 (IH, s), 7.60 (2H, d with small coupling, J = 8.2 Hz), 7.28 (2H, d, J= 8.5 Hz), 7.21 (2H, d, J= 8.0 Hz), 7.08 (IH, d, J= 2.5 Hz), 6.93 (IH, dd, J= 2.2 and 9.1 Hz), 5.90 (IH, d, J= 6.1 Hz), 4.71 (IH, t, J= 5.6 Hz), 4.42 (2H, t, J = 6.6 Hz), 4.33 (2H, t, J= 6.6 Hz), 4.33 (IH, dd, J= 3.2 and 5.1 Hz), 4.14 (IH, q, J= 3.0 Hz), 3.90 (IH, dd, J= 2.8 and 12.6 Hz), 3.74 (IH, dd, J= 3.2 and 12.5 Hz), 3.00 (2H, t, J = 6.6 Hz), 2.34 (3H, s), 2.29 (3H, s); HRMS (ESI-MS m/z) calcd for C34H35N6Oi0S2 (M+H)+ 751.1856, found 751.1819.
2-(3"-(5"-FIuoro-l"-(p-toluenesulfonyl)indolyl)ethyloxy)adenosine (121)
[0155] The yield is 55 %. 1H NMR (CD3OD) δ 8.13 (IH, s), 7.91 (IH, dd, J= 4.4 and
9.1 Hz), 7.70 (2H, d with small coupling, J= 8.5 Hz), 7.64 (IH, s), 7.29 (IH, dd, J= 2.5 and 8.8 Hz), 7.16 (2H, d, J= 8.0 Hz), 7.05 (IH, dt, J= 2.5 and 9.1 Hz), 5.89 (IH, d, J= 6.0 Hz), 4.73 (IH, t, J= 5.5 Hz), 4.54 (2H, t, J= 6.2 Hz), 4.33 (IH, dd, J= 3.3 and 5.2 Hz), 4.12 (IH, q, J= 3.0 Hz), 3.89 (IH, dd, J= 2.7 and 12.4 Hz), 3.74 (IH, dd, J = 3.2 and 12.5 Hz), 3.10 (2H, t, J= 6.3 Hz), 2.26 (3 H, s); HRMS (ESI-MS m/z) calcd for C27H28N6O7SF (M+H)+ 599.1724, found 599.1714.
2-(3"-(6"-Chloro-l"-(/7-toluenesulfonyl)indolyl)ethyloxy)adenosine (122)
[0156] The yield is 51 %. 1H NMR (CD3OD) δ 8.13 (1 H, s), 7.92 (1 H, d, J = 1.7 Hz),
7.72 (2H, d with small coupling, J= 8.2 Hz), 7.61 (IH, s), 7.58 (IH, d, J= 8.5 Hz), 7.25 (IH, dd, J= 1.9 and 8.5 Hz), 7.21 (2H, dd, J= 0.7 and 8.7 Hz), 5.89 (IH, d, J= 6.1 Hz), 4.72 (IH, t, J= 5.5 Hz), 4.55 (2H, m), 4.33 (IH, dd, J= 3.3 and 5.2 Hz), 4.12 (IH, q, J= 2.9 Hz), 3.88 (IH, dd, J= 2.8 and 12.4 Hz), 3.74 (IH, dd, J= 3.3 and 12.4 Hz), 3.12 (2H, t, J= 6.5 Hz), 2.28 (3H, s); HRMS (ESI-MS m/z) calcd for C27H28N6O7SCl (M+H)+ 615.1429, found 615.1413.
2-(3"-(6"-Bromo-l"-(p-toluenesu]fonyl)indolyl)ethyloxy)adenosine (123)
[0157] The yield is 50 %. 1H NMR (CD3OD) δ 8.13 (IH, s), 8.08 (IH, d, J= 1.7 Hz),
7.71 (2H, dt, J= 2.1 and 8.5 Hz), 7.60 (IH, s), 7.53 (IH, d, J= 8.2 Hz), 7.38 (IH, dd, J= 1.7 and 8.2 Hz), 7.21 (2H, d with small coupling, J= 8.2 Hz), 5.89 (IH, d, J= 6.1 Hz), 4.72 (IH, t, J= 5.5 Hz), 4.54 (2H, m), 4.33 (IH, dd, J= 3.3 and 5.2 Hz), 4.12 (IH, q, J= 3.0 Hz), 3.88 (IH, dd, J= 2.7 and 12.4 Hz), 3.74 (IH, dd, J= 3.3 and 12.4 Hz), 3.12 (2H, t, J= 6.5 Hz), 2.28 (3H, s); HRMS (ESI-MS m/z) calcd for C27H28N6O7BrS (M+H)+ 659.0924, found 659.0910.
2-(3"-(5"-Chloro-l"-(/?-toIuenesulfonyl)indolyl)ethyloxy)adenosine (124) [0158] The yield is 62 %. 1H NMR (CD3OD) 8 8.13 (IH, s), 7.90 (IH, d, J= 8.8), 7.71 (2H, d with small coupling, J= 8.8 Hz), 7.64 (IH, s), 7.57 (IH, d, J= 1.9 Hz), 7.27 (IH, dd, J = 1.9 and 8.8 Hz), 7.18 (2H, d, J= 8.2 Hz), 5.89 (IH, d, J= 5.8 Hz), 4.73 (IH, t, J= 5.6 Hz), 4.55 (2H, t, J= 6.5 Hz), 4.33 (IH, dd, J= 3.3 and 5.2 Hz), 4.12 (IH, q, J= 3.1 Hz), 3.89 (IH, dd, J= 2.7 and 12.6 Hz), 3.74 (IH, dd, J= 3.2 and 12.5 Hz), 3.11 (2H, t, J= 6.3 Hz), HRMS (ESI-MS m/z) calcd for C27H28N6O7SCl (M+H)+ 615.1429, found 615.1401.
2-(3"-(5"-Iodo-l"-(p-toluenesulfonyl)indolyl)ethyloxy)adenosine (125)
[0159] The yield is 71 %. 1H NMR (CD3OD) δ 8.14 (IH, s), 7.89 (IH, d, J= 1.7 Hz),
7.73 (IH, d, J= 9.1 Hz), 7.71 (2H, d with small coupling, J= 8.5 Hz), 7.58 (IH, s), 7.57 (IH, dd, J= 1.8 and 8.7 Hz), 7.18 (2H, d, J= 8.2 Hz), 5.89 (IH, d, J= 5.8 Hz), 4.73 (IH, t, J= 5.5 Hz), 4.54 (2H, t, J= 6.3 Hz), 4.34 (IH, dd, J= 3.3 and 5.2 Hz), 4.12 (IH, q, J= 3.0 Hz), 3.89 (IH, dd, J= 2.9 and 12.5 Hz), 3.75 (IH, dd, J= 3.3 and 12.4 Hz), 3.10 (2H, t, J= 6.3 Hz), 3.27 (3H, s); APCI-MS (m/z) 707.0 (M+H)+.
2-(3"-(4"-Bromo-l"-(p-toluenesulfonyl)indolyl)ethyloxy)adeiiosine (126)
[0160] The yield is 43 %. 1H NMR (CD3OD) 8 8.13 (IH, s), 7.95 (IH, dd, J= 0.7 and
8.4 Hz), 7.73 (2H, d with small coupling, J = 8.5 Hz), 7.69 (IH, s), 7.39 (IH, dd, J= 0.8 and 7.7 Hz), 7.19 (2H, dd, J= 0.6 and 8.5 Hz), 7.15 (IH, t, J= 8.1 Hz), 5.89 (IH, d, J= 6.1 Hz),
4.74 (IH, t, J= 5.5 Hz), 4.61 (2H, m), 4.33 (IH, dd, J= 3.3 and 5.2 Hz), 4.12 (IH, q, J= 3.1 Hz), 3.89 (IH, dd, J= 2.8 and 12.4 Hz), 3.75 (IH, dd, J= 3.3 and 12.4 Hz), 3.44 (2H, m), 2.27 (3H, s); APCI-MS (m/z) 659.1 (M+H)+.
2-(3"-(l"-(^-toluenesulfonyl)pyrrolyl)ethyloxy)-adenosine (127)
[0161] The yield is 69 %. IH NMR (CD3OD) D 8.12 (IH, s), 7.72 (2H, d with small coupling, J = 8.5 H), 7.29 (2H, d, J = 8.5Hz), 7.08-7.14 (2H, m), 6.30 (IH, dd, J = 1.7 and 3.2 Hz), 5.88 (IH, d, J = 6.0 Hz), 4.71 (IH, t, J = 5.5 Hz), 4.41 (2H, t, J = 6.7 Hz), 4.32 (IH, dd, J = 3.4 and 5.1 Hz), 4.11 (IH, q, J = 3.2 Hz), 3.86 (IH, dd, J = 2.6 and 12.5 Hz), 3.73 (IH, dd, J = 3.3 and 12.7 Hz), 2.85 (2H, t, J = 6.5 Hz), 2.36 (3H, s); HRMS (ESI-MS m/z) calcd for C23H27N6O7S (M+H)+ 531.1662, found 531.1667. EXAMPLE 18
[0162] This example demonstrates a synthesis of (2R, 3S, 4S, 5R)-2-(2'-amino-6'- chloropurin-9'-yl)-5-hydroxymethyl-3, 4-<9-isopropylidene-tetrahydrofuran (128) used in the preparation of compounds in an embodiment of the invention. See Figure 6. [0163] To a solution of 2-amino-6-chloropurine-9-riboside (100 mg, 0.331 mmol) in N5N- dimethylformamide (2 mL) are added 2,2-dimethoxypropane (0.242 ml, 1.97 mmol) andp- toluenesulfonic acid monohydrate (188 mg, 0.993 mmol). The reaction mixture is stirred overnight at room temperature. The reaction is diluted with ethyl acetate, washed with water, dried over MgSO4, and filtered. The filtrate is evaporated to give an oil, which is subjected to preparative TLC developed with a mixture of toluene and acetone (1:1) to give 128 (56 mg, 50 %). 1H ΝMR (CDCl3) δ 7.81 (IH, s), 5.79 (IH, d, J = 4.9 Hz), 5.68 (IH, dd, J= 1.4 and 11.3 Hz), 5.14 - 5.24 (3H, m), 5.08 (IH, dd, J= 1.4 and 6.0 Hz), 4.51 (IH, d, J= 1.7 Hz), 3.97 (IH, d with small coupling, J= 12.6 Hz), 3.78 (IH, ddd, J= 1.9, 11.3 and 13.2 Hz), 1.64 (3H, s), 1.38 (3H, s); HRMS (ESI-MS m/z) calcd for C13H]7N5O4Cl (M+H)+ 342.0969, found 342.0979.
EXAMPLE 19
[0164] This example demonstrates a synthesis of (2R, 3S, 4S, 5R)-2-(2'-amino-6'- chloropurin-9'-yl)-5-carboxy-3, 4-<9-isopropylidene-tetrahydrofuran (129) used in the preparation of compounds in an embodiment of the invention. See Figure 6. [0165] To a solution of 128 (16.9 mg, 0.0494 mmol) in water (4.5 mL) are added potassium permanganate (70.3 mg, 0.445 mmol) and potassium hydroxide (25 mg, 0.444 mmol), and the reaction mixture is stirred for 1 h. After addition of isopropanol the reaction mixture is filtered. The filtrate is neutralized with 0.1N hydrochloric acid aqueous solution and evaporated to give a crude solid, which is subjected to preparative TLC developed with a mixture of chloroform, methanol, and saturated aqueous ammonia (2: 1 : 0.3) to give 129 (9 mg, 51 %). 1H NMR (CD3OD) δ 8.29 (IH, s), 6.18 (IH, d, J= 1.2 Hz), 5.52 (IH, dd, J= 1.7 and 6.2 Hz), 5.37 (IH, d, J= 6.0 Hz), 4.59 (IH, d, J= 1.7 Hz), 1.55 (3H, s), 1.39 (3H, S); HRMS (ESI-MS m/z) calcd for Ci3H13N5O5Cl (M-H)- 354.0605, found 354.0622. EXAMPLE 20
[0166] This example demonstrates a synthesis of (2R, 3 S, 4S, 5R)-2-(2'-amino-6'- chloropurin-9'-yl)-5-ethoxycarboxyamide-3, 4-isopropylidene-tetrahydrofuran (130) used in the preparation of compounds in an embodiment of the invention. See Figure 6. [0167] To a solution of 129 (11.9 mg, 0.0334 mmol) in N, N-dimethylformamide (0.8 mL) are added ethylamine hydrochloride (8.1 mg, 0.100 mmol), N, N-diisopropylethylamine (0.035 ml, 0.200 mmol), and (benzotriazol-l-yloxy)tripyrrolidinophosphonium hexafluorophosphate (22.5 mg, 0.0434 mmol). The reaction mixture is stirred overnight. The mixture is diluted with ethyl acetate, washed with water, dried over MgSO4, and filtered. The filtrate is evaporated to give a crude oil, which is subjected to preparative TLC developed with a mixture of chloroform and methanol (10:1) to give 130 (10 mg, 78 %). 1H ΝMR (CD3OD) δ 8.16(1H, s), 6.27 (IH, s), 5.73 (IH, dd, J= 1.9 and 6.3 Hz), 5.43 (IH, d, J = 6.3 Hz), 4.62 (IH, d, J= 1.7 Hz), 2.91 (IH, dt, J= 6.0 and 13.3 Hz), 2.80 (IH, dt, J= 6.0 and 13.3 Hz), 1.55 (3H, s), 1.40 (3H,s), 0.61 (3H, t, J= 7.3 Hz); HRMS (ESI- MS m/z) calcd for Ci5H20N6O4Cl (M+H)+383.1235, found 383.1229.
EXAMPLE 21
[0168] This example demonstrates a synthesis of (2R, 3S, 4S, 5R)-5- ethoxycarboxyamide-2-(2'-hydroxy-6'-chloropurin-9'-yl)-3, 4-O-isopropylidene- tetrahydrofuran (131) used in the preparation of compounds in an embodiment of the invention. See Figure 6.
[0169] To a solution of 130 (10 mg, 0.026 mmol) in a mixture of 2-propanol (0.4 mL) and water (0.4 mL) is added ^-butylnitrite (13.3 μl, 0.115 mmol) at 4 0C. The reaction mixture is stirred at room temperature overnight. The reaction mixture is diluted with ethyl acetate, washed with water, dried over MgSO4, and filtered. The filtrate is evaporated to give a crude oil, which is subjected to preparative TLC developed with a mixture of chloroform and methanol (10:1) to give 131 (5 mg, 50 %). 1H NMR (CDCl3) δ 7.99 (IH, s), 6.37 (IH, br t, J = 6.1Hz), 6.11 (IH, d, J = 1.6 Hz), 5.72 (IH, dd, J = 1.7 and 6.1 Hz), 5.36 (IH, dd, J = 1.7 and 6.0 Hz), 4.75 (IH, s), 3.05 (2H, m), 1.61 (3H, s), 1.41 (3H, s), 0.77 (3H, t, J = 7.3 Hz); APCI-MS (m/z) 384.1 (M+H)+. EXAMPLE 22
[0170] This example demonstrates a synthesis of (2R, 3 S, 4S, 5R)-5-ethylcarboxyamide- 2-2'-(3"-(l"-(p-toluenesufonyl)indolyl)ethyloxy)-6'-chloropurin-9'-yl)-3, 4-isopropylidene- tetrahydrofuran (132) used in the preparation of compounds in an embodiment of the invention. See Figure 6.
[0171] To a solution of 131 (19.4 mg, 0.0505 mmol) in N, N-dimethylformamide (0.8 mL) is added iodide 47 (43 mg, 0.101 mmol) and cesium carbonate (49.3 mg, 0.151 mmol), and the reaction mixture is stirred at room temperature overnight. The reaction mixture is diluted with ethyl acetate, washed with water, dried over MgSO4, and filtered. The filtrate is evaporated to give a crude oil, which is subjected to preparative TLC developed with a mixture of toluene and acetone (4:1) to give 132 (24 mg, 70 %). 1H ΝMR (CDCl3) δ 7.99 (IH, s), 7.98 (IH, d, J= 8.2 Hz), 7.77 (2H, d with small coupling, J= 8.5 Hz), 7.61 (IH, d with small coupling, J= 7.7 Hz), 7.56 (IH, s), 7.24-7.35 (2H, m), 7.21 (2H, d, J= 8.5 Hz), 6.27 (IH, t, J= 6.0 Hz), 6.14 (IH, d, J= 2.2 Hz), 5.52 (IH, dd, J= 1.9 and 6.1 Hz), 5.39 (IH, dd, J= 2.1 and 6.2 Hz), 4.60-4.80 (3H, m), 3.27 (2H, t, J= 6.7 Hz), 2.97 (2H, m), 2.32 (3H, s), 1.61 (3H, s), 1.35 (3H, s), 0.69 (3H, t, J = 7.3Hz); HRMS (ESI-MS m/z) calcd for C32H33N6O7SCl Na (M+Na)+ 703.1718, found 703.1732.
EXAMPLE 23
[0172] This example demonstrates a synthesis of (2R, 3S, 4S, 5R)-5-ethylcarboxyamide- 2-(6'-amino -2'-(3"-(I "-(p-toluenesufonyl)indolyl)ethyloxy)-purin-9'-yl)-3, 4-0- isopropylidene-tetrahydrofuran (133) used in the preparation of compounds in an embodiment of the invention. See Figure 6.
[0173] A solution of 132 in saturated ammonia ethanol solution was stirred at 120 0C overnight. The solvent is evaporated to give an oil, which is subjected to preparative TLC developed with a mixture of chloroform and methanol (10:1) to give 133 (17 mg, 88 % yield). 1H NMR (CDCl3) δ 7.96 (IH, d, J= 7.7 Hz), 7.77 (2H, d, J= 8.2 Hz), 7.68 (IH, s), 7.54-7.60 (2H, m), 7.30 (IH, dt, J= 1.6 and 7.7 Hz), 7.24 (IH, overlapped with CHCl3), 7.19 (2H, d, J= 8.5 Hz), 6.44 (IH, t, J= 5.9 Hz), 6.07 (IH, d, J= 1.9 Hz), 5.59 (IH, br s), 5.51 (IH, dd, J= 1.9 and 6.0 Hz), 5.43 (IH, dd, J= 1.8 and 6.2 Hz), 4.70 (IH, d, J= 1.9 Hz), 4.61 (2H, m), 3.18 (2H, t, J= 6.7 Hz), 2.96 (2H, m), 2.31 (3H, s), 1.64 (3H, s), 1.31 (3H, s), 0.69 (3H, t, J= 7.3 Hz); HRMS (ESI-MS m/z) calcd for C32H36N7O7S (M+H)+ 662.2397, found 662.2374.
EXAMPLE 24
[0174] This example demonstrates a synthesis of (2R, 3S, 4S, 5R)-5-ethylcarboxyamide- 2-(6'-amino -2'-(3 "-( 1 " -(p-toluenesufonyl)mdolyl)ethyloxy)-purin-9'-yl)-tetrahydrofuran (134) in an embodiment of the invention. See Figure 6.
[0175] A solution of 133 (13.4 mg, 0.0202 mmol) in 80 % acetic acid aqueous solution is stirred at 80 0C for 63 h and evaporated to give an oil, which is subjected to preparative TLC developed with a mixture of chloroform and methanol (8:1) to give 134 (9 mg, recovery yield 85 %). 1H NMR (CD3OD) δ 8.09 (IH, s), 7.94 (IH, d, J= 7.7 Hz), 7.69 (2H, d with small coupling, J= 8.5 Hz), 7.56 (IH, d with small coupling, J= 1.1 Hz), 7.52 (IH, s), 7.30 (IH, dt, J= 1.3 and 7.7 Hz), 7.16-7.26 (3H, m), 5.93 (IH, d, J= 7.4 Hz), 4.54-4.76 (2H, m), 4.42 (IH, d, J= 1.9 Hz), 4.31 (IH, dd, J= 1.8 and 4.8 Hz), 3.00-3.18 (4H, m), 2.28 (3H, s), 0.83 (3H, t, J= 7.1 Hz); HRMS (ESI-MS m/z) calcd for C29H32N7O7S (M+H)+ 622.2084, found 622.2095.
2-Phenylpropoxyadenosine (8)
[0176] The yield is 66 %. 1H NMR (CD3OD) 5 8.11 (IH, s), 7.10 - 7.28 (5H, m), 5.88
(IH, d, J= 6.1 Hz), 4.72 (IH, t, J= 5.5 Hz), 4.30 -4.34 (IH overlaped with CH2), 4.29 (2H, t, J= 6.6 Hz), 4.10 (IH, q, J= 3.2 Hz), 3.85 (IH, dd, J= 2.9 and 12.5 Hz), 3.72 (IH, dd, J = 3.3 and 12.4 Hz), 2.78 (2H, t, J= 7.7 Hz), 2.06 (2H, dt, J= 6.4 and 15.3 Hz); HRMS (ESI- MS m/z) calcd for Ci9H24N5O5(M+H)+ 402.1777, found 402.1771; HPLC (system A) 14.1 min (99%), (system C) 10.7 min (99%).
2-(3"-Indolylethyloxy)adenosine (17)
[0177] The yield is 72 %. 1H NMR (CD3OD) δ 8.12 (IH, s), 7.60 (IH, d with small coupling, J= 7.7 Hz), 7.32 (IH, d with small coupling, J= 7.7 Hz), 7.15 (IH, s), 7.07 (IH, dt, J= 1.2 and 7.5 Hz), 7.01 (IH, dt, J= 1.2 and 7.3 Hz), 5.90 (IH, d, J = 5.5 Hz), 4.71 (IH, t, J= 5.5 Hz), 4.58 (2H, m), 4.31 (IH, dd, J= 3.6 and 5.2 Hz), 4.11 (IH, q, J= 3.2 Hz), 3.85 (IH, dd, J= 2.8 and 12.4 Hz), 3.73 (IH, dd, J= 3.4 and 12.2 Hz), 3.24 (2H, t, J= 7.3 Hz); HRMS (ESI-MS m/z) calcd for C20H23N6O5 (M+H)+ 427.1730, found 427.17111; HPLC (system A) 11.4 min (99 %), (system C) 9.3 min (99 %). 2-(3"-(l"-(p-Toluenesulfonyl)indolyl)ethyloxy)adenosine (18)
[0178] The yield is 60 %. 1H NMR (CD3OD) δ 8.14 (IH, s), 7.93 (IH, d with small coupling, J= 7.4 Hz), 7.70 (2H, d with small coupling, J= 8.2 Hz), 7.59 (2H, m), 7.29 (IH, dt, J= 1.7 and 8.1 Hz), 7.23 (IH, dt, J= 1.5 and 8.1 Hz), 7.17 (2H, d, J= 8.0 Hz), 5.90 (IH, d, J= 6.0 Hz), 4.73 (IH, t, J= 5.6 Hz), 4.57 (2H, m), 4.33 (IH, dd, J= 3.2 and 5.1 Hz), 4.12 (IH, q, J= 3.0 Hz), 3.88 (IH, dd, J= 2.8 and 12.4 Hz), 3.74 (IH, dd, J= 3.3 and 12.4 Hz), 3.14 (2H, t, J= 6.3 Hz); HRMS (ESI-MS m/z) calcd for C27H29N6O7S (M+H)+ 581.1818, found 581.1797; HPLC (system B) 16.5 min (99%), (system C) 16.7 min(99%).
2-(3"-Pyrrolylethyloxy)adenosine (19)
[0179] The yield is 57 %. 1H NMR (CD3OD) δ 8.11(1H, s), 6.63(2H, d, J= 2.2 Hz), 6.04
(IH, t, J= 2.1 Hz), 5.88 (IH, d, J= 5.8 Hz), 4.72 (IH, t, J= 5.5 Hz), 4.42 (2H, t, J= 7.4 Hz), 4.31 (IH, dd, J= 3.4 and 5.1 Hz), 4.10 (IH, q, J=3.2 Hz). 3.85 (IH, dd, J= 3.0 and 12.4 Hz), 3.73 (IH, dd, J = 3.6 and 12.4 Hz), 2.91 (2H, t, J= 7.3 Hz); HRMS (ESI-MS m/z) calcd for C16H2IN6O5 (M+H)+ 377.1573, found 377.1577; HPLC (system A) 4.5 min (99%), (system C) 4.7 min (99%).
2-(2"-Indolylethyloxy)adenosine (20)
[0180] The yield is 32 %. 1H NMR (CD3OD) δ 8.08 (IH, s), 7.37 (IH, d with small couplings, J= 7.4 Hz), 7.25 (IH, d with small coupling, J= 8.0 Hz), 6.97 (IH, dt, J= 1.4 and 7.1 Hz), 6.88 (IH, dt, J= 1.1 and 7.2 Hz), 6.21 (IH, d, J= 0.8 Hz), 5.86 (IH, d, J= 5.8 Hz), 4.69 (IH, t, J= 5.5 Hz), 4.57 (2H, t, J= 6.6 Hz), 4.30 (IH, dd, J= 3.3 and 5.2 Hz), 4.08 (IH, q, J= 3.3 Hz), 3.83 (IH, dd, J= 2.9 and 12.2 Hz), 3.72 (IH, dd, J= 3.3 and 12.4 Hz), 3.18 (2H, t, J= 6.7 Hz); HRMS (ESI-MS m/z) calcd for C20H23N6O5 (M+H)+ 427.1730, found 427.1735; HPLC (system A) 14.6 min (99 %), (system C) 10.9 min (99 %).
2-(2"-(l "-fø-Tøluenesulfonyl)indolyl)ethyloxy)adenosine (21)
[0181] The yield is 55 %. 1H NMR (CDCl3) δ 8.10 (IH, d, J= 8.2 Hz), 7.57-7.64 (3H, m), 7.37 (IH, d, J= 7.7 Hz), 7.11-7.26 (4H, m), 6.52 (IH, s), 5.71 (IH, d, J= 6.9 Hz), 5.66 (2H, br s), 5.03 (IH, t, J= 6.9 Hz), 4.62 (IH, m), 4.62 (IH, m), 4.46 (2H, m), 4.27 (IH, s), 3.89 (IH, d,J= 11.5 Hz), 3.74 (IH, d, J= 11.5 Hz), 3.43 (2H, m), 3.19 (IH, br s), 2.30 (3H, s); HRMS (ESI-MS m/z) calcd for C27H29N6O7S (M+H)+ 581.1818, found 581.1802; HPLC (system A) 23.0 min (99%), (system C) 16.7 min (99%).
2-(3"-(5"-Fluoro-indolyl)ethyloxy)adenosine (22)
[0182] The yield is 80 %. 1H NMR (CD3OD) δ 8.12 (IH, s), 7.20 ~ 7.30 (3H, m), 6.83
(IH, dt, J= 2.5 and 9.1 Hz), 5.89 (IH, d, J= 5.8 Hz), 4.72 (IH, t, J= 5.5 Hz), 4.55 (2H, t, J= 7.0 Hz), 4.31 (IH, dd, J= 3.3 and 5.2 Hz), 4.11 (IH, q, J= 3.3 Hz), 3.86 (IH, dd, J= 2.8 and 12.4 Hz), 3.73 (IH, dd, J= 3.6 and 12.4 Hz), 3.16 (2H, t, J= 7.1 Hz); HRMS (ESI-MS m/z) calcd for C20H2IN6O5FNa (M+Na)+ 467.1455, found 467.1479; HPLC (system A) 13.1 min (99%), (system C) 10.2 min (99 %).
2-(3"-(6"-Chloro-indolyl)ethyloxy)adenosine (23)
[0183] The yield is 54 %. 1H NMR (CD3OD) δ 8.08 (IH, s), 7.52 (IH, d, J= 8.5 Hz),
7.27 (IH, d, J= 1.7 Hz), 7.13 (IH, s), 6.95 (IH, dd, J= 1.9 and 8.5 Hz), 5.86 (IH, d, J= 6.0 Hz), 4.67 (IH, t, J= 5.5 Hz), 4.51 (2H, m), 4.28 (IH, dd, J= 3.4 and 5.1 Hz), 4.07 (IH, q, J=
3.2 Hz), 3.81 (IH, dd, J= 2.8 and 12.4 Hz), 3.69 (IH, dd, J= 3.3 and 12.4 Hz), 3.14 (2H, t, J = 7.0 Hz); HRMS (ESI-MS m/z) calcd for C20H22N6O5Cl (M+H)+ 461.1340, found 461.1339; HPLC (system A) 15.7 min (99 %), (system C) 11.9 min (99 %).
2-(3"-(5"-Bromo-indolyl)ethyloxy)adenosine (24)
[0184] The yield is 70 %. 1H NMR (CD3OD) δ 8.12 (IH, s), 7.70 (IH, d, J= 1.9 Hz),
7.24 (IH, d, J= 8.8 Hz), 7.20 (IH, s), 7.15 (IH, dd, J= 1.8 and 8.7 Hz), 5.89 (IH, d, J= 5.8 Hz), 4.71 (IH, t, J= 5.5 Hz), 4.56 (2H, t, J= 7.0 Hz), 4.32 (IH, dd, J= 3.6 and 5.2 Hz), 4.11 (IH, q, J= 3.2 Hz), 3.86 (IH, dd, J= 2.8 and 12.4 Hz), 3.73 (IH, dd, J= 3.6 and 12.4 Hz), 3.17 (2H, t, J= 6.9 Hz); HRMS (ESI-MS m/z) calcd for C20H22N6O5Br (M+H)+ 505.0835, found 505.0822; HPLC (system A) 15.2 min(98 %), (system C) 12.2 min (98%).
2-(3"-(5"-Iodo-indolyl)ethyloxy)adenosine (25)
[0185] The yield is 56 %. 1H NMR (CD3OD) δ 8.12 (IH, s) 7.89 (IH, dd, J= 0.6 and 1.7
Hz), 7.32 (IH, dd, J= 1.7 and 8.5 Hz), 7.16 (IH, s), 7.15 (IH, dd, J= 0.6 and 8.5 Hz), 5.89 (IH, d, J= 5.8 Hz), 4.71 (IH, t, J= 5.5 Hz), 4.55 (2H, t, J= 7.0 Hz), 4.32 (IH, dd, J= 3.6 and 5.2 Hz), 4.11 (IH, q, J= 3.2 Hz), 3.86 (IH, dd, J= 2.8 and 12.4 Hz), 3.73 (IH, dd, J-
3.3 and 12.4 Hz), 3.16 (2H, t, J= 6.9 Hz); HRMS (ESI-MS m/z) calcd for C20H22N6O5I (M+H)+ 553.0696, found 553.0681; HPLC (system A) 19.0 min (98 %), (system C) 13.1 min (98 %).
2-(3"-(5"-Bromo-l"-(/>-toluenesulfonyl)indolyl)ethyloxy)adenosine (26)
[0186] The yield is 68 %. 1H NMR (CD3OD) δ 8.14 (IH, s), 7.86 (IH, dd, J= 0.6 and
8.8 Hz), 7.68 - 7.74 (3H, m), 7.63 (IH, s), 7.40 (IH, dd, J= 1.8 and 8.8 Hz), 7.18 (2H, d with small coupling, J= 8.5 Hz), 5.89 (IH, d, J= 6.0 Hz), 4.73 (IH, t, J= 5.6 Hz), 4.55 (2H, t, J = 6.3 Hz), 4.33 (IH, dd, J= 3.3 and 5.2 Hz), 4.12 (IH, q, J= 3.0 Hz), 3.89 (IH, dd, J= 2.9 and 12.5 Hz), 3.74 (IH, dd, J= 3.3 and 12.4 Hz), 3.11 (2H, t, J= 6.2 Hz), 2.27 (3H, s); HRMS (ESI MS m/z) calcd for C27H28N6O7BrS (M+H)+ 659.0924, found 659.0921.
2-(3"-(5"-Chloro-indolyl)ethyloxy)adenosine (27)
[0187] The yield is 54 %. 1H NMR (CD3OD) δ 8.12 (IH, s), 7.55 (IH, d, J= 1.9 Hz),
7.28 (IH, dd, J= 0.5 and 8.5 Hz), 7.22 (IH, s), 7.03 (IH, dd, J= 1.9 and 8.5 Hz), 5.89 (IH, d, J= 6.0 Hz), 4.71 (IH, t, J= 5.5 Hz), 4.56 (2H, t, J= 7.0 Hz), 4.31 (IH, dd, J= 3.6 and 5.2 Hz), 4.11 (IH, q, J= 3.2 Hz), 3.86 (IH, dd, J= 2.9 and 12.5 Hz), 3.73 (IH, dd, J= 3.3 and 12.4 Hz), 3.17 (2H, t, J= 7.0 Hz); HRMS (ESI-MS m/z) calcd for C20H22N6O5Cl (M+H)+ 461.1340, found 461.1332; HPLC (system A) 16.6 min (99 %), (system C) 11.8 min (99 %).
2-(3"-(6"-Bromo-indolyl)ethyloxy)adenosine (28)
[0188] The yield is 71 %. 1H NMR (CD3OD) δ 8.12 (IH, s), 7.52 (IH, d, J= 8.5 Hz),
7.48 (IH, d, J= 1.7 Hz), 7.17 (IH, s), 7.12 (IH, dd,J= 1.7 and 8.5 Hz), 5.90 (IH, d, J= 5.8 Hz), 4.71 (IH, t, J= 5.5 Hz), 4.56 (2H, m), 4.31 (IH, dd, J= 3.3 and 5.2 Hz), 4.11 (IH, q, J= 3.2 Hz), 3.85 (IH, dd, J= 3.4 and 12.2 Hz), 3.73 (IH, dd, J =3.4 and 12.2 Hz), 3.21 (2H, t, J = 7.2 Hz); HRMS (ESI-MS m/z) calcd for C20H22N6O5Br (M+H)+ 505.0835 found 505.0840; HPLC (system A) 18.1 min (98 %), (system C) 12.6 min (98 %).
2-(3"-(4"-Bromo-indo]yl)ethyloxy)adenosine (29)
[0189] The yield is 44 %. 1H NMR (CD3OD) δ 8.11 (IH, s), 7.31 (IH, dd, J= 0.8 and
8.0 Hz), 7.24 (IH, s), 7.16 (IH, dd, J= 0.8 and 7.4 Hz), 6.93 (IH, t, J= 7.8 Hz), 5.89 (IH, d, J= 6.0 Hz), 4.71 (IH, t, J= 5.5 Hz), 4.60 (2H, m), 4.31 (IH, dd, J= 3.6 and 5.0 Hz), 4.10 (IH, q, J= 3.2 Hz), 3.85 (IH, dd, J= 2.9 and 12.5 Hz), 3.73 (IH, dd, J= 3.4 and 12.5 Hz), 3.47 (IH, t, J= 7.1 Hz); HRMS (ESI-MS m/z) calcd for C20H22N6O5Br (M+H)+ 505.0835, found 505.0843; HPLC (system A) 15.3 min (99 %), (system C) 11.7 min (99 %).
2-(3"-(7"-Bromo-indolyl)ethyloxy)adenosine (30)
[0190] The yield is 27 %. 1H NMR (CD3OD) δ 8.12 (IH, s), 7.60 (IH, d, J= 8.0 Hz),
7.25 (IH, d, J= IA Hz), 7.24 (IH, s), 6.95 (IH, t, J= 7.8 Hz), 5.90 (IH, d, J= 5.8 Hz), 4.71 (IH, t, J = 5.6 Hz), 4.58 (2H, m), 4.31 (IH, dd, J= 3.6 and 4.9 Hz), 4.11 (IH, q, J= 3.2 Hz), 3.85 (IH, dd, J= 2.9 and 12.2 Hz), 3.73 (IH, dd, J= 3.3 and 12.4 Hz), 3.20 (2H, t, J= 6.9 Hz); HRMS (ESI-MS m/z) calcd for C20H22N6O5Br (M+H)+ 505.0835, found 505.0837; HPLC (system A) 15.9 min (99 %), (system C) 12.1 min (99 %).
2-(3"-(5"-Methoxy-2"-methylindolyl)ethyloxy)adenosine (31)
[0191] The yield is 29 %. 1H NMR (CD3OD) δ 8.13 (IH, s), 7.10 (IH, dd, J= 0.6 and 8.8
Hz), 6.96 (IH, d, J= 2.2 Hz), 6.65 (IH, dd, J= 2.3 and 8.7 Hz), 5.90 (IH, d, J= 5.8 Hz), 4.68 (IH, t, J= 5.5 Hz), 4.47 (2H, m), 4.30 (IH, dd,J= 3.6 and 5.2 Hz), 4.10 (IH, q,J= 3.2 and 6.5 Hz), 3.84 (IH, dd, J= 2.9 and 12.2 Hz), 3.72 (IH, dd, J= 3.3 and 12.4 Hz), 3.13 (2H, t, J= 7.3 Hz), 2.37 (3H, s); HRMS (ESI-MS m/z) calcd for C22H26N6O8Na (M+Na)+ 493.1812, found 493.1792; HPLC (system A) 11.8 min (98%), (system C) 9.4 min (98 %).
2-(3"-(5"-Methoxy-2"-methyl-l"-(p-toluenesulfonyl)indolyl)ethyloxy)adenosine (32) [0192] The yield is 71 %. 1H NMR (CD3OD) δ 8.12 (lH,s), 7.96 (IH, d, J= 9.1 Hz),
7.52 (2H, d, J= 8.5 Hz), 7.11 (2H, d, J= 8.5 Hz), 6.92 (IH, d, J= 2.5 Hz), 6.82 (IH, dd, J= 2.5 and 9.1 Hz), 5.87 (IH, d, J= 6.0 Hz), 4.66 (IH, t, J= 5.6 Hz), 4.41 (2H, m), 4.29 (IH, dd, J= 3.2 and 5.1 Hz), 4.10 (IH, q, J=2.9 Hz), 3.82 (IH, dd, J= 2.8 and 12.4 Hz), 3.77 (3H, s), 3.70 (IH, dd, J= 3.2 and 12.5 Hz), 3.06 (2H, t, J= 6.6 Hz), 2.56 (3H, s), 2.21 (3H, s); HRMS (ESI-MS m/z) calcd for C29H33N6O8S (M+H)+ 625.2081, found 625.2079; HPLC (system B) 17.3 min (99 %), (system C) 17.6 min (99 %).
2-(3"-(5"-Methoxy-indoIyI)ethyIoxy)adenosine (33)
[0193] The yield is 54 %. 1H NMR (CD3OD) δ 8.12 (IH, s), 7.20 (IH, d, J= 8.8 Hz),
7.12 (IH, s), 7.05 (IH, d, J= 2.5 Hz), 6.73 (IH, dd,J= 2.5 and 8.8 Hz), 5.89 (IH, d,J= 5.8 Hz), 4.71 (IH, t, J= 5.5 Hz), 4.56 (2H, t, J=7.1 Hz), 4.31 (IH, dd, J=3.3 and 5.2 Hz), 4.10 (IH5 q, J= 3.2 Hz), 3.84 (IH, dd, J= 2.8 and 12.4 Hz), 3.72 (IH, dd, J= 3.3 and 12.4 Hz), 3.18 (2H, t, J= 7.1 Hz), HRMS (ESI-MS m/z) calcd for C2iH25N6O6 (M+H)+ 457.1836, found 457.1815; HPLC (system A) 10.7 min (98 %), (system C) 8.8 min (98%).
2-(3"-(5"-Hydroxyindolyl)ethyloxy)adenosine (34)
[0194] The yield is 31 %. 1H NMR (CD3OD) δ 8.12 (IH, s), 7.15 (IH, dd, J= 0.6 and
8.8 Hz), 7.09 (IH, s), 7.01 (IH, dd, J= 0.5 and 2.5 Hz), 6.65 (IH, dd, J= 2.3 and 8.4 Hz), 5.90 (IH, d, J= 5.8 Hz), 4.72 (IH, t, J= 5.6 Hz), 4.54 (2H, t, J= 7.4 Hz), 4.31 (IH, dd, J = 3.3 and 5.2 Hz), 4.11 (IH, q, J= 3.2 Hz), 3.86 (IH, dd, J= 2.7 and 12.4 Hz), 3.74 (IH, dd, J = 3.3 and 12.4 Hz), 3.13 (2H, t, J= 7.3 Hz); HRMS (ESI-MS m/z) calcd for C20H22N6O6Na (M+Na)+ 465.1499, found 465.1471; HPLC (system A) 5.5 min (98%), (system C) 5.3 min (98 %).
2-(3"-(Benzoimidazole-l"-yl)ethyloxy)adenosine (35)
[0195] The yield is 57 %. 1H NMR (CD3OD) δ 8.22 (IH, s), 8.10 (IH, s), 7.62 (2H, d with small coupling, J= 8.2 Hz), 7.31 (IH, dt, J= 1.2 and 7.6 Hz), 7.24 (IH, dt, J= 1.3 and 7.6 Hz), 5.84 (IH, d, J= 5.8 Hz), 4.63 - 4.74 (5H, m), 4.31 (IH, dd, J= 3.2 and 5.1 Hz), 4.11 (IH, q, J= 3.0 Hz), 3.86 (IH, dd, J= 2.8 and 12.4 Hz), 3.73 (IH, dd, J= 3.0 and 12.4 Hz); HRMS (ESI-MS m/z) calcd for Ci9H22N7O5 (M+H)+ 428.1682, found 428.1691; HPLC (system A) 4.9 min (99%), (system D) 8.3 min (99 %).
2-(3"-(Benzotriazole-l"-yI)ethyloxy)adenosine (36)
[0196] The yield is 40 %. 1H NMR (CD3OD) δ 8.10 (IH, s), 7.93(1H, dt, J=LO and 7.4
Hz), 7.80 (IH, dt, J=0.8 and 8.2 Hz), 7.52 (IH, ddd, J= 1.0, 7.1 and 8.1 Hz), 7.38 (IH, ddd, J=LO, 7.1 and 8.1 Hz), 5.83 (IH, d, J= 5.8 Hz), 5.13 (2H, m), 4.82-4.92 (2H, m, overlapped with HDO), 4.64 (IH, t, J= 5.6 Hz), 4.31 (IH, dd, J= 3.3 and 5.2 Hz), 4.10 (IH, q, J= 3.1 Hz), 3.83 (IH, dd, J= 2.8 and 12.6 Hz), 3.72 (IH, dd, J= 3.3 and 12.4 Hz); HRMS (ESI- MS m/z) calcd for Ci8H2iN8O5 (M+H)+ 429.1635, found 429.1642; HPLC (system A) 4.7 min (99 %), (system C) 4.8 min (99 %).
EXAMPLE 25
[0197] This example demonstrates a synthesis of 6-guanidino-2-(3"- indolylethyloxy)adenosine (37) and 6-guanidino-2-(3"-(l"-(p- toluenesulfonyl)indolyl)ethyloxy)adenosine (38) in an embodiment of the invention. See Figure 5.
[0198] To a solution of guanidine hydrochloride (98 mg, 1.02 mmol) in acetonitrile (2.2 mL) and N, N-dimethylformamide (1.1 mL) is added sodium hydride (60 %) (41.2 mg, 1.02 mmol) at room temperature, and the reaction mixture is stirred overnight. This guanidine solution is added to a mixture of compound 102 (46 mg, 0.0633 mmol) and 1,4- diazabicyclo[2.2.2]octane (14 mg, 0.0126 mmol), and the resulting mixture is stirred overnight at 110 0C and filtered. The filtrate is evaporated to give a crude oil, which is subjected to preparative TLC developed with a mixture of chloroform, methanol and saturated aqueous ammonia (2:1:0.3) to give 37 (2.3 mg, 8 %) and 38 (9 mg, 23 %) as an amorphous solid.
[0199] Compound 37: 1B ΝMR (CD3OD) £8.09 (IH, s), 7.60 (IH, d with small coupling, J= 7.7 Hz), 7.32 (IH, d with small coupling, J= 7.2 Hz), 7.16 (IH, s), 7.08 (IH, dt, J= 1.3 and 7.6 Hz), 7.01 (IH, dt, J = 1.2 and 7.3 Hz), 5.90 (IH, d, J= 6.0 Hz), 4.73 (IH, X, J = 5.5 Hz), 4.54 (2H, t, J= 7.1 Hz), 4.32 (IH, dd, J= 3.3 and 4.9 Hz), 4.12 (IH5 q, J= 3.0 Hz), 3.87 (IH, dd, J= 2.8 and 12.6 Hz), 3.73 (IH, dd, J= 3.3 and 12.4 Hz), 3.24 (2H, t, J =
7.4 Hz); HRMS (ESI-MS m/z) calcd for C2IH25N8O5 (M+H)+ 469.1948, found 469.1952; HPLC (system B) 8.9 min (97 %), (system D) 7.5 min (97 %).
[0200] Compound 38: 1H NMR (CD3OD) δ 8.25 (IH, s), 7.93 (IH, dd, J= 1.4 and 7.4 Hz), 7.71 (2H, d with small coupling, J= 8.5 Hz), 7.61 (IH, dd, J= 1.4 and 7.1 Hz), 7.59 (IH, s), 7.30 (IH, dt, J= 1.2 and 7.6 Hz), 7.24 (IH, dt, J= 1.2 and 7.6 Hz), 7.18 (2H, d with small coupling, J= 8.5 Hz), 5.96 (IH, d, J= 6.0 Hz), 4.74 (IH, t, J= 5.5 Hz), 4.59 (2H, t, J =
6.5 Hz), 4.35 (IH, dd, J= 3.4 and 5.1 Hz), 4.13 (IH, q, J= 3.2 Hz), 3.89 (IH, dd, J= 2.9 and 12.5 Hz), 3.76 (IH, dd, J= 3.6 and 12.4 Hz), 3.18 (2H, t, J= 6.5 Hz), 3.26 (3H, s); HRMS (ESI-MS m/z) calcd for C28H31N8O7S (M+H)+ 623.2036, found 623.2045; HPLC (system A) 16.3 min (97 %), (system C) 8.7 min (97 %).
EXAMPLE 26
[0201] This example demonstrates a synthesis of 6-ethylamino-2-(3"- indolyl)ethyloxy)adenosine (39) in an embodiment of the invention. See Figure 5. [0202] To a solution of 102 (28.3 mg, 0.0389 mmol) in DMF (1.4 mL) in sealed tube are added ethylamine hydrochloride (63.5 mg, 0.779 mmol) and N, N-diisopropylethylamine (0.271 niL), and the reaction mixture is stirred at 140 0C overnight and evaporated to give an oil. The oil is dissolved in methanol (1 mL), KOH (14.6 mg, 0.261 mmol) is added, and the reaction mixture is stirred for 42 h at 80 0C. The solvent is evaporated to give a crude solid which is subjected to preparative TLC developed with a mixture of chloroform and methanol (5:1) to give 39 (2.2 mg, 28 % yield in two steps). 1B NMR (CD3OD) £8.05 (IH, s), 7.60 (IH, d with small coupling, J= 7.1 Hz), 7.32 (IH, d with small coupling, J= 7.7 Hz), 7.14 (IH, s), 7.07 (IH, dt, J= 1.3 and 7.6 Hz), 7.00 (IH, dt, J= 1.1 and 7.4 Hz), 5.87 (IH, d, J = 6.0 Hz), 4.71 (IH, t, J= 5.6 Hz), 4.60 (2H, t, J= 7.0 Hz), 4.30 (IH, dd, J= 3.2 and 5.1 Hz), 4.11 (IH, q, J= 3.0 Hz), 3.86 (IH, dd, J= 2.6 and 12.5 Hz), 3.73 (IH, dd, J= 3.2 and 12.5 Hz), 3.50-3.66 (2H, br m), 3.22 (2H, t, J= 7.1 Hz), 1.26 (3H, t, J= 7.3 Hz); HRMS (ESI-MS m/z) calcd for C22H27N6O5 (M+H)+ 455.2043, found 455.2063; HPLC (system A) 19.5 min (99 %), (system C) 13.5 min (99 %).
EXAMPLE 27
[0203] This example demonstrates a synthesis of (2R, 3S, 4S, 5R)-6-amino-5- ethylcarboxyamide-2-(3"-indolyl)ethyloxy)-purin-9-yl)-tetrahydrofuran (40) in an embodiment of the invention. See Figure 6.
[0204] Potassium hydroxide (12.6 mg, 0.025 mmol) is added to a solution of 134 (7.0 mg, 0.0112 mmol) in methanol (1.5 mL), and the reaction mixture is stirred at 70 °C overnight. The mixture is evaporated to a small amount of solution, which is subjected to preparative TLC developed with a mixture of chloroform and methanol (5:1) to give 40 (1.7 mg, 33% yield). 1H NMR (CD3OD) δ 8.06 (IH, s), 7.56 (IH, d with small coupling, J= 8.0 Hz), 7.32 (IH, d with small coupling, J= 8.0 Hz), 7.11 (IH, s), 7.08 (IH, dt, J= 1.2 and 8.1 Hz), 7.00 (IH, dt, J = Ll and 8.1 Hz), 5.91 (IH, d, J= 7.4 Hz), 4.74 (IH, dd, J= 4.8 and 7.3 Hz), 4.61 (2H, m), 4.41 (IH, d, J= 1.9 Hz), 4.30 (IH, dd, J= 1.7 and 4.9 Hz), 3.15-3.26 (4H, m), 0.99 (3H, t, J= 7.2 Hz); HRMS (ESI MS m/z) calcd for C22H26N7O5(M+H)+ 468.1995, found 468.2015; HPLC (system A) 15.7 min (98 %), (system C) 11.4 min (98 %).
EXAMPLE 28
[0205] This example demonstrates cell culture and membrane preparation used in the testing of compounds in accordance with an embodiment of the invention. [0206] CHO (Chinese hamster ovary) cells expressing the recombinant human ARs (Gao et al., Biochem. Pharmacol. 2004, 68, 1985-1993) are cultured in DMEM supplemented with 10% fetal bovine serum, 100 units/mL penicillin, 100 μg/mL streptomycin, 2 μmol/mL glutamine, and 800 μg/mL geneticin. Cells are harvested by trypsinization. After homogenization and suspension, cells are centrifuged at 500 g for 10 min, and the pellet is re- suspended in 50 mM Tris-HCl buffer (pH 8.0) containing 10 mM MgCl2, 1 mM EDTA, and 0.1 mg/mL CHAPS. The suspension is homogenized with an electric homogenizer for 10 sec, and is then re-centrifuged at 20,000 g for 20 min at 4 0C. The resultant pellets are re- suspended in buffer in the presence of 3 Units/ml adenosine deaminase, and the suspension is stored at -80 0C until the binding experiments. The protein concentration is measured using the Bradford assay (Bradford et al., Anal. Biochem. 1976, 72, 248-254).
EXAMPLE 29
[0207] This example illustrates binding assays that can be carried out on compounds in accordance with an embodiment of the invention.
Human Ai and A2A Receptors
[0208] For binding to human A1 receptors, [3H]CCPA (1 nM) is incubated with membranes (40 μg/tube) from CHO cells stably expressing human Aj receptors at 25 0C for 60 min in 50 mM Tris-HCl buffer (pH 7.4; MgCl2, 10 mM) in a total assay volume of 200 μL. Nonspecific binding is determined using 10 μM of CPA. For human A2A receptor binding, membranes (20 μg/tube) from HEK-293 cells stably expressing human A2A receptors are incubated with 15 nM [3H]CGS21680 at 25 0C for 60 min in 200 μl 50 mM Tris-HCl, pH 7.4, containing 10 mM MgCl2. NECA (10 μM) is used to define nonspecific binding. The reaction is terminated by filtration with GF/B filters.
Human A3 Receptor
[0209] For competitive binding assay, each tube contains 100 μL membrane suspension (20 μg protein), 50 μL of [125I]I-AB-MECA (0.5 nM), and 50 μL of increasing concentrations of the nucleoside derivative in Tris-HCl buffer (50 mM, pH 7.4) containing 10 mM MgCl2, 1 mM EDTA. Nonspecific binding is determined using 10 μM of Cl-IB-MECA in the buffer. The mixtures are incubated at 25 0C for 60 min. Binding reactions are terminated by filtration through Whatman GF/B filters under reduced pressure using a MT-24 cell harvester (Brandell, Gaithersburgh, MD, USA). Filters are washed three times with 9 mL ice-cold buffer. Radioactivity is determined in a Beckman 5500B γ-counter. [0210] The adenosine receptor (AR) binding affinities of compounds 8 and 17 - 40 are investigated in comparison to compounds 1-7 and 9-16 (Table 1).
Table la
Figure imgf000051_0001
Figure imgf000051_0002
H, R2 = CH2OH, unless noted
ECso at K1 at
Ki at A1AR K1 at A3AR Efficacy,
A28AR (nM) A2AAR
Compound Name/Substitution (nM) or % (nM) or % A3AR or (% κ (nM) or % inhibition" ,. inhibition" % aaccttiivvaattiioonn)) inhibition
Reference
Agonists
R-Λ^-φhenylisopropyl)
1 1680±500 2.0±0.3 884±188 8.7±0.9 102±6 adenosine (R-PIA)
5'-JV-
2 ethylcarboxamido- 140+19 6.8±2.4 2.2+0.6 16.0±5.4 100 adenosine (NECA) 6-guanidino-NECA 54.5±13.3 7.0+1.0 628+39 5.1+1.3 100
(S)-2-(3-hydroxy-3- phenyl- 1 -propyn- 1 -
220 2.1 2.0 0.75 yl)NECA (OS)-PHP-NECA) -Ethers
5e (-1%) 2640±540 360+139 568+205 99±4
(36%) (36%) 579±250 578±182 52±3
3490±1490 221±57 9.3±2.9 54.2±14.3 71±3
(29%) 960±95 500±50 66 ± 3 42±8
%) 2060 ± 630 519 ± 41 352 ± 66 37±8
10e (13%) 1560 ± 250 413 ± 37 312 ± 47 18±8
lle (12%) 331+22 58.1+24.9 77.8+13.5 45±5
12e (64%) 312±24 69.3 ±4.7 119±8 50±7
13e (11%) 467±100 56.8+16.3 112+16 74±5
14e 1440±70 141±51 16.1±7.0 130+8 45±9
Figure imgf000052_0001
15e -O 1780±260 174±20 10.9+4.8 93.3±16.8 80±5
16e 280±72 13.3±4.1 101+34 62±15
17° 299±45 148±19 45.0±11.6 232±54 17±3
Figure imgf000052_0002
Figure imgf000053_0001
31 (48%) 579±88 (64*2%) 599±3 13*4
32 (24%) 20±2% (26*4%) (66*5%) 3±2
33 1250 1820±330 1400±300 360±50 -3*3
34 896 310 ± 90 450±8 120 ± 20 24±3
35 (18*4%) 3870±497 2070 ± 700 3±2
36 (41*5%) (46*2%) 1920 ± 470 13*4
37 (40%) 73.6 ± 8.0 277 ± 74 90 ± 10 58±8
Figure imgf000054_0001
R4 = C(NH)NH2
38 (42%) 52±2% 344 ± 72 457 ± 40 24±7
Figure imgf000054_0002
R4 = C(NH)NH2
39 3270 640±110 40±4% 30±10 54±12
Figure imgf000054_0003
R4 = Et
40 989 190±20 250±30 110*20 102±2
Figure imgf000054_0004
R2 = CONHEt aPotency of various adenosine derivatives in activation of the human A28AR expressed in CHO cells, with values expressed either the EC50 (nM) or the percent stimulation at 10 μM (in parentheses). For comparison, binding affinities of the adenosine derivatives at human Ai , A2A, and A3 ARs expressed in CHO cells (expressed as Kj value or percent displacement at 10 μM) and maximal agonist effects at 10 μM at the A3AR. Values for compounds 5 - 7 and 9 - 16 are from Gao et al., Biochem. Pharmacol. 2004, 68, 1985-1993; bAll experiments are performed using adherent CHO cells stably transfected with cDNA encoding a human AR. Percent activation of the human A2B or A3AR is determined at 10 μM. Binding at A1, A2A and A3ARs was carried out as described in Experimental Procedures. The A3 receptor activation results were from three separate experiments. The K1 and EC50 values from the present study are expressed as mean±s.e.m., N = 3-5; cCompounds 3, MRS3218; 17, MRS3534; 24, MRS3854; 28, MRS3997; dData from references Volpini et al., J. Med. Chem. 2002, 45, 3271-3279 and Vittori et al., Nucleosides Nucleotides Nucleic Acids 2004, 23, 471-481; and eData from Gao et al., Biochem. Pharmacol. 2004, 68, 1985-1993.
[0211] For several decades NECA (S'-N-ethylcarboxaminocarbonyladenosine) 2 was considered to be the most potent known agonist at the A2B AR, with an EC50 of 140 nM (Hide et al., MoI. Pharmacol. 1992, 41, 352-359; Gao et al., Biochem. Pharmacol. 2004, 68, 1985- 1993; and Klotz et al., Naunyn-Schmiedeberg's Arch. Pharmacol. 1998, 357, 1-9).
Figure imgf000055_0001
The functional effects of compound 28 compared to compound 2 on adenylate cyclase in CHO cells stably expressing the human ARs are depicted in Figure 7. Compound 28 is a full agonist at the A2A and A2B adenosine receptors. In the curves shown, the EC50 values for 2 are 21.9 nM (A2A, Figure 7B) and 110 nM (A2B, Figure 7C). The EC50 values of compound 28 are 39.7 nM (A2A, Figure 7B) and 109 nM (A2B, Figure 7C) are obtained. The relative maximal efficacy of 28 at the Ai and A3 adenosine receptors is 31.8% and 20.2±1.0% of the full agonist 2, respectively. Compound 28 is more selective to A2B adeonosine receptors than compound 2.
EXAMPLE 30
[0212] This example illustrates an anti-infarct effect of compound 28 in an isolated rabbit heart model of ischemia/reperfusion injury in accordance with an embodiment of the invention.
[0213] New Zealand White rabbits are anesthetized with pentobarbital sodium (30 mg/kg i.v.) and ventilated with 100% oxygen. A suture is passed around a coronary arterial branch.
The excised heart is perfused on a Langendorff apparatus with Krebs-Henseleit bicarbonate buffer bubbled with 95% O2/5% CO2 to a pH of 7.35 - 7.45 at 38 0C. A fluid-filled latex balloon measures pressure in the left ventricle as the heart spontaneously beats in an isometric fashion on the balloon.
[0214] A prominent branch of the left coronary artery is occluded (regional ischemia) for
30 min and reperfused for 2 hr in both the control and the treated groups. In the control group no other treatment is given. In the drug-treated group, 200 nM of compound 28 is given for one hour starting 10 min prior to reperfusion. Figure 8 shows the infarct sizes for the 3 groups. Both of the treated groups were protected.
[0215] The percentage of the ischemic region that infarcts in a rabbit heart is a function of the absolute size of the risk zone. Therefore, infarct size is examined by plotting it against the risk zone to make sure that the apparent salvage seen in the drug is not an artifact resulting from smaller risk zones in the treatment group. As shown in Figure 9, a line is fit to the control data with an intercept at about 0.35 cm3. This result means that no infarction would occur in an untreated heart with a 30-minute insult if the risk zone is less than 0.35 cm3. Figure 9 shows that all of the drug-treated hearts fell well below the line. Thus for any given risk zone volume, the infarcts are clearly smaller in the treated groups. In addition, a single high dose (e.g., 160 μg/kg) bolus plus a continued administration (e.g., 1920 μg/kg/hr) of compound 28 provides a reduction in the size of the infarcted area. [0216] The foregoing shows that compound 28 provides treatment/prevention of myocardial ischemia and/or myocardial ischemia/reperfusion injury.
[0217] All references, including publications, patent applications, and patents, cited herein are hereby incorporated by reference to the same extent as if each reference were individually and specifically indicated to be incorporated by reference and were set forth in its entirety herein.
[0218] The use of the terms "a" and "an" and "the" and similar referents in the context of describing the invention (especially in the context of the following claims) are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context. The terms "comprising," "having," "including," and "containing" are to be construed as open-ended terms (i.e., meaning "including, but not limited to,") unless otherwise noted. Recitation of ranges of values herein are merely intended to serve as a shorthand method of referring individually to each separate value falling within the range, unless otherwise indicated herein, and each separate value is incorporated into the specification as if it were individually recited herein. AU methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g., "such as") provided herein, is intended merely to better illuminate the invention and does not pose a limitation on the scope of the invention unless otherwise claimed. No language in the specification should be construed as indicating any non-claimed element as essential to the practice of the invention.
[0219] Preferred embodiments of this invention are described herein, including the best mode known to the inventors for carrying out the invention. Variations of those preferred embodiments may become apparent to those of ordinary skill in the art upon reading the foregoing description. The inventors expect skilled artisans to employ such variations as appropriate, and the inventors intend for the invention to be practiced otherwise than as specifically described herein. Accordingly, this invention includes all modifications and equivalents of the subject matter recited in the claims appended hereto as permitted by applicable law. Moreover, any combination of the above-described elements in all possible variations thereof is encompassed by the invention unless otherwise indicated herein or otherwise clearly contradicted by context.

Claims

CLAIM(S):
1. A compound of formula I or pharmaceutically acceptable salt thereof
Figure imgf000058_0001
(I) wherein
R1 is selected from the group consisting of aryl, heteroaryl, heterocycloalkyl, cycloalkylaryl, heterocycloalkylaryl, arylheteroaryl, and arylheterocycloalkyl, each of which is optionally substituted with 1 to 3 substituents selected from the group consisting of Ci-I2 alkyl, hydroxyl, CM2 alkoxy, halo, tosyl (Ts), CN, -C(O)OH, aminocarbonyl, amino, Ci-I2 dialkylamino, and Ci-I2 alkylamino;
Z is a bond or -CH2-;
n is an integer of 1 to 4;
R2 is selected from the group consisting Of -CH2OH, aminocarbonyl, Ci-I2 alkylaminocarbonyl, and di-Ci-i2 alkylaminocarbonyl; and
R3 and R4 are the same or different and each is selected from the group consisting of hydrogen, CM2 alkyl, C3-8 cycloalkyl, Cό-3o aryl, and imidamido;
provided that when R2 is -CH2OH, and R3 and R4 are hydrogen, then -0-(CH2)H-Z-R1 is not benzyloxy, phenylalkoxy or a substituted phenylalkoxy, thiophenylalkoxy or a substituted thiophenylalkoxy, pyridylalkoxy, indolylalkoxy, naphthylalkoxy, biphenylalkoxy, or indolylalkoxy.
2. The compound or pharmaceutically acceptable salt of claim 1, wherein R1 is selected from the group consisting of
C6-30 aryl, a heteroaryl consisting of one or two oxygen or sulfur atoms and/or from one to four nitrogen atoms provided that the total number of heteroatoms in each ring is four or less and each ring has at least one carbon atom, a heterocycloalkyl consisting of a 5 or 6- membered monocyclic ring that contains one, two, or three heteroatoms selected from nitrogen, oxygen, and/or sulfur, Cs-7 cycloalkyl-fused phenyl, heterocycloalkyl-fused phenyl, wherein the heterocycloalkyl consists of a 5 or 6-membered monocyclic ring that contains one, two, or three heteroatoms selected from nitrogen, oxygen, and/or sulfur, a benzo-fused heteroaryl, wherein the heteroaryl consists of one or two oxygen or sulfur atoms and/or from one to four nitrogen atoms provided that the total number of heteroatoms in each ring is four or less and each ring has at least one carbon atom, and a benzo-fused heterocycloalkyl, wherein the heterocycloalkyl consists of a 5 or 6-membered monocyclic ring that contains one, two, or three heteroatoms selected from N, O, and/or S.
3. The compound or pharmaceutically acceptable salt of claim 1 or 2, wherein R1 is selected from the group consisting of phenyl, biphenyl, naphthyl, anthracenyl, pyrenyl, pyridinyl, pyridazinyl, pyrimidyl, pyrazinyl, triazinyl, pyrrolyl, pyrazolyl, imidazolyl, (1,2,3,)- and (l,2,4)-triazolyl, pyrimidinyl, tetrazolyl, furyl, thiophenyl, isothiazolyl, thiazolyl, isoxazolyl, oxadiazolyl, oxazolyl, pyrrolo[2,3-c]pyridinyl, pyrrolo[3,2-c]pyridinyl, pyrrolo[2,3-b]pyridinyl, pyrrolo[3,2-b]pyridinyl, pyrrolo[3,2-d]pyrimidinyl, pyrrolo[2,3- d]pyrimidinyl), pyrrolinyl, pyranyl, piperidyl, tetrahydrofuranyl, tetrahydrothiopheneyl, morpholinyl), 5-, 6-, 7-, or 8-1,2,3,4-tetrahydronaphthalenyl, 4-, 5-, 6-, or 7-2,3-dihydro-lH- indenyl), 5-, 6-, 7-, or 8-1,2,3,4-tetrahydroquinolinyl, 5-, 6-, 7-, or 8-1,2,3,4- tetrahydroquinoxalinyl, 5-, 6-, 7-, or 8-2,3-dihydrobenzo[b][l,4]dioxinyl), benzimidazolyl, indolyl, indazolyl, benzo-l,2,3-triazolyl, 2-benzo[d]oxazolyl, 2-benzo[d]thiazolyl, 1,- 3-,or 4- isoquinolinyl, 2-, 3-, or 4-quinolinyl, 2- or 3-quinoxalinyl, 2,3-dihydrobenzo[b][l,4]dioxinyl, 2-indolinyl, and 3-indolinyl.
4. The compound or pharmaceutically acceptable salt of any of claims 1 -3, wherein R1 is selected from the group consisting of
Figure imgf000060_0001
wherein
R and R are the same or different and are selected from the group consisting of hydrogen, Ci-I2 alkyl, hydroxyl, Ci-I2 alkoxy, halo, tosyl, CN, -C(O)OH, aminocarbonyl, amino, Ci-I2 dialkylamino, and Ci-I2 alkylamino, and
R6 is selected from the group consisting of hydrogen, CM2 alkyl, C3-8 cycloalkyl, and C6-3o aryl.
5. The compound or pharmaceutically acceptable salt of claim 4, wherein R1 is selected from the group consisting of
Figure imgf000061_0001
6. The compound or pharmaceutically acceptable salt of claim 5, wherein R1 is selected from the group consisting of
Figure imgf000061_0002
7. The compound or pharmaceutically acceptable salt of any of claims 4-6, wherein R5 and R5 are the same or different and each is selected from the group consisting of hydrogen, Ci-Cj2 alkyl, hydroxyl, Cj-Ci2 alkoxy, halo, and tosyl.
8. The compound or pharmaceutically acceptable salt of any of claims 1 -5, wherein R1 is selected from the group consisting of
Figure imgf000061_0003
Figure imgf000062_0001
9. The compound or pharmaceutically acceptable salt of any of claims 1 -8, wherein n is 2 or 3.
10. The compound or pharmaceutically acceptable salt of any of claims 1 -9, wherein n is 2.
11. The compound or pharmaceutically acceptable salt of any of claims 1-10, wherein R2 is selected from the group consisting Of -CH2OH and Ci-I2 alkylaminocarbonyl.
12. The compound or pharmaceutically acceptable salt of any of claims 1-11, wherein R2 is -CH2OH.
13. The compound or pharmaceutically acceptable salt of any of claims 1-11, wherein R2 is N-ethylcarboxaminocarbonyl.
14. The compound or pharmaceutically acceptable salt of any of claims 1-13, wherein R3 and R4 are the same or different and each is selected from the group consisting of hydrogen, Ci-I2 alkyl, and imidamido.
15. The compound or pharmaceutically acceptable salt of any of claims 1-14, wherein R3 and R4 are hydrogen.
16. The compound or pharmaceutically acceptable salt of any of claims 1-14, wherein R3 is hydrogen and R4 is Ci-I2 alkyl or imidamido.
17. The compound or pharmaceutically acceptable salt of claim 16, wherein R4 is CM2 alkyl.
18. The compound or pharmaceutically acceptable salt of claim 16, wherein R4 is imidamido.
19. A pharmaceutical composition comprising the compound or pharmaceutically acceptable salt of any of claims 1-18 and a pharmaceutically acceptable carrier.
20. A method for activating an A2B adenosine receptor in a mammal comprising administering to the mammal an effective amount of a compound or pharmaceutically acceptable salt of any of claims 1-18.
21. A method for activating an A2B adenosine receptor in a cell comprising contacting the cell with a compound or pharmaceutically acceptable salt of any of claims 1- 18.
22. The method of claim 21 , wherein said contacting is carried out in vivo.
23. The method of claim 21, wherein said contacting is carried out in vitro.
24. The method of any of claims 20-23, further comprising activating an A2A adenosine receptor.
25. A method of treating or preventing (a) myocardial ischemia or (b) myocardial ischemia/reperfusion injury in a mammal comprising administering a therapeutically effective amount of a compound or pharmaceutically acceptable salt of any of claims 1-18 to the mammal, whereupon the (a) myocardial ischemia or (b) myocardial ischemia/reperfusion injury is treated or prevented.
26. The method of claim 25, wherein the compound or pharmaceutically acceptable salt thereof is administered for at least the first 24 hrs following reperfusion.
27. The method of claim 25 or 26, wherein the compound or pharmaceutically acceptable salt thereof is administered during reperfusion.
28. The method of any of claims 25-27, wherein the compound is 2-(3"-(6"- bromo-indolyl)ethyloxy)adenosine or a pharmaceutically acceptable salt thereof.
29. Use of a compound or pharmaceutically acceptable salt of any of claims 1-18 in the preparation of a medicament for activating A2B adenosine receptors of a mammal.
30. Use of a compound or pharmaceutically acceptable salt of any of claims 1-18 in the preparation of a medicament for treating or preventing myocardial ischemia or myocardial ischemia/reperfusion injury in a mammal.
PCT/US2008/067978 2007-06-29 2008-06-24 A2 adenosine receptor agonists WO2009006089A2 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US94706607P 2007-06-29 2007-06-29
US60/947,066 2007-06-29
US95025007P 2007-07-17 2007-07-17
US60/950,250 2007-07-17

Publications (2)

Publication Number Publication Date
WO2009006089A2 true WO2009006089A2 (en) 2009-01-08
WO2009006089A3 WO2009006089A3 (en) 2009-04-09

Family

ID=39952379

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2008/067978 WO2009006089A2 (en) 2007-06-29 2008-06-24 A2 adenosine receptor agonists

Country Status (1)

Country Link
WO (1) WO2009006089A2 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011133871A3 (en) * 2010-04-22 2012-03-08 Alnylam Pharmaceuticals, Inc. 5'-end derivatives
WO2014024195A1 (en) * 2012-08-09 2014-02-13 Can-Fite Biopharma Ltd. A3 adenosine receptor ligands for use in treatment of a sexual dysfunction

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996002553A2 (en) * 1994-07-14 1996-02-01 Glaxo Group Limited AMINO PURINE-β-D-RIBOFURANURONAMIDE DERIVATIVES

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996002553A2 (en) * 1994-07-14 1996-02-01 Glaxo Group Limited AMINO PURINE-β-D-RIBOFURANURONAMIDE DERIVATIVES

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
ADACHI, H. ET AL.: "Structure-activity relationship of 2,N6,5'-substituted adenosine derivatives with potent activity at the A2B adenosine receptor" J. MED. CHEM., vol. 50, April 2004 (2004-04), pages 1810-1827, XP002504703 *
BARALDI ET AL: "N6-[(Hetero)aryl/(cyclo)alkyl-carbamoyl- methoxy-phenyl]-(2-chloro)- 5'-N-ethylcarboxamido-adenosines: The first example of adenosine-related structures with potent agonist activity at the human A2B adenosine receptor" BIOORGANIC & MEDICINAL CHEMISTRY, ELSEVIER SCIENCE LTD, GB, vol. 15, no. 7, 12 March 2007 (2007-03-12), pages 2514-2527, XP005922442 ISSN: 0968-0896 *
BARALDI, P.G. ET AL.: "Synthesis and Biological Evaluation of Novel 1-Deoxy-1-[6-[((hetero)arylcarbonyl) hydrazino]- 9H-purin-9-yl]-N-ethyl-?-d-ribo furanuronamide Derivatives as Useful Templates for the Development of A2B Adenosine Receptor Agonists" J. MED. CHEM., vol. 50, 2007, page 374, XP002513993 *
UEEDA M ET AL: "2-Aralkoxyadenosines: potent and selective agonists at the coronary artery A2 adenosine receptor" JOURNAL OF MEDICINAL CHEMISTRY, US AMERICAN CHEMICAL SOCIETY. WASHINGTON, vol. 34, no. 4, 1 April 1991 (1991-04-01), pages 1340-1344, XP002961133 ISSN: 0022-2623 *
ZHAN-GUO GAO ET AL: "2-Substituted adenosine derivatives: affinity and efficacy at four subtypes of human adenosine receptors" BIOCHEMICAL PHARMACOLOGY, PERGAMON, OXFORD, GB, vol. 68, 2004, pages 1985-1993, XP002453822 ISSN: 0006-2952 cited in the application *

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011133871A3 (en) * 2010-04-22 2012-03-08 Alnylam Pharmaceuticals, Inc. 5'-end derivatives
US9725479B2 (en) 2010-04-22 2017-08-08 Ionis Pharmaceuticals, Inc. 5′-end derivatives
WO2014024195A1 (en) * 2012-08-09 2014-02-13 Can-Fite Biopharma Ltd. A3 adenosine receptor ligands for use in treatment of a sexual dysfunction
KR20150042232A (en) * 2012-08-09 2015-04-20 캔-파이트 바이오파마 리미티드 A3 adenosine receptor ligands for use in treatment of a sexual dysfunction
AU2013301125B2 (en) * 2012-08-09 2018-03-15 Can-Fite Biopharma Ltd. A3 adenosine receptor ligands for use in treatment of a sexual dysfunction
KR101971762B1 (en) 2012-08-09 2019-08-13 캔-파이트 바이오파마 리미티드 A3 adenosine receptor ligands for use in treatment of a sexual dysfunction
EP3530273A3 (en) * 2012-08-09 2020-02-26 Can-Fite Biopharma Ltd. A3 adenosine receptor ligands for use in treatment of a sexual dysfunction

Also Published As

Publication number Publication date
WO2009006089A3 (en) 2009-04-09

Similar Documents

Publication Publication Date Title
JP5431476B2 (en) A3 adenosine receptor antagonist and A3 adenosine receptor partial agonist
CN110240596B (en) Therapeutic agents for the treatment of HBV infection
RU2622104C2 (en) Macrocyclic lrrk2 kinase inhibitors
CA3121408A1 (en) Heteroaromatic derivatives for use as regulator, preparation method therefor and use thereof
KR20070050484A (en) Purine derivatives as a3 and a1 adenosine receptor agonists
US8916570B2 (en) A3 adenosine receptor agonists and antagonists
CA2987019A1 (en) Pyrido[3,4-d]pyrimidine derivative and pharmaceutically acceptable salt thereof
JP2004520273A (en) Thieno [2,3-C] isoquinoline for use as an inhibitor of PARP
PL187635B1 (en) Derivatives of adenosine n6-substituted with heterocyclic groups
CA2899646A1 (en) Substituted xanthines and methods of use thereof
KR20030008151A (en) Imidazopyridine and imidazopyrimidine antiviral agents
CA3110576A1 (en) Compounds for treating certain leukemias
US7199127B2 (en) Purine nucleosides
EP3567028B1 (en) Biphenyl compound as ccr2/ccr5 receptor antagonist
EP3240783B1 (en) New benzimidazole derivatives as antihistamine agents
CN116444492A (en) P38MAPK/MK2 pathway regulator, and composition, preparation method and application thereof
WO2009006089A2 (en) A2 adenosine receptor agonists
JPH0269496A (en) Imidazo (4, 5-b) pyridine derivative
CN116462662A (en) Aromatic ring Nav1.8 inhibitor and application thereof
EP0366171B1 (en) Nucleoside derivatives
WO2024012565A1 (en) Memantine derivative, pharmaceutical composition thereof and use thereof
CN114149457B (en) Benzo [ c ] [1,2] oxaborole-1 (3H) -alcohol compound and application thereof
RU2792034C2 (en) Tetrahydroisoquinoline compound, its production method, pharmaceutical composition containing such a compound, and its use
WO2022232810A1 (en) Purine nucleosides, their intermediates, and methods of preparation thereof
WO2020083089A1 (en) Compound of 5- or 6-membered heterocyclic pyrimidines and use thereof

Legal Events

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

Ref document number: 08771786

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase in:

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 08771786

Country of ref document: EP

Kind code of ref document: A2