WO2014042433A2 - Compounds and compositions for modulating adenosine a3 receptor activity - Google Patents

Compounds and compositions for modulating adenosine a3 receptor activity Download PDF

Info

Publication number
WO2014042433A2
WO2014042433A2 PCT/KR2013/008229 KR2013008229W WO2014042433A2 WO 2014042433 A2 WO2014042433 A2 WO 2014042433A2 KR 2013008229 W KR2013008229 W KR 2013008229W WO 2014042433 A2 WO2014042433 A2 WO 2014042433A2
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
compound
aryl
amino
independently
Prior art date
Application number
PCT/KR2013/008229
Other languages
French (fr)
Other versions
WO2014042433A3 (en
Inventor
MinJeong CHOI
Su-Sung OH
Original Assignee
Kainos Medicine, Inc.
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 Kainos Medicine, Inc. filed Critical Kainos Medicine, Inc.
Publication of WO2014042433A2 publication Critical patent/WO2014042433A2/en
Publication of WO2014042433A3 publication Critical patent/WO2014042433A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system

Landscapes

  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The present invention provides for Adenosine A3 receptor agonists, and also methods and use of the compounds of the invention, by themselves or in combination with other therapies, for treating a disease, disorder, or condition.

Description

DESCRIPTION
COMPOUNDS AND COMPOSITIONS FOR MODULATING ADENOSINE A3 RECEPTOR ACTIVITY CROSS-REFERENCE TO RELATED APPLICATION
This application claims priority to, and the benefit of, U.S. provisional application No. 61/700,924, filed September 14, 2012, the content of which is incorporated herein by reference in its entirety.
FIELD OF THE INVENTION
The present invention relates to certain compounds which can modulate the activity of adenosine A3 receptor; and relates to processes for their preparation, as well as pharmaceutical compositions containing them as an active ingredient, and their use as medicaments.
BACKGROUND OF THE INVENTION
Extracellular adenosine can bind to a number of receptors on the cell membranes, including, among others, adenosine Al receptor, adenosine A2A receptor, adenosine A2B receptor, and adenosine A3 receptor ("A3 receptor"). These receptors are G protein-coupled receptors that are involved in a variety of intracellular signaling pathways and physiological functions. The A3 receptor is widely expressed in human tissues with the most abundant expression in the lung and liver. It has been reported that A3 receptors are primarily expressed on inflammatory cells such as eosinophils in human lung, and also expressed on mast cells and neutrophils involved in inflammation, whereas low expression is found in other normal tissues. This pattern of expression on inflammatory cells is supportive of a role of A3 receptor in inflammation. A3 receptor was also found to be highly expressed in tumor cells and tissues, with low expression levels in normal cells or adjacent tissue. Further, A3 receptor expression in the tumor tissues was directly correlated to the severity of the cancer. This pattern of expression on tumor cells and tissue, but not surrounding normal cells and tissue, is supportive of a role of A3 receptor in tumor pathology.
Further studies support the roles of A3 receptor in diseases involving inflammation. For example, it is known that inflammation plays a role in the pathogenesis of early brain injury following subarachnoid hemorrhage, and that a selective A3 receptor agonist (e.g., 2-chloro-N6-(3-iodobenzyl)-adenosine-5'-N- methyluronamide (Cl-IB-MECA) significantly attenuated brain injury after administration of the selective A3 receptor agonist. Thus, activation of the A3 receptor by the selective A3 receptor agonist resulted in anti-inflammatory effects. In another example, selective A3 receptor agonist, methyl l-[N6-(3-iodobenzyl)- adenin-9-yl] -b-D-ribofuronamide (IB-MECA) was shown in Phase I and Phase II trials in humans to have an anti-inflammatory effect when administered to individuals suffering from rheumatoid arthritis. In a further example, a selective A3 receptor agonist was tested in standard experimental models of liver inflammation. It was found that activating A3 receptor by an agonist resulted in protection of the liver against inflammation by a number of mechanisms, including preventing apoptosis of hepatocytes. A3 receptor agonists have also been reported to inhibit inflammation in a number of other experimental models of human diseases such as osteoarthritis, psoriasis, and inflammatory bowel disease. Thus, activation of A3 receptor by an A3 receptor agonist induces an anti-inflammatory effect, and such agonists are potential inhibitors of inflammatory diseases, including autoimmune diseases.
A3 receptors are also expressed on cardiac ventricular cells. Several studies have shown that activation of A3 receptor with one of a variety of A3 receptor agonists resulted in a cardioprotective effect against the deleterious effect of subsequent cardiac ischemia and reperfusion injury. Thus, activation of A3 receptor by an A3 receptor agonist induces a cardioprotective effect, and such agonists are potential inhibitors of cardiac ischemia and reperfusion injury.
High expression of A3 receptor in tumor cells and tumor tissues is thought to be, at least in part, attributable to overexpression of NF-kappaB, known to act as an A3 receptor transcription factor. Treatment of tumor cells, or individuals with tumor, with A3 receptor agonists has shown to induce inhibition of tumor growth. Selective A3 receptor agonist, Cl-IB-MECA (at an IC 50 of 5 μΜ, and 14μΜ, respectively), was shown to inhibit growth of melanoma cells and lung carcinoma cells. A synergistic effect against tumor cell growth and metastasis has been observed when an A3 receptor agonist was combined with another chemotherapeutic agent (e.g., cyclophosphamide). Additionally, an A3 receptor agonist was shown to prevent the myelotoxic effects of cyclophosphamide during combinatorial therapy by increasing the number of white blood cells and the percentage of neutrophils. Thus, an A3 agonist can additionally function as a chemoprotective agent. Selective A3 receptor agonist, Cl-IB-MECA was shown (at nanomolar to micromolar levels) to inhibit growth of human hepatocellular carcinoma cells, human prostate carcinoma cells, human ovarian carcinoma cells, colon carcinoma cells, and lymphoma cells. Taken together, A3 receptors abundantly expressed in tumor cells may be targeted by A3 agonists, leading to inhibition of tumor growth.
SUMMARY OF THE INVENTION
The invention includes compounds comprising a C-nucleoside that selectively activates Adenosine A3 receptor. The compounds of the present invention, comprising A3 receptor agonists, surprisingly may be more chemically stable in body under physiological conditions, as compared to N-nucleoside compounds (e.g., C-C bond chemically more stable than C-N bond), as well as more stable to purine nucleoside phosphorylase enzyme activity. It is an object of the present invention to provide a novel compound which is effective as an A3 receptor agonist.
It is another object of the present invention to provide a pharmaceutical composition for treating or preventing a disease, disorder, or condition modulated by activating adenosine A3 receptor on cells. It is yet another object of the present invention to provide a method for treating or preventing a disease, disorder, or condition modulated by activating adenosine A3 receptor on cells in a mammal.
It is still another object of the present invention to provide a use of the inventive compound. In accordance with one aspect of the present invention, there is provided a compound of Formula (I), or a pharmaceutically acceptable salt, a prodrug, a hydrate, a solvate, or an isomer thereof, wherein formula (I) is as defined herein.
In accordance with another aspect of the present invention, there is provided a pharmaceutical composition comprising the compound of formula (I) as an active ingredient, or a pharmaceutically acceptable salt, a prodrug, a hydrate, a solvate, or an isomer thereof, and a pharmaceutically acceptable carrier.
In accordance with yet another aspect of the present invention, there is provided a method for treating or preventing a disease, disorder, or condition modulated by activating adenosine A3 receptor on cells in a mammal, which comprises administering the compound of formula (I), or a pharmaceutically acceptable salt, a prodrug, a hydrate, a solvate, or an isomer thereof to the mammal.
In accordance with a further aspect of the present invention, there is provided a use of the compound of formula (I), or a pharmaceutically acceptable salt, a prodrug, a hydrate, a solvate, or an isomer thereof for the manufacture of a medicament for treating or preventing a disease, disorder, or condition modulated by activating adenosine A3 receptor on cells.
Other aspects, objects and features of the invention will be apparent from the following description.
DETAILED DESCRIPTION OF THE INVENTION
While the terms used in the description of the invention are believed to be well understood by one of ordinary skill in the pharmaceutical arts, definitions, where provided herein, are set forth to facilitate description of the invention, and to provide illustrative examples for use of the terms.
The terms "a," "an," and "the" may mean one or more, and may be used to reference both the singular and the plural. The term "alkyl" is used herein to refer to a hydrocarbon containing normal, secondary, tertiary, or cyclic carbon atoms (e.g., linear saturated aliphatic hydrocarbon groups, branched saturated aliphatic hydrocarbon groups, or a saturated or unsaturated non-aromatic hydrocarbon mono or multi- ring system (e.g., cycloalkyl)). When the term "alkyl" is used without reference to a number of carbon atoms, it is to be understood to refer to a C)-C10 alkyl, preferably Ci-8 alkyl, more preferably Cj-4 alkyl; e.g., a Q, C2, C3, C4, C5, C6, C7, C8, C9 or C10 alkyl.
The term "aminoalkyl" is used herein to refer to at least one amino group that is appended to the parent molecular moiety through an alkyl group, as defined herein.
The term "alkylamino" is used herein to refer to a straight or branched chain hydrocarbon containing from 1 to 10 carbon atoms, preferably from 1 to about 6 carbon atoms, connected to the parent molecular moiety through an NH group. Representative examples of alkylamino include, but are not limited to, methylamino, ethylamino, isopropylamino and tert-butylamino.
The term "alkylamido" is used herein to refer to an alkyl group as defined above, which is attached via— C(O)NH— or— NHC(O)— .
The term "aryl" is used herein to refer to cyclic, aromatic hydrocarbon groups which have 1 to 3 aromatic rings, for example phenyl or naphthyl. The aryl group may have fused thereto a second or third ring which is a heterocyclo, cycloalkyl, or heteroaryl ring, provided in that case the point of attachment will be to the aryl portion of the ring system. "Heteroaryl" refers to an aryl group in which at least one of the carbon atoms in the aromatic ring has been replaced by a heteroatom selected from oxygen, nitrogen and sulphur. The nitrogen and/or sulfur heteroatoms may optionally be oxidized and the nitrogen heteroatoms may optionally be quaternized. The heteroaryl group may be a 5 to 6 membered monocyclic, 7 to 11 membered bicyclic, or 10 to 16 membered tricyclic ring system.
The term "alkenyl" is used herein to refer to a straight or branched chain hydrocarbon containing from 2 to 10 carbons, preferably from 2 to 8 carbon atoms, more preferably from 2 to 4 carbon atoms, and containing at least one carbon-carbon double bond formed by the removal of two hydrogens. The term "alkynyl" is used herein to refer to a straight or branched chain hydrocarbon group containing from 2 to 10 carbon atoms, preferably from 2 to 8 carbon atoms, more preferably from 2 to 4 carbon atoms, and containing at least one carbon-carbon triple bond.
The term "BOC" is used herein to refer to tert-butyloxycarbonyl.
The term "carbocyclyl" (alone or in combination with another term(s)) is used herein to refer to a saturated cyclic (i.e., "cycloalkyl"), partially saturated cyclic (i.e., "cycloalkenyl"), or completely unsaturated (i.e., "aryl") hydrocarbyl substituent containing from 3 to 14 carbon ring atoms ("ring atoms" being the atoms bound together to form the ring or rings of a cyclic substituent). A carbocyclyl may be a single ring, which typically contains from 3 to 6 ring atoms. The term "carbocyclylalkyl" is used herein to refer to an alkyl group substituted with a carbocycle group.
The term "cycloalkyl" is used herein to refer to monocyclic or multicyclic (e.g., bicyclic, tricyclic, etc.) hydrocarbons containing from 3 to 12 carbon atoms, preferably from 3 to 10 carbon atoms, more preferably from 3 to 6 carbon atoms, that is completely saturated or has one or more unsaturated bonds but does not amount to an aromatic group.
The term "cyano" as used herein means a— CN group.
The term "halo" or "halogen" means—CI,—Br,—I, or— F.
The term "haloalkyl" means that at least one hydrogen atom on an alkyl group is replaced with a halogen atom selected from fluorine, chlorine, bromine, iodine, and combinations thereof. The degree of halogenation may range from one hydrogen atom on the alkyl group being replaced by a halogen atom (e.g., a monofluoromethyl group) to full halogenation (e.g., perhalogenation) wherein all hydrogen atoms on the alkyl group have been replaced by a halogen atom (e.g., trifluoromethyl or perfluoromethyl). The haloalkyl groups useful in embodiments of the invention may be partially or fully halogenated and may be linear or branched.
The term "heterocyclyl" is used herein to include non-aromatic, ring systems, including, but not limited to, monocyclic, bicyclic and tricyclic rings, which can be completely saturated, and have 3 to 12 atoms including at least one heteroatom, such as nitrogen, oxygen, or sulfur. The term "hydroxyalkyl" is used herein to refer to an alkyl that is substituted with at least one hydroxy substituent. Examples of hydroxyalkyl include hydroxymethyl, hydroxyethyl, hydroxypropyl and hydroxybutyl.
The terms "first" and "second" are used herein for purposes of distinguishing between two compounds, or between two compositions, as will be clearer from the description.
The phrase "medically effective amount" means an amount of a composition or compound that treats the particular disease, condition or disorder; ameliorates, relieves, or decreases one or more symptoms associated with the particular disease, condition or disorder; and/or delays or prevents the onset of symptoms of, or a pathological process associated, with the particular disease, condition or disorder described herein in more detail.
The terms "prevent", "prevents", or "preventing", as used herein, embrace one or more of preventative (prophylactically).
The terms "treat", "treats", or "treating" , as used herein, embrace one or more of therapeutically (palliative).
The term "pharmaceutically acceptable carrier" is used herein to mean any compound or composition or carrier medium useful in any one or more of administration, delivery, storage, stability of a composition or compound described herein. These carriers are known in the art to include, but are not limited to, a diluent, water, saline, suitable vehicle (e.g., liposome, microparticle, nanoparticle, emulsion, capsule), buffer, medical parenteral vehicle, excipient, aqueous solution, suspension, solvent, emulsions, detergent, chelating agent, solubilizing agent, salt, colorant, polymer, hydrogel, surfactant, emulsifier, adjuvant, filler, preservative, stabilizer, oil, binder, disintegrant, absorbant, flavor agent, and the like as broadly known in the pharmaceutical art.
Hereinafter, the present invention is described in detail.
The present invention provides a compound selected from the group consisting of a compound of Formula 1 below, and pharmaceutically acceptable salts, a prodrug, a hydrate, a solvate, and an isomer thereof:
Figure imgf000009_0001
wherein,
Y is oxygen, sulfur, or carbon;
X! is H, alkyl, aRbNC(=O)- or HORc-, wherein said Ra and Rb are each independently hydrogen, alkyl, amino, haloalkyl, aminoalkyl, BOC-aminoalkyl, or cycloalkyl, or optionally fuse together to form a heterocyclic ring containing two to five carbon atoms, and said Rc is alkyl, amino, haloalkyl, aminoalkyl, BOC-aminoalkyl, or cycloalkyl;
X2 is H, hydroxyl, alkylamino, alkylamido or hydroxyalkyl;
X3 and X4 are each independently hydrogen, hydroxyl, amino, amido, azido, halo, alkyl, carboxy, nitrile, nitro, trifluoro, aryl, alkaryl, thio, thioester, thioether, -OCOPh or -OC(=S)OPh, or both X3 and 4 are optionally oxygens connected to >C=S to form a 5-membered ring, or X2 and X3 optionally form the ring of
Figure imgf000009_0002
wherein R' and R" represent independently an alkyl group;
X5 is H, cyano, Cr8 alkyl, C2-8 alkenyl, or C2-8 alkynyl;
Z is nitrogen or carbon;
Rj is halogen, NRUR , N(Ru)OR , NR NR R , N3, NO, NO2, CHO, CN, -CH(=NRn), -CH=NNHRn, -CH=N(ORu), -CH(ORn)2, -C(=O)NRnR12, - C(=S)NRnR12, -C(=O)ORn, Ci-8 alkyl, C2-8 alkenyl, C2-8 alkynyl, C4-8 carbocyclylalkyl, C6-20 aryl, C2-20 heterocyclyl, heteroaryl, -C(=O)-C]-8 alkyl, - S(O)n-C1-8 alkyl, aryl-Cr8 alkyl, OR11 or SR11, and said n is 0, 1, or 2; and R2 and R3 are each independently H, halogen, NRnR12, N(Rn)ORn, NRNNR R12, N3, NO, NO2, CHO, CN, -CH(=NRN), -CH=NNHRU, - CH=N(OR ), -CH(OR")2, -C(=O)NRnR12, -C(=S)NR"R12, -C(=O)ORn, R11, ORn, or SRu,
1 1 12
wherein said R and R are each independently H, Cr8 alkyl, C2-8 alkenyl,
C2-8 alkynyl, C3-8 carbocyclyl, C4-8 carbocyclylalkyl, aryl-C[-8 alkyl, heterocyclyl- C 8 alkyl, C6-20 aryl, C2-2o heterocyclyl, heteroaryl, -C(=O)-Cr8 alkyl, or -S(O)n- Cr8 alkyl, or R and R optionally fuse together with a nitrogen to which they are both attached to form 3 to 7 membered heterocyclic ring, wherein any carbon atom of said heterocyclic ring is optionally replaced with -O-, -S(O) n- or -NRd-, wherein, optionally, said Cr8 alkyl, C2-8 alkenyl, C2-8 alkynyl, C3-8 carbocyclyl, C4-8 carbocyclylalkyl, aryl-C1-8 alkyl, heterocyclyl-Crs alkyl, C6-20 aryl, C2-20 heterocyclyl, and heteroaryl are each independently substituted with one or more halogen, hydroxyl, CN, N3, N(Rd)2, NH(Rd), NH2, C(O)N(Rd)2, C(O)NH(Rd), C(O)NH2, OC(O)N(Rd)2, OC(O)NH(Rd), OC(O)NH2, C(O)ORd, OC(O)Rd, C(O)Rd, OC(O)Rd, S(O)nRd, S(O)2N(Rd)2, S(O)2NH(Rd), S(O)2NH2, ORd or Rd; wherein Rd is C 8 alkyl, C2-8 alkenyl, C2-8 alkynyl, C3-8 carbocyclyl, C4-8 carbocyclylalkyl, aryl-Cr8 alkyl, heterocyclyl-CR8 alkyl, C6-2o aryl, C -20 heterocyclyl, or heteroaryl.
(Note: Ph is used to represent the phenyl group, C6H5).
In a preferred embodiment of the present invention, there is provided the compound of Formula (I), as defined below:
Y is oxygen;
X! is H or RaRbNC(=O)-, wherein said Ra and Rb are each independently hydrogen, Ci-8 alkyl, amino, haloalkyl, aminoalkyl, BOC-aminoalkyl, or cycloalkyl;
X2 is H, hydroxyl, alkylamino, alkylamido or hydroxyalkyl;
X3 and X4 are each independently hydrogen, hydroxyl, amino, amido, azido, halo, alkyl, carboxy, nitrile, nitro, trifluoro, aryl, alkaryl, thio, thioester, thioether, -OCOPh or -OC(=S)OPh;
X5 is H, cyano, Cr8 alkyl, C2-8 alkenyl, or C2-8 alkynyl;
Z is nitrogen or carbon; R, is halogen, NRnR12, N(Rn)ORn, NRnNRnR12, -CH(=NRn), - CH=NNHRU, -CH=N(ORn), -CH(OR )2, -C(=O)NRuR12, or -C(=S)NRnR12; and
R2 and R3 are each independently H, halogen, NRnR12, N(R11)OR11, NRnNRnR12, -CH(=NR ), -CH=NNHRn, -CH=N(ORn), -CH(ORn)2, - C(=O)NRuR12, or -C(=S)NRUR12,
1 1 1
wherein said R and R are each independently H, Cr8 alkyl, C2-8 alkenyl, C2-8 alkynyl, C3-8 carbocyclyl, C4-8 carbocyclylalkyl, aryl-Cr8 alkyl, heterocyclyl- Cr8 alkyl, C6-2o ryl, C2-2o heterocyclyl, heteroaryl, -C(=0)-Cr8 alkyl, or -S(O)n- Cr8 alkyl,
wherein, optionally, said Q-g alkyl, C2-8 alkenyl, and C2-8 alkynyl are each independently substituted with one or more halogen, hydroxyl, CN, or N3.
In a more preferred embodiment of the present invention, there is provided the compound of Formula (I), as defined below:
Y is oxygen;
X! is RaRbNC(=O)-, wherein said Ra and Rb are each independently hydrogen or Ci-8 alkyl;
X2 is H;
X3 and X4 are hydroxyl;
X5 is H or cyano;
Z is nitrogen or carbon;
R! is NRnR12; and
R2 and R3 are each independently H or halogen, wherein said R11 and R12 are each independently H or phenyl-d-8 alkyl.
Compounds especially useful in the present invention are selected from the group consisting of:
(1) (2S,3S,4R,5S)-3,4-dihydroxy-5-(4-((3- iodobenzyl)amino)imidazo[2, 1 -J] [ 1 ,2,4]triazin-7-yl)-N-methyltetrahydrofuran-2- carboxamide;
(2) (2S,3S,4R,5R)-5-cyano-3,4-dihydroxy-5-(4-((3- iodobenzyl)amino)imidazo[2, 1 -j] [ 1 ,2,4]triazin-7-yl)-N-methyltetrahydrofuran-2- carboxamide; and (3) (2S,3S,4R,5S)-3,4-dihydroxy-5-(4-((3- iodobenzyl)amino)pyrrolo[2, 1 -f [ 1 ,2,4]triazin-7-yl)-N-methyltetrahydrofuran-2- carboxamide. The compounds of Formula (I) can form salts, and salts of the compounds are included within the scope of the invention. The terms "salt" or pharmaceutically acceptable salt", as used herein, refers to inorganic or organic salts of a compound. These salts can be prepared, for example, by reacting a compound of Formula (I) with an amount of acid or base, such as an equivalent amount, and in a medium such as one in which the salt formed then precipitates, or in an aqueous medium followed by lyophilization. Representative salts include bisulfate, sulfate, benzene sulfonate, camphorsulfonate, laurylsulphonate, methanesulfonate, toluenesulfonate, naphthalenesulformate, acetate, trifluoracetate, benzoate, borate, butyrate, citrate, formate, fumarate, hydorbromide, hydrochloride, hydroiodide, lactate, laurate, maleate, malonate, mesylate, nitrate, oxalate, phosphate, hexafluorophosphate, propionate, salicylate, stearate, succinate, tartrate, thiocyanate, and the like. The salts may include base salts based on the alkali and alkaline earth metals, such as calcium, sodium, lithium, magnesium, and potassium; or with organic bases such as with organic amines (e.g., dicyclohexylamine, t-butyl amine, methylamine, dimethylamine, triethylamine, ethylamine, procaine, morpholine, N-methylpiperidine, dibenzylamine, and the like); or as an ammonium salt.
Prodrugs of the compounds of Formula (I), or salts thereof, are included within the scope of the invention. The term "prodrug", as used herein, refers to a compound that is transformed in vivo (e.g., by a metabolic, physiological, or chemical process) to yield a compounds of Formula (I), or a pharmaceutically acceptable salt, hydrate or solvate of the compound. Prodrugs, made by synthesizing one or more prodrug moieties as part of an active compound, can serve to enhance one or more of solubility, absorption, lipophilicity, pharmacodynamics, pharmacokinetics, and efficacy, as compared to the active compound without the one or more prodrug moieties. Various forms of prodrugs are known in the art. Examples of prodrugs of the compounds of the invention include an in vivo cleavable ester of a carboxy group (e.g., lower alkyl esters, cycloalkyl esters, lower alkenyl esters, benzyl esters, mono-or di-substituted lower alkyl esters, and the like); or S-acyl and O-acyl derivatives of thiols, alcohols, or phenols. "Prodrug moiety" refers to a labile functional group, including but not limited to a protective group, which can be removed or reduced from the active compound during a process elected from one or more of metabolism, systemic circulation, intracellular, hydrolysis, or enzymatic cleavage. Enzymes which are capable of an enzymatically activating a phosphonate prodrug include, but are not limited to, amidases, esterases, phospholipases, cholinesterases, and phosphases. Prodrug moieties can serve to enhance solubility, absorption and lipophilicity to optimize drug delivery, bioavailability and efficacy. A prodrug moiety may include an active metabolite or drug itself. Exemplary prodrug moieties include the hydrolytically sensitive or labile acyloxymethyl esters -CH2OC(=O)R3 and acyloxymethyl carbonates esters - CH2OC(=O)OR3 where R3 is CrC6 alkyl, CrC6 substituted alkyl, C6-C20 aryl or C6-C2o substituted aryl. Phosphonate prodrug moieties may include, but are not limited, to groups such as phosphate esters, phosphonate esters, phosphonamidate esters, wherein the ester may be further substituted with groups that confer a pharmaceutical advantage such as one or more of improved solubility in aqueous solutions, or prolonged in vivo exposure, or enhanced absorption through a mucosa of a compound according to Formula (I). Thio-containing prodrug moieties, reported to be useful for the intracellular delivery of drugs, contain an ethylthio group in which the thiol group is either esterified with an acyl group or combined with another thiol group to form a disulfide. Following deesterification or reduction of the disulfide, generated is a free thio intermediate which, in turn, breaks down.
The compounds of Formula (I) may exist in a solvated form or unsolvated form. Solvates of a compound of the invention may be formed in the synthetic process in which the compound becomes physically associated with one or more solvent molecules (e.g., such as by ionic and/or covalent bonding) or, optionally, may be converted to a solvate such as by dissolving the compound in desired amounts of a solvent of choice (e.g., organic solvent, water, or mixtures thereof) in forming a solution, heating the solution to a temperature higher that ambient temperature, and cooling the solution at a rate sufficient to form crystals of the solvate, which may then be further isolated using methods known the art. Examples of suitable solvents include methanolates, ethanolates, hydrates (where the solvent molecule is water), and the like.
The terms "purified" or "isolated" for a compound according to Formula
(I) refers to the physical state of the compound following isolation from a synthetic process or purification step described herein or well known to those in the art, and in sufficient purity to be characterizable by standard analytical methods described herein or well known in the art.
The compounds of Formula (I) may contain asymmetric or chiral centers, and thus exist in different stereoisomeric forms. All stereoisomers (e.g., geometric isomers, optical isomers, and the like), enantiomeric forms, diastereomeric forms, tautomeric forms, positional isomers, of the compounds of the invention are embraced within the scope of the invention. A first conformational form of the compound can be separated from a second and different conformational form of the compound using methods well known in the chemical arts such as by chromatography, crystallization, and methods of synthesis which selectively result in a particular desired conformational form.
Further, the present invention provides a pharmaceutical composition for preventing or treating cancer, inflammation, cardiac ischemia or reperfusion injury, comprising a compound of Formula (I), or a pharmaceutically acceptable salt, a prodrug, a hydrate, a solvate, or an isomer thereof, and a pharmaceutically acceptable carrier.
The pharmaceutical composition according to the present invention may comprise a medically effective amount (e.g., therapeutically effective amount, or prophylactic ally effective amount, or a combination thereof, as the medical condition, disease or disorder warrants and as known to a skilled medical practitioner) of a compound according to the present invention, as an A3 receptor agonist, for activating A3 receptor activity in modulating (for inhibiting, ameliorating, treating, preventing) a disease, disorder, or condition in which A3 receptor activation inhibits the disease, disorder, or condition. The pharmaceutical composition, in addition to comprising a compound according to the present invention, may also contain at least one additional pharmaceutical agent (i.e., other than an A3 receptor agonist) in a medically effective amount. For example, one or more compounds having A3 receptor agonist activity, when used to treat cancer, may be used in combination with one or more additional chemotherapeutic agents, with the potential for synergistically enhancing one or more of apoptosis of cancer cells, growth inhibition of cancer cells, or reducing the myelotoxic effect of the one or more additional chemotherapeutic agents in the combination. Examples of chemotherapeutic agents which can be used in combination with an A3 receptor agonist of the invention include, but are not limited to, LSD1 blockers PPAR (peroxisome proliferating-activator receptor) ligands (e.g., rosiglitazone); alkylating agents (e.g., nitrogen mustards, such as mechlorethamine, chlorambucil, cyclophosphamide, ifosfamide, and melphalan; nitrosoureas, such as streptozocin, carmustine, and lomustine; alkyl sulfonates, such as busulfan; triazines, such as dacarbazine and temozolomide; ethylenimines, such as thiotepa and altretamine; and platinum-based drugs, such as cisplatin, carboplatin, and oxalaplatin); antimetabolites (e.g., 5-fluorouracil, 6-mercaptopurine, capecitabine, cladribine, clofarabine, cytarabine, floxuridine, fludarabine, gemcitabine, hydroxyurea, methotrexate, pemetrexed, pentostatin, and thioguanine); anti-tumor antibiotics (e.g., anthracyclines, such as daunorubicin, doxorubicin, epirubicin, and idarubicin; and actinomycin-D, bleomycin, mitomycin-C, and mitoxantrone); topoisomerase inhibitors (e.g., topoisomerase I inhibitors such as topotecan and irinotecan; and topoisomerase II inhibitors, such as etoposide, teniposide, and mitoxantrone); mitotic inhibitors (e.g., taxanes, such as paclitaxel and docetaxel; epothilones such as ixabepilone; Vinca alkaloids, such as vinblastine, vincristine, and vinorelbine; and estramustine); corticosteroids (e.g., prednisone, methylprednisolone, and dexamethasone); proteosome inhibitors (e.g., bortezomib); targeted therapies (e.g., imatinib, gefitinib, sunitinib, rituximab, alemtuzumab, trastuzumab, and bortezomib); differentiating agents (e.g., retinoids, tretinoin, and bexarotene); and hormonal agents (e.g., anti-estrogens, such as fulvestrant, tamoxifen, and toremifene; aromatase inhibitors, such as anastrozole, exemestane, and letrozole; progestins, such as megestrol acetate; estrogens; anti-androgens, such as bicalutamide, flutamide, and nilutamde; gonadotropin-releasing hormone (GnRH), also known as luteinizing hormone- releasing hormone (LHRH) agonists or analogs, such as leuprolide and goserelin). In another example, one or more compounds of the present invention having A3 receptor agonist activity, when used to treat an inflammatory disease or condition (e.g., comprising osteoarthritis, rheumatoid arthritis, psoriasis, inflammatory bowel disease, liver inflammation, lung inflammation, brain inflammation) may be used in combination with one or more agents that include, but are not limited to, non-steroidal anti-inflammatory drugs (e.g., aspirin, ibuprofen, naproxen, ketoprofen, flurbiprofen, misoprostol, diclofenac potassium, dicolfenac sodium, sulindac, oxaprozin, diflunisal, piroxicam, indomethacin, etodolac, meclofenamate, meloxicam, fenoprofen, mefanamic acid, nabumetone, and tolmetin); and COX-2 inhibitors (e.g., celecoxib).
In another example, one or more compounds of the invention having A3 receptor agonist activity, when used to treat an ischemic disease or condition (e.g., comprising cardiac ischemia and reperfusion injury) may be used in combination with one or more agents that include, but are not limited to, aspirin, nitroglycerin, beta-blockers (e.g., acebutolol, atenolol, bisoprolol, metoprolol, nadolol, nebivolol, and propranolol), cholesterol lowering medications (statins (e.g., lovastatin, amlodipine and atorvastatin, rosuvastatin, sitagliptin/simvastatin, fluvastatin, atorvastatin, pitavastatin, and pravastatin); niacin; fibrates; and bile acid sequestrants), calcium channel blockers (e.g., amlodipine, diltiazem, felodipine, isradipine, nicardipine, nifedipine, nisoldipine, and verapamil); angiotensin converting enzyme inhibitors (e.g., benazepril, captopril, enalapril, fosinopril, lisinopril, moexipril, perindopril, quinapril, ramipril, and trandolapril); and ranolazine.
The pharmaceutical composition according to the present invention may be administered once, or multiple times, as needed, to deliver a medically effective amount of the composition, e.g., an amount effective to mediate modulation of a disease, disorder, or condition by activating A3 receptor on cells in the individual receiving the composition. For example, a medically effective amount of the composition comprising the compound of the present invention may be an amount that enters into cells which are contacted with the compound, and which results in activating A3 receptor on the cells. Such a medically effective amount of the composition will depend on such factors as the mode of administration, the formulation for administration, disease to be modulated, the size and health of the individual to receive such a composition, and other factors which can be taken into consideration by a medical practitioner whom is skilled in the art of determining appropriate dosages for treatment. An amount of compound of the present invention in the composition to be administered may vary from 0.01 milligrams to about 500 milligrams, and more typically from about 1 milligram per day to about 200 milligram per day. One skilled in the art can apply known principles and models of drug delivery and pharmacokinetics to ascertain a likely range of dosages to be tested in preclinical and clinical studies for determining a medically effective amount of a compound of the invention. The pharmaceutically acceptable carrier, used in the composition of the present invention may facilitate one or more of storage, stability, administration, and delivery, of the composition. The carrier may be particulate, so that the composition may be in, for example, powder or solid form. The carrier may be in a semi-solid, gel, or liquid formula, so that the composition may be ingested, injected, applied, or otherwise administered. The carrier may be gaseous, so that the composition may be inhaled.
For oral administration of the composition containing the compound of the present invention, suitable formulations may be presented in the form of tablets, caplets, capsules, and the like, in which typically the compound of the invention may be present in a predetermined amount as a powder, granules, solution, or suspension as the sole active agent, or in combination with an additional one or more pharmaceutical agents. As known in the art, such oral formulations typically involve one or more of a binder (e.g., syrup, sorbitol, gum, corn starch, gelatin, and acacia), a filler (e.g., lactose, sugar, starch, calcium phosphate), an excipient (e.g., dicalcium phosphate), a disintegrating agent (e.g., vegetable starch, alginic acid), a lubricant (e.g., magnesium stearate), a flavoring agent (sweetening agent, natural or artificial flavors). Such oral formulations may be coated or uncoated to modify their disintegration and/or absorption. Coating may be performed using conventional coating agents and methods known in the art.
The mode of administration of the compound or composition of the present invention to an individual (such as a human) in need of such composition or compound may be any mode known in the art to be suitable for delivering a pharmaceutical composition, and particularly suitable for treating a disease, disorder or condition by activating A3 receptors, and may include but is not limited to, intravenously, intraperitoneally, orally, subcutaneously, intramuscularly, intranasally, transdermally, by perfusion, and by peristaltic techniques. The compositions of the present invention may also be combined with other therapies, such as one or more additional pharmaceutical agents, to treat a disease, disorder or condition. Such combination therapy may be administered in concurrently, sequentially, or in regimen alternating between the composition of the invention and the other therapy. Such combination therapies may include the following treatments: a compound of Formula (I) with one or more additional chemotherapeutic agents, for treating cancer; a compound of Formula (I) with one or more anti-inflammatory agents (e.g., non-steroidal anti- inflammatory drugs, COX-2 inhibitors, and a combination thereof) for treating inflammation; a compound of Formula (I) with one or more agents for treating cardiac ischemia and reperfusion injury that include, but are not limited to, aspirin, nitroglycerin, beta-blockers, cholesterol lowering medications, calcium channel blockers, angiotensin converting enzyme inhibitors, ranolazine, and a combination thereof. The structure of the agents, for combination with the compound of Formula (I), identified herein, and their generic or trademark names, are readily available to those skilled in the art, such as from the standard compendium of drugs (e.g., The Merck Index) or from the applicable pharmaceutical company's web site, as well as dosages applicable for treatment (see also The Physician's Desk Reference). Alternatively, the doses and dosage regimen of an additional pharmaceutical agent, used in conjunction with the compound of the present invention in combination therapy, can be determined by a physician, taking into account the medical literature, the health, age and sex of the patient, the disease or condition or disorder to be treated, the mode of administration and dosing schedule of the pharmaceutical agent, and other relevant considerations. Generally, dosages of such agents can range from about 0.1 mg to 1000 mg per day, with more specific dosages dependent on the aforementioned factors.
Accordingly, provided herein is a pharmaceutical composition or medicament comprising a medically effective amount of the compound of one or more of Formula (I), in combination with a medically effective amount of one or more chemotherapeutic agents, anti-inflammatory drugs, agents for treating cardiac ischemia and reperfusion injury; and optionally further comprising a pharmaceutically acceptable carrier. Also provided herein is a pharmaceutical composition or medicament comprising a medically effective amount of the compound of one or more of Formula (I), and a pharmaceutically acceptable carrier.
Further, the present invention provides a method for preventing or treating cancer, inflammation, cardiac ischemia or reperfusion injury in a mammal, which comprises administering the compound of formula (I), or a pharmaceutically acceptable salt, a prodrug, a hydrate, a solvate, or an isomer thereof to the mammal.
Furthermore, the present invention provides a method for preventing or treating cancer, inflammation, cardiac ischemia or reperfusion injury in a mammal, comprising the steps of: (i) administering to the mammal in need thereof a first composition comprising a compound of Formula (I), and a pharmaceutically acceptable carrier; and (ii) administering to the mammal in need thereof a second composition comprising at least one additional pharmaceutical agent comprising one or more selected from the group consisting of a chemotherapeutic agent, anti-inflammatory agent, and an agent for treating cardiac ischemia and reperfusion injury.
In these methods, one or more compounds of the invention may be administered in a medically effective amount as the sole pharmaceutical agent, or may be administered in combination therapy wherein a medically effective amount of a compound of the invention is administered with a medically effective amount of at least one additional pharmaceutical agent. Such combination therapy may comprise (a) a single pharmaceutical composition comprised of a compound of formula (I), at least one additional pharmaceutical agent, and a pharmaceutically acceptable carrier; or (b) two separate pharmaceutical compositions, which can be administered simultaneously or sequentially, comprising a first composition comprising a compound of the invention and a pharmaceutically acceptable carrier; and a second composition comprising at least one additional pharmaceutical agent and a pharmaceutically acceptable carrier.
In addition, the present invention provides a use of a compound of
Formula (I), or a pharmaceutically acceptable salt, a prodrug, a hydrate, a solvate, or an isomer thereof for the manufacture of a medicament for preventing or treating a cancer, inflammation, cardiac ischemia or reperfusion injury. General Methods of Synthesis and Testing
The general methods for preparing the compounds of this invention are illustrated in the following schemes (in case Ri is NRnR12)
<Scheme I>
Figure imgf000020_0001
(X = H or CN)
,Si Si,
CD'
Figure imgf000020_0002
EXAMPLES
Herein after, the present invention is described in more detail. The following Examples are given for the purpose of illustration only, and are not intended to limit the scope of the invention. Example 1: Preparation of (2^,3^,4if,55 -3,4-dihydroxy-5-(4-((3- iodobenzyl)amino)imidazo[2,l-/ [l,2,4]triazin-7-yl)-N- methyltetrahydrofuran-2-carboxamide
<!-!> Preparation of (3R,4R,5RV2-(4-aminoimidazor2.1-/1[L2,41triazin- 7-yl -3,4-bis(benzyloxy)-5-((benzyloxy)methyl)tetrahvdrofuran-2-ol (2)
Figure imgf000021_0001
1 2
A flask equipped with a thermocouple was added with 7- bromoimidazo[2, l- |[l ,2,4]triazin-4-arnine (500 mg, 2.34 mmol, PharmaBlock) and anhydrous THF (5 mL). The reaction mixture was cooled down to 0 °C and treated dropwise with a solution of MeMgCl in THF (3 M, 0.78 mL). The rate of addition was controlled to maintain internal temperature below 10 °C. After completion of addition, the reaction mixture was cooling to 0 °C, treated with 1 ,2- bis(chlorodimethylsilyl)ethane (503 mg, 2.34 mmol) in a single portion and dropwise with the second portion of MeMgCl solution (3 M, 0.78 mL) while internal temperature was kept below 10 °C. The reaction mixture was cooled down to 0 °C again and treated with a solution of i-PrMgCl-LiCl in THF (1.3 M, 2.0 mL). The reaction mixture was stirred at room temperature, and the disappearance of 7-bromoimidazo[2,l- |[l,2,4]triazin-4-amine was checked with TLC. When about 95% of starting material was disappeared, the reaction mixture was cooled down to 0 °C and treated with a solution of (3R,4R,5R)-3,4- bis(benzyloxy)-5-((benzyloxy)methyl)dihydrofuran-2(3H)-one (1.96 g, 4.67 mmol) in THF (2.5 mL) via cannula. The resulting orange solution was stirred at room temperature for 12 h. The reaction was quenched with aq. 13% NH4C1 (5 mL), and resulting mixture was diluted with EtOAc (20 mL). Phases were separated, and the organic layer was dried (Na2SO4) and concentrated under reduced pressure to provide crude product. Flash chromatography (SiO2, EtOAc) afforded 2 as a colorless oil (560 mg, 45%).
MS (EI) for C31H31N5O5 (M+H) +, (calcd: 554.2); found 554.3.
<l-2> Preparation of 7-((2S,3S,4R,5R)-3,4-bis(benzyloxy)-5-
((benzyloxy)methyl)tetrahvdrofuran-2-yl)imidazor2. \-f\\l .2.41triazin-4-amine (3)
Figure imgf000022_0001
2 3
To a stirred solution of the compound 2 (1.0 g, 1.81 mmol) in CH2C12 (25 mL) at 0 °C was added boron trifluoride etherate (1.3 mL, 10.84 mmol) and triethylsilane (2.3 mL, 14.45 mmol). The reaction mixture was stirred at room temperature for 16 h and diluted with CH2C12 (30 mL) and saturated aq. NaHCO3 (30 mL). Phases were separated, and the organic layer was washed with water, saturated aq. NH4C1 and brine, dried over MgSO4 and concentrated under reduced pressure to provide crude product. Flash chromatography (SiO2, 3 : 1 EtOAc:hexane) to give the compound 3 as a yellow solid (675 mg, 70%).
1H NMR (400 MHz, methanol-cL,): S 7.98 (s, 1H), 7.46 (s, 1H), 7.36-7.26
(m, 1 1H), 7.21-7.19 (m, 4H), 5.45 (d, J = 5.2 Hz, 1H), 4.68-4.44 (m, 7H), 4.44 (t, J = 5.0 Hz, 1H), 4.29-4.27 (m, 1H), 4.20 (t, J = 5.2 Hz, 1H), 3.71 (dd, J = 10.8, 4.0 Hz, 1H) ), 3.62 (dd, J= 10.8, 4.4 Hz, 1H). <l-3> Preparation of 7-((2S,3S,4R,5R)-3,4-bis(benzyloxy)-5-
((benzyloxy)methyl)tetrahvdrofuran-2-yl)-4-chloroimidazor2, 1 -f\ \ 1 ,2,41triazine
Figure imgf000022_0002
3 4
To a solution of the compound 3 (400 mg, 0.74 mmol) in CH2C12 (10 mL) at 0 °C was added dropwise TMSCI (0.19 mL, 1.49 mmol) and t-butyl nitrite (0.28 mL, 3.72 mmol). The reaction mixture allowed to warm at room temperature 12 h, and the reaction was quenched saturated aq. NaHCO3 (30 mL). Resulting mixture was extracted with CH2C12 (50 mL). Organic layer was washed with brine, dried over Na2SO4 and concentrated under reduced pressure. The residue was purified with flash chromatography (SiO2, 2: 1 EtOAc:hexane) to provide the compound 4 as a yellow solid (100 mg, 24%).
1H NMR (400 MHz, DMSO-dg) : δ 8.80 (s, IH), 8.00 (s, IH), 7.38-7.27 (m, 1 1H), 7.26-7.21 (m, 4H), 5.46 (d, J = 5.2 Hz, IH), 4.64-4.50 (m, 7H), 4.26- 4.24 (m, IH), 4.18 (t, J = 5.2 Hz, IH), 3.69 (dd, J= 10.8, 3.6 Hz, IH) ), 3.60 (dd, J= 10.8, 4.4 Hz, IH).
<l-4> Preparation of 7-((2S.3S,4R,5RV3.4-bis(benzyloxy)-5- ((benzyloxy)methyl)tetrahydrofuran-2-yl)-N-(3-iodobenzyl)imidazo|"2,l- /ir i,2,41triazin-4-amine (5
Figure imgf000023_0001
4 5
To a solution of the compound 4 (25 mg, 0.04 mmol) in ethanol (5 mL) was added 3-iodobenzylamine (63 mg, 0.27 mmol). The reaction mixture was stirred at 81 °C for 3 h. The reaction mixture concentrated under reduced pressure, and the residue was purified with flash chromatography (SiO2, 1 :5 EtOAc:hexane) to afford the compound 5 as a colorless oil (22 mg, 65%).
1H NMR (400 MHz, DMSO-d6): δ 9.39 (t, J= 5.2 Hz, IH), 8.12 (s, IH), 7.75 (brs, IH), 7.61 (d, J= 8.0 Hz, IH), 7.50 (s, IH), 7.38-7.19 (m, 16H), 7.12 (t, J = 8.0 Hz, IH), 5.35 (d, J = 5.2 Hz, IH), 4.68-4.48 (m, 8H), 4.45 (t, J = 5.0 Hz, IH), 4.21-4.15 (m, 2H), 3.65 (dd, J = 10.8, 3.6 Hz, IH) ), 3.58 (dd, J = 10.8, 4.4 Hz, IH).
<l-5> Preparation of (2R,3S,4R,5S)-2-(hydroxymethvn-5-(4-((3- iodobenzyl)amino)imidazo|"2, 1 -f\ \ 1 ,2,41triazin-7-yl)tetrahydrofuran-3 ,4-diol (6)
Figure imgf000024_0001
A solution of the compound 5 (148 mg, 0.20 mmol) in anhydrous CH2C12 (5 mL) at -78 °C under N2 was treated with a solution of boron trichloride in CH2C12 (IN, 0.8 mL). The reaction mixture stirred at -78 °C for 1 h, and the reaction was quenched with methanol (5 mL). Resulting mixture was warm to room temperature and concentrated under reduced pressure. The residue was purified with flash chromatography (SiO2, 1 : 10 MeOH:CH2Cl2) to give the compound 6 as a white powder (53 mg, 56%).
Ή NMR (400 MHz, methanol^) : S 8.27. (s, 1H), 7.83-7.80 (m, 2H), 7.65 (d, J = 8.0 Hz, 1H), 7.43 (d, J = 8.0 Hz, 1H), 7.13 (t, J = 8.0 Hz, 1H), 5.25 (d, J= 6.0 Hz, 1H), 4.83 (s, 2H), 4.50 (t, J= 5.8 Hz, 1H), 4.20 (t, J= 4.8 Hz, lH), 4.05-4.04 (m, 1H), 3.81 (dd, J = 12.0, 3.2 Hz, 1H) ), 3.70 (dd, J = 12.0, 4.0 Hz, 1H). <l-6> Preparation of ((3aR.4R,6S.6aSV6-i4-(Y3- iodobenzyl)amino imidazof2, 1 -f\\ 1 ,2,4 triazin-7-yl -2,2-
Figure imgf000024_0002
To a solution of the compound 6 (53 mg, 0.1 1 mmol) in acetone (5 mL) at room temperature were added 2,2-dimethoxy propane (206 mg, 1.97 mmol) and camphor- 10-sulfonic acid (25 mg, 0.11 mmol). The reaction mixture was stirred for 2 h and quenched with NaHCO3 (28 mg, 0.33 mmol). Resulting mixture was stirred for lh and diluted with H2O (30 mL) and chloroform (20 mL). Phases were separated, and organic phase was washed with saturated aq. NaHCO3 (30 mL) and brine, dried over Na2SO4, and concentrated under reduced pressure. The residue was purified with flash chromatography (SiO2, 1 :1 EtOAc:hexane) to afford the compound 7 as a white powder (53 mg, 93%).
1H NMR (400 MHz, methanol-cL : δ 8.13 (s, 1H), 7.78 (brs, 1H), 7.64-
7.61 (m, 2H), 7.40 (d, J = 7.6 Hz, 1H), 7.11 (t, J = 7.8 Hz, 1H), 5.31 (d, J = 4.8 Hz, 1H), 5.16-5.13 (m, 1H), 4.86-4.84 (m, 1H), 4.77 (s, 2H), 4.17 (q, J = 4.1 Hz, 1H), 3.74-3.64 (m, 2H), 1.59 (s, 3H), 1.37 (s, 3H). <\-l> Preparation of (3aS,4S,6S.6aS)-6-('4-rf3- iodobenzyl)amino)imidazo[2, l-f]\ l,2,4]triazin-7-yl -2,2- dimethyltetrahvdrofuror3,4-<f1[l,31dioxole-4-carboxylic acid (8)
Figure imgf000025_0001
A mixture of the compound 7 (53 mg, 0.10 mmol), bis(acetoxy)iodobenzene (BAIB, 72 mg, 0.22 mmol) and 2,2,6,6- tetramethylpiperidine-l-oxyl (TEMPO, 3 mg, 0.02 mmol) in 1 : 1 CH3CN:H2O (4 mL) was stirred at room temperature for 5 h. The reaction mixture was diluted with CH2C12 (15 mL) and water (15 mL). Phases were separated, and the aqueous layer was extracted with CH2C1 (2 x 15 mL). The combined organic layers were dried over Na2SO4 and concentrated under reduced pressure to provide the compound 8 as yellow oil (54 mg, 100%). 1H NMR (400 MHz, methanol-d4): δ 8.09 (s, 1H), 7.78 (s, 1H), 7.72 (dd, J = 8 Hz, 1.2 Hz, 1H), 7.63 (d, 1H), 7.41 (d, J = 7.6 Hz, 1H), 7.13 (m, 1H), 5.54 (m, 1H), 5.37 (m, 1H), 5.37 (dd, J= 6.0 Hz, 2.0 Hz, 1H), 5.26 (dd, J= 6.4 Hz, 1.6 Hz, 1H), 4.77 (s, 2H), 4.53 (m, 1H), 1.56 (s, 3H), 1.38 (s, 3H).
<l-8> Preparation of (2S,3S,4R,5S)-ethyl 3,4-dihvdroxy-5-(4-(Y3- iodobenzyl)amino)imidazo 2, 1 -f] \ 1 ,2,4]triazin-7-yl')tetrahydrofuran-2-carboxylate
Figure imgf000026_0001
8 9
To a solution of the compound 8 (40 mg, 0.07 mmol) in anhydrous ethanol (5 mL) at 0 °C was added SOCl2 (43 mg, 0.37 mmol). The reaction mixture was stirred at room temperature for 15 h and concentrated under reduced pressure. The residue was purified with flash chromatography (SiO2, 1 : 1 EtOAc:hexane) to give the compound 9 as a colorless oil (25 mg, 64%).
1H NMR (400 MHz, methanol-d4) : S 8.12 (s, 1H), 7.77 (brs, 1H), 7.74
(s, 1H), 7.61 (d, J = 8.0 Hz, 1H), 7.40 (d, J = 7.2 Hz, 1H), 7.10 (t, J = 7.8 Hz, 1H), 5.42 (d, J = 5.6 Hz, 1H), 4.77 (s, 2H), 4.50-4.4 (m, 2H), 4.39 (t, J = 4.6 Hz, 1H), 4.24 (dd, J= 7.2 Hz, 1.6 Hz, 2H), 1.29 (t, J= 7.2 Hz, 3H). <l-9> Preparation of (2S,3£4R,5S)-3,4-dihvdroxy-5-(4-(T3- iodobeiizyl)amino)imidazor2, 1 -f \ 1 ,2,41triazin-7-yl)-N-methyltetrahvdrofuran-2- carboxamide (10)
Figure imgf000027_0001
10
A flask containing the compound 9 (25 mg, 0.05 mmol) was treated with a solution of methylamine in methanol (2M, 2 mL). The reaction mixture was stirred at -20 °C for 3 h. Volatiles were removed under reduced pressure, and the residue was purified with flash chromatography (SiO2, 1 :20 MeOH:CH2Cl2) to give the compound 10 as a white powder (15 mg, 63%).
1H NMR (400 MHz, methanol-d4) : δ 8.21 (s, 1H), 7.78 (brs, 1H), 7.66 (s, 1H), 7.62 (d, J = 8.0 Hz, 1H), 7.41 (d, J = 8.0 Hz, 1H), 7.1 1 (t, J = 7.8 Hz, 1H), 5.23 (d, J = 8.8 Hz, 1H), 4.79 (d, J = 2.8 Hz, 2H), 4.63-4.59 (m, 1H), 4.39 (d, J = 3.6 Hz, 1H), 4.29 (dd, J = 5.0, 1.8 Hz, 1H), 2.84 (s, 3H). MS (EI) for C9H! ,DN2O5 (M+H) +, (calcd: 51 1.0); found 511.0.
Example 2: Preparation of (2S,3S,4R,5R)-5-cyano-3,4-dihydroxy-5- (4-((3-iodobenzyI)amino)imidazo[2,l-/ [l,2,4]triazin-7-yl)-N- methylt
Figure imgf000027_0002
<2-l> Preparation of (3R,4R,5R -2-(4-aminoimidazor2q- f] 1 ,2,41triazin-7-yl -3 ,4-bis(benzyloxy -5-((benzyloxy methyl tetrahvdrofuran-2- carbonitrile (1 1)
To a stirred solution of the compound 2 (2.2 g, 3.97 mmol) in CH2C12 (50 mL) at 0 °C was added trimethylsilyl trifluoromethanesulfonate (4.3 mL, 23.82 mmol) and trimethylsilyl cyanide (2.5 mL, 19.85 mmol). The reaction mixture was stirred at room temperature for 16 h and diluted with CH2C12 (30 mL) and saturated aq. NaHC03 (50 mL). Phases were separated, and the organic layer is washed sequentially with water, saturated aq. NH4C1 and brine, dried over MgSO4, and concentrated under reduced pressure. The residue was purified with flash chromatography (SiO2, 3:1 :1 EtOAc:hexane:CH2Cl2) to result in an isomeric mixture of the compound 11 (1.4 g, 62%).
MS (EI) for C32H30N6O4 (M+H) +, (calcd: 563.2); found 563.0.
<2-2> Preparation of (3R,4R,5RV3.4-bis(benzyloxyV5- ((benzyloxy methyl)-2-(4-chloroimidazo[2, 1 -f \ 1 ,2,41triazin-7-
Figure imgf000028_0001
To a solution of the compound 11 (144 mg, 0.25 mmol) in anhydrous CH2C12 (4.5 mL) at 0 °C was added dropwise TMSCI (0.12 mL, 1.02 mmol) and t-butyl nitrite (0.18 mL, 2.56 mmol). The reaction mixture was stirred at room temperature for 12 h, and the reaction was quenched with saturated aq. NaHCO3 (15 mL). The resulting mixture was extracted with CH2C12 (10 mL). The organic extract was washed with brine, dried over Na2SO4, and concentrated under reduced pressure. The residue was purified with flash chromatography (SiO2, 1 : 1 EtOAc:CH2Cl2) to give the compound 12 as an isomeric mixture (78 mg, 52%).
MS (EI) for C32H28ClN5O4 (M+H) +, (calcd: 582.1); found 582.4.
<2-3> Preparation of (3R,4R,5R)-3,4-bis(benzyloxy)-5- ((benzyloxy)methyl -2-(4-(f3-iodobenzyl)amino)imidazo|"2,l- 1 l,2,41triazin-7- yl)tetrahydrofuran-2-carbonitrile (13)
Figure imgf000029_0001
13
To a solution of the compound 12 (634 mg, 1.1 mmol) in ethanol (150 mL) was added 3-iodobenzylamine (0.87 mL, 6.5 mmol). The reaction mixture was stirred at 80 °C for 1 h and concentrated under reduced pressure. The residue was purified with flash chromatography (SiO2, 1 :2 EtOAc:hexane) to give the compound 13 as an isomeric mixture (950 mg, 88%).
MS (EI) for C39H35lN6O4 ((M+H)+, (calcd: 779.2) found 779.0.
<2-4> Preparation of (2R3R,4S,5R -3,4-dihydroxy-5-(hvdroxymethyl)- 2-(4-((3-iodobenzyl)amino)imidazo[2, 1 -f] \ 1 ,2,4]triazin-7-yl)tetrahydrofuran-2- carbonitrile (14)
Figure imgf000029_0002
A solution of the compound 13 (950 mg, 1.23 mmol) in anhydrous CH2C12 (50 mL) at -78 °C under N2 was treated with a solution of boron trichloride in CH2C12 (1 M, 7 mL). The reaction mixture was stirred at same temperature for 1 h, and the reaction was quenched with methanol (10 mL). The resulting mixture was allowed to warm to room temperature and concentrated under reduced pressure. The residue was purified with flash chromatography (SiO2, 1 :19 methanol :CH2C12) to afford the compound 14 as a white powder (53 mg, 56%). *
1H NMR (400 MHz, methanol-cLj: δ 8.14 (s, 1H), 7.77 (s, 1H), 7.75 (s, 1H), 7.60 (d, 1H), 7.39 (d, 1H), 7.08 (t, , J = 8.0 Hz, 1H), 4.76 (s, 2H), 4.25-4.22 (m, 2H), 3.82 (dd, 1H), 3.73 (dd, 1H). <2-5> Preparation of (3aR.4R.6R.6aRV6-(hvdroxymethylV4-(4-((3- iodobenzyl)amino)imidazo[2, 1 -f] \ 1 ,2,4]triazin-7-yl)-2,2- dimethyltetrahydrofuror3,4-dirL31dioxole-4-carbonitrile (15)
Figure imgf000030_0001
To a solution of the compound 14 (370 mg, 0.73 mmol) in acetone (50 mL) at room temperature were added 2,2-dimethoxy propane (1.6 mL, 13 mmol) and camphor- 10-sulfonic acid (170 mg, 0.73 mmol). The reaction mixture was stirred at room temperature for 8 h, and the reaction mixture was quenched with saturated aq. NaHCO3. The resulting mixture was diluted with chloroform (20 mL), and phases were separated. Organic layer was washed with brine, dried over Na2SO4 and concentrated under reduced pressure. The residue was purified with flash chromatography (SiO2, 3% methanol-CH2Cl2) to provide the compound 15 as a white powder (280 mg, 70%).
1H NMR (400 MHz, CDC13): δ 8.15 (s, 1H), 7.81 (s, 1H), 7.69 (s, 1H), 7.63 (d, J= 4 A Hz, 1H), 7.29 (d, J= 8.4 Hz, 1H), 7.07 (t, J= 7.6 Hz, 1H), 5.30 (m, 1H), 5.21 (dd, / = 6.8 Hz, 2.4 Hz, 1H), 4.78 (d, 2H), 4.66 (d, 1H), 4.22 (d, 1H), 3.97 (d, 1H), 3.84 (d, 1H), 1.78 (s, 3H), 1.37 (s, 3H).
<2-6> Preparation of (3aS,4S.6R.6aR)-6-cvano-6-i4-((3- iodobenzyl)amino)imidazor2J- irL2,4]triazin-7-yl -2.2- dimethyltetrahydrofuro[3,4-dl[1.31dioxole-4-carboxylic acid (16)
Figure imgf000031_0001
15 16
A mixture of the compound 15 (280 mg, 0.51 mmol), BAIB (360 mg, 1.12 mmol) and TEMPO (16 mg, 0.10 mmol) in 1 : 1 CH3CN:H2O (5 mL) was stirred at room temperature for 1 h. The reaction mixture was diluted with CH2C12 (15 mL) and treated with water (15 mL). Phases were separated, and the aqueous layer was extracted with CH2C12 (2 x 15 mL). Combined organic layers were dried over Na2SO4 and concentrated under reduced pressure to provide the crude the compound 16 as a yellow oil (470 mg, 100%), which used in the next step without purification.
1H NMR (400 MHz, methanol-d4) : δ 8.12 (s, 1H), 7.74 (s, 1H), 7.59 (m,
1H), 7.39 (m, 1H), 7.32 (t, 1H), 7.09 (t, 1H), 5.51 (m, 1H), 5.48 (m, 1H), 4.79 (m, 1H), 4.73 (m, 2H), 1.65 (s, 3H), 1.41 (s, 3H).
<2-7> Preparation of (2S,3S.4R,5S)-ethyl 3,4-dihvdroxy-5-(4-(Y3- iodobenzyl)amino)imidazo[2, 1 - I Γ 1 ,2,41triazin-7-yl tetrahvdrofuran-2-carboxylate
(17)
Figure imgf000031_0002
To a solution of crude the compound 16 (470 mg, 0.84 mmol) in anhydrous ethanol (80 mL) at 0 °C was added SOCl2 (0.3 mL, 4.2 mmol). The reaction mixture was stirred at room temperature for 15 h. Volatiles were removed under reduced pressure, and the residue was purified with flash chromatography (SiO2, 1 :2 EtOAc.hexane) to afford the compound 17 as a colorless oil (292 mg, 63%).
Ή NMR (400 MHz, CDC13): δ 8.30 (s, 1H), 8.09 (s, 1H), 7.95 (brs, 1H), 7.75 (s, 1H), 7.65 (d, 1H), 7.38 (d, 1H), 7.10 (t, 1H), 4.96 (s, 1H), 4.87 (m, 2H), 4.70 (m, 1H), 4.60 (m, 1H), 4.20 (q, J = 7.2 Hz, 2H), 2.88 (s, 3H), 1.25 (t, J= 7.2 Hz, 3H).
<2-8> Preparation of S.SS^R.SRVS-cvano^-dihvdroxy-S-^-iO- iodobenzyl)amino imidazo 2, 1 - | \ 1 ,2,41triazin-7-yl)-N-methyltetrahvdrofuran-2- carboxamide (18)
Figure imgf000032_0001
17 18
The compound 17 (292 mg, 0.53 mmol) in a flask was treated with a solution of methylamine in methanol (2 M, 40 mL) and stirred at -20 °C for 3 h. Upon completion of reaction, volatiles were removed under reduced pressure, and the residue was purified with flash chromatography (SiO2, 1 : 19 methanol:CH2Cl2) to give the compound 18 as a white powder(123 mg, 43%).
Ή NMR (400 MHz, methanol-d4) : δ 8.19 (s, 1H), 7.83 (s, 1H), 7.75 (s, 1H), 7.59 (d, J = 8.0 Hz, 1H), 7.39 (d, J = 7.6 Hz, 1H), 7.08 (t, J = 8.0 Hz, 1H), 4.98 (d, J = 5.2 Hz, 1H), 4.77 (d, J = 2.8 Hz, 2H), 4.60 (d, J = 2.8 Hz, 1H), 4.38 (dd, J = 5.2 Hz, 2.4 Hz, 1H), 2.78 (s, 3H). MS (EI) for C19H18IN704 (M+H) +, (calcd; 536.05); found 536.05. Example 3: Preparation of (2S,3S,4R,5S)-3,4-dihydroxy-5-(4-((3- iodobenzyl)amino)pyrrolo[2,l-/1[l,2,4]triazin-7-yl)-N- methyltetrahydrofuran-2-carboxamide
<3- l> Preparation of ('3R,4R,5RV2-(4-aminopyrrolor2J- 1|" L2.41triazm- 7-yl)-3^-bis(benzyloxy)-5-((benzyloxy)methyl)tetrahvdrofuran-2-ol (20)
Figure imgf000033_0001
19 20
A flask equipped with a thermocouple was added with 7- bromopyrole[2,l- |[ l,2,4]triazin-4-amine (1 g, 4.69 mmol) and anhydrous THF (10 mL). The reaction mixture was cooled down to 0 °C and treated dropwise with a solution of MeMgCl in THF (3 M, 1.57 mL). The rate of addition was controlled to maintain internal temperature below 10 °C. After completion of the · addition, the reaction mixture was cooled to 0 °C, treated with 1 ,2- bis(chlorodimethylsilyl)ethane (1.0 g, 4.69 mmol) in a single portion and dropwise with the second portion of MeMgCl solution (3 M, 1.57 mL) while internal temperature was kept below 10 °C. The reaction mixture was cooled down to 0 °C again and treated with a solution of /-PrMgCLLiCl in THF (1.3 M, 4.71 mL). The resulting dark solution was allowed to warm to room temperature, and the conversion was checked by TLC. Once the conversion of 7- bromoimidazo[2,l ][l,2,4]triazin-4-amine was > 95% complete (6 h), the solution was cooled down to 0 °C, and treated with a solution of (3R,4R,5R)-3,4- bis(benzyloxy)-5-((benzyloxy)methyl)dihydrofuran-2(3H)-one (2.56 g, 6.1 mmol) in THF (5 mL) via cannula. The resulting orange solution was stirred at room temperature for 12 h, and the reaction was found to be complete by TLC. At this point, aq. 13% NH4C1 (10 mL) was added, and phased were separated. Organic layer was added with EtOAc (40 mL), dried over NaSO4 and concentrated under reduced pressure. The residue was purified with flash chromatography (SiO2, 100% EtOAc) to afford the compound 20 (650 mg, 26%) as a yellow oil.
MS (EI) for C32H32N4O5 (M+H)+, (calcd; 553.2); found 553.3. <3-2> Preparation of 7-((2S,3S,4R,5R -3,4-bis(benzyloxy -5- ((benzyloxy)methyl)tetrahydrofuran-2-yl)pyrrolo[2, 1 -f] \ 1 ,2,41triazin-4-amine (21)
Figure imgf000034_0001
To a stirred solution of the compound 20 (0.65 g, 1.21 mmol) in CH2C12
(17 mL) at -78 °C was added boron trifluoride etherate (0.89 mL, 7.26 mmol) and triethylsilane (1.55 mL, 9.68 mmol), and the reaction mixture was allowed to warm to room temperature for 16 h. The reaction mixture was diluted with CH2C12 (20 mL) and saturated aq. NaHCO3 (20 mL). Phases were separate, and the organic layer is washed with water, saturated aq. NH4C1 and brine, dried over MgSO4, and concentrated. The residue was purified with flash chromatography (SiO2, 2:1 EtOAc: hexane) to give the compound 21 as a yellow oil (324 mg, 50%).
1H NMR (400 MHz, methanol-d4): δ 7.74 (s, 1H), 7.31-7.23 (m, 15H), 6.77 (d, J= 4.4 Hz, 1H), 6.61 (d, J = 4.4 Hz, 1H), 5.55 (d, J = 4.8 Hz, 1H), 4.61 (s, 2H), 4.54 (m, 2H), 4.47 (m, 2H), 4.30 (t, J = 4.8 Hz, 1H) ), 4.12 (t, J = 4.4 Hz, 1H), 3.67 (dd, J= 10.8 Hz, 3.6 Hz, 1H), 3.59 (ddt J= 10.8 Hz, 3.2 Hz, 1H).
<3-2> Preparation of 7-((2S S.4R,5R)-3,4-bis(benzyloxy -5- ((benzyloxy)methyl)tetrahvdrofuran-2-yl)-4-chloropyrrolof 2, 1 -f] [ 1 ,2,4]triazine (22)
Figure imgf000034_0002
To a solution of the compound 21 (1 g, 1.86 mmol) in anhydrous CH2C12 (40 mL) at 0 °C was added dropwise TMSCI (0.5 mL, 3.72 mmol) and t-butyl nitrite (0.70 mL, 9.3 mmol). The reaction mixture was allowed to warm at room temperature for 12 h. Upon completion of reaction, the reaction mixture was quenched with saturated aq. NaHCO3, and the resulting mixture was extracted with CH2C12. The organic layer was washed brine, dried over Na2SO4, and concentrated under reduced pressure. The residue was purified with flash chromatography (Si02, 1 :2 EtOAcrhexane) to afford the compound 22 as an oil (273 mg, 30%).
1H NMR (400 MHz, methanol-d4) : δ 8.22 (s, 1H), 7.89 (s, 1H), 7.34- 7.20 (m, 16H), 7.09 (d, J= 4.4 Hz, 1H), 6.94 (d, J= 4.8 Hz, 1H), 5.62 (d, J = 4.8 Hz, 1H), 4.67-4.48 (m, 6H), 4.34 (t, J = 4.8 Hz, 1H), 4.30 (m, 1H), 4.17 (t, J- 5.2 Hz, lH), 3.74 (dd, J= 10.8 Hz, 3.6 Hz, 1H), 3.64 (dd, J = 10.8 Hz, 4.0 Hz, 1H).
<3-3> Preparation of 7-(,(2S,3S,4R,5R)-3,4-bis(benzyloxy)-5- ((benzyloxy)methyl)tetrahydrofuran-2-yD-N-(3-iodobenzyl)pyrrolo[2,l-
Figure imgf000035_0001
To a solution of the compound 22 (734 mg, 1.32 mmol) in ethanol (250 mL) was added 3-iodobenzylamine (1.05 mL, 7.92 mmol). The reaction mixture was stirred at 81 °C for 1 h. Upon completion of reaction, the reaction mixture concentrated under reduced pressure. The residue was purified with flash chromatography (SiO2, 1 :4 EtOAcrhexane) to give the compound 23 as yellow oil (486 mg, 50%).
Ή NMR (400 MHz, CDC13) : δ 7.96 (s, 1H), 7.70 (s, 1H), 7.63 (d, J = 7.6 Hz, 1H), 7.34-7.23 (m, 11H), 7.07 (t, J= 7.6 Hz, 1H), 6.63 (d, J= 4.4 Hz, 1H), 6.42 (d, J = 4.4 Hz, 1H), 5.67 (d, 1H), 5.49 (brs, 1H), 4.77-4.72 (m, 3H), 4.54- 4.49 (m, 2H), 4.40- 4.36 (m, 2H), 4.27 (t, J = 4.8 Hz, 1H), 4.12-4.09 (m, 1H), 3.77 (dd, J= 10.8 Hz, 3.2 Hz, 1H), 3.65 (dd, J= 4.0 Hz, 10.8 Hz, 1H). <3-4> Preparation of (2R3S,4R¾5S)-2-(hvdroxymethyl)-5-(4-(Y3-
Figure imgf000036_0001
23 24
A solution of the compound 23 (31 1 mg, 0.41 mmol) in anhydrous
CH2C12 (20 mL) under N2 at -78 °C was treated with a solution of boron trichloride in CH2C12 (IN, 1.23 mL). The reaction mixture stirred at -78 °C for 10 min. When the reaction was complete by TLC, methanol (5 mL) was added to quench the reaction. The resulting mixture was allowed to room temperature and concentrated under reduced pressure. The residue was purified with flash chromatography (SiO2, 1 : 1 EtOAc:hexane) to give the compound 24 as a white solid (148 mg, 75%).
1H NMR (400 MHz, methanol-d4) : δ 7.96 (s, 1H), 7.79 (s, 1H), 7.72 (m, 1H), 7.42 (m, 2 H), 7.19 (m, 1H), 5.59 (d, 1H), (s, 2H), 4.25-4.22 (m, 2H), 3.82 (dd, 1H), 3.73 (dd, 1H).
<3-5> Preparation of (Y3aR,4R,6S.6aS)-6-(4-((3- iodobenzyl)amino)pyrrolo[2, 1 -f\\ 1 ,2,4"|triazin-7-yl)-2,2- dimethyltetrahydrofuro 3,4-dl l ,31dioxol-4-yl)methanol (25)
Figure imgf000037_0001
24 25
To a solution of the compound 24 (148 mg, 0.30 mmol) in acetone (30 mL) at room temperature was added 2,2-dimethoxy propane (0.2 mL, 1.53 mmol) and camphor- 10-sulfonic acid (70 mg, 0.30 mmol). The reaction mixture was stirred at room temperature for 1 h. Upon completion of reaction, the reaction mixture was quenched with saturated aq. NaHCO3. The resulting mixture was diluted with chloroform (20 mL), and phases were separated. The organic layer was washed with brine, dried over Na2SO4 and concentrated under reduced pressure. The residue was purified with flash chromatography (SiO2, 1 : 1 EtOAc:hexane) to give the compound 25 as yellow oil (63.2 mg, 40%).
1H NMR (400 MHz, CDC13): δ 7.96 (s, 1H), 7.69 (s, 1H), 7.61 (d, 1H), 7.29 (d, J = 7.6 Hz, 1H), 7.08 (t, J = 7.6 Hz, 1H), 6.87 (d, J = 4.8 Hz, 1H), 6.57 (d, J = 4.4 Hz, 1H), 5.68 (d, 1H), 5.53 (brs, 1H), 5.02 (m, 1H), 4.78-4.69 (m, 3H), 4.33 (m, 1H), 3.78-3.70 (m, 2H), 1.49 (s, 3H), 1.30 (s, 3H).
<3-6> Preparation of (3aS,4S,6S,6aS -6-(4-rf3- iodobenzyl)amino)pyrrolo[2J- L2,41triazin-7-yl)-2,2- dimethyltetrahydrofuro|"3,4-dl[l,31dioxole-4-carboxylic acid (26)
Figure imgf000038_0001
25 26
A mixture of the compound 25 (63 mg, 0.12 mmol), BAIB (147 mg, 0.24 mmol) and TEMPO (6.4 mg, 0.0024 mmol) in 1.5 mL of mixture solvent (CH3CN:H2O, 1 :1) was stirred at room temperature for 1 hour. Upon completion of reaction, the reaction mixture was diluted with CH2C12 (2 mL) and then added with water (2 mL). Phases were separated, and the aqueous layer was extracted with CH2C12 (2 5 mL). The combined organic layers were dried with Na2SO4 and concentrated under reduced pressure to provide give crude the compound 26 as yellow oil (32 mg), which was used in the next step without purification.
MS (EI) for C21H21IN4O5 (M+H) +, calcd; 537.1); found 537.1.
<3-7> Preparation of (3aS,4S,6S,6aS)-methyl 6-(4-((3- iodobenzyl)amino)pyrrolo|"2, 1 -f] \ 1 ,2,4]triazin-7-yl)-2,2-
Figure imgf000038_0002
26 27
To a solution of the compound 26 (32 mg, 0.06 mmol) in 1 : 1 THF:MeOH (1 mL) was added a solution of TMSCHN2 in hexane (1M, 0.26 mL), and the reaction mixture was stirred at room temperature for 3 h. The solvent was removed under reduced pressure, and the residue was purified with flash chromatography (SiO2, 1 : 1 EtOAc.hexane) to give the compound 27 as yellow oil (7 mg, 21%).
1H NMR (400 MHz, CDC13) : δ 7.93 (s, 1H), 7.64 (s, 1H), 7.58 d, J = 8.0 Hz 1H), 7.26 (d, J= 7.6 Hz, 1H), 7.03 (t, J= 7.6 Hz, 1H), 6.81 (d, J= 4.4 Hz, 1H), 6.52 (d, J= 4.8 Hz, 1H), 5.68 (d, J= 3.6 Hz, 1H), 5.50 (brs, 1H), 5.08 (d, J = 6.0 Hz, 1H), 4.99-4.96 (m, 1H), 4.74-4.65 (m, 3H), 3.73 (s, 3H), 1.40 (s, 3H), 1.26 (s, 3H).
<3-8> Preparation of (3aS,4S,6S,6aS)-6-(4-((3- iodobenzyl)amino)pyrrolo[2, 1 -f[ [ 1 ,2,4]triazin-7-yl)-N,2,2- trimethyltetrahvdrofuro 3,4-d1[L31dioxole-4-carboxamide (28)
Figure imgf000039_0001
27
The compound 27 (7 mg, 0.012 mmol) was treated with a solution of methylamine in methanol (2M, 0.5 mL) and stirred at -20 °C for 3 h. Upon completion of the reaction, volatiles were removed under reduced pressure, and the residue was purified with flash chromatography (SiO2, 1 :1 EtOAcihexane) to give the compound 28 as a yellow powder (7 mg, 100%).
Ή NMR (400 MHz, CDC13) : δ 7.93 (s, 1H), 7.64 (s, 1H), 7.59 (d, J = 8.4 Hz, 1H), 7.26 (d, J = 7.6 Hz, 1H), 7.09 (brs, 1H), 7.04 (t, J = 8.0 Hz, 1H), 6.93 (d, J= 4.4 Hz, 1H), 5.55 (brs, 1H), 5.28-5.23 (m, 3H), 4.91 (m, 1H), 4.73 (m, 2H), 4.51 (s, 1H), 2.83 (d, J = 5.2 Hz, 1H), 1.45 (s, 3H), 1.29 (s, 3H).
<3-9> Preparation of (2S.3S,4R,5S)-3,4-dihvdroxy-5-(4-(T3- iodobenzyl)amino)pyrrolo 2, 1 -f Γ 1 ,2,41triazin-7-yl)-N-methyltetrahvdrofuran-2- carboxamide (29)
Figure imgf000040_0001
The compound 28 (7mg, 0.012 mmol) was dissolved in 1 :1 TFA:H2O (2 mL) and stirred at room temperature for 3 h. Upon completion of reaction, the reaction mixture was diluted with CH2C12 (2 mL), and the reaction was quenched with saturated aq. NaHCO3 (2 mL). Phases were separated, and the organic layer was washed with brine, dried over Na2SO4 and concentrated under reduced pressure. The residue was purified with flash chromatography (SiO2, 3% MeOH- CH2C12) to give the compound 29 as a white solid (3 mg, 50%).
1H NMR (400 MHz, CDC13) : δ 7.92 (s, 1H), 7.64 (s, 1H), 7.59 (d, J = 8.0 Hz, 1H), 7.27 (d, J = 7.2 Hz, 1H), 7.04 (t, J = 7.6 Hz, 1H), 6.82 (d, J = 4.4 Hz, 1H), 6.54 (d, J = 4.4 Hz, 1H), 5.76 (m, 2H), 4.74 (m, 2H), 4.49-4.44 (m, 3H), 3.97 (brs 1H), 3.26 (brs, 1H) 2.79 (m, 3H). MS (EI) for Οι9Η2οΓΝ5Ο4 (M+H) +, (calcd: 510.1); found 510.06. Experimental Examples
Adenosine receptor assays were done in DiscoverX in California, USA. The activities of the compounds as agonist on adenosine receptors were assay as followings.
For Al and A3 receptors, the inhibition of folkskolin-induced cAMP production was measured. Cyclopentyladenosine (Discover X) and chloro-IB- MECA (Discover X) were used as standards, respectively. (1) cAMP Hunter cell lines (DiscoverX) expressing the Al or A3 receptor were seeded in a total volume of 20 \L into white walled, 384- well microplates and incubated at 37°C.
(2) Media (DMEM w/10% serum, DiscoverX) was aspirated from cells and replaced with 15 \i 2: 1 HBSS/1 OmM Hepes : cAMP XS+ Ab reagent.
(3) Prepare a series of dilution of sample stocks was made to 4X containing EC80 folkskolin in media, and added 5 and incubated at 37°C for 60 minutes. Final assay vehicle concentration was 1%.
(4) Cells were lysed through incubation with 20 cAMP XS+ ED/CL lysis cocktail for one hour followed by incubation with 20 cAMP XS+ EA reagent for three hours at room temperature to generate signals. Microplates were read for chemiluminescence using the PerkinElmer EnvisionTM instrument.
For A2A receptor, calcium mobilization was measured using NECA (DiscoverX) as a standard.
Cells were seeded in a total volume of 20 μΕ into blackwalled, clear bottom, Poly-D-lysine coated 384-well microplates and incubated at 37°C.
Cells were loaded with dye prior to testing by incubation with 20 \\L Dye Loading Buffer consisting of lx Dye, lx Additive A and 2.5 mM Probenecid in HBSS/20 mM Hepes (Probenicid was prepared fresh) for 30-60 minutes at 37°C.
Prepare a series of dilution of sample stocks were made to 3X, and added 10 ul to cells. Calcium mobilization was monitored on a FLIPR Tetra (MDS).
For A2B receptor, stimulation of cAMP production was measured using NECA as a standard.
(1) cAMP Hunter cell line expressing the A2B receptor were seeded in a total volume of 20 μΕ into white walled, 384-well microplates and incubated at 37°C.
(2) Media (DMEM w/10% serum, DiscoverX) was aspirated from cells and replaced with 15 μΐ 2: 1 HBSS/lOmM Hepes : cAMP XS+ Ab reagent.
(3) Prepare a series of dilution of sample stocks was made to 4X in media, and added 5 μΐ, and incubated at 37°C for 60 minutes. Final assay vehicle concentration was 1%. (4) Cells were lysed through incubation with 20 JL cAMP XS+ ED/CL lysis cocktail for one hour followed by incubation with 20 μΕ cAMP XS+ EA reagent for three hours at room temperature to generate signals. Microplates were read for chemiluminescence using the PerkinElmer EnvisionTM instrument.
Data were analyzed using CBIS data analysis suite (Chemlnnovation,
CA).
For A2A and A2B receptor agonist mode assays, percentage activity was calculated using the following formula:
% Activity -100% x (mean RLU of test sample— mean RLU of vehicle control) / (mean RLU of MAX control— mean RLU of vehicle control). For Al and A3 Gi agonist mode assays, percentage activity was calculated using the following formula:
% Activity = 100% x (1— (mean RLU of test sample— mean RLU of MAX control) / (mean RLU of vehicle control— mean RLU of MAX control)).
Hep-3B Cell proliferation assays were done in Crown Biosciences in Beijing, China.
Hep-3B cells (Crown Biosciences) were cultured in the media supplemented with 10% FBS at 37 °C , 5% CO2 and 95% humidity.
(1) Harvest cells and adjust cell concentrations to 5.56 * 104 cells/ml, and add 90μ1 cell suspensions to two 96-well plates (A and B) with the final cell density of 5x 103 cells/well. Incubate seeded plates at 37°C.
(2) Remove cell culture medium next day and replace with 90μ1 culture medium containing 1% serum. Starve the cells overnight.
(3) Dissolve the test article or reference controls with DMSO. Prepare 200x solution of test article (2 mM) using DMSO. Then, dilute DMSO solution 20-fold with culture medium containing 1% serum to 10x working solution. Prepare 10x reference control solution with culture medium containing 1%» serum. (4) Dispense ΙΟμΙ drug solution (10x) to each well (triplicate for each concentration) of the plate B. Incubate the test plates for 48h in the humidified incubator at 37 °C with 5% CO2.
(5) Equilibrate the plate and its contents at room temperature for approximately 30 minutes. Add a volume of CellTiter-Glo® Reagent equal to the volume of cell culture medium present in each well, mix contents for 2 minutes on an orbital shaker to induce cell lysis, and incubate at room temperature for 10 minutes to stabilize luminescent signal.
(6) Record luminescence using EnVision Multi Label Reader. (7) In order to calculate IC50, a dose-responsive curve will be fitted using nonlinear regression model with a sigmoidal dose response.
The formula of surviving rate is shown below, and the IC50 was automatically produced by GraphPad Prism 5.0.
The surviving rate (%) = (LumTest articie-LumMedium controi)/ (LumNone treated-
LumMedium control) X 100%.
[Table 1]
Figure imgf000043_0001
Mouse pharmacokinetics experiments were done in Crown Biosciences in Taican, China, using ICR male mice (DiscoverX). The compounds were formulated in appropriate solutions, and IV and oral (PO.) administration were performed. Blood samples were collected by cardiac puncture over 24 hr time course, and the compounds were quantified by analyzing on LC/MS/MS. The pharmacokinetic parameters were calculated using the software WinNonlin 5.3. [Table 2]
Figure imgf000044_0001
As shown in Table 1, the compounds of the present invention selectively activated Adenosine A3 receptor. Pharmacokinetic data showed that the compounds of the present invention demonstrated improved oral bioavailability but moderate Cm and Tl/2 (Table 2). The results indicate that the compounds of the present invention selectively activated Adenosine A3 receptor.

Claims

What is claimed is:
1. A compound selected from the group consisting of a compound of Formula 1 below, and pharmaceutically acceptable salts, a prodrug, a hydrate, a solvate, and an isomer thereof:
Figure imgf000045_0001
wherein,
Y is oxygen, sulfur, or carbon;
Xj is H, alkyl, RaRbNC(=O)- or HORc-, wherein said Ra and Rb are each independently hydrogen, alkyl, amino, haloalkyl, aminoalkyl, BOC-aminoalkyl, or cycloalkyl, or optionally fuse together to form a heterocyclic ring containing two to five carbon atoms, and said Rc is alkyl, amino, haloalkyl, aminoalkyl, BOC-aminoalkyl, or cycloalkyl;
X2 is H, hydroxyl, alkylamino, alkylamido or hydroxyalkyl;
X3 and X4 are each independently hydrogen, hydroxyl, amino, amido, azido, halo, alkyl, carboxy, nitrile, nitro, trifluoro, aryl, alkaryl, thio, thioester, thioether, -OCOPh or -OC(=S)OPh, or both X3 and X4 are optionally oxygens connected to >C=S to form a 5-membered ring, or X2 and X3 optionally form the ring of
Figure imgf000045_0002
wherein R' and R" represent independently an alkyl group;
X5 is H, cyano, Cr8 alkyl, C2-8 alkenyl, or C2-8 alkynyl;
Z is nitrogen or carbon; Ri is halogen, NRNR12, N(RN)ORN, NRNNRNR12, N3, NO, NO2, CHO, CN, -CH(=NRN), -CH=NNHRN, -CH=N(ORN), -CH(ORN)2, -C(=O)NRNR12, - C(=S)NRNR12, -C(=O)ORN, d-8 alkyl, C2-8 alkenyl, C2-8 alkynyl, C4-8 carbocyclylalkyl, C6-2o aryl, C2-20 heterocyclyl, heteroaryl, -C(=0)-C1 -8 alkyl, - S(O)N-C1-8 alkyl, aryl-Cr8 alkyl, OR11 or SRn, and said n is 0, 1 , or 2; and
R2 and R3 are each independently H, halogen, NRNR12, N(RN)ORH, NR NRNR12, N3, NO, NO2, CHO, CN, -CH(=NRN), -CH=NNHRN, - CH=N(ORN), -CH(OR")2, -C(=O)NRUR12, -C(=S)NR"R12, -C(=O)ORN, RN, OR1 1, or SR1 1,
wherein said 1 1 12
R and R are each independently H, CR8 alkyl, C2-8 alkenyl, C2-8 alkynyl, C3-8 carbocyclyl, C4-8 carbocyclylalkyl, aryl-C 8 alkyl, heterocyclyl- CR8 alkyl, C6-20 aryl, C2-20 heterocyclyl, heteroaryl, -C(=O)-CR8 alkyl, or -S(O)N- CR8 alkyl, or R1 1 and R12 optionally fuse together with a nitrogen to which they are both attached to form 3 to 7 membered heterocyclic ring, wherein any carbon atom of said heterocyclic ring is optionally replaced with -O-, -S(O)„- or -NRD-, wherein, optionally, said Cr8 alkyl, C2-8 alkenyl, C2-8 alkynyl, C3-8 carbocyclyl, C4-8 carbocyclylalkyl, aryl-C1 -8 alkyl, heterocyclyl-CR8 alkyl, C6-20 aryl, C2-20 heterocyclyl, and heteroaryl are each independently substituted with one or more halogen, hydroxyl, CN, N3, N(RD)2, NH(RD), NH2, C(O)N(RD)2, C(O)NH(RD), C(O)NH2, OC(O)N(RD)2, OC(O)NH(RD), OC(O)NH2, C(O)ORD, OC(O)RD, C(O)RD, OC(O)RD, S(O)NRD, S(O)2N(RD)2, S(O)2NH(RD), S(O)2NH2, ORD or RD; wherein RD is CR8 alkyl, C2-8 alkenyl, C2-8 alkynyl, C3-8 carbocyclyl, C4-8 carbocyclylalkyl, aryl-Ci-8 alkyl, heterocyclyl-C 8 alkyl, C6-2o aryl, C2-20 heterocyclyl, or heteroaryl.
2. The compound of claim 1,
wherein,
Y is oxygen;
X] is H, or RARBNC(=O)-, wherein said RA and RB are each independently hydrogen, Ci-8 alkyl, amino, haloalkyl, aminoalkyl, BOC-aminoalkyl, or cycloalkyl;
* X2 is H, hydroxyl, alkylamino, alkylamido or hydroxyalkyl; X3 and X4 are each independently hydrogen, hydroxyl, amino, amido, azido, halo, alkyl, carboxy, nitrile, nitro, trifluoro, aryl, alkaryl, thio, thioester, thioether, -OCOPh or -OC(=S)OPh;
X5 is H, cyano, Cr8 alkyl, C2-8 alkenyl, or C2-8 alkynyl;
Z is nitrogen or carbon;
Ri is halogen, NRUR12, N(Ru)ORu, NRuNRnR12, -CH(=NRn), - CH=NNHRn, -CH=N(ORn), -CH(ORn)2, -C(=O)NRnR12, or -C(=S)NRnR12; and
R2 and R3 are each independently H, halogen, NRUR12, N(Rn)OR", NRnNRnR12, -CH(=NRn), -CH=NNHRU, -CH=N(ORu), -CH(OR )2, - C(=O)NRnR12, or -C(=S)NRuR12,
1 1 1 9
wherein said R and R are each independently H, C 8 alkyl, C -8 alkenyl, C2-8 alkynyl, C3-8 carbocyclyl, C4-8 carbocyclylalkyl, aryl-Cr8 alkyl, heterocyclyl- Cr8 alkyl, C6-20 aryl, C2-20 heterocyclyl, heteroaryl, -C(=O)-Cr8 alkyl, or -S(O)n- Cr8 alkyl,
wherein, optionally, said C 8 alkyl, C2-8 alkenyl, and C2-8 alkynyl are each independently substituted with one or more halogen, hydroxyl, CN, or N3.
3. The compound of claim 1 ,
wherein,
Y is oxygen;
X] is RaRbNC(=O)-, wherein said Ra and Rb are each independently hydrogen or C1-8 alkyl;
X2 is H;
X3 and X4 are hydroxyl;
X5 is H or cyano;
Z is nitrogen or carbon;
Ri is NRnR12; and
R 2z and R 3J are each independently H or halogen, wherein said R 11 and R12 are each independently H or phenyl-C 8 alkyl.
4. The compound of claim 1, which is selected from the group consisting of:
(1) (2S,3S,4R,5S)-3,4-dihydroxy-5-(4-((3-iodobenzyl)amino)imidazo[2,l- f\ [ 1 ,2,4]triazin-7-yl)-N-methyltetrahydrofuran-2-carboxamide; (2) (2S,3S,4R,5R)-5-cyano-3,4-dihydroxy-5-(4-((3- iodobenzyl)amino)imidazo[2, 1 -fj[ 1 ,2,4]triazin-7-yl)-N-methyltetrahydrofuran-2- carboxamide; and
(3) (2S,3S,4R,5S)-3,4-dihydroxy-5-(4-((3-iodobenzyl)amino)pyrrolo[2,l- fj [ 1 ,2,4]triazin-7-yl)-N-methyltetrahydrofuran-2-carboxamide.
5. A pharmaceutical composition for preventing or treating cancer, inflammation, cardiac ischemia or reperfusion injury, comprising the compound of claim 1, and a pharmaceutically acceptable carrier.
6. The pharmaceutical composition of claim 5, which further comprises at least one additional pharmaceutical agent.
7. A method for preventing or treating cancer, inflammation, cardiac ischemia or reperfusion injury in a mammal, which comprises administering the compound of claim 1 to the mammal.
8. A method for preventing or treating cancer, inflammation, cardiac ischemia or reperfusion injury in a mammal, comprising the steps of:
(i) administering to the mammal in need thereof a first composition comprising the compound of claim 1 , and a pharmaceutically acceptable carrier; and
(ii) administering to the mammal in need thereof a second composition comprising at least one additional pharmaceutical agent comprising one or more selected from the group consisting of a chemotherapeutic agent, antiinflammatory agent, and an agent for treating cardiac ischemia and reperfusion injury.
9. The method of claim 8, wherein the first composition and second composition are administered simultaneously.
10. The method of claim 8, wherein the first composition and second composition are administered sequentially and in any order.
1 1. The use of the compound of claim 1 for the manufacture of a medicament for preventing or treating a cancer, inflammation, cardiac ischemia or reperfusion injury.
PCT/KR2013/008229 2012-09-14 2013-09-11 Compounds and compositions for modulating adenosine a3 receptor activity WO2014042433A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201261700924P 2012-09-14 2012-09-14
US61/700,924 2012-09-14

Publications (2)

Publication Number Publication Date
WO2014042433A2 true WO2014042433A2 (en) 2014-03-20
WO2014042433A3 WO2014042433A3 (en) 2014-05-08

Family

ID=50278812

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2013/008229 WO2014042433A2 (en) 2012-09-14 2013-09-11 Compounds and compositions for modulating adenosine a3 receptor activity

Country Status (1)

Country Link
WO (1) WO2014042433A2 (en)

Cited By (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8957078B2 (en) 2013-03-15 2015-02-17 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8969360B2 (en) 2013-03-15 2015-03-03 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9309250B2 (en) 2011-06-22 2016-04-12 Vertex Pharmaceuticals Incorporated Substituted pyrrolo[2,3-b]pyrazines as ATR kinase inhibitors
WO2016069825A1 (en) * 2014-10-29 2016-05-06 Gilead Sciences, Inc. Methods for the preparation of ribosides
US9340546B2 (en) 2012-12-07 2016-05-17 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9663519B2 (en) 2013-03-15 2017-05-30 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9670215B2 (en) 2014-06-05 2017-06-06 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9981975B2 (en) 2016-03-28 2018-05-29 Incyte Corporation Pyrrolotriazine compounds as tam inhibitors
US10160760B2 (en) 2013-12-06 2018-12-25 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US10251904B2 (en) 2015-09-16 2019-04-09 Gilead Sciences, Inc. Methods for treating arenaviridae and coronaviridae virus infections
CN109748921A (en) * 2017-11-03 2019-05-14 中国科学院上海药物研究所 Heterocycle compound, preparation method and its method for being used to prepare C- nucleoside analog of N- tertbutyloxycarbonyl protection
CN111233869A (en) * 2020-03-12 2020-06-05 杭州新博思生物医药有限公司 Novel compound for preparing Rudexilvir key intermediate and preparation method thereof
US10675296B2 (en) 2017-07-11 2020-06-09 Gilead Sciences, Inc. Compositions comprising an RNA polymerase inhibitor and cyclodextrin for treating viral infections
US10682368B2 (en) 2017-03-14 2020-06-16 Gilead Sciences, Inc. Methods of treating feline coronavirus infections
US10696679B2 (en) 2010-07-22 2020-06-30 Gilead Sciences, Inc. Methods and compounds for treating paramyxoviridae virus infections
US10836787B2 (en) 2017-05-01 2020-11-17 Gilead Sciences, Inc. Crystalline forms of (S)-2-ethylbutyl 2-(((S)-(((2R,3S,4R,5R)-5- (4-aminopyrrolo[2,1-f] [1,2,4]triazin-7-yl)-5-cyano-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)(phenoxy) phosphoryl)amino)propanoate
CN112321589A (en) * 2020-02-18 2021-02-05 山东科巢生物制药有限公司 Synthesis method of antiviral drug Reidesciclovir and intermediate thereof
US10988498B2 (en) 2009-09-21 2021-04-27 Gilead Sciences, Inc. Processes and intermediates for the preparation of 1′-substituted carba-nucleoside analogs
US11179394B2 (en) 2014-06-17 2021-11-23 Vertex Pharmaceuticals Incorporated Method for treating cancer using a combination of Chk1 and ATR inhibitors
WO2022012630A1 (en) 2020-07-17 2022-01-20 四川大学 Method for synthesizing c-nucleoside compound
US11464774B2 (en) 2015-09-30 2022-10-11 Vertex Pharmaceuticals Incorporated Method for treating cancer using a combination of DNA damaging agents and ATR inhibitors
US11491169B2 (en) 2020-05-29 2022-11-08 Gilead Sciences, Inc. Remdesivir treatment methods
US11613553B2 (en) 2020-03-12 2023-03-28 Gilead Sciences, Inc. Methods of preparing 1′-cyano nucleosides
US11660307B2 (en) 2020-01-27 2023-05-30 Gilead Sciences, Inc. Methods for treating SARS CoV-2 infections
US11701372B2 (en) 2020-04-06 2023-07-18 Gilead Sciences, Inc. Inhalation formulations of 1'-cyano substituted carba-nucleoside analogs
US11780844B2 (en) 2022-03-02 2023-10-10 Gilead Sciences, Inc. Compounds and methods for treatment of viral infections
US11814406B2 (en) 2020-08-27 2023-11-14 Gilead Sciences, Inc. Compounds and methods for treatment of viral infections
US11939347B2 (en) 2020-06-24 2024-03-26 Gilead Sciences, Inc. 1′-cyano nucleoside analogs and uses thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009132123A1 (en) * 2008-04-23 2009-10-29 Gilead Sciences, Inc. Carba-nucleoside analogs for antiviral treatment
WO2011035250A1 (en) * 2009-09-21 2011-03-24 Gilead Sciences, Inc. Processes and intermediates for the preparation of 1'-substituted carba-nucleoside analogs
WO2012012776A1 (en) * 2010-07-22 2012-01-26 Gilead Sciences, Inc. Methods and compounds for treating paramyxoviridae virus infections

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009132123A1 (en) * 2008-04-23 2009-10-29 Gilead Sciences, Inc. Carba-nucleoside analogs for antiviral treatment
WO2009132135A1 (en) * 2008-04-23 2009-10-29 Gilead Sciences, Inc. 1' -substituted carba-nucleoside analogs for antiviral treatment
WO2011035250A1 (en) * 2009-09-21 2011-03-24 Gilead Sciences, Inc. Processes and intermediates for the preparation of 1'-substituted carba-nucleoside analogs
WO2012012776A1 (en) * 2010-07-22 2012-01-26 Gilead Sciences, Inc. Methods and compounds for treating paramyxoviridae virus infections

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
SAMMY E. METOBO ET AL.: ''Practical synthesis of l' -substituted Tubercidin C-nucleoside analogs'' TETRAHEDRON LETTERS vol. 53, 2012, pages 484 - 486 *

Cited By (64)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10988498B2 (en) 2009-09-21 2021-04-27 Gilead Sciences, Inc. Processes and intermediates for the preparation of 1′-substituted carba-nucleoside analogs
US10696679B2 (en) 2010-07-22 2020-06-30 Gilead Sciences, Inc. Methods and compounds for treating paramyxoviridae virus infections
US11492353B2 (en) 2010-07-22 2022-11-08 Gilead Sciences, Inc. Methods and compounds for treating Paramyxoviridae virus infections
US9309250B2 (en) 2011-06-22 2016-04-12 Vertex Pharmaceuticals Incorporated Substituted pyrrolo[2,3-b]pyrazines as ATR kinase inhibitors
US9340546B2 (en) 2012-12-07 2016-05-17 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9650381B2 (en) 2012-12-07 2017-05-16 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US10787452B2 (en) 2012-12-07 2020-09-29 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9718827B2 (en) 2012-12-07 2017-08-01 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US11117900B2 (en) 2012-12-07 2021-09-14 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US11370798B2 (en) 2012-12-07 2022-06-28 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US10392391B2 (en) 2012-12-07 2019-08-27 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8969360B2 (en) 2013-03-15 2015-03-03 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9663519B2 (en) 2013-03-15 2017-05-30 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8957078B2 (en) 2013-03-15 2015-02-17 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US10160760B2 (en) 2013-12-06 2018-12-25 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US10815239B2 (en) 2013-12-06 2020-10-27 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US11485739B2 (en) 2013-12-06 2022-11-01 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9670215B2 (en) 2014-06-05 2017-06-06 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US10093676B2 (en) 2014-06-05 2018-10-09 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US10800781B2 (en) 2014-06-05 2020-10-13 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US11179394B2 (en) 2014-06-17 2021-11-23 Vertex Pharmaceuticals Incorporated Method for treating cancer using a combination of Chk1 and ATR inhibitors
US9949994B2 (en) 2014-10-29 2018-04-24 Gilead Sciences, Inc. Methods for treating Filoviridae virus infections
JP2017533903A (en) * 2014-10-29 2017-11-16 ギリアード サイエンシーズ, インコーポレイテッド Process for the preparation of riboside
US11344565B2 (en) 2014-10-29 2022-05-31 Gilead Sciences, Inc. Methods for the preparation of ribosides
US11266666B2 (en) 2014-10-29 2022-03-08 Gilead Sciences, Inc. Methods for treating Filoviridae virus infections
US10695357B2 (en) 2014-10-29 2020-06-30 Gilead Sciences, Inc. Methods for treating filoviridae virus infections
US9724360B2 (en) 2014-10-29 2017-08-08 Gilead Sciences, Inc. Methods for treating Filoviridae virus infections
TWI698444B (en) * 2014-10-29 2020-07-11 美商基利科學股份有限公司 Methods for the preparation of ribosides
CN107074902A (en) * 2014-10-29 2017-08-18 吉利德科学公司 The method for preparing ribonucleotide
AU2015339222B2 (en) * 2014-10-29 2018-11-22 Gilead Sciences, Inc. Methods for the preparation of ribosides
AU2021201474B2 (en) * 2014-10-29 2023-02-02 Gilead Sciences, Inc. Methods for the preparation of ribosides
US10251898B2 (en) 2014-10-29 2019-04-09 Gilead Sciences, Inc. Methods for treating Filoviridae virus infections
AU2019201232B2 (en) * 2014-10-29 2020-12-10 Gilead Sciences, Inc. Methods for the preparation of ribosides
TWI740546B (en) * 2014-10-29 2021-09-21 美商基利科學股份有限公司 Methods for the preparation of ribosides
WO2016069825A1 (en) * 2014-10-29 2016-05-06 Gilead Sciences, Inc. Methods for the preparation of ribosides
US11382926B2 (en) 2015-09-16 2022-07-12 Gilead Sciences, Inc. Methods for treating Arenaviridae and Coronaviridae virus infections
US10695361B2 (en) 2015-09-16 2020-06-30 Gilead Sciences, Inc. Methods for treating arenaviridae and coronaviridae virus infections
US11007208B2 (en) 2015-09-16 2021-05-18 Gilead Sciences, Inc. Methods for treating arenaviridae and coronaviridae virus infections
US10251904B2 (en) 2015-09-16 2019-04-09 Gilead Sciences, Inc. Methods for treating arenaviridae and coronaviridae virus infections
US11464774B2 (en) 2015-09-30 2022-10-11 Vertex Pharmaceuticals Incorporated Method for treating cancer using a combination of DNA damaging agents and ATR inhibitors
US9981975B2 (en) 2016-03-28 2018-05-29 Incyte Corporation Pyrrolotriazine compounds as tam inhibitors
US11260070B2 (en) 2017-03-14 2022-03-01 Gilead Sciences, Inc. Methods of treating feline coronavirus infections
US10682368B2 (en) 2017-03-14 2020-06-16 Gilead Sciences, Inc. Methods of treating feline coronavirus infections
US11597742B2 (en) 2017-05-01 2023-03-07 Gilead Sciences, Inc. Crystalline forms of (S)-2-ethylbutyl 2-(((S)-(((2R,3S,4R,5R)-5-(4-aminopyrrolo[2,1-f] [1,2,4]triazin-7-yl)-5-cyano-3,4-dihydroxytetrahydrofuran-2-yl)methoxy) (phenoxy) phosphoryl)amino)propanoate
US10836787B2 (en) 2017-05-01 2020-11-17 Gilead Sciences, Inc. Crystalline forms of (S)-2-ethylbutyl 2-(((S)-(((2R,3S,4R,5R)-5- (4-aminopyrrolo[2,1-f] [1,2,4]triazin-7-yl)-5-cyano-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)(phenoxy) phosphoryl)amino)propanoate
US11266681B2 (en) 2017-07-11 2022-03-08 Gilead Sciences, Inc. Compositions comprising an RNA polymerase inhibitor and cyclodextrin for treating viral infections
US10675296B2 (en) 2017-07-11 2020-06-09 Gilead Sciences, Inc. Compositions comprising an RNA polymerase inhibitor and cyclodextrin for treating viral infections
CN109748921B (en) * 2017-11-03 2022-03-22 中国科学院上海药物研究所 N-tert-butyloxycarbonyl protected heterocyclic compound, preparation method thereof and method for preparing C-nucleoside analogue by using N-tert-butyloxycarbonyl protected heterocyclic compound
CN109748921A (en) * 2017-11-03 2019-05-14 中国科学院上海药物研究所 Heterocycle compound, preparation method and its method for being used to prepare C- nucleoside analog of N- tertbutyloxycarbonyl protection
US11660307B2 (en) 2020-01-27 2023-05-30 Gilead Sciences, Inc. Methods for treating SARS CoV-2 infections
CN112321589B (en) * 2020-02-18 2022-05-03 山东科巢生物制药有限公司 Synthesis method of antiviral drug Reidesciclovir and intermediate thereof
CN112321589A (en) * 2020-02-18 2021-02-05 山东科巢生物制药有限公司 Synthesis method of antiviral drug Reidesciclovir and intermediate thereof
CN111233869A (en) * 2020-03-12 2020-06-05 杭州新博思生物医药有限公司 Novel compound for preparing Rudexilvir key intermediate and preparation method thereof
US11613553B2 (en) 2020-03-12 2023-03-28 Gilead Sciences, Inc. Methods of preparing 1′-cyano nucleosides
US11701372B2 (en) 2020-04-06 2023-07-18 Gilead Sciences, Inc. Inhalation formulations of 1'-cyano substituted carba-nucleoside analogs
US11903953B2 (en) 2020-05-29 2024-02-20 Gilead Sciences, Inc. Remdesivir treatment methods
US11491169B2 (en) 2020-05-29 2022-11-08 Gilead Sciences, Inc. Remdesivir treatment methods
US11939347B2 (en) 2020-06-24 2024-03-26 Gilead Sciences, Inc. 1′-cyano nucleoside analogs and uses thereof
WO2022012630A1 (en) 2020-07-17 2022-01-20 四川大学 Method for synthesizing c-nucleoside compound
US11814406B2 (en) 2020-08-27 2023-11-14 Gilead Sciences, Inc. Compounds and methods for treatment of viral infections
US11926645B2 (en) 2020-08-27 2024-03-12 Gilead Sciences, Inc. Compounds and methods for treatment of viral infections
US11845755B2 (en) 2022-03-02 2023-12-19 Gilead Sciences, Inc. Compounds and methods for treatment of viral infections
US11851438B2 (en) 2022-03-02 2023-12-26 Gilead Sciences, Inc. 1′-cyano nucleoside analogs and methods for treatment of viral infections
US11780844B2 (en) 2022-03-02 2023-10-10 Gilead Sciences, Inc. Compounds and methods for treatment of viral infections

Also Published As

Publication number Publication date
WO2014042433A3 (en) 2014-05-08

Similar Documents

Publication Publication Date Title
WO2014042433A2 (en) Compounds and compositions for modulating adenosine a3 receptor activity
TWI663169B (en) Cdk2/4/6 inhibitors
WO2014035140A2 (en) Compounds and compositions for modulating histone methyltransferase activity
EP3129021B1 (en) Treatment of b-cell malignancies by a combination jak and pi3k inhibitor
EP3423443B1 (en) Cyano-substituted indole compounds and uses thereof as lsd1 inhibitors
US9580432B2 (en) Fused pyrimidine compound or salt thereof
KR20200036922A (en) Selective inhibitor of protein arginine methyltransferase 5 (PRMT5)
JP6404332B2 (en) Novel 3- (1H-pyrazol-4-yl) -1H-pyrrolo [2,3-c] pyridine derivatives as NIK inhibitors
JP6769000B2 (en) A novel compound of 4&#39;-thionucleoside, its preparation method, its pharmaceutical composition and its use
US10604502B2 (en) Substituted 5-cyanoindole compounds and uses thereof
ES2910071T3 (en) Aminopyrazine diol compounds as PI3K-Y inhibitors
JP5890043B2 (en) Novel acetate of 2-deoxy monosaccharide having anticancer activity
JP2016538281A (en) Protein phosphatase inhibitors that cross the blood-brain barrier
CN101534835B (en) L-OddC prodrugs for cancer
US9724331B2 (en) Use of maleimide derivatives for preventing and treating leukemia
JP2022521491A (en) Selective inhibitor of protein arginine methyltransferase 5 (PRMT5)
EP4174068A1 (en) Novel compound, and pharmaceutical composition for preventing or treating resistant cancer, comprising same
RU2530887C2 (en) Quinazoline compounds
CN117529321A (en) Combination of ERK inhibitor and KRAS inhibitor and use thereof
CN108368117B (en) Substituted imidazoquinazoline compound and pharmaceutical composition thereof
US20220281910A1 (en) Dinucleotide compounds for treating cancers and medical uses thereof
RU2710013C2 (en) Polymorphic forms of icotinib and uses thereof
ES2248351T3 (en) HALOGENUROS OF 7,9-GUANINIO DISUSTITUTED AS INHIBITORS OF TOLEMERASA.
TW202045162A (en) Methods of inhibiting kinases
EA043542B1 (en) HEMISULFATE SALT OF NUCLEOTIDE FOR TREATMENT OF HEPATITIS C VIRUS

Legal Events

Date Code Title Description
32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC (EPO FORM 1205 DATED 03.07.2015)

122 Ep: pct application non-entry in european phase

Ref document number: 13836662

Country of ref document: EP

Kind code of ref document: A2