US20070185064A1 - 2-Aminopurine Analogs Having HSP90-Inhibiting Activity - Google Patents

2-Aminopurine Analogs Having HSP90-Inhibiting Activity Download PDF

Info

Publication number
US20070185064A1
US20070185064A1 US11/531,221 US53122106A US2007185064A1 US 20070185064 A1 US20070185064 A1 US 20070185064A1 US 53122106 A US53122106 A US 53122106A US 2007185064 A1 US2007185064 A1 US 2007185064A1
Authority
US
United States
Prior art keywords
compound
substituents
tautomer
pharmaceutically acceptable
prodrug
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/531,221
Inventor
Srinivas Kasibhatla
Kevin Hong
Marcus Boehm
Marco Biamonte
Lin Zhang
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Conforma Therapeutics Corp
Original Assignee
Conforma Therapeutics Corp
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 Conforma Therapeutics Corp filed Critical Conforma Therapeutics Corp
Priority to US11/531,221 priority Critical patent/US20070185064A1/en
Publication of US20070185064A1 publication Critical patent/US20070185064A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/04Drugs for skeletal disorders for non-specific disorders of the connective tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/08Antiepileptics; Anticonvulsants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D473/00Heterocyclic compounds containing purine ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D473/00Heterocyclic compounds containing purine ring systems
    • C07D473/02Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6
    • C07D473/18Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6 one oxygen and one nitrogen atom, e.g. guanine
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D473/00Heterocyclic compounds containing purine ring systems
    • C07D473/02Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6
    • C07D473/24Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6 one nitrogen and one sulfur atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D473/00Heterocyclic compounds containing purine ring systems
    • C07D473/26Heterocyclic compounds containing purine ring systems with an oxygen, sulphur, or nitrogen atom directly attached in position 2 or 6, but not in both
    • C07D473/32Nitrogen atom

Definitions

  • This application also relates to three other United States Utility Applications, entitled PYRAZOLOPYRIDIMES AND RELATED ANALOGS AS HSP90 INHIBITORS, PYRROLOPYRIMIDINES AND RELATED ANALOGS AS HSP90 INHIBITORS, and TRIAZOPYRIMIDES AND RELATED ANALOGS AS HSP90 INHIBITORS, which will be filed on the same date by the same entity.
  • This application further relates to International Application PCT/US02/35069, filed Oct. 30, 2002, entitled PURINE ANALOGS HAVING HSP90-INHIBITING ACTIVITY. All the. above cited U.S. utility applications, provisional applications and international application are expressly incorporated herein by reference in their entirety.
  • the invention relates in general to 2-aminopurine analogs and their broad-spectrum utility, e.g., in inhibiting heat shock protein 90 (HSP90) to thereby treat or prevent HSP90-mediated diseases.
  • HSP90 heat shock protein 90
  • HSP90s are ubiquitous chaperone proteins that are involved in folding, activation and assembly of a wide range of proteins, including key proteins involved in signal transduction, cell cycle control and transcriptional regulation.
  • HSP90 chaperone proteins are associated with important signaling proteins, such as steroid hormone receptors and protein kinases, including, e.g., Raf-1, EGFR, v-Src family kinases, Cdk4, and ErbB-2 (Buchner J. TIBS 1999,24, 136-141; Stepanova, L. et al. Genes Dev. 1996,10, 1491-502; Dai, K. et al. J. Biol. Chem. 1996, 271, 22030-4).
  • HSP70 e.g., HSP70, p60/Hop/Sti1, Hip, Bag1, HSP40/Hdj2/Hsj1, immunophilins, p23, and p50
  • HSP90 may assist HSP90 in its function (see, e.g., Caplan, A. Trends in Cell Biol 1999, 9, 262-68).
  • Ansamycin antibiotics e.g., herbimycin A (HA), geldanamycin (GM), and 17-allylaminogeldanamycin (17-AAG) are thought to exert their anticancerous effects by tight binding of the N-terminus pocket of HSP90, thereby destabilizing substrates that normally interact with HSP90 (Stebbins, C. et al. Cell 1997, 89, 239-250).
  • This pocket is highly conserved and has weak homology to the ATP-binding site of DNA gyrase (Stebbins, C. et al., supra; Grenert, J. P. et al. J. Biol. Chem. 1997, 272, 23843-50).
  • ATP and ADP have both been shown to bind this pocket with low affinity and to have weak ATPase activity (Proromou, C. et al. Cell 1997, 90, 65-75; Panaretou, B. et al. EMBO J. 1998, 17, 4829-36).
  • In vitro and in vivo studies have demonstrated that occupancy of this N-terminal pocket by ansamycins and other HSP90 inhibitors alters HSP90 function and inhibits protein folding.
  • ansamycins and other HSP90 inhibitors have been shown to prevent binding of protein substrates to HSP90 (Scheibel, T. H. et al. Proc. Natl. Acad. Sci.
  • the substrates are degraded by a ubiquitin-dependent process in the proteasome (Schneider, C. L., supra; Sepp-Lorenzino, L., et al. J. Biol. Chem. 1995, 270, 16580-16587; Whitesell, L. et al. Proc. Natl. Acad. Sci. USA 1994, 91, 8324-8328).
  • HSP90 substrate destabilization occurs in tumor and non-transformed cells alike and has been shown to be especially effective on a subset of signaling regulators, e.g., Raf (Schulte, T. W. et al. Biochem. Biophys. Res. Commun. 1997, 239, 655-9; Schulte, T. W., et al. J. Biol. Chem. 1995, 270, 24585-8), nuclear steroid receptors (Segnitz, B.; U. Gehring J. Biol. Chem. 1997, 272, 18694-18701; Smith, D. F. et al. Mol. Cell. Biol.
  • Ansamycins thus hold great promise for the treatment and/or prevention of many types of cancers and proliferative disorders, and also hold promise as traditional antibiotics.
  • their relative insolubility makes them difficult to formulate and administer, and they are not easily synthesized and currently must, at least in part, be generated through fermentation.
  • the dose limiting toxicity of ansamycins is hepatic.
  • HSP90 inhibitors have also been implicated in a wide variety of other utilities, including use as anti-inflammation agents, anti-infectious disease agents, agents for treating autoimmunity, agents for treating stroke, ischemia, multiple sclerosis, cardiac disorders, central nervous system related disorders and agents usefull in promoting nerve regeneration (See, e.g., Rosen et al. WO 02/09696 (PCT/US01/23640); Degranco et al. WO 99/51223 (PCT/US99/07242); Gold, U.S. Pat. No. 6,210,974 B1; DeFranco et al., U.S. Pat. No. 6,174,875.
  • fibrogenetic disorders including but not limited to scleroderma, polymyositis, systemic lupus, rheumatoid arthritis, liver cirrhosis, keloid formation, interstitial nephritis, and pulmonary fibrosis also may be treatable with HSP90 inhibitors.
  • Still further HSP90 modulation, modulators and uses thereof are reported in Application Nos.
  • the present invention is directed towards heterocyclic compounds, in particular towards 2-aminopures and related compounds that show broad utility, e.g., in inhibiting HSP90 and/or treating and preventing diseases that are HSP90-dependent.
  • the invention comprises the heterocyclic compounds as specified below in Formulae A and I, and compounds that are produced by a process of the invention.
  • stereoisomeric forms including the individual enantiomers and diastereomers, racemic mixtures, and diastereomeric mixtures, as well as polymorphs, solvates, esters, tautomers, pharmaceutically acceptable salts and prodrugs of these compounds.
  • Stereoisomers of the compounds of the present invention may be isolated by standard resolution techniques such as, for example, fractional crystallization and chiral column chromatography.
  • the invention provides compounds of Formula A, or a polymorph, solvate, ester, tautomer, diastereomer, enantiomer, pharmaceutically acceptable salt or prodrug thereof, which show utility by inhibiting HSP90 and treating and/or preventing diseases that are HSP90-dependent.
  • the invention provides compounds of Formula I, or a polymorph, solvate, ester, tautomer, enantiomer, diastereomer, pharmaceutically acceptable salt or prodrug thereof, which show utility for inhibiting HSP90 and for treating and/or preventing diseases that are HSP90-dependent, wherein:
  • the invention provides compounds, or a polymorph, solvate, ester, tautomer, diastereomer, enantiomer, pharmaceutically acceptable salt or prodrug thereof, which show utility for inhibiting HSP90 and treating and/or preventing diseases that are HSP90-dependent, that are prepared by the process comprising:
  • Another aspect of the invention is a method of treating an individual having an HSP90 mediated disorder comprising administering to said individual a pharmaceutical composition comprising a pharmaceutically effective amount of a compound of Formula I: or a polymorph, solvate, ester, tautomer, enantiomer, diastereomer, pharmaceutically acceptable salt or prodrug thereof, wherein:
  • the invention provides a method for treating an individual having a disorder selected from the group of inflammatory diseases, infections, autoimmune disorders, stroke, ischemia, cardiac disorders, neurological disorders, fibrogenetic disorders, proliferative disorders, tumors, leukemias, neoplasms, cancers, carcinomas, metabolic diseases, and malignant disease.
  • the invention provides a method for treating an individual having a fibrogenetic disorder, such as, for example, scleroderma, polymyositis, systemic lupus, rheumatoid arthritis, liver cirrhosis, keloid formation, interstitial nephritis and pulmonary fibrosis.
  • a fibrogenetic disorder such as, for example, scleroderma, polymyositis, systemic lupus, rheumatoid arthritis, liver cirrhosis, keloid formation, interstitial nephritis and pulmonary fibrosis.
  • the invention provides a combination therapy comprising the administration of a pharmaceutically effective amount of a compound of Formulae A, I or II, or a solvate, tautomer, pharmaceutically acceptable salt, polymorph, or prodrug thereof according to any of the preceding aspects or embodiments, and at least one therapeutic agent selected from the group of cytotoxic agents, anti-angiogenesis agents and anti-neoplastic agents.
  • the anti-neoplastic agent may be selected from the group of alkylating agents, anti-metabolites, epidophyllotoxins antineoplastic enzymes, topoisomerase inhibitors, procarbazines, mitoxantrones, platinum coordination complexes, biological response modifiers and growth inhibitors, honnonal/anti-hormonal therapeutic agents, and haematopoietic growth factors.
  • the present invention is directed to pharmaceutical compositions comprising the compounds of the invention, in particular, the compounds of Formulae A, I and II, and compounds formed by the process of the invention, and their polymorphs, solvates, esters, tautomers, diastereomer, enantiomers, pharmaceutically acceptable salts and prodrugs thereof, and one or more pharmaceutical excipients, for use in treatment or prevention of diseases that are HSP90-dependent.
  • Advantages of the invention depend on the specific aspect and embodiment and may include one or more of: ease of synthesis and/or formulation, solubility, and IC 50 relative to previously existing compounds in the same or different classes of HSP90 inhibitors.
  • a “pharmaceutically acceptable derivative or prodrug” means any pharmaceutically acceptable salt, ester, salt of an ester or other derivative of a compound of this invention, which, upon administration to a recipient, is capable of providing, either directly or indirectly, a compound of this invention or a pharmaceutically active metabolite or residue thereof.
  • Particularly favored derivatives or prodrugs are those that increase the bioavailability of the compounds of this invention when such compounds are administered to a patient (e.g., by allowing orally administered compound to be more readily absorbed into blood) or which enhance delivery of the parent compound to a biological compartment (e.g., the brain or lymphatic system).
  • a “pharmaceutically acceptable salt” may be prepared for any compound of the invention having a functionality capable of forming a salt, for example, an acid or base functionality.
  • Pharmaceutically acceptable salts may be derived from organic or inorganic acids and bases.
  • Compounds of the invention that contain one or more basic functional groups, e.g., amino or alkylamino, are capable of forming pharmaceutically acceptable salts with pharmaceutically acceptable organic and inorganic acids.
  • These salts can be prepared in situ during the final isolation and purification of the compounds of the invention, or by separately reacting a purified compound of the invention in its free base form with a suitable organic or inorganic acid, and isolating the salt thus formed.
  • Suitable acid salts include acetate, adipate, alginate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, flimarate, glucoheptanoate, glycerophosphate, glycolate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate, lactate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oxalate, palmoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate
  • compositions of the present invention that contain one or more acidic functional groups are capable of forming pharmaceutically acceptable salts with pharmaceutically acceptable bases.
  • pharmaceutically acceptable salts refers to the relatively non-toxic, inorganic and organic base addition salts of compounds of the present invention. These salts can likewise be prepared in situ during the final isolation and purification of the compounds, or by separately reacting the purified compound in its free acid form with a suitable base, such as the hydroxide, carbonate or bicarbonate of a pharmaceutically acceptable metal cation, with ammonia, or with a pharmaceutically acceptable organic primary, secondary or tertiary amine.
  • suitable base such as the hydroxide, carbonate or bicarbonate of a pharmaceutically acceptable metal cation, with ammonia, or with a pharmaceutically acceptable organic primary, secondary or tertiary amine.
  • alkali or alkaline earth salts include the lithium, sodium, potassium, calcium, magnesium, and aluminum salts and the like.
  • Illustrative examples of some of the bases that can be used include sodium hydroxide, potassium hydroxide, choline hydroxide, sodium carbonate, N + (C 1-4 alkyl) 4 , and the like.
  • Representative organic amines useful for the formation of base addition salts include ethylamine, diethylamine, ethylenediamine, ethanolamine, diethanolamine, piperazine and the like.
  • This invention also envisions the quaternization of any basic nitrogen-containing groups of the compounds disclosed herein. Water or oil-soluble or dispersible products may be obtained by such quaternization. See, for example, Berge et al., supra.
  • prodrugs of the compounds of this invention include, but are not limited to, esters, carbonates, thiocarbonates, N-acyl derivatives, N-acyloxyalkyl derivatives, quaternary derivatives of tertiary amines, N-Mannich bases, Schiff bases, aminoacid conjugates, phosphate esters, metal salts and sulfonate esters.
  • Suitable positions for derivatization of the compounds of the invention to create “prodrugs” include but are not limited, 2-amino substitution. Those of ordinary skill in the art have the knowledge and means to accomplish this without undue experimentation. Various forms of prodrugs are well known in the art. For examples of such prodrug derivatives, see, e.g.,
  • Bundgaard H. “Design and Application of Prodrugs” in A Textbook of Drug Design and Development , Krosgaard-Larsen and H. Bundgaard, Ed., 1991, Chapter 5, p. 113-191; and
  • prodrugs as employed herein includes, but is not limited to, the following groups and combinations of these groups:
  • alkyl refers to an optionally substituted straight-chain, or optionally substituted branched-chain saturated hydrocarbon radical having from one to about thirty carbons, more preferably one to twelve carbons.
  • alkyl radicals include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, tert-amyl, pentyl, hexyl, heptyl, octyl and the like.
  • cycloalkyl embraces cyclic alkyl radicals which include monocyclic, bicyclic, tricyclic, and higher multicyclic alkyl radicals wherein each cyclic moiety has from three to about eight carbon atoms.
  • examples of cycloalkyl radicals include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and the like.
  • a “lower alkyl” is a shorter alkyl, e.g., one containing from one to about six carbon atoms.
  • alkenyl refers to an optionally substituted straight-chain, or optionally substituted branched-chain hydrocarbon radical having one or more carbon-carbon double-bonds and having from two to about thirty carbon atoms, more preferably two to about eighteen carbons.
  • alkenyl radicals include ethenyl, propenyl, butenyl, 1,3-butadienyl and the like.
  • cycloalkenyl refers to cyclic alkenyl radicals which include monocyclic, bicyclic, tricyclic, and higher multicyclic alkenyl radicals wherein each cyclic moiety has from three to about eight carbon atoms.
  • a “lower alkenyl” refers to an alkenyl having from two to about six carbons.
  • alkynyl refers to an optionally substituted straight-chain or optionally substituted branched-chain hydrocarbon radical having one or more carbon-carbon triple-bonds and having from two to about thirty carbon atoms, more preferably from two to about twelve carbon atoms, from two to about six carbon atoms as well as those having from two to about four carbon atoms.
  • alkynyl radicals include ethynyl, 2-propynyl, 2-butynyl, 1,3-butadiynyl and the like.
  • cycloalkynyl refers to cyclic alkynyl radicals which include monocyclic, bicyclic, tricyclic, and higher multicyclic alkynyl radicals wherein each cyclic moiety has from three to about eight carbon atoms.
  • a “lower alkynyl” refers to an alkynyl having from two to about six carbons.
  • heteroalkyl, heteroalkenyl and heteroalkynyl include optionally substituted alkyl, alkenyl and alkynyl structures, as described above, and which have one or more skeletal chain atoms selected from an atom other than carbon, e.g., oxygen, nitrogen, sulfur, phosphorous or combinations thereof.
  • carbon chain embraces any alkyl, alkenyl, alkynyl, or heteroalkyl, heteroalkenyl, or heteroalkynyl group, which are linear, cyclic, or any combination thereof. If the chain is part of a linker and that linker comprises one or more rings as part of the core backbone, for purposes of calculating chain length, the “chain” only includes those carbon atoms that compose the bottom or top of a given ring and not both, and where the top and bottom of the ring(s) are not equivalent in length, the shorter distance shall be used in determining the chain length. If the chain contains heteroatoms as part of the backbone, those atoms are not calculated as part of the carbon chain length.
  • membered ring can embrace any cyclic structure, including aromatic, heteroaromatic, alicyclic, heterocyclic and polycyclic fused ring systems as described below.
  • membered is meant to denote the number of skeletal atoms that constitute the ring.
  • pyridine, pyran, and pyrimidine are six-membered rings and pyrrole, tetrahydrofuran, and thiophene are five-membered rings.
  • aryl refers to an optionally substituted aromatic hydrocarbon radical of six to about twenty ring atoms, and includes mono-aromatic rings and fused aromatic ring.
  • a fused aromatic ring radical contains from two to four fused rings where the ring of attachment is an aromatic ring, and the other individual rings within the fused ring may be aromatic, heteroaromatic, alicyclic or heterocyclic.
  • aryl includes mono-aromatic ring and fused aromatic rings containing from six to about twelve carbon atoms, as well as those containing from six to about ten carbon atoms.
  • aryl groups include, without limitation, phenyl, naphthyl, anthryl, chrysenyl, and benzopyrenyl ring systems.
  • lower aryl refers to an aryl having six to about ten skeletal ring carbons, e.g., phenyl and naphthyl ring systems.
  • heteroaryl refers to optionally substituted aromatic radicals containing from about five to about twenty skeletal ring atoms and where one or more of the ring atoms is a heteroatom such as, for example, oxygen, nitrogen, sulfur, selenium and phosphorus.
  • heteroaryl includes optionally substituted mono-heteroaryl radicals and fused heteroaryl radicals having at least one heteroatom (e.g., quinoline, benzothiazole).
  • a fused heteroaryl radical may contain from two to four fused rings and where the ring of attachment is a heteroaromatic ring, the other individual rings within the fused ring system may be aromatic, heteroaromatic, alicyclic or heterocyclic.
  • heteroaryl also includes mono-heteroaryls or fused heteroaryls having from five to about twelve skeletal ring atoms, as well as those having from five to about ten skeletal ring atoms.
  • heteroaryls include, without limitation, furanyl, benzofuranyl, chromenyl, pyridyl, pyrrolyl, indolyl, quinolinyl, pyridyl-N-oxide, pyrimidyl, pyrazinyl, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, benzothiozole, benzimidazole, benzoxazoles, benzothiadiazole, benzoxadiazole, benzotriazole, quinolines, isoquinolines, indoles, purinyl, indolizinyl, thienyl and the like and their oxides.
  • lower heteroaryl refers to a heteroaryl having five to about ten skeletal ring atoms, e.g., pyridyl, thienyl, pyrimidyl, pyrazinyl, pyrrolyl, or furanyl.
  • alicyclic alone or in combination, refers to an optionally substituted saturated or unsaturated nonaromatic hydrocarbon ring system containing from three to about twenty ring atoms.
  • the term alicyclic includes mono-alicyclic and fused alicyclic radicals.
  • a fused alicyclic may contain from two to four fused rings where the ring of attachment is an alicyclic ring, and the other individual rings within the fused-alicyclic radical may be aromatic, heteroaromatic, alicyclic and heterocyclic.
  • the term alicyclic also includes mono-alicyclic and fused alicyclic radicals containing from three to about twelve carbon atoms, as well as those containing from three to about ten carbon atoms.
  • alicyclics include, without limitation, cyclopropyl, cyclopropenyl, cyclobutyl, cyclopentyl, cyclodecyl, cyclododecyl, cyclopentadienyl, indanyl, and cyclooctatetraenyl ring systems.
  • the term “lower alicyclic” refers to an alicyclic having three to about ten skeletal ring carbons, e.g., cyclopropyl, cyclopropenyl, cyclobutyl, cyclopentyl, decalinyl, and cyclohexyl.
  • heterocyclic refers to optionally substituted saturated or unsaturated nonaromatic ring radicals containing from five to about twenty ring atoms where one or more of the ring atoms are heteroatoms such as, for example, oxygen, nitrogen, sulfur, and phosphorus.
  • alicyclic includes mono-heterocyclic and fused heterocyclic ring radicals.
  • a fused heterocyclic radical may contain from two to four fused rings where the attaching ring is a heterocyclic, and the other individual rings within the fused heterocyclic radical may be aromatic, heteroaromatic, alicyclic or heterocyclic.
  • heterocyclic also includes mono-heterocyclic and fused alicyclic radicals having from five to about twelve skeletal ring atoms, as well as those having from five to about ten skeletal ring atoms.
  • Example of heterocyclics include without limitation, tetrahydrofuranyl, benzodiazepinyl, tetrahydroindazolyl, dihyroquinolinyl, and the like.
  • the term “lower heterocyclic” refers to a heterocyclic ring system having five to about ten skeletal ring atoms, e.g., dihydropyranyl, pyrrolidinyl, indolyl, piperidinyl, piperazinyl, and the like.
  • alkylaryl refers to an aryl radical as defined above in which one H atom is replaced by an alkyl radical as defined above, such as, for example, tolyl, xylyl and the like.
  • arylalkyl refers to an alkyl radical as defined above in which one H atom is replaced by an aryl radical as defined above, such as, for example, benzyl, 2-phenylethyl and the like.
  • heteroarylalkyl refers to an alkyl radical as defined above in which one H atom is replaced by a heteroaryl radical as defined above, each of which may be optionally substituted.
  • alkoxy refers to an alkyl ether radical, alkyl-O—, wherein the term alkyl is defined as above.
  • alkoxy radicals include methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, iso-butoxy, sec-butoxy, tert-butoxy and the like.
  • aryloxy refers to an aryl ether radical wherein the term aryl is defined as above.
  • aryloxy radicals include phenoxy, benzyloxy and the like.
  • alkylthio refers to an alkyl thio radical, alkyl-S—, wherein the term alkyl is as defined above.
  • arylthio refers to an aryl thio radical, aryl-S—, wherein the term aryl is as defined above.
  • heteroarylthio refers to the group heteroaryl-S—, wherein the term heteroaryl is as defined above.
  • acyl refers to a radical —C(O)R where R includes alkyl, alkenyl, alkynyl, aryl, heteroaryl, alicyclic, heterocyclic, arylalkyl or heteroarylalkyl wherein the alkyl, alkenyl, alkynyl, aryl, heteroaryl, alicyclic, heterocyclic, arylalkyl or heteroaryl alkyl groups may be optionally substituted.
  • acyloxy refers to the ester group —OC(O)R, where R is H, alkyl, alkenyl, alkynyl, aryl, heteroaryl, alicyclic, heterocyclic, arylalkyl, or heteroarylalkyl wherein the alkyl, alkenyl, alkynyl, aryl, heteroaryl, alicyclic, heterocyclic, arylalkyl or heteroarylalkyl may be optionally substituted.
  • carboxy esters refers to —C(O)OR where R is alkyl, aryl or arylalkyl, wherein the alkyl, aryl and arylalkyl groups may be optionally substituted.
  • R and R′ are independently selected from the group consisting of H, alkyl, aryl, heteroaryl, alicyclic, heterocyclic, arylalkyl and heteroarylalkyl, wherein the alkyl, aryl, heteroaryl, alicyclic, heterocyclic, or arylalkyl groups may be optionally substituted.
  • halogen includes F, Cl, Br and I.
  • haloalkyl, haloalkenyl, haloalkynyl and haloalkoxy include alkyl, alkenyl, alkynyl and alkoxy structures, as described above, that are substituted with one or more fluorines, chlorines, bromines or iodines, or with combinations thereof.
  • perhaloalkyl, perhaloalkyloxy and perhaloacyl refer to alkyl, alkyloxy and acyl radicals as described above, that all the H atoms are substituted with fluorines, chlorines, bromines or iodines, or combinations thereof.
  • cycloalkyl, arylalkyl, aryl, heteroaryl, alicyclic, heterocyclic, alkyl, alkynyl, alkenyl, haloalkyl, and heteroalkyl include optionally substituted cycloalkyl, arylalkyl, aryl, heteroaryl, alicyclic, heterocyclic, alkyl alkynyl, alkenyl, haloalkyl and heteroalkyl groups.
  • alkylamino refers to the group —NHR where R is independently selected from alkyl.
  • dialkylamino refers to the group —NRR′ where R and R′ are alkyls.
  • sulfuride refers to a sulfur atom covalently linked to two atoms; the formal oxidation state of said sulfur is (II).
  • thioether may be used interchangeably with the term “sulfide.”
  • sulfoxide refers to a sulfur atom covalently linked to three atoms, at least one of which is an oxygen atom; the formal oxidation state of said sulfur atom is (IV).
  • sulfurone refers to a sulfur atom covalently linked to four atoms, at least two of which are oxygen atoms; the formal oxidation state of said sulfur atom is (VI).
  • aryl optionally mono- or di-substituted with an alkyl means that the alkyl may but need not be present, or either one alkyl or two may be present, and the description includes situations where the aryl is substituted with one or two alkyls and situations where the aryl is not substituted with an alkyl.
  • “Optionally substituted” groups may be substituted or unsubstituted.
  • the substituents of an “optionally substituted” group may include, without limitation, one or more substituents independently selected from the following groups or designated subsets thereof: lower alkyl, lower alkenyl, lower alkynyl, lower aryl, heteroaryl, alicyclic, heterocyclic, arylalkyl, heteroarylalkyl, lower alkoxy, lower aryloxy, amino, alkylamino, dialkylamino, diarylalkylamino, alkylthio, arylthio, heteroarylthio, oxo, oxa, carbonyl (—C(O)), carboxyesters (—C(O)OR), carboxamido (—C(O)NH 2 ), carboxy, acyloxy, —H, halo, —CN, —NO 2 , —N 3 , —SH, —OH, —C(O)
  • An optionally substituted group may be unsubstituted (e.g., —CH 2 CH 3 ), fully substituted (e.g., —CF 2 CF 3 ), monosubstituted (e.g., —CH 2 CH 2 F) or substituted at a level anywhere in-between fully substituted and monosubstituted (e.g., —CH 2 CF 3 ).
  • pyridine-1-oxy also means “pyridine—N-oxy.”
  • Some of the compounds of the present invention may contain one or more chiral centers and therefore may exist in enantiomeric and diastereomeric forms.
  • the scope of the present invention is intended to cover all isomers per se, as well as mixtures of cis and trans isomers, mixtures of diastereomers and racemic mixtures of enantiomers (optical isomers) as well. Further, it is possible using well known techniques to separate the various forms, and some embodiments of the invention may feature purified or enriched species of a given enantiomer or diastereomer.
  • a “pharmacological composition” refers to a mixture of one or more of the compounds described herein, or pharmaceutically acceptable salts thereof, with other chemical components, such as pharmaceutically acceptable carriers and/or excipients.
  • the purpose of a pharmacological composition is to facilitate administration of a compound to an organism.
  • pharmaceutically acceptable carrier means a pharmaceutically-acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting the subject agent from one organ, or portion of the body, to another organ, or portion of the body.
  • a pharmaceutically-acceptable material such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting the subject agent from one organ, or portion of the body, to another organ, or portion of the body.
  • Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient.
  • materials which can serve as pharmaceutically-acceptable carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydrox
  • excipient refers to an inert substance added to a pharmacological composition to further facilitate administration of a compound.
  • excipients include but are not limited to calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils and polyethylene glycols.
  • a “pharmaceutically effective amount” means an amount which is capable of providing a therapeutic and/or prophylactic effect.
  • the specific dose of compound administered according to this invention to obtain therapeutic and/or prophylactic effect will, of course, be determined by the particular circumstances surrounding the case, including, for example, the specific compound administered, the route of administration, the condition being treated, and the individual being treated.
  • a typical daily dose (administered in single or divided doses) will contain a dosage level of from about 0.01 mg/kg to about 50-100 mg/kg of body weight of an active compound of the invention.
  • Preferred daily doses generally will be from about 0.05 mg/kg to about 20 mg/kg and ideally from about 0.1 mg/kg to about 10 mg/kg.
  • Factors such as clearance rate, half-life and maximum tolerated dose (MTD) have yet to be determined but one of ordinary skill in the art can determine these using standard procedures.
  • the preferred therapeutic effect is the inhibition, to some extent, of the growth of cells characteristic of a proliferative disorder, e.g., breast cancer.
  • a therapeutic effect will also normally, but need not, relieve to some extent one or more of the symptoms other than cell growth or size of cell mass.
  • a therapeutic effect may include, for example, one or more of 1) a reduction in the number of cells; 2) a reduction in cell size; 3) inhibition (i.e., slowing to some extent, preferably stopping) of cell infiltration into peripheral organs, e.g., in the instance of cancer metastasis; 3) inhibition (i.e., slowing to some extent, preferably stopping) of tumor metastasis; 4) inhibition, to some extent, of cell growth; and/or 5) relieving to some extent one or more of the symptoms associated with the disorder.
  • IC 50 refers to an amount, concentration or dosage of a particular test compound that achieves a 50% inhibition of a maximal response in an assay that measures such response.
  • the “IC 50 ” value of a compound of the invention can be greater for normal cells than for cells exhibiting a proliferative disorder, e.g., breast cancer cells. The value depends on the assay used.
  • a “standard” is meant a positive or negative control.
  • a negative control in the context of HER2 expression levels is, e.g., a sample possessing an amount of HER2 protein that correlates with a normal cell.
  • a negative control may also include a sample that contains no HER2 protein.
  • a positive control does contain HER2 protein, preferably of an amount that correlates with overexpression as found in proliferative disorders, e.g., breast cancers.
  • the controls may be from cell or tissue samples, or else contain purified ligand (or absent ligand), immobilized or otherwise. In some embodiments, one or more of the controls may be in the form of a diagnostic “dipstick.”
  • selective targeting is meant affecting one type of cell to a greater extent than another, e.g., in the case of cells with high as opposed to relatively low or normal HER2 levels.
  • One embodiment of the compounds of the invention is of Formula A: or a polymorph, solvate, ester, tautomer, diastereomer, enantiomer, pharmaceutically acceptable salt or prodrug thereof, wherein:
  • the compound, tautomer, pharmaceutically acceptable salt thereof, or prodrug thereof of Formula A, X 1 and X 2 are the same or different and each is nitrogen or —CR 6 ;
  • R 1 is halogen, —OR 8 , —SR 8 , or lower alkyl;
  • R 2 is —NR 8 R 10 ;
  • R 3 is hydrogen, —OH or keto tautomer, —OR 8 , halogen, —CN, lower alkyl, or —C(O)R 9 ;
  • R 5 is alkyl, aromatic, heteroaromatic, alicyclic, heterocyclic, each of which is optionally bi- or tricyclic, and optionally substituted with H, halogen, lower alkyl, —SR 8 , —OR 8 , —CN,
  • R 1 is selected from halogen, hydroxyl, lower alkoxy, lower thioalkyl and C 1-4 alkyl; and R 2 is —NH 2 .
  • R 4 is selected from halogen, hydroxyl, lower alkoxy, lower thioalkyl or C 1-4 alkyl; optionally wherein R 2 is NH 2 .
  • R 1 is halogen, hydroxyl, lower alkoxy, lower thioalkyl, or C 1-4 alkyl; and R 2 is optionally NH 2 , R 4 is —(CH 2 )—, and R 5 is phenyl, benzyl, or pyridyl, all optionally substituted with H, halogen, lower alkyl, —SR 8 , —OR 8 (or cyclic ethers such as methylenedioxy), —CN, —C0 2 R 9 , —NO 2 , or —NR 8 R 10 ; R 8 is hydrogen, lower alkyl, lower aryl or —(CO)R 9 ; R 9 is lower alkyl, lower aryl, lower heteroaryl, —NR 8 R 10 or —OR 11 ; R 11 is lower alkyl or lower aryl; and R 10 is hydrogen or lower alkyl.
  • R 1 is halogen
  • R 2 is —NH 2
  • R 4 is —CH 2 —
  • R 6 is H or halogen
  • R 5 is phenyl optionally substituted with H, halogen, C 1-4 alkyl, C 1-4 alkoxy, C 1-4 alkylthio, perhaloalkyl, perhaloalkyloxy, —CN, —NO 2 , —NH 2 or —CO 2 R 11 .
  • R 1 is halogen
  • R 2 is —NH 2
  • R 4 is —CH 2 —
  • R 6 is H
  • R 5 is 2-halo-3,5-dimethoxyphenyl optionally substituted with H, halogen, C 1-4 alkyl, C 1-4 alkoxy, C 1-4 alkylthio, perhaloalkyl, perhaloalkyloxy, —CN, —NO 2 , —NH 2 , or —CO 2 R 11 at the para (4 ⁇ ) position.
  • R 1 is chloro
  • R 2 is —NH 2
  • R 4 is —CH 2 —
  • R 6 is H
  • R 5 is 2-chloro-3,4,5-trimethoxyphenyl.
  • R 1 is chloro
  • R 2 is —NH 2
  • R 4 is —CH 2 —
  • R 6 is H
  • R 5 is 2-bromo-3,4,5-trimethoxyphenyl.
  • R 5 is selected from 2-iodo-3,4,5-trimethoxyphenyl, 2-fluoro-3,4,5-trimethoxyphenyl, and 2-bromo-3,4,5-trimethoxyphenyl.
  • the invention provides compounds of Formula A1: or a polymorph, solvate, ester, tautomer, diastereomer, enantiomer, pharmaceutically acceptable salt or prodrug thereof, wherein:
  • R 1 is halogen, hydroxyl, lower alkoxy, lower thioalkyl, or C 1-4 alkyl; and R 2 is NH 2 .
  • R 1 is halogen;
  • R 2 is NH 2 ,
  • R 4 is —CH 2 —.
  • R 4 is —(CH 2 )—;
  • R 5 is phenyl, benzyl, or pyridyl, and is independently optionally substituted with H, halogen, lower alkyl, —SR 8 , —OR 8 (or cyclic ethers such as methylenedioxy), —CN, —CO 2 R 9 , —NO 2 , or —NR 8 R 10 ;
  • R 8 is hydrogen, lower alkyl, lower aryl or —(CO)R 9 ;
  • R 9 is lower alkyl, lower aryl, lower heteroaryl, —NR 8 R 10 or —OR 11 ;
  • R 11 is lower alkyl or lower aryl; and
  • R 10 is hydrogen or lower alkyl.
  • R 1 is halogen
  • R 2 is —NH 2
  • R 4 is —CH 2 —
  • R 6 is H or halogen
  • R 5 is phenyl optionally substituted with H, halogen, C 1-4 alkyl, C 1-4 alkoxy, C 1-4 alkylthio, perhaloalkyl, perhaloalkyloxy, —CN, —NO 2 , —NH 2 or —CO 2 R 11 .
  • R 1 is halogen
  • R 2 is —NH 2
  • R 4 is —CH 2 —
  • R 6 is H
  • R 5 is 2-halo-3,5-dimethoxyphenyl optionally substituted with H, halogen, C 1-4 alkyl, C 1-4 alkoxy, C 1-4 alkylthio, perhaloalkyl, perhaloalkyloxy, —CN, —NO 2 , —NH 2 , or —CO 2 R 11 at the para (4 ⁇ ) position.
  • R 1 is chloro
  • R 2 is —NH 2
  • R 4 is —CH 2 —
  • R 6 is H
  • R 5 is 2-chloro-3,4,5 -trimethoxyphenyl.
  • R 1 is chloro
  • R 2 is —NH 2
  • R 4 is —CH 2 —
  • R 6 is H
  • R 5 is 2-bromo-3,4,5-trimethoxyphenyl.
  • R 1 is chloro
  • R 2 is —NH 2
  • R 4 is —CH 2 —
  • R 6 is H
  • R 5 is 2-iodo-3,4,5-trimethoxyphenyl.
  • R 1 is chloro
  • R 2 is —NH 2
  • R 4 is —CH 2 —
  • R 6 is H
  • R 5 is 2-fluoro-3,4,5-trimethoxyphenyl.
  • R 1 is bromo
  • R 2 is —NH 2
  • R 4 is —CH 2 —
  • R 6 is H
  • R 5 is 2-bromo-3,4,5-trimethoxyphenyl.
  • R 1 is bromo
  • R 2 is —NH 2
  • R 4 is —CH 2 —
  • R 6 is H
  • R 5 is 2-iodo-3,4,5-trimethoxyphenyl.
  • R 1 is bromo
  • R 2 is —NH 2
  • R 4 is —CH 2 —
  • R 6 is H
  • R 5 is 2-iodo-3,4,5-trimethoxyphenyl.
  • R 1 is bromo
  • R 2 is —NH 2
  • R 4 is —CH 2 —
  • R 6 is H
  • R 5 is 2-fluoro-3,4,5-trimethoxyphenyl.
  • R 1 is chloro
  • R 2 is —NH 2
  • R 4 is —CH 2 —
  • R 6 is halo
  • R 5 is 2-chloro-3,4,5-trimethoxyphenyl.
  • R 1 is chloro
  • R 2 is NH 2
  • R 4 is —CH 2 —
  • R 6 is halo
  • R 5 is 2-bromo-3,4,5-trimethoxyphenyl.
  • R 1 is bromo
  • R 2 is —NH 2
  • R 4 is —CH 2 —
  • R 6 is halo
  • R 5 is 2-chloro-3,4,5-trimethoxyphenyl.
  • R 1 is bromo
  • R 2 is —NH 2
  • R 4 is —CH 2 —
  • R 6 is halo
  • R 5 is 2-bromo-3,4,5-trimethoxyphenyl.
  • the invention provides compounds of Formula I: or a polymorph, solvate, ester, enantiomer, diastereomer, tautomer, pharmaceutically acceptable salt or prodrug thereof, wherein:
  • R 1 is halogen or methyl
  • R 2 is —NHR 8 , where R 8 is hydrogen or —C(O)R 9 .
  • R 1 is halogen;
  • R 2 is —NH 2 and R 3 is hydrogen.
  • R 4 is —(CH 2 )—.
  • R 1 is halogen;
  • R 2 is —NH 2 ;
  • R 4 is lower alkyl;
  • R 3 is hydrogen;
  • R 5 is aryl or heteroaryl, wherein each of said aryl and heteroaryl groups is monocyclic or bicyclic, the aryl group is substituted with 4 to 5 substituents, the heteroaryl group is substituted with 2 to 5 substituents, wherein when the heteroaryl is substituted with only two substituents, the two substituents must form part of an optionally substituted fused ring.
  • R 1 is halogen;
  • R 2 is —NH 2 ;
  • R 3 is hydrogen;
  • R 5 is aryl or heteroaryl, wherein each of said aryl and heteroaryl groups is monocyclic or bicyclic, the aryl group is substituted with 4 to 5 substituents, the heteroaryl group is substituted with 2 to 5 substituents, wherein when the heteroaryl is substituted with only two substituents, the two substituents must form part of an optionally substituted fused ring.
  • R 1 is chloro or bromo
  • R 2 is —NH 2
  • R 5 is a phenyl having 3 to 5 substituents.
  • R 1 is chloro or bromo
  • R 2 is —NH 2
  • R 5 is a pyridyl having 3 to 5 substituents.
  • R 1 is chloro or bromo
  • R 2 is —NH 2
  • R 5 is an 1-oxy-pyridyl (N-oxy-pyridyl) having 3 to 5 substituents.
  • each of the aryl, heteroaryl, alicyclic or heterocyclic group is monocyclic or bicyclic.
  • the invention provides compounds of Formula II, which are compounds of Formula I where R 4 is —CH 2 —, or a polymorph, solvate, ester, tautomer, enantiomer, diastereomer, pharmaceutically acceptable salt or prodrug thereof, wherein:
  • each of the aryl, heteroaryl, alicyclic or heterocyclic group is monocyclic or bicyclic.
  • R 1 is halogen or methyl; and R 2 is —NHR 8 , where R 8 is hydrogen or —C(O)R 9 .
  • R 1 is halogen
  • R 2 is —NH 2 and R 3 is hydrogen.
  • R 1 is halogen;
  • R 2 is —NH 2 ;
  • R 3 is hydrogen;
  • R 5 is aryl or heteroaryl, wherein each of the aryl and heteroaryl groups is monocyclic or bicyclic, the aryl group is substituted with 4 to 5 substituents, the heteroaryl group is substituted with 2 to 5 substituents, wherein when the heteroaryl is substituted with only two substituents, the two substituents must form part of an optionally substituted fused ring.
  • R 1 is chloro or bromo
  • R 2 is —NH 2
  • R 5 is a phenyl having 3 to 5 substituents.
  • R 1 is chloro or bromo
  • R 2 is —NH 2
  • R 5 is a pyridyl having 3 to 5 substituents.
  • R 1 is chloro or bromo
  • R 2 is —NH 2
  • R 5 is an 1-oxy-pyridyl (N-oxy-pyridyl) having 3 to 5 substituents.
  • the invention provides compounds, or polymorphs, solvates, esters, tautomers, pharmaceutically acceptable salts or prodrugs thereof, prepared by the process comprising:
  • Y is represented by any of the following formulae:
  • R 5 is aryl, heteroaryl, alicyclic, or heterocyclic, each of which is optionally mono- or bicyclic.
  • R 4 is —CH 2 —.
  • L 1 is —Cl, —Br or NH 2 ;
  • R 5 is aryl or heteroaryl, wherein the aryl group is substituted with 4 to 5 substituents and the heteroaryl group is substituted with 2 to 5 substituents wherein when the heteroaryl is substituted with only two substituents, the two substituents must form part of an optionally substituted fused ring.
  • Y is a substituted purine.
  • the reaction is performed in a solvent comprising a member selected from the group of DMF, THF and DMSO.
  • the reaction is performed in a solvent that comprises DMF.
  • HSP90 inhibitors of the following Formula I, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof, wherein:
  • R 4 R 5 is not a ribose or derivative thereof, or a sugar or derivative thereof;
  • R 3 is hydrogen, halogen or —CN;
  • R 5 is aryl, heteroaryl, alicyclic, or heterocyclic, all optionally substituted with halogen, lower alkyl, lower alkenyl, lower alkynyl, lower aryl, lower alicyclic, arylalkyl, aryloxy, aryloxyalkyl, alkoxyalkyl, perhaloalkyl, perhaloalkyloxy, perhaloacyl, —N 3 , —SR 8 , —OR 8 , —CN, —C(O)R 9 , —NO 2 , or —NR 8 R 10 .
  • R 1 is halogen or methyl and R 2 is —NHR 8 , where R 8 is hydrogen or —C(O)R 9 .
  • R 2 is —NH 2 and R 3 is hydrogen.
  • R 4 is —CH 2 —.
  • R 1 is halogen;
  • R 2 is —NH 2 ;
  • R 3 is hydrogen;
  • R 4 is —CH 2 —;
  • R 5 is aryl or heteroaryl, the aryl and heteroaryl groups are monocyclic or bicyclic, the aryl group is substituted with 4 to 5 substituents, the heteroaryl group is substituted with 2 to 5 substituents, wherein when the heteroaryl is substituted with two substituents, the two substituents must form part of an optionally substituted fused ring.
  • R 1 is chloro or bromo
  • R 2 is —NH 2
  • R 5 is a phenyl having 3 to 5 substituents.
  • R 1 is chloro or bromo
  • R 2 is —NH 2
  • R 5 is a pyridyl having 3 to 5 substituents.
  • R 1 is chloro or bromo
  • R 2 is —NH 2
  • R 5 is an 1-oxy-pyridyl (N-oxy-pyridyl) having 3 to 5 substituents.
  • compounds of interest in Table 1 are compounds 2, 3, 11, 44, 82, 83, 242, 243, 245, 248, 249, 254, 255, 260, 266, 272, 273, 278, 279, 284, 286, 287, 308, 309, 343, 348, 349, 354, 366, 367, 372, 373, 671 and 697 with those selected being 2, 242, 243, 248, 254, 260, 266, 272, 278, 284, 286, 287, 308, 343, 348, 349, 354, 372, 373, 671 and 697.
  • Synthesis of compounds of the present invention may be accomplished by various methods the art, including those described in, for example, Montgomery, J. Med. Pharm. Chem., 1962, 5, 15-24; Sircar, U.S. Pat. No. 4,772,606 (1988); Sircar, U.S. Pat. No. 4,748,177 (1988); Hans, U.S. Pat. No. 5,110,818 (1992); Gillespie, WO 02/055521; Matsuda, JP 10025294 A2, (1998).
  • Scheme 1 For the synthesis of compounds of Formulae I and II, a general strategy is outlined in Scheme 1. It should be understood that other methods can also be used.
  • the starting materials or the intermediates of the Formula 2, or/and 4 can exist in tautomeric forms as shown in FIG. 1. Both forms are indiscriminately described in the specifications.
  • the compounds of Formula I can be synthesized from the commercially available starting heterocycle, for example compounds of Formula 2 where R 6 is —Cl, —Br or —OH, R 7 is —NH 2 and R 8 is —H are commercially available from Aldrich, AlfaAesar, etc. Accordingly, Formula 2 can be alkylated in the presence of a base such as K 2 CO 3 , NaH, Cs 2 CO 3 , DBU etc. with/without the presence of halide such as NaI, KI, (Bu) 3 NI etc., and in a polar solvent such as DMF, THF, DMSO etc.
  • a base such as K 2 CO 3 , NaH, Cs 2 CO 3 , DBU etc.
  • halide such as NaI, KI, (Bu) 3 NI etc.
  • a polar solvent such as DMF, THF, DMSO etc.
  • Leaving groups include but are not limited to, e.g., halogen, triflate, tosylate , mesylate etc. (See Kasibhatla, WO 03/037860)
  • Compounds of Formula I, wherein R 1 is —OH can be converted to halides using standard conditions POCl 3 , POBr 3 etc. with/without the presence of base such as Et 3 N, N,N-diethylaniline, (i-Pr) 2 NEt etc. in polar solvents such as CH 3 CN, CH 2 Cl 2 etc.
  • R 1 is —OR 11 , —SR 11 , or —NHR 8 , where R 11 is alkyl, R 8 is hydrogen, lower alkyl, lower aryl, or —C(O)R 9 , where R 9 is lower alkyl, lower aryl, lower heteroaryl, —NR 10 R 10 , or —OR 11 , where R 10 is independently hydrogen or lower alkyl, can be prepared from compounds of Formula I wherein R 1 is halogen by reacting with HOR 11 , HSR 11 or NH 2 R 8 in presence of a base such as K 2 CO 3 or NaH and a polar solvent such as DMF or THF.
  • Compounds of Formula I where R 8 is —C(O)R 9 can be prepared when R 1 is hydroxyl by simple acylation.
  • Compounds of Formula I where R 1 is alkyl can be prepared from compounds of Formula I where R 1 is halogen and trialkyl aluminum or dialkyl zinc. See Holy, J. Med. Chem. 1999, 42, 2064.
  • R 5 especially when it is aryl or heteroaryl can be further modified as needed, for example by halogenation, nitration, palladium coupling of halogen, Friedel-Crafts alkylation/acylation, etc. or these modifications can also be done before alkylation, see Jerry March, Advanced Organic Chemistry .
  • the heteroaromatic rings can also be oxidized to their corresponding N-oxides using various oxidizing agents such as H 2 O 2 , O 3 , MCPBA etc. in polar solvents such as CH 2 Cl 2 , CHCl 3 , CF 3 COOH etc. See Jerry March, Advanced Organic Chemistry, 4th edition, Chapter 19.
  • Compounds of Formula I where R 3 is halogen can be prepared from Formulae 1 or 2 using halogenating agents such as Br 2 , NBS, NCS, NIS etc. in polar solvents such as DMF, water, or suitable buffer solution. See Herdewijn, J. Med Chem. 1995, 38, 3838.
  • compounds of Formula 2 where R 8 is iodo can also be made using procedures known in the literature, e.g., Burger, J. Org. Chem. 2000, 65, 7825. These can be further modified as needed; for example, where R 3 is —N 3 , or —CN by reacting with an azide such as NaN 3 , LiN 3 etc.
  • compounds of Formula I can also be synthesized from Formula 7, 2-amino-4, 6-dichloro pyrimidine (see Scheme 4).
  • Reduction of the azo compound with zinc in acetic acid to give compounds of Formula 8, where R 16 is —NH 2 , see, Meier, U.S. Pat. No. 5,204,353, 1993.
  • compounds of Formula I can also be prepared from the substituted imidazoles as shown in Scheme 6. Accordingly, compounds of Formula 4,wherein R 14 is NH 2 , R 13 is C(O)NH 2 and R 15 is H, can be alkylated in the presence of a base such as KOH, NaOH, K 2 CO 3 , NaH, Cs 2 CO 3 , DBU etc. with/without the presence of halide such as NaI, KI, (Bu) 3 NI etc., and in a polar solvent such as DMF, THF, DMSO etc. using electrophiles such as L 1 —R 4 —R 5 where L 1 is a leaving group.
  • a base such as KOH, NaOH, K 2 CO 3 , NaH, Cs 2 CO 3 , DBU etc.
  • halide such as NaI, KI, (Bu) 3 NI etc.
  • a polar solvent such as DMF, THF, DMSO etc.
  • electrophiles such as L 1 —R 4
  • Leaving groups include but are not limited to, e.g., halogen, triflate, tosylate, mesylate etc. to give compounds of Formula 10.
  • the ring closure can be achieved using many methods reported in the literature, Alhede, J. Org. Chem., 1991, 2139 and references cited therein to give guanines of Formula I, wherein R 1 is OH.
  • These compounds can be converted to the compounds of Formula I, wherein R 1 is Cl using POCl 3 as described earlier.
  • these steps can be reversed as shown in Scheme 6 via Formula 11.
  • the electrophiles can be prepared from the substituted benzene derivatives using various methods reported in the literature, see Jerry March, Advanced Organic Chemistry, 4 th edition; Larock, Comprehensive Organic Transformations, 1989, VCH, New York.
  • the L 1 is —Br can be prepared by reduction followed by halogenation of the benzoic acid or aldehyde derivatives.
  • These benzyl derivatives can also be prepared by benzylic oxidation or benzylic halogenation. Further modification of the benzyl ring can be done before or after the purine alkylation step, for example the halogenation was done in both ways.
  • the corresponding amines where L 1 is —NH 2 can be prepared from the compounds where L 1 is leaving group such as chloride, bromide, tosylate, mesylate etc. using ammonia.
  • the present invention is directed to the clinical use of the heterocyclics, in particular, 2-aminopurine and related analogs of Formulae A, I and II, and their polymorphs, solvates, esters, tautomers, enantiomers, diastereomers, pharmaceutically acceptable salts and prodrugs thereof, for use in treatment or prevention of diseases that are HSP90-dependent.
  • a disorder such as inflammatory diseases, infections, autoimmune disorders, stroke, ischemia, cardiac disorders, neurological disorders, fibrogenetic disorders, proliferative disorders, tumors, leukemias, neoplasms, cancers, carcinomas, metabolic diseases, and malignant disease.
  • the fibrogenetic disorders include but are not limited to scleroderma, polymyositis, systemic lupus, rheumatoid arthritis, liver cirrhosis, keloid formation, interstitial nephritis and pulmonary fibrosis.
  • the present invention features pharmaceutical compositions comprising the compound of Formulae A, I and II, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt thereof, or prodrug thereof, of any of the preceding aspect and embodiments and one or more pharmaceutical excipients.
  • the compounds utilized in the methods of the instant invention may be administered either alone or in combination with pharmaceutically acceptable carriers, excipients or diluents, in a pharmaceutical composition, according to standard pharmaceutical practice.
  • the compounds can be administered orally or parenterally, including the intravenous, intramuscular, intraperitoneal, subcutaneous, rectal and topical routes of administration.
  • the therapeutic or pharmaceutical compositions of the invention can be administered locally to the area in need of treatment.
  • This may be achieved by, for example, but not limited to, local infusion during surgery, topical application, e.g., cream, ointment, injection, catheter, or implant, said implant made, e.g., out of a porous, non-porous, or gelatinous material, including membranes, such as sialastic membranes, or fibers.
  • the administration can also be by direct injection at the site (or former site) of a tumor or neoplastic or pre-neoplastic tissue.
  • the compounds or compositions of the invention can be delivered in a vesicle, e.g., a liposome (see, for example, Langer, Science 1990, 249,1527-1533; Treat et al., Liposomes in the Therapy of Infectious Disease and Cancer , Lopez-Bernstein and Fidler, Ed., Liss, N.Y., pp. 353-365, 1989).
  • a liposome see, for example, Langer, Science 1990, 249,1527-1533; Treat et al., Liposomes in the Therapy of Infectious Disease and Cancer , Lopez-Bernstein and Fidler, Ed., Liss, N.Y., pp. 353-365, 1989).
  • the compounds and pharmaceutical compositions used in the methods of the present invention can also be delivered in a controlled release system.
  • a pump may be used (see, Sefton, 1987, CRC Crit. Ref. Biomed. Eng. 14:201; Buchwald et al. Surgery, 1980 88, 507; Saudek et al. N. Engl. J. Med. 1989, 321, (574).
  • a controlled release system can be placed in proximity of the therapeutic target. (See, Goodson, Medical Applications of Controlled Release, 1984, Vol. 2, pp. 115-138).
  • compositions used in the methods of the instant invention can also contain the active ingredient in a form suitable for oral use, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs.
  • Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions, and such compositions may contain one or more agents selected from the group consisting of sweetening agents, flavoring agents, coloring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations. Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets.
  • excipients may be, for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, such as microcrystalline cellulose, sodium crosscarmellose, corn starch, or alginic acid; binding agents, for example starch, gelatin, polyvinyl-pyrrolidone or acacia, and lubricating agents, for example, magnesium stearate, stearic acid or talc.
  • the tablets may be un-coated or coated by known techniques to mask the taste of the drug or delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • a water soluble taste masking material such as hydroxypropylmethyl-cellulose or hydroxypropylcellulose, or a time delay material such as ethyl cellulose, or cellulose acetate butyrate may be employed as appropriate.
  • Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water soluble carrier such as polyethyleneglycol or an oil medium, for example peanut oil, liquid paraffin, or olive oil.
  • an inert solid diluent for example, calcium carbonate, calcium phosphate or kaolin
  • water soluble carrier such as polyethyleneglycol or an oil medium, for example peanut oil, liquid paraffin, or olive oil.
  • Aqueous suspensions contain the active material in admixture with excipients suitable for the manufacture of aqueous suspensions.
  • excipients are suspending agents, for example sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethyl-cellulose, sodium alginate, polyvinyl-pyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents may be a naturally-occurring phosphatide, for example lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethylene-oxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example polyethylene sorbitan mono
  • the aqueous suspensions may also contain one or more preservatives, for example ethyl, or n-propyl p-hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents, such as sucrose, saccharin or aspartame.
  • preservatives for example ethyl, or n-propyl p-hydroxybenzoate
  • coloring agents for example ethyl, or n-propyl p-hydroxybenzoate
  • flavoring agents such as sucrose, saccharin or aspartame.
  • sweetening agents such as sucrose, saccharin or aspartame.
  • Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in mineral oil such as liquid paraffin.
  • the oily suspensions may contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol.
  • Sweetening agents such as those set forth above, and flavoring agents may be added to provide a palatable oral preparation.
  • These compositions may be preserved by the addition of an anti-oxidant such as butylated hydroxyanisol or alpha-tocopherol.
  • Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives.
  • Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients, for example sweetening, flavoring and coloring agents, may also be present. These compositions may be preserved by the addition of an anti-oxidant such as ascorbic acid.
  • the compounds and pharmaceutical compositions used in the methods of the instant invention may also be in the form of an oil-in-water emulsions.
  • the oily phase may be a vegetable oil, for example olive oil or arachis oil, or a mineral oil, for example liquid paraffin or mixtures of these.
  • Suitable emulsifying agents may be naturally-occurring phosphatides, for example soy bean lecithin, and esters or partial esters derived from fatty acids and hexitol anhydrides, for example sorbitan monooleate, and condensation products of the said partial esters with ethylene oxide, for example polyoxyethylene sorbitan monooleate.
  • the emulsions may also contain sweetening agents, flavoring agents, preservatives and antioxidants.
  • Syrups and elixirs may be formulated with sweetening agents, for example glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative, flavoring and coloring agents and antioxidant.
  • sweetening agents for example glycerol, propylene glycol, sorbitol or sucrose.
  • Such formulations may also contain a demulcent, a preservative, flavoring and coloring agents and antioxidant.
  • compositions may be in the form of a sterile injectable aqueous solution.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • the sterile injectable preparation may also be a sterile injectable oil-in-water microemulsion where the active ingredient is dissolved in the oily phase.
  • the active ingredient may be first dissolved in a mixture of soybean oil and lecithin. The oil solution then introduced into a water and glycerol mixture and processed to form a microemulsion.
  • the injectable solutions or microemulsions may be introduced into a patient's blood-stream by local bolus injection.
  • a continuous intravenous delivery device may be utilized.
  • An example of such a device is the Deltec CADD-PLUSTM model 5400 intravenous pump.
  • the pharmaceutical compositions may be in the form of a sterile injectable aqueous or oleagenous suspension for intramuscular and subcutaneous administration.
  • This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example as a solution in 1,3-butane diol.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid find use in the preparation of injectables.
  • compositions can be prepared by mixing the inhibitors with a suitable non-irritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • suitable non-irritating excipient include cocoa butter, glycerinated gelatin, hydrogenated vegetable oils, mixtures of polyethylene glycols of various molecular weights and fatty acid esters of polyethylene glycol.
  • creams, ointments, jellies, solutions or suspensions, etc., containing a compound or composition of the invention can be used.
  • topical application can include mouth washes and gargles.
  • the compounds used in the methods of the present invention can be administered in intranasal form via topical use of suitable intranasal vehicles and delivery devices, or via transdermal routes, using those forms of transdermal skin patches well known to those of ordinary skill in the art.
  • the dosage administration will, of course, be continuous rather than intermittent throughout the dosage regimen.
  • the methods, compounds and compositions of the instant invention may also be used in conjunction with other well known therapeutic agents that are selected for their particular usefulness against the condition that is being treated.
  • the instant compounds may be useful in combination with known anti-cancer and cytotoxic agents.
  • the instant methods and compounds may also be useful in combination with other inhibitors of parts of the signaling pathway that links cell surface growth factor receptors to nuclear signals initiating cellular proliferation.
  • the methods of the present invention may also be useful with other agents that inhibit angiogenesis and thereby inhibit the growth and invasiveness of tumor cells, including, but not limited to VEGF receptor inhibitors, including ribozymes and antisense targeted to VEGF receptors, angiostatin and endostatin.
  • VEGF receptor inhibitors including ribozymes and antisense targeted to VEGF receptors, angiostatin and endostatin.
  • antineoplastic agents examples include, in general, and as appropriate, alkylating agents, anti-metabolites, epidophyllotoxins, an antineoplastic enzyme, a topoisomerase inhibitor, procarbazine, mitoxantrone, platinum coordination complexes, biological response modifiers and growth inhibitors, hormonal/anti-hormonal therapeutic agents and haematopoietic growth factors.
  • alkylating agents include, in general, and as appropriate, alkylating agents, anti-metabolites, epidophyllotoxins, an antineoplastic enzyme, a topoisomerase inhibitor, procarbazine, mitoxantrone, platinum coordination complexes, biological response modifiers and growth inhibitors, hormonal/anti-hormonal therapeutic agents and haematopoietic growth factors.
  • alkylating agents examples include, in general, and as appropriate, alkylating agents, anti-metabolites, epidophyllotoxins, an antineoplastic enzyme, a topoisomerase inhibitor, procarbazine
  • Particularly useful members of those classes include, for example, carminomycin, daunorubicin, aminopterin, methotrexate, methopterin, dichloromethotrexate, mitomycin C, porfiromycin, 5-fluorouracil, 6-mercaptopurine, gemcitabine, cytosine arabinoside, podophyllotoxin or podo-phyllotoxin derivatives such as etoposide, etoposide phosphate or teniposide, melphalan, vinblastine, vincristine, leurosidine, vindesine, leurosine, paclitaxel and the like.
  • antineoplastic agents include estramustine, carboplatin, cyclophosphamide, bleomycin, gemcitibine, ifosamide, melphalan, hexamethyl melamine, thiotepa, cytarabin, idatrexate, trimetrexate, dacarbazine, L-asparaginase, canptothecin, CPT-11, topotecan, ara-C, bicalutamide, flutamide, leuprolide, pyridobenzoindole derivatives, interferons and interleukins.
  • the daily dosage will normally be determined by the prescribing physician with the dosage generally varying according to the age, weight, and response of the individual patient, as well as the severity of the patient's symptoms.
  • a suitable amount of compound is administered to a mammal undergoing treatment for cancer, for example, breast cancer.
  • Administration typically occurs in an anount of between about 0.01 mg/kg of body weight to about 100 mg/kg of body weight per day (administered in single or divided doses), more preferably at least about 0.1 mg/kg of body weight per day.
  • a particular therapeutic dosage can include, e.g., from about 0.01 mg to about 1000 mg of compound, and preferably includes, e.g., from about 1 mg to about 1000 mg.
  • the quantity of active compound in a unit dose of preparation may be varied or adjusted from about 0.1 mg to 1000 mg, preferably from about 1 mg to 300 mg, more preferably 10 mg to 200 mg, according to the particular application.
  • the amount administered will vary depending on the particular IC 50 value of the compound used and the judgment of the attending clinician taking into consideration factors such as health, weight, and age. In combinational applications in which the compound is not the sole active ingredient, it may be possible to administer lesser amounts of compound and still have therapeutic or prophylactic effect.
  • the pharmaceutical preparation is in unit dosage form.
  • the preparation is subdivided into unit doses containing appropriate quantities of the active component, e.g., an effective amount to achieve the desired purpose.
  • the actual dosage employed may be varied depending upon the requirements of the patient and the severity of the condition being treated. Determination of the proper dosage for a particular situation is within the skill of 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 amounts 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 amount and frequency of administration of the compounds and compositions of the present invention used in the methods of the present invention, and if applicable other chemotherapeutic agents and/or radiation therapy, will be regulated according to the judgment of the attending clinician (physician) considering such factors as age, condition and size of the patient as well as severity of the disease being treated.
  • the chemotherapeutic agent and/or radiation therapy can be administered according to therapeutic protocols well known in the art. It will be apparent to those skilled in the art that the administration of the chemotherapeutic agent and/or radiation therapy can be varied depending on the disease being treated and the known effects of the chemotherapeutic agent and/or radiation therapy on that disease. Also, in accordance with the knowledge of the skilled clinician, the therapeutic protocols (e.g., dosage amounts and times of administration) can be varied in view of the observed effects of the administered therapeutic agents (i.e., antineoplastic agent or radiation) on the patient, and in view of the observed responses of the disease to the administered therapeutic agents.
  • the administered therapeutic agents i.e., antineoplastic agent or radiation
  • the compounds of the invention need not be administered in the same pharmaceutical composition as a chemotherapeutic agent, and may, because of different physical and chemical characteristics, be administered by a different route.
  • the compounds/compositions may be administered orally to generate and maintain good blood levels thereof, while the chemotherapeutic agent may be administered intravenously.
  • the determination of the mode of administration and the advisability of administration, where possible, in the same pharmaceutical composition, is well within the knowledge of the skilled clinician.
  • the initial administration can be made according to established protocols known in the art, and then, based upon the observed effects, the dosage, modes of administration and times of administration can be modified by the skilled clinician.
  • compositions of the invention may be administered concurrently (e.g., simultaneously, essentially simultaneously or within the same treatment protocol) or sequentially, depending upon the nature of the proliferative disease, the condition of the patient, and the actual choice of chemotherapeutic agent and/or radiation to be administered in conjunction (i.e., within a single treatment protocol) with the compound/composition.
  • the compound/composition and the chemotherapeutic agent and/or radiation need not be administered simultaneously or essentially simultaneously, and the initial order of administration of the compound/composition, and the chemotherapeutic agent and/or radiation, may not be important.
  • the compounds/compositions of the invention may be administered first followed by the administration of the chemotherapeutic agent and/or radiation; or the chemotherapeutic agent and/or radiation may be administered first followed by the administration of the compounds/compositions of the invention.
  • This alternate administration may be repeated during a single treatment protocol.
  • the determination of the order of administration, and the number of repetitions of administration of each therapeutic agent during a treatment protocol is well within the knowledge of the skilled physician after evaluation of the disease being treated and the condition of the patient.
  • the chemotherapeutic agent and/or radiation may be administered first, especially if it is a cytotoxic agent, and then the treatment continued with the administration of the compounds/compositions of the invention followed, where determined advantageous, by the administration of the chemotherapeutic agent and/or radiation, and so on until the treatment protocol is complete.
  • the practicing physician can modify each protocol for the administration of a compound/composition for treatment according to the individual patient's needs, as the treatment proceeds.
  • the attending clinician in judging whether treatment is effective at the dosage administered, will consider the general well-being of the patient as well as more definite signs such as relief of disease-related symptoms, inhibition of tumor growth, actual shrinkage of the tumor, or inhibition of metastasis. Size of the tumor can be measured by standard methods such as radiological studies, e.g., CAT or MRI scan, and successive measurements can be used to judge whether or not growth of the tumor has been retarded or even reversed. Relief of disease-related symptoms such as pain, and improvement in overall condition can also be used to help judge effectiveness of treatment.
  • HSP90 competitive binding assays and functional assays can be performed as known in the art substituting in the compounds of the invention. Chiosis et al. Chemistry & Biology 2001, 8, 289-299, describe some of the known ways in which this can be done.
  • competition binding assays using, e.g., geldanamycin or 17-AAG as a competitive binding inhibitor of HSP90 can be used to determine relative HSP90 affinity of the compounds of the invention by immobilizing the compound of interest or other competitive inhibitor on a gel or solid matrix, preincubating HSP90 with the other inhibitor, passing the preincubated mix over the gel or matrix, and then measuring the amount of HSP90 that retains or does not retain on the gel or matrix.
  • Downstream effects can also be evaluated based on the known effect of HSP90 inhibition on function and stability of various steroid receptors and signaling proteins including, e.g., Raf1 and HER2.
  • Compounds of the present invention induce dose-dependent degradation of these molecules, which can be measured using standard techniques. Inhibition of HSP90 also results in up-regulation of HSP90 and related chaperone proteins that can similarly be measured.
  • Antiproliferative activity on various cancer cell lines can also be measured, as can morphological and functional differentiation related to HSP90 inhibition.
  • Indirect techniques include nucleic acid hybridization and amplification using, e.g., polymerase chain reaction (PCR). These techniques are known to the person of skill and are discussed, e.g., in Sambrook, Fritsch & Maniatis Molecular Cloning: A Laboratory Manual, 2nd ed., Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y., 1989; Ausubel, et al. Current Protocols in Molecular Biology , John Wiley & Sons, NY, 1994, and, as specifically applied to the quantification, detection, and relative activity of HER2/Neu in patient samples, e.g., in U.S. Pat. No. Nos. 4,699,877, 4,918,162, 4,968,603, and 5,846,749. A brief discussion of two generic techniques that can be used follows.
  • HER2 expression in breast cancer cells can be determined with the use of an immunohistochemical assay, such as the Dako HercepTM test (Dako Corp., Carpinteria, Calif.).
  • the HercepTM test is an antibody staining assay designed to detect HER2 overexpression in tumor tissue specimens. This particular assay grades HER2 expression into four levels: 0, 1, 2, and 3, with level 3 representing the highest level of HER2 expression.
  • Accurate quantitation can be enhanced by employing an Automated Cellular Imaging System (ACIS) as described, e.g., by Press, M. et al. Modern Pathology 2000, 13, 225A.
  • ACIS Automated Cellular Imaging System
  • Antibodies polyclonal or monoclonal, can be purchased from a variety of commercial suppliers, or may be manufactured using well-known methods, e.g., as described in Harlow et al. Antibodies: A Laboratory Manual, 2nd ed; Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y., 1988.
  • HER2 overexpression can also be determined at the nucleic acid level since there is a reported high correlation between overexpression of the HER2 protein and amplification of the gene that codes for it.
  • One way to test this is by using RT-PCR.
  • the genomic and cDNA sequences for HER2 are known.
  • Specific DNA primers can be generated using standard, well-known techniques, and can then be used to amplify template already present in the cell. An example of this is described in Kurokawa, H. et al. Cancer Res. 2000, 60, 5887-5894.
  • PCR can be standardized such that quantitative differences are observed as between normal and abnormal cells, e.g., cancerous and noncancerous cells.
  • Well known methods employing, e.g., densitometry can be used to quantitate and/or compare nucleic acid levels amplified using PCR.
  • FISH fluorescent in situ hybridization
  • other assays can be used, e.g., Northern and/or Southern blotting.
  • FISH fluorescent in situ hybridization
  • this nucleic acid probe can be conjugated to a fluorescent molecule, e.g., fluorescein and/or rhodamine, that preferably does not interfere with hybridization, and which fluorescence can later be measured following hybridization.
  • Immuno and nucleic acid detection can also be directed against proteins other than HSP90 and HER2, which proteins are nevertheless affected in response to HSP90 inhibition.
  • the final compounds were usually purified by preparative TLC (silica gel 60 ⁇ , Whatman Partisil PK6F) or flash chromatography (silica gel 60 ⁇ , EMD) using EtOAc/hexane or MeOH/CH 2 Cl 2 as eluents. Rf's were measured using sicila gel TLC plates (silica gel 60 ⁇ , EMD).
  • the 6-halo purine derivative was dissolved in acetic acid and a catalytic amount of 5% Pd/C was added, and the mixture stirred under H 2 atmosphere (1 psi) at r.t. for 1 h., evaporation, and purification afforded the dehalogenated derivatives.
  • the 2-amino purine derivative was dissolved in acetic anhydride, treated with a catalytic amount of concentrated sulfuric acid at r.t. for 1 hour. Work-up (EtOAc), evaporation, and purification afforded the 2-acetamido-pyridine.
  • Method 1 The free base (40 mmol) was heated in MeOH (1:6, 300 mL) until it dissolved. A solution of H 3 PO 4 in MeOH (40 mmol) was added dropwise at r.t. The mixture was stirred for 10 min., and the solvent was evaporated to give the pyridinium phosphate as a glassy white solid.
  • the following solvents could also be used: THF, EtOH, or i-PrOH.
  • Method 2 The free base (50 mmol) was heated in i-PrOH (6 L) until it dissolved. The solution was allowed to cool to r.t. and a solution of HCl in i-PrOH (75 mmol) was added slowly dropwise. The hydrochloride crystallized out of solution within a few minutes, and was collected by filtration, washed (acetone) and dried. The sulfate and mesylate salts were also prepared in this manner.
  • the hydrochloride salts can also be made by adding CH 3 COCl to the alcoholic solution of the pyridine or free base derivative.
  • Variant 1 A solution of the aromatic compound in MeOH/THF/acetate buffer (1N in each AcOH and AcONa) was treated with Br 2 (1.3 equiv) at r.t. for 5 min. The excess bromine and solvent were removed on a rotary evaporator. Work-up (CHCl 3 ) and flash chromatography afforded the desired bromobenzene.
  • Variant 2 A solution of the aromatic compound (7 mmol) and N-halosuccinimide (NCS, NBS, or NIS, 1.06 equiv) in acetic acid (40 mL) was heated to 40-90° C. for 0.3-1 h. Evaporation, work-up (EtOAc) and flash chromatography afforded the desired halogenated benzene.
  • Benzoic acid derivatives were reduced to the corresponding benzylic alcohols according to the procedure given by Bhaskar et al. J. Org. Chem. 1991, 56, 5964-5965.
  • the alkylating agent 4-bromo-2-bromomethyl-3,5-dimethyl-pyridine, could itself be prepared by any of the following three methods:
  • Step 3 Acetic acid 4-bromo-3,5-dimethyl-pyridin-2-yl methyl ester
  • Step 4 4-Bromo-3,5-dimethyl-pyridin-2-yl methanol
  • Step 2 4-bromo-2-chloromethyl-3,5-dimethyl pyridine
  • Step 1 4-bromo-2-chloromethyl-3,5-dimethyl pyridine
  • Step 1 4-Chloro-2-chloromethyl-3,5-dimethyl pyridine
  • Step 2 6-chloro-9-(4-chloro-3,5-dimethyl-1-oxy-pyridin-2-yl methyl) -9H-purin-2-ylamine
  • Step 1 Acetic acid 3,5-dimethyl-pyridin-2-yl methyl ester
  • the title compound was obtained by alkylating 6-chloro-9H-purin-2-ylamine with 2-chloromethyl-4-methoxy-3,5-dimethylpyridine (or its HCl salt) according to the general procedure 1.1.
  • the title compound was obtained by alkylation of 6-chloro-9H-purin-2-ylamine with 2-chloro-6-chloromethyl-4-methoxy-3,5-dimethylpyridine according to the general procedure 1.1.
  • Step 6 6-Chloro-9-(3-methoxy-5-methoxymethyl-4-methyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine
  • the title compound was obtained by alkylation of 2-amino-6-chloropurine with 3,4,5-trimethoxybenzyl according to the general procedure 1.1.
  • the title compound was also obtained by treating a solution of 6-chloro-N-4-(3,4,5-trimethoxy-benzyl)-pyrimidine-2,4,5-triamine in triethyl orthoformate with a catalytic amount of conc. HCl at r.t. for 20 min.
  • HPLC Rt 4.906 min.
  • Step 4 Acetic acid 4-(2-amino-6-chloro-purin-9-ylmethyl)-3-bromo-2,6-dimethoxy-phenyl ester
  • the title compound was obtained by O-methylation of 4-(2-amino-6-chloro-purin-9-ylmethyl)-3-bromo-2,6-dimethoxy-phenol (see previous example) according to the general procedure 2.6.
  • the title compound could also be obtained by bromination of 6-chloro-9-(3,4,5-trimethoxy-benzyl)-9H-purin-2-ylamine (see example 48) in acetate buffer according to the general procedure 3.1.
  • the title compound was obtained from (2,4-dimethoxy-3-methyl-phenyl)-methanol according to the general procedure 2.5.
  • the title compound was obtained from (2-chloro-3,4-dimethoxy-phenyl)-methanol according to the general procedure 2.5.
  • Step 1 2-Bromo-1-chloromethyl-3,5-dimethoxy-benzene and 2,4-dibromo-3-chloromethyl-1,5-dimethoxy-benzene
  • the title compound was obtained from (3,5-dichloro-phenyl)-methanol according to the general procedure 2.5.
  • Step 8 6-Chloro-9-(4-iodo-3, S-dimethyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine
  • Step 5 6-Chloro-9-(3,5-dimethyl-4-methylsulfanyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine
  • Step 7 6-Chloro-9-(3,4,5-trimethyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine
  • the compound was obtained by reacting 2-bromomethyl-3,4,5-trimethyl-pyridine (see example 117) with 6-bromo-9H-purin-2-ylamine in the presence of K 2 CO 3 in DMF for 0.5 h at 50° C. as described in the general procedure 1.1.
  • rHSP90 protein Stressgen, BC, Canada, #SPP-770
  • PBS phosphated buffered saline
  • biotin-GM biotinylated-geldanamycin
  • biotin-GM biotinylated-geldanamycin
  • Relative fluorescence units was measured using a SpectraMax Gemini XS Spectrofluorometer (Molecular Devices, Sunnyvale, Calif.) with an excitation at 485 nm and emission at 580 nm; data was acquired using SOFTmax®PRO software (Molecular Devices Corporation, Sunnyvale, Calif.).
  • the background was defined as the RFU generated from wells that were not coated with HSP90 but were treated with the biotin-GM and streptavidin-PE.
  • MCF7 breast carcinoma cell lysates were prepared by douncing in lysing buffer (20 mM HEPES, pH 7.3, 1 mM EDTA, 5 mM MgCl 2 , 100 mM KCl), and then incubated with or without test compound for 30 mins at 4° C., followed by incubation with biotin-GM linked to BioMagTM streptavidin magnetic beads (Qiagen) for 1 hr at 4° C. The tubes were placed on a magnetic rack, and the unbound supernatant removed. The magnetic beads were washed three times in lysis buffer and boiled for 5 mins at 95° C. in SDS-PAGE sample buffer.
  • the lysate binding ability of selected compounds of the invention based on the above assay is summarized in Table 2.
  • the IC 50 reported is the concentration of test compound needed to achieve 50% inhibition of the biotin-GM binding to rHSP90 in the MCF7 cell lysates.
  • MCF7 breast carcinoma cells were grown in Dulbecco's modified Eagle's medium (DMEM) containing 10% fetal bovine serum (FBS) and 10 mM HEPES, and plated in 24 well plates (50% confluent). Twenty-four hrs later (cells are 65-70% confluent), test compounds were added and incubated overnight for 16 h. For the less potent compounds, the amounts added were 100 ⁇ M, 30 ⁇ M, 10 ⁇ M and 1 ⁇ M, and for more potent compounds, the amounts added were 1 ⁇ M, 0.3 ⁇ M, 0.1 ⁇ M, 0.03 ⁇ M, 0.01 ⁇ M and 0.003 ⁇ M.
  • DMEM Dulbecco's modified Eagle's medium
  • FBS fetal bovine serum
  • HEPES fetal bovine serum
  • the wells were washed with 1 mL phosphate buffered saline (PBS), and 200 ⁇ L trypsin was added to each well. After trypsinization was complete, 50 ⁇ L of FBS was added to each well. Then 200 ⁇ L cells was transferred to 96 well plates. The cells were pipetted up and down to obtain a single cell suspension. The plates were centrifuged at 2,500 rpm for 1 min using a Sorvall Legend RTTM tabletop centrifuge (Kendro Laboratory Products, Asheville, N.C.). The cells were then washed once in PBS containing 0.2% BSA and 0.2% sodium azide (BA buffer).
  • PBS phosphate buffered saline
  • PE conjugated anti HER2/Neu antibody Becton Dickinson, #340552
  • PE conjugated anti-keyhole limpet hemacyanin [KLH] Becton Dickinson, #340761
  • control antibody was added at a dilution of 1:20 and 1:40 respectively (final concentration was 1 ⁇ g/mL) and the cells were pipetted up and down to form a single cell suspension, and incubated for 15 mins. The cells were washed twice with 200 ⁇ L BA buffer, and resuspended in 200 ⁇ L BA buffer, and transferred to FACSCAN tubes with an additional 250 ⁇ L BA buffer.
  • IC 50 is defined as the concentration at which there was 50% degradation of the HER2/Neu protein.
  • MTS assays measures the cytotoxicity of geldanamycin derivatives.
  • MTS (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)2-(4-sulfophenyl)-2H-tetrazolium is a tetrazolium dye that is converted to a formazan product by dehydrogenase enzymes of metabolically active cells (Corey, A. et al. “Use of an aqueous soluble tetrazolium/formazan assay for cell growth assays in culture,” Cancer Commun. 1991, 3, 207-212).
  • % viable cells (Abs at 490 nm treated cells/Abs at 490 nm untreated cells) ⁇ 100

Abstract

2-Aminopurine analogs are described and demonstrated or predicted to have utility as Heat Shock Protein 90 (HSP90) inhibiting agents in the treatment and prevention of various HSP90 mediated disorders, e.g., proliferative disorders. Method of synthesis and use of such compounds are also described and claimed.

Description

    RELATED APPLICATIONS
  • This application relates and claims priority to U.S. Provisional Application Ser. No. 60/504,135, filed Sept. 18, 2003, entitled NOVEL HETEROCYCLIC COMPOUNDS AS HSP90 INHIBITORS and U.S. Provisional Application Ser. No. 60/591,467, filed Jul. 26, 2004, entitled 2-AMINOPURINE ANALOGS HAVING HSP90-INHIBITING ACTIVITY. This application also relates to three other United States Utility Applications, entitled PYRAZOLOPYRIDIMES AND RELATED ANALOGS AS HSP90 INHIBITORS, PYRROLOPYRIMIDINES AND RELATED ANALOGS AS HSP90 INHIBITORS, and TRIAZOPYRIMIDES AND RELATED ANALOGS AS HSP90 INHIBITORS, which will be filed on the same date by the same entity. This application further relates to International Application PCT/US02/35069, filed Oct. 30, 2002, entitled PURINE ANALOGS HAVING HSP90-INHIBITING ACTIVITY. All the. above cited U.S. utility applications, provisional applications and international application are expressly incorporated herein by reference in their entirety.
  • FIELD OF THE INVENTION
  • The invention relates in general to 2-aminopurine analogs and their broad-spectrum utility, e.g., in inhibiting heat shock protein 90 (HSP90) to thereby treat or prevent HSP90-mediated diseases.
  • BACKGROUND
  • HSP90s are ubiquitous chaperone proteins that are involved in folding, activation and assembly of a wide range of proteins, including key proteins involved in signal transduction, cell cycle control and transcriptional regulation. Researchers have reported that HSP90 chaperone proteins are associated with important signaling proteins, such as steroid hormone receptors and protein kinases, including, e.g., Raf-1, EGFR, v-Src family kinases, Cdk4, and ErbB-2 (Buchner J. TIBS 1999,24, 136-141; Stepanova, L. et al. Genes Dev. 1996,10, 1491-502; Dai, K. et al. J. Biol. Chem. 1996, 271, 22030-4). Studies further indicate that certain co-chaperones, e.g., HSP70, p60/Hop/Sti1, Hip, Bag1, HSP40/Hdj2/Hsj1, immunophilins, p23, and p50, may assist HSP90 in its function (see, e.g., Caplan, A. Trends in Cell Biol 1999, 9, 262-68).
  • Ansamycin antibiotics, e.g., herbimycin A (HA), geldanamycin (GM), and 17-allylaminogeldanamycin (17-AAG) are thought to exert their anticancerous effects by tight binding of the N-terminus pocket of HSP90, thereby destabilizing substrates that normally interact with HSP90 (Stebbins, C. et al. Cell 1997, 89, 239-250). This pocket is highly conserved and has weak homology to the ATP-binding site of DNA gyrase (Stebbins, C. et al., supra; Grenert, J. P. et al. J. Biol. Chem. 1997, 272, 23843-50). Further, ATP and ADP have both been shown to bind this pocket with low affinity and to have weak ATPase activity (Proromou, C. et al. Cell 1997, 90, 65-75; Panaretou, B. et al. EMBO J. 1998, 17, 4829-36). In vitro and in vivo studies have demonstrated that occupancy of this N-terminal pocket by ansamycins and other HSP90 inhibitors alters HSP90 function and inhibits protein folding. At high concentrations, ansamycins and other HSP90 inhibitors have been shown to prevent binding of protein substrates to HSP90 (Scheibel, T. H. et al. Proc. Natl. Acad. Sci. USA 1999, 96, 1297-302; Schulte, T. W. et al. J. Biol. Chem. 1995, 270, 24585-8; Whitesell, L., et al. Proc. Natl. Acad. Sci. USA 1994, 91, 8324-8328). Ansamycins have also been demonstrated to inhibit the ATP-dependent release of chaperone-associated protein substrates (Schneider, C. L. et al. Proc. Natl. Acad. Sci., USA 1996, 93, 14536-41; Sepp-Lorenzino et al. J. Biol. Chem. 1995, 270, 16580-16587). In either event, the substrates are degraded by a ubiquitin-dependent process in the proteasome (Schneider, C. L., supra; Sepp-Lorenzino, L., et al. J. Biol. Chem. 1995, 270, 16580-16587; Whitesell, L. et al. Proc. Natl. Acad. Sci. USA 1994, 91, 8324-8328).
  • HSP90 substrate destabilization occurs in tumor and non-transformed cells alike and has been shown to be especially effective on a subset of signaling regulators, e.g., Raf (Schulte, T. W. et al. Biochem. Biophys. Res. Commun. 1997, 239, 655-9; Schulte, T. W., et al. J. Biol. Chem. 1995, 270, 24585-8), nuclear steroid receptors (Segnitz, B.; U. Gehring J. Biol. Chem. 1997, 272, 18694-18701; Smith, D. F. et al. Mol. Cell. Biol. 1995, 15, 6804-12), v-Src (Whitesell, L., et al. Proc. Natl. Acad. Sci. USA 1994, 91, 8324-8328) and certain transmembrane tyrosine kinases (Sepp-Lorenzino, L. et al. J. Biol. Chem. 1995, 270, 16580-16587) such as EGF receptor (EGFR) and HER2/Neu (Hartmann, F., et al. Int. J. Cancer 1997, 70, 221-9; Miller, P. et al. Cancer Res. 1994, 54, 2724-2730; Mimnaugh, E. G., et al. J. Biol. Chem. 1996, 271, 22796-801; Schnur, R. et al. J. Med Chem. 1995, 38, 3806-3812), CDK4, and mutant p53. Erlichman et al. Proc. AACR 2001, 42, abstract 4474. The ansamycin-induced loss of these proteins leads to the selective disruption of certain regulatory pathways and results in growth arrest at specific phases of the cell cycle (Muise-Heimericks, R. C. et al. J. Biol. Chem. 1998, 273, 29864-72), and apoptosis, and/or differentiation of cells so treated (Vasilevskaya, A. et al. Cancer Res., 1999, 59, 3935-40). Ansamycins thus hold great promise for the treatment and/or prevention of many types of cancers and proliferative disorders, and also hold promise as traditional antibiotics. However, their relative insolubility makes them difficult to formulate and administer, and they are not easily synthesized and currently must, at least in part, be generated through fermentation. Further, the dose limiting toxicity of ansamycins is hepatic.
  • In addition to anti-cancer and antitumorgenic activity, HSP90 inhibitors have also been implicated in a wide variety of other utilities, including use as anti-inflammation agents, anti-infectious disease agents, agents for treating autoimmunity, agents for treating stroke, ischemia, multiple sclerosis, cardiac disorders, central nervous system related disorders and agents usefull in promoting nerve regeneration (See, e.g., Rosen et al. WO 02/09696 (PCT/US01/23640); Degranco et al. WO 99/51223 (PCT/US99/07242); Gold, U.S. Pat. No. 6,210,974 B1; DeFranco et al., U.S. Pat. No. 6,174,875. Overlapping somewhat with the above, there are reports in the literature that fibrogenetic disorders including but not limited to scleroderma, polymyositis, systemic lupus, rheumatoid arthritis, liver cirrhosis, keloid formation, interstitial nephritis, and pulmonary fibrosis also may be treatable with HSP90 inhibitors. Strehlow, WO 02/02123 (PCT/US01/20578). Still further HSP90 modulation, modulators and uses thereof are reported in Application Nos. PCT/US03/04283, PCT/US02/35938, PCT/US02/16287, PCT/US02/06518, PCT/US98/09805, PCT/US00/09512, PCT/US01/09512, PCT/US01/23640, PCT/US01/46303, PCT/US01/46304, PCT/US02/06518, PCT/US02/29715, PCT/US02/35069, PCT/US02/35938, PCT/US02/39993, 60/293,246, 60/371,668, 60/335,391, 60/128,593, 60/337,919, 60/340,762, 60/359,484 and 60/331,893.
  • Recently, purine derivatives showing HSP90 inhibitory activity have been reported, e.g., in PCT/US02/35069; PCT/US02/36075. Purine moieties are well accepted bioisosteres for a variety of ATP-dependent molecular targets, see, JP 10025294; U.S. Pat. Nos. 4,748,177; 4,772,606; 6,369,092; WO 00/06573; WO 02/055521; WO 02/055082; WO 02/055083; European Patent 0178178; Eur. J. Med. Chem. 1994, 29(1), 3-9; and J. Het. Chem. 1990, 27(5), 1409. However, compounds having the desired potency, selectivity and pharmaceutical properties required for effective HSP90 inhibition in vivo have not been reported. Therefore, a need remains for additional novel and potent HSP90 inhibitors that meet the demanding biological and pharmaceutical criteria required to proceed towards human clinical trials.
  • SUMMARY OF THE INVENTION
  • The present invention is directed towards heterocyclic compounds, in particular towards 2-aminopures and related compounds that show broad utility, e.g., in inhibiting HSP90 and/or treating and preventing diseases that are HSP90-dependent.
  • In one aspect, the invention comprises the heterocyclic compounds as specified below in Formulae A and I, and compounds that are produced by a process of the invention. Also included in the scope of the present invention are stereoisomeric forms, including the individual enantiomers and diastereomers, racemic mixtures, and diastereomeric mixtures, as well as polymorphs, solvates, esters, tautomers, pharmaceutically acceptable salts and prodrugs of these compounds. Stereoisomers of the compounds of the present invention may be isolated by standard resolution techniques such as, for example, fractional crystallization and chiral column chromatography.
  • In one embodiment, the invention provides compounds of Formula A, or a polymorph, solvate, ester, tautomer, diastereomer, enantiomer, pharmaceutically acceptable salt or prodrug thereof, which show utility by inhibiting HSP90 and treating and/or preventing diseases that are HSP90-dependent.
    Figure US20070185064A1-20070809-C00001

    wherein:
      • X1 and X2 are the same or different and each is nitrogen or —CR6;
      • X3 is nitrogen or —CR3 wherein R3 is hydrogen, OH, a keto tautomer, —OR8, —CN, halogen, lower alkyl, or —C(O)R9;
      • X4 is nitrogen or —CR6 when X3 is nitrogen, and X4 is —CR6R7 when X3 is —CR3;
      • R1 is halogen, —OR8, —SR8, or lower alkyl;
      • R2 is —NR8R10;
      • R4 is —(CH2)n— wherein n=0-3, —C(O), —C(S), —SO2—, or —SO2N—; and
      • R5 is alkyl, aromatic, heteroaromatic, alicyclic, or heterocyclic, each of which is optionally bi- or tri-cyclic, and optionally substituted with H, halogen, lower alkyl, lower alkenyl, lower alkynyl, lower aryl, lower alicyclic, aralkyl, aryloxyalkyl, alkoxyalkyl, perhaloalkyl, perhaloalkyloxy, perhaloacyl, —N3, —SR8, —OR8, —CN, —CO2R9, —NO2, or —NR8R10.
  • In certain embodiments, there are exclusionary provisos with respect to compounds disclosed in JP 10025294; U.S. Pat. Nos. 4,748,177; 4,748,177; 6,369,092; WO 00/06573; WO 02/055521; WO 02/055082; WO 02/055083; Eur. J. Med. Chem. 1994, 29(1), 3-9; and J. Het. Chem. 1990, 27(5), 1409, which disclose compounds with —R4R5 comprising ribose or a derivative thereof, or a sugar or derivative thereof; compounds where —R4R5 is a phosphonate or phosphonic acid, or is substituted with a phosphonate or phosphonic acid; or compounds where R4 is —CH2— or —(CH2)n— that are connected through an oxygen atom to another group.
  • In another embodiment, the invention provides compounds of Formula I, or a polymorph, solvate, ester, tautomer, enantiomer, diastereomer, pharmaceutically acceptable salt or prodrug thereof, which show utility for inhibiting HSP90 and for treating and/or preventing diseases that are HSP90-dependent,
    Figure US20070185064A1-20070809-C00002

    wherein:
      • R1 is halogen or lower alkyl;
      • R2 is —NR8R10;
      • R4 is —CHR12—;
      • R3 is hydrogen, halogen, or —CN;
      • R5 is aryl, heteroaryl, alicyclic, or heterocyclic, wherein
        • the aryl group is substituted with 3 to 5 substituents,
        • the heteroaryl group is substituted with 2 to 5 substituents, wherein when the heteroaryl is substituted with only two substituents, the two substituents must form part of an optionally substituted fused ring,
        • the alicyclic group is substituted with 3 to 5 substituents,
        • the heterocyclic group is substituted with 3 to 5 substituents, and
        • the substituents are selected from the group of halogen, lower alkyl, lower alkenyl, lower alkynyl, lower aryl, lower alicyclic, arylalkyl, aryloxy, aryloxyalkyl, alkoxyalkyl, perhaloalkyl, perhaloalkyloxy, perhaloacyl, —N3, —SR8, —OR8, —CN, —C(O)R9, —NO2, —NR8R10, phosphonate and phosphonic acid;
      • R8 is hydrogen, lower alkyl, lower aryl, or —C(O)R9;
      • R9 is lower alkyl, lower aryl, lower heteroaryl, —NR10R10 or —OR11;
      • R10 is independently hydrogen or lower alkyl;
      • R11 is lower alkyl, lower aryl or lower heteroaryl;
      • R12 is hydrogen or lower alkyl; provided that
      • when R5 is aryl, R5 is not an organo-metallic cyclopentadiene;
      • when R5 is phenyl, the substituents are not 3,5 di-halo;
      • when R5 is alicyclic, the ring system does not contain any tetra-substituted sp3 ring carbons; and
      • when R5 is heterocyclic, the ring system does not contain any tetra-substituted sp3 ring carbons or the ring system is not a tetra-substituted pyrrolidine.
  • In another embodiment, the invention provides compounds, or a polymorph, solvate, ester, tautomer, diastereomer, enantiomer, pharmaceutically acceptable salt or prodrug thereof, which show utility for inhibiting HSP90 and treating and/or preventing diseases that are HSP90-dependent, that are prepared by the process comprising:
      • reacting a compound of Formula Y and a compound of Formula Z, wherein:
      • Y is represented by any of the following formulae:
        Figure US20070185064A1-20070809-C00003
      • Z is L1—R4—R5; wherein:
        • L1 is halogen, NR8R10, triflate, tosylate, or mesylate;
        • R4 is —CHR12—,
        • R5 is aryl, heteroaryl, alicyclic, or heterocyclic, wherein:
          • the aryl group is substituted with 3 to 5 substituents,
          • the heteroaryl group is substituted with 2 to 5 substituents, wherein when the heteroaryl is substituted with only two substituents, the two substituents must form part of an optionally substituted fused ring,
          • the alicyclic group is substituted with 3 to 5 substituents,
          • the heterocyclic group is substituted with 3 to 5 substituents, and
          • the substituents are selected from the group of halogen, lower alkyl, lower alkenyl, lower alkynyl, lower aryl, lower alicyclic, arylalkyl, aryloxy, aryloxyalkyl, alkoxyalkyl, perhaloalkyl, perhaloalkyloxy, perhaloacyl, —N3, —SR8, —OR8, —CN, —C(O)R9, —NO2, —NR8R10, phosphonate and phosphonic acid;
        • R8 is hydrogen, lower alkyl, lower aryl, or —C(O)R9;
        • R9 is lower alkyl, lower aryl, lower heteroaryl, —NR10OR10 or —OR11;
        • R10 is independently hydrogen or lower alkyl;
        • R11 is lower alkyl, lower aryl or lower heteroaryl;
        • R12 is hydrogen or lower alkyl;
        • R21 is halogen, lower alkyl or —OH;
        • R22 is —NR8R10;
        • R23 is hydrogen, halogen, or —CN;
        • R24 is —NH2, —NO2 or —NO;
        • R25 is halogen or —OH;
        • R26 is —C(O)NH2 or C(O)OEt; and
        • R27 is —NH2, —OH or halogen;
          provided that:
      • when R5 is aryl, R5 is not an organo-metallic cyclopentadiene;
      • when R5 is phenyl, the substituents are not 3, 5 di-halo;
      • when R5 is alicyclic, the ring system does not contain any tetra-substituted sp3 ring carbons; or
      • when R5 is heterocyclic, the ring system does not contain any tetra-substituted sp3 ring carbons or the ring system is not a tetra-substituted pyrrolidine.
  • Another aspect of the invention is a method of treating an individual having an HSP90 mediated disorder comprising administering to said individual a pharmaceutical composition comprising a pharmaceutically effective amount of a compound of Formula I:
    Figure US20070185064A1-20070809-C00004

    or a polymorph, solvate, ester, tautomer, enantiomer, diastereomer, pharmaceutically acceptable salt or prodrug thereof, wherein:
      • R1 is halogen, —OR11, —SR11, —NHR8, hydrogen, or lower alkyl;
      • R2 is —NR8R10;
      • R3 is hydrogen, halogen, —N3, or —CN;
      • R4 is —(CHR12)n— where n=0, 1 or 2, —C(O)—, —C(S)—, or —S(O)—;
      • R5 is alkyl, aryl, heteroaryl, alicyclic, or heterocyclic, all optionally substituted with halogen, lower alkyl, lower alkenyl, lower alkynyl, lower aryl, lower alicyclic, arylalkyl, aryloxy, aryloxyalkyl, alkoxyalkyl, perhaloalkyl, perhaloalkyloxy, perhaloacyl, —N3, —SR8, —OR8, —CN, —C(O)R9, —NO2, or —NR8R10;
      • R8 is hydrogen, lower alkyl, lower aryl, or —C(O)R9;
      • R9 is lower alkyl, lower aryl, lower heteroaryl, —NR10R10, or —OR11;
      • R10 is independently hydrogen or lower alkyl;
      • R11 is lower alkyl, lower aryl or lower heteroaryl;
      • R12 is hydrogen or lower alkyl;
        provided that:
      • —R4R5 is not a ribose or derivative thereof, or a sugar or derivative thereof;
      • —R4R5 is not a phosphonate or phosphonic acid, or substituted with phosphonate or phosphonic acid; and
      • when R4 is —(CH2)n— where n=1 or 2, then R4 and R5 are not connected through an ether linkage.
  • In one embodiment, the invention provides a method for treating an individual having a disorder selected from the group of inflammatory diseases, infections, autoimmune disorders, stroke, ischemia, cardiac disorders, neurological disorders, fibrogenetic disorders, proliferative disorders, tumors, leukemias, neoplasms, cancers, carcinomas, metabolic diseases, and malignant disease.
  • In another embodiment, the invention provides a method for treating an individual having a fibrogenetic disorder, such as, for example, scleroderma, polymyositis, systemic lupus, rheumatoid arthritis, liver cirrhosis, keloid formation, interstitial nephritis and pulmonary fibrosis.
  • In another embodiment, the invention provides a combination therapy comprising the administration of a pharmaceutically effective amount of a compound of Formulae A, I or II, or a solvate, tautomer, pharmaceutically acceptable salt, polymorph, or prodrug thereof according to any of the preceding aspects or embodiments, and at least one therapeutic agent selected from the group of cytotoxic agents, anti-angiogenesis agents and anti-neoplastic agents. The anti-neoplastic agent may be selected from the group of alkylating agents, anti-metabolites, epidophyllotoxins antineoplastic enzymes, topoisomerase inhibitors, procarbazines, mitoxantrones, platinum coordination complexes, biological response modifiers and growth inhibitors, honnonal/anti-hormonal therapeutic agents, and haematopoietic growth factors.
  • In another aspect, the present invention is directed to pharmaceutical compositions comprising the compounds of the invention, in particular, the compounds of Formulae A, I and II, and compounds formed by the process of the invention, and their polymorphs, solvates, esters, tautomers, diastereomer, enantiomers, pharmaceutically acceptable salts and prodrugs thereof, and one or more pharmaceutical excipients, for use in treatment or prevention of diseases that are HSP90-dependent.
  • Any of the above described aspects and embodiments of the invention can be combined where practical.
  • The individual compounds, methods and compositions prescribed do not preclude the utilization of other, unspecified steps and agents, and those of ordinary skill in the art will appreciate that additional steps and compounds may also be combined usefully within the spirit of various aspects and embodiments of the invention.
  • Advantages of the invention depend on the specific aspect and embodiment and may include one or more of: ease of synthesis and/or formulation, solubility, and IC50 relative to previously existing compounds in the same or different classes of HSP90 inhibitors.
  • DETAILED DESCRIPTION OF THE INVENTION
  • I. Definitions
  • A “pharmaceutically acceptable derivative or prodrug” means any pharmaceutically acceptable salt, ester, salt of an ester or other derivative of a compound of this invention, which, upon administration to a recipient, is capable of providing, either directly or indirectly, a compound of this invention or a pharmaceutically active metabolite or residue thereof. Particularly favored derivatives or prodrugs are those that increase the bioavailability of the compounds of this invention when such compounds are administered to a patient (e.g., by allowing orally administered compound to be more readily absorbed into blood) or which enhance delivery of the parent compound to a biological compartment (e.g., the brain or lymphatic system).
  • A “pharmaceutically acceptable salt” may be prepared for any compound of the invention having a functionality capable of forming a salt, for example, an acid or base functionality. Pharmaceutically acceptable salts may be derived from organic or inorganic acids and bases. Compounds of the invention that contain one or more basic functional groups, e.g., amino or alkylamino, are capable of forming pharmaceutically acceptable salts with pharmaceutically acceptable organic and inorganic acids. These salts can be prepared in situ during the final isolation and purification of the compounds of the invention, or by separately reacting a purified compound of the invention in its free base form with a suitable organic or inorganic acid, and isolating the salt thus formed. Examples of suitable acid salts include acetate, adipate, alginate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, flimarate, glucoheptanoate, glycerophosphate, glycolate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate, lactate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oxalate, palmoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, salicylate, succinate, sulfate, tartrate, thiocyanate, tosylate and undeconate. Other acids, such as oxalic, while not in themselves pharmaceutically acceptable, may be employed in the preparation of salts useful as intermediates in obtaining the compounds of the invention and their pharmaceutically acceptable acid addition salts. See, e.g., Berge et al. “Pharmaceutical Salts”, J. Pharm. Sci. 1977, 66:1-19.
  • Compounds of the present invention that contain one or more acidic functional groups are capable of forming pharmaceutically acceptable salts with pharmaceutically acceptable bases. The term “pharmaceutically acceptable salts” in these instances refers to the relatively non-toxic, inorganic and organic base addition salts of compounds of the present invention. These salts can likewise be prepared in situ during the final isolation and purification of the compounds, or by separately reacting the purified compound in its free acid form with a suitable base, such as the hydroxide, carbonate or bicarbonate of a pharmaceutically acceptable metal cation, with ammonia, or with a pharmaceutically acceptable organic primary, secondary or tertiary amine. Representative alkali or alkaline earth salts include the lithium, sodium, potassium, calcium, magnesium, and aluminum salts and the like. Illustrative examples of some of the bases that can be used include sodium hydroxide, potassium hydroxide, choline hydroxide, sodium carbonate, N+(C1-4 alkyl)4, and the like. Representative organic amines useful for the formation of base addition salts include ethylamine, diethylamine, ethylenediamine, ethanolamine, diethanolamine, piperazine and the like. This invention also envisions the quaternization of any basic nitrogen-containing groups of the compounds disclosed herein. Water or oil-soluble or dispersible products may be obtained by such quaternization. See, for example, Berge et al., supra.
  • Pharmaceutically acceptable prodrugs of the compounds of this invention include, but are not limited to, esters, carbonates, thiocarbonates, N-acyl derivatives, N-acyloxyalkyl derivatives, quaternary derivatives of tertiary amines, N-Mannich bases, Schiff bases, aminoacid conjugates, phosphate esters, metal salts and sulfonate esters.
  • Suitable positions for derivatization of the compounds of the invention to create “prodrugs” include but are not limited, 2-amino substitution. Those of ordinary skill in the art have the knowledge and means to accomplish this without undue experimentation. Various forms of prodrugs are well known in the art. For examples of such prodrug derivatives, see, e.g.,
  • a) Design of Prodrugs, Bundgaard, A. Ed., Elseview, 1985 and Method in Enzymology, Widder, K. et al., Ed.; Academic, 1985, vol. 42, p. 309-396;
  • b) Bundgaard, H. “Design and Application of Prodrugs” in A Textbook of Drug Design and Development, Krosgaard-Larsen and H. Bundgaard, Ed., 1991, Chapter 5, p. 113-191; and
  • c) Bundgaard, H., Advanced Drug Delivery Review, 1992, 8, 1-38.
  • Each of which is incorporated herein by reference.
  • The term “prodrugs” as employed herein includes, but is not limited to, the following groups and combinations of these groups:
  • Hydroxy Prodrugs:
      • Acyloxyalkyl esters;
      • Alcoxycarbonyloxyalkyl esters;
      • Alkyl esters;
      • Aryl esters;
      • Disulfide containing esters.
  • Amine Prodrugs:
    Figure US20070185064A1-20070809-C00005
  • The term “alkyl,” alone or in combination, refers to an optionally substituted straight-chain, or optionally substituted branched-chain saturated hydrocarbon radical having from one to about thirty carbons, more preferably one to twelve carbons. Examples of alkyl radicals include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, tert-amyl, pentyl, hexyl, heptyl, octyl and the like. The term “cycloalkyl” embraces cyclic alkyl radicals which include monocyclic, bicyclic, tricyclic, and higher multicyclic alkyl radicals wherein each cyclic moiety has from three to about eight carbon atoms. Examples of cycloalkyl radicals include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and the like. A “lower alkyl” is a shorter alkyl, e.g., one containing from one to about six carbon atoms.
  • The term “alkenyl,” alone or in combination, refers to an optionally substituted straight-chain, or optionally substituted branched-chain hydrocarbon radical having one or more carbon-carbon double-bonds and having from two to about thirty carbon atoms, more preferably two to about eighteen carbons. Examples of alkenyl radicals include ethenyl, propenyl, butenyl, 1,3-butadienyl and the like. The term “cycloalkenyl” refers to cyclic alkenyl radicals which include monocyclic, bicyclic, tricyclic, and higher multicyclic alkenyl radicals wherein each cyclic moiety has from three to about eight carbon atoms. A “lower alkenyl” refers to an alkenyl having from two to about six carbons.
  • The term “alkynyl,” alone or in combination, refers to an optionally substituted straight-chain or optionally substituted branched-chain hydrocarbon radical having one or more carbon-carbon triple-bonds and having from two to about thirty carbon atoms, more preferably from two to about twelve carbon atoms, from two to about six carbon atoms as well as those having from two to about four carbon atoms. Examples of alkynyl radicals include ethynyl, 2-propynyl, 2-butynyl, 1,3-butadiynyl and the like. The term “cycloalkynyl” refers to cyclic alkynyl radicals which include monocyclic, bicyclic, tricyclic, and higher multicyclic alkynyl radicals wherein each cyclic moiety has from three to about eight carbon atoms. A “lower alkynyl” refers to an alkynyl having from two to about six carbons.
  • The terms “heteroalkyl, heteroalkenyl and heteroalkynyl” include optionally substituted alkyl, alkenyl and alkynyl structures, as described above, and which have one or more skeletal chain atoms selected from an atom other than carbon, e.g., oxygen, nitrogen, sulfur, phosphorous or combinations thereof.
  • The term “carbon chain” embraces any alkyl, alkenyl, alkynyl, or heteroalkyl, heteroalkenyl, or heteroalkynyl group, which are linear, cyclic, or any combination thereof. If the chain is part of a linker and that linker comprises one or more rings as part of the core backbone, for purposes of calculating chain length, the “chain” only includes those carbon atoms that compose the bottom or top of a given ring and not both, and where the top and bottom of the ring(s) are not equivalent in length, the shorter distance shall be used in determining the chain length. If the chain contains heteroatoms as part of the backbone, those atoms are not calculated as part of the carbon chain length.
  • The term “membered ring” can embrace any cyclic structure, including aromatic, heteroaromatic, alicyclic, heterocyclic and polycyclic fused ring systems as described below. The term “membered” is meant to denote the number of skeletal atoms that constitute the ring. Thus, for example, pyridine, pyran, and pyrimidine are six-membered rings and pyrrole, tetrahydrofuran, and thiophene are five-membered rings.
  • The term “aryl,” alone or in combination, refers to an optionally substituted aromatic hydrocarbon radical of six to about twenty ring atoms, and includes mono-aromatic rings and fused aromatic ring. A fused aromatic ring radical contains from two to four fused rings where the ring of attachment is an aromatic ring, and the other individual rings within the fused ring may be aromatic, heteroaromatic, alicyclic or heterocyclic. Further, the term aryl includes mono-aromatic ring and fused aromatic rings containing from six to about twelve carbon atoms, as well as those containing from six to about ten carbon atoms. Examples of aryl groups include, without limitation, phenyl, naphthyl, anthryl, chrysenyl, and benzopyrenyl ring systems. The term “lower aryl” refers to an aryl having six to about ten skeletal ring carbons, e.g., phenyl and naphthyl ring systems.
  • The term “heteroaryl” refers to optionally substituted aromatic radicals containing from about five to about twenty skeletal ring atoms and where one or more of the ring atoms is a heteroatom such as, for example, oxygen, nitrogen, sulfur, selenium and phosphorus. The term heteroaryl includes optionally substituted mono-heteroaryl radicals and fused heteroaryl radicals having at least one heteroatom (e.g., quinoline, benzothiazole). A fused heteroaryl radical may contain from two to four fused rings and where the ring of attachment is a heteroaromatic ring, the other individual rings within the fused ring system may be aromatic, heteroaromatic, alicyclic or heterocyclic. The term heteroaryl also includes mono-heteroaryls or fused heteroaryls having from five to about twelve skeletal ring atoms, as well as those having from five to about ten skeletal ring atoms. Examples of heteroaryls include, without limitation, furanyl, benzofuranyl, chromenyl, pyridyl, pyrrolyl, indolyl, quinolinyl, pyridyl-N-oxide, pyrimidyl, pyrazinyl, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, benzothiozole, benzimidazole, benzoxazoles, benzothiadiazole, benzoxadiazole, benzotriazole, quinolines, isoquinolines, indoles, purinyl, indolizinyl, thienyl and the like and their oxides. The term “lower heteroaryl” refers to a heteroaryl having five to about ten skeletal ring atoms, e.g., pyridyl, thienyl, pyrimidyl, pyrazinyl, pyrrolyl, or furanyl.
  • The term “alicyclic” alone or in combination, refers to an optionally substituted saturated or unsaturated nonaromatic hydrocarbon ring system containing from three to about twenty ring atoms. The term alicyclic includes mono-alicyclic and fused alicyclic radicals. A fused alicyclic may contain from two to four fused rings where the ring of attachment is an alicyclic ring, and the other individual rings within the fused-alicyclic radical may be aromatic, heteroaromatic, alicyclic and heterocyclic. The term alicyclic also includes mono-alicyclic and fused alicyclic radicals containing from three to about twelve carbon atoms, as well as those containing from three to about ten carbon atoms. Examples of alicyclics include, without limitation, cyclopropyl, cyclopropenyl, cyclobutyl, cyclopentyl, cyclodecyl, cyclododecyl, cyclopentadienyl, indanyl, and cyclooctatetraenyl ring systems. The term “lower alicyclic” refers to an alicyclic having three to about ten skeletal ring carbons, e.g., cyclopropyl, cyclopropenyl, cyclobutyl, cyclopentyl, decalinyl, and cyclohexyl.
  • The term “heterocyclic” refers to optionally substituted saturated or unsaturated nonaromatic ring radicals containing from five to about twenty ring atoms where one or more of the ring atoms are heteroatoms such as, for example, oxygen, nitrogen, sulfur, and phosphorus. The term alicyclic includes mono-heterocyclic and fused heterocyclic ring radicals. A fused heterocyclic radical may contain from two to four fused rings where the attaching ring is a heterocyclic, and the other individual rings within the fused heterocyclic radical may be aromatic, heteroaromatic, alicyclic or heterocyclic. The term heterocyclic also includes mono-heterocyclic and fused alicyclic radicals having from five to about twelve skeletal ring atoms, as well as those having from five to about ten skeletal ring atoms. Example of heterocyclics include without limitation, tetrahydrofuranyl, benzodiazepinyl, tetrahydroindazolyl, dihyroquinolinyl, and the like. The term “lower heterocyclic” refers to a heterocyclic ring system having five to about ten skeletal ring atoms, e.g., dihydropyranyl, pyrrolidinyl, indolyl, piperidinyl, piperazinyl, and the like.
  • The term “alkylaryl,” alone or in combination, refers to an aryl radical as defined above in which one H atom is replaced by an alkyl radical as defined above, such as, for example, tolyl, xylyl and the like.
  • The term “arylalkyl,” or “aralkyl,” alone or in combination, refers to an alkyl radical as defined above in which one H atom is replaced by an aryl radical as defined above, such as, for example, benzyl, 2-phenylethyl and the like.
  • The term “heteroarylalkyl” refers to an alkyl radical as defined above in which one H atom is replaced by a heteroaryl radical as defined above, each of which may be optionally substituted.
  • The term “alkoxy,” alone or in combination, refers to an alkyl ether radical, alkyl-O—, wherein the term alkyl is defined as above. Examples of alkoxy radicals include methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, iso-butoxy, sec-butoxy, tert-butoxy and the like.
  • The term “aryloxy,” alone or in combination, refers to an aryl ether radical wherein the term aryl is defined as above. Examples of aryloxy radicals include phenoxy, benzyloxy and the like.
  • The term “alkylthio,” alone or in combination, refers to an alkyl thio radical, alkyl-S—, wherein the term alkyl is as defined above.
  • The term “arylthio,” alone or in combination, refers to an aryl thio radical, aryl-S—, wherein the term aryl is as defined above.
  • The term “heteroarylthio” refers to the group heteroaryl-S—, wherein the term heteroaryl is as defined above.
  • The term “acyl” refers to a radical —C(O)R where R includes alkyl, alkenyl, alkynyl, aryl, heteroaryl, alicyclic, heterocyclic, arylalkyl or heteroarylalkyl wherein the alkyl, alkenyl, alkynyl, aryl, heteroaryl, alicyclic, heterocyclic, arylalkyl or heteroaryl alkyl groups may be optionally substituted.
  • The term “acyloxy” refers to the ester group —OC(O)R, where R is H, alkyl, alkenyl, alkynyl, aryl, heteroaryl, alicyclic, heterocyclic, arylalkyl, or heteroarylalkyl wherein the alkyl, alkenyl, alkynyl, aryl, heteroaryl, alicyclic, heterocyclic, arylalkyl or heteroarylalkyl may be optionally substituted.
  • The term “carboxy esters” refers to —C(O)OR where R is alkyl, aryl or arylalkyl, wherein the alkyl, aryl and arylalkyl groups may be optionally substituted.
  • The term “carboxamido” refers to
    Figure US20070185064A1-20070809-C00006
  • where each of R and R′ are independently selected from the group consisting of H, alkyl, aryl, heteroaryl, alicyclic, heterocyclic, arylalkyl and heteroarylalkyl, wherein the alkyl, aryl, heteroaryl, alicyclic, heterocyclic, or arylalkyl groups may be optionally substituted.
  • The term “oxo” refers to ═O.
  • The term “halogen” includes F, Cl, Br and I.
  • The terms “haloalkyl, haloalkenyl, haloalkynyl and haloalkoxy” include alkyl, alkenyl, alkynyl and alkoxy structures, as described above, that are substituted with one or more fluorines, chlorines, bromines or iodines, or with combinations thereof.
  • The terms “perhaloalkyl, perhaloalkyloxy and perhaloacyl” refer to alkyl, alkyloxy and acyl radicals as described above, that all the H atoms are substituted with fluorines, chlorines, bromines or iodines, or combinations thereof.
  • The terms “cycloalkyl, arylalkyl, aryl, heteroaryl, alicyclic, heterocyclic, alkyl, alkynyl, alkenyl, haloalkyl, and heteroalkyl” include optionally substituted cycloalkyl, arylalkyl, aryl, heteroaryl, alicyclic, heterocyclic, alkyl alkynyl, alkenyl, haloalkyl and heteroalkyl groups.
  • The terms “alkylamino”, refers to the group —NHR where R is independently selected from alkyl.
  • The terms “dialkylamino”, refers to the group —NRR′ where R and R′ are alkyls.
  • The term “sulfide” refers to a sulfur atom covalently linked to two atoms; the formal oxidation state of said sulfur is (II). The term “thioether” may be used interchangeably with the term “sulfide.”
  • The term “sulfoxide” refers to a sulfur atom covalently linked to three atoms, at least one of which is an oxygen atom; the formal oxidation state of said sulfur atom is (IV).
  • The term “sulfone” refers to a sulfur atom covalently linked to four atoms, at least two of which are oxygen atoms; the formal oxidation state of said sulfur atom is (VI).
  • The terms “optional” or “optionally” mean that the subsequently described event or circumstance may but need not occur, and that the description includes instances where the event or circumstance occurs and instances in which it does not. For example, “aryl optionally mono- or di-substituted with an alkyl” means that the alkyl may but need not be present, or either one alkyl or two may be present, and the description includes situations where the aryl is substituted with one or two alkyls and situations where the aryl is not substituted with an alkyl.
  • “Optionally substituted” groups may be substituted or unsubstituted. The substituents of an “optionally substituted” group may include, without limitation, one or more substituents independently selected from the following groups or designated subsets thereof: lower alkyl, lower alkenyl, lower alkynyl, lower aryl, heteroaryl, alicyclic, heterocyclic, arylalkyl, heteroarylalkyl, lower alkoxy, lower aryloxy, amino, alkylamino, dialkylamino, diarylalkylamino, alkylthio, arylthio, heteroarylthio, oxo, oxa, carbonyl (—C(O)), carboxyesters (—C(O)OR), carboxamido (—C(O)NH2), carboxy, acyloxy, —H, halo, —CN, —NO2, —N3, —SH, —OH, —C(O)CH3, perhaloalkyl, perhaloalkoxy, perhaloacyl, guanidine, pyridinyl, thiophene, furanyl, indole, indazole, esters, amides, phosphonates, phosphonic acid, phosphates, phosphoramides, sulfonates, sulfones, sulfates, sulphonamides, carbamates, ureas, thioureas and thioamides, thioalkyls. An optionally substituted group may be unsubstituted (e.g., —CH2CH3), fully substituted (e.g., —CF2CF3), monosubstituted (e.g., —CH2CH2F) or substituted at a level anywhere in-between fully substituted and monosubstituted (e.g., —CH2CF3).
  • The term “pyridine-1-oxy” also means “pyridine—N-oxy.”
  • Some of the compounds of the present invention may contain one or more chiral centers and therefore may exist in enantiomeric and diastereomeric forms. The scope of the present invention is intended to cover all isomers per se, as well as mixtures of cis and trans isomers, mixtures of diastereomers and racemic mixtures of enantiomers (optical isomers) as well. Further, it is possible using well known techniques to separate the various forms, and some embodiments of the invention may feature purified or enriched species of a given enantiomer or diastereomer.
  • A “pharmacological composition” refers to a mixture of one or more of the compounds described herein, or pharmaceutically acceptable salts thereof, with other chemical components, such as pharmaceutically acceptable carriers and/or excipients. The purpose of a pharmacological composition is to facilitate administration of a compound to an organism.
  • The phrase “pharmaceutically acceptable carrier” as used herein means a pharmaceutically-acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting the subject agent from one organ, or portion of the body, to another organ, or portion of the body. Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient. Some examples of materials which can serve as pharmaceutically-acceptable carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydroxide; (15) alginic acid; (16) pyrogen-free water; (17) isotonic saline; (18) Ringer's solution; (19) ethyl alcohol; (20) phosphate buffer solutions; and (21) other non-toxic compatible substances employed in pharmaceutical formulations. A physiologically acceptable carrier should not cause significant irritation to an organism and does not abrogate the biological activity and properties of the administered compound.
  • An “excipient” refers to an inert substance added to a pharmacological composition to further facilitate administration of a compound. Examples of excipients include but are not limited to calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils and polyethylene glycols.
  • A “pharmaceutically effective amount” means an amount which is capable of providing a therapeutic and/or prophylactic effect. The specific dose of compound administered according to this invention to obtain therapeutic and/or prophylactic effect will, of course, be determined by the particular circumstances surrounding the case, including, for example, the specific compound administered, the route of administration, the condition being treated, and the individual being treated. A typical daily dose (administered in single or divided doses) will contain a dosage level of from about 0.01 mg/kg to about 50-100 mg/kg of body weight of an active compound of the invention. Preferred daily doses generally will be from about 0.05 mg/kg to about 20 mg/kg and ideally from about 0.1 mg/kg to about 10 mg/kg. Factors such as clearance rate, half-life and maximum tolerated dose (MTD) have yet to be determined but one of ordinary skill in the art can determine these using standard procedures.
  • In some method embodiments, the preferred therapeutic effect is the inhibition, to some extent, of the growth of cells characteristic of a proliferative disorder, e.g., breast cancer. A therapeutic effect will also normally, but need not, relieve to some extent one or more of the symptoms other than cell growth or size of cell mass. A therapeutic effect may include, for example, one or more of 1) a reduction in the number of cells; 2) a reduction in cell size; 3) inhibition (i.e., slowing to some extent, preferably stopping) of cell infiltration into peripheral organs, e.g., in the instance of cancer metastasis; 3) inhibition (i.e., slowing to some extent, preferably stopping) of tumor metastasis; 4) inhibition, to some extent, of cell growth; and/or 5) relieving to some extent one or more of the symptoms associated with the disorder.
  • As used herein, the term IC50 refers to an amount, concentration or dosage of a particular test compound that achieves a 50% inhibition of a maximal response in an assay that measures such response. In some method embodiments of the invention, the “IC50” value of a compound of the invention can be greater for normal cells than for cells exhibiting a proliferative disorder, e.g., breast cancer cells. The value depends on the assay used.
  • By a “standard” is meant a positive or negative control. A negative control in the context of HER2 expression levels is, e.g., a sample possessing an amount of HER2 protein that correlates with a normal cell. A negative control may also include a sample that contains no HER2 protein. By contrast, a positive control does contain HER2 protein, preferably of an amount that correlates with overexpression as found in proliferative disorders, e.g., breast cancers. The controls may be from cell or tissue samples, or else contain purified ligand (or absent ligand), immobilized or otherwise. In some embodiments, one or more of the controls may be in the form of a diagnostic “dipstick.”
  • By “selectively targeting” is meant affecting one type of cell to a greater extent than another, e.g., in the case of cells with high as opposed to relatively low or normal HER2 levels.
  • II Compounds of the Invention
  • Compounds of the invention and their polymorphs, solvates, esters, tautomers, diastereomers, enantiomers, pharmaceutically acceptable salts or prodrugs show utility for inhibiting HSP90 and treating and/or preventing diseases that are HSP90-dependent.
  • One embodiment of the compounds of the invention is of Formula A:
    Figure US20070185064A1-20070809-C00007

    or a polymorph, solvate, ester, tautomer, diastereomer, enantiomer, pharmaceutically acceptable salt or prodrug thereof, wherein:
      • X1 and X2 are the same or different and each is nitrogen or —CR6;
      • X3 is nitrogen or —CR3 wherein R3 is hydrogen, OH, a keto tautomer, —OR8, —CN, halogen, lower alkyl, or —C(O)R9;
      • X4 is nitrogen or a group CR6 when X3 is nitrogen, and X4 is —CR6R7 when X3 is —CR3;
      • R1 is halogen, —OR8, —SR8, or lower alkyl;
      • R2 is —NR8R10;
      • R4 is —(CH2)n— wherein n=0-3, —C(O), —C(S), —SO2—, or —SO2N—; and
      • R5 is alkyl, aryl, heteroaryl, alicyclic, or heter ocyclic, each of which is optionally bi- or tricyclic, and optionally substituted with H, halogen, lower alkyl, lower alkenyl, lower alkynyl, lower aryl, lower alicyclic, arylalkyl, aryloxyalkyl, alkoxyalkyl, perhaloalkyl, perhaloalkyloxy, perhaloacyl, —N3, —SR8, —OR8, —CN, —CO2R9, —NO2, or —NR8R10;
      • with the provisos that:
      • the one not found or described in one or more of JP 10025294; U.S. Pat. Nos. 4,748,177; 4,748,177; 6,369,092; WO 00/06573; WO 02/055521; WO 02/055082; WO 02/055083; Eur. J. Med. Chem., 1994, 29(1), 3-9; and J. Het. Chem. 1990, 27(5), 1409;
      • —R4R5 is not a ribose or derivative thereof, or a sugar or derivative thereof;
      • —R4R5 is not a phosphonate or phosphonic acid, or substituted with phosphonate or phosphonic acid; and
      • when R4 is (CH2)n where n=0 or 1, then R4 and R5 are not connected with ‘O’, e.g., —CH2—O—CH2— or —CH2—CH2—O—CH2—.
  • In one embodiment, the compound, tautomer, pharmaceutically acceptable salt thereof, or prodrug thereof of Formula A, X1 and X2 are the same or different and each is nitrogen or —CR6; R1 is halogen, —OR8, —SR8, or lower alkyl; R2 is —NR8R10; R3 is hydrogen, —OH or keto tautomer, —OR8, halogen, —CN, lower alkyl, or —C(O)R9; R4 is —(CH2)n— wherein n=0-3, —C(O), —C(S), —SO2—, or —SO2N—; and R5 is alkyl, aromatic, heteroaromatic, alicyclic, heterocyclic, each of which is optionally bi- or tricyclic, and optionally substituted with H, halogen, lower alkyl, —SR8, —OR8, —CN, —CO2R9, —NO2 or —NR8R10; R8 is hydrogen, lower alkyl, lower aryl or —(CO)R9; R9 is lower alkyl, lower aryl, lower heteroaryl, —NR8R10 or OR11; R11 is lower alkyl or lower aryl; and R10 is hydrogen or lower alkyl.
  • In one embodiment, the compound, tautomer, pharmaceutically acceptable salt thereof, or prodrug thereof of Formula A, R1 is selected from halogen, hydroxyl, lower alkoxy, lower thioalkyl and C1-4 alkyl; and R2 is —NH2.
  • In another embodiment, R4 is —(CH2)n—, wherein n=0-3.
  • In another embodiment, R4 is selected from halogen, hydroxyl, lower alkoxy, lower thioalkyl or C1-4 alkyl; optionally wherein R2 is NH2.
  • In another embodiment, R4 is —(CH2)n—, wherein n=0-3.
  • In another embodiment, R4 is —(CH2)n—, wherein n=0-3, R1 is selected from halogen, hydroxyl, lower alkoxy, lower thioalkyl, and C1-4 alkyl, and R2 is optionally NH2.
  • In another embodiment, R1 is halogen, hydroxyl, lower alkoxy, lower thioalkyl, or C1-4 alkyl; and R2 is optionally NH2, R4 is —(CH2)—, and R5 is phenyl, benzyl, or pyridyl, all optionally substituted with H, halogen, lower alkyl, —SR8, —OR8 (or cyclic ethers such as methylenedioxy), —CN, —C02R9, —NO2, or —NR8R10; R8 is hydrogen, lower alkyl, lower aryl or —(CO)R9; R9 is lower alkyl, lower aryl, lower heteroaryl, —NR8R10 or —OR11; R11 is lower alkyl or lower aryl; and R10 is hydrogen or lower alkyl.
  • In another embodiment R1 is halogen, R2 is —NH2, R4 is —CH2—, R6 is H or halogen, and R5 is phenyl optionally substituted with H, halogen, C1-4 alkyl, C1-4 alkoxy, C1-4 alkylthio, perhaloalkyl, perhaloalkyloxy, —CN, —NO2, —NH2 or —CO2R11.
  • In another embodiment, R1 is halogen, R2 is —NH2, R4 is —CH2—, R6 is H, and R5 is 2-halo-3,5-dimethoxyphenyl optionally substituted with H, halogen, C1-4 alkyl, C1-4 alkoxy, C1-4 alkylthio, perhaloalkyl, perhaloalkyloxy, —CN, —NO2, —NH2, or —CO2R11 at the para (4−) position.
  • In another embodiment, R1 is chloro, R2 is —NH2, R4 is —CH2—, R6 is H and R5 is 2-chloro-3,4,5-trimethoxyphenyl.
  • In another embodiment, R1 is chloro, R2 is —NH2, R4 is —CH2—, R6 is H and R5 is 2-bromo-3,4,5-trimethoxyphenyl. In other embodiments, R5 is selected from 2-iodo-3,4,5-trimethoxyphenyl, 2-fluoro-3,4,5-trimethoxyphenyl, and 2-bromo-3,4,5-trimethoxyphenyl.
  • Any of the forgoing embodiments can be combined where feasible and appropriate.
  • In another embodiment, the invention provides compounds of Formula A1:
    Figure US20070185064A1-20070809-C00008

    or a polymorph, solvate, ester, tautomer, diastereomer, enantiomer, pharmaceutically acceptable salt or prodrug thereof, wherein:
      • X1 and X2 are the same or different and each is nitrogen or —CR6;
      • R1 is halogen, —OR8, —SR8, or lower alkyl;
      • R2 is —NR8R10;
      • R4 is —(CH2)n— wherein=0-3, —C(O), —C(S), —SO2—, or —SO2N—;
      • R6 is hydrogen, halogen, lower alkyl, —SR8, —OR8, —NR8R10, —N3, —CN, or —C(O)R9;
      • R5 is alkyl, aryl, heteroaryl, alicyclic, or heterocyclic, all optionally bi- or tri-cyclic, and all optionally substituted with H, halogen, lower alkyl, —SR8, —OR8, —CN, —CO2R9, —NO2, or —NR8R10;
      • R8 is hydrogen, lower alkyl, lower aryl, or —(CO)R9;
      • R9 is lower alkyl, lower aryl, lower heteroaryl, —NR8R10 or —OR11;
      • R10 is hydrogen or lower alkyl,
      • R11 is lower alkyl or lower aryl.
  • In one embodiment of the compounds of Formula A1, or tautomer, pharmaceutically acceptable salt, or prodrug thereof, R1 is halogen, hydroxyl, lower alkoxy, lower thioalkyl, or C1-4 alkyl; and R2 is NH2.
  • In another embodiment of the compounds of Formula A1, or tautomer, pharmaceutically acceptable salt, or prodrug thereof, R4 is —(CH2)n—, where n=0-3.
  • In another embodiment of the compounds of Formula A1, or tautomer, pharmaceutically acceptable salt, or prodrug thereof, R1 is halogen, hydroxyl, lower alkoxy, lower thioalkyl, or C1-4 alkyl; and R2 is NH2; R4 is —(CH2)n—, wherein n=0-3.
  • In another embodiment of the compounds of Formula A1, or tautomer, pharmaceutically acceptable salt thereof, R1 is halogen; R2 is NH2, R4 is —CH2—.
  • In another embodiment of the compounds of Formula A1, or tautomer, pharmaceutically acceptable salt, or prodrug thereof, R4 is —(CH2)—; R5 is phenyl, benzyl, or pyridyl, and is independently optionally substituted with H, halogen, lower alkyl, —SR8, —OR8 (or cyclic ethers such as methylenedioxy), —CN, —CO2R9, —NO2, or —NR8R10; R8 is hydrogen, lower alkyl, lower aryl or —(CO)R9; R9 is lower alkyl, lower aryl, lower heteroaryl, —NR8R10 or —OR11; R11 is lower alkyl or lower aryl; and R10 is hydrogen or lower alkyl.
  • In another embodiment of the compounds of Formula A1, or tautomer, pharmaceutically acceptable salt, or prodrug thereof, R1 is halogen, R2 is —NH2, R4 is —CH2—, R6 is H or halogen, R5 is phenyl optionally substituted with H, halogen, C1-4 alkyl, C1-4 alkoxy, C1-4 alkylthio, perhaloalkyl, perhaloalkyloxy, —CN, —NO2, —NH2 or —CO2R11.
  • In another embodiment of the compounds of Formula A1, or tautomer, pharmaceutically acceptable salt, or prodrug thereof, R1 is halogen, R2 is —NH2, R4 is —CH2—, R6 is H, wherein R5 is 2-halo-3,5-dimethoxyphenyl optionally substituted with H, halogen, C1-4 alkyl, C1-4 alkoxy, C1-4 alkylthio, perhaloalkyl, perhaloalkyloxy, —CN, —NO2, —NH2, or —CO2R11 at the para (4−) position.
  • In another embodiment of the compounds of Formula A1, or tautomer, pharmaceutically acceptable salt, or prodrug thereof, R1 is chloro, R2 is —NH2, R4 is —CH2—, R6 is H and R5 is 2-chloro-3,4,5 -trimethoxyphenyl.
  • In another embodiment of the compounds of Formula A1, or tautomer, pharmaceutically acceptable salt, or prodrug thereof, R1 is chloro, R2 is —NH2, R4is —CH2—, R6 is H and R5is 2-bromo-3,4,5-trimethoxyphenyl.
  • In another embodiment of the compounds of Formula A1, or tautomer, pharmaceutically acceptable salt, or prodrug thereof, R1 is chloro, R2 is —NH2, R4 is —CH2—, R6 is H and R5 is 2-iodo-3,4,5-trimethoxyphenyl.
  • In another embodiment of the compounds of Formula A1, or tautomer, pharmaceutically acceptable salt, or prodrug thereof, R1 is chloro, R2 is —NH2, R4 is —CH2—, R6 is H and R5 is 2-fluoro-3,4,5-trimethoxyphenyl.
  • In another embodiment of the compounds of Formula A1, or tautomer, pharmaceutically acceptable salt, or prodrug thereof, R1 is bromo, R2 is —NH2, R4 is —CH2—, R6 is H, and R5 is 2-bromo-3,4,5-trimethoxyphenyl.
  • In another embodiment of the compounds of Formula A1, or tautomer, pharmaceutically acceptable salt, or prodrug thereof, R1 is bromo, R2 is —NH2, R4 is —CH2—, R6 is H, and R5 is 2-iodo-3,4,5-trimethoxyphenyl.
  • In another embodiment of the compounds of Formula A1, or tautomer, pharmaceutically acceptable salt, or prodrug thereof, R1 is bromo, R2 is —NH2, R4 is —CH2—, R6 is H and R5 is 2-iodo-3,4,5-trimethoxyphenyl.
  • In another embodiment ofthe compounds of Formula A1, or tautomer, pharmaceutically acceptable salt, or prodrug thereof, R1 is bromo, R2 is —NH2, R4 is —CH2—, R6 is H and R5 is 2-fluoro-3,4,5-trimethoxyphenyl.
  • In another embodiment of the compounds of Formula A1, or tautomer, pharmaceutically acceptable salt, or prodrug thereof, R1 is chloro, R2 is —NH2, R4 is —CH2—, R6 is halo and R5 is 2-chloro-3,4,5-trimethoxyphenyl.
  • In another embodiment of the compounds of Formula A1, or tautomer, pharmaceutically acceptable salt, or prodrug thereof, R1 is chloro, R2 is NH2, R4 is —CH2—, R6 is halo, and R5 is 2-bromo-3,4,5-trimethoxyphenyl.
  • In another embodiment of the compounds of Formula A1, or tautomer, pharmaceutically acceptable salt, or prodrug thereof, R1 is bromo, R2 is —NH2, R4 is —CH2—, R6 is halo and R5 is 2-chloro-3,4,5-trimethoxyphenyl.
  • In another embodiment of the compounds of Formula A1, or tautomer, pharmaceutically acceptable salt, or prodrug thereof, R1 is bromo, R2 is —NH2, R4 is —CH2—, R6 is halo and R5 is 2-bromo-3,4,5-trimethoxyphenyl.
  • In another embodiment, the invention provides compounds of Formula I:
    Figure US20070185064A1-20070809-C00009

    or a polymorph, solvate, ester, enantiomer, diastereomer, tautomer, pharmaceutically acceptable salt or prodrug thereof, wherein:
      • R1 is halogen or lower alkyl;
      • R2 is —NR8R10;
      • R4 is —CHR12—;
      • R3 is hydrogen, halogen, or —CN;
      • R5 is aryl, heteroaryl, alicyclic, or heterocyclic, wherein
        • the aryl group is substituted with 3 to 5 substituents,
        • the heteroaryl group is substituted with 2 to 5 substituents, wherein when the heteroaryl is substituted with only two substituents, the two substituents must form part of an optionally substituted fused ring,
        • the alicyclic group is substituted with 3 to 5 substituents,
        • the heterocyclic group is substituted with 3 to 5 substituents, and
        • the substituents are selected from the group of halogen, lower alkyl, lower alkenyl, lower alkynyl, lower aryl, lower alicyclic, arylalkyl, aryloxy, aryloxyalkyl, alkoxyalkyl, perhaloalkyl, perhaloalkyloxy, perhaloacyl, —N3, —SR8, —OR8, —CN, —C(O)R9, —NO2, —NR8R10 phosphonate and phosphonic acid;
      • R8 is hydrogen, lower alkyl, lower aryl, or —C(O)R9;
      • R9 is lower alkyl, lower aryl, lower heteroaryl, —NR10R10 or —OR11;
      • R10 is independently hydrogen or lower alkyl;
      • R11 is lower alkyl, lower aryl or lower heteroaryl;
      • R12 is hydrogen or lower alkyl;
        provided that
      • when R5 is aryl, R5 is not an organo-metallic cyclopentadiene;
      • when R5 is phenyl, the substituents are not 3,5 di-halo;
      • when R5 is alicyclic, the ring system does not contain any tetra-substituted sp3 ring carbons;
      • when R5 is heterocyclic, the ring system does not contain any tetra-substituted sp3 ring carbons or the ring system is not a tetra-substituted pyrrolidine.
  • In one embodiment of the compounds of Formula I or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof, wherein: R1 is halogen or methyl; and R2 is —NHR8, where R8 is hydrogen or —C(O)R9.
  • In another embodiment of the compounds of Formula I, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof, R1 is halogen; R2 is —NH2 and R3 is hydrogen.
  • In another embodiment of the compounds of Formula I, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof, R4 is —(CH2)—.
  • In another embodiment of the compounds of Formula I, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof, R1 is halogen; R2 is —NH2; R4is lower alkyl; R3 is hydrogen; and R5 is aryl or heteroaryl, wherein each of said aryl and heteroaryl groups is monocyclic or bicyclic, the aryl group is substituted with 4 to 5 substituents, the heteroaryl group is substituted with 2 to 5 substituents, wherein when the heteroaryl is substituted with only two substituents, the two substituents must form part of an optionally substituted fused ring.
  • In another embodiment of the compounds of Formula I, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof, R1 is halogen; R2 is —NH2; R3 is hydrogen; R5 is aryl or heteroaryl, wherein each of said aryl and heteroaryl groups is monocyclic or bicyclic, the aryl group is substituted with 4 to 5 substituents, the heteroaryl group is substituted with 2 to 5 substituents, wherein when the heteroaryl is substituted with only two substituents, the two substituents must form part of an optionally substituted fused ring.
  • In another embodiment of the compounds of Formula I, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof, R1 is chloro or bromo, R2 is —NH2, and R5 is a phenyl having 3 to 5 substituents.
  • In another embodiment of the compounds of Formula I, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof, R1 is chloro or bromo, R2 is —NH2, and R5 is a pyridyl having 3 to 5 substituents.
  • In another embodiment of the compounds of Formula I, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof, R1 is chloro or bromo, R2 is —NH2, and R5 is an 1-oxy-pyridyl (N-oxy-pyridyl) having 3 to 5 substituents.
  • In another embodiment of the compounds of Formula I, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof, each of the aryl, heteroaryl, alicyclic or heterocyclic group is monocyclic or bicyclic.
  • In another embodiment, the invention provides compounds of Formula II, which are compounds of Formula I where R4 is —CH2—,
    Figure US20070185064A1-20070809-C00010

    or a polymorph, solvate, ester, tautomer, enantiomer, diastereomer, pharmaceutically acceptable salt or prodrug thereof, wherein:
      • R1 is halogen or lower alkyl;
      • R2 is —NR8R10;
      • R3 is hydrogen, halogen, or —CN;
      • R5 is aryl, heteroaryl, alicyclic, or heterocyclic, wherein
        • the aryl group is substituted with 3 to 5 substituents,
        • the heteroaryl group is substituted with 2 to 5 substituents, wherein when the heteroaryl is substituted with only two substituents, the two substituents must form part of an optionally substituted fused ring,
        • the alicyclic group is substituted with 3 to 5 substituents,
        • the heterocyclic group is substituted with 3 to 5 substituents, and
        • the substituents are selected from the group of halogen, lower alkyl, lower alkenyl, lower alkynyl, lower aryl, lower alicyclic, arylalkyl, aryloxy, aryloxyalkyl, alkoxyalkyl, perhaloalkyl, perhaloalkyloxy, perhaloacyl, —N3, —SR8, —OR8, —CN, —C(O)R9, —NO2, —NR8R10, phosphonate and phosphonic acid;
      • R8 is hydrogen, lower alkyl, lower aryl, or —C(O)R9;
      • R9 is lower alkyl, lower aryl, lower heteroaryl, —NR10R10 or —OR11;
      • R10 is independently hydrogen or lower alkyl;
      • R11 is lower alkyl, lower aryl or lower heteroaryl;
        provided that
      • when R5 is aryl, R5 is not an organo-metallic cyclopentadiene;
      • when R5 is phenyl, the substituents are not 3,5 di-halo;
      • when R5 is alicyclic, the ring system does not contain any tetra-substituted sp3 ring carbons;
      • when R5 is heterocyclic, the ring system does not contain any tetra-substituted sp3 ring carbons or the ring system is not a tetra-substituted pyrrolidine.
  • In another embodiment of the compounds of Formula II, each of the aryl, heteroaryl, alicyclic or heterocyclic group is monocyclic or bicyclic.
  • In another embodiment of the compounds of Formula II, or a polymorph, solvate, ester, tautomer, enantiomer, pharmaceutically acceptable salt or prodrug thereof, R1 is halogen or methyl; and R2 is —NHR8, where R8 is hydrogen or —C(O)R9.
  • In another embodiment of the compounds of Formula II, or a polymorph, solvate, ester, tautomer, enantiomer, pharmaceutically acceptable salt or prodrug thereof, R1 is halogen.
  • In another embodiment of the compounds of Formula II, or a polymorph, solvate, ester, tautomer, enantiomer, pharmaceutically acceptable salt or prodrug thereof, R2 is —NH2 and R3 is hydrogen.
  • In another embodiment of the compounds of Formula II, or a polymorph, solvate, ester, tautomer, enantiomer, pharmaceutically acceptable salt or prodrug thereof, R1 is halogen; R2 is —NH2; R3 is hydrogen; R5 is aryl or heteroaryl, wherein each of the aryl and heteroaryl groups is monocyclic or bicyclic, the aryl group is substituted with 4 to 5 substituents, the heteroaryl group is substituted with 2 to 5 substituents, wherein when the heteroaryl is substituted with only two substituents, the two substituents must form part of an optionally substituted fused ring.
  • In another embodiment of the compounds of Formula II, or a polyrnorph, solvate, ester, tautomer, enantiomer, pharmaceutically acceptable salt or prodrug thereof, R1 is chloro or bromo, R2 is —NH2, and R5 is a phenyl having 3 to 5 substituents.
  • In another embodiment of the compounds of Formula II, or a polymorph, solvate, ester, tautomer, enantiomer, pharmaceutically acceptable salt or prodrug thereof, R1 is chloro or bromo, R2 is —NH2, and R5 is a pyridyl having 3 to 5 substituents.
  • In another embodiment of the compounds of Formula II, or a polymorph, solvate, ester, tautomer, enantiomer, pharmaceutically acceptable salt or prodrug thereof, R1 is chloro or bromo, R2 is —NH2, and R5 is an 1-oxy-pyridyl (N-oxy-pyridyl) having 3 to 5 substituents.
  • It should be understood that any of the foregoing embodiments can be combined where feasible and appropriate.
  • In another embodiment, the invention provides compounds, or polymorphs, solvates, esters, tautomers, pharmaceutically acceptable salts or prodrugs thereof, prepared by the process comprising:
  • reacting a compound of formula Y and a compound of formula Z, wherein:
  • Y is represented by any of the following formulae:
    Figure US20070185064A1-20070809-C00011

    and
      • Z is L1—R4—R5; wherein:
        • L1 is halogen, NR8R10, triflate, tosylate, or mesylate;
        • R4 is —CHR12—,
        • R5 is aryl, heteroaryl, alicyclic, or heterocyclic, wherein:
          • the aryl group is substituted with 3 to 5 substituents,
          • the heteroaryl group is substituted with 2 to 5 substituents, wherein when the heteroaryl is substituted with only two substituents, the two substituents must form part of an optionally substituted fused ring,
          • the alicyclic group is substituted with 3 to 5 substituents,
          • the heterocyclic group is substituted with 3 to 5 substituents, and
          • the substituents are selected from the group of halogen, lower alkyl, lower alkenyl, lower alkynyl, lower aryl, lower alicyclic, arylalkyl, aryloxy, aryloxyalkyl, alkoxyalkyl, perhaloalkyl, perhaloalkyloxy, perhaloacyl, —N3, —SR8, —OR8, —CN, —C(O)R9, —NO2, —NR8R10, phosphonate and phosphonic acid;
        • R8 is hydrogen, lower alkyl, lower aryl, or —C(O)R9;
        • R9 is lower alkyl, lower aryl, lower heteroaryl, —NR10R10 or —OR11;
        • R10 is independently hydrogen or lower alkyl;
        • R11 is lower alkyl, lower aryl or lower heteroaryl;
        • R12 is hydrogen or lower alkyl;
        • R21 is halogen, lower alkyl or —OH;
        • R22 is —NR8R10;
        • R23 is hydrogen, halogen, or —CN;
        • R24 is —NH2, —NO2 or —NO;
        • R25 is halogen or —OH;
        • R26 is —C(O)NH2 or C(O)OEt; and
        • R27 is —NH2, —OH or halogen;
          provided that:
      • when R5 is aryl, R5 is not an organo-metallic cyclopentadiene;
      • when R5 is phenyl, the substituents are not 3,5 di-halo;
      • when R5 is alicyclic, the ring system does not contain any tetra-substituted sp3 ring carbons; or
      • when R5 is heterocyclic, the ring system does not contain any tetra-substituted sp3 ring carbons or the ring system is not a tetra-substituted pyrrolidine.
  • In one embodiment of the compounds prepared by the process of the invention, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof, R5 is aryl, heteroaryl, alicyclic, or heterocyclic, each of which is optionally mono- or bicyclic.
  • In another embodiment of the compounds prepared by the process of the invention, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof, R4 is —CH2—.
  • In another embodiment of the compounds prepared by the process of the invention, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof, L1 is —Cl, —Br or NH2; R5 is aryl or heteroaryl, wherein the aryl group is substituted with 4 to 5 substituents and the heteroaryl group is substituted with 2 to 5 substituents wherein when the heteroaryl is substituted with only two substituents, the two substituents must form part of an optionally substituted fused ring.
  • In another embodiment of the compounds prepared by the process of the invention, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof, Y is a substituted purine.
  • In another embodiment of the compounds prepared by the process of the invention, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof, the reaction is performed in a solvent comprising a member selected from the group of DMF, THF and DMSO.
  • In another embodiment of the compounds prepared by the process of the invention, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof, the reaction is performed in a solvent that comprises DMF.
  • Compounds for use in the method of treatment of the invention are HSP90 inhibitors of the following Formula I, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof,
    Figure US20070185064A1-20070809-C00012

    wherein:
      • R1 is halogen, —OR11, —SR11, —NHR8, hydrogen, or lower alkyl;
      • R2 is —NR8R10;
      • R3 is hydrogen, halogen, —N3, or —CN;
      • R4 is —(CHR12)n— where n=0, 1 or 2, —C(O)—, —C(S)—, or —S(O)—;
      • R5 is alkyl, aryl, heteroaryl, alicyclic, or heterocyclic, all optionally substituted with halogen, lower alkyl, lower alkenyl, lower alkynyl, lower aryl, lower alicyclic, arylalkyl, aryloxy, aryloxyalkyl, alkoxyalkyl, perhaloalkyl, perhaloalkyloxy, perhaloacyl, —N3, —SR8, —OR8, —CN, —C(O)R9, —NO2, or —NR8R10;
      • R8 is hydrogen, lower alky, lower aryl, or —C(O)R9;
      • R9 is lower alkyl, lower aryl, lower heteroaryl, —NR10R10, or —OR11;
      • R10 is independently hydrogen or lower alkyl;
      • R11 is lower alkyl, lower aryl or lower heteroaryl;
      • R12 is hydrogen or lower alkyl;
        provided that:
  • —R4R5 is not a ribose or derivative thereof, or a sugar or derivative thereof;
      • —R4R5 is not a phosphonate or phosphonic acid, or substituted with phosphonate or phosphonic acid; and
      • when R4 is —(CH2)n— where n=1 or 2, then R4 and R5 are not connected through an ether linkage.
  • In one embodiment of the compounds of Formula I for the method of the invention, R3 is hydrogen, halogen or —CN; R5 is aryl, heteroaryl, alicyclic, or heterocyclic, all optionally substituted with halogen, lower alkyl, lower alkenyl, lower alkynyl, lower aryl, lower alicyclic, arylalkyl, aryloxy, aryloxyalkyl, alkoxyalkyl, perhaloalkyl, perhaloalkyloxy, perhaloacyl, —N3, —SR8, —OR8, —CN, —C(O)R9, —NO2, or —NR8R10.
  • In another embodiment of the compounds of Formula I for the method of the invention, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof, R1 is halogen or methyl and R2 is —NHR8, where R8 is hydrogen or —C(O)R9.
  • In another embodiment of the compounds of Formula I for the method of the invention, R2 is —NH2 and R3 is hydrogen.
  • In another embodiment of the compounds of Formula I for the method of the invention, R4 is —CH2—.
  • In another embodiment of the compounds of Formula I for the method of the invention, R1 is halogen; R2 is —NH2; R3 is hydrogen; R4 is —CH2—; R5 is aryl or heteroaryl, the aryl and heteroaryl groups are monocyclic or bicyclic, the aryl group is substituted with 4 to 5 substituents, the heteroaryl group is substituted with 2 to 5 substituents, wherein when the heteroaryl is substituted with two substituents, the two substituents must form part of an optionally substituted fused ring.
  • In another embodiment of the compounds of Formula I for the method of the invention, R1 is chloro or bromo, R2 is —NH2, and R5 is a phenyl having 3 to 5 substituents.
  • In another embodiment of the compounds of Formula I for the method of the invention, R1 is chloro or bromo, R2 is —NH2, and R5 is a pyridyl having 3 to 5 substituents.
  • In another embodiment of the compounds of Formula I for the method of the invention, R1 is chloro or bromo, R2 is —NH2, and R5 is an 1-oxy-pyridyl (N-oxy-pyridyl) having 3 to 5 substituents.
  • It should be understood that any of the foregoing embodiments can be combined where feasible and appropriate.
  • Compounds of the invention that are based on the following Formula I have illustrative species as described in TABLE 1. Suitable prodrugs which can be employed by these compounds include, but are not limited to, those listed in the Definition section.
    TABLE 1
    Exemplary Compounds of the Invention of Formula I
    I
    Figure US20070185064A1-20070809-C00013
    No. Ex. R1 R3 R4 R5
    1 48 Cl H CH2 3,4,5-Trimethoxyphenyl
    2 98 Cl H CH2 2-Chloro-3,4,5-trimethoxyphenyl
    3 51 Cl H CH2 2-Bromo-3,4,5-trimethoxyphenyl
    4 106 Cl H CH2 2-Iodo-3,4,5-trimethoxyphenyl
    5 Cl H CH2 2-Fluoro-3,4,5-trimethoxyphenyl
    6 Cl H CH2 3,4,5-Trimethylphenyl
    7 Cl H CH2 2-Chloro-3,4,5-trimethylphenyl
    8 Cl H CH2 2-Bromo-3,4,5-trimethylphenyl
    9 Cl H CH2 2-Iodo-3,4,5-trimethylphenyl
    10 Cl H CH2 2-Fluoro-3,4,5-trimethylphenyl
    11 43 Cl H CH2 3,5-Dimethoxy-4-methylphenyl
    12 Cl H CH2 2-Chloro-3,5-dimethoxy-4-methylphenyl
    13 44 Cl H CH2 2-Bromo-3,5-dimethoxy-4-methylphenyl
    14 Cl H CH2 2-Iodo-3,5-dimethoxy-4-methylphenyl
    15 Cl H CH2 2-Fluoro-3,5-dimethoxy-4-methylphenyl
    16 Cl H CH2 3,5-Dichloro-4-methylphenyl
    17 Cl H CH2 2,3,5-Trichloro-4-methylphenyl
    18 Cl H CH2 2-Bromo-3,5-dichloro-4-methylphenyl
    19 Cl H CH2 2-Iodo-3,5-dichloro-4-methylphenyl
    20 Cl H CH2 2-Fluoro-3,5-dichloro-4-methylphenyl
    21 Cl H CH2 3,5-Dibromo-4-methylphenyl
    22 Cl H CH2 2-Chloro-3,5-dibromo-4-methylphenyl
    23 Cl H CH2 2,3,5-Tribromo-4-methylphenylphenyl
    24 Cl H CH2 2-Iodo-3,5-dibromo-4-methylphenyl
    25 Cl H CH2 2-Fluoro-3,5-dibromo-4-methylphenyl
    26 Cl H CH2 3,5-Dichloro-4-methoxyphenyl
    27 Cl H CH2 2,3,5-Trichloro-4-methoxyphenyl
    28 Cl H CH2 2-Bromo-3,5-dichloro-4-methoxyphenyl
    29 Cl H CH2 2-Iodo-3,5-dichloro-4-methoxyphenyl
    30 Cl H CH2 2-Fluoro-3,5-dichloro-4-methoxyphenyl
    31 Cl H CH2 3,5-Dibromo-4-methoxyphenyl
    32 Cl H CH2 2-Chloro-3,5-dibromo-4-methoxyphenyl
    33 Cl H CH2 2,3,5-Tribromo-4-methoxyphenyl
    34 Cl H CH2 2-Iodo-3,5-dibromo-4-methoxyphenyl
    35 Cl H CH2 2-Fluoro-3,5-dibromo-4-methoxyphenyl
    36 Cl H CH2 3-Chloro-5-bromo-4-methylphenyl
    37 Cl H CH2 2,3-Dichloro-5-bromo-4-methylphenyl
    38 Cl H CH2 2,5-Dibromo-3-chloro-4-methylphenyl
    39 Cl H CH2 2-Iodo-3-chloro-5-bromo-4-methylphenyl
    40 Cl H CH2 2-Fluoro-3-chloro-5-bromo-4-methylphenyl
    41 Cl H CH2 3-Chloro-5-bromo-4-methoxyphenylphenyl
    42 Cl H CH2 2,3-Dichloro-5-bromo-4-methoxyphenyl
    43 Cl H CH2 2,5-Dibromo-3-chloro-4-methoxyphenyl
    44 Cl H CH2 2-Iodo-3-chloro-5-bromo-4-methoxyphenyl
    45 Cl H CH2 2-Fluoro-3-chloro-5-bromo-4-methoxyphenyl
    46 Cl H CH2 3-Bromo-5-chloro-4-methylphenyl
    47 Cl H CH2 2,5-Dichloro-3-bromo-4-methylphenyl
    48 Cl H CH2 2,3-Dibromo-5-chloro-4-methylphenyl
    49 Cl H CH2 2-Iodo-3-bromo-5-chloro-4-methylphenyl
    50 Cl H CH2 2-Fluoro-3-bromo-5-chloro-4-methylphenyl
    51 Cl H CH2 3-Bromo-5-chloro-4-methoxyphenyl
    52 Cl H CH2 2,5-Dichloro-3-bromo-4-methoxyphenyl
    53 Cl H CH2 2,3-Dibromo-5-chloro-4-methoxyphenyl
    54 Cl H CH2 2-Iodo-3-bromo-5-chloro-4-methoxyphenyl
    55 Cl H CH2 2-Fluoro-3-bromo-5-chloro-4-methoxyphenyl
    56 Cl H CH2 3,5-Dimethoxy-4-trifluoromethylphenyl
    57 Cl H CH2 2-Chloro-3,5-dimethoxy-4-trifluoromethylphenyl
    58 Cl H CH2 2-Bromo-3,5-dimethoxy-4-trifluoromethylphenyl
    59 Cl H CH2 2-Iodo-3,5-dimethoxy-4-trifluoromethylphenyl
    60 Cl H CH2 2-Fluoro-3,5-dimethoxy-4-trifluoromethylphenyl
    61 Cl H CH2 3,5-dibromo-4-trifluoromethoxyphenyl
    62 Cl H CH2 2-Chloro-3,5-dibromo-4-trifluoromethoxyphenyl
    63 Cl H CH2 2,3,5-Tribromo-4-trifluoromethoxyphenyl
    64 Cl H CH2 2-Iodo-3,5-dibromo-4-trifluoromethoxyphenyl
    65 Cl H CH2 2-Fluoro-3,5-dibromo-4-trifluoromethoxyphenyl
    66 Cl H CH2 3,5-Dimethyl-4-methoxyphenyl
    67 Cl H CH2 2-Chloro-3,5-dimethyl-4-methoxyphenyl
    68 Cl H CH2 2-Bromo-3,5-dimethyl-4-methoxyphenyl
    69 Cl H CH2 2-Iodo-3,5-dimethyl-4-methoxyphenyl
    70 Cl H CH2 2-Fluoro-3,5-dimethyl-4-methoxyphenyl
    71 Cl H CH2 3,5-Dimethyl-4-bromophenyl
    72 Cl H CH2 2-Chloro-3,5-dimethyl-4-bromophenyl
    73 Cl H CH2 2,4-Dibromo-3,5-dimethylphenyl
    74 Cl H CH2 2-Iodo-3,5-dimethyl-4-bromophenyl
    75 Cl H CH2 2-Fluoro-3,5-dimethyl-4-bromophenyl
    76 Cl H CH2 3,5-Dimethyl-4-chlorophenyl
    77 Cl H CH2 2,4-Dichloro-3,5-dimethylphenyl
    78 Cl H CH2 2-Bromo-3,5-dimethyl-4-chlorophenyl
    79 Cl H CH2 2-Iodo-3,5-dimethyl-4-chlorophenyl
    80 Cl H CH2 2-Fluoro-3,5-dimethyl-4-chlorophenyl
    81 101 Br H CH2 3,4,5-Trimethoxyphenyl
    82 77 Br H CH2 2-Chloro-3,4,5-trimethoxyphenyl
    83 102 Br H CH2 2-Bromo-3,4,5-trimethoxyphenyl
    84 Br H CH2 2-Iodo-3,4,5-trimethoxyphenyl
    85 Br H CH2 2-Fluoro-3,4,5-trimethoxyphenyl
    86 Br H CH2 3,4,5-Trimethylphenyl
    87 Br H CH2 2-Chloro-3,4,5-trimethylphenyl
    88 Br H CH2 2-Bromo-3,4,5-trimethylphenyl
    89 Br H CH2 2-Iodo-3,4,5-trimethylphenyl
    90 Br H CH2 2-Fluoro-3,4,5-trimethylphenyl
    91 Br H CH2 3,5-Dimethoxy-4-methylphenyl
    92 Br H CH2 2-Chloro-3,5-dimethoxy-4-methylphenyl
    93 Br H CH2 2-Bromo-3,5-dimethoxy-4-methylphenyl
    94 Br H CH2 2-Iodo-3,5-dimethoxy-4-methylphenyl
    95 Br H CH2 2-Fluoro-3,5-dimethoxy-4-methylphenyl
    96 Br H CH2 3,5-Dichloro-4-methylphenyl
    97 Br H CH2 2,3,5-Trichloro-4-methylphenyl
    98 Br H CH2 2-Bromo-3,5-dichloro-4-methylphenyl
    99 Br H CH2 2-Iodo-3,5-dichloro-4-methylphenyl
    100 Br H CH2 2-Fluoro-3,5-dichloro-4-methylphenyl
    101 Br H CH2 3,5-Dibromo-4-methylphenyl
    102 Br H CH2 2-Chloro-3,5-dibromo-4-methylphenyl
    103 Br H CH2 2,3,5-Tribromo-4-methylphenylphenyl
    104 Br H CH2 2-Iodo-3,5-dibromo-4-methylphenyl
    105 Br H CH2 2-Fluoro-3,5-dibromo-4-methylphenyl
    106 Br H CH2 3,5-Dichloro-4-methoxyphenyl
    107 Br H CH2 2,3,5-Trichloro-4-methoxyphenyl
    108 Br H CH2 2-Bromo-3,5-dichloro-4-methoxyphenyl
    109 Br H CH2 2-Iodo-3,5-dichloro-4-methoxyphenyl
    110 Br H CH2 2-Fluoro-3,5-dichloro-4-methoxyphenyl
    111 Br H CH2 3,5-Dibromo-4-methoxyphenyl
    112 Br H CH2 2-Chloro-3,5-dibromo-4-methoxyphenyl
    113 Br H CH2 2,3,5-Tribromo-4-methoxyphenyl
    114 Br H CH2 2-Iodo-3,5-dibromo-4-methoxyphenyl
    115 Br H CH2 2-Fluoro-3,5-dibromo-4-methoxyphenyl
    116 Br H CH2 3-Chloro-5-bromo-4-methylphenyl
    117 Br H CH2 2,3-Dichloro-5-bromo-4-methylphenyl
    118 Br H CH2 2,5-Dibromo-3-chloro-4-methylphenyl
    119 Br H CH2 2-Iodo-3-chloro-5-bromo-4-methylphenyl
    120 Br H CH2 2-Fluoro-3-chloro-5-bromo-4-methylphenyl
    121 Br H CH2 3-Chloro-5-bromo-4-methoxyphenylphenyl
    122 Br H CH2 2,3-Dichloro-5-bromo-4-methoxyphenyl
    123 Br H CH2 2,5-Dibromo-3-chloro-4-methoxyphenyl
    124 Br H CH2 2-Iodo-3-chloro-5-bromo-4-methoxyphenyl
    125 Br H CH2 2-Fluoro-3-chloro-5-bromo-4-methoxyphenyl
    126 Br H CH2 3-Bromo-5-chloro-4-methylphenyl
    127 Br H CH2 2,5-Dichloro-3-bromo-4-methylphenyl
    128 Br H CH2 2,3-Dibromo-5-chloro-4-methylphenyl
    129 Br H CH2 2-Iodo-3-bromo-5-chloro-4-methylphenyl
    130 Br H CH2 2-Fluoro-3-bromo-5-chloro-4-methylphenyl
    131 Br H CH2 3-Bromo-5-chloro-4-methoxyphenyl
    132 Br H CH2 2,5-Dichloro-3-bromo-4-methoxyphenyl
    133 Br H CH2 2,3-Dibromo-5-chloro-4-methoxyphenyl
    134 Br H CH2 2-Iodo-3-bromo-5-chloro-4-methoxyphenyl
    135 Br H CH2 2-Fluoro-3-bromo-5-chloro-4-methoxyphenyl
    136 Br H CH2 3,5-Dimethoxy-4-trifluoromethylphenyl
    137 Br H CH2 2-Chloro-3,5-dimethoxy-4-trifluoromethylphenyl
    138 Br H CH2 2-Bromo-3,5-dimethoxy-4-trifluoromethylphenyl
    139 Br H CH2 2-Iodo-3,5-dimethoxy-4-trifluoromethylphenyl
    140 Br H CH2 2-Fluoro-3,5-dimethoxy-4-trifluoromethylphenyl
    141 Br H CH2 3,5-Dibromo-4-trifluoromethoxyphenyl
    142 Br H CH2 2-Chloro-3,5-dibromo-4-trifluoromethoxyphenyl
    143 Br H CH2 2,3,5-Tribromo-4-trifluoromethoxyphenyl
    144 Br H CH2 2-Iodo-3,5-dibromo-4-trifluoromethoxyphenyl
    145 Br H CH2 2-Fluoro-3,5-dibromo-4-trifluoromethoxyphenyl
    146 Br H CH2 3,5-Dimethyl-4-methoxyphenyl
    147 Br H CH2 2-Chloro-3,5-dimethyl-4-methoxyphenyl
    148 Br H CH2 2-Bromo-3,5-dimethyl-4-methoxyphenyl
    149 Br H CH2 2-Iodo-3,5-dimethyl-4-methoxyphenyl
    150 Br H CH2 2-Fluoro-3,5-dimethyl-4-methoxyphenyl
    151 Br H CH2 3,5-Dimethyl-4-bromophenyl
    152 Br H CH2 2-Chloro-3,5-dimethyl-4-bromophenyl
    153 Br H CH2 2,4-Dibromo-3,5-dimethylphenyl
    154 Br H CH2 2-Iodo-3,5-dimethyl-4-bromophenyl
    155 Br H CH2 2-Fluoro-3,5-dimethyl-4-bromophenyl
    156 Br H CH2 3,5-Dimethyl-4-chlorophenyl
    157 Br H CH2 2,4-Dichloro-3,5-dimethylphenyl
    158 Br H CH2 2-Bromo-3,5-dimethyl-4-chlorophenyl
    159 Br H CH2 2-Iodo-3,5-dimethyl-4-chlorophenyl
    160 F H CH2 3,4,5-Trimethoxyphenyl
    161 F H CH2 2-Chloro-3,4,5-trimethoxyphenyl
    162 F H CH2 2-Bromo-3,4,5-trimethoxyphenyl
    163 F H CH2 2-Iodo-3,4,5-trimethoxyphenyl
    164 F H CH2 2-Fluoro-3,4,5-trimethoxyphenyl
    165 F H CH2 3,4,5-Trimethylphenyl
    166 F H CH2 2-Chloro-3,4,5-trimethylphenyl
    167 F H CH2 2-Bromo-3,4,5-trimethylphenyl
    168 F H CH2 2-Iodo-3,4,5-trimethylphenyl
    169 F H CH2 2-Fluoro-3,4,5-trimethylphenyl
    170 F H CH2 3,5-Dimethoxy-4-methylphenyl
    171 F H CH2 2-Chloro-3,5-dimethoxy-4-methylphenyl
    172 F H CH2 2-Bromo-3,5-dimethoxy-4-methylphenyl
    173 F H CH2 2-Iodo-3,5-dimethoxy-4-methylphenyl
    174 F H CH2 2-Fluoro-3,5-dimethoxy-4-methylphenyl
    175 F H CH2 3,5-Dichloro-4-methylphenyl
    176 F H CH2 2,3,5-Trichloro-4-methylphenyl
    177 F H CH2 2-Bromo-3,5-dichloro-4-methylphenyl
    178 F H CH2 2-Iodo-3,5-dichloro-4-methylphenyl
    179 F H CH2 2-Fluoro-3,5-dichloro-4-methylphenyl
    180 F H CH2 3,5-Dibromo-4-methylphenyl
    181 F H CH2 2-Chloro-3,5-dibromo-4-methylphenyl
    182 F H CH2 2,3,5-Tribromo-4-methylphenylphenyl
    183 F H CH2 2-Iodo-3,5-dibromo-4-methylphenyl
    184 F H CH2 2-Fluoro-3,5-dibromo-4-methylphenyl
    185 F H CH2 3,5-Dichloro-4-methoxyphenyl
    186 F H CH2 2,3,5-Trichloro-4-methoxyphenyl
    187 F H CH2 2-Bromo-3,5-dichloro-4-methoxyphenyl
    188 F H CH2 2-Iodo-3,5-dichloro-4-methoxyphenyl
    189 F H CH2 2-Fluoro-3,5-dichloro-4-methoxyphenyl
    190 F H CH2 3,5-Dibromo-4-methoxyphenyl
    191 F H CH2 2-Chloro-3,5-dibromo-4-methoxyphenyl
    192 F H CH2 2,3,5-Tribromo-4-methoxyphenyl
    193 F H CH2 2-Iodo-3,5-dibromo-4-methoxyphenyl
    194 F H CH2 2-Fluoro-3,5-dibromo-4-methoxyphenyl
    195 F H CH2 3-Chloro-5-bromo-4-methylphenyl
    196 F H CH2 2,3-Dichloro-5-bromo-4-methylphenyl
    197 F H CH2 2,5-Dibromo-3-chloro-4-methylphenyl
    198 F H CH2 2-Iodo-3-chloro-5-bromo-4-methylphenyl
    199 F H CH2 2-Fluoro-3-chloro-5-bromo-4-methylphenyl
    200 F H CH2 3-Chloro-5-bromo-4-methoxyphenylphenyl
    201 F H CH2 2,3-Dichloro-5-bromo-4-methoxyphenyl
    202 F H CH2 2,5-Dibromo-3-chloro-4-methoxyphenyl
    203 F H CH2 2-Iodo-3-chloro-5-bromo-4-methoxyphenyl
    204 F H CH2 2-Fluoro-3-chloro-5-bromo-4-methoxyphenyl
    205 F H CH2 3-Bromo-5-chloro-4-methylphenyl
    206 F H CH2 2,5-Dichloro-3-bromo-4-methylphenyl
    207 F H CH2 2,3-Dibromo-5-chloro-4-methylphenyl
    208 F H CH2 2-Iodo-3-bromo-5-chloro-4-methylphenyl
    209 F H CH2 2-Fluoro-3-bromo-5-chloro-4-methylphenyl
    210 F H CH2 3-Bromo-5-chloro-4-methoxyphenyl
    211 F H CH2 2,5-Dichloro-3-bromo-4-methoxyphenyl
    212 F H CH2 2,3-Dibromo-5-chloro-4-methoxyphenyl
    213 F H CH2 2-Iodo-3-bromo-5-chloro-4-methoxyphenyl
    214 F H CH2 2-Fluoro-3-bromo-5-chloro-4-methoxyphenyl
    215 F H CH2 3,5-Dimethoxy-4-trifluoromethylphenyl
    216 F H CH2 2-Chloro-3,5-dimethoxy-4-trifluoromethylphenyl
    217 F H CH2 2-Bromo-3,5-dimethoxy-4-trifluoromethylphenyl
    218 F H CH2 2-Iodo-3,5-dimethoxy-4-trifluoromethylphenyl
    219 F H CH2 2-Fluoro-3,5-dimethoxy-4-trifluoromethylphenyl
    220 F H CH2 3,5-Dibromo-4-trifluoromethoxyphenyl
    221 F H CH2 2-Chloro-3,5-dibromo-4-trifluoromethoxyphenyl
    222 F H CH2 2,3,5-Tribromo-4-trifluoromethoxyphenyl
    223 F H CH2 2-Iodo-3,5-dibromo-4-trifluoromethoxyphenyl
    224 F H CH2 2-Fluoro-3,5-dibromo-4-trifluoromethoxyphenyl
    225 F H CH2 3,5-Dimethyl-4-methoxyphenyl
    226 F H CH2 2-Chloro-3,5-dimethyl-4-methoxyphenyl
    227 F H CH2 2-Bromo-3,5-dimethyl-4-methoxyphenyl
    228 F H CH2 2-Iodo-3,5-dimethyl-4-methoxyphenyl
    229 F H CH2 2-Fluoro-3,5-dimethyl-4-methoxyphenyl
    230 F H CH2 3,5-Dimethyl-4-bromophenyl
    231 F H CH2 2-Chloro-3,5-dimethyl-4-bromophenyl
    232 F H CH2 2,4-Dibromo-3,5-dimethylphenyl
    233 F H CH2 2-Iodo-3,5-dimethyl-4-bromophenyl
    234 F H CH2 2-Fluoro-3,5-dimethyl-4-bromophenyl
    235 F H CH2 3,5-Dimethyl-4-chlorophenyl
    236 F H CH2 2,4-Dichloro-3,5-dimethylphenyl
    237 F H CH2 2-Bromo-3,5-dimethyl-4-chlorophenyl
    238 F H CH2 2-Iodo-3,5-dimethyl-4-chlorophenyl
    239 F H CH2 2-Fluoro-3,5-dimethyl-4-chlorophenyl
    240 25 Cl H CH2 3,5-Dimethyl-4-methoxypyridin-2-yl
    241 31 Cl H CH2 3,5-Dimethyl-4-methoxy-1-oxypyridin-2-yl
    242 Cl H CH2 6-Bromo-3,5-dimethyl-4-methoxypyridin-2-yl
    243 33 Cl H CH2 6-Chloro-3,5-dimethyl-4-methoxypyridin-2-yl
    244 Cl H CH2 6-Chloro-3,5-dimethyl-4-methoxy-1-oxypyridin-2-yl
    245 Cl H CH2 6-Bromo-3,5-dimethyl-4-methoxy-1-oxypyridin-2-yl
    246 1 Cl H CH2 3,5-Dimethyl-4-bromopyridin-2-yl
    247 4 Cl H CH2 3,5-Dimethyl-4-bromo-1-oxypyridin-2-yl
    248 Cl H CH2 6-Bromo-3,5-dimethyl-4-bromopyridin-2-yl
    249 Cl H CH2 6-Chloro-3,5-dimethyl-4-bromopyridin-2-yl
    250 Cl H CH2 6-Chloro-3,5-dimethyl-4-bromo-1-oxypyridin-2-yl
    251 Cl H CH2 4,6-Dibromo-3,5-dimethyl-1-oxypyridin-2-yl
    252 18 Cl H CH2 3,5-Dimethyl-4-chloropyridin-2-yl
    253 19 Cl H CH2 3,5-Dimethyl-4-chloro-1-oxypyridin-2-yl
    254 Cl H CH2 6-Bromo-3,5-dimethyl-4-chloropyridin-2-yl
    255 Cl H CH2 6-Chloro-3,5-dimethyl-4-chloropyridin-2-yl
    256 Cl H CH2 4,6-Dichloro-3,5-dimethyl-1-oxypyridin-2-yl
    257 Cl H CH2 6-Bromo-3,5-dimethyl-4-chloro-1-oxypyridin-2-yl
    258 111 Cl H CH2 3,5-Dimethyl-4-iodopyridin-2-yl
    259 Cl H CH2 3,5-Dimethyl-4-iodo-1-oxypyridin-2-yl
    260 Cl H CH2 6-Bromo-3,5-dimethyl-4-iodopyridin-2-yl
    261 Cl H CH2 6-Chloro-3,5-dimethyl-4-iodopyridin-2-yl
    262 Cl H CH2 6-Chloro-3,5-dimethyl-4-iodo-1-oxypyridin-2-yl
    263 Cl H CH2 6-Bromo-3,5-dimethyl-4-iodo-1-oxypyridin-2-yl
    264 115 Cl H CH2 3,5-Dimethyl-4-thiomethyl-pyridin-2-yl
    265 Cl H CH2 3,5-Dimethyl-4-thiomethyl-1-oxypyridin-2-yl
    266 Cl H CH2 6-Bromo-3,5-dimethyl-4-thiomethyl-pyridin-2-yl
    267 Cl H CH2 6-Chloro-3,5-dimethyl-4-thiomethyl-pyridin-2-yl
    268 Cl H CH2 6-Chloro-3,5-dimethyl-4-thiomethyl-1-oxypyridin-2-yl
    269 Cl H CH2 6-Bromo-3,5-dimethyl-4-thiomethyl-1-oxypyridin-2-yl
    270 117 Cl H CH2 3,4,5-Trimethyl-pyridin-2-yl
    271 Cl H CH2 3,4,5-Trimethyl-1-oxypyridin-2-yl
    272 Cl H CH2 6-Bromo-3,4,5-trimethyl-pyridin-2-yl
    273 Cl H CH2 6-Chloro-3,4,5-trimethyl-pyridin-2-yl
    274 Cl H CH2 6-Chloro-3,4,5-trimethyl-1-oxypyridin-2-yl
    275 Cl H CH2 6-Bromo-3,4,5-trimethyl-1-oxypyridin-2-yl
    276 Cl H CH2 4,5,6-Trimethoxypyridin-2-yl
    277 Cl H CH2 4,5,6-Trimethoxy-1-oxypyridin-2-yl
    278 Cl H CH2 3-Bromo-4,5,6-trimethoxypyridin-2-yl
    279 Cl H CH2 3-Chloro-4,5,6-trimethoxypyridin-2-yl
    280 Cl H CH2 3-Chloro-4,5,6-trimethoxy-1-oxypyridin-2-yl
    281 Cl H CH2 3-Bromo-4,5,6-trimethoxy-1-oxypyridin-2-yl
    282 Cl H CH2 3,4,5-Trimethoxy-pyridin-2-yl
    283 Cl H CH2 3,4,5-Trimethoxy-1-oxypyridin-2-yl
    284 Cl H CH2 3-Bromo-3,4,5-trimethoxy-pyridin-2-yl
    285 Cl H CH2 3-Chloro-3,4,5-trimethoxy-pyridin-2-yl
    286 Cl H CH2 3-Chloro-3,4,5-trimethoxy-1-oxypyridin-2-yl
    287 Cl H CH2 3-Bromo-3,4,5-trimethoxy-1-oxypyridin-2-yl
    288 Cl H CH2 4,5,6-Trimethyl-pyridin-2-yl
    289 Cl H CH2 4,5,6-Trimethyl-1-oxypyridin-2-yl
    290 Cl H CH2 3-Bromo-4,5,6-trimethyl-pyridin-2-yl
    291 Cl H CH2 3-Chloro-4,5,6-trimethyl-pyridin-2-yl
    292 Cl H CH2 3-Chloro-4,5,6-trimethyl-1-oxypyridin-2-yl
    293 Cl H CH2 3-Bromo-4,5,6-trimethyl-1-oxypyridin-2-yl
    294 Cl H CH2 4,6-Dimethyl-5-methoxy-pyridin-2-yl
    295 Cl H CH2 4,6-Dimethyl-5-methoxy-1-oxypyridin-2-yl
    296 Cl H CH2 3-Bromo-4,6-dimethyl-5-methoxy-pyridin-2-yl
    297 Cl H CH2 3-Chloro-4,6-dimethyl-5-methoxy-pyridin-2-yl
    298 Cl H CH2 3-Chloro-4,6-dimethyl-5-methoxy-1-oxypyridin-2-yl
    299 Cl H CH2 3-Bromo-4,6-dimethyl-5-methoxy-1-oxypyridin-2-yl
    300 Cl H CH2 4-Bromo-5,6-dimethoxy-pyridin-2-yl
    301 Cl H CH2 4-Bromo-5,6-dimethoxy-1-oxypyridin-2-yl
    302 Cl H CH2 3,4-Dibromo-5,6-dimethoxy-pyridin-2-yl
    303 Cl H CH2 3-Chloro-4-bromo-5,6-dimethoxy-pyridin-2-yl
    304 Cl H CH2 3-Chloro-4-bromo-5,6-dimethoxy-1-oxypyridin-2-yl
    305 Cl H CH2 3,4-Dibromo-5,6-dimethoxy-1-oxypyridin-2-yl
    306 41 Cl H CH2 4,6-Dimethyl-5-methoxypyridin-3-yl
    307 Cl H CH2 4,6-Dimethyl-5-methoxy-1-oxypyridin-3-yl
    308 Cl H CH2 4,6-Dimethyl-5-bromopyridin-3-yl
    309 Cl H CH2 4,6-Dimethyl-5-chloropyridin-3-yl
    310 Cl H CH2 5,6-Dimethyl-4-bromopyridin-3-yl
    311 Cl H CH2 5,6-Dimethyl-4-chloropyridin-3-yl
    312 Cl H CH2 4,6-Dimethyl-5-bromo-1-oxypyridin-pyridin-3-yl
    313 Cl H CH2 4,6-Dimethyl-5-chloro-1-oxypyridin-pyridin-3-yl
    314 Cl H CH2 5,6-Dimethyl-4-bromo-1-oxypyridin-pyridin-3-yl
    315 Cl H CH2 5,6-Dimethyl-4-chloro-1-oxypyridin-pyridin-3-yl
    316 Cl H CH2 2,6-Dimethyl-3-methoxypyridin-4-yl
    317 Cl H CH2 2,6-Dimethyl-pyridin-4-yl
    318 Cl H CH2 2,3,6-Trimethyl-pyridin-4-yl
    319 Cl H CH2 2,3,6-Trimethoxy-pyridin-4-yl
    320 Cl H CH2 2,6-Dimethyl-3-bromopyridin-4-yl
    321 Cl H CH2 2,6-Dimethyl-3-chloropyridin-4-yl
    322 Cl H CH2 2,6-Dichloro-3-bromopyridin-4-yl
    323 Cl H CH2 2,6-Dibromo-3-chloropyridin-4-yl
    324 Cl H CH2 2,3,6-Trichloro-pyridin-4-yl
    325 Cl H CH2 2,3,6-Tribromo-pyridin-4-yl
    326 Cl H CH2 2,6-Dimethyl-3-methoxy-1-oxy-pyridin-4-yl
    327 Cl H CH2 2,6-Dimethyl-1-oxy-pyridin-4-yl
    328 Cl H CH2 2,3,6-Trimethyl-1-oxy-pyridin-4-yl
    329 Cl H CH2 2,3,6-Trimethoxy-1-oxy-pyridin-4-yl
    330 Cl H CH2 2,6-Dimethyl-3-bromo1-oxy-pyridin-4-yl
    331 Cl H CH2 2,6-Dimethyl-3-chloro1-oxy-pyridin-4-yl
    332 Cl H CH2 2,6-Dichloro-3-bromo-1-oxy-pyridin-4-yl
    333 Cl H CH2 2,6-Dibromo-3-chloro-1-oxy-pyridin-4-yl
    334 Cl H CH2 2,3,6-Trichloro-1-oxy-pyridin-4-yl
    335 Cl H CH2 2,3,6-Tribromo-1-oxy-pyridin-4-yl
    336 Cl H CH2 4,6-Dimethyl-5-iodopyridin-3-yl
    337 Cl H CH2 5,6-Dimethyl-4-iodopyridin-3-yl
    338 Cl H CH2 4,5,6-Trichloropyridin-3-yl
    339 Cl H CH2 4,5,6-Tribromopyridin-3-yl
    340 21 Br H CH2 3,5-Dimethyl-4-methoxypyridin-2-yl
    341 24 Br H CH2 3,5-Dimethyl-4-methoxy-1-oxypyridin-2-yl
    342 Br H CH2 6-Bromo-3,5-dimethyl-4-methoxypyridin-2-yl
    343 Br H CH2 6-Chloro-3,5-dimethyl-4-methoxypyridin-2-yl
    344 Br H CH2 6-Chloro-3,5-dimethyl-4-methoxy-1-oxypyridin-2-yl
    345 Br H CH2 6-Bromo-3,5-dimethyl-4-methoxy-1-oxypyridin-2-yl
    346 5 Br H CH2 3,5-Dimethyl-4-bromopyridin-2-yl
    347 6 Br H CH2 3,5-Dimethyl-4-bromo-1-oxypyridin-2-yl
    348 Br H CH2 6-Bromo-3,5-dimethyl-4-bromopyridin-2-yl
    349 Br H CH2 6-Chloro-3,5-dimethyl-4-bromopyridin-2-yl
    350 Br H CH2 6-Chloro-3,5-dimethyl-4-bromo-1-oxypyridin-2-yl
    351 Br H CH2 4,6-Dibromo-3,5-dimethyl-1-oxypyridin-2-yl
    352 Br H CH2 3,5-Dimethyl-4-chloropyridin-2-yl
    353 Br H CH2 3,5-Dimethyl-4-chloro-1-oxypyridin-2-yl
    354 Br H CH2 6-Bromo-3,5-dimethyl-4-chloropyridin-2-yl
    355 Br H CH2 6-Chloro-3,5-dimethyl-4-chloropyridin-2-yl
    356 Br H CH2 4,6-Dichloro-3,5-dimethyl-1-oxypyridin-2-yl
    357 Br H CH2 6-Bromo-3,5-dimethyl-4-chloro-1-oxypyridin-2-yl
    358 Br H CH2 3,5-Dimethyl-4-iodopyridin-2-yl
    359 Br H CH2 3,5-Dimethyl-4-iodo-1-oxypyridin-2-yl
    360 Br H CH2 6-Bromo-3,5-dimethyl-4-iodopyridin-2-yl
    361 Br H CH2 6-Chloro-3,5-dimethyl-4-iodopyridin-2-yl
    362 Br H CH2 6-Chloro-3,5-dimethyl-4-iodo-1-oxypyridin-2-yl
    363 Br H CH2 6-Bromo-3,5-dimethyl-4-iodo-1-oxypyridin-2-yl
    364 Br H CH2 3,5-Dimethyl-4-thiomethyl-pyridin-2-yl
    365 Br H CH2 3,5-Dimethyl-4-thiomethyl-1-oxypyridin-2-yl
    366 Br H CH2 6-Bromo-3,5-dimethyl-4-thiomethyl-pyridin-2-yl
    367 Br H CH2 6-Chloro-3,5-dimethyl-4-thiomethyl-pyridin-2-yl
    368 Br H CH2 6-Chloro-3,5-dimethyl-4-thiomethyl-1-oxypyridin-2-yl
    369 Br H CH2 6-Bromo-3,5-dimethyl-4-thiomethyl-1-oxypyridin-2-yl
    370 118 Br H CH2 3,4,5-Trimethyl-pyridin-2-yl
    371 119 Br H CH2 3,4,5-Trimethyl-1-oxypyridin-2-yl
    372 Br H CH2 6-Bromo-3,4,5-trimethyl-pyridin-2-yl
    373 Br H CH2 6-Chloro-3,4,5-trimethyl-pyridin-2-yl
    374 Br H CH2 6-Chloro-3,4,5-trimethyl-1-oxypyridin-2-yl
    375 Br H CH2 6-Bromo-3,4,5-trimethyl-1-oxypyridin-2-yl
    376 Br H CH2 3,4,5-Trimethoxy-pyridin-2-yl
    377 Br H CH2 3,4,5-Trimethoxy-1-oxypyridin-2-yl
    378 Br H CH2 6-Bromo-3,4,5-trimethoxy-pyridin-2-yl
    379 Br H CH2 6-Chloro-3,4,5-trimethoxy-pyridin-2-yl
    380 Br H CH2 6-Chloro-3,4,5-trimethoxy-1-oxypyridin-2-yl
    381 Br H CH2 6-Bromo-3,4,5-trimethoxy-1-oxypyridin-2-yl
    382 Br H CH2 4,5,6-Trimethoxypyridin-2-yl
    383 Br H CH2 4,5,6-Trimethoxy-1-oxypyridin-2-yl
    384 Br H CH2 3-Bromo-4,5,6-trimethoxypyridin-2-yl
    385 Br H CH2 3-Chloro-4,5,6-trimethoxypyridin-2-yl
    386 Br H CH2 3-Chloro-4,5,6-trimethoxy-1-oxypyridin-2-yl
    387 Br H CH2 3-Bromo-4,5,6-trimethoxy-1-oxypyridin-2-yl
    388 Br H CH2 4,5,6-Trimethoxypyridin-2-yl
    389 Br H CH2 4,5,6-Trimethoxy-1-oxypyridin-2-yl
    390 Br H CH2 3-Bromo-4,5,6-trimethyl-pyridin-2-yl
    391 Br H CH2 3-Chloro-4,5,6-trimethyl-pyridin-2-yl
    392 Br H CH2 3-Chloro-4,5,6-trimethyl-1-oxypyridin-2-yl
    393 Br H CH2 3-Bromo-4,5,6-trimethyl-1-oxypyridin-2-yl
    394 Br H CH2 4,6-Dimethyl-5-methoxy-pyridin-2-yl
    395 Br H CH2 4,6-Dimethyl-5-methoxy-1-oxypyridin-2-yl
    396 Br H CH2 3-Bromo-4,6-dimethyl-5-methoxy-pyridin-2-yl
    397 Br H CH2 3-Chloro-4,6-dimethyl-5-methoxy-pyridin-2-yl
    398 Br H CH2 3-Chloro-4,6-dimethyl-5-methoxy-1-oxypyridin-2-yl
    399 Br H CH2 3-Bromo-4,6-dimethyl-5-methoxy-1-oxypyridin-2-yl
    400 Br H CH2 4-Bromo-5,6-dimethoxy-pyridin-2-yl
    401 Br H CH2 4-Bromo-5,6-dimethoxy-1-oxypyridin-2-yl
    402 Br H CH2 3,4-Dibromo-5,6-dimethoxy-pyridin-2-yl
    403 Br H CH2 3-Chloro-4-bromo-5,6-dimethoxy-pyridin-2-yl
    404 Br H CH2 3-Chloro-4-bromo-5,6-dimethoxy-1-oxypyridin-2-yl
    405 Br H CH2 3,4-Dibromo-5,6-dimethoxy-1-oxypyridin-2-yl
    406 Br H CH2 4,6-Dimethyl-5-methoxypyridin-3-yl
    407 Br H CH2 4,6-Dimethyl-5-methoxy-1-oxypyridin-3-yl
    408 Br H CH2 4,6-Dimethyl-5-bromopyridin-3-yl
    409 Br H CH2 4,6-Dimethyl-5-chloropyridin-3-yl
    410 Br H CH2 5,6-Dimethyl-4-bromopyridin-3-yl
    411 Br H CH2 5,6-Dimethyl-4-chloropyridin-3-yl
    412 Br H CH2 4,6-Dimethyl-5-bromo-1-oxypyridin-pyridin-3-yl
    413 Br H CH2 4,6-Dimethyl-5-chloro-1-oxypyridin-pyridin-3-yl
    414 Br H CH2 5,6-Dimethyl-4-bromo-1-oxypyridin-pyridin-3-yl
    415 Br H CH2 5,6-Dimethyl-4-chloro-1-oxypyridin-pyridin-3-yl
    416 Br H CH2 2,6-Dimethyl-3-methoxypyridin-4-yl
    417 Br H CH2 2,6-Dimethyl-pyridin-4-yl
    418 Br H CH2 2,3,6-Trimethyl-pyridin-4-yl
    419 Br H CH2 2,3,6-Trimethoxy-pyridin-4-yl
    420 Br H CH2 2,6-Dimethyl-3-bromopyridin-4-yl
    421 Br H CH2 2,6-Dimethyl-3-chloropyridin-4-yl
    422 Br H CH2 2,6-Dichloro-3-bromopyridin-4-yl
    423 Br H CH2 2,6-Dibromo-3-chloropyridin-4-yl
    424 Br H CH2 2,3,6-Trichloro-pyridin-4-yl
    425 Br H CH2 2,3,6-Tribromo-pyridin-4-yl
    426 Br H CH2 2,6-Dimethyl-3-methoxy-1-oxy-pyridin-4-yl
    427 Br H CH2 2,6-Dimethyl-1-oxy-pyridin-4-yl
    428 Br H CH2 2,3,6-Trimethyl-1-oxy-pyridin-4-yl
    429 Br H CH2 2,3,6-Trimethoxy-1-oxy-pyridin-4-yl
    430 Br H CH2 2,6-Dimethyl-3-bromo1-oxy-pyridin-4-yl
    431 Br H CH2 2,6-Dimethyl-3-chloro1-oxy-pyridin-4-yl
    432 Br H CH2 2,6-Dichloro-3-bromo1-oxy-pyridin-4-yl
    433 Br H CH2 2,6-Dibromo-3-chloro1-oxy-pyridin-4-yl
    434 Br H CH2 2,3,6-Trichloro-1-oxy-pyridin-4-yl
    435 Br H CH2 2,3,6-Tribromo-1-oxy-pyridin-4-yl
    436 Br H CH2 4,6-Dimethyl-5-iodopyridin-3-yl
    437 Br H CH2 5,6-Dimethyl-4-iodopyridin-3-yl
    438 Br H CH2 4,5,6-Trichloropyridin-3-yl
    439 Br H CH2 4,5,6-Tribromopyridin-3-yl
    440 F H CH2 3,5-Dimethyl-4-methoxypyridin-2-yl
    441 F H CH2 3,5-Dimethyl-4-methoxy-1-oxypyridin-2-yl
    442 F H CH2 6-Bromo-3,5-dimethyl-4-methoxypyridin-2-yl
    443 F H CH2 6-Chloro-3,5-dimethyl-4-methoxypyridin-2-yl
    444 F H CH2 6-Chloro-3,5-dimethyl-4-methoxy-1-oxypyridin-2-yl
    445 F H CH2 6-Bromo-3,5-dimethyl-4-methoxy-1-oxypyridin-2-yl
    446 F H CH2 3,5-Dimethyl-4-bromopyridin-2-yl
    447 F H CH2 3,5-Dimethyl-4-bromo-1-oxypyridin-2-yl
    448 F H CH2 6-Bromo-3,5-dimethyl-4-bromopyridin-2-yl
    449 F H CH2 6-Chloro-3,5-dimethyl-4-bromopyridin-2-yl
    450 F H CH2 6-Chloro-3,5-dimethyl-4-bromo-1-oxypyridin-2-yl
    451 F H CH2 4,6-Dibromo-3,5-dimethyl-1-oxypyridin-2-yl
    452 F H CH2 3,5-Dimethyl-4-chloropyridin-2-yl
    453 F H CH2 3,5-Dimethyl-4-chloro-1-oxypyridin-2-yl
    454 F H CH2 6-Bromo-3,5-dimethyl-4-chloropyridin-2-yl
    455 F H CH2 6-Chloro-3,5-dimethyl-4-chloropyridin-2-yl
    456 F H CH2 4,6-Dichloro-3,5-dimethyl-1-oxypyridin-2-yl
    457 F H CH2 6-Bromo-3,5-dimethyl-4-chloro-1-oxypyridin-2-yl
    458 F H CH2 3,5-Dimethyl-4-iodopyridin-2-yl
    459 F H CH2 3,5-Dimethyl-4-iodo-1-oxypyridin-2-yl
    460 F H CH2 6-Bromo-3,5-dimethyl-4-iodopyridin-2-yl
    461 F H CH2 6-Chloro-3,5-dimethyl-4-iodopyridin-2-yl
    462 F H CH2 6-Chloro-3,5-dimethyl-4-iodo-1-oxypyridin-2-yl
    463 F H CH2 6-Bromo-3,5-dimethyl-4-iodo-1-oxypyridin-2-yl
    464 F H CH2 3,5-Dimethyl-4-thiomethyl-pyridin-2-yl
    465 F H CH2 3,5-Dimethyl-4-thiomethyl-1-oxypyridin-2-yl
    466 F H CH2 6-Bromo-3,5-dimethyl-4-thiomethyl-pyridin-2-yl
    467 F H CH2 6-Chloro-3,5-dimethyl-4-thiomethyl-pyridin-2-yl
    468 F H CH2 6-Chloro-3,5-dimethyl-4-thiomethyl-1-oxypyridin-2-yl
    469 F H CH2 6-Bromo-3,5-dimethyl-4-thiomethyl-1-oxypyridin-2-yl
    470 F H CH2 3,4,5-Trimethyl-pyridin-2-yl
    471 F H CH2 3,4,5-Trimethyl-1-oxypyridin-2-yl
    472 F H CH2 6-Bromo-3,4,5-trimethyl-pyridin-2-yl
    473 F H CH2 6-Chloro-3,4,5-trimethyl-pyridin-2-yl
    474 F H CH2 6-Chloro-3,4,5-trimethyl-1-oxypyridin-2-yl
    475 F H CH2 6-Bromo-3,4,5-trimethyl-1-oxypyridin-2-yl
    476 F H CH2 3,4,5-Trimethoxy-pyridin-2-yl
    477 F H CH2 3,4,5-Trimethoxy-1-oxypyridin-2-yl
    478 F H CH2 6-Bromo-3,4,5-trimethoxy-pyridin-2-yl
    479 F H CH2 6-Chloro-3,4,5-trimethoxy-pyridin-2-yl
    480 F H CH2 6-Chloro-3,4,5-trimethoxy-1-oxypyridin-2-yl
    481 F H CH2 6-Bromo-3,4,5-trimethoxy-1-oxypyridin-2-yl
    482 F H CH2 4,5,6-Trimethoxy-pyridin-2-yl
    483 F H CH2 4,5,6-Trimethoxy-1-oxypyridin-2-yl
    484 F H CH2 3-Bromo-4,5,6-trimethoxy-pyridin-2-yl
    485 F H CH2 3-Chloro-4,5,6-trimethoxy-pyridin-2-yl
    486 F H CH2 3-Chloro-4,5,6-trimethoxy-1-oxypyridin-2-yl
    487 F H CH2 3-Bromo-4,5,6-trimethoxy-1-oxypyridin-2-yl
    488 F H CH2 4,5,6-Trimethyl-pyridin-2-yl
    489 F H CH2 4,5,6-Trimethyl-1-oxypyridin-2-yl
    490 F H CH2 3-Bromo-4,5,6-trimethyl-pyridin-2-yl
    491 F H CH2 3-Chloro-4,5,6-trimethyl-pyridin-2-yl
    492 F H CH2 3-Chloro-4,5,6-trimethyl-1-oxypyridin-2-yl
    493 F H CH2 3-Bromo-4,5,6-trimethyl-1-oxypyridin-2-yl
    494 F H CH2 4,6-Dimethyl-5-methoxy-pyridin-2-yl
    495 F H CH2 4,6-Dimethyl-5-methoxy-1-oxypyridin-2-yl
    496 F H CH2 3-Bromo-4,6-dimethyl-5-methoxy-pyridin-2-yl
    497 F H CH2 3-Chloro-4,6-dimethyl-5-methoxy-pyridin-2-yl
    498 F H CH2 3-Chloro-4,6-dimethyl-5-methoxy-1-oxypyridin-2-yl
    499 F H CH2 3-Bromo-4,6-dimethyl-5-methoxy-1-oxypyridin-2-yl
    500 F H CH2 4-Bromo-5,6-dimethoxy-pyridin-2-yl
    501 F H CH2 4-Bromo-5,6-dimethoxy-1-oxypyridin-2-yl
    502 F H CH2 3,4-Dibromo-5,6-dimethoxy-pyridin-2-yl
    503 F H CH2 3-Chloro-4-bromo-5,6-dimethoxy-pyridin-2-yl
    504 F H CH2 3-Chloro-4-bromo-5,6-dimethoxy-1-oxypyridin-2-yl
    505 F H CH2 3,4-Dibromo-5,6-dimethoxy-1-oxypyridin-2-yl
    506 F H CH2 4,6-Dimethyl-5-methoxypyridin-3-yl
    507 F H CH2 4,6-Dimethyl-5-methoxy-1-oxypyridin-3-yl
    508 F H CH2 4,6-Dimethyl-5-bromopyridin-3-yl
    509 F H CH2 4,6-Dimethyl-5-chloropyridin-3-yl
    510 F H CH2 5,6-Dimethyl-4-bromopyridin-3-yl
    511 F H CH2 5,6-Dimethyl-4-chloropyridin-3-yl
    512 F H CH2 4,6-Dimethyl-5-bromo-1-oxypyridin-pyridin-3-yl
    513 F H CH2 4,6-Dimethyl-5-chloro-1-oxypyridin-pyridin-3-yl
    514 F H CH2 5,6-Dimethyl-4-bromo-1-oxypyridin-pyridin-3-yl
    515 F H CH2 5,6-Dimethyl-4-chloro-1-oxypyridin-pyridin-3-yl
    516 F H CH2 2,6-Dimethyl-3-methoxypyridin-4-yl
    517 F H CH2 2,6-Dimethyl-pyridin-4-yl
    518 F H CH2 2,3,6-Trimethyl-pyridin-4-yl
    519 F H CH2 2,3,6-Trimethoxy-pyridin-4-yl
    520 F H CH2 2,6-Dimethyl-3-bromopyridin-4-yl
    521 F H CH2 2,6-Dimethyl-3-chloropyridin-4-yl
    522 F H CH2 2,6-Dichloro-3-bromopyridin-4-yl
    523 F H CH2 2,6-Dibromo-3-chloropyridin-4-yl
    524 F H CH2 2,3,6-Trichloro-pyridin-4-yl
    525 F H CH2 2,3,6-Tribromo-pyridin-4-yl
    526 F H CH2 2,6-Dimethyl-3-methoxy-1-oxy-pyridin-4-yl
    527 F H CH2 2,6-Dimethyl-1-oxy-pyridin-4-yl
    528 F H CH2 2,3,6-Trimethyl-1-oxy-pyridin-4-yl
    529 F H CH2 2,3,6-Trimethoxy-1-oxy-pyridin-4-yl
    530 F H CH2 2,6-Dimethyl-3-bromo1-oxy-pyridin-4-yl
    531 F H CH2 2,6-Dimethyl-3-chloro1-oxy-pyridin-4-yl
    532 F H CH2 2,6-Dichloro-3-bromo1-oxy-pyridin-4-yl
    533 F H CH2 2,6-Dibromo-3-chloro1-oxy-pyridin-4-yl
    534 F H CH2 2,3,6-Trichloro-1-oxy-pyridin-4-yl
    536 F H CH2 2,3,6-Tribromo-1-oxy-pyridin-4-yl
    537 F H CH2 4,6-Dimethyl-5-iodopyridin-3-yl
    538 F H CH2 5,6-Dimethyl-4-iodopyridin-3-yl
    539 F H CH2 4,5,6-Trichloropyridin-3-yl
    540 Cl Cl CH2 3,5-Dimethyl-4-methoxypyridin-2-yl
    541 Br Cl CH2 3,5-Dimethyl-4-methoxypyridin-2-yl
    542 Cl Br CH2 3,5-Dimethyl-4-methoxypyridin-2-yl
    543 Br Br CH2 3,5-Dimethyl-4-methoxypyridin-2-yl
    544 Cl N3 CH2 3,5-Dimethyl-4-methoxypyridin-2-yl
    545 Br N3 CH2 3,5-Dimethyl-4-methoxypyridin-2-yl
    546 F Cl CH2 3,5-Dimethyl-4-methoxypyridin-2-yl
    547 F Br CH2 3,5-Dimethyl-4-methoxypyridin-2-yl
    548 Cl CN CH2 3,5-Dimethyl-4-methoxypyridin-2-yl
    549 Br CN CH2 3,5-Dimethyl-4-methoxypyridin-2-yl
    550 Cl Cl CH2 3,5-Dimethyl-4-bromopyridin-2-yl
    551 Br Cl CH2 3,5-Dimethyl-4-bromopyridin-2-yl
    552 Cl Br CH2 3,5-Dimethyl-4-bromopyridin-2-yl
    553 Br Br CH2 3,5-Dimethyl-4-bromopyridin-2-yl
    554 Cl N3 CH2 3,5-Dimethyl-4-bromopyridin-2-yl
    555 Br N3 CH2 3,5-Dimethyl-4-bromopyridin-2-yl
    556 F Cl CH2 3,5-Dimethyl-4-bromopyridin-2-yl
    557 F Br CH2 3,5-Dimethyl-4-bromopyridin-2-yl
    558 Cl CN CH2 3,5-Dimethyl-4-bromopyridin-2-yl
    559 Br CN CH2 3,5-Dimethyl-4-bromopyridin-2-yl
    560 Cl Cl CH2 3,5-Dimethyl-4-chloropyridin-2-yl
    561 Br Cl CH2 3,5-Dimethyl-4-chloropyridin-2-yl
    562 Cl Br CH2 3,5-Dimethyl-4-chloropyridin-2-yl
    563 Br Br CH2 3,5-Dimethyl-4-chloropyridin-2-yl
    564 Cl N3 CH2 3,5-Dimethyl-4-chloropyridin-2-yl
    565 Br N3 CH2 3,5-Dimethyl-4-chloropyridin-2-yl
    566 F Cl CH2 3,5-Dimethyl-4-chloropyridin-2-yl
    567 F Br CH2 3,5-Dimethyl-4-chloropyridin-2-yl
    568 Cl CN CH2 3,5-Dimethyl-4-chloropyridin-2-yl
    569 Br CN CH2 3,5-Dimethyl-4-chloropyridin-2-yl
    570 Cl Cl CH2 3,5-Dimethyl-4-iodopyridin-2-yl
    571 Br Cl CH2 3,5-Dimethyl-4-iodopyridin-2-yl
    572 Cl Br CH2 3,5-Dimethyl-4-iodopyridin-2-yl
    573 Br Br CH2 3,5-Dimethyl-4-iodopyridin-2-yl
    574 Cl N3 CH2 3,5-Dimethyl-4-iodopyridin-2-yl
    575 Br N3 CH2 3,5-Dimethyl-4-iodopyridin-2-yl
    576 F Cl CH2 3,5-Dimethyl-4-iodopyridin-2-yl
    577 F Br CH2 3,5-Dimethyl-4-iodopyridin-2-yl
    578 Cl CN CH2 3,5-Dimethyl-4-iodopyridin-2-yl
    579 Br CN CH2 3,5-Dimethyl-4-iodopyridin-2-yl
    580 Cl Cl CH2 4,5,6-Trimethoxy-3-chloropyridin-2-yl
    581 Br Cl CH2 4,5,6-Trimethoxy-3-chloropyridin-2-yl
    582 Cl Br CH2 4,5,6-Trimethoxy-3-chloropyridin-2-yl
    583 Br Br CH2 4,5,6-Trimethoxy-3-chloropyridin-2-yl
    584 Cl Cl CH2 4,5,6-Trimethoxy-3-bromopyridin-2-yl
    585 Br Cl CH2 4,5,6-Trimethoxy-3-bromopyridin-2-yl
    586 Cl Br CH2 4,5,6-Trimethoxy-3-bromopyridin-2-yl
    587 Br Br CH2 4,5,6-Trimethoxy-3-bromopyridin-2-yl
    588 7 Cl H CH2 3,5-Dimethyl-4-hydroxypyridin-2-yl
    589 8 Cl H CH2 3,5-Dimethyl-4-ethoxypyridin-2-yl
    590 9 Cl H CH2 3,5-Dimethyl-4-allyloxypyridin-2-yl
    591 10 Cl H CH2 3,5-Dimethyl-4-(2-ethoxy-ethoxy)pyridin-2-yl
    592 11 Cl H CH2 3,5-Dimethyl-4isopropoxypyridin-2-yl
    593 12 Cl H CH2 3,5-Dimethyl-4-cyclopropylmethoxypyridin-2-yl
    594 13 Cl H CH2 3,5-Dimethyl-4-(3-methyl-butoxy)pyridin-2-yl
    595 14 Cl H CH2 3,5-Dimethyl-4-isobutoxypyridin-2-yl
    596 15 Cl H CH2 3,5-Dimethyl-4-(2-acetyloxy-ethoxy)pyridin-2-yl
    597 16 Cl H CH2 3,5-Dimethyl-4-(3-acetyloxy-propoxy)pyridin-2-yl
    598 17 Cl H CH2 3,5-Dimethyl-4-propoxypyridin-2-yl
    599 20 Cl H CH2 3,5-Dimethylpyridin-2-yl
    600 32 Cl H CH2 3,5-Dimethyl-4-methoxy-1-methoxypyridin-2-yl
    601 34 Cl H CH2 5-Methoxy-4-methoxymethyl-6-methylpyridin-3-yl
    602 35 Cl H CH2 5-Ethoxy-4-hydroxymethyl-6-methylpyridin-3-yl
    604 36 Cl H CH2 3,5-Dimethyl-4-aminopyridin-2-yl
    605 37 Cl H CH2 3-Methoxy-5-methoxymethyl-4-methylpyridin-2-yl
    606 38 Cl H CH2 5-Chloro-6-methoxypyridin-3-yl
    607 39 Cl H CH2 3,4-Dimethoxypyridin-2-yl
    608 40 Cl H CH2 3-Methoxy-6-methylpyridin-2-yl
    609 42 H H CH2 4-Methoxy-3,5-dimethylpyridin-2-yl
    610 45 Cl Br CH2 2-Bromo-3,5-dimethoxy-4-methylphenyl
    611 47 Cl Bu CH2 3,4,5-Trimethoxyphenyl
    612 49 Cl H CH2 2-Bromo-3,5-dimethoxy-4-acetyloxyphenyl
    613 50 Cl H CH2 2-Bromo-3,5-dimethoxy-4-hydroxyphenyl
    614 52 Cl H CH2 2-Bromo-3,5-dimethoxy-4-allyloxyphenyl
    615 53 Cl H CH2 2-Bromo-3,5-dimethoxy-4-chloromethyloxyphenyl
    616 54 Cl H CH2 2-Bromo-3,5-dimethoxy-4-chloroethyloxyphenyl
    617 55 Cl H CH2 2-Bromo-3,5-dimethoxy-4-cyclopropylmethoxyphenyl
    618 56 Cl H CH2 2-Bromo-3,5-dimethoxy-4-ethoxyphenyl
    619 57 Cl H CH2 2-Bromo-3,5-dimethoxy-4-propoxyphenyl
    620 58 Cl H CH2 2-Bromo-3,5-dimethoxy-4-butoxyphenyl
    621 59 Cl H CH2 3-Methoxymethyloxy-6-imethylpyridin-2-yl
    622 60 Cl H CH2 3H-Benzothiazole-2-thione
    623 61 Cl H CH2 2,5-Dimethylphenyl
    624 62 Cl H CH2 Isoquinolin-1-yl
    625 63 Cl H CH2 Benzo[1,2,5]thiadiazol-5-yl
    626 64 Cl H CH2 1-Methyl-1H-benzotriazol-5-yl
    627 65 Cl H CH2 6-Chloro-benzo[1,2,5]thiadiazol-5-yl
    628 66 Cl H CH2 Benzo[1,2,5]thiadiazol-4-yl
    629 67 Cl H CH2 6-Fluoro-4a,8a-dihydro-4H-benzo[1,3]dioxin-8-yl
    630 68 Cl H CH2 2-Methoxy-4-acetylphenyl
    631 69 Cl H CH2 3-Trifluoromethoxyphenyl
    632 70 Cl H CH2 2-Fluoro-3-trifluoromethylphenyl
    633 71 Cl H CH2 2-Fluoro-4,5-dimethoxyphenyl
    634 72 Cl H CH2 2,3-Dimethoxyphenyl
    635 73 Cl H CH2 3,4-Dimethoxyphenyl
    636 74 Me H CH2 2-Chloro-3,4,5-trimethoxyphenyl
    637 75 Cl H CH2 2-Chloro-4,5-dimethoxyphenyl
    638 76 Cl H CH2 2-Iodo-4,5-dimethoxyphenyl
    639 78 Cl H CH2 6-Chloro-benzo[1,3]dioxol-5-yl
    640 79 Cl H CH2 2,4-Dimethoxy-3-methylphenyl
    641 80 Cl H CH2 2-Chloro-3,4-dimethoxyphenyl
    642 81 Cl H CH2 3-Methoxyphenyl
    643 82 Cl H CH2 2,6-Dibromo-3,5-dimethoxyphenyl
    644 83 Cl H CH2 2-Bromo-3,5-dimethoxyphenyl
    645 84 Cl H CH2 3,5-Dimethoxyphenyl
    646 86 Cl H CH2 2,5-Dimethoxyphenyl
    647 87 Cl Br CH2 2,5-Dimethoxyphenyl
    648 88 Cl H CH2 2-Nitro-4,5-dimethoxyphenyl
    649 89 Cl Br CH2 2-Nitro-4,5-dimethoxyphenyl
    650 90 Cl H CH2 2,5-Dichlorophenyl
    651 91 Cl H CH2 2,3,5-Trifluorophenyl
    652 92 Cl H C(O) 3,4,5-Trimethoxyphenyl
    653 95 Cl H CH2 3,5-Dichlorophenyl
    654 96 Cl H CH2 3,4-Dichlorophenyl
    655 97 Cl Br CH2 3,4-Dichlorophenyl
    656 99 Cl H CH2 2,6-Dichloro-3,4,5-trimethoxyphenyl
    657 100 OH H CH2 2-Chloro-3,4,5-trimethoxyphenyl
    658 103 SEt H CH2 2-Bromo-3,4,5-trimethoxyphenyl
    659 104 OMe H CH2 2-Bromo-3,4,5-trimethoxyphenyl
    660 105 NH2 H CH2 2-Bromo-3,4,5-trimethoxyphenyl
    661 107 Cl H CH2 2-Bromo-3,5-dimethoxy-4-methoxymethyloxyphenyl
    662 108 Cl H CH2 Benzothiazol-2-yl
    663 109 Cl H CH2 4-Methoxyphenyl
    664 110 Cl H CH2 2-Bromo-4,5-dimethoxyphenyl
    665 112 Cl H CH2 4-Methylquinolin-2-yl
    666 113 Br H CH2 4-Methylquinolin-2-yl
    667 114 Br H CH2 4-Methyl-1-oxy-quinolin-2-yl
    668 116 Cl H CH2 6-Chloro-benzothiazol-2-yl
    669 Cl H CH2 4,5,6-Trimethoxy-pyridin-3-yl
    670 Cl H CH2 4,5,6-Trimethoxy-1-oxy-pyridin-3-yl
    671 Cl H CH2 2-Bromo-4,5,6-trimethoxy-pyridin-3-yl
    672 Cl H CH2 2-Chloro-4,5,6-trimethoxy-pyridin-3-yl
    673 Cl H CH2 2-Chloro-4,5,6-trimethoxy-1-oxy-pyridin-3-yl
    674 Cl H CH2 2-Bromo-4,5,6-trimethoxy-1-oxy-pyridin-3-yl
    675 Cl H CH2 4,5,6-Trimethyl-pyridin-3-yl
    676 Cl H CH2 4,5,6-Trimethyl-1-oxy-pyridin-3-yl
    677 Cl H CH2 2-Bromo-4,5,6-trimethyl-pyridin-3-yl
    678 Cl H CH2 2-Chloro-4,5,6-trimethyl-pyridin-3-yl
    679 Cl H CH2 2-Chloro-4,5,6-trimethyl-1-oxy-pyridin-3-yl
    680 Cl H CH2 2-Bromo-4,5,6-trimethyl-1-oxy-pyridin-3-yl
    681 Cl H CH2 2-Iodo-4,5,6-trimethyl-pyridin-3-yl
    682 Cl H CH2 2-Iodo-4,5,6-trimethyl-pyridin-3-yl
    683 Br H CH2 4,5,6-Trimethoxy-pyridin-3-yl
    684 Br H CH2 4,5,6-Trimethoxy-1-oxy-pyridin-3-yl
    685 Br H CH2 2-Bromo-4,5,6-trimethoxy-pyridin-3-yl
    686 Br H CH2 2-Chloro-4,5,6-trimethoxy-pyridin-3-yl
    687 Br H CH2 2-Chloro-4,5,6-trimethoxy-1-oxy-pyridin-3-yl
    688 Br H CH2 2-Bromo-4,5,6-trimethoxy-1-oxy-pyridin-3-yl
    689 Br H CH2 4,5,6-Trimethyl-pyridin-3-yl
    690 Br H CH2 4,5,6-Trimethyl-1-oxy-pyridin-3-yl
    691 Br H CH2 2-Bromo-4,5,6-trimethyl-pyridin-3-yl
    692 Br H CH2 2-Chloro-4,5,6-trimethyl-pyridin-3-yl
    693 Br H CH2 2-Chloro-4,5,6-trimethyl-1-oxy-pyridin-3-yl
    694 Br H CH2 2-Bromo-4,5,6-trimethyl-1-oxy-pyridin-3-yl
    695 I H CH2 3,5-Dimethyl-4-methoxypyridin-2-yl
    696 I H CH2 3,5-Dimethyl-4-methoxy-1-oxypyridin-2-yl
    697 I H CH2 3-Bromo-4,5,6-trimethoxypyridin-2-yl
    698 I H CH2 4,5,6-Trimethoxypyridin-2-yl
    699 I H CH2 6-Chloro-3,5-dimethyl-4-methoxy-pyridin-2-yl
    700 I H CH2 3,5-dimethyl-4-thiomethyl-1-oxypyridin-2-yl
    701 I H CH2 3,5-Dimethyl-4-bromopyridin-2-yl
    702 I H CH2 3,5-Dimethyl-4-bromo-1-oxypyridin-2-yl
    703 I H CH2 3-Chloro-4,5,6-trimethoxypyridin-2-yl
    704 I H CH2 3,4,5-Trimethoxyphenyl
    705 I H CH2 2-Chloro-3,4,5-trimethoxyphenyl
    706 I H CH2 2-Bromo-3,4,5-trimethoxyphenyl
    707 I H CH2 2-Iodo-3,4,5-trimethoxyphenyl
    708 I H CH2 3,4,5-Trimethylpyridin-2-yl
    709 I H CH2 4,5,6-Trimethylpyridin-3-yl
    710 I H CH2 3,5-dimethyl-4-thiomethylpyridin-2-yl
    711 I H CH2 4,6-Dimethyl-5-methoxypyridin-3-yl
    712 I H CH2 4,6-Dimethyl-5-methoxy-1-oxypyridin-3-yl
    713 I H CH2 3,5-Dimethyl-4-chloropyridin-2-yl
    714 I H CH2 3,5-Dimethyl-4-iodopyridin-2-yl
  • compounds of interest in Table 1 are compounds 2, 3, 11, 44, 82, 83, 242, 243, 245, 248, 249, 254, 255, 260, 266, 272, 273, 278, 279, 284, 286, 287, 308, 309, 343, 348, 349, 354, 366, 367, 372, 373, 671 and 697 with those selected being 2, 242, 243, 248, 254, 260, 266, 272, 278, 284, 286, 287, 308, 343, 348, 349, 354, 372, 373, 671 and 697.
  • III. Synthesis of the Compounds of the Invention
  • Synthesis of compounds of the present invention may be accomplished by various methods the art, including those described in, for example, Montgomery, J. Med. Pharm. Chem., 1962, 5, 15-24; Sircar, U.S. Pat. No. 4,772,606 (1988); Sircar, U.S. Pat. No. 4,748,177 (1988); Hans, U.S. Pat. No. 5,110,818 (1992); Gillespie, WO 02/055521; Matsuda, JP 10025294 A2, (1998). For the synthesis of compounds of Formulae I and II, a general strategy is outlined in Scheme 1. It should be understood that other methods can also be used.
    Figure US20070185064A1-20070809-C00014
  • The starting materials or the intermediates of the Formula 2, or/and 4 can exist in tautomeric forms as shown in FIG. 1. Both forms are indiscriminately described in the specifications.
    Figure US20070185064A1-20070809-P00001
  • Method 1: From Purines:
  • The compounds of Formula I (see, Scheme 2) can be synthesized from the commercially available starting heterocycle, for example compounds of Formula 2 where R6 is —Cl, —Br or —OH, R7 is —NH2 and R8 is —H are commercially available from Aldrich, AlfaAesar, etc. Accordingly, Formula 2 can be alkylated in the presence of a base such as K2CO3, NaH, Cs2CO3, DBU etc. with/without the presence of halide such as NaI, KI, (Bu)3NI etc., and in a polar solvent such as DMF, THF, DMSO etc. using electrophiles such as L1—R4—R5 where —L1 is a leaving group. Leaving groups include but are not limited to, e.g., halogen, triflate, tosylate , mesylate etc.
    Figure US20070185064A1-20070809-C00015

    (See Kasibhatla, WO 03/037860) Compounds of Formula I, wherein R1 is —OH can be converted to halides using standard conditions POCl3, POBr3 etc. with/without the presence of base such as Et3N, N,N-diethylaniline, (i-Pr)2NEt etc. in polar solvents such as CH3CN, CH2Cl2 etc.
  • The compounds of Formula I wherein R1 is —OR11, —SR11, or —NHR8, where R11 is alkyl, R8 is hydrogen, lower alkyl, lower aryl, or —C(O)R9, where R9 is lower alkyl, lower aryl, lower heteroaryl, —NR10R10, or —OR11, where R10 is independently hydrogen or lower alkyl, can be prepared from compounds of Formula I wherein R1 is halogen by reacting with HOR11, HSR11 or NH2R8 in presence of a base such as K2CO3 or NaH and a polar solvent such as DMF or THF. Compounds of Formula I where R8 is —C(O)R9 can be prepared when R1 is hydroxyl by simple acylation.
  • Compounds of Formula I where R1 is alkyl can be prepared from compounds of Formula I where R1 is halogen and trialkyl aluminum or dialkyl zinc. See Holy, J. Med. Chem. 1999, 42, 2064.
  • R5 especially when it is aryl or heteroaryl, can be further modified as needed, for example by halogenation, nitration, palladium coupling of halogen, Friedel-Crafts alkylation/acylation, etc. or these modifications can also be done before alkylation, see Jerry March, Advanced Organic Chemistry. The heteroaromatic rings can also be oxidized to their corresponding N-oxides using various oxidizing agents such as H2O2, O3, MCPBA etc. in polar solvents such as CH2Cl2, CHCl3, CF3COOH etc. See Jerry March, Advanced Organic Chemistry, 4th edition, Chapter 19.
  • Compounds of Formula I where R3 is halogen, can be prepared from Formulae 1 or 2 using halogenating agents such as Br2, NBS, NCS, NIS etc. in polar solvents such as DMF, water, or suitable buffer solution. See Herdewijn, J. Med Chem. 1995, 38, 3838. Alternatively, compounds of Formula 2 where R8 is iodo can also be made using procedures known in the literature, e.g., Burger, J. Org. Chem. 2000, 65, 7825. These can be further modified as needed; for example, where R3 is —N3, or —CN by reacting with an azide such as NaN3, LiN3 etc. or cynide such as KCN or NaCN in polar solvents such as DMF, DMSO etc. See Halbfinger, J. Med. Chem. 1999, 42, 1625; Jacobson, J. Med. Chem. 1999, 42, 5325.
  • Method 2: From Pyrimidines:
  • Compounds of Formula I can also be prepared from the substituted pyrimidines of Formula 5 (see, Scheme 3). Accordingly, reaction of commercially available compounds of Formula 5, where R16 is hydrogen or —NO2, (see J. Chem. Soc. 1962, 4186,for the preparation of R16=—NO2 compound) with NH2—R4—R5 in solvents such as EtOH, BuOH etc. in the presence of organic
    Figure US20070185064A1-20070809-C00016

    bases such as Et3N, (i-Pr)2NEt etc. followed by nitrosation (when R16=—H) using nitrous acid, then reduction with sodium dithionite or Zn/HCOOH etc. of Formula 6 (R16=—NO or —NO2) to yield compounds of Formula 6 where R16 is —NH2. Condensation of Formula 6,where R16 is —NH2 using standard conditions such as triethylorthoformate, formic acid, cyanogenbromide etc. as described in J. Chem. Soc. 1963, 4186; Sircar, U.S. Pat. No. 4,748,177, 1988; and Dang, WO 98/39344,followed by reaction with POCl3 to give compounds of Formula I, Scheme 3. These compounds of Formula I can be further modified as necessary.
  • Similarly, compounds of Formula I can also be synthesized from Formula 7, 2-amino-4, 6-dichloro pyrimidine (see Scheme 4). Reaction of Formula 7 with NH2—R4—R5 in solvents such as EtOH, BuOH etc. in presence of organic bases such as Et3N, (i-Pr)2NEt, etc. followed by reaction with diazonium salt prepared from 4-chloroaniline
    Figure US20070185064A1-20070809-C00017

    and NaNO2 in aq. HCl to give compound Formula of 8,where R16 is azo-(4-chlorobenzene). Reduction of the azo compound with zinc in acetic acid to give compounds of Formula 8, where R16 is —NH2, see, Meier, U.S. Pat. No. 5,204,353, 1993. Condensation of these compounds using standard conditions such as triethylorthoformate, formic acid, cyanogenbromide etc. as described in J. Chem. Soc. 1963, 4186; Sircar, U.S. Pat. No. 4,748,177, 1988, and Dang, WO 98/39344, followed by reaction with POCl3 to give compounds of Formula I, Scheme 4. These compounds of Formula I can be further modified as necessary.
  • Likewise compounds of Formula 8 where R16 is NH2 can be made from the commercially available 2,5-diamino-4,6-dihydroxy pyrimidine as described in Daluge, U.S. Pat. No. 5,917,042, 1999 (see Scheme 5).
    Figure US20070185064A1-20070809-C00018

    Method 3: From Imidazoles:
  • Compounds of Formula I can also be prepared from the substituted imidazoles as shown in Scheme 6. Accordingly, compounds of Formula 4,wherein R14 is NH2, R13 is C(O)NH2 and R15 is H, can be alkylated in the presence of a base such as KOH, NaOH, K2CO3, NaH, Cs2CO3, DBU etc. with/without the presence of halide such as NaI, KI, (Bu)3NI etc., and in a polar solvent such as DMF, THF, DMSO etc. using electrophiles such as L1—R4—R5 where L1 is a leaving group. Leaving groups include but are not limited to, e.g., halogen, triflate, tosylate, mesylate etc. to give compounds of Formula 10. The ring closure can be achieved using many methods reported in the literature, Alhede, J. Org. Chem., 1991, 2139 and references cited therein to give guanines of Formula I, wherein R1 is OH. These compounds can be converted to the
    Figure US20070185064A1-20070809-C00019

    compounds of Formula I, wherein R1 is Cl using POCl3 as described earlier. Advantageously, these steps can be reversed as shown in Scheme 6 via Formula 11. Alternately, we can also construct these 2-aminopyrimidine rings from Formula 4, wherein R14 is —OH or halide, R13 is —C(O)OEt and R15 is —H by reacting with guanidine hydrochoride as described in Chowdhury, J. Med. Chem. 1999, 42, 4300.
    Preparation of Electrophiles L1—R4—R5 wherein L— is a Leaving Group and of Nucleophiles NH2—R4—R5
    Synthesis of Benzyl Type Electrophile:
    Figure US20070185064A1-20070809-P00002
  • The electrophiles can be prepared from the substituted benzene derivatives using various methods reported in the literature, see Jerry March, Advanced Organic Chemistry, 4th edition; Larock, Comprehensive Organic Transformations, 1989, VCH, New York. For example the L1 is —Br can be prepared by reduction followed by halogenation of the benzoic acid or aldehyde derivatives. These benzyl derivatives can also be prepared by benzylic oxidation or benzylic halogenation. Further modification of the benzyl ring can be done before or after the purine alkylation step, for example the halogenation was done in both ways. The corresponding amines where L1 is —NH2 can be prepared from the compounds where L1 is leaving group such as chloride, bromide, tosylate, mesylate etc. using ammonia.
  • Synthesis of Pyridyl Methyl Type Electrophile:
    Figure US20070185064A1-20070809-P00003
  • These compounds can be prepared from many methods reported in the literature. Morisawa et al. J. Med. Chem. 1974, 17, 1083; Klaus, W. et al. J. Med. Chem. 1992, 35, 438; Abramovitch, R. A.; Smith, E. M. “Pyridine-1-oxide in Pyridine and its Derivatives” in The Chemistry of Heterocyclic Compounds; Weissberger, A., Taylor, E. C., Eds.; John Wieley, New York, 1974, Pt. 2, pp 1-261; Jeromin, G. E. et al. Chem. Ber. 1987, 120, 649. Blanz, E. J., et al. J. Med. Chem. 1970, 13, 1124; Smith, Kline and French, EP 0184322, 1986; Abblard, J. et al. Bull. Soc. Chim. Fr. 1972, 2466; Fisher, B. E. et al. “The Structure of Isomaltol” J Org. Chem. 1964, 29, 776. De Cat, A. et al. Bull. Soc. Chim. Belg. 1965, 74, 270; Looker, J. H. et al. J. Org. Chem. 1979, 44, 3407. Ackerman, J. F. Ph.D. Dissertation, University of Notre Dame, June, 1949. These methods can be applied to the synthesis of quinoline and isoquinolines type compounds.
  • Further modification of the pyridyl ring can be done after the purine alkylation see Scheme 7.
    Figure US20070185064A1-20070809-C00020
    Figure US20070185064A1-20070809-C00021

    IV. Pharmaceutical Compositions, Dosing, and Modes of Administration
  • The present invention is directed to the clinical use of the heterocyclics, in particular, 2-aminopurine and related analogs of Formulae A, I and II, and their polymorphs, solvates, esters, tautomers, enantiomers, diastereomers, pharmaceutically acceptable salts and prodrugs thereof, for use in treatment or prevention of diseases that are HSP90-dependent. For example, a disorder such as inflammatory diseases, infections, autoimmune disorders, stroke, ischemia, cardiac disorders, neurological disorders, fibrogenetic disorders, proliferative disorders, tumors, leukemias, neoplasms, cancers, carcinomas, metabolic diseases, and malignant disease. The fibrogenetic disorders include but are not limited to scleroderma, polymyositis, systemic lupus, rheumatoid arthritis, liver cirrhosis, keloid formation, interstitial nephritis and pulmonary fibrosis.
  • The present invention features pharmaceutical compositions comprising the compound of Formulae A, I and II, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt thereof, or prodrug thereof, of any of the preceding aspect and embodiments and one or more pharmaceutical excipients.
  • Those of ordinary skill in the art are familiar with formulation and administration techniques that can be employed with the compounds and methods of the invention, e.g., as discussed in Goodman and Gilman, The Pharmacological Basis of Therapeutics, current ed.; Pergamon; and Remington's, Pharmaceutical Sciences (current edition), Mack Publishing Co., Easton, Pa.
  • The compounds utilized in the methods of the instant invention may be administered either alone or in combination with pharmaceutically acceptable carriers, excipients or diluents, in a pharmaceutical composition, according to standard pharmaceutical practice. The compounds can be administered orally or parenterally, including the intravenous, intramuscular, intraperitoneal, subcutaneous, rectal and topical routes of administration.
  • For example, the therapeutic or pharmaceutical compositions of the invention can be administered locally to the area in need of treatment. This may be achieved by, for example, but not limited to, local infusion during surgery, topical application, e.g., cream, ointment, injection, catheter, or implant, said implant made, e.g., out of a porous, non-porous, or gelatinous material, including membranes, such as sialastic membranes, or fibers. The administration can also be by direct injection at the site (or former site) of a tumor or neoplastic or pre-neoplastic tissue.
  • Still further, the compounds or compositions of the invention can be delivered in a vesicle, e.g., a liposome (see, for example, Langer, Science 1990, 249,1527-1533; Treat et al., Liposomes in the Therapy of Infectious Disease and Cancer, Lopez-Bernstein and Fidler, Ed., Liss, N.Y., pp. 353-365, 1989).
  • The compounds and pharmaceutical compositions used in the methods of the present invention can also be delivered in a controlled release system. In one embodiment, a pump may be used (see, Sefton, 1987, CRC Crit. Ref. Biomed. Eng. 14:201; Buchwald et al. Surgery, 1980 88, 507; Saudek et al. N. Engl. J. Med. 1989, 321, (574). Additionally, a controlled release system can be placed in proximity of the therapeutic target. (See, Goodson, Medical Applications of Controlled Release, 1984, Vol. 2, pp. 115-138).
  • The pharmaceutical compositions used in the methods of the instant invention can also contain the active ingredient in a form suitable for oral use, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs. Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions, and such compositions may contain one or more agents selected from the group consisting of sweetening agents, flavoring agents, coloring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations. Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets. These excipients may be, for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, such as microcrystalline cellulose, sodium crosscarmellose, corn starch, or alginic acid; binding agents, for example starch, gelatin, polyvinyl-pyrrolidone or acacia, and lubricating agents, for example, magnesium stearate, stearic acid or talc. The tablets may be un-coated or coated by known techniques to mask the taste of the drug or delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a water soluble taste masking material such as hydroxypropylmethyl-cellulose or hydroxypropylcellulose, or a time delay material such as ethyl cellulose, or cellulose acetate butyrate may be employed as appropriate.
  • Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water soluble carrier such as polyethyleneglycol or an oil medium, for example peanut oil, liquid paraffin, or olive oil.
  • Aqueous suspensions contain the active material in admixture with excipients suitable for the manufacture of aqueous suspensions. Such excipients are suspending agents, for example sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethyl-cellulose, sodium alginate, polyvinyl-pyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents may be a naturally-occurring phosphatide, for example lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethylene-oxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example polyethylene sorbitan monooleate. The aqueous suspensions may also contain one or more preservatives, for example ethyl, or n-propyl p-hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents, such as sucrose, saccharin or aspartame.
  • Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in mineral oil such as liquid paraffin. The oily suspensions may contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol. Sweetening agents such as those set forth above, and flavoring agents may be added to provide a palatable oral preparation. These compositions may be preserved by the addition of an anti-oxidant such as butylated hydroxyanisol or alpha-tocopherol.
  • Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients, for example sweetening, flavoring and coloring agents, may also be present. These compositions may be preserved by the addition of an anti-oxidant such as ascorbic acid.
  • The compounds and pharmaceutical compositions used in the methods of the instant invention may also be in the form of an oil-in-water emulsions. The oily phase may be a vegetable oil, for example olive oil or arachis oil, or a mineral oil, for example liquid paraffin or mixtures of these. Suitable emulsifying agents may be naturally-occurring phosphatides, for example soy bean lecithin, and esters or partial esters derived from fatty acids and hexitol anhydrides, for example sorbitan monooleate, and condensation products of the said partial esters with ethylene oxide, for example polyoxyethylene sorbitan monooleate. The emulsions may also contain sweetening agents, flavoring agents, preservatives and antioxidants.
  • Syrups and elixirs may be formulated with sweetening agents, for example glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative, flavoring and coloring agents and antioxidant.
  • The pharmaceutical compositions may be in the form of a sterile injectable aqueous solution. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • The sterile injectable preparation may also be a sterile injectable oil-in-water microemulsion where the active ingredient is dissolved in the oily phase. For example, the active ingredient may be first dissolved in a mixture of soybean oil and lecithin. The oil solution then introduced into a water and glycerol mixture and processed to form a microemulsion.
  • The injectable solutions or microemulsions may be introduced into a patient's blood-stream by local bolus injection. Alternatively, it may be advantageous to administer the solution or microemulsion in such a way as to maintain a constant circulating concentration of the instant compound. In order to maintain such a constant concentration, a continuous intravenous delivery device may be utilized. An example of such a device is the Deltec CADD-PLUS™ model 5400 intravenous pump.
  • The pharmaceutical compositions may be in the form of a sterile injectable aqueous or oleagenous suspension for intramuscular and subcutaneous administration. This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example as a solution in 1,3-butane diol. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid find use in the preparation of injectables.
  • The compounds of the present invention used in the methods of the present invention may also be administered in the form of suppositories for rectal administration of the drug. These compositions can be prepared by mixing the inhibitors with a suitable non-irritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug. Such materials include cocoa butter, glycerinated gelatin, hydrogenated vegetable oils, mixtures of polyethylene glycols of various molecular weights and fatty acid esters of polyethylene glycol.
  • For topical use, creams, ointments, jellies, solutions or suspensions, etc., containing a compound or composition of the invention can be used. As used herein, topical application can include mouth washes and gargles.
  • The compounds used in the methods of the present invention can be administered in intranasal form via topical use of suitable intranasal vehicles and delivery devices, or via transdermal routes, using those forms of transdermal skin patches well known to those of ordinary skill in the art. To be administered in the form of a transdermal delivery system, the dosage administration will, of course, be continuous rather than intermittent throughout the dosage regimen.
  • The methods, compounds and compositions of the instant invention may also be used in conjunction with other well known therapeutic agents that are selected for their particular usefulness against the condition that is being treated. For example, the instant compounds may be useful in combination with known anti-cancer and cytotoxic agents. Further, the instant methods and compounds may also be useful in combination with other inhibitors of parts of the signaling pathway that links cell surface growth factor receptors to nuclear signals initiating cellular proliferation.
  • The methods of the present invention may also be useful with other agents that inhibit angiogenesis and thereby inhibit the growth and invasiveness of tumor cells, including, but not limited to VEGF receptor inhibitors, including ribozymes and antisense targeted to VEGF receptors, angiostatin and endostatin.
  • Examples of antineoplastic agents that can be used in combination with the compounds and methods of the present invention include, in general, and as appropriate, alkylating agents, anti-metabolites, epidophyllotoxins, an antineoplastic enzyme, a topoisomerase inhibitor, procarbazine, mitoxantrone, platinum coordination complexes, biological response modifiers and growth inhibitors, hormonal/anti-hormonal therapeutic agents and haematopoietic growth factors. Exemplary classes of antineoplastic include the anthracyclines, vinca drugs, mitomycins, bleomycins, cytotoxic nucleosides, epothilones, discodermolide, pteridines, diynenes and podophyllotoxins. Particularly useful members of those classes include, for example, carminomycin, daunorubicin, aminopterin, methotrexate, methopterin, dichloromethotrexate, mitomycin C, porfiromycin, 5-fluorouracil, 6-mercaptopurine, gemcitabine, cytosine arabinoside, podophyllotoxin or podo-phyllotoxin derivatives such as etoposide, etoposide phosphate or teniposide, melphalan, vinblastine, vincristine, leurosidine, vindesine, leurosine, paclitaxel and the like. Other useful antineoplastic agents include estramustine, carboplatin, cyclophosphamide, bleomycin, gemcitibine, ifosamide, melphalan, hexamethyl melamine, thiotepa, cytarabin, idatrexate, trimetrexate, dacarbazine, L-asparaginase, canptothecin, CPT-11, topotecan, ara-C, bicalutamide, flutamide, leuprolide, pyridobenzoindole derivatives, interferons and interleukins.
  • When a compound or composition of the invention is administered into a human subject, the daily dosage will normally be determined by the prescribing physician with the dosage generally varying according to the age, weight, and response of the individual patient, as well as the severity of the patient's symptoms.
  • In one exemplary application, a suitable amount of compound is administered to a mammal undergoing treatment for cancer, for example, breast cancer. Administration typically occurs in an anount of between about 0.01 mg/kg of body weight to about 100 mg/kg of body weight per day (administered in single or divided doses), more preferably at least about 0.1 mg/kg of body weight per day. A particular therapeutic dosage can include, e.g., from about 0.01 mg to about 1000 mg of compound, and preferably includes, e.g., from about 1 mg to about 1000 mg. The quantity of active compound in a unit dose of preparation may be varied or adjusted from about 0.1 mg to 1000 mg, preferably from about 1 mg to 300 mg, more preferably 10 mg to 200 mg, according to the particular application. The amount administered will vary depending on the particular IC50 value of the compound used and the judgment of the attending clinician taking into consideration factors such as health, weight, and age. In combinational applications in which the compound is not the sole active ingredient, it may be possible to administer lesser amounts of compound and still have therapeutic or prophylactic effect.
  • Preferably, the pharmaceutical preparation is in unit dosage form. In such form, the preparation is subdivided into unit doses containing appropriate quantities of the active component, e.g., an effective amount to achieve the desired purpose.
  • The actual dosage employed may be varied depending upon the requirements of the patient and the severity of the condition being treated. Determination of the proper dosage for a particular situation is within the skill of 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 amounts 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 amount and frequency of administration of the compounds and compositions of the present invention used in the methods of the present invention, and if applicable other chemotherapeutic agents and/or radiation therapy, will be regulated according to the judgment of the attending clinician (physician) considering such factors as age, condition and size of the patient as well as severity of the disease being treated.
  • The chemotherapeutic agent and/or radiation therapy can be administered according to therapeutic protocols well known in the art. It will be apparent to those skilled in the art that the administration of the chemotherapeutic agent and/or radiation therapy can be varied depending on the disease being treated and the known effects of the chemotherapeutic agent and/or radiation therapy on that disease. Also, in accordance with the knowledge of the skilled clinician, the therapeutic protocols (e.g., dosage amounts and times of administration) can be varied in view of the observed effects of the administered therapeutic agents (i.e., antineoplastic agent or radiation) on the patient, and in view of the observed responses of the disease to the administered therapeutic agents.
  • Also, in general, the compounds of the invention need not be administered in the same pharmaceutical composition as a chemotherapeutic agent, and may, because of different physical and chemical characteristics, be administered by a different route. For example, the compounds/compositions may be administered orally to generate and maintain good blood levels thereof, while the chemotherapeutic agent may be administered intravenously. The determination of the mode of administration and the advisability of administration, where possible, in the same pharmaceutical composition, is well within the knowledge of the skilled clinician. The initial administration can be made according to established protocols known in the art, and then, based upon the observed effects, the dosage, modes of administration and times of administration can be modified by the skilled clinician.
  • The particular choice of compound (and where appropriate, chemotherapeutic agent and/or radiation) will depend upon the diagnosis of the attending physicians and their judgment of the condition of the patient and the appropriate treatment protocol.
  • The compounds/compositions of the invention (and where appropriate chemotherapeutic agent and/or radiation) may be administered concurrently (e.g., simultaneously, essentially simultaneously or within the same treatment protocol) or sequentially, depending upon the nature of the proliferative disease, the condition of the patient, and the actual choice of chemotherapeutic agent and/or radiation to be administered in conjunction (i.e., within a single treatment protocol) with the compound/composition.
  • In combinational applications and uses, the compound/composition and the chemotherapeutic agent and/or radiation need not be administered simultaneously or essentially simultaneously, and the initial order of administration of the compound/composition, and the chemotherapeutic agent and/or radiation, may not be important. Thus, the compounds/compositions of the invention may be administered first followed by the administration of the chemotherapeutic agent and/or radiation; or the chemotherapeutic agent and/or radiation may be administered first followed by the administration of the compounds/compositions of the invention. This alternate administration may be repeated during a single treatment protocol. The determination of the order of administration, and the number of repetitions of administration of each therapeutic agent during a treatment protocol, is well within the knowledge of the skilled physician after evaluation of the disease being treated and the condition of the patient. For example, the chemotherapeutic agent and/or radiation may be administered first, especially if it is a cytotoxic agent, and then the treatment continued with the administration of the compounds/compositions of the invention followed, where determined advantageous, by the administration of the chemotherapeutic agent and/or radiation, and so on until the treatment protocol is complete.
  • Thus, in accordance with experience and knowledge, the practicing physician can modify each protocol for the administration of a compound/composition for treatment according to the individual patient's needs, as the treatment proceeds.
  • The attending clinician, in judging whether treatment is effective at the dosage administered, will consider the general well-being of the patient as well as more definite signs such as relief of disease-related symptoms, inhibition of tumor growth, actual shrinkage of the tumor, or inhibition of metastasis. Size of the tumor can be measured by standard methods such as radiological studies, e.g., CAT or MRI scan, and successive measurements can be used to judge whether or not growth of the tumor has been retarded or even reversed. Relief of disease-related symptoms such as pain, and improvement in overall condition can also be used to help judge effectiveness of treatment.
  • V. Assays for Determining HSP90 Binding and Downstream Effect
  • A variety of in vitro and in vivo assays are available to test the effect of the compounds of the invention on HSP90. HSP90 competitive binding assays and functional assays can be performed as known in the art substituting in the compounds of the invention. Chiosis et al. Chemistry & Biology 2001, 8, 289-299, describe some of the known ways in which this can be done. For example, competition binding assays using, e.g., geldanamycin or 17-AAG as a competitive binding inhibitor of HSP90 can be used to determine relative HSP90 affinity of the compounds of the invention by immobilizing the compound of interest or other competitive inhibitor on a gel or solid matrix, preincubating HSP90 with the other inhibitor, passing the preincubated mix over the gel or matrix, and then measuring the amount of HSP90 that retains or does not retain on the gel or matrix.
  • Downstream effects can also be evaluated based on the known effect of HSP90 inhibition on function and stability of various steroid receptors and signaling proteins including, e.g., Raf1 and HER2. Compounds of the present invention induce dose-dependent degradation of these molecules, which can be measured using standard techniques. Inhibition of HSP90 also results in up-regulation of HSP90 and related chaperone proteins that can similarly be measured. Antiproliferative activity on various cancer cell lines can also be measured, as can morphological and functional differentiation related to HSP90 inhibition.
  • Many different types of methods are known in the art for determining protein concentrations and measuring or predicting the level of proteins within cells and in fluid samples. Indirect techniques include nucleic acid hybridization and amplification using, e.g., polymerase chain reaction (PCR). These techniques are known to the person of skill and are discussed, e.g., in Sambrook, Fritsch & Maniatis Molecular Cloning: A Laboratory Manual, 2nd ed., Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y., 1989; Ausubel, et al. Current Protocols in Molecular Biology, John Wiley & Sons, NY, 1994, and, as specifically applied to the quantification, detection, and relative activity of HER2/Neu in patient samples, e.g., in U.S. Pat. No. Nos. 4,699,877, 4,918,162, 4,968,603, and 5,846,749. A brief discussion of two generic techniques that can be used follows.
  • The determination of whether cells overexpress or contain elevated levels of HER2 can be determined using well known antibody techniques such as immunoblotting, radioimmunoassays, western blotting, immunoprecipitation, enzyme-linked immunosorbant assays (ELISA), and derivative techniques that make use of antibodies directed against HER2. As an example, HER2 expression in breast cancer cells can be determined with the use of an immunohistochemical assay, such as the Dako Hercep™ test (Dako Corp., Carpinteria, Calif.). The Hercep™ test is an antibody staining assay designed to detect HER2 overexpression in tumor tissue specimens. This particular assay grades HER2 expression into four levels: 0, 1, 2, and 3, with level 3 representing the highest level of HER2 expression. Accurate quantitation can be enhanced by employing an Automated Cellular Imaging System (ACIS) as described, e.g., by Press, M. et al. Modern Pathology 2000, 13, 225A.
  • Antibodies, polyclonal or monoclonal, can be purchased from a variety of commercial suppliers, or may be manufactured using well-known methods, e.g., as described in Harlow et al. Antibodies: A Laboratory Manual, 2nd ed; Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y., 1988.
  • HER2 overexpression can also be determined at the nucleic acid level since there is a reported high correlation between overexpression of the HER2 protein and amplification of the gene that codes for it. One way to test this is by using RT-PCR. The genomic and cDNA sequences for HER2 are known. Specific DNA primers can be generated using standard, well-known techniques, and can then be used to amplify template already present in the cell. An example of this is described in Kurokawa, H. et al. Cancer Res. 2000, 60, 5887-5894. PCR can be standardized such that quantitative differences are observed as between normal and abnormal cells, e.g., cancerous and noncancerous cells. Well known methods employing, e.g., densitometry, can be used to quantitate and/or compare nucleic acid levels amplified using PCR.
  • Similarly, fluorescent in situ hybridization (FISH) assays and other assays can be used, e.g., Northern and/or Southern blotting. These rely on nucleic acid hybridization between the HER2 gene or mRNA and a corresponding nucleic acid probe that can be designed in the same or a similar way as for PCR primers, above. See, e.g., Mitchell MS, and Press M. F. Oncol, Suppl. 1999, 12, 108-116. For FISH, this nucleic acid probe can be conjugated to a fluorescent molecule, e.g., fluorescein and/or rhodamine, that preferably does not interfere with hybridization, and which fluorescence can later be measured following hybridization. See, e.g., Kurokawa, H et al, Cancer Res. 2000, 60, 5887-5894 (describing a specific nucleic acid probe having sequence 5′-FAM-NucleicAcid-TAMRA-p-3′ sequence). ACIS-based approaches as described above can be employed to make the assay more quantitative (de la Torre-Bueno, J., et al. Modern Pathology 2000, 13, 221A).
  • Immuno and nucleic acid detection can also be directed against proteins other than HSP90 and HER2, which proteins are nevertheless affected in response to HSP90 inhibition.
  • The following examples are offered by way of illustration only and are not intended to be limiting of the full scope and spirit of the invention.
  • EXAMPLES
  • I. Materials and Methods
  • The chemical reagents used to create the novel products of the invention below are all available commercially, e.g., from Aldrich Chemical Co., Milwaukee, Wis., USA. Otherwise their preparation is facile and known to one of ordinary skill in the art, or it is referenced or described herein.
  • The final compounds were usually purified by preparative TLC (silica gel 60 Å, Whatman Partisil PK6F) or flash chromatography (silica gel 60 Å, EMD) using EtOAc/hexane or MeOH/CH2Cl2 as eluents. Rf's were measured using sicila gel TLC plates (silica gel 60 Å, EMD).
  • 1H-NMR spectra were recorded on a Bruker Avance 400 MHz spectrometer. Analytical HPLC chromatograms were obtained using a C18 column (Agilent Zorbax 300SB-C18; 5 microns; 4.6 mm×150 mm). A gradient was applied between solvent A (0.1% TFA in H2O) and solvent B (0.5% TFA in CH3CN) increasing the proportion of A linearly from 5% (t=0) to 100% (t=7.00 min), with a constant flow rate of 1 mL/min. The samples were diluted to typically 0.1-1 mg/mL in MeOH or CH3CN and the injection volumes were typically 10 μL. The column was not heated, and UV detection was effected at 254 nm.
  • The chemical names were generated using the Beilstein Autonom 2.1 software.
  • 1. General Procedures to Manipulate the Purine Ring
  • General Procedure 1.1: Alkylation of Purines at N-9
  • A suspension of the purine (3 mmol), (hetero)aryl-CH2-halide (3 mmol, if needed it can be added in batches) and K2CO3 (3.3 mmol) in dry DMF (15 mL) was heated to 40-60° C. for 3 to 10 h. Work-up (EtOAc) and purification by preparative TLC or flash chromatography (EtOAc/hexane or MeOH/CH2Cl2) yielded the pure N-9 alkylated product.
  • General Procedure 1.2: Halogenation of Purines at C-8
  • A solution of purine in MeOH/THF/acetate buffer (1N in each AcOH and AcONa) was treated with Br2 (1.3-equiv. 1M in CHCl3) at room temperature (r.t.) for 16 h. Evaporation, work-up (EtOAc), drying (MgSO4) and flash chromatography afforded the desired 8-bromopurine.
  • General Procedure 1.3: Nucleophilic Substitution of Purines at C-6
  • For details, see J Med. Chem. 1999, 42, 2064-2086.
  • A) Sulfanyl derivatives: A suspension of 6-chloropurine and sodium or potassium thiolate in THF was heated to reflux for 6-24 hours. Work-up (EtOAc) and flash chromatography afforded the desired 6-sulfanylpurine.
  • B) Alkoxy derivatives: A suspension of 6-chloropurine and alkoxide in the appropriate alcohol was heated to reflux for 1-16 hours before quenching with water. Evaporation, work-up (EtOAc), and flash chromatography afforded the desired 6-alkoxypurine.
  • C) Amino derivatives: A solution of 6-chloropurine and alkylamine in MeOH was heated in a sealed tube to 100° C. for 16 hours. Evaporation, work-up (EtOAc), and flash chromatography afforded the desired 6-alkoxypurine.
  • General Procedure 1.4: Methylation of Purines at C-6
  • A suspension of 6-chloropurine (0.2 mmol) and tetrakis(triphenylphosphino)-palladium (0.02 mmol) in dry THF (3 mL) was treated with trimethylaluminum (2M in toluene, 0.45 mmol) under nitrogen. The resulting solution was heated to reflux for 3 h, cooled to r.t., diluted with toluene (5 mL), and quenched with methanol (0.5 mL) followed by ammonium chloride (1 mmol). The mixture was heated to reflux for 2 h and filtered while hot through a Celite plug. Evaporation and purification by prep TLC afforded the 6-methylpurine. See J. Med. Chem. 1999, 42, 2064-2086.
  • General Procedure 1.5: Reductive Dehalogenation of 6-halopurine
  • The 6-halo purine derivative was dissolved in acetic acid and a catalytic amount of 5% Pd/C was added, and the mixture stirred under H2 atmosphere (1 psi) at r.t. for 1 h., evaporation, and purification afforded the dehalogenated derivatives.
  • General Procedure 1.6: Acetylation of 2-amino-purines
  • The 2-amino purine derivative was dissolved in acetic anhydride, treated with a catalytic amount of concentrated sulfuric acid at r.t. for 1 hour. Work-up (EtOAc), evaporation, and purification afforded the 2-acetamido-pyridine.
  • 2. General Procedures to Manipulate the Pyridine Ring
  • General Procedure 2.1: Preparation of Pyridine N-oxides
  • A solution of the pyridine derivative (1.0 mmol) in dichloromethane or chloroform (5 mL) was cooled by means of an ice-bath, treated with m-CPBA (1.1 to 3 mmol) in three portions, and allowed to warm to r.t. The mixture was extracted with dichloromethane and washed with aqueous NaOH, followed by water. Drying (Na2SO4) and concentration afforded the pyridine N-oxide.
  • General Procedure 2.2: Preparation of 2-(Acetoxymethy)Pyridines
  • A solution of the 2-methyl pyridine N-oxide (1.0 mmol) in acetic anhydride (5 mmol) was heated to reflux for 0.5 h. Work-up (EtOAc), drying (MgSO4), evaporation and purification by preparative TLC or flash chromatography afforded the 2-(acetoxymethyl) pyridine.
  • General Procedure 2.3: Preparation of 2-(Hydroxymethyl)Pyridines
  • A suspension of 2-acetoxymethyl-pyridine derivative and solid K2CO3 in methanol was heated to 50° C. for 5-30 min. Evaporation, work-up (EtOAc), and drying (MgSO4) afforded the 2-(hydroxymethyl)pyridine.
  • General Procedure 2.4: Preparation of 2-(Chloromethyl)Pyridines
  • A suspension of 2-hydroxymethyl-pyridine (10 g) in POCl3 (30 mL) was stirred at 110° C. for 1.5 h. The resulting viscous oil was cooled to r.t. and poured onto ice water (500 g). The pH was adjusted to 10 with solid KOH. Work-up (CHCl3), drying (MgSO4) and evaporation gave the 2-(chloromethyl)pyridine, usually as a purple oil or solid, which was used without purification.
  • General Procedure 2.5: Preparation of 2-(Bromomethyl)Pyridines
  • A solution of 2-(hydroxymethyl)pyridine (1.0 mmol) and triphenyl phosphine (1.2 mmol) in dichloromethane or chloroform (5 mL) was cooled to 0° C. A solution of CBr4 (1.5 mmol) in dichloromethane or chloroform was added dropwise, and the resulting mixture was stirred at 0° C. for 0.5-1 h. Work up followed and purification by flash chromatography afforded the 2-(bromomethyl)pyridine.
  • General Procedure 2.6: O-alkylation of 3- or 4-hydroxypyridines
  • A mixture of hydroxypyridine, alkyl halide (1.1 equiv.), base (K2CO3, KOH or NaH 1.2-2 equiv.) and solvent (THF or DMF) was stirred at 23-80° C. for 5-30 min. Work-up (EtOAc), drying (Na2SO4) and evaporation gave the crude 4-(alkoxy)pyridine, which was purified by preparative TLC or flash chromatography.
  • General Procedure 2.7: Preparation of Salts.
  • Method 1: The free base (40 mmol) was heated in MeOH (1:6, 300 mL) until it dissolved. A solution of H3PO4 in MeOH (40 mmol) was added dropwise at r.t. The mixture was stirred for 10 min., and the solvent was evaporated to give the pyridinium phosphate as a glassy white solid. The following solvents could also be used: THF, EtOH, or i-PrOH.
  • Method 2: The free base (50 mmol) was heated in i-PrOH (6 L) until it dissolved. The solution was allowed to cool to r.t. and a solution of HCl in i-PrOH (75 mmol) was added slowly dropwise. The hydrochloride crystallized out of solution within a few minutes, and was collected by filtration, washed (acetone) and dried. The sulfate and mesylate salts were also prepared in this manner.
  • The hydrochloride salts can also be made by adding CH3COCl to the alcoholic solution of the pyridine or free base derivative.
  • Method 3: To a suspension of free base (5 mmol) in MeOH (50 mL) was added a solution of methane sulfonic acid in MeOH (75 mmol) was added slowly dropwise. The mixture became clear within a few minutes, upon addition of i-PrOH (50-100 mL) the salt precipitated out and was collected by filtration, washed with i-PrOH, ether and dried.
  • These methods can be applied to prepare all other salts.
  • 3. General Procedure to Manipulate Benzene Rings
  • General Procedure 3.1: Halogenation of Benzene Rings.
  • Variant 1: A solution of the aromatic compound in MeOH/THF/acetate buffer (1N in each AcOH and AcONa) was treated with Br2 (1.3 equiv) at r.t. for 5 min. The excess bromine and solvent were removed on a rotary evaporator. Work-up (CHCl3) and flash chromatography afforded the desired bromobenzene.
  • Variant 2: A solution of the aromatic compound (7 mmol) and N-halosuccinimide (NCS, NBS, or NIS, 1.06 equiv) in acetic acid (40 mL) was heated to 40-90° C. for 0.3-1 h. Evaporation, work-up (EtOAc) and flash chromatography afforded the desired halogenated benzene.
  • General Procedure 3.2: Preparation of Benzylic Alcohols
  • Benzoic acid derivatives were reduced to the corresponding benzylic alcohols according to the procedure given by Bhaskar et al. J. Org. Chem. 1991, 56, 5964-5965.
  • 4. SPECIFIC EXAMPLES Example 1 9-(4-Bromo-3,5-dimethyl-pyridin-2-yl)-6-chloro-9H-purin-2-ylamine
  • The title compound was obtained by alkylating 2-amino-6-chloropurine with 4-bromo-2-bromomethyl-3,5-dimethyl-pyridine according to the general procedure 1.1. HPLC Rt: 5.301 min.
  • 1H-NMR (CDCl3): δ 8.19 (s, 1H), 7.88 (s, 1H), 5.41 (s, 2H), 5.06 (s, 2H), 2.53 (s, 3H), 2.39 (s, 3H). m/z (%) 367.1 (M+1, 75%), 369.1 (M+3, 100%), 371.1 (M+5, 25%).
  • The alkylating agent, 4-bromo-2-bromomethyl-3,5-dimethyl-pyridine, could itself be prepared by any of the following three methods:
  • Method 1 Step 1: 2,3,5-Collidine-N-oxide
  • Oxidation of 2,3,5-collidine according to the general procedure 2.1 gave 2,3,5-collidine-N-oxide. Yield: 70%. HPLC Rt: 3.96 min. 1H-NMR (CDCl3): δ 8.03 (s, 1H), 6.90 (s, 1H), 2.47 (s, 3H), 2.31 (s, 3H), 2.24 (s, 3H). m/z (%) 138.2 (M+1, 100%). Rf (20% MeOH/EtOAc): 0.35.
  • Step 2: 4-Bromo-2,3,5-collidine-N-oxide
  • 2,3,5-collidine-N-oxide (1.3 g, 10 mmol) and K2CO3 (2.9 g, 20 mmol) were suspended in 10 mL of CCl4. Bromine (1 mL, 20 mmol) was added dropwise, and the reaction mixture was heated to reflux for 2 h. Work-up (EtOAc) and flash chromatography (10% MeOH/EtOAc) afforded the product as a solid (1.05 g, 51% yield). HPLC Rt: 5.239 min. 1H-NMR (CDCl3): δ 8.06 (s, 1H), 2.56 (s, 3H), 2.43 (s, 3H), 2.31 (s, 3H). m/z (%) 216.2 (M+1, 100%), 218.2 (M+3, 100%). Rf(20% MeOH/EtOAc): 0.45.
  • Step 3: Acetic acid 4-bromo-3,5-dimethyl-pyridin-2-yl methyl ester
  • 4-Bromo-2,3,5-collidine-N-oxide (0.25 g, 11 mmol) was dissolved in acetic anhydride (5 mL) and the solution was heated to reflux for 30 min. Work-up and flash chromatography (50% Hexane/EtOAc) afforded the product (0.27 g, 96% yield). Rf (50% Hexane/EtOAc): 0.70. HPLC Rt: 4.759 min. 1H-NMR (CDCl3): δ 8.26 (s, 1H), 5.27 (s, 2H), 2.46 (s, 3H), 2.41 (s, 3H), 2.14 (s, 3H).
  • Step 4: 4-Bromo-3,5-dimethyl-pyridin-2-yl methanol
  • A suspension of acetic acid 4-bromo-3,5-dimethyl-pyridin-2-yl methyl ester (0.26 g, 1.0 mmol) and K2CO3 (excess) in MeOH (5 mL) was heated to 50° C. for 15 min. Work-up (CHCl3), filtration through a silica gel pad (eluent: 100% EtOAc) and evaporation gave the title compound as a white solid (0.19 g, 88% yield). Rf (50% Hexane/EtOAc): 0.5. HPLC Rt: 3.80 min. 1H-NMR (CDCl3): δ 8.23 (s, 1H), 4.70 (s, 2H), 2.46 (s, 3H), 2.30 (s, 3H).
  • Step 5: 4-Bromo-2-bromomethyl-3,5-dimethyl-pyridine
  • The title compound was obtained from 4-bromo-3,5-dimethyl-pyridin-2-yl methanol according to the general procedure 2.5. HPLC Rt: 6.323 min. 1H-NMR (CDCl13): δ 8.22 (s, 1H), 4.63 (s, 2H), 2.52 (s, 3H), 2.40 (s, 3H).
  • Method 2 Step 1: 2-chloromethyl-3,5-dimethyl-pyridin-4-ol
  • The title compound was obtained by following the procedure described in the patent by Tarbit, et al. WO 99/10326.
  • Step 2: 4-bromo-2-chloromethyl-3,5-dimethyl pyridine
  • A neat mixture of 2-chloromethyl-3,5-dimethyl-pyridin-4-ol (8.2 g, 47.8 mmol) and POBr3 (60 g, 209 mmol) was stirred at 130° C. for 3 h. The resulting viscous oil was cooled to r.t. and poured onto ice water. The pH was adjusted to 10 with solid KOH. Work-up (CHCl3), drying (MgSO4) and evaporation afforded the title compound as a purple solid (8.7 g, 78% yield) which was used without purification. HPLC Rt: 6.028 min. 1H-NMR (CDCl3): 8.20 (s, 1H), 4.62 (s, 2H), 2.50 (s, 3H), 2.38 (s, 3H).
  • Method 3 Step 1: 4-bromo-2-chloromethyl-3,5-dimethyl pyridine
  • A suspension of 2-chloromethyl-4-methoxy-3,5-dimethyl-pyridine (3.24 g, 14.6 mmol) in PBr3 (8.0 ml, 85.1 mmol, 5.8 equiv.) was heated to 80° C. under nitrogen. A catalytic amount of DMF (0.50 ml, 6.4 mmol, 0.44 equiv.) was added, whereupon the suspension rapidly turned into an orange solution. After 40 min., the reaction was still incomplete as judged by HPLC. The temperature was raised to 110° C. and the reaction was prolonged for 30 min, at which point it was complete. The mixture was poured over ice, made basic with conc. aq. NH4OH and extracted into EtOAc. Washing with water, drying (brine, MgSO4) and concentration gave the title compound as a pink solid (1.51 g, 44%) containing 10% of an impurity by 1H-NMR. The crude was used without further purification. 1H-NMR (CDCl3) δ 8.19 (s, 1H), 4.59 (s, 2H), 2.48 (s, 3H), 2.37 (s, 3H).
  • Example 2 9-(4-bromo-3,5-dimethyl-pyridin-2-yl methyl)-6-chloro-9H-purin-2-ylamine, phosphate salt
  • The title compound was obtained by treating 9-(4-bromo-3,5-dimethyl-pyridin-2-yl methyl)-6-chloro-9H-purin-2-ylamine with H3PO4 according to the general procedure 2.7. HPLC Rt: 5.294 min. 1H-NMR (d6-DMSO): δ 8.12 (s, 1H), 8.09 (s, 1H), 6.83 (s, 2H), 5.47 (s, 2H), 2.49 (s, 3H), 2.29 (s, 3H)
  • Example 3 9-(4-bromo-3,5-dimethyl-pyridin-2-yl methyl)-6-chloro-9H-purin-2-ylamine, hydrochloric acid salt
  • The title compound was obtained by treating 9-(4-bromo-3,5-dimethyl-pyridin-2-yl methyl)-6-chloro-9H-purin-2-ylamine with HCl according to the general procedure 2.7. HPLC Rt: 5.294 min. 1H-NMR (d6-DMSO): δ 8.13 (s, 1H), 8.12 (s, 1H), 5.47 (s, 2H), 5.47 (s, 2H), 2.49 (s, 3H), 2.30 (s, 3H).
  • Example 4 9-(4-bromo-3,5-dimethyl-1-oxy-pyridin-2-yl methyl)-6-chloro-9H-purin-2-ylamine
  • The title compound was obtained by oxidation of 9-(4-bromo-3,5-dimethyl-pyridin-2-yl)-6-chloro-9H-purin-2-ylamine according to the general procedure 2.1. HPLC Rt: 4.916 min. 1H-NMR (CDCl3): δ 8.46 (s, 1H), 8.07 (s, 1H), 5.57 (s, 2H), 5.03 (s, 2H), 2.81 (s, 3H), 2.35 (s, 3H).
  • Example 5 6-bromo-9-(4-bromo-3,5-dimethyl-pyridin-2-yl methyl)-9H-purin-2-ylamine
  • A mixture of 6-bromo-9H-purin-2-ylamine (2.4 g, 11 mmol), 4-bromo-2-chloromethyl-3,5-dimethyl pyridine (3.5 g, 15 mmol), K2CO3 (2.07 g, 15 mmol) and DMF (50 mL) was stirred at 50° C. for 2 h. Work-up and flash chromatography gave the title compound as a white solid (2.6 g, 56%). HPLC Rt: 5.415 min. 1H-NMR (CDCl3): δ 8.17 (s, 1H), 7.88 (s, 1H), 5.38 (s, 2H), 5.05 (s, 2H), 2.51(s, 3H), 2.37 (s, 3H).
  • Example 6 6-bromo-9-(4-bromo-3,5-dimethyl-1-oxy-pyridin-2-yl methyl) -9H-purin-2-ylamine
  • The title compound was obtained by oxidation of 6-bromo-9-(4-bromo-3,5-dimethyl-pyridin-2-yl methyl) according to the general procedure 2.1 (52% yield). Rf (100% EtOAc): 0.1. HPLC Rt: 4.978 min. 1H-NMR (CDCl3): δ 8.47 (s, 1H), 8.07 (s, 1H), 5.56 (s, 2H), 5.06 (s, 2H), 2.81(s, 3H), 2.35 (s, 3H).
  • Example 7 2-(2-Amino-6-chloro-purin-9-ylmethyl)-3,5-dimethyl-pyridin-4-ol
  • The title compound was obtained by alkylating 6-chloro-9H-purin-2-ylamine with 2-chloromethyl-3,5-dimethylpyridin-4-ol according to the general procedure 1.1. HPLC Rt: 3.624 min. 1H-NMR (d6-DMSO): δ 8.07 (s, 1H), 7.47 (s, 1H), 6.90 (s, 2H), 5.20 (s, 2H), 2.00 (s, 3H), 1.86 (s, 3H).
  • Example 8 6-Chloro-9-(4-ethoxy-3,5-dimethyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine
  • The title compound was obtained by O-alkylation of 2-(2-amino-6-chloro-purin-9-ylmethyl)-3,5-dimethyl-pyridin-4-ol with ethyl iodide using the general procedure 2.6. HPLC Rt: 4.321 min. 1H-NMR (CDCl3): δ 8.21 (s, 1H), 7.90 (s, 1H), 5.34 (s, 2H), 5.12 (s, 2H), 3.90 (q, 2H), 2.31 (s, 3H) 2.26 (s, 3H), 1.44 (t, 3H).
  • Example 9 9-(4-Allyloxy-3,5-dimethyl-pyridin-2-ylmethyl)-6-chloro-9H-purin-2-ylamine
  • The title compound was obtained by O-alkylation of 2-(2-amino-6-chloro-purin-9-ylmethyl)-3,5-dimethyl-pyridin-4-ol with allyll chloride using the general procedure 2.6. HPLC Rt: 4.417 min. 1H-NMR (CDCl3): δ 8.20 (s, 1H), 7.90 (s, 1H), 6.03-6.10 (m, 1H), 5.40-5.44 (dd, 1H), 5.34 (s, 2H), 5.29-5.32 (dd, 1H), 5.19 (s, 2H), 4.34-4.36 (m, 2H), 2.30 (s, 3H) 2.25 (s, 3H).
  • Example 10 6-Chloro-9-[4-(2-ethoxy-ethoxy)-3,5-dimethyl-pyridin-2-ylmethyl]-9H-purin-2-ylamine
  • The title compound was obtained by O-alkylation of 2-(2-amino-6-chloro-purin-9-ylmethyl)-3,5-dimethyl-pyridin-4-ol with ethoxyethyl chloride, using the general procedure 2.6. HPLC Rt: 4.388 min. 1H-NMR (CDCl3): δ 8.19 (s, 1H), 7.89 (s, 1H), 5.33 (s, 2H), 5.14 (s, 2H), 3.97-4.00 (t, 2H), 3.73-3.76 (t, 2H), 3.56-3.61 (q, 2H), 2.33 (s, 3H), 2.26 (s, 3H), 1.22-1.26 (t, 3H).
  • Example 11 6-Chloro-9-(4-isopropoxy-3,5-dimethyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine
  • The title compound was obtained by 0-alkylation of 2-(2-amino-6-chloro-purin-9-ylmethyl)-3,5-dimethyl-pyridin-4-ol with isopropyl iodide using the general procedure 2.6. HPLC Rt: 4.571 min. 1H-NMR (CDCl3): δ 8.17 (s, 1H), 7.89 (s, 1H), 5.32 (s, 2H), 5.06 (s, 2H), 4.20 (m, 1H), 2.26 (s, 3H) 2.22 (s, 3H), 1.28-1.30 (d, 3H).
  • Example 12 6-Chloro-9-(4-cyclopropylmethoxy-3,5-dimethyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine
  • The title compound was obtained by O-alkylation of 2-(2-amino-6-chloro-purin-9-ylmethyl)-3,5-dimethyl-pyridin-4-ol with cyclopropylmethyl iodide using the general procedure 2.6. HPLC Rt: 4.709 min. 1H-NMR (CDCl3): δ 8.20 (s, 1H), 7.90 (s, 1H), 5.34 (s, 2H) 5.09 (s, 2H), 3.68-3.70 (d, 2H), 2.32(s, 3H) 2.27 (s, 3H), 1.23-1.31 (m, 1H), 0.63-0.68 (m, 2H), 0.30-0.33 (m, 2H).
  • Example 13 6-Chloro-9-[3,5-dimethyl-4-(3-methyl-butoxy)-pyridin-2-ylmethyl]-9H-purin-2-ylamine
  • The title compound was obtained by O-alkylation of 2-(2-amino-6-chloro-purin-9-ylmethyl)-3,5-dimethyl-pyridin-4-ol with 3-methylbutyl bromide using the general procedure 2.6. HPLC Rt: 5.425 min. 1H-NMR (CDCl3): δ 8.21 (s, 1H), 7.90 (s, 1H), 5.34 (s, 2H), 5.07 (s, 2H), 3.82-3.86 (t, 2H), 2.31 (s, 3H) 2.26 (s, 3H), 1.84-1.91 (m, 1H), 1.71-1.74 (q, 2H), 1.00 (s, 3H), 0.98 (s, 3H).
  • Example 14 6-Chloro-9-(4-isobutoxy-3,5-dimethyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine
  • The title compound was obtained by O-alkylation of 2-(2-amino-6-chloro-purin-9-ylmethyl)-3,5-dimethyl-pyridin-4-ol with isobutyl bromide using the general procedure 2.6. HPLC Rt: 4.321 min. 1H-NMR (CDCl3): δ 8.21 (s, 1H), 7.90 (s, 1H), 5.34 (s, 2H), 5.12 (s, 2H), 3.58-3.56 (d, 2H), 2.30 (s, 3H) 2.26 (s, 3H), 1.09 (s, 3H), 1.08 (s, 3H).
  • Example 15 Acetic acid 2-[2-(2-amino-6-chloro-purin-9-ylmethyl)-3,5-dimethyl-pyridin-4-yloxy]-ethyl ester
  • The title compound was obtained by O-alkylation of 2-(2-amino-6-chloro-purin-9-ylmethyl)-3,5-dimethyl-pyridin-4-ol with 2-bromoethyl acetate using the general procedure 2.6. HPLC Rt: 4.103 min. 1H-NMR (CDCl3): δ 8.21 (s, 1H), 7.90 (s, 1H), 5.35 (s, 2H), 5.10 (s, 2H), 4.42 (t, 2H), 4.05 (t, 2H), 2.34 (s, 3H) 2.27 (s, 3H), 2.13 (s, 3H).
  • Example 16 Acetic acid 3-[2-(2-amino-6-chloro-purin-9-ylmethyl)-3,5-dimethyl-pyridin-4-yloxy]-propyl ester
  • The title compound was obtained by O-alkylation of 2-(2-amino-6-chloro-purin-9-ylmethyl)3,5-dimethyl-pyridin-4-ol with 2-chloropropyl acetate the general procedure 2.6. HPLC Rt: 4.414 min. 1H-NMR (CDCl3): δ 8.21 (s, 1H), 7.91 (s, 1H), 5.34 (s, 2H), 5.12 (s, 2H), 4.34 (t, 2H), 3.90 (t, 2H), 2.31 (s, 3H), 2.25 (s, 3H), 2.15 (m, 1H), 2.09 (s, 3H).
  • Example 17 6-Chloro-9-(3,5-dimethyl-4-propoxy-pyridin-2-ylmethyl)-9H-purin-2-ylamine
  • The title compound was obtained by O-alkylation of 2-(2-amino-6-chloro-purin-9-ylmethyl)-3,5-dimethyl-pyridin-4-ol according to the general procedure 2.6. HPLC Rt: 4.644 min. 1H-NMR (CDCl3): δ 8.21 (s, 1H), 7.91 (s, 1H), 5.34 (s, 2H), 5.12 (s, 2H), 3.80-3.76 (t, 2H), 2.31 (s, 3H), 2.26 (s, 3H), 1.85 (m, 2H), 1.07-1.10 (t, 3H).
  • Example 18 6-Chloro-9-(4-chloro-3,5-dimethyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine Step 1: 4-Chloro-2-chloromethyl-3,5-dimethyl pyridine
  • The title compound was obtained by treating 2-chloromethyl-3,5-dimethyl-pyridin-4-ol with POCl3 according to the general procedure 2.4 (74% yield). HPLC Rt: 5.543 min. 1H-NMR (CDCl3): 8.24 (s, 1H), 4.71 (s, 2H), 2.48 (s, 3H), 2.36 (s, 3H).
  • Step 2: 6-chloro-9-(4-chloro-3,5-dimethyl-1-oxy-pyridin-2-yl methyl) -9H-purin-2-ylamine
  • A mixture of 6-chloro-9H-purin-2-ylamine (7 g, 41 mmol), 4-chloro-2-chloromethyl-3,5-dimethyl pyridine (8.2 g, 43 mmol), K2CO3 (10 g, 72 mmol) and DMF (200 mL) was heated to 50° C. for 2 h. The reaction mixture was diluted with water (200 mL) and the resulting precipitate was collected by filtration, washed with water, and dried to give the title compound as a beige solid (11.7 g, 88% yield, 90% purity). HPLC Rt: 5.167 min. 1H-NMR (CDCl3): δ 8.24 (s, 1H), 7.90 (s, 1H), 5.40 (s, 2H), 5.07 (s, 2H), 2.49 (s, 3H), 2.37 (s, 3H).
  • Example 19 6-Chloro-9-(4-chloro-3,5-dimethyl-1-oxy-pyridin-2-yl methyl) -9H-purin-2-ylamine
  • The title compound was obtained by oxidation of 6-chloro-9-(4-chloro-3,5-dimethyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine according to the general procedure 2.1 (56% yield). HPLC Rt: 4.813 min. 1H-NMR (d6-DMSO): δ 8.31 (s, 1H), 8.20 (s, 1H), 6.91 (s, 2H), 5.41 (s, 2H), 2.73 (s, 3H), 2.26 (s, 3H).
  • Examples 20 6-Chloro-9-(3,5-dimethyl-pyridin-2-yl methyl)-9H-purin-2-ylamine Step 1: Acetic acid 3,5-dimethyl-pyridin-2-yl methyl ester
  • The title compound was prepared from 2,3,5-collidine-N-oxide (see example 1) according to the general procedure 2.2. HPLC Rt: 2.916 min. 1H-NMR (CDCl3): δ 8.30 (s, 1H), 7.33 (s, 1H), 5.22 (s, 2H), 2.34 (s, 3H), 2.32 (s, 3H), 2.13 (s, 3H).
  • Step 2: 3,5-Dimethyl-pyridin-2-yl methanol
  • The title compound was obtained by deacetylation of acetic acid 3,5-dimethyl-pyridin-2-yl methyl ester according to the general procedure 2.3. HPLC Rt: 2.909 min. 1H-NMR (CDCl3): δ 8.24 (s, 1H), 7.30 (s, 1H), 4.85 (broad s, 1H), 4.67 (s, 2H), 2.33 (s, 3H), 2.20 (s, 3H).
  • Step 3: 2-Bromomethyl-3,5-dimethyl pyridine
  • The title compound was obtained from 3,5-dimethyl-pyridin-2-yl methanol according to the general procedure 2.5. HPLC Rt: 3.895 min. 1H-NMR (CDCl3): δ 8.3 (s, 1H), 7.3 (s, 1H), 4.61 (s, 2H), 2.41 (s, 3H), 2.33 (s, 3H).
  • Step 4: 6-Chloro-9-(3,5-dimethyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 2-bromomethyl-3,5-dimethyl pyridine according to the general procedure 1.1. HPLC Rt: 3.760 min. 1H-NMR (CDCl3): δ 8.21 (s, 1H), 7.89 (s, 1H), 7.30 (s, 1H), 5.32 (s, 2H), 5.05 (s, 2H), 2.36 (s, 3H), 2.29 (s, 3H).
  • Example 21 6-Bromo-9-(4-methoxy-3,5-dimethyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 6-bromo-9H-purin-2-ylamine with 2-chloromethyl-4-methoxy-3,5-dimethylpyridine according to the general procedure 1.1. PLC Rt: 4.138. 1H-NMR (CDCl3): δ 8.21 (s, 1H), 7.91 (s, 1H), 5.34 (s, 2H), 5.12 (s, 2H), 3.77 (s, 3H), 2.32 (s, 3H), 2.27 (s, 3H).). m/z (%) 363.2 (M+1, 100%), 365.2 (M+3, 100%).
  • Example 22 6-Bromo-9-(4-methoxy-3,5-dimethyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine, phosphate salt
  • The title compound was obtained by treating 6-bromo-9-(4-methoxy-3,5-dimethyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine with H3PO4 according to the general procedure 2.7. HPLC Rt: 4.138. 1H-NMR (d6-DMSO): δ 8.07 (s, 1H), 8.02 (s, 1H), 6.84 (s, 2H), 5.35 (s, 2H), 3.73 (s, 3H), 2.29 (s, 3H), 2.15 (s, 3H). ). m/z (%) 363.2(M+1, 100%), 365.2 (M+3, 100%).
  • Example 23 6-Bromo-9-(4-methoxy-3,5-dimethyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine, hydrochloride salt
  • The title compound was obtained by treating 6-bromo-9-(4-methoxy-3,5-dimethyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine with HCl according to the general procedure 2.7. HPLC Rt: 4.138. 1H-NMR (d6-DMSO): δ 8.44 (s, 1H), 8.26 (s, 1H), 5.57 (s, 2H), 3.96 (s, 3H), 2.35 (s, 3H), 2.34 (s, 3H).
  • Example 24 6-Bromo-9-(4-methoxy-3,5-dimethyl-1-oxy-pyridin-2-ylmethyl)-9H-purin-2-ylamine
  • The title compound was obtained by oxidation of 6-bromo-9-(4-methoxy-3,5-dimethyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine according to the general procedure 2.1. HPLC Rt: 4.439 min. 1H-NMR (CDCl3): δ 8.55 (s, 1H), 8.06 (s, 1H), 5.50 (s, 2H), 5.12 (s, 2H), 3.76 (s, 3H), 2.60 (s, 3H), 2.25 (s, 3H). m/z (%) 379.1 (M+1, 100%), 381.1 (M+3, 100%).
  • Example 25 6-Chloro-9-(4-methoxy-3,5-dimethyl-pyridin-2-ylmethyl) 9H-purin-2-ylamine Method 1
  • The title compound was obtained by alkylating 6-chloro-9H-purin-2-ylamine with 2-chloromethyl-4-methoxy-3,5-dimethylpyridine (or its HCl salt) according to the general procedure 1.1.
  • Method 2
  • The title compound could also be obtained by O-methylation of 6-chloro-9-(4-hydroxy-3,5-dimethyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine according to the procedure 2.6 (KOH, MeI, DMF, 80° C., 5 min). HPLC Rt: 3.980 min. 1H-NMR (CDCl3): δ 8.19 (s, 1H), 7.88 (s, 1H), 5.32 (s, 2H), 5.07 (s, 2H), 3.75 (s, 3H), 2.29 (s, 3H), 2.24 (s, 3H).
  • Method 3 Step 1: (4-Methoxy-3,5-dimethyl-pyridin-2-yl)-methylamine
  • A solution of 2-chloromethyl-4-methoxy-3,5-dimethyl-pyridine hydrochloride in 7N methanolic ammonia was placed in a pressure vessel and to 100° C. for 16 h. Concentration and flash chromatography gave the title compound as a greenish solid (76% yield). HPLC Rt: 3.773 min. 1H-NMR (CDCl3): δ 8.19 (s, 1H), 4.35 (s, 2H), 3.76 (s, 3H), 2.24 (s, 3H), 2.18 (s, 3H).
  • Step 2: 6-Chloro-4-(4-methoxy-3,5-dimethyl-pyridin-2-ylmethyl)-pyrimidine-2,4,5-triamine
  • A solution of 4,6-dichloro-pyrimidine-2,5-diamine (see Seela et al. Helv. Chim. Acta. 1986, 69, 1602-1613 and U.S. Pat. No. 5,917,042), 4-methoxy-3,5-dimethyl-pyridin-2-yl)-methylamine, and Et3N in butanol was heated to reflux for 1 h to give the title compound. HPLC Rt: 3.761 min. 1H-NMR (CDCl3): δ 8.24 (s, 1H), 7.15 (s, 1H), 4.60 (s, 2H), 4.56-4.55 (d, 2H), 3.80 (s, 3H), 3.00 (s, 2H), 2.28 (s, 3H), 2.27 (s, 3H).
  • Step 3: Synthesis of 6-chloro-9-(4-methoxy-3,5-dimethyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine
  • The cyclization with trimethylorthoformate in presence of acid to prepare purines can be done. See similar reaction, example 48.
  • Example 26 6-chloro-9-(4-methoxy-3,5-dimethyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine, phosphate salt
  • The title compound was obtained treating 6-chloro-9-(4-methoxy-3,5-dimethyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine with H3PO4 according to the general procedure 2.7. HPLC Rt: 4.003 min. 1H-NMR (d6-DMSO): δ 8.06 (s, 1H), 8.03 (s, 1H), 6.83 (s, 2H), 5.36 (s, 2H), 3.74 (s, 3H), 2.29 (s, 3H), 2.16 (s, 3H).
  • Example 27 6-Chloro-9-(4-methoxy-3,5-dimethyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine, sulphate salt
  • The title compound was obtained treating 6-chloro-9-(4-methoxy-3,5-dimethyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine with H2SO4 according to the general procedure 2.7. HPLC Rt: 3.999 min. 1H-NMR (d6-DMSO): δ 8.36 (s, 1H), 8.15 (s, 1H), 5.52 (s, 2H), 3.93 (s, 3H), 2.31 (s, 3H), 2.30 (s, 3H).
  • Example 28 6-Chloro-9-(4-methoxy-3,5-dimethyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine, hydrochloride salt
  • The title compound was obtained treating 6-chloro-9-(4-methoxy-3,5-dimethyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine with HCl according to the general procedure 2.7. HPLC Rt: 4.093 min. 1H-NMR (d6-DMSO): δ 8.38 (s, 1H), 8.23 (s, 1H), 5.55 (s, 2H), 3.93 (s, 3H), 2.34 (s, 3H), 2.31 (s, 3H). Example 29: 6-Chloro-9-(4-methoxy-3,5-dimethyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine, mesylate salt
  • The title compound was obtained treating 6-chloro-9-(4-methoxy-3,5-dimethyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine with MeSO3H according to the general procedure 2.7. HPLC Rt: 4.093 min. 1H-NMR (d6-DMSO): δ 8.38 (s, 1H), 8.17 (s, 1H), 5.54 (s, 2H), 3.95 (s, 3H), 2.36 (s, 3H), 2.34 (s, 3H), 2.33 (s, 3H).
  • Example 30 N-[6-Chloro-9-(4-methoxy-3,5-dimethyl-pyridin-2-ylmethyl)-9H-purin-2yl]-acetamide
  • A suspension of 6-chloro-9-(4-methoxy-3,5-dimethyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine (80 mg, 0.25 mmol) in acetic acid anhydride (2.2 g) was treated with one drop of concentrated H2SO4 and stirred at r.t. for 5 min. Work-up (EtOAc), drying (MgSO4) and evaporation gave the title compound as a white solid. HPLC Rt: 4.093 min. 1H-NMR (CDCl3): δ 8.20 (s, 1H), 8.10 (s, 1H), 5.46 (s, 2H), 3.78 (s, 3H), 2.54 (s, 3H), 2.38 (s, 3H), 2.27 (s, 3H).
  • Example 31 6-Chloro-9-(4-methoxy-3,5-dimethyl-1-oxy-pyridin-2-ylmethyl)-9H-purin-2-ylamine
  • The title compound was obtained by oxidation of 6-chloro-9-(4-methoxy-3,5-dimethyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine according to the general procedure 2.1. HPLC Rt: 4.435 min. 1H-NMR (CDCl3): δ 8.55 (s, 1H), 8.06 (s, 1H), 5.52 (s, 2H), 5.07 (s, 2H), 3.76 (s, 3H), 2.61 (s, 3H), 2.25 (s, 3H).). m/z (%) 335.1 (M+1, 100%), 337.1 (M+3, 34%).
  • Example 32 6-Chloro-9-(4-methoy-3,5-dimethyl-1-methoxy-pyridinium-2-methyl)-9H-purin-2-ylamine methyl sulfate salt
  • A suspension of 6-chloro-9-(4-methoxy-3,5-dimethyl-1-oxy-pyridin-2-ylmethyl)-9H-purin-2-ylamine (0.2 g, 0.56 mmol) in DCM (10 ml) was heated to reflux. Dimethyl sulfate (1.12 mmol) was added dropwise (Tarbit WO 99/10326) and heating was continued for 3 h. Filtration and washing (hot acetone) gave the title compound as a beige solid. HPLC Rt: 3.379 min. 1H-NMR (d6-DMSO): δ 9.68 (s, 1H), 9.40 (s, 1H), 5.85 (s, 2H), 4.42 (s, 3H), 4.15 (s, 3H), 4.12 (s, 3H), 2.70 (s, 3H), 2.47 (s, 3H).
  • Example 33 6-Chloro-9-(6-chloro-4-metho-3,5-dimethyl-pyridin-2ylmethyl)-9H-purin-2-ylamine Method 1
  • 6-Chloro-9-(4-methoxy-3,5-dimethyl-1-oxy-pyridin-2-ylmethyl)-9H-purin-2-ylamine was treated with POCl3 according to the general procedure 2.4. Flash chromatography gave the title compound as a white solid. HPLC Rt: 5.741 min. 1H-NMR (CDCl3): δ 7.94 (s, 1H), 5.29 (s, 2H), 5.05 (s, 2H), 3.74 (s, 3H), 2.30 (s, 3H), 2.28 (s, 3H).
  • Method 2 Step 1: 2-Chloromethyl-4-methoxy-3,5-dimethylpyridine-1-oxide
  • The title compound was obtained by oxidation of 2-chloromethyl-4-methoxy-3,5-dimethylpyridine according to the general procedure 2.1. HPLC Rt: 4.462 min. 1H-NMR (CDCl3): δ 8.05 (s, 1H), 4.93 (s, 2H), 3.77 (s, 3H), 2.37 (s, 3H), 2.24 (s, 3H).
  • Step 2: 2-Chloro-6-chloromethyl-4-methoxy-3,5-dimethylpyridine
  • The title compound was obtained by treating 2-chloromethyl-4-methoxy-3,5-dimethylpyridine-1-oxide with POCl3 according to the general procedure 2.4. HPLC Rt: 6.757 min. 1H-NMR (CDCl3): δ 4.64 (s, 2H), 3.79 (s, 3H), 2.35 (s, 3H), 2.33 (s, 3H).
  • Step 3: 6-Chloro-9-(6-chloro-4-methoxy-3,5-dimethyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 6-chloro-9H-purin-2-ylamine with 2-chloro-6-chloromethyl-4-methoxy-3,5-dimethylpyridine according to the general procedure 1.1.
  • Example 34 6-Chloro-9-(5-methoxy4-methoxymethyl-6-methyl-pyridin-3-ylmethyl)-9H-purin-2-ylamine Step 1: Acetic acid 3-acetoxy-5-hydroxymethyl-2-methyl-pyridin-4-ylmethyl ester
  • The title compound was obtained by following the procedure reported by Morisawa et al. J. Med. Chem. 1974, 17, 1083-1086. HPLC Rt: 3.08 min. 1H-NMR (CDCl3): δ 8.41 (s, 1H), 5.20 (s, 2H), 4.80 (s, 2H), 2.40 (s, 3H), 2.38 (s, 3H), 2.03 (s, 3H).
  • Step 2: Acetic acid 3-acetoxy-5-bromomethyl-2-methyl-pyridin-4-ylmethyl ester
  • The title compound was obtained from Acetic acid 3-acetoxy-5-hydroxymethyl-2-methyl-pyridin-4-ylmethyl ester according to the general procedure 2.5. HPLC Rt: 5.332 min. 1H-NMR (CDCl3): δ 8.43 (s, 1H), 5.22 (s, 2H), 4.70 (s, 2H), 2.43 (s, 3H), 2.41 (s, 3H), 2.06 (s, 3H).
  • Step 3: 6-Chloro-9-(5-acetoxy-4-acetoxymethyl-6-methyl-pyridin-3-ylmethyl)-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with acetic acid 3-acetoxy-5-bromomethyl-2-methyl-pyridin-4-ylmethyl ester according to the general procedure 1.1. HPLC Rt: 4.498 min. 1H-NMR (CDCl3): δ 8.42 (s, 1H), 7.74 (s, 1H), 5.43 (s, 2H), 5.10 (s, 2H), 5.07 (s, 2H), 2.42 (s, 3H), 2.39 (s, 3H), 1.96 (s, 3H).
  • Step 4: 6-Chloro-9-(5-hydroxy-4-hydroxymethyl-6-methyl-pyridin-3-ylmethyl)-9H-purin-2-ylamine
  • A suspension of 6-chloro-9-(5-acetoxy-4-acetoxymethyl-6-methyl-pyridin-3-ylmethyl)-9H-purin-2-ylamine and K2CO3 (excess) in MeOH was heated to 50° C. for 15 min. Filtration, work-up (EtOAc) and purification by preparative TLC gave the title compound. HPLC Rt: 4.498 min. 1H-NMR (d6-DMSO) δ 8.08 (s, 1H), 7.74 (s, 1H), 6.95 (s, 1H), 5.31 (s, 2H), 4.74 (s, 2H), 2.31 (s, 3H).
  • Step 5: 6-Chloro-9-(5-methoxy-4-methoxymethyl-6-methyl-pyridin-3-ylmethyl)-9H-purin-2ylamine
  • A mixture of 6-chloro-9-(5-hydroxy-4-hydroxymethyl-6-methyl-pyridin-3-ylmethyl)-9H-purin-2-ylamine, MeI, K2CO3 (excess) and DMF was heated to 50° C. for 15 min. Work-up (EtOAc), drying (MgSO4), evaporation, and purification by preparative TLC gave the title compound. HPLC Rt: 5.446 min. 1H-NMR (CDCl3) δ 8.35 (s, 1H), 7.70 (s, 1H), 5.35 (s, 2H), 5.25 (s, 2H), 5.15 (s, 2H), 3.80 (s, 3H) 2.59 (s, 3H), 1.970 (s, 3H).
  • Example 35 Synthesis of 6-Chloro-9-(5-ethoxy-4-hydroxymethyl-6-methyl-pyridin-3-ylmethyl)-9H-purin-2-ylamine
  • A mixture of 6-chloro-9-(5-hydroxy4-hydroxymethyl-6-methyl-pyridin-3-ylmethyl)-9H-purin-2-ylamine (see previous example), EtI (excess), K2CO3 (excess) and DMF was heated to 50° C. for 15 min. Work-up (EtOAc), drying (MgSO4), evaporation, and purification by preparative TLC gave the title compound. HPLC Rt: 3.720 min. 1H-NMR (CDCl3) δ 8.27 (s, 1H), 7.90 (s, 1H), 5.40 (s, 2H), 5.11 (s, 2H), 4.85 (d, 2H), 4.50 (t, 1H), 3.95 (q, 2H), 2.51 (s, 3H) 1.45 (t, 3H).
  • Example 36 6-Chloro-9-(3,5-dimethyl-4-amino-pyridin-2-ylmethyl)-9H-purin-2-ylamine Step 1: 2-Bromomethyl-3,5-dimethyl-4-nitro-pyridine
  • The title compound was obtained from (3,5-dimethyl-4-nitro-pyridin-2-yl)-methanol according to the general procedure 2.5. HPLC Rt: 6.206 min. 1H-NMR (CDCl3) δ 8.46 (s, 1H), 4.64 (s, 2H), 2.38 (s, 3H), 2.33 (s, 3H).
  • Step 2: 6-Chloro-9-(3,5-dimethyl-4-nitro-pyridin-2-ylmethyl)-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 2-bromomethyl-3,5-dimethyl-4-nitro-pyridine according to the general procedure 1.1 HPLC Rt: 6.206 min. 1H-NMR (CDCl3) δ 8.40 (s, 1H), 7.94 (s, 1H), 5.40 (s, 2H), 5.05 (s, 2H), 2.40 (s, 3H), 2.27 (s, 3H).
  • Step 3: 6-Chloro-9-(3,5-dimethyl-4-amino-pyridin-2-ylmethyl)-9H-purin-2-ylamine
  • A suspension of 6-chloro-9-(4,6-dimethyl-5-nitro-pyridine-3-ylmethyl)-9H-purin-2-ylamine and excess of sodium hydrosulfite (Na2S2O4) in methanol was stirred for 2 days at r.t. The MeOH was evaporated before extracting with EtOAc. Evaporation and purification by preparative TLC (100% EtOAc) gave the title compound. HPLC Rt: 3.544 min. 1H-NMR (CDCl3) δ 8.05 (s, 1H), 7.83 (s, 1H), 5.31 (s, 2H), 5.05 (s, 2H), 4.08 (s, 2H), 2.12 (s, 6H).
  • Example 37 6-Chloro-9-(3-methoxy-5-methoxymethyl-4-methyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine Step 1: 3-Methoxy-5-methoxymethyl-2,4-dimethyl-pyridine
  • The title compound was obtained by treating a solution of 5-hydroxymethyl-2,4-dimethyl-pyridin-3-ol hydrochloride (1 g, 5.2 mmol) in DMF with Mel (2.28 g, 15 mmol) and NaH (0.6 g, 50 mmol) for 1 h at 0° C. HPLC Rt: 2.835 min. 1H-NMR (CDCl3) δ 8.16 (s, 1H), 4.44 (s, 2H), 3.75 (s, 3H), 3.41 (s, 3H), 2.53 (s, 3H), 2.32 (s, 3H).
  • Step 2: 3-Methoxy-5-methoxymethyl-2,4-dimethyl-pyridine 1-oxide
  • The title compound was obtained by oxidation of 3-methoxy-5-methoxymethyl-2,4-dimethyl-pyridine according to the general procedure 2.1. HPLC Rt: 4.181 min. 1H-NMR (CDCl3) δ 8.18 (s, 1H), 4.39 (s, 2H), 3.76 (s, 3H), 3.43 (s, 3H), 2.52 (s, 3H), 2.46 (s, 3H).
  • Step 3: Acetic acid 3-methoxy-5-methoxymethyl-4-methyl-pyridin-2-ylmethyl ester
  • The title compound was obtained by treating 3-methoxy-5-methoxymethyl-2,4-dimethyl-pyridine 1-oxide with Ac2O according to the general procedure 2.2. HPLC Rt: 4.062 min. 1H-NMR (CDCl3) δ 8.32 (s, 1H), 5.27 (s, 2H), 4.47 (s, 2H), 3.80 (s, 3H), 3.43 (s, 3H), 2.34 (s, 3H), 2.25 (s, 3H).
  • Step 4: (3-Methoxy-5-methoxymethyl-4-methyl-pyridin-2-yl)-methanol
  • The title compound was obtained from acetic acid 3-methoxy-5-methoxymethyl-4-methyl-pyridin-2-ylmethyl ester according to the general procedure 2.3. HPLC Rt: 3.465 min. 1H-NMR (CDCl3) δ 8.22(s, 1H), 4.75 (d, 2H), 4.47 (s, 2H), 4.20 (t, 1H), 3.77 (s, 3H), 3.43 (s, 3H), 2.34 (s, 3H).
  • Step 5: 2-Bromomethyl-3-methoxy-5-methoxymethyl-4-methyl-pyridine
  • The title compound was obtained from (3-methoxy-5-methoxymehtyl-4-methyl-pyridine-2-yl)-methanol according to the general procedure 2.5. HPLC Rt: 4.498 min. 1H-NMR (CDCl3) δ 8.22 (s, 1H), 4.695 (s, 2H), 4.42 (s, 2H), 3.86 (s, 3H), 3.40 (s, 3H), 2.31 (s, 3H).
  • Step 6: 6-Chloro-9-(3-methoxy-5-methoxymethyl-4-methyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 2-bromomethyl-3-methoxy-5-methoxymethyl-4-methyl-pyridine according to the general procedure 1.1. HPLC Rt: 4.254 min. 1H-NMR (CDCl3) δ 8.210 (s, 1H), 7.96 (s, 1H), 5.40 (s, 2H) 5.05 (s, 2H), 4.42 (s, 2H ), 3.78 (s, 3H), 3.40 (s, 3H), 2.31 (s, 3H).
  • Example 38 6-Chloro-9-(5-chloro-6-methoxy-pyridin-3-ylmethyl)-9H-purin-2-ylamine Step 1: (5-Chloro-6-methoxy-pyridin-3-yl)-methanol
  • (5,6-Dichloro-pyridin-3-yl)-methanol was dissolved in a saturated solution of NaOMe in MeOH and heated to reflux overnight. Evaporation of MeOH, work-up (EtOAc) and evaporation gave the title compound. 1H-NMR (CDCl3) δ 8.03 (d, 1H), 7.72 (d, 1H), 4.65 (s, 2H), 4.04 (s, 3H).
  • Step 2: 5-Bromomethyl-3-chloro-2-methoxy-pyridine
  • The title compound was obtained from (5-chloro-6-methoxy-pyridine-3-yl)-methanol according to the general procedure 2.5. 1H-NMR (CDCl3) δ 8.05 (d, 1H), 7.70 (d, 1H), 4.42 (s, 2H), 4.02 (s, 3H).
  • Step 3: 6-Chloro-9-(5-chloro-6-methoxy-pyridin-3-ylmethyl)-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 5-bromomethyl-3-chloro-2-methoxy-pyridine according to the general procedure 1.1. HPLC Rt: 5.256 min. 1H-NMR (CDCl3) δ 8.11 (d, 1H), 7.78 (s, 1H), 7.63 (d, 1H), 5.19 (s, 2H), 4.04 (s, 3H).
  • Example 39 6-chloro-9-(3,4-dimethoxy-pyridin-2-ylmethyl)-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 2-chloromethyl-3,4-dimethoxylpyridine hydrochloride according to the general procedure 1.1. HPLC Rt: 3.777 min. 1H-NMR (CDCl3): δ 8.19 (d, 1H), 7.95 (s, 1H), 6.82 (d, 1H) 5.39 (s, 2H), 5.09 (s, 2H), 3.92 (s, 3H), 3.87 (s, 3H).
  • Example 40 6-chloro-9-(3-methoxy-6-methyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine Step 1: (3-Methoxy-6-methyl-pyridin-2-yl) -methanol
  • The title compound was obtained by O-methylation of 2-hydroxymethyl-6-methyl-pyridin-3-ol according to the general procedure 2.6. HPLC Rt: 2.304 min. 1H-NMR (CDCl3) δ 7.05-7.11 (m, 2H), 4.72-4.71 (d, 2H), 4.47-4.49 (t, 1H), 3.84 (s, 3H), 2.51 (s, 3H).
  • Step 2: 2-Bromomethyl-3-methoxy-6-methyl-pyridine
  • The title compound was obtained from (3-Methoxy-6-methyl-pyridine-2-yl)-methanol according to the general procedure 2.5. HPLC Rt: 4.361. 1H-NMR (CDCl3) δ 7.06-7.12 (m, 2H), 4.61 (s, 2H), 3.89 (s, 3H), 2.49 (s, 3H).
  • Step 3: 6-chloro-9-(3-methoxy-6-methyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2amino-6-chloropurine with 2bromomethyl-3-methoxy-6-methylpyridine according to the general procedure 1.1. HPLC Rt: 3.777 min. 1H-NMR (CDCl3): δ 7.92 (s, 1H), 7.11 (m, 2H), 5.39 (s, 2H), 5.15 (s, 2H), 3.85 (s, 3H), 2.45 (s, 3H).
  • Example 41 6-Chloro-9-(5-methoxy-4,6-dimethyl-pyridin-3-ylmethyl)-9H-purin-2-ylamine Step 1: (5-Methoxy-4,6-dimethyl-pyridin-3-yl)-methanol
  • The title compound was obtained by O-methylation of 5-hydroxymethyl-2,4-dimethyl-pyridin-3-ol according to the general procedure 2.6. HPLC Rt: 3.114 min. 1H-NMR (CDCl3) δ 8.08 (s, 1H), 4.67 (s, 2H), 3.74 (s, 3H), 2.49 (s, 3H), 2.33 (s, 3H).
  • Step 2: 5-Bromomethyl-3-methoxy-2,4-dimethyl-pyridine
  • The title compound was obtained from (5-methoxy-4,6-dimethyl-pyridine-3-yl)-methanol according to the general procedure 2.5. HPLC Rt: 2.873. 1H-NMR (CDCl3) δ 8.22 (s, 1H), 4.50 (s, 2H), 3.764 (s, 3H), 2.54 (s, 3H), 2.37 (s, 3H).
  • Step 3: 6-Chloro-9-(5-methoxy-4,6-dimethyl-pyridin-3-ylmethyl)-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 5-bromomethyl-3-methoxy-2,4-methylpyridine according to the general procedure 1.1. HPLC Rt: 3.7387 min. 1H-NMR (CDCl3): δ 8.25 (s, 1H), 7.63 (s, 1H), 5.39 (s, 2H), 5.11 (s, 2H), 3.73 (s, 3H), 2.55 (s, 3H), 2.22 (s, 3H).
  • Example 42 9-Methoxy-3,5-dimethyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 2-chloromethyl-4-methoxy-3,5-dimethylpyridine hydrochloride according to the general procedure 1.1. HPLC Rt: 3.434 min. 1H-NMR (CDCl3): δ 8.75 (s, 1H), 8.25 (s, 1H), 7.90 (s, 1H), 5.37 (s, 2H), 5.07 (s, 2H), 3.77 (s, 3H), 2.32 (s, 3H), 2.26 (s, 3H).
  • Example 43 6-Chloro-9-(3,5-dimethoxy-4-methyl-benzyl)-9H-purin-2-ylamine Step 1: (3,5-Dimethoxy-4-methyl-phenyl)-methanol
  • The title compound was obtained by reducing 3,5-dimethoxy-4-methyl-benzoic acid as described by Bhaskar et al. J. Org. Chem. 1991, 56, 5964-5965. HPLC Rt: 5.352. 1H-NMR (CDCl3): δ 6.58 (s, 2H), 4.68 (s, 2H), 3.85 (s, 6H), 2.10 (s, 3H).
  • Step 2: 5-Bromomethyl-1,3-dimethoxy-2-methyl-benzene
  • The title compound was obtained from (3,5-dimethoxy-4-methyl-phenyl)-methanol according to the general procedure 2.5. HPLC Rt: 7.200. 1H-NMR (CDCl3): δ 6.59 (s, 2H), 4.51 (s, 2H), 3.86 (s, 3H), 3.85 (s, 3H), 2.22 (s, 3H).
  • Step 3: 6-Chloro-9-(3,5-dimethoxy-4-methyl-benzyl)-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 5-bromomethyl-1,3-dimethoxy-4-methyl benzene according to the general procedure 1.1. HPLC Rt: 5.841 min. 1H-NMR (CDCl3): δ 7.75 (s, 1H), 6.46 (s, 1H), 5.21 (s, 2H), 5.07(s, 2H), 3.80 (s, 6H), 2.09 (s, 3H).
  • Example 44 9-(2-Bromo-3,5-dimethoxy-4-methyl-benzyl)-6-chloro-9H-purin-2-ylamine
  • The title compound was obtained by brominating 6-chloro-9-(3,5-dimethoxy-4-methyl-benzyl)-9H-purin-2-ylamine with bromine according to the general procedure 3.1. HPLC Rt: 6.222 min. 1H-NMR (CDCl3): δ 7.85 (s, 1H), 6.60 (s, 1H), 5.36 (s, 2H), 5.07 (s, 2H), 3.80 (s, 3H), 3.74 (s, 3H), 2.20 (s, 3H).
  • Example 45 8-Bromo-9-(2-bromo-3,5-dimethoxy-4-methyl-benzyl)-6-chloro-9H-purin-2-ylamine
  • The title compound was obtained by brominating 6-chloro-9-(3,5-dimethoxy-4-methyl-benzyl)-9H-purin-2-ylamine with excess bromine according to the general procedure 3.1. HPLC Rt: 7.040 min. 1H-NMR (CDCl3): δ 5.88 (s, 1H), 5.38 (s, 2H), 5.14 (s, 2H), 3.83 (s, 3H), 3.57 (s, 3H), 2.19 (s, 3H).
  • Example 46 2,6-Dichloro-9-(4-methoxy-3,5-dimethyl-pyridin-2-ylmethyl)-9H-purine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 2-chloromethyl-4-methoxy-3,5-dimethylpyridine (or its HCl salt) according to the general procedure 1.1. HPLC Rt: 5.081 min. 1H-NMR (CDCl3): δ 8.30 (s, 1H), 8.17 (s, 1H), 5.50 (s, 2H) 3.80 (s, 3H), 2.38 (s, 3H), 2.27 (s, 3H).
  • Example 47 8-butyl-6-chloro-9-(3,4,5-trimethoxy-benzyl)-9H-purin-2-ylamine Step 1: 6-Chloro-N4-(3,4,5-trimethoxy-benzyl)-pyrimidine-2,4,5-triamine
  • The title compound was obtained by refluxing 4,6-dichloro-pyrimidine-2,5-diamine (see Seela et al. Helv. Chim. Acta. 1986, 69, 1602-1613 and U.S. Pat. No. 5,917,042), 3,4,-5-trimethoxybenzylamine, and Et3N in butanol or ethanol for 1 to 14 h. HPLC Rt: 4.327 min. 1H-NMR (CDCl3): δ 6.57 (s, 2H), 5.65 (t, 1H), 4.75 (s, 2H), 4.54 (d, 2H), 3.86-3.87 (d, 9H).
  • Step 2: 8-Butyl-6-chloro-9-(3,4,5-trimethoxy-benzyl)-9H-purin-2-ylamine
  • The title compound was obtained by following the procedure given in WO 98/39344. HPLC Rt: 5.971 min. 1H-NMR (CDCl3): δ 6.34 (s, 2H), 5.19 (s, 2H), 5.04 (s, 2H), 3.81 (s, 3H), 3.77 (s, 6H), 2.71-2.75 (t, 2H), 1.68-1.74 (m, 2H), 1.35-1.41 (m, 2H), 0.88-0.92 (t, 2H).
  • Example 48 6-Chloro-9-(3,4,5-trimethoxy-benzyl)-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 3,4,5-trimethoxybenzyl according to the general procedure 1.1. The title compound was also obtained by treating a solution of 6-chloro-N-4-(3,4,5-trimethoxy-benzyl)-pyrimidine-2,4,5-triamine in triethyl orthoformate with a catalytic amount of conc. HCl at r.t. for 20 min. HPLC Rt: 4.906 min. 1H-NMR (CDCl3): δ 7.76 (s, 1H), 6.51 (s, 2H), 5.18 (s, 2H), 5.12 (s, 2H), 3.85 (s, 3H), 3.84 (s, 6H).
  • Example 49 Acetic acid 4-(2-amino-6-chloro-purin-9-ylmethyl)-3-bromo-2,6-dimethoxy-phenyl ester Step 1: Acetic acid 4-hydroxymethyl-2,6-dimethoxy-phenyl ester
  • A solution of acetic acid 4-formyl-2,6-dimethoxy-phenyl ester (25 mmol) in MeOH (100 mL) was treated with NaBH4 (1 equiv.) at 0° C. for 15 min. After quenching with acetone and evaporating the solvent, work-up (CH2Cl2) and evaporation gave the title compound as a white solid (85% yield). Rf (in EtOAc/Hexane 1:1): 0.5. 1H-NMR (CDCl3): δ 6.66 (s, 1H), 4.68-4.70 (d, 2H), 3.85 (s, 3H), 2.36 (s, 3H), 1.74 (t, 1H).
  • Step 2: Acetic acid 3-bromo-4-hydroxymethyl-2,6-dimethoxy-phenyl ester
  • The title compound was obtained by bromination of acetic acid 4-hydroxymethyl-2,6-dimethoxy-phenyl ester in AcOH/AcONa buffer according to the general procedure 3.1. Rf (EtOAc/Hexane 1:3): 0.2. 1H-NMR (CDCl3): δ 7.01 (s, 1H), 4.75-4.76 (d, 2H), 3.86 (s, 3H), 3.85 (s, 3H ), 2.38 (s, 3H), 2.05 (t, 1H).
  • Step 3: Acetic acid 3-bromo-4-bromomethyl-2,6-dimethoxy-phenyl ester
  • The title compound was obtained from Acetic acid 3-bromo-4-hydroxymethyl-2,6-dimethoxy-phenyl ester according to the general procedure 2.5. Rf (EtOAc/Hexane 1:3): 0.8. 1H-NMR (CDCl3): δ 6.87 (s, 1H), 4.60 (s, 2H), 3.84 (s, 3H), 3.83 (s, 3H ), 2.36 (s, 3H).
  • Step 4: Acetic acid 4-(2-amino-6-chloro-purin-9-ylmethyl)-3-bromo-2,6-dimethoxy-phenyl ester
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with acetic acid 3-bromo-4-bromomethyl-2,6-dimethoxy-phenyl ester according to the general procedure 1.1. HPLC Rt: 5.081 min. 1H-NMR (CDCl3): δ 8.30 (s, 1H), 8.17 (s, 1H), 5.50 (s, 2H), 3.80 (s, 3H), 2.38 (s, 3H), 2.27 (s, 3H).
  • Example 50 4-(2-amino-6-chloro-purin-9-ylmethyl)-3-bromo-2,6-dimethoxy-phenol
  • The title compound was obtained by deacetylation of Acetic acid 4-(2-amino-6-chloro-purin-9-ylmethyl)-3-bromo-2,6-dimethoxy-phenyl ester (see previous example) in NH3/MeOH at r.t. for 0.5 h or K2CO3 in methanol according to the general procedure 2.3. HPLC Rt: 4.912 min. 1H-NMR (CDCl3): δ 7.85 (s, 1H), 6.72 (s, 1H), 5.70 (s, 1H), 5.33 (s, 1H), 5.07 (s, 2H), 3.94 (s, 3H), 3.82 (s, 3H).
  • Example 51 9-(2-bromo-3,4,5-trimethoxy-benzyl)-6-chloro-9H-purin-2-ylamine
  • The title compound was obtained by O-methylation of 4-(2-amino-6-chloro-purin-9-ylmethyl)-3-bromo-2,6-dimethoxy-phenol (see previous example) according to the general procedure 2.6. The title compound could also be obtained by bromination of 6-chloro-9-(3,4,5-trimethoxy-benzyl)-9H-purin-2-ylamine (see example 48) in acetate buffer according to the general procedure 3.1. HPLC Rt: 5.742 min. 1H-NMR (CDCl3): δ 7.85 (s, 1H), 6.66 (s, 1H), 5.32 (s, 2H), 5.11 (s, 2H), 3.90 (s, 3H), 3.87 (s, 3H), 3.76 (s, 3H).
  • Example 52 9-(4-allyloxy-2-bromo-3,5-dimethoxy-benzyl)-6-chloro-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 4-(2-amino-6-chloro-purin-9-ylmethyl)-3-bromo-2,6-dimethoxy-phenol (see example 50) with 3-bromo-propene in DMF in the presence of K2CO3 at 70° C. for 0.25-1 h. HPLC Rt: 6.309 min. 1H-NMR (CDCl3): δ 7.84 (s, 1H), 6.64 (s, 1H), 6.00-6.10 (m, 2H), 5.36-5.36 (m, 1H), 5.31 (s, 2H), 5.20-5.21 (m, 2H), 4.52-4.54 (m, 2H), 3.90 (s, 3H), 3.74 (s, 2H).
  • Example 53 9-(2-bromo-4-chloromethoxy-3,5-dimethoxy-benzyl)-6-chloro-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 4-(2-amino-6-chloro-purin-9-ylmethyl)-3-bromo-2,6-dimethoxy-phenol (see example 50) with chloroiodomethane in DMF in the presence of K2CO3 at 70° C. for 0.25-1 h. HPLC Rt: 6.109 min. 1H-NMR (CDCl3): δ 7.87 (s, 1H), 6.66 (s, 1H), 5.91 (s, 2H), 5.34 (s, 2H), 5.08 (s, 2H), 3.90 (s, 3H), 3.76 (s, 3H).
  • Example 54 9-[2-bromo-4-(2-chloro-ethoxy)-3,5-dimethoxy-benzyl]-6-chloro-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 4-(2-amino-6-chloro-purin-9-ylmethyl)-3-bromo-2,6-dimethoxy-phenol (see example 50) with 1-bromo-2-chloro-ethane in DMF in the presence of K2CO3 at 70° C. for 0.25-1 h. HPLC Rt: 6.285 min. 1H-NMR (CDCl3): δ 7.86 (s, 1H), 6.67 (s, 1H), 5.37 (s, 2H), 5.32 (s, 2H), 4.22-4.25 (t, 2H), 3.91 (s, 3H), 3.77-3.78 (t, 2H), 3.75 (s, 3H).
  • Example 55 9-(2-bromo-4-cyclopropylmethoxy-3,5-dimethoxy-benzyl)-6-chloro-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 4-(2-amino-6-chloro-purin-9-ylmethyl)-3-bromo-2,6-dimethoxy-phenol (see example 50) with bromomethyl-cyclopropane in DMF in the presence of K2CO3 at 70° C. for 0.25-1 h. HPLC Rt: 6.512 min. 1H-NMR (CDCl3): δ 7.86 (s, 1H), 6.67 (s, 1H), 5.34 (s, 2H), 5.17 (s, 2H), 3.95 (s, 3H), 3.83-3.84 (d, 2H), 3.77 (s, 3H), 1.27 (m, 1H), 0.58-0.62 (m, 2H), 0.28-0.32 (m, 2H).
  • Example 56 9-(2-bromo-4-ethoxy-3,5-dimethoxy-benzyl)-6-chloro-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 4-(2-amino-6-chloro-purin-9-ylmethyl)-3-bromo-2,6-dimethoxy-phenol (see example 50) with EtI in DMF in the presence of K2CO3 at 70° C. for 0.25-1 h. HPLC Rt: 6.112 min. 1H-NMR (CDCl3): δ 7.84 (s, 1H), 6.65 (s, 1H), 5.31 (s, 2H), 5.13 (s, 2H), 4.04-4.09 (q, 2H), 3.90(s, 3H), 3.82 (s, 3H), 1.32-1.38 (t, 3H).
  • Example 57 9-(2-bromo-3,5-dimethoxy-4-propoxy-benzyl)-6-chloro-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 4-(2-amino-6-chloro-purin-9-ylmethyl)-3-bromo-2,6-dimethoxy-phenol (see example 50) with PrI in DMF in the presence of K2CO3 at 70° C. for 0.25-1 h. HPLC Rt: 6.594 min. 1H-NMR (CDCl3): δ 7.84 (s, 1H), 6.65 (s, 1H), 5.30 (s, 2H), 5.14 (s, 2H), 3.93-3.97 (t, 2H), 3.89 (s, 3H), 3.74 (s, 3H), 1.72-1.81 (m, 2H), 1.00-1.04 (t, 3H).
  • Example 58 9-(2-bromo-4-butoxy-3,5-dimethoxy-benzyl)-6-chloro-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 4-(2-amino-6-chloro-purin-9-ylmethyl)-3-bromo-2,6-dimethoxy-phenol (see example 50) with BuI in DMF in the presence of K2CO3 at 70° C. for 0.25-1 h. HPLC Rt: 6.594 min. 1H-NMR (CDCl3): δ 7.84 (s, 1H), 6.65 (s, 1H), 5.30 (s, 2H), 5.14 (s, 2H), 3.97-4.00 (t, 2H), 3.89 (s, 3H), 3.74 (s, 3H), 1.68-1.76 (m, 2H), 1.44-1.53 (m,
  • Example 59 6-chloro-9-(3-methoxymethoxy-6-methyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine Step 1: (3-methoxymethoxy-6-methyl-pyridin-2-yl)-methanol
  • The title compound was obtained by O-alkylation of 2-hydroxymethyl-6-methyl-pyridin-3-ol with chloromethyl methyl ether according to the general procedure 2.6. 1H-NMR (CDCl3) δ 7.28-7.30 (d, 1H), 6.98-7.00 (d, 1H), 5.17 (s, 2H), 4.71 (s, 2H), 4.50 (s, 2H), 3.45 (s, 3H), 2.49 (s, 3H).
  • Step 2: 2-Bromomethyl-3-methoxymethoxy-6-methyl-pyridine
  • The title compound was obtained from (3-methoxymethyl-6-methyl-pyridine-2-yl)-methanol according to the general procedure 2.5. 1H-NMR (CDCl3) δ 7.32-7.40 (d 1H), 7.08-7.10 (d, 1H), 5.30 (s, 2H), 4.67 (s, 2H), 3.55 (s, 3H), 2.54 (s, 3H).
  • Step 3: 6-Chloro-9-(3-methoxymethoxy-6-methyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 2-bromomethyl-3-methoxymethoxy-6-methyl-pyridine according to the general procedure 1.1. HPLC Rt: 3.884 min. 1H-NMR (CDCl3): δ 7.92 (s, 1H), 7.34-7.36 (d, 1H), 7.05-7.07 (d, 1H), 5.39 (s, 2H), 5.17 (s, 2H), 5.06 (s, 2H), 3.40 (s, 3H), 2.44 (s, 3H).
  • Example 60 3-(2-Amino-6-chloro-purin-9-ylmethyl)-3H-benzothiazole-2-thione
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 3-chloromethyl-3H-benzothiazole-2-thione according to the general procedure 1.1. HPLC Rt: 5.982 min. 1H-NMR (d6-DMSO): δ 8.37 (s, 1H), 8.25-8.28 (d, 1H) 7.79-7.81 (d, 1H), 7.54-7.56 (t, 1H), 7.39-7.43 (m 1H), 7.20 (s, 2H), 6.62 (s, 2H), 3.34 (s, 3H).
  • Example 61 6-Chloro-9-(2,5-dimethyl-benzyl)-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 2-chloromethyl-1,4-dimethyl-benzene according to the general procedure 1.1. HPLC Rt: 5.920 min. 1H-NMR (d6-DMSO): δ 8.10 (s, 1H), 7.10 (d, 1H) 7.04 (d, 1H), 6.95 (s, 2H), 6.65 (s, 1H), 5.23, (s, 2H), 3.34 (s, 3H), 2.30 (s, 3H), 2.17 (s, 3H).
  • Example 62 6-Chloro-9-isoquinolin-1-ylmethyl-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 1-bromomethyl-isoquinoline according to the general procedure 1.1. HPLC Rt: 4.306 min. 1H-NMR (d6-DMSO): δ 8.43-8.45 (d, 1H) 8.27-8.28 (d, 1H), 8.20 (s, 1H), 8.03-8.05 (d 1H), 7.85-7.89 (m, 1H), 7.77-7.82 (m, 2H), 6.83 (s, 2H), 6.04 (s, 2H).
  • Example 63 9-benzo[1,2,5]thiadiazol-5-ylmethyl-6-chloro-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 5-bromomethyl-benzo[1,2,5]thiadiazole according to the general procedure 1.1. HPLC Rt: 4.931 min. 1H-NMR (d6-DMSO): δ 8.31 (s, 1H), 8.11 (d, 1H), 7.87 (br. s, 1H), 7.70-7.67 (dd, 1H), 6.98 (br, s, 2H), 5.52 (s, 2H).
  • Example 64 9-(1-methyl-1H-benzotriazol-5-ylmethyl)-6-chloro-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 6-bromomethyl-1-methyl-1H-benzotriazole according to the general procedure 1.1. HPLC Rt: 4.295 min. 1H-NMR (d6-DMSO): δ 8.29 (s, 1H), 7.95 (s, 1H), 7.86-7.84 (d, 1H), 7.55-7.53 (dd, 1H), 6.97 (s, 2H), 5.45 (s, 2H), 4.3 (s, 3H).
  • Example 65 6-chloro-9-(6-chloro-benzo[1,2,5]thiadiazol-5-ylmethyl)-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 5-bromomethyl-6-chloro-benzo[1,2,5]thiadiazole according to the general procedure 1.1. HPLC Rt: 5.400 min. 1H-NMR (d6-DMSO): δ 8.45 (s, 1H), 8.20 (s, 1H), 7.64 (s, 1H), 6.97 (s, 2H), 5.55 (s, 2H).
  • Example 66 9-benzo[1,2,5]thiadiazol-4-ylmethyl-6-chloro-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 4-bromomethyl-benzo[1,2,5]thiadiazole according to the general procedure 1.1. HPLC Rt: 5.027 min. 1H-NMR (d6-DMSO): δ 8.26 (s, 1H), 8.04-8.07 (d, 1H), 7.68-7.64 (dd, 1H), 7.22-7.20 (dd, 1H), 6.93 (s, 2H), 5.79 (s, 2H).
  • Example 67 6-chloro-9-(6-fluoro-4a,8a-dihydro-4H-benzo [1,3]dioxin-8-ylmethyl)-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 8-chloromethyl-6-fluoro-4a,8a-dihydro-4H-benzo[1,3]dioxine according to the general procedure 1.1. HPLC Rt: 5.172 min. 1H-NMR (CDCl3): δ 7.84 (s, 1H), 6.92-6.89 (dd, 1H), 6.70-6.67 (dd, 1H), 5.31 (s, 2H), 5.22 (s, 2H), 5.07 (s, 2H), 4.90 (s, 2H).
  • Example 68 1-[3-(2-Amino-6-chloro-purin-9-ylmethyl)-4-methoxy-phenyl]-ethanone
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 1-(3-chloromethyl-4-methoxy-phenyl)-ethanone according to the general procedure 1.1. HPLC Rt: 4.887 min. 1H-NMR (CDCl3): δ 8.03-8.02 (d, 1H), 7.97-7.95 (dd, 1H), 7.81 (s, 1H), 6.96-6.93 (d, 1H), 5.25 (s, 2H), 5.08 (s, 2H), 3.93 (s, 3H), 2.54 (s, 3H).
  • Example 69 6-chloro-9-(3-trifluoromethoxy-benzyl)-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 1-chloromethyl-3-trifluoromethoxy-benzene according to the general procedure 1.1. HPLC Rt: 5.965 min. 1H-NMR (CDCl3): δ 7.76 (s, 1H), 7.39-7.37 (t, 1H), 7.21-7.15 (m, 3H), 5.27 (s, 2H), 5.12 (s, 2H).
  • Example 70 6-chloro-9-(2-fluoro-3-trifluoromethyl-benzyl)-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 1-bromomethyl-2-fluoro-3-trifluoromethyl-benzene according to the general procedure 1.1. HPLC Rt: 4.841 min. 1H-NMR (CDCl3): δ 7.83 (s, 1H), 7.63-7.59 (t, 1H), 7.48-7.45 (t, 1H), 7.25-7.22 (t, 1H), 5.36 (s, 2H), 5.12 (s, 2H).
  • Example 71 6-Chloro-9-(2-fluoro-4,5-dimethoxy-benzyl)-9H-purin-2-ylamine Step 1: (2-Fluoro-4,5-dimethoxy-phenyl)-methanol
  • A solution of 2-fluoro-4,5-dimethoxy-benzaldehyde (6.0 mmol) in MeOH (10 mL) was treated with NaBH4 (1.2 equiv.) at 0-23° C. for 0.5 h. After quenching with acetone and evaporating the solvent, work-up (CH2Cl2), drying (MgSO4) and evaporation gave the title compound as a crude oily product (94% yield). 1H-NMR (CDCl3): δ 6.90-6.88 (d, 1H), 6.61-6.64 (d, 1H), 4.65-4.63 (d, 2H), 3.86 (s, 3H), 3.84 (s, 3H), 2.00-1.94 (t, 1H).
  • Step 2: 1-Bromomethyl-2-fluoro-4,5-dimethoxy-benzene
  • The title compound was obtained from (2-fluoro-4,5-dimethoxy-phenyl)-methanol according to the general procedure 2.5. 1H-NMR (CDCl3): δ 6.84-6.81 (d, 1H), 6.64-6.61 (d, 1H), 4.52 (s, 2H), 3.89 (s, 3H), 3.84 (s, 3H).
  • Step 3: 6-Chloro-9-(2-fluoro-4,5-dimethoxy-benzyl)-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 1-bromomethyl-2-fluoro-4,5-dimethoxy-benzene according to the general procedure 1.1. HPLC Rt: 4.939 min. 1H-NMR (CDCl3): δ 7.79 (s, 1H), 6.88-6.85 (d, 1H), 6.69-6.66 (d, 1H), 5.22 (s, 2H), 5.12 (s, 2H), 3.89 (s, 3H), 3.82 (s, 3H).
  • Example 72 6-chloro-9-(2,3-dimethoxy-benzyl)-9H-purin-2-ylamine Step 1: 1-Bromomethyl-2,3-dimethoxy-benzene
  • The title compound was obtained from (2,3-dimethoxy-phenyl)-methanol according to the general procedure 2.5. 1H-NMR (CDCl3): δ 7.02-7.08 (t, 1H), 6.95-6.98 (dd, 1H), 6.87-6.91 (dd, 1H), 4.57 (s, 2H), 3.92 (s, 3H), 3.86 (s, 3H).
  • Step 2: 6-Chloro-9-(2,3-dimethoxy-benzyl)-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 1-bromomethyl-2,3-dimethoxy-benzene according to the general procedure 1.1. HPLC Rt: 5.200. 1H-NMR (CDCl3): δ 7.81 (s, 1H), 7.02-7.08 (t, 1H), 6.95-6.98 (dd, 1H), 6.87-6.91 (dd, 1H), 5.28 (s, 2H), 5.12 (s, 2H), 3.92 (s, 3H), 3.87 (s, 3H).
  • Example 73 6-chloro-9-(3,4-dimethoxy-benzyl)-9H-purin-2-ylamine Step 1: 4-Bromomethyl-1,2-dimethoxy-benzene
  • The title compound was obtained from (3,4-dimethoxy-phenyl)-methanol according to the general procedure 2.5. 1H-NMR (CDCl3): δ 6.95-6.98 (dd, 1H), 6.92-6.94 (d, 1H), 6.78-6.81 (d, 1H), 4.51 (s, 2H), 3.92 (s, 3H), 3.86 (s, 3H).
  • Step 2: 6-Chloro-9-(3,4-dimethoxy-benzy1)-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 4-bromomethyl-1,2-dimethoxy-benzene according to the general procedure 1.1. HPLC Rt: 4.753.
  • 1H-NMR (CDCl3): δ 7.71 (s, 1H), 6.87 (s, 2H), 6.82 (s, 1H), 5.20 (s, 2H), 5.12 (s, 2H), 3.92 (s, 3H), 3.87 (s, 3H).
  • Example 74 9-(2-chloro-3,4,5-trimethoxy-benzyl)-6-methyl-9H-purin-2-ylamine
  • A suspension of 6-chloro-9-(2-chloro-3,4,5-trimethoxy-benzyl)-9H-purin-2-ylamine, see example 97) (0.2 mmol) and tetrakis(triphenylphosphino)-palladium (0.02 mmol) in dry THF (3 mL) was treated with trimethylaluminum (2M in toluene, 0.45 mmol) under nitrogen and heated to reflux for 3 h. The reaction mixture was cooled to r.t., diluted with toluene (5 mL) and quenched with methanol (0.5 mL) followed by ammonium chloride (1 mmol). The mixture was heated to reflux for 2 h and filtered, while hot, on Celite. See J. Med. Chem. 1999, 42, 2064-2086. TLC (100% EtOAc) Rf was 0.2. HPLC Rt: 4.800 min.
  • Example 75 6-chloro-9-(2-chloro-4,5-dimethoxy-benzyl)-9H-purin-2-ylamine Step 1: (2-Chloro-4,5-dimethoxy-phenyl)-methanol
  • The title compound was obtained by chlorination of (3,4-dimethoxy-phenyl)-methanol according to the general procedure 3.1. 1H-NMR (CDCl3): δ 7.01 (s, 1H), 6.88 (s, 1H), 4.75-4.73 (d, 2H), 3.91 (s, 3H), 3.90 (s, 3H), 1.95-1.92 (t, 1H).
  • Step 2: 1-Bromomethyl-2-chloro-4, 5-dimethoxy-benzene
  • The title compound was obtained from (2-chloro-4,5-dimethoxy-phenyl)-methanol according to the general procedure 2.5. 1H-NMR (CDCl3): δ 6.92 (s, 1H), 6.88 (s, 1H), 4.60 (s, 2H), 3.91 (s, 3H), 3.90 (s, 3H).
  • Step 3: 6-Chloro-9-(2-chloro-4,5-dimethoxy-benzyl)-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 1-bromomethyl-2-chloro-4,5-dimethoxy-benzene according to the general procedure 1.1. HPLC Rt: 5.366 min. 1H-NMR (CDCl3): δ 7.81 (s, 1H), 7.28 (s, 1H), 6.92 (s, 1H), 5.30 (s, 2H), 5.15 (s, 2H), 3.90 (s, 3H), 3.84 (s, 3H).
  • Example 76 6-chloro-9-(2-iodo-4,5-dimethoxy-benzyl)-9H-purin-2-ylamine Step 1: 4-Bromomethyl-1,2-dimethoxy-benzene
  • The title compound was obtained from (3,4-dimethoxy-phenyl)-methanol according to the general procedure 2.5. 1H-NMR (CDCl3): δ 6.95-6.98 (dd, 1H), 6.92-6.94 (d, 1H), 6.78-6.81 (d, 1H), 4.51 (s, 2H), 3.92 (s, 3H), 3.86 (s, 3H).
  • Step 2: 1-Bromomethyl-2-iodo-4, 5-dimethoxy-benzene
  • The title compound was obtained by iodination of 4-bromomethyl-1,2-dimethoxy-benzene according to the general procedure 3.1. 1H-NMR (CDCl3): δ 7.04 (s, 1H), 6.95 (s, 1H), 4.61 (s, 2H), 3.91 (s, 3H), 3.90 (s, 3H).
  • Step 3: 6-Chloro-9-(2-iodo-4,5-dimethoxy-benzyl)-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 1-bromomethyl-2-iodo-4,5-dimethoxy-benzene according to the general procedure 1.1. HPLC Rt: 5.470 min. 1H-NMR (CDCl3): δ 7.84 (s, 1H), 7.08 (s, 1H), 6.93 (s, 1H), 5.30 (s, 2H), 5.15 (s, 2H), 3.91 (s, 3H), 3.82 (s, 3H).
  • Example 77 6-Bromo-9-(2-chloro-3,4,5-trimethoxy-benzyl)-9H-purin-2-ylamine Step 1: 2-Chloro-1-chloromethyl-3,4,5-trimethoxy-benzene
  • The title compound was obtained by chlorination of 5-chloromethyl-1,2,3-trimethoxy-benzene according to the general procedure 3.1. 1H-NMR (CDCl3): δ 6.82 (s, 1H), 4.70 (s, 1H), 3.93 (s, 3H), 3.90 (s, 3H) 3.87 (s, 3H).
  • Step 2: Synthesis of 6-chloro-9-(2-chloro-3,4,5-trimethoxy-benzyl)-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 6-bromoguanine with 2-chloro-1-chloromethyl-3,4,5-trimethoxy-benzene according to the general procedure 1.1. HPLC Rt: 5.676 min. 1H-NMR (CDCl3): δ 7.82 (s, 1H), 6.70 (s, 1H), 5.32 (s, 2H), 5.15 (s, 2H), 3.93 (s, 3H), 3.91 (s, 3H) 3.79 (s, 3H).
  • Example 78 6-chloro-9-(6-chloro-benzol[1,3]dioxol-5-ylmethyl)-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 5-chloro-6-chloromethyl-benzo[1,3]dioxole according to the general procedure 1.1. HPLC Rt: 5.506 min. 1H-NMR (CDCl3): δ 7.81 (s, 1H), 6.88 (s, 1H), 6.79 (s, 1H), 5.98 (s, 2H), 5.25 (s, 2H), 5.13 (s, 2H).
  • Example 79 6-chloro-9-(2,4-dimethoxy-3-methyl-benzyl)-9H-purin-2-ylamine Step 1: 1-Bromomethyl-2,4-dimethoxy-3-methyl-benzene
  • The title compound was obtained from (2,4-dimethoxy-3-methyl-phenyl)-methanol according to the general procedure 2.5.
  • Step 2: 6-Chloro-9-(2,4-dimethoxy-3-methyl-benzyl)-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 1-bromomethyl-2,4-dimethoxy-3-methyl-benzene according to the general procedure 1.1. HPLC Rt: 5.433 min. 1H-NMR (CDCl3): δ 7.76 (s, 1H), 7.08-7.06 (d, 1H), 6.60-6.62 (d, 1H), 5.20 (s, 2H), 5.07 (s, 2H), 3.82 (s, 3H), 3.72 (s, 3H), 2.17 (s, 3H).
  • Example 80 6-Chloro-9-(2-chloro-3,4-dimethoxy-benzyl)-9H-purin-2-ylamine Step 1: 1-Bromomethyl-2-chloro-3, 4-dimethoxy-benzene
  • The title compound was obtained from (2-chloro-3,4-dimethoxy-phenyl)-methanol according to the general procedure 2.5.
  • Step 2: 6-Chloro-9-(2-chloro-3,4-dimethoxy-benzyl)-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 1-bromomethyl-2-chloro-3,4-dimethoxy-benzene according to the general procedure 1.1. HPLC Rt: 5.633 min. 1H-NMR (CDCl3): δ 7.80 (s, 1H), 7.00-6.98 (d, 1H), 6.82-6.79 (d, 1H), 5,31 (s, 2H), 5.08 (s, 2H), 3.88 (s, 3H), 3.88 (s, 3H).
  • Example 81 6-chloro-9-(3-methoxy-benzyl)-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 1-bromomethyl-3-methoxybenzene according to the general procedure 1.1. HPLC Rt: 5.136 min. 1H-NMR (CDCl3): δ 7.75 (s, 1H), 7.30-7.28 (m. 1H), 6.88-6.85 (dd, 1H), 6.84-6.82 (dd, 1H), 6.80-6.79 (m, 1H), 5.28 (s, 2H), 5.15 (s, 2H), 3.78 (s, 3H).
  • Example 82 6-Chloro-9-(2,6-dibromo-3,5-dimethoxy-benzyl)-9H-purin-2-ylamine Step 1: 2-Bromo-1-chloromethyl-3,5-dimethoxy-benzene and 2,4-dibromo-3-chloromethyl-1,5-dimethoxy-benzene
  • Bromination of 1-chloromethyl-3,5-dimethoxy-benzene according to the general procedure 3.1 gave a mixture of the two title compounds, which were separated by flash chromatography. 1H-NMR of 2,4-dibromo-3-chloromethyl-1,5-dimethoxy-benzene (CDCl3): δ 6.52 (s, 1H), 5.02 (s, 2H), 3.93 (s, 6H). 2-bromo-1-chloromethyl-3,5-dimethoxy-benzene 1H-NMR (CDCl3): δ 6.67-6.67 (d, 1H), 6.47-6.46 (d, 1H), 4.80 (s, 2H), 3.85 (s, 3H), 3.82 (s, 3H).
  • Step 2: 6-Chloro-9-(2,6-dibromo-3,5-dimethoxy-benzyl)-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 2,4-dibromo-3-chloromethyl-1,5-dimethoxy-benzene according to the general procedure 1.1. HPLC Rt: 6.022 min. 1H-NMR (CDCl3): δ 7.46 (s, 1H), 6.64 (s, 1H), 5.64 (s, 2H), 5.14 (s, 2H), 3.99 (s, 6H).
  • Example 83 9-(2-Bromo-3,5-dimethoxy-benzyl)-6-chloro-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 2-bromo-1-chloromethyl-3,5-dimethoxy-benzene (see previous example, step 1) according to the general procedure 1.1. HPLC Rt: 6.026 min. 1H-NMR (CDCl3): δ 7.82 (s, 1H), 6.48-6.47 (d, 1H), 6.32-6.32 (d, 1H), 5.35 (s, 2H), 5.09 (s, 2H), 3.90 (s, 3H), 3.73 (s, 3H).
  • Example 84 6-chloro-9-(3,5-dimethoxy-benzyl)-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 1-chloromethyl-3,5-dimethoxy-benzene according to the general procedure 1.1. HPLC Rt: 5.257 min. 1H-NMR (CDCl3): δ 7.79 (s, 1H), 6.44-6.42 (t, 1H), 6.41-6.39 (d, 2H), 5.22 (s, 2H), 5.15 (s, 2H), 3.80 (s, 6H).
  • Example 85 N-[6-chloro-9-(3,4,5-trimethoxy-benzyl)-9H-purin-2-yl]-acetamide
  • A solution of 6-chloro-9-(3,4,5-trimethoxy-benzyl)-9H-purin-2-ylamine in acetic acid was treated with fuming HNO3 at 0° C. for 15 min. Work-up and preparative TLC (EtOAc:hexane 1:1) gave N-[6-chloro-9-(3,4,5-trimethoxy-benzyl)-9H-purin-2-yl]-acetamide. HPLC Rt: 5.744 min. 1H-NMR (CDCl3): δ 8.09 (s, 1H), 6.58 (s, 2H), 5.33 (s, 2H), 3.85 (s, 3H), 3.85 (s, 6H), 2.43 (s, 3H).
  • Example 86 6-chloro-9-(2,5-dimethoxy-benzyl)-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 1-chloromethyl-2,5-dimethoxy-benzene according to the general procedure 1.1. HPLC Rt: 5.291 min. 1H-NMR (CDCl3): δ 7.82 (s, 1H), 6.85-6.84 (d, 1H), 6.82-6.82 (d, 2H), 5.18 (s, 2H), 5.16 (s, 2H), 3.80 (s, 3H), 3.75 (s, 3H).
  • Example 87 8-bromo-6-chloro-9-(2,5-dimethoxy-benzyl)-9H-purin-2-ylamine
  • The title compound was obtained by bromination of 6-chloro-9-(2,5-dimethoxy-benzyl)-9H-purin-2-ylamine according to the general procedure 1.2. HPLC Rt: 6.150 min. 1H-NMR (CDCl3): δ 6.83-6.78 (m, 2H), 6.37-6.36 (d, 1H), 5.31 (s, 2H), 5.13 (s, 2H), 3.83 (s, 3H), 3.70 (s, 3H).
  • Example 88 6-chloro-9-(4,5-dimethoxy-2-nitro-benzyl)-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 1-bromomethyl-4,5-dimethoxy-2-nitro-benzene according to the general procedure 1.1. HPLC Rt: 5.194 min. 1H-NMR (CDCl3): δ 7.98 (s, 1H), 7.74 (s, 1H), 6.79 (s, 1H), 5.67 (s, 2H), 5.15 (s, 2H), 3.98 (s, 3H), 3.85 (s, 3H).
  • Example 89 8-bromo-6-chloro-9-(4,5-dimethoxy-2-nitro-benzyl)-9H-purin-2-ylamine
  • The title compound was obtained by brominating 6-chloro-9-(4,5-dimethoxy-2-nitro-benzyl)-9H-purin-2-ylamine (see previous example) according to the general procedure 1.2. HPLC Rt: 6.040 min. 1H-NMR (CDCl3): δ 7.74 (s, 1H), 6.13 (s, 1H), 5.78 (s, 2H), 5.16 (s, 2H), 3.99 (s, 3H), 3.71 (s, 3H).
  • Example 90 6-chloro-9-(2,5-dichloro-benzyl)-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 2-bromomethyl-1,4-dichloro-benzene according to the general procedure 1.1. HPLC Rt: 5.846 min. 1H-NMR (CDCl3): δ 7.82 (s, 1H), 7.38-7.36 (d, 1H), 7.28-7.26 (dd, 1H), 7.18-7.18 (d, 1H), 5.32 (s, 2H), 5.17 (s, 2H).
  • Example 91 6-chloro-9-(2,3,5-trifluoro-benzyl)-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 1-bromomethyl-2,3,5-trifluoro-benzene according to the general procedure 1.1. HPLC Rt: 5.414 min. 1H-NMR (CDCl3): δ 7.82 (s, 1H), 6.98-6.89 (m, 1H), 6.82-6.75 (m, 1H), 5.30 (s, 2H), 5.13 (s, 2H).
  • Example 92 (2-amino-6-chloro-purin-9-yl)-(3,4,5-trimethoxy-phenyl)-methanone
  • A solution of 6-chloro-9H-purin-2-ylamine in pyridine was treated with 3,4,5-trimethoxybenzoyl chloride at r.t. for 2 h. Work-up and purification by preparative TLC (EtOAc:hexane 1:1) gave the title compound. HPLC Rt: 5.305 min. 1H-NMR (CDCl3): δ 8.24 (s, 1H), 7.13 (s, 2H), 5.36 (s, 2H), 3.99 (s, 3H), 3.88 (s, 6H).
  • Example 93 N-[9-(2-Bromo-3,4,5-trimethoxy-benzyl)-6-chloro-9H-purin-2-yl]-acetamide
  • A suspension of 9-(2-bromo-3,4,5-trimethoxy-benzyl)-6-chloro-9H-purin-2-ylamine (example 51) in acetic anhydride was treated with a catalytic amount of conc. H2SO4 at r.t. for 3 h. Work-up and purification by preparative TLC (EtOAc:hexane 9:1) gave the title compound. HPLC Rt: 5.603 min. 1H-NMR (CDCl3): δ 8.20 (s, 1H), 8.10 (s, 1H), 7.00 (s, 1H), 5.47 (s, 2H), 3.92 (s, 3H), 3.90 (s, 3H), 3.86 (s, 3H), 2.51 (s, 3H).
  • Example 94 N-[9-(2-bromo-3,4,5-trimethoxy-benzyl)-6-chloro-9H-purin-2yl]-N-methyl-acetamide
  • A mixture of N-[9-(2-bromo-3,4,5-trimethoxy-benzyl)-6-chloro-9H-purin-2-yl]-acetamide and NaH in DMF was stirred at r.t. for 15 min, before adding MeI. Stirring was prolonged for 2 h at 50° C. Work-up and purification by preparative TLC (EtOAc:hexane 1:1) gave the title compound. HPLC Rt: 6.422 min. 1H-NMR (CDCl3): δ 8.20 (s, 1H), 6.80 (s, 1H), 5.45 (s, 2H), 3.93 (s, 3H), 3.90 (s, 3H), 3.82 (s, 3H), 3.57 (s, 3H), 2.51 (s, 3H).
  • Example 95 6-chloro-9-(3,5-dichloro-benzyl)-9H-purin-2-ylamine Step 1: 1-Bromomethyl-3,5-dichloro-benzene
  • The title compound was obtained from (3,5-dichloro-phenyl)-methanol according to the general procedure 2.5.
  • Step 2: 6-chloro-9-(3,5-dichloro-benzyl)-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 1-bromomethyl-3,5-dichloro-benzene. HPLC Rt: 6.074 min. 1H-NMR (Acetone-d6): δ 8.12 (s, 1H), 7.45-7.43 (t, 1H), 7.72-7.42 (d, 2H), 6.30 (s, 2H), 5.40 (s, 2H).
  • Example 96 6-chloro-9-(3,4-dichloro-benzyl)-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 4-bromomethyl-1,2-dichloro-benzene according to the general procedure 1.1. HPLC Rt: 5.982 min. 1H-NMR (CDCl3): δ 7.76 (s, 1H), 7.48-7.45 (d, 1H), 7.40-7.39 (d, 1H), 7.12-7.10 (dd, 1H), 5.23 (s, 2H), 5.12 (s, 2H).
  • Example 97 8-bromo-6-chloro-9-(3,4-dichloro-benzyl)-9H-purin-2-ylamine
  • The title compound was obtained by bromination of 6-chloro-9-(3,4-dichloro-benzyl)-9H-purin-2-ylamine (see previous example) according to the general procedure 1.2. HPLC Rt: 6.878 min. 1H-NMR (CDCl3): δ 7.45-7.43 (m, 2H), 7.17-7.14 (dd, 1H), 5.23 (s, 2H), 5.12 (s, 2H).
  • Example 98 6-chloro-9-(2-chloro-3,4,5-trimethoxy-benzyl)-9H-purin-2-ylamine
  • Chlorination of 6-chloro-9-(3,4,5-trimethoxy-benzyl)-9H-purin-2-ylamine according to the general procedure 3.1 gave a mixture of the title compound and of 6-chloro-9-(2,6-dichloro-3,4,5-trimethoxy-benzyl)-9H-purin-2-ylamine. The two compounds were isolated by preparative TLC. HPLC Rt: 5.626 min. 1H-NMR (CDCl3): δ 7.83 (s, 1H), 6.68 (s, 1H), 5.51 (s, 2H), 5.23 (s, 2H), 3.94 (s, 3H), 3.90 (s, 3H), 3.80 (s, 3H).
  • Example 99 6-chloro-9-(2,6-dichloro-3,4,5-trimethoxy-benzyl)-9H-purin-2-ylamine
  • See the previous example. HPLC Rt: 6.099 min. 1H-NMR (CDCl3): δ 7.57 (s, 1H), 5.48 (s, 2H), 5.12 (s, 2H), 3.99 (s, 3H), 3.92 (s, 6H).
  • Example 100 2-amino-9-(2-chloro-3,4,5-trimethoxy-benzyl)-9H-purin-6-ol
  • The title compound was obtained by heating a solution of 6-chloro-9-(2-chloro-3,4,5-trimethoxy-benzyl)-9H-purin-2-ylamine (see example 98in 1N HCl for 4 h. Solvent was evaporated and the residue was washed with EtOAc to give the title compound. HPLC Rt: 4.603 min. 1H-NMR (DMSO-d6): δ 7.82 (s, 1H), 6.65 (s, 1H), 6.60 (s, 2H), 5.15 (s, 2H), 3.81 (s, 3H), 3.77 (s, 3H), 3.69 (s, 3H).
  • Example 101 6-bromo-9-(3,4,5-trimethoxy-benzyl)-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 6-bromoguanine with 3-chloromethyl-3,4,5-trimethoxy benzene according to the general procedure 1.1. HPLC Rt: 4.947 min. 1H-NMR (CDCl3): δ 7.80 (s, 1H), 6.50 (s, 2H), 5.20 (s, 2H), 5.18 (s, 2H), 3.85 (s, 3H), 3.84 (s, 6H).
  • Example 102 6-bromo-9-(2-bromo-3,4,5-trimethoxy-benzyl)-9H-purin-2-ylamine
  • The title compound was obtained by bromination of 6-bromo-9-(3,4,5-trimethoxy-benzyl)-9H-purin-2-ylamine (see previous example) according to the general procedure 3.1. HPLC Rt: 5.793 min. 1H-NMR (CDCl3): δ 7.88 (s, 1H), 6.68 (s, 1H), 5.33 (s, 2H), 5.19 (s, 2H), 3.93 (s, 3H), 3.90 (s, 3H), 3.79 (s, 3H).
  • Example 103 9-(2-bromo-3,4,5-trimethoxy-benzyl)-6-ethylsulfanyl-9H-purin-2-ylamine
  • A mixture of 6-bromo-9-(2-bromo-3,4,5-trimethoxy-benzyl)-9H-purin-2-ylamine (see previous example), EtSH, K2CO3 and THF was placed in a pressure vessel and heated to 70° C. for 6 h. Work-up and purification by preparative TLC (EtOAc:hexane 1:1) gave the title compound. HPLC Rt: 6.039 min. 1H-NMR (CDCl3): δ 7.72 (s, 1H), 6.62 (s, 1H), 5.30 (s, 2H), 4.98 (s, 2H), 3.91 (s, 3H), 3.88 (s, 3H), 3.74 (s, 3H), 3.35-3.29 (q, 2H), 1.44-1.41 (t, 3H).
  • Example 104 9-(2-bromo-3,4,5-trimethoxy-benzyl)-6-methoxy-9H-purin-2-ylamine
  • A solution of 9-(2-bromo-3,4,5-trimethoxy-benzyl)-6-chloro-9H-purin-2-ylamine (see example 51) in MeOH was treated with MeONa at reflux for 1 h. Work-up and purification by preparative TLC (EtOAc:hexane 1:1) gave the title compound. HPLC Rt: 5.229 min. 1H-NMR (CDCl3): δ 7.69 (s, 1H), 6.58 (s, 1H), 5.33 (s, 2H), 4.88 (s, 2H), 4.11 (s, 3H), 3.93 (s, 3H), 3.89 (s, 3H), 3.74 (s, 3H).
  • Example 105 9-(2-bromo-3,4,5-trimethoxy-benzyl)-9H-purine-2,6-diamine
  • A solution of 9-(2-bromo-3,4,5-trimethoxy-benzyl)-6-chloro-9H-purin-2-ylamine (see example 51) in MeOH was treated with NH3 (7N in MeOH) in a pressure vessel at 90° C. for 16 h. Work-up and purification by preparative TLC (EtOAc:hexane 3:1) gave the title compound. HPLC Rt: 4.884 min. 1H-NMR (DMSO-d6): δ 7.67 (s, 1H), 6.71 (s, 2H), 6.60 (s, 1H), 5.84 (s, 2H), 5.17 (s, 2H), 3.80 (s, 3H), 3.76 (s, 3H), 3.66 (s, 3H).
  • Example 106 6-chloro-9-(2-iodo-3,4,5-trimethoxy-benzyl)-9H-purin-2-ylamine
  • The title compound was obtained by iodination of 6-chloro-9-(3,4,5-trimethoxy-benzyl)-9H-purin-2-ylamine (see example 48) according to the general procedure 3.1. HPLC Rt: 5.887 min. 1H-NMR (CDCl3): δ 7.87 (s, 1H), 6.67 (s, 1H), 5.33 (s, 2H), 5.22 (s, 2H), 3.91 (s, 3H), 3.88 (s, 3H), 3.77 (s, 3H).
  • Example 107 9-(2-bromo-3,5-dimethoxy-4-methoxymethoxy-benzyl)-6-chloro-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 4-(2-amino-6-chloro-purin-9-ylmethyl)-3-bromo-2,6-dimethoxy-phenol (see example 50) chloromethyl methyl ether according to the general procedure 2.6 (NaOH, THF, r.t.) HPLC Rt: 5.817 min. 1H-NMR (CDCl3): δ 7.90 (s, 1H), 6.70 (s, 1H), 5.35 (s, 2H), 5.32 (s, 2H), 5.17 (s, 2H), 3.93 (s, 3H), 3.78 (s, 3H), 3.62 (s, 3H).
  • Example 108 9-benzothiazol-2-ylmethyl-6-chloro-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 2-bromomethyl-benzothiazole according to the general procedure 1.1. HPLC Rt: 5.055 min. 1H-NMR (DMSO-d6): δ 8.34 (s,1H), 8.09-8.07 (dd, 1H), 7.98-7.96 (d, 1H), 7.53-7.50 (m, 1H), 7.47-7.43 (m, 1H), 7.01 (s, 2H), 5.81 (s, 2H).
  • Example 109 6-chloro-9-(4-methoxy-benzyl)-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 1-chloromethyl-4-methoxy-benzene according to the general procedure 1.1. HPLC Rt: 5.067 min. 1H-NMR (CDCl3): δ 7.69 (s, 1H), 7.22-7.20 (d, 2H), 6.88-6.86 (d, 2H), 5.22 (s, 2H), 5.17 (s, 2H), 3.79 (s, 3H).
  • Example 110 9-(2-bromo-4,5-dimethoxy-benzyl)-6-chloro-9H-purin-2-ylamine Step 1: (2-Bromo-4,5-dimethoxy-phenyl)-methanol
  • The title compound was obtained by bromination of (3,4-dimethoxy-phenyl)-methanol according to the general procedure 3.1. 1H-NMR (CDCl3): δ 7.03 (s, 1H), 7.03 (s, 1H), 4.70 (s, 2H), 3.91 (s, 3H), 3.89 (s, 3H)
  • Step 2: 1-Bromomethyl-2-chloro-4,5-dimethoxy-benzene
  • The title compound was obtained from (2-bromo-4,5-dimethoxy-phenyl)-methanol according to the general procedure 2.5. 1H-NMR (CDCl3): δ 7.03 (s, 1H), 6.94 (s, 1H), 4.60 (s, 2H), 3.89 (s, 3H), 3.86 (s, 3H)
  • Step 3: 9-(2-Bromo-4,5-dimethoxy-benzyl)-6-chloro-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 1-bromomethyl-2-chloro-4,5-dimethoxy-benzene according to the general procedure 1.1. HPLC Rt: 5.458 min. 1H-NMR (CDCl3): δ 7.84 (s, 1H), 7.08 (s, 1H), 6.93 (s, 1H), 5.30 (s, 2H), 5.15 (s, 2H), 3.90 (s, 3H), 3.82 (s, 3H).
  • Example 111 6-Chloro-9-(4-iodo-3,5-dimethyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine Step 1: 2,3,5-Collidine-N-oxide
  • The title compound was obtained by oxidation of 2,3,5-collidine according to the general procedure 2.1 (yield 70%). HPLC Rt: 3.964 min. 1H-NMR (CDCl3): δ 8.03 (s, 1H), 6.90 (s, 1H), 2.47 (s, 3H), 2.31 (s, 3H), 2.24 (s, 3H). m/z (%) 138.2 (M+1, 100%). Rf(20% MeOH/EtOAc) was 0.35.
  • Step 2: 2,3,5-Trimethyl-4-nitro-pyridine 1-oxide
  • A suspension of 2,3,5-collidine-N-oxide (3.77 g, 28 mmol) in conc. H2SO4 (8 mL) was cooled to 0° C. and fuming HNO3 (5 mL, 100 mmol) was added dropwise. The resulting transparent solution was stirred at 100° C. for 24 h, cooled to r.t., poured onto ice, and the pH was adjusted to 10. Work-up (CHCl3), drying (MgSO4), and evaporation gave the title compound, (97% yield, 97% purity). Rf (MeOH/EtOAc 1:9): 0.7. HPLC Rt: 4.756 min. 1H-NMR (CDCl3): δ 8.08 (s, 1H), 2.50 (s, 3H), 2.27 (s, 3H), 2.23 (s, 3H). m/z (%) 183.1 (M+1, 100%).
  • Step 3: 2,3,5-Trimethyl-pyridin-4-ylamine-1-oxide hydrochloride
  • A suspension of 2,3,5-trimethyl-4-nitro-pyridine 1-oxide (4.2 g, 23 mmol) and 10% Pd/C (0.42 g) in conc. aq. HCl/EtOH (1 :11) was treated with H2 (60 psi) at r.t. for 3 h. Filtration and evaporation gave the title compound as a slightly yellow solid. HPLC Rt: 4.756 min. 1H-NMR (DMSO-d6): δ 8.28 (s, 1H), 7.24 (s, 2H), 2.50 (s, 3H), 2.12 (s, 3H), 2.11 (s, 3H). m/z (%) 153.2 (M+1, 100%).
  • Step 4: 4-Iodo-23,5-trimethyl-pyridine-1-oxide
  • A solution of 2,3,5-Trimethyl-pyridin-4-ylamine-1-oxide hydrochloride (1.9 g, 10 mmol) HBF4 (20 mmol) in water (50 mL) was cooled to 0° C. A solution of NaNO2 (0.76 g, 11 mmol) in water (5 mL) was added dropwise to give a dark yellow solution which gradually gave a precipitate within 15 min. Potassium iodide (2.3 g, 1.39×10−2 mol) was added slowly in several portion to give a dark brown precipitate. The reaction mixture was stirred at r.t. for 5 min, and then heated to 60° C. for 10 min. The mixture was cooled to r.t. and the pH was adjusted to 10. Work-up (CHCl3), drying (MgSO4), evaporation, and flash chromatography gave the title compound. HPLC Rt: 5.579 min. 1H-NMR (CDCl3): δ 8.07 (s, 1H), 2.62 (s, 3H), 2.56 (s, 3H), 2.38 (s,3H). m/z (%) 264.1 (M+1, 100%).
  • Step 5: Acetic acid 4-iodo-3,5-dimethyl-pyridin-2-ylmethyl ester
  • The title compound was obtained by treating 4-iodo-3,5-dimethyl-pyridine 1-oxide was with Ac2O according to the general procedure 2.2. HPLC Rt: 2.913 min. 1H-NMR (CDCl3): δ 8.26 (s, 1H), 5.32 (s, 2H), 2.47 (s, 3H), 2.41 (s, 3H), 2.24 (s, 3H). m/z (%) 306.0 (M+1, 100%).
  • Step 6: (4-Iodo-3,5-dimethyl-pyridin-2-yl)-methanol
  • The title compound was obtained by deacetylation of 4-iodo-3,5-dimethyl-pyridin-2-ylmethyl ester according to the general procedure 2.3. HPLC Rt: 3.773 min. 1H-NMR (CDCl3): δ 8.15 (s, 1H), 4.70 (s, 2H), 2.46 (s, 3H), 2.40 (s, 3H). m/z (%) 264.1 (M+1, 100%).
  • Step 7: 2-Bromomethyl-4-iodo-3, 5-dimethyl-pyridine
  • The title compound was obtained from (4-Iodo-3,5-dimethyl-pyridin-2-yl)-methanol according to the general procedure 2.5. HPLC Rt: 5.957 min. 1H-NMR (CDCl3): δ 8.14 (s, 1H), 4.67 (s, 2H), 2.59 (s, 3H), 2.45 (s, 3H). m/z (%) 326.07 (M+1, 100%), 328.07 (M+1, 100%).
  • Step 8: 6-Chloro-9-(4-iodo-3, S-dimethyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 2-bromomethyl-4-iodo-3,5-dimethyl-pyridine according to the general procedure 1.1. HPLC Rt: 5.361 min. 1H-NMR (CDCl3): δ 8.08 (s, 1H), 7.86 (s, 1H), 5.41 (s, 2H), 5.04 (s, 2H), 2.57 (s, 3H), 2.41 (s, 3H).
  • Example 112 6-chloro-9-(4-methyl-quinolin-2-ylmethyl)-9H-purin-2-ylamine Step 1: 2,4-Dimethyl-quinoline 1-oxide
  • The title compound was obtained by oxidation of 2,4-dimethyl-quinoline according to the general procedure 2.1. HPLC Rt: 4.489 min. 1H-NMR (CDCl3): δ 8.89-8.07 (dd, 1H), 8.00-7.97 (dd, 1H), 7.82-7.79 (m, 1H), 7.69-7.65 (m, 1H), 7.20 (s, 1H), 2.73 (s, 3H), 2.69 (s, 3H).
  • Step 2: Acetic acid 4-methyl-quinolin-2-ylmethyl ester
  • The title compound was obtained by treating 2,4-dimethyl-quinoline-1-oxide with Ac2O according to the general procedure 2.2. HPLC Rt: 3.158 min. Rf (EtOAc/Hexane 1:1): 0.8.
  • Step 3: (4-Methyl-quinolin-2-yl)-methanol
  • The title compound was obtained by deacetylation of acetic acid 4-methyl-quinolin-2-ylmethyl ester according to the general procedure 2.3. HPLC Rt: 3.715 min. 1H-NMR (CDCl3): δ 8.08-8.06 (dd, 1H), 8.00-7.97 (dd, 1H), 7.73-7.69 (m, 1H), 7.57-7.54 (m, 1H), 7.12 (s, 1H), 4.87 (s, 2H), 4.52 (s, 1H), 2.70 (s, 3H).
  • Step 4: 2-bromomethyl-4-methyl-quinoline
  • The title compound was obtained from (4-methyl-quinolin-2-yl)-methanol according to the general procedure 2.5. HPLC Rt: 4.516 min. 1H-NMR (CDCl3): δ 8.07-8.05 (dd, 1H), 7.98-7.96 (dd, 1H), 7.73-7.69 (m, 1H), 7.58-7.54 (m, 1H), 7.40 (d, 1H), 4.66 (s, 2H), 2.70 (s, 3H).
  • Step 5: 6-chloro-9-(4-methyl-quinolin-2-ylmethyl)-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 2-bromomethyl-4-methyl-quinoline according to the general procedure 1.1. HPLC Rt: 4.387 min. 1H-NMR (DMSO-d6): δ 8.31 (s, 1H), 8.11-8.09 (dd, 1H), 7.90-7.88 (dd, 1H), 7.77-7.74 (m, 1H), 7.66-7.62 (m, 1H), 7.27 (s, 1H), 6.91 (s, 2H) 5.58 (s, 2H), 2.68 (s, 3H).
  • Example 113 6-bromo-9-(4-methyl-quinolin-2-ylmethyl)-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 6-bromoguanine with 2-bromomethyl-4-methyl-quinoline (see previous example) according to the general procedure 1.1. HPLC Rt: 4.489 min. 1H-NMR (DMSO-d6): δ 8.27 (s, 1H), 8.06-8.04 (dd, 1H), 7.85-7.83 (dd, 1H), 7.73-7.69 (m, 1H), 7.61-7.57 (m, 1H), 7.22 (s, 1H), 6.89 (s, 2H), 5.52 (s, 2H), 2.64 (s, 3H)
  • Example 114 6-bromo-9-(4-methyl-1-oxy-quinolin-2-ylmethyl)-9H-purin-2-ylamine
  • The title compound was obtained by oxidation of 6-bromo-9-(4-methyl-quinolin-2-ylmethyl)-9H-purin-2-ylamine (see previous example) according to the general procedure 2.1. HPLC Rt: 4.698 min. 1H-NMR (DMSO-d6): δ 8.62-8.60 (dd, 1H), 8.27 (s, 1H), 8.12-8.10 (dd, 1H), 7.88-7.85 (m, 1H), 7.79-7.75 (m, 1H), 6.93 (s, 2H), 6.89 (s, 1H) 5.57 (s, 2H), 2.64 (s, 3H).
  • Example 115 6-chloro-9-(3,5-dimethyl-4-methylsulfanyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine Step 1: 2,3,5-Trimethyl-4-methylsulfanyl-pyridine 1-oxide
  • A solution of 4-bromo-2,3,5-trimethyl-pyridine 1-oxide in THF was treated in a pressure vessel with NaSMe at 110° C. for 16 h. HPLC Rt: 5.303 min. 1H-NMR (CDCl3): δ 8.07 (s, 1H), 2.57 (s, 3H), 2.52 (s, 3H), 2.42 (s, 3H), 2.23 (s, 3H).
  • Step 2: Acetic acid 3,5-dimethyl-4-methylsulfanyl-pyridin-2-ylmethyl ester
  • The title compound was obtained by treating 2,3,5-trimethyl-4-methylsulfanyl-pyridine 1-oxide with Ac2O according to the general procedure 2.2. HPLC Rt: 4.341 min. 1H-NMR (CDCl3): δ 8.27 (s, 1H), 5.20 (s, 2H), 2.57 (s, 3H), 2.46 (s, 3H), 2.25 (s, 3H), 2.10 (s, 3H).
  • Step 3: (3,5-Dimethyl-4-methylsulfanyl-pyridin-2-yl)-methanol
  • The title compound was obtained by deacetylation of acetic acid 3,5-dimethyl-4-methylsulfanyl-pyridin-2-ylmethyl ester according to the general procedure 2.3. HPLC Rt: 3.921 min.
  • Step 4: 2-Bromomethyl-3,5-dimethyl-4-methylsulfanyl-pyridine
  • The compound was obtained from (3,5-dimethyl-4-methylsulfanyl-pyridin-2-yl)-methanol according to the general procedure 2.5. HPLC Rt: 4.905 min. 1HNMR (CDCl3): δ 8.26 (s, 1H), 4.59 (s, 2H), 2.62 (s, 3H), 2.47 (s, 3H), 2.27 (s, 3H), 2.10 (s, 3H). m/z (%): 246.13 (M+1, 96%), 248.09 (M+3, 100%).
  • Step 5: 6-Chloro-9-(3,5-dimethyl-4-methylsulfanyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine
  • The title compound was obtained by alkylation of 2-amino-6-chloropurine with 2-bromomethyl-3,5-dimethyl-4-methylsulfanyl-pyridine according to the general procedure 1.1. HPLC Rt: 4.611 min. 1H-NMR (CDCl3): δ 8.24 (s, 1H), 7.87 (s, 1H), 5.36 (s, 2H), 5.00 (s, 2H), 2.61 (s, 3H), 2.47 (s, 3H), 2.26 (s, 3H).
  • Example 116 6-chloro-9-(7-chloro-benzothiazol-2-ylmethyl)-9H-purin-2-ylamine Step 1: (2,3-dichloro-phenyl)-acetamide
  • A solution of 2,3-dichloroaniline (5.00 g, 30.86 mmol) in pyridine (20 ml) was cooled to 0° C. and treated with AcCl (4.85 g, 62 mmol). The reaction mixture was stirred at r.t. for 1 h and then concentrated under reduced pressure. The residue was dissolved in ethyl acetate, washed with 1% aqueous hydrochloric acid, water and brine, and dried over MgSO4. The solution was concentrated and recrystallized from EtOAc/hexane to give (2,3-dichloro-phenyl)-acetamide (4.50 g, 22 mmol). HPLC Rt: 5.52 min. 1H-NMR (CDCl3): δ 8.35 (br. s, 1H), 7.7 (1H), 7.24 (1H), 7.23 (1H), 2.28 (s, 3H).
  • Step 2: (2,3-dichloro-phenyl)-thioacetamide
  • A solution of (2,3-dichloro-phenyl)-acetamide (4.50 g, 22 mmol,) in toluene (50 ml) was treated at r.t. with P2S5 (9.80 g, 22 mmol). The reaction mixture was heated to 90° C. for 1.5 h, cooled to r.t., and filtered. The solid was washed with ether and the washings were combined with the filtrate. The combined solutions were extracted twice with 10% aq. NaOH. The combined aqueous extracts were acidified at 0° C. with HCl. The precipitate was collected and recrystallized from ethyl acetate/hexane to afford (2,3-dichloro-phenyl)-thioacetamide (3.40 g, 16 mmol). HPLC Rt: 5.91 min. 1H-NMR (CDCl3): δ 8.80 (br. s, 1H, NH), 8.5 (d, 1H), 7.42 (d, 1H), 7.30 (t, 1H), 2.82 (s, 3H).
  • Step 3: 7-Chloro-2-methyl-benzothiazole
  • A solution of (2,3-dichloro-phenyl)-thioacetamide (3.4 g, 15 mmol) in N-methyl-2-pyrrolidinone (25 ml) was treated with NaH (60% oil suspension, 0.74 g, 19 mmol) at r.t. The reaction mixture was heated to 150° C. for 30 min. Work-up (EtOAc), drying (brine, MgSO4), evaporation, and purification by flash chromatography afforded the title compound (2.4 g, 13 mmol). HPLC Rt: 6.65 min. 1H-NMR (CDCl3): δ 7.87 (d, 1H, J=7.9 Hz, ph-H), 7.44 (t, 1H, J=7.9 Hz, ph-H), 7.35 (d, 1H, J=8.0 Hz, ph-H), 2.87 (s, 3H, CH3).
  • Step 4: 2-Bromomethyl-7-chloro-benzothiazole
  • A mixture of 7-chloro-2-methyl-benzothiazole (1.00 g, 5.45 mmol), N-bromosuccinimide (1.26 g, 7.08 mmol), benzoyl peroxide (0.1 g), and CCl4 (10 mL was heated to reflux under irradiation of a UV lamp for 14 h. The reaction mixture was cooled and filtered to remove the succinimide formed in the reaction, and the filtrate was evaporated to dryness. The resulting solid was purified by flash chromatography to give the title lamp (400 mg, 1.5 mmol). 1H-NMR (CDCl3): δ 7.94 (d, 1H), 7.47 (t, 1H), 7.42 (d, 1H), 4.82 (s, 2H, CH2). HPLC Rt: 7.19 min.
  • Step 5: 6-Chloro-9-(7-chloro-benzothiazol-2-ylmethyl)-9H-purin-2-ylamine
  • A mixture of 2-bromomethyl-7-chloro-benzothiazole (60 mg, 0.2286 mmol), 2-amino-6-chloropurine (32 mg, 0.19 mmol), Cs2CO3 (67.86 mg, 0.208 mmol), and DMF (2 ml) was heated to 40° C. for 1 h. The reaction was cooled to r.t. and the solvent was removed on a rotary evaporator. The resulting solid was purified by preparative TLC to give the title compound (50 mg, 0.14 mmol). HPLC Rt: 5.81 min. 1H-NMR (CDCl3): δ 8.0 (s, 1H), 7.95 (d, 1H), 7.48 (t, 1H), 7.43 (d, 1H), 5.69 (s, 2H), 5.17 (s, 2H).
  • Example 117 6-Chloro-9-(3,4,5-trimethyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine Step 1: 2,3,5-Collidine-N-oxide
  • See Example 1, Method 1, step 1.
  • Step 2: 4-Bromo-2,3,5-collidine-N-oxide
  • See Example 1, Method 1, step 2.
  • Step 3: 2,3,4,5-Tetramethyl-pyridine 1-oxide
  • 4-Bromo-2,3,5-trimethyl-pyridine 1-oxide (2 g, 9.2 mmole) and catalytic tetrakis(triphenylphosphino)-palladium (80 mg, 4% by wt) in 20 mL of dry THF before treating with trimethylaluminum (2M in toluene, 15.2 mmole) under nitrogen. The solution was heated to reflux for 3 h, diluted with toluene (20 mL) before quenching the reaction with 4 mL of methanol followed by ammonium chloride (15 mmole). The mixture was refluxed for 2 h and filtered, while hot, on Celite. See J. Med Chem. 1999, 42(12), 2064-2086. HPLC Rt: 4.183 min. 1H-NMR (CDCl3): δ 8.05 (s, 1H), 2.55 (s, 3H), 2.27 (s, 3H), 2.22 (s, 3H), 2.20 (s, 3H).
  • Step 4: Acetic acid 3,4,5-trimethyl-pyridin-2-ylmethyl ester
  • The compound was obtained by dissolving in acetic anhydride before bringing the reaction to reflux for 0.5 h as described in the general procedure 2.2. Quenching the reaction with water and extracting with chloroform yielded the titled product. HPLC Rt: 3.843 min. 1H-NMR (CDCl3): δ 8.20 (s, 1H), 5.22 (s, 2H), 2.26 (s, 3H), 2.25 (s, 3H), 2.21 (s, 3H), 2.11 (s, 3H).
  • Step 5: (3,4,5-Trimethyl-pyridin-2-yl)-methanol
  • The compound was obtained by hydrolysis of acetic acid 3,4,5-trimethyl-pyridin-2-ylmethyl ester in MeOH and K2CO3 at 50° C. for 0.5 h as described in the general procedure 2.3. After removing MeOH, the residue was dissolved in water and extracted with chloroform. HPLC Rt: 3.405 min. 1H-NMR (CDCl3): δ 8.18 (s, 1H), 5.00 (s, 1H), 4.67 (s, 2H), 2.28 (s, 3H), 2.23 (s, 3H), 2.12 (s, 3H).
  • Step 6: 2-bromomethyl-3,4,5-trimethyl-pyridine
  • The compound was obtained from reacting (3,4,5-trimethyl-pyridin-2-yl)-methanol with triphenyl phosphine and carbon tetrabromide in dichloromethane as described in the general procedure 2.5. HPLC Rt: 3.979 min. 1H-NMR (CDCl3): δ 8.18 (s, 1H), 4.63 (s, 2H), 2.35 (s, 3H), 2.48 (s, 3H), 2.24 (s, 3H).
  • Step 7: 6-Chloro-9-(3,4,5-trimethyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine
  • The compound was obtained by reacting 2-bromomethyl-3,4,5-trimethyl-pyridine with 6-chloro-9H-purin-2-ylamine in the presence of K2CO3 at 50° C. for 0.5 h as described in the general procedure 1.1. HPLC Rt: 3.903 min. 1H-NMR (CDCl3): δ 8.18 (s, 1H), 7.84 (s, 1H), 5.38 (s, 2H), 5.08 (s, 2H), 2.29 (s, 3H), 2.27 (s, 3H), 2.22 (s, 3H).
  • Example 118 6-Bromo-9-(3,4,5-trimethyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine
  • The compound was obtained by reacting 2-bromomethyl-3,4,5-trimethyl-pyridine (see example 117) with 6-bromo-9H-purin-2-ylamine in the presence of K2CO3 in DMF for 0.5 h at 50° C. as described in the general procedure 1.1. HPLC Rt: 4.045 min. 1H-NMR (CDCl3): δ 8.18 (s, 1H), 7.85 (s, 1H), 5.37 (s, 2H), 5.10 (s, 2H), 2.29 (s, 3H), 2.27 (s, 3H), 2.22 (s, 3H).
  • Example 119 6-Bromo-9-(3,4,5-trimethyl-1-oxy-pyridin-2-ylmethyl)-9H-purin-2-ylamine
  • The compound was obtained by the oxidation of 6-bromo-9-(3,4,5-trimethyl-pyridin-2-ylmethyl)-9H-purin-2-ylamine with m-CPBA in dichloromethane as described in the general procedure 2.1. HPLC Rt: 5.611 min. 1H-NMR (CDCl3): δ 9.13 (s, 1H), 8.08 (s, 1H), 5.91 (s, 2H), 2.70 (s, 3H), 2.27 (s, 3H), 2.22 (s, 3H).
  • BIOLOGY EXAMPLES Example A
  • rHSP90 Competitive Binding Assay
  • Five microgram of purified rHSP90 protein (Stressgen, BC, Canada, #SPP-770) in phosphated buffered saline (PBS) was coated on 96 well plates by incubating overnight at 4° C. Unbound protein was removed and the coated wells were washed twice with 200 μL PBS. DMSO controls (considered as untreated samples) or test compounds were then added at 100-30-10-3-1-0.3 μM dilutions (in PBS), the plates mixed for 30 seconds on the plate shaker, and then incubated for 60 min. at 37° C. The wells were washed twice with 200 μL PBS, and 10 μM biotinylated-geldanamycin (biotin-GM) was added and incubated for 60 min. at 37° C. The wells were washed again twice with 200 μL PBS, before the addition of 20 μg/mL streptavidin-phycoerythrin (streptavidin-PE) (Molecular Probes, Eugene, Oreg.) and incubation for 60 min. at 37° C. The wells were washed again twice with 200 μL PBS. Relative fluorescence units (RFU) was measured using a SpectraMax Gemini XS Spectrofluorometer (Molecular Devices, Sunnyvale, Calif.) with an excitation at 485 nm and emission at 580 nm; data was acquired using SOFTmax®PRO software (Molecular Devices Corporation, Sunnyvale, Calif.). The background was defined as the RFU generated from wells that were not coated with HSP90 but were treated with the biotin-GM and streptavidin-PE. The background measurements were substrated from each sample treated with biotin-GM and streptavidin-PE measurements before other computation. Percent inhibition of binding for each sample was calculated from the background subtracted values as follows:
    % binding inhibition=[RFU untreated−RFU treated]/RFU untreated]×100.
  • Example B Cell Lysate Binding Assay
  • MCF7 breast carcinoma cell lysates were prepared by douncing in lysing buffer (20 mM HEPES, pH 7.3, 1 mM EDTA, 5 mM MgCl2, 100 mM KCl), and then incubated with or without test compound for 30 mins at 4° C., followed by incubation with biotin-GM linked to BioMag™ streptavidin magnetic beads (Qiagen) for 1 hr at 4° C. The tubes were placed on a magnetic rack, and the unbound supernatant removed. The magnetic beads were washed three times in lysis buffer and boiled for 5 mins at 95° C. in SDS-PAGE sample buffer. Samples were analyzed on SDS protein gels, and Western blots done for rHSP90. Bands in the Western Blots were quantitated using the Bio-rad Fluor-S Multihnager, and the % inhibition of binding of rHSP90 to the biotin-GM was calculated.
  • The lysate binding ability of selected compounds of the invention based on the above assay is summarized in Table 2. The IC50 reported is the concentration of test compound needed to achieve 50% inhibition of the biotin-GM binding to rHSP90 in the MCF7 cell lysates.
  • Example C HER2 Degradation Assay
  • MCF7 breast carcinoma cells (ATCC) were grown in Dulbecco's modified Eagle's medium (DMEM) containing 10% fetal bovine serum (FBS) and 10 mM HEPES, and plated in 24 well plates (50% confluent). Twenty-four hrs later (cells are 65-70% confluent), test compounds were added and incubated overnight for 16 h. For the less potent compounds, the amounts added were 100 μM, 30 μM, 10 μM and 1 μM, and for more potent compounds, the amounts added were 1 μM, 0.3 μM, 0.1 μM, 0.03 μM, 0.01 μM and 0.003 μM. The wells were washed with 1 mL phosphate buffered saline (PBS), and 200 μL trypsin was added to each well. After trypsinization was complete, 50 μL of FBS was added to each well. Then 200 μL cells was transferred to 96 well plates. The cells were pipetted up and down to obtain a single cell suspension. The plates were centrifuged at 2,500 rpm for 1 min using a Sorvall Legend RT™ tabletop centrifuge (Kendro Laboratory Products, Asheville, N.C.). The cells were then washed once in PBS containing 0.2% BSA and 0.2% sodium azide (BA buffer). Phycoerythrin (PE) conjugated anti HER2/Neu antibody (Becton Dickinson, #340552), or PE conjugated anti-keyhole limpet hemacyanin [KLH] (Becton Dickinson, #340761) control antibody was added at a dilution of 1:20 and 1:40 respectively (final concentration was 1 μg/mL) and the cells were pipetted up and down to form a single cell suspension, and incubated for 15 mins. The cells were washed twice with 200 μL BA buffer, and resuspended in 200 μL BA buffer, and transferred to FACSCAN tubes with an additional 250 μL BA buffer. Samples were analyzed using a FACSCalibur™ flow cytometer (Becton Dickinson, San Jose, Calif.) equipped with Argon-ion laser that emits 15 mW of 488 nm light for excitation of the PE fluorochrome. 10,000 events were collected per sample. A fluorescence histogram was generated and the mean fluorescence intensity (MFI) of each sample was determined using Cellquest software. The background was defined as the MFI generated from cells incubated with control IgG-PE, and was subtracted from each sample stained with the HER2/Neu antibody. Cells incubated with DMSO was always done as untreated controls since the compounds were resuspended in DMSO. Percent degradation of HER2 was calculated as follows:
    % HER2 degraded=[(MFl untreated cells−MFl treated cells)/MFl untreated cell]×100
  • The HER2 degradation ability of selected compounds of the invention based on this assay is summarized in Table 2. IC50 is defined as the concentration at which there was 50% degradation of the HER2/Neu protein.
  • Example D MTS Assay
  • MTS assays measures the cytotoxicity of geldanamycin derivatives. MTS (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)2-(4-sulfophenyl)-2H-tetrazolium is a tetrazolium dye that is converted to a formazan product by dehydrogenase enzymes of metabolically active cells (Corey, A. et al. “Use of an aqueous soluble tetrazolium/formazan assay for cell growth assays in culture,” Cancer Commun. 1991, 3, 207-212). Cells were seeded in 96 well plates at 2000 cells/well and allowed to adhere overnight in Dulbecco's modified Eagle's medium supplemented with 10% fetal bovine serum. The final culture volume was 100 μl. Viable cell number was determined by using the Celltiter 96 AQueousNon-radioactive Cell Proliferation Assay (Promega, Madison Wis.). The MTS/PMS (phenazine methosulfate) solution was mixed at a ratio of 20:1, and 20 μL was added per well to 100 μl of culture medium. After 2-4 hours, the formation of the formazan product was measured at 490 nm absorbance using a multiwell plate spectrophotometer. Background was determined by measuring the Abs 490 nm of cell culture medium and MTS-PMS in the absence of cells and was subtracted from all values. Percent viable cells was calculated as follows:
    % viable cells=(Abs at 490 nm treated cells/Abs at 490 nm untreated cells)×100
  • The effect of selected compounds of the invention on MCF7 breast carcinoma cells according to the MTS assay is summarized in Table 2. IC50 was defined as the concentration of the compound which gave rise to 50% viable cell number.
    TABLE 2
    Biological Activities of Selected Compounds of Formula II,
    where R2 = NH2, R3 = H
    II
    Figure US20070185064A1-20070809-C00022
    Figure US20070185064A1-20070809-P00899
    Figure US20070185064A1-20070809-P00899
    Figure US20070185064A1-20070809-P00899
    Figure US20070185064A1-20070809-P00899
    Figure US20070185064A1-20070809-P00899
    Figure US20070185064A1-20070809-P00899
    Figure US20070185064A1-20070809-P00899
    Figure US20070185064A1-20070809-P00899
    1 51
    Figure US20070185064A1-20070809-C00023
    Cl 0.7 0.025 0.055 0.2
    2 98
    Figure US20070185064A1-20070809-C00024
    Cl 0.7 0.03 0.025 0.1
    3 102
    Figure US20070185064A1-20070809-C00025
    Br 0.9 0.022 0.055 0.1
    4 77
    Figure US20070185064A1-20070809-C00026
    Br ND ND 0.10 0.3
    5 44
    Figure US20070185064A1-20070809-C00027
    Cl ND ND 0.08 2.0
    6 25
    Figure US20070185064A1-20070809-C00028
    Cl 0.9 0.021 0.030 0.2
    7 31
    Figure US20070185064A1-20070809-C00029
    Cl 0.8 0.030 0.060 5.0
    8 21
    Figure US20070185064A1-20070809-C00030
    Br 0.7 0.030 0.045 0.3
    9 41
    Figure US20070185064A1-20070809-C00031
    Cl ND ND 0.035 0.2
    10 33
    Figure US20070185064A1-20070809-C00032
    Cl ND ND 0.09 0.5
    11 18
    Figure US20070185064A1-20070809-C00033
    Cl ND ND 0.055 0.1
    12 1
    Figure US20070185064A1-20070809-C00034
    Cl ND ND 0.015 0.02
    13 4
    Figure US20070185064A1-20070809-C00035
    Cl ND ND 0.020 0.13
    14 24
    Figure US20070185064A1-20070809-C00036
    Br ND ND 0.060 0.4
    15 19
    Figure US20070185064A1-20070809-C00037
    Cl ND ND 0.080 0.4
    16 5
    Figure US20070185064A1-20070809-C00038
    Br ND 0.008 0.014 0.02
    17 6
    Figure US20070185064A1-20070809-C00039
    Br ND ND 0.060 0.05
    18 111
    Figure US20070185064A1-20070809-C00040
    Cl ND 0.003 0.009 0.02
    19 115
    Figure US20070185064A1-20070809-C00041
    Cl ND ND 0.060 ND

    ND = not determined
  • The foregoing examples are not limiting and are merely illustrative of various aspects and embodiments of the present invention. All documents cited herein are indicative of the levels of skill in the art to which the invention pertains and are incorporated by reference herein in their entireties. None, however, is admitted to be prior art.
  • One skilled in the art will readily appreciate that the present invention is well adapted to carry out the objects and obtain the ends and advantages mentioned, as well as those inherent therein. The methods and compositions described illustrate preferred embodiments, are exemplary, and are not intended as limitations on the scope of the invention. Certain modifications and other uses will occur to those skilled in the art, and are encompassed within the spirit of the invention, as defined by the scope of the claims.
  • The invention illustratively described herein suitably may be practiced in the absence of any element or elements, limitation or limitations which is not specifically disclosed herein. The terms and expressions which have been employed are used as terms of description and not of limitation, and there is no intention in the use of such terms and expressions of excluding any equivalents of the features shown and described, or portions thereof. It is recognized that various modifications are possible within the scope of the invention claimed. Thus, it should be understood that although the present invention has been specifically disclosed by preferred embodiments, optional features, modifications and variations of the concepts herein disclosed may be resorted to by those skilled in the art, and that such modifications and variations are considered to be within the scope of this invention as defined by the description and the appended claims.
  • In addition, where features or aspects of the invention are described in terms of Markush groups or other grouping of alternatives, e.g., genuses, those skilled in the art will recognize that the invention is also thereby described in terms of any individual member or subgroup of members of the Markush group or subgenus, and exclusions of individual members as appropriate, e.g, by proviso.
  • Other embodiments are within the following claims.

Claims (72)

1. A compound represented by Formula I, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof:
Figure US20070185064A1-20070809-C00042
wherein:
R1 is halogen or lower alkyl;
R2 is —NR8R10;
R4 is —CHR12—;
R3is hydrogen, halogen, or —CN;
R5 is aryl, heteroaryl, alicyclic, or heterocyclic, wherein:
the aryl group is substituted with 3 to 5 substituents,
the heteroaryl group is substituted with 2 to 5 substituents, wherein when the heteroaryl is substituted with only two substituents, the two substituents must form part of an optionally substituted fused ring,
the alicyclic group is substituted with 3 to 5 substituents,
the heterocyclic group is substituted with 3 to 5 substituents, and
the substituents are selected from the group of halogen, lower alkyl, lower alkenyl, lower alkynyl, lower aryl, lower alicyclic, arylalkyl, aryloxy, aryloxyalkyl, alkoxyalkyl, perhaloalkyl, perhaloalkyloxy, perhaloacyl, —N3, —SR8, —OR8, —CN, —C(O)R9, —NO2, —NR8R10, phosphonate and phosphonic acid;
R8 is hydrogen, lower alkyl, lower aryl, or —C(O)R9;
R9 is lower alkyl, lower aryl, lower heteroaryl, —NR10R10 or —OR11;
R10 is independently hydrogen or lower alkyl;
R11 is lower alkyl, lower aryl or lower heteroaryl;
R12 is hydrogen or lower alkyl;
provided that
when R5 is aryl, R5 is not an organo-metallic cyclopentadiene;
when R5 is phenyl, the substituents are not 3,5 di-halo;
when R5 is alicyclic, the ring system does not contain any tetra-substituted sp3 ring carbons; and
when R5 is heterocyclic, the ring system does not contain any tetra-substituted sp3 ring carbons or the ring system is not a tetra-substituted pyrrolidine.
2. The compound of claim 1, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof, wherein:
R1 is halogen or methyl; and
R2 is —NHR8, where R8 is hydrogen or —C(O)R9.
3. The compound of claim 1, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof, wherein:
R1 is halogen;
R2 is —NH2;
R3 is hydrogen; and
R5 is aryl or heteroaryl, wherein
each of said aryl and heteroaryl groups is monocyclic or bicyclic,
the aryl group is substituted with 4 to 5 substituents,
the heteroaryl group is substituted with 2 to 5 substituents, wherein when the heteroaryl is substituted with only two substituents, the two substituents must form part of an optionally substituted fused ring.
4. The compound of claim 1, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof, wherein:
R1 is halogen;
R2 is —NH2;
R4 is lower alkyl;
R3 is hydrogen; and
R5 is aryl or heteroaryl, wherein
each of said aryl and heteroaryl groups is monocyclic or bicyclic,
the aryl group is substituted with 4 to 5 substituents,
the heteroaryl group is substituted with 2 to 5 substituents, wherein when the heteroaryl is substituted with only two substituents, the two substituents must form part of an optionally substituted fused ring.
5. The compound of claim 1, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof, wherein
R1 is chloro or bromo,
R2 is —NH2, and
R5 is a phenyl having 3 to 5 substituents, a pyridyl having 3 to 5 substituents or an 1-oxy-pyridyl (N-oxy-pyridyl) having 3 to 5 substituents.
6. A compound represented by Formula II, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof:
Figure US20070185064A1-20070809-C00043
wherein:
R1 is halogen or lower alkyl;
R2 is —NR8R10;
R3 is hydrogen, halogen, or —CN;
R5 is aryl, heteroaryl, alicyclic, or heterocyclic, wherein
the aryl group is substituted with 3 to 5 substituents,
the heteroaryl group is substituted with 2 to 5 substituents, wherein when the heteroaryl is substituted with only two substituents, the two substituents must form part of an optionally substituted fused ring,
the alicyclic group is substituted with 3 to 5 substituents,
the heterocyclic group is substituted with 3 to 5 substituents, and
the substituents are selected from the group of halogen, lower alkyl, lower alkenyl, lower alkynyl, lower aryl, lower alicyclic, arylalkyl, aryloxy, aryloxyalkyl, alkoxyalkyl, perhaloalkyl, perhaloalkyloxy, perhaloacyl, —N3, —SR8, —OR8, —CN, —C(O)R9, —NO2, —NR8R10, phosphonate and phosphonic acid;
R8 is hydrogen, lower alkyl, lower aryl, or —C(O)R9;
R9 is lower alkyl, lower aryl, lower heteroaryl, —NR10R10 or —OR11;
R10 is independently hydrogen or lower alkyl; and
R11 is lower alkyl, lower aryl or lower heteroaryl;
provided that
when R5 is aryl, R5 is not an organo-metallic cyclopentadiene;
when R5 is phenyl, the substituents are not 3,5 di-halo;
when R5 is alicyclic, the ring system does not contain any tetra-substituted sp3 ring carbons;
when R5 is heterocyclic, the ring system does not contain any tetra-substituted sp3 ring carbons or the ring system is not a tetra-substituted pyrrolidine.
7. The compound of claim 6, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof, wherein each of said aryl, heteroaryl, alicyclic or heterocyclic group is monocyclic or bicyclic.
8. The compound of claim 6, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof, wherein:
R1 is halogen or methyl; and
R2 is —NHR8, where R8 is hydrogen or —C(O)R9.
9. The compound of claim 8, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof, wherein R1 is halogen.
10. The compound of claim 9, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof, wherein R2 is —NH2 and R3 is hydrogen.
11. The compound of claim 6, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof, wherein:
R1 is halogen;
R2 is —NH2;
R3 is hydrogen; and
R5 is aryl or heteroaryl, wherein
each of the aryl and heteroaryl groups is monocyclic or bicyclic,
the aryl group is substituted with 4 to 5 substituents,
the heteroaryl group is substituted with 2 to 5 substituents, wherein when the heteroaryl is substituted with only two substituents, the two substituents must form part of an optionally substituted fused ring.
12. The compound of claim 6, or a polymorph, solvate , ester, tautomer, pharmaceutically acceptable salt or prodrug thereof, wherein R1 is chloro or bromo, R2 is —NH2, and R5 is a phenyl having 3 to 5 substituents.
13. The compound of claim 6, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof, wherein R1 is chloro or bromo, R2 is —NH2, and R5 is a pyridyl having 3 to 5 substituents.
14. The compound of claim 6, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof, wherein R1 is chloro or bromo, R2 is —NH2, and R5 is an 1-oxy-pyridyl (N-oxy-pyridyl) having 3 to 5 substituents.
15. The compound of claim 11, wherein said compound is a member selected from the group below, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof:
Figure US20070185064A1-20070809-C00044
Figure US20070185064A1-20070809-C00045
16. The compound of claim 11, wherein said compound is a member selected from the group below, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof:
Figure US20070185064A1-20070809-C00046
Figure US20070185064A1-20070809-C00047
17. The compound of claim 11, wherein said compound is a member selected from the group below, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof:
Figure US20070185064A1-20070809-C00048
18. The compound of claim 11, wherein said compound is represented by the formula below, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof:
Figure US20070185064A1-20070809-C00049
19. The compound of claim 11, wherein said compound is represented by the formula below, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof:
Figure US20070185064A1-20070809-C00050
20. The compound of claim 11, wherein said compound is represented by the formula below, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof:
Figure US20070185064A1-20070809-C00051
21. The compound of claim 11, wherein said compound is represented by the formula below, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof:
Figure US20070185064A1-20070809-C00052
22. The compound of claim 11, wherein the compound is a member selected from the group of compounds below, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof:
Figure US20070185064A1-20070809-C00053
Figure US20070185064A1-20070809-C00054
Figure US20070185064A1-20070809-C00055
Figure US20070185064A1-20070809-C00056
23. The compound of claim 11, wherein said compound is a member selected from the group below, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof:
Figure US20070185064A1-20070809-C00057
24. The compound of claim 11, wherein said compound is a member selected from the group below, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof:
Figure US20070185064A1-20070809-C00058
25. The compound of claim 11, wherein said compound is a member selected from the group below, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof:
Figure US20070185064A1-20070809-C00059
26. The compound of claim 11, wherein said compound is a member selected from the group below, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof:
Figure US20070185064A1-20070809-C00060
27. The compound of claim 11, wherein said compound is a member selected from the group below, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof:
Figure US20070185064A1-20070809-C00061
28. The compound of claim 11, wherein said compound is a member selected from the group below, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof:
Figure US20070185064A1-20070809-C00062
29. The compound of claim 11, wherein said compound is represented by the formula below, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof:
Figure US20070185064A1-20070809-C00063
30. The compound of claim 11, wherein said compound is represented by the formula below, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof:
Figure US20070185064A1-20070809-C00064
31. The compound of claim 11, wherein said compound is represented by the formula below, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof:
Figure US20070185064A1-20070809-C00065
32. The compound of claim 11, wherein said compound is represented by the formula below, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof:
Figure US20070185064A1-20070809-C00066
33. The compound of claim 11, wherein said compound is represented by the formula below, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof:
Figure US20070185064A1-20070809-C00067
34. The compound of claim 11, wherein said compound is represented by the formula below, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof:
Figure US20070185064A1-20070809-C00068
35. The compound of claim 11, wherein said compound is represented by the formula below, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof:
Figure US20070185064A1-20070809-C00069
36. The compound of claim 11, wherein said compound is represented by the formula below, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof:
Figure US20070185064A1-20070809-C00070
37. The compound of claim 11, wherein said compound is represented by the formula below, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof:
Figure US20070185064A1-20070809-C00071
38. The compound of claim 11, wherein said compound is represented by the formula below, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof:
Figure US20070185064A1-20070809-C00072
39. The compound of claim 11, wherein said compound is represented by the formula below, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof:
Figure US20070185064A1-20070809-C00073
40. The compound of claim 11, wherein said compound is represented by the formula below, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof:
Figure US20070185064A1-20070809-C00074
41. The compound of claim 11, wherein said compound is a member selected from the group below, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof:
Figure US20070185064A1-20070809-C00075
42. The compound of claim 11, wherein said compound is a member selected from the group below, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof:
Figure US20070185064A1-20070809-C00076
43. The compound of claim 11, wherein said compound is a member selected from the group below, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof:
Figure US20070185064A1-20070809-C00077
44. The compound of claim 11, wherein said compound is represented by the formula below, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof:
Figure US20070185064A1-20070809-C00078
45. The compound of claim 11, wherein said compound is represented by the formula below, or a polymorph, solvate, ester tautomer, pharmaceutically acceptable salt or prodrug thereof:
Figure US20070185064A1-20070809-C00079
46. The compound of claim 11, wherein said compound is represented by the formula below, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof:
Figure US20070185064A1-20070809-C00080
47. The compound of claim 11, wherein said compound is represented by the formula below, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof:
Figure US20070185064A1-20070809-C00081
48. The compound of claim 11, wherein said compound is represented by the formula below, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof:
Figure US20070185064A1-20070809-C00082
49. The compound of claim 11, wherein said compound is represented by the formula below, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof:
Figure US20070185064A1-20070809-C00083
50. The compound of claim 11, wherein said compound is represented by the formula below, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof:
Figure US20070185064A1-20070809-C00084
51. The compound of claim 11, wherein said compound is represented by the formula below, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof:
Figure US20070185064A1-20070809-C00085
52. The compound of claim 11, wherein said compound is a member selected from the group below, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof:
Figure US20070185064A1-20070809-C00086
53. The compound of claim 11, wherein said compound is a member selected from the group below, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof:
Figure US20070185064A1-20070809-C00087
54. The compound of claim 11, wherein said compound is a member selected from the group below, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof:
Figure US20070185064A1-20070809-C00088
55. A pharmaceutical composition comprising one or more pharmaceutical acceptable excipients and at least one compound represented by Formula I below, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof:
Figure US20070185064A1-20070809-C00089
wherein:
R1 is halogen or lower alkyl;
R2 is —NR8R10;
R4 is —CHR12—;
R3 is hydrogen, halogen, or —CN;
R5 is aryl, heteroaryl, alicyclic, or heterocyclic, wherein
the aryl group is substituted with 3 to 5 substituents,
the heteroaryl group is substituted with 2 to 5 substituents, wherein when the heteroaryl is substituted with only two substituents, the two substituents must form part of an optionally substituted fused ring,
the alicyclic group is substituted with 3 to 5 substituents,
the heterocyclic group is substituted with 3 to 5 substituents, and
the substituents are selected from the group of halogen, lower alkyl, lower alkenyl, lower alkynyl, lower aryl, lower alicyclic, arylalkyl, aryloxy, aryloxyalkyl, alkoxyalkyl, perhaloalkyl, perhaloalkyloxy, perhaloacyl, —N3, —SR8, —OR8, —CN, —C(O)R9, —NO2, —NR8R10, phosphonate and phosphonic acid;
R8 is hydrogen, lower alkyl, lower aryl, or —C(O)R9;
R9 is lower alkyl, lower aryl, lower heteroaryl, —NR10R10 or —OR11;
R10 is independently hydrogen or lower alkyl;
R11 is lower alkyl, lower aryl or lower heteroaryl;
R12 is hydrogen or lower alkyl; and
provided that
when R5 is aryl, R5 is not an organo-metallic cyclopentadiene;
when R5 is phenyl, the substituents are not 3,5 di-halo;
when R5 is alicyclic, the ring system does not contain any tetra-substituted sp3 ring carbons; and
when R5 is heterocyclic, the ring system does not contain any tetra-substituted sp3 ring carbons or the ring system is not a tetra-substituted pyrrolidine.
56. The pharmaceutical composition of claim 55 wherein R4 is —CH2—.
57. The pharmaceutical composition of claim 55, wherein:
R1 is halogen;
R2 is —NH2;
R3 is hydrogen; and
R5 is aryl or heteroaryl, wherein
each of the aryl and heteroaryl groups is monocyclic or bicyclic,
the aryl group is substituted with 4 to 5 substituents,
the heteroaryl group is substituted with 2 to 5 substituents, wherein when the heteroaryl is substituted with only two substituents, the two substituents must form part of an optionally substituted fused ring.
58. The pharmaceutical composition of claim 56 wherein, wherein
R1 is chloro or bromo;
R2 is —NH2; and
R5 is selected from a phenyl having 3 to 5 substituents, a pyridyl having 3 to 5 substituents and an 1-oxy-pyridyl (N-oxy-pyridyl) having 3 to 5 substituents.
59. A method of treating an gingival having an HSP90 mediated disorder comprising administering to said individual a pharmaceutical composition comprising a pharmaceutically effective amount of a compound of Formula I:
Figure US20070185064A1-20070809-C00090
or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof, wherein:
R1 is halogen, —OR11, SR11, —NHR8, hydrogen, or lower alkyl;
R2 is —NR8R10;
R3 is hydrogen, halogen, —N3 or —CN;
R1 is —(CHR12)n— where n=0, 1 or 2, —C(O)—, —C(S)—, or —S(O)—;
R5 is alkyl, aryl, heteroaryl, alicyclic, or heterocyclic, all optionally substituted with halogen, lower alkyl, lower alkenyl, lower alkynyl, lower aryl, lower alicyclic, arylalkyl, aryloxy, aryloxyalkyl, alkoxyalkyl, perhaloalkyl, perhaloalkyloxy, perhaloacyl, —N3, —SR8, —OR8, —CN, —C(O)R9, —NO2, or —NR8R10;
R8 is hydrogen, lower alkyl, lower aryl, or —C(O)R9;
R9 is lower alkyl, lower aryl, lower heteroaryl, —NR10R10, or —OR11;
R10 is independently hydrogen or lower alkyl;
R11 is lower alkyl, lower aryl or lower heteroaryl;
R12 is hydrogen or lower alkyl; and
provided that:
—R4R5 is not a ribose or derivative thereof, or a sugar or derivative thereof;
—R4R5 is not a phosphonate or phosphonic acid, or substituted with phosphonate or phosphonic acid; and
when R4 is —(CH2)n— where n=1 or 2, then R4 and R5 are not connected through an ether linkage.
60. The method of claim 59, wherein:
R3 is hydrogen, halogen or —CN; and
R5 is aryl, heteroaryl, alicyclic, or heterocyclic, all optionally substituted with halogen, lower alkyl, lower alkenyl, lower alkynyl, lower aryl, lower alicyclic, arylalkyl, aryloxy, aryloxyalkyl, alkoxyalkyl, perhaloalkyl, perhaloalkyloxy, perhaloacyl, —N3, —SR8, —OR8, —CN, —C(O)R9, —NO2, or —NR8R10.
61. The method of claim 59, wherein:
R1 is halogen;
R is —NH2;
R3 is hydrogen;
R4 is —CH2; and
R5 is aryl or heteroaryl, wherein:
the aryl and heteroaryl groups are monocyclic or bicyclic,
the aryl group is substituted with 4 to 5 substituents,
the heteroaryl group is substituted with 2 to 5 substituents, wherein when
the heteroaryl is substituted with two substituents, the two substituents must form part of an optionally substituted fused ring.
62. The method of claim 60, wherein R1 is chloro or bromo, R2 is —NH2, and R5 is a phenyl having 3 to 5 substituents, a pyridyl having 3 to 5 substituents or an 1-oxy-pyridyl (N-oxy-pyridyl) having 3 to 5 substituents.
63. The method of claim 60, wherein the HSP90 mediated disorder is selected from the group of inflammatory diseases, infections, autoimmune disorders, stroke, ischemia, cardiac disorders, neurological disorders, fibrogenetic disorders, proliferative disorders, tumors, leukemias, neoplasms, cancers, carcinomas, metabolic diseases, and malignant disease.
64. The method of claim 63 wherein the fibrogenetic disorder is further selected from the group of scleroderma, polymyositis, systemic lupus, rheumatoid arthritis, liver cirrhosis, keloid formation, interstitial nephritis and pulmonary fibrosis.
65. The method of claim 60, further comprising administering at least one therapeutic agent selected from the group of cytotoxic agents, anti-angiogenesis agents and anti-neoplastic agents.
66. The method of claim 65, wherein the at least one anti-neoplastic agents is selected from the group of alkylating agents, anti-metabolites, epidophyllotoxins, antineoplastic enzymes, topoisomerase inhibitors, procarbazines, mitoxantrones, platinum coordination complexes, biological response modifiers and growth inhibitors, hormonal/anti-hormonal therapeutic agents, and haematopoietic growth factors.
67. A compound, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof, prepared by the process comprising:
reacting a compound of formula Y and a compound of formula Z, wherein:
Y is represented by any of the following formulae, respectively:
Figure US20070185064A1-20070809-C00091
and
Z is L1—R4—R5; wherein:
L1 is halogen, NR8R10, triflate, tosylate, or mesylate;
R4 is —CHR2—,
R5 is aryl, heteroaryl, alicyclic, or heterocyclic, wherein:
the aryl group is substituted with 3 to 5 substituents,
the heteroaryl group is substituted with 2 to 5 substituents, wherein when the heteroaryl is substituted with only two substituents, the two substituents must form part of an optionally substituted fused ring,
the alicyclic group is substituted with 3 to 5 substituents,
the heterocyclic group is substituted with 3 to 5 substituents, and
the substituents are selected from the group of halogen, lower alkyl, lower alkenyl, lower alkynyl, lower aryl, lower alicyclic, arylalkyl, aryloxy, aryloxyalkyl, alkoxyalkyl, perhaloalkyl, perhaloalkyloxy, perhaloacyl, —N3, —SR8, —OR8, —CN, —C(O)R9, —NO2, —NR R8R10, phosphonate and phosphonic acid;
R8 is hydrogen, lower alkyl, lower aryl, or —C(O)R9;
R9 is lower alkyl, lower aryl, lower heteroaryl, —NR10R10 or —OR11;
R10 is independently hydrogen or lower alkyl;
R11 is lower alkyl, lower aryl or lower heteroaryl;
R12 is hydrogen or lower alkyl;
R21 is halogen, lower alkyl or —OH;
R22 is —NR8R10;
R23 is hydrogen, halogen, or —CN;
R24 is —NH2, —NO2, or —NO;
R25 is halogen or —OH;
R26 is —C(O)NH2 or C(O)OEt; and
R27 is —NH2, —OH or halogen;
provided that:
when R5 is aryl, R5 is not an organo-metallic cyclopentadiene;
when R5 is phenyl, the substituents are not 3, 5 di-halo;
when R5 is alicyclic, the ring system does not contain any tetra-substituted sp3 ring carbons; or
when R5 is heterocyclic, the ring system does not contain any tetra-substituted sp3 ring carbons or the ring system is not a tetra-substituted pyrrolidine.
68. The compound of claim 67 wherein R4 is —CH2—.
69. The compound of claim 68, wherein:
L1 is —Cl, —Br or NH2; and
R5 is aryl or heteroaryl, wherein the aryl group is substituted with 4 to 5 substituents, the heteroaryl group is substituted with 2 to 5 substituents, wherein when the heteroaryl is substituted with two substituents, the two substituents must form part of an optionally substituted fused ring.
70. The compound of claim 69, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof, wherein Y is a substituted purine.
71. The compound of claim 68, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof, wherein said reaction is performed in a solvent comprising a member selected from the group of DMF, THF, and DMSO.
72. The compound of claim 70, or a polymorph, solvate, ester, tautomer, pharmaceutically acceptable salt or prodrug thereof, wherein said reaction is performed in a solvent that comprises DMF.
US11/531,221 2003-09-18 2006-09-12 2-Aminopurine Analogs Having HSP90-Inhibiting Activity Abandoned US20070185064A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/531,221 US20070185064A1 (en) 2003-09-18 2006-09-12 2-Aminopurine Analogs Having HSP90-Inhibiting Activity

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US50413503P 2003-09-18 2003-09-18
US59146704P 2004-07-26 2004-07-26
US10/946,645 US7138401B2 (en) 2003-09-18 2004-09-20 2-aminopurine analogs having HSP90-inhibiting activity
US11/531,221 US20070185064A1 (en) 2003-09-18 2006-09-12 2-Aminopurine Analogs Having HSP90-Inhibiting Activity

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/946,645 Continuation US7138401B2 (en) 2003-09-18 2004-09-20 2-aminopurine analogs having HSP90-inhibiting activity

Publications (1)

Publication Number Publication Date
US20070185064A1 true US20070185064A1 (en) 2007-08-09

Family

ID=34381103

Family Applications (8)

Application Number Title Priority Date Filing Date
US10/946,645 Active 2024-10-13 US7138401B2 (en) 2003-09-18 2004-09-20 2-aminopurine analogs having HSP90-inhibiting activity
US10/946,628 Active US7129244B2 (en) 2003-09-18 2004-09-20 Triazolopyrimidines and related analogs as HSP90-inhibitors
US10/945,851 Active 2024-11-17 US7138402B2 (en) 2002-10-30 2004-09-20 Pyrrolopyrimidines and related analogs as HSP90-inhibitors
US10/946,637 Active US7148228B2 (en) 2003-09-18 2004-09-20 Pyrazolopyrimidines and related analogs as HSP90-inhibitors
US11/531,218 Abandoned US20070173483A1 (en) 2002-10-30 2006-09-12 Pyrrolopyrimidines and Related Analogs as HSP90-Inhibitors
US11/531,221 Abandoned US20070185064A1 (en) 2003-09-18 2006-09-12 2-Aminopurine Analogs Having HSP90-Inhibiting Activity
US11/537,017 Abandoned US20070111997A1 (en) 2003-09-18 2006-09-29 Triazolopyrimidines and related analogs as HSP90-inhibitors
US11/537,002 Abandoned US20070111996A1 (en) 2003-09-18 2006-09-29 Pyrazolopyrimidines and related analogs as HSP90-inhibitors

Family Applications Before (5)

Application Number Title Priority Date Filing Date
US10/946,645 Active 2024-10-13 US7138401B2 (en) 2003-09-18 2004-09-20 2-aminopurine analogs having HSP90-inhibiting activity
US10/946,628 Active US7129244B2 (en) 2003-09-18 2004-09-20 Triazolopyrimidines and related analogs as HSP90-inhibitors
US10/945,851 Active 2024-11-17 US7138402B2 (en) 2002-10-30 2004-09-20 Pyrrolopyrimidines and related analogs as HSP90-inhibitors
US10/946,637 Active US7148228B2 (en) 2003-09-18 2004-09-20 Pyrazolopyrimidines and related analogs as HSP90-inhibitors
US11/531,218 Abandoned US20070173483A1 (en) 2002-10-30 2006-09-12 Pyrrolopyrimidines and Related Analogs as HSP90-Inhibitors

Family Applications After (2)

Application Number Title Priority Date Filing Date
US11/537,017 Abandoned US20070111997A1 (en) 2003-09-18 2006-09-29 Triazolopyrimidines and related analogs as HSP90-inhibitors
US11/537,002 Abandoned US20070111996A1 (en) 2003-09-18 2006-09-29 Pyrazolopyrimidines and related analogs as HSP90-inhibitors

Country Status (14)

Country Link
US (8) US7138401B2 (en)
EP (2) EP2145888A1 (en)
JP (1) JP2007505933A (en)
KR (1) KR20060070572A (en)
CN (1) CN101906106A (en)
AU (2) AU2004274507B2 (en)
BR (1) BRPI0414533A (en)
CA (1) CA2539548A1 (en)
EA (1) EA010160B1 (en)
IL (1) IL174375A0 (en)
MX (1) MXPA06002997A (en)
NO (1) NO20061396L (en)
NZ (1) NZ546611A (en)
WO (1) WO2005028434A2 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060223797A1 (en) * 2005-03-30 2006-10-05 Conforma Therapeutics Corporation Alkynyl pyrrolopyrimidines and related analogs as hsp90-inhibitors
US20070105874A1 (en) * 2005-09-23 2007-05-10 Conforma Therapeutics Corporation Anti-Tumor Methods Using Multi Drug Resistance Independent Synthetic HSP90 Inhibitors
US20070253896A1 (en) * 2006-02-07 2007-11-01 Conforma Therapeutics Corporation 7,9-Dihydro-Purin-8-One and Related Analogs as HSP90-Inhibitors

Families Citing this family (177)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6992188B1 (en) * 1995-12-08 2006-01-31 Pfizer, Inc. Substituted heterocyclic derivatives
JP2005519848A (en) * 2000-11-02 2005-07-07 スローン−ケッタリング・インスティテュート・フォー・キャンサー・リサーチ Small molecule composition for binding to HSP90
AU2002315389A1 (en) * 2001-06-21 2003-01-08 Ariad Pharmaceuticals, Inc. Novel pyrazolo-and pyrrolo-pyrimidines and uses thereof
US20070129334A1 (en) * 2001-10-30 2007-06-07 Conforma Therapeutics Corporation Orally Active Purine-Based Inhibitors of Heat Shock Protein 90
EP1440072A4 (en) 2001-10-30 2005-02-02 Conforma Therapeutic Corp Purine analogs having hsp90-inhibiting activity
CN101906106A (en) 2003-09-18 2010-12-08 康福玛医药公司 New heterogeneous ring compound as the HSP90-inhibitor
WO2006050457A2 (en) * 2004-11-02 2006-05-11 Conforma Therapeutics Corporation Methods and compositions for treating chronic lymphocytic leukemia
WO2006050333A2 (en) * 2004-11-02 2006-05-11 The Regents Of The University Of California Methods and compositions for modulating apoptosis
US9120774B2 (en) 2004-11-03 2015-09-01 University Of Kansas Novobiocin analogues having modified sugar moieties
US8212012B2 (en) * 2004-11-03 2012-07-03 University Of Kansas Novobiocin analogues having modified sugar moieties
AU2006235735C1 (en) * 2005-04-13 2013-10-31 Astex Therapeutics Limited Hydroxybenzamide derivatives and their use as inhibitors of Hsp90
KR20080006614A (en) * 2005-04-14 2008-01-16 노바티스 백신즈 앤드 다이아그노스틱스 인코포레이티드 2-amino-quinazolin-5-ones as hsp90 inhibitors useful in treating proliferation diseases
WO2006124897A2 (en) * 2005-05-13 2006-11-23 Lexicon Genetics Incorporated Methods and compositions for improving cognition
EP1915377A1 (en) * 2005-07-22 2008-04-30 Sunesis Pharmaceuticals, Inc. Pyrazolo pyrimidines useful as aurora kinase inhibitors
US20090324551A1 (en) * 2005-08-22 2009-12-31 The Regents Of The University Of California Office Of Technology Transfer Tlr agonists
EP1926734A1 (en) 2005-08-22 2008-06-04 Amgen Inc. Pyrazolopyridine and pyrazolopyrimidine compounds useful as kinase enzymes modulators
AU2006283511A1 (en) * 2005-08-24 2007-03-01 Lexicon Pharmaceuticals, Inc. Pyrrolopyridine, pyrrolopyrimidine and pyrazolopyridine compounds, compositions comprising them, and methods of their use
US20090227616A1 (en) * 2005-11-10 2009-09-10 Smithkline Beecham Corporation, A Corporation Inhibitors of akt activity
JP2009515884A (en) * 2005-11-10 2009-04-16 スミスクライン・ビーチャム・コーポレイション Inhibitor of AKT activity
JP2009521446A (en) * 2005-12-22 2009-06-04 コンフォーマ・セラピューティクス・コーポレイション Orally active purine-based inhibitors of heat shock protein 90
WO2007097839A2 (en) * 2006-02-16 2007-08-30 Massachusetts Eye And Ear Infirmary Ansamycin analogs or heat shock 90 inhibitors in combination with pdt treatin conditions of the eye
AU2007249194B2 (en) * 2006-05-12 2012-11-22 Myrexis, Inc. Therapeutic compounds and their use in cancer
WO2007142755A2 (en) * 2006-05-31 2007-12-13 The Regents Of The University Of California Purine analogs
CA2656202C (en) 2006-06-30 2018-01-16 Sloan-Kettering Institute For Cancer Research Treatment of neurodegenerative diseases through inhibition of hsp90
US20080009508A1 (en) * 2006-07-10 2008-01-10 Lucie Szucova 6,9-Disubstituted Purine Derivatives And Their Use For Treating Skin
ATE414281T1 (en) * 2006-08-03 2008-11-15 Cellzome Ag METHOD FOR IDENTIFYING MOLECULES THAT INTERACT WITH PI3K AND METHOD FOR PURIFYING PI3K
EP2065388B1 (en) 2006-09-19 2011-09-07 Daiichi Sankyo Company, Limited Pyrazolopyrimidine derivative
WO2008045529A1 (en) * 2006-10-12 2008-04-17 Serenex, Inc. Purine and pyrimidine derivatives for treatment of cancer and inflammatory diseases
CL2007002994A1 (en) * 2006-10-19 2008-02-08 Wyeth Corp HETEROCICLIC DERIVATIVE COMPOUNDS CONTAINING SULFAMOIL, INHIBITORS OF HSP90; PHARMACEUTICAL COMPOSITION; AND USE FOR THE TREATMENT OF CANCER, SUCH AS CANCER OF BREAST, COLON AND PROSTATE, BETWEEN OTHERS.
FR2907453B1 (en) * 2006-10-24 2008-12-26 Sanofi Aventis Sa NOVEL FLUORENE DERIVATIVES, COMPOSITIONS CONTAINING SAME AND USE THEREOF
GB0622084D0 (en) * 2006-11-06 2006-12-13 Chroma Therapeutics Ltd Inhibitors of HSP90
US20110281821A9 (en) * 2007-01-30 2011-11-17 Biogen Idec Ma Inc. Modulators of Mitotic Kinases
EP2109612B1 (en) * 2007-02-01 2010-11-10 AstraZeneca AB 5,6,7,8-tetrahydropteridine derivatives as hsp90 inhibitors
PL2125007T3 (en) 2007-02-07 2014-07-31 Univ California Conjugates of synthetic tlr agonists and uses therefor
AU2008229216B2 (en) * 2007-03-20 2013-03-21 Curis, Inc. Fused amino pyridine as HSP90 inhibitors
NZ580313A (en) 2007-04-12 2011-11-25 Joyant Pharmaceuticals Inc Smac mimetic dimers and trimers useful as anti-cancer agents
US8143265B2 (en) * 2007-04-16 2012-03-27 Meharry Medical College Method of treating atherosclerosis
US7960353B2 (en) * 2007-05-10 2011-06-14 University Of Kansas Novobiocin analogues as neuroprotective agents and in the treatment of autoimmune disorders
DE102007028521A1 (en) * 2007-06-21 2008-12-24 Merck Patent Gmbh Indazolamidderivate
US8119616B2 (en) * 2007-09-10 2012-02-21 Curis, Inc. Formulation of quinazoline based EGFR inhibitors containing a zinc binding moiety
WO2009036092A2 (en) 2007-09-10 2009-03-19 University Of Massachusetts Mitochondria-targeted anti-tumor agents
GB2467670B (en) 2007-10-04 2012-08-01 Intellikine Inc Chemical entities and therapeutic uses thereof
WO2009086170A1 (en) 2007-12-21 2009-07-09 Joyant Pharmaceuticals, Inc. Diazonamide analogs with improved solubility
US8895701B2 (en) * 2008-01-05 2014-11-25 Sloan-Kettering Institute For Cancer Research Peptide-conjugated oligonucleotide therapeutic and method of making and using same
BRPI0907907A2 (en) * 2008-02-07 2015-07-28 Univ California Method to inhibit or treat superficial bladder cancer in a mammal, and use of a tlr agonist 7
US8190543B2 (en) * 2008-03-08 2012-05-29 Tokyo Electron Limited Autonomous biologically based learning tool
WO2009114470A2 (en) * 2008-03-10 2009-09-17 Curis, Inc. Tetrahydroindole and tetrahdyroindazole as hsp90 inhibitors containing a zinc binding moiety
US8637542B2 (en) 2008-03-14 2014-01-28 Intellikine, Inc. Kinase inhibitors and methods of use
WO2009114874A2 (en) 2008-03-14 2009-09-17 Intellikine, Inc. Benzothiazole kinase inhibitors and methods of use
WO2009134938A1 (en) 2008-04-29 2009-11-05 Joyant Pharmaceuticals, Inc. Indoline anti-cancer agents
LT5623B (en) 2008-04-30 2010-01-25 Biotechnologijos Institutas, , 5-aryl-4-(5-substituted 2,4-dihydroxyfenil)-1,2,3-thiadiazoles as inhibitors of hsp90 chaperone and the intermediates for production thereof
WO2009139834A1 (en) * 2008-05-13 2009-11-19 Poniard Pharmaceuticals, Inc. Bioactive compounds for treatment of cancer and neurodegenerative diseases
WO2009143485A1 (en) 2008-05-22 2009-11-26 Joyant Pharmaceuticals, Inc. Diazonamide analogs
BRPI0915231A2 (en) 2008-07-08 2018-06-12 Intellikine Inc kinase inhibitor compounds and methods of use
WO2010014617A1 (en) * 2008-07-28 2010-02-04 University Of Kansas Heat shock protein 90 inhibitor dosing methods
GB0819102D0 (en) * 2008-10-17 2008-11-26 Chroma Therapeutics Ltd Pyrrolo-pyrimidine compounds
US8476282B2 (en) 2008-11-03 2013-07-02 Intellikine Llc Benzoxazole kinase inhibitors and methods of use
CA2743864A1 (en) * 2008-11-25 2010-06-10 Merck Sharp & Dohme Corp. Soluble guanylate cyclase activators
US8722703B2 (en) 2009-01-16 2014-05-13 Curis, Inc. Fused amino pyridines for the treatment of brain tumors
WO2010088924A1 (en) 2009-02-06 2010-08-12 Telormedix Sa Pharmaceutical compositions comprising imidazoquinolin(amines) and derivatives thereof suitable for local administration
US8729088B2 (en) * 2009-02-11 2014-05-20 The Regents Of The University Of California Toll-like receptor modulators and treatment of diseases
RU2520966C2 (en) * 2009-02-25 2014-06-27 Дайити Санкио Компани, Лимитед Tricyclic pyrazolopyrimidine derivative
EP2770327B1 (en) * 2009-03-30 2017-06-14 Nordic Bioscience A/S Fibrosis biomarker assay
WO2010117425A1 (en) * 2009-03-31 2010-10-14 Biogen Idec Ma Inc. Certain substituted pyrimidines, pharmaceutical compositions thereof, and methods for their use
ME01838B (en) 2009-06-29 2014-12-20 Lncyte Holdings Corp Pyrimidinones as pi3k inhibitors
KR101609412B1 (en) * 2009-07-10 2016-04-05 다이호야쿠힌고교 가부시키가이샤 Azabicyclo compound and salt thereof
EA026693B1 (en) 2009-08-17 2017-05-31 Интелликайн ЭлЭлСи Benzoxazole and benzothiazole derivatives as pi3 kinase inhibitors
WO2011041593A1 (en) * 2009-09-30 2011-04-07 University Of Kansas Novobiocin analogues and treatment of polycystic kidney disease
BR112012008019A2 (en) 2009-10-07 2016-03-01 Sloan Kettering Inst Cancer purine derivatives useful as hsp90 inhibitors
CN102958928B (en) 2009-10-28 2015-05-13 焦阳特制药股份有限公司 Dimeric SMAC Mimetics
US20110130711A1 (en) * 2009-11-19 2011-06-02 Follica, Inc. Hair growth treatment
US8680108B2 (en) * 2009-12-18 2014-03-25 Incyte Corporation Substituted fused aryl and heteroaryl derivatives as PI3K inhibitors
WO2011075643A1 (en) 2009-12-18 2011-06-23 Incyte Corporation Substituted heteroaryl fused derivatives as pi3k inhibitors
WO2011076725A1 (en) 2009-12-21 2011-06-30 Bayer Cropscience Ag Thienylpyri (mi) dinylazole and their use for controlling phytopathogenic fungi
US9193721B2 (en) 2010-04-14 2015-11-24 Incyte Holdings Corporation Fused derivatives as PI3Kδ inhibitors
US9050319B2 (en) 2010-04-30 2015-06-09 Telormedix, Sa Phospholipid drug analogs
CA2797315C (en) 2010-04-30 2018-09-11 Telormedix Sa Phospholipid drug analogs
WO2011146313A1 (en) 2010-05-19 2011-11-24 The University Of North Carolina At Chapel Hill Pyrazolopyrimidine compounds for the treatment of cancer
US9062055B2 (en) 2010-06-21 2015-06-23 Incyte Corporation Fused pyrrole derivatives as PI3K inhibitors
KR20130098982A (en) 2010-08-23 2013-09-05 다이이찌 산쿄 가부시키가이샤 Crystal of tricyclic pyrazolopyrimidine derivative
TW201213334A (en) 2010-08-23 2012-04-01 Daiichi Sankyo Co Ltd Crystal forms of free form of tricyclic pyrazolopyrimidine derivative
AU2011326071A1 (en) 2010-11-08 2013-05-23 Lycera Corporation N- sulfonylated tetrahydroquinolines and related bicyclic compounds inhibition of RORy activity and the treatment of diseases
WO2012083181A1 (en) * 2010-12-16 2012-06-21 Indiana University Research And Technology Corporation Alpha helix mimetics and methods for using
PT2651951E (en) 2010-12-16 2015-01-14 Hoffmann La Roche Tricyclic pi3k inhibitor compounds and methods of use
EP2655374B1 (en) 2010-12-20 2019-10-23 Incyte Holdings Corporation N-(1-(substituted-phenyl)ethyl)-9h-purin-6-amines as pi3k inhibitors
US8754114B2 (en) 2010-12-22 2014-06-17 Incyte Corporation Substituted imidazopyridazines and benzimidazoles as inhibitors of FGFR3
US8362023B2 (en) 2011-01-19 2013-01-29 Hoffmann-La Roche Inc. Pyrazolo pyrimidines
JP5766820B2 (en) 2011-02-09 2015-08-19 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Heterocyclic compounds as PI3 kinase inhibitors
AR085397A1 (en) 2011-02-23 2013-09-25 Intellikine Inc COMBINATION OF QUINASA INHIBITORS AND THEIR USES
US9108984B2 (en) 2011-03-14 2015-08-18 Incyte Corporation Substituted diamino-pyrimidine and diamino-pyridine derivatives as PI3K inhibitors
WO2012135009A1 (en) 2011-03-25 2012-10-04 Incyte Corporation Pyrimidine-4,6-diamine derivatives as pi3k inhibitors
ES2647889T3 (en) 2011-04-05 2017-12-27 Sloan-Kettering Institute For Cancer Research Hsp90 inhibitors
MX360390B (en) 2011-04-05 2018-10-31 Sloan Kettering Inst Cancer Res Hsp90 inhibitors.
WO2012145255A2 (en) 2011-04-22 2012-10-26 Joyant Pharmaceuticals, Inc. Diazonamide analogs
EP2714688B1 (en) 2011-05-26 2016-02-24 Daiichi Sankyo Company, Limited Heterocyclic compounds as protein kinase inhibitors
WO2012178125A1 (en) 2011-06-22 2012-12-27 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
ITTO20110652A1 (en) * 2011-07-20 2013-01-21 Medestea Res & Production S P A COMPOUND DERIVED FROM THE ADENINE HAVING IMMUNO-MODULATING ACTION, ANTI-INFLAMMATORY AND ANTI-DOLUTION ACTION
MX2020004502A (en) 2011-09-02 2022-01-20 Incyte Holdings Corp Heterocyclylamines as pi3k inhibitors.
BR112014007788A2 (en) 2011-10-03 2017-04-18 Univ North Carolina Chapel Hill Pyrrolopyrimidine compounds for cancer treatment
AR090548A1 (en) 2012-04-02 2014-11-19 Incyte Corp BICYCLIC AZAHETEROCICLOBENCILAMINS AS PI3K INHIBITORS
JP6236068B2 (en) 2012-05-08 2017-11-22 メルク・シャープ・アンド・ドーム・コーポレーションMerck Sharp & Dohme Corp. Tetrahydronaphthyridine and related bicyclic compounds for inhibiting RORγ activity and treating diseases
EP2847198B1 (en) 2012-05-08 2016-12-14 Lycera Corporation Tetrahydro[1,8]naphthyridine sulfonamide and related compounds for use as agonists of ror-gamma and the treatment of disease
WO2013172872A1 (en) * 2012-05-15 2013-11-21 Calasia Pharmaceuticals, Inc. Pyrimidine diamine derivatives as inhibitors of cytosolic hsp90
CN104302627A (en) 2012-05-22 2015-01-21 北卡罗来纳大学教堂山分校 Pyrimidine compounds for the treatment of cancer
UA117347C2 (en) 2012-06-13 2018-07-25 Інсайт Холдинґс Корпорейшн Substituted tricyclic compounds as fgfr inhibitors
WO2014026125A1 (en) 2012-08-10 2014-02-13 Incyte Corporation Pyrazine derivatives as fgfr inhibitors
EP2904119B1 (en) 2012-10-02 2020-06-17 The General Hospital Corporation d/b/a Massachusetts General Hospital Methods relating to dna-sensing pathway related conditions
CA2885259A1 (en) 2012-10-04 2014-04-10 Pfizer Limited Pyrrolo[3,2-c]pyridine tropomyosin-related kinase inhibitors
CN103724269B (en) * 2012-10-11 2016-12-21 中国科学院上海药物研究所 Phenyl 1,2-isoxazole or phenyl 1,2-pyrazole compound and application thereof
WO2014062774A1 (en) 2012-10-17 2014-04-24 The University Of North Carolina At Chapel Hill Pyrazolopyrimidine compounds for the treatment of cancer
EP2925752A4 (en) 2012-11-27 2016-06-01 Univ North Carolina Pyrimidine compounds for the treatment of cancer
PT3808749T (en) 2012-12-07 2023-06-07 Vertex Pharma Pyrazolo[1,5-a]pyrimidines useful as inhibitors of atr kinase for the treatment of cancer diseases
US9266892B2 (en) 2012-12-19 2016-02-23 Incyte Holdings Corporation Fused pyrazoles as FGFR inhibitors
AU2014228822A1 (en) 2013-03-15 2015-10-01 Memorial Sloan-Kettering Cancer Center HSP90-targeted cardiac imaging and therapy
JP2016512815A (en) 2013-03-15 2016-05-09 バーテックス ファーマシューティカルズ インコーポレイテッドVertex Pharmaceuticals Incorporated Condensed pyrazolopyrimidine derivatives useful as inhibitors of ATR kinase
US8969360B2 (en) 2013-03-15 2015-03-03 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
WO2014145576A2 (en) 2013-03-15 2014-09-18 Northwestern University Substituted pyrrolo(2,3-d)pyrimidines for the treatment of cancer
WO2014143241A1 (en) 2013-03-15 2014-09-18 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
JP6360881B2 (en) 2013-03-22 2018-07-18 ミレニアム ファーマシューティカルズ, インコーポレイテッドMillennium Pharmaceuticals, Inc. Combination of catalytic MTORC1 / 2 inhibitor and selective inhibitor of Aurora A kinase
CN105263931B (en) 2013-04-19 2019-01-25 因赛特公司 Bicyclic heterocycle as FGFR inhibitor
US9993460B2 (en) 2013-07-26 2018-06-12 Race Oncology Ltd. Compositions to improve the therapeutic benefit of bisantrene and analogs and derivatives thereof
JP6539275B2 (en) 2013-08-16 2019-07-03 メモリアル スローン ケタリング キャンサー センター Selective GRP94 inhibitors and uses thereof
HUE046727T2 (en) 2013-12-06 2020-03-30 Vertex Pharma 2-amino-6-fluoro-n-[5-fluoro-pyridin-3-yl]pyrazolo[1,5-a]pyrimidin-3-carboxamide compound useful as atr kinase inhibitor, its preparation, different solid forms and radiolabelled derivatives thereof
US9663502B2 (en) 2013-12-20 2017-05-30 Lycera Corporation 2-Acylamidomethyl and sulfonylamidomethyl benzoxazine carbamates for inhibition of RORgamma activity and the treatment of disease
US9809561B2 (en) 2013-12-20 2017-11-07 Merck Sharp & Dohme Corp. Tetrahydronaphthyridine, benzoxazine, aza-benzoxazine and related bicyclic compounds for inhibition of RORgamma activity and the treatment of disease
WO2015095792A1 (en) 2013-12-20 2015-06-25 Merck Sharp & Dohme Corp. Carbamate benzoxaxine propionic acids and acid derivatives for modulation of rorgamma activity and the treatment of disease
JP2017507950A (en) 2014-02-27 2017-03-23 リセラ・コーポレイションLycera Corporation Adoptive cell therapy using retinoic acid receptor-related orphan receptor gamma agonists and related therapeutic methods
ES2548927B1 (en) * 2014-03-21 2016-08-11 Universidad De Granada Purine derivatives as Dapk-1 inhibitors
WO2015157125A1 (en) 2014-04-11 2015-10-15 The University Of North Carolina At Chapel Hill Therapuetic uses of selected pyrazolopyrimidine compounds with anti-mer tyrosine kinase activity
US10189777B2 (en) 2014-05-05 2019-01-29 Lycera Corporation Benzenesulfonamido and related compounds for use as agonists of RORγ and the treatment of disease
US9896441B2 (en) 2014-05-05 2018-02-20 Lycera Corporation Tetrahydroquinoline sulfonamide and related compounds for use as agonists of RORγ and the treatment of disease
NZ764151A (en) 2014-06-05 2023-12-22 Vertex Pharma Radiolabelled derivatives of a 2-amino-6-fluoro-n-[5-fluoro-pyridin-3-yl]- pyrazolo[1,5-a]pyrimidin-3-carboxamide compound useful as atr kinase inhibitor, the preparation of said compound and different solid forms thereof
WO2015191677A1 (en) 2014-06-11 2015-12-17 Incyte Corporation Bicyclic heteroarylaminoalkyl phenyl derivatives as pi3k inhibitors
RU2736219C2 (en) 2014-06-17 2020-11-12 Вертекс Фармасьютикалз Инкорпорейтед Method of treating cancer using a combination of chk1 and atr inhibitors
AU2015317632B2 (en) 2014-09-17 2022-01-13 Memorial Sloan Kettering Cancer Center Hsp90-targeted inflammation and infection imaging and therapy
US10851105B2 (en) 2014-10-22 2020-12-01 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
KR101713638B1 (en) * 2014-11-18 2017-03-08 한국과학기술연구원 Novel pyrrolo pyrimidine derivatives and composition for preventing or treating cancer comprising the same
CA2975997A1 (en) 2015-02-11 2016-08-18 Merck Sharp & Dohme Corp. Substituted pyrazole compounds as rorgammat inhibitors and uses thereof
UA121669C2 (en) 2015-02-20 2020-07-10 Інсайт Корпорейшн Bicyclic heterocycles as fgfr inhibitors
WO2016134294A1 (en) 2015-02-20 2016-08-25 Incyte Corporation Bicyclic heterocycles as fgfr4 inhibitors
MA41551A (en) 2015-02-20 2017-12-26 Incyte Corp BICYCLIC HETEROCYCLES USED AS FGFR4 INHIBITORS
CR20170389A (en) 2015-02-27 2018-01-26 Incyte Corp P13K INHIBITOR SALTS AND PREPARATION PROCESSES
CA2982847A1 (en) 2015-05-05 2016-11-10 Lycera Corporation Dihydro-2h-benzo[b][1,4]oxazine sulfonamide and related compounds for use as agonists of ror.gamma. and the treatment of disease
WO2016183063A1 (en) 2015-05-11 2016-11-17 Incyte Corporation Crystalline forms of a pi3k inhibitor
WO2016183060A1 (en) 2015-05-11 2016-11-17 Incyte Corporation Process for the synthesis of a phosphoinositide 3-kinase inhibitor
KR20180025894A (en) 2015-06-11 2018-03-09 라이세라 코퍼레이션 Benzo [B] [1,4] oxazinesulfonamides and related compounds for use as agonists of RORY and the treatment of diseases
CA2993018A1 (en) 2015-07-20 2017-01-26 Genzyme Corporation Colony stimulating factor-1 receptor (csf-1r) inhibitors
AU2016331955B2 (en) 2015-09-30 2022-07-21 Vertex Pharmaceuticals Incorporated Method for treating cancer using a combination of DNA damaging agents and ATR inhibitors
US10344000B2 (en) 2015-10-27 2019-07-09 Merck Sharp & Dohme Corp. Substituted bicyclic pyrazole compounds as RORgammaT inhibitors and uses thereof
EP3368539B1 (en) 2015-10-27 2020-12-02 Merck Sharp & Dohme Corp. Substituted indazole compounds as ror gamma t inhibitors and uses thereof
JP2018531958A (en) 2015-10-27 2018-11-01 メルク・シャープ・アンド・ドーム・コーポレーションMerck Sharp & Dohme Corp. Heteroaryl-substituted benzoic acids and their use as RORγT inhibitors
US10709708B2 (en) 2016-03-17 2020-07-14 The University Of North Carolina At Chapel Hill Method of treating cancer with a combination of MER tyrosine kinase inhibitor and an epidermal growth factor receptor (EGFR) inhibitor
US11697851B2 (en) 2016-05-24 2023-07-11 The Regents Of The University Of California Early ovarian cancer detection diagnostic test based on mRNA isoforms
KR101896616B1 (en) * 2016-10-11 2018-09-07 한국과학기술연구원 2-Amino-purine derivative compound, preparing method and use thereof
WO2018079988A1 (en) * 2016-10-26 2018-05-03 울산과학기술원 Compound useful as trap1 inhibitor, and anticancer composition containing same
DK3483164T3 (en) 2017-03-20 2020-03-23 Forma Therapeutics Inc PYRROLOPYRROL COMPOSITIONS AS PYRUVAT KINASE (PKR) ACTIVATORS
AR111960A1 (en) 2017-05-26 2019-09-04 Incyte Corp CRYSTALLINE FORMS OF A FGFR INHIBITOR AND PROCESSES FOR ITS PREPARATION
US10472361B2 (en) 2017-08-16 2019-11-12 Bristol-Myers Squibb Company Toll-like receptor 7 (TLR7) agonists having a benzotriazole moiety, conjugates thereof, and methods and uses therefor
JOP20180094A1 (en) 2017-10-18 2019-04-18 Hk Inno N Corp Heterocyclic compound as a protein kinase inhibito
CN107857763A (en) * 2017-11-28 2018-03-30 江苏杏睿生物科技有限公司 A kind of adjacent nitro aroylation derivatives quasi-compound, preparation method and applications
FR3075794A1 (en) 2017-12-21 2019-06-28 Galderma Research & Development NEW MTOR INHIBITOR COMPOUNDS
FR3075795A1 (en) 2017-12-21 2019-06-28 Galderma Research & Development NEW MTOR INHIBITOR COMPOUNDS
IL276802B2 (en) 2018-03-09 2023-09-01 Recurium Ip Holdings Llc Substituted 1,2-dihydro-3h-pyrazolo[3,4-d]pyrimidin-3-ones
SG11202010636VA (en) 2018-05-04 2020-11-27 Incyte Corp Solid forms of an fgfr inhibitor and processes for preparing the same
MA52493A (en) 2018-05-04 2021-03-10 Incyte Corp FGFR INHIBITOR SALTS
WO2020061255A1 (en) 2018-09-19 2020-03-26 Forma Therapeutics, Inc. Activating pyruvate kinase r
WO2020061378A1 (en) 2018-09-19 2020-03-26 Forma Therapeutics, Inc. Treating sickle cell disease with a pyruvate kinase r activating compound
US11628162B2 (en) 2019-03-08 2023-04-18 Incyte Corporation Methods of treating cancer with an FGFR inhibitor
WO2021007269A1 (en) 2019-07-09 2021-01-14 Incyte Corporation Bicyclic heterocycles as fgfr inhibitors
PE20221085A1 (en) 2019-10-14 2022-07-05 Incyte Corp BICYCLIC HETEROCYCLES AS FGFR INHIBITORS
WO2021076728A1 (en) 2019-10-16 2021-04-22 Incyte Corporation Bicyclic heterocycles as fgfr inhibitors
CN115151539A (en) 2019-12-04 2022-10-04 因赛特公司 Derivatives of FGFR inhibitors
WO2021113479A1 (en) 2019-12-04 2021-06-10 Incyte Corporation Tricyclic heterocycles as fgfr inhibitors
KR102516260B1 (en) * 2020-07-10 2023-03-31 울산과학기술원 Compounds as a TRAP1 selective inhibitor, and composition for preventing or treating cancer comprising the same
EP4267573A1 (en) 2020-12-23 2023-11-01 Genzyme Corporation Deuterated colony stimulating factor-1 receptor (csf-1r) inhibitors
CA3220274A1 (en) 2021-06-09 2022-12-15 Incyte Corporation Tricyclic heterocycles as fgfr inhibitors
CN116332940A (en) * 2023-02-14 2023-06-27 广西民族大学 7-deazapurine derivative and preparation method and application thereof

Citations (47)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4495190A (en) * 1980-12-22 1985-01-22 Astra Lakemedel Aktiebolag Derivatives of guanine for combating herpes virus infections
US4547573A (en) * 1983-12-02 1985-10-15 Ici Pharma Process for preparing cephalosporin derivatives
US4617304A (en) * 1984-04-10 1986-10-14 Merck & Co., Inc. Purine derivatives
US4699877A (en) * 1982-11-04 1987-10-13 The Regents Of The University Of California Methods and compositions for detecting human tumors
US4748177A (en) * 1984-03-26 1988-05-31 Warner-Lambert Company Guanine derivatives
US4772606A (en) * 1985-08-22 1988-09-20 Warner-Lambert Company Purine derivatives
US4774325A (en) * 1984-09-20 1988-09-27 Pierrel Spa New 8-substituted nucleoside and purine derivatives, the process for the preparation thereof and the pharmaceutical compositions containing them
US4806642A (en) * 1984-10-05 1989-02-21 Warner-Lambert Company Purine derivatives
US4921859A (en) * 1983-10-31 1990-05-01 Warner-Lambert Company Purine derivatives
US4923385A (en) * 1986-07-09 1990-05-08 Krauss-Maffei A.G. Blow molding machine
US4968603A (en) * 1986-12-31 1990-11-06 The Regents Of The University Of California Determination of status in neoplastic disease
US5002950A (en) * 1986-10-24 1991-03-26 Warner-Lambert Co. 7-deazaguanines as immunomodulators
US5098906A (en) * 1983-10-31 1992-03-24 Warner-Lambert Company Purine derivatives
US5110818A (en) * 1988-10-06 1992-05-05 Ciba-Geigy Corporation Anticonvulsive substituted-9-benzyl-9h-purines
US5204353A (en) * 1987-04-07 1993-04-20 Ciba-Geigy Corporation 3-benzyl-3H-1,2,3-triazolo[4,5-d]pyrimidines, compositions thereof, and method of treating epilepsy therewith
US5217866A (en) * 1985-03-15 1993-06-08 Anti-Gene Development Group Polynucleotide assay reagent and method
US5369008A (en) * 1991-05-24 1994-11-29 Board Of Regents, The University Of Texas System Methods for the detection of BCR-ABL and abnormal ABL proteins in leukemia patients
US5602156A (en) * 1993-09-17 1997-02-11 The United States Of America As Represented By The Department Of Health And Human Services Method for inhibiting metalloproteinase expression
US5656629A (en) * 1995-03-10 1997-08-12 Sanofi Winthrop, Inc. 6-substituted pyrazolo (3,4-d)pyrimidin-4-ones and compositions and methods of use thereof
US5789394A (en) * 1992-12-23 1998-08-04 Nguyen-Ba; Nghe Anti-viral compounds
US5846749A (en) * 1994-10-12 1998-12-08 The Regents Of The University Of California Quantitative measurement of tissue protein identified by immunohistochemistry and standardized protein determination
US5861503A (en) * 1997-04-30 1999-01-19 The Regents Of The University Of California Process for producing 8-fluoropurines
US5917042A (en) * 1994-02-04 1999-06-29 Glaxo Wellcome Inc. Process for the preparation of 2,5-diamino-4,6-dichloropyrimidine
US5932566A (en) * 1994-06-16 1999-08-03 Pfizer Inc. Ansamycin derivatives as antioncogene and anticancer agents
US6005107A (en) * 1992-12-23 1999-12-21 Biochem Pharma, Inc. Antiviral compounds
US6025126A (en) * 1991-10-28 2000-02-15 Arch Development Corporation Methods and compositions for the detection of chromosomal aberrations
US6174875B1 (en) * 1999-04-01 2001-01-16 University Of Pittsburgh Benzoquinoid ansamycins for the treatment of cardiac arrest and stroke
US6177460B1 (en) * 1995-04-12 2001-01-23 The Procter & Gamble Company Method of treatment for cancer or viral infections
US6210974B1 (en) * 1997-10-24 2001-04-03 Oregon Health Sciences University Compositions and methods for promoting nerve regeneration
US6369092B1 (en) * 1998-11-23 2002-04-09 Cell Pathways, Inc. Method for treating neoplasia by exposure to substituted benzimidazole derivatives
US6444656B1 (en) * 1992-12-23 2002-09-03 Biochem Pharma, Inc. Antiviral phosphonate nucleotides
US20020156277A1 (en) * 2001-04-20 2002-10-24 Fick David B. Synthesis and methods of use of purine analogues and derivatives
US20020161014A1 (en) * 2000-04-25 2002-10-31 Chanchal Sadhu Inhibitors of human phosphatidylinositol 3-kinase delta
US20030022864A1 (en) * 2001-04-24 2003-01-30 Ishaq Khalid S. 9-[(5-dihydroxyboryl)-pentyl] purines, useful as an inhibitor of inflammatory cytokines
US20030078413A1 (en) * 2001-09-12 2003-04-24 Epoch Biosciences, Inc. Process for the synthesis of pyrazolopyrimidines
US20030113331A1 (en) * 1999-01-06 2003-06-19 Brooks Peter C. Method and composition for angiogenesis inhibition
US20040063668A1 (en) * 2001-01-19 2004-04-01 Jong-Ryoo Choi Novel acyclic nucleoside phosphonate derivatives, salts thereof and process for the preparation of the same
US20040097526A1 (en) * 2001-01-10 2004-05-20 Gillespie Roger John Triazolo[4,5-d]pyrimidine derivatives and their use as purinergic receptor antagonists
US20040102458A1 (en) * 2000-11-02 2004-05-27 Gabriela Chiosis Small molecule compositions for binding to hsp90
US20040242490A1 (en) * 2003-02-20 2004-12-02 Brooks Peter C. CLK-peptide and SLK-peptide
US20040241706A1 (en) * 2002-09-13 2004-12-02 Irm, Llc Highly specific modulators of GTPases for target validation
US20050043239A1 (en) * 2003-08-14 2005-02-24 Jason Douangpanya Methods of inhibiting immune responses stimulated by an endogenous factor
US20050049263A1 (en) * 2001-10-30 2005-03-03 Kasibhatla Srinivas Rao Purine analogs having hsp90-inhibiting activity
US20050107343A1 (en) * 2003-09-18 2005-05-19 Conforma Therapeutics Corporation Pyrrolopyrimidines and related analogs as HSP90-inhibitors
US20050256183A1 (en) * 2001-11-09 2005-11-17 Kasibhatla Srinivas R Hsp90-inhibiting zearalanol compounds and methods of producing and using same
US20060035909A1 (en) * 2002-08-23 2006-02-16 Kveta Fuksova Azapurine derivatives
US7087754B2 (en) * 2001-01-10 2006-08-08 Vernalis Research Limited Pyrazolo[3,4-d]pyrimidine derivatives and their use as purinergic receptor antagonists

Family Cites Families (72)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB884151A (en) 1957-12-06 1961-12-06 Ciba Ltd New pyrazolo-pyrimidines substituted in the pyrazole nucleus, and process for their manufacture
EP0151528B1 (en) 1984-02-02 1990-07-04 Merck & Co. Inc. 5-(amino or substituted amino)-1,2,3-triazoles
ZA857647B (en) * 1984-10-12 1986-05-28 Warner Lambert Co Purine derivatives
IL76546A (en) 1984-10-12 1988-12-30 Warner Lambert Co 9-(heteroarylalkyl)-6-purine(thi)one derivatives,their preparation and pharmaceutical compositions containing them
JO1406B1 (en) 1984-11-02 1986-11-30 سميث كلاين اند فرينش لابوراتوريز ليمتد Chemical compounds
DE3687030T2 (en) 1985-03-15 1993-03-11 James Summerton STEREO-REGULAR POLYNUCLEOTID-BINDING POLYMERS.
IL78643A0 (en) 1985-05-02 1986-08-31 Wellcome Found Purine derivatives,their preparation and pharmaceutical compositions containing them
GB8515934D0 (en) 1985-06-24 1985-07-24 Janssen Pharmaceutica Nv (4-piperidinomethyl and-hetero)purines
US4918162A (en) * 1986-05-06 1990-04-17 The Regents Of The University Of California Assays and antibodies for N-MYC proteins
JPH0821101B2 (en) 1987-07-21 1996-03-04 三洋電機株式会社 Coin sorter
SE8801729D0 (en) 1988-05-06 1988-05-06 Astra Ab PURINE DERIVATIVES FOR USE IN THERAPY
US4923885A (en) * 1988-08-19 1990-05-08 Merck & Co., Inc. 5-amino-1-(4-naphthoylbenzyl)-1,2,3-triazole-4-carboxamides and analogs as antiproliferative agents
US5332744A (en) 1989-05-30 1994-07-26 Merck & Co., Inc. Substituted imidazo-fused 6-membered heterocycles as angiotensin II antagonists
ATE133678T1 (en) 1990-07-04 1996-02-15 Merrell Dow Pharma 9-PURINYL PHOSPHONIC ACID DERIVATIVES
GB9020931D0 (en) 1990-09-26 1990-11-07 Wellcome Found Heterocyclic compounds
DE69230349T2 (en) 1991-03-05 2000-07-27 Ajinomoto Kk Cyclopropane derivative
US5939420A (en) 1991-04-08 1999-08-17 Duquesne University Of The Holy Ghost Pyrrolo 2,3d!derivatives
CA2093403C (en) 1992-04-08 1999-08-10 Fumio Suzuki Therapeutic agent for parkinson's disease
JPH0680670A (en) 1992-09-03 1994-03-22 Ajinomoto Co Inc Cyclopropane derivative and its production
US5744492A (en) 1993-09-17 1998-04-28 United States Of America Method for inhibiting angiogenesis
JP3769737B2 (en) 1994-03-30 2006-04-26 味の素株式会社 Cyclopropane derivative and process for producing the same
US6110923A (en) 1994-06-22 2000-08-29 Biochem Pharma Inc. Method for treating cancer using novel substituted purinyl derivatives with immunomodulating activity
US5525606A (en) 1994-08-01 1996-06-11 The United States Of America As Represented By The Department Of Health And Human Services Substituted 06-benzylguanines and 6(4)-benzyloxypyrimidines
JPH0841035A (en) 1994-08-05 1996-02-13 Ajinomoto Co Inc Cyclopropane derivative and its production
JPH08208687A (en) 1994-11-25 1996-08-13 Sankyo Co Ltd Glyceryl oligonucleotide
JPH0920776A (en) 1995-06-30 1997-01-21 Nippon Paper Ind Co Ltd New purine nucleoside derivative, its production and antiviral agent with the same as active ingredient
JPH09169758A (en) 1995-10-18 1997-06-30 Nippon Paper Ind Co Ltd New purine nucleoside derivative, its production and antivirus using the same
JPH1025294A (en) 1996-03-26 1998-01-27 Akira Matsuda Condensed heterocyclic derivative, its production and malignant tumor therapeutic agent containing the same
EP0882727B9 (en) 1996-07-03 2005-06-15 Sumitomo Pharmaceuticals Company, Limited Novel purine derivatives
US6723727B1 (en) 1996-12-20 2004-04-20 Hoechst Aktiengesellschaft Substituted purine derivatives, processes for their preparation, their use, and compositions comprising them
US6284748B1 (en) 1997-03-07 2001-09-04 Metabasis Therapeutics, Inc. Purine inhibitors of fructose 1,6-bisphosphatase
DE69841549D1 (en) 1997-05-14 2010-04-22 Sloan Kettering Inst Cancer PROCESS AND PREPARATIONS FOR DESTRUCTION OF CERTAIN PROTEINS
HRP980375A2 (en) 1997-07-03 1999-04-30 Argyrios Georgios Arvanitis Imidazopyrimidines and imidazopyridines for the treatment of neurological disorders
JP2001509483A (en) 1997-07-12 2001-07-24 カンサー リサーチ キャンペーン テクノロジー リミテッド Cyclin-dependent kinase inhibitor purine derivatives
GB9717849D0 (en) 1997-08-23 1997-10-29 Seal Sands Chemicals Limited Preparation of pyridene derivatives
AUPO912997A0 (en) 1997-09-11 1997-10-02 Commonwealth Scientific And Industrial Research Organisation Antiviral agents
WO1999024432A1 (en) 1997-11-12 1999-05-20 Mitsubishi Chemical Corporation Purine derivatives and medicine containing the same as the active ingredient
TW572758B (en) 1997-12-22 2004-01-21 Sumitomo Pharma Type 2 helper T cell-selective immune response inhibitors comprising purine derivatives
AU3378599A (en) 1998-04-03 1999-10-25 University Of Pittsburgh Benzoquinoid ansamycins for the treatment of cardiac arrest and stroke
US5985060A (en) 1998-07-25 1999-11-16 Breed Automotive Technology, Inc. Gas generant compositions containing guanidines
SI20022A (en) 1998-07-29 2000-02-29 Kemijski inštitut Alkyl substituted purine derivatives and their preparation
JP2000072773A (en) 1998-08-28 2000-03-07 Zeria Pharmaceut Co Ltd Purine derivative
CZ27399A3 (en) 1999-01-26 2000-08-16 Ústav Experimentální Botaniky Av Čr Substituted nitrogen heterocyclic derivatives process of their preparation, the derivatives employed as medicaments, pharmaceutical composition and a compound pharmaceutical preparation in which these derivatives are comprised as well as use of these derivatives for preparing medicaments
DE60023268T2 (en) 1999-02-01 2006-06-01 CV Therapeutics, Inc., Palo Alto INHIBITEURS PURIQUES DE KINASE 2 DEPENDANTS OF CYCLINES et IkB-alpha
FR2790702B1 (en) 1999-03-08 2001-07-20 Sidel Sa MOLDING UNIT AND EXTRUSION-BLOWING MACHINE PROVIDED WITH SUCH A UNIT
AU4589800A (en) 1999-05-05 2000-11-21 Darwin Discovery Limited 9-(1,2,3,4-tetrahydronaphthalen-1-yl)-1,9-dihydropurin-6-one derivatives as pde7inhibitors
US6660845B1 (en) 1999-11-23 2003-12-09 Epoch Biosciences, Inc. Non-aggregating, non-quenching oligomers comprising nucleotide analogues; methods of synthesis and use thereof
CO5271715A1 (en) 1999-12-21 2003-04-30 Sugen Inc 7-AZA-INDOLIN-2-WAVES SUBSTITUTED IN 4 AND ITS USE AS PROTEIUNA QUINASA INHIBITORS
JP2003519232A (en) 2000-01-07 2003-06-17 ウニフェルジテーレ・インステリング・アントウェルペン Purine derivatives, their production and their use
WO2001072779A1 (en) 2000-03-24 2001-10-04 Duke University Characterization of grp94-ligand interactions and purification, screening, and therapeutic methods relating thereto
ATE502941T1 (en) 2000-04-25 2011-04-15 Icos Corp HUMAN PHOSPHATIDYL-INOSITOL-3-KINASE DELTA INHIBITORS
WO2002002123A1 (en) 2000-06-29 2002-01-10 Trustees Of Boston University Use of geldanamycin and related compounds for prophylaxis or treatment of fibrogenic disorders
US6946456B2 (en) 2000-07-28 2005-09-20 Sloan-Kettering Institute For Cancer Research Methods for treating cell proliferative disorders and viral infections
US6613089B1 (en) 2000-10-25 2003-09-02 Sdgi Holdings, Inc. Laterally expanding intervertebral fusion device
KR20030065502A (en) 2000-11-02 2003-08-06 슬로안-케테링인스티튜트퍼캔서리서치 Methods for enhancing the efficacy of cytotoxic agents through the use of hsp90 in-hibitors
GB0100623D0 (en) 2001-01-10 2001-02-21 Vernalis Res Ltd Chemical compounds IV
WO2002069900A2 (en) 2001-03-01 2002-09-12 Conforma Therapeutics Corp. Methods for treating genetically-defined proliferative disorders with hsp90 inhibitors
CA2377054C (en) * 2001-03-16 2006-09-05 Susan M. Boyetchko Biocontrol of weeds using pseudomonas compositions
ES2314042T3 (en) 2001-04-17 2009-03-16 Dainippon Sumitomo Pharma Co., Ltd. NEW DERIVATIVES OF ADENINA.
WO2002088079A2 (en) 2001-05-01 2002-11-07 Bristol-Myers Squibb Company Dual inhibitors of pde 7 and pde 4
EP1404871A4 (en) 2001-05-23 2006-10-04 Sloan Kettering Inst Cancer Method for treatment of cancer associated with elevated her 2 levels
PE20030008A1 (en) 2001-06-19 2003-01-22 Bristol Myers Squibb Co DUAL INHIBITORS OF PDE 7 AND PDE 4
BR0210594A (en) 2001-06-22 2005-11-01 Pharmasset Ltd (beta) -d or (beta) -1,3-halonucleoside
EP1399446B1 (en) 2001-06-27 2005-08-03 Cyclacel Limited 2,6,9-substituted purine derivatives and their use in the treatment of proliferative disorders
CN100378079C (en) 2001-09-24 2008-04-02 康福玛医药公司 Process for preparing 17-allyl amino geldanamycin (17-AAG) and other ansamycins
BR0212985A (en) 2001-09-24 2005-08-30 Imp College Innovations Ltd Use of pyy or an agonist in the preparation of medicaments for treating disorders associated with overweight
JP2003113181A (en) 2001-10-04 2003-04-18 Sumika Fine Chemicals Co Ltd Method for producing 6-halopurine
NZ545058A (en) * 2003-07-16 2008-05-30 Janssen Pharmaceutica Nv Triazolopyrimidine derivatives as glycogen synthase kinase 3 inhibitors
US7449465B2 (en) * 2003-07-16 2008-11-11 Janssen Pharmaceutica, N.V. Triazolopyrimidine derivatives as glycogen synthase kinase 3 inhibitors
US20080081817A1 (en) * 2004-07-23 2008-04-03 Tanabe Seiyaku Co., Ltd. Nitrogenous Fused Bicyclic Compound
EP1937258A2 (en) * 2005-09-23 2008-07-02 Conforma Therapeutics Corporation Anti-tumor methods using multi drug resistance independent synthetic hsp90 inhibitors
TWI640539B (en) 2015-03-13 2018-11-11 朱寶麒 Zinc binding fusion protein of glutathione s-transferase and metallothionein

Patent Citations (52)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4495190A (en) * 1980-12-22 1985-01-22 Astra Lakemedel Aktiebolag Derivatives of guanine for combating herpes virus infections
US4699877A (en) * 1982-11-04 1987-10-13 The Regents Of The University Of California Methods and compositions for detecting human tumors
US5098906A (en) * 1983-10-31 1992-03-24 Warner-Lambert Company Purine derivatives
US4921859A (en) * 1983-10-31 1990-05-01 Warner-Lambert Company Purine derivatives
US4547573A (en) * 1983-12-02 1985-10-15 Ici Pharma Process for preparing cephalosporin derivatives
US4748177A (en) * 1984-03-26 1988-05-31 Warner-Lambert Company Guanine derivatives
US4617304A (en) * 1984-04-10 1986-10-14 Merck & Co., Inc. Purine derivatives
US4774325A (en) * 1984-09-20 1988-09-27 Pierrel Spa New 8-substituted nucleoside and purine derivatives, the process for the preparation thereof and the pharmaceutical compositions containing them
US4806642A (en) * 1984-10-05 1989-02-21 Warner-Lambert Company Purine derivatives
US5217866A (en) * 1985-03-15 1993-06-08 Anti-Gene Development Group Polynucleotide assay reagent and method
US4772606A (en) * 1985-08-22 1988-09-20 Warner-Lambert Company Purine derivatives
US4923385A (en) * 1986-07-09 1990-05-08 Krauss-Maffei A.G. Blow molding machine
US5002950A (en) * 1986-10-24 1991-03-26 Warner-Lambert Co. 7-deazaguanines as immunomodulators
US4968603A (en) * 1986-12-31 1990-11-06 The Regents Of The University Of California Determination of status in neoplastic disease
US5204353A (en) * 1987-04-07 1993-04-20 Ciba-Geigy Corporation 3-benzyl-3H-1,2,3-triazolo[4,5-d]pyrimidines, compositions thereof, and method of treating epilepsy therewith
US5110818A (en) * 1988-10-06 1992-05-05 Ciba-Geigy Corporation Anticonvulsive substituted-9-benzyl-9h-purines
US5369008A (en) * 1991-05-24 1994-11-29 Board Of Regents, The University Of Texas System Methods for the detection of BCR-ABL and abnormal ABL proteins in leukemia patients
US6025126A (en) * 1991-10-28 2000-02-15 Arch Development Corporation Methods and compositions for the detection of chromosomal aberrations
US6005107A (en) * 1992-12-23 1999-12-21 Biochem Pharma, Inc. Antiviral compounds
US5955610A (en) * 1992-12-23 1999-09-21 Biochem Pharma, Inc. Antiviral compounds
US5789394A (en) * 1992-12-23 1998-08-04 Nguyen-Ba; Nghe Anti-viral compounds
US6444656B1 (en) * 1992-12-23 2002-09-03 Biochem Pharma, Inc. Antiviral phosphonate nucleotides
US5602156A (en) * 1993-09-17 1997-02-11 The United States Of America As Represented By The Department Of Health And Human Services Method for inhibiting metalloproteinase expression
US5917042A (en) * 1994-02-04 1999-06-29 Glaxo Wellcome Inc. Process for the preparation of 2,5-diamino-4,6-dichloropyrimidine
US5932566A (en) * 1994-06-16 1999-08-03 Pfizer Inc. Ansamycin derivatives as antioncogene and anticancer agents
US5846749A (en) * 1994-10-12 1998-12-08 The Regents Of The University Of California Quantitative measurement of tissue protein identified by immunohistochemistry and standardized protein determination
US5656629A (en) * 1995-03-10 1997-08-12 Sanofi Winthrop, Inc. 6-substituted pyrazolo (3,4-d)pyrimidin-4-ones and compositions and methods of use thereof
US6177460B1 (en) * 1995-04-12 2001-01-23 The Procter & Gamble Company Method of treatment for cancer or viral infections
US6262254B1 (en) * 1997-04-30 2001-07-17 The Regents Of Univ. Of California 8-fluoropurine compounds
US5861503A (en) * 1997-04-30 1999-01-19 The Regents Of The University Of California Process for producing 8-fluoropurines
US6210974B1 (en) * 1997-10-24 2001-04-03 Oregon Health Sciences University Compositions and methods for promoting nerve regeneration
US6369092B1 (en) * 1998-11-23 2002-04-09 Cell Pathways, Inc. Method for treating neoplasia by exposure to substituted benzimidazole derivatives
US20030113331A1 (en) * 1999-01-06 2003-06-19 Brooks Peter C. Method and composition for angiogenesis inhibition
US6174875B1 (en) * 1999-04-01 2001-01-16 University Of Pittsburgh Benzoquinoid ansamycins for the treatment of cardiac arrest and stroke
US20020161014A1 (en) * 2000-04-25 2002-10-31 Chanchal Sadhu Inhibitors of human phosphatidylinositol 3-kinase delta
US20040102458A1 (en) * 2000-11-02 2004-05-27 Gabriela Chiosis Small molecule compositions for binding to hsp90
US7087754B2 (en) * 2001-01-10 2006-08-08 Vernalis Research Limited Pyrazolo[3,4-d]pyrimidine derivatives and their use as purinergic receptor antagonists
US20040097526A1 (en) * 2001-01-10 2004-05-20 Gillespie Roger John Triazolo[4,5-d]pyrimidine derivatives and their use as purinergic receptor antagonists
US20040063668A1 (en) * 2001-01-19 2004-04-01 Jong-Ryoo Choi Novel acyclic nucleoside phosphonate derivatives, salts thereof and process for the preparation of the same
US20020156277A1 (en) * 2001-04-20 2002-10-24 Fick David B. Synthesis and methods of use of purine analogues and derivatives
US20030022864A1 (en) * 2001-04-24 2003-01-30 Ishaq Khalid S. 9-[(5-dihydroxyboryl)-pentyl] purines, useful as an inhibitor of inflammatory cytokines
US20030078413A1 (en) * 2001-09-12 2003-04-24 Epoch Biosciences, Inc. Process for the synthesis of pyrazolopyrimidines
US20050049263A1 (en) * 2001-10-30 2005-03-03 Kasibhatla Srinivas Rao Purine analogs having hsp90-inhibiting activity
US20050256183A1 (en) * 2001-11-09 2005-11-17 Kasibhatla Srinivas R Hsp90-inhibiting zearalanol compounds and methods of producing and using same
US20060035909A1 (en) * 2002-08-23 2006-02-16 Kveta Fuksova Azapurine derivatives
US20040241706A1 (en) * 2002-09-13 2004-12-02 Irm, Llc Highly specific modulators of GTPases for target validation
US20040242490A1 (en) * 2003-02-20 2004-12-02 Brooks Peter C. CLK-peptide and SLK-peptide
US20050043239A1 (en) * 2003-08-14 2005-02-24 Jason Douangpanya Methods of inhibiting immune responses stimulated by an endogenous factor
US20050107343A1 (en) * 2003-09-18 2005-05-19 Conforma Therapeutics Corporation Pyrrolopyrimidines and related analogs as HSP90-inhibitors
US20050113339A1 (en) * 2003-09-18 2005-05-26 Kasibhatla Srinivas R. Triazolopyrimidines and related analogs as HSP90-inhibitors
US20050119282A1 (en) * 2003-09-18 2005-06-02 Conforma Therapeutics Corporation Pyrazolopyrimidines and related analogs as HSP90-inhibitors
US7138401B2 (en) * 2003-09-18 2006-11-21 Conforma Therapeutics Corporation 2-aminopurine analogs having HSP90-inhibiting activity

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060223797A1 (en) * 2005-03-30 2006-10-05 Conforma Therapeutics Corporation Alkynyl pyrrolopyrimidines and related analogs as hsp90-inhibitors
US7544672B2 (en) 2005-03-30 2009-06-09 Conforma Therapeutics Corporation Alkynyl pyrrolo[2,3-d]pyrimidines and related analogs as HSP90-inhibitors
US8093229B2 (en) 2005-03-30 2012-01-10 Conforma Therapeutics Corporation Alkynyl pyrrolo[2,3-d]pyrimidines and related analogs as HSP90-inhibitors
US20070105874A1 (en) * 2005-09-23 2007-05-10 Conforma Therapeutics Corporation Anti-Tumor Methods Using Multi Drug Resistance Independent Synthetic HSP90 Inhibitors
US20070253896A1 (en) * 2006-02-07 2007-11-01 Conforma Therapeutics Corporation 7,9-Dihydro-Purin-8-One and Related Analogs as HSP90-Inhibitors

Also Published As

Publication number Publication date
CA2539548A1 (en) 2005-03-31
IL174375A0 (en) 2006-08-01
US7129244B2 (en) 2006-10-31
CN101906106A (en) 2010-12-08
AU2010202750B2 (en) 2011-09-01
US7138401B2 (en) 2006-11-21
BRPI0414533A (en) 2006-11-07
EA010160B1 (en) 2008-06-30
MXPA06002997A (en) 2007-02-08
US20070111997A1 (en) 2007-05-17
US7138402B2 (en) 2006-11-21
US20050113340A1 (en) 2005-05-26
WO2005028434A2 (en) 2005-03-31
EA200600594A1 (en) 2006-10-27
US20070173483A1 (en) 2007-07-26
AU2004274507A1 (en) 2005-03-31
US20070111996A1 (en) 2007-05-17
WO2005028434A3 (en) 2006-03-23
EP2145888A1 (en) 2010-01-20
EP1670802A4 (en) 2010-07-07
US7148228B2 (en) 2006-12-12
AU2004274507B2 (en) 2010-08-19
NZ546611A (en) 2010-02-26
NO20061396L (en) 2006-06-16
AU2010202750A1 (en) 2010-07-22
KR20060070572A (en) 2006-06-23
US20050119282A1 (en) 2005-06-02
US20050107343A1 (en) 2005-05-19
US20050113339A1 (en) 2005-05-26
JP2007505933A (en) 2007-03-15
EP1670802A2 (en) 2006-06-21

Similar Documents

Publication Publication Date Title
US7138401B2 (en) 2-aminopurine analogs having HSP90-inhibiting activity
US20080096903A1 (en) Sulfamoyl-containing derivatives and uses thereof
US8093229B2 (en) Alkynyl pyrrolo[2,3-d]pyrimidines and related analogs as HSP90-inhibitors
US7241890B2 (en) Purine analogs having HSP90-inhibiting activity
US20070253896A1 (en) 7,9-Dihydro-Purin-8-One and Related Analogs as HSP90-Inhibitors
ZA200602786B (en) Novel heterocyclic compounds as HSP90-inhibitors

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION