WO2005012323A2 - Compounds useful as a3 adenosine receptor agonists - Google Patents

Compounds useful as a3 adenosine receptor agonists Download PDF

Info

Publication number
WO2005012323A2
WO2005012323A2 PCT/GB2004/003283 GB2004003283W WO2005012323A2 WO 2005012323 A2 WO2005012323 A2 WO 2005012323A2 GB 2004003283 W GB2004003283 W GB 2004003283W WO 2005012323 A2 WO2005012323 A2 WO 2005012323A2
Authority
WO
WIPO (PCT)
Prior art keywords
ring
product
formula
cycle
compound
Prior art date
Application number
PCT/GB2004/003283
Other languages
French (fr)
Other versions
WO2005012323A3 (en
Inventor
Robin Havard Davies
Original Assignee
Trigen Limited
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 Trigen Limited filed Critical Trigen Limited
Publication of WO2005012323A2 publication Critical patent/WO2005012323A2/en
Publication of WO2005012323A3 publication Critical patent/WO2005012323A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/23Heterocyclic radicals containing two or more heterocyclic rings condensed among themselves or condensed with a common carbocyclic ring system, not provided for in groups C07H19/14 - C07H19/22
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/16Purine radicals

Definitions

  • D is N or CH
  • R c is hydrogen or alkyl
  • X 2 is mono-N- or di- ,N((C ⁇ -C4) alkylaminocarbonyl, mono-N- or di-, N-(C3-C5) cycloalkylaminocarbonyl or N-(C ⁇ -C4) alkyl-N- (C3-C5) cycloalkylaminocarbonyl and especially mono-N-(C ⁇ -C4) alkylaminocarbonyl;
  • halogen herein includes reference to F, CI, Br and I, of which CI is often preferred.
  • D is N or CH
  • D and R 2 are as defined previously (e.g D may be N and R 2 CI or other halogen);
  • E is 0, S or CH2 (e.g. E is O and optionally D is N and R 2 is CI or other halogen); R 1 is C1-C4 alkyl; and X 3a is -OH or -NH 2 .
  • any ring A shown above may be fused with any ring B shown above (e.g. ring A of structure 5 may be fused with ring B of structures 2, 6 or 10), e.g. to form a 5/6 (ring A is 5- membered, ring B 6-membered), 6/6 or 6/5 fused ring.
  • Structures 7 and 8 are particularly preferred CYCLE moieties. Accordingly, a class of compounds have a CYCLE moiety of formula (IX)
  • the above compounds may be used to treat malignant hypotension.
  • the administration of IB-MECA results in a significant increase (e.g., about 10-30%) in basal or systemic blood pressure (e.g., from about 70 mm Hg to about 90 mm Hg).
  • Another aspect of this invention is directed to methods for treating arteriosclerosis comprising administering to a mammal a therapeutically effective amount of a product of the invention.
  • Another aspect of this invention is directed to methods for treating angina pectoris comprising administering to a mammal a therapeutically effective amount of a product of the invention.
  • the compounds of the invention have the advantage that they may be more efficacious, be less toxic, be longer acting, have a broader range of activity, be more potent, produce fewer side effects, be more easily absorbed than, or that they may have other useful pharmacological properties over, compounds known in the prior art.
  • the 2 iodo uronamide is then reacted with triethylamine, bis(triphenylphosphine)palladium dichloride and Cul in catalytic amount using as solvent a mixture of acetonitrile/DMF 2:1 and to this mixture is added the terminal alkyne and the reaction takes place under N2 atmosphere at room temperature (ref J. Med. Chem. 1995, vol 38, 1462-1472) to result in compound E.
  • the coupling of the benzoxazole reactant with the 6-chloro derivative is then accomplished as described above.
  • the alkyne used in the phenyl alkyne; other alkynes can be used in analogous manner.
  • the alkenyl compounds may be prepared analogously.

Landscapes

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

Abstract

Compounds useful as A3 Adenosine Receptor Agonists. Adenosine analogue-type A3 receptor agonists having an N6 substituent of the formula CR20R21CYCLE where CYCLE is a specified heterocycle, e.g. a substituted pyridyl group or a substituted oxazolyl-containing bicyclic ring. 10

Description

TITLE OF THE INVENTION
COMPOUNDS USEFUL AS A3 ADENOSINE RECEPTOR AGONISTS
BACKGROUND OF THE INVENTION
This invention relates to compounds useful as A3 adenosine receptor agonists and methods of selectively activating an A3 adenosine receptor in a mammal, particularly a human. The present invention also relates to methods of treating various medical disorders with A3 receptor agonists, in particular post-infarct patients, patients with severe angina and related cardiovascular disorders.
Adenosine, an endogenous purine nucleoside, is ubiquitous in mammalian cell types. Adenosine present in the plasma and other extracellular fluids mediates many of its physiological effects via cell surface receptors and is an important regulatory species. Adenosine has the formula:
Figure imgf000002_0001
Adenosine receptors are generally divided into three major subclasses, Al, A2 and A3, on the basis of the differential affinities of a number of adenosine receptor agonists and antagonists for the receptors, their primary structures and the secondary messenger systems to which they couple.
Various adenosine A3 receptor agonists and uses therefor are taught in the prior art.
WO 95/02604 (the contents of which are incorporated herein by reference) discloses A3 adenosine receptor agonists and their use as locomotor depressants, hypotensive agents, anxiolytic agents, cerebroprotectants and antiseizure agents.
US 5573772 and related US 5443836 claim the use of adenosine A3 agonists for applications where ischaemic preconditioning is beneficial, for example cardioprotection. WO 98/50047 and WO 99/20284 also relate to ischaemic protection. WO 98/50047 claims methods of administering a compound having A3 agonist activity and a compound (whether the same compound or a different one) having Al agonist activity or A2 antagonist activity. WO 99/20284 claims a method for preventing or reducing ischaemic heart damage by administration of at least two cardioprotectants, of which one may be an A3 agonist.
WO 01/19360 claims the use of A3 receptor agonists to achieve the following effect: induce G-CSF secretion • induce proliferation or differentiation of bone marrow or white blood cells prevent or treat leukopenia prevent or treat toxic side effects of a drug (e.g. drug-induce leukopenia or weight loss) inhibiting abnormal cell growth treating cancer.
WO 01/083152 relates to the use of adenosine A3 receptor agonists to activate natural killer (NK) cells whilst WO 02/055085 teaches their use to inhibit viral replication.
WO 02/066020 proposes the use of adenosine A3 receptor agonists to modulate the activity of glycogen synthase kinase 3β.
Adenosine receptor ligands are described in the following documents: US 6048865 (Al ligands) WO 01/60835 (A2 antagonists) • WO 00/23447 (Al and A2 ligands) WO 92/05177 (A1/A2 agonists) WO 92/05177 (A1/A2 agonists) WO 95/28160 (A1/A2 agonists) EP 277917 (A2 ligands) • WO 86/00310 (A2 ligands) EP 1241176 (A3 agonists) WO 01/23399 (A3 agonists) WO 02/055521 (A2a antagonists) WO 93/23418 (A1/A2 antagonists) • WO 95/07921 (Al agonists) WO 98/16539 (Al ligands) WO 02/055085 (A3 agonists) WO 96/12496 (Al agonist). The article "Adenosine Receptor Ligands-Recent Developments Part I. Agonists", C.E. Muller, Current Medicinal Chemistry 2000, 7, 1269-1288 reports on the developments in the field of adenosine receptor agonists, including A3 receptor agonists.
The above publications are all included herein by reference. The art therefore includes adenosine receptor agonists which are adenosine analogues characterised by specific variations which make the compounds capable of binding to and acting on one or more adenosine receptors. More particularly, the skilled person knows that there exists a class of adenosine analogue-type A3 receptor agonists.
Adenosine analogue-type A3 receptor agonists are familiar to the skilled reader and will require no further explanation to the skilled reader. Nonetheless, it may be of assistance to describe that adenosine analogue-type A3 receptor agonists may have an N6 nitrogen which may be identified with the N6 nitrogen of adenosine and is usually substituted by at least one substituent. Such agonists include without limitation compounds of the formula (I):
Figure imgf000004_0001
wherein D is N or CH;
E is O, S or CH2;
X1 is an N6 substituent;
X2 (the 4' substituent) is hydroxymethyl, (Cι-C3)alkoxymethyI, (C3-C5)cycloalkoxy methyl, carboxy, (Cι-C3)alkoxycarbonyl, (C3-C5)cycloalkoxycarbonyl, 1,1-aminoiminomethyl, l,l-(mono- N- or di-N,N-(Cι-C4)aIkylamino)iminomethyI, l,l-(mono-N- or di-N,N-(C3-C5)cycloalkyl- amino)iminomethyl, carbamoyl, mono-N- or di-N,N-(Cι-C4)alkylaminocarbonyl, mono-N- or di- N,N-(C3-C5)cycloalkylaminocarbonyl or -(Cι-C4)alkyl-N-(C3-C5)cycloalkylaminocarbonyl;
X3 and X4 are each independently hydrogen, alkyl, hydroxyalkyl, alkoxyalkyl, ORa NRaRb, where
Ra and R^ are independently hydrogen (most preferably X3 and X4 are OH), alkyl, aralkyl, carbamoyl, alkyl carbamoyl, dialkylcarbamoyl, acyl, alkoxycarbonyl, aralkoxycarbonyl, aryloxycarbonyl, or, when X3 and X4 are both ORa, the two Ra groups together may form
Figure imgf000005_0001
where Rc is hydrogen or alkyl,
Figure imgf000005_0002
where R0" and Re are independently hydrogen, alkyl, or together with the carbon atom to which they are attached may form a 1,1-cycloalkyl group;
X5 is H, halogen, (Cι-Cιg)alkyl, fluorinated (Ci-C^g) alkyl (e.g. trifluoromethyl), (C^-C^Q) alkoxyalkyl, (Cj-Cinjal oxy, (Ci-Cjo alkylether, (Cj_-Cj_o)thioalkoxy,
Figure imgf000005_0003
amino,
(C1-Cιo)alkylamino, -COX6R25 where X6 is 0 or NH and R25 is (Cι-C4)alkyl optionally terminally substituted by an aryl or a heteroaryl group [for example phenyl or a 5- or 6-membered heteroaryl group] and additionally or alternatively terminally substituted by hydroxy, C2- Cιo)alkenyl, (C2-Cιg)alkynyl, or is ^-Cjnjalkenyl or (C2-Cιo)alkynyl in either case terminally substituted by an aryl or heteroaryl group [for example phenyl or a 5- or 6-membered heteroaryl group] and, when having a terminal methylic carbon atom, optionally further terminally substituted by hydroxy. Alkyl groups comprised in X5 substituents are preferably linear.
Preferred values for the above-defined symbols are as follows: D is N; E is O;
X2 is mono-N- or di- ,N((Cι-C4) alkylaminocarbonyl, mono-N- or di-, N-(C3-C5) cycloalkylaminocarbonyl or N-(Cι-C4) alkyl-N- (C3-C5) cycloalkylaminocarbonyl and especially mono-N-(Cι-C4) alkylaminocarbonyl;
X3 is OH or NH2;
X4 is OH; X5 is H, halogen, ((Ci-C^g alkyl and especially (C1-C4) alkyl, trifluoromethyl, (C2-C10) alkenyl,
(c2"clθ) alkynyl or either of the latter two groups where terminally substituted as described above, X5 more preferably being H, chloro, bromo, iodo, (C1-C4) alkyl and especially methyl, or trifluoromethyl.
WO 03/061670 describes adenosine analogue-type A3 receptor agonist having an N6 nitrogen substituted by a group of the formula -CR20R21-CYCLE, where R2u and R21 are the same or different and H, F or CH3 and CYCLE is: (I) a 2-pyridyl, or an analogue thereof in which the C3 and/or C5 carbon atoms are replaced by nitrogen, optionally substituted at the 4-position with CH3, 1, Br, CI, CF3, OH or H2 and/or at the 6-position by OR11, CO2R11, COR11 or CONR11 where R11 is C2-C4 alkyl; or (II) A bicyclic (fused) heteroaromatic ring of the formula
Figure imgf000006_0001
wherein ring A is a 5- or 6- membered ring characterised by the following features (in which ring positions are numbered relative to the linkage to -CR20R21-): i. a carbon atom at the 1-position; ii. carbon atom as CH or a nitrogen atom at position 2; iii. it is 3, 4 fused to ring B; iv. the 5-position ring atom is substituted by a moiety R5 which is H, CH3, 1, Br, CI, CF3 or less preferably OH or NH2- v. if a 6-membered ring, it has at the 6-position a nitrogen, or -CM- where M is H, CH3 or F; ring B is a 5 or 6 membered ring characterised by the following features:
(a) an in-ring heteroatom including O, N or S joined to the 4-position of ring A;
(b) said in-ring heteroatom is joined within the ring secondly to a carbon which is substituted by a moiety R^ which is H or another moiety wherein the number of atoms which are not hydrogen or halogen is no more than 10;
(c) an in-ring atom joined to the 3-position of ring A which is N,0, or less preferably S or C, said C being in the form of a CH or CO group;
(d) in the case of a 6-membered ring, the remaining ring member is nitrogen or carbon in the form of CH. It will be appreciated that any one or more of D, E, X2, X3, X4 and X5 may be one of the preferred species listed above; most desirably all are preferred.
BRIEF SUMMARY OF THE INVENTION
The present invention provides in one aspect an adenosine analogue-type A3 receptor agonist having an N6 nitrogen substituted by a group of the formula -CR20R21-CYCLE where R20 and R21 are the same or different and H, F or CH3; and CYCLE is of formula (II):
Figure imgf000007_0001
where: R5 is iodine, bromine, methyl or trifluoromethyl; R8 is -NR9R10 or -COR9, where R9 and R*0 are each independently methyl or ethyl; and W is N or CH; or CYCLE is of formula (III):
Figure imgf000007_0002
R5 where: R5 is iodine, bromine, methyl or trifluoromethyl; and R7 is H, halogen, CI~CIQ acyl, OR11, CO2R11 or CONR11 where R11 is
Figure imgf000007_0003
hydrocarbyl optionally containing one or more in-chain and/or in-ring -O- linkages.
The present invention provides in a further aspect an adenosine analogue-type A3 receptor agonist comprising a purine residue or purine analogue residue which in either case is 2- substituted by halogen, e.g. bromine or especially chlorine, and having an N6 nitrogen substituted by a group of the formula -CR20R21-CYCLE, where CYCLE is of formula (I) or formula (II) above.
Included also are adenosine analogue-type A3 receptor agonists having an N6 nitrogen substituted by a group of the formula -CR20R21-CYCLE where R20 and R21 are the same or different and H, F or CH3; and CYCLE is a bicyclic (fused) heteroaromatic ring of the formula
Figure imgf000008_0001
wherein ring A is a 5- or 6- membered ring characterised by the following features (in which ring positions are numbered relative to the linkage to -CR20R2:1~): i. a carbon atom at the 1-position; ii. carbon atom as CH or a nitrogen atom at position 2; iii. it is 3, 4 fused to ring B; iv. the 5-position ring atom is substituted by a moiety R5 which is H or an organic moiety having from 1 to 6 plurally valent atoms in addition to monovalent atoms selected from hydrogen and halogen. v. if a 6-membered ring, it has at the 6-position a nitrogen, or -CM- where M is H, CH3 or F; ring B is a 5 or 6 membered ring characterised by the following features:
(a) an in-ring heteroatom including 0, N or S joined to the 4-position of ring A;
(b) said in-ring heteroatom is joined within the ring secondly to a carbon which is substituted by a moiety R8 which is -N(C2H5 2; (c) an in-ring atom joined to the 3-position of ring A which is N,0, S or C, said C being in the form of a CH or CO group;
(d) in the case of a 6-membered ring, the remaining ring member is nitrogen or carbon in the form of CH.
Included, therefore, are compounds of formula (I) in which X1 is a -CR20R21-CYCLE group as described herein.
As discussed below, the products of the invention include any compound capable of resulting in the delivery of such agonists to adenosine A3 receptors in vivo and include, therefore, salts and prodrugs of such agonists as well as the salts of such prodrugs. In this application the term "product of the invention" is to be understood accordingly.
The invention includes but is not limited to adenosine-5'-uronamides which are N6- monosubstituted by -CR20R2:1~CYCLE. T e uronamides may be ethyl or methyl uronamides. The adenosine-5'-uronamides may, for example, be 2-substituted by, amongst others, a small substituent such as CI, Br, I, CH3 or CF3. The present invention also provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and an effective amount, e.g. a therapeutically effective amount, including a prophylactically effective amount, of one or more products of the invention.
In addition, the present invention provides a method of selectively activating A3 adenosine receptors in a mammal, which method comprises acutely or chronically administering to a mammal in need of selective activation of its A3 adenosine receptors a therapeutically effective amount, including a prophylactically effective amount, of one or more products of the invention.
Throughout the description and claims of this specification, the words "comprise" and "contain" and variations of the words, for example "comprising" and "comprises", mean "including but not limited to", and are not intended to (and do not) exclude other moieties, additives, components, integers or steps.
DETAILED DESCRIPTION OF THE INVENTION
The present invention includes adenosine A3 receptor agonists having a normally mono- substituted N6 nitrogen wherein the substituent is -CR2uR21-CYCLE where R20 and R21 are the same or different and are H, F and CH3 and CYCLE is: (I) an oxazolyl moiety selected from benzoxazolyl and pyridyloxazolyl moieties and having a specified substituent pattern described below; (II) a pyridyl moiety having a specified substituent pattern described below; or (III) a bicyclic moiety as described below.
Group -CR20R21-CYCLE therefore corresponds to substituent X1 in relation to the compounds of formula (I).
Preferably the -CR 0R21- is -CH2-.
Glossary
The term "alkyl" in this specification includes linear and branched alkyl groups, for example methyl, ethyl, n-propyl, iso-propyl, tert-butyl, n-pentyl and n-hexyl. Similarly, the term "alkoxy" includes groups of which the alkyl part may be linear or branched, for example one of those groups listed in the preceding sentence; alkylene groups may likewise be linear or branched and may, for example, correspond to one of those alkyl groups listed in the preceding sentence. The alkyl groups may be (but preferably are not) interrupted by one or more ether linkages. Where not otherwise specified, the term "alkyl" includes 1C-10C alkyl, e.g. IC, 2C, 3C or 4C alkyl; this applies equally to the alkyl part of larger moieties, for example alkoxy. Similarly, the terms "alkenyl" and "alkynyl" include 2C-10C moieties, e.g. 2C, 3C or 4C.
The term "halogen" herein includes reference to F, CI, Br and I, of which CI is often preferred.
Oxazolyl N6 Compounds
The oxazolyl compounds have a group CYCLE of formula (II):
Figure imgf000010_0001
where:
R5 is iodine, bromine, methyl or trifluoromethyl;
R8 is -NR9R10 or -COR9, where R9 and R10 are each independently methyl or ethyl; and W is N or CH.
In one class of compounds, W is N. In another class of compounds, W is CH.
Included is a class of compounds in which R8 is dimethylamino and a class of compounds in which R8 is diethylamino.
In embodiments of the compounds, R8 is -COR9. In a sub-class of these compounds, R9 is acetyl.
One class of compounds comprises those in which R8 is diethylamino or -COR9.
The invention therefore includes compounds having the following CYCLE moieties:
Figure imgf000010_0002
(1) (2) (5)
Figure imgf000011_0001
(4) (5) (6)
Figure imgf000011_0002
(12) (13) (14)
Figure imgf000011_0003
(15) (16) (17)
Figure imgf000011_0004
(18) (19)
Figure imgf000011_0005
Figure imgf000012_0001
Included is an embodiment in which CYCLE is of formula 1, 2, 3 or 4; other compounds are those in which CYCLE is of formula 5, 6 or 7; further compounds are those in which CYCLE is of formula 8, 9, 10 or 11. In certain compounds, CYCLE is of formula 12, 13, 14 or 15; in certain others it is of formula 16, 17, 18 or 19, but in yet other compounds CYCLE is of formula 20, 21, 22 or 23.. In some of the compounds mentioned in this paragraph, X^ is H whereas in others it is halogen, for example chlorine.
Pyridyl N6 Compounds
The pyridyl compounds have a group CYCLE of the formula (III):
Figure imgf000012_0002
R5 where:
R5 is iodine, bromine, methyl or trifluoromethyl; and
R7 is H, halogen, Cj-Ci acyl, OR11, CO2 11 or CONR11 where R11 is Cι-C10 hydrocarbyl optionally containing one or more in-chain and/or in-ring -0- linkages.
One class of compounds comprise R7 groups which are Ci-C^g acyl wherein acyl is -COR12 and R 2 is hydrocarbyl optionally containing one or more in-chain and/or in-ring -O- linkages.
Where R7 is C^-C^g acyl, OR11, CO2R11 or CONR11, the acyl or R11 group may contain from 1 to 6 carbon atoms. Included in the invention are compounds in which acyl has 2, 3, 4 or 5 carbon atoms. Also included are compounds in which R11 has 1, 2, 3 or 4 carbon atoms. R11 and R12 may be a straight chain. R11 and R12 may contain one or more (e.g. one) carbon- carbon double or triple bonds, whether or not they also contain any -0- linkages. In some embodiments, R11 and R12 contain one carbon-carbon double bond.
Accordingly, one class of compounds has an R7 group which is -COR12 wherein R12 is Cj-Cjg hydrocarbyl optionally containing one or more -O- linkages and/or having one or more unsaturated carbon-carbon linkages. In some of these compounds, R12 has 1, 2, 3 or 4 carbon atoms. The invention includes a class of compounds in which R12 is alkyl or alkenyl, e.g. methyl, ethyl or peπtenyl; a particular sub-class has ethyl as R 2; another subclass has alkenyl as R12, for example CnH nCH=CH2 (in which n is from 1 to 8, e.g. 1, 2, 3 or 4), of which C2H4CH=CH2 may be mentioned.
The invention therefore includes compounds having the following CYCLE moieties:
Figure imgf000013_0001
Figure imgf000014_0001
T e structure of remainder of the molecule is not critical. The invention includes compounds of formula (I) in which X1 is -CR20R21-CYCLE as described above, e.g. -CH2-CYCLE.
Included is an embodiment in which CYCLE is of formula 29, 30, 31 or 32; other compounds are those in which CYCLE is of formula 33, 34, 35 or 36; further compounds are those in which CYCLE is of formula 37, 38, 39 or 40. In some of the compounds mentioned in this paragraph, R2 is H whereas in others it is halogen, for example chlorine.
The compounds of the invention may comprise a purine or purine analogue ring which is 2- substituted by halogen, e.g. bromine or especially chlorine. Accordingly, the invention includes compounds comprising an N6-substituted purine (analogue) moiety of formula (IV):
Figure imgf000014_0002
where:
-CR20R21-CYCLE is as defined previously;
D is N or CH; and
R2 is H or halogen, e.g. bromine or chlorine (e.g D may be N and R2 CI or other halogen).
In one class of structure (IV) compounds, -CR20R21-is -CH2--
Whether or not -CR20R 1-is -CH2-, D is N in one embodiment. The invention includes a class of compounds in which R2 is H. The invention includes a class of compounds in which R2 is halogen. The invention includes a class of compounds in which R2 is chlorine.
The invention includes variants of formula (IV) in which R2 is any X5 moiety as defined with reference to formula (I).
A class of compounds are of formula (V):
Figure imgf000015_0001
X3a OH where:
-CR20R21-CYCLE, D and R2 are as defined previously (e.g D may be N and R2 CI or other halogen);
E is 0, S or CH2 (e.g. E is O and optionally D is N and R2 is CI or other halogen); R1 is C1-C4 alkyl; and X3a is -OH or -NH2.
In some embodiments E is O.
A sub-class of formula (V) compounds has methyl or ethyl, particularly methyl, as R1.
The invention includes compounds of formula (VI)
Figure imgf000015_0002
where CYCLE is a group of formula (II), e.g., selected from structures 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22 and 23.
The invention includes compounds of formula (VI) where CYCLE is a group of formula (III), e.g., selected from structures 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39 and 40.
The invention includes compounds of formula (VII)
Figure imgf000016_0001
where CYCLE is a group of formula (II), e.g., selected from structures 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22 and 23.
The invention includes compounds of formula (VII) where CYCLE is a group of formula (III), e.g., selected from structures 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39 and 40.
Other compounds of the invention are those of formulae (VI) and (VII) in which the 3' -OH group is replaced by another X3 or X3a group, notably -NH2.
One class of compounds excludes the compound:
Figure imgf000016_0002
Another class of compounds excludes compounds of the formula:
Figure imgf000017_0001
A further class of compounds excludes compounds of the formula
Figure imgf000017_0002
A yet further class excludes compounds having an N6 substituent which is
Figure imgf000017_0003
Another class comprises CYCLE group of formula (II) in which W is N, and R^ and R8 are both as defined above in relation to formula (II), or in which W is CH and R5 is iodine, bromine, methyl or trifluoromethyl and R8 is diethylamino or -COR9, for example is -COR9,
R8 is -NR9R10 or -COR9, where R9 and R10 are each independently methyl or ethyl; and W is N or CH.
In one class of compounds, W is N. In another class of compounds, W is CH.
Included is a class of compounds in which R8 is dimethylamino and a class of compounds in which R8 is diethylamino.
In embodiments of the compounds, R8 is -COR9. In a sub-class of these compounds, R9 is acetyl.
One class of compounds comprises those in which R8 is diethylamino or -COR9. In embodiments of the invention, the compounds of the invention exclude compounds of the formula
Figure imgf000018_0001
where CYCLE is of formula 24 or 27. In other embodiments, there are excluded compounds of the above formula in which CYCLE is of formula 24, 25, 27 or 28.
The invention also includes embodiments in which there are excluded compounds of the formula:
Figure imgf000018_0002
where CYCLE is of formula 24 or 27. In other embodiments, there are excluded compounds of the above formula in which CYCLE is of formula 24, 25, 27 or 28.
The invention additionally includes embodiments in which there are excluded compounds of the formula:
Figure imgf000018_0003
where CYCLE is of formula 24 or 27. In other embodiments, there are excluded compounds of the above formula in which CYCLE is of formula 24, 25, 27 or 28. A class of compounds excludes compounds having an N6 substituent which is -CR2DR21-CYCLE wherein CYCLE is of formula 24 or 27. In other embodiments, there are excluded compounds of the aforesaid formula in which CYCLE is of formula 24, 25, 27 or 28.
The Bicyclic N6 Substituent Compounds
The compounds described in this section are adenosine analogue-type A3 receptor agonists having an N6 nitrogen substituted by a group of the formula -CRR21-CYCLE where R2u and R21 are the same or different and H, F or CH3; and CYCLE is a bicyclic (fused) heteroaromatic ring of the formula
Figure imgf000019_0001
wherein ring A is a 5- or 6- membered ring characterised by the following features (in which ring positions are numbered relative to the linkage to -CR20R21-): i. a carbon atom at the 1-position; ii. carbon atom as CH or a nitrogen atom at position 2; iii. it is 3, 4 fused to ring B; iv. the 5-position ring atom is substituted by a moiety R5 which is (a) H or halogen, or (b) an organic moiety having from 1 to 6 plurally valent atoms in addition to monovalent atoms selected from hydrogen and halogen. v. if a 6-membered ring, it has at the 6-position a nitrogen, or -CM- where M is H, CH3 or F; ring B is a 5 or 6 membered ring characterised by the following features:
(a) an in-ring heteroatom including O, N or S joined to the 4-position of ring A;
(b) said in-ring heteroatom is joined within the ring secondly to a carbon which is substituted by a moiety R8 which is -H O^Λ^)-,
(c) an in-ring atom joined to the 3-position of ring A which is N,0, S or C, said C being in the form of a CH or CO group;
(d) in the case of a 6-membered ring, the remaining ring member is nitrogen or carbon in the form of CH.
R5 may contain from 1 to 4 plurally valent atoms. Included are compounds in which the plurally valent atoms are selected from carbon, oxygen, sulfur and nitrogen, for example R5 may be CH3, CF3, OH or NH2. Other moieties containing such plurally valent atoms are nitro and thiol, for example. A class of the bicyclic N6 substituent compounds have R5 groups which are not H or CI; a larger class of excluded R5 groups additionally includes OH and H2. Included is a class of compounds in which R5 is a monovalent atom, for example H, I, Br or CI.
In a class of compounds, R20 and R21 are both hydrogen.
As examples of CYCLE moieties may be mentioned the following, in which R8 is diethylamino:
) 2)
Figure imgf000021_0001
Figure imgf000021_0002
In the above structures R^a comprises a plurally valent atom directly bonded to the ring, e.g. it is CH3 or less preferably CF3_
In principle, any ring A shown above may be fused with any ring B shown above (e.g. ring A of structure 5 may be fused with ring B of structures 2, 6 or 10), e.g. to form a 5/6 (ring A is 5- membered, ring B 6-membered), 6/6 or 6/5 fused ring. Structures 7 and 8 are particularly preferred CYCLE moieties. Accordingly, a class of compounds have a CYCLE moiety of formula (IX)
Figure imgf000022_0001
where W is N or CH.
Included are compounds in which: D is N; E is O;
X2 is mono-N- or di-N,N(Cι-C4)alkylaminocarbonyl, mono-N- or di-, N-(C3-C5)cycloalkylaminocarbonyl or N-(Cι-C4)aikyl-N- (C3-C5)cycloaIkylaminocarbonyl;
X3 is OH or NH2;
X4 is OH; X5 is H, halogen, (Cι-Cι )alkyl, trifluoromethyl, (C2"Ci )alkenyl, (C2-Cιg)alkynyl, or either of the latter two groups where terminally substituted as defined previously.
As other classes of compounds may be mentioned those of formula (X) and (XI):
Figure imgf000022_0002
Figure imgf000023_0001
where CYCLE is a bicyclic heteroaromatic ring as disclosed above.
The disclosed compounds can exist in different forms, such as salts and esters, for example, and the invention includes all variant forms of the compounds. In particular, the compounds may be in the form of acid addition salts which, for those compounds for pharmaceutical use, will be pharmaceutically acceptable. Exemplary acids include HBr, HCI and HSO2CH3.
Certain compounds of the invention exist in different tautomeric forms and the invention includes all such tautomers. Compounds in which ring B contains an imidazole ring are tautomeric and, in the case of such compounds, it is highly desirable that more than 50% of the molecule is in the form of the isomer in which the nitrogen atom on the same side of CYCLE as R^ (the nitrogen atom joined to the 4-position of ring A should be in the unprotonated form -N=. More desirably, at least 75%, e.g. at least 90% of the compound is in this form.
The invention includes prodrugs for the active pharmaceutical species of the invention, for example in which one or more functional groups are protected or derivatised but can be converted in vivo to the functional group, as in the case of esterified hydroxy groups, for example. The term "prodrug," as used herein, represents compounds which are transformed in vivo to the parent compound, for example, by hydrolysis in blood. A thorough discussion is provided in T. Higuchi and V. Stella, Pro-drugs as Novel Delivery Systems, Vol. 14 of the A.C.S. Symposium Series, Edward B. Roche, ed., Bioreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon Press, 1987, and Judkins, et al. Synthetic Communications, 26(23), 4351-4367 (1996), each of which is incorporated herein by reference. The term "prodrug" is to be widely interpreted and includes, inter alia, salts of covalent prodrug molecules.
The use of protecting groups is fully described in Protective Groups in Organic Chemistry , edited by J W F McOmie, Plenum Press (1973), and " Protective Groups in Organic Synthesis', 2nd edition, T W Greene &. P G M Wutz, Wiley-Interscience (1991). Thus, it will be appreciated by those skilled in the art that, although protected derivatives of compounds of the invention may not possess pharmacological activity as such, they may be administered, for example parenterally or orally, and thereafter metabolised in the body to form compounds of the invention which are pharmacologically active. Such derivatives and their salts are therefore examples of "prodrugs". All prodrugs of the described compounds are included within the scope of the invention.
The pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound (active compound or prodrug) which contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred. Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, Pa., US, 1985, p. 1418, the disclosure of which is hereby incorporated by reference.
The invention thus includes pharmaceutically-acceptable salts of the disclosed compounds and their covalent prodrug molecules wherein the parent compound is modified by making acid or base salts thereof, for example the conventional non-toxic salts or the quaternary ammonium salts which are formed, e.g., from inorganic or organic acids or bases. Examples of such acid addition salts include acetate, adipate, alginate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate, lactate, maleate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, oxalate, palmoate, pectinate, persulfate, 3-phenylpropionate, picrate, pivalate, propionate, succinate, tartrate, thiocyanate, tosylate, and undecanoate. Base salts include ammonium salts, alkali metal salts such as sodium and potassium salts, alkaline earth metal salts such as calcium and magnesium salts, salts with organic bases such as dicyclohexylamine salts, N-methyl-D-glucamine, and salts with amino acids such as arginine, lysine, and so forth. Also, the basic nitrogen-containing groups may be quatemized with such agents as lower alkyl halides, such as methyl, ethyl, propyl, and butyl chloride, bromides and iodides; dialkyl sulfates like dimethyl, diethyl, dibutyl; and diamyl sulfates, long chain halides such as decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides, aralkyl halides like benzyl and phenethyl bromides and others.
The phrase "pharmaceutically acceptable" is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings or animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit risk ratio.
Geometric isomers may exist in the products of the present invention. The present invention contemplates the various geometric isomers and mixtures thereof resulting from the arrangement of substituents around a carbon—carbon double bond and designates such isomers as of the Z or E configuration, wherein the term "Z" represents substituents on the same side of the carbon- carbon double bond and the term "E" represents substituents on opposite sides of the carbon- carbon double bond.
The invention therefore includes all variant forms of the defined compounds, for example any substance which, upon administration, is capable of providing directly or indirectly a compound as defined above or providing a species which is capable of existing in equilibrium with such a compound.
It will be understood that the invention specifically includes variants of preferred or exemplary compounds in which one or more moieties (e.g. substituents) have been replaced by alternatives described in this application.
The products of the invention are useful for treating mammals, especially a human (male or female) as described next.
Methods of Use
Another aspect of this invention, therefore, resides in methods of treating a mammal having a disease or condition mediated by an A3 adenosine receptor by administering a therapeutically effective amount of a product of the invention to the mammal.
In addition, the present invention provides a method of selectively activating A3 adenosine receptors in a mammal, which method comprises acutely or chronically administering to a mammal in need of selective activation of its A3 adenosine receptors a therapeutically effective amount, including a prophylactically effective amount, of a compound which binds with the A3 receptor so as to stimulate an A3 receptor-dependent response.
The method of the present invention has particular usefulness in vivo applications. For example, A3 adenosine receptor agonists can be used in the treatment of any disease, state or condition involving the release of cyclic adenosine monophosphate or the release of inositol-1,4,5- triphosphate (IP3), diacylglycerol (DAG), and free radicals and subsequent arachidonic acid cascades. Thus, high blood pressure, locomotor hyperactivity, hypertension, acute hypoxia, depression, and infertility can be treated in accordance with the present inventive method, wherein one of the above-described compounds is acutely administered, e.g., within about a few minutes to about an hour of the onset or realization of symptoms. The method also has utility in the treatment of chronic disease states and conditions, in particular those conditions and disease states wherein prophylactic or therapeutic administration of one of the above-described compounds will prevent the further onset of symptoms or will reduce recovery time. Examples of disease states and conditions that may be treated in accordance with the present inventive method include inflammatory disorders, such as vascular inflammation and arthritis, allergies, asthma, wound healing, stroke, cardiac infarct, cardiac failure, acute spinal cord injury, acute head injury or trauma, seizure, neonatal hypoxia (cerebral palsy; prophylactic treatment involves chronic exposure through placental circulation), hypoxia and chronic hypoxia due to arteriovenous malformations and occlusive cerebral artery disease, severe neurological disorders related to excitotoxicity, Parkinson's disease, Huntington's chorea, and other diseases of the central nervous system (CNS), cardiac disease, kidney disease, and contraception. Particular disease states which may be treated with the compounds of the invention are cardiac infarct and hypoxia.
Moreover, the above compounds may be used to treat malignant hypotension. For example, the administration of IB-MECA results in a significant increase (e.g., about 10-30%) in basal or systemic blood pressure (e.g., from about 70 mm Hg to about 90 mm Hg).
The above compounds may also be used to treat and/or protect against a variety of disorders, including, for example, seizures, transient ischemic shock, strokes, focal ischemia originating from thrombus or cerebral hemorrhage, global ischemia originating from cardiac arrest, trauma, neonatal palsy, hypovolemic shock, and hyperglycemia and associated neuropathies.
The present inventive method includes the administration to an animal, such as a mammal, particularly a human, in need of the desired A3 receptor-dependent response of an effective amount, e.g., a therapeutically effective amount, of one or more of the aforementioned present inventive compounds or pharmaceutically acceptable salts or derivatives thereof, alone or in combination with one or more other pharmaceutically active compounds.
The compounds of the invention will normally be administered orally, intravenously, subcutaneously, buccally, rectally, dermally, nasally, tracheally, bronchially, by any other parenteral route, as an oral or nasal spray or via inhalation, The compounds may be administered in the form of pharmaceutical preparations comprising prodrug or active compound either as a free compound or, for example, a pharmaceutically acceptable non-toxic organic or inorganic acid or base addition salt, in a pharmaceutically acceptable dosage form. Depending upon the disorder and patient to be treated and the route of administration, the compositions may be administered at varying doses.
The most preferred routes of administration are injection and infusion, especially intravenous administration.
The compounds of the invention may be combined and/or co-administered with any antithrombotic agent, such as the antiplatelet agents acetylsalicylic acid, ticlopidine, clopidogrel, thromboxane receptor and/or synthetase inhibitors, fibrinogen receptor antagonists, prostacyclin mimetics and phosphodiesterase inhibitors and ADP-receptor (P2 T) antagonists.
The compounds of the invention may be combined and/or co-administered with thrombolytics such as tissue plasminogen activator (natural, recombinant or modified), streptokinase, urokinase, prourokinase, anisoylated plasminogen-streptokinase activator complex (APSAC), animal salivary gland plasminogen activators, and the like, in the treatment of thrombotic diseases, in particular myocardial infarction.
Typically, therefore, the pharmaceutical compounds of the invention may be administered orally or parenterally ("parenterally" as used herein, refers to modes of administration which include intravenous, intramuscular, intraperitoneal, intrasternal, subcutaneous and intraarticular injection and infusion of which intavenous is most preferred.) to a host to obtain a desired effect, for example protection against ischaemia or a cardioprotectant effect. In the case of larger animals, such as humans, the compounds may be administered alone or as compositions in combination with pharmaceutically acceptable diluents, excipients or carriers.
Actual dosage levels of active ingredients in the pharmaceutical compositions of this invention may be varied so as to obtain an amount of the active compoϋnd(s) that is effective to achieve the desired therapeutic response for a particular patient, compositions, and mode of administration. The selected dosage level will depend upon the activity of the particular compound, the route of administration, the severity of the condition being treated and the condition and prior medical history of the patient being treated. However, it is within the skill of the art to start doses of the compound at levels lower than required for to achieve the desired therapeutic effect and to gradually increase the dosage until the desired effect is achieved.
Another aspect of this invention is directed to methods of reducing tissue damage (e.g., substantially preventing tissue damage, inducing tissue protection) resulting from ischemia or hypoxia comprising administering to a mammal in need of such treatment a therapeutically effective amount of a product of the invention. Preferred ischemic/hypoxic tissues taken individually or as a group are cardiac, brain, liver, kidney, lung, gut, skeletal muscle, spleen, pancreas, nerve, spinal cord, retina tissue, the vasculature, or intestinal tissue, an especially preferred ischemic/hypoxic tissue is cardiac tissue.
It is especially preferred that the products of the invention are administered to prevent perioperative myocardial ischemic injury.
Preferably, the products of this invention are administered prophylactically.
The ischemic/hypoxic damage may occur during organ transplantation. Preferably, the compounds of this invention are administered prior to, during or shortly after, cardiac surgery or non-cardiac surgery (e.g., a three to four day infusion).
In one aspect of this invention a product of the invention is administered locally.
Another aspect of this invention is directed to methods of reducing myocardial tissue damage (e.g., substantially preventing tissue damage, inducing tissue protection) during surgery (e.g., coronary artery bypass grafting (CABG) surgeries, vascular surgeries, percutaneous transluminal coronary angioplasty (PTCA) or any percutaneous transluminal coronary intervention (PTCI), organ transplantation, or other non-cardiac surgeries) comprising administering to a mammal a therapeutically effective amount of a product of the invention. Another aspect of this invention is directed to methods of reducing myocardial tissue damage (e.g., substantially preventing tissue damage, inducing tissue protection) in patients presenting with ongoing cardiac syndromes (acute coronary syndromes, e.g., myocardial infarction or unstable angina) or cerebral ischemic events (e.g., stroke) comprising administering to a mammal a therapeutically effective amount of a product of the invention.
Another aspect of this invention is directed to chronic methods of reducing myocardial tissue damage (e.g., substantially preventing tissue damage, inducing tissue protection) in a patient with diagnosed coronary heart disease (e.g., previous myocardial infarction or unstable angina) or patients who are at high risk for myocardial infarction (e.g. age > 65 and two or more risk factors for coronary heart disease) comprising administering to a mammal a therapeutically effective amount of a product of the invention.
Another aspect of this invention is directed to methods of preventing ischemic/hypoxic damage comprising the chronic oral administration to a mammal in need of such treatment of a therapeutically effective amount of a product of the invention. Another aspect of this invention is directed to methods for treating cardiovascular diseases comprising administering to a mammal a therapeutically effective amount of a product of the invention.
Another aspect of this invention is directed to methods for treating arteriosclerosis comprising administering to a mammal a therapeutically effective amount of a product of the invention.
Another aspect of this invention is directed to methods for treating arrhythmia comprising administering to a mammal a therapeutically effective amount of a product of the invention.
Another aspect of this invention is directed to methods for treating angina pectoris comprising administering to a mammal a therapeutically effective amount of a product of the invention.
Another aspect of this invention is directed to methods for treating cardiac hypertrophy comprising administering to a mammal a therapeutically effective amount of a product of the invention.
Another aspect of this invention is directed to methods for treating renal diseases comprising administering to a mammal a therapeutically effective amount of a product of the invention.
Another aspect of this invention is directed to methods for treating diabetic complications comprising administering to a mammal a therapeutically effective amount of a product of the invention.
Another aspect of this invention is directed to methods for treating restenosis comprising administering to a mammal a therapeutically effective amount of a product of the invention.
Another aspect of this invention is directed to methods for treating organ hypertrophies or hyperplasias comprising administering to a mammal a therapeutically effective amount of a product of the invention.
Another aspect of this invention is directed to methods for treating septic shock and other inflammatory diseases (septicemia, endotoxcemia) comprising administering to a mammal a therapeutically effective amount of a product of the invention.
Another aspect of this invention is directed to methods for treating cerebro ischemic disorders comprising administering to a mammal a therapeutically effective amount of a product of the invention. Another aspect of this invention is directed to methods for treating myocardial stunning comprising administering to a mammal a therapeutically effective amount of a product of the invention.
Another aspect of this invention is directed to methods for treating myocardial dysfunction comprising administering to a mammal a therapeutically effective amount of a product of the invention.
Another aspect of this invention is directed to methods for treating cerebrovascular diseases comprising administering to a mammal a therapeutically effective amount of a product of the invention.
Further applications of the products of the invention are described in the prior art documents mentioned under the heading "Background of the Invention".
Pharmaceutical Compositions
The present invention also provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and an effective amount, e.g., a therapeutically effective amount, including a prophylactically effective amount, of one or more of the aforesaid compounds. The products of the invention may be formulated into pharmaceutical compositions as described in WO 95/02604, the contents of which are incorporated herein by reference.
According to a further aspect of the invention there is thus provided a pharmaceutical composition including a compound of the invention, in admixture with a pharmaceutically acceptable adjuvant, diluent or carrier. Proposed compositions are intavenous formulations. These formulations typically contain a compound of the invention or a salt thereof.
Pharmaceutical compositions of this invention for parenteral injection suitably comprise pharmaceutically acceptable sterile aqueous or nonaqueous solutions, dispersions, suspensions or emulsions as well as sterile powders for reconstitution into sterile injectable solutions or dispersions just prior to use. Examples of suitable aqueous and nonaqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol and the like), and suitable mixtures thereof, vegetable oils (such as olive oil) and injectable organic esters such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of coating materials such as lecithin, by the maintenance of the required particle size in the case of dispersions and by the use of surfactants. These compositions may also contain adjuvants such as preservative, wetting agents, emulsifying agents and dispersing agents. Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol or phenol sorbic acid. It may also be desirable to include isotonic agents such as sugars or sodium chloride, for example. Prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents (for example aluminum monostearate and gelatin) which delay absorption.
In some cases, in order to prolong the effect of the drug, it is desirable to slow the absorption of the drug from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of the drug then depends upon its rate of dissolution which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle.
Injectable depot forms are suitably made by forming microencapsule matrices of the drug in biodegradable polymers, for example polylactide-polyglycolide. Depending upon the ratio of drug to polymer and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations may also prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissues. The injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable media just prior to use.
Solid dosage forms for oral administration include capsules, tablets, pills, powders and granules. In such solid dosage forms, the active compound is typically mixed with at least one inert, pharmaceutically acceptable excϊpieπt or carrier such as sodium citrate or dicalcium phosphate and/or one or more: a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol and silicic acid; b) binders such as carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose and acacia; c) humectants such as glycerol; d) disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginϊc acid, certain silicates and sodium carbonate; e) solution retarding agents such as paraffin; f) absorption accelerators such as quaternary ammonium compounds; g) wetting agents such as cetyl alcohol and glycerol monostearate; h) absorbents such as kaolin and bentonite clay and i) lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate and mixtures thereof. In the case of capsules, tablets and pills, the dosage form may also comprise buffering agents. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycol, for example.
Suitably, oral formulations contain a dissolution aid. The dissolution aid is not limited as to its identity so long as it is pharmaceutically acceptable. Examples include nonionic surface active agents, such as sucrose fatty acid esters, glycerol fatty acid esters, sorbitan fatty acid esters (e.g., sorbitan trioleate), polyethylene glycol, polyoxyethylene hydrogenated castor oil, polyoxyethylene sorbitan fatty acid esters, polyoxyethylene alkyl ethers, methoxypolyoxyethylene alkyl ethers, polyoxyethylene alkylphenyl ethers, polyethylene glycol fatty acid esters, polyoxyethylene alkylamines, polyoxyethylene alkyl thioethers, polyoxyethylene polyoxypropylene copolymers, polyoxyethylene glycerol fatty acid esters, pentaerythritol fatty acid esters, propylene glycol monofatty acid esters, polyoxyethylene propylene glycol monofatty acid esters, polyoxyethylene sorbitol fatty acid esters, fatty acid alkylolamides, and alkylamine oxides; bile acid and salts thereof (e.g., chenodeoxycholic acid, cholic acid, deoxycholic acid, dehydrocholic acid and salts thereof, and glycine or taurine conjugate thereof); ionic surface active agents, such as sodium laurylsulfate, fatty acid soaps, alkylsulfonates, alkylphosphates, ether phosphates, fatty acid salts of basic amino acids; triethanolamine soap, and alkyl quaternary ammonium salts; and amphoteric surface active agents, such as betaines and amiπocarboxylic acid salts.
The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and may also be of a composition such that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, and/or in delayed fashion. Examples of embedding compositions which can be used include polymeric substances and waxes.
The products of the invention may also be in micro-encapsulated form, if appropriate, with one or more of the above-mentioned excipients.
The active compound may be in finely divided form, for example it may be micronised.
Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups and elixirs. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly used in the art such as water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethyl formamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan and mixtures thereof. Besides inert diluents, the oral compositions may also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring and perfuming agents. Suspensions, in addition to the active compounds, may contain suspending agents such as ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar, and tragacanth and mixtures thereof.
Compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of this invention with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at room temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.
Compounds of the present invention can also be administered in the form of liposomes. As is known in the art, liposomes are generally derived from phospholipids or other lipid substances. Liposomes are formed by mono- or multi-lamellar hydrated liquid crystals which are dispersed in an aqueous medium. Any non-toxic, physiologically acceptable and metabolisable lipid capable of forming liposomes can be used. The present compositions in liposome form can contain, in addition to a compound of the present invention, stabilisers, preservatives, excipients and the like. The preferred lipids are the phospholipids and the phosphatidyl cholines (lecithins), both natural and synthetic. Methods to form liposomes are known in the art, for example, Prescott, Ed., Methods in Cell Biology, Volume XIV, Academic Press, New York, N.Y. (1976), p 33 et seq.
Dosage forms for topical administration of a compound of this invention include powders, sprays, ointments and inhalants. The active compound is mixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives, buffers or propellants which may be required. Ophthalmic formulations, eye ointments, powders and solutions are also contemplated as being within the scope of this invention.
Advantageously, the compounds of the invention are orally active, have rapid onset of activity and low toxicity.
The compounds of the invention have the advantage that they may be more efficacious, be less toxic, be longer acting, have a broader range of activity, be more potent, produce fewer side effects, be more easily absorbed than, or that they may have other useful pharmacological properties over, compounds known in the prior art.
Suitable doses and dosage regimens can be determined by conventional range-finding techniques known to those of ordinary skill in the art. Generally, treatment is initiated with smaller dosages, which are less than the optimum dose of the compound. Thereafter, the dosage is increased by small increments until the optimum effect under the circumstances is reached. For convenience, the total daily dosage may be divided and administered in portions during the day if desired. The compounds of the invention may be administered by any suitable means. A preferred method of administration is by IV injection. In proper doses and with suitable administration of certain compounds, the present invention provides for a wide range of selective A3 receptor-dependent responses. Exemplary dosages range from about 0.1 to about 100 mg/kg body weight of the animal being treated/day. Therapeutically effective dosages range from about 0.01 to about 10 mg/kg body weight day .The invention will now be illustrated by the following non-limiting examples.
This invention is also directed to pharmaceutical compositions which comprise a therapeutically effective amount of a product of the invention and a pharmaceutically acceptable carrier, vehicle or diluent.
This invention is also directed to pharmaceutical compositions for the reduction of tissue damage resulting from ischemia or hypoxia which comprise a therapeutically effective amount of a product of the invention.
This invention is also directed to a kit for use in treating a mammal having or at risk of having a disease or condition resulting from, for example, ischemia or hypoxia which may be ameliorated by an A3 agonist. The kit comprises a) a suitable dosage form, such as, for example, an injectable parenteral solution particularly adapted for intravenous or intramuscular injection, comprising a compound of Formula I; and b) instructions describing a method of using the dosage form to reduce tissue damage resulting from ischemia or hypoxia.
Yet another aspect of this invention is combinations of a product of the invention and one or more other compounds as described below.
This invention is also directed to a pharmaceutical combination composition comprising: a therapeutically effective amount of a composition comprising a first product, said first product being a product of the invention; a second compound, said second compound being a cardiovascular agent; and, optionally, a pharmaceutical carrier, vehicle or diluent.
Another aspect of this invention are methods of reducing tissue damage (e.g., substantially preventing tissue damage, inducing tissue protection) resulting from or which could result from ischemia or hypoxia comprising administering to a mammal a first product, said first compound being a product of the invention; and a second product, said second product being a cardiovascular agent wherein the amounts of the first and second compounds result in a therapeutic effect. Another aspect of this invention are kits comprising: a. a product of the invention and a pharmaceutically-acceptable carrier, vehicle or diluent in a first unit dosage form; . b. a cardiovascular agent and a pharmaceutically acceptable carrier, vehicle or diluent in a second unit dosage form; and c. means for containing said first and second dosage forms wherein the amounts of the first and second compounds result in a therapeutic effect.
The invention therefore includes methods of treatment in which a product of the invention and one or more other therapeutic agents are administered to a mammal. Also included are products including both a product of the invention and one or more other therapeutic agents. Said other therapuetic agent(s) (e.g., agents having a cardiovascular effect) are, for example, β-blockers (e.g., acebutolol, atenolol, bopindolol, labetolol, mepindolol, nadolol, oxprenol, pindolol, propranolol, sotalol), calcium channel blockers (e.g., amlodipine, nifedipine, nisoldipine, nitrendipine, verapamil), potassium channel openers, adenosine, adenosine agonists, sodium- hydrogen exchanger type 1 (NHE-1) inhibitors, ACE inhibitors (e.g., captopril, enalapril), nitrates (e.g., isosorbide dϊnϊtrate, isosorbide 5-mononitrate, glyceryl trinitrate), diuretics (e.g., hydrochlorothiazide, indapamide, piretanide, xipamide), glycosides (e.g., digoxin, metildigoxin), thrombolytics as described above, platelet inhibitors (e.g., repro), aspirin, dipyridamol, potassium chloride, clonidine, prazosin, pyruvate dehydrogenase kinase inhibitors (e.g., dichloroacetate), pyruvate dehydrogenase complex activators, biguanides (e.g. metformin) or other 5 adenosine A3 receptor agonists. Other cardiovascular agents include angiotensin II (All) receptor antagonists, C5a inhibitors, soluble complement receptor type 1 (sCRl) or analogues, partial fatty acid oxidation (PFOX) inhibitors (specifically, ranolazine), acetyl CoA carboxylase activators, malonyl CoA decarboxylase inhibitors, 5' AMP-activated protein kinase (AMPK) inhibitors, adenosine πucleoside inhibitors, anti-apoptotic agents (e.g., caspase inhibitors), monophosphoryl lipid A or analogues, nitric oxide synthase activators/inhibitors, protein kinase C activators (specifically, protein kinase E), protein kinase delta inhibitor, poly (ADP ribose) synthetase (PARS, PARR) inhibitors, metformin (gluconeogenesis inhibitors, insulin sensitizers), endothelin converting enzyme (ECE) inhibitors, endothelin ETA receptor antagonists, thrombin activated fibrinolytic inhibitor TAFT inhibitors and Na/Ca exchanger modulators.
In one very preferred method, a patient is administered, in effective amounts, a product of the invention and a thrombolytic. Sometimes, but not always, one of these two active agents is administered more or less immediately after the other. This invention is also directed to a pharmaceutical combination composition comprising: a therapeutically effective amount of a composition comprising a first product, said first product being a product of the invention; a second product, said second product being a glycogen phosphorylase inhibitor; and, optionally, a pharmaceutical carrier, vehicle or diluent.
Another aspect of this invention resides in methods of reducing tissue damage (e.g., substantially preventing tissue damage, inducing tissue protection) resulting from or which could result from ischemia or hypoxia comprising administering to a mammal a. a first compound, said first compound being a product of the invention said second compound being a glycogen phosphorylase inhibitor wherein the amounts of the first and second compounds result in a therapeutic effect.
Another aspect of this invention are kits comprising: a. a product of the invention and a pharmaceutically acceptable carrier, vehicle or diluent in a first unit dosage form; b. a glycogen phosphorylase inhibitor and a pharmaceutically acceptable carrier, vehicle or diluent in a second unit dosage form; and b. means for containing said first and second dosage forms wherein the amounts of the first and second compounds result in a therapeutic effect.
This invention is also directed to a pharmaceutical combination composition comprising: a therapeutically effective amount of a composition comprising a product of the invention; an aldose reductase inhibitor; and, optionally, a pharmaceutical carrier, vehicle or diluent.
Another aspect of this invention are methods of reducing tissue damage (e.g., substantially preventing tissue damage, inducing tissue protection) resulting from or which could result from ischemia or hypoxia comprising administering to a mammal a. a product of the invention and c. an aldose reductase inhibitor wherein the amounts of said product and said inhibitor result in a therapeutic effect.
Another aspect of this invention are kits comprising: a. a product of the invention and a pharmaceutically acceptable carrier, vehicle or diluent in a first unit dosage form; b. an aldose reductase inhibitor and a pharmaceutically acceptable carrier, vehicle or diluent in a second unit dosage form; and c. means for containing said first and second dosage forms wherein the amounts of the first and second compounds result in a therapeutic effect.
In the above combination compositions, combination methods and kits a preferred aldose reductase inhibitor is zopolrestat:, 3,4-dihydro-4-oxo-3-[[5-trifluoromethyl)-2-benzothiazolyl] methyl]-l-phthalazineacetic acid.
In the methods of treatment as applied to the combinations described above the following are preferred administration routes, modes, etc.
Preferred ischemic or hypoxic tissues taken individually or as a group are wherein the ischemic/hypoxic tissue is cardiac, brain, liver, kidney, lung, gut, skeletal muscle, spleen, pancreas, nerve, spinal cord, retina tissue, the vasculature, or intestinal tissue.
An especially preferred ischemic or hypoxic tissue is cardiac tissue.
It is especially preferred that the combinations are administered to prevent perioperative myocardial ischemic injury.
Preferably, the combinations of this invention are administered prophylactically.
The ischemic/hypoxic damage may occur during organ transplantation. Preferably, the combinations of this invention are administered prior to, during and/or shortly after, cardiac surgery or non-cardiac surgery.
In one aspect of this invention the combinations are administered locally.
In one aspect of this invention myocardial tissue damage is reduced during or after surgery.
In another aspect of this inventor myocardial tissue damage is reduced in patients presenting with ongoing cardiac or cerebral ischemic events.
In yet another aspect of this inventor myocardial tissue damage is reduced by chronic admin istratioπ of the above combinations in a patient with diagnosed coronary heart disease. The term "reduction" is intended to include partial prevention or prevention which, although greater than that which would result from taking no compound or from taking a placebo, is less than 100% in addition to substantially total prevention. The term "damage resulting from ischemia or hypoxia "as employed herein refers to conditions directly associated with reduced blood flow or oxygen delivery to tissue, for example due to a clot or obstruction of blood vessels which supply blood to the subject tissue and which result, inter alia, in lowered oxygen transport to such tissue, impaired tissue performance, tissue dysfunction and/or necrosis and/or apoptosis. Alternatively, where blood flow or organ perfusion may be quantitatively adequate, the oxygen carrying capacity of the blood or organ perfusion medium may be reduced, e.g., in a hypoxic environment, such that oxygen supply to the tissue is lowered, and impaired tissue performance, tissue dysfunction, and/or tissue necrosis and/or apoptosis ensues.
The term "treating", "treat" or "treatment" as used herein includes preventative (e.g., prophylactic) and palliative treatment.
Compound Synthesis
Compounds of the invention may be synthesized by any suitable means. In this respect, synthesis of adenosine analogues is well known in the art and is described in the documents listed above under the heading "Background of the Invention". For example, guidance may be found in the "Compound Synthesis" sections of WO 95/02604 as well as of corresponding US 5773423 and US 5688774, which sections are included herein by reference. The reader is also referred to reaction schemes A to I and examples of WO 92/05177 and corresponding US 5561134 and US 5736554, all of which disclosures are incorporated herein by reference. Further assistance may be found in EP 1241176 and corresponding USSN 60/276411, including Schemes I, II and III thereof and the related text, all of which disclosures are incorporated herein by reference. The following methods and reactants (intermediates) are novel and part of the invention.
A first method comprises reacting a compound of the formula L-CR20R21-CYCLE, where L is a leaving group, with a compound H2 -ARA, where the nitrogen of H2N- is the N6 nitrogen of an adenosine A3 receptor agonist and ARA represents the remainder of the adenosine A3 receptor agonist.
A second method comprises reacting a compound of the formula H2N -CR20R21-CYCLE with a compound of the formula
C6-L-ARA, where ARA again represents the residue of an adenosine A3 receptor agonist, excluding the N6 nitrogen, and C6-L represents a leaving group substituted on the C6 carbon of
ARA. Especially but not exclusively in this second method, reactive functional groups of ARA (e.g. hydroxy or amino groups constituting X3 and X4 of formula III) may be protected. Suitable leaving groups include chloro and bromo. Chloro is often convenient for the second method, as in the case of an ARA residue as illustrated by formula IV in which R2 is H and R1 is Me.
One suitable method for synthesising amongst others, the compounds of formula I of the invention in which R2 is hydrogen is illustrated by following reaction scheme A in which R^ is H:
Figure imgf000039_0001
This method uses as a starting material the known 5'-/IAaIkyIcarboxamidoadenosines such as 5'-N- methylcarboxamidoadenosine, which may be protected as necessary prior to reaction. The 2- picolyl reactant may be made using the reaction scheme of Figure 1, which may be generalised where necessary. The 2-alkenyl substituted compounds of the invention may be synthesised using the synthetic reaction scheme illustrated in Figure 2, which may be generalized where necessary. In this scheme, the 6-chIoro-2-iodopurine-9-riboside (Compound A) is already known, J. Med. Chem., 2000, vol43, page 4137. In step 1, this is protected in a manner known per se to yield compound B which is then oxidised to give acid C Acid £ is reacted with the R1 amine (R1 H2) to give uronamide D. The 2-iodo uronamide is then reacted with triethylamine, bis- (triphenylphosphine)palladium dichloride and Cul in catalytic amount using as solvent a mixture of acetonitrile/DMF 2:1 and to this mixture is added the terminal alkyne and the reaction takes place under 2 atmosphere at room temperature (ref J. Med. Chem. 1995, vol 38, 1462-1472) to result in compound E. The coupling of the 2-pϊcolylamine with the 6-chloro derivative is then accomplished as described above (compound F), and deprotection with HCI IN at 70°C yields the final compound, G. In the example shown, the alkyne used in the phenyl alkyne; other alkynes can be used in analogous ,manner. The alkenyl compounds may be prepared analogously.
Another method, suited for example for synthesising the compounds of formula II of the invention in which R2 is hydrogen is illustrated by following reaction scheme B in which Z is -N= and Y is -0-:
Figure imgf000041_0001
The above scheme is applicable equally to compounds having a pyridyloxazolyl group in place of a benzoxazolyl group.
Each P is a protecting group, which may be taken together to represent a bridging protecting group such as an isopropylidene radical. The protecting group may be removed by conventional means, for example by treatment with an acid. L is a leaving group which may for example be selected from chloro, bromo or iodo or tosylates. Preferably the leaving group is chloro.
This method thus uses as a starting material the known 2',3'-CMsopropylidene-6-chloropurine-5'- alkyluronamide and equivalents. This reaction step is also shown more specifically in the reaction scheme of Figure 4. The benzoxazole reactant may be made using the reaction scheme of Figure 3, which may be generalised where necessary.
The 2-alkynyl substituted compounds of the invention may be synthesised using the synthetic reaction scheme illustrated in Figure 5 which may be generalized where necessary. I'β'-O- IsopropyIideneguanosine-5'-carboxylic acid A (J. Org. Chem. 1999, 64, 293-295)is reacted with triethylamine, isopropenylchloroformate and methylamine at 0°C yielding compound B. This is reacted with phosphoryl chloride to obtain compound C. C is treated with isoamyl nitrite, Cul, CH2I2 and I2 to give compound D. The 2 iodo uronamide is then reacted with triethylamine, bis(triphenylphosphine)palladium dichloride and Cul in catalytic amount using as solvent a mixture of acetonitrile/DMF 2:1 and to this mixture is added the terminal alkyne and the reaction takes place under N2 atmosphere at room temperature (ref J. Med. Chem. 1995, vol 38, 1462-1472) to result in compound E. The coupling of the benzoxazole reactant with the 6-chloro derivative is then accomplished as described above. In the example shown, the alkyne used in the phenyl alkyne; other alkynes can be used in analogous manner. The alkenyl compounds may be prepared analogously.
In the case of compounds which are 2-substituted by halogen (i.e. R2 is halogen), the procedure of Reaction scheme A or B may be followed using a correspondingly halogenated starting material, to produce a mixture of products, which may then be separated, for example by HPLC or another conventional technique.
Examples
Example 1
/\t5-(4-methyl-2-picolyl)-adenosine-5,-/\Λmethyluronamide
Figure imgf000043_0001
To a solution of 5'-/V-methylcarboxamidoadenosine (100 mg, 0.34 mmol) in DMF (1 L) was added 2-(bromomethyl)-4-methylpyridine (95 mg, 0.51 mmol), and the solution stirred for 3 days at 40 °C. The solvent was evaporated and the residue treated with methanol (1.5 mL) and concentrated NH4OH (3.0 mL). The mixture was warmed in a closed vessel at 90 °C for 2 h with stirring. After evaporating the solvent, the residue was chromatographied (6% MeOH in CH2CI2) and 60 mg (44%) of the title compound (a white solid) were obtained. Rf (CHzCI^MeOH 9:1) 0.3; H NMR (CD3OD) δ 8.3-8.5 (m, 3H, H-8, H-2, H-6pyπdyι), 7.31 (s, IH, H-3pyridy)), 7.19 (d, IH, J=5.0 Hz, H-5pyridyι), 6.07 (d, IH, ^=7.8 Hz, H-1Q, 4.9 (not observed, 2H, CH2), 4.79 (dd, IH, Jχ,v= 7.8 Hz, J2',3<=4.7 Hz, H-2 , 4.52 (s, IH, H-41), 4.35 (d, IH, Jr,2'=4.7 Hz, H-3 , 2.91 (s, 3H, 5'-N- methyl), 2.37 (s, 3H, CH3); 13C NMR (CD3OD) δ 173.2 (C-50, 159.6 (Cpyridyr2), 156.6 (C-6), 154.3 (C-2), 151.1 (C-4), 149.8 (Cpyridy,-6), 143.0 (Cpyridyl-4), 142.8 (C-8), 125.1 (Cpyπdy,-3), 123.8 (Cpyridyl- 5), 122.2 (C-5), 90.9 (C-l1), 86.9 (C-47), 75.3, 73.8 (C-3', C-2 , 46.5 (CH2), 26.4 (5'-/V-methyl), 21.5 (CH3). High-resolution MS calculated for .(d8 H21 N7 04 Na) 422.1553, found 422.1556.
2-(BromomethyO-4-methy[pyridine:
Figure imgf000043_0002
To a mixture of the alcohol 2-(Hydroxymβthyl)-4-methylpyridine (560 mg, 4.55 mmol) and CBR4 (2.43 g, 7.33 mmol) in CH2CI2 (10.6 mL) was added triphenylphosphine (1.4 g, 5.34 mmol) in several portions at 0 °C. The reaction mixture was stirred for 20 min at the same temperature and then directly passed through a short silica gel column using 10% EtOAc in hexane as an eluent to give the title compound as a white solid (640 mg) in 76% yield. XH NMR (CDCI3) δ 8.34 (d, IH, J=4.9 Hz, H-6); 7.17 (s, IH, H-3); 6.94 (d, IH, J-4.9 Hz, H-5); 4.43 (s, 2H, CH2); 2.26 (s, 3H); 13C NMR (CDCI3) δ 156.9 (C-2), 149.8 (C-6), 148.8 (C-4), 124.7 (C-3), 124.4 (C-5), 34.4 (CH2), 21.4 (Me). 2-(HydroxymethvO-4-methylpyridine:
Figure imgf000044_0001
Compound 2f4-Di ethylpyridine N-Oxide (4.47 g, 36.3 mmol) was dissolved in Ac20 (11 mL) and added dropwise to acetic anhydride (108 mL) heated to 110 °C. The resulting solution was stirred at 110 °C for 1 hour and 15 minutes. Excess reagent was evaporated leaving the corresponding 2-[(acetyloxy)-methyl]pyridine which was used without further purification. To a solution of the protected alcohol in MeOH (5 mL) was added NaOH 2N (15 mL) at room temperature. The mixture was stirred at room temperature for 1.5 hours, extracted with EtOAc and washed with water. The solvent was evaporated giving a residue that was purified by column chromatography (4% MeOH in CH2CI2) affording 1.54 g (34% overall yield) of colorless oil. H NMR (CD3OD) δ 8.29 (d, IH, J=4.2 Hz, H-6); 7.40 (s, IH, H-3); 7.11 (d, IH, J=4.2 Hz, H-5); 5.10 (s, IH, OH); 4.68 (s, 2H, CH2); 2.38 (s, 3H); 13C NMR (CD3OD) δ 162.4 (C-2), 151.0 (C-4), 149.4 (C-6), 125.0 (C-3), 123.3 (C-5), 65.8 (CH2), 21.7 (Me).
2,4-Dimethylpyridine /lOxide:
Figure imgf000044_0002
Aqueous H202 (30%, 2.6 mL) was added to 2,4-lutidine (5 mL, 43.2 mmol) in acetic acid (15 mL), and the mixture was stirred for 3 hours at 90 °C. The mixture was cooled, and a second portion of aqueous H202 (30%, 1.1 mL) was added, after which the mixture was stirred for another 20 hours at 90 °C. The solvent was evaporated (toluene was used to remove remaining traces of acetic acid by means of azeotropic destination). The pH was adjusted to 10 with NaOH 10 M, CH3CN was added (10 mL) and precipitated materials were filtered off. The filtrate was evaporated leaving 4.47 g (84%) of the title compound as a white solid. XH NMR (CDC!3) δ 8.10 (d, IH, J=6.5 Hz, H-6); 7.04 (s, IH, H-3); 6.92 (d, IH, J=6.5 Hz, H-5); 2.45 (s, 3H), 2.28 (s, 3H); 13C NMR (CDCI3) δ 148,6 (C-4), 139.1 (C-6), 137.6 (C-2), 127.6 (C-3), 124.8 (C-5), 20.6 (MeC4), 18.2 (Me∞).
Example 2 l/3-(4-iodo-2-picolyl)-adenosine-5,-ΛAmethyluronamide
Figure imgf000045_0001
To a solution of 5'-/\/-methylcarboxamidoadenosine (150 mg, 0.51 mmol) in DMF (1.5 mL) was added 2-(Bromomethyl)-4-iodopyridine (152 mg, 0.51 mmol), and the solution stirred for 3 days at 40 °C. The solvent was evaporated and the residue treated with methanol (1.5 mL) and concentrated NH4OH (3.0 mL). The mixture was warmed in a closed vessel at 90 °C for 2 h with stirring. After evaporating the solvent, the residue was chromatographied (4% MeOH in CH2CI2) and 140 mg (54%) of the title compound (a white solid) were obtained. Rf (CH2Cl2/MeOH 9:1)
0.37; m.p. (MeOH): 128°C; H NMR (CD3OD) δ 8.40 (s, IH, H-8), 8.38 (s, IH, H-2), 8.26 (d, IH,
J=5.2 Hz, Hpyridy,-6), 7.92 (s, IH, Hpyridyr3), 7.79 (dd, IH, J=5.2 Hz, J=1.2 Hz, Hpyridyl-5), 6.13 (d,
IH, Jr,2'=7.7 Hz, H-1 , 4.97 (s, 2H, CH2), 4.86 (dd, IH, Jr,r= 7.7 Hz, ^=4. Hz, H-27), 4.59 (s,
IH, H-4 , 4.43 (dd, IH, -7^=4.7 Hz, ^=1.2 Hz, H-3 , 2.97 (s, 3H, 5'-/IAmethyl); 13C NMR
(DMSO-oQ δ 170.2 (C-51), 160.5 (Cpyridyl-2), 155.9 (C-6), 152.9 (C-2), 150.0 (Cpyridyr6), 148.2 (C-
4), 141.3 (C-8), 131.3 (Cpyridy,-3), 129.9 (Cpyridyl-5), 120.0 (C-5), 107.3 (Cpyridyl-4), 88.1 (C-1 , 85.0
(C-4 , 73.4, 72.5 (C-3', C-2 48.9 (CH2), 25.7 (5'-/lAmethyl).
2-(Bromomethyl)-4-iodopyridine:
Figure imgf000045_0002
To a mixture of the alcohol (4-Iodo-2-pyridyl)methanol '(245 mg, 1.04 mmol) and CBR4 (556 mg, 1.68 mmol) in CH2C!2 (4.6 mL) was added triphenylphosphine (320 mg, 1.22 mmol) in several portions at 0 °C. The reaction mixture was stirred for 20 min at the same temperature and then directly passed through a short silica gel column using 10% EtOAc in hexane as an eluent to give the title compound as white crystals (220 mg) in 71% yield. M.p.(CH2CI2) 74-76°C; *H NMR (CDCI3) δ 8.29 (d, IH, J=5.2 Hz, H-6); 7.89 (d, IH, J=1.5 Hz, H-3); 7.66 (dd, IH, Jι=5.2, . =1.5 Hz, H-5); 4.52 (s, 2H, CH2); 13C NMR (CDCI3) δ 158.1 (C-2), 150.3 (C-6), 133.1 (C-3), 132.7 (C-5), 106.5 (C-4), 33.0 (CH2).
(4-Iodo-2-pyridyDmethanol :
Figure imgf000046_0001
A solution of (4-iodo-2-pyridyl)methyIacetate ((4-iodo-2-pyridyl)methylacetatέ in 2.0 mL (MeOH/NaOH 2N 1:3) was stirred at room temperature for 40 minutes. The mixture was extracted with EtOAc, washed with NaCI sat., dried and evaporated, affording 245 mg of the title compound (100 %) as a colourless oil. XH NMR (CDCI3) δ 8.18 (d, IH, =4.1 Hz, H-6); 7.76 (s, IH, H-3); 7.59 (d, IH, 4.9 Hz, H-5); 4.72 (s, 2H, CH2); 4.45 (bs, IH, OH); 13C NMR (CDCI3) δ 161.2 (C-2), 149.3 (C-6), 132.1 (C-3), 130.5 (C-5), 106.9 (C-4), 64.3 (CH2).
4 odo-2-methylpyridine and (4-iodo-2-pyridyl)methylacetate:
Figure imgf000046_0002
2-(((Tert-butyldimethylsilyl)oxy)methyl)-4-chloropyridine (600 mg, 2.33 mmol), dry sodium iodide (5 g) and freshly distilled acetyl chloride (0.7. mL, 9.78 mmol) in 6 mL of anhydrous acetonitrile were refluxed under nitrogen for 33 hours. Aqueous 10% K2C03/ 5% NaHS03 was added and the mixture extracted three times with chloroform. After drying (Na2S04) and evaporation of the chloroform, flash chromatography (hexane/EtOAc 9:1) yielded 110 mg (22%) of 4-Iodo-2- methylpyridine as a white solid and 290 mg (45%) of (4-iodo-2-pyridyl)methylacetate as a white solid.
4-Iodo-2-methyIpyridine: XH NMR (CDCI3) δ 8.06 (d, IH, J=5.2 Hz, H-6); 7.48 (d, IH, J=l.l Hz, H- 3); 7.38 (dd, IH, =5.2, . =1.4 Hz, H-5); 2.41 (s, 3H, CH3); 13C NMR (CDCI3) δ 159.8 (C-2), 149.8 (C-6), 133.0 (C-3), 130.4 (C-5), 106.3 (C-4), 24.5 (CH3).
(4-iodo-2-pyridyI)methylacetate: XH NMR (CDCI3) δ 8.16 (d, IH, =5.2 Hz, H-6); 7.66 (d, IH, J=1.0 Hz, H-3); 7.54 (dd, IH, =5.2, J,=1.6 Hz, H-5); 5.09 (s, 2H, CH2); 2.11 (s, 3H, CH3); 13C NMR (CDCI3) δ 170.9 (CO), 157.1 (C-2), 150.1 (C-6), 132.5 (C-3), 131.3 (C-5), 106.6 (C-4), 66.3 (CH2), 21.3 (CH3).
2-(((re/ -butyldimethylsilyl)oxy)methylV4-chloropyridine:
Figure imgf000047_0001
To a mixture of 4-chloro-2-(hydroxymethyl)pyridine (360 mg, 2.51 mmol) and imidazole (684 mg, 10.04 mmol) in DMF (2 mL) was added fBuMe2SiCI (452 mg, 3.0 mmol) in several portions at room temperature. The mixture was stirred for 15 min and extracted with ether (50 mL). The extract was washed with water (5 mL x 3), dried over MgS04, and concentrated under reduced pressure. The residue was purified by column chromatography on silica gel eluted with CH2CI2 to give the title compound (600 mg, 93%) as a colourless oil. H NMR (CDCI3) δ 8.30 (d, IH, J=5.3 Hz, H-6); 7.44 (d, IH, J=l.l Hz, H-3); 7.07 (dd, IH, =5.3, J,=2.0 Hz, H-5); 4.73 (s, 2H, CH2); 0.88 (s, 9H); 0.05 (s, 6H); 13C NMR (CDCI3) δ 163.7 (C-2), 150.0 (C-6), 145.3 (C-4), 122.6 (C-3), 120.8 (C-5), 65.9 (CH2), 26.3
Figure imgf000047_0002
18.7 ((CH3 3C), -4.6 ((Me)2Si).
4-Chloro-2-(hydroxymethyl)pyridine:
Figure imgf000047_0003
A solution of 4-chloropyridine ΛAoxide (5 g, 38.6 mmol) and trimethyloxonium tetrafluoroborate (5.94 g, 40.1 mmol) in CH2CI2 (115 mL) was stirred for two hours at ambient temperature. The solvent was evaporated and the residue taken up in MeOH (115 mL) and heated to near boiling. Ammonium persulfate (1.76 g, 7.72 mmol) dissolved in H20 (7.7 mL) was added and the mixture was heated to reflux for 30 min. A second portion of ammonium persulfate (0.88 g) in H20 (3.9 mL) was added and the mixture was refluxed for another 30 min. The solvent was evaporated and the residue was partitioned between CH2CI2 and aqueous Na2C0 (10% w/v). The organic layer was washed with H20, dried over MgS04, and evaporated leaving 2.4 g (43%) of the title compound. i NMR (CDCI3) δ 8.20 (d, IH, J=5.0 Hz, H-6); 7.31 (s, IH, H-3); 7.04 (d, IH, J=5.0 Hz, H-5); 5.46 (s, IH, OH); 4.61 (s, 2H, CH2).
Example 3
Ag-(2-picotyl) adenosine-5'-/\Λmethyluronamide :
Figure imgf000048_0001
Procedure A
To a solution of 5'-ΛAmethylcarboxamidoadenosine (64 mg, 0.22 mmol) in DMF (1 mL) was added 2-picolyl chloride hydrochloride (129 mg, 0.79 mmol) and triethylamine (0.11 mL, 0.79 mmol). The solution was stirred for 3 days at 40 °C. DMF was removed under vacuum giving a syrup that crystallized when acetone and ether were added. The solvent was removed using a Pasteur pipette. The residue was treated with methanol (1.5 mL), and concentrated NH4OH (3.0 mL) was added. The mixture was warmed in a closed tube at 90 °C for 2 h with stirring. The solvent was evaporated and the residue purified by column chromatography (CH^I^MeOH 9:1) yielding 17 mg (20% yield overall) of the title compound as a white foaming solid and 40 mg of starting material.
Procedure B
2',3'-- Isopropylidene-6-chloropurine-5'-methyluronamide (150 mg, 0.43 mmol), 2-(aminomethyl) pyridine (44 μL, 0.43 mmol), and triethylamine (0.18 mL, 1.26 mmol) were dissolved in absolute ethanol (1.5 mL). The solution was stirred at 65°C for 16 h in a sealed vessel. The solvent was removed under nitrogen. HCI (1 N) (1.0 mL) was added and the solution stirred at 70°C for 45 min. After cooling, NaHC03 was added until pH 7, and a white solid precipitated. The solid was filtered and washed with cold water yielding 94 mg (57% yield overall) of the title compound. Rf (CH2Cl2/MeOH 9:1) 0.3; m.p. 84-86 °C; H NMR (CD3OD) δ 8.54 (m, IH, Hpyridyr6), 8.35 (s, IH, H- 8), 8.31 (s, IH, H-2), 7.81 (dt, IH, J=7.8 Hz, J=1.8 Hz, HpyridyI-4), 7.47 (d, IH, J=7.8 Hz, Hpyridv)- 3), 7.34 (dd, IH, J=6.6 Hz, J=5.1 Hz, Hpyridy|-5), 6.05 (d, IH, J ,2'=7.8 Hz, H-l1), 4.93 (not observed, 2H, CH2), 4.78 (dd, IH, Jr,2'= 7.8 Hz, J,-,^ -8 Hz, H-2 , 4.52 (d, IH, ^-=1.0 Hz, H- 4 , 4.35 (dd, IH, Jy,2-=4.8 Hz, ^-=1.0 Hz, H-3 , 2.90 (s, 3H, 5'- V-methyl). 13C NMR (CD3OD) δ 173.2 0-5 160.0 (Cpyridyr2), 156.7 (C-6), 154.3 (C-2), 150.2 (Cpyridyr6), 142.8 (C-8), 139.2 (Cpyridyι-4), 124.2 (Cpyridyr3), 123.2 (Cpyridyl-5), 122.2 (C-5), 90.9 (C-1 , 86.9 (C-4 , 75.3, 73.8 (C-3', C-27), 46.7 (CH2), 26.4 (5'-/V-methyl). High-resolution MS calcd for (C17 H19 N7 04 Na) 408.1396, found 408.1392. Anal. (C17H19N704) C, H, N.
Example 4 /Vg-(6-acetyl-2-picolyl)adenosine-5'-/\Λmethyluronamide:
Figure imgf000049_0001
To a solution of 5'-ΛAmethylcarboxamidoadenosine (75 mg, 0.26 mmol) in DMF (1.0 mL) was added 2-(acetyl)-6-bromomethylpyridine (83.5 mg, 0.39 mmol), and the solution stirred for 3 days at 40 °C. The solvent was evaporated and the residue treated with methanol (1.0 mL) and concentrated NH40H (2.0 mL). The mixture was warmed in a closed vessel at 90 °C for 3 h with stirring. After evaporating the solvent, the residue was chromatographied (4% MeOH in CH2CI2) and 63 mg (57%) of the title compound (a white solid) were obtained. Rf (CH2Cl2/MeOH 9:1) 0.6; m.p.(MeOH): 226-228°C; *H NMR (CD3OD) δ 8.35 (s, IH, H-8), 8.32 (s, IH, H-2), 7.91 (m, 2H, Hpyridyr3,5), 7.63 (dd, IH, J=5.5 Hz, J=3.3 Hz, Hpyridyr4), 6.05 (d, IH, Jl -=7.8 Hz, W-V), 4.9 (not observed, 2H, CH2), 4.77 (dd, IH, J,<r= 7.8 Hz, -^-,3-=4.8 Hz, H-2 , 4.50 (s, IH, H-4 , 4.33 (dd, IH, J3',2'=4.8 Hz, J3',4'=1.4 Hz, H-3 , 2.89 (s, 3H, 5'-/V-methyl), 2.67 (s, 3H, CH3CO); 13C NMR (DMSO- ) δ 199.8 (CH3CO), 170.2 (C-5 , 159.3 (Cpyridyr2), 155.0 (C-6), 152.9 (Cpyridyr6), 152.7 (C-2), 148.7 (C-4), 141.3 (C-8), 138.4 (Cpyridyl-4), 124.8 (Cpyridyr3), 120.5 (C-5), 119.7 (Cpyridy,-5), 88.1 (C-1 , 85.0 (C-4 , 73.4, 72.5 (C-3', C-20, 45.4 (CH2), 25.9, 25.7 (5'-/V-methyl, CH3CO). High- resolution MS calcd for (C19 H21 N7 Os Na) 450.1502, found 450.1503.
2-Acetyl-6-bromomethylpyridine:
Figure imgf000049_0002
To a mixture of the alcohol 2-Acetyl-6-(hydroxymethyl)pyridine (450 mg, 2.98 mmol) and CBR4 (1.59 g, 4.8 mmol) in CH2CI2 (8.5 mL) was added triphenylphosphine (923 mg, 3.5 mmol) in several portions at 0 °C. The reaction mixture was stirred for 20 min at the same temperature and then directly passed through a short silica gel column using 10% EtOAc in hexane as an eluent to give the title compound as a transparent oil (300 mg) in 47% yield. Rf (hexane/EtOAc 9:1) 0.44; H NMR (CDCI3) δ 7.92 (d, IH, J=7.7 Hz, H-3), 7.82 (t, IH, J=7.7 Hz, H-4), 7.61 (d, IH, .7=7.7 Hz, H-5), 4.58 (s, 2H, CH2Br), 2.70 (s, 3H, CH3CO); 13C NMR (CDCI3) δ 200.3 (CO), 156.8 (C-6), 153.6 (C-2), 138.3 (C-4), 127.3 (C-5), 121.1 (C-3), 33.8 (CH2Br), 26.2 (CH3); mass spectrum (ES+): m/e 213.8 (M++l).
2-Acetyl-6-(hydroxymethyl)pyridine:
Figure imgf000050_0001
To a THF (10 mL) solution of the silyl ether 2-Acetyl-6-(((tert- butyldimethylsilyl)oxy)methyl)pyridine (0.8 g, 3.0 mmol) was added tetrabutylammonium fluoride hydrate (870 mg, 3.3 mmol) at room temperature, and the mixture was stirred for 40 min. It was extracted with EtOAc (150 mL) and washed with water (6 mLx3) and brine (6 mL). The extract was dried over MgS04 and evaporated under reduced pressure. The residue was purified by column chromatography on silica gel eluted with hexane/EtOAc 4:1 to give the title compound (450 mg, 99%) as an oil. H NMR (CDCI3) δ 8.00 ( d, IH, J=7.2 Hz, H-3), 7.88 (m, IH, H-4), 7.49 (d, IH, J=7.7 Hz, H-5), 4.88 (d, 2H, J= 5.0 Hz, CH2), 3.84 (d, IH, J= 3.2 Hz, OH), 2.78 (s, 3H, CH3); 13C NMR (CDCI3) δ 200.0 (CO), 159.1 (C-6), 152.7 (C-2), 138.0 (C-4), 124.4 (C-5), 120.8 (C- 3), 64.3 (CH2), 26.3 (CH3).
2-Acetyl-6-(((fe/t-butyldimethylsilyDoxy)methy0pyridine:
Figure imgf000050_0002
To a stirred solution of 2-Bromo-6-(((tert-butyldimethylsilyl)oxy)methyi)pyridine (1.59 g, 5.26 mmol) in a mixture of ether, hexane and THF (2:1:1, 105 mL) was added n-BuLi (3.29 mL, 5.26 mmol, 1.6 M in hexane solution) dropwise at -78°C during 5-10 min. To the resulting dark brown solution was dropped anhydrous DMA (0.74 mL, 7.89 mmol) at the same temperature. The reaction mixture was stirred for 30 min and then quenched with water (8 mL) and extracted with EtOAc. The organic layer was washed with water and brine and dried over MgS04. After purification by column chromatography on silica gel (hexane/EtOAc 98:2) 0.78 g (56%) of the title compound as an oil were obtained. XH NMR (CDCI3) δ 7.95 ( d, IH, J=7.5 Hz, H-3), 7.88 (t, IH, J=7.6 Hz, H-4), 7.73 (t, IH, J=7.6 Hz, H-5), 4.93 (s, 2H, CH2), 2.75 (s, 3H, CH3), 1.03 (s, 9H, (CH3)3C), 0.20 (s, 6H, (Me)2Si); 13C NMR (CDCI3) δ 200.8 (CO), 161.4 (C-6), 153.0 (C-2), 139.4 (C- 4), 124.0 (C-5), 118.0 (C-3), 66.4 (CH2), 26.3 ((Ch^C), 18.8 ((CHs)^), -4.96 ((Me)2Si).
2-Bromo-6-(((te t-butyldimethylsilyπoxy)methyl)pyridine:
Figure imgf000051_0001
To a mixture of 2-bromo-6-hydroxymethylpyridine (1.03 g, 5.48 mmol) and imidazole (1.49 g, 21.92 mmol) in DMF (5 mL) was added fBuMe2SiCI (991 mg, 6.58 mmol) in several portions at room temperature. The mixture was stirred for 15 min and extracted with ether (50 mL). The extract was washed with water (5 mL x 3), dried over MgS04, and concentrated under reduced pressure. The residue was purified by column chromatography on silica gel eluted with 2.5% of EtOAc in hexane to give the title compound (1.59 g, 96%) as an oil. H NMR (CDCI3) δ 7.65-7.40 (m, 3H), 4.89 (s, 2H, CH2), 1.04 (s, 9H), 0.20 (s, 6H); 13C NMR (CDCI3) δ 163.6 (C-6), 141.3 (C-2), 139.4 (C-4), 126.4 (C-3), 119.1 (C-5), 65.9 (CH2), 26.3 ((CH^C), 18.8 ((O^aC), -5.0 ((Me)2Si).
2-Bromo-6-(methylaminomethylcarbonyl)pyridine and 2-bromo-6-hydroxymethyl--pyridine:
Figure imgf000051_0002
To a stirred solution of 2,6-dibromopyridine (3 g, 12.7 mmol) in a mixture of THF (8 mL), ether (16 mL), and hexane (8 mL) was added n-BuLi (7.94 mL, 12.7 mmol, 1.6 M in hexane) drop-by- drop at -78°C over 5 min. After the mixture was stirred for 5 min, DMF (2.01 mL, 26.3 mmol) was slowly added dropwise to the mixture over 10 min at the same temperature. The reaction mixture was warmed to -50°C and quenched with MeOH (15 mL). Then, NaBH4 (487 mg, 12.9 mmol) was added at room temperature. After addition of acetone (1.5 mL), the mixture was diluted with EtOAc (300 mL). The whole was washed with water (6 mL x 3) and brine (6 mL) and dried over MgS04. Solvent was evaporated and the residue was purified by column chromatography on silica gel eluted with 20% EtOAc in hexane to give 380 mg (13%) of 2-Bromo-6- (methylaminomethylcarbonyl)pyridine and 1.11 g (46%) of the oil 2-bromo-6- hydroxymethylpyridine.
Bromo-6-(methylaminomethylcarbonyl)pyridine: H NMR (CDCI3) δ 7.53 (m, IH, H-4), 7.38 (d, IH, J=7.9 Hz), 7.28 (d, IH, J=7.5 Hz), 5.14 (s, 2H, CH2), 2.12 (s, 3H, Me); 13C NMR (CDCI3) δ 170.9 (CO), 157.7 (C-6), 142.1 (C-2), 139.5 (C-4), 127.7 (C-3), 120.9 (C-5), 66.4 (CH2), 21.3 (Me). Mass spectrum (ES+): m/e 230 (M++l).
2-bromo-6-hydroxymethylpyridine: *H NMR (CDCI3) δ 7.56 (t, IH, J=7.7 Hz), 7.46 (d, IH, j=7.7 Hz), 7.37 (d, IH, .7=7.7 Hz), 4.85 (s, 2H, CH2); 13C NMR (CDCI3) δ 162.0 (C-6), 141.7 (C-2), 139.6 (C-4), 124.8 (C-3), 119.8 (C-5), 64.6 (CH2). Example 5
Λ^-(4-iodo-2-picolyl)-2-(2-phenyl-l-ethynyl)-adenosine-5'- y methyluronamide:
Figure imgf000052_0001
N-Methyl-l'-deoxy-l'-[6-chloro-2-(2-phenyl-l-ethynyl)-9H-purin-9-yl]-2',3'-0-isopropylidene-β-D- ribofuranuronamide (210 mg, 0.47 mmol), (4-iodo-2-pyridyl)methylamine (219 mg, 0.93 mmol), and triethylamine (0.2 mL, 1.41 mmol) were dissolved in absolute ethanol (3.0 mL). The solution was stirred at 65°C for 16 h in a sealed vessel. The solvent was removed under nitrogen and the 2',3'-CMsopropylidene derivative was purified by column chromatography (2% MeOH in CH2CI2) yielding 230 mg . HCI (1 N) (3.0 mL) were added and the solution stirred at 70°C for 45 min. A white precipitate appeared immediately. After cooling, NaHC03 was added until pH 7, and a white solid precipitated. The solid was filtered and washed with water yielding 138 mg (48% yield overall) of the title compound. H NMR (DMSO- ) δ 8.69 (s, IH, NH), 8.60 (s, 2H, NHCO, H-8), 8.26 (d, IH, J=5.0 Hz, H-6py), 7.87 (s, IH, H-3py), 7.78 (d, IH, J=5.0 Hz, H-5py), 7.62 (2H, m, C6H5), 7.47 (3H, m, CsHs), 6.02 (d, IH, J=7.5 Hz, H-1 , 4.82 (bs, 2H, CH2-N), 4.63 (dd, IH, Jι=7.5, J2=4.6 Hz, H-2 , 4.35 (s, IH, H-40, 4.18 (d, IH, J=4.1 Hz, H-3 , 2.78 (d, 3H, J=4.5 Hz, 5-/lAmethyl). 13C NMR (DMSO-d6) δ 170.2 (CO), 159.6 C-2py), 154.5 (C-6), 149.3(C-6py), 148.9 (C- 4), 145.5 (C-2), 142.4 (C-8), 132.2 (C-Ha), 131.7 (C-5py), 130.8 (para-C), 130.1 (C-3py), 129.3 (C- Hb), 121.2 (ipsσ-C), 90.0 (β-alkynyl), 88.0 (C-1 , 85.0 (C-4 , 84.1 (α-alkynyl), 73.4 (C-3 , 72.5, (C-2 , 44.4 (CH2), 26.0 (5'-/IAmethyl). Anal. (QSH^C I) C, H, N. High-resolution MS calcd for (CzsH^N^Nal) 634.0684, found 634.0676.
iV-methyl-l'-deoxy-l'- 6-chloro-2-(2-phenyl-l-ethynyl)-9 j purin-9-yl]-2'f3'-t isopropylidene-β-D- ribofuranuronamide:
Figure imgf000053_0001
To a solution of 500 mg (1.04 mmol) of N-methyl-l'-deoxy-l'-(6-chloro-2-iodo-9H-purin-9-yl)- 2',3'-0-isopropylidene-β-D-ribofuranuronamide in 10 mL of dry acetonitrile, and 4.5 mL of triethylamine under an argon atmosphere was added 14.5 mg (0.0206 mmol) of bϊs(triphenylphosphine)palladium dichloride and 1.0 mg (0.0054 mmol) of cuprous iodide. To the mixture was added phenylacetylene (0.17 mL, 1.56 mmol), and the reaction mixture was stirred at room temperature for 1 hour. The solvent was removed in vacuo, and the residue was chromatographed on a silica gel column (2% MeOH in CH2C!2) to give 454 mg (96%) of the title compound. H NMR (CDCI3) δ 8.29 (s, IH, H-8), 7.73 (2H, m, H5), 7.47 (3H, m, C6H5), 7.13 (m, IH, NH), 6.17 (d, IH, J=3.5 Hz, H-1 , 5.35 (m, 2H, H-3', H-2 , 4.82 (d, IH, J=1.8 Hz, H-4 , 2.86 (d, 3H, J=4.9 Hz, 5'-Λ<-methyl), 1.70, 1.44 (2 s, 2 3H, 2 CH3, isopropylidene). 13C NMR (CDCI3) δ 169.3 (CO), 152.3, 151.3, 146.6 (C-2), 145.6 (C-8), 133.0 (C-Ha), 132.1 (C-5), 130.6 (para-C), 129.1 (C-Hb), 121.2 (ipso-C), 115.6 (Cq, isopropylidene), 92.8 (C-10, 89.5 (β-alkynyl), 87.8 αalkynyl), 85.6 (C-4*), 83.7, 82.7 (C-3', C-2 , 27.7, 26.5, 25.6 (2 CH3, isopropylidene, 5'-/V- methyl). Mass spectrum (ES+): m/e 454 (M++l).
Λt-methyl-l'-deoxy-l,-(6-chloro-2-iodo-9 purin-9-yl)-2'f3'-r isopropylidene-Jg-D- ribofuranuronamide:
Figure imgf000053_0002
Freshly isoamyl nitrite (0.74 mL, 5.47 mmol) was added to a mixture of N-methyl- -deoxy-l'-(2- amino-6 hloro-9H-purin-9-ylJ-2/3-0-isopropylidene-β-D-ribofuranuronamide (618 mg, 1.68 mmol), Cul (339 mg, 1.78 mmol), CH2I2 (1.35 mL, 16.8 mmol), and I2 (426 mg, 1.68 mmol) in dry THF (6.0 mL) at room temperature and under argon. The reaction mixture was refluxed for 30 min, cooled to ambient temperature, filtered to remove insolubles, and then evaporated. The product was purified by flash chromatography. Iodine was first eluted from the column with 100% of CH2CI2 and then the title compound was eluted with 2% MeOH in CH2CI2 in 62% yield. *H NMR (CDCI3) δ 8.25 (s, IH, H-8), 6.60 (m, IH, NH), 6.19 (d, IH, J=2.8 Hz, H-l7), 5.37, 5.29 (m, 2H, H- 3', H-27), 4.74 (d, IH, J=2.1 Hz, H-47), 2.75 (d, 3H, J=4.9 Hz, 5'-/IAmethyI), 1.63, 1.40 (2 s, 2 3H, 2 CH3, isopropylidene). 13C NMR (CDCI3) δ 169.1 (CO), 152.1, 151.5, 145.2 (C-8), 132.7 (C-5), 117.4 (C-2), 115.4 (Cq, isopropylidene), 92.2 (C-1Q, 86.5 (C-47), 83.6, 83.3 (C-3', C-27), 27.4, 26.8, 25.6 (2 CH3, isopropylidene, 5'-/V-methyl).
N-methyl-l,-deoxy-l'-(2-amino-6-chloro-9H-purin-9-yl)-2'.3'-Q-isopropylidene-β-D- ribofuranuronamide:
Figure imgf000054_0001
To a mixture of pre-dried 5'-N-Methyl-2,'3'-0-isoprapylidenecarboxamidoguanosine (950 mg, 2.71 mmol), Et4NCI (898 mg, 5.42 mmol, pre-dried in vacuo at 90 °C overnight over P205), N, N- dimethylaniline (0.34 mL, 2.71 mmol) in anhydrous acetonitrile (20 mL), 1.5 mL (16.26 mmol) of phosphoryl chloride were added at room temperature. The flask was placed in an oil bath pre- heated to 100°C and the solution was heated with stirring at reflux for 10 minutes. Volatile materials were flash evaporated immediately in vacuo. The resulting yellow foam was dissolved in 15 mL of CHCI3 and stirred vigorously with crushed ice for 15 minutes. The layers were separated and the aqueous phase was extracted with 5 x 5 mL of CHC13. The combined organic phase was kept cold by addition of crushed ice. It was washed with cold water, 5% of NaHC03 to pH~7, dried over MgS04, and filtered. The product was purified by column chromatography (3% MeOH in CH2CI2) to give 840 mg (84%) of the title compound as a white crystalline product. XH NMR (CDCI3) δ 7.87 (s, IH, H-8), 6.35 (m, IH, NH), 6.12 (d, IH, .7=1.2 Hz, H-l7), 5.70 (s, 2H, NH2), 5.66 (dd, IH,
Figure imgf000054_0002
Hz, H-37), 5.29 (d, IH, J=l.l Hz, H-27), 4.73 (d, IH, 1.6 Hz, H-47), 2.52 (d, 3H, .7=4.9 Hz, 5'-Λ/-methyl), 1.59, 1.39 (2 s, 2 3H, 2 CH3, isopropylidene). 13C NMR (CDCI3) δ 170.3 (CO), 151.9, 159.6 (C-2), 153.0, 152.1, 142.0 (C-8), 125.5 (C-5), 114.3 (Cq, isopropylidene), 91.9 (C-l7), 88.5 (C-47), 84.1, 84.2 (C-37, C-27), 27.1, 25.5 ( 2 CH3, isopropylidene), 26.1 (5'-/V-methyl).
57-N-Methyl-27 r3 -Q-isopropylidenecarboxamidoguaπosine:
Figure imgf000055_0001
2',3'-c Isopropylideneguanosine-5'-carboxylic acid (1.6 g, 4.6 mmol) was coevaporated with DMF (3 x 20 mL) and taken up in DMF (20 mL) with Et3N (0.94 mL, 6.9 mmol) under N2 atmosphere. After the solution was cooled to 0 °C, the coupling reagent isopropenyl chloroformate (1.0 g, 8.30 mmol) and methylamine 2.0 M in THF (5.3 mL, 10.6 mmol) were added subsequently. The reaction mixture was stirred for 15 min at 0°C. The mixtures were concentrated under reduce pressure, redissolved in CH2CI2 washed with NaHC03 solution (10%) and water, dried on MgS04 and concentrated. The remaining oils were purified by column chromatography (eluent CHzClz/MeOH 9:1) and 693 mg (43%) of the title compound were obtained. H NMR (CD3OD) δ 7.87 (s, IH, H-8), 6.21 (s, IH, H-l7), 5.64 (dd, IH,
Figure imgf000055_0002
1.9 Hz, H-27), 5.37 (d, IH, 6.1 Hz, H-37), 4.63 (s, IH, H-47), 2.47 (s, 3H, 5'- V-methyl), 1.56, 1.40 (2 s, 2 3H, 2 CH3, isopropylidene).
Example 6
//6-r(2-dimethylamino-7-iodo-lf3-benzoxazol-5-yl)-methyll- adenosine-57t-methyl uronamide:
Figure imgf000055_0003
2',3'-c Isopropylidene-6-chloropurine-57-methyluronamide (72 mg, 0.2 mmol), N2, N2-dimethyl-5- (aminomethyl)-7-iodo-l,3-benzoxazol-2-amine (64 mg, 0.2 mmol), and triethylamine (0.084 mL, 0.6 mmol) were dissolved in absolute ethanol (1.0 mL). The solution was stirred at 65°C for 16 h in a sealed vessel. The solvent was removed under nitrogen. HCI (1 N) (1.0 mL) was added and the solution stirred at 70°C for 45 min. After cooling, NaHC03 was added until pH 7, and a white solid precipitated. The solid was filtered and washed with water yielding 60 mg (50% yield overall) of the title compound. M.p. 196-198 °C; XH NMR (DMSO-fl δ 8.92 (bs, IH, NHCO), 8.61 (s, IH, NH), 8.46 (s, IH, H-8), 8.33 (s, IH, H-2), 7.32 (s, IH, Hbenzoχazor6), 7.20 (s, IH, Hb-4), 5.98 (d, IH, Jr,2.=7.6 Hz, H-l7), 5.77 (d, IH, J= 2.5 Hz, OH-37), 5.58 (d, IH, J= 5.1 Hz, OH-27), 4.70 (s, 2H, -CH2N-), 4.60 (bs, IH, H-27), 4.32 (s, IH, H-47), 4.15 (bs, IH, H-37), 3.11 (s, 6H, (CH3)2N), 2.72 (d, 3H, J=4.5 Hz , 5'-/V-methyl). 13C NMR (DMSO-cQ δ 168.5 (C-57), 160.6 (Cb-2), 153.0 (C- 6), 151.2 (C-2), 147.8 (C-4), 146.1 (Cb-7a), 141.6 (Cb-3a), 139.4 (C-8), 136.7 (Cb-5), 125.8 (Cb-6), 118.7 (C-5), 113.0 (Cb-4), 86.4 (C-l7), 83.3 (C-47), 71.7, 70.7 (C-37, C-27), 69.7 (Cb-7), 47.2 (CH2), 35.9 ((CH3)2N), 24.0 (5'-/V-methyl). Anal. (C^HaNsOsI) C, H, N.
Synthesis of N2, 7V -dimethyl-5-(aminomethyl)-7-iodo-lr3-benzoxazol-2-amine
The N2, /\t2-dimethyl-5-(aminomethyl)-7-iodo-l,3-benzoxazol-2-amine used in the above synthesis was made by the following procedure.
3-Bromo-4-hydroxy-5-nitrobenzonitrile:
Figure imgf000056_0001
To a mixture of 5 g (30.5 mmol) of 4-hydroxy-3-nitrobenzonitrile in H2S04 solution (50 mL of concentrated H2S04 + 50 mL of H20) at 25 °C, 7.9 g (47.3 mmol) of potassium bromate were added in small portions cooling the flask with an ice-bath and maintaining the temperature between 25 and 35 °C. After the addition was completed, the reaction was stirred at room temperature for 22 h and then filtered. The pale yellow solid was washed with water and dried to give 4.2 g (57%) of the title compound. M.p.: 162-164 °C; *H NMR (CD3OD) δ 8.54 (d, IH, J=2.0 Hz, H-6), 8.32 (d, IH, J=2.0 Hz, H-2); 13C NMR (DMSO-ok) δ 153.9 (C-4), 140.8 (C-2), 138.2 (C- 5), 130.1 (C-6), 117.0 (CN), 115.9 (C-3), 101.6 (C-l).
2-amino-4-(aminomethyl)-6-bromophenol:
Figure imgf000056_0002
A solution of 3-bromo-4-hydroxy-5-nitrobenzonitrile (1.0 g, 4.11 mmol) in 20.0 mL of dry THF was added dropwise to a solution of BHs/THF 1.0 M in THF (12.3 mL) at 0°C under N2. The reaction mixture was stirred at room temperature for 24 hours. Excess borane was decomposed by the cautious dropwise addition of 1 N HCI at 0°C. It was extracted with EtOAc, washed with NaHC03, NaCI saturated solution and dried (Na2S04 anh.) to give a crude. After purification by column chromatography (CH2Cl2/MeOH 8:2+ 1% NH4OH), 170 mg (19%) of 2-amino-4-(aminόmethyl)-6- bromophenol were obtained. H NMR (DMSO-ck) δ 6.67 (s, IH, H-5); 6.53 (s, IH, H-3); 3.52 (s, 2H, CH2). 13C NMR (DMSO-tf6) δ 139.7, 139.4 (C-l, C-4), 136.0 (C-2), 118.5 (C-5), 113.0 (C-3), 111.5 (C-6), 44.9 (CH2).
re/f-butyl/if(3-amino-5-bromo-4-hydroxybenzyl)carbamate:
Figure imgf000057_0001
To a solution of 2-amino-4-(aminomethyI)-6-bromophenoI (560 mg, 2.59 mmol) in 15 mL of DMF, triethylamine (0.37 mL, 2.63 mmol) and di-te -butyldicarbonate (0.59 mL, 2.59 mmol) were added. The mixture was stirred at room temperature for 1 hour and 15 minutes. The solvent was evaporated and the residue was suspended in EtOAc and filtered. The solution was extracted with 0.5 M NaH2P04 (3x) and dried. The product was purified by column chromatography (2% MeOH in CH2CI2) yielding 304 mg (37%) of fetf-butyl/lA(3-amino-5-bromo-4-hydroxybenzyl)carbamate. *H NMR (CDCI3) δ 6.76 (d, IH, J=1.8 Hz, H-6); 6.57 (d, IH, J=1.2 Hz, H-2); 4.13 (d, 2H, J=5.8 Hz, CH2); 1.49 (s, 9H, (CH3)3C); 13C NMR (CDCI3) δ 156.5 (CO), 140.1 (C-4), 136.9 (C-l), 133.0 (C-3), 120.3 (C-6), 114.3 (C-2), 110.7 (C-5), 79.8 (CH^), 44.2 (CH2), 28.8 ((CHj)3C).
7aη7--butyl ι/-fr7-bromo-2-(dimethylamino)-lf3-benzoxazol-5-yπmethyl carbamate :
Figure imgf000057_0002
A mixture of te/f-butyl/V-(3-amino-5-bromo-4-hydroxybenzyI)carbamate (300 mg, 0.95 mmol) and dichloromethylenedimethylammonium chloride (phosgene iminium chloride, 240 mg, 1.48 mmol) in 10 mL of dry CH2CI2 was refluxed for 6 h. After cooling the solution was extracted with EtOAc, washed with NaHC03, brine and dried (MgS04). After purification by column chromatography (1% MeOH in CH2CI2) 192 mg (55%) of te/t-butyl/V-{[7-bromo-2-(dimethylamino)-l,3-benzoxazol-5- yl]methyl} carbamate were obtained. XH NMR (CDCI3) δ 7.14 (s, IH, H-6); 7.04 (s, IH, H-4); 5.11 (bs, IH, NH); 4.31 (d, 2H, J=5.6 Hz, CH2); 3.23, 3.22 (s, 6H, NCH3 2x); 1.48 (s, 9H, (CH3)3C); 13C NMR (CDCI3) δ 163.3 (C-2), 156.2 (CO), 146.6 (C-7a), 145.1 (C-3a), 137.0 (C-5), 122.8 (C-6), 114.3 (C-4), 100.6 (C-7), 79.9 ((CH3)3C , 44.7 (CH2), 38.1 (NCH3 x2), 28.8 ((CH^C).
rerf:-butyl l/-f[2-(dimethylamino)-7-iodo-lf3-benzoxazol-5-yl]methyl}carbamate :
Figure imgf000058_0001
mixture of te/f-butyl/V-{[7-bromo-2-(dimethylamino)-l,3-benzoxazol-5-yI]methyl} carbamate (192 mg, 0.52 mmol), Kl (1.3 g, 7.8 mmol), Cul (495 mg, 2.6 mmol) in 1.6 mL of HMPA was stirred overnight (16 h) at 135°C. After cooling, the mixture was extracted with EtOAc, washed with Na2S203 0.5 M solution, and brine (x3). After purification by column chromatography
(hexane/EtOAc 6:4), 120 mg (55%) of te/f-butyl V-{[2-(dimethylamino)-7-iodo-l,3-benzoxazol-5- yl]methyl}carbamate were obtained. *H NMR (CDCI3) δ 7.32 (s, IH, H-6); 7.27 (s, IH, H-4); 5.02 (bs, IH, NH); 4.40 (d, 2H, J=5.4 Hz, CH2); 3.32 (s, 6H, NCH3 2x); 1.57 (s, 9H, (CH3)3C); 13C NMR (CDCI3) δ 162.7 (C-2), 156.2 (CO), 150.4 (C-7a), 143.8 (C-3a), 137.4 (C-5), 128.4 (C-6), 115.3 (C- 4), 80.0 ((CH3)3C), 70.5 (C-7), 44.6 (CH2), 38.1 (NCH3 x2), 28.8 ((CHVKC).
NI, /ιg-dimethyl-5-(aminomethyl)-7-iodo-lf3-benzoxazol-2-amine:
Figure imgf000058_0002
To a solution of te*-butyl/V-{[2-(dimethylamino)-7-iodo-l,3-benzoxazol-5-yl]methyl}carbamate (120 mg, 0.29 mmol) in 4 ml of CH2CI2 was added slowly 0.5 mL of trifluoroacetic acid and it was stirred at room temperature for 30 min. The excess of TFA was removed under vacuum. The product was purified by column chromatography (CHzCI^MeOH 9:1) yielding 64 mg (70%) of NI, Λβ-dimethyl-5-(aminomethyl)-7-iodo-l,3-benzoxazol-2-amine as a white solid. l NMR (CD3OD) δ 7.46 (d, IH, J=1.5 Hz, H-6); 7.30 (d, IH, J=1.5 Hz, H-4); 4.12 (s, 2H, CH2); 3.23, 3.24 (s, 6H, CH3X2); 13C NMR (CD3OD) δ 164.5 (C-2), 152.7 (C-7a), 144.6 (C-3a), 132.9 (C-5), 131.6 (C-6), 117.3 (C-4), 72.0 (C-7), 44.2 (CH2), 38.3 (CH3).
Example 7
Λt5-r(2-Dimethylamino-1.3-benzoxazol-5-yl)-methyl]- adenosine-57-/l/-methyluronamide:
Figure imgf000059_0001
2',37-0ϊsopropylidene-6-chloropurine-57-methyluronamide (100 mg, 0.28 mmol), N2, N2-dimethyl- 5-(aminomethyl)-l,3-benzoxazol-2-amine (80.3 mg, 0.42 mmol), and triethylamine (0.117 mL, 0.84 mmol) were dissolved in absolute ethanol (1.5 mL). The solution was stirred at 65°C for 16 h in a sealed vessel. The solvent was removed under nitrogen. The 27,37-CMsopropylidene derivative was purified by column chromatography (3% MeOH in CH2CI2). HCI (1 N) (1.2 mL) was added and the solution stirred at 70°C for 45 min. After cooling, NaHC03 was added until pH 7, and the solution was left overnight crystallizing. The white solid was filtered and washed with water yielding 68 mg (52%) of the title compound. M.p.: 239-241°C; XH NMR (DMSO-cQ δ 8.94 (bs, IH, NHCO), 8.57 (s, IH, NH), 8.44 (s, IH, H-8), 8.33 (s, IH, H-2), 7.29 (d, IH, J= 8.2 Hz, Hbenzoxazor 7), 7.24 (s, IH, Hb-4), 6.99 (d, IH, J= 8.2 Hz, Hb-6), 5.98 (d, IH, Jr,2-=7.5 Hz, H-l7), 5.75 (s, 2H, OH-2',37), 4.73 (s, 2H, -CH2N-), 4.61 (dd, IH, J2-,r=7.5 Hz, J2',3'=4.6 Hz, H-27), 4.33 (s, IH, H-47), 4.15 (d, IH, J3',2'=4.6 Hz, H-37), 3.1 (s, 6H, (CH3)2N), 2.73 (d, 3H, J=3.9 Hz , 5'-/V-methyl); 13C NMR (DMSO-cQ δ 168.4 (C-57), 161.4 (Cb-2), 153.0 (C-4), 151.0 (C-2), 146.7 (C-6), 146.1 (Cb-7a), 142.0 (Cb-3a), 139.2 (C-8), 134.2 (Cb-5), 118.5 (C-5), 117.6 (Cb-6), 112.9 (Cb-4), 106.7 (Cb-7), 86.3 (C-l7), 83.2 (C-47), 71.6, 70.5 (C-37, C-27), 41.5 (CH2), 35.7 ((CH3)2N), 23.9 (5'-/V-methyl). High-resolution MS calcd for (C21 H24 N8 05Na) 491.1767, found 491.1749. Anal. (C21H24N805) C, H, N.
N2, /V2-Dimethyl-5-(aminomethyl)-lf3-benzoxazol-2-amine:
Figure imgf000059_0002
To a mixture of 2-(Dimethylamino)-lf3-benzoxazole-5-carbonitrile (1.5 g, 8.0 mmol) in 150 mL of dry ether, 640 mg (16.0 mmol) of LiAIH4 were slowly added. The mixture was stirred at room temperature for 1.5 h and the excess of reagent was decomposed by careful addition of the minimum amount of water and filtered. The solids were washed with hot EtOAc (3x100 mL) giving a residue after evaporation. The purification by column chromatography yield 622 mg (41%) of the title compound as a white solid. Rf (CH2Cl2/MeOH 9:1 + 0.25% NH3) 0.2; XH NMR (CD3OD) δ 7.23 (s, IH, H-4), 7.15 (d, IH, J=8.1 Hz, H-7), 6.89 (d, IH, J=8.1 Hz, H-6), 3.83 (s, 2H, CH2), 3.14 (s, 6H, CH3X2); 13C NMR (CD3OD) δ 163.7 (C-2), 148.4 (C-7a), 144.2 (C-3a), 139.8 (C-5), 119.6 (C-6), 114.9 (C-4), 108.7 (C-7), 47.1 (CH2), 38.0 (CH3); mass spectrum (ES+): m/e 192.0 (M++l).
2-(Dimethylamino)-lf3-benzoxazole-5-carbonitrile:
Figure imgf000060_0001
A mixture of 3-Amino-4-hydroxybenzonitrile (2 g, 14.9 mmol) and dichloromethylene- dimethylammonium chloride (phosgene iminium chloride, 2.42 g, 14.9 mmol) in 50 mL of dry CH2CI2 was refluxed for 6 h. After cooling the solution was extracted with EtOAc, washed with NaHC03, brine and dried (MgS04). The residue was crystallized in MeOH yielding 2.51 g (90%) of the title compound as a white solid. M.p.: 198-200 °C, *H NMR (CD3OD) δ 7.61 (d, IH, J=2.8 Hz, H-4), 7.36-7.32 (m, 2H, H-6, H-7), 3.28 (s, 6H, CH3X2); 13C NMR (CD3OD) δ 164.4 (C-2), 152.2 (C- 7a), 145.0 (C-3a), 125.4 (C-6), 120.0 (CN), 119.9 (C-4), 109.8 (C-7), 108.0 (C-5), 38.2 (CH3).
3-Amino-4-hydroxybenzonitrile:
Figure imgf000060_0002
To a mixture of 5 g (30.5 mmol) of 4-hydroxy-3-nitrobenzonitrile, 18.1 g of powder tin metal (152.5 mmol, 325 mesh), and 45 mL of ethanol was added with stirring a solution of 10 mL of concentrated HCI in 30 mL of H20. The suspension was heated at reflux for 45 min and the resulting hot solution was poured into 100 mL of H20. Saturated aqueous NaHC03 solution was slowly added to bring the pH to ca. 7. The suspension was filtered, and the residue was washed with MeOH giving the title compound (3.15 g, 77%) as a white powder. Rf (CH2CI;>/MeOH 9:1)0.47; *H NMR (CD3OD) δ 6.96 (d, IH, J=1.3 Hz, H-2), 6.92 (d, IH, =B.l Hz, H-6), 6.75 (d, IH, J=8.1 Hz, H-5); 13C NMR (CD3OD) δ 151.1 (C-4), 138.6 (C-3), 124.7 (C-6), 121.6 (CN), 119.1 (C- 2), 115.9 (C-5), 103.6 (C-l).
Example 8
Λβ-[(2-dimethylamino-7-iodo-l),3-benzoxazol-5-yl)-methyll-2-(2-phenyl-l-ethynyl)-adenosine-5-ΛA methyluronamide:
Figure imgf000061_0001
N-Methyl- -deoxy-lHS-chloro^-C∑-phenyl-l-ethynyO-ΘH-purin-θ-ylJ^S'-O-isopropylidene-β-D- ribofuranuronamide (170 mg, 0.37 mmol), NZ, /Ifi-dimethyl-S-faminomethy ^-iodo-l/S- benzoxazol-2-amine (120 mg, 0.38 mmol), and triethylamine (0.16 mL, 1.11 mmol) were dissolved in absolute ethanol (2.5 mL). The solution was stirred at 65°C for 16 h in a sealed vessel. The solvent was removed under nitrogen and the 2,37-<_ -isopropylidene derivative was purified by column chromatography (2% MeOH in CH2CI2) yielding 170 mg. HCI (1 N) (3.0 mL) were added and the solution stirred at 70°C for 45 min. A white precipitate appeared immediately. After cooling, NaHC03 was added until pH 7, and a white solid precipitated. The solid was filtered and washed with water yielding 90 mg (35% yield overall) of the title compound. *H NMR (DMSO- ) δ 8.74 (s, IH, NH), 8.61 (s, IH, NHCO), 8.55 (s, IH, H-8), 7.64 (2H, m, Hs), 7.47 (3H, m, Hs), 7.37 (s, IH, Hbenzoχazoi-6), 7.22 (s, IH, Hb-4), 6.00 (d, IH, .7=7.6 Hz, H-l7), 5.79 (d, IH, J= 4.3 Hz, OH-37), 5.61 (d, IH, J= 6.3 Hz, OH-27), 4.72 (d, 2H, J= 4.5 Hz, CH2-N), 4.63 (m, IH, H-27), 4.34 (s, IH, H-47), 4.18 (m, IH, H-37), 3.11 (s, 6H, (CH3)2N), 2.78 (d, 3H, J=4.5 Hz , 5'-ΛAmethyl). 13C NMR (DMSO-cfc) δ 168.9 (CO), 161.0 (Cb-2), 153.2 (C-6), 148.3, 147.5, 144.3 (C-2), 142.0 (Cb-3a), 141.0 (C-8), 136.7 (0,-5), 131.0 (C-Ha), 128.8 (para-C), 128.1 (C-Hb), 126.5 (Cb-6), 120.0 (ipso-C), 118.8 (C-5), 113.5 (Cb-4), 88.9 (β-alkynyl), 86.8 (C-l7), 83.8 (C-47), 82.7 (α-alkynyl), 72.1 (C-37), 72.1 (C-20, 70.2 (Cb-7), 41.6 (CH2), 36.4, 36.3 ((CH3)2N), 24.8 (5'-ΛAmethyl). Anal. (C29H27N8OsI) C, H, N. High-resolution MS calcd for (C29H27N805NaI) 717.1047, found 717.1060.
/i/-methyl-l'-deoxy-l'-[6-chloro-2-(2-phenyl-l-ethynyl)-9/^purin-9-yl 2'f37-t -isopropylidene- ?-D- ribofuranuronamide:
The above compound was prepared as described in example 5.
NI, UZ-dimethy[-5-(aminomethyl)-7-iodo-l,3-benzoxazo'-2-amine:
Figure imgf000062_0001
To a solution of Tert-butylN-{[2-(dimethylamino)-7-iodo-l/3-benzoxazol-5-yl]methyl}carbamate (120 mg, 0.29 mmol) in 4 ml of CH2CI2 was added slowly 0.5 mL of trifluoroacetic acid and it was stirred at room temperature for 30 min. The excess of TFA was removed under vacuum. The product was purified by column chromatography (CH2Cl2/MeOH 9:1) yielding 64 mg (70%) of the title compound as a white solid. XH NMR (CD3OD) δ 7.46 (d, IH, J=1.5 Hz, H-6); 7.30 (d, IH, .7=1.5 Hz, H-4); 4.12 (s, 2H, CH2); 3.23, 3.24 (s, 6H, CH3X2); 13C NMR (CD3OD) δ 164.5 (C-2), 152.7 (C-7a), 144.6 (C-3a), 132.9 (C-5), 131.6 (C-6), 117.3 (C-4), 72.0 (C-7), 44.2 (CH2), 38.3 (CH3).
Tert-butylN-^ 2-(dimethylamino)-7-iodo-lf3-benzoxazol-5-yl]methyl>carbamate:
Figure imgf000062_0002
A mixture of te t-butyl/V-[[7-bromo-2-(dimethylamino)-l,3-benzoxazol-5-yl]methyl} carbamate Tert-butylN-{[7-bro o-2-(dimethylamino)-lr3-benzoxazol-5-yl]methyl} carbamate (192 mg, 0.52 mmol), Kl (1.3 g, 7.8 mmol), Cul (495 mg, 2.6 mmol) in 1.6 mL of HMPA was stirred overnight (16 h) at 135°C. After cooling, the mixture was extracted with EtOAc, washed with Na2S203 0.5 M solution, and brine (x3). After purification by column chromatography (hexane/EtOAc 6:4), 120 mg (55%) of the title compound were obtained. *H NMR (CDCI3) δ 7.32 (s, IH, H-6); 7.27 (s, IH, H-4); 5.02 (bs, IH, NH); 4.40 (d, 2H, 7=5.4 Hz, CH2); 3.32 (s, 6H, NCH3 2x); 1.57 (s, 9H, (CH3)3C); 13C NMR (CDCI3) δ 162.7 (C-2), 156.2 (CO), 150.4 (C-7a), 143.8 (C-3a), 137.4 (C-5), 128.4 (C-6), 115.3 (C-4), 80.0 ((CH3)3C), 70.5 (C-7), 44.6 (CH2), 38.1 (NCH3 x2), 28.8 ((CH.)^C).
7e^-butyl/y-f[7-bromo-2-(dimethylamino)-lf3-benzoxazol-5-yπmethyl carbamate:
Figure imgf000062_0003
A mixture of Tert-butylN-(3-amino-5-bromo-4-hydroxybenzyl)carbamate (300 mg, 0.95 mmol) and dichloromethylenedimethyiammonium chloride (phosgene iminium chloride, 240 mg, 1.48 mmol) in 10 mL of dry CH2CI2 was refluxed for 6 h. After cooling the solution was extracted with EtOAc, washed with NaHC0 , brine and dried (MgS04). After purification by column chromatography (1% MeOH in CH2CI2) 192 mg (55%) of the title compound were obtained. XH NMR (CDCI3) δ 7.14 (s, IH, H-6); 7.04 (s, IH, H-4); 5.11 (bs, IH, NH); 4.31 (d, 2H, J=5.6 Hz, CHz); 3.23, 3.22 (s, 6H, NCH3 2x); 1.48 (s, 9H, (CH3)3C); 13C NMR (CDCI3) δ 163.3 (C-2), 156.2 (CO), 146.6 (C-7a), 145.1 (C-3a), 137.0 (C-5), 122.8 (C-6), 114.3 (C-4), 100.6 (C-7), 79.9 ((CH^sC), 44.7 (CH2), 38.1 (NCH3 χ2), 28.8 ((CH^C).
re/f-butyl/IA(3-amino-5-bromo-4-hydroxybenzyl)carbamate:
Figure imgf000063_0001
To a solution of 2-amino-4-(aminomethyl)-6-bromophenol(560 mg, 2.59 mmol) in 15 mL of DMF, triethylamine (0.37 mL, 2.63 mmol) and di-te/t-butyldicarbonate (0.59 mL, 2.59 mmol) were added. The mixture was stirred at room temperature for 1 hour and 15 minutes. The solvent was evaporated and the residue was suspended in EtOAc and filtered. The solution was extracted with 0.5 M NaH2P04 (3x) and dried. The product was purified by column chromatography (2% MeOH in CH2CI2) yielding 304 mg (37%) of the title compound. *H NMR (CDCI3) δ 6.76 (d, IH, J=1.8 Hz, H-6); 6.57 (d, IH, J=1.2 Hz, H-2); 4.13 (d, 2H, J=5.8 Hz, CH2); 1.49 (s, 9H, (CH3)3C); 13C NMR (CDCI3) δ 156.5 (CO), 140.1 (C-4), 136.9 (C-l), 133.0 (C-3), 120.3 (C-6), 114.3 (C-2), 110.7 (C- 5), 79.8 ((CH3)3C), 44.2 (CH2), 28.8 ((C±b)^).
2-amino-4-(aminomethyl)-6-bromophenol:
Figure imgf000063_0002
A solution of 3-Bromo-4-hydroxy-5-nitrobenzonitrile (1.0 g, 4.11 mmol) in 20.0 mL of dry THF was added dropwise to a solution of BH3/THF 1.0 M in THF (12.3 mL) at 0°C under N2. The reaction mixture was stirred at room temperature for 24 hours. Excess borane was decomposed by the cautious dropwise addition of 1 N HCI at 0°C. It was extracted with EtOAc, washed with NaHC03, NaCI saturated solution and dried (Na2S04 anh.) to give a crude. After purification by column chromatography (CH^I^MeOH 8:2+ 1% NH4OH), 170 mg (19%) of the title compound were obtained. *H NMR (DMSO-cQ δ 6.67 (s, IH, H-5); 6.53 (s, IH, H-3); 3.52 (s, 2H, CH2). 13C NMR (DMSO- ) δ 139.7, 139.4 (C-l, C-4), 136.0 (C-2), 118.5 (C-5), 113.0 (C-3), 111.5 (C-6), 44.9 (CH2).
3-Bromo-4-hydroxy-5-nitrobenzonitrile:
Figure imgf000064_0001
To a mixture of 5 g (30.5 mmol) of 4-hydroxy-3-nitrobenzonitrile in H2S04 solution (50 mL of concentrated H2S04 + 50 mL of H20) at 25 °C, 7.9 g (47.3 mmol) of potassium bromate were added in small portions cooling the flask with an ice-bath and maintaining the temperature between 25 and 35 °C. After the addition was completed, the reaction was stirred at room temperature for 22 h and then filtered. The pale yellow solid was washed with water and dried to give 4.2 g (57%) of the title compound. M.p.: 162-164 °C; XH NMR (CD3OD) δ 8.54 (d, IH, .7=2.0 Hz, H-6), 8.32 (d, IH, J=2.0 Hz, H-2); 13C NMR (DMSO-fifc) δ 153.9 (C-4), 140.8 (C-2), 138.2 (C- 5), 130.1 (C-6), 117.0 (CN), 115.9 (C-3), 101.6 (C-l).
Pharmacological data
Binding to Aι, A? and A^ receptors
The compounds of Examples 1 to 8 were evaluated for their pharmacological effect.
These compounds were firstly evaluated for binding to human A3 receptors expressed in Chinese Hamster Ovary cells (CHO cells). [125I]-AB-MECA (0.3nM) binding to membrane preparations was examined using 60 min incubation at room temperature. The displacement of binding by the adenosine analogues was determined, non-specific binding being measured from the displacement by IB-MECA (10"7M).
Compounds are identified in the following Tables by their Example number. Values for A3 receptor binding are the IC50 values (nM) for displacement of [125I]-AB-MECA binding, the concentration for 50% inhibition. These are compared with IB-MECA which has an approximate IC50 value of 0.31nM. Thus, it can be seen from table 1 that compounds 2-4 are approximately equipotent with IB-MECA and that compound 6 is approximately two orders of magnitude more potent.
The selectivity of compounds 2 and 6 for A3 receptors has been evaluated by determining their functional activity at Ai and A2 receptors in preliminary experiments. For compound 2, Ax receptor functional activity was determined from the negative inotropic response of guinea-pig isolated paced left atria set up in tissue baths containing Krebs- bicarbonate solution gassed with 5% C02 in oxygen at 37°C. Dose-related inhibition of atrial developed tension (negative inotropy) was observed commencing at 300nM. The IC50 value (concentration for 50% inhibition of contractions) was 7500nM.
Compound 27s A2 receptor functional activity was determined from the relaxation response of guinea-pig isolated tracheal spirals set up in tissue baths containing Krebs-bicarbonate solution gassed with 5% C02 in oxygen at 37°C. The tissue was pre-contracted with carbachol (lOOnM) and when the tension had reached a plateau, increasing concentrations of compound 9 were introduced cumulatively. There were dose-related relaxations indicative of A2 receptor activity commencing at lμM. The IC50 (concentration for 50% inhibition of the carbachol-induced contraction) was 20,000nM. These values compare with IC50 values for the non-selective Aι/A2 receptor agonist of lOOnM in the atria and 500nM in the trachea.
Thus, the selectivity of compound 2 for A3 receptors (radioligand binding) against Ai receptors (negative inotropy) is 16,600 and against A2 receptors (tracheal relaxation) is 44,000.
The results obtained are set out in Table 1, with the IC50 for IB-MECA included for comparison.
Table 1
Figure imgf000066_0001
The lowest IC50 value for A3 receptor binding exhibited by the test compounds was found to be that of compound 6. However, subsequent experiments have indicated that the IC50 for compound 6 is in the nanomolar range, not the indicated picomolar value. Table 2 below shows displacement of [125I]-AB-MECA (0.3nM) from human A3 receptors transfected into Chinese Hamster Ovary (CHO) cells by IB-MECA and compound 6.
Table 2
Figure imgf000067_0001
Total binding in the presence of each concentration of IB-MECA or compound 6 is expressed as a percentage of the total binding in their absence.
Table 3 shows the activity of compound 6 at Ai receptors of guinea-pig atria compared with the standard non-selective agonist, NECA (N-ethylcarboxamidoadenosine).
Table 3
Figure imgf000067_0002
Activity is expressed as the negative inotropic action (reduction in the tension development of paced isolated atria) obtained in cumulative concentration-response curves. At the maximum concentration of compound 6, a maximum effective concentration of NECA was added and the responses to compound 6 were expressed as a percentage of this maximum response. The IC50 and IC25 concentrations (concentrations for 50 and 25% of the maximum response to NECA) were then calculated. After washout of the tissues, a full concentration-response curve for NECA was obtained and the IC25 and IC50 values for NECA calculated. This data is displayed graphically in Figure 6.
Table 4 shows the activity of compound 6 at A2 receptors of guinea-pig trachea compared with the standard non-selective agonist, NECA (N-ethylcarboxamidoadenosine). Table 4
Figure imgf000068_0001
The activity is expressed as the relaxation of the trachea precontracted with carbachol (lOOnM) obtained in cumulative concentration-response curves. At the maximum concentration of compound 6, a maximum effective concentration of NECA was added and the responses to compound 6 were expressed as a percentage of this maximum response. The IC25 concentration (concentration for 25% of the maximum response to NECA) was then calculated. After washout of the tissues and contraction again with carbachol, a full concentration-response curve for NECA was obtained and the IC25 value for NECA calculated. This data is represented graphically in Figure 7.
Compound 6 has picomolar potency for binding to the human A3 receptor. Based on functional tests in atrial and tracheal tissues, the selectivity over Ai and A2 receptors is 6.25xl06 and 5.6xl06, respectively.
Further tests were carried out on compound 6 to evaluate its usefulness as a therapeutic compound. Efficacy of compound 6 in protecting against myocardial contractile dysfunction of isolated atria
The following experiment was carried out to evaluate the efficacy of compound 6 in protecting against myocardial contractile dysfunction (stunning)after simulated ischaemia of isolated atria. Guinea-pig isolated left atria were set up in tissue baths containing Krebs-bicarbonate solution gassed with 5% C02 in oxygen at 37°C and electrically paced at 2Hz with pulses of threshold voltage +50% and 5ms pulse width. Developed tension was recorded. After equilibrium, they were exposed to 30 min of simulated ischaemia by gassing with 5% C02 in nitrogen and removing the glucose substrate which was replaced with choline chloride (7mM) to maintain isotonicity. Pacing was continued throughout. After 30 min, the tissues were reoxygenated and glucose was returned. Contractions were virtually abolished during ischaemia but on reoxygenation contractile function was partially restored and reached 38.0+2% of the pre-ischaemic developed tension after lOmin of reoxygenation. This partial recovery was the index of myocardial stunning. The test compound (compound 6 (5xlO"9M)) or the reference A3 receptor ligand, IB-MECA (3χlO"7M) was introduced at reoxygenation. This timing was to simulate administration at the time of reperfusion induced by thrombolyic therapy following myocardial infarction. Table 5 below shows a comparison of the effects of IB-MECA and compound 6 on atrial contractile function after 30 min simulated ischaemia. This data is also represented graphically in Figure 8.
Table 5
Figure imgf000069_0001
As can be seen from the above table, when IB-MECA at a concentration of 3χlO"7M was introduced at regassing, the contractile tension recovered to 66.5+6.5% of its pre-ischaemic value. This value is significantly greater than in the control tissues and indicates a reversal of the myocardial stunning. A concentration of 5χlO"9M of compound 6 was selected for these experiments based on its potency in the radioligand binding experiments. When this concentration was introduced at the onset of regassing, the recovery of developed tension was to 84.0+18.0% (n=3). Thus at a concentration 60 times less than that of IB-MECA, compound 6 produced a greater degree of recovery of contractile tension following simulated ischaemia.
Efficacy of compound 6 in protecting against myocardial contractile dysfunction of perfused hearts The following experiment was carried out to evaluate the efficacy of compound 6 in protecting against myocardial contractile dysfunction (stunning) after no-flow global ischaemia of guinea-pig perfused hearts.
Guinea pig hearts were perfused by the Langendorff method. The cut aortic stump was perfused reterogradely with Krebs solution gassed with 5% C02 in oxygen at 37°C at a constant flow rate of 7ml/min to perfuse the coronary circulation. The heart was jacketed at 37°C and the spontaneous force of contraction was measured by attaching a clip to the apex of the heart which was connected to a tension transducer. Coronary perfusion pressure was also monitored.
After equilibration, global ischaemia was produced by stopping perfusion and clamping the infusion line. Spontaneous contractions ceased. After 22 minutes, flow was restored to 30 % of the pre-ischaemic level. After a further 10 minutes, flow was restored to the pre-ischaemic level (7ml/min). Spontaneous contractions resumed after about 10 minutes. These contractions reached a maximum recovery of 18.8+7.4% of the pre-ischaemic level. The adenosine receptor ligand infusion was commenced just prior to resumption of 30% coronary perfusion, to mimic administration at reperfusion after thrombolytic therapy, and was continued until restoration of flow to the pre-ischaemic level. The results are presented graphically in Figure 9.
Significantly improved results were obtained by standardisation of pre-conditioning. Male Dunkin- Harley guinea-pigs (360-390g) were killed by cervical dislocation and their hearts removed and perfused with krebs-bicarbonate solution via a constant flow Langendorff heart preparation. Between cervical dislocation and perfusion of the isolated heart a period of 2.5 minutes elapsed where the heart was not perfused. During this period the isolated heart was mounted onto the cannula in readiness for perfusion. The experiment not was performed on any preparation that could not be mounted and perfused within 2.5 minutes. In addition the pulmonary artery was cut to assist out flow of perfusate from the heart. Data shown is the developed tension measured as a percentage of the pre-ischaemic level. Error bars represent S.E.M. * denotes a significant difference from control (p>0.05) using a unpaired f-test. For the control n=6 and for the drug treated group n=4. The results are presented graphically in Figure 10.
Pharmacodynamic considerations
The pKa values for compounds 6 and 7 are given in Table 6 below. The partition coefficient profile for compound 6 is given in Table 7 below. The partition measurements were based on a long chain ester (propylene glycol dipelargonate-PGDP)/water model. Table 6 shows pKa values of compounds 6 and 7 calculated using methanol and dimethylformamide co-solvents respectively. All experiments carried out in ionic strength water (0.15 KCI).
Table 6
Figure imgf000072_0001
*"goodness of fit" of data to chosen ionisation model. A high GOF indicates poor fit; values close to unity (<2) are considered to provide a highly reliable resul ** Linear regression coefficient for Yasuda-Shediovsky Plot for multiple titrations in presence of different % co-solvent (aqueous pl , obtained by extrapolation
Table 7 shows Log P value of compound 6 (partition solvent: propylene glycol dipelargonate (PGDP). Table 7
Figure imgf000073_0001
* "goodness of fit77 of data to chosen ionisation model. A high GOF indicates poor fit; values close to unity (<2) are considered to provide a highly reliable result
No 2nd pKg was reported. A p g of 3.7 would be expected for the benzoxazole part and a pKg of 3.2 for the adenine moiety (protonation expected at NI). The reported pl , value of 3.66+0.03 therefore, very probably refers to the benzoxazole moiety. The partition coefficient of 1.48+0.03 on the logio scale indicates that the compound is not too lipophilic. The precise model for optimal pharmacodynamic properties is believed to be a value of unity (i.e. a value of zero on the logio scale) or a little over for a hydrocarbon/water model. Empiric evidence shows that propranolol has an effective partition after allowing for protonation of some 0.8 on the logio PGDP/H20 scale. The presence of a sugar moiety in the adenosine system may increase the relative value of compound 10 on this particular scale. Even so, the difference from this chosen optimum of a little over 0.6 logω units is equivalent to the presence of only one methyl group. Intravenous injection is likely to reduce any significance in the optimal properties.

Claims

1. A product which is a compound of the formula:
Figure imgf000074_0001
wherein
D is N or CH;
E is 0, S or CH2;
X1 is a group of the formula -CR 0R21-CYCLE, where R20 and R21 are the same or different and H, F or CH3; CYCLE is of formula (II) or formula (III):
Figure imgf000074_0002
(II) (III) where:
R5 is iodine, bromine, methyl or trifluoromethyl;
R7 is H, halogen, Ci-C^g acyl, OR11, CO2R11 or CONR11 where R11 is Ci-Cig hydrocarbyl optionally containing one or more in-chain and/or in-ring -O- linkages;
R8 is -NR9R10 or -COR9, where R9 and R10 are each independently methyl or ethyl; and W is N or CH;
.X2 is hydroxymethyl, (Cι-C3)alkoxymethyI, (C3-C5)cycloalkoxy methyl, carboxy, (C^- C3)alkoxycarbonyI, (C3-C5)cycloalkoxy-carbonyl, 1,1-aminoiminomethyl, l,l-(mono-N- or di-N,N- (Cι-C4)alkylamino)iminomethyl, l,l-(mono-N- or di-N,N-(C3-C5)cycloalkylamino)iminomethyl, carbamoyl, mono-N- or di-N,N-(Cι-C4)alkylaminocarbonyl, mono-N- or di-N,N-(C3-C5)cycloalkyl- aminocarbonyl or N-(Cι-C4)alkyl-N-(C3-C5)cycloalkylamino-carbonyl;
X3 and X4 are each independently hydrogen, alkyl, hydroxyalkyl, alkoxyalkyl, ORa or NRaRD, where Ra and RD are independently hydrogen, alkyl, aralkyl, carbamoyl, alkyl carbamoyl, dialkylcarbamoyl, acyl, alkoxycarbonyl, aralkoxycarbonyl, aryloxycarbonyl, or, when X3 and X4 are both ORa, the two Ra groups together may form
Figure imgf000075_0001
where Rc is hydrogen or alkyl,
Figure imgf000075_0002
where Rd and Re are independently hydrogen, alkyl, or together with the carbon atom to which they are attached may form a 1,1-cycloalkyl group;
X5 is H, halogen, (Cι-Cιg)alkyl, fluorinated (C^-Cio) alkyl (e.g. trifluoromethyl), (C^-C^g) alkoxyalkyl, (Cι-Cιg)alkoxy, (Ci-C^gjalkylether, (Cι-Cιg)thioalkoxy, (Cι-Cιg)alkylthio, amino,
(Cι-Cιg)alkylamino, -COX6R25 where X6 is 0 or NH and R25 is (Ci- ^alkyl optionally terminally substituted by an aryl or a heteroaryl group and additionally or alternatively terminally substituted by hydroxy, (C2-Cιg)alkenyl, (C2-Cιg)alkynyl, or is (C2-Cιg)alkenyl, (C2-Cιg)alkynyl in either case terminally substituted by an aryl or heteroaryl group and, when having a terminal methylic carbon atom, optionally further terminally substituted by hydroxy,
or a pharmaceutically acceptable salt or prodrug thereof or a pharmaceutically acceptable salt of such a prodrug.
2. A product of claim 1, wherein
D is N; E is O;
X2 is mono-N- or di-N,N(Cι-C4)alkylaminocarbonyl, mono-N- or di-, N-(C3-C5)cycloa!kylaminocarbonyl or N-fCi-GaJalkyl-N- (C3-C5)cycloalkylaminocarbonyl; X3 is OH or NH ;
X4 is OH;
X5 is H, halogen, (Cι-Cιg)alkyl, trifluoromethyl, (C2"Cιg)alkenyl, (C2-Cιg)al ynyl, or either of the latter two groups where terminally substituted as defined in claim 1.
3. A product of claim 1 or claim 2 wherein X^ is halogen.
4. A product of claim 3 wherein X5 is bromine or chlorine.
5. A product of any preceding claim wherein R and R2 are both H.
6. A product of claim 1 wherein the compound is of formula (V):
Figure imgf000076_0001
where: -CR20R21-CYCLE, D and R2 are as defined in claim 1;
E is 0, S or CH2 (e.g. E is O and optionally D is N and R2 is CI or other halogen);
R1 is C1-C4 alkyl; and
X3a is -OH or -NH2.
7. A product of claim 6 wherein E is O.
8. A product of any preceding claim wherein CYCLE is of formula (II).
9. A product of claim 8 wherein W is N.
10. A product of claim 8 or claim 9 wherein R8 is dimethylamino or diethylamino.
11. A product of claim 8 wherein CYCLE is selected from the group consisting of the following moieties:
Figure imgf000077_0001
CD (2) (3)
Figure imgf000077_0002
(12) (13) (14)
Figure imgf000077_0003
(15) (16) (i7)
Figure imgf000078_0001
(18) (19)
Figure imgf000078_0002
Figure imgf000078_0003
12. A product of claim 11 wherein CYCLE is of formula 1, 2, 3 or 4; or of formula 12, 13, 14 or 15.
13. A product of any of claims 8, 11 and 12 wherein the compound is of formula (VI):
Figure imgf000078_0004
where CYCLE is a group of formula (II).
14. A product of any of claims 8, 11 and 12 wherein the compound is of formula (VII):
Figure imgf000078_0005
where CYCLE is a group of formula (II).
15. A modification of a product of claim 13 or claim 14 in which the 3' -OH group is replaced by another X3 or X3a group.
16. A product of any preceding claim in which the compound is not
Figure imgf000079_0001
17. A product of any of claims 1 to 7 wherein CYCLE is of formula (III).
18. A product of claim 17 wherein R7 is C^-C^g acyl wherein acyl is -COR12 in which R12 is hydrocarbyl or hydrocarbyl containing one or ore in-chain and/or in-ring -0- linkages.
19. A product of claim 17 wherein CYCLE is selected from the group consisting of the following moieties:
The invention therefore includes compounds having the following CYCLE moieties:
Figure imgf000079_0002
Figure imgf000080_0001
20. A compound of formula (VIII):
Figure imgf000080_0002
wherein
D is N or CH;
E is 0, S or CH2;
X1 is of the formula -CR20R21-CYCLE where R20 and R21 are the same or different and H, F or CH3; and CYCLE is a bicyclic (fused) heteroaromatic ring of the formula
Figure imgf000081_0001
wherein ring A is a 5- or 6- membered ring characterised by the following features (in which ring positions are numbered relative to the linkage to -CR20R21-): i. a carbon atom at the 1-position; ii. carbon atom as CH or a nitrogen atom at position 2; iii. it is 3, 4 fused to ring B; iv. the 5-position ring atom is substituted by a moiety R5 which is H, halogen, or an organic moiety having from 1 to 6 plurally valent atoms in addition to monovalent atoms selected from hydrogen and halogen. v. if a 6-membered ring, it has at the 6-position a nitrogen, or -CM- where M is H, CH3 or
F; ring B is a 5 or 6 membered ring characterised by the following features:
(a) an in-ring heteroatom including O, N or S joined to the 4-position of ring A;
(b) said in-ring heteroatom is joined within the ring secondly to a carbon which is substituted by a moiety R8 which is -N^Hs
(c) an in-ring atom joined to the 3-position of ring A which is N,0, S or C, said C being in the form of a CH or CO group;
(d) in the case of a 6-membered ring, the remaining ring member is nitrogen or carbon in the form of CH; X2 (the 4' substituent) is hydroxymethyl, (Cι-C3)alkoxymethyl, (C3-C5)cycloalkoxy methyl, carboxy, (C]i-C3)alkoxycarbonyl, (C3-C5)cycloalkoxycarbonyI, 1,1-aminoiminomethyl, l,l-(mono-
N- or di-N,N-(Cι-C4)alkylamino)iminomethyl, l,l-(mono-N- or di-N,N-(C3-C5)cycloalkyl- amino)iminomethyl, carbamoyl, mono-N- or di-N,N-(Cι-C4)alkylaminocarbonyl, mono-N- or di-
N,N-(C3-C5)cycloalkylaminocarbonyl or N-(Cι-C4)aIkyl-N-(C3-C5)cycloalkylaminocarbonyl; X3 and X4 are each independently hydrogen, alkyl, hydroxyalkyl, alkoxyalkyl, ORa NRaRD, where Ra and RD are independently hydrogen (most preferably X3 and X4 are OH), alkyl, aralkyl, carbamoyl, alkyl carbamoyl, dialkylcarbamoyl, acyl, alkoxycarbonyl, aralkoxycarbonyl, aryloxycarbonyl, or, when X3 and X4 are both ORa, the two Ra groups together may form
Figure imgf000082_0001
where Rc is hydrogen or alkyl,
Figure imgf000082_0002
where R^ and Re are independently hydrogen, alkyl, or together with the carbon atom to which they are attached may form a 1,1-cycloalkyl group;
X5 is H, halogen, (C^-Cigjalk l, fluorinated (C^-C^g) alkyl (e.g. trifluoromethyl), (C^-Cig) alkoxyalkyl,
Figure imgf000082_0003
(Cι-Cιg)thioalkoxy, (Cι-Cιg)alkylthio, amino,
(Ci-Cigjalkylamino, -COX6R25 where X6 is O or NH and R25 is (C1-C4)alkyl optionally terminally substituted by an aryl or a heteroaryl group [for example phenyl or a 5- or 6-membered heteroaryl group] and additionally or alternatively terminally substituted by hydroxy, (C2-
Cιg)alkenyl, C2-Cιg)alkynyl, or is (C2-Cjg)alkenyl or (C2-Cιg)alkynyl in either case terminally substituted by an aryl or heteroaryl group [for example phenyl or a 5- or 6-membered heteroaryl group] and, when having a terminal methylic carbon atom, optionally further terminally substituted by hydroxy.
21. A compound of claim 20 wherein R5 has from 1 to 4 plurally valent atoms.
22. A compound of claim 21 wherein the plurally valent atoms are selected from carbon, oxygen, sulfur and nitrogen.
23. A compound of claim 22 wherein R^ is CH3, CF3, OH or NH2-
24. A compound of claim 20 wherein R^ is H, I, Br or CI.
25. A compound of any of claims 20 to 24 wherein CYCLE is of formula (IX)
Figure imgf000083_0001
26. A compound of any of claims 20 to 25 wherein where R20 and R21 are both hydrogen.
27. An adenosine analogue-type A3 receptor agonist having an N6 nitrogen substituted by a group of the formula -CR20R 1-CYCLE where R20 and R21 are the same or different and H, F or CH3; and CYCLE is of formula (II) or formula (III):
Figure imgf000083_0002
where:
R5 is iodine, bromine, methyl or trifluoromethyl;
R7 is H, halogen, C^-C^ acyl, OR11, CO2R11 or CONR11 where R11 is Cι-C10 hydrocarbyl optionally containing one or more in-chain and/or in-ring -O- linkages;
R8 is -NR9R10 or -COR9, where R9 and R10 are each independently methyl or ethyl; and
W is N or CH.
28. An adenosine analogue-type A3 receptor agonist having an N6 nitrogen substituted by a group of the formula -CR20R21-CYCLE where R20 and R21 are the same or different and H, F or CH3; and CYCLE is a bicyclic (fused) heteroaromatic ring of the formula
Figure imgf000083_0003
wherein ring A is a 5- or 6- membered ring characterised by the following features (in which ring positions are numbered relative to the linkage to -CR20R21-): a carbon atom at the 1-position; carbon atom as CH or a nitrogen atom at position 2; iii. it is 3, 4 fused to ring B; iv. the 5-position ring atom is substituted by a moiety R5 which is H, halogen or an organic moiety having from 1 to 6 plurally valent atoms in addition to monovalent atoms selected from hydrogen and halogen. v. if a 6-membered ring, it has at the 6-position a nitrogen, or -CM- where M is H, CH3 or F; ring B is a 5 or 6 membered ring characterised by the following features:
(a) an in-ring heteroatom including 0, N or S joined to the 4-position of ring A;
(b) said in-ring heteroatom is joined within the ring secondly to a carbon which is substituted by a moiety R8 which is -N(C2H5)2; (c) an in-ring atom joined to the 3-position of ring A which is N,0, S or C, said C being in the form of a CH or CO group;
(d) in the case of a 6-membered ring, the remaining ring member is nitrogen or carbon in the form of CH.
29. A product of any one of claims 1 to 26 or an agonist of claim 27 or claim 28 for use as a medicament.
30. A product of any one of claims 1 to 26 or an agonist of claim 27 or claim 28 for use in a method for selectively activating A3 adenosine receptors in a mammal.
31. The use of a product of any one of claims 1 to 26 or an agonist of claim 27 or claim 28 for the manufacture of a medicament for use in a method for selectively activating A3 adenosine receptors in a mammal.
32. The use of a product of any one of claims 1 to 26 or an agonist of claim 27 or claim 28 for the manufacture of a medicament for use for the reduction of tissue damage resulting from ischaemia or hypoxia.
33. The use of a product of any one of claims 1 to 26 or an agonist of claim 27 or claim 28 for the manufacture of a medicament for use for preconditioning the heart to protect it from ischaemic damage.
34. The use of any of claims 31 to 33 wherein the medicament is for intravenous administration.
35. The use of any of claims 31 to 34 wherein the medicament is for use in combination therapy with another cardiovacular drug.
36. A pharmaceutical composition comprising a product of any one of claims 1 to 26 or an agonist of claim 27 or claim 28.
37. A pharmaceutical composition of claim 36 which is an intravenous formulation.
38. The use in a medicament of an N6 substituent as defined in claim 27 or claim 28 to increase the A3 receptor specificity of an adenosine analogue of which the remainder is compatible with the adenosine A3 receptor for agonist use.
39. A method of stimulating adenosine A3 receptors, comprising administering to a mammal in need of such treatment a therapeutically effective amount of a product of any one of claims 1 to 26 or an agonist of claim 27 or claim 28.
40. A method of reducing tissue or organ damage (e.g., substantially preventing tissue or organ damage, inducing tissue or organ protection) resulting from ischaemia or hypoxia, comprising administering to a mammal in need of such treatment a therapeutically effective amount of an agent selected from a product of any one of claims 1 to 26 and agonist of claim 27 or claim 28.
PCT/GB2004/003283 2003-07-31 2004-07-30 Compounds useful as a3 adenosine receptor agonists WO2005012323A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB0317951.2 2003-07-31
GBGB0317951.2A GB0317951D0 (en) 2003-07-31 2003-07-31 Compounds

Publications (2)

Publication Number Publication Date
WO2005012323A2 true WO2005012323A2 (en) 2005-02-10
WO2005012323A3 WO2005012323A3 (en) 2005-05-26

Family

ID=27799566

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2004/003283 WO2005012323A2 (en) 2003-07-31 2004-07-30 Compounds useful as a3 adenosine receptor agonists

Country Status (2)

Country Link
GB (1) GB0317951D0 (en)
WO (1) WO2005012323A2 (en)

Cited By (38)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7737126B2 (en) 2004-05-24 2010-06-15 Glaxo Group Limited Purine derivative
US7985740B2 (en) 2005-07-19 2011-07-26 Glaxo Group Limited Purine derivatives as agonists of the adenosine A2A receptor
US8193182B2 (en) 2008-01-04 2012-06-05 Intellikine, Inc. Substituted isoquinolin-1(2H)-ones, and methods of use thereof
US8476282B2 (en) 2008-11-03 2013-07-02 Intellikine Llc Benzoxazole kinase inhibitors and methods of use
US8604032B2 (en) 2010-05-21 2013-12-10 Infinity Pharmaceuticals, Inc. Chemical compounds, compositions and methods for kinase modulation
US8637542B2 (en) 2008-03-14 2014-01-28 Intellikine, Inc. Kinase inhibitors and methods of use
US8642604B2 (en) 2006-04-04 2014-02-04 The Regents Of The University Of California Substituted pyrazolo[3,2-d]pyrimidines as anti-cancer agents
US8697709B2 (en) 2008-10-16 2014-04-15 The Regents Of The University Of California Fused ring heteroaryl kinase inhibitors
US8703778B2 (en) 2008-09-26 2014-04-22 Intellikine Llc Heterocyclic kinase inhibitors
US8703777B2 (en) 2008-01-04 2014-04-22 Intellikine Llc Certain chemical entities, compositions and methods
US8785454B2 (en) 2009-05-07 2014-07-22 Intellikine Llc Heterocyclic compounds and uses thereof
US8785470B2 (en) 2011-08-29 2014-07-22 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US8809349B2 (en) 2011-01-10 2014-08-19 Infinity Pharmaceuticals, Inc. Processes for preparing isoquinolinones and solid forms of isoquinolinones
US8828998B2 (en) 2012-06-25 2014-09-09 Infinity Pharmaceuticals, Inc. Treatment of lupus, fibrotic conditions, and inflammatory myopathies and other disorders using PI3 kinase inhibitors
US8901133B2 (en) 2010-11-10 2014-12-02 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US8940742B2 (en) 2012-04-10 2015-01-27 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US8969363B2 (en) 2011-07-19 2015-03-03 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US8980899B2 (en) 2009-10-16 2015-03-17 The Regents Of The University Of California Methods of inhibiting Ire1
US8993580B2 (en) 2008-03-14 2015-03-31 Intellikine Llc Benzothiazole kinase inhibitors and methods of use
US9056877B2 (en) 2011-07-19 2015-06-16 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9096611B2 (en) 2008-07-08 2015-08-04 Intellikine Llc Kinase inhibitors and methods of use
US9295673B2 (en) 2011-02-23 2016-03-29 Intellikine Llc Combination of mTOR inhibitors and P13-kinase inhibitors, and uses thereof
US9321772B2 (en) 2011-09-02 2016-04-26 The Regents Of The University Of California Substituted pyrazolo[3,4-D]pyrimidines and uses thereof
US9359349B2 (en) 2007-10-04 2016-06-07 Intellikine Llc Substituted quinazolines as kinase inhibitors
US9359365B2 (en) 2013-10-04 2016-06-07 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9481667B2 (en) 2013-03-15 2016-11-01 Infinity Pharmaceuticals, Inc. Salts and solid forms of isoquinolinones and composition comprising and methods of using the same
US9512125B2 (en) 2004-11-19 2016-12-06 The Regents Of The University Of California Substituted pyrazolo[3.4-D] pyrimidines as anti-inflammatory agents
US9629843B2 (en) 2008-07-08 2017-04-25 The Regents Of The University Of California MTOR modulators and uses thereof
US9708348B2 (en) 2014-10-03 2017-07-18 Infinity Pharmaceuticals, Inc. Trisubstituted bicyclic heterocyclic compounds with kinase activities and uses thereof
US9751888B2 (en) 2013-10-04 2017-09-05 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9775844B2 (en) 2014-03-19 2017-10-03 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US10131668B2 (en) 2012-09-26 2018-11-20 The Regents Of The University Of California Substituted imidazo[1,5-a]pYRAZINES for modulation of IRE1
US10160761B2 (en) 2015-09-14 2018-12-25 Infinity Pharmaceuticals, Inc. Solid forms of isoquinolinones, and process of making, composition comprising, and methods of using the same
CN110799517A (en) * 2017-01-27 2020-02-14 中央研究院 Compound having analgesic effect for preventing and treating pain
US10759806B2 (en) 2016-03-17 2020-09-01 Infinity Pharmaceuticals, Inc. Isotopologues of isoquinolinone and quinazolinone compounds and uses thereof as PI3K kinase inhibitors
US10919914B2 (en) 2016-06-08 2021-02-16 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US11110096B2 (en) 2014-04-16 2021-09-07 Infinity Pharmaceuticals, Inc. Combination therapies
US11147818B2 (en) 2016-06-24 2021-10-19 Infinity Pharmaceuticals, Inc. Combination therapies

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5688774A (en) * 1993-07-13 1997-11-18 The United States Of America As Represented By The Department Of Health And Human Services A3 adenosine receptor agonists
EP1241176A1 (en) * 2001-03-16 2002-09-18 Pfizer Products Inc. Purine derivatives for the treatment of ischemia
WO2003061670A1 (en) * 2002-01-25 2003-07-31 Muscagen Limited Compounds useful as a3 adenosine receptor agonists

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5688774A (en) * 1993-07-13 1997-11-18 The United States Of America As Represented By The Department Of Health And Human Services A3 adenosine receptor agonists
EP1241176A1 (en) * 2001-03-16 2002-09-18 Pfizer Products Inc. Purine derivatives for the treatment of ischemia
WO2003061670A1 (en) * 2002-01-25 2003-07-31 Muscagen Limited Compounds useful as a3 adenosine receptor agonists

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
AFIFY H M N M ET AL: "A NOVEL AND FACILE REACTION TO N6-ALKYLATED ADENOSINE VIA BENZOTRIAZOLE AS A SYNTHETIC AUXILIARY" JOURNAL OF HETEROCYCLIC CHEMISTRY, HETEROCORPORATION. PROVO, US, vol. 37, no. 2, March 2000 (2000-03), pages 339-341, XP001147847 ISSN: 0022-152X *
SIDDIQI S M ET AL: "SEARCH FOR NEW PURINE- AND RIBOSE-MODIFIED ADENOSINE ANALOGUES AS SELECTIVE AGONISTS AND ANTAGONISTS AT ADENOSINE RECEPTORS" JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY. WASHINGTON, US, vol. 38, no. 7, 1995, pages 1174-1188, XP000882724 ISSN: 0022-2623 *
Z.-G. GAO ET AL.: "N6-Substituted adenosine derivatives: selectivity, efficacy, and species differences at A3 adenosine receptors" BIOCHEMICAL PHARMACOLOGY, vol. 65, 2003, pages 1675-1684, XP002309313 *

Cited By (80)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7737126B2 (en) 2004-05-24 2010-06-15 Glaxo Group Limited Purine derivative
US9512125B2 (en) 2004-11-19 2016-12-06 The Regents Of The University Of California Substituted pyrazolo[3.4-D] pyrimidines as anti-inflammatory agents
US7985740B2 (en) 2005-07-19 2011-07-26 Glaxo Group Limited Purine derivatives as agonists of the adenosine A2A receptor
US9493467B2 (en) 2006-04-04 2016-11-15 The Regents Of The University Of California PI3 kinase antagonists
US8642604B2 (en) 2006-04-04 2014-02-04 The Regents Of The University Of California Substituted pyrazolo[3,2-d]pyrimidines as anti-cancer agents
US9359349B2 (en) 2007-10-04 2016-06-07 Intellikine Llc Substituted quinazolines as kinase inhibitors
US11433065B2 (en) 2008-01-04 2022-09-06 Intellikine Llc Certain chemical entities, compositions and methods
US9216982B2 (en) 2008-01-04 2015-12-22 Intellikine Llc Certain chemical entities, compositions and methods
US8785456B2 (en) 2008-01-04 2014-07-22 Intellikine Llc Substituted isoquinolin-1(2H)-ones, and methods of use thereof
US8193182B2 (en) 2008-01-04 2012-06-05 Intellikine, Inc. Substituted isoquinolin-1(2H)-ones, and methods of use thereof
US9655892B2 (en) 2008-01-04 2017-05-23 Intellikine Llc Certain chemical entities, compositions and methods
US8703777B2 (en) 2008-01-04 2014-04-22 Intellikine Llc Certain chemical entities, compositions and methods
US9822131B2 (en) 2008-01-04 2017-11-21 Intellikine Llc Certain chemical entities, compositions and methods
US8637542B2 (en) 2008-03-14 2014-01-28 Intellikine, Inc. Kinase inhibitors and methods of use
US8993580B2 (en) 2008-03-14 2015-03-31 Intellikine Llc Benzothiazole kinase inhibitors and methods of use
US9637492B2 (en) 2008-03-14 2017-05-02 Intellikine Llc Benzothiazole kinase inhibitors and methods of use
US9629843B2 (en) 2008-07-08 2017-04-25 The Regents Of The University Of California MTOR modulators and uses thereof
US9096611B2 (en) 2008-07-08 2015-08-04 Intellikine Llc Kinase inhibitors and methods of use
US9828378B2 (en) 2008-07-08 2017-11-28 Intellikine Llc Kinase inhibitors and methods of use
US8703778B2 (en) 2008-09-26 2014-04-22 Intellikine Llc Heterocyclic kinase inhibitors
US9790228B2 (en) 2008-09-26 2017-10-17 Intellikine Llc Heterocyclic kinase inhibitors
US9296742B2 (en) 2008-09-26 2016-03-29 Intellikine Llc Heterocyclic kinase inhibitors
US8697709B2 (en) 2008-10-16 2014-04-15 The Regents Of The University Of California Fused ring heteroaryl kinase inhibitors
US8476431B2 (en) 2008-11-03 2013-07-02 Itellikine LLC Benzoxazole kinase inhibitors and methods of use
US8476282B2 (en) 2008-11-03 2013-07-02 Intellikine Llc Benzoxazole kinase inhibitors and methods of use
US8785454B2 (en) 2009-05-07 2014-07-22 Intellikine Llc Heterocyclic compounds and uses thereof
US9315505B2 (en) 2009-05-07 2016-04-19 Intellikine Llc Heterocyclic compounds and uses thereof
US9522146B2 (en) 2009-07-15 2016-12-20 Intellikine Llc Substituted Isoquinolin-1(2H)-one compounds, compositions, and methods thereof
US9206182B2 (en) 2009-07-15 2015-12-08 Intellikine Llc Substituted isoquinolin-1(2H)-one compounds, compositions, and methods thereof
US8569323B2 (en) 2009-07-15 2013-10-29 Intellikine, Llc Substituted isoquinolin-1(2H)-one compounds, compositions, and methods thereof
US8980899B2 (en) 2009-10-16 2015-03-17 The Regents Of The University Of California Methods of inhibiting Ire1
US8604032B2 (en) 2010-05-21 2013-12-10 Infinity Pharmaceuticals, Inc. Chemical compounds, compositions and methods for kinase modulation
US9738644B2 (en) 2010-05-21 2017-08-22 Infinity Pharmaceuticals, Inc. Chemical compounds, compositions and methods for kinase modulation
US9181221B2 (en) 2010-05-21 2015-11-10 Infinity Pharmaceuticals, Inc. Chemical compounds, compositions and methods for kinase modulation
US9388183B2 (en) 2010-11-10 2016-07-12 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US8901133B2 (en) 2010-11-10 2014-12-02 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
USRE46621E1 (en) 2011-01-10 2017-12-05 Infinity Pharmaceuticals, Inc. Processes for preparing isoquinolinones and solid forms of isoquinolinones
US9840505B2 (en) 2011-01-10 2017-12-12 Infinity Pharmaceuticals, Inc. Solid forms of (S)-3-(1-(9H-purin-6-ylamino)ethyl)-8-chloro-2-phenylisoquinolin-1 (2H)-one and methods of use thereof
US10550122B2 (en) 2011-01-10 2020-02-04 Infinity Pharmaceuticals, Inc. Solid forms of (S)-3-(1-(9H-purin-6-ylamino)ethyl)-8-chloro-2-phenylisoquinolin-1(2H)-one and methods of use thereof
US8809349B2 (en) 2011-01-10 2014-08-19 Infinity Pharmaceuticals, Inc. Processes for preparing isoquinolinones and solid forms of isoquinolinones
US11312718B2 (en) 2011-01-10 2022-04-26 Infinity Pharmaceuticals, Inc. Formulations of (S)-3-(1-(9H-purin-6-ylamino)ethyl)-8-chloro-2-phenylisoquinolin-1(2H)-one
US9290497B2 (en) 2011-01-10 2016-03-22 Infinity Pharmaceuticals, Inc. Processes for preparing isoquinolinones and solid forms of isoquinolinones
US9295673B2 (en) 2011-02-23 2016-03-29 Intellikine Llc Combination of mTOR inhibitors and P13-kinase inhibitors, and uses thereof
US9718815B2 (en) 2011-07-19 2017-08-01 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9056877B2 (en) 2011-07-19 2015-06-16 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9605003B2 (en) 2011-07-19 2017-03-28 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US8969363B2 (en) 2011-07-19 2015-03-03 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US8785470B2 (en) 2011-08-29 2014-07-22 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9546180B2 (en) 2011-08-29 2017-01-17 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9115141B2 (en) 2011-08-29 2015-08-25 Infinity Pharmaceuticals, Inc. Substituted isoquinolinones and methods of treatment thereof
US9321772B2 (en) 2011-09-02 2016-04-26 The Regents Of The University Of California Substituted pyrazolo[3,4-D]pyrimidines and uses thereof
US9895373B2 (en) 2011-09-02 2018-02-20 The Regents Of The University Of California Substituted pyrazolo[3,4-D]pyrimidines and uses thereof
US8940742B2 (en) 2012-04-10 2015-01-27 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9255108B2 (en) 2012-04-10 2016-02-09 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US8828998B2 (en) 2012-06-25 2014-09-09 Infinity Pharmaceuticals, Inc. Treatment of lupus, fibrotic conditions, and inflammatory myopathies and other disorders using PI3 kinase inhibitors
US9527847B2 (en) 2012-06-25 2016-12-27 Infinity Pharmaceuticals, Inc. Treatment of lupus, fibrotic conditions, and inflammatory myopathies and other disorders using PI3 kinase inhibitors
US10131668B2 (en) 2012-09-26 2018-11-20 The Regents Of The University Of California Substituted imidazo[1,5-a]pYRAZINES for modulation of IRE1
US10822340B2 (en) 2012-09-26 2020-11-03 The Regents Of The University Of California Substituted imidazolopyrazine compounds and methods of using same
US11613544B2 (en) 2012-09-26 2023-03-28 The Regents Of The University Of California Substituted imidazo[1,5-a]pyrazines for modulation of IRE1
US9481667B2 (en) 2013-03-15 2016-11-01 Infinity Pharmaceuticals, Inc. Salts and solid forms of isoquinolinones and composition comprising and methods of using the same
US9828377B2 (en) 2013-10-04 2017-11-28 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9751888B2 (en) 2013-10-04 2017-09-05 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US10329299B2 (en) 2013-10-04 2019-06-25 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9359365B2 (en) 2013-10-04 2016-06-07 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US11541059B2 (en) 2014-03-19 2023-01-03 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US10675286B2 (en) 2014-03-19 2020-06-09 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9775844B2 (en) 2014-03-19 2017-10-03 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US11944631B2 (en) 2014-04-16 2024-04-02 Infinity Pharmaceuticals, Inc. Combination therapies
US11110096B2 (en) 2014-04-16 2021-09-07 Infinity Pharmaceuticals, Inc. Combination therapies
US9708348B2 (en) 2014-10-03 2017-07-18 Infinity Pharmaceuticals, Inc. Trisubstituted bicyclic heterocyclic compounds with kinase activities and uses thereof
US10253047B2 (en) 2014-10-03 2019-04-09 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US10941162B2 (en) 2014-10-03 2021-03-09 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US11247995B2 (en) 2015-09-14 2022-02-15 Infinity Pharmaceuticals, Inc. Solid forms of isoquinolinones, and process of making, composition comprising, and methods of using the same
US11939333B2 (en) 2015-09-14 2024-03-26 Infinity Pharmaceuticals, Inc. Solid forms of isoquinolinones, and process of making, composition comprising, and methods of using the same
US10160761B2 (en) 2015-09-14 2018-12-25 Infinity Pharmaceuticals, Inc. Solid forms of isoquinolinones, and process of making, composition comprising, and methods of using the same
US10759806B2 (en) 2016-03-17 2020-09-01 Infinity Pharmaceuticals, Inc. Isotopologues of isoquinolinone and quinazolinone compounds and uses thereof as PI3K kinase inhibitors
US10919914B2 (en) 2016-06-08 2021-02-16 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US11147818B2 (en) 2016-06-24 2021-10-19 Infinity Pharmaceuticals, Inc. Combination therapies
CN110799517A (en) * 2017-01-27 2020-02-14 中央研究院 Compound having analgesic effect for preventing and treating pain
US11753432B2 (en) 2017-01-27 2023-09-12 Academia Sinica Compound with analgesic effect for use in prevention and treatment of pain

Also Published As

Publication number Publication date
GB0317951D0 (en) 2003-09-03
WO2005012323A3 (en) 2005-05-26

Similar Documents

Publication Publication Date Title
WO2005012323A2 (en) Compounds useful as a3 adenosine receptor agonists
US7414036B2 (en) Compounds useful as A3 adenosine receptor agonists
US6448236B1 (en) Purine derivatives
JP2003502339A (en) Purine derivatives
CZ20021223A3 (en) Purine derivatives
CA3070004A1 (en) Piperazine heteroaryl derivative, preparation method therefor and use of same in medicine
TW201811775A (en) Ectonucleotidase inhibitors and methods of use thereof
HUT76980A (en) Novel naphthyridine derivative and medicinal composition thereof
CA3081751A1 (en) Substituted pyrrolopyrimidine jak inhibitors and methods of making and using the same
KR20030036138A (en) Purine Derivatives
EP3962897A1 (en) Substituted pyrrolopyridines as jak inhibitors
WO2018140648A1 (en) Pyrrolopyrimidine itk inhibitors for treating inflammation and cancer
KR20230144550A (en) Inhibitors of cGAS activity as therapeutic agents
NZ252109A (en) Adenosine derivatives and pharmaceutical compositions
JP2022531899A (en) Modified cyclic dinucleoside compound as a STING modulator
WO2002002550A1 (en) Novel pyrazinone derivatives
SK280336B6 (en) Pharmaceutical compositions containing pyridazino[4,5-b]quinolines, methods for their preparation
EP1469864A1 (en) Compounds useful as a-3 adenosine receptor agonists
WO2021022178A1 (en) Substituted sulfonamide pyrrolopyridines as jak inhibitors
WO2001049291A1 (en) 10,11-difluoromethylenedioxycamptothecin compounds with topoisomerase i inhibition
JP2023532027A (en) Novel compound and pharmaceutical composition for prevention or treatment of resistant cancer containing the same
AU2003202076A1 (en) Compounds useful as A3 adenosine receptor agonists
CN113924293B (en) P2X3 and/or P2X2/3 receptor antagonists, pharmaceutical compositions containing them and their use
JP7125714B2 (en) 2&#39;-Deoxy-7-deazapurine nucleoside derivatives with antiviral activity
CN118076610A (en) Macrocyclic compounds with ENT1 inhibitory activity

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
122 Ep: pct application non-entry in european phase