WO1997049706A1 - SUBSTITUTED 7-AMINO-PYRROLO[3,2-d]PYRIMIDINES AND THE USE THEREOF - Google Patents

SUBSTITUTED 7-AMINO-PYRROLO[3,2-d]PYRIMIDINES AND THE USE THEREOF Download PDF

Info

Publication number
WO1997049706A1
WO1997049706A1 PCT/EP1997/003115 EP9703115W WO9749706A1 WO 1997049706 A1 WO1997049706 A1 WO 1997049706A1 EP 9703115 W EP9703115 W EP 9703115W WO 9749706 A1 WO9749706 A1 WO 9749706A1
Authority
WO
WIPO (PCT)
Prior art keywords
pyrrolo
phenyl
unsubstituted
substituted
pyrimidin
Prior art date
Application number
PCT/EP1997/003115
Other languages
French (fr)
Inventor
Eva Altmann
Martin Missbach
Leo Widler
Jürgen Klaus MAIBAUM
Original Assignee
Novartis Ag
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 Novartis Ag filed Critical Novartis Ag
Priority to AU31762/97A priority Critical patent/AU3176297A/en
Publication of WO1997049706A1 publication Critical patent/WO1997049706A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Definitions

  • the invention relates to compounds of formula I
  • R is unsubstituted or substituted lower alkyl or hetero-substituted lower alkyl, unsubstituted or substituted cyclo-lower hydrocarbyl or cyclo-lower hydrocarbyl-lower alkyl, unsubstituted or substituted aryl or aryl-lower alkyl, or unsubstituted or substituted hetero-lower cyclyl or hetero-lower cyclyl-lower alkyl;
  • R 2 is hydrogen, lower alkyl or halogen
  • R 3 is unsubstituted or substituted lower alkyl or hetero-substituted lower alkyl, unsubstituted or substituted cyclo-lower hydrocarbyl or cyclo-lower hydrocarbyl-lower alkyl, unsubstituted or substituted aryl or aryl-lower alkyl, or unsubstituted or substituted hetero-lower cyclyl or hetero-lower cyclyl-lower alkyl; and salts thereof.
  • lower denotes a radical having up to and including 7, and especially up to and including 6, carbon atoms.
  • Lower alkyl is, for example, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, ferf-butyl, n-pentyl, neopentyl, n-hexyl or n-heptyl, preferably ethyl or methyl.
  • Alkyl is straight-chain or branched.
  • lower alkyl On its own, for example lower alkyl, or as a constituent of other groups, for example lower alkoxy, lower alkylcarbonyl, lower alkoxycarbonyl, lower alkylaminocarbonyl, di-lower alkylaminocarbonyl, it may be unsubstituted or mono- or poly- substituted, for example by halogen, hydroxy, lower alkoxy, t ⁇ fluoromethyl or by imidazolyl, and is preferably unsubstituted or substituted by halogen, hydroxy, lower alkoxy, ammo, N- lower alkylamino or by N,N-d ⁇ -lower alkylamino.
  • hetero-substituted alkyl the carbon chain is interrupted by one or more, identical or different hetero atoms, for example by oxygen, sulfur or S(O 2 ), especially oxygen
  • Cyclo-lower hydrocarbyl denotes saturated or mono- or poly-unsaturated carbocyciic rings, preferably having from 3 up to and including 7 ring members, especially 5 or 6 ring mem ⁇ bers, for example cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclopentadienyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl.
  • Cyclo-lower hydrocarbyl-lower alkyl is, for example, cyclopentylmethyl, cyclopentenylmethyl, cyclopentylethyl, cyclopentylpropyl, cyclohexylmethyl, cyclohexenylethyl
  • Cyclo-lower hydrocarbyl and cyclo-lower hydrocarbyl- lower alkyl may be unsubstituted or mono- or poly-substituted, for example by hydroxy, lower alkyl, lower alkoxy, hydroxy-lower alkyl, hydroxy-lower alkoxy, halo-lower alkyl, halo- lower alkoxy, lower alkoxy-lower alkoxy, ammo-lower alkyl, ammo-lower alkoxy, lower alkylamino-lower alkyl, lower alkylamino-lower alkoxy, N,N-d ⁇ -lower alkylamino-lower alkyl,
  • Aryl is, for example, phenyl or naphthyl, each of which is unsubstituted or substituted, for example as indicated hereinafter for phenyl
  • Aryl is preferably phenyl unsubstituted or substituted by one or more, for example from one to three, especially one or two, substi ⁇ tuents from the group consisting of hydroxy, lower alkyl, halo-lower alkyl, (hydroxy or lower alkanoyloxy)-lower alkyl, lower alkoxy-lower alkyl, (hydroxy, lower alkoxy or lower alkanoyl- oxy)-lower alkoxy-lower alkyl, (ammo, lower alkylamino, di-lower alkylamino or lower alkanoylam ⁇ no)-lower alkoxy-lower alkyl, (ammo or lower aikanoylam ⁇ no)-lower alkyl, lower alkylamino-lower al
  • the two hetero atoms are preferably separated from one another by at least two carbon atoms; in other words, the lower alkyl moiety is preferably so selected that there are at least two carbon atoms between the two hetero atoms.
  • Halogen is, for example, chlorine, bromine or fluorine, but may also be iodine.
  • Hetero-lower cyclyl is a saturated, mono- or poly-unsaturated or aromatic radical having from 3 up to and including 7 ring atoms, especially from 3 up to and including 6 ring atoms, wherein at least one of the ring atoms is a hetero atom, preferably nitrogen, oxygen or sulfur.
  • a hetero atom preferably nitrogen, oxygen or sulfur.
  • aza-lower cycloalkyl and aza-heteroaryl that is to say hetero-lower cyclyl radicals that contain at least one nitrogen ring atom.
  • the hetero atoms may be identical or different; one ring hetero atom is preferred.
  • Hetero-lower cyclyl is, for example, piperidmyl, piperazinyl, morpholinyl, pyrrolidinyl, furanyl, tetrahydrofuran, thiophenyl, imidazolyl, oxazolyl, thiazolyl, triazolyl, pyranyl, thiopyranyl, pyridyl, pyrrolyl or tetrazolyl.
  • the compounds I and their salts may be in the form of one of the possible isomers, for example stereoisomers, diastereoisomers or tautomers, or in the form of a mixture thereof.
  • isomers for example, pure enantiomers, pure diastereo- isomers or, where appropriate, pure tautomers.
  • isomeric mixtures may be in the form of, for example, racemates or diastereoisomeric mixtures.
  • Salts of compounds of formula I are especially pharmaceutically acceptable salts, especially acid addition salts with suitable mineral acids, such as hydrohalic acids, sulfuric acid or phosphoric acid, for example hydrochlorides, hydrobromides, sulfates, hydrogen sulfates or phosphates, salts with suitable aliphatic or aromatic sulfonic acids or N-substituted sulfamic acids, for example methanesulfonates, benzenesulfonates, p-toluenesulfonates or N-cyclo- hexylsulfamates (cyclamates), or salts with strong organic carboxylic acids, such as lower alkanecarboxylic acids or saturated or unsaturated or hydroxylated aliphatic dicarboxylic acids, for example acetates, oxalates, malonates, maleates, fumarates, tartrates or citrates.
  • suitable mineral acids such as hydrohalic acids, sulfuric acid or phosphoric
  • salts with bases for example corresponding alkali metal or alkaline earth metal salts, for example sodium, potassium or magnesium salts, pharmaceutically acceptable transition metal salts, such as zinc or copper salts, or salts with ammonia or organic amines, such as cyclic amines, mono-, di- or tri-lower alkylamines, hydroxy-lower alkylamines, for example mono-, di- or tri-hydroxy-lower alkylamines, hydroxy-lower alkyl-lower alkyl-amines or poly- hydroxy-lower alkylamines.
  • bases for example corresponding alkali metal or alkaline earth metal salts, for example sodium, potassium or magnesium salts
  • pharmaceutically acceptable transition metal salts such as zinc or copper salts
  • salts with ammonia or organic amines such as cyclic amines, mono-, di- or tri-lower alkylamines, hydroxy-lower alkylamines, for example mono-, di- or tri-hydroxy-low
  • Cyclic amines are, for example, morpholine, thiomorpholine, piperidine or pyrrolidine.
  • Suitable mono-lower alkylamines are, for example, ethyl- and tert- butyl-amine;
  • suitable di-lower alkylamines are, for example, diethyl- and diisopropyl-amine and
  • suitable tri-lower alkylamines are, for example, trimethyl- and triethyl-amine.
  • Suitable hydroxy-lower alkylamines are, for example, mono-, di- and tri-ethanolamine; hydroxy-lower alkyl-lower alkyl-amines are, for example, N,N-dimethylamino- and N,N-diethylamino- ethanol.
  • Compounds of formula I having an acid group, for example carboxy, and a basic group, for example amino may also be in the form of, for example, internal salts, i.e. in zwitterionic form, or part of the molecule may be in the form of an internal salt and another part in the form of a normal salt.
  • Pharmaceutically unacceptable salts are also included, since they can be used, for example, for the isolation and/or purification of free compounds I and the pharmaceutically acceptable salts thereof.
  • the compounds of formula I have valuable pharmacological properties. In particular, they inhibit the activity of tyrosine protein kinase pp60 c src in concentrations of from approximately 0.001 to approximately 10 ⁇ M [test description: K. Farley et al., Anal. Biochem. 203 (1992) 151-157; purified enzyme - as described in N. B. Lydon er a/., Biochem. J. 287 (1992) 985-
  • the compounds of formula I are therefore capable of inhibiting the bone absorption ability of osteoclasts. That can be demonstrated, for example, in the bone slice assay on bovine cortical bone platelets with rat osteoclasts in concentrations of from approx. 0.001 to approx 10 ⁇ M. [The "bone slice assay" is described, for example, in Biochem. Biophys. Res. Comm 188 (1992) 1097- 1103] In that assay, the compounds of formula I inhibit the formation of characteristic absorption holes in bone platelets in vitro
  • the effectiveness of compounds of formula I can be demonstrated, for example, in the Hock model in the rat In that test, the compounds of formula I - when administered once a day per os in concentrations of from approx 1 to approx 100 mg/kg of body weight - for from 3 to 4 weeks completely or at least partially inhibit the bone loss produced as a result of ovanectomy in rats [the "Hock model" is described, for example, in Metab. Bone
  • the in vivo activity of compounds of formula I can also be demonstrated, for example, via calcium metabolism in intact rats.
  • acute hypocalcaemia is induced within from 1 to 4 hours; it is demonstrated by determining the concentration of calcium in the blood plasma.
  • the observation of acute hypocalcaemia can be interpreted as indirect evidence that the test compound inhibits bone absorption
  • the compounds of formula I are therefore very suitable for the treatment of diseases that are responsive to inhibition of the activity of tyrosine protein kinase pp60 c src .
  • the com ⁇ pounds of formula I are useful in the treatment of benign or malignant tumours that are responsive to inhibition of tyrosine protein kinase pp60 c"src , such as breast cancer (mammary carcinoma) or intestinal cancer (colon carcinoma). They are capable of effecting tumour regression and of preventing the formation of tumour metastases and the growth of micrometastases.
  • the compounds of formula I are also useful in the treatment of cardio ⁇ vascular disorders, such as thrombosis.
  • the compounds of formula I also inhibit the activity of other non-receptor tyrosine protein kmases, such as (a) other members of the src family, for example Ick and fyn, (b) abl kinase and (c) ZAP70 kinase Furthermore, the compounds of formula I also inhibit the activity of receptor tyrosine protein kmases, such as (a) the EGF family, for example the EGF receptor, c-erbB2, c-erbB3 and c-erbB4, and (b) the PDGF family, for example the PDGF receptor, CSF-1 , Kit, VEGF and FGF.
  • other non-receptor tyrosine protein kmases such as (a) other members of the src family, for example Ick and fyn, (b) abl kinase and (c) ZAP70 kinase
  • the compounds of formula I also inhibit the activity of receptor tyrosine protein kmases,
  • the compounds of formula I can also be used in immunomodulation and in the treatment of diseases of the immune system, for example in the case of inflammations or organ transplants They are also suitable for the treatment of (hyper)prol ⁇ ferat ⁇ ve diseases, such as psoriasis, tumours, carcinomas and leukaemias, and in fibrosis and restenosis.
  • the compounds of formula I can also be used in the treatment of diseases of the central or the peripheral nervous system where signal transmission by at least one tyrosine protein kinase is involved.
  • the invention relates preferably to compounds of formula I wherein
  • R is unsubstituted or substituted lower alkyl or hetero-substituted lower alkyl, unsubstituted or substituted cyclo-lower hydrocarbyl, unsubstituted or substituted aryl, unsubstituted or substituted aryl-lower alkyl, or unsubstituted or substituted hetero-lower cyclyl;
  • R 2 is hydrogen, lower alkyl or halogen
  • R 3 is unsubstituted or substituted lower alkyl or hetero-substituted lower alkyl, unsubstituted or substituted cyclo-lower hydrocarbyl, unsubstituted or substituted aryl or aryl-lower alkyl, or unsubstituted or substituted hetero-lower cyclyl; and salts thereof
  • the invention relates especially to compounds of formula I wherein
  • R is unsubstituted or substituted lower alkyl or hetero-substituted lower alkyl, unsubstituted or substituted cyclo-lower hydrocarbyl, unsubstituted or substituted aryl, unsubstituted or substituted aryl-lower alkyl, or unsubstituted or substituted hetero-lower cyclyl;
  • R 2 is hydrogen, lower alkyl or halogen
  • R 3 is unsubstituted or substituted lower alkyl or hetero-substituted lower alkyl, unsubstituted or substituted cyclo-lower hydrocarbyl, or unsubstituted or substituted aryl; and salts thereof.
  • the invention relates more especially to compounds of formula I wherein
  • R is unsubstituted or substituted lower alkyl or hetero-substituted lower alkyl, unsubstituted or substituted cyclo-lower hydrocarbyl, unsubstituted or substituted aryl, unsubstituted or substituted aryl-lower alkyl, or unsubstituted or substituted hetero-lower cyclyl;
  • R 2 is hydrogen
  • R 3 is unsubstituted or substituted lower alkyl or hetero-substituted lower alkyl, unsubstituted or substituted cyclo-lower hydrocarbyl, or unsubstituted or substituted aryl; and salts thereof.
  • the invention relates most especially to compounds of formula I wherein
  • Ri is unsubstituted or substituted lower alkyl or hetero-substituted lower alkyl, unsubstituted or substituted cyclo-lower hydrocarbyl, unsubstituted or substituted aryl, unsubstituted or substituted aryl-lower alkyl, or unsubstituted or substituted hetero-lower cyclyl;
  • R 2 is hydrogen
  • R 3 is unsubstituted or substituted lower alkyl, unsubstituted or substituted cyclo-lower hydrocarbyl, or unsubstituted or substituted aryl; and pharmaceutically acceptable salts thereof.
  • the invention relates especially to the specific compounds described in the Examples and to salts thereof.
  • the compounds of formula I can be prepared in a manner known perse, for example by a) subjecting a compound of formula II wherein
  • R,, R 2 and R 3 are as defined above, and R 4 is lower alkyl, to a ring-closure reaction, with the formation of the pyrimidine ring, to give a compound of formula III
  • R 1 t R 2 and R 3 are as defined above, to a ring-closure reaction with the formation of the pyrimidine ring; or
  • R 2 and R 3 are as defined above, with a compound of formula VI wherein Ri is as defined above and X is a leaving group;
  • Suitable cyclisation reagents are the sequence 1. N,N-di-lower alkylformamide di-lower alkyl acetal and 2. ammonia.
  • Preferred N,N-di-lower alkylformamide di-lower alkyl acetals are di-lower alkyl acetals of N,N-dimethylformamide, especially N,N-di-lower alkylformamide dineopentyl acetal.
  • the reaction of compounds of formula II is preferably effected at elevated tempera ⁇ tures, for example at the reflux temperature of the reaction mixture.
  • the reaction can be carried out with or without solvent, but preferably takes place in an aprotic solvent or solvent mixture.
  • the resulting dialkylamino-methyleneaminoalkoxycarbonyl-pyrroles are then cyclised with ammonia to form a compound of formula III.
  • the latter reaction is preferably effected in a polar solvent or solvent mixture in a bomb tube at ambient temperature or slightly elevated temperature whereby also one of the reactants may serve as solvent or component of a solvent mixture.
  • the 4-oxo group can then be replaced in a manner known per se by halogen, especially chlorine, by reaction with a phosphorus oxytrihalide, prefer ⁇ ably phosphorus oxytrichloride.
  • the reaction can be carried out with or without solvent, but preferably takes place without solvent at distinctly elevated temperature, for example at 100°C to 120°C.
  • the 4-halide can be replaced by -NH 2 by treating the reaction product with ammonia. That reaction is generally carried out at relatively high temperatures, for example at 130°C in an autoclave.
  • the compounds of formula II are preferably prepared using one of the known methods of pyrrole synthesis. They are obtained, for example, by cyclising a compound of formula I la
  • a base for example sodium ethanolate/ethanol.
  • Process (b) corresponds to the cyclisation known perse of 2-am ⁇ no-3-cyano-pyrroles to 4-am ⁇ no-pyrrolo[2,3-d]pyr ⁇ m ⁇ d ⁇ nes.
  • Suitable cyclisa ⁇ tion reagents are, for example, (1) formamide or (2) 1. trialkyi orthoformate/2. ammonia.
  • the cyclisation of compounds of formula IV with formamide is preferably carried out at elevated temperature, for example at 160°C, and advantageously with the addition of a small amount of dimethylformamide and formic acid.
  • the reaction of compounds of formula IV with trialkyi orthoformate to give the corresponding alkoxy formimidates formed as intermediates norm ⁇ ally takes place at less elevated temperatures, for example at from 80 to 140°C
  • the reac ⁇ tion of the latter with ammonia can then be carried out generally at relatively low tempera ⁇ tures, for example at ambient temperature.
  • the cyclisation then takes place preferably with base catalysis, especially using sodium ethanolate/ethanol, once again at elevated tem ⁇ perature, for example at the reflux temperature of the reaction mixture.
  • Suitable leaving groups are, for example, methanesulfonates or p-toluenesulf- onates of hydroxy compounds and halogen.
  • the preparation of suitable pyrrolo[3,2-d]- py ⁇ midmes of formula V is known from the literature or can be carried out analogously to the methods described in the literature.
  • the reaction of compounds of formula V with compounds of formula VI is carried out in a manner known per se. For example, a methane- sulfonate of formula VI is reacted with a pyrrolo[3,2-d]pyr ⁇ m ⁇ d ⁇ ne of formula V in the presence of a base, for example potassium carbonate.
  • the reaction is preferably carried out at elevated temperature, for example at from 50°C to the reflux temperature of the reaction mixture, especially at from 60 to 80°C, and advantageously in an inert solvent or solvent mixture.
  • the reaction can be accelerated in an advantageous manner by the addition of a suitable crown ether.
  • the reaction takes place in a manner known per se under the conditions of phase transfer catalysis (E.V. Dehmlow and S. S. Dehmlow, Phase Transfer Catalysis, 3rd ed., VCH, Weinheim, 1993).
  • the reactants of formulae V and VI are dissolved in a suitable inert solvent or solvent mixture, and the second phase is formed by a concentrated aqueous alkali metal hydroxide solution, for example 30 % sodium hydroxide solution.
  • a phase transfer catalyst for example a quaternary ammonium halide, such as tetrabutylammonium bromide, is added.
  • halo-lower alkyl e.g. chloromethyl
  • halo-lower alkyl e.g. chloromethyl
  • Hydroxy can be reacted, for example, with unsubstituted or substituted halo-lower alkanes, yielding unsubstituted or substituted lower alkoxy. Hydroxy can, for example, also be reacted first with a di-halo-lower alkane, for example 1 -bromo-2-chloroethane, yielding ⁇ -halo-lower alkoxy; the latter can be reacted in a manner analogous to that described above with unsubstituted or substititued lower alkanols, lower alkanethiols or lower alkyl ⁇ amines in accordance with a nucleophilic substitution reaction, yielding unsubstituted or substituted lower alkoxy-lower alkoxy, lower alkylthio-lower alkoxy or lower alkylamino-lower alkoxy, respectively.
  • a di-halo-lower alkane for example 1 -bromo-2-chloroethan
  • mercapto can also be alkylated as described in the preceding paragraph.
  • Lower alkylthio groups can be converted by controlled oxidation both into lower alkylsulfinyl groups and into lower alkylsulfonyl groups.
  • Amino groups and hydroxy groups can be acylated in known manner, yielding, for example, lower alkanoylar ⁇ ino and lower alkanoyloxy groups, respectively.
  • Carboxylic acid radicals can be converted in accordance with known derivatisation methods, such as esterification or amide formation, into carboxylic acid derivatives, such as lower alkoxycarbonyl, aminocarbonyl, N-lower alkylaminocarbonyl, N,N-di-lower alkylamino ⁇ carbonyl, cyano or amidino.
  • carboxylic acid derivatives can also be converted into free carboxylic acids, for example by hydrolysis.
  • hydroxy groups can be esterified with organic or inorganic acids or etherified with alcohols or organic halides or they can be removed by reduction.
  • any of the intermediates contain troublesome reactive groups, for example carboxy, hydroxy, mercapto or amino groups, those groups can be protected temporarily by readily removable protecting groups.
  • suitable protecting groups their introduction and their removal are known perse and are described, for example, in J.F.W. McOmie, Protective Groups in Organic Chemistry, Plenum Press, London, New York 1973 and, for example, in T.W. Greene and P.G.M. Wuts, Protective Groups in Organic Synthesis, Wiley, New York et al., 1991 , or also in PJ. Kocienski, Protecting Groups, Thieme, Stuttgart, New York 1994.
  • Salts of compounds I can be prepared in a manner known per se.
  • acid addition salts of compounds I are obtained by treatment with a suitable acid or a suitable ion exchange reagent and salts with bases by treatment with a suitable base or a suitable ion exchange reagent.
  • Salts of compounds of formula I can be converted into the free compounds I in customary manner: acid addition salts, for example, by treatment with a suitable basic agent or a suitable ion exchange reagent and salts with bases, for example, by treatment with a suitable acid or a suitable ion exchange reagent.
  • Salts of compounds I can be converted in a manner known per se into other salts of compounds I: acid addition salts can be converted, for example, into other acid addition salts, for example by treatment of a salt of an inorganic acid, such as a hydrochloride, with a suitable metal salt, such as a sodium, barium or silver salt, of an acid, for example with silver acetate, in a suitable solvent in which an inorganic salt that forms, for example silver chloride, is insoluble and thus precipitates from the reaction mixture.
  • a salt of an inorganic acid such as a hydrochloride
  • a suitable metal salt such as a sodium, barium or silver salt
  • any reference to the free compound I or its salts should be understood as including also the corresponding salts or the free compound I, respect ⁇ ively, as approp ⁇ ate and expedient
  • the compounds I including the salts of salt-forming compounds, can also be obtained in the form of their hydrates and/or may include other solvents, for example solvents that may have been used for the crystallisation of compounds in solid form
  • the compounds I and their salts may be in the form of one of the possible isomers or in the form of a mixture thereof.
  • isomers for example, pure diastereoisomers
  • isome ⁇ c mixtures may be in the form of, for example, diastereoisomenc mixtures.
  • Isome ⁇ c mixtures of compounds I in free form or in salt form obtainable in accordance with the process or by another method can be separated into their components in customary manner, for example on the basis of the physico-chemical differences between the con ⁇ stituents in known manner by fractional crystallisation, distillation and/or chromatography
  • the more active isomer is isolated.
  • the invention relates also to those forms of the process in which a compound obtainable as intermediate at any stage of the process is used as starting materiai and the remaining steps are carried out or a starting material is used in the form of a derivative or salt or, especially, is formed under the reaction conditions.
  • the invention relates also to the use of compounds I and their pharmaceutically acceptable salts in the treatment of allergic conditions and diseases, preferably in the form of pharma ⁇ ceutically acceptable preparations, especially in a method for the therapeutic treatment of the animal or human body, and to such a method of treatment.
  • the invention relates also to pharmaceutical compositions comprising as active ingredient a compound I or a pharmaceutically acceptable salt thereof, and to processes for their preparation.
  • Those pharmaceutical compositions are, for example, for enteral, such as especially oral, also rectal, administration, for parenteral administration and for local administration to warm-blooded animals, especially humans, the compositions comprising the pharmacological active ingredient on its own or together with customary pharmaceutical excipients.
  • the pharmaceutical compositions comprise (in percent by weight) for example from approximately 0.001 % to 100 %, preferably from approximately 0.1 % to approxi ⁇ mately 50 %, active ingredient.
  • compositions for enteral or parenteral administration are, for example, those in unit dose forms, such as dragees, tablets, capsules or suppositories, and also ampoules. They are prepared in a manner known perse, for example by means of conventional mixing, granulating, confectioning, dissolving or lyophilising procedures.
  • pharmaceutical compositions for oral administration can be obtained by combining the active ingredient with solid carriers, if desired granulating a resulting mixture, and processing the mixture or granules, if desired or necessary after the addition of appropriate excipients, into tablets or dragee cores.
  • Suitable carriers are especially fillers, such as sugars, for example lactose, saccharose, mannitol or sorbitol, cellulose preparations and/or calcium phosphates, for example tricalcium phosphate or calcium hydrogen phosphate, and also binders, such as starch pastes using, for example, corn, wheat, rice or potato starch, gelatin, tragacanth, methyl- cellulose and/or polyvinylpyrrolidone, and, if desired, disintegrators, such as the above- mentioned starches, also carboxymethyl starch, crosslinked polyvinylpyrrolidone, agar or alginic acid or a salt thereof, such as sodium alginate.
  • fillers such as sugars, for example lactose, saccharose, mannitol or sorbitol, cellulose preparations and/or calcium phosphates, for example tricalcium phosphate or calcium hydrogen phosphate
  • binders such as starch pastes using, for example, corn
  • Excipients are especially flow conditioners and lubricants, for example silicic acid, talc, stearic acid or salts thereof, such as magnesium or calcium stearate, and/or polyethylene glycol.
  • Dragee cores are provided with suitable, optionally enteric, coatings, there being used, inter alia, concentrated sugar solutions which may comprise gum arabic, talc, polyvinylpyrrolidone, polyethylene glycol and/or titanium dioxide, or coating solutions in suitable organic solvents or solvent mixtures, or, for the preparation of enteric coatings, solutions of suitable cellulose preparations, such as acetylcellulose phthalate or hydroxypropylmethylcellulose phthalate. Dyes or pigments may be added to the tablets or dragee coatings, for example for identification purposes or to indicate different doses of active ingredient.
  • compositions are hard gelatin capsules, and also soft, sealed capsules consisting of gelatin and a plasticiser, such as glycerol or sorbitol.
  • the hard gelatin capsules may comprise the active ingredient in the form of granules, for example in admixture with fillers, such as lactose, binders, such as starches, and/or glidants, such as talc or magnesium stearate, and optionally stabilisers.
  • the active ingredient is preferably dissolved or suspended in suitable liquids, such as fatty oils, paraffin oil or liquid polyethylene glycols, to which stabilisers may also have been added.
  • Suitable rectally administrable pharmaceutical compositions are, for example, suppositories that consist of a combination of the active ingredient with a suppository base material.
  • Suitable suppository base materials are, for example, natural or synthetic triglycerides, paraffin hydrocarbons, polyethylene glycols or higher alkanols. It is also possible to use gelatin rectal capsules which comprise a combination of the active ingredient with a base material.
  • Suitable base materials are, for example, liquid triglycerides, polyethylene glycols or paraffin hydrocarbons.
  • aqueous solutions of an active ingredient in water-soluble form for example in the form of a water-soluble salt
  • suspensions of the active ingredient such as corresponding oily injection suspensions
  • suitable lipophilic solvents or vehicles such as fatty oils, for example sesame oil, or synthetic fatty acid esters, for example ethyl ol ⁇ ate or triglycerides
  • aqueous injection suspensions which comprise viscosity-increasing substances, for example sodium carboxymethylcellulose, sorbitol and/or dextran, and optionally also stabilisers.
  • compositions for local administration are, for example for topical treatment of the skin, lotions, creams and ointments, i.e. liquid or semi-solid oil-in-water or water-in-oil emulsions, fatty ointments, which are anhydrous, pastes, i.e. creams and ointments having secretion-absorbing powder constituents, gels, which are aqueous, of low water content or anhydrous and consist of swellable, gel-forming materials, foams, i.e.
  • liquid oil-in-water emulsions in aerosol form which are administered from pressurised containers, and tinctures having an aqueous-ethanolic base and may comprise other customary pharma ⁇ ceutical excipients, such as preservatives.
  • the pharmaceutical compositions for local administration are prepared in a manner known per se by mixing the active ingredient with the pharmaceutical excipients, for example by dissolving or suspending the active ingred ⁇ ient in the base or in a portion thereof, if necessary.
  • the active ingredient is normally dissolved therein prior to emulsification; in order to prepare suspensions in which the active ingredient is suspended in the emulsion, the active ingredient is mixed with a portion of the base after emulsification and then added to the remainder of the formulation.
  • the dosage of the active ingredient can depend upon various factors, such as the effective ⁇ ness and duration of action of the active ingredient, the severity of the disease to be treated and of its symptoms, the mode of administration, the species of warm-blooded animal, and the sex, age, weight and/or individual condition of the warm-blooded animal.
  • the, for example oral, daily dose for a warm-blooded animal weighing approximately 75 kg is estimated to be from approximately 1 mg to approximately 1000 mg, especially from approximately 5 mg to approximately 200 mg. It can be administered, for example, as a single dose or in several part doses of, for example, from 10 to 100 mg.
  • the following Examples are intended to illustrate the invention described hereinbefore, but without limiting the invention thereto.
  • M.p.: melting point
  • CDCI 3 deuterochloroform
  • DMSO-d 6 hexadeuterodimethyl sulfoxide
  • CD 3 OD deuteromethanol
  • C 2 D 2 CI 4 deuterotetrachloroethane
  • Example 1 5-lsopropyl-7-phenyl-5H-pyrrolof3.2-d]pyrimidin-4-ylamine 0.252 g of 4-chloro-5-isopropyl-7-phenyl-5H-pyrrolo[3,2-d]pyrimidine is stirred in a bomb tube with 15 ml of ammonia at 130°C for 24 hours. The ammonia is evaporated off and the residue is purified by flash chromatography (methylene chloride/methanol 10:0.4). M.p.: 96-98°C.
  • Example 3 5-(2-Methoxy-ethoxymethyl)-7-phenyl-5H-Pyrrolof3.2-d]pyrimidin-4-ylamine M.p.: 161 -162°C.
  • Example 4 5-f2-(2-Methoxy-ethoxy)-ethyl1-7-phenyl-5H-pyrrolof3.2-dlpyrimidin-4-ylamine M.p.: 150-151 °C. 'H-NMR (CDCI 3 , ppm): 8.5 (s, 1H), 8.05 (m, 2H), 7.45 (m, 3H), 7.2 (s, 1H), 5.8 (s, 2H), 4.55 (t, 2H), 3.95 (t, 2H), 3.62 (m, 2H), 3.48 (m, 2H), 3.3 (s, 3H).
  • f(3-Nitrilo-2-phenyl-propenyl)-phenyl-aminol-acetic acid ethyl ester 4.48 g of phenyl- ammo-acetic acid ethyl ester, 3.60 g of ⁇ -formyl-phenylacetonitrile and 0.100 g of toluenesulfonic acid are dissolved in 200 ml of toluene and stirred under reflux using a water separator for 1.5 hours. The reaction mixture is cooled to room temperature and concentrated by evaporation using a rotary evaporator, and the residue is dried under a high vacuum.
  • Phenylammo-acetic acid ethyl ester (CAS Re ⁇ .No.: 2216-92-4] ⁇ s prepared according to W.K. Anderson et al., J. Med. Chem. 1989, 32, page 1 19.
  • Example 7 The following Examples are prepared analogously to Example 7 using the variously substituted phenylaminoacetic acid ethyl ester or benzylaminoacetic acid ethyl ester and the appropriate ⁇ -formyl-acetonitrile derivative as starting materials.
  • Example 12 3-(4-Amino-7-phenyl-pyrrolof3.2-d1pyrimidin-5-yl)-phenol
  • Example 13 7-[4-(2-Benzyloxy-ethoxy)-phenyl1-5-phenyl-5H-pyrrolo[3.2-d1pyrimidin-4- ylamine
  • Example 14 2-f4-(4-Amino-5-phenyl-5H-Pyrrolor3.2-dlpyrimidin-7-y ⁇ -phenoxyl-ethanol ⁇ -NMR (DMSO-d 6 , ppm): 8.32 (s, 1H), 8.15 (m, 3H), 7.60 (m, 6H), 7.0 (d, 2H), 6.8 (s, 2H), 4.89 (t, 1 H), 4.0 (t, 2H), 3.72 (m, 2H). M.p.: 190-192°C.
  • the oily residue is pre-purified by flash chromatography (hexane/ethyl acetate 10:1). Unreacted N-(3-methoxy- phenyl)glycine ethyl ester is then substantially removed from the resulting crude product by bulb tube distillation (170-180°C/0.06 mbar). The crude title compound is obtained in the form of a coloured oil.
  • Example 17 7-Cvclohexyl-5-(3-methoxy-phenvO-5H-pyrrolof3.2-d]pyrimidin-4-ylamine
  • a solution of 0.22 g of 7-cyclohex-1-enyl-5-(3-methoxyphenyl)-5H-pyrrolo[3,2-d]pyrimidin-4- ylamine in 10 ml of methanol is hydrogenated in the presence of 0.025 g of Pd/C (10 %) at room temperature under normal pressure for 3 days.
  • the reaction mixture is filtered over Celite 545 and the crude product obtained after concentration of the filtrate is purified by flash chromatography (methylene chloride/methanol 97:3).
  • 0.27 ml of boron tribromide is added in portions in the course of 15 minutes at 0-5°C to a solution of 0.30 g of 7-cyclohex-1-enyl-5-(3-methoxyphenyl)-5H-pyrrolo[3,2-d]pyr ⁇ m ⁇ din-4- ylamine in 10 ml of methylene chloride and, after 2.5 hours, a further 0.27 ml of boron tribromide is added. Stirring is then carried out for a further hour, 1.5 ml of water are then added dropwise and the mixture is stirred at 0-5°C for 30 minutes.
  • the crude product obtained after customary working-up is purified by flash chromatography (methylene chlonde/methanol 95:5).
  • Example 19 5-Phenyl-7-(pyr ⁇ d ⁇ n-3-yl)-5H-pyrrolo[3.2-dlpyr ⁇ mid ⁇ n-4-ylam ⁇ ne
  • a solution of 0.25 g of 5-phenyl-7-(pyridin-3-yl)-5/-/-pyrrolo[3,2-d]pyr ⁇ m ⁇ d ⁇ n-4-ol in 2 ml of phosphoryl chloride is stirred at 120°C for 2 hours.
  • the reaction mixture is concentrated using a rotary evaporator and the residue is dried under a high vacuum.
  • the crude product so obtained is stirred in 15 ml of liquid ammonia at 130°C in an autoclave for 24 hours.
  • Example 21 3-(4-Am ⁇ no-7-r4-(2- ⁇ m ⁇ dazol-1-yl-ethoxy)-phenyll-pyrrolo[3.2-d]pyrimidin-5-yll- phenol
  • Example 22 7-f4-(2-lmidazol-1 -yl-ethoxy)-phenvn-5-(4-methoxy-phenvh-5H-Pyrrolof3.2-dl- pyrimidin-4-ylamine
  • Example 27 7-[4-(2-Amino-ethoxy)-Phenyl1-5-(4-methoxy-phenyl,-5/-/-pyrrolof3.2-dlpyr ⁇ m ⁇ - d ⁇ n-4-ylam ⁇ ne
  • Example 28 7-[4-(2-Am ⁇ no-ethoxy)-phenvn-5-(3-methoxy-phenyl)-5/-/-pyrrolof3.2-dlpy ⁇ m ⁇ - d ⁇ n-4-ylam ⁇ ne
  • Example 30 4-(4-Am ⁇ no-7-(4-[2-(2-hvdroxy-ethylam ⁇ no)-ethoxyl-phenyl)-pyrrolo[3.2-d]- pyr ⁇ m ⁇ d ⁇ n-5-yl-phenol
  • Examole 32 2-2- ⁇ 4-[4-Amino-5-(4-methoxy-phenyl)-5H-pyrrolor3.2-d]pyrimidin-7-yl]-phen- oxy ⁇ -ethylamino)-ethanol
  • Example 46 7-(3.4-Difluoro-cvclopentyl-5-phenyl-5H-pyrrolor3.2-d1pyrimidin-4-ylamine
  • Example 47 3-(4-Amino-5-phenyl-5H-pyrrolo[3.2-d]pyrimidin-7-yl)-cvclohexanol
  • Example 56 1 -f3-(4-Amino-5-phenyl-5/-/-pyrrolor3.2-dlPyrimidin-7-yl)-pyrrolidin-1 -yll- ethanone
  • Example 61 7-(1-Methyl-piperidin-4-v ⁇ -5-phenyl-5/-/-pyrrolof3,2-d1pyrimidin-4-ylamine
  • Example 62 1-f4-(4-Amino-5-phenyl-5H-pyrrolor3.2-dlpyrimidin-7-yl)-piperidin-1-vn- ethanone
  • Example 66 7-(1.1-Dioxo-tetrahvdro-thiophen-3-yl)-5-phenyl-5H-pyrrolo[3.2-dlpyrimidin-4-yl- amine
  • Example 67 7-Cvclopent-1 -enyl-5-(3-fluorophenyl)-5H-pyrrolo[3.2-d]pyrimidin-4-ylamine
  • a solution of 0.07 g of sodium in 10 ml of absolute ethanol is added dropwise to a suspension of 0.81 g of N-[2-cyano-4-cyclopent-1-enyl-1-(3-fluorophenyl)-7H-pyrrolo-3-yl]- formamidme in 3 ml of ethanol and the mixture is stirred overnight at room temperature. It is then heated to reflux over 2 hours and, after cooling, the solvent is removed and the residue is partitioned between water and methylene chloride.
  • Sodium 2-cvclopent-1 -enyl-3-oxido-acrylonitrile 25.2 g of sodium hydride (80 % dispersion in oil) are washed twice with absolute tetrahydrofuran and then suspended in 1000 ml of tetrahydrofuran. 57.1 g of (1-cyclopent-1-enyl)acetonitrile are added and the mixture is heated to 50°C. After 15 minutes, 204 ml of ethyl formate are added with stirring, vigorous gas evolution commencing after a short time. After stirring at 50°C for 1 hour, the coloured suspension is filtered and the precipitate is washed with tetrahydrofuran and dried under a high vacuum.
  • a viscous slurry is obtained, to which 50 ml of toluene are added followed by heating to 80°C. 19.7 g of (3- fluorophenyl)-acetonitrile are then added and the reaction mixture is heated under reflux using a water separator. After 1 hour, the mixture is allowed to cool, the insoluble residue is removed by filtration and the filtrate is concentrated. The oily residue is pre-purified by flash chromatography on silica gel (hexane/ethyl acetate/toluene 6:1 :7).
  • the crude product so obtained is taken up in 40 ml of a saturated solution of ammonia in methanol, diluted with 30 ml of methylene chloride and stirred at room temperature for several days until the reaction comes to a standstill (monitoring by TLC).
  • the reaction mixture is concentrated and the residue is purified by means of flash chromatography on silica gel (methylene chloride/methanol 98:2).
  • the title compound is obtained in the form of a solid.
  • Example 68 7-Cvclopent-1 -enyl-5-(3-methoxyphenyl)-5H-pyrrolo[3.2-d)pyrimidin-4-ylamine
  • the title compound is obtained from 2.96 g of 7-cyclopent-1-enyl-5-(3-methoxyphenyl)-5/-/- pyrrolo[3,2-d]pyrimidin-4-ol in a manner analogous to that described in Examples 1 ) and 1e), after recrystallisation from hexane/ethyl acetate.
  • reaction mixture is then stirred for a further 30 minutes and, after cooling, the precipitate is removed by filtration and the filtrate is washed with saturated sodium hydrogen carbonate solution.
  • the aqueous phase is back-extracted with toluene and the combined organic phases are dried over magnesium suifate and concentrated.
  • the majority of the unreacted N-(3-methoxyphenyl)giycine ethyl ester is removed from the resulting crude product by solid distillation under a high vacuum (0.15 mbar; bath temperature 160-180°C).
  • Example 69 7-Cvclopent-1-enyl-5-(4-methoxyphenv ⁇ -5H-pyrrolor3.2-dlpyrimidin-4-ylamine
  • the title compound is obtained in the form of a solid from 5.1 g of 7-cyclopent-1 -enyl-5-(4- methoxyphenyl)-5H-pyrrolo[3,2-d]pyrimidin-4-ol in a manner analogous to that described in Examples 1 ) and 1e), after recrystallisation from methylene chloride/hexane.
  • FAB-MS 307 [M+H] + .
  • Example 70 7-Cyclopent-1 -enyl-5-(3-hvdroxyphenv ⁇ -5H-pyrrolof3.2-d1pyr ⁇ m ⁇ d ⁇ n-4-ylam ⁇ ne 0 47 ml of boron tribromide is added dropwise at 0-5°C, with stirring, to a suspension of 0.5 g of 7-cyclopent-1-enyl-5-(3-methoxyphenyl)-5H-pyrrolo[3,2-d]pyr ⁇ m ⁇ d ⁇ n-4-ylam ⁇ ne in 10 ml of methylene chloride. After 45 minutes, a further 0.16 ml of boron tribromide is added and stirring is subsequently carried out for 30 minutes.
  • Example 71 7-Cvclopent-1-enyl-5-(4-hvdroxyphenyl)-5H-pyrrolo[3.2-dlpyr ⁇ mid ⁇ n-4-ylam ⁇ ne
  • the title compound is obtained in the form of a solid from 1.0 g of 7-cyclopent-1-enyl-5-(4- methoxyphenyl)-5H-pyrrolo[3,2-d]pyr ⁇ m ⁇ d ⁇ n-4-ylam ⁇ ne in a manner analogous to that described in Example 70).
  • Example 72 7-Cvclopentyl-5-(4-hvdroxyphenv ⁇ -5H-pyrrolor3.2-d]pyrimidin-4-ylamine
  • a solution of 0.22 g of 7-cyclopent-1-enyl-5-(4-hydroxyphenyl)-5H-pyrrolo[3,2-d]pyrimidin-4- ylamine in 20 ml of methanol is hydrogenated in the presence of 0.06 g of 10 % palladium- on-carbon for 24 hours at room temperature.
  • the crude product obtained after customary working-up is purified by flash chromatography on silica gel (methylene chloride/methanol 95:5).
  • the title compound is obtained in the form of a solid.
  • Example 73 7-Cvclopentyl-5-(3-hvdroxyphenv ⁇ -5H-pyrrolof3,2-d]pyrimidin-4-ylamine
  • the title compound is obtained in the form of a solid from 0.30 g of 7-cyclopent-1 -enyl-5-(3- hydroxyphenyl)-5H-pyrrolo[3,2-d]pyrimidin-4-ylamine in a manner analogous to that described in Example 72).
  • FAB-MS 295 [M+H]+.
  • Example 74 7-Cvclopentyl-5-(3-fluorophenyl)-5H-pyrrolo[3.2-dlPyrimidin-4-ylamine Obtained in the form of a solid from 0.22 g of 7-cyclopent-1-enyl-5-(3-fluorophenyl)-5H- pyrrolo[3,2-d]pyrimidin-4-ylamine in a manner analogous to that described in Example 30), after recrystallisation from methylene chloride/hexane.
  • FAB-MS 297 [M+H]+.
  • Example 75 7-lsopropyl-5-(3-methoxyphenyl)-5H-pyrrolof3.2-dlpyrimidin-4-ylamine Reaction of 2.0 g of 7-isopropyl-5-(3-methoxyphenyl)-5H-pyrrolo[3,2-d]pyrimidin-4-ol in 10 ml of phosphorus oxychloride at 120°C for 2 hours, working-up and further reaction in methanolic ammonia under pressure at 130°C in a manner analogous to that described in Example 19) yield the title compound in the form of a white solid after recrystallisation from 1 :3 hexane/ethyl acetate.
  • f2-Cvano-3-methyl-but-1-enylH3-methoxyphenyl)aminol-acetic acid ethyl ester A mixture of 10.0 g of isovaleronitrile and ethanolic sodium ethanolate solution (2.76 g of sodium in 33 ml of absolute ethanol) in 36 ml of toluene is stirred at 50°C and 40-50 bar pressure under a carbon monoxide atmosphere for 10 hours analogously to a known process (DE 2753322 A1 ). After concentration of the reaction mixture by evaporation and drying under a high vacuum, the crude sodium 2-isopropyl-3-oxido-acrylonitrile is obtained in the form of a beige solid.
  • the crude product is dissolved in about 45 ml of water and adjusted to pH 6 by the addition of 1N hydrochloric acid. Extraction is then carried out with toluene, and the combined organic phases are washed with saturated sodium chloride solution, dried over magnesium suifate and filtered. The filtrate is added to a solution, heated to 50°C, of 8.95 g of N-(3-methoxyphenyl)glycine ethyl ester in 50 ml of toluene, a catalytic amount of p-toluenesulfonic acid is added to the reaction mixture and heating is then carried out under reflux for 1 hour.
  • N-(3-methoxyphenyl)- glycine ethyl ester contained in the crude product are substantially removed by bulb tube distillation at 220°C/5.5 mbar. Further purification by means of flash chromatography on silica gel (hexane/ethyl acetate/toluene 5:1 :5) yields the title compound in the form of an oil.
  • Example 76 5-(3-Hvdroxyphenyl)-7-isopropyl-5r-/-pyrrolo[3.2-dlpyrimidin-4-ylamine 0.5 ml of boron tribromide is added in portions over 20 minutes at 0°C to a solution of 0.5 g of 7-isopropyl-5-(3-methoxyphenyl)-5H-pyrrolo[3,2-d]pyrimidin-4-ylamine in 30 ml of methylene chloride. The resulting suspension is further stirred at 0-5°C for 2 hours and then, with ice cooling, 10 ml of water are added. The mixture is allowed to warm to room temperature and is subsequently stirred for a further hour.
  • Example 77 7-Cvclopent-3-enyl-5-(3-methoxyphenyl)-5H-pyrrolo[3.2-d]pyrimidin-4-ylamine 2.1 g of 7-cyclopent-3-enyl-5-(3-methoxyphenyl)-5H-pyrrolo[3,2-d]pyrimidin-4-ol and 10 ml of phosphorus oxychloride are stirred at 120°C for 2 hours. After concentration of the reaction mixture and drying under a high vacuum, 4-chloro-7-cyclopent-3-enyl-5-(3- methoxyphenyl)-5H-pyrrolo[3,2-d]pyrimidine is obtained as the crude product.
  • a solution of 10.7 g of the crude product so obtained in 30 ml of water is adjusted to pH 6 with 1N hydrochloric acid and extracted twice with 100 ml of toluene.
  • the combined organic phases are dried over magnesium suifate, filtered and then added, at 50°C, to a solution of 7.15 g of N-(3-methoxyphenyl)glycine ethyl ester in 50 ml of toluene.
  • the reaction mixture is stirred under reflux for 60 minutes and, after cooling, the organic phase is washed with 5 % sodium hydrogen carbonate solution and saturated sodium chloride solution, dried over magnesium suifate and concentrated.
  • the residue obtained after bulb tube distillation (210°C/2 mbar) is purified by means of flash chromatography on silica gel (hexane/ethyl acetate/toluene
  • Potassium 2-cvclopent-3-enyl-3-oxido-acrylonitrile 5.0 g of cyclopent-3-enyl-acetonitrile are added, with stirring, to a suspension of potassium hydride (obtained from 28.1 g of potassium hydride 20 % dispersion in oil by washing twice with n-hexane) in 1 :1 diethyl ether/hexane, gas evolution taking place. The brown suspension is stirred at room temperature for 1 hour and then 30.4 ml of ethyl formate are added dropwise over 15 minutes.
  • the initially viscous suspension is stirred at room temperature for 12 hours and then filtered over a glass frit, and the coloured precipitate is washed with a small amount of hexane.
  • the filter residue is dried at 50°C under a high vacuum for 3 hours and is then reacted, without further purification, with N-(3-methoxyphenyl)glyc ⁇ ne ethyl ester as described in Example 44d).
  • the oily residue is purified by means of flash chromatography on silica gel (hexane/ethyl acetate/- toluene, gradient 3:1 :4 to 1:1:4). Reaction of 5.72 g of the resulting product in 150 ml of a saturated ammonia solution in methanol at 40-45°C for 48 hours and subsequent purifi ⁇ cation by means of flash chromatography on silica gel (methylene chlonde/methanol 98:2) yield the title compound in the form of a solid.
  • Example 78 7-Cvclopent-3-enyl-5-(3-hvdro ⁇ yphenyl)-5H-pyrrolo[3.2-d1pyrimid ⁇ n-4-ylam ⁇ ne 0.47 ml of boron tribromide is added dropwise at 0-5 c C over 20 minutes, with stirring, to a solution of 0.5 g of 7-cyclopent-3-enyl-5-(3-methoxyphenyl)-5H-pyrrolo[3,2-d]pyr ⁇ mid ⁇ n-4- ylamme in 30 ml of methylene chloride.
  • the title compound contains 50% 7-[4-(2-chloro-ethoxy)- phenyl]-5-(3-methoxy-phenyl)-5H-pyrrolo[2,3-d]pyr ⁇ m ⁇ d ⁇ n-4-ylam ⁇ ne.
  • Example 83 3-(4-Am ⁇ no-7-r4-(2-hvdroxy-ethoxy)-phenyl]-pyrrolo[3,2-d1pyr ⁇ m ⁇ d ⁇ n-5-ylr-phenol
  • the compound is prepared analogously to Example 81.
  • Example 85 7-(4-[2-(2-Methoxy-ethylam ⁇ no)-ethoxyl-phenyll-5-(3-methoxy-phenyl)-5r-y-pyr- rolof3.2-d1pynm ⁇ d ⁇ n-4-ylam ⁇ ne
  • the compound is prepared analogously to Example 81. M.p.:152 - 154°C.
  • Example 87 7-(4-[2-(4-Fluoro-benzylam ⁇ no)-ethoxy1-phenyl)-5-(3-methoxy-phenyl)-5/-/-pyr- rolo[3,2-dlpyr ⁇ m ⁇ d ⁇ n-4-ylam ⁇ ne
  • the compound is prepared analogously to Example 81. M.p/ 137 -140°C.
  • Example 91 1-(2-(4-[4-Amino-5-(3-methoxy-phenyl)-ffH-pyrrolor3.2-d]pyrimidin-7-yl]-phen- oxy>-ethyl)-pyrrolidin-3-ol
  • the compound is prepared analogously to Example 81. M.p.: 132 -135°C.
  • the compound is prepared analogously to Example 81. M.p.: 212 - 216°C.
  • the compound is prepared analogously to Example 81. M.p.: 166 - 168°C.
  • the compound is prepared analogously to Example 81. M.p.: 152 -154°C.
  • Example 96 2-2-(4-f4-Am ⁇ no-5-(3-fluoro-phenyl)-5H-pyrrolo[3,2-d]pyr ⁇ mid ⁇ n-7-v ⁇ -phenoxy>- ethylaminoVethanol
  • the compound is prepared analogously to Example 81 . M.p.: 148-151 °C.
  • the compound is prepared analogously to Example 81. M.p.: 164 - 168°C.
  • Example 99 4-f4-Amino-5-(3-hvdroxy-phenyl)-5/-/-pyrrolo[3.2-d1pyrimidin-7-yl]-phenol
  • the compound is prepared analogously to Example 81.
  • Example 100 3-r4-Amino-7-(4-(2-f(2-hvdroxy-ethvO-methyl-amino1-ethoxy)-phenyl)-pyrrolo- f3.2-dlpyrimidin-5-yl1-phenol
  • the compound is prepared analogously to Example 81. M.p.: 212 - 215°C.
  • the compound is prepared analogously to Example 81. M.p.: 212 - 216°C.
  • Example 102 1 -(2-(4-[4-Amino-5-(3-hvdroxy-phenyl)-5/-/-pyrrolo[3.2-dlpyrimidin-7-v ⁇ -phen- oxVf-ethyl)-pyrrolid ⁇ n-3-ol
  • the compound is prepared analogously to Example 81. M.p.: 218 - 222°C.
  • Example 104 2-(2-(3-[4-Amino-5-(3-methoxy-phenyl)-5/-/-pyrrolof3.2-d1pyrimidin-7-yl]-phen- oxy
  • the compound is prepared analogously to Example 81. M.p.: 166 - 168 C C.
  • Example 106 7-f3-(2-lmidazol-1 -yl-ethoxy)-phenv ⁇ -5-(3-methoxy-phenyl)-5/-/-pyrrolo[3.2-d]- pyrimidin-4-ylam ⁇ ne
  • Example 107 5-(3-Methoxy-phenyl)-7-r3-(2-piperaz ⁇ n-1 -yl-ethoxy)-phenv ⁇ -5H-pyrrolor3.2-dl- pyr ⁇ m ⁇ d ⁇ n-4-ylam ⁇ ne hydrochloride
  • Example A Tablets, each comprising 50 mg of active ingredient:
  • composition (10 000 tablets) active ingredient 500.0 g lactose 500.0 g potato starch 352.0 g gelatin 8.0 g talc 60.0 g magnesium stearate 10 O g silicon dioxide (highly dispersed) 20.0 g ethanol q s
  • the active ingredient is mixed with the lactose and 292 g of the potato starch and the mixture is moistened with an ethanohc solution of the gelatin and granulated through a sieve. After drying, the remaining potato starch, the magnesium stearate, the talc and the silicon dioxide are mixed in and the mixture is compressed to form tablets each weighing 145 mg and comprising 50 mg of active ingredient, which may, if desired, be provided with dividing notches for finer adaptation of the dose
  • composition 1000 film-coated tablets
  • active ingredient 100.0 g lactose 100 O g corn starch 70.0 g talc 8.5 g calcium stearate 1.5 g hydroxypropyimethylcellulose 2.36 g shellac 0.64 g water q.s. dichloromethane q.s.
  • the active ingredient, the lactose and 40 g of the corn starch are mixed, and the mixture is moistened with a paste, prepared from 15 g of the corn starch and water (with heating), and granulated.
  • the granules are dried, and the remaining corn starch, the talc and the calcium stearate are mixed with the granules.
  • the mixture is compressed to form tablets (each weighing 280 mg), which are then coated with a solution of the hydroxypropyimethylcellul ⁇ ose and the shellac in dichloromethane (final weight of each film-coated tablet: 283 mg).

Abstract

Pyrrolo[3,2-d]pyrimidines of formula (I) wherein R1, R2 and R3, are as defined in the description, are described. The compounds have valuable pharmaceutical properties and are effective especially as tyrosine protein kinase inhibitors. They can be used in warm-blooded animals in the treatment of bone diseases and other diseases that are favourably influenced by the inhibition of tyrosine protein kinase.

Description

Substituted 7-amino-pyrrolof3,2-d]pyrimidines and the use thereof
The invention relates to compounds of formula I
Figure imgf000003_0001
wherein
R, is unsubstituted or substituted lower alkyl or hetero-substituted lower alkyl, unsubstituted or substituted cyclo-lower hydrocarbyl or cyclo-lower hydrocarbyl-lower alkyl, unsubstituted or substituted aryl or aryl-lower alkyl, or unsubstituted or substituted hetero-lower cyclyl or hetero-lower cyclyl-lower alkyl;
R2 is hydrogen, lower alkyl or halogen;
R3 is unsubstituted or substituted lower alkyl or hetero-substituted lower alkyl, unsubstituted or substituted cyclo-lower hydrocarbyl or cyclo-lower hydrocarbyl-lower alkyl, unsubstituted or substituted aryl or aryl-lower alkyl, or unsubstituted or substituted hetero-lower cyclyl or hetero-lower cyclyl-lower alkyl; and salts thereof.
Within the context of the present Application, the general terms used hereinbefore and hereinafter preferably have the following definitions:
The term "lower " denotes a radical having up to and including 7, and especially up to and including 6, carbon atoms.
Lower alkyl is, for example, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, ferf-butyl, n-pentyl, neopentyl, n-hexyl or n-heptyl, preferably ethyl or methyl. Alkyl is straight-chain or branched. On its own, for example lower alkyl, or as a constituent of other groups, for example lower alkoxy, lower alkylcarbonyl, lower alkoxycarbonyl, lower alkylaminocarbonyl, di-lower alkylaminocarbonyl, it may be unsubstituted or mono- or poly- substituted, for example by halogen, hydroxy, lower alkoxy, tπfluoromethyl or by imidazolyl, and is preferably unsubstituted or substituted by halogen, hydroxy, lower alkoxy, ammo, N- lower alkylamino or by N,N-dι-lower alkylamino. In hetero-substituted alkyl, the carbon chain is interrupted by one or more, identical or different hetero atoms, for example by oxygen, sulfur or S(O2), especially oxygen
Cyclo-lower hydrocarbyl denotes saturated or mono- or poly-unsaturated carbocyciic rings, preferably having from 3 up to and including 7 ring members, especially 5 or 6 ring mem¬ bers, for example cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclopentadienyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl. Cyclo-lower hydrocarbyl-lower alkyl is, for example, cyclopentylmethyl, cyclopentenylmethyl, cyclopentylethyl, cyclopentylpropyl, cyclohexylmethyl, cyclohexenylethyl Cyclo-lower hydrocarbyl and cyclo-lower hydrocarbyl- lower alkyl may be unsubstituted or mono- or poly-substituted, for example by hydroxy, lower alkyl, lower alkoxy, hydroxy-lower alkyl, hydroxy-lower alkoxy, halo-lower alkyl, halo- lower alkoxy, lower alkoxy-lower alkoxy, ammo-lower alkyl, ammo-lower alkoxy, lower alkylamino-lower alkyl, lower alkylamino-lower alkoxy, N,N-dι-lower alkylamino-lower alkyl, N,N-dι-lower alkylamino-lower alkoxy, N,N -hydroxy-lower alkyl-lower alkylamino-lower alkyl or by N,N-hydroxy-lower alkyl-lower alkylamino-lower alkoxy, and preferably is unsubsti¬ tuted
Aryl is, for example, phenyl or naphthyl, each of which is unsubstituted or substituted, for example as indicated hereinafter for phenyl Aryl is preferably phenyl unsubstituted or substituted by one or more, for example from one to three, especially one or two, substi¬ tuents from the group consisting of hydroxy, lower alkyl, halo-lower alkyl, (hydroxy or lower alkanoyloxy)-lower alkyl, lower alkoxy-lower alkyl, (hydroxy, lower alkoxy or lower alkanoyl- oxy)-lower alkoxy-lower alkyl, (ammo, lower alkylamino, di-lower alkylamino or lower alkanoylamιno)-lower alkoxy-lower alkyl, (ammo or lower aikanoylamιno)-lower alkyl, lower alkylamino-lower alkyl, di-lower alkylamino-lower alkyl; azacycloalkyl-lower alkyl, for example (pipendinyl, piperazinyl, morpholinyl or pyrrolιdιnyl)-lower alkyl; azaheteroaryl-lower alkyl, for example (imidazolyl, tπazolyl, pyridyl, pyπmidinyl or pyrrolyl)-lower alkyl, (hydroxy, lower alkoxy or lower alkanoyloxy)-lower alkylamino-lower alkyl, (ammo, lower alkylamino, dι-lower alkylamino or lower alkanoylamιno)-lower alkylamino-lower alkyl; azacycloalkyl- lower alkylamino-lower alkyl, for example (pipeπdmyl, piperazinyl, morpholinyl or pyrrol- ιdιnyl)-lower alkyiammo-lower alkyl; azaheteroaryl-lower alkylamino-lower alkyl, for example (imidazolyl, tπazolyl, pyπdyl, pyπmidinyl or pyrrolyl)-lower alkylamino-lower alkyl; mercapto- lower alkyl, lower alkyl-(thιo, sulfinyl or sulfonyl)-lower alkyl, (hydroxy, lower alkoxy or lower alkanoyloxy)-lower alkyl-(thio, sulfinyl or sulfonyl)-lower alkyl, (ammo, lower alkylamino, di- lower alkylamino or lower alkanoylamιno)-lower alkyl-(thιo, sulfinyl or sulfonyl)-lower alkyl, carboxy-lower alkyl, lower alkoxycarbonyl-lower alkyl, aminocarbonyl-lower alkyl, N-lower alkylammocarbonyl-lower alkyl, N,N-dι-lower alkylaminocarbonyl-lower alkyl, hydroxy, lower alkoxy, lower alkanoyloxy, Cι-C3alkylenedιoxy, phenyl-lower alkoxy, (hydroxy, lower alkoxy or lower alkanoyloxy)-lower alkoxy, (amino or lower alkanoylamιno)-lower alkoxy, lower alkylammo-lower alkoxy, di-lower alkylamino-lower alkoxy; azacycloalkyl-lower alkoxy, for example (piperidmyl, piperazinyl, morpholinyl or pyrrolιdιnyl)-lower alkoxy; azaheteroaryl- lower alkoxy, for example (imidazolyl, tnazolyl, pyndyl, pyπmidinyl or pyrrolyl)-lower alkoxy, (hydroxy, lower alkoxy or lower alkanoyloxy)-lower alkylamino-lower alkoxy, (ammo, lower alkylamino, di-lower alkylamino or lower alkanoylamιno)-lower alkyiammo-lower alkoxy; aza¬ cycloalkyl-lower alkylamino-lower alkoxy, for example (piperidmyl, piperazinyl, morpholinyl or pyrrolιdιnyl)-lower alkylamino-lower alkoxy; azaheteroaryl-lower alkylamino-lower alkoxy, for example (imidazolyl, tnazolyl, pyndyl, pynmidinyl or pyrrolyl)-lower alkylamino-lower alkoxy, (hydroxy, lower alkoxy or lower alkanoyloxy)-lower alkoxy-lower alkoxy, (ammo, lower alkyl¬ amino, di-lower alkylamino or lower alkanoylamιno)-lower alkoxy-lower alkoxy, (hydroxy, lower alkoxy or lower alkanoyloxy)-lower alkyl-(thιo, sulfinyl or sulfonyl)-lower alkoxy, (ammo, lower alkylamino, di-lower alkylamino or lower alkanoylamιno)-lower alkyl-(thιo, sulfinyl or sulfonyl)-lower alkoxy, hydroxysulfonyl-lower alkoxy, carboxy-lower alkoxy, lower alkoxy¬ carbonyl-lower alkoxy, aminocarbonyl-lower alkoxy, N-lower alkylammocarbonyl-lower alkoxy, N,N-dι-lower alkylammocarbonyl-lower alkoxy, ammo, lower alkylamino, di-lower alkylamino, lower alkanoylamino; azacycloalkyl, for example piperidmyl, piperazinyl, morpholinyl or pyrrolidinyl; azaheteroaryl, for example imidazolyl, tnazolyl, pyndyl, pynmi¬ dinyl or pyrrolyl, mercapto, lower alkyl-(thιo, sulfinyl or sulfonyl), (hydroxy, lower alkoxy or lower alkanoyloxy)-lower alkyl-(thιo, sulfinyl or sulfonyl), (ammo, lower alkylamino, di-lower alkylamino or lower alkanoylamιno)-lower alkyl-(thιo, sulfinyl or sulfonyl), (hydroxy, lower alkoxy or lower alkanoyioxy)-lower alkoxy-lower alkyl-(thιo, sulfinyl or sulfonyl), (ammo, lower alkylamino, di-lower alkylamino or lower alkanoylamιno)-lower alkoxy-lower alkyl-(thιo, sulfinyl or sulfonyl), (hydroxy, lower alkoxy or lower alkanoyloxy)-lower alkylamino-lower alkyl-(thio, sulfinyl or sulfonyl), (amino, lower alkylamino, di-lower alkylamino or lower alkanoylamino)-lower alkylamino-lower alkyl-(thio, sulfinyl or sulfonyl), carboxy-lower alkyl- thio, lower alkoxycarbonyl-lower alkylthio, aminocarbonyl-lower alkylthio, N-lower alkyl- aminocarbonyl-lower alkylthio, N,N-di-lower alkylaminocarbonyl-lower alkylthio, halogen, carboxy, lower alkoxycarbonyl, aminocarbonyl, N-lower alkylaminocarbonyl, N-[(hydroxy, lower alkoxy or lower alkanoyloxy)-lower alkyl]-aminocarbonyl, N-[(amino, lower alkylamino, di-lower alkylamino or lower alkanoylamino)-lower alkyl]-aminocarbonyl; [azacycloalkyl-lower alkyl]-aminocarbonyl, for example N-[(piperidinyl, piperazinyl, morpholinyl or pyrrolidinyl)- lower alkyl]-aminocarbonyl; [azaheteroaryl-lower alkyl]-aminocarbonyl, for example N-[(imid- azolyl, triazolyl, pyridyl, pyrimidinyl or pyrrolyl)-lower alkyl]-aminocarbonyl; N-(hydroxy- sulfonyl-lower alkyl)-aminocarbonyl, N,N-di-lower alkylaminocarbonyl, cyano, amidino, formamidino and guanidino and, for example, nitro, lower alkanoyl and benzoyl.
In substituents containing groups such as, for example, hydroxy-lower alkoxy, amino-lower alkoxy, hydroxy-lower alkylamino, amino-lower alkylamino, hydroxy-lower alkylthio or amino- lower alkylthio, the two hetero atoms are preferably separated from one another by at least two carbon atoms; in other words, the lower alkyl moiety is preferably so selected that there are at least two carbon atoms between the two hetero atoms.
Halogen is, for example, chlorine, bromine or fluorine, but may also be iodine.
Hetero-lower cyclyl is a saturated, mono- or poly-unsaturated or aromatic radical having from 3 up to and including 7 ring atoms, especially from 3 up to and including 6 ring atoms, wherein at least one of the ring atoms is a hetero atom, preferably nitrogen, oxygen or sulfur. Especially preferred are aza-lower cycloalkyl and aza-heteroaryl, that is to say hetero-lower cyclyl radicals that contain at least one nitrogen ring atom. When a hetero- cyclyl radical contains more than one ring hetero atom, the hetero atoms may be identical or different; one ring hetero atom is preferred. Hetero-lower cyclyl is, for example, piperidmyl, piperazinyl, morpholinyl, pyrrolidinyl, furanyl, tetrahydrofuran, thiophenyl, imidazolyl, oxazolyl, thiazolyl, triazolyl, pyranyl, thiopyranyl, pyridyl, pyrrolyl or tetrazolyl.
The compounds I and their salts may be in the form of one of the possible isomers, for example stereoisomers, diastereoisomers or tautomers, or in the form of a mixture thereof. There are obtainable as pure isomers, for example, pure enantiomers, pure diastereo- isomers or, where appropriate, pure tautomers. Accordingly, isomeric mixtures may be in the form of, for example, racemates or diastereoisomeric mixtures.
Salts of compounds of formula I are especially pharmaceutically acceptable salts, especially acid addition salts with suitable mineral acids, such as hydrohalic acids, sulfuric acid or phosphoric acid, for example hydrochlorides, hydrobromides, sulfates, hydrogen sulfates or phosphates, salts with suitable aliphatic or aromatic sulfonic acids or N-substituted sulfamic acids, for example methanesulfonates, benzenesulfonates, p-toluenesulfonates or N-cyclo- hexylsulfamates (cyclamates), or salts with strong organic carboxylic acids, such as lower alkanecarboxylic acids or saturated or unsaturated or hydroxylated aliphatic dicarboxylic acids, for example acetates, oxalates, malonates, maleates, fumarates, tartrates or citrates.
Also possible, where the compounds of formula I contain an acid group, are corresponding salts with bases, for example corresponding alkali metal or alkaline earth metal salts, for example sodium, potassium or magnesium salts, pharmaceutically acceptable transition metal salts, such as zinc or copper salts, or salts with ammonia or organic amines, such as cyclic amines, mono-, di- or tri-lower alkylamines, hydroxy-lower alkylamines, for example mono-, di- or tri-hydroxy-lower alkylamines, hydroxy-lower alkyl-lower alkyl-amines or poly- hydroxy-lower alkylamines. Cyclic amines are, for example, morpholine, thiomorpholine, piperidine or pyrrolidine. Suitable mono-lower alkylamines are, for example, ethyl- and tert- butyl-amine; suitable di-lower alkylamines are, for example, diethyl- and diisopropyl-amine and suitable tri-lower alkylamines are, for example, trimethyl- and triethyl-amine. Suitable hydroxy-lower alkylamines are, for example, mono-, di- and tri-ethanolamine; hydroxy-lower alkyl-lower alkyl-amines are, for example, N,N-dimethylamino- and N,N-diethylamino- ethanol. Compounds of formula I having an acid group, for example carboxy, and a basic group, for example amino, may also be in the form of, for example, internal salts, i.e. in zwitterionic form, or part of the molecule may be in the form of an internal salt and another part in the form of a normal salt. Pharmaceutically unacceptable salts are also included, since they can be used, for example, for the isolation and/or purification of free compounds I and the pharmaceutically acceptable salts thereof.
The compounds of formula I have valuable pharmacological properties. In particular, they inhibit the activity of tyrosine protein kinase pp60c src in concentrations of from approximately 0.001 to approximately 10 μM [test description: K. Farley et al., Anal. Biochem. 203 (1992) 151-157; purified enzyme - as described in N. B. Lydon er a/., Biochem. J. 287 (1992) 985-
Figure imgf000008_0001
It is known that both targeted modification of the c-src gene, leading to the elimination of c-src, and inhibition of the activity of tyrosine protein kinase pp60c src affect the bone absorption ability of osteoclasts [for elimination of c-src by gene manipulation: see, for example, P Soriano et al., Ce//64 (1991) 693-702; for inhibition of the activity of tyrosine protein kinase pp60c src: see, for example, B.F. Boyce et al., J Clin. Invest. 90 (1992) 1622- 1627, T. Yoneda er a/., J. Clin. Invest. 91 (1993) 2791 -2795]
Owing to their inhibitory activity against tyrosine protein kinase pp60c src, the compounds of formula I are therefore capable of inhibiting the bone absorption ability of osteoclasts. That can be demonstrated, for example, in the bone slice assay on bovine cortical bone platelets with rat osteoclasts in concentrations of from approx. 0.001 to approx 10 μM. [The "bone slice assay" is described, for example, in Biochem. Biophys. Res. Comm 188 (1992) 1097- 1103] In that assay, the compounds of formula I inhibit the formation of characteristic absorption holes in bone platelets in vitro
In vivo, the effectiveness of compounds of formula I can be demonstrated, for example, in the Hock model in the rat In that test, the compounds of formula I - when administered once a day per os in concentrations of from approx 1 to approx 100 mg/kg of body weight - for from 3 to 4 weeks completely or at least partially inhibit the bone loss produced as a result of ovanectomy in rats [the "Hock model" is described, for example, in Metab. Bone
Figure imgf000008_0002
The in vivo activity of compounds of formula I can also be demonstrated, for example, via calcium metabolism in intact rats. In that method, after i.v. injection of the test compound acute hypocalcaemia is induced within from 1 to 4 hours; it is demonstrated by determining the concentration of calcium in the blood plasma. The observation of acute hypocalcaemia can be interpreted as indirect evidence that the test compound inhibits bone absorption
The compounds of formula I are therefore very suitable for the treatment of diseases that are responsive to inhibition of the activity of tyrosine protein kinase pp60c src. Special mention may be made here of osteoporosis, and of other diseases in the course of which the absorption of bone by osteoclasts plays a role, such as tumour-induced hypercalcaemia or Paget's disease, or the treatment of bone metastases, and also inflammatory processes in joints and bones and degenerative processes in cartilage tissue. In addition, the com¬ pounds of formula I are useful in the treatment of benign or malignant tumours that are responsive to inhibition of tyrosine protein kinase pp60c"src, such as breast cancer (mammary carcinoma) or intestinal cancer (colon carcinoma). They are capable of effecting tumour regression and of preventing the formation of tumour metastases and the growth of micrometastases. The compounds of formula I are also useful in the treatment of cardio¬ vascular disorders, such as thrombosis.
The compounds of formula I also inhibit the activity of other non-receptor tyrosine protein kmases, such as (a) other members of the src family, for example Ick and fyn, (b) abl kinase and (c) ZAP70 kinase Furthermore, the compounds of formula I also inhibit the activity of receptor tyrosine protein kmases, such as (a) the EGF family, for example the EGF receptor, c-erbB2, c-erbB3 and c-erbB4, and (b) the PDGF family, for example the PDGF receptor, CSF-1 , Kit, VEGF and FGF. Owing to those actions, the compounds of formula I can also be used in immunomodulation and in the treatment of diseases of the immune system, for example in the case of inflammations or organ transplants They are also suitable for the treatment of (hyper)prolιferatιve diseases, such as psoriasis, tumours, carcinomas and leukaemias, and in fibrosis and restenosis. The compounds of formula I can also be used in the treatment of diseases of the central or the peripheral nervous system where signal transmission by at least one tyrosine protein kinase is involved.
The invention relates preferably to compounds of formula I wherein
R, is unsubstituted or substituted lower alkyl or hetero-substituted lower alkyl, unsubstituted or substituted cyclo-lower hydrocarbyl, unsubstituted or substituted aryl, unsubstituted or substituted aryl-lower alkyl, or unsubstituted or substituted hetero-lower cyclyl;
R2 is hydrogen, lower alkyl or halogen;
R3 is unsubstituted or substituted lower alkyl or hetero-substituted lower alkyl, unsubstituted or substituted cyclo-lower hydrocarbyl, unsubstituted or substituted aryl or aryl-lower alkyl, or unsubstituted or substituted hetero-lower cyclyl; and salts thereof The invention relates especially to compounds of formula I wherein
R, is unsubstituted or substituted lower alkyl or hetero-substituted lower alkyl, unsubstituted or substituted cyclo-lower hydrocarbyl, unsubstituted or substituted aryl, unsubstituted or substituted aryl-lower alkyl, or unsubstituted or substituted hetero-lower cyclyl;
R2 is hydrogen, lower alkyl or halogen;
R3 is unsubstituted or substituted lower alkyl or hetero-substituted lower alkyl, unsubstituted or substituted cyclo-lower hydrocarbyl, or unsubstituted or substituted aryl; and salts thereof.
The invention relates more especially to compounds of formula I wherein
R, is unsubstituted or substituted lower alkyl or hetero-substituted lower alkyl, unsubstituted or substituted cyclo-lower hydrocarbyl, unsubstituted or substituted aryl, unsubstituted or substituted aryl-lower alkyl, or unsubstituted or substituted hetero-lower cyclyl;
R2 is hydrogen;
R3 is unsubstituted or substituted lower alkyl or hetero-substituted lower alkyl, unsubstituted or substituted cyclo-lower hydrocarbyl, or unsubstituted or substituted aryl; and salts thereof.
The invention relates most especially to compounds of formula I wherein
Ri is unsubstituted or substituted lower alkyl or hetero-substituted lower alkyl, unsubstituted or substituted cyclo-lower hydrocarbyl, unsubstituted or substituted aryl, unsubstituted or substituted aryl-lower alkyl, or unsubstituted or substituted hetero-lower cyclyl;
R2 is hydrogen,
R3 is unsubstituted or substituted lower alkyl, unsubstituted or substituted cyclo-lower hydrocarbyl, or unsubstituted or substituted aryl; and pharmaceutically acceptable salts thereof.
The invention relates especially to the specific compounds described in the Examples and to salts thereof.
The compounds of formula I can be prepared in a manner known perse, for example by a) subjecting a compound of formula II
Figure imgf000011_0001
wherein
R,, R2 and R3 are as defined above, and R4 is lower alkyl, to a ring-closure reaction, with the formation of the pyrimidine ring, to give a compound of formula III
Figure imgf000011_0002
and replacing the oxygen by NH2 by means of an exchange reaction; or
b) subjecting a compound of formula IV
Figure imgf000011_0003
wherein
R1 t R2 and R3 are as defined above, to a ring-closure reaction with the formation of the pyrimidine ring; or
c) reacting a compound of formula V
Figure imgf000011_0004
wherein R2 and R3 are as defined above, with a compound of formula VI
Figure imgf000012_0001
wherein Ri is as defined above and X is a leaving group;
and, if desired, converting a compound of formula I into a different compound of formula I, and/or, if desired, converting a resulting salt into the free compound or into a different salt, and/or, if desired, converting a resulting free compound of formula I having salt-forming properties into a salt.
In the more detailed description of the processes which follows, unless indicated otherwise each of the symbols R1-R4 is as defined for formulae I to VI.
Process (a): The reaction according to process (a) corresponds to the cyclisation known per se of 3-amino-2-alkoxycarbonyl-pyrroles to 4-oxo-pyrrolo[3,2-d]pyrimidines. Suitable cyclisation reagents are the sequence 1. N,N-di-lower alkylformamide di-lower alkyl acetal and 2. ammonia. Preferred N,N-di-lower alkylformamide di-lower alkyl acetals are di-lower alkyl acetals of N,N-dimethylformamide, especially N,N-di-lower alkylformamide dineopentyl acetal. The reaction of compounds of formula II is preferably effected at elevated tempera¬ tures, for example at the reflux temperature of the reaction mixture. The reaction can be carried out with or without solvent, but preferably takes place in an aprotic solvent or solvent mixture. The resulting dialkylamino-methyleneaminoalkoxycarbonyl-pyrroles are then cyclised with ammonia to form a compound of formula III. The latter reaction is preferably effected in a polar solvent or solvent mixture in a bomb tube at ambient temperature or slightly elevated temperature whereby also one of the reactants may serve as solvent or component of a solvent mixture. The 4-oxo group can then be replaced in a manner known per se by halogen, especially chlorine, by reaction with a phosphorus oxytrihalide, prefer¬ ably phosphorus oxytrichloride. The reaction can be carried out with or without solvent, but preferably takes place without solvent at distinctly elevated temperature, for example at 100°C to 120°C. Subsequently, the 4-halide can be replaced by -NH2 by treating the reaction product with ammonia. That reaction is generally carried out at relatively high temperatures, for example at 130°C in an autoclave. The compounds of formula II are preferably prepared using one of the known methods of pyrrole synthesis. They are obtained, for example, by cyclising a compound of formula I la
x
Figure imgf000013_0001
preferably in the presence of a base, for example sodium ethanolate/ethanol.
Process (b). The reaction according to process (b) corresponds to the cyclisation known perse of 2-amιno-3-cyano-pyrroles to 4-amιno-pyrrolo[2,3-d]pyrιmιdιnes. Suitable cyclisa¬ tion reagents are, for example, (1) formamide or (2) 1. trialkyi orthoformate/2. ammonia. The cyclisation of compounds of formula IV with formamide is preferably carried out at elevated temperature, for example at 160°C, and advantageously with the addition of a small amount of dimethylformamide and formic acid. The reaction of compounds of formula IV with trialkyi orthoformate to give the corresponding alkoxy formimidates formed as intermediates norm¬ ally takes place at less elevated temperatures, for example at from 80 to 140°C The reac¬ tion of the latter with ammonia can then be carried out generally at relatively low tempera¬ tures, for example at ambient temperature. The cyclisation then takes place preferably with base catalysis, especially using sodium ethanolate/ethanol, once again at elevated tem¬ perature, for example at the reflux temperature of the reaction mixture.
Process (c): Suitable leaving groups are, for example, methanesulfonates or p-toluenesulf- onates of hydroxy compounds and halogen. The preparation of suitable pyrrolo[3,2-d]- pyπmidmes of formula V is known from the literature or can be carried out analogously to the methods described in the literature. The reaction of compounds of formula V with compounds of formula VI is carried out in a manner known per se. For example, a methane- sulfonate of formula VI is reacted with a pyrrolo[3,2-d]pyrιmιdιne of formula V in the presence of a base, for example potassium carbonate. The reaction is preferably carried out at elevated temperature, for example at from 50°C to the reflux temperature of the reaction mixture, especially at from 60 to 80°C, and advantageously in an inert solvent or solvent mixture. The reaction can be accelerated in an advantageous manner by the addition of a suitable crown ether. In a further process, the reaction takes place in a manner known per se under the conditions of phase transfer catalysis (E.V. Dehmlow and S. S. Dehmlow, Phase Transfer Catalysis, 3rd ed., VCH, Weinheim, 1993). The reactants of formulae V and VI are dissolved in a suitable inert solvent or solvent mixture, and the second phase is formed by a concentrated aqueous alkali metal hydroxide solution, for example 30 % sodium hydroxide solution. Advantageously, a phase transfer catalyst, for example a quaternary ammonium halide, such as tetrabutylammonium bromide, is added.
In a variant of process (c), a compound of formula III wherein Ri is hydrogen, or a corres¬ ponding compound obtainable by replacing the 4-oxo group by halogen, is used as starting material and reacted with a compound of formula VI, and then the 4-amino group is intro¬ duced as described in process (a).
Compounds of formula I can be converted into other compounds of formula I. For example, in a manner known per se substituents in the aryl radical Ri can be converted into one another.
For example, halo-lower alkyl, e.g. chloromethyl, can be reacted, for example, with unsubstituted or substituted lower alkanols, lower alkanethiols or lower alkylamines in accordance with a nucleophilic substitution reaction, yielding unsubstituted or substituted lower alkoxy-lower alkyl, lower alkylthio-lower alkyl or lower alkylamino-lower alkyl, respectively.
Hydroxy can be reacted, for example, with unsubstituted or substituted halo-lower alkanes, yielding unsubstituted or substituted lower alkoxy. Hydroxy can, for example, also be reacted first with a di-halo-lower alkane, for example 1 -bromo-2-chloroethane, yielding Ω-halo-lower alkoxy; the latter can be reacted in a manner analogous to that described above with unsubstituted or substititued lower alkanols, lower alkanethiols or lower alkyl¬ amines in accordance with a nucleophilic substitution reaction, yielding unsubstituted or substituted lower alkoxy-lower alkoxy, lower alkylthio-lower alkoxy or lower alkylamino-lower alkoxy, respectively.
Analogously to hydroxy, mercapto can also be alkylated as described in the preceding paragraph. Lower alkylthio groups can be converted by controlled oxidation both into lower alkylsulfinyl groups and into lower alkylsulfonyl groups.
Amino groups and hydroxy groups can be acylated in known manner, yielding, for example, lower alkanoylarπino and lower alkanoyloxy groups, respectively.
Carboxylic acid radicals can be converted in accordance with known derivatisation methods, such as esterification or amide formation, into carboxylic acid derivatives, such as lower alkoxycarbonyl, aminocarbonyl, N-lower alkylaminocarbonyl, N,N-di-lower alkylamino¬ carbonyl, cyano or amidino. Conversely, carboxylic acid derivatives can also be converted into free carboxylic acids, for example by hydrolysis.
Compounds of formula I wherein R2 is hydrogen can be converted by reaction with a halogenating agent, for example a N-halosuccinimide, into compounds of formula I wherein R2 is halogen.
For example, hydroxy groups can be esterified with organic or inorganic acids or etherified with alcohols or organic halides or they can be removed by reduction.
If any of the intermediates contain troublesome reactive groups, for example carboxy, hydroxy, mercapto or amino groups, those groups can be protected temporarily by readily removable protecting groups. The choice of suitable protecting groups, their introduction and their removal are known perse and are described, for example, in J.F.W. McOmie, Protective Groups in Organic Chemistry, Plenum Press, London, New York 1973 and, for example, in T.W. Greene and P.G.M. Wuts, Protective Groups in Organic Synthesis, Wiley, New York et al., 1991 , or also in PJ. Kocienski, Protecting Groups, Thieme, Stuttgart, New York 1994.
Salts of compounds I can be prepared in a manner known per se. For example, acid addition salts of compounds I are obtained by treatment with a suitable acid or a suitable ion exchange reagent and salts with bases by treatment with a suitable base or a suitable ion exchange reagent. Salts of compounds of formula I can be converted into the free compounds I in customary manner: acid addition salts, for example, by treatment with a suitable basic agent or a suitable ion exchange reagent and salts with bases, for example, by treatment with a suitable acid or a suitable ion exchange reagent.
Salts of compounds I can be converted in a manner known per se into other salts of compounds I: acid addition salts can be converted, for example, into other acid addition salts, for example by treatment of a salt of an inorganic acid, such as a hydrochloride, with a suitable metal salt, such as a sodium, barium or silver salt, of an acid, for example with silver acetate, in a suitable solvent in which an inorganic salt that forms, for example silver chloride, is insoluble and thus precipitates from the reaction mixture.
Depending upon the procedure and reaction conditions, compounds I having salt-forming properties can be obtained in free form or in the form of salts
Owing to the close relationship between the compound I in free form and in the form of its salts, hereinbefore and hereinafter any reference to the free compound I or its salts should be understood as including also the corresponding salts or the free compound I, respect¬ ively, as appropπate and expedient
The compounds I, including the salts of salt-forming compounds, can also be obtained in the form of their hydrates and/or may include other solvents, for example solvents that may have been used for the crystallisation of compounds in solid form
Depending upon the starting materials and procedures chosen, the compounds I and their salts may be in the form of one of the possible isomers or in the form of a mixture thereof. There are obtainable as pure isomers, for example, pure diastereoisomers Accordingly, isomeπc mixtures may be in the form of, for example, diastereoisomenc mixtures. Isomeπc mixtures of compounds I in free form or in salt form obtainable in accordance with the process or by another method can be separated into their components in customary manner, for example on the basis of the physico-chemical differences between the con¬ stituents in known manner by fractional crystallisation, distillation and/or chromatography Advantageously, the more active isomer is isolated.
The invention relates also to those forms of the process in which a compound obtainable as intermediate at any stage of the process is used as starting materiai and the remaining steps are carried out or a starting material is used in the form of a derivative or salt or, especially, is formed under the reaction conditions.
In the process of the present invention it is preferable to use those starting materials and intermediates, in each case in free form or in salt form, which result in the compounds I described at the beginning as being especially valuable or the salts thereof. The invention relates also to novel starting materials and intermediates, in each case in free form or in salt form, for the preparation of compounds I or the salts thereof, to the use thereof and to processes for their preparation, the variable R being as defined for compounds I.
The invention relates also to the use of compounds I and their pharmaceutically acceptable salts in the treatment of allergic conditions and diseases, preferably in the form of pharma¬ ceutically acceptable preparations, especially in a method for the therapeutic treatment of the animal or human body, and to such a method of treatment.
The invention relates also to pharmaceutical compositions comprising as active ingredient a compound I or a pharmaceutically acceptable salt thereof, and to processes for their preparation. Those pharmaceutical compositions are, for example, for enteral, such as especially oral, also rectal, administration, for parenteral administration and for local administration to warm-blooded animals, especially humans, the compositions comprising the pharmacological active ingredient on its own or together with customary pharmaceutical excipients. The pharmaceutical compositions comprise (in percent by weight) for example from approximately 0.001 % to 100 %, preferably from approximately 0.1 % to approxi¬ mately 50 %, active ingredient.
Pharmaceutical compositions for enteral or parenteral administration are, for example, those in unit dose forms, such as dragees, tablets, capsules or suppositories, and also ampoules. They are prepared in a manner known perse, for example by means of conventional mixing, granulating, confectioning, dissolving or lyophilising procedures. For example, pharmaceutical compositions for oral administration can be obtained by combining the active ingredient with solid carriers, if desired granulating a resulting mixture, and processing the mixture or granules, if desired or necessary after the addition of appropriate excipients, into tablets or dragee cores. Suitable carriers are especially fillers, such as sugars, for example lactose, saccharose, mannitol or sorbitol, cellulose preparations and/or calcium phosphates, for example tricalcium phosphate or calcium hydrogen phosphate, and also binders, such as starch pastes using, for example, corn, wheat, rice or potato starch, gelatin, tragacanth, methyl- cellulose and/or polyvinylpyrrolidone, and, if desired, disintegrators, such as the above- mentioned starches, also carboxymethyl starch, crosslinked polyvinylpyrrolidone, agar or alginic acid or a salt thereof, such as sodium alginate. Excipients are especially flow conditioners and lubricants, for example silicic acid, talc, stearic acid or salts thereof, such as magnesium or calcium stearate, and/or polyethylene glycol. Dragee cores are provided with suitable, optionally enteric, coatings, there being used, inter alia, concentrated sugar solutions which may comprise gum arabic, talc, polyvinylpyrrolidone, polyethylene glycol and/or titanium dioxide, or coating solutions in suitable organic solvents or solvent mixtures, or, for the preparation of enteric coatings, solutions of suitable cellulose preparations, such as acetylcellulose phthalate or hydroxypropylmethylcellulose phthalate. Dyes or pigments may be added to the tablets or dragee coatings, for example for identification purposes or to indicate different doses of active ingredient.
Further orally administrable pharmaceutical compositions are hard gelatin capsules, and also soft, sealed capsules consisting of gelatin and a plasticiser, such as glycerol or sorbitol. The hard gelatin capsules may comprise the active ingredient in the form of granules, for example in admixture with fillers, such as lactose, binders, such as starches, and/or glidants, such as talc or magnesium stearate, and optionally stabilisers. In soft capsules, the active ingredient is preferably dissolved or suspended in suitable liquids, such as fatty oils, paraffin oil or liquid polyethylene glycols, to which stabilisers may also have been added.
Suitable rectally administrable pharmaceutical compositions are, for example, suppositories that consist of a combination of the active ingredient with a suppository base material. Suitable suppository base materials are, for example, natural or synthetic triglycerides, paraffin hydrocarbons, polyethylene glycols or higher alkanols. It is also possible to use gelatin rectal capsules which comprise a combination of the active ingredient with a base material. Suitable base materials are, for example, liquid triglycerides, polyethylene glycols or paraffin hydrocarbons. For parenteral administration there are suitable, especially, aqueous solutions of an active ingredient in water-soluble form, for example in the form of a water-soluble salt, and also suspensions of the active ingredient, such as corresponding oily injection suspensions, there being used suitable lipophilic solvents or vehicles, such as fatty oils, for example sesame oil, or synthetic fatty acid esters, for example ethyl olβate or triglycerides, or aqueous injection suspensions which comprise viscosity-increasing substances, for example sodium carboxymethylcellulose, sorbitol and/or dextran, and optionally also stabilisers.
Pharmaceutical compositions for local administration are, for example for topical treatment of the skin, lotions, creams and ointments, i.e. liquid or semi-solid oil-in-water or water-in-oil emulsions, fatty ointments, which are anhydrous, pastes, i.e. creams and ointments having secretion-absorbing powder constituents, gels, which are aqueous, of low water content or anhydrous and consist of swellable, gel-forming materials, foams, i.e. liquid oil-in-water emulsions in aerosol form which are administered from pressurised containers, and tinctures having an aqueous-ethanolic base and may comprise other customary pharma¬ ceutical excipients, such as preservatives. The pharmaceutical compositions for local administration are prepared in a manner known per se by mixing the active ingredient with the pharmaceutical excipients, for example by dissolving or suspending the active ingred¬ ient in the base or in a portion thereof, if necessary. In order to prepare emulsions in which the active ingredient is dissolved in one of the liquid phases, the active ingredient is normally dissolved therein prior to emulsification; in order to prepare suspensions in which the active ingredient is suspended in the emulsion, the active ingredient is mixed with a portion of the base after emulsification and then added to the remainder of the formulation.
The dosage of the active ingredient can depend upon various factors, such as the effective¬ ness and duration of action of the active ingredient, the severity of the disease to be treated and of its symptoms, the mode of administration, the species of warm-blooded animal, and the sex, age, weight and/or individual condition of the warm-blooded animal. In a normal case, the, for example oral, daily dose for a warm-blooded animal weighing approximately 75 kg is estimated to be from approximately 1 mg to approximately 1000 mg, especially from approximately 5 mg to approximately 200 mg. It can be administered, for example, as a single dose or in several part doses of, for example, from 10 to 100 mg. The following Examples are intended to illustrate the invention described hereinbefore, but without limiting the invention thereto. (Hereinbefore and hereinafter, unless otherwise indicated the meanings of the following abbreviations are: M.p.: = melting point; CDCI3 = deuterochloroform; DMSO-d6 = hexadeuterodimethyl sulfoxide; CD3OD = deuteromethanol; C2D2CI4 = deuterotetrachloroethane).
Example 1 : 5-lsopropyl-7-phenyl-5H-pyrrolof3.2-d]pyrimidin-4-ylamine 0.252 g of 4-chloro-5-isopropyl-7-phenyl-5H-pyrrolo[3,2-d]pyrimidine is stirred in a bomb tube with 15 ml of ammonia at 130°C for 24 hours. The ammonia is evaporated off and the residue is purified by flash chromatography (methylene chloride/methanol 10:0.4). M.p.: 96-98°C. 1H-NMR (CDCI3, ppm): 8.50 (s, 1H), 8.0 (d, 2H), 7.65 (s, 1 H), 7.45 (m, 2H), 7.25 (m, 1 H), 5.1 (s, 2H), 4.7 (qq, 1 H), 1.68 (d, 6H).
a) (3-Nitrilo-2-phenyl-propenylamino)-acetic acid ethyl ester: 2.50 g of glycine ethyl ester hydrochloride are suspended in 200 ml of toluene, and 2.50 ml of triethylamine, 2.60 g of α-formyl-phenylacetonitrile and 0.050 g of toluenesulfonic acid are added and stirring is carried out under reflux using a water separator for 1.5 hours. The reaction mixture is cooled to room temperature and filtered. The filtrate is concentrated by evaporation using a rotary evaporator and the residue is dried under a high vacuum. 1H-NMR (CDCI3, ppm): 7.42-7.32 (m, 4H), 7.18 (m, 1 H), 7.12 (s, 1 H), 5.45 (m (broad), 1 H), 4.3 (q, 2H), 4.08 (d, 2H), 1.30 (t, 3H).
α-Formyl-phenylacetonitrile (CAS Rea.No.:5841-70-3l\s prepared according to J.D. Albright et al. US 79-34060.
b) 3-Amino-4-phenyl-7H-pyrrole-2-carboxylic acid ethyl ester: 0.430 g of sodium is dissolved in 20 ml of ethanol (absolute) and then 4.00 g of (3-nitrilo-2-phenyl-propenylamino)-acetic acid ethyl ester are added. The reaction mixture is stirred at room temperature for 48 hours and then poured into 300 ml of water. The suspension is filtered and the solid is dissolved in methylene chloride, the organic phase is extracted with saturated sodium chloride solution and dried over sodium suifate, and the solvent is removed using a rotary evaporator. The residue is purified by flash chromatography (diethyl ether/petroleum ether 1 :1 ). 1H-NMR (CDCI3, ppm): 8.30 (s (broad),1 H), 7.50-7.30 (m, 5H), 6.85 (d, 1H), 4.6 (s (broad), 2H), 4.3 (q, 2H), 1.38 (t, 3H).
c) 3-(Dtmethylammo-methyleneamιnoM-phenyl-7H-pyrrole-2-caιtx>xylιc acid ethyl ester: 2.10 g of 3-amιno-4-phenyl-τΗ-pyrrole-2-carboxylic acid ethyl ester and 12.0 ml of dimethylformamide dineopentyl acetal are dissolved in 12.0 ml of chloroform and stirring is carried out at 70°C for 4 days The reaction mixture is concentrated by evaporation using a rotary evaporator and the residue is purified by flash chromatography (diethyl ether/- petroleum ether 3:1 ) 1H-NMR (CDCI3, ppm): 8.70 (s (broad), 1 H), 7.75 (d, 2H), 7 52 (s, 1 H), 7 45 (m, 2H), 7 18 (m, 1 H), 7.05 (d, 1 H), 4.30 (q, 2H), 3.08 (s (broad), 6H), 1.35 (t, 3H)
d) 7-Phenyl-3.5-dιhvdro-pyrrolof3.2-d]pyrιmιdιn-4-one 1.00 g of 3-(dιmethylamιno- methyleneamιno)-4-phenyl-7H-pyrrole-2-carboxylιc acid ethyl ester is dissolved in 100 ml of ammonia/methanol and stirring is carried out at 45°C in a bomb tube. Cooling to room temperature is then carried out and the suspension is filtered 1H-NMR (DMSO-d6, ppm). 12 2 (s (broad), 2H), 8.12 (d, 2H), 7.90 (d, 2H), 7.38 (m, 2H), 7 19 (m, 1 H).
e) 4-Chloro-7-phenyl-5H-pyrrolo[3.2-d]pyrιmιdιne: 1.00 g of 7-phenyl-3,5-dιhydro-pyrrolo- [3,2-d]pyrιmιdιn-4-one is stirred in 20 ml of phosphoryl chloride at 120°C for 15 hours. Phosphoryl chloride is removed using a rotary evaporator and the residue is dissolved in water and rendered alkaline with 25 % ammonia. The suspension is filtered and the product is dried Η-NMR (DMSO-d6, ppm): 12.7 (s, 1H), 8.8 (s, 1 H), 8.55 (d, 1H), 8.25 (d, 2H), 7.45 (m, 2H), 7.30 (t, 1 H)
f) 4-Chloro-5-ιsopropyl-7-phenyl-5H-pyrrolof3.2-d]pyrιmιdιne: 0.250 g of 4-chloro-7-phenyl- 5/-τ-pyrrolo[3,2-d]pyrιmιdιne is dissolved in 5 ml of dimethylformamide, and 0.300 g of potassium carbonate and 0.332 g of 18-crown-6 ether are added. The reaction mixture is stirred at 70°C for 10 minutes, then 0.200 ml of 2-bromopropane is added and stirring is again carried out at 70°C for 5 hours. The suspension is poured into water and extracted with ethyl acetate. The organic phase is separated and dried over sodium suifate, and the solvent is removed using a rotary evaporator The residue is purified by flash chromato- graphy (petroleum ether/ethyl acetate 6:1). 1H-NMR (CDCI3, ppm): 8.8 (s, 1H), 8.08 (d, 2H), 7.92 (s, 1 H), 7.50 (m, 2H), 7.40 (m, 1 H), 5.10 (m, 1H), 1.70 (d, 6H).
The following Examples are prepared analogously using 4-chloro-7-phenyl-5H-pyrrolo- [3,2-d]pyrimidine and the appropriate bromides as starting materials:
Example 2: 5-Cvclopentyl-7-phenyl-5H-pyrrolof3.2-d1pyrimidin-4-ylamine
M.p.: 211-212°C. 1H-NMR (CDCI3, ppm): 8.5 (s, 1H), 8.05 (m, 2H), 7.45 (m, 3H), 7.2 (s,1 H),
5.1 (s, 2H), 4.95 (m, 1H), 2.4-2.2 (m, 8H).
Example 3: 5-(2-Methoxy-ethoxymethyl)-7-phenyl-5H-Pyrrolof3.2-d]pyrimidin-4-ylamine M.p.: 161 -162°C. 1H-NMR (CDCI3, ppm): 8.5 (s, 1 H), 8.0 (m, 2H), 7.45 (m, 3H), 7.2 (s, 1 H), 5.8 (s, 2H), 5.6 (s, 2H), 3.7 (m, 2H), 3.59 (m, 2H), 3.4 (s, 3H).
Example 4: 5-f2-(2-Methoxy-ethoxy)-ethyl1-7-phenyl-5H-pyrrolof3.2-dlpyrimidin-4-ylamine M.p.: 150-151 °C. 'H-NMR (CDCI3, ppm): 8.5 (s, 1H), 8.05 (m, 2H), 7.45 (m, 3H), 7.2 (s, 1H), 5.8 (s, 2H), 4.55 (t, 2H), 3.95 (t, 2H), 3.62 (m, 2H), 3.48 (m, 2H), 3.3 (s, 3H).
Example 5: (4R/2S) 4-(4-Amino-7-phenyl-pyrrolof3.2-dlpyrimidin-5-yl)-2-carbamoyl- pyrrolidine-1 -carboxylic acid (erf-butyl ester
'H-NMR (CDCI3, ppm): 8.5 (s, 1 H), 7.98 (d, 2H), 7.5 (m, 3H), 7.22 (s, 1 H), 6.0 (s, 2H), 5.50 (m, 1 H), 4.98 (d, 1 H), 4.12 (t, 1 H), 3.90 (t, 1 H), 2.99 (m, 1 H), 2.1 (m, 1H), 1.1 (s, 9H).
Example 6: (4R/2R) 4-(4-Amino-7-phenyl-pyrrolof3.2-d1pyrimidin-5-yl)-2-carbamoyl- pyrrolidine-1 -carboxylic acid fe/t-butyl ester
'H-NMR (C2D2CI4, ppm): 8.5 (s, 1 H), 7.98 (d, 2H), 7.5 (m, 3H), 7.22 (s, 1 H), 6.0 (s, 2H), 5.50 (m, 1 H), 4.98 (d, 1 H), 4.12 (t, 1 H), 3.90 (t, 1 H), 2.99 (m, 1 H), 2.1 (m, 1H), 1.1 (s, 9H).
Example 7: 5.7-Diphenyl-5H-pyrrolof3.2-d]pyrimidin-4-ylamine
0.200 g of 4-chloro-5,7-diphenyl-5H-pyrrolo[3,2-d]pyrimidine is stirred in a bomb tube with 15 ml of ammonia at 40°C for 7 days. Unreacted ammonia is evaporated off and the residue is recrystallised from ethanol. M.p.:199-201 °C. 1H-NMR (CDCI3, ppm): 8.55 (s, 1H), 8.08 (d, 2H), 7.65-7.4 (m, 8H), 7.27 (s, 1 H), 4.80 (s, 2H). a) f(3-Nitrilo-2-phenyl-propenyl)-phenyl-aminol-acetic acid ethyl ester: 4.48 g of phenyl- ammo-acetic acid ethyl ester, 3.60 g of α-formyl-phenylacetonitrile and 0.100 g of toluenesulfonic acid are dissolved in 200 ml of toluene and stirred under reflux using a water separator for 1.5 hours. The reaction mixture is cooled to room temperature and concentrated by evaporation using a rotary evaporator, and the residue is dried under a high vacuum. Η-NMR (CDCI3, ppm): 7.40-7.20 (m, 11 H), 4.88 (s, 2H), 4.30 (q, 2H), 1.35 (t, 3H).
α-Formyl-phenylacetonitrile ICAS Rea.No.:5841 -70-31 is prepared according to J.D. Albright et al. US 79-34060.
Phenylammo-acetic acid ethyl ester (CAS Reα.No.: 2216-92-4]\s prepared according to W.K. Anderson et al., J. Med. Chem. 1989, 32, page 1 19.
b) 3-Amιno-1 ,4-dιphenyl- 7Η-pyrrolo-2-carboxylic acid ethyl ester: 0.115 g of sodium is dissolved in 5 ml of absolute ethanol, and 1.39 g of [(3-nιtrilo-2-phenyl-propenyl)-phenyl- amιno]-acetιc acid ethyl ester are added, and the reaction mixture is subsequently stirred at room temperature for 1 hour. It is then poured into water and extracted with ethyl acetate. The organic phase is separated and dried over sodium suifate, and the solvent is removed using a rotary evaporator. The residue is purified by flash chromatography (petroleum ether/diethyl ether 2:1). Η-NMR (CDCI3, ppm): 7.52-7.22 (m, 10H), 6.88 (s, 1 H), 4.85 (s, 2H), 4.12 (q, 2H), 1.1 (t, 3H).
c) 3-(Dιmethylamιno-methyleneamino)-4-phenyl-7H-pyrrolo-2-carboxylιc acid ethyl ester: 2.10 g of 3-amιno-1 ,4-dιphenyl-ϊH-pyrrolo-2-carboxylιc acid ethyl ester are dissolved in 7 ml of chloroform, and 5.97 ml of dimethylformamide dineopentyl acetal are added. The reaction mixture is stirred at 70°C for 5 days. It is then concentrated by evaporation using a rotary evaporator and the residue is solidified with petroleum ether. 1 H-NMR (CDCI3, ppm): 7.72 (d, 2H), 7.51 (s, 1 H), 7.42-7.1 (m, 8H), 7.05 (s, 1 H), 4.05 (q, 2H), 3.05 (s (broad), 6H), 1.05 (t, 3H) d) 7.5-Diphenyl-3,5-dihvdro-pyrrolo[3.2-d1pyrimidin-4-one: A solution of 0.750 g of 3-(di- methylamino-methyleneamino)-4-phenyl-7H-pyrrolo-2-carboxylic acid ethyl ester in ammonia/methanol is stirred at room temperature for 3 days. Ammonia/methanol are removed using a rotary evaporator and the product is solidified with diethyl ether. Η-NMR (CDCI3, ppm): 11.7 (s, 1 H), 7.98 (d, 2H), 7.8 (s, 1 H), 7.62 (s, 1 H), 7.58-7.32 (m, 8H).
e) 4-Chloro-5.7-diphenyl-5H-pyrrolo[3.2-dlpyrimidine: 0.440 g of 7,5-diphenyl-3,5-dihydro- pyrrolo[3,2-d)pyrimidin-4-one is stirred in 1 ml of phosphoryl chloride at 1 15°C. Phosphoryl chloride is removed using a rotary evaporator and the product is solidified with petroleum ether. Η-NMR (CD3OD, ppm): 8.9 (s, 1 H), 8.5 (s, 1 H), 7.95 (d, 2H), 7.60-7.38 (m, 8H).
The following Examples are prepared analogously to Example 7 using the variously substituted phenylaminoacetic acid ethyl ester or benzylaminoacetic acid ethyl ester and the appropriate α-formyl-acetonitrile derivative as starting materials.
Example 8: 5-Benzyl-7-phenyl-5H-pyrrolof3.2-d1pyrimidin-4-ylamine
'H-NMR (CDCI3, ppm): 8.48 (s, 1 H), 8.02 (d, 2H), 7.60-7.10 (m, 9H), 5.02 (s, 2H), 4.70 (s
(broad), 2H).
Example 9: 5-(4-Methoxy-phenyl)-7-phenyl-5H-pyrrolo[3,2-d]pyrimidin-4-ylamine M.p.: 240-244°C.
Example 10: 5-(3-Methoxy-phenyl)-7-phenyl-5/-/-pyrrolof3.2-d]pyrimidin-4-ylamine
Example 11 : 4-,4-Amino-7-phenyl-pyrrolo[3.2-dlpyrimidin-5-yl,-phenol
Η-NMR (DMSO-de, ppm): 9.98 (s, 1 H), 8.29 (s, 1 H), 8.22 (d, 2H), 8.10 (s, 1H), 7.40 (m,
4H), 7.20 (m, 1 H), 6.92 (d, 2H), 5.75 (s (broad), 2H).
Example 12: 3-(4-Amino-7-phenyl-pyrrolof3.2-d1pyrimidin-5-yl)-phenol Example 13: 7-[4-(2-Benzyloxy-ethoxy)-phenyl1-5-phenyl-5H-pyrrolo[3.2-d1pyrimidin-4- ylamine
1H-NMR (CDCI3, ppm): 8.55 (s, 1 H), 8.00 (d, 2H), 7.68-7.20 (m, 11 H), 7.03 (d, 2H), 4.78 (s,
2H), 4.75 (s, 2H), 4.22 (t, 2H), 3.89 (t, 2H). M.p.: 139-141°C.
Example 14: 2-f4-(4-Amino-5-phenyl-5H-Pyrrolor3.2-dlpyrimidin-7-yπ-phenoxyl-ethanol Η-NMR (DMSO-d6, ppm): 8.32 (s, 1H), 8.15 (m, 3H), 7.60 (m, 6H), 7.0 (d, 2H), 6.8 (s, 2H), 4.89 (t, 1 H), 4.0 (t, 2H), 3.72 (m, 2H). M.p.: 190-192°C.
Example 15: 7-f4-(2-lmidazol-1 -yl-ethoxy)-phenyll-5-phenyl-5H-pyrrolof3.2-dlPyrimidin-4-yl- amine
Η-NMR (CDCl3, ppm): 8.52 (s, 1 H), 7.99 (d, 2H), 7.65 (m, 7H), 7.28 (s, 1 H), 7.09 (m, 2H),
6.98 (d, 2H), 4.70 (s, 2H), 4.40 (t, 2H), 4.28 (t, 2H).
Example 16: 7-Cvclohexen-1-enyl-5-(3-methoxy-phenyl)-5H-pyrrolo[3.2-d]pyrimidin-4- ylamine
The title compound is obtained from 1.08 g of 7-cyclohex-1-enyl-5-(3-methoxyphenyl)-5H- pyrrolo[3,2-d]pyrimidin-4-ol in a manner analogous to that described in Examples 1 and 1e), after re crystallisation from hexane/ethyl acetate. Η-NMR (CDCI3, ppm): 8.55 (br. s, 1 H),
7.44 (m, 1 H), 7.29 (m, 1H), 7.15 (m, 1 H), 7.05-6.95 (m, 2H), 6.93 (s, 1 H), 4.78 (br. s, 2H),
3.87 (s, 3H), 2.45 (m, 2H), 2.32 (m, 2H), 1.81 (m, 2H), 1.71 (m, 2H). FAB-MS: 321 [M+H]+.
a) Sodium 2-cvclohex-1 -enyl-3-oxido-acrylonitrile: 10.3 g of sodium hydride (80 % dispersion in oil) are washed twice with absolute tetrahydrofuran and then suspended in
1.2 I of tetrahydrofuran. With stirring at room temperature, 83 ml of ethyl formate are added thereto and then a solution of 25 ml of (1-cyclohex-1-enyl)acetonitrile in 50 ml of tetrahydro¬ furan is added dropwise over 10 minutes. The reaction mixture is heated to 55-60°C and stirred until no further gas evolution is observed (approx. 2 hours). The viscous slurry is filtered off with suction, washed with tetrahydrofuran and dried overnight at 45°C under a high vacuum. The crude title compound is obtained in the form of a beige solid. Η-NMR (DMSO-De, ppm): 8.42 (s, 1 H), 4.03 (m, 1 H), 2.06 (m, 2H), 1.99 (m, 2H), 1.6-1.45 (m, 4H).
b) [(3-Nitrilo-2-(cvclohex-1 -enyl)-propenyl)-(3-methoxyphenyl)aminol-acetic acid ethyl ester:
7.3 ml of 4M hydrogen chloride in dioxane are added at room temperature, with stirring, to a mixture of 5.0 g of the sodium salt of 2-cyclohex-1-enyl-3-hydroxy-acrylonitrile in 50 ml of toluene. After 5 minutes, 6.1 g of N-(3-methoxyphenyl)glycine ethyl ester and a small amount of p-toluenesulfonic acid hydrate are added and the suspension is heated under reflux using a water separator. After 1 hour, the reaction mixture is left to cool, the undis- solved residue is removed by filtration and the filtrate is concentrated. The oily residue is pre-purified by flash chromatography (hexane/ethyl acetate 10:1). Unreacted N-(3-methoxy- phenyl)glycine ethyl ester is then substantially removed from the resulting crude product by bulb tube distillation (170-180°C/0.06 mbar). The crude title compound is obtained in the form of a coloured oil. Η-NMR (CDCI3, ppm): 7.28 (m, 1H), 6.96 (s, 1H), 6.75-6.6 (m, 3H), 5.98 (m, 1 H), 4.79 (s, 2H), 4.28 (q, J=7Hz, 2H), 3.81 (s, 3H), 2.2-2.1 (m, 4H), 1.71 (m, 2H), 1.61 (m, 2H), 1.31 (t, J=7Hz, 3H).
c) 3-Amino-1 -(3-methoχyphenyl)-4-(cyclohex-1 -enyl)-7/-/-pyrrole-2-carboxylic acid ethyl ester: The title compound is obtained from 2.18 g of [(3-nitrilo-2-(cyclohex-1-enyl)-propenyl)- (3-methoxyphenyl)amino]-acetic acid ethyl ester in a manner analogous to that described in Example 1b). Η-NMR (CDCI3, ppm): 7.26 (m, 1 H), 6.9-6.75 (m, 3H), 6.68 (s, 1 H), 5.96 (m, 1 H), 4.86 (br. s, 2H), 4.12 (q, J=7Hz, 2H), 3.81 (s, 3H), 2.3-2.15 (m, 4H), 1.74 (m, 2H), 1.66 (m, 2H), 1.07 (t, J=7Hz, 3H).
d) 7-Cvclohex-1 -enyl-5-(3-methoxyphenyl)-5H-pyrrolof3.2-dlpyrimidin-4-ol: The title compound is obtained from 2.18 g of 3-amino-1 -(3-methoxyphenyl)-4-(cyclohex-1-enyl)-7H- pyrrole-2-carboxylic acid ethyl ester in a manner analogous to that described in Examples 1c) and 1d). Η-NMR (DMSO-D6, ppm): 12.1 (s, 1H), 7.90 (m, 1H), 7.56 (s, 1H), 7.35 (m, 1 H), 7.1-6.9 (m, 4H), 3.78 (s, 3H), 2.39 (m, 2H), 2.18 (m, 2H), 1.70 (m, 2H), 1.62 (m, 2H). FAB-MS: 322 [M+H]+.
Example 17: 7-Cvclohexyl-5-(3-methoxy-phenvO-5H-pyrrolof3.2-d]pyrimidin-4-ylamine A solution of 0.22 g of 7-cyclohex-1-enyl-5-(3-methoxyphenyl)-5H-pyrrolo[3,2-d]pyrimidin-4- ylamine in 10 ml of methanol is hydrogenated in the presence of 0.025 g of Pd/C (10 %) at room temperature under normal pressure for 3 days. The reaction mixture is filtered over Celite 545 and the crude product obtained after concentration of the filtrate is purified by flash chromatography (methylene chloride/methanol 97:3). Η-NMR (CD3OD, ppm): 8.20 (s, 1H), 7.47 (m, 1 H), 7.35 (s, 1 H), 7.08 (m, 1 H), 7.05-6.95 (m, 2H), 3.86 (s, 3H), 2.96 (m, 1 H), 2.15-2.05 (m, 2H), 1.95-1.7 (m, 3H), 1.6-1.25 (m, 5H). FAB-MS: 322 M+, 321 [M-H]+.
Example 18: 7-Cvclohexen-1 -enyl-5-(3-hvdroxy-phenyl)-5H-pyrrolo[3.2-d]pyπmidιn-4- ylamine
0.27 ml of boron tribromide is added in portions in the course of 15 minutes at 0-5°C to a solution of 0.30 g of 7-cyclohex-1-enyl-5-(3-methoxyphenyl)-5H-pyrrolo[3,2-d]pyrιmιdin-4- ylamine in 10 ml of methylene chloride and, after 2.5 hours, a further 0.27 ml of boron tribromide is added. Stirring is then carried out for a further hour, 1.5 ml of water are then added dropwise and the mixture is stirred at 0-5°C for 30 minutes. The crude product obtained after customary working-up is purified by flash chromatography (methylene chlonde/methanol 95:5). Η-NMR (DMSO-D6, ppm): 10.0 (s, 1H), 8.24 (s, 1 H), 7.55 (s, 1H), 7.36 (m, 1 H), 7.16 (m, 1 H), 6.89 (m, 2H), 6.80 (m, 1 H), 5.80 (br. s, 2H), 2.39 (m, 2H), 2.20 (m, 2H), 1.72 (m, 2H), 1.52 (m, 2H).
Example 19: 5-Phenyl-7-(pyrιdιn-3-yl)-5H-pyrrolo[3.2-dlpyrιmidιn-4-ylamιne A solution of 0.25 g of 5-phenyl-7-(pyridin-3-yl)-5/-/-pyrrolo[3,2-d]pyrιmιdιn-4-ol in 2 ml of phosphoryl chloride is stirred at 120°C for 2 hours. The reaction mixture is concentrated using a rotary evaporator and the residue is dried under a high vacuum. The crude product so obtained is stirred in 15 ml of liquid ammonia at 130°C in an autoclave for 24 hours. After the ammonia has been evaporated off, the solid residue is taken up in methylene chlonde/- methanol 2:1 and filtered, and the residue obtained after concentration of the filtrate is purified by flash chromatography (methylene chlonde/methanol 95:5). The title compound is obtained. Η-NMR (CDCI3, ppm): 9.11 (br. s, 1 H), 8.65-8.5 (m, 3H), 7.69 (s, 1 H), 7.65-7.5 (m, 5H), 7.40 (m, 1 H), 4.83 (br. s, 2H).
a) r(3-Nitrιlo-2-(pyrιdιn-3-vπ-propenyl)-phenylamιnol-acetιc acid ethyl ester: A mixture of 1.0 g of 3-oxo-2-(pyrιdιn-3-yl)-propionitrile (prepared according to M. Caπou, Bull. Soc. Chim. France (1979), 11-12, 651 -656), 1.12 g of phenylammo-acetic acid ethyl ester and a catalytic amount of p-toluenesulfonic acid hydrate in 50 ml of toluene and 7 ml of dimethyl sulfoxide is stirred under reflux using a water separator for 3 hours. The reaction mixture is filtered and the filtrate is then washed with saturated sodium carbonate solution and brine, dried over magnesium suifate and concentrated. The residue is purified by means of flash chromatography (hexane/ethyl acetate 2:1 ). 1H-NMR (CDCI3, ppm): 8.63 (s, 1 H), 8.44 (m, 1 H), 7.71 (m, 1 H), 7.42 (m, 2H), 7.35-7.2 (m, 5H), 4.86 (s, 2H), 4.32 (q, 2H), 1.33 (t, 3H).
b) 3-Amino-1-phenyl-4-(pyridin-3-vD-7H-pyrrole-2-carboxylic acid ethyl ester: 0.38 g of [(3- nitrilo-2-(pyridin-3-yl)-propenyl)-phenylamino]-acetic acid ethyl ester, dissolved in 8 ml of absolute ethanol, is added to a solution of 32 mg of sodium in 15 ml of absolute EtOH. The reaction mixture is stirred at room temperature for 60 minutes and then concentrated using a rotary evaporator, and the residue is taken up in 40 ml of ethyl acetate. The organic phase is washed with saturated sodium carbonate solution and brine, dried over magnesium suifate and concentrated. 1H-NMR (CDCI3, ppm): 8.77 (m, 1 H), 8.52 (m, 1H), 7.84 (m, 1 H), 7.5-7.25 (m, 6H), 6.90 (s, 1H), 4.33 (br s, 2H), 4.14 (q, 2H), 1.06 (t, 3H).
c) 3-(Dιmethylamιno-methyleneamιno)-1-phenyl-4-(pyridιn-3-yl)-7/-/-Pyrrole-2-carboxylic acid ethyl ester: 1.64 ml of N,N-dιmethylformamide dineopentyl acetal are added to a solution of 0.58 g of 3-amιno-1 -phenyl-4-(pyrιdιn-3-yl)-7H-pyrrole-2-carboxyltc acid ethyl ester in 5 ml of chloroform and the mixture is stirred at 70°C for 4 days. It is concentrated using a rotary evaporator and the residue is purified by flash chromatography (hexane/ethyl acetate 1 :2). 'H-NMR (CDCI3, ppm): 8.98 (s, 1H), 8.41 (m, 1 H), 8.09 (m, 1 H), 7.58 (s, 1H), 7.45-7.2 (m, 6H), 7.10 (s, I H), 4.13 (q, 2H), 3.09 (br s, 3H), 3.03 (br s, 3H), 1.02 (t, 3H).
d) 5-Phenyl-7-(pyrιdιn-3-yl)-5/-/-pyrrolo[3.2-d]pyrιmιdιn-4-ol: A mixture of 0.66 g of 3-(dι- methylamιno-methyleneamιno)-1 -phenyl-4-(pyridιn-3-yl)-7H-pyrrole-2-carboxylιc acid ethyl ester and 40 ml of saturated methanohc ammonia solution is stirred in a bomb tube at 40-45°C for 65 hours. The whitish suspension is filtered with suction and the residue is washed with methanol and dried at 50°C under a high vacuum. Η-NMR (DMSO-D6, ppm): 8.20 (s, 1 H), 7.65-7.5 (m, 2H), 7.08 (m, 1 H), 6.84 (m, 1H), 6.7-6.6 (m, 6H), 6.49 (s, 1 H).
Example 20: 4-{4-Amιno-7-[4-(2-ιmidazol-1 -yl-ethoxy)-phenyll-pyrrolof3.2-dlPyrimidin-5-yl I- phenol
Example 21 : 3-(4-Amιno-7-r4-(2-ιmιdazol-1-yl-ethoxy)-phenyll-pyrrolo[3.2-d]pyrimidin-5-yll- phenol Example 22: 7-f4-(2-lmidazol-1 -yl-ethoxy)-phenvn-5-(4-methoxy-phenvh-5H-Pyrrolof3.2-dl- pyrimidin-4-ylamine
Example 23: 7-f4-(2-lmidazol-1 -yl-ethoxy)-phenvn-5-(3-methoxy-phenyl)-5/-/-pyrrolof3.2-d1- pyrimidιn-4-ylamιne
'H-NMR (CD3OD, ppm): 8.28 (s, 1 H), 7.85 (d, 2H), 7.75 (d, 2H), 7.50 (t, 1H), 7.25 (s, 1 H),
7.12 - 6.90 (m, 6H), 4.45 (t, 2H), 4.28 (t, 2H), 3.87 (s, 3H).
Example 24: 7-f4-(2-Amino-ethoxy)-phenvn-5-phenyl-5H-pyrrolof3.2-d1pyrιmidιn-4-ylamine
Example 25: 4-(4-Amιno-7-f4-(2-amιno-ethoxy)-phenvn-pyrrolo[3.2-dlpyrιmidιn-5-yll-phenol
Example 26: 3-(4-Amιno-7-[4-(2-amιno-ethoxy)-phenvπ-pyrrolo[3,2-d1pyrιmιdιn-5-yl)-phenol
Example 27: 7-[4-(2-Amino-ethoxy)-Phenyl1-5-(4-methoxy-phenyl,-5/-/-pyrrolof3.2-dlpyrιmι- dιn-4-ylamιne
Example 28: 7-[4-(2-Amιno-ethoxy)-phenvn-5-(3-methoxy-phenyl)-5/-/-pyrrolof3.2-dlpyπmι- dιn-4-ylamιne
'H-NMR (DMSO-d6, ppm): 8.30 (s, 1 H), 8.10 (m, 3H), 7.50 (t, 1 H), 7.18 - 6.97 (m, 5H), 5.8
(s, 2H), 3.98 (t, 2H), 3.85 (s, 3H), 2.90 (t, 2H).
Example 29: 2-(2-r4-(4-Amino-5-phenyl-5H-pyrrolof3,2-dlpyrιmιdιn-7-yl)-phenoxyl-ethyl- aminol-ethanol
Example 30: 4-(4-Amιno-7-(4-[2-(2-hvdroxy-ethylamιno)-ethoxyl-phenyl)-pyrrolo[3.2-d]- pyrιmιdιn-5-yl-phenol
Example 31 : 3-(4-Amιno-7-(4-f2-(2-hydroxy-ethylamιno)-ethoxy1-phenyl,-pyrrolo[3.2-d1- pyπmιdιn-5-yl-phenol
'H-NMR (DMSO-d6, ppm): 8.32 (s, 1 H), 8.20 - 8.08 (m, 3H), 7.40 (t, 1 H), 7.00 - 6.80 (m, 5H),
5.80 (s, 2H), 4.50 (s (broad), 1 H), 4.03 (m, 2H), 3.50 (m, 2H), 2.90 (m, 2H), 2.65 (m, 2H). Examole 32: 2-2-{4-[4-Amino-5-(4-methoxy-phenyl)-5H-pyrrolor3.2-d]pyrimidin-7-yl]-phen- oxy}-ethylamino)-ethanol
Example 33: 2-2-f4-f4-Amino-5-(3-methoxy-phenyl)-5H-pyrrolof3.2-dlpyrimidin-7-yll-phen- oxy}-ethylamino)-ethanol
Η-NMR (DMSO-d6l ppm): 8.30 (s, 1 H), 8.12 (m, 3H), 7.50 (t, 1H), 7.19 - 6.95 (m, 5H), 5.90
(s, 2H), 4.55 (s, 1 H), 4.05 (t, 2H), 3.85 (s, 3H), 3.5 (m, 2H), 2.92 (m, 2H), 2.70 (m, 2H).
Example 34: 7-Cvclohexen-1 -enyl-5-(3-fluoro-phenyl)-5H-pyrrolo[3.2-d]pyrimidin-4-ylamine
Example 35: 7-Cvclopentyl-5-phenyl-5H-pyrrolo[3.2-dlpyrimidin-4-ylamine
Example 36: 7-Cvclopent-1 -enyl-5-phenyl-5H-pyrrolof3.2-dlpyrimidin-4-ylamine
Example 37: 7-Cvclopent-3-enyl-5-phenyl-5H-pyrrolof3.2-dlpyrimidin-4-ylamine
Example 38: 3-(4-Amino-5-phenyl-5H-pyrrolor3.2-d1pyrimidin-7-yl)-cvclopentanol
Example 39: 3-K-Amino-5-phenyl-5H-pyrrolor3.2-d1pyrimidin-7-yl)-cyclopent-2-enol
Example 40: 7-(3-Methoxy-cvclopentyl-5-phenyl-5H-pyrrolo[3.2-d]pyrimidin-4-ylamine
Example 41 : 7-(3-Methoxy-cvclopent-1 -enyl-5-phenyl-5H-pyrrolof3,2-d]pyrimidin-4-ylamine
Example 42: [3-(4-Amino-5-phenyl-5/-/-pyrrolo[3.2-d1pyrimidin-7-yl)-cvclopentyl]-methanol
Example 43: 7-(3-Methoxymethyl-cvclopentyl-5-phenyl-5H-pyrrolor3.2-dlpyrimidin-4-ylamine
Example 44: 4-l4-Amino-5-phenyl-5H-pyrrolof3,2-d]pyrimidin-7-yl)-cvclopentane-1 ,2-diol
Example 45: 7-(3-Fluoro-cvclopentyl-5-phenyl-5H-pyrrolo[3.2-d]pyrimidin-4-ylamine
Example 46: 7-(3.4-Difluoro-cvclopentyl-5-phenyl-5H-pyrrolor3.2-d1pyrimidin-4-ylamine Example 47: 3-(4-Amino-5-phenyl-5H-pyrrolo[3.2-d]pyrimidin-7-yl)-cvclohexanol
Example 48: 3-(4-Amino-5-phenyl-5H-pyrrolof3.2-dlpyrimidin-7-yl,-cvclohex-2-enol
Example 49: 7-(3-Methoxy-cvclohexyl-5-phenyl-5H-pyrrolo[3.2-d]pyrimidin-4-ylamine
Example 50: 7-(3-Methoxymethyl-cvclohexyl-5-phenyl-5/-/-pyrrolor3.2-dlpyrimidin-4-ylamine
Example 51 : 4-(4-Amino-5-phenyl-5H-pyrrolo[3.2-d]pyrimidin-7-yl)-cvclohexanol
Example 52: 7-(4-Methoxy-cvclohexyl-5-phenyl-5H-pyrrolo[3.2-dlpyrimidin-4-ylamine
Example 53: 7-(4-Methoxymethyl-cvclohexyl-5-phenyl-5H-pyrrolo[3.2-dlPyrimidin-4-ylamine
Example 54: 5-Phenyl-7-pyrrolidin-3-yl-5H-pyrrolof3.2-dlpyrimidin-4-ylamine
Example 55: 7-(1 -Methyl-piperidin-3-vπ-5-phenyl-5H-pyrrolo[3.2-d]pyrimidin-4-ylamine
Example 56: 1 -f3-(4-Amino-5-phenyl-5/-/-pyrrolor3.2-dlPyrimidin-7-yl)-pyrrolidin-1 -yll- ethanone
Example 57: 5-Phenyl-7-piperidin-3-yl-5H-pyrrolor3.2-d1pyrimidin-4-ylamine
Example 58: 7-(1 -Methyl-piperidin-3-vπ-5-phenyl-5H-pyrrolof3.2-dlpyrimidin-4-ylamine
Example 59: 1-f3-(4-Amino-5-phenyl-5/-/-pyrrolo[3.2-d]pyrimidin-7-vπ-piperidin-1 -yll- ethanone
Example 60: 5-Phenyl-7-piperidin-4-yl-5/-y-pyrrolo[3.2-d1pyrimidin-4-ylamine
Example 61 : 7-(1-Methyl-piperidin-4-vπ-5-phenyl-5/-/-pyrrolof3,2-d1pyrimidin-4-ylamine Example 62: 1-f4-(4-Amino-5-phenyl-5H-pyrrolor3.2-dlpyrimidin-7-yl)-piperidin-1-vn- ethanone
Example 63: 7-(5-Methoxymethyl-pyrrolidin-3-yl)-5-phenyl-5H-pyrrolo[3.2-d]pyrimidin-4- ylamine
Example 64: 5-Phenyl-7-(tetrahvdrofuran-3-yl)-5H-pyrrolo[3,2-dlpyrimidin-4-ylamine
Example 65: 5-Phenyl-7-(tetrahvdro-thiophen-3-vπ-5H-pyrrolor3.2-dlpyrimidin-4-ylamine
Example 66: 7-(1.1-Dioxo-tetrahvdro-thiophen-3-yl)-5-phenyl-5H-pyrrolo[3.2-dlpyrimidin-4-yl- amine
Example 67: 7-Cvclopent-1 -enyl-5-(3-fluorophenyl)-5H-pyrrolo[3.2-d]pyrimidin-4-ylamine A solution of 0.07 g of sodium in 10 ml of absolute ethanol is added dropwise to a suspension of 0.81 g of N-[2-cyano-4-cyclopent-1-enyl-1-(3-fluorophenyl)-7H-pyrrolo-3-yl]- formamidme in 3 ml of ethanol and the mixture is stirred overnight at room temperature. It is then heated to reflux over 2 hours and, after cooling, the solvent is removed and the residue is partitioned between water and methylene chloride. Toluene is added to the combined organic phases, concentration is carried out using a rotary evaporator and the crude product is purified by means of flash chromatography on silica gel (methylene chloride/methanol 99:1 ). The title compound is obtained in the form of a white solid after recrystalhsation from methylene chloride/hexane. Η-NMR (CDCI3, ppm): 8.53 (s, 1 H), 7.53 (m, 1 H), 7.3-7.1 (m, 4H), 6.91 (m, 1 H), 4.78 (br. s, 2H), 2.8-2.55 (m, 4H), 2.1 -1.95 (m, 2H). FAB-MS: 295 [M+H]+.
a) Sodium 2-cvclopent-1 -enyl-3-oxido-acrylonitrile: 25.2 g of sodium hydride (80 % dispersion in oil) are washed twice with absolute tetrahydrofuran and then suspended in 1000 ml of tetrahydrofuran. 57.1 g of (1-cyclopent-1-enyl)acetonitrile are added and the mixture is heated to 50°C. After 15 minutes, 204 ml of ethyl formate are added with stirring, vigorous gas evolution commencing after a short time. After stirring at 50°C for 1 hour, the coloured suspension is filtered and the precipitate is washed with tetrahydrofuran and dried under a high vacuum. The crude title compound is obtained in the form of a beige solid. b) 2-(Cvclopent-1-enyl)-propenvπ-3-f(3-fluorophenyl)-(2-nitriloethvπ-amino]-acrylonitrile: 32.8 ml of 4M hydrogen chloride in dioxane and a small amount of p-toluenesulfonic acid hydrate are added at room temperature, with stirring, to a solution of 22.5 g of the sodium salt of 2-cyclopent-1-enyl-3-hydroxy-acrylonitrile in 150 ml of toluene. A viscous slurry is obtained, to which 50 ml of toluene are added followed by heating to 80°C. 19.7 g of (3- fluorophenyl)-acetonitrile are then added and the reaction mixture is heated under reflux using a water separator. After 1 hour, the mixture is allowed to cool, the insoluble residue is removed by filtration and the filtrate is concentrated. The oily residue is pre-purified by flash chromatography on silica gel (hexane/ethyl acetate/toluene 6:1 :7). Unreacted N-(3- fluorophenyl)glycine ethyl ester is then substantially removed from the resulting crude product by bulb tube distillation (160°C/0.04 mbar). The crude title compound is obtained in the form of a coloured oil.
c) 3-Amino-4-(cvclopent-1-enyl)-1-(3-fluorophenvπ-7H-pyrrole-2-carbonitrile: A solution of 2.45 g of 2-(cyclopent-1-enyl)-propenyl)-3-[(3-fluorophenyl)-(2-nitriloethyl)-amino]-acrylo- nitrile in 7 ml of ethanol is added at room temperature to a solution of 0.23 g of sodium in 20 ml of absolute ethanol and stirring is carried out for 30 minutes. The reaction mixture is concentrated, water is added to the residue and extraction is carried out with methylene chloride. The combined organic phases are dried over magnesium suifate and concen¬ trated, and the coloured residue is purified by means of flash chromatography on silica gel (hexane/ethyl acetate, gradient of 5:1 to 1 :1 ). The title compound is obtained in the form of a solid. Η-NMR (CDCI3, ppm): 7.42 (m, 1 H), 7.24 (m, 1 H), 7.14 (m, 1 H), 7.04 (m, 1 H), 6.82 (s, 1 H), 5.87 (m, 1 H), 4.03 (br, s, 2H), 2.7-2.5 (m, 4H), 2.05-1.9 (m, 2H).
d) N-[2-Cvano-4-(cvclopent-1-enyl)-1-(3-fluorophenyl)-7H-pyrrolo-3-yl1-formamidine: A few drops of acetic anhydride are added at room temperature, with stirring, to a mixture of 1.93 g of 3-amino-4-(cyclopent-1-enyl)-1-(3-fluorophenyl)-7H-pyrrole-2-carbonitrile and 20 ml of triethyl orthoformate. The reaction mixture is heated to 80°C and subsequently stirred for 3 hours. It is then concentrated and the residue is dried briefly under a high vacuum. The crude product so obtained is taken up in 40 ml of a saturated solution of ammonia in methanol, diluted with 30 ml of methylene chloride and stirred at room temperature for several days until the reaction comes to a standstill (monitoring by TLC). The reaction mixture is concentrated and the residue is purified by means of flash chromatography on silica gel (methylene chloride/methanol 98:2). The title compound is obtained in the form of a solid. Η-NMR (CDCI3, ppm): 8.53 (s, 1H), 7.55 (m, 1 H), 7.35-7.1 (m, 4H), 6.92 (m, 1 H), 4.76 (br. s, 2H), 2.8-2.55 (m, 4H), 2.1 -1.95 (m, 2H).
Example 68: 7-Cvclopent-1 -enyl-5-(3-methoxyphenyl)-5H-pyrrolo[3.2-d)pyrimidin-4-ylamine The title compound is obtained from 2.96 g of 7-cyclopent-1-enyl-5-(3-methoxyphenyl)-5/-/- pyrrolo[3,2-d]pyrimidin-4-ol in a manner analogous to that described in Examples 1 ) and 1e), after recrystallisation from hexane/ethyl acetate. Η-NMR (CDCI3, ppm): 8.52 (s, 1 H), 7.45 (m, 1 H), 7.26 (m, 1H), 7.05-6.85 (m, 4H), 4.87 (br. s, 2H), 3.88 (s, 3H), 2.8-2.55 (2m, 4H), 2.1-1.95 (m, 2H). FAB-MS: 307 [M+H]+.
a) r(3-Nitrilo-2-(cvclopent-1-envh-propenvn-(3-methoχyphenyl)amino]-acetic acid ethyl ester: 62 ml of 4M hydrogen chloride in dioxane are added in portions over 3 hours to a mixture of 18.4 g of N-(3-nπethoxyphenyl)glycine ethyl ester, 36.8 g of the sodium salt of 2-cyclopent- 1-enyl-3-hydroxy-acryionitrile (Example 34a) and a small amount of p-toluenesulfonic acid in 250 ml of toluene under reflux using a water separator. The reaction mixture is then stirred for a further 30 minutes and, after cooling, the precipitate is removed by filtration and the filtrate is washed with saturated sodium hydrogen carbonate solution. The aqueous phase is back-extracted with toluene and the combined organic phases are dried over magnesium suifate and concentrated. The majority of the unreacted N-(3-methoxyphenyl)giycine ethyl ester is removed from the resulting crude product by solid distillation under a high vacuum (0.15 mbar; bath temperature 160-180°C). Further purification of the crude product is carried out by means of flash chromatography (hexane/ethyl acetate 9:1 ) and, if necessary, by bulb tube distillation once more (180°C/0.08 bar). The crude title compound is obtained in the form of a coloured oil. Η-NMR (CDCI3, ppm): 7.29 (m, 1 H), 6.9-6.85 (m, 3H), 6.71 (s, 1 H), 5.81 (m, 1 H), 4.91 (br. s, 2H), 4.13 (q, J=7Hz, 2H), 3.81 (s, 3H), 2.65-2.5 (m, 4H), 2.0- 1.9 (m, 2H), 1.08 (t, J=7Hz, 3H).
b) 3-Amino-1 -(3-methoxyphenvO-4-(cvclopent-1-enyl)-7H-pyrrole-2-carboxylic acid ethyl ester: The title compound is obtained from 6.75 g of [(3-nitrilo-2-(cyclopent-1-enyl)- propenyl)-(3-methoxyphenyl)amino]-acetic acid ethyl ester in a manner analogous to that described in Example 1b). c) 7-Cvclopent-1-enyl-5-(3-methoxyphenyl)-5H-pyrrolor3.2-d]pyrimidin-4-ol: The title compound is obtained from 6.74 g of 3-amino-1-(3-methoxyphenyl)-4-(cyclopent-1-enyl)-7H- pyrrole-2-carboxylic acid ethyl ester in a manner analogous to that described in Examples 1c) and 1d). Η-NMR (DMSO-D6, ppm): 12.1 (s, 1H), 7.95 (s, 1 H), 7.60 (s, 1 H), 7.36 (m, 1 H), 7.15-6.9 (m, 3H), 6.66 (m, 1H), 3.80 (s, 3H), 2.75-2.6 (m, 2H), 2.6-2.4 (m, 2H; superimposed by solvent signal), 2.0-1.85 (m, 2H). FAB-MS: 308 [M+H]+.
Example 69: 7-Cvclopent-1-enyl-5-(4-methoxyphenvπ-5H-pyrrolor3.2-dlpyrimidin-4-ylamine The title compound is obtained in the form of a solid from 5.1 g of 7-cyclopent-1 -enyl-5-(4- methoxyphenyl)-5H-pyrrolo[3,2-d]pyrimidin-4-ol in a manner analogous to that described in Examples 1 ) and 1e), after recrystallisation from methylene chloride/hexane. Η-NMR (CDCI3, ppm): 8.49 (s, 1H), 7.45-6.95 (m, 5H), 6.87 (m, 1 H), 4.80 (br. s, 2H), 3.89 (s, 3H), 2.8-2.5 (m, 4H), 2.1-1.95 (m, 2H). FAB-MS: 307 [M+H]+.
a) [(3-Nitrilo-2-(cvclopent-1-enyl)-propenylW4-methoχyphenyl)amino1-acetic acid ethyl ester: 60.0 g of the sodium salt of 2-cyclopent-1-enyl-3-hydroxy-acrylonitrile (Example 34) and a small amount of p-toluenesulfonic acid hydrate are added at 80°C, with stirring, to a solution of 73.3 g of N-(4-methoxyphenyl)glycine ethyl ester in 500 ml of toluene and 87.6 ml of 4M hydrogen chloride in dioxane, and the reaction mixture is heated under reflux using a water separator. After 1 hour, the reaction mixture is allowed to cool, the undissolved residue is removed by filtration and the filtrate is concentrated. The oily residue is pre-purified by flash chromatography on silica gel (hexane/ethyl acetate/toluene 6:1 :7). Unreacted N-(4- methoxyphenyl)glycine ethyl ester is then substantially removed from the resulting crude product by bulb tube distillation (210°C/0.07 mbar). The title compound is obtained in the form of a solid after recrystallisation from methylene chloride/hexane. Η-NMR (CDCI3, ppm): 7.2-7.1 (m, 2H), 6.95-6.85 (m, 2H), 6.75 (s, 1 H), 5.75 (m, 1 H), 4.66 (s, 2H), 4.27 (q, J=7Hz, 2H), 3.81 (s, 3H), 2.5-2.3 (m, 4H), 2.0-1.9 (m, 2H), 1.31 (t, J=7Hz, 3H).
b) 3-Amino-1 -(4-methoxyphenyl)-4-(cvclopent-1-envπ-7H-pyrrole-2-carboxylic acid ethyl ester: The title compound is obtained in the form of a solid (recrystallised from ethyl acetate/hexane) from 34.6 g of [(3-nitrilo-2-(cyclopent-1-enyl)-propenyl)-(4-methoxy- phenyl)amino]-acetic acid ethyl ester in a manner analogous to that described in Example 1 b). M.p. 1 13.5-115°C. 1H-NMR (CDCI3, ppm): 7.18 (m, 2H), 6.90 (m, 2H), 6.65 (s, 1 H), 5.83 (m, 1 H), 4.87 (br. s, 2H), 4.12 (q, J=7Hz, 2H), 3.84 (s, 3H), 2.65-2.5 (m, 4H), 2.0-1.85 (m, 2H), 1.09 (t, J=7Hz, 3H). FAB-MS: 327 [M+H]+.
c) 7-Cvclopent-1-enyl-5-(4-methoxyphenyl)-5H-pyrrolo[3.2-d]pyrimιdin-4-ol: The title compound is obtained in the form of a solid from 7.0 g of 3-amιno-1-(4-methoxyphenyl)-4- (cyclopent-1 -enyl)-7H-pyrrole-2-carboxylιc acid ethyl ester in a manner analogous to that described in Examples 1c) and 1d). M.p. >253°C (decomp.). Η-NMR (DMSO-D6, ppm): 12.1 (s, 1 H), 7.91 (s, 1 H), 7 49 (s, 1 H), 7.40 (m, 2H), 7.00 (m, 2H), 6.63 (m, 1H), 3.81 (s, 3H), 2.75-2.6 (m, 2H), 2.6-2.4 (m, 2H, superimposed by solvent signal), 1.97-1.8 (m, 2H). FAB- MS: 308 [M+H]+
Example 70 7-Cyclopent-1 -enyl-5-(3-hvdroxyphenvπ-5H-pyrrolof3.2-d1pyrιmιdιn-4-ylamιne 0 47 ml of boron tribromide is added dropwise at 0-5°C, with stirring, to a suspension of 0.5 g of 7-cyclopent-1-enyl-5-(3-methoxyphenyl)-5H-pyrrolo[3,2-d]pyrιmιdιn-4-ylamιne in 10 ml of methylene chloride. After 45 minutes, a further 0.16 ml of boron tribromide is added and stirring is subsequently carried out for 30 minutes. 3.5 ml of water are added to the reaction mixture which is then allowed to warm slowly to room temperature and the coloured suspension is subsequently stirred for 90 minutes. The mixture is then neutralised with sodium hydroxide solution, and the resulting precipitate is filtered off and washed with a small amount of water and methylene chloride. The crude product is purified by recrystal¬ lisation from methanol. M.p. >225°C (decomp.). Η-NMR (DMSO-D6> ppm)- 10.0 (s, 1 H), 8.28 (s, 1 H), 7.58 (s, 1 H), 7.37 (m, 1 H), 6.95-6.8 (m, 3H), 6.76 (m, 1 H), 5.86 (br. s, 2H), 2.75-2.6 (m, 2H), 2.6-2 4 (m, 2H; superimposed by solvent signal), 1.95-1.8 (m, 2H). FAB- MS. 293 [M+H]+.
Example 71 7-Cvclopent-1-enyl-5-(4-hvdroxyphenyl)-5H-pyrrolo[3.2-dlpyrιmidιn-4-ylamιne The title compound is obtained in the form of a solid from 1.0 g of 7-cyclopent-1-enyl-5-(4- methoxyphenyl)-5H-pyrrolo[3,2-d]pyrιmιdιn-4-ylamιne in a manner analogous to that described in Example 70). Η-NMR (DMSO-D6, ppm): 9.94 (s, 1 H), 8.24 (s, 1 H), 7.48 (s, 1 H), 7.30 (m, 2H), 6.91 (m, 2H), 6.73 (m, 1 H), 5.66 (br. s, 2H), 2.75-2.6 (m, 2H), 2.6-2.4 (m, 2H; superimposed by solvent signal), 1.95-1.8 (m, 2H). FAB-MS: 293 [M+H]+. Example 72: 7-Cvclopentyl-5-(4-hvdroxyphenvπ-5H-pyrrolor3.2-d]pyrimidin-4-ylamine A solution of 0.22 g of 7-cyclopent-1-enyl-5-(4-hydroxyphenyl)-5H-pyrrolo[3,2-d]pyrimidin-4- ylamine in 20 ml of methanol is hydrogenated in the presence of 0.06 g of 10 % palladium- on-carbon for 24 hours at room temperature. The crude product obtained after customary working-up is purified by flash chromatography on silica gel (methylene chloride/methanol 95:5). The title compound is obtained in the form of a solid. 1H-NMR (CD3OD, ppm): 8.16 (s, 1 H), 7.3-7.25 (m, 3H), 6.9-7.0 (m, 2H), 2.25-2.1 (m, 2H), 1.9-1.6 (m, 7H). FAB-MS: 295 [M+H]+.
Example 73: 7-Cvclopentyl-5-(3-hvdroxyphenvπ-5H-pyrrolof3,2-d]pyrimidin-4-ylamine The title compound is obtained in the form of a solid from 0.30 g of 7-cyclopent-1 -enyl-5-(3- hydroxyphenyl)-5H-pyrrolo[3,2-d]pyrimidin-4-ylamine in a manner analogous to that described in Example 72). Η-NMR (DMSO-D, ppm): 9.97 (s, 1 H), 8.19 (s, 1 H), 7.43 (s, 1 H), 7.34 (m, 1 H), 6.9-6.8 (m, 2H), 6.77 (m, 1H), 5.77 (br. s, 2H), 3.19 (m, 1H), 2.1-1.95 (m, 2H), 1.85-1.55 (m, 6H). FAB-MS: 295 [M+H]+.
Example 74: 7-Cvclopentyl-5-(3-fluorophenyl)-5H-pyrrolo[3.2-dlPyrimidin-4-ylamine Obtained in the form of a solid from 0.22 g of 7-cyclopent-1-enyl-5-(3-fluorophenyl)-5H- pyrrolo[3,2-d]pyrimidin-4-ylamine in a manner analogous to that described in Example 30), after recrystallisation from methylene chloride/hexane. Η-NMR (DMSO-D6, ppm): 8.22 (s, 1 H), 7.55 (m, 1 H), 7.53 (s, 1 H), 7.4-7.2 (m, 3H), 5.92 (br. s, 2H), 3.20 (m, 1 H), 2.1 -1.95 (m, 2H), 1.85-1.55 (m, 6H). FAB-MS: 297 [M+H]+.
Example 75: 7-lsopropyl-5-(3-methoxyphenyl)-5H-pyrrolof3.2-dlpyrimidin-4-ylamine Reaction of 2.0 g of 7-isopropyl-5-(3-methoxyphenyl)-5H-pyrrolo[3,2-d]pyrimidin-4-ol in 10 ml of phosphorus oxychloride at 120°C for 2 hours, working-up and further reaction in methanolic ammonia under pressure at 130°C in a manner analogous to that described in Example 19) yield the title compound in the form of a white solid after recrystallisation from 1 :3 hexane/ethyl acetate. Η-NMR (CDCI3, ppm): 8.43 (s, 1 H), 7.45 (m, 1 H), 7.21 (s, 1 H), 7.05-6.9 (m, 3H), 5.28 (br. s, 2H), 3.88 (s, 3H), 3.78 (m, 1H), 1.40 (d, J=7Hz, 6H). FAB-MS: 283 [M+H]+. a) f2-Cvano-3-methyl-but-1-enylH3-methoxyphenyl)aminol-acetic acid ethyl ester: A mixture of 10.0 g of isovaleronitrile and ethanolic sodium ethanolate solution (2.76 g of sodium in 33 ml of absolute ethanol) in 36 ml of toluene is stirred at 50°C and 40-50 bar pressure under a carbon monoxide atmosphere for 10 hours analogously to a known process (DE 2753322 A1 ). After concentration of the reaction mixture by evaporation and drying under a high vacuum, the crude sodium 2-isopropyl-3-oxido-acrylonitrile is obtained in the form of a beige solid. Without further purification, the crude product is dissolved in about 45 ml of water and adjusted to pH 6 by the addition of 1N hydrochloric acid. Extraction is then carried out with toluene, and the combined organic phases are washed with saturated sodium chloride solution, dried over magnesium suifate and filtered. The filtrate is added to a solution, heated to 50°C, of 8.95 g of N-(3-methoxyphenyl)glycine ethyl ester in 50 ml of toluene, a catalytic amount of p-toluenesulfonic acid is added to the reaction mixture and heating is then carried out under reflux for 1 hour. After cooling, washing is carried out with 5 % sodium hydrogen carbonate solution and saturated sodium chloride solution, followed by drying over magnesium suifate and concentration. Contents of N-(3-methoxyphenyl)- glycine ethyl ester contained in the crude product are substantially removed by bulb tube distillation at 220°C/5.5 mbar. Further purification by means of flash chromatography on silica gel (hexane/ethyl acetate/toluene 5:1 :5) yields the title compound in the form of an oil. Η-NMR (CDCI3, ppm): 7.25 (m, 1H), 6.80 (s, 1 H), 6.7-6.55 (m, 3H), 4.72, (s, 2H), 4.28 (q, J=7Hz, 2H), 3.81 (s, 3H), 2.43 (m, 1 H), 1.31 (t, J=7Hz, 3H), 1.17 (d, J=7Hz, 6H).
b) 3-Amino-4-isopropyl-1-(3-methoχyphenyl)-7/-/-pyrrole-2-carboxylic acid ethyl ester: The title compound is obtained from 2.46 g of [2-cyano-3-methyl-but-1-enyl)-(3-methoxyphenyl)- amino]-acetic acid ethyl ester in a manner analogous to that described in Example 1b). 'H-NMR (CDCI3, ppm): 7.25 (m, 1 H), 6.85-6.75 (m, 3H), 6.57 (s, 1 H), 4.55 (br. s, 2H), 4.14 (q, J=7Hz, 2H), 3.81 (s, 3H), 2.75 (m, 1H), 1.23 (d, J=7Hz, 6H), 1.09 (t, J=7Hz, 3H).
c) 7-lsopropyl-5-(3-methoxyphenyl)-5H-pyrrolof3.2-d1pyrimidin-4-ol: A mixture of 2.1 g of 3- amino-4-isopropyl-1-(3-methoxyphenyl)-7H-pyrrole-2-carboxylic acid ethyl ester and 6.05 ml of N,N-dimethylformamide dineopentyl acetal in 30 ml of chloroform is stirred under reflux at 70CC for 4 days. The reaction mixture is concentrated and dried briefly under a high vacuum, and the oily residue is purified by means of flash chromatography on silica gel (methylene chloride/methanol 95:5). Further reaction of the resulting oil in a saturated solution of ammonia in methanol at 40-45°C for 24 hours yields the title compound in the form of a white solid. 1H-NMR (CD3OD, ppm): 8.88 (s, 1 H), 7.35-7.25 (m, 2H), 7.05-6.9 (m, 3H), 3.81 (s, 3H), 3.25 (m, 1H), 1.34 (d, J=7Hz, 6H). FAB-MS: 284 [M+H]+.
Example 76: 5-(3-Hvdroxyphenyl)-7-isopropyl-5r-/-pyrrolo[3.2-dlpyrimidin-4-ylamine 0.5 ml of boron tribromide is added in portions over 20 minutes at 0°C to a solution of 0.5 g of 7-isopropyl-5-(3-methoxyphenyl)-5H-pyrrolo[3,2-d]pyrimidin-4-ylamine in 30 ml of methylene chloride. The resulting suspension is further stirred at 0-5°C for 2 hours and then, with ice cooling, 10 ml of water are added. The mixture is allowed to warm to room temperature and is subsequently stirred for a further hour. The reaction mixture is rendered neutral by the addition of 1N sodium hydroxide solution, after 5 minutes is filtered over a glass frit and the residue is washed with a small amount of water and methylene chloride. The crude product is recrystallised from 2:1 ethyl acetate/methanol. White solid: m.p. >273°C (decomp.). Η-NMR (CD3OD, ppm): 8.19 (s, 1 H), 7.4-7.3 (m, 2H), 6.95-6.8 (m, 3H), 3.29 (m, 1 H), 1.36 (d, J=7Hz, 6H). FAB-MS: 269 [M+H]+.
Example 77: 7-Cvclopent-3-enyl-5-(3-methoxyphenyl)-5H-pyrrolo[3.2-d]pyrimidin-4-ylamine 2.1 g of 7-cyclopent-3-enyl-5-(3-methoxyphenyl)-5H-pyrrolo[3,2-d]pyrimidin-4-ol and 10 ml of phosphorus oxychloride are stirred at 120°C for 2 hours. After concentration of the reaction mixture and drying under a high vacuum, 4-chloro-7-cyclopent-3-enyl-5-(3- methoxyphenyl)-5H-pyrrolo[3,2-d]pyrimidine is obtained as the crude product. Η-NMR (CDCI3, ppm): 9.05 (s, 1 H), 7.71 (s, 1H), 7.46 (t, J=8.5Hz, 1 H), 7.13 (dd, J=2.4 and 8.5Hz, 1 H), 6.98 (m, 1 H), 6.93 (d, J=2.4Hz, 1 H), 5.83 (m, 2H), 4.13 (m, 1 H), 3.88 (s, 3H), 3.2-3.0 (m, 2H), 2.55-2.4 (m, 2H). Reaction of 4.03 g of the oil so obtained in 50 ml of liquid ammonia at 130°C for 24 hours yields the title compound in the form of a solid after recrystallisation from 1 :10 methylene chlonde/methanol. M.p. 175.5-176°C. Η-NMR (CDCI3, ppm): 8.49 (br. s, 1 H), 7.43 (m, 1 H), 7.21 (s, 1 H), 7.05-6.9 (m, 3H), 5.83 (m, 2H), 4.88 (br. s, 2H), 3.95-3.8 (m, 4H), 3.0-2.85 (m, 2H), 2.65-2.45 (m, 2H). FAB-MS: 307 [M+H]+.
a) Toluene-4-sulfonic acid cvclopent-3-enyl-methyl ester: 145 g of p-toluenesulfonyl chloride, dissolved in 200 ml of pyridine, are added dropwise, with stirring at 0-5°C, to a solution of 62 g of cyclopent-3-enyl-methanol (prepared according to the method described by A. Hutchison et al. in J. Heterocyclic Chem. (1989), 26, pages 451-452) in 400 ml of pyridine. Stirring is then carried out at 0-5°C for 16 hours, and the reaction mixture is subsequently poured into 600 ml of ice-water and extracted with fert-butyl methyl ether. The combined organic phases are washed with water, 1 N hydrochloric acid and saturated sodium chloride solution, dried over magnesium suifate and concentrated. The crude title compound is obtained in the form of an oil. Η-NMR (CDCI3, ppm): 7.79 (m, 2H), 7.33 (m, 2H), 5.58 (m, 2H), 3.91 (d, J=7.5Hz, 2H), 2.61 (m, 1H), 2.5-2.35 (m, 5H; s at 2.44 ppm), 2.1- 1.95 (m, 2H).
b) Cvclopent-3-enyl-acetonitrile: A mixture of 115.7 g of toluene-4-sulfonic acid cyciopent-3- enyl -methyl ester and 60 g of potassium cyanide in 800 ml of 90 % aqueous ethanol is stirred under reflux for 12 hours. After cooling, the reaction mixture is poured into 500 ml of ice-water and extracted with fert-butyl methyl ether, and the combined organic phases are washed with water and saturated sodium chloride solution and concentrated after drying over magnesium suifate. The title compound is obtained in the form of a colourless liquid after distillation of the crude product. Η-NMR (CDCI3, ppm): 5.64 (m, 2H), 2.65-2.5 (m, 3H), 2.33 (m, 2H), 2.7-2.0 (m, 2H).
c) [(2-Cvclopent-3-enyl-3-nitrilo-propenyl)-(3-methoxyphenvπamino]acetic acid ethyl ester: A mixture of 20.0 g of cyclopent-3-enyl-acetonitrile and sodium ethanolate (prepared from 4.3 g of sodium in 50 ml of absolute ethanol) in 50 ml of toluene is stirred under a carbon monoxide atmosphere at 50°C and 40-50 bar pressure for 10 hours. Subsequent concentration of the reaction mixture and drying under a high vacuum yield the crude sodium 2-cyclopent-3-enyl-3-oxido-acrylonitrile in the form of a beige solid. A solution of 10.7 g of the crude product so obtained in 30 ml of water is adjusted to pH 6 with 1N hydrochloric acid and extracted twice with 100 ml of toluene. The combined organic phases are dried over magnesium suifate, filtered and then added, at 50°C, to a solution of 7.15 g of N-(3-methoxyphenyl)glycine ethyl ester in 50 ml of toluene. After the addition of a catalytic amount of p-toluenesulfonic acid, the reaction mixture is stirred under reflux for 60 minutes and, after cooling, the organic phase is washed with 5 % sodium hydrogen carbonate solution and saturated sodium chloride solution, dried over magnesium suifate and concentrated. The residue obtained after bulb tube distillation (210°C/2 mbar) is purified by means of flash chromatography on silica gel (hexane/ethyl acetate/toluene
3:1 :4). The crude title compound is obtained in the form of an oil. Η-NMR (CDCI3, ppm): 7.25 (m, 1 H), 6.82 (s, 1 H), 6.7-6.55 (m, 3H), 5.70 (m, 2H), 4.73 (s, 2H), 4.28 (q, J=7Hz, 2H), 3.81 (s, 3H), 2.98 (m, 1 H), 2.65-2.3 (m, 4H), 1.32 (t, J=7Hz, 3H). According to an alternative method, the title compound is obtained in a manner analogous to that described above from potassium 2-cyclopent-3-enyl-3-oxido-acrylonitrile:
Potassium 2-cvclopent-3-enyl-3-oxido-acrylonitrile: 5.0 g of cyclopent-3-enyl-acetonitrile are added, with stirring, to a suspension of potassium hydride (obtained from 28.1 g of potassium hydride 20 % dispersion in oil by washing twice with n-hexane) in 1 :1 diethyl ether/hexane, gas evolution taking place. The brown suspension is stirred at room temperature for 1 hour and then 30.4 ml of ethyl formate are added dropwise over 15 minutes. The initially viscous suspension is stirred at room temperature for 12 hours and then filtered over a glass frit, and the coloured precipitate is washed with a small amount of hexane. The filter residue is dried at 50°C under a high vacuum for 3 hours and is then reacted, without further purification, with N-(3-methoxyphenyl)glycιne ethyl ester as described in Example 44d).
d) 3-Amιno-1 -(3-methoxyphenyl)-4-(cyclopent-3-envπ- 7H-pyrrole-2-carboxylιc acid ethyl ester. A solution of 18.2 g of [(2-cyclopent-3-enyl-3-nιtrilo-propenyl)-(3-methoxyphenyl)- amιno]acetιc acid ethyl ester in 40 ml of absolute ethanol is added dropwise at room temperature, with stirring, to a solution of 1.41 g of sodium in 80 ml of ethanol. After 1 hour, the reaction mixture is concentrated, the residue is taken up in ethyl acetate and the organic phase is washed with 5 % sodium carbonate solution and saturated sodium chloride solution, dried over magnesium suifate and concentrated. The crude title compound is obtained in the form of an oil. Η-NMR (CDCI3, ppm): 7.3-7.2 (m, 1 H), 6.85-6.75 (m, 3H), 6.59 (s, 1 H), 5.80 (m, 2H), 4.13 (q, J=7Hz, 2H), 3.81 (s, 3H), 3.35 (m, 1 H), 2.85-2.75 (m, 2H), 2.5-2.35 (m, 2H), 1.27 (t, J=7Hz, 3H).
e) 7-Cyclopent-3-enyl-5-(3-methoxyphenyl)-5H-pyrrolof3.2-dlpyrimidιn-4-ol: A mixture of 15.2 g of 3-amιno-1-(3-methoxyphenyl)-4-(cyclopent-3-enyl)- 7H-pyrrole-2-carboxylic acid ethyl ester and 40 ml of N,N-dimethylformamide dineopentyl acetal in 130 ml of chloroform is stirred at 70°C for 12 hours and then concentrated using a rotary evaporator. The oily residue is purified by means of flash chromatography on silica gel (hexane/ethyl acetate/- toluene, gradient 3:1 :4 to 1:1:4). Reaction of 5.72 g of the resulting product in 150 ml of a saturated ammonia solution in methanol at 40-45°C for 48 hours and subsequent purifi¬ cation by means of flash chromatography on silica gel (methylene chlonde/methanol 98:2) yield the title compound in the form of a solid. Η-NMR (CDCI3, ppm): 7.94 (s, 1 H), 7.36 (m, 1 H), 7.22 (s, 1 H), 7.1-7.0 (m, 2H), 6.93 (m, 1 H), 5.83 (m, 2H), 3.86 (s, 3H), 3.80 (m, 1 H), 3.0-2.85 (m, 2H), 2.6-2.45 (m, 2H).
Example 78: 7-Cvclopent-3-enyl-5-(3-hvdroχyphenyl)-5H-pyrrolo[3.2-d1pyrimidιn-4-ylamιne 0.47 ml of boron tribromide is added dropwise at 0-5cC over 20 minutes, with stirring, to a solution of 0.5 g of 7-cyclopent-3-enyl-5-(3-methoxyphenyl)-5H-pyrrolo[3,2-d]pyrιmidιn-4- ylamme in 30 ml of methylene chloride. After 2 hours, a further 0.25 ml of boron tribromide is added dropwise and the mixture is stirred at 0-5°C for a further 2 hours. Subsequently, 5 ml of water are added to the reaction mixture, which is left to stand at room temperature for 2 hours. The coloured, two-phase mixture is filtered, the filtrate is adjusted to pH 7-8 by the addition of 1 N sodium hydroxide solution and the resulting white precipitate is filtered off The title compound is obtained in the form of a solid after recrystallisation from diethyl ether Η-NMR (CD3OD, ppm): 8.20 (s, 1 H), 7.45-7.35 (m, 2H), 7.0-6.0 (m, 3H), 5.80, (m, 2H), 3.80 (m, 1 H), 2.95-2.80 (m, 2H), 2.6-2.45 (m, 2H). FAB-MS. 293 [M+H]+.
Example 79 3-(4-Amιno-7-cvclohexyl-pyrrolor3.2-d]pyrιmιdιn-5-yl)-phenol
'H-NMR (DMSO-d6, ppm): 9.98 (s, 1 H), 8.20 (s, 1 H), 7.35 (m, 2H), 6.87 (m, 2H), 6.79 (s,
1 H), 5.70 (s (broad), 2H), 2.83 (m, 1 H), 2.08 (m, 2H), 1.78 (m, 3H), 1.59 - 1.20 (m, 5H).
Example 80 7-Cvclopentyl-5-(3-methoxy-phenyl)-5/-/-pyrrolor3,2-d]pyrιmιdιn-4-ylamιne hydrochloride
'H-NMR (DMSO-d6, ppm): 8.54 (s, 1 H), 7.89 (s, 1 H), 7.50 (t, 1 H), 7 10 (m, 3H), 3.83 (s, 3H),
3.29 (m, 1 H), 2 12 (m, 3H), 1 .73 - 1.58 (m, 7H)
Example 81. 2-f(2-f4-[4-Ammo-5-(3-methoxy-phenvO-5H-pyrrolof3.2-d]pyrimidin-7-vn-phen- oxy)-ethyl)-methyl-amιno1-ethanol
A solution of 0.300 g 7-[4-(2-ιodo-ethoxy)-phenyl]-5-(3-methoxy-phenyl)-5H-pyrrolo[2,3-d]- pyrιmιdιn-4-ylamιne in 2 ml methylammo-ethanol is stirred at 80 CC for 1.5 hours. The reaction mixture is concentrated using a rotary evaporator and the residue is triturated with diethyl ether. The title compound is obtained as white solid Η-NMR (DMSO-d6, ppm): 8.30 (s, 1 H), 8 12 (m, 3H), 7.50 (t, 1 H), 7.18 - 6.92 (m, 5H), 5.80 (s (broad), 2H), 4.10 (m, 2H), 3.86 (s, 3H), 3.86 (s, 3H), 3.64 (m, 1 H), 3.49 (m, 2H), 2.97 (m, 1 H), 2.80 (m, 2H), 2.30 (s, 3H) a) 2-(4-f4-Amιno-5-(3-methoxy-phenyl)-5H-pyrrolof3.2-dlpyrιmιdιn-7-yn-phenoxy>-ethanol: The title compound is prepared analogously to Example 14. 1 H-NMR (DMSO-d6, ppm): 8,60 (s, 1 H), 8.40 (s, 1H), 7.85 (d, 2H), 7.53 (t, 1H), 7,28 - 7.01 (m, 5H), 4.05 (t,2H), 3.84 (s, 3H), 3.75 (t, 2H).
b) 7-l4-(2-lodo-ethoxy)-phenyll-5-(3-methoxy-phenyl)-5H-pyrrolor2.3-dlPvnmidin-4-ylamιn: 8.26 g 2-{4-[4-Amιno-5-(3-methoxy-phenyl)-5H-pyrrolo[3,2-d]pyrιmιdιn-7-yl]-phenoxy}- ethanol, 3.42 ml dnsopropyl ethylamine and 10.85 g methyl tnphenoxyphosphonium iodide are added to 270 ml dimethylformamide and stirred at room temperatur for 2 hours. Then, the reaction mixture is poured onto water and extracted with ethylacetate. The organic layer is separated, extracted with saturated sodium chloride solution and dried with sodium suifate The solvent is removed using a rotary evaporator whereby the title compound is obtained as colourless crystalls. The title compound contains 50% 7-[4-(2-chloro-ethoxy)- phenyl]-5-(3-methoxy-phenyl)-5H-pyrrolo[2,3-d]pyrιmιdιn-4-ylamιne. 1 H-NMR (CDCI3, ppm): 8.53 (s, 1 H), 7 99 (m, 2H), 7.58 (s, 1 H), 7.49 (t, 1 H), 7.08 - 6.95 (m, 5H), 4.88 (s, 2H), 4.30 (t, 2H), 3.82 (t, 2H), 3.90 (s, 3H), 3,85 (t, 2H).
Example 82 2-(4-f4-Ammo-5-(3-methoxy-phenyO-5H-pyrrolo[3,2-dlpyrιmιdιn-7-yl]-phenoxy}- ethanol
The compound is prepared analogously to Example 81 'H-NMR (DMSO-d6, ppm): 8.60 (s,
1 H), 8.40 (s, 1 H), 7.85 (d, 2H), 7.55 (t, 1H), 7.28 - 7.05 (m, 5H), 4.20 (t, 2H), 3.85 (s, 3H),
3.75 (t, 2H).
Example 83. 3-(4-Amιno-7-r4-(2-hvdroxy-ethoxy)-phenyl]-pyrrolo[3,2-d1pyrιmιdιn-5-ylr-phenol The compound is prepared analogously to Example 81. Η-NMR (DMSO-d6, ppm): 10.2 (s, 1 H), 8.25 - 8.00 (m, 4H), 7.40 (m, 1 H), 7.0 - 6.80 (m, 5H), 5.92 (s (broad), 2H), 4.00 (t, 2H), 3.7 (t, 2H) Example 84: 2-(4-f4-Amιno-5-(3-methoxy-phenyl)-5H-pyrrolo[3,2-dlPyrιmidin-7-vn-phenoxy)- ethanol hydrochloride
The compound is prepared analogously to Example 81. Η-NMR (DMSO-d6, ppm): 8.59 (s, 1 H), 8.49 (s, 1 H), 7.85 (d, 2H), 7.54 (t, 1 H), 7.25 (m, 1 H), 7.16 (m, 2H), 7.07 (d, 2H), 4.05 (t, 2H), 3.85 (s, 3H), 3.75 (t, 2H).
Example 85: 7-(4-[2-(2-Methoxy-ethylamιno)-ethoxyl-phenyll-5-(3-methoxy-phenyl)-5r-y-pyr- rolof3.2-d1pynmιdιn-4-ylamιne
The compound is prepared analogously to Example 81. M.p.: 145 - 148°C.
Example 86: 5-(3-Methoxy-phenyl)-7-f4-(2-pyrrolιdιn-1-yl-ethoxy)-phenyn-5H-pyrrolof3.2-d]- pyπmιdιn-4-ylamιne
The compound is prepared analogously to Example 81. M.p.:152 - 154°C.
Example 87: 7-(4-[2-(4-Fluoro-benzylamιno)-ethoxy1-phenyl)-5-(3-methoxy-phenyl)-5/-/-pyr- rolo[3,2-dlpyrιmιdιn-4-ylamιne
The compound is prepared analogously to Example 81. M.p/ 137 -140°C.
Example 88- 4-(2-M-f4-Amιno-5-(3-methoxy-phenvO-5H-pyrrolor3,2-d1pyrιmιdιn-7-vH-phen- oxy)-ethylamιno)-cvclohexanol
The compound is prepared analogously to Example 81. M.p 189 - 192°C.
Example 89: 5-(3-Methoxy-phenyl)-7-[4-(2-pιperazιn-1 -yl-ethoxy)-phenyll-5H-pyrrolof3.2-d1- pyrιmιdιn-4-ylamιne dihvdrochlonde
The compound is prepared analogously to Example 81 ' H-NMR (CD3OD, ppm): 8.59 (s, 1 H), 8.09 (s, 1 H), 7.69 (d, 2H), 7.58 (t, 1 H), 7.20 (m, 5H), 4.52 (t, 2H), 3.90 (s, 3H), 3.82 (m, 4H), 3.70 (m, 6H).
Example 90- 7-f4-(2-D[methylamιno-ethoxy)-phenyl1-5-(3-methoxy-phenyl)-5H-pyrrolo[3.2-d1- pyrιmιdιn-4-ylamιne
The compound is prepared analogously to Example 81. M.p.: 170 - 173°C. Example 91 : 1-(2-(4-[4-Amino-5-(3-methoxy-phenyl)-ffH-pyrrolor3.2-d]pyrimidin-7-yl]-phen- oxy>-ethyl)-pyrrolidin-3-ol
The compound is prepared analogously to Example 81. M.p.: 132 -135°C.
Example 92: 2-f4-f4-Amino-5-(3-fluoro-phenyl^5H-pyrrolo[3.2-d]pyrimidin-7-yll-phenoxyr- ethanol hydrochloride
The compound is prepared analogously to Example 81. M.p.: 212 - 216°C.
Example 93: 7-f4-(2-Amino-ethoxy)-phenvn-5-(3-fluoro-phenyl)-5H-pyrrolof3.2-dlpyrimidin-4- ylamine
The compound is prepared analogously to Example 81. M.p.: 166 - 168°C.
Example 94: 5-(3-Fluoro-phenyl)-7-(4-f2-(2-methoxy-ethylamino)-ethoxy]-phenyl)-5H- pyrrolof3.2-d1pyrιmidιn-4-ylamιne
The compound is prepared analogously to Example 81. M.p.: 99.5 - 101.5°C.
Example 95: 5-(3-Fluoro-phenvπ-7-(4-(2-f(2-methoxy-ethyl)-methyl-amino]-ethoxy)-phenyl)-
5H-pyrrolo[3.2-d]pyrιmιdιn-4-ylamιne
The compound is prepared analogously to Example 81. M.p.: 152 -154°C.
Example 96: 2-2-(4-f4-Amιno-5-(3-fluoro-phenyl)-5H-pyrrolo[3,2-d]pyrιmidιn-7-vπ-phenoxy>- ethylaminoVethanol
The compound is prepared analogously to Example 81 . M.p.: 148-151 °C.
Example 97: 5-(3-Fluoro-phenyl)-7-f4-(2-pyrrolιdιn-1 -yl-ethoxy)-phenyl]-5H-pyrrolo[3.2-d1- pyrimidιn-4-ylamιne
The compound is prepared analogously to Example 81. M.p.: 164 - 168°C.
Example 98: 5-(3-Fluoro-phenyπ-7-[4-(2-pιperazιn-1 -yl-ethoxy)-phenvπ-5H-pyrrolor3.2-d1- pyrιmidιn-4-ylamιne
The compound is prepared analogously to Example 81. M.p.: 172 - 175°C. Example 99: 4-f4-Amino-5-(3-hvdroxy-phenyl)-5/-/-pyrrolo[3.2-d1pyrimidin-7-yl]-phenol The compound is prepared analogously to Example 81. Η-NMR (DMSO-d6, ppm): 9.33 (s, 1H), 8.31 (s, 1 H), 8.00 (m, 1 H), 7.40 (m, 1 H), 6.98 (m, 6H), 6.03 (s (broad), 2H).
Example 100: 3-r4-Amino-7-(4-(2-f(2-hvdroxy-ethvO-methyl-amino1-ethoxy)-phenyl)-pyrrolo- f3.2-dlpyrimidin-5-yl1-phenol
The compound is prepared analogously to Example 81. M.p.: 212 - 215°C.
Example 101 : 3-(4-Amino-7-f4-(2-pyrrolidin-1 -yl-ethoxy)-phenyl1-pyrrolof3.2-dlpyrimidin-5-yll- phenol
The compound is prepared analogously to Example 81. M.p.: 212 - 216°C.
Example 102: 1 -(2-(4-[4-Amino-5-(3-hvdroxy-phenyl)-5/-/-pyrrolo[3.2-dlpyrimidin-7-vπ-phen- oxVf-ethyl)-pyrrolidιn-3-ol
The compound is prepared analogously to Example 81. M.p.: 218 - 222°C.
Example 103: 2-(3-f4-Amino-5-(3-methoxy-phenyl)-5H-pyrrolof3,2-d]pyrimidin-7-yl|- phenoxy)-ethanol hydrochloride
The compound is prepared analogously to Example 81. Η-NMR (DMSO-d6, ppm): 8.62 (s, 1 H), 8.50 (s, 1 H), 7.69 - 6.92 (m, 8H), 4.08 (t, 2H), 3.86 (s, 3H), 3.77 (t, 2H).
Example 104: 2-(2-(3-[4-Amino-5-(3-methoxy-phenyl)-5/-/-pyrrolof3.2-d1pyrimidin-7-yl]-phen- oxy|-ethylamιno)-ethanol
The compound is prepared analogously to Example 81. M.p.: 166 - 168CC.
Example 105: 5-(3-Methoxy-phenyl)-7-[3-(2-pyrrolidin-1 -yl-ethoxy)-phenyl)-5/-/-pyrrolofι3.2-d]- pyrimιdin-4-ylamιne
The compound is prepared analogously to Example 81. M.p.: 168 - 170°C.
Example 106: 7-f3-(2-lmidazol-1 -yl-ethoxy)-phenvπ-5-(3-methoxy-phenyl)-5/-/-pyrrolo[3.2-d]- pyrimidin-4-ylamιne
The compound is prepared analogously to Example 81. M.p.: 161 - 164°C. Example 107: 5-(3-Methoxy-phenyl)-7-r3-(2-piperazιn-1 -yl-ethoxy)-phenvπ-5H-pyrrolor3.2-dl- pyrιmιdιn-4-ylamιne hydrochloride
The compound is prepared analogously to Example 81. Η-NMR (CD3OD, ppm): 8.53 (s, 1H), 8.19 (s, 1 H), 7.22 - 7.10 (m, 8H), 4.53 (t, 2H), 3.92 (s, 3H), 3.80 (t, 2H), 3.72 (m, 4H), 3.31 (m, 4H).
Examples A-B. Pharmaceutical Compositions
Example A: Tablets, each comprising 50 mg of active ingredient:
Composition (10 000 tablets) active ingredient 500.0 g lactose 500.0 g potato starch 352.0 g gelatin 8.0 g talc 60.0 g magnesium stearate 10 O g silicon dioxide (highly dispersed) 20.0 g ethanol q s
The active ingredient is mixed with the lactose and 292 g of the potato starch and the mixture is moistened with an ethanohc solution of the gelatin and granulated through a sieve. After drying, the remaining potato starch, the magnesium stearate, the talc and the silicon dioxide are mixed in and the mixture is compressed to form tablets each weighing 145 mg and comprising 50 mg of active ingredient, which may, if desired, be provided with dividing notches for finer adaptation of the dose
Example B- Film-coated tablets, each comprising 100 mg of active ingredient:
Composition (1000 film-coated tablets) active ingredient 100.0 g lactose 100 O g corn starch 70.0 g talc 8.5 g calcium stearate 1.5 g hydroxypropyimethylcellulose 2.36 g shellac 0.64 g water q.s. dichloromethane q.s.
The active ingredient, the lactose and 40 g of the corn starch are mixed, and the mixture is moistened with a paste, prepared from 15 g of the corn starch and water (with heating), and granulated. The granules are dried, and the remaining corn starch, the talc and the calcium stearate are mixed with the granules. The mixture is compressed to form tablets (each weighing 280 mg), which are then coated with a solution of the hydroxypropyimethylcellul¬ ose and the shellac in dichloromethane (final weight of each film-coated tablet: 283 mg).

Claims

What is claimed is:
1. A compound of formula
Figure imgf000049_0001
wherein
Ri is unsubstituted or substituted lower alkyl or hetero-substituted lower alkyl, unsubstituted or substituted cyclo-lower hydrocarbyl or cyclo-lower hydrocarbyl-lower alkyl, unsubstituted or substituted aryl or aryl-lower alkyl, or unsubstituted or substituted hetero-lower cyclyl or hetero-lower cyclyl-lower alkyl;
R2 is hydrogen, lower alkyl or halogen;
R3 is unsubstituted or substituted lower alkyl or hetero-substituted lower alkyl, unsubstituted or substituted cyclo-lower hydrocarbyl or cyclo-lower hydrocarbyl-lower alkyl, unsubstituted or substituted aryl or aryl-lower alkyl, or unsubstituted or substituted hetero-lower cyclyl or hetero-lower cyclyl-lower alkyl; or a salt thereof
2. A compound of formula I according to claim 1 , wherein
RT is unsubstituted or substituted lower alkyl or hetero-substituted lower alkyl, unsubstituted or substituted cyclo-lower hydrocarbyl, unsubstituted or substituted aryl, unsubstituted or substituted aryl-lower alkyl, or unsubstituted or substituted hetero-lower cyclyl;
R2 is hydrogen, lower alkyl or halogen;
R3 is unsubstituted or substituted lower alkyl or hetero-substituted lower alkyl, unsubstituted or substituted cyclo-lower hydrocarbyl, unsubstituted or substituted aryl or aryl-lower alkyl, or unsubstituted or substituted hetero-lower cyclyl; or a salt thereof.
3. A compound of formula I according to claim 1 , wherein
Ri is unsubstituted or substituted lower alkyl or hetero-substituted lower alkyl, unsubstituted or substituted cyclo-lower hydrocarbyl, unsubstituted or substituted aryl, unsubstituted or substituted aryl-lower alkyl, or unsubstituted or substituted hetero-lower cyclyl;
R2 is hydrogen, lower alkyl or halogen;
R3 is unsubstituted or substituted lower alkyl or hetero-substituted lower alkyl, unsubstituted or substituted cyclo-lower hydrocarbyl, or unsubstituted or substituted aryl; or a salt thereof.
4. A compound of formula I according to claim 1 , wherein
Rt is unsubstituted or substituted lower alkyl or hetero-substituted lower alkyl, unsubstituted or substituted cyclo-lower hydrocarbyl, unsubstituted or substituted aryl, unsubstituted or substituted aryl-lower alkyl, or unsubstituted or substituted hetero-lower cyclyl;
R2 is hydrogen;
R3 is unsubstituted or substituted lower alkyl or hetero-substituted lower alkyl, unsubstituted or substituted cyclo-lower hydrocarbyl, or unsubstituted or substituted aryl; or a salt thereof.
5. A compound of formula I according to claim 1 , wherein
Ri is unsubstituted or substituted lower alkyl or hetero-substituted lower alkyl, unsubstituted or substituted cyclo-lower hydrocarbyl, unsubstituted or substituted aryl, unsubstituted or substituted aryl-lower alkyl, or unsubstituted or substituted hetero-lower cyclyl;
R2 is hydrogen;
R3 is unsubstituted or substituted lower alkyl, unsubstituted or substituted cyclo-lower hydrocarbyl, or unsubstituted or substituted aryl; or a pharmaceutically acceptable salt thereof.
6. A compound from the group consisting of 5-isopropyl-7-phenyl-5H-pyrrolo[3,2-d]pyrimidin-4-ylamine; 5-cyclopentyl-7-phenyl-5/-/-pyrrolo[3,2-d]pyrimidin-4-ylamine; 5-(2-methoxy-ethoxymethyl)-7-phenyl-5H-pyrrolo[3,2-d]pyrimidin-4-ylamine; 5-[2-(2-methoxy-ethoxy)-ethyl]-7-phenyl-5H-pyrrolo[3,2-d]pyrimidin-4-ylamine; (4fl/2S) 4-(4-amino-7-phenyl-pyrrolo[3,2-d]pyrimidin-5-yl)-2-carbamoyl-pyrrolidine-1- carboxylic acid fert-butyl ester;
(4fl/2R)-4-(4-amιno-7-phenyl-pyrrolo[3,2-d]pyrimidin-5-yl)-2-carbamoyl-pyrrolidine-1- carboxylic acid rert-butyl ester;
5,7-diphenyl-5/-/-pyrrolo[3,2-d]pyrimidin-4-ylamine;
5-benzyl-7-phenyl-5H-pyrrolo[3,2-d]pyrimidin-4-ylamine;
5-(4-methoxy-phenyl)-7-phenyl-5H-pyrrolo[3,2-d]pyrimidin-4-ylamine;
5-(3-methoxy-phenyl)-7-phenyl-5H-pyrrolo[3,2-d]pyrimidin-4-ylamine;
4-(4-amino-7-phenyl-pyrrolo[3,2-d]pyrimidin-5-yl)-phenol;
3-(4-amino-7-phenyl-pyrrolo[3,2-d]pyrimidin-5-yl)-phenol;
7-[4-(2-benzyloxy-ethoxy)-phenyl]-5-phenyl-5H-pyrrolo[3,2-d]pyrimidin-4-ylamine;
2-[4-(4-amιno-5-phenyl-5H-pyrrolo[3,2-d]pyrimidin-7-yl)-phenoxy]-ethanol;
7-[4-(2-imidazol-1 -yl-ethoxy)-phenyl]-5-phenyl-5H-pyrrolo[3,2-d]pyrimidin-4-ylamine;
7-cyclohexen-1 -enyl-5-(3-methoxy-phenyl)-5H-pyrrolo[3,2-d]pyrimidin-4-ylamine;
7-cyclohexyi-5-(3-methoxy-phenyl)-5/-/-pyrrolo[3,2-d]pyrimidin-4-ylamine;
7-cyclohexen-1 -enyl-5-(3-hydroxy-phenyl)-5H-pyrrolo[3,2-d]pyrimidin-4-ylamine;
5-phenyl)-7-(pyrιdιn-3-yl)-5H-pyrrolo[3,2-d]pyrimidin-4-ylamιne;
4-{4-amιno-7-[4-(2-ιmιdazol-1 -yl-ethoxy)-phenyl]-pyrrolo[3,2-d]pyrimidin-5-yl}-phenol;
3-{4-amιno-7-[4-(2-ιmιdazol-1-yl-ethoxy)-phenyl]-pyrrolo[3,2-d]pyrimidin-5-yl}-phenol;
7-[4-(2-ιmιdazol-1 -yl-ethoxy)-phenyl]-5-(4-methoxy-phenyl)-5H-pyrrolo[3,2-d]pyrimidin-4-yl- amme;
7-[4-(2-ιmιdazol-1 -yl-ethoxy)-phenyl]-5-(3-methoxy-phenyl)-5/-/-pyrrolo[3,2-d]pyrimidin-4-yl- amine;
7-[4-(2-amιno-ethoxy)-phenyl]-5-phenyl-5H-pyrrolo[3,2-d]pyrimidin-4-ylamine;
4-{4-amιno-7-[4-(2-amιno-ethoxy)-phenyl]-pyrrolo[3,2-d]pyrimidin-5-yl}-phenol;
3-{4-amino-7-[4-(2-amιno-ethoxy)-phenyl]-pyrrolo[3,2-d]pyrimidin-5-yl}-phenol;
7-[4-(2-amιno-ethoxy)-phenyl]-5-(4-methoxy-phenyl)-5H-pyrrolo[3,2-d]-pyrimidin-4-ylamine;
7-[4-(2-amιno-ethoxy)-phenyl]-5-(3-methoxy-phenyl)-5H-pyrrolo[3,2-d]-pyrimidin-4-ylamine;
2-{2-[4-(4-amιno-5-ρhenyl-5H-pyrrolo[3,2-d]pyrimιdin7-yl)-phenoxy]-ethylamino}-ethanol;
4-(4-amιno-7-{4-[2-(2-hydroxy-ethylamino)-ethoxy]-phenyl}-pyrrolo[3,2-d]pyrimidin-5-yl- phenol;
3-(4-amino-7-{4-[2-(2-hydroxy-ethylamino)-ethoxy]-phenyl}-pyrrolo[3,2-d]pyrimidin-5-yl- phenol; 2-2-{4-[4-amιno-5-(4-methoxy-phenyl)-5H-pyrrolo[3,2-d]pyπmιdιn-7-yl]-phenoxy}-ethyl- amιno)-ethanol;
2-2-{4-[4-amιno-5-(3-methoxy-phenyl)-5H-pyrrolo[3,2-d]pyrιmιdιn-7-yl]-phenoxy}-ethyl- amino)-ethanol,
7-cyclohexen-1-enyl-5-(3-fluoro-phenyl)-5H-pyrrolo[3,2-d]pyrιmidin-4-ylamine;
7-cyclopentyl-5-phenyl-5H-pyrrolo[3,2-d]pyrιmιdιn-4-ylamιne;
7-cyclopent-1 -enyl-5-phenyl-5H-pyrrolo[3,2-d]pyrιmιdιn-4-ylamιne;
7-cyclopent-3-enyl-5-phenyl-5H-pyrrolo[3,2-d]pyπmιdιn-4-ylamιne;
3-(4-amιno-5-phenyl-5/-/-pyrrolo[3,2-d]pyrιmιdιn-7-yl)-cyclopentanol;
3-(4-amιno-5-phenyl-5H-pyrrolo[3,2-d]pyrιmιdιn-7-yl)-cyclopent-2-enol;
7-(3-methoxy-cyclopentyl-5-phenyl-5H-pyrrolo[3,2-d]pyrιmιdιn-4-ylamιne;
7-(3-methoxy-cyclopent-1-enyl-5-phenyl-5H-pyrrolo[3,2-d]pyrιmιdιn-4-ylamιne;
[3-(4-amιno-5-phenyl-5/-/-pyrrolo[3,2-d]pyπmιdιn-7-yl)-cyclopentyl]-methanol;
7-(3-methoxymethyl-cyclopentyl-5-phenyl-5/-/-pyrrolo[3,2-d]pyrιmιdιn-4-ylamιne;
4-(4-amιno-5-phenyl-5/-/-pyrrolo[3,2-d]pyπmιdιn-7-yl)-cyclopentane-1 ,2-dιol;
7-(3-fluoro-cyclopentyl-5-phenyl-5/-/-pyrrolo[3,2-d]pyrιmιdιn-4-ylamιne;
7-(3,4-dιfluoro-cyclopentyl-5-phenyl-5H-pyrrolo[3,2-d]pyrιmιdιn-4-ylamιne;
3-(4-amιno-5-phenyl-5/-/-pyrrolo[3,2-d]pyrιmιdιn-7-yl)-cyclohexanol,
3-(4-amιno-5-phenyl-5H-pyrrolo[3,2-d]pyrιmιdιn-7-yl)-cyclohex-2-enol;
7-(3-methoxy-cyclohexyl-5-phenyl-5/-/-pyrrolo[3,2-d]pyrιmιdιn-4-ylamιne,
7-(3-methoxymethy!-cyclohexyl-5-phenyl-5/-/-pyrrolo[3,2-d]pyrιmιdιn-4-ylamιne;
4-(4-amιno-5-phenyl-5H-pyrrolo[3,2-d]pyrιmιdιn-7-yl)-cyclohexanol;
7-(4-methoxy-cyclohexyl-5-phenyl-5H-pyrrolo[3,2-d]pyrιmιdιn-4-ylamιne,
7-(4-methoxymethyl-cyclohexyl-5-phenyl-5/-/-pyrrolo[3,2-d]pyrιmιdιn-4-ylamιne;
5-phenyl-7-pyrrolιdιn-3-yl-5H-pyrrolo[3,2-d]pyrιmιdιn-4-ylamιne;
7-(1-methyl-pιpeπdιn-3-yl)-5-phenyl-5H-pyrrolo[3,2-d]pyrιmιdιn-4-ylamιne;
1-[3-(4-amιno-5-ρhenyl-5H-pyrrolo[3,2-d]pyπmιdιn-7-yl)-pyrrolιdιn-1 -yl]-ethanone;
5-phenyl-7-pιperιdιn-3-yl-5H-pyrrolo[3,2-d]pyrιmιdιn-4-ylamιne;
7-(1 -methyl-pιperιdιn-3-yl)-5-phenyl-5H-pyrrolo[3,2-d]pyπmιdιn-4-ylamιne;
1 -[3-(4-amιno-5-phenyl-5/-/-pyrrolo[3,2-d]pyπmιdιn-7-yl)-pιperιdιn-1-yl]-ethanone;
5-phenyl-7-pιpeπdιn-4-yl-5/-/-pyrrolo[3,2-d]pyrιmιdm-4-ylamιne;
7-(1 -methyl-pιperιdιn-4-yl)-5-phenyl-5/-/-pyrrolo[3,2-d]pyπmιdιn-4-ylamιne;
1-[4-(4-amιno-5-phenyl-5H-pyrrolo[3,2-d]pyrιmιdιn-7-yl)-pιpeπdιn-1-yl]-ethanone; 7-(5-methoxymethyl-pyrrolidιn-3-yl)-5-phenyl-5H-pyrrolo[3,2-d]pyrimidin-4-ylamine;
5-phenyl-7-(tetrahydrofuran-3-y!)-5H-pyrrolo[3,2-d]pyrimidin-4-ylamine;
5-phenyl-7-(tetrahydro-thiophen-3-yl)-5H-pyrrolo[3,2-d]pyrimidin-4-ylamine;
7-(1 ,1-dioxo-tetrahydro-thiophen-3-yl)-5-phenyl-5H-pyrrolo[3,2-d]pyrimidin-4-ylamine;
7-cyclopent-1-enyl-5-(3-fluorophenyl)-5H-pyrrolo[3,2-d]pyrimidin-4-ylamine;
7-cyclopent-1 -enyl-5-(3-methoxyphenyl)-5/-/-pyrrolo[3,2-d]pyrimidin-4-ylamine;
7-cyclopent-1 -enyl-5-(4-methoxyphenyl)-5/-/-pyrrolo[3,2-d]pyrιmidin-4-ylamine;
7-cyclopent-1 -enyl-5-(3-hydroxyphenyl)-5H-pyrrolo[3,2-d]pyrιmidin-4-ylamine;
7-cyclopent-1 -enyl-5-(4-hydroxyphenyl)-5/-/-pyrrolo[3,2-d]pyrimidιn-4-ylamine;
7-cyclopentyl-5-(4-hydroxyphenyl)-5/-/-pyrrolo[3,2-d]pyrimιdιn-4-ylamine;
7-cyclopentyl-5-(3-hydroxyphenyl)-5H-pyrrolo[3,2-d]pyrιmιdιn-4-ylamιne;
7-cyclopentyl-5-(3-fluorophenyl)-5H-pyrrolo[3,2-d]pyrιmιdιn-4-ylamine;
7-ιsopropy!-5-(3-methoxyphenyl)-5/-/-pyrrolo[3,2-d]pyπmιdιn-4-ylamine;
5-(3-hydroxyphenyl)-7-ιsopropyl-5H-pyrrolo[3,2-d]pyrimidιn-4-ylamine;
7-cyclopent-3-enyl-5-(3-methoxyphenyl)-5H-pyrrolo[3,2-d]pyrιmιdιn-4-ylamιne;
7-cyclopent-3-enyl-5-(3-hydroxyphenyl)-5H-pyrrolo[3,2-d]pyrιmιdin-4-ylamine;
2-[(2-{4-[4-amιno-5-(3-methoxy-phenyl)-5H-pyrrolo[3,2-d]pyrιmιdιn-7-yl]-phenoxy}-ethyl)- methyl-amιno]-ethanol,
2-{4-[4-amιno-5-(3-methoxy-phenyl)-5/-/-pyrrolo[3,2-d]pyπmιdιn-7-yl]-phenoxy}-ethanol; and
3-{4-amιno-7-[4-(2-hydroxy-ethoxy)-phenyl]-pyrroto[3,2-d]pyπmιdιn-5-yl}-phenol.
7. A pharmaceutical composition comprising a compound according to any one of claims 1 to 6 and at least one pharmaceutically acceptable carrier.
8. A compound according to any one of claims 1 to 6 for use in a method for the therapeutic treatment of the animal or human body.
9. A compound according to any one of claims 1 to 6 for use in the treatment of diseases that are responsive to inhibition of the activity of tyrosine protein kinase pp60c'src.
10. The use of a compound according to any one of claims 1 to 6 in the preparation of a pharmaceutical composition
11. The use of a compound according to any one of claims 1 to 6 in the preparation of a pharmaceutical composition for the treatment of diseases that are responsive to inhibition of the activity of tyrosine protein kinase pp60c'src .
12. A process for the preparation of a compound of formula I according to claim 1 , which comprises a) subjecting a compound of formula II
Figure imgf000054_0001
wherein
Rι, R2 and R3 are as defined in claim 1 , and R4 is lower alkyl, to a ring-closure reaction, with the formation of the pyrimidine ring, to give a compound of formula III
Figure imgf000054_0002
and replacing the oxygen by NH2 by means of an exchange reaction; or
b) subjecting a compound of formula IV
Figure imgf000054_0003
wherein
Ri, R2 and R3 are as defined in claim 1 , to a ring-closure reaction with the formation of the pyrimidine ring, or c) reacting a compound of formula V
Figure imgf000055_0001
wherein R2 and R3 are as defined in claim 1 , with a compound of formula VI
Figure imgf000055_0002
wherein R, is as defined in claim 1 and X is a leaving group;
and, if desired, converting a compound of formula I into a different compound of formula I, and/or, if desired, converting a resulting salt into the free compound or into a different salt, and/or, if desired, converting a resulting free compound of formula I having salt-forming properties into a salt
PCT/EP1997/003115 1996-06-25 1997-06-16 SUBSTITUTED 7-AMINO-PYRROLO[3,2-d]PYRIMIDINES AND THE USE THEREOF WO1997049706A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU31762/97A AU3176297A (en) 1996-06-25 1997-06-16 Substituted 7-amino-pyrrolo{3,2-d}pyrimidines and the use thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CH159196 1996-06-25
CH1591/96 1996-06-25

Publications (1)

Publication Number Publication Date
WO1997049706A1 true WO1997049706A1 (en) 1997-12-31

Family

ID=4214005

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP1997/003115 WO1997049706A1 (en) 1996-06-25 1997-06-16 SUBSTITUTED 7-AMINO-PYRROLO[3,2-d]PYRIMIDINES AND THE USE THEREOF

Country Status (2)

Country Link
AU (1) AU3176297A (en)
WO (1) WO1997049706A1 (en)

Cited By (72)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6187777B1 (en) 1998-02-06 2001-02-13 Amgen Inc. Compounds and methods which modulate feeding behavior and related diseases
WO2001072751A1 (en) * 2000-03-29 2001-10-04 Knoll Gesellschaft Mit Beschraenkter Haftung Pyrrolopyrimidines as tyrosine kinase inhibitors
US6316464B1 (en) 1997-10-20 2001-11-13 Syntex (U.S.A.) Llc P38 MAP kinase inhibitors
WO2002006223A1 (en) * 2000-07-13 2002-01-24 Abbott Laboratories 1,3-disubstituted and 1,3,3-trisubstituted pyrrolidines as histamine-3 receptor ligands and their therapeutic applications
US6515013B2 (en) 2000-07-13 2003-02-04 Abbott Laboratories 1,3-disubstituted and 1,3,3-trisubstituted pyrrolidines as histamine-3 receptor ligands and their therapeutic applications
US6610847B2 (en) 1998-06-19 2003-08-26 Pfizer Inc. Pyrrolo[2,3-d]pyrimidine compounds
US6627754B2 (en) 1999-12-10 2003-09-30 Pfizer Inc. Pyrrolo[2,3-d]pyrimidine compounds
US6635762B1 (en) 1998-06-19 2003-10-21 Pfizer Inc. Monocyclic-7H-pyrrolo[2,3-d]pyrimidine compounds, compositions, and methods of use
US6696567B2 (en) 2000-06-26 2004-02-24 Pfizer Inc. Pyrrolo[2,3-d]pyrimidine compounds
US6713474B2 (en) 1998-09-18 2004-03-30 Abbott Gmbh & Co. Kg Pyrrolopyrimidines as therapeutic agents
WO2005019219A1 (en) * 2003-08-26 2005-03-03 Teijin Pharma Limited Pyrrolopyrimidinone derivative
JP2005526816A (en) * 2002-04-01 2005-09-08 アルミラル プロデスファルマ ソシエダッド アノニマ Azaindolyl piperidine derivatives as antihistamines and antiallergic agents
US7071199B1 (en) * 1999-09-17 2006-07-04 Abbott Gmbh & Cco. Kg Kinase inhibitors as therapeutic agents
US7125875B2 (en) 1999-04-15 2006-10-24 Bristol-Myers Squibb Company Cyclic protein tyrosine kinase inhibitors
US7132426B2 (en) 2003-07-14 2006-11-07 Arena Pharmaceuticals, Inc. Fused-aryl and heteroaryl derivatives as modulators of metabolism and the prophylaxis and treatment of disorders related thereto
US7250420B2 (en) 2002-11-26 2007-07-31 Pfizer Inc. Method of treatment of transplant rejection
US7301023B2 (en) 2001-05-31 2007-11-27 Pfizer Inc. Chiral salt resolution
US7326699B2 (en) 2001-05-14 2008-02-05 Novartis Ag 4-Amino-5-phenyl-7-cyclobutyl-pyrrolo(2,3-d)pyrimidine derivatives
US7427616B2 (en) 2002-08-06 2008-09-23 Astrazeneca Ab Condensed pyridines and pyrimidines with tie2 (TEK) activity
US7557113B2 (en) 2003-08-26 2009-07-07 Teijin Pharma Limited Substituted pyrrolo[3,2-d]pyrimidine derivatives
EP2258371A1 (en) 2001-08-10 2010-12-08 Novartis AG Use of c-Src inhibitors alone or in combination with STI571 for the treatment of leukaemia
WO2011029915A1 (en) 2009-09-10 2011-03-17 Novartis Ag Ether derivatives of bicyclic heteroaryls
EP2308855A1 (en) 2002-03-15 2011-04-13 Novartis AG 2,4-Diaminopyrimidine derivatives
WO2011157793A1 (en) 2010-06-17 2011-12-22 Novartis Ag Piperidinyl substituted 1,3-dihydro-benzoimidazol-2-ylideneamine derivatives
WO2011157787A1 (en) 2010-06-17 2011-12-22 Novartis Ag Biphenyl substituted 1,3-dihydro-benzoimidazol-2-ylideneamine derivatives
EP2409969A1 (en) 2004-06-24 2012-01-25 Novartis AG Pyrimidine urea derivatives as kinase inhibitors
US8133899B2 (en) 2008-08-20 2012-03-13 Pfizer Inc. Pyrrolo[2,3-d]pyrimidine compounds
WO2012120469A1 (en) 2011-03-08 2012-09-13 Novartis Ag Fluorophenyl bicyclic heteroaryl compounds
WO2012122499A2 (en) 2011-03-09 2012-09-13 Pestell Richard G Prostate cancer cell lines, gene signatures and uses thereof
WO2013052417A1 (en) * 2011-10-03 2013-04-11 The University Of North Carolina At Chapel Hill Pyrrolopyrimidine compounds for the treatment of cancer
JP2013518098A (en) * 2010-01-29 2013-05-20 ハンミ ファーム. シーオー., エルティーディー. Bicyclic heteroaryl derivatives having protein kinase inhibitory activity
CN103328482A (en) * 2011-01-03 2013-09-25 韩美药品株式会社 New bicyclic compound for modulating G protein-coupled receptors
JP2013253089A (en) * 2007-03-28 2013-12-19 Pharmacyclics Inc Inhibitor of bruton's tyrosine kinase
JP2014510113A (en) * 2011-04-04 2014-04-24 ファーマサイエンス・インコーポレイテッド Protein kinase inhibitor
JP2014510114A (en) * 2011-04-04 2014-04-24 ファーマサイエンス・インコーポレイテッド Protein kinase inhibitor
WO2014081645A1 (en) 2012-11-20 2014-05-30 Glaxosmithkline Llc Novel compounds
WO2014081643A1 (en) * 2012-11-20 2014-05-30 Glaxosmithkline Llc Novel compounds
WO2014081644A1 (en) * 2012-11-20 2014-05-30 Glaxosmithkline Llc Novel compounds
US8933083B2 (en) 2003-01-14 2015-01-13 Arena Pharmaceuticals, Inc. 1,2,3-trisubstituted aryl and heteroaryl derivatives as modulators of metabolism and the prophylaxis and treatment of disorders related thereto such as diabetes and hyperglycemia
US8940750B2 (en) 2007-03-28 2015-01-27 Pharmacyclics, Inc. Inhibitors of bruton's tyrosine kinase
WO2015058084A1 (en) * 2013-10-18 2015-04-23 Medivation Technologies, Inc. Heterocyclic compounds and methods of use
WO2015074135A1 (en) * 2013-11-19 2015-05-28 Pharmascience Inc. Protein kinase inhibitors
US9096604B2 (en) 2012-11-15 2015-08-04 Pharmacyclics, Inc. Pyrrolopyrimidine compounds as kinase inhibitors
WO2016016894A1 (en) 2014-07-30 2016-02-04 Yeda Research And Development Co. Ltd. Media for culturing pluripotent stem cells
US9273051B2 (en) 2011-12-30 2016-03-01 Pharmacyclics Llc Pyrazolo[3,4-d]pyrimidine and pyrrolo[2,3-d]pyrimidine compounds as kinase inhibitors
US9296753B2 (en) 2012-06-04 2016-03-29 Pharmacyclics Llc Crystalline forms of a Bruton's tyrosine kinase inhibitor
US9453836B2 (en) 2012-05-14 2016-09-27 Richard G. Pestell Use of modulators of CCR5 in the treatment of cancer and cancer metastasis
US9555030B2 (en) 2014-04-11 2017-01-31 The University Of North Carolina At Chapel Hill Therapeutic uses of selected pyrazolopyrimidine compounds with anti-Mer tyrosine kinase activity
US9555036B2 (en) 2012-08-24 2017-01-31 Glaxosmithkline Llc Pyrazolopyrimidine compounds
US9562047B2 (en) 2012-10-17 2017-02-07 The University Of North Carolina At Chapel Hill Pyrazolopyrimidine compounds for the treatment of cancer
US9567326B2 (en) 2012-05-22 2017-02-14 The University Of North Carolina At Chapel Hill Pyrimidine compounds for the treatment of cancer
US9624224B2 (en) 2013-09-30 2017-04-18 Pharmacyclics Llc Inhibitors of Bruton's tyrosine kinase
US9744172B2 (en) 2010-05-19 2017-08-29 The University Of North Carolina At Chapel Hill Pyrazolopyrimidine compounds for the treatment of cancer
US9771330B2 (en) 2012-11-27 2017-09-26 The University Of North Carolina At Chapel Hill Pyrimidine compounds for the treatment of cancer
US9801881B2 (en) 2010-06-03 2017-10-31 Pharmacyclics Llc Use of inhibitors of bruton's tyrosine kinase (BTK)
US9862722B2 (en) 2011-07-13 2018-01-09 Pharmacyclics Llc Inhibitors of Bruton's tyrosine kinase
US9877968B2 (en) 2008-08-11 2018-01-30 Glaxosmithkline Llc 6-amino-purin-8-one compounds
US20180028537A1 (en) 2014-08-07 2018-02-01 Pharmacyclics Llc Novel Formulations of a Bruton's Tyrosine Kinase Inhibitor
US9885086B2 (en) 2014-03-20 2018-02-06 Pharmacyclics Llc Phospholipase C gamma 2 and resistance associated mutations
US10010507B1 (en) 2015-03-03 2018-07-03 Pharmacyclics Llc Pharmaceutical formulations of a bruton's tyrosine kinase inhibitor
US10112946B2 (en) 2011-07-22 2018-10-30 Glaxosmithkline Llc Composition
WO2019119486A1 (en) 2017-12-21 2019-06-27 中国科学院合肥物质科学研究院 Class of pyrimidine derivative kinase inhibitors
WO2020115481A1 (en) * 2018-12-05 2020-06-11 Imperial College Of Science, Technology And Medicine Map4k4 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
WO2020152686A1 (en) 2019-01-23 2020-07-30 Yeda Research And Development Co. Ltd. Culture media for pluripotent stem cells
JP2020536937A (en) * 2017-10-13 2020-12-17 インペリアル カレッジ オブ サイエンス,テクノロジー アンド メディスン MAP4K4 inhibitor
US10894787B2 (en) 2010-09-22 2021-01-19 Arena Pharmaceuticals, Inc. Modulators of the GPR119 receptor and the treatment of disorders related thereto
US10954567B2 (en) 2012-07-24 2021-03-23 Pharmacyclics Llc Mutations associated with resistance to inhibitors of Bruton's Tyrosine Kinase (BTK)
US11007175B2 (en) 2015-01-06 2021-05-18 Arena Pharmaceuticals, Inc. Methods of treating conditions related to the S1P1 receptor
WO2021097256A1 (en) 2019-11-14 2021-05-20 Cohbar, Inc. Cxcr4 antagonist peptides
US11034669B2 (en) 2018-11-30 2021-06-15 Nuvation Bio Inc. Pyrrole and pyrazole compounds and methods of use thereof
US11884626B2 (en) 2015-06-22 2024-01-30 Arena Pharmaceuticals, Inc. Crystalline L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclo-penta [b]indol-3-yl)acetic acid(Compound1) for use in S1P1 receptor-associated disorders

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996010028A1 (en) * 1994-09-29 1996-04-04 Novartis Ag PYRROLO[2,3-d]PYRIMIDINES AND THEIR USE
WO1996040142A1 (en) * 1995-06-07 1996-12-19 Pfizer Inc. Heterocyclic ring-fused pyrimidine derivatives

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996010028A1 (en) * 1994-09-29 1996-04-04 Novartis Ag PYRROLO[2,3-d]PYRIMIDINES AND THEIR USE
WO1996040142A1 (en) * 1995-06-07 1996-12-19 Pfizer Inc. Heterocyclic ring-fused pyrimidine derivatives

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
CHEMICAL ABSTRACTS, vol. 98, no. 13, 1983, Columbus, Ohio, US; abstract no. 107247m, O.S. SIZOVA ET AL.: "Pyrrolo[3,2-d]pyrimidine derivatives. IV. Synthesis, antibacterial and antitumor activity of 2,4,7-substituted pyrrolo[3,2-d]pyrimidines" page 598; XP002041395 *
KHIM.-FARM. ZH., vol. 16, no. 11, 1982, pages 1338 - 43 *

Cited By (163)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6316464B1 (en) 1997-10-20 2001-11-13 Syntex (U.S.A.) Llc P38 MAP kinase inhibitors
US6479507B2 (en) 1997-10-20 2002-11-12 Syntex (U.S.A.) Llc p38 MAP kinase inhibitors
US6187777B1 (en) 1998-02-06 2001-02-13 Amgen Inc. Compounds and methods which modulate feeding behavior and related diseases
US6583154B1 (en) 1998-02-06 2003-06-24 Amgen Inc. Compounds and methods which modulate feeding behavior and related diseases
US7687507B2 (en) 1998-06-19 2010-03-30 Pfizer Inc. Pyrrolo[2,3-d]pyrimidine compounds
US6610847B2 (en) 1998-06-19 2003-08-26 Pfizer Inc. Pyrrolo[2,3-d]pyrimidine compounds
US7569569B2 (en) 1998-06-19 2009-08-04 Pfizer Inc. Pyrrolo[2,3-d]pyrimidine compounds
US6635762B1 (en) 1998-06-19 2003-10-21 Pfizer Inc. Monocyclic-7H-pyrrolo[2,3-d]pyrimidine compounds, compositions, and methods of use
US6890929B2 (en) 1998-06-19 2005-05-10 Pfizer, Inc. Pyrrolo [2,3-D] pyrimidine compounds
US6713474B2 (en) 1998-09-18 2004-03-30 Abbott Gmbh & Co. Kg Pyrrolopyrimidines as therapeutic agents
US7125875B2 (en) 1999-04-15 2006-10-24 Bristol-Myers Squibb Company Cyclic protein tyrosine kinase inhibitors
US7071199B1 (en) * 1999-09-17 2006-07-04 Abbott Gmbh & Cco. Kg Kinase inhibitors as therapeutic agents
US7842699B2 (en) 1999-12-10 2010-11-30 Pfizer Inc. Pyrrolo[2,3-D]pyrimidine compounds
US7601727B2 (en) 1999-12-10 2009-10-13 Pfizer, Inc. Pyrrolo[2,3-d]pyrimidine compounds
USRE41783E1 (en) 1999-12-10 2010-09-28 Pfizer Inc. Pyrrolo[2,3-D]pyrimidine compounds
US6627754B2 (en) 1999-12-10 2003-09-30 Pfizer Inc. Pyrrolo[2,3-d]pyrimidine compounds
US6956041B2 (en) 1999-12-10 2005-10-18 Pfizer Inc. Pyrrolo[2,3-d]pyrimidine compounds
US7265221B2 (en) 1999-12-10 2007-09-04 Pfizer, Inc. Pyrrolo[2,3-d]pyrimidine compounds
US7091208B2 (en) 1999-12-10 2006-08-15 Pfizer Inc. Pyrrolo[2,3-D]pyrimidine compounds
WO2001072751A1 (en) * 2000-03-29 2001-10-04 Knoll Gesellschaft Mit Beschraenkter Haftung Pyrrolopyrimidines as tyrosine kinase inhibitors
US7192963B2 (en) 2000-06-26 2007-03-20 Pfizer Inc Pyrrolo[2,3-d]pyrimidine compounds
US6696567B2 (en) 2000-06-26 2004-02-24 Pfizer Inc. Pyrrolo[2,3-d]pyrimidine compounds
US6962993B2 (en) 2000-06-26 2005-11-08 Pfizer Inc. Pyrrolo[2,3-d]pyrimidine compounds
US6515013B2 (en) 2000-07-13 2003-02-04 Abbott Laboratories 1,3-disubstituted and 1,3,3-trisubstituted pyrrolidines as histamine-3 receptor ligands and their therapeutic applications
WO2002006223A1 (en) * 2000-07-13 2002-01-24 Abbott Laboratories 1,3-disubstituted and 1,3,3-trisubstituted pyrrolidines as histamine-3 receptor ligands and their therapeutic applications
US7326699B2 (en) 2001-05-14 2008-02-05 Novartis Ag 4-Amino-5-phenyl-7-cyclobutyl-pyrrolo(2,3-d)pyrimidine derivatives
US7301023B2 (en) 2001-05-31 2007-11-27 Pfizer Inc. Chiral salt resolution
US7432370B2 (en) 2001-05-31 2008-10-07 Pfizer Inc. Chiral salt resolution
US8119649B2 (en) 2001-08-10 2012-02-21 Novartis Ag Use of c-Src inhibitors alone or in combination with STI571 for the treatment of leukaemia
EP2258371A1 (en) 2001-08-10 2010-12-08 Novartis AG Use of c-Src inhibitors alone or in combination with STI571 for the treatment of leukaemia
US8268837B2 (en) 2001-08-10 2012-09-18 Novartis Ag Use of c-Src inhibitors alone or in combination with STI571 for the treatment of leukaemia
EP2308855A1 (en) 2002-03-15 2011-04-13 Novartis AG 2,4-Diaminopyrimidine derivatives
JP2005526816A (en) * 2002-04-01 2005-09-08 アルミラル プロデスファルマ ソシエダッド アノニマ Azaindolyl piperidine derivatives as antihistamines and antiallergic agents
US7427616B2 (en) 2002-08-06 2008-09-23 Astrazeneca Ab Condensed pyridines and pyrimidines with tie2 (TEK) activity
US7250420B2 (en) 2002-11-26 2007-07-31 Pfizer Inc. Method of treatment of transplant rejection
US8933083B2 (en) 2003-01-14 2015-01-13 Arena Pharmaceuticals, Inc. 1,2,3-trisubstituted aryl and heteroaryl derivatives as modulators of metabolism and the prophylaxis and treatment of disorders related thereto such as diabetes and hyperglycemia
US7625906B2 (en) 2003-07-14 2009-12-01 Arena Pharmaceuticals, Inc. Fused-aryl and heteroaryl derivatives as modulators of metabolism and the prophylaxis and treatment of disorders related thereto
EP2287165A2 (en) 2003-07-14 2011-02-23 Arena Pharmaceuticals, Inc. Fused-aryl and heteroaryl derivatives as modulators of metabolism and the prophylaxis and treatment of disorders related thereto
EP2287166A2 (en) 2003-07-14 2011-02-23 Arena Pharmaceuticals, Inc. Fused-aryl and heteroaryl derivatives as modulators of metabolism and the prophylaxis and treatment of disorders related thereto
EP2292620A2 (en) 2003-07-14 2011-03-09 Arena Pharmaceuticals, Inc. Fused-aryl and heteroaryl derivatives as modulators of metabolism and the prohylaxis and treatment of disorders related thereto
US7132426B2 (en) 2003-07-14 2006-11-07 Arena Pharmaceuticals, Inc. Fused-aryl and heteroaryl derivatives as modulators of metabolism and the prophylaxis and treatment of disorders related thereto
US8410119B2 (en) 2003-07-14 2013-04-02 Arena Pharmaceuticals, Inc. Fused-aryl and heteroaryl derivatives as modulators of metabolism and the prophylaxis and treatment of disorders related thereto
WO2005019219A1 (en) * 2003-08-26 2005-03-03 Teijin Pharma Limited Pyrrolopyrimidinone derivative
AU2004266975B2 (en) * 2003-08-26 2009-11-12 Teijin Limited Pyrrolopyrimidinone derivative
WO2005019218A1 (en) * 2003-08-26 2005-03-03 Teijin Pharma Limited Pyrrolopyrimidinethione derivative
US7557113B2 (en) 2003-08-26 2009-07-07 Teijin Pharma Limited Substituted pyrrolo[3,2-d]pyrimidine derivatives
EP2409969A1 (en) 2004-06-24 2012-01-25 Novartis AG Pyrimidine urea derivatives as kinase inhibitors
EP2418205A1 (en) 2004-06-24 2012-02-15 Novartis AG Pyrimidine urea derivatives as kinase inhibitors
US9139591B2 (en) 2007-03-28 2015-09-22 Pharmacyclics Llc Inhibitors of bruton's tyrosine kinase
US9556182B2 (en) 2007-03-28 2017-01-31 Pharmacylics LLC Inhibitors of Bruton's tyrosine kinase
US9079908B2 (en) 2007-03-28 2015-07-14 Pharmacyclics, Inc. Inhibitors of Bruton'S tyrosine kinase
US8940750B2 (en) 2007-03-28 2015-01-27 Pharmacyclics, Inc. Inhibitors of bruton's tyrosine kinase
JP2013253089A (en) * 2007-03-28 2013-12-19 Pharmacyclics Inc Inhibitor of bruton's tyrosine kinase
US10117873B2 (en) 2008-08-11 2018-11-06 Glaxosmithkline Llc 6-amino-purin-8-one compounds
US9877968B2 (en) 2008-08-11 2018-01-30 Glaxosmithkline Llc 6-amino-purin-8-one compounds
US9161939B2 (en) 2008-08-20 2015-10-20 Zoetis Services Llc Pyrrolo[2,3-d]pyrimidine compounds
US9271981B2 (en) 2008-08-20 2016-03-01 Zoetis Services Llc Pyrrolo[2,3-D]pyrimidine compounds
US8133899B2 (en) 2008-08-20 2012-03-13 Pfizer Inc. Pyrrolo[2,3-d]pyrimidine compounds
US8987283B2 (en) 2008-08-20 2015-03-24 Zoetis Llc Pyrrolo[2,3-D]pyrimidine compounds
WO2011029915A1 (en) 2009-09-10 2011-03-17 Novartis Ag Ether derivatives of bicyclic heteroaryls
JP2013518098A (en) * 2010-01-29 2013-05-20 ハンミ ファーム. シーオー., エルティーディー. Bicyclic heteroaryl derivatives having protein kinase inhibitory activity
US10112954B2 (en) 2010-01-29 2018-10-30 Hanmi Pharm. Co., Ltd. Bicyclic heteroaryl derivatives having inhibitory activity for protein kinase
US9744172B2 (en) 2010-05-19 2017-08-29 The University Of North Carolina At Chapel Hill Pyrazolopyrimidine compounds for the treatment of cancer
US10751342B2 (en) 2010-06-03 2020-08-25 Pharmacyclics Llc Use of inhibitors of Bruton's tyrosine kinase (Btk)
US10004745B2 (en) 2010-06-03 2018-06-26 Pharmacyclics Llc Use of inhibitors of Bruton'S tyrosine kinase (Btk)
US9801881B2 (en) 2010-06-03 2017-10-31 Pharmacyclics Llc Use of inhibitors of bruton's tyrosine kinase (BTK)
US9814721B2 (en) 2010-06-03 2017-11-14 Pharmacyclics Llc Use of inhibitors of bruton'S tyrosine kinase (BTK)
US10004746B2 (en) 2010-06-03 2018-06-26 Pharmacyclics Llc Use of inhibitors of Bruton's tyrosine kinase (Btk)
US10016435B2 (en) 2010-06-03 2018-07-10 Pharmacyclics Llc Use of inhibitors of Bruton's tyrosine kinase (Btk)
US10478439B2 (en) 2010-06-03 2019-11-19 Pharmacyclics Llc Use of inhibitors of bruton's tyrosine kinase (Btk)
US10653696B2 (en) 2010-06-03 2020-05-19 Pharmacyclics Llc Use of inhibitors of bruton's tyrosine kinase (BTK)
US11672803B2 (en) 2010-06-03 2023-06-13 Pharmacyclics Llc Use of inhibitors of Brutons tyrosine kinase (Btk)
WO2011157787A1 (en) 2010-06-17 2011-12-22 Novartis Ag Biphenyl substituted 1,3-dihydro-benzoimidazol-2-ylideneamine derivatives
WO2011157793A1 (en) 2010-06-17 2011-12-22 Novartis Ag Piperidinyl substituted 1,3-dihydro-benzoimidazol-2-ylideneamine derivatives
US10894787B2 (en) 2010-09-22 2021-01-19 Arena Pharmaceuticals, Inc. Modulators of the GPR119 receptor and the treatment of disorders related thereto
EP2661438A2 (en) * 2011-01-03 2013-11-13 Hanmi Pharm. Co., Ltd. New bicyclic compound for modulating g protein-coupled receptors
CN103328482A (en) * 2011-01-03 2013-09-25 韩美药品株式会社 New bicyclic compound for modulating G protein-coupled receptors
US8853213B2 (en) 2011-01-03 2014-10-07 Hanmi Pharm. Co., Ltd Bicyclic compound for modulating G protein-coupled receptors
EP2661438A4 (en) * 2011-01-03 2014-06-11 Hanmi Pharm Ind Co Ltd New bicyclic compound for modulating g protein-coupled receptors
WO2012120469A1 (en) 2011-03-08 2012-09-13 Novartis Ag Fluorophenyl bicyclic heteroaryl compounds
US10952415B2 (en) 2011-03-09 2021-03-23 Richard G. Pestell Prostate cancer cell lines, gene signatures and uses thereof
EP3597761A1 (en) 2011-03-09 2020-01-22 Richard G. Pestell Prostate cancer cell lines, gene signatures and uses thereof
WO2012122499A2 (en) 2011-03-09 2012-09-13 Pestell Richard G Prostate cancer cell lines, gene signatures and uses thereof
JP2014510114A (en) * 2011-04-04 2014-04-24 ファーマサイエンス・インコーポレイテッド Protein kinase inhibitor
JP2014510113A (en) * 2011-04-04 2014-04-24 ファーマサイエンス・インコーポレイテッド Protein kinase inhibitor
US9862722B2 (en) 2011-07-13 2018-01-09 Pharmacyclics Llc Inhibitors of Bruton's tyrosine kinase
US10112946B2 (en) 2011-07-22 2018-10-30 Glaxosmithkline Llc Composition
US9273056B2 (en) 2011-10-03 2016-03-01 The University Of North Carolina At Chapel Hill Pyrrolopyrimidine compounds for the treatment of cancer
WO2013052417A1 (en) * 2011-10-03 2013-04-11 The University Of North Carolina At Chapel Hill Pyrrolopyrimidine compounds for the treatment of cancer
JP2014532060A (en) * 2011-10-03 2014-12-04 ザ・ユニヴァーシティ・オヴ・ノース・キャロライナ・アト・チャペル・ヒル Pyrrolopyrimidine compounds for the treatment of cancer
RU2631655C2 (en) * 2011-10-03 2017-09-26 Зэ Юниверсити Оф Норд Каролина Эт Чапель Хилл Pyrrolopyrimidine compounds for malignant tumour treatment
US10179133B2 (en) 2011-10-03 2019-01-15 The University Of North Carolina At Chapel Hill Pyrrolopyrimidine compounds for the treatment of cancer
JP2017101043A (en) * 2011-10-03 2017-06-08 ザ・ユニヴァーシティ・オヴ・ノース・キャロライナ・アト・チャペル・ヒル Pyrrolopyrimidine compound for treating cancer
US9273051B2 (en) 2011-12-30 2016-03-01 Pharmacyclics Llc Pyrazolo[3,4-d]pyrimidine and pyrrolo[2,3-d]pyrimidine compounds as kinase inhibitors
US9546172B2 (en) 2011-12-30 2017-01-17 Pharmacyclics Llc Pyrazolo[3,4-d]pyrimidine and pyrazolo[2,3-d]pyrimidine compounds as kinase inhibitors
US11633397B2 (en) 2012-05-14 2023-04-25 Richard G. Pestell Use of modulators of CCR5 in the treatment of cancer and cancer metastasis
US9453836B2 (en) 2012-05-14 2016-09-27 Richard G. Pestell Use of modulators of CCR5 in the treatment of cancer and cancer metastasis
US9567326B2 (en) 2012-05-22 2017-02-14 The University Of North Carolina At Chapel Hill Pyrimidine compounds for the treatment of cancer
US9540382B2 (en) 2012-06-04 2017-01-10 Pharmacyclics Llc Crystalline forms of a Bruton's tyrosine kinase inhibitor
US9713617B2 (en) 2012-06-04 2017-07-25 Pharmacyclics Llc Crystalline forms of a Bruton's tyrosine kinase inhibitor
US9296753B2 (en) 2012-06-04 2016-03-29 Pharmacyclics Llc Crystalline forms of a Bruton's tyrosine kinase inhibitor
US10752634B2 (en) 2012-06-04 2020-08-25 Pharmacyclics Llc Crystalline forms of a brutons tyrosine kinase inhibitor
US10294232B2 (en) 2012-06-04 2019-05-21 Pharmacyclics Llc Crystalline forms of a Bruton's tyrosine kinase inhibitor
US10294231B2 (en) 2012-06-04 2019-05-21 Pharmacyclics Llc Crystalline forms of a Bruton's tyrosine kinase inhibitor
US10266540B2 (en) 2012-06-04 2019-04-23 Pharmacyclics Llc Crystalline forms of a Bruton's tyrosine kinase inhibitor
US9828383B1 (en) 2012-06-04 2017-11-28 Pharmacyclic s LLC Crystalline forms of a bruton's tyrosine kinase inhibitor
US10961251B1 (en) 2012-06-04 2021-03-30 Pharmacyclics Llc Crystalline forms of a Bruton's tyrosine kinase inhibitor
US10125140B1 (en) 2012-06-04 2018-11-13 Pharmacyclics Llc Crystalline forms of a bruton's tyrosine kinase inhibitor
US9725455B1 (en) 2012-06-04 2017-08-08 Pharmacyclics Llc Crystalline forms of a bruton's tyrosine kinase inhibitor
US10106548B2 (en) 2012-06-04 2018-10-23 Pharmacyclics Llc Crystalline forms of a Bruton's tyrosine kinase inhibitor
US10065968B2 (en) 2012-06-04 2018-09-04 Pharmacyclics Llc Crystalline forms of a bruton's tyrosine kinase inhibitor
US10954567B2 (en) 2012-07-24 2021-03-23 Pharmacyclics Llc Mutations associated with resistance to inhibitors of Bruton's Tyrosine Kinase (BTK)
US9555036B2 (en) 2012-08-24 2017-01-31 Glaxosmithkline Llc Pyrazolopyrimidine compounds
US10022442B2 (en) 2012-08-24 2018-07-17 Glaxosmithkline Llc Pyrazolopyrimidine compounds
US9662336B2 (en) 2012-08-24 2017-05-30 Glaxosmithkline Llc Pyrazolopyrimidine compounds
US9562047B2 (en) 2012-10-17 2017-02-07 The University Of North Carolina At Chapel Hill Pyrazolopyrimidine compounds for the treatment of cancer
US9096604B2 (en) 2012-11-15 2015-08-04 Pharmacyclics, Inc. Pyrrolopyrimidine compounds as kinase inhibitors
US9540385B2 (en) 2012-11-15 2017-01-10 Pharmacyclics Llc Pyrrolopyrimidine compounds as kinase inhibitors
AU2013348216B2 (en) * 2012-11-20 2016-10-13 Glaxosmithkline Llc Novel compounds
CN104780922A (en) * 2012-11-20 2015-07-15 葛兰素史克有限责任公司 Novel compounds
RU2643371C2 (en) * 2012-11-20 2018-02-01 ГЛАКСОСМИТКЛАЙН ЭлЭлСи New compounds
WO2014081645A1 (en) 2012-11-20 2014-05-30 Glaxosmithkline Llc Novel compounds
US9907847B2 (en) 2012-11-20 2018-03-06 Glaxosmithkline Llc Pyrrolopyrimidines as therapeutic agents for the treatment of diseases
US9428512B2 (en) 2012-11-20 2016-08-30 Glaxosmithkline Llc Compounds
US9550785B2 (en) 2012-11-20 2017-01-24 Glaxosmithkline Llc Pyrrolopyrimidines as therapeutic agents for the treatment of diseases
EP2922549A4 (en) * 2012-11-20 2016-04-06 Glaxosmithkline Llc Novel compounds
WO2014081643A1 (en) * 2012-11-20 2014-05-30 Glaxosmithkline Llc Novel compounds
CN104780922B (en) * 2012-11-20 2016-09-07 葛兰素史克有限责任公司 Interferon-induced immunomodulator compounds
CN104780924B (en) * 2012-11-20 2016-09-14 葛兰素史克有限责任公司 Interferon-induced immunomodulator compounds
RU2640200C2 (en) * 2012-11-20 2017-12-27 ГЛАКСОСМИТКЛАЙН ЭлЭлСи New compounds
EA028480B1 (en) * 2012-11-20 2017-11-30 ГЛАКСОСМИТКЛАЙН ЭлЭлСи Novel compounds
WO2014081644A1 (en) * 2012-11-20 2014-05-30 Glaxosmithkline Llc Novel compounds
JP2016500067A (en) * 2012-11-20 2016-01-07 グラクソスミスクライン・リミテッド・ライアビリティ・カンパニーGlaxoSmithKline LLC New compounds
JP2016500066A (en) * 2012-11-20 2016-01-07 グラクソスミスクライン・リミテッド・ライアビリティ・カンパニーGlaxoSmithKline LLC New compounds
AU2013348218B2 (en) * 2012-11-20 2016-10-13 Glaxosmithkline Llc Novel compounds
US9540383B2 (en) 2012-11-20 2017-01-10 Glaxosmithkline Llc Pyrrolopyrimidines as therapeutic agents for the treatment of diseases
CN104780924A (en) * 2012-11-20 2015-07-15 葛兰素史克有限责任公司 Novel compounds
US9771330B2 (en) 2012-11-27 2017-09-26 The University Of North Carolina At Chapel Hill Pyrimidine compounds for the treatment of cancer
US9624224B2 (en) 2013-09-30 2017-04-18 Pharmacyclics Llc Inhibitors of Bruton's tyrosine kinase
WO2015058084A1 (en) * 2013-10-18 2015-04-23 Medivation Technologies, Inc. Heterocyclic compounds and methods of use
WO2015074135A1 (en) * 2013-11-19 2015-05-28 Pharmascience Inc. Protein kinase inhibitors
US9885086B2 (en) 2014-03-20 2018-02-06 Pharmacyclics Llc Phospholipase C gamma 2 and resistance associated mutations
US9603850B2 (en) 2014-04-11 2017-03-28 The University Of North Carolina At Chapel Hill MerTK-specific pyrazolopyrimidine compounds
US10004755B2 (en) 2014-04-11 2018-06-26 The University Of North Carolina At Chapel Hill Therapeutic uses of selected pyrrolopyrimidine compounds with anti-mer tyrosine kinase activity
US9649309B2 (en) 2014-04-11 2017-05-16 The University Of North Carolina At Chapel Hill Therapeutic uses of selected pyrimidine compounds with anti-Mer tyrosine kinase activity
US9555030B2 (en) 2014-04-11 2017-01-31 The University Of North Carolina At Chapel Hill Therapeutic uses of selected pyrazolopyrimidine compounds with anti-Mer tyrosine kinase activity
US9555031B2 (en) 2014-04-11 2017-01-31 The University Of North Carolina At Chapel Hill Therapeutic uses of selected pyrrolopyrimidine compounds with anti-mer tyrosine kinase activity
WO2016016894A1 (en) 2014-07-30 2016-02-04 Yeda Research And Development Co. Ltd. Media for culturing pluripotent stem cells
US20180028537A1 (en) 2014-08-07 2018-02-01 Pharmacyclics Llc Novel Formulations of a Bruton's Tyrosine Kinase Inhibitor
US11007175B2 (en) 2015-01-06 2021-05-18 Arena Pharmaceuticals, Inc. Methods of treating conditions related to the S1P1 receptor
US10828259B2 (en) 2015-03-03 2020-11-10 Pharmacyclics Llc Pharmaceutical formulations of a Bruton's tyrosine kinase inhibitor
US10010507B1 (en) 2015-03-03 2018-07-03 Pharmacyclics Llc Pharmaceutical formulations of a bruton's tyrosine kinase inhibitor
US10213386B2 (en) 2015-03-03 2019-02-26 Pharmacyclics Llc Pharmaceutical formulations of a Bruton's tyrosine kinase inhibitor
US11884626B2 (en) 2015-06-22 2024-01-30 Arena Pharmaceuticals, Inc. Crystalline L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclo-penta [b]indol-3-yl)acetic acid(Compound1) for use in S1P1 receptor-associated disorders
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
US11795169B2 (en) 2017-10-13 2023-10-24 Imperial College Innovations Limited MAP4K4 inhibitors
JP2020536937A (en) * 2017-10-13 2020-12-17 インペリアル カレッジ オブ サイエンス,テクノロジー アンド メディスン MAP4K4 inhibitor
US11602534B2 (en) 2017-12-21 2023-03-14 Hefei Institutes Of Physical Science, Chinese Academy Of Sciences Pyrimidine derivative kinase inhibitors
WO2019119486A1 (en) 2017-12-21 2019-06-27 中国科学院合肥物质科学研究院 Class of pyrimidine derivative kinase inhibitors
US11034669B2 (en) 2018-11-30 2021-06-15 Nuvation Bio Inc. Pyrrole and pyrazole compounds and methods of use thereof
WO2020115481A1 (en) * 2018-12-05 2020-06-11 Imperial College Of Science, Technology And Medicine Map4k4 inhibitors
WO2020152686A1 (en) 2019-01-23 2020-07-30 Yeda Research And Development Co. Ltd. Culture media for pluripotent stem cells
WO2021097256A1 (en) 2019-11-14 2021-05-20 Cohbar, Inc. Cxcr4 antagonist peptides

Also Published As

Publication number Publication date
AU3176297A (en) 1998-01-14

Similar Documents

Publication Publication Date Title
WO1997049706A1 (en) SUBSTITUTED 7-AMINO-PYRROLO[3,2-d]PYRIMIDINES AND THE USE THEREOF
US6051577A (en) N-7-heterocyclyl pyrrolo[2,3-D]pyrimidines and the use thereof
CA2667487C (en) Imidazo[1,2-b]pyridazine and pyrazolo[1,5-a]pyrimidine derivatives and their use as protein kinase inhibitors
US6133273A (en) Pyrazolopyrimidine-2,4-dione sulfonamides
US5686457A (en) Pyrrolopyrimidine derivatives having pharmacological activity
EP0911333B1 (en) Pyrazolo[4,3-d]pyrimidine derivatives and pharmaceutical compositions containing them
US7326699B2 (en) 4-Amino-5-phenyl-7-cyclobutyl-pyrrolo(2,3-d)pyrimidine derivatives
EP0783505B1 (en) PYRROLO[2,3-d]PYRIMIDINES AND THEIR USE
WO1997028161A1 (en) Novel pyrrolo[2,3-d]pyrimidines and their use as tyrosine kinase inhibitors
Novinson et al. 2-(Alkylthio)-1, 2, 4-triazolo [1, 5-a] pyrimidines as adenosine 3', 5'-monophosphate phosphodiesterase inhibitors with potential as new cardiovascular agents
KR20120097508A (en) Benzodiazepine bromodomain inhibitor
WO2005028434A2 (en) Novel heterocyclic compounds as hsp90-inhibitors
WO2001083485A1 (en) Imidazopyrimidine derivatives and triazolopyrimidine derivatives
JP2002518395A (en) Condensed azepinone-type cyclin-dependent kinase inhibitors
JPH05140167A (en) Substituted pyrazolopyrimidine and imidazopyridazine as angiotensin ii antagonist
KR900005836B1 (en) 1,2,4-triazolo(4,3-c)pyrimidines
US6166016A (en) Amide derivatives
JP3937367B2 (en) Nitric oxide synthase inhibitor
US4528288A (en) Substituted triazolo[1,5-c]pyrimidines
CA1254558A (en) Pyrazolo¬4,3-d|pyrimidine derivative, process for its production, antihyperlipidemic agent containing it, its intermediate and process for the production of the intermediate
US5116841A (en) Imidazoquinoxalines and their preparation and use
EP0121341B1 (en) Triazolo(4,3-c)pyrimidines and triazolo(1,5-c)pyrimidines substituted by nitrogen-containing heterocyclic rings
EP1966213A1 (en) Tricyclic lactam derivatives, their manufacture and use as pharmaceutical agents
WO2004041826A1 (en) Imidazopyrazinones and imidazotriazinones derivates as gaba-a receptor anxiolytic
US4532242A (en) Substituted triazolo[4,3-c]pyrimidines

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GE GH HU IL IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK TJ TM TR TT UA UG US UZ VN YU ZW AM AZ BY KG KZ MD RU TJ TM

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH KE LS MW SD SZ UG ZW AT BE CH DE DK ES FI FR GB GR IE IT LU MC NL PT

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
NENP Non-entry into the national phase

Ref country code: JP

Ref document number: 98502234

Format of ref document f/p: F

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: CA