WO2006107851A1 - 1-substituted pyrazolo (3,4-c) ring compounds as modulators of cytokine biosynthesis for the treatment of viral infections and neoplastic diseases - Google Patents

1-substituted pyrazolo (3,4-c) ring compounds as modulators of cytokine biosynthesis for the treatment of viral infections and neoplastic diseases Download PDF

Info

Publication number
WO2006107851A1
WO2006107851A1 PCT/US2006/012263 US2006012263W WO2006107851A1 WO 2006107851 A1 WO2006107851 A1 WO 2006107851A1 US 2006012263 W US2006012263 W US 2006012263W WO 2006107851 A1 WO2006107851 A1 WO 2006107851A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
alkyl
hydrogen
compound
aryl
Prior art date
Application number
PCT/US2006/012263
Other languages
French (fr)
Inventor
David S. Hays
Ryan B. Prince
Chad A. Haraldson
Jason D. Bonk
Original Assignee
Coley Pharmaceutical Group, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Coley Pharmaceutical Group, Inc. filed Critical Coley Pharmaceutical Group, Inc.
Priority to EP06749140A priority Critical patent/EP1863814A1/en
Priority to AU2006232375A priority patent/AU2006232375A1/en
Priority to CA002602590A priority patent/CA2602590A1/en
Priority to JP2008504494A priority patent/JP2008538550A/en
Priority to US11/887,492 priority patent/US7943636B2/en
Publication of WO2006107851A1 publication Critical patent/WO2006107851A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/12Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains three hetero rings
    • C07D471/14Ortho-condensed systems

Definitions

  • the present invention provides a new class of compounds that are useful in inducing cytokine biosynthesis in animals.
  • Such compounds are of the following Formula
  • R 1 , R 2 , RA, RB 3 X, Z, and m are as defined below.
  • the compounds of Formula I are useful as immune response modifiers due to their ability to induce cytokine biosynthesis (e.g., induces the synthesis of at least one cytokine) and otherwise modulate the immune response when administered to animals. This makes the compounds useful in the treatment of a variety of conditions such as viral diseases and tumors that are responsive to such changes in the immune response.
  • the invention further provides pharmaceutical compositions containing an effective amount of a compound of Formula I and methods of inducing cytokine biosynthesis in an animal, treating a viral infection or disease and/or treating a neoplastic disease in an animal by administering an effective amount of a compound of Formula I to the animal.
  • the present invention provides a compound of the Formula I:
  • Ri is selected from the group consisting of: hydrogen, hydroxy, fluorine, alkoxy,
  • Ri is other than hydrogen
  • m is an integer from 1 to 5
  • RA and R B are each independently selected from the group consisting of: hydrogen, halogen, alkyl, alkenyl, alkoxy, alkylthio, and
  • R 8 is selected from the group consisting of hydrogen, alkyl, alkoxyalkylenyl, and arylalkylenyl;
  • the present invention provides a compound of the Formula XIII:
  • haloalkyl is inclusive of groups that are substituted by one or more halogen atoms, including perfluorinated groups. This is also true of other groups that include the prefix “halo-.” Examples of suitable haloalkyl groups are chloromethyl, trifluoromethyl, and the like.
  • heterocyclyl groups include pyrrolidinyl, tetrahydrofuranyl, morpholinyl, thiomorpholinyl, 1,1- dioxothiomorpholinyl, piperidinyl, piperazinyl, thiazolidinyl, imidazolidinyl, isothiazolidinyl, tetrahydropyranyl, quinuclidinyl, homopiperidinyl (azepanyl), 1,4- oxazepanyl, homopiperazinyl (diazepanyl), 1,3-dioxolanyl, aziridinyl, azetidinyl, dihydroisoquinolin-(lH)-yl, octahydroisoquinolin-(lH)-yl, dihydroquinolin-(2H) ⁇ yl, octahydroquinolin-(2H)-yl, dihydro-lH-l
  • heterocyclyl includes bicyclic and tricyclic heterocyclic ring systems. Such ring systems include fused and/or bridged rings and spiro rings. Fused rings can include, in addition to a saturated or partially saturated ring, an aromatic ring, for example, a benzene ring. Spiro rings include two rings joined by one spiro atom and three rings joined by two spiro atoms.
  • fused aryl ring includes fused carbocyclic aromatic rings or ring systems. Examples of fused aryl rings include benzo, naphtho, fluoreno, and indeno.
  • fused heteroaryl ring includes the fused forms of 5 or 6 membered aromatic rings that contain one heteroatom selected from S and N.
  • each group is independently selected, whether explicitly stated or not.
  • each R 9 group is independently selected for the formula -N(R ⁇ - each R 9 group.
  • each R 4 group is independently selected for the formula -N(R ⁇ - each R 9 group.
  • each R 4 group is independently selected for the formula -N(R ⁇ - each R 9 group.
  • each R 4 group is independently selected for the formula -N(R ⁇ - each R 9 group.
  • each R 4 group is independently selected.
  • tautomer or tautomeric form
  • proton tautomers include interconversions via migration of a proton, such as keto-enol and imine-enamine isomerizations.
  • proton migration between the 2 and 3 positions may occur.
  • any of its embodiments can be combined with any one or more of the other variables in any of their embodiments and associated with any one of the formulas described herein, as would be understood by one oflskill in the art.
  • Each of the resulting combinations of variables is an embodiment of the present invention.
  • RA and R B are each independently selected from the group consisting of hydrogen, halogen, alkyl, alkenyl, alkoxy, alkylthio, and -N(R 9 ) 2 ; or when taken together, RA and RB form a fused aryl ring or heteroaryl ring containing one heteroatom selected from the group consisting of N and S wherein the aryl or heteroaryl ring is unsubstituted or substituted by one or more R groups; or when taken together, R A and RB form a fused 5 to 7 membered saturated ring, optionally containing one heteroatom selected from the group consisting of N and S, and unsubstituted or substituted by one or more R groups.
  • m is an integer from 1 to 5.
  • n is an integer from 1 to 3. In certain embodiments, m is 1.
  • X is a bond
  • R 1 is selected from the group consisting of hydrogen, hydroxy, fluorine, alkoxy, -N(Rg) 2 , -NH-Q-R 4 , -S(0)o -2 -alkyl, -S(O) 2 -NH-R 9 , -C(Re)-N(Rg)-R 4 , -0-C(Rg)-N(Rs)-R 4 , -C(R 6 )-O-alkyl, -0-C(Re)-R 4 , and
  • Rj is fluoro, except where Rj is otherwise defined.
  • Ri is selected from the group consisting of -NH 2 , -NH-Q-R 4 , -C(O)-NH 2 , and -C(O)-N(Rs)-R 4 , wherein Q is selected from the group consisting of -C(O)-, -S(O) 2 -, -C(O)-O-, and -C(O)-NH-
  • R 8 is selected from the group consisting of hydrogen and alkyl
  • R 4 is selected from the group consisting of alkyl and alkoxyalkylenyl, except where R 1 is otherwise defined.
  • Z is selected from the group consisting of:
  • Z is selected from the group consisting of a bond and C 1-3 alkylene.
  • X is a bond
  • Ri is hydrogen
  • Z is
  • Q is selected from the group consisting of -C(O)-, -C(O)-O-, -S(O) 2 -, and -C(Re)-N(R 8 )-.
  • R 2 is selected from the group consisting of hydrogen, alkyl, alkoxyalkylenyl, hydroxyalkylenyl, haloalkylenyl, and R 4 -C(ReO-O-Ci -4 alkylenyl.
  • R 2 is selected from the group consisting of Cj -4 alkyl, Cj -4 alkyl-O-C 2-4 alkylenyl, and hydroxyC 2-4 alkylenyl.
  • R 2 is selected from the group consisting of methyl, ethyl, ⁇ -propyl, «-butyl, 2-methoxy ethyl, and 2-hydroxyethyl.
  • R 4 is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, aryl, arylalkylenyl, aryloxyalkylenyl, alkylarylenyl, heteroaryl, heteroarylalkylenyl, heteroaryloxyalkylenyl, alkylheteroarylenyl, and heterocyclyl, wherein the alkyl, alkenyl, alkynyl, aryl, arylalkylenyl, aryloxyalkylenyl, alkylarylenyl, heteroaryl, heteroarylalkylenyl, heteroaryloxyalkylenyl, alkylheteroarylenyl, and heterocyclyl groups can be unsubstituted or substituted by one or more substituents independently selected from the group consisting of alkyl, alkoxy, hydroxyalkyl, haloalkyl, haloalkoxy
  • R 9 is selected from the group consisting of hydrogen and alkyl. In certain embodiments, R 9 is alkyl. In certain embodiments, R 9 is hydrogen.
  • Q is selected from the group consisting of -C(O)-, -C(O)-O-, -S(O) 2 -, and -C(Re)-N(R 8 )- .
  • Q is a bond.
  • V is selected from the group consisting Of-C(R 6 )-, -0-C(R 6 )-, -N(Rs)-C(R 6 )-, and -S(O) 2 -.
  • V is -C(R 6 )-.
  • V is -N(Rg)-C(Re)-.
  • G 2 is selected from the group consisting of -X 2 -C(O)-R', ⁇ -aminoacyl, ⁇ -aminoacyl- ⁇ -aminoacyl, -X 2 -C(O)-O-R', -C(0)-N(R")R', and -S(O) 2 -R'.
  • Y' is selected from the group consisting of hydrogen, Ci -6 alkyl, and benzyl.
  • Y 0 is selected from the group consisting of Cj -6 alkyl, carboxy-Ci- 6 alkylenyl, amino-Cu alkylenyl, mono-N-Ci- 6 alky lamino-C I-4 alkylenyl, and di-N,N-Cu 6 alkylamino-Ci_ 4 alkylenyl.
  • XIII, Gi is selected from the group consisting of -C(O)-R', ⁇ -aminoacyl, and -C(O)-O-R'.
  • R' contains one to ten carbon atoms.
  • ⁇ -aminoacyl is an ⁇ -C 2 - ⁇ aminoacyl group derived from an ⁇ -amino acid selected from the group consisting of racemic, D-, and L-amino acids containing a total of at least 2 carbon atoms and a total of up to 11 carbon atoms, and may also include one or more heteroatoms selected from the group consisting of O, S, and N.
  • the present invention provides a method of inducing cytokine biosynthesis in an animal comprising administering an effective amount of a compound or salt of any one of Formulas I 5 H 5 III, IV, V, VI, VII, VIII, IX, X, XI 5 XII 5 XIII, XIV, or any one of the above embodiments or administering any one of the above pharmaceutical compositions to the animal.
  • the cytokine is selected from the group consisting of IFN- ⁇ , TNF- ⁇ , IL-6, IL-IO, and IL- 12.
  • the cytokine is IFN- ⁇ or TNF- ⁇ .
  • the present invention provides a method of treating a viral disease in an animal comprising administering a therapeutically effective amount of a compound or salt of any one of Formulas I 5 H 5 III, IV 5 V 5 VI 5 VII, VIII, IX, X, XI, XII, XIII 5 XIV 5 or any one of the above embodiments or administering any one of the above pharmaceutical compositions to the animal.
  • compounds of the invention can be prepared according to Reaction Scheme II, where R, R 2 , Z, n and m are as defined above.
  • compounds of the invention can also be prepared according to Reaction Scheme III, where Rj, R 2 , X, Z, and m are as defined above, and RA 2 and RB 2 taken together form a fused benzene ring or fused pyridine ring wherein the benzene ring or pyridine ring is unsubstituted or substituted by one or more R groups.
  • a ketone of Formula XXX is condensed with diethyl oxalate under Claisen condensation conditions to provide a ketoester of Formula XXXI.
  • the reaction can be carried out by adding sodium f ⁇ rt-butoxide to a solution of diethyl oxalate and the ketone of Formula XXX in ethanol at ambient temperature.
  • a ketoester of Formula XXXI reacts with a hydrazine of Formula R 2 NHNH 2 to provide a pyrazole carboxylate of Formula XXXII.
  • the reaction is conveniently carried out by slowly adding the hydrazine to a solution of a compound of Formula XXXI in a suitable solvent such as acetic acid. The reaction can be carried out at ambient temperature.
  • step (3) of Reaction Scheme III the ester group of a pyrazole carboxylate of Formula XXXII is converted to an amide.
  • the amination can be carried out by adding ammonium hydroxide to the pyrazole carboxylate of Formula XXXII in a suitable solvent such as methanol and heating at an elevated temperature such as 100 0 C.
  • the reaction can be carried out in a pressure vessel.
  • step (3) can be carried out by first hydrolyzing a pyrazole carboxylate of Formula XXXII to a carboxylic acid and then converting the carboxylic acid to an amide.
  • the ester hydrolysis can be carried out under basic conditions by combining a pyrazole carboxylate of Formula XXXII with lithium hydroxide or sodium hydroxide in water and in a suitable solvent such as methanol or ethanol.
  • the reaction can be carried out at ambient temperature, and the carboxylic acid product can be isolated using conventional methods.
  • the conversion of the carboxylic acid to a pyrazole carboxamide of Formula XXXIII can be carried out under coupling conditions by adding 1-hydroxybenzotriazole and l-(3-dimethylaminopropyl)-3- ethylcarbodiimide hydrochloride to a solution of the carboxylic acid in a suitable solvent such as jV,7V-dimethylformamide (DMF) at ambient temperature and then adding concentrated ammonium hydroxide.
  • a suitable solvent such as jV,7V-dimethylformamide (DMF)
  • a pyrazole carboxamide of Formula XXXIII is dehydrated to a pyrazole carbonitrile of Formula XXXIV.
  • Suitable dehydrating agents include thionyl chloride, trifluoroacetic anhydride, and phosphorous oxychloride.
  • the reaction is conveniently carried out by treating the pyrazole carboxamide of Formula XXXIII with phosphorous oxychloride and heating the reaction at an elevated temperature such as 90 °C.
  • a pyrazole carbonitrile of Formula XXXIV is brominated to provide a bromo-substituted pyrazole carbonitrile of Formula XXXV.
  • the bromination is conveniently carried out by adding bromine to a solution of the pyrazole carbonitrile of Formula XXXIV and potassium acetate in acetic acid. The reaction can be carried out at ambient temperature.
  • a Suzuki coupling reaction is conveniently carried out by heating a mixture of the bromo-substituted pyrazole of Formula XXX, dichlorobis(triphenylphosphine)palladiurn(II) and a boron reagent of Formula XXXVI 5 where M is -B(OH) 2 or -B(0-alkyl) 2 , in the presence of a base such as potassium carbonate.
  • the reaction is carried out in a suitable solvent such as 1 ,2-dimethoxyethane and can be heated at an elevated temperature such as 75 - 95 0 C.
  • step (7) of Reaction Scheme III the amine and nitrile functionalities of a pyrazole of Formula XXXVII react under acidic conditions to form a pyrazolo [3 ,4- c]quinoline or pyrazolo [3 ,4-c]naphthyridine of Formula XXXVIII.
  • the intramolecular addition is conveniently carried out by stirring acetyl chloride in ethanol and adding the resulting acidic solution to the pyrazole of Formula XXXVIII.
  • the reaction is then heated at reflux to provide the pyrazolo [3 ,4-c]quinoline or pyrazolo[3,4-c]naphthyridine of Formula XXXVIII.
  • Reaction Scheme IV where R], R 2 , X, Z, and m are as defined above and RA I and RB I are as defined below.
  • step (1) of Reaction Scheme IV a bromo-substituted pyrazole carbonitrile of Formula XXXV undergoes a Sonogashira coupling reaction with (trimethylsilyl)acetylene to provide a pyrazole carbonitrile of Formula XXXIX.
  • the reaction can be carried out according to the literature procedure, Sonogashira, K.; Tohda, Y.; Hagihara, N., Tetrahedron Lett, 4467 (1975).
  • the iodo analog may be used as a starting material for Reaction Scheme IV.
  • the iodo analog can be prepared from a pyrazole carbonitrile of Formula XXIV 5 shown in Reaction Scheme III.
  • the iodination can be carried out by treating a pyrazole carbonitrile of Formula XXXIV with iodine monochloride in a suitable solvent such as dichloromethane in the presence of a base such as potassium carbonate.
  • the reaction can be carried out at ambient temperature.
  • step (2) of Reaction Scheme IV the trimethylsilyl group of the pyrazole of Formula XXXIX is removed to provide the pyrazole of Formula XL.
  • Potassium carbonate in methanol or tetrabutylammonium fluoride in tetrahydrofuran can be used to carry out the transformation.
  • step (3) of Reaction Scheme IV the acetylene of the pyrazole of Formula XL is alkylated using conventional synthetic methods, Jacobs, T. L. in Organic Reactions, 5, 1, (1949), to provide a pyrazole of Formula XLI.
  • the reaction can be carried out by deprotonation of the compound of Formula XL with a base and reaction of the resulting carbanion with an electrophile of Formula Rei-Halide, for example, iodomethane.
  • Step (3) can be omitted when RBI is hydrogen.
  • steps (1) through (3) of Reaction Scheme IV may be replaced with one step from the iodo analog using a Sonogashira coupling reaction.
  • the coupling can be carried out by combining an alkyne of Formula RBI -G ⁇ C-H, copper(I) iodide, dichlorobis(triphenyl ⁇ hosphine)palladium(II), and triethylamine in a suitable solvent such as acetom ' trile and then heating at an elevated temperature, such as the reflux temperature of the solvent.
  • step (6) of Reaction Scheme IV a bromo-substituted pyrazolo[3,4-c]pyridin-4- amine of Formula XLIII undergoes a transition metal catalyzed coupling reaction with a reagent of Formula RAI-M, where R A I is alkenyl, alkoxy, and -N(R ⁇ 2 to provide a pyrazolo[3,4-c]pyridine-4-amine of Formula XLIV.
  • step (6) can be carried out by coupling a compound of Formula XLIII with an alkyne under Sonogashira conditions as described in step (1) of this reaction scheme.
  • the resulting alkyne can be reduced under conventional hydrogenation conditions to provide a compound of Formula XLIV, where RA 1 is alkenyl or alkyl.
  • Step (6) may also be carried out by (i) protecting the amino group of the compound of Formula XLIII, for example, with a Boc group; (ii) performing a lithium-halogen exchange; (iii) treating with an electrophile of the Formula R A i-Halide, for example iodomethane; and (iv) deprotecting the amine to provide a compound of Formula XLIV.
  • compounds of the invention can be prepared according to Reaction Scheme V, where Rj b and R 2! , are subsets of Rj and R 2 as defined above that do not include those substituents which would be susceptible to reduction under the acidic hydrogenation conditions of the reaction and R, X, Z, and n are as defined above.
  • Reaction Scheme V a pyrazolo[3,4-c]quinoline of Formula XLV is reduced to provide a 6,7,8,9-tetrahydro-2i/-pyrazolo[3,4-c]quinoline of Formula XLVI, which is a subgenus of Formula IV.
  • the reaction may be carried out under heterogeneous hydrogenation conditions by adding platinum (IV) oxide to a solution or suspension of a pyrazolo[3,4-c]quinoline of Formula XLV in a suitable solvent such as trifluoroacetic acid and placing the reaction under hydrogen pressure.
  • the reduction may be carried out at an earlier stage in the synthetic pathway.
  • Pyrazolo[3,4 ⁇ c]naphthyridines of the invention can be prepared by using an azaindole as the starting material in Reaction Scheme I. Azaindoles are known compounds. Some are commercially available and others can be prepared using known synthetic methods. Alternatively, pyrazolo[3,4-c]naphthyridines of the invention can be prepared by using an aminopyridine boronic acid in Reaction Scheme III. Aminopyridine boronic acids can be prepared using known methods, for example, by directed ortho metalation of Boc-protected aminopyridines and subsequent electr ⁇ philic substitution. Alternatively, for some isomers, halogen-lithium exchange and subsequent electrophilic substitution can be used.
  • halogen-lithium exchange can be carried out on a 2-bromopyridine that has a protected amino group in the 3 -position; subsequent electrophilic substitution with tributyltin chloride and deprotection of the amino group provides 3 -amino-2-tri-n-butylstannylpyridme.
  • 6 7,8,9-Tetrahydro-2H-pyrazolo[3,4-c]naphthyridines can be prepared by reducing pyrazolo[3,4-c]naphthyridines using the method of Reaction Scheme V.
  • compounds can be further elaborated using conventional synthetic methods.
  • a compound of Formula XLVII 5 can undergo acid mediated cleavage of the Boc group in step (1) to give a secondary amine that can be functionalized in step (2) with an acid chloride of Formula R 4 C(O)Cl 5 an acid anhydride of Formula (R 4 C(O)) I O, an alkyl chloroformate of Formula R 4 OC(O)Cl 5 a sulfonyl chloride of Formula R 4 S(O) 2 Cl, a sulfonic anydride of Formula (R 4 S(O) 2 ⁇ O 5 an isocyanate of formula R 4 NCO, or an isothiocyanate of formula R 4 NCS to provide a compound of Formula XLIX where R 4 is defined as above and Q is -C(O)-, -C(O)O-, -S(O) 2 -, -C(O)NH-, or -C(S)NH-.
  • a compound of Formula XLVIII in Reaction Scheme VI can undergo alkylation of the secondary amine.
  • the compound of Formula XLVIII may be reacted with aldehydes, alkyl halides or triflates to provide a compound Formula L in which R 8 is defined as above.
  • a compound of Formula XLVIII with aqueous formaldehyde and a reducing agent such as sodium cyanoborohydride in an appropriate solvent such as methanol yields a compound of Formula L, where R 8 is a methyl group.
  • an olefin of Formula LII is derivatized using conventional methods.
  • a compound of Formula LII where X is a bond and R 1 is hydrogen can be prepared by reducing the olefin using conventional heterogeneous hydrogenation conditions.
  • a compound of Formula LII can be treated with pivalonitrile in the presence of titanium tetrachloride; the resulting nitrile-substituted compound can be converted by convention methods to a compound of Formula LIII where X is a bond and Ri is -C(O)-NH 2 .
  • a compound of Formula LII can also be treated with ammonium hydroxide followed by di ⁇ tert-butyl dicarbonate to provide a compound of Formula LIII where X is a bond and Rj is -NHBoc, which can be deprotected and treated according to the methods of Reaction Scheme VI to provide a variety of other compounds.
  • compounds of the invention can be prepared according to Reaction Scheme VIII, where RA 2 , RB 23 R2, and Z are as defined above.
  • step (1) of Reaction Scheme VIII an olefin of Formula LII is condensed with diethyl oxalate to provide a ketoester of Formula LIV.
  • the reaction can be carried out as described in step (1) of Reaction Scheme III.
  • step (2) of Reaction Scheme VIII a ketoester of Formula LIV reacts with a hydrazine of Formula R 2 NHNH 2 to provide a pyrazole carboxylate of Formula LV.
  • the reaction can be carried out as described in step (2) of Reaction Scheme III.
  • step (3) of Reaction Scheme VIII a pyrazole carboxylate of Formula LV is converted to a pyrazole carboxamide of Formula LVL
  • the reaction can be carried out as described in step (3) of Reaction Scheme III.
  • step (4) of Reaction Scheme VIII the olefinic bond in a compound of Formula LVI is oxidized to provide an epoxide of Formula LVII.
  • the reaction can be carried out by treating a suspension of a compound of Formula LVI in a suitable solvent such as chloroform with 3-chloroperoxybenzoic acid. The reaction can be carried out at ambient temperature.
  • step (5) of Reaction Scheme VIII the epoxide ring in a compound of Formula LVII is cleaved to provide a hydroxy substituted pyrazole carboxamide of Formula LVIII.
  • the reaction can be carried out by treating a solution of a compound of Formula LVII in a suitable solvent such as ethanol with palladium on carbon and ammonium formate. The reaction can be carried out at ambient temperature.
  • a pyrazole carbonitrile of Formula LIX is converted to a pyrazolo[3,4 ⁇ c]quinoline or pyrazolo[3,4-c]naphthyridine of Formula LX.
  • the conversion can be carried out using the methods described in steps (5) through (7) of Reaction Scheme III.
  • compounds of the invention can be prepared according to Reaction Scheme IX, where R A2 , RB2, R 2 , and Z are as defined above.
  • step (1) of Reaction Scheme IX 5 the hydroxy group in a pyrazolo carbonitrile of Formula LIX is replaced with a fluoro group to provide a pyrazolo carbonitrile of Formula LXI.
  • the reaction can be carried out by treating a solution of a compound of Formula LIX in a suitable solvent such as dichloromethane with [bis(2-methoxyethyl)amino]sulfur trifiuoride.
  • the trifluoride is added in a controlled fashion at a sub-ambient temperature such as 0 0 C.
  • a pyrazole carbonitrile of Formula LXI is converted to a pyrazolo[3,4-c]quinoline or pyrazolo[3,4-c]naphthyridine of Formula LXII.
  • the conversion can be carried out using the methods described in steps (5) through (7) of Reaction Scheme III.
  • Compounds of the invention can also be prepared using variations of the routes shown in Reaction Schemes I through IX that would be apparent to one of skill in the art.
  • a compound of Formula LII wherein Z is -N(Boc)- can be readily prepared from l-Boc-4-piperidinone according to the method of step (1) of Reaction Scheme VII.
  • Steps (1) through (6) of Reaction Scheme VIII can then be used to prepare a compound of Formula LIX wherein Z is -N(Boc)-.
  • Boc group can then be cleaved and the resulting amine can be treated with an acid chloride, alkyl chloroformate, sulfonyl chloride, sulfonic anhydride, isocyanate, or isothiocyanate according to the methods described in steps (1) and (2) of Reaction Scheme VI.
  • Boc removal and amine functionalization steps can conveniently be carried out after the bromination in step (7) of Reaction Scheme VIII or after steps (1) and (2) of Reaction Scheme IX.
  • compounds of the invention can be prepared according to Reaction Scheme X, where Ri, R 2 , RA 5 RB, G b Z, and m are as defined above.
  • the amino group of a pyrazolo compound of Formula I can be converted by conventional methods to a functional group such as an amide, carbamate, urea, amidine, or another hydrolyzable group.
  • reaction can be carried out, for example, by combining a compound of Formula I with a chloroformate or acid chloride, such as ethyl chloroformate or acetyl chloride, in the presence of a base such as triethylamine in a suitable solvent such as dichloromethane at room temperature.
  • a chloroformate or acid chloride such as ethyl chloroformate or acetyl chloride
  • compounds of the invention can be prepared according to Reaction Scheme XI, where R 2 , RA 5 RB, G 2 , Z, and m are as defined above.
  • the hydrogen atom of the alcohol group of Formula LXIII can be replaced using conventional methods with a group such as Ci -6 alkanoyloxymethyl, 1-(Ci -6 alkanoyloxy)ethyl, 1 -methyl- 1-(C 1-6 alkanoyloxy)ethyl, Ci -6 alkoxycarbonyloxymethyl, iV-(C 1-6 alkoxycarbonyl)aminomethyl, succinoyl, Ci -6 alkanoyl, ⁇ -aminoCi- 4 alkanoyl, arylacyl, -P(O)(OH) 2 , -P(O)(O-C 1-6 alkyl) 2 , Ci -6 alkoxycarbonyl, C 1-6 alkylcarbamoyl, and ⁇ -aminoacyl or
  • a therapeutically effective amount and “effective amount” mean an amount of the compound or salt sufficient to induce a therapeutic or prophylactic effect, such as cytokine induction, immunomodulation, antitumor activity, and/or antiviral activity.
  • cytokine induction cytokine induction
  • immunomodulation antitumor activity
  • antiviral activity cytokine induction
  • amount of compound or salt used in a pharmaceutical composition of the invention will vary according to factors known to those of skill in the art, such as the physical and chemical nature of the compound or salt, the nature of the carrier, and the intended dosing regimen.
  • the method includes administering sufficient compound to provide a dose of from about 0.1 mg/m 2 to about 2.0 mg/ m 2 to the subject, for example, a dose of from about 0.4 mg/m 2 to about 1.2 mg/m 2 .
  • dosage forms such as tablets, lozenges, capsules, parenteral formulations, syrups, creams, ointments, aerosol formulations, transdermal patches, transmucosal patches and the like.
  • These dosage forms can be prepared with conventional pharmaceutically acceptable carriers and additives using conventional methods, which generally include the step of bringing the active ingredient into association with the carrier.
  • the compounds or salts of the invention can be administered as the single therapeutic agent in the treatment regimen, or the compounds or salts described herein may be administered in combination with one another or with other active agents, including additional immune response modifiers, antivirals, antibiotics, antibodies, proteins, peptides, oligonucleotides, etc.
  • Cytokines whose production may be induced by the administration of compounds or salts of the invention generally include interferon- ⁇ (IFN- ⁇ ) and tumor necrosis factor- ⁇ (TNF- ⁇ ) as well as certain interleukins (IL). Cytokines whose biosynthesis may be induced by compounds or salts of the invention include IFN- ⁇ , TNF- ⁇ , IL-I, IL-6, IL-IO and IL- 12, and a variety of other cytokines. Among other effects, these and other cytokines can inhibit virus production and tumor cell growth, making the compounds or salts useful in the treatment of viral diseases and neoplastic diseases. Accordingly, the invention provides a method of inducing cytokine biosynthesis in an animal comprising administering an effective amount of a compound or salt of the invention to the animal.
  • compounds or salts described herein can affect other aspects of the innate immune response. For example, natural killer cell activity may be stimulated, an effect that may be due to cytokine induction.
  • the compounds or salts may also activate macrophages, which in turn stimulate secretion of nitric oxide and the production of additional cytokines. Further, the compounds or salts may cause proliferation and differentiation of B-lymphocytes.
  • T helper type 1 cytokine IFN- ⁇
  • T helper type 2 cytokines IL- 4, IL-5 and IL- 13
  • the compound or salt or composition may be administered alone or in combination with one or more active components as in, for example, a vaccine adjuvant.
  • the compound or salt or composition and other component or components may be administered separately; together but independently such as in a solution; or together and associated with one another such as (a) covalently linked or (b) non-covalently associated, e.g., in a colloidal suspension.
  • Conditions for which compounds or salts or compositions identified herein may be used as treatments include, but are not limited to:
  • viral diseases such as, for example, diseases resulting from infection by an adenovirus, a herpesvirus (e.g., HSV-I, HSV-II, CMV, or VZV), a poxvirus (e.g., an orthopoxvirus such as variola or vaccinia, or molluscum contagiosum), a picornavirus (e.g., rhinovirus or enterovirus), an orthomyxovirus (e.g., influenzavirus), a paramyxovirus (e.g., parainfluenzavirus, mumps virus, measles virus, and respiratory syncytial virus (RSV)), a coronavirus (e.g., SARS), a papovavirus (e.g., papillomaviruses, such as those that cause genital warts, common warts, or plantar warts), a hepadnavirus (e.g., hepatitis B virus),
  • atopic diseases such as atopic dermatitis or eczema, eosinophilia, asthma, allergy, allergic rhinitis, and Ommen's syndrome;
  • autoimmune diseases such as systemic lupus erythematosus, essential thrombocythaemia, multiple sclerosis, discoid lupus, alopecia areata; and (g) diseases associated with wound repair such as, for example, inhibition of keloid formation and other types of scarring (e.g., enhancing wound healing, including chronic wounds).
  • Compounds or salts identified herein may be particularly helpful in individuals having compromised immune function.
  • compounds or salts may be used for treating the opportunistic infections and tumors that occur after suppression of cell mediated immunity in, for example, transplant patients, cancer patients and HIV patients.
  • one or more of the above diseases or types of diseases for example, a viral disease or a neoplastic disease may be treated in an animal in need thereof (having the disease) by administering a therapeutically effective amount of a compound or salt of the invention to the animal.
  • An animal may also be vaccinated by administering an effective amount of a compound or salt described herein, as a vaccine adjuvant.
  • a method of vaccinating an animal comprising administering an effective amount of a compound or salt described herein to the animal as a vaccine adjuvant.
  • the amount is expected to be a dose of, for example, from about 0.01 mg/m to about 5.0 mg/m , (computed according to the Dubois method as described above) although in some embodiments the induction or inhibition of cytokine biosynthesis may be performed by administering a compound or salt in a dose outside this range.
  • the method includes administering sufficient compound or salt or composition to provide a dose of from about 0.1 mg/m 2 to about 2.0 mg/ m 2 to the subject, for example, a dose of from about 0.4 mg/m 2 to about 1.2 mg/m 2 .
  • the invention also provides a method of treating a viral infection in an animal and a method of treating a neoplastic disease in an animal comprising administering an effective amount of a compound or salt of the invention to the animal.
  • An amount effective to treat or inhibit a viral infection is an amount that will cause a reduction in one or more of the manifestations of viral infection, such as viral lesions, viral load, rate of virus production, and mortality as compared to untreated control animals.
  • the precise amount that is effective for such treatment will vary according to factors known in the art but is expected to be a dose of about 100 ng/kg to about 50 mg/kg, preferably about 10 ⁇ g/kg to about 5 mg/kg.
  • An amount of a compound or salt effective to treat a neoplastic condition is an amount that will cause a reduction in tumor size or in the number of tumor foci. Again, the precise amount will vary according to factors known in the art but is expected to be a dose of about 100 ng/kg to about 50 mg/kg, preferably about 10 ⁇ g/kg to about 5 mg/kg. In other embodiments, the amount is expected to be a dose of, for example, from about 0.01 mg/m 2 to about 5.0 mg/m 2 , (computed according to the Dubois method as described above) although in some embodiments either of these methods may be performed by administering a compound or salt in a dose outside this range.
  • the method includes administering sufficient compound or salt to provide a dose of from about 0.1 mg/m 2 to about 2.0 mg/ m 2 to the subject, for example, a dose of from about 0.4 mg/m 2 to about 1.2 mg/m 2 .
  • Tetrahydro-4/f-pyran-4-one (901 mg, 9.0 mmol) was added, and reaction mixture was stirred for 10 minutes.
  • the -78 0 C bath was replaced with a 0 0 C bath, and then saturated aqueous ammonium chloride (30 mL) was added.
  • the aqueous layer was separated and extracted three times with ter/-butyl methyl ether, and the combined organic fractions were dried over magnesium sulfate, filtered, and concentrated under reduced pressure.
  • the residue (1.3 g) was purified by automated flash chromatography (eluting with ethyl acetate) and heated in refluxing 1 M hydrogen chloride in ethanol (50 mL) for one hour.
  • the vessel was purged with nitrogen, and 10% palladium on carbon (400 mg) was added.
  • the vessel was shaken under hydrogen pressure (50 psi, 3.4 X 10 5 Pa) for approximately ten minutes, and the reaction mixture was filtered through a layer of CELITE filter agent. The filter cake was washed with ethyl acetate. The filtrate was concentrated under reduced pressure, and the residue was dried under a stream of nitrogen to provide 3.80 g of l-tetrahydro-2H-pyran-4-ylacetone as a colorless oil.
  • 2-Aminophenylboronic acid hydrochloride (1.39 g, 8.0 mmol) and dichlorobis(tri ⁇ henylphosphine)palladium(II) (140 mg, 0.20 mmol) were sequentially added to a mixture of 4-bromo-l-propyl-5-(tetrahydro-2H-pyran-4-ylmethyl)-lH- pyrazole-3-carbonitrile (1.25 g, 4.00 mmol), potassium carbonate (1.82 g, 13.2 mmol), 1,2-dimethoxyethane (DME) (15 mL), and water (7.5 mL). The flask was placed under vacuum and back-filled with nitrogen four times.
  • dichlorobis(tri ⁇ henylphosphine)palladium(II) 140 mg, 0.20 mmol
  • Part A tert-B ⁇ xXy ⁇ iV-(2-pyridyi)carbamate is available from the literature procedure (Moraczewski, A. L. et al, J. Org. Chem., 1998, 63, 7258) or can be prepared by the following method. Under a nitrogen atmosphere, sodium bis(trimethylsilyl)arnide (225 mL of a 1.0 M solution in tetrahydrofuran) was added over a period of 20 minutes to a solution of 2-aminopyridine (10.61 g, 108.0 mmol) in dry THF (150 mL). The solution was stirred for 15 minutes and then cooled to 0 0 C.
  • Lithium hydroxide monohydrate (2.94 g, 70.2 mmol) was added to a solution of ethyl 1 -(2-methoxyethyl)-5-(tetrahydro-2i7 ⁇ pyran-4-ylmethyl)- 1 i/-pyrazole-3-carboxylate (5.2 g, 17.5 mmol) in methanol (60 mL) and water (20 niL). The mixture was stirred for overnight. Most of the volatiles were removed under reduced pressure, and acetic acid (40 mL) and water were added. The solution was cooled to approximately 0 0 C and stirred for one hour. The volatiles were removed under reduced pressure, and the residue was partitioned between water and chloroform.
  • a solution of sodium tert-butoxide (5.29 g, 1.1 eq) in ethanol (51 mL) was added to a mixture of the material from Part A (LO eq) and diethyl oxalate (7.45 mL, 1.1 eq).
  • the vessel containing the material from part A and diethyl oxalate was rinsed with additional ethanol (27 mL) and the rinse was added to the reaction mixture.
  • the reaction mixture was stirred for 2 hours and then cooled to 0 °C.
  • Acetic acid (57 mL) was added and the reaction mixture was stirred for 5 minutes.
  • Methylhydrazine (2.64 mL, 1.0 eq) was added dropwise.
  • Triethylamine (5.13 mL, 3.0 eq) was added to a stirred suspension of a portion of the material from Part F (2.91 g, 1.0 eq). The mixture was cooled to 0 °C and trifluoroacetic anhydride (5.14 mL, 3.0 eq) was added dropwise over a period of 5 minutes. The reaction mixture was stirred for 2 hours, quenched with saturated sodium carbonate (50 mL), and then allowed to warm to ambient temperature. Water (50 mL) and dichloromethane (200 mL) were added sequentially. The organic layer was separated, dried over magnesium sulfate, and then concentrated under reduced pressure to provide a yellow oil.
  • the oil was dissolved in methanol (80 mL). Solid potassium carbonate (420 mg, 0.25 eq) was added and the mixture was stirred for 30 minutes. Aqueous hydrochloric acid (1.7 mL of 7 M, 1.0 eq) was added, the solution was stirred for 10 minutes, and then the bulk of the methanol was removed under reduced pressure. The residue was partitioned between dichloromethane (200 mL) and water (50 mL). The pH of the aqueous layer was adjusted to 7-8 with saturated sodium bicarbonate. The layers were separated and the aqueous layer was back extracted with dichloromethane (2 x 75 mL).
  • Part H A solution of material from Part G (lots 1 and 2, 1.0 eq) in dichloromethane (41 mL) was cooled to 0 0 C. [Bis(2-methoxyethyl)amino]sulfur trifluoride (1.14 mL, 1.5 eq) was added dropwise. The reaction mixture was stirred for 45 minutes, quenched with saturated sodium bicarbonate (20 mL), and allowed to warm to ambient temperature. Saturated sodium bicarbonate (50 mL) and dichloromethane (50 mL) were added sequentially. The layers were separated and the aqueous layer was back extracted with dichloromethane (30 mL).
  • the resulting foamy suspension was combined with dichloromethane (50 mL).
  • the aqueous layer was adjusted to about pH 8 with 5% sodium hydroxide.
  • the layers were separated and the aqueous layer was back extracted with dichloromethane (50 mL).
  • the combined organics were dried over magnesium sulfate and then concentrated under reduced pressure to provide 0.59 g of a light yellow solid.
  • This material was triturated with hot ethanol (about 8 mL), isolated by filtration, rinsed with ethanol (3 x 5 mL), and then dried (0.15 torr (20 Pa), 130 0 C, 2 hours) to provide 143 mg of l-[(4-fluorotetrahydro-2H ' -pyran-4-yl)methyl]-2-methyl-2H- ⁇ yrazolo[3,4-c]quinolin-4-amine as a tan powder, mp 252-254 0 C.
  • the material from Part A (1.0 eq) was coupled with 2-aminophenylboronic acid hydrochloride (1.8 eq) and then cyclized using the general method of Example 7 Part J.
  • the crude product was purified by automated flash chromatography (eluting with 20% CMA in chloroform for 3 column volumes, a gradient of 20-40% CMA in chloroform over 10 column volumes, and 40% CMA in chloroform for 3 column volumes) to provide a tan solid.
  • This material was triturated with hot ethyl acetate (about 8 mL), isolated by filtration, rinsed with ethyl acetate (3 x 5 mL), and dried under high vacuum to provide 106 mg of 4-[(4-amino-2-methyl-2i/-pyrazolo[3,4-c]quinolin- 1 -yl)methyl]tetrahydro-2H- pyran-4-ol as a tan powder, mp 260-262 (dec) °C.
  • This material was triturated with methanol (about 12 mL), isolated by filtration, rinsed with methanol (3 x 5 mL), and dried under high vacuum to provide 112 mg of 2-ethyl-l-[(4-fluorotetrahydro-2/J-pyran-4- yl)methyl]-2//-pyrazolo[3,4-c]quinolin-4-amine as a tan powder, mp 275-277 °C.
  • Part A 4-[(4-Amino-2-ethyl-2/f-pyrazolo[3,4-c]quinolin-l -yl)methyl]piperidin-4-ol was prepared according to the method of Example 1 Part F using di(tert-butyl) 2-ethyl-l- methyl-2H-pyrazolo[3,4-c]quinolin-4-ylimidodicarbonate in lieu of di(tert-bntyl) 1- methyl-2 ⁇ propyl-2H-pyrazolo[3,4-c]quinolin-4-ylimidodicarbonate and tert-butyl A- oxopiperidine-1-carboxylate in lieu of cyclobutanone.
  • Methanesulfonic anhydride (0.160 g, 0.922 mmol) was added to a slurry of 4-[(4- amino-2-ethyl-2H-pyrazolo[3,4-c]quinolin-l-yl)methyl]piperidin-4-ol (0.300 g, 0.922 mmol) in chloroform (10 mL). After 16 hours, 2M aqueous sodium carbonate was added and the biphasic mixture was stirred for 30 minutes resulting in a white precipitate. The mixture was extracted with 10% methanol in dichloromethane. The solution was concentrated.
  • ⁇ -Propyl isocyanate (86 ⁇ L, 0.922 mmol) was added to a slurry of 4-[(4-amino-2- ethyl-2H-pyrazolo[3,4-c]quinolin-l-yl)methyl]piperidin-4-ol (0.300 g, 0.922 mmol) in chloroform (10 mL). After 16 hours, the solution was purified via automated flash chromatography eluting with a linear gradient of 2-25% CMA in chloroform.
  • the acidic mixture was brought to pH 14 using 50% aqueous sodium hydroxide.
  • the mixture was extracted with 10% methanol in dichloromethane, dried over anhydrous sodium sulfate, and concentrated.
  • the material was purified via automated flash chromatography eluting with a linear gradient of 2-20% CMA in chloroform.
  • the resulting white solid was triturated in acetonitrile and then isolated by filtration to provide 0.085 g of l-[(4-amino-2-ethyl- 6 5 7,8,9-tetrahydro-2H-pyrazolo[3,4-c]quinolin-l-yl)methyl]cyclohexanol as a white solid, m.p. 232-234 °C.
  • MS (ESI) m/z 329.44 (M + H) +
  • the material from Part C was treated according to the methods of Parts D, E, and F of Example 7.
  • the crude solid obtained from Part F was partitioned between dichloromethane (200 mL) and water (100 mL).
  • the aqueous solution was separated and extracted with chloroform (3 x 100 mL).
  • the organic fractions were combined, dried over magnesium sulfate, and filtered.
  • the filter cake was washed with chloroform (5 x 60 mL).
  • the material from Part D was treated with triethylamine and trifluoroacetic anhydride according to the method of Part G of Example 7.
  • the crude yellow oil that was obtained was purified by automated flash chromatography (eluting with a gradient of 30% hexanes in tert-butyl methyl ether to 100% tert-butyl methyl ether over 4 column volumes and then 100% tert-butyl methyl ether for 3 column volumes) and dried under high vacuum to provide 3.62 of the trifluoroacetate ester of tert-butyl 4-[(5-cyano-2-methyl- 27i-pyrazol-3-yl)methyl]-4-hydroxypiperidine-l -carboxylate as a colorless oil.
  • Triethylamine (3.4 mL, 3.0 eq.) was added to a stirred suspension of 4-bromo-5- [(4-fluoropiperidin-4-yl)methyl]-l -methyl- lH-pyrazole-3-carbonitrile hydrochloride (2.7 g, 8.0 mmol) in dichloromethane (80 mL), and the mixture was cooled to 0 0 C.
  • Acetyl chloride (0.74 mL, 1.3 eq.) was added, and the resulting solution was stirred for 1.5 hours and concentrated under reduced pressure.
  • Triethylamine (1.5 mL, 1.5 eq.) was added to a stirred suspension of 4-bromo-5- [(4-fluoropiperidin-4-yl)methyl]-l -methyl- li?-pyrazole-3-carbonitrile hydrochloride (2.28 g, 6.75 mmol) in dichloromethane (67 mL), and isopropyl isocyanate (1.34 mL, 2.0 eq.) was added to the resulting solution. The reaction was stirred for 1.5 hours and concentrated under reduced pressure.
  • Triethylamine (11.1 mL, 79.5 mmol) and methanesulfonyl chloride (1.24 mL, 15.9 mmol) were sequentially added to a mixture of the material from Part B in dichloromethane (80 mL), and the reaction was stirred for 5 minutes. Brine (40 mL) was added, and the aqueous layer was separated and extracted with dichloromethane (2 x 100 mL). The combined organic fractions were dried over magnesium sulfate, filtered, and concentrated under reduced pressure to provide a brown foam.
  • the foam was purified by automated flash chromatography (eluting with a gradient of 0- 10% CMA in chloroform over 8 column volumes) to provide 1.9 g of 4-bromo-l-ethyl-5- ⁇ [4-fluoro-l- (methylsulfonyl)pi ⁇ eridin-4-yl]methyl ⁇ -lH-pyrazole-3-carbonitrile as a tan foam.
  • Triethylamine (3.35 niL, 24.0 mmol) was added to a stirred suspension of 4- bromo-l-ethyl-5-[(4-fluoropiperidin-4-yl)methyl-lH-pyrazole-3-carbonitrile hydrochloride (2.52 g, 8.0 mmol) in dichloromethane (80 niL), and the resulting solution was cooled to 0 0 C.
  • Acetyl chloride (0.74 mL, 10.4 mmol) was added dropwise, and the resulting solution was stirred for 1 hour. Water (50 mL) was added, and then the aqueous layer was separated and extracted with dichloromethane (2 x 100 mL).
  • Triethylamine (2.23 mL, 16.0 mmol) and isopropyl isocyanate (1.01 niL, 10.4 mmol) were sequentially added to a mixture of 4-bromo-l-ethyl-5-[(4-fluoropiperidin-4- yl)methyl-l/7-pyrazole-3-carbonitrile hydrochloride (2.52 g, 8.0 mmol) and dichloromethane (40 mL). The reaction was stirred for 1.5 hours and diluted with dichloromethane (250 mL), washed with water (WO mL), dried over magnesium sulfate, filtered, and concentrated under reduced pressure to provide a brown foam.
  • the foam was purified by automated flash chromatography (eluting with a gradient of 0-10% CMA in chloroform over 8 column volumes). The resulting off-white foam was concentrated from 1,2-dimethoxyethane (50 mL) and dried under high vacuum to provide 1.6 g of 4-[(4- bromo-5-cyano-2-ethyl-2i ⁇ -pyrazol-3-yl)methyl]-4-fluoro-N-isopropylpiperidine-l- carboxamide. Part B
  • the foam was purified by automated flash chromatography (eluting with a gradient of 0-15% CMA in chloroform) to provide 1.5 g of 4-bromo-5- ⁇ [4-fluoro- 1 -(methylsulfonyl) ⁇ iperidin-4-yl]methyl ⁇ - 1 -propyl- lH-pyrazole- 3-carbonitrile as a white foam.
  • the foam was concentrated from 1,2-dimethoxyethane (40 mL) before it was used in Part D. Part D
  • the reaction conditions and purification methods described in Part J of Example 7 were used to treat the material from Part C.
  • Isopropyl isocyanate (0.46 niL, 1.3 eq.) was added dropwise to a portion of the solution from Part B of Example 26 (21 mL) at room temperature. The resulting solution was stirred for 1 hour, diluted with dichloromethane (100 mL), washed with water (70 mL), dried over magnesium sulfate, filtered, and concentrated under reduced pressure to provide 1.48 g of a tan foam.
  • the foam was purified by automated flash chromatography (eluting with a gradient of 0-10% CMA in chloroform over 8 column volumes) to provide 1.05 g of 4-(4-bromo-5-cyano-2-propyl-2#-pyrazol ⁇ 3-ylmemyl)-4-fluoro-iV- isopropylpiperidine- 1 -carboxamide as a white foam.
  • Part B 4-(4-bromo-5-cyano-2-propyl-2#-pyrazol ⁇ 3-ylmemyl)-4-fluoro-iV- isopropylpiperidine- 1 -carboxamide as a white foam.
  • Certain exemplary compounds including some of those described above in the Examples, have the following Formulas (IH-I 5 IV-I, V-I, or VIII-I) and the following R 2 , Z, Rj, and m substituents or variables, wherein each line of the table is matched with
  • Certain exemplary compounds including some of those described above in the Examples, have the following Formulas (III-2. IV-2, V-2, and VIII-2) wherein R 2 , Q, R 4 , R 1 , and m are defined immediately below in the table. Each row of the table is matched . with Formula III-2, IV-2, V-2, or VIII-2 to represent a specific embodiment of the invention.
  • Certain exemplary compounds including some of those described above in the Examples, have the following Formulas (III-4. IV-4, V-4, and VIII-4) wherein R 2 and m are defined immediately below in the table. Each row of the table is matched with Formula III-4, IV-4, V-4, or VIII-4 to represent a specific embodiment of the invention.
  • Compounds of the invention have been found to modulate cytokine biosynthesis by inducing the production of interferon ⁇ and/or tumor necrosis factor ⁇ in human cells when tested using one of the methods described below.
  • PBMC Peripheral blood mononuclear cells
  • DPBS Dulbecco's Phosphate Buffered Saline
  • HBSS Hank's Balanced Salts Solution
  • whole blood is placed in Accuspin (Sigma) or LeucoSep (Greiner Bio-One, Inc., Longwood, FL) centrifuge frit tubes containing density gradient medium.
  • Accuspin Sigma
  • LeucoSep Garnier Bio-One, Inc., Longwood, FL centrifuge frit tubes containing density gradient medium.
  • the PBMC layer is collected and washed twice with DPBS or HBSS and re-suspended at 4 x 10 6 cells/mL in RPMI complete.
  • the PBMC suspension is added to 96 well flat bottom sterile tissue culture plates containing an equal volume of RPMI complete media containing test compound.
  • the compounds are solubilized in dimethyl sulfoxide (DMSO).
  • DMSO concentration should not exceed a final concentration of 1% for addition to the culture wells.
  • the compounds are generally tested at concentrations ranging from 30-0.014 ⁇ M.
  • Controls include cell samples with media only, cell samples with DMSO only (no compound), and cell samples with reference compound.
  • test compound is added at 60 ⁇ M to the first well containing RPMI complete and serial 3 fold dilutions are made in the wells.
  • the PBMC suspension is then added to the wells in an equal volume, bringing the test compound concentrations to the desired range (usually 30-0.014 ⁇ M).
  • the final concentration of PBMC suspension is 2 x
  • IFN- ⁇ concentration is determined with a human multi-subtype colorimetric sandwich ELISA (Catalog Number 41105) from PBL Biomedical Laboratories, Piscataway, NJ. Results are expressed in pg/mL.
  • the data output of the assay consists of concentration values of TNF- ⁇ and
  • IFN- ⁇ (y-axis) as a function of compound concentration (x-axis).
  • the reference compound used is 2-[4-amino-2-ethoxymethyl-6,7,8,9-tetrahydro- ⁇ , ⁇ - dimethyl-lH-imidazo[4,5-c]qumolin-l-yl]ethanol hydrate (U.S. Patent No. 5,352,784; Example 91) and the expected area is the sum of the median dose values from the past 61 experiments.
  • the minimum effective concentration is calculated based on the background- subtracted, reference-adjusted results for a given experiment and compound.
  • the minimum effective concentration ( ⁇ molar) is the lowest of the tested compound concentrations that induces a response over a fixed cytokine concentration for the tested cytokine (usually 20 pg/mL for IFN- ⁇ and 40 pg/mL for TNF- ⁇ ).
  • the maximal response is the maximal amount of cytokine (pg/ml) produced in the dose-response.
  • PBMC Peripheral blood mononuclear cells
  • the compounds are solubilized in dimethyl sulfoxide (DMSO).
  • DMSO dimethyl sulfoxide
  • Controls include cell samples with media only, cell samples with DMSO only (no compound), and cell samples with a reference compound 2-[4 ⁇ amino-2-ethoxymethyl-6,7,8,9-tetrahydro- ⁇ , ⁇ -dimethyl- lH-imidazo[4,5-c]quinolin-l-yl]ethanol hydrate (U.S. Patent No. 5,352,784; Example 91) on each plate.
  • test compound is added at 7.5 mM to the first well of a dosing plate and serial 3 fold dilutions are made for the 7 subsequent concentrations in DMSO.
  • RPMI Complete media is then added to the test compound dilutions in order to reach a final compound concentration of 2-fold higher (60 - 0.028 ⁇ M) than the final tested concentration range. Incubation
  • test compound solution is then added to the wells containing the PBMC suspension bringing the test compound concentrations to the desired range (usually 30 ⁇ M - 0.014 ⁇ M) and the DMSO concentration to 0.4%.
  • the final concentration of PBMC suspension is 2x10 6 cells/mL.
  • the plates are covered with sterile plastic lids, mixed gently and then incubated for 18 to 24 hours at 37°C in a 5% carbon dioxide atmosphere.
  • the data output of the assay consists of concentration values of TNF- ⁇ or
  • IFN- ⁇ (y-axis) as a function of compound concentration (x-axis).
  • a plate-wise scaling is performed within a given experiment aimed at reducing plate-to-plate variability associated within the same experiment.
  • the greater of the median DMSO (DMSO control wells) or the experimental background (usually 20 pg/mL for IFN- ⁇ and 40 pg/mL for TNF- ⁇ ) is subtracted from each reading. Negative values that may result from background subtraction are set to zero.
  • Each plate within a given experiment has a reference compound that serves as a control. This control is used to calculate a median expected area under the curve across all plates in the assay.
  • a plate- wise scaling factor is calculated for each plate as a ratio of the area of the reference compound on the particular plate to the median expected area for the entire experiment.
  • the data from each plate are then multiplied by the plate-wise scaling factor for all plates. Only data from plates bearing a scaling factor of between 0.5 and 2.0 (for both cytokines IFN- ⁇ , TNF- ⁇ ) are reported. Data from plates with scaling factors outside the above- mentioned interval are retested until they bear scaling factors inside the above mentioned interval. The above method produces a scaling of the y- values without altering the shape of the curve.
  • the reference compound used is 2-[4-amino-2-ethoxymethyl-6,7,8,9- tetrahydro- ⁇ , ⁇ -dimethyl-lH-imidazo[4,5-c]quinolin-l-yl]ethanol hydrate (U.S. Patent No. 5,352,784; Example 91).
  • the median expected area is the median area across all plates that are part of a given experiment.
  • a second scaling may also be performed to reduce inter-experiment variability
  • the adjustment ratio is the area of the reference compound in the new experiment divided by the expected area of the reference compound based on an average of previous experiments (unadjusted readings). This results in the scaling of the reading (y-axis) for the new data without changing the shape of the dose-response curve.
  • the reference compound used is 2-[4- amino-2-ethoxymethyl-6,7,8,9-tetrahydro-a,a-dimethyl-lH-imidazo[4,5-c]quinolin-l- yljethanol hydrate (U.S. Patent No. 5,352,784; Example 91) and the expected area is the sum of the median dose values from an average of previous experiments.
  • the minimum effective concentration is calculated based on the background- subtracted, reference-adjusted results for a given experiment and compound.
  • the minimum effective concentration ( ⁇ molar) is the lowest of the tested compound concentrations that induces a response over a fixed cytokine concentration for the tested cytokine (usually 20 pg/mL for IFN- ⁇ and 40 pg/mL for TNF- ⁇ ).
  • the maximal response is the maximal amount of cytokine (pg/ml) produced in the dose-response.

Abstract

Pyrazolo[3,4-c] ring compounds of Formula (I), e.g., pyrazolo[3,4-c]pyridines, pyrazolo[3,4-c]quinolines, 6,7,8,9-tetrahydro pyrazolo[3,4-c]quinolines, and pyrazolo[3,4-c]naphthyridines, substituted at the 1-position, pharmaceutical compositions containing the compounds, intermediates, methods of making and methods of use of these compounds as immunomodulators, for inducing cytokine biosynthesis in animals and in the treatment of diseases including viral and neoplastic diseases are disclosed.

Description

1-SUBSTITUTED PYRAZOLO (3 , 4-C) RING COMPOUNDS AS MODULATORS OF CYTOKINE BIOSYNTHESIS FOR THE TREATMENT OF VIRAL INFECTIONS AND NEOPLASTIC DISEASES
CROSS REFERENCE TO RELATED APPLICATIONS
The present invention claims priority to U.S. Provisional Application Serial No. 60/667,869, filed April 1, 2005, and U.S. Provisional Application Serial No. 60/733,037, filed November 3, 2005, both of which are incorporated herein by reference.
BACKGROUND
Certain compounds have been found to be useful as immune response modifiers (IRMs), rendering them useful in the treatment of a variety of disorders. However, there continues to be interest in and a need for compounds that have the ability to modulate the immune response, by induction of cytokine biosynthesis or other mechanisms.
SUMMARY
The present invention provides a new class of compounds that are useful in inducing cytokine biosynthesis in animals. Such compounds are of the following Formula
I:
Figure imgf000002_0001
I wherein R1, R2, RA, RB3 X, Z, and m are as defined below.
The compounds of Formula I are useful as immune response modifiers due to their ability to induce cytokine biosynthesis (e.g., induces the synthesis of at least one cytokine) and otherwise modulate the immune response when administered to animals. This makes the compounds useful in the treatment of a variety of conditions such as viral diseases and tumors that are responsive to such changes in the immune response. The invention further provides pharmaceutical compositions containing an effective amount of a compound of Formula I and methods of inducing cytokine biosynthesis in an animal, treating a viral infection or disease and/or treating a neoplastic disease in an animal by administering an effective amount of a compound of Formula I to the animal.
In addition, methods of synthesizing compounds of Formula I and intermediates useful in the synthesis of these compounds are provided.
As used herein, "a," "an," "the," "at least one," and "one or more" are used interchangeably. The terms "comprises" and variations thereof do not have a limiting meaning where these terms appear in the description and claims.
The above summary of the present invention is not intended to describe each disclosed embodiment or every implementation of the present invention. The description that follows more particularly exemplifies illustrative embodiments. In several places throughout the description, guidance is provided through lists of examples, which examples can be used in various combinations. In each instance, the recited list serves only as a representative group and should not be interpreted as an exclusive list.
DETAILED DESCRIPTION OF ILLUSTRATIVE EMBODIMENTS OF THE INVENTION
The present invention provides compounds of the following Formula I:
Figure imgf000003_0001
I and more specifically compounds of the following Formulas II, III, IV, V, VI, VII, VIII, XIII, and XIV:
Figure imgf000004_0001
II
Figure imgf000004_0002
IV
Figure imgf000004_0003
10 V
Figure imgf000005_0001
VI
Figure imgf000005_0002
VII
Figure imgf000005_0003
VIII
Figure imgf000006_0001
XIII
Figure imgf000006_0002
XIV wherein RA, RB5 RA'> RB1, R, Rh R2, m, n, p, X, Z, G1, and G2 are as defined below; and pharmaceutically acceptable salts thereof.
In one embodiment, the present invention provides a compound of the Formula I:
Figure imgf000006_0003
wherein:
Z is selected from the group consisting of:
Figure imgf000006_0004
Figure imgf000007_0001
X is selected from the group consisting of a bond, -C2-3 alkylene-, and
-O-C2-3 alkylene-;
Ri is selected from the group consisting of: hydrogen, hydroxy, fluorine, alkoxy,
Figure imgf000007_0002
Figure imgf000008_0001
with the proviso that when Z is a bond, C1-5 alkylene,
Figure imgf000008_0002
, and X is a bond, then Ri is other than hydrogen; m is an integer from 1 to 5; RA and RB are each independently selected from the group consisting of: hydrogen, halogen, alkyl, alkenyl, alkoxy, alkylthio, and
-N(Re)2; or when taken together, RA and RB form a fused aryl ring or heteroaryl ring containing one heteroatom selected from the group consisting of N and S wherein the aryl or heteroaryl ring is unsubstituted or substituted by one or more R groups; or when taken together, RA and RB form a fused 5 to 7 membered saturated ring, optionally containing one heteroatom selected from the group consisting of N and S, and unsubstituted or substituted by one or more R groups;
R is selected from the group consisting of: halogen, hydroxy, alkyl, haloalkyl, alkoxy, and -N(R9)2;
R2 is selected from the group consisting of: hydrogen, alkyl, alkoxyalkylenyl, hydroxyalkylenyl, haloalkylenyl, and R4-C(Re)-O-C1-4 alkylenyl;
R4 is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, aryl, aryklkylenyl, aryloxyalkylenyl, alkylarylenyl, heteroaryl, heteroarylalkylenyl, heteroaryloxyalkylenyl, alkylheteroarylenyl, and heterocyclyl, wherein the alkyl, alkenyl, alkynyl, aryl, arylalkylenyl, aryloxyalkylenyl, alkylarylenyl, heteroaryl, heteroarylalkylenyl, heteroaryloxyalkylenyl, alkylheteroarylenyl, and heterocyclyl groups can be unsubstituted or substituted by one or more substituents independently selected from the group consisting of alkyl, alkoxy, hydroxyalkyl, haloalkyl, haloalkoxy, halogen, nitro, hydroxy, mercapto, cyano, aryl, aryloxy, arylalkyleneoxy, heteroaryl, heteroaryloxy, heteroarylalkyleneoxy, heterocyclyl, amino, acetylamino, alkylamino, dialkylamino, (dialkylamino)alkyleneoxy, and in the case of alkyl, alkenyl, alkynyl, and heterocyclyl, oxo; R6 is selected from the group consisting of =0 and =S;
R8 is selected from the group consisting of hydrogen, alkyl, alkoxyalkylenyl, and arylalkylenyl;
R9 is selected from the group consisting of hydrogen and alkyl; R11 is selected from the group consisting of hydrogen, alkyl, halogen, and trifiuoromethyl;
A is selected from the group consisting of -O-, -C(O)-, -S(O)0-2-, -CH2-, and -N(R4)-;
Q is selected from the group consisting of a bond, -C(R6)-, -C(R6)-C(R6)-, -S(O)2-, -C(Rg)-N(Rg)-W-, -S(O)2-N(R8)-, -C(Re)-O-, and -C(Re)-N(OR9)-; V is selected from the group consisting of -C(R6)-, -0-C(R6)-, -N(Rs)-C(R6)-, and
-S(O)2-;
W is selected from the group consisting of a bond, -C(O)-, and -S(O)2-; and a and b are independently integers from 1 to 6 with the proviso that a + b is < 7; or a pharmaceutically acceptable salt thereof.
In another embodiment, the present invention provides a compound of the Formula II:
Figure imgf000010_0001
II wherein:
Z, X, Ri, R2, and m are as defined in Formula I; and RA 1 and RB 1 are each independently selected from the group consisting of: hydrogen, halogen, alkyl, alkenyl, alkoxy, alkylthio, and
-N(Re)2; or a pharmaceutically acceptable salt thereof.
In another embodiment, the present invention provides a compound of the Formula III:
Figure imgf000010_0002
wherein:
Z, X, R], R-2, R, and m are as defined in Formula I; and n is an integer from O to 4; or a pharmaceutically acceptable salt thereof.
In another embodiment, the present invention provides a compound of the Formula IV:
Figure imgf000011_0001
IV wherein:
Z, X, Ri, R2, R, and m are as defined in Formula I; and n is an integer from 0 to 4; or a pharmaceutically acceptable salt thereof.
In another embodiment, the present invention provides a compound selected from the group consisting of the Formulas V, VI, VII, and VIII:
Figure imgf000011_0002
v VI VII
Figure imgf000012_0001
VIII wherein:
Z, X, Ri, R2, R, and m are as defined in Formula I; and p is an integer from O to 3; or a pharmaceutically acceptable salt thereof.
In another embodiment, the present invention provides a compound selected from the group consisting of the Formulas IX, X, XI, and XII:
Figure imgf000012_0002
XII wherein:
Z, X, R1, R2, R, and m are as defined in Formula I; and p is an integer from 0 to 3; or a pharmaceutically acceptable salt thereof.
In another embodiment, the present invention provides a compound of the Formula XIII:
Figure imgf000013_0001
XIII wherein:
R1, R2, RA, RB, X5 Z, and m are as defined in Formula I, G1 is selected from the group consisting of:
-C(O)-R', α-aminoacyl, α-aminoacyl-α-aminoacyl,
Figure imgf000013_0002
R' and R" are independently selected from the group consisting of CMO alkyl, C3-7 cycloalkyl, phenyl, and benzyl, each of which may be unsubstituted or substituted by one or more substituents selected from the group consisting of halogen, hydroxy, nitro, cyano, carboxy, Ci-6 alkyl, C^ alkoxy, aryl, heteroaryl, aryl-Ci-4 alkylenyl, heteroary 1-Ci-4 alky lenyl, halo-Ci-4 alkylenyl, halo-Ci-4 alkoxy, -0-C(O)-CH3, -C(O)-O-CH3, -C(O)-NH2, -0-CH2-C(O)-NH2, -NH2, and -S(O)2-NH2, with the proviso that R" can also be hydrogen; α-aminoacyl is an α-aminoacyl group derived from an amino acid selected from the group consisting of racemic, D-, and L-amino acids;
Y' is selected from the group consisting of hydrogen, C]-6 alkyl, and benzyl; Y0 is selected from the group consisting of Ci-6 alkyl, carboxy-Ci-6 alkylenyl, amino-C i -4 alkylenyl, mono-iV-C i -6 alkylamino-C i -4 alkylenyl, and di--YiV-Ci-6 alkylamino-C1-4 alkylenyl; and
Y2 is selected from the group consisting of mono-N-C1-6 alkylamino, di-N, N-Ci-6 alkylamino, morpholin-4-yl, piperidin-1-yl, pyrrolidin-1-yl, and 4-Ci-4 alkylpiperazin-l-yl; or a pharmaceutically acceptable salt thereof.
In another embodiment, the present invention provides a compound of the Formula XIV:
Figure imgf000014_0001
XIV wherein:
RA, RB, R2, Z, and m are as defined in Formula I, G2 is selected from the group consisting of: -X2-C(O)-R1, α-aminoacyl, α-aminoacyl-α-aminoacyl,
-X2-C(O)-O-R1, -C(O)-N(R")R'5 and -S(O)2-R1;
X2 is selected from the group consisting of a bond; -CH2-O-; -CH(CH3)-O-; -C(CH3)2-O-; and, in the case of -X2-C(O)-O-R1, -CH2-NH-;
R' and R" are independently selected from the group consisting OfC1-10 alkyl, C3-7 cycloalkyl, phenyl, and benzyl, each of which may be unsubstituted or substituted by one or more substituents independently selected from the group consisting of halogen, hydroxy, nitro, cyano, carboxy, Cj.β alkyl, Ci^ alkoxy, aryl, heteroaryl, aryl-C^ alkylenyl, heteroaryl-Ci-4 alkylenyl, halo-C1-4 alkylenyl, halo-C1-4 alkoxy, -0-C(O)-CH3, -C(O)-O-CH3, -C(O)-NH2, -0-CH2-C(O)-NH2, -NH2, and -S(O)2-NH2, with the proviso that R" can also be hydrogen; and α-aminoacyl is an α-aminoacyl group derived from an amino acid selected from the group consisting of racemic, D-, and L-amino acids; or a pharmaceutically acceptable salt thereof.
As used herein, the terms "alkyl", "alkenyl", "alkynyl" and the prefix "alk-" are inclusive of both straight chain and branched chain groups and of cyclic groups, e.g., cycloalkyl and cycloalkenyl. Unless otherwise specified, these groups contain from 1 to 20 carbon atoms, with alkenyl groups containing from 2 to 20 carbon atoms, and alkynyl groups containing from 2 to 20 carbon atoms. In some embodiments, these groups have a total of up to 10 carbon atoms, up to 8 carbon atoms, up to 6 carbon atoms, or up to 4 carbon atoms. Cyclic groups can be monocyclic or polycyclic and preferably have from 3 to 10 ring carbon atoms. Exemplary cyclic groups include cyclopropyl, cyclopropylmethyl, cyclopentyl, cyclohexyl, adamantyl, and substituted and unsubstituted bornyl, norbornyl, and norbornenyl.
Unless otherwise specified, "alkylene", "-alkylene-", "alkenylene", "-alkenylene-", "alkynylene", and "-alkynylene-" are the divalent forms of the "alkyl", "alkenyl", and "alkynyl" groups defined above. The terms "alkylenyl", "alkenylenyl", and "alkynylenyl" are used when "alkylene", "alkenylene", and "alkynylene", respectively, are substituted. For example, an arylalkylenyl group comprises an "alkylene" moiety to which an aryl group is attached.
The term "haloalkyl" is inclusive of groups that are substituted by one or more halogen atoms, including perfluorinated groups. This is also true of other groups that include the prefix "halo-." Examples of suitable haloalkyl groups are chloromethyl, trifluoromethyl, and the like.
The term "aryl" as used herein includes carbocyclic aromatic rings or ring systems. Examples of aryl groups include phenyl, naphthyl, biphenyl, fluorenyl and indenyl. Unless otherwise indicated, the term "heteroatom" refers to the atoms O, S, or N. The term "heteroaryl" includes aromatic rings or ring systems that contain at least one ring heteroatom (e.g., O, S5 N). In some embodiments, the term "heteroaryl" includes a ring or ring system that contains 2-12 carbon atoms, 1-3 rings, 1-4 heteroatoms, and O, S, and N as the heteroatoms. Exemplary heteroaryl groups include furyl, thienyl, pyridyl, quinolinyl, isoquinolinyl, indolyl, isoindolyl, triazolyl, pyrrolyl, tetrazolyl, imidazolyl, pyrazolyl, oxazolyl, thiazolyl, benzofuranyl, benzothiophenyl, carbazolyl, benzoxazolyl, pyrimidinyl, benzimidazolyl, quinoxalinyl, benzothiazolyl, naphthyridinyl, isoxazolyl, isothiazolyl, purinyl, quinazolinyl, pyrazinyl, 1-oxidopyridyl, pyridazinyl, triazinyl, tetrazinyl, oxadiazolyl, thiadiazolyl, and so on.
The term "heterocyclyl " includes non-aromatic rings or ring systems that contain at least one ring heteroatom (e.g., O, S, N) and includes all of the fully saturated and partially unsaturated derivatives of the above mentioned heteroaryl groups. In some embodiments, the term "heterocyclyl" includes a ring or ring system that contains 2-12 carbon atoms, 1-3 rings, 1-4 heteroatoms, and O, S, and N as the heteroatoms. Exemplary heterocyclyl groups include pyrrolidinyl, tetrahydrofuranyl, morpholinyl, thiomorpholinyl, 1,1- dioxothiomorpholinyl, piperidinyl, piperazinyl, thiazolidinyl, imidazolidinyl, isothiazolidinyl, tetrahydropyranyl, quinuclidinyl, homopiperidinyl (azepanyl), 1,4- oxazepanyl, homopiperazinyl (diazepanyl), 1,3-dioxolanyl, aziridinyl, azetidinyl, dihydroisoquinolin-(lH)-yl, octahydroisoquinolin-(lH)-yl, dihydroquinolin-(2H)~yl, octahydroquinolin-(2H)-yl, dihydro-lH-imidazolyl, 3-azabicyclo[3.2.2]non-3-yl, and the like.
The term "heterocyclyl" includes bicyclic and tricyclic heterocyclic ring systems. Such ring systems include fused and/or bridged rings and spiro rings. Fused rings can include, in addition to a saturated or partially saturated ring, an aromatic ring, for example, a benzene ring. Spiro rings include two rings joined by one spiro atom and three rings joined by two spiro atoms.
When "heterocyclyl" contains a nitrogen atom, the point of attachment of the heterocyclyl group may be the nitrogen atom. The terms "arylene", "heteroaryl ene", and "heterocyclylene" are the divalent forms of the "aryl", "heteroaryl", and "heterocyclyl" groups defined above. The terms, "arylenyl", "heteroarylenyl", and "heterocyclylenyl" are used when "arylene", "heteroarylene", and "heterocyclylene", respectively, are substituted. For example, an alkylarylenyl group comprises an arylene moiety to which an alkyl group is attached.
The term "fused aryl ring" includes fused carbocyclic aromatic rings or ring systems. Examples of fused aryl rings include benzo, naphtho, fluoreno, and indeno. The term "fused heteroaryl ring" includes the fused forms of 5 or 6 membered aromatic rings that contain one heteroatom selected from S and N.
The term "fused 5 to 7 membered saturated ring" includes rings which are fully saturated except for the bond where the ring is fused.
When a group (or substituent or variable) is present more than once in any formula described herein, each group (or substituent or variable) is independently selected, whether explicitly stated or not. For example, for the formula -N(R^- each R9 group is independently selected. In another example, when an Rj and a Z group both contain an R4 group, each R4 group is independently selected.
The invention is inclusive of the compounds described herein (including intermediates) in any of their pharmaceutically acceptable forms, including isomers (e.g., diastereomers and enantiomers), salts, solvates, polymorphs, prodrugs, and the like. In particular, if a compound is optically active, the invention specifically includes each of the compound's enantiomers as well as racemic mixtures of the enantiomers. It should be understood that the term "compound" includes any or all of such forms, whether explicitly stated or not (although at times, "salts" are explicitly stated).
The term "prodrug" means a compound that can be transformed in vivo to yield an immune response modifying compound, including any of the salt, solvated, polymorphic, or isomeric forms described above. The prodrug, itself, may be an immune response modifying compound, including any of the salt, solvated, polymorphic, or isomeric forms described above. The transformation may occur by various mechanisms, such as through a chemical (e.g., solvolysis or hydrolysis, for example, in the blood) or enzymatic biotransformation. A discussion of the use of prodrugs is provided by T. Higuchi and W. Stella, "Pro-drugs as Novel Delivery Systems," Vol. 14 of the A. C. S. Symposium Series, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987.
Compounds (including intermediates) of the present invention may exist in different tautomeric forms, and all such forms are embraced within the scope of the invention. The term "tautomer" or "tautomeric form" refers to structural isomers of different energies which are interconvertible via a low energy barrier. For example, proton tautomers (prototropic tautomers) include interconversions via migration of a proton, such as keto-enol and imine-enamine isomerizations. When compounds of the present invention have a hydrogen atom at the 2 position, proton migration between the 2 and 3 positions may occur.
The compounds of the present invention may exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like. The present invention embraces both solvated and unsolvated forms. For any of the compounds presented herein, each one of the following variables
(e.g., RA, RB5 RAJ 5 RB1, R5 RI, Ra5 m, n, p, A3 Q5 X, Z, and so on) in any of its embodiments can be combined with any one or more of the other variables in any of their embodiments and associated with any one of the formulas described herein, as would be understood by one oflskill in the art. Each of the resulting combinations of variables is an embodiment of the present invention.
In certain embodiments (e.g., of Formula I), RA and RB are each independently selected from the group consisting of hydrogen, halogen, alkyl, alkenyl, alkoxy, alkylthio, and -N(R9)2; or when taken together, RA and RB form a fused aryl ring or heteroaryl ring containing one heteroatom selected from the group consisting of N and S wherein the aryl or heteroaryl ring is unsubstituted or substituted by one or more R groups; or when taken together, RA and RB form a fused 5 to 7 membered saturated ring, optionally containing one heteroatom selected from the group consisting of N and S, and unsubstituted or substituted by one or more R groups. In certain embodiments (e.g., of Formula I)5 RA and RB are each independently selected from the group consisting of hydrogen, halogen, alkyl, alkenyl, alkoxy, alkylthio, and -N(R9)2.
In certain embodiments (e.g., of Formula I), RA and RB form a fused aryl or heteroaryl ring. In certain embodiments (e.g., of Formula I)5 RA and RB form a fused aryl ring. In certain embodiments, the fused aryl ring is benzo. In certain embodiments (e.g., of Formula I), RA and RB form a fused heteroaryl ring. In certain embodiments, the fused heteroaryl ring is pyrido or thieno. In certain embodiments, the fused heteroaryl ring is pyrido. In certain of these embodiments, the pyrido ring is
Figure imgf000019_0001
or wherein the highlighted bond indicates the position where the ring is fused.
In certain embodiments (e.g., of Formula I), RA and RB form a fused 5 to 7 membered saturated ring. In certain embodiments, the ring is a cyclohexene ring. In certain embodiments (e.g., of Formula I), RA and RB form a fused 5 to 7 membered saturated ring containing one heteroatom selected from the group consisting of N and S. In certain embodiments, the ring is tetrahydropyrido or dihydrothieno. In certain embodiments the heteroatom is N. In certain embodiments, the ring is tetrahydropyrido.
In certain of these embodiments, the ring is
Figure imgf000019_0002
or wherein the highlighted bond indicates the position where the ring is fused. In certain embodiments (e.g., of Formula II), RA1 and RB 1 are each independently selected from the group consisting of hydrogen, halogen, alkyl, alkenyl, alkoxy, alkylthio, and -N(R9)2.
In certain embodiments (e.g., of Formula II), RA1 and RB 1 are independently hydrogen or alkyl. In certain embodiments (e.g., of Formula II), RA and RB 1 are both methyl.
In certain embodiments (e.g., of any one of Formulas IX through XII), R is selected from the group consisting of alkyl and haloalkyl.
In certain embodiments (e.g., of any one of the above embodiments of Formulas I, III through VIII, XIII, and XIV where R is present), R is selected from the group consisting of alkyl, alkoxy, halogen, and hydroxy.
In certain embodiments (e.g., of any one of the above embodiments of Formulas I, III through VIII, XIII, and XIV where R is present), R is hydroxy.
In certain embodiments (e.g., of Formulas III or IV), n is 0.
In certain embodiments (e.g., of any one of Formulas V through XII), p is 0. In certain embodiments, the compound or salt selected from the group consisting of Formulas V, VI, VII, and VIII is the compound of Formula V or Formula VIII:
Figure imgf000020_0001
v VIII; or a pharmaceutically acceptable salt thereof.
In certain embodiments (e.g., of any one of Formulas I through XIV), m is an integer from 1 to 5.
In certain embodiments, including any one of the above embodiments, m is an integer from 1 to 3. In certain embodiments, m is 1.
In certain embodiments (e.g., of any one of Formulas I through XIII), X is selected from the group consisting of a bond, -C2-3 alkylene-, and -0-C2-3 alkylene-.
In certain embodiments, including any one of the above embodiments, X is a bond.
In certain embodiments (e.g., of any one of Formulas I through XIII), R1 is selected from the group consisting of hydrogen, hydroxy, fluorine, alkoxy, -N(Rg)2, -NH-Q-R4, -S(0)o-2-alkyl, -S(O)2-NH-R9, -C(Re)-N(Rg)-R4, -0-C(Rg)-N(Rs)-R4, -C(R6)-O-alkyl, -0-C(Re)-R4, and
Figure imgf000020_0002
wjtø tøe prOyjsoat when Z is a bond, CJ-5 alkylene,
Figure imgf000020_0003
, and X is a bond, then R1 is other than hydrogen.
In certain embodiments, including any one of the above embodiments wherein R1 is present, R1 is selected from the group consisting of hydroxy and methoxy.
In certain embodiments, including any one of the above embodiments wherein Ri is present, Rj is fluoro, except where Rj is otherwise defined. In certain embodiments, including any one of the above embodiments wherein Rj is present, Ri is selected from the group consisting of -NH2, -NH-Q-R4, -C(O)-NH2, and -C(O)-N(Rs)-R4, wherein Q is selected from the group consisting of -C(O)-, -S(O)2-, -C(O)-O-, and -C(O)-NH-, R8 is selected from the group consisting of hydrogen and alkyl, and R4 is selected from the group consisting of alkyl and alkoxyalkylenyl, except where R1 is otherwise defined.
In certain embodiments (e.g., of any one of Formulas I through XIV), Z is selected from the group consisting of:
Figure imgf000021_0003
bond, C1-5 alkylene,
Figure imgf000021_0001
In certain embodiments, including any one of the above embodiments, Z is selected from the group consisting of a bond and C1-3 alkylene.
In certain embodiments, including any one of the above embodiments, Z is selected from the group consisting of:
Figure imgf000021_0004
where Z is otherwise defined. In certain of these embodiments, Z is
Figure imgf000021_0002
In certain embodiments, including any one of the above embodiments wherein X is a bond and Z is ^ R] is hydrogen.
Figure imgf000022_0001
In certain embodiments, including embodiments of Formula III wherein n is 0, and embodiments of Formulas V, VI, VII, or VIII wherein p is 0, and embodiments of Formulas IX, X, XI, or XI wherein p is O5 Z is
Figure imgf000022_0002
, X is a bond, and Ri is hydrogen,
In certain embodiments, including any one of the above embodiments, Z is
Figure imgf000022_0003
9 except where Z is otherwise defined.
In certain embodiments, including any one of the above embodiments wherein Q is present, Q is selected from the group consisting of -C(O)-, -C(O)-O-, -S(O)2-, and -C(Re)-N(R8)-.
In certain embodiments, including any one of the above embodiments wherein R4 is present, R4 is selected from the group consisting of alkyl, aryl, arylalkylene, heteroaryl, and heterocyclyl, wherein the aryl group can be unsubstituted or substituted by one or more substituents independently selected from the group consisting of acetylamino, alkyl, alkoxy, cyano, and halogen.
In certain embodiments (e.g., of any one of Formulas I through XIV), R2 is selected from the group consisting of hydrogen, alkyl, alkoxyalkylenyl, hydroxyalkylenyl, haloalkylenyl, and R4-C(ReO-O-Ci-4 alkylenyl.
In certain embodiments, including any one of the above embodiments, R2 is selected from the group consisting of Cj-4 alkyl, Cj-4 alkyl-O-C2-4 alkylenyl, and hydroxyC2-4 alkylenyl.
In certain embodiments, including any one of the above embodiments, R2 is selected from the group consisting of methyl, ethyl, ^-propyl, «-butyl, 2-methoxy ethyl, and 2-hydroxyethyl. For certain embodiments, R4 is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, aryl, arylalkylenyl, aryloxyalkylenyl, alkylarylenyl, heteroaryl, heteroarylalkylenyl, heteroaryloxyalkylenyl, alkylheteroarylenyl, and heterocyclyl, wherein the alkyl, alkenyl, alkynyl, aryl, arylalkylenyl, aryloxyalkylenyl, alkylarylenyl, heteroaryl, heteroarylalkylenyl, heteroaryloxyalkylenyl, alkylheteroarylenyl, and heterocyclyl groups can be unsubstituted or substituted by one or more substituents independently selected from the group consisting of alkyl, alkoxy, hydroxyalkyl, haloalkyl, haloalkoxy, halogen, nitro, hydroxy, mercapto, cyano, aryl, aryloxy, arylalkyleneoxy, heteroaryl, heteroaryloxy, heteroarylalkyleneoxy, heterocyclyl, amino, acetylamino, alkylamino, dialkylamino, (dialkylamino)alkyleneoxy, and in the case of alkyl, alkenyl, alkynyl, and heterocyclyl, oxo.
For certain embodiments, R4 is selected from the group consisting of alkyl and alkoxyalkylenyl .
For certain embodiments, R4 is selected from the group consisting of alkyl, aryl, arylalkylene, heteroaryl, and heterocyclyl, wherein the aryl group can be unsubstituted or substituted by one or more substituents independently selected from the group consisting of acetylamino, alkyl, alkoxy, cyano, and halogen. For certain embodiments, R6 is selected from the group consisting of =0 and =S.
In certain embodiments, R6 is =0. In certain embodiments, R6 is =S.
For certain embodiments, R8 is selected from the group consisting of hydrogen, alkyl, alkoxyalkylenyl, and arylalkylenyl. In certain embodiments, R8 is selected from the group consisting of hydrogen and alkyl. In certain embodiments, R8 is hydrogen. In certain embodiments, R8 is alkyl.
For certain embodiments, R9 is selected from the group consisting of hydrogen and alkyl. In certain embodiments, R9 is alkyl. In certain embodiments, R9 is hydrogen.
For certain embodiments, R11 is selected from the group consisting of hydrogen, alkyl, halogen, and trifluoromethyl. For certain embodiments, A is selected from the group consisting of -O-, -C(O)-,
-S(O)0-2-, -CH2-, and -N(R4)-. For certain embodiments, A is -0-.
For certain embodiments, Q is selected from the group consisting of a bond, -C(R6)-, -C(R6)-C(R6)-, -S(O)2-, -C(Re)-N(Re)-W-, -S(O)2-N(R8)-, -C(Re)-O-, and -C(R6)-N(OR9)-. For certain embodiments, Q is selected from the group consisting of -C(O)-, -S(O)2-, -C(O)-O-, and -C(O)-NH-. For certain embodiments, Q is selected from the group consisting of -C(O)-, -C(O)-O-, -S(O)2-, and -C(Re)-N(R8)- . For certain embodiments, Q is a bond. For certain embodiments, V is selected from the group consisting Of-C(R6)-, -0-C(R6)-, -N(Rs)-C(R6)-, and -S(O)2-. In certain embodiments, V is -C(R6)-. In certain embodiments, V is -N(Rg)-C(Re)-.
For certain embodiments, W is selected from the group consisting of a bond, -C(O)-, and -S(O)2-. In certain embodiments, W is selected from the group consisting of a bond and -C(O)-. In certain embodiments, W is a bond.
For certain embodiments, a and b are independently integers from 1 to 6 with the proviso that a + b is < 7. For certain embodiments, a and b are each the integer 2.
For certain embodiments, Gi is selected from the group consisting of -C(O)-R', α- aminoacyl, α-aminoacyl-α-aminoacyl, -C(O)-O-R', -C(O)-N(R")R', -C(=NY')-R', -CH(OH)-C(O)-OY', -CH(OCi-4 alkyl)Y0, -CH2Y2, and -CH(CH3)Y2.
For certain embodiments, G2 is selected from the group consisting of -X2-C(O)-R', α-aminoacyl, α-aminoacyl-α-aminoacyl, -X2-C(O)-O-R', -C(0)-N(R")R', and -S(O)2-R'.
For certain embodiments, R' is selected from the group consisting OfCi-10 alkyl, C3-7 cycloalkyl, phenyl, and benzyl, each of which may be unsubstituted or substituted by one or more substituents independently selected from the group consisting of halogen, hydroxy, nitro, cyano, carboxy, C1-6 alkyl, Ci-4alkoxy, aryl, heteroaryl, aryl-Ci-4 alkylenyl, heteroaryl-C1-4 alkylenyl, halo-C1-4 alkylenyl, halo-Cj-4 alkoxy, -0-C(O)-CH3, -C(O)-O-CH3, -C(O)-NH2, -0-CH2-C(O)-NH2, -NH2, and -S(O)2-NH2. For certain embodiments, R" is selected from the group consisting of hydrogen,
Ci-io alkyl, C3-7 cycloalkyl, phenyl, and benzyl, each of which may be unsubstituted or substituted by one or more substituents independently selected from the group consisting of halogen, hydroxy, nitro, cyano, carboxy, Ci-6 alkyl, Ci-4 alkoxy, aryl, heteroaryl, aryl-Ci-4 alkylenyl, heteroaryl-Ci-4 alkylenyl, halo-Ci-4 alkylenyl, halo-Ci-4 alkoxy, -0-C(O)-CH3, -C(O)-O-CH3, -C(O)-NH2, -0-CH2-C(O)-NH2, -NH2, and -S(O)2-NH2.
For certain embodiments, X2 is selected from the group consisting of a bond; -CH2-O-; -CH(CH3)-0-; -C(CH3)2-O-; and, in the case Of -X2-C(O)-O-R', -CH2-NH-.
For certain embodiments, α-aminoacyl is an α-aminoacyl group derived from an amino acid selected from the group consisting of racemic, D-, and L-amino acids. For certain of these embodiments, the amino acid is a naturally occurring amino acid.
For certain embodiments, Y' is selected from the group consisting of hydrogen, Ci-6 alkyl, and benzyl. For certain embodiments, Y0 is selected from the group consisting of Cj-6 alkyl, carboxy-Ci-6 alkylenyl, amino-Cu alkylenyl, mono-N-Ci-6 alky lamino-C I-4 alkylenyl, and di-N,N-Cu6 alkylamino-Ci_4 alkylenyl.
For certain embodiments, Y2 is selected from the group consisting of mono-JV-Ci-6 alkylamino, di-JV,iV-Ci-6 alkylamino, morpholin-4-yl, piperidin-1-yl, pyrrolidin-1-yl, and 4-Ci-4 alkylpiperazin-l-yl.
For certain embodiments, including any one of the above embodiments of Formula
XIII, Gi is selected from the group consisting of -C(O)-R', α-aminoacyl, and -C(O)-O-R'. For certain of these embodiments, R' contains one to ten carbon atoms. For certain of these embodiments, α-aminoacyl is an α-C2-π aminoacyl group derived from an α-amino acid selected from the group consisting of racemic, D-, and L-amino acids containing a total of at least 2 carbon atoms and a total of up to 11 carbon atoms, and may also include one or more heteroatoms selected from the group consisting of O, S, and N.
For certain embodiments, including any one of the above embodiments of Formula XIV, G2 is selected from the group consisting of -C(O)-R' and α-aminoacyl, wherein R' is C1-6 alkyl or phenyl which is unsubstituted or substituted by one or more substituents independently selected from the group consisting of halogen, hydroxy, nitro, cyano, carboxy, C1-6 alkyl, C1-4 alkoxy, aryl, heteroaryl, aryl-C1-4 alkylenyl, heteroaryl-C1-4 alkylenyl, halo-Ci-4 alkylenyl, halo-C1-4 alkoxy, -0-C(O)-CH3, -C(O)-O-CH3, -C(O)-NH2, -0-CH2-C(O)-NH2, -NH2, and -S(O)2-NH2.
For certain embodiments, including any one of the above embodiments of Formula
XIV, G2 is selected from the group consisting of α-amino-C2.5 alkanoyl, C2-6 alkanoyl, Ci-6alkoxycarbonyl, and C1-6 alkylcarbamoyl.
For certain embodiments, including any one of the above embodiments which include an α-aminoacyl group, α-aminoacyl is an α-aminoacyl group derived from a naturally occuring α-amino acid selected from the group consisting of racemic, D-, and L- amino acids.
For certain embodiments, including any one of the above embodiments which include an α-aminoacyl group, α-aminoacyl is an α-aminoacyl group derived from an α- amino acid found in proteins, wherein the the amino acid is selected from the group consisting of racemic, D-, and L-amino acids. In some embodiments, the present invention provides a pharmaceutical composition comprising a therapeutically effective amount of a compound or salt of any one of Formulas I5 H5 III, IV, V, VI5 VII5 VIII5 IX5 X5 XI5 XII, XIII5 XIV, or any one of the above embodiments in combination with a pharmaceutically acceptable carrier. In some embodiments, the present invention provides a method of inducing cytokine biosynthesis in an animal comprising administering an effective amount of a compound or salt of any one of Formulas I5 H5 III, IV, V, VI, VII, VIII, IX, X, XI5 XII5 XIII, XIV, or any one of the above embodiments or administering any one of the above pharmaceutical compositions to the animal. For certain of these embodiments, the cytokine is selected from the group consisting of IFN-α, TNF-α, IL-6, IL-IO, and IL- 12. For certain of these embodiments, the cytokine is IFN-α or TNF-α.
In some embodiments, the present invention provides a method of treating a viral disease in an animal comprising administering a therapeutically effective amount of a compound or salt of any one of Formulas I5 H5 III, IV5 V5 VI5 VII, VIII, IX, X, XI, XII, XIII5 XIV5 or any one of the above embodiments or administering any one of the above pharmaceutical compositions to the animal.
In some embodiments, the present invention provides a method of treating a neoplastic disease in an animal comprising administering a therapeutically effective amount of a compound or salt of any one of Formulas I, II, III, IV5 V, VI, VII, VIII, IX5 X, XI5 XII, XIII5 XIV, or any one of the above embodiments or administering any one of the above pharmaceutical compositions to the animal.
Preparation of the Compounds
Compounds of the invention may be synthesized by synthetic routes that include processes analogous to those well known in the chemical arts, particularly in light of the description contained herein. The starting materials are generally available from commercial sources such as Aldrich Chemicals (Milwaukee, Wisconsin, USA) or are readily prepared using methods well known to those skilled in the art (e.g. prepared by methods generally described in Louis F. Fieser and Mary Fieser, Reagents for Organic Synthesis, v. 1-19, Wiley, New York, (1967-1999 ed.); Alan R. Katritsky, Otto Meth-
Cohn, Charles W. Rees, Comprehensive Organic Functional Group Transformations, v 1- 6, Pergamon Press, Oxford, England, (1995); Barry M. Trost and Ian Fleming, Comprehensive Organic Synthesis, v. 1-8, Pergamon Press, Oxford, England, (1991); or Beilsteins Handbuch der organischen Chemie, 4, Aufl. Ed. Springer- Verlag, Berlin, Germany, including supplements (also available via the Beilstein online database)).
For illustrative purposes, the reaction schemes depicted below provide potential routes for synthesizing the compounds of the present invention as well as key intermediates. For more detailed description of the individual reaction steps, see the EXAMPLES section below. Those skilled in the art will appreciate that other synthetic routes may be used to synthesize the compounds of the invention. Although specific starting materials and reagents are depicted in the reaction schemes and discussed below, other starting materials and reagents can be easily substituted to provide a variety of derivatives and/or reaction conditions. In addition, many of the compounds prepared by the methods described below can be further modified in light of this disclosure using conventional methods well known to those skilled in the art.
In the preparation of compounds of the invention it may sometimes be necessary to protect a particular functionality while reacting other functional groups on an intermediate. The need for such protection will vary depending on the nature of the particular functional group and the conditions of the reaction step. Suitable amino protecting groups include acetyl, trifluoroacetyl, fert-butoxycarbonyl (Boc), benzyloxycarbonyl, and 9- fiuorenylmethoxycarbonyl (Fmoc). Suitable hydroxy protecting groups include acetyl and silyl groups such as the tert-buty\ dimethylsilyl group. For a general description of protecting groups and their use, see T, W. Greene and P. G. M. Wuts, Protective Groups in Organic Synthesis, John Wiley & Sons, New York, USA5 1991.
Conventional methods and techniques of separation and purification can be used to isolate compounds of the invention, as well as various intermediates related thereto. Such techniques may include, for example, all types of chromatography (high performance liquid chromatography (HPLC), column chromatography using common absorbents such as silica gel, and thin layer chromatography, recrystallization, and differential (i.e., liquid- liquid) extraction techniques.
For some embodiments, compounds of the invention can be prepared according to Reaction Scheme I, where R, R2, Z, n and m are as defined above and Boc is tert- butoxycarbonyl. In step (1) of Reaction Scheme I, an indole of Formula XX is acylated to provide an oxalated indole of Formula XXI. The reaction can be carried out by adding ethyl chlorooxoacetate to a solution of an indole of Formula XX in a suitable solvent such as diethyl ether in the presence of pyridine. The reaction can be carried out at a sub-ambient temperature such as 0 0C. Many indoles of Formula XX are known. Some are commercially available and others can be readily prepared using known synthetic methods.
In step (2) of Reaction Scheme I, an oxalated indole of Formula XXI is rearranged to a pyrazolo[3,4-c]quinolin-4-one of Formula XXII. The reaction can be carried out by adding a hydrazine of Formula R2NHNH2 to a solution of an oxalated indole of Formula
XXI in a solvent or solvent mix such as ethanol/acetic acid in the presence of hydrochloric acid. The reaction can be carried out at an elevated temperature such as at reflux.
If step (2) is carried out using hydrazine, the resulting pyrazolo[3,4-c]quinolin-4- one of Formula XXII where R2 is hydrogen can be further elaborated using known synthetic methods. For example, a pyrazolo[3,4-c]quinolin-4-one of Formula XXII where R2 is hydrogen can alkylated. The alkylation is conveniently carried out by treating a solution of a pyrazolo[3,4-c]quinolin-4-one of Formula XXII, where R2 is hydrogen, with a base such as sodium ethoxide followed by the addition of an alkyl halide. The reaction can be run in a suitable solvent such as ethanol and can be carried out at an elevated temperature, for example, the reflux temperature of the solvent, or at ambient temperature. Alternatively, a pyrazolo[3,4-c]quinolin-4-one of Formula XXII where R2 is hydrogen can undergo a Buchwald amination with an aryl halide or heteroaryl halide. Numerous alkyl halides, aryl halides, and heteroaryl halides are commercially available; others can be prepared using known synthetic methods. In step (3) of Reaction Scheme I, a pyrazolo[3,4-c]quinolin-4-one of Formula XXII is chlorinated to provide a 4-chloropyrazolo[3,4-c]quinoline of Formula XXIII. The reaction can be carried out by combining a pyrazolo[3,4-c]quinolin-4-one of Formula
XXII with phosphorous oxychloride and heating.
In step (4) of Reaction Scheme I, the chloro group of a 4-chloropyrazolo[3,4- cjquinoline of Formula XXIII is displaced to provide a pyrazolo[3,4-c]quinolin-4-amine of Formula XXIV. The reaction can be carried out by combining a compound of Formula
XXIII with a solution of ammonia in methanol and heating the mixture in a sealed reactor. In step (5) of Reaction Scheme I, the amino group of a pyrazolo[3,4-c]quinolin-4- amine of Formula XXIV is protected with two Boc groups using conventional methods.
In step (6) of Reaction Scheme I, a compound of Formula XXV undergoes a lithiation-substitution reaction to provide a compound of Formula XXVI, which is a subgenus of Formula XIII. The reaction can be carried out by treating a compound of Formula XXV with fer^-butyllithium and subsequently adding a cyclic ketone. Other organolithium reagents may be used. The reaction can be carried out below room temperature in a suitable solvent such as diethyl ether, THF, or tert-bxAy\ methyl ether. Conveniently, the reaction can be carried out at -78 0C. Numerous cyclic ketones useful in this transformation are commercially available, such as cyclohexanone, cyclopentanone, tetrahydro-4H~pyran-4-one, and l-Boc-4-piperidone. Others can be made by known methods.
In step (7) of Reaction Scheme I, a Boc protected pyrazolo[3,4-c]quinolin-4-amine of Formula XXVI undergoes acid mediated cleavage to remove the Boc group to provide a compound of Formula XXVII which is a subgenus of Formulas I and III.
Reaction Scheme I
Figure imgf000030_0001
(6)
Figure imgf000030_0002
XXVI XXVII
For some embodiments, compounds of the invention can be prepared according to Reaction Scheme II, where R, R2, Z, n and m are as defined above.
In Reaction Scheme II, compounds of Formula XXIX are prepared from 7- azaindoles of Formula XXVIII using the methods of steps (1) through (7) of Reaction Scheme I. Reaction Scheme II
Figure imgf000031_0001
For some embodiments, compounds of the invention can also be prepared according to Reaction Scheme III, where Rj, R2, X, Z, and m are as defined above, and RA2 and RB2 taken together form a fused benzene ring or fused pyridine ring wherein the benzene ring or pyridine ring is unsubstituted or substituted by one or more R groups.
In step (1) of Reaction Scheme III, a ketone of Formula XXX is condensed with diethyl oxalate under Claisen condensation conditions to provide a ketoester of Formula XXXI. The reaction can be carried out by adding sodium ført-butoxide to a solution of diethyl oxalate and the ketone of Formula XXX in ethanol at ambient temperature.
In step (2) of Reaction Scheme III, a ketoester of Formula XXXI reacts with a hydrazine of Formula R2NHNH2 to provide a pyrazole carboxylate of Formula XXXII. The reaction is conveniently carried out by slowly adding the hydrazine to a solution of a compound of Formula XXXI in a suitable solvent such as acetic acid. The reaction can be carried out at ambient temperature.
In step (3) of Reaction Scheme III, the ester group of a pyrazole carboxylate of Formula XXXII is converted to an amide. The amination can be carried out by adding ammonium hydroxide to the pyrazole carboxylate of Formula XXXII in a suitable solvent such as methanol and heating at an elevated temperature such as 100 0C. The reaction can be carried out in a pressure vessel.
Alternatively, step (3) can be carried out by first hydrolyzing a pyrazole carboxylate of Formula XXXII to a carboxylic acid and then converting the carboxylic acid to an amide. The ester hydrolysis can be carried out under basic conditions by combining a pyrazole carboxylate of Formula XXXII with lithium hydroxide or sodium hydroxide in water and in a suitable solvent such as methanol or ethanol. The reaction can be carried out at ambient temperature, and the carboxylic acid product can be isolated using conventional methods. The conversion of the carboxylic acid to a pyrazole carboxamide of Formula XXXIII can be carried out by first treating the carboxylic acid with oxalyl chloride at ambient temperature in a suitable solvent such as dichloromethane to generate an acid chloride, which can then be treated with ammonium hydroxide at a sub-ambient temperature such as 0 0C. Alternatively, the conversion of the carboxylic acid to a pyrazole carboxamide of Formula XXXIII can be carried out under coupling conditions by adding 1-hydroxybenzotriazole and l-(3-dimethylaminopropyl)-3- ethylcarbodiimide hydrochloride to a solution of the carboxylic acid in a suitable solvent such as jV,7V-dimethylformamide (DMF) at ambient temperature and then adding concentrated ammonium hydroxide.
In step (4) of Reaction Scheme III, a pyrazole carboxamide of Formula XXXIII is dehydrated to a pyrazole carbonitrile of Formula XXXIV. Suitable dehydrating agents include thionyl chloride, trifluoroacetic anhydride, and phosphorous oxychloride. The reaction is conveniently carried out by treating the pyrazole carboxamide of Formula XXXIII with phosphorous oxychloride and heating the reaction at an elevated temperature such as 90 °C. The reaction can also be carried out by combining the pyrazole carboxamide of Formula XXXIII with trifluoroacetic anhydride in the presence of a base such as triethylamine and in a suitable solvent such as dichloromethane. The reaction can be carried out at ambient temperature or at a sub-ambient temperature such as 0 0C.
In step (5) of Reaction Scheme III, a pyrazole carbonitrile of Formula XXXIV is brominated to provide a bromo-substituted pyrazole carbonitrile of Formula XXXV. The bromination is conveniently carried out by adding bromine to a solution of the pyrazole carbonitrile of Formula XXXIV and potassium acetate in acetic acid. The reaction can be carried out at ambient temperature.
In step (6) of Reaction Scheme III, a bromo-substituted pyrazole of Formula XXXV undergoes a transition-metal catalyzed cross coupling reaction with a reagent of Formula XXXVI. Reagents of Formula XXXVI, where M is, for example, -B(OH)2, -B(O-alkyl)2, -Sn(alkyl)3, and -Zn-Halide, are known to undergo coupling reactions. Several reagents of Formula XXXVI or their protected analogs are commercially available, for example, including 2-aminophenylboronic acid, 2- aminophenylboronic acid hydrochloride, and (2,2-dimethyl-N-[3~(454,5,5-tetramethyl- l,3,2-dioxaborolan-2-yl)pyridin-4-yl]propanamide; others can be prepared using known synthetic methods. For example, fert-butoxycarbonyl (Boc)-protected anilines undergo directed ortho metalation in the presence of butyllithium reagents. The resulting organolithium intermediate reacts with electrophiles such as B(O-alkyl)3 and ClSn(alkyl)3 to provide compounds of Formula XXXVI, where M is -B(O-alkyl)2 or -B(OH)2 and -Sn(alkyl)3, respectively, after removal of the Boc protecting group.
In step (6), a Suzuki coupling reaction is conveniently carried out by heating a mixture of the bromo-substituted pyrazole of Formula XXX, dichlorobis(triphenylphosphine)palladiurn(II) and a boron reagent of Formula XXXVI5 where M is -B(OH)2 or -B(0-alkyl)2, in the presence of a base such as potassium carbonate. The reaction is carried out in a suitable solvent such as 1 ,2-dimethoxyethane and can be heated at an elevated temperature such as 75 - 95 0C.
In step (7) of Reaction Scheme III, the amine and nitrile functionalities of a pyrazole of Formula XXXVII react under acidic conditions to form a pyrazolo [3 ,4- c]quinoline or pyrazolo [3 ,4-c]naphthyridine of Formula XXXVIII. The intramolecular addition is conveniently carried out by stirring acetyl chloride in ethanol and adding the resulting acidic solution to the pyrazole of Formula XXXVIII. The reaction is then heated at reflux to provide the pyrazolo [3 ,4-c]quinoline or pyrazolo[3,4-c]naphthyridine of Formula XXXVIII.
Reaction Scheme III
Figure imgf000034_0001
In some embodiments, compounds of the invention can be prepared according to
Reaction Scheme IV where R], R2, X, Z, and m are as defined above and RAI and RBI are as defined below.
In step (1) of Reaction Scheme IV, a bromo-substituted pyrazole carbonitrile of Formula XXXV undergoes a Sonogashira coupling reaction with (trimethylsilyl)acetylene to provide a pyrazole carbonitrile of Formula XXXIX. The reaction can be carried out according to the literature procedure, Sonogashira, K.; Tohda, Y.; Hagihara, N., Tetrahedron Lett, 4467 (1975).
Alternatively, the iodo analog may be used as a starting material for Reaction Scheme IV. The iodo analog can be prepared from a pyrazole carbonitrile of Formula XXIV5 shown in Reaction Scheme III. The iodination can be carried out by treating a pyrazole carbonitrile of Formula XXXIV with iodine monochloride in a suitable solvent such as dichloromethane in the presence of a base such as potassium carbonate. The reaction can be carried out at ambient temperature.
In step (2) of Reaction Scheme IV, the trimethylsilyl group of the pyrazole of Formula XXXIX is removed to provide the pyrazole of Formula XL. Potassium carbonate in methanol or tetrabutylammonium fluoride in tetrahydrofuran can be used to carry out the transformation.
In step (3) of Reaction Scheme IV, the acetylene of the pyrazole of Formula XL is alkylated using conventional synthetic methods, Jacobs, T. L. in Organic Reactions, 5, 1, (1949), to provide a pyrazole of Formula XLI. The reaction can be carried out by deprotonation of the compound of Formula XL with a base and reaction of the resulting carbanion with an electrophile of Formula Rei-Halide, for example, iodomethane. Step (3) can be omitted when RBI is hydrogen.
For some embodiments, steps (1) through (3) of Reaction Scheme IV may be replaced with one step from the iodo analog using a Sonogashira coupling reaction. The coupling can be carried out by combining an alkyne of Formula RBI -G≡C-H, copper(I) iodide, dichlorobis(triphenylρhosphine)palladium(II), and triethylamine in a suitable solvent such as acetom'trile and then heating at an elevated temperature, such as the reflux temperature of the solvent.
In step (4) of Reaction Scheme IV, a pyrazole of Formula XLI reacts with ammonia to provide a pyrazolo[3;4-c]pyridin-4-amine of Formula XLII. The reaction can be carried out by adding a solution of ammonia in methanol to the pyrazole of Formula XLI and heating at an elevated temperature, such as 150 0C. The reaction may be carried out in a pressure vessel.
Steps (5) and (6) may be carried out to provide a compound of Formula XLIV in which RA1 is other than hydrogen. In step (5) of Reaction Scheme IV, a pyrazolo[3,4- c]pyridin-4-amine of Formula XLII is brominated under conventional bromination conditions to provide a bromo-substituted pyrazolo[3,4-c]pyridine-4-amine of Formula XLIII. The reaction can be carried out as described in step (5) of Reaction Scheme III.
In step (6) of Reaction Scheme IV, a bromo-substituted pyrazolo[3,4-c]pyridin-4- amine of Formula XLIII undergoes a transition metal catalyzed coupling reaction with a reagent of Formula RAI-M, where RAI is alkenyl, alkoxy, and -N(R^2 to provide a pyrazolo[3,4-c]pyridine-4-amine of Formula XLIV. Reagents of Formula RAI-M, where M is, for example, -B(OH)2, -B(O-alkyl)2, -Sn(alkyl)3, and -Zn-Halide, are known to undergo coupling reactions. The transformation can be carried out by first protecting the amino group of the compound of Formula XLIII, treating the protected compound with a reagent of Formula RAI-M in the presence of a transition metal catalyst using conditions described in step (6) of Reaction Scheme III, and deprotecting the amine to provide the pyrazolo[354-c]pyridin-4-amine of Formula XLIV, which is a subgenus of Formula II. Alternatively, step (6) can be carried out by coupling a compound of Formula XLIII with an alkyne under Sonogashira conditions as described in step (1) of this reaction scheme. The resulting alkyne can be reduced under conventional hydrogenation conditions to provide a compound of Formula XLIV, where RA1 is alkenyl or alkyl. Step (6) may also be carried out by (i) protecting the amino group of the compound of Formula XLIII, for example, with a Boc group; (ii) performing a lithium-halogen exchange; (iii) treating with an electrophile of the Formula RAi-Halide, for example iodomethane; and (iv) deprotecting the amine to provide a compound of Formula XLIV.
Reaction Scheme IV
Figure imgf000037_0001
(3)
Figure imgf000037_0002
(6)
Figure imgf000037_0003
For some embodiments, compounds of the invention can be prepared according to Reaction Scheme V, where Rj b and R2!,, are subsets of Rj and R2 as defined above that do not include those substituents which would be susceptible to reduction under the acidic hydrogenation conditions of the reaction and R, X, Z, and n are as defined above. In Reaction Scheme V, a pyrazolo[3,4-c]quinoline of Formula XLV is reduced to provide a 6,7,8,9-tetrahydro-2i/-pyrazolo[3,4-c]quinoline of Formula XLVI, which is a subgenus of Formula IV. The reaction may be carried out under heterogeneous hydrogenation conditions by adding platinum (IV) oxide to a solution or suspension of a pyrazolo[3,4-c]quinoline of Formula XLV in a suitable solvent such as trifluoroacetic acid and placing the reaction under hydrogen pressure.
Alternatively, the reduction may be carried out at an earlier stage in the synthetic pathway.
Reaction Scheme V
Figure imgf000038_0001
Pyrazolo[3,4~c]naphthyridines of the invention can be prepared by using an azaindole as the starting material in Reaction Scheme I. Azaindoles are known compounds. Some are commercially available and others can be prepared using known synthetic methods. Alternatively, pyrazolo[3,4-c]naphthyridines of the invention can be prepared by using an aminopyridine boronic acid in Reaction Scheme III. Aminopyridine boronic acids can be prepared using known methods, for example, by directed ortho metalation of Boc-protected aminopyridines and subsequent electrόphilic substitution. Alternatively, for some isomers, halogen-lithium exchange and subsequent electrophilic substitution can be used. For example, halogen-lithium exchange can be carried out on a 2-bromopyridine that has a protected amino group in the 3 -position; subsequent electrophilic substitution with tributyltin chloride and deprotection of the amino group provides 3 -amino-2-tri-n-butylstannylpyridme. 657,8,9-Tetrahydro-2H-pyrazolo[3,4-c]naphthyridines can be prepared by reducing pyrazolo[3,4-c]naphthyridines using the method of Reaction Scheme V. For some embodiments, compounds can be further elaborated using conventional synthetic methods. For example, as shown in Scheme VI, a compound of Formula XLVII5 can undergo acid mediated cleavage of the Boc group in step (1) to give a secondary amine that can be functionalized in step (2) with an acid chloride of Formula R4C(O)Cl5 an acid anhydride of Formula (R4C(O))IO, an alkyl chloroformate of Formula R4OC(O)Cl5 a sulfonyl chloride of Formula R4S(O)2Cl, a sulfonic anydride of Formula (R4S(O)2^O5 an isocyanate of formula R4NCO, or an isothiocyanate of formula R4NCS to provide a compound of Formula XLIX where R4 is defined as above and Q is -C(O)-, -C(O)O-, -S(O)2-, -C(O)NH-, or -C(S)NH-. Numerous acid chlorides, alkyl chloroformates, sulfonyl chlorides, sulfonic anhydrides, isocyanates, and isothiocyanates are commercially available; others can be prepared readily using known synthetic methods. The reaction can be conveniently carried out by adding the acid chloride, alkyl chloroformate, sulfonyl chloride, sulfonic anhydride, isocyanate, or isothiocyanate to a solution or suspension of an amine of Formula XLVIII, in a suitable solvent such as chloroform. The reaction can be carried out at ambient temperature.
In addition, a compound of Formula XLVIII in Reaction Scheme VI can undergo alkylation of the secondary amine. In step (3) the compound of Formula XLVIII may be reacted with aldehydes, alkyl halides or triflates to provide a compound Formula L in which R8 is defined as above. For example, treatment of a compound of Formula XLVIII with aqueous formaldehyde and a reducing agent such as sodium cyanoborohydride in an appropriate solvent such as methanol yields a compound of Formula L, where R8 is a methyl group.
Reaction Scheme VI
Figure imgf000040_0001
Intermediates of Formula LIII can be prepared according to Reaction Scheme VII, where R1, X, and Z are as defined above.
In step (1) of Reaction Scheme VII, a ketone of the Formula LI is reacted with diethyl (2-oxopropyl)phosphonate to provide an olefin of Formula LII. The reaction can be carried out by adding a ketone of Formula LI and diethyl (2-oxopropyl)phosphonate to a solution of a base, such as potassium hydroxide, in a suitable solvent or solvent mix such as ethanol and water. The reaction can be carried out at a sub-ambient temperature such as O 0C.
In step (2) of Reaction Scheme VII, an olefin of Formula LII is derivatized using conventional methods. A compound of Formula LII where X is a bond and R1 is hydrogen can be prepared by reducing the olefin using conventional heterogeneous hydrogenation conditions. Also, a compound of Formula LII can be treated with pivalonitrile in the presence of titanium tetrachloride; the resulting nitrile-substituted compound can be converted by convention methods to a compound of Formula LIII where X is a bond and Ri is -C(O)-NH2. A compound of Formula LII can also be treated with ammonium hydroxide followed by di~tert-butyl dicarbonate to provide a compound of Formula LIII where X is a bond and Rj is -NHBoc, which can be deprotected and treated according to the methods of Reaction Scheme VI to provide a variety of other compounds.
Reaction Scheme VII
Figure imgf000041_0001
For some embodiments, compounds of the invention can be prepared according to Reaction Scheme VIII, where RA2, RB23 R2, and Z are as defined above.
In step (1) of Reaction Scheme VIII, an olefin of Formula LII is condensed with diethyl oxalate to provide a ketoester of Formula LIV. The reaction can be carried out as described in step (1) of Reaction Scheme III.
In step (2) of Reaction Scheme VIII, a ketoester of Formula LIV reacts with a hydrazine of Formula R2NHNH2 to provide a pyrazole carboxylate of Formula LV. The reaction can be carried out as described in step (2) of Reaction Scheme III.
In step (3) of Reaction Scheme VIII, a pyrazole carboxylate of Formula LV is converted to a pyrazole carboxamide of Formula LVL The reaction can be carried out as described in step (3) of Reaction Scheme III.
In step (4) of Reaction Scheme VIII, the olefinic bond in a compound of Formula LVI is oxidized to provide an epoxide of Formula LVII. The reaction can be carried out by treating a suspension of a compound of Formula LVI in a suitable solvent such as chloroform with 3-chloroperoxybenzoic acid. The reaction can be carried out at ambient temperature.
In step (5) of Reaction Scheme VIII, the epoxide ring in a compound of Formula LVII is cleaved to provide a hydroxy substituted pyrazole carboxamide of Formula LVIII. The reaction can be carried out by treating a solution of a compound of Formula LVII in a suitable solvent such as ethanol with palladium on carbon and ammonium formate. The reaction can be carried out at ambient temperature.
In step (6) of Reaction Scheme VIII, a pyrazole carboxamide of Formula LVIII is dehydrated to a pyrazole carbonitrile of Formula LIX. The reaction can be carried out as described in step (4) of Reaction Scheme III.
In steps (7) through (9) of Reaction Scheme VIII, a pyrazole carbonitrile of Formula LIX is converted to a pyrazolo[3,4~c]quinoline or pyrazolo[3,4-c]naphthyridine of Formula LX. The conversion can be carried out using the methods described in steps (5) through (7) of Reaction Scheme III.
Reaction Scheme VIII
Figure imgf000042_0001
For some embodiments, compounds of the invention can be prepared according to Reaction Scheme IX, where RA2, RB2, R2, and Z are as defined above.
In step (1) of Reaction Scheme IX5 the hydroxy group in a pyrazolo carbonitrile of Formula LIX is replaced with a fluoro group to provide a pyrazolo carbonitrile of Formula LXI. The reaction can be carried out by treating a solution of a compound of Formula LIX in a suitable solvent such as dichloromethane with [bis(2-methoxyethyl)amino]sulfur trifiuoride. The trifluoride is added in a controlled fashion at a sub-ambient temperature such as 0 0C.
In steps (2) through (4) of Reaction Scheme IX, a pyrazole carbonitrile of Formula LXI is converted to a pyrazolo[3,4-c]quinoline or pyrazolo[3,4-c]naphthyridine of Formula LXII. The conversion can be carried out using the methods described in steps (5) through (7) of Reaction Scheme III.
Reaction Scheme IX
Figure imgf000043_0001
Compounds of the invention can also be prepared using variations of the routes shown in Reaction Schemes I through IX that would be apparent to one of skill in the art. For example, a compound of Formula LII wherein Z is -N(Boc)- can be readily prepared from l-Boc-4-piperidinone according to the method of step (1) of Reaction Scheme VII. Steps (1) through (6) of Reaction Scheme VIII can then be used to prepare a compound of Formula LIX wherein Z is -N(Boc)-. The Boc group can then be cleaved and the resulting amine can be treated with an acid chloride, alkyl chloroformate, sulfonyl chloride, sulfonic anhydride, isocyanate, or isothiocyanate according to the methods described in steps (1) and (2) of Reaction Scheme VI. These Boc removal and amine functionalization steps can conveniently be carried out after the bromination in step (7) of Reaction Scheme VIII or after steps (1) and (2) of Reaction Scheme IX. Finally, the coupling and ring-closing methods of steps (8) and (9) of Reaction Scheme VIII or steps (3) and (4) of Reaction Scheme IX can be used to provide compounds of Formulas LX and LXII, respectively, wherein Z is -N(Q-R4)-.
For some embodiments, compounds of the invention can be prepared according to Reaction Scheme X, where Ri, R2, RA5 RB, Gb Z, and m are as defined above. The amino group of a pyrazolo compound of Formula I can be converted by conventional methods to a functional group such as an amide, carbamate, urea, amidine, or another hydrolyzable group. A compound of this type can be made by the replacement of a hydrogen atom in the amino group with a group such as -C(O)-R', α-aminoacyl, α-aminoacyl-α-aminoacyl, -C(O)-O-R1, -C(O)-N(R")-R', -C(=NY')-R', -CH(OH)-C(O)-OY', -CH(OCi-4 alkyl)Y0, -CH2Yi5 or -CH(CH3)Yi; wherein R1 and R" are each independently C1-Io alkyl, C3-7 cycloalkyl, phenyl, or benzyl, each of which may be unsubstituted or substituted by one or more substituents independently selected from the group consisting of halogen, hydroxy, nitro, cyano, carboxy, C1-6 alkyl, C1-4 alkoxy, aryl, heteroaryl, arylCi-4 alkylenyl, heteroarylQ.4 alkylenyl, haloCM alkylenyl, haloCM alkoxy, -0-C(O)-CH3, -C(O)-O-CH3, -C(O)-NH2, -0-CH2-C(O)-NH2, -NH2, and -S(O)2-NH2;, with the proviso that R" may also be hydrogen; each α-aminoacyl group is independently selected from racemic, D, or L- amino acids; Y' is hydrogen, Ci-6 alkyl, or benzyl; Y0 is C1-6 alkyl, carboxyCi-6 alkylenyl, aminoCi-4 alkylenyl, mono-JV-Ci-6 alkylaminoCi.4 alkylenyl, or di-iV,iV-Ci-6 alkylaminoCi-4 alkylenyl; and Yi is mono-7V-Ci-6 alkylamino, di-N,N-C\.6 alkylamino, morpholin-4-yl, piperidin-1-yl, pyrrolidin-1-yl, or 4-C1-4alkylpiperazin-l-yl. Particularly useful compounds of Formula XIII are amides derived from carboxylic acids containing one to ten carbon atoms, amides derived from amino acids, and carbamates containing one to ten carbon atoms. The reaction can be carried out, for example, by combining a compound of Formula I with a chloroformate or acid chloride, such as ethyl chloroformate or acetyl chloride, in the presence of a base such as triethylamine in a suitable solvent such as dichloromethane at room temperature. Reaction Scheme X
Figure imgf000045_0001
I XIII
For some embodiments, compounds of the invention can be prepared according to Reaction Scheme XI, where R2, RA5 RB, G2, Z, and m are as defined above. The hydrogen atom of the alcohol group of Formula LXIII can be replaced using conventional methods with a group such as Ci-6 alkanoyloxymethyl, 1-(Ci-6 alkanoyloxy)ethyl, 1 -methyl- 1-(C1-6 alkanoyloxy)ethyl, Ci-6 alkoxycarbonyloxymethyl, iV-(C1-6 alkoxycarbonyl)aminomethyl, succinoyl, Ci-6alkanoyl, α-aminoCi-4 alkanoyl, arylacyl, -P(O)(OH)2, -P(O)(O-C1-6 alkyl)2, Ci-6 alkoxycarbonyl, C1-6 alkylcarbamoyl, and α-aminoacyl or α-aminoacyl-α-aminoacyl, where each α-aminoacyl group is independently selected from racemic, D, and L-amino acids. Particularly useful compounds of Formula XIV are esters made from carboxylic acids containing one to six carbon atoms, unsubstituted or substituted benzoic acid esters, or esters made from naturally occurring amino acids.
Reaction Scheme XI
Figure imgf000045_0002
For some embodiments, compounds of the invention can be prepared using the synthetic routes described in the EXAMPLES below.
Pharmaceutical Compositions and Biological Activity Pharmaceutical compositions of the invention contain a therapeutically effective amount of a compound or salt described above in combination with a pharmaceutically acceptable carrier.
The terms "a therapeutically effective amount" and "effective amount" mean an amount of the compound or salt sufficient to induce a therapeutic or prophylactic effect, such as cytokine induction, immunomodulation, antitumor activity, and/or antiviral activity. The exact amount of compound or salt used in a pharmaceutical composition of the invention will vary according to factors known to those of skill in the art, such as the physical and chemical nature of the compound or salt, the nature of the carrier, and the intended dosing regimen. In some embodiments, the compositions of the invention will contain sufficient active ingredient or prodrug to provide a dose of about 100 nanograms per kilogram (ng/kg) to about 50 milligrams per kilogram (mg/kg), preferably about 10 micrograms per kilogram (μg/kg) to about 5 mg/kg, of the compound or salt to the subject.
In other embodiments, the compositions of the invention will contain sufficient active ingredient or prodrug to provide a dose of, for example, from about 0.01 mg/m2 to about 5.0 mg/m2, computed according to the Dubois method, in which the body surface area of a subject (m2) is computed using the subject's body weight: m2 = (wt kg0425 x height cm0725) x 0.007184, although in some embodiments the methods may be performed by administering a compound or salt or composition in a dose outside this range. In some of these embodiments, the method includes administering sufficient compound to provide a dose of from about 0.1 mg/m2 to about 2.0 mg/ m2 to the subject, for example, a dose of from about 0.4 mg/m2 to about 1.2 mg/m2.
A variety of dosage forms may be used, such as tablets, lozenges, capsules, parenteral formulations, syrups, creams, ointments, aerosol formulations, transdermal patches, transmucosal patches and the like. These dosage forms can be prepared with conventional pharmaceutically acceptable carriers and additives using conventional methods, which generally include the step of bringing the active ingredient into association with the carrier.
The compounds or salts of the invention can be administered as the single therapeutic agent in the treatment regimen, or the compounds or salts described herein may be administered in combination with one another or with other active agents, including additional immune response modifiers, antivirals, antibiotics, antibodies, proteins, peptides, oligonucleotides, etc.
Compounds or salts of the invention have been shown to induce the production of certain cytokines in experiments performed according to the tests set forth below. These results indicate that the compounds or salts are useful for modulating the immune response in a number of different ways, rendering them useful in the treatment of a variety of disorders.
Cytokines whose production may be induced by the administration of compounds or salts of the invention generally include interferon-α (IFN-α) and tumor necrosis factor-α (TNF-α) as well as certain interleukins (IL). Cytokines whose biosynthesis may be induced by compounds or salts of the invention include IFN-α, TNF-α, IL-I, IL-6, IL-IO and IL- 12, and a variety of other cytokines. Among other effects, these and other cytokines can inhibit virus production and tumor cell growth, making the compounds or salts useful in the treatment of viral diseases and neoplastic diseases. Accordingly, the invention provides a method of inducing cytokine biosynthesis in an animal comprising administering an effective amount of a compound or salt of the invention to the animal.
The animal to which the compound or salt is administered for induction of cytokine biosynthesis may have a disease as described infra, for example a viral disease or a neoplastic disease, and administration of the compound or salt may provide therapeutic treatment. Alternatively, the compound or salt may be administered to the animal prior to the animal acquiring the disease so that administration of the compound or salt may provide a prophylactic treatment.
In addition to the ability to induce the production of cytokines, compounds or salts described herein can affect other aspects of the innate immune response. For example, natural killer cell activity may be stimulated, an effect that may be due to cytokine induction. The compounds or salts may also activate macrophages, which in turn stimulate secretion of nitric oxide and the production of additional cytokines. Further, the compounds or salts may cause proliferation and differentiation of B-lymphocytes.
Compounds or salts described herein can also have an effect on the acquired immune response. For example, the production of the T helper type 1 (THI) cytokine IFN- γ may be induced indirectly and the production of the T helper type 2 (TH2) cytokines IL- 4, IL-5 and IL- 13 may be inhibited upon administration of the compounds or salts.
Whether for prophylaxis or therapeutic treatment of a disease, and whether for effecting innate or acquired immunity, the compound or salt or composition may be administered alone or in combination with one or more active components as in, for example, a vaccine adjuvant. When administered with other components, the compound or salt or composition and other component or components may be administered separately; together but independently such as in a solution; or together and associated with one another such as (a) covalently linked or (b) non-covalently associated, e.g., in a colloidal suspension. Conditions for which compounds or salts or compositions identified herein may be used as treatments include, but are not limited to:
(a) viral diseases such as, for example, diseases resulting from infection by an adenovirus, a herpesvirus (e.g., HSV-I, HSV-II, CMV, or VZV), a poxvirus (e.g., an orthopoxvirus such as variola or vaccinia, or molluscum contagiosum), a picornavirus (e.g., rhinovirus or enterovirus), an orthomyxovirus (e.g., influenzavirus), a paramyxovirus (e.g., parainfluenzavirus, mumps virus, measles virus, and respiratory syncytial virus (RSV)), a coronavirus (e.g., SARS), a papovavirus (e.g., papillomaviruses, such as those that cause genital warts, common warts, or plantar warts), a hepadnavirus (e.g., hepatitis B virus), a flavivirus (e.g., hepatitis C virus or Dengue virus), or a retrovirus (e.g., a lentivirus such as HIV);
(b) bacterial diseases such as, for example, diseases resulting from infection by bacteria of, for example, the genus Escherichia, Enterobacter, Salmonella, Staphylococcus, Shigella, Listeria, Aerobacter, Helicobacter, Klebsiella, Proteus, Pseudomonas, Streptococcus, Chlamydia, Mycoplasma, Pneumococcus, Neisseria, Clostridium, Bacillus, Corynebacterium, Mycobacterium, Campylobacter, Vibrio, Serratia, Providencia, Chromobacterium, Brucella, Yersinia, Haemophilus, or Bordetella; (c) other infectious diseases, such as chlamydia, fungal diseases including but not limited to candidiasis, aspergillosis, histoplasmosis, cryptococcal meningitis, or parasitic diseases including but not limited to malaria, Pneumocystis carnii pneumonia, leishmaniasis, cryptosporidiosis, toxoplasmosis, and trypanosome infection; (d) neoplastic diseases, such as intraepithelial neoplasias, cervical dysplasia, actinic keratosis, basal cell carcinoma, squamous cell carcinoma, renal cell carcinoma, Kaposi's sarcoma, melanoma, leukemias including but not limited to acute myeloid leukemia, acute lymphocytic leukemia, chronic myeloid leukemia, chronic lymphocytic leukemia, multiple myeloma, Hodgkin's lymphoma, non-Hodgkin's lymphoma, cutaneous T-cell lymphoma, B-cell lymphoma, and hairy cell leukemia, and other cancers;
(e) TH2 -mediated, atopic diseases, such as atopic dermatitis or eczema, eosinophilia, asthma, allergy, allergic rhinitis, and Ommen's syndrome;
(f) certain autoimmune diseases such as systemic lupus erythematosus, essential thrombocythaemia, multiple sclerosis, discoid lupus, alopecia areata; and (g) diseases associated with wound repair such as, for example, inhibition of keloid formation and other types of scarring (e.g., enhancing wound healing, including chronic wounds).
Additionally, a compound or salt identified herein may be useful as a vaccine adjuvant for use in conjunction with any material that raises either humoral and/or cell mediated immune response, such as, for example, live viral, bacterial, or parasitic immunogens; inactivated viral, tumor-derived, protozoal, organism-derived, fungal, or bacterial immunogens; toxoids; toxins; self-antigens; polysaccharides; proteins; glycoproteins; peptides; cellular vaccines; DNA vaccines; autologous vaccines; recombinant proteins; and the like, for use in connection with, for example, BCG, cholera, plague, typhoid, hepatitis A, hepatitis B, hepatitis C, influenza A, influenza B, parainfluenza, polio, rabies, measles, mumps, rubella, yellow fever, tetanus, diphtheria, hemophilus influenza b, tuberculosis, meningococcal and pneumococcal vaccines, adenovirus, HIV, chicken pox, cytomegalovirus, dengue, feline leukemia, fowl plague, HSV-I and HSV-2, hog cholera, Japanese encephalitis, respiratory syncytial virus, rotavirus, papilloma virus, yellow fever, and Alzheimer's Disease.
Compounds or salts identified herein may be particularly helpful in individuals having compromised immune function. For example, compounds or salts may be used for treating the opportunistic infections and tumors that occur after suppression of cell mediated immunity in, for example, transplant patients, cancer patients and HIV patients. Thus, one or more of the above diseases or types of diseases, for example, a viral disease or a neoplastic disease may be treated in an animal in need thereof (having the disease) by administering a therapeutically effective amount of a compound or salt of the invention to the animal.
An animal may also be vaccinated by administering an effective amount of a compound or salt described herein, as a vaccine adjuvant. In one embodiment, there is provided a method of vaccinating an animal comprising administering an effective amount of a compound or salt described herein to the animal as a vaccine adjuvant.
An amount of a compound or salt effective to induce cytokine biosynthesis is an amount sufficient to cause one or more cell types, such as monocytes, macrophages, dendritic cells and B-cells to produce an amount of one or more cytokines such as, for example, IFN-α, TNF-α, IL-I, IL-6, IL-IO and IL- 12 that is increased (induced) over a background level of such cytokines. The precise amount will vary according to factors known in the art but is expected to be a dose of about 100 ng/kg to about 50 mg/kg, preferably about 10 μg/kg to about 5 mg/kg. In other embodiments, the amount is expected to be a dose of, for example, from about 0.01 mg/m to about 5.0 mg/m , (computed according to the Dubois method as described above) although in some embodiments the induction or inhibition of cytokine biosynthesis may be performed by administering a compound or salt in a dose outside this range. In some of these embodiments, the method includes administering sufficient compound or salt or composition to provide a dose of from about 0.1 mg/m2 to about 2.0 mg/ m2 to the subject, for example, a dose of from about 0.4 mg/m2 to about 1.2 mg/m2. The invention also provides a method of treating a viral infection in an animal and a method of treating a neoplastic disease in an animal comprising administering an effective amount of a compound or salt of the invention to the animal. An amount effective to treat or inhibit a viral infection is an amount that will cause a reduction in one or more of the manifestations of viral infection, such as viral lesions, viral load, rate of virus production, and mortality as compared to untreated control animals. The precise amount that is effective for such treatment will vary according to factors known in the art but is expected to be a dose of about 100 ng/kg to about 50 mg/kg, preferably about 10 μg/kg to about 5 mg/kg. An amount of a compound or salt effective to treat a neoplastic condition is an amount that will cause a reduction in tumor size or in the number of tumor foci. Again, the precise amount will vary according to factors known in the art but is expected to be a dose of about 100 ng/kg to about 50 mg/kg, preferably about 10 μg/kg to about 5 mg/kg. In other embodiments, the amount is expected to be a dose of, for example, from about 0.01 mg/m2 to about 5.0 mg/m2, (computed according to the Dubois method as described above) although in some embodiments either of these methods may be performed by administering a compound or salt in a dose outside this range. In some of these embodiments, the method includes administering sufficient compound or salt to provide a dose of from about 0.1 mg/m2 to about 2.0 mg/ m2 to the subject, for example, a dose of from about 0.4 mg/m2 to about 1.2 mg/m2.
In addition to the formulations and uses described specifically herein, other formulations, uses, and administration devices suitable for compounds of the present invention are described in, for example, International Publication Nos. WO 03/077944 and WO 02/036592, U.S. Patent No. 6,245,776, and U.S. Publication Nos. 2003/0139364, 2003/185835, 2004/0258698, 2004/0265351, 2004/076633, and 2005/0009858.
Objects and advantages of this invention are further illustrated by the following examples, but the particular materials and amounts thereof recited in these examples, as well as other conditions and details, should not be construed to unduly limit this invention.
EXAMPLES In the examples below automated flash chromatography on silica gel was carried out using a HORIZON HPFC system (an automated high-performance flash purification product available from Biotage, Inc, Charlottesville, Virginia, USA) or an INTELLIFLASH Flash Chromatography System (an automated flash purification system available from AnaLogix, Inc, Burlington, Wisconsin, USA). The eluent used for each purification is given in the example. In some chromatographic separations, the solvent mixture 80/18/2 v/v/v chloroform/methanol/concentrated ammonium hydroxide (CMA) was used as the polar component of the eluent. In these separations, CMA was mixed with chloroform in the indicated ratio. Example 1
1 -[(4-Amino-2-propyl-2H-pyrazolo[3,4-c]quinolin- 1 -yl)methyl]cyclobutanol
Figure imgf000052_0001
Part A
A solution of ethyl chlorooxoacetate (22.53 g, 0.165 mol) in diethyl ether (100 mL) was added over a period of 15 minutes to a cooled (0 0C) solution of 2-methylindole (19.7 g, 0.15 mol) and pyridine (14.2 g, 0.18 mol) in diethyl ether (200 mL). The reaction was stirred for two hours at 0 0C under a nitrogen atmosphere. Most of the diethyl ether had evaporated by the end of the two-hour reaction time, and a solid was present. Water (100 mL) was added, and the solid was isolated by filtration and washed with 1:1 diethyl ether/hexane. The solid (27.9 g) was then dissolved in boiling toluene (250 mL) and recrystallized upon cooling to 6 0C. The crystals were isolated by filtration, washed with toluene, dried for two hours on the vacuum filter funnel, triturated with water at 75 0C for five minutes, isolated by filtration, and dried for three hours on the filter funnel to provide 17.8 g ethyl (2-methyl-lH-indol-3-yl)(oxo)acetate of as a rust-colored powder. Part B
Ethyl (2-methyl-lH-indol-3-yl)(oxo)acetate (6.94 g, 30.0 mmol) and propylhydrazine oxalate (10.8 g, 66.0 mmol) were added to a solution of acetyl chloride (5.18 g, 66.0 mmol) in acetic acid (5 mL) and ethanol (150 mL), and the reaction was heated at reflux under nitrogen for 42.5 hours. The ethanol was removed under reduced pressure, and 2 M aqueous sodium carbonate was added. The mixture was stirred, and the resulting solid was isolated by filtration, washed with water, and dried for 90 minutes on the vacuum filter funnel to provide a dark semi-solid. The crude product was stirred with tert-butyl methyl ether (50 mL) and isolated by filtration, washed with tert-bntyl methyl ether, and dried on the vacuum filter funnel to provide 6.1O g of an orange solid, which was stirred with boiling acetonitrile (50 mL), isolated by filtration, and purified by automated flash chromatography to provide 5.11 g of l-methyl-2-propyl-2H-pyrazolo[3,4- e]quinolin-4-ol as a white solid. Part C
A solution of l-methyl-2-propyl-2H-pyrazolo[3,4-c]quinolin-4--ol (5.11 g, 21.2 mmol) in phosphorus oxychloride (50 niL) was heated at reflux for 30 minutes, allowed to cool to room temperature, and stirred for two days. The reaction mixture was poured into ice water (500 niL) with stirring, and concentrated ammonium hydroxide (169 mL) and ice were added. A solid was present and was isolated by filtration, washed with water, and purified by automated flash chromatography (eluting with 5% to 25% CMA in chloroform). The resulting orange solid (5.5 g) was recrystallized from acetonitrile (25 mL). The crystals were washed with acetonitrile and dried for five hours to provide 3.85 g of 4-chloro-l-methyl-2-propyl-2/J~pyrazolo[3,4-c]quinoline as a white solid, mp 145-147 0C.
Anal. Calcd for Ci4H14N3Cl: C, 64.74; H, 5.43; N, 16.18. Found: C, 64.50; H, 5.64; N, 16.20. This product was combined with material from another run. Part D
A Parr vessel was charged with 4-chloro-l-methyl-2-proρyl-2H-pyrazolo[3,4- c]quinoline (4.31 g) and a solution of ammonia in methanol (50 mL of 7 N). The reaction was heated at 150 0C for 24 hours and allowed to cool to room temperature. Most of the methanol was removed under reduced pressure, and water was added. A precipitate formed and was isolated by filtration, washed with water, and dried on the vacuum filter funnel to provide 3.8 g of l-methyl-2-propyl-2H-pyrazolo[3,4-c]quinolin-4-amine as a white solid. Part E
A solution of l-methyl-2-propyl-2//-pyrazolo[3,4-c]quinolin-4-amine (3.78 g, 15.7 mmol), di-tert-butyl dicarbonate (8.6 g, 39.3 mmol), and 4-dimethylaminopyridine
(DMAP) (96 mg, 0.79 mmol) in tetrahydrofuran (THF) (100 mL) was heated at reflux for 14 hours and allowed to cool to room temperature. The THF was removed under reduced pressure, and the residue was purified by chromatography using a HORIZON HPFC system (eluting with 50% ethyl acetate in hexane) followed by recrystallization from tert- butyl methyl ether/hexane. The crystals were washed with hexane and dried on the vacuum filter funnel for two hours to provide 6.46 g of di(tørt-butyl) l-methyl-2-propyl- 27f-pyrazolo[3,4-c]quinolin-4-ylimidodicarbonate as an off-white solid. Part F
A solution of di(fert-butyl) l-memyl-2-propyl-2H-pyrazolo[3,4-c]quinolin-4- ylimidodicarbonate (1.32 g, 3.00 mmol) in anhydrous tetrahydrofuran (THF) (30 mL) was cooled to -78 0C under an argon atmosphere. tert-Butyllithium (7.06 mL of a 1.7 M solution in pentane) was added over a period of eight minutes, and the resulting dark- colored solution was stirred at -78 0C for 40 minutes. Cyclobutanone (1.05 g, 15 mmol) was added over a period of two minutes, and the -78 0C bath was replaced with a 0 0C bath. The reaction mixture was stirred for 10 minutes, and then saturated aqueous ammonium chloride (30 mL) was added. The aqueous layer was separated and extracted three times with tert-butyl methyl ether, and the combined organic fractions were dried over magnesium sulfate, filtered, and concentrated under reduced pressure. The residue (1.3 g) was purified by automated flash chromatography and then boiled in 2 M hydrochloric acid for 30 minutes. The solution was allowed to cool to room temperature overnight and then made basic with the addition of 2 M aqueous sodium carbonate. The resulting solution was extracted four times with chloroform, and the combined extracts were dried over magnesium sulfate, filtered, and concentrated under reduced pressure. The residue (1.2 g) was purified twice by automated flash chromatography and recrystallized from acetonitrile to provide 297 mg of 1 -[(4-amino-2 -propyl -2H- pyrazolo[3,4-c]quinolin-l-yl)methyl]cyclobutanol as a white solid, mp 191-193 0C. MS (APCI) m/z 311 (M + H)+;
Anal. Calcd for Ci8H22N4O: C, 69.65; H, 7.14; N, 18.05. Found: C, 69.35; H, 7.14; N, 18.11.
Example 2 4-[(4-Amino-2-propyl-2H-pyrazolo[3,4-c]quinolin-l-yl)methyl]tetrahydro-2H-pyran-4-ol
Figure imgf000054_0001
A solution of di(tert-butyl) l-methyl-2-propyl-2H-ρyrazolo[3,4-c]quinolin-4- ylimidodicarbonate (1.32 g, 3.00 mmol) in anhydrous tetrahydrofuran (THF) (30 mL) was cooled to -78 0C under an argon atmosphere. fø/Y-Butyllithium (4.4 rnL of a 1.7 M solution in pentane) was added over a period of five minutes, and the resulting dark- colored solution was stirred at -78 0C for 30 minutes. Tetrahydro-4/f-pyran-4-one (901 mg, 9.0 mmol) was added, and reaction mixture was stirred for 10 minutes. The -78 0C bath was replaced with a 0 0C bath, and then saturated aqueous ammonium chloride (30 mL) was added. The aqueous layer was separated and extracted three times with ter/-butyl methyl ether, and the combined organic fractions were dried over magnesium sulfate, filtered, and concentrated under reduced pressure. The residue (1.3 g) was purified by automated flash chromatography (eluting with ethyl acetate) and heated in refluxing 1 M hydrogen chloride in ethanol (50 mL) for one hour. The solution was allowed to cool to room temperature and then made basic with the addition of 2 M aqueous sodium carbonate. The resulting solution was extracted four times with chloroform, and the combined extracts were dried over magnesium sulfate, filtered, and concentrated under reduced pressure. The residue (1.2 g) was purified by automated flash chromatography and recrystallized from acetonitrile to provide 430 mg of 4-[(4-amino-2-propyl-2H- pyrazolo[3,4-c]quinolin-l-yl)methyl]tetrahydro-2H-pyran-4-ol as a white solid, mp 209-
211 0C.
MS (APCI) m/z 341 (M + H)+;
Anal. Calcd for C19H24N4O2: C, 67.04; H, 7.11; N, 16.46. Found: C, 66.98; H3 7.17; N, 16.39.
Example 3
2-Propyl-l-(tetrahydro-2ϋ/-pyran-4-ylmethyl)-2H-pyrazolo[3,4-c]quinolin-4-amine
Figure imgf000055_0001
Part A
A solution of potassium hydroxide (4.66 g, 83.1 mmol) in ethanol (181 mL) and water (45 mL) was cooled to approximately 0 0C. Tetrahydro-4/i-pyran-4-one (7.56 g, 75.5 mmol) and diethyl (2-oxopropyl)phosphonate (16.1 g, 83.1 mmol) were sequentially added. The reaction was allowed to warm to room temperature and stirred for five hours. The reaction mixture was washed with brine and the organic layer was separated. The aqueous layer was extracted three times with tert-butyl methyl ether. The combined organic tractions were dried over magnesium sulfate, filtered, and concentrated under reduced pressure. The residue was dissolved in a mixture of 20% ethyl acetate in hexane and chloroform, dried again over magnesium sulfate, filtered, concentrated under reduced pressure, and purified by automated flash chromatography (eluting with 35% to 45% ethyl acetate in hexane). The resulting colorless oil was dried under a stream of nitrogen to provide 8.03 g of l-tetrahydro-4H-pyran-4-ylideneacetone. Part B A solution of 1 -tetrahydro-4//-pyran-4-ylideneacetone (4.0 g, 28.5 mmol) in ethyl acetate was added to a Parr vessel. The vessel was purged with nitrogen, and 10% palladium on carbon (400 mg) was added. The vessel was shaken under hydrogen pressure (50 psi, 3.4 X 105 Pa) for approximately ten minutes, and the reaction mixture was filtered through a layer of CELITE filter agent. The filter cake was washed with ethyl acetate. The filtrate was concentrated under reduced pressure, and the residue was dried under a stream of nitrogen to provide 3.80 g of l-tetrahydro-2H-pyran-4-ylacetone as a colorless oil. Part C
A neat mixture of 1 -tetrahydro-2H-pyran-4-ylacetone (3.80 g, 26.7 mmol) and diethyl oxalate (4.30 g, 29.4 mmol) was added in one portion, followed by an ethanol rinse (10 mL), to a stirred solution of sodium tert-butoxide (2.83 g, 29.4 mmol) in ethanol (20 mL). The mixture was stirred for one hour, cooled to approximately 0 0C, and treated with acetic acid (30.0 mL). After the mixture was stirred for five minutes, propylhydrazine oxalate (4.38 g, 26.7 mmol) was added in one portion. The reaction was allowed to warm to room temperature slowly and stirred overnight. Most of the volatiles were removed under reduced pressure, and 2 M aqueous sodium carbonate was added. The mixture was extracted four times with tert-butyl methyl ether. The organic layers were combined, dried over magnesium sulfate, filtered, and concentrated to yield 8.5 g of a yellow oil. The oil was purified twice by automated flash chromatography (eluting with ethyl acetate in hexane), and the resulting pale yellow oil was dried under a stream of nitrogen to provide 6.43 g of ethyl l-propyl-5-(tetrahydro-2H-ρyran-4-ylmethyl)-lH-pyrazole-3- carboxylate. Part D
Lithium hydroxide monohydrate (3.8 g, 92 mmol) was added to a solution of ethyl l-propyl-5-(tetralτ.ydro-2H-pyran-4-ylmethyl)-lH-pyrazole-3-carboxylate (6.43 g, 22.9 mmol) in methanol (30 niL) and water (10 mL). The mixture was stirred for 20 hours. Most of the volatiles were removed under reduced pressure, and water (100 mL) and acetic acid (52 mL, 910 mmol) were sequentially added. The solution was cooled to approximately 0 0C. After five minutes, a white solid formed, and more water (100 mL) was added. The mixture was stirred for 30 minutes, and the solid was isolated by filtration, washed with water, and dried on the vacuum filter funnel to provide 4.88 g of 1- ρropyl-5-(tetrahydro-2/i-pyran-4-ylmethyl)-l//-pyrazole-3-carboxylic acid as a white solid. Part E l-(3-Dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (4.06 g, 21.2 mmol) was added to a solution of l-propyl-5-(tetrahydro-2H-pyran-4-ylmethyl)-lH- pyrazole-3-carboxylic acid (4.88 g, 19.3 mmol) and 1-hydroxybenzotriazole (2.87 g, 21.2 mmol) in A^iV-dimethylformamide (DMF) (24.4 mL) at room temperature. The mixture was stirred for two hours, cooled in an ice bath, and treated with concentrated ammonium hydroxide (3.9 mL). A precipitate formed, and the mixture was stirred 30 minutes at 0 0C. Water (100 mL) was added, and the mixture was stirred for an additional ten minutes. The precipitate was isolated by filtration, washed with water, and dried on the vacuum filter funnel to provide 4.05 g of l-propyl-5-(tetrahydro-2H-pyran-4-ylmethyl)-lH'-pyrazole-3- carboxamide as white crystals. Part F
A solution of trifluoroacetic anhydride (2.50 mL, 17.7 mmol) in dichloromethane (32 mL) was added over a period often minutes to a 0 °C solution of l-propyl-5-
(tetrahydro-2H-pyran-4-ylmethyl)-lH-pyrazole-3-carboxamide (4.05 g, 16.1 mmol) and triethylamine (4.89 g, 48.3 mmol) in dichloromethane (32 mL). After the addition was complete, the cooling bath was removed and the solution was stirred for three hours. The solution was washed with 2 M aqueous sodium carbonate. The aqueous layer was extracted three times with tert-butyl methyl ether. The organic layers were combined, dried over magnesium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by automated flash chromatography (eluting with 40% to 70% ethyl acetate in hexane) to provide 3.98 g of l-propyl-5-(tetrahydro-2i:/-pyran-4- ylmethyl)~lH-pyrazole-3-carbonitrile as a pale yellow oil. Part G
Potassium acetate (3.93 g, 40.0 mmol) and bromine (3.58 g, 22.4 mmol) were sequentially added to a solution of 1 -propyl-5-(tetrahydro-2H-pyran-4-ylmethyl)-lH- pyrazole-3-carbonitrile (3.98 g, 16 mmol) in acetic acid (32 rnL). The reaction was stirred for 40 hours at room temperature. Saturated aqueous sodium hydrogensulfite was added until the reaction became colorless. Most of the acetic acid was removed under reduced pressure, and 2M aqueous sodium carbonate was added. A precipitate formed, was isolated by filtration, washed with water, and dried on the vacuum filter funnel to provide 4.91 g of 4-bromo- 1 -propyl-5-(tetrahydro-2H~pyran-4-ylniethyl)- 1 /7-pyrazole-3 - carbonitrile as a white solid. Part H
2-Aminophenylboronic acid hydrochloride (1.39 g, 8.0 mmol) and dichlorobis(triρhenylphosphine)palladium(II) (140 mg, 0.20 mmol) were sequentially added to a mixture of 4-bromo-l-propyl-5-(tetrahydro-2H-pyran-4-ylmethyl)-lH- pyrazole-3-carbonitrile (1.25 g, 4.00 mmol), potassium carbonate (1.82 g, 13.2 mmol), 1,2-dimethoxyethane (DME) (15 mL), and water (7.5 mL). The flask was placed under vacuum and back-filled with nitrogen four times. The reaction was heated under a nitrogen atmosphere at 95 °C for one hour. The reaction was allowed to cool to room temperature, and water and tert-bxtiyl methyl ether were added. The aqueous layer was separated and extracted with førf-butyl methyl ether three times. The organic fractions were combined, dried over magnesium sulfate, filtered, and concentrated to yield a brown solid (1.75 g). The crude product was purified by automated flash chromatography (eluting with ethyl acetate in hexane) to provide 1.10 g of 4-(2-aminophenyl)- 1 -propyl-5- (tetrahydro-2H-pyran-4-ylmethyl)-lH-pyrazole-3-carbonitrile as a pale orange solid. Part i
Acetyl chloride (50 mmol) was stirred with anhydrous ethanol (50 ml), and the resulting solution was added to the material from Part H. The resulting solution was heated at reflux for 16 hours. Aqueous sodium carbonate (30 mL of 2 M) was added, and then most of the ethanol was removed under reduced pressure. Water was added; a solid was present and was isolated by filtration and washed with water. The crude product was purified by automated flash chromatography (eluting with 5% to 25% CMA in chloroform) followed by recystallization from ethyl acetate. The crystals were dried on the vacuum filter funnel for 2.5 hours to provide 853 mg of 2-propyl-l-(tetrahydro-2//- pyran-4-ylmethyl)-2H-pyrazolo[3,4-c]qumolin-4-amine as a white solid, mp 218-220 °,C. MS (APCI) m/z 325 (M + H)+;
Anal. Calcd for C19H24N4O: C, 70.34; H5 7.46; N, 17.27. Found: C, 70.11; H, 7.72; N, 17.28.
Example 4 2-Propyl- 1 -(tetrahydro-2H-pyran-4-ylmethyl)-2H-pyrazolo[3,4-c] [1 ,8]naphthyridin-4- amine
Figure imgf000059_0001
Part A tert-B\xXy\ iV-(2-pyridyi)carbamate is available from the literature procedure (Moraczewski, A. L. et al, J. Org. Chem., 1998, 63, 7258) or can be prepared by the following method. Under a nitrogen atmosphere, sodium bis(trimethylsilyl)arnide (225 mL of a 1.0 M solution in tetrahydrofuran) was added over a period of 20 minutes to a solution of 2-aminopyridine (10.61 g, 108.0 mmol) in dry THF (150 mL). The solution was stirred for 15 minutes and then cooled to 0 0C. A solution of di-fert-butyl dicarbonate (24.60 g, 112.7 mmol) in THF (50 mL) was added slowly, and the reaction was allowed to warm to ambient temperature slowly and stirred overnight. The THF was removed under reduced pressure, and the residue was partitioned between ethyl acetate (500 mL) and 0.1 M hydrochloric acid (250 mL). The organic layer was separated; washed sequentially with 0.1 M hydrochloric acid (250 mL), water (250 mL), and brine (250 mL); dried over magnesium sulfate; filtered; and concentrated under reduced pressure. The crude product was purified by automated flash chromatography (eluting with 80:20 hexanes/ethyl acetate) to provide 17.43 g of tert-hutyl iV-(2-pyridyl)carbamate as a white solid. Part B
Under a nitrogen atmosphere, a solution of tert-butyl iV-(2-ρyridyl)carbamate (15.71 g, 80.9 mmol) and iV.ΛζN'.N'-tetramethylethylenediamine (TMEDA) (25.3 g, 218 mmol) in THF (400 mL) was cooled to -78 0C. M-Butyllithium (81 mL of a 2.5 M solution in hexanes) was added dropwise over a period of 20 minutes. The solution was stirred for ten minutes, and then the addition funnel was rinsed with additional THF (20 mL). The solution was warmed to -6 0C, stirred for two hours, and cooled again to -78 0C. Triisopropyl borate (57.7 g, 307 mmol) was added over a period often minutes. The resulting solution was warmed to 00C and then poured into saturated aqueous ammonium chloride (500 mL). A yellow solid formed and was stirred with diethyl ether (300 mL), isolated by filtration, washed with diethyl ether and water, and air-dried overnight to provide 2-ført-butoxycarbonylamino-3-pyridylboronic acid as a yellow solid. Part C
Hydrochloric acid (10 mL of IM) was added to a solution of 2-tert- butoxycarbonylammo-3-pyridylboronic acid (2.59 g, 10.9 mmol), and the resulting mixture was heated at 80 0C for 45 minutes and allowed to cool to room temperature. Potassium carbonate (3.87 g, 27.95 mmol) was added with stirring, and then DME (20 mL), 4-bromo- 1 -propyl-5-(tetrahydro-2H~pyran-4-ylmethyl)- 1 H-pyrazole-3 -carbonitrile (1.70 g, 5.44 mmol), and dichlorobis(triphenylphosphine)palladium(II) (190 mg, 0.27 mmol) were added. The flask was placed under vacuum and back-filled with nitrogen three times. The reaction was heated under a nitrogen atmosphere at 95 °C overnight. The reaction was allowed to cool to room temperature, and the volatiles were removed under reduced pressure. The residue was dissolved in chloroform (100 mL), and the resulting solution was washed with water (100 mL), dried over magnesium sulfate, filtered, and concentrated to yield a light yellow solid. The crude product was purified by automated flash chromatography (eluting with 0% to 30% CMA in chloroform) followed by recrystallization from acetonitrile (30 mL). The crystals were washed with cold acetonitrile and dried overnight in a vacuum oven at 60 0C to provide 0.43 g of 2-propyl- l-(tetrahydro-2H-ρyran-4-ylmethyl)-2/i-pyrazolo[3,4-c][l,8]naphthyridin-4-amine as white needles, mp 252-255 0C.
Anal, calcd for C18H23N5O: C, 66.44; H, 7.12; N, 21.52. Found: C, 66.21; H, 7.35; N, 21.54. Example 5 2-(2-Methoxyethyl)-l-(tetrahydro-2/-r-pyran-4-ylmethyl)-2i7-pyrazolo[3,4-(?]quinolin-4- amine
Figure imgf000061_0001
Part A
The method described in Part B of Example 3 was repeated with l-tetrahydro-4H- pyran-4-ylideneacetone (4.0 g, 28.5 mmol), and the resulting l~tetrahydro-2//-pyran-4- ylacetone was mixed with diethyl oxalate (4.66 g, 31.9 mmol) and added to a solution of sodium /er/-butoxide (3.07 g, 31.9 mmol) in ethanol (22 mL). The reaction was carried out according to the method described in Part C of Example 3 with the following modifications. Ηydroxyethylhydrazine (2.43 g, 31.9 mmol) was used instead of ethylhydrazine oxalate. Extractions were carried out seven times with chloroform, and the crude product was not purified. Ethyl l-(2-hydroxyethyl)-5-(tetrahydro-2//-pyran-4- ylmethyl)-lH-pyrazole-3-carboxylate (8.2 g) was obtained as a viscous yellow oil. Part B
A solution of the material from Part A in TΗF (50 mL) was cooled to approximately 0 0C under nitrogen, and iodomethane (4.12 g, 29 mmol) was added. Sodium hydride (1.16 g of 60% in mineral oil, 29 mmol) was added over a period of two minutes. The mixture was stirred at 0 °C for 20 minutes, allowed to warm to room temperature, and stirred overnight. Saturated aqueous ammonium chloride was added, and the mixture was extracted four times with tert-bxxty\ methyl ether. The combined extracts were dried over magnesium sulfate, filtered, and concentrated under reduced pressure to provide 1O g of a yellow oil. The oil was purified by automated flash chromatography (eluting with ethyl acetate) to provide 5.20 g of ethyl l-(2-methoxyethyl)-5-(tetrahydro- 2i/-pyran-4-ylmethyl)-l/f-ρyrazole-3-carboxylate as a pale yellow oil. Part C
Lithium hydroxide monohydrate (2.94 g, 70.2 mmol) was added to a solution of ethyl 1 -(2-methoxyethyl)-5-(tetrahydro-2i7~pyran-4-ylmethyl)- 1 i/-pyrazole-3-carboxylate (5.2 g, 17.5 mmol) in methanol (60 mL) and water (20 niL). The mixture was stirred for overnight. Most of the volatiles were removed under reduced pressure, and acetic acid (40 mL) and water were added. The solution was cooled to approximately 0 0C and stirred for one hour. The volatiles were removed under reduced pressure, and the residue was partitioned between water and chloroform. The aqueous layer was separated and extracted four times with chloroform and then adjusted to pH 4 with the addition of 1 M hydrochloric acid. The aqueous fraction was extracted again four times with chloroform. The combined organic fractions were dried over magnesium sulfate and sodium sulfate, filtered, and concentrated under reduced pressure. The residue was twice dissolved in heptane and concentrated and twice dissolved in toluene and concentrated to provide 5.42 g of 1 -(2-methoxyethyl)-5-(tetrahydro-2/-r-pyran-4-ylmethyl)-l/f-pyrazole-3-carboxylic acid containing some toluene and 3 mol% acetic acid. Part D
1 -(3 -Dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (3.69 g, 19.3 mmol) was added to a solution of the material from Part C and 1 -hydroxybenzotriazole (2.61 g, 19.3 mmol) in DMF (27.1 mL) at room temperature. The mixture was stirred for one hour, cooled in an ice bath, and treated with concentrated ammonium hydroxide (3.5 mL). A precipitate formed, and the mixture was stirred 15 minutes at 0 0C. Water (150 mL) was added, and the mixture was stirred. The precipitate was isolated by filtration, washed with water, and dried on the vacuum filter funnel to provide 1.44 g of l-(2- methoxyethyl)-5-(tetrahydro-2H-pyran-4-ylmethyl)-lH-pyrazole-3-carboxamide as a white solid. The filtrate was extracted six times with chloroform, and the combined extracts were dried over sodium sulfate, filtered, and concentrated under reduced pressure. The residue was twice dissolved in xylenes and concentrated under reduced pressure and then recrystallized from a mixture of 50% ethyl acetate in hexane (50 mL) and ethyl acetate (150 mL). The crystals were dried overnight on the vacuum filter funnel to provide an additional 2.027 g of l-(2-methoxyethyl)-5-(tetrahydro-2f/-pyran-4-ylmethyl)- lH-pyrazole-3-carboxamide as white plates. Anal. Calcd for C13H19N3O3: C, 58.41; H, 7.92; N9 15.72. Found: C, 58.52; H, 7.89; N, 15.86. Part E
The method described in Part F of Example 3 was used to treat l-(2- methoxyethyl)-5 -(tetrahydro-2H-pyran-4-ylmethyl)- 1 H-pyrazole-3 -carboxamide (3.41 g, 12.7 mmol) with trifluoroacetic anhydride (1.98 mL, 14.0 mmol) in the presence of triethylamine (3.86 g, 38.1 mmol) with the following modifications. Extractions were carried out four times with chloroform, and the automated flash chromatography column was eluted with ethyl acetate. l-(2-Methoxyethyl)-5-(tetrahydro-2H-pyran-4-ylmethyl)- l/f-pyrazole-3-carbonitrile (3.16 g) was obtained as a colorless oil. Part F Potassium acetate (3.12 g, 31.7 mmol) and bromine (2.84 g, 17.7 mmol) were sequentially added to a solution of l~(2-methoxyethyl)-5-(tetrahydro-2/i-pyran-4- ylmethyl)-lH-pyrazole-3-carbonitrile (3.16 g, 12.7 mmol) in acetic acid (25 mL). The reaction was stirred for 16 hours at room temperature. Saturated aqueous sodium hydrogensulfite was added until the reaction became colorless. Water was added, and a precipitate formed, was isolated by filtration, washed with water, and dried on the vacuum filter funnel to provide 3.78 g of 4-bromo-l-(2-methoxyethyl)-5-(tetrahydro-2H-pyran-4- ylmethyl)-lH-pyrazole-3-carbonitrile as a white solid containing about 2 mol% acetic acid. Part G Potassium carbonate ( 1.82 g, 13.2 mmol), 1 ,2-dimethoxyethane (DME) ( 15 mL), water (7.5 mL), and dichlorobis(triphenylphosphine)palladium(II) (140 mg, 0.20 mmol) were added to a mixture of 2-aminophenylboronic acid hydrochloride (1.39 g, 8.0 mmol) and 4-bromo- 1 -(2-methoxy ethyl)-5 -(tetrahydro-2H-pyran-4-ylmethy I)- 1 H-pyr azole-3 - carbonitrile (1.31 g, 4.00 mmol). The flask was placed under vacuum and back-filled with nitrogen four times. The reaction was heated under a nitrogen atmosphere at 95 °C for 90 minutes. The aqueous layer was separated and extracted twice with ført-butyl methyl ether. The organic fractions were combined, dried over magnesium sulfate, filtered, and concentrated to yield a black oil (2.2 g). The crude product was purified by automated flash chromatography (eluting with ethyl acetate) to provide 1.14 g of 4-(2-aminophenyl)- l-(2-methoxyethyl)-5-(tetrahydro-2H-pyran-4-ylmethyl)-lH-pyrazole-3-carbonitrile as a yellow, waxy solid. Part H
The method described in Part I of Example 3 was followed. Following recrystallization from ethyl acetate (40 mL), the crystals were dried on the vacuum filter funnel for 19 hours to provide 736 mg of 2-(2-methoxy ethyl)- 1 -(tetrahydro-2#-pyran-4- ylmethyl)-2H-pyrazolo[3,4-c]quinolin-4-amine as white crystals, mp 189-190 °C. MS (APCI) m/z 341 (M + H)+;
Anal. Calcd for C19H24N4O2: C, 67.04; H5 7.11; N5 16.46. Found: C, 67.03; H5 7.22; N, 16.55.
Example 6
2-(2-Methoxyethyl)- 1 -(tetrahydro-2H-pyran-4-ylmethyl)-2#-pyrazolo [3 ,4- c] [ 1 ,8]naρhthyridin-4-amine
Figure imgf000064_0001
Hydrochloric acid (10 mL of IM) was added to a solution of 2-fert-butoxycarbonylamino- 3-pyridylboronic acid (2.59 g, 10.9 mmol), and the resulting mixture was heated at 80 °C for 45 minutes and allowed to cool to room temperature. Potassium carbonate (3.60 g, 26.0 mmol) was added with stirring, and then DME (20 mL), 4-bromo-l-(2- methoxyethyl)-5-(tetrahydro-2H-pyran-4-ylmethyl)-lH-pyrazole-3-carbonitrile (1.75 g, 5.33 mmol), and dichlorobis(triphenylphosphine)palladium(II) (190 mg, 0.27 mmol) were added. The reaction was carried out as described in Part C of Example 4. The crude product was purified by automated flash chromatography (eluting with 0% to 35% CMA in chloroform) followed by recrystallization from acetonitrile (30 mL) after hot filtration. The crystals were washed with cold acetonitrile and dried overnight in a vacuum oven at 60 0C to provide 0.13 g of 2-(2-methoxyethyl)-l-(tetrahydro-2H-ρyran-4-ylmethyl)-2Jf- pyrazolo[3,4-c][l ,8]naphthyridin-4-arnine as light yellow needles, mp 230-233 °C.
Anal, calcd for Ci8H23N5O2: C, 63.32; H, 6.79; N5 20.51. Found: C, 63.37; H, 6.84; N, 20.59. Example 7 l-[(4-Fluorotetrahydro-2H-pyran-4-yl)methyl]-2-methyl- 2H-pyrazolo[3,4-c]quinolin-4~arnme
Figure imgf000065_0001
Part A
Diethyl 2-oxopropylphosphonate (12.8 niL, 1.1 eq) was added to a stirred solution of tetrahydropyran-4-one (6.06 g, 1.0 eq) in ethanol (148 mL) and then the mixture was cooled to 0 °C with an ice bath. A solution of potassium hydroxide (3.74 g, 1.1 eq) in water (38 mL) was added over a period of about 2 minutes. The ice bath was removed and the reaction mixture was stirred for 3 hours. The bulk of the ethanol was removed under reduced pressure while maintaining the temperature at <20 °C to provide about 32 g of a yellow liquid. The liquid was diluted with water (150 mL) and then extracted with dichloromethane (2 x 150 mL). The combined organics were dried over magnesium sulfate and then concentrated under reduced pressure while maintaining the temperature at <20 0C to provide 12.05 g of a pale yellow liquid. This material was purified by automated flash chromatography (eluting with 20% ethyl acetate in hexanes for 2 column volumes, with a gradient of 20-40% ethyl acetate in hexanes over 5 column volumes, and with 40% ethyl acetate in hexanes for 2 column volumes) to provide 7.02 g of 1- tetrahydro-4/i-pyran-4-ylideneacetone. Analysis by 1H NMR indicated clean product with residual ethyl acetate (14%). Part B
A solution of sodium tert-butoxide (5.29 g, 1.1 eq) in ethanol (51 mL) was added to a mixture of the material from Part A (LO eq) and diethyl oxalate (7.45 mL, 1.1 eq). The vessel containing the material from part A and diethyl oxalate was rinsed with additional ethanol (27 mL) and the rinse was added to the reaction mixture. The reaction mixture was stirred for 2 hours and then cooled to 0 °C. Acetic acid (57 mL) was added and the reaction mixture was stirred for 5 minutes. Methylhydrazine (2.64 mL, 1.0 eq) was added dropwise. After 15 minutes the ice bath was removed and the reaction mixture was stirred overnight. The reaction mixture was concentrated under reduced pressure and the residue was partitioned between 50% aqueous sodium carbonate (450 niL) and dichloromethane (250 mL). The aqueous layer was back extracted with dichloromethane (100 mL). The combined organics were dried over magnesium sulfate and then concentrated under reduced pressure at ambient temperature to provide 11.76 g of a yellow oil. This material was purified by automated flash chromatography (eluting with 30% ethyl acetate in hexanes for 1 column volume, with a gradient of 30-70% ethyl acetate in hexanes over 6 column volumes, and with 70% ethyl acetate in hexanes for 1 column volume) to provide 4.99 g of ethyl l-methyl-5-(tetrahydro-4H-pyran-4- ylidenemethyl)-l/i-pyrazole-3-carboxylate as a yellow oil. Analysis by HPLC, LCMS, TLC, and 1H NMR indicated clean product with residual ethyl acetate (14%). Part C
A solution of sodium hydroxide (1.99 g, 2.5 eq) in water (5 mL) was added to a stirred solution of the material from Part B (1.0 eq) in ethanol (50 mL). The reaction mixture was stirred for 30 minutes and then the bulk of the ethanol was removed under reduced pressure. The residue was diluted with dichloromethane (250 mL) and water (50 mL). The aqueous layer was acidified (pH about 1-2) with hydrochloric acid (50 mL of 1 M). The layers were separated and the aqueous layer was back extracted with dichloromethane (4 x 125 mL). The combined organics were dried over magnesium sulfate and then concentrated under reduced pressure to provide 4.82 g of l-methyl-5- (tetrahydro-4H"-pyran-4-ylidenemethyl)-l/f-ρyrazole-3-carboxylic acid. Part D
1-Hydroxybenzotriazole hydrate (3.23 g, 1.2 eq) and l-(3-dimethylarninopropyl)- 3-ethylcarbodiimide hydrochloride (EDC, 4.58 g, 1.2 eq) were added sequentially to a solution of the material from Part C (1.0 eq) in DMF (25 mL). After 25 minutes the EDC was dissolved. After 1 hour the reaction mixture was cooled to 0 0C and concentrated ammonium hydroxide (5.3 mL, 4.0 eq) was added. Solids formed several minutes later. The reaction mixture was stirred for 30 minutes, diluted with water (100 mL), and then stirred for 20 minutes. The solids were isolated by filtration, rinsed with water (2 x 25 mL), and then dried to provide 3.69 g of l-methyl-5-(tetrahydro-4H-pyran-4- ylidenemethyl)-lH-pyrazole-3-carboxamide as a white powder. Part E
3-Chloroperoxybenzoic acid (4.51 g, 1.1 eq based on 70% titer) was added to a stirred suspension of the material from Part D (1.0 eq) in chloroform (83 ml). After several minutes a solution was obtained. The solution was stirred overnight, diluted with additional chloroform (275 mL), and then washed with a solution of saturated sodium bicarbonate:5% sodium hydroxide (20:1, 1 x 150 mL, then 1 x 100 mL). The organic layer was dried over magnesium sulfate and then concentrated under reduced pressure to provide a voluminous white foam. This material was concentrated from ethanol to provide 3.94 g of 5-(l,6-dioxaspiro[2.5]oct-2-yI)-l-methyl-lH-pyrazole-3-carboxamide as a white solid. Part F
A suspension of the material from Part E (1.0 eq) in ethanol (170 mL) was warmed until most of the solid was dissolved and then cooled to 35 0C. 10% Palladium on carbon (400 mg) and ammonium formate (5.24 g, 5.0 eq) were added sequentially. The reaction mixture was allowed to cool to ambient temperature and stirred for 4 hours. The reaction mixture was filtered through a layer of CELITE filter agent. The filter cake was rinsed sequentially with ethanol (3 x 25 mL), methanol (4 x 25 mL), and 1:1 methanol :chloroform (total of about 500 mL). The filtrate was concentrated under reduced pressure to provide 5.74 g of a white solid. This material was triturated with ethanol (45 mL), isolated by filtration, rinsed with ethanol (3 x 8 mL), and then dried to provide 3.13 g of 5-[(4-hydroxytetrahydro-2H-pyran-4-yl)methyl]-l-methyl-lH-pyrazole- 3-carboxamide as a white crystalline solid. Part G
Triethylamine (5.13 mL, 3.0 eq) was added to a stirred suspension of a portion of the material from Part F (2.91 g, 1.0 eq). The mixture was cooled to 0 °C and trifluoroacetic anhydride (5.14 mL, 3.0 eq) was added dropwise over a period of 5 minutes. The reaction mixture was stirred for 2 hours, quenched with saturated sodium carbonate (50 mL), and then allowed to warm to ambient temperature. Water (50 mL) and dichloromethane (200 mL) were added sequentially. The organic layer was separated, dried over magnesium sulfate, and then concentrated under reduced pressure to provide a yellow oil. The oil was dissolved in methanol (80 mL). Solid potassium carbonate (420 mg, 0.25 eq) was added and the mixture was stirred for 30 minutes. Aqueous hydrochloric acid (1.7 mL of 7 M, 1.0 eq) was added, the solution was stirred for 10 minutes, and then the bulk of the methanol was removed under reduced pressure. The residue was partitioned between dichloromethane (200 mL) and water (50 mL). The pH of the aqueous layer was adjusted to 7-8 with saturated sodium bicarbonate. The layers were separated and the aqueous layer was back extracted with dichloromethane (2 x 75 mL). The combined organics were dried over magnesium sulfate and then concentrated under reduced pressure to provide 2.37 g of a light yellow semisolid. This material was triturated with 5:95 methanol: chloroform (10 mL). A solid was isolated by filtration and rinsed with 5:95 methanol :chloroform (1 x 6 mL, then 1 x 2 mL) to provide 536 mg of 5- [(4-hydroxytetrahydro-2i/-pyran-4-yl)methyl]-l-methyl-li/-pyrazole-3-carbonitrile as a white solid (lot 1). The filtrate was concentrated to provide a viscous yellow oil which was crystallized from warm ethanol (about 15 mL). A solid was isolated by filtration, rinsed with ethanol (2 x 5 mL), and then dried to provide 380 mg of 5-[(4- hydroxytetrahydro-2H-pyran-4-yl)methyl] - 1 -methyl- 1 H-pyrazole-3 -carbonitrile as prisms (lot 2). The filtrate was concentrated to a semisolid. This material was purified by automated flash chromatography (eluting with 5:95 methanolxhloroform for 8 column volumes) to provide 0.47 g of 5-[(4-hydroxytetrahydro-2H-pyran-4-yl)methyl]-l-methyl- l/f-pyrazole-3 -carbonitrile (lot 3) as a white solid. Part H A solution of material from Part G (lots 1 and 2, 1.0 eq) in dichloromethane (41 mL) was cooled to 0 0C. [Bis(2-methoxyethyl)amino]sulfur trifluoride (1.14 mL, 1.5 eq) was added dropwise. The reaction mixture was stirred for 45 minutes, quenched with saturated sodium bicarbonate (20 mL), and allowed to warm to ambient temperature. Saturated sodium bicarbonate (50 mL) and dichloromethane (50 mL) were added sequentially. The layers were separated and the aqueous layer was back extracted with dichloromethane (30 mL). The combined organics were dried over magnesium sulfate and then concentrated under reduced pressure at ambient temperature to provide 1.19 g of a light orange oil. This material was purified by automated flash chromatography (eluting with 10% ethyl acetate in 2:1 hexanes: dichloromethane for 2 column volumes, a gradient of 10-30% ethyl acetate in 2:1 hexanes: dichloromethane over 8 column volumes, and 30% ethyl acetate in 2:1 hexanes:dichloromethane for 4 column volumes) to provide 401 mg of 5- [(4-fluorotetrahydro-2H-pyran-4-yl)methyl]- 1 -methyl- 1 /7-pyrazole-3 -carbonitrile as a clear colorless oil which crystallized on standing.
Part i
Potassium acetate (437 mg, 2.5 eq) was added to a stirred solution of the material from Part H (1.0 eq) in acetic acid (4 mL). After the potassium acetate had dissolved, bromine (0.13 mL, 1.4 eq) was added dropwise. The resulting red solution was stirred for 18 hours. Additional potassium acetate (2 eq), acetic acid (0.5 mL), and bromine (1.4 eq) were added sequentially. The reaction mixture was stirred for 2.5 hours and then quenched with saturated sodium thiosulfate until colorless (about 4 mL). The reaction mixture was concentrated under reduced pressure and then diluted with water (10 mL) and saturated sodium carbonate (10 mL added dropwise). The resulting foamy suspension was combined with dichloromethane (50 mL). The aqueous layer was adjusted to about pH 8 with 5% sodium hydroxide. The layers were separated and the aqueous layer was back extracted with dichloromethane (50 mL). The combined organics were dried over magnesium sulfate and then concentrated under reduced pressure to provide 0.59 g of a light yellow solid. The bulk of the material was dissolved in 10:90 methanol :dichloromethane (40 mL), loaded onto silica gel, and then purified by automated flash chromatography (eluting with dichloromethane for 3 column volumes, a gradient of 0-10% ethyl acetate in dichloromethane over 5 column volumes, and then 10% ethyl acetate in dichloromethane for 10 column volumes) to provide 474 mg of 4-bromo-5-[(4- fluorotetrahydro-2H-pyran-4-yl)methyl]-l -methyl- lH-pyrazole-3-carbonitrile as a white solid. Part J
Water (2.5 mL) was added dropwise to a stirred solution of the material from Part I (1.0 eq) in 1,2-dimethoxyethane (7.5 mL). 2-Aminophenylboronic acid hydrochloride
(460 mg, 1.7 eq) and potassium carbonate (712 mg, 3.3 eq) were added sequentially. The reaction mixture was purged with nitrogen and then dichlorobis(triphenylphosphine)palladium(II) (22 mg, 0.02 eq) was added. The reaction mixture was heated at 75 0C for 9 hours and then allowed to cool to ambient temperature over the weekend. The reaction mixture was concentrated under reduced pressure and the residue was partitioned between dichloromethane (50 mL) and water (30 mL). The organic layer was separated, dried over magnesium sulfate, and then concentrated under reduced pressure to provide 2 g of a brown oil. This material was dissolved in ethanol (10 mL) and a precipitate formed immediately. Hydrochloric acid (0.90 mL of 7 M5 4.0 eq) was added and the resulting solution was heated at 75 °C for 4 hours. The reaction mixture was cooled to ambient temperature and then concentrated under reduced pressure. The residue was partitioned between dichloromethane (75 mL) and an aqueous mixture of saturated sodium bicarbonate (25 mL) and water (25 mL). The aqueous layer was separated and then extracted with dichloromethane (2 x 40 mL). The combined organics were dried over magnesium sulfate and then concentrated under reduced pressure to provide 0.66 g of a brown foam, This material was purified by automated flash chromatography (eluting with a gradient of 0-20% CMA in chloroform over 10 column volumes and 20% CMA in chloroform for 3 column volumes) to provide 200 mg of a tan solid. This material was triturated with hot ethanol (about 8 mL), isolated by filtration, rinsed with ethanol (3 x 5 mL), and then dried (0.15 torr (20 Pa), 130 0C, 2 hours) to provide 143 mg of l-[(4-fluorotetrahydro-2H'-pyran-4-yl)methyl]-2-methyl-2H- ρyrazolo[3,4-c]quinolin-4-amine as a tan powder, mp 252-254 0C. 1H NMR (500 MHz, DMSO-de): δ 8.16 (d, J=7.8 Hz, IH), 7.47 (dd, J=1.2, 8.1 Hz, IH), 7.32 (m, IH), 7.17 (m, IH), 6.65 (br s, 2H), 4.12 (s, 3H), 3.73 (m, 4H)5 3.45 (m, 2H), 2.08 (m, IH), 2.00 (m, IH), 1.65 (m, 2H); MS (ESI) 315 m/z (M+H)+; Anal, calcd for C17Hi9FN4O0.04 EtOH»0.25 H2O: C3 63.97; H, 6.20; N, 17.47. Found: C5 63.97; H5 6.10; N, 17.49.
Example 8
4-[(4-Amino-2-methyl-2H-pyrazolo[3,4-c]quinolin-l-yl)methyl]tetrahydiO-2H-pyran-4-ol
Figure imgf000070_0001
Part A
Potassium acetate (0.52 g, 2.5 eq) was added to a stirred solution of 5-[(4- hydroxytetrahydro-2H-pyran-4-yl)methyl]-l-methyl-liϊ-pyrazole-3-carbonitrile (Example 7, Part G, lot 3, 0.47 g, 1.0 eq) in acetic acid (7 mL). The reaction mixture was stirred until all of the potassium acetate was dissolved. Bromine (0.15 niL, 1.4 eq) was added dropwise and the reaction was stirred for 16 hours. The reaction mixture was quenched with saturated sodium thiosulfate (4 mL) and then the bulk of the acetic acid was removed under reduced pressure. The residue was diluted with water (15 mL) and saturated sodium carbonate (about 10-12 mL) and then extracted with dichloromethane (3 x 25 mL). The combined organics were dried over magnesium sulfate and then concentrated under reduced pressure to provide 0.72 g of a yellow solid. This material was purified by automated flash chromatography (eluting with 30% CMA in chloroform for 7 column volumes) to provide 528 mg of 4-bromo-5-[(4-hydroxytetrahydro-2H-pyran-4-yl)methyl]- 1 -methyl- lH-pyrazole-3-carbonitrile as a colorless sticky semisolid. Part B
The material from Part A (1.0 eq) was coupled with 2-aminophenylboronic acid hydrochloride (1.8 eq) and then cyclized using the general method of Example 7 Part J. The crude product was purified by automated flash chromatography (eluting with 20% CMA in chloroform for 3 column volumes, a gradient of 20-40% CMA in chloroform over 10 column volumes, and 40% CMA in chloroform for 3 column volumes) to provide a tan solid. This material was triturated with hot ethyl acetate (about 8 mL), isolated by filtration, rinsed with ethyl acetate (3 x 5 mL), and dried under high vacuum to provide 106 mg of 4-[(4-amino-2-methyl-2i/-pyrazolo[3,4-c]quinolin- 1 -yl)methyl]tetrahydro-2H- pyran-4-ol as a tan powder, mp 260-262 (dec) °C. 1H NMR (500 MHz, DMSOd6): 58.19 (dd, J=1.0, 8.0 Hz, IH), 7.46 (dd, J=1.2, 8.1 Hz, IH), 7.30 (m, IH), 7.17 (m, IH)5 6.61 (s, 2H), 4.77 (s, IH), 4.16 (s, 3H), 3.55 (m, 4H), 3.41 (s, 2H)5 1.84 (m, 2H), 1.44 (br s, IH), 1.39 (br s, IH); MS (APCI) 313 m/z (M+H)+; Anal, calcd for C17H20N4O2O^ H2O: C, 64.44; H, 6.52; N, 17.68. Found: C, 64.11; H, 6.32; N, 17.60
Example 9
2-Ethyl- 1 -[(4-fluorotetrahydro-2J7-pyran-4-yl)methylj- 2#-pyrazolo[3,4-c]quinolin-4-amine
Figure imgf000072_0001
2-Ethyl- 1 -[(4-fluorotetrahydro-2/f-pyran-4-yl)methyl]-2//-ρyrazolo[354- cJquinolin-4-amine was prepared according to the general method of Example 7 Parts A through J using 1.3 eq of ethylhydrazine oxalate in lieu of methylhydrazine in part B. The crude product was purified by automated flash chromatography (eluting with a gradient of 0-20% CMA in chloroform over 5 column volumes and 20% CMA in chloroform for 5 column volumes) to provide 170 mg of a tan solid. This material was triturated with methanol (about 12 mL), isolated by filtration, rinsed with methanol (3 x 5 mL), and dried under high vacuum to provide 112 mg of 2-ethyl-l-[(4-fluorotetrahydro-2/J-pyran-4- yl)methyl]-2//-pyrazolo[3,4-c]quinolin-4-amine as a tan powder, mp 275-277 °C. 1H NMR (500 MHz, DMSOd6): δ 8.15 (d5 J=7.6 Hz, IH), 7.48 (dd, J=I .2, 8.1 Hz, IH), 7.20 (m, IH), 7.17 (m, IH), 6.64 (br s, 2H), 4.44 (q, J=7.2 Hz, 2H), 3.73 (m, 4H), 3.44 (t, J=I 1.3 Hz, 2H), 2.10 (ddd, J=5.4, 13.5, 13.5 Hz, IH), 1.97 (ddd, J=5.6, 13.6, 13.6 Hz, IH), 1.64 (m, 2H), 1.48 (t, J=7.1 Hz, 3H); MS (ESI) 329 m/z (M+H)+; Anal, calcd for Ci8H2IFN4O: C, 65.84; H, 6.45; N, 17.06. Found: C, 65.56; H, 6.24; N, 16.93.
Example 10
4-[(4-Amino-2-ethyl-2/Z-pyrazolo[3,4-c]quinolin-l-yl)methyl]tetrahydro-2H-pyran-4-ol
Figure imgf000072_0002
Part A
Di(fer/-butyl) 2-ethyl-l-methyl-2H-pyrazolo[3,4-c]quinolin-4-ylimidodicarbonate was prepared according to the method of Example 1 Part E using 2-ethyl-l-methyl-2H- pyrazolo[3,4-c]quinolin-4-amine (International Publication Number WO 2005/079195, Example 35) in lieu of l-memyl-2-propyl-2H~pyrazolo[3,4-c]quinolin-4-amine. The crude product was triturated with hexanes to provide 13.34 g di(ført-butyl) 2-ethyl-l- memyl-2H-pyrazolo[3,4-c]quinolin~4-ylimidodicarbonate as a white granular solid.
Part B
Di(jfert-butyl) 2-ethyl-l-methyl-2/i-pyrazoIo[3,4-c]quinolin-4-ylimidodicarbonate was reacted with tetrahydro-4/i-pyran-4-one according to the method of Example 2 using di(ført-butyl) 2-ethyl-l-methyl-2H-pyrazolo[3,4-c]quinolin-4-ylimidodicarbonate in lieu of di(fert-butyl) 1 -methyl-2~propyl-2H~-pyrazolo [3 ,4-c]quinolin-4-ylimidodicarbonate.
The crude product was recrystaUized acetonitrile provide 0.349 g 4-[(4-amino-2-ethyl-2i7- pyrazolo[3,4-c]quinolin-l-yl)methyl]tetrahydro-2/f-pyran-4-ol as off-white crystals, mp 253-255 0C.
MS (ESI) m/z 327.28 (M + H)+
Anal. Calcd for C]8H22N4O2: C5 66.24; H, 6.79; N, 17.16. Found: C, 66.45; H, 7.05; N,
17.24.
Example 11 l-[(4-Amino-2-ethyl-2H-pyrazolo[3,4-c]quinolin-l-yl)methyl]cyclohexanol
Figure imgf000073_0001
l-[(4-Amino-2-ethyl-2H-pyrazolo[3,4-c]quinolin-l-yl)methyl]cyclohexanol was prepared according to the method of Example 1 Part F using di(tert-butyX) 2-ethyl-l- methyl-2//-pyrazolo[3,4-c]quino]in-4-ylimidodicarbonate in lieu of di(tert-butyϊ) 1- methyl-2-propyl-2/f-pyrazolo[3,4-c]quinolin-4-ylimidodicarbonate and cyclohexanone in lieu of cyclobutanone. The crude product was recrystaUized from acetonitrile to provide 0.560 g of l-[(4-amino-2-ethyl-2/-r-pyrazolo[3,4-c]quinolin-l-yl)methyl]cyclohexanol as a white powder, mp 211.5-213 °C. MS (ESI) m/z 325.23 (M + H)+
Anal. Calcd for Ci9H24N4O: C, 70.34; H3 7.46; N5 17.27. Found: C, 70.19; H5 7.57; N5 17.35.
Example 12 l-[(4-Amino-2-ethyl-2H-pyrazolo[3,4-c]quinolin-l-yl)methyl]cyclopentanol
Figure imgf000074_0001
l-[(4-Amino-2-ethyl-2H-pyrazolo[3,4-c]quinolm-l-yl)methyl]cyclopentanol was prepared according to the method of Example 1 Part F using di(tert-butyl) 2-ethyl-l- methyl-2H-pyrazolo[3,4-c]quinolin-4-ylimidodicarbonate in lieu of di(fert-butyl) 1- methyl-2-propyl-2/f-pyrazolo[3,4-c]quinolin-4-ylimidodicarbonate and cycpentanone in lieu of cyclobutanone. The crude product was triturated with acetonitrile and isolated by filtration to provide 0.414 g of l-[(4-amino-2-ethyl-2H-pyrazoIo[3,4-c]quinolin-l- yl)methyl]cyclopentanol as a white powder, mp 256-259 0C. MS (ESI) m/z 311.32 (M + H)+
Anal. Calcd for C18H22N4OO^CHCl3: C5 69.20; H5 7.10; N5 17.91. Found: C, 68.95; H5 6.79; N, 17.83.
Example 13 l-[(4-Amino-2-ethyl-2i/-pyrazolo[354-c]quinolin-l-yl)methyl]cyclobutanol
Figure imgf000074_0002
1 -[(4-Amino-2-ethyl-2#-pyrazolo [3 ,4-c]quinolin- 1 -yl)methyl]cyclobutanol was prepared according to the method of Example 1 Part F using di(tert-butyl) 2-ethyl-l- methyl-2#-pyrazolo[3,4-c]quinolin-4-ylimidodicarbonate in lieu of di<7e/Y-butyl) 1- methyl-2-propyl-2H-pyrazolo[3,4-c]quinolin-4-ylimidodicarbonate. The crude product was triturated with acetonitrile and isolated by filtration to provide 0.396 g of l-[(4-amino- 2-ethyl-2H-pyrazolo[3,4-c]quinolin-l-yl)methyl]cyclobutanol as a white solid, mp 245- 247 0C,
MS (ESI) m/z 297.26 (M + H)+
Anal. Calcd for Ci7H20N4O: C5 68.90; H, 6.80; N5 18.90. Found: C, 68.66; H5 6.72; N, 18.83.
Example 14
4-[(4-Amino-2-ethyl-2H-pyrazolo[354-c]quinolin-l-yl)methyl]tetrahydro-
2H-thiopyran-4-ol
Figure imgf000075_0001
4-[(4-Amino-2-ethyl-2H-pyrazolo[354-c]quinolin-l-yl)methyl]tetrahydro-2H- thiopyran-4-ol was prepared according to the method of Example 1 Part F using di(tert- butyl) 2-ethyl-l-methyl-2H-pyrazolo[3,4-c]quinolin-4-ylimidodicarbonate in lieu of di(tert-butyl) l-methyl-2-propyl-2//-pyrazolo[3,4-c]quinolin-4-ylimidodicarbonate and 4- oxothiane in lieu of cyclobutanone. The crude product was recrystallized from acetonitrile to provide 0.475 g of 4-[(4-amino-2-ethyl-2H-pyrazolo[3,4-c]quinolin-l- yl)methyl]tetrahydro-2H-thiopyran-4-ol as a white powder, mp 251.5-255 °C. MS (ESI) m/z 343.29 (M + H)+ Anal. Calcd for Ci8H22N4OS: C5 63.13; H5 6.48; N5 16.36; S5 9.36. Found: C5 63.02; H5 6.67; N5 16.37; S, 9.36. Example 15 l-Acetyl-4-[(4-amino-2-ethyl-2/i-pyrazolo[354-c]quinolin.-l-yl)methyl]piperidin-4-ol
Figure imgf000076_0001
Part A 4-[(4-Amino-2-ethyl-2/f-pyrazolo[3,4-c]quinolin-l -yl)methyl]piperidin-4-ol was prepared according to the method of Example 1 Part F using di(tert-butyl) 2-ethyl-l- methyl-2H-pyrazolo[3,4-c]quinolin-4-ylimidodicarbonate in lieu of di(tert-bntyl) 1- methyl-2~propyl-2H-pyrazolo[3,4-c]quinolin-4-ylimidodicarbonate and tert-butyl A- oxopiperidine-1-carboxylate in lieu of cyclobutanone. The crude product was recrystallized from acetonitrile to provide 1.09 g of l-[(4-amino-2-ethyl-27i-pyrazolo[3,4- c]quinolin-l-yl)methyl]piperidin-4-ol as an off-white solid. Part B
Acetic anhydride (88 μL, 0.922 mmol) was added to a slurry of 4-[(4-amino-2- ethyl-2//-pyrazolo[3,4-c]quinolin-l-yl)methyl]piperidin-4-ol (0.300 g, 0.922 mmol) in chloroform (10 niL). After 16 hours, the solution was purified via automated flash chromatography eluting with a linear gradient of 2-25% CMA in chloroform. The residue was triturated in acetonitrile and isolated by filtration to provide 0.209 g of l-acetyl-4-[(4- amino-2-ethyl-2H-pyrazolo[3,4-c]quinolin-l-yl)methyl]piperidin-4-ol as a white solid, mp 231-232.5 0C. MS (ESI) m/z 368.20 (M + H)+
Anal. Calcd for C20H25N5O2: C, 65.37; H, 6.86; N, 19.06. Found: C, 65.14; H, 7.03; N, 19.25. Example 16
4-[(4-Amino-2-ethyl-2//-pyrazolo[3,4-c?]quinolin-l-yl)methyl]- l-(methylsulfonyl)piperidin-4-ol
Figure imgf000077_0001
Methanesulfonic anhydride (0.160 g, 0.922 mmol) was added to a slurry of 4-[(4- amino-2-ethyl-2H-pyrazolo[3,4-c]quinolin-l-yl)methyl]piperidin-4-ol (0.300 g, 0.922 mmol) in chloroform (10 mL). After 16 hours, 2M aqueous sodium carbonate was added and the biphasic mixture was stirred for 30 minutes resulting in a white precipitate. The mixture was extracted with 10% methanol in dichloromethane. The solution was concentrated. The residue was purified via automated flash chromatography and recrystallized from acetonitrile to provide 0.165 g of 4-[(4-amino-2-ethyl-2H- pyrazolo[3,4-c]quinolin-l-yl)methyl]-l-(methylsulfonyl)piperidin-4-ol as a white solid, mp 268-270 0C. MS (ESI) m/z 404.30 (M + H)+ Anal. Calcd for C19H25N5O3S^OJ H2OO.15 CH3CN: C5 55.97; H, 6.32; N, 17.43; S, 7.74. Found: C, 56.33; H, 6.23; N, 17.56; S, 7.73.
Example 17
4-[(4-Amino-2-ethyl-2/f.pyrazolo[3,4-c]quinolm-l-yl)methyl]- 4-hydroxy-iV-propylpiperidine- 1 -carboxamide
Figure imgf000078_0001
^-Propyl isocyanate (86 μL, 0.922 mmol) was added to a slurry of 4-[(4-amino-2- ethyl-2H-pyrazolo[3,4-c]quinolin-l-yl)methyl]piperidin-4-ol (0.300 g, 0.922 mmol) in chloroform (10 mL). After 16 hours, the solution was purified via automated flash chromatography eluting with a linear gradient of 2-25% CMA in chloroform. The residue was triturated in acetonitrile and isolated by filtration to provide 0.277 g of 4-[(4-amino-2- ethyl-2//-pyrazolo [3 ,4-c]quinoIin- 1 -yl)methyl]-4-hydroxy-JV-propylpiperidine- 1 - carboxamide as a flocculent white solid, mp 208.5-210 0C. MS (ESI) m/z 411.28 (M + H)+
Anal. Calcd for C22H30N6O2O-S H2O: C, 63.53; H, 7.42; N, 20.21. Found: C, 63.49; H9 7.16; N, 20.09.
Example 18
2-Ethyl-l-(tetrahydro-2H-pyran-4-ylmethyl)-2H-pyrazolo[3,4-c]quinolin-4-amine
Figure imgf000078_0002
2-Ethyl-l-(tetrahydro-2H-pyran-4-ylmethyl)-2H-pyrazolo[3,4-c]quinolin-4-amine was prepared according to the methods of Example 3 parts A-I. Ethylhydrazine oxalate was used in lieu of propylhydrazine oxalate in part C. The crude product was recrystallized from acetonitrile to provide 0.371 g of 2-ethyl-l-(tetrahydro-2H-pyran-4- ylmethyl)~2H-pyrazolo[3,4-c]quinolin-4-amine as tan crystals, m.p. 226.0-228.0 0C. MS (ESI) m/z 311.29 (M + H)+ Anal. Calcd for Ci8H22N4O: C5 69.65; H5 7.14; N5 18.05. Found: C5 69.81; H5 7.26; N5 18.29.
Example 19 l-[(4-Amino-2-ethyl-657,859-tetrahydro-2H-pyrazolo[354-c]quinolin-l- yl)methyl]cyclohexanol
Figure imgf000079_0001
1 -[(4-Amino-2-ethyl-2ϋ/-pyrazolo[3,4-c]quinolin-l -yl)methyl]cyclohexanol (0.226 g, 0.70 mmol) and platinum(IV) oxide (0.107 g, 0.47 mmol) were slurried in trifluoroacetic acid (10 mL). The flask was degassed three times and charged to 50 psi hydrogen (3.45 x 105 Pa). After 16 hours, the catalyst was removed via filtration through a bed of CELITE filter agent, rinsing with methanol. The filtrate was concentrated. The oily residue was dissolved in 6N hydrochloric acid (3 mL). The acidic mixture was brought to pH 14 using 50% aqueous sodium hydroxide. The mixture was extracted with 10% methanol in dichloromethane, dried over anhydrous sodium sulfate, and concentrated. The material was purified via automated flash chromatography eluting with a linear gradient of 2-20% CMA in chloroform. The resulting white solid was triturated in acetonitrile and then isolated by filtration to provide 0.085 g of l-[(4-amino-2-ethyl- 657,8,9-tetrahydro-2H-pyrazolo[3,4-c]quinolin-l-yl)methyl]cyclohexanol as a white solid, m.p. 232-234 °C. MS (ESI) m/z 329.44 (M + H)+
Anal. Calcd for C19H28N4O: C, 69.48; H, 8.59; N5 17.06. Found: C, 69.28; H, 8.51; N, 17.06. Example 20
1 -{ [4-Fluoro- 1 -(methylsulfonyl)piperidin-4-yl]methyl }-2-methyl-2H-pyrazolo[3,4- c] quinolin-4-amine
Figure imgf000080_0001
Part A
The method described in Part A of Example 3 was followed using l-Boc-4- piperidinone (46.6 g, 0.234 mol) instead of tetrahydro-4H-pyran~4-one. The crude product was purified by column chromatography on silica gel (eluting with 5% ethyl acetate in hexanes) and then concentrated from ethyl acetate/hexanes to provide 23.7 g of tert-butyl 4-(2-oxopropylidene)piperidine- 1 -carboxylate as a white solid. Part B
A solution oftert-butyl 4-(2-oxopropyIidene)piperidine-l -carboxylate (20.0 g, 83.6 mmol) and diethyl oxalate (24.9 mL, 2.2 equivalents (eq.)) in ethanol (15 mL) was added to a solution of potassium ethoxide (15.47 g, 2.2 eq.) in ethanol (170 mL) at room temperature. The addition vessel was rinsed with ethanol (2x5 mL), and the rinses were added to the reaction. The reaction was stirred for 1 hour and then cooled in an ice bath to 4 °C. Acetic acid (100 mL) was added, and the reaction was stirred for several minutes to allow internal temp to return to 4 0C. Methyl hydrazine (19.4 mL, 4.4 eq) was added dropwise over a period of five minutes. After 15 minutes the ice bath was removed, and the reaction mixture was stirred for 1 hour. The reaction mixture was concentrated under reduced pressure and the residue was partitioned between water (300 mL) and dichloromethane (400 mL). Saturated aqueous sodium carbonate (100 mL) was added portionwise followed by 50% aqueous sodium hydroxide (40 mL) and 5% aqueous sodium hydroxide (100 mL) to adjust the mixture to pH 8. The aqueous layer was separated and extracted with dichloromethane (2 x 300 mL). The combined organics were dried over magnesium sulfate and then concentrated under reduced pressure to provide 38 g of brown oil. This material was purified by automated flash chromatography (eluting with tert-butyl methyl ether) to provide 62 g of tert-butyl 4-[(5-ethoxycarbonyl-2-methyl- 2H-pyrazol-3-yl)rnethylene]piperidine-l-carboxylate as a yellow oil. Part C
The material from Part B (6.03 g, 17.2 mmol) was treated according to the method of Part C of Example 7 with the modification that at the end of the reaction, the reaction mixture was cooled to 00C, and hydrochloric acid (40 mL of 1 M) was added dropwise. Approximately half the solvent was removed under reduced pressure, and a precipitate formed and was collected by filtration, washed with cold water (2 x 25 mL), and dried under high vacuum to provide 3.71 g of tert-butyl 4-[(5-carboxy-2-methyl-2H-pyrazol-3- yl)methylene]piperidine- 1 -carboxylate as a white solid. Part D
The material from Part C was treated according to the methods of Parts D, E, and F of Example 7. The crude solid obtained from Part F was partitioned between dichloromethane (200 mL) and water (100 mL). The aqueous solution was separated and extracted with chloroform (3 x 100 mL). The organic fractions were combined, dried over magnesium sulfate, and filtered. The filter cake was washed with chloroform (5 x 60 mL). The combined filtrates were concentrated under reduced pressure to provide 6.33 g of white foam, which was suspended in chloroform (125 mL), isolated by filtration, washed with chloroform (2 x 10 mL), and dried to provide 3.8 g of tert-butyl 4-[(5-carbamoyI-2- methyl-2H-pyrazol-3-yl)methyl]-4-hydroxypiρeri dine- 1 -carboxylate as a white solid. The filtrate was concentrated and dried to give an additional 3.0 g of product. Both solids contained chloroform and a trace impurity. Part E
The material from Part D was treated with triethylamine and trifluoroacetic anhydride according to the method of Part G of Example 7. The crude yellow oil that was obtained was purified by automated flash chromatography (eluting with a gradient of 30% hexanes in tert-butyl methyl ether to 100% tert-butyl methyl ether over 4 column volumes and then 100% tert-butyl methyl ether for 3 column volumes) and dried under high vacuum to provide 3.62 of the trifluoroacetate ester of tert-butyl 4-[(5-cyano-2-methyl- 27i-pyrazol-3-yl)methyl]-4-hydroxypiperidine-l -carboxylate as a colorless oil. A portion of the oil (3.43 g) was dissolved in methanol (80 mL), and concentrated ammonium hydroxide (1.6 mL) was added. The reaction was stirred for 1 hour and concentrated under reduced pressure. The residue was concentrated from chloroform (2 x 100 mL) and purified by automated flash chromatography (eluting with 20% CMA in chloroform) to provide 2.50 g of tert-buty\ 4-[(5-cyano-2-memyl-2i7-pyrazol-3-yl)rnethyl]-4- hydroxypiperidine-1-carboxylate as a sticky white solid, which was converted to tert-bxxtyl 4- [(4-bromo-5 -cyano-2-methyl-2H-pyrazol-3 -yl)methyl] -4-fluoropiperidine- 1 -carboxylate according to the methods of Parts H and I of Example 7. Part F
Hydrochloric acid (0.55 mL of 6 M) was added to a suspension of tert-butyl 4-[(4- bromo-5-cyano-2-methyl-2//-pyrazol-3-yl)methyl]-4-fluoropiperidine- 1 -carboxylate (446 mg, 1.11 mmol) in ethanol (10 mL), and the reaction was heated at 70 0C for 1.5 hours, allowed to cool to room temperature, and concentrated under reduced pressure to provide 4-bromo-5-[(4-fluoropiperidin-4-yl)methyl]-l-methyl-lH-pyrazole-3-carbonitrile hydrochloride as a white solid. Part G Triethylamine (0.62 mL, 4.0 eq.) was added to a stirred suspension of the material from Part F in dichloromethane (11 mL), and the mixture was cooled to 0 0C. Methanesulfonyl chloride (0.095 mL, 1.1 eq.) was added, and the resulting solution was stirred for 1.5 hours. Saturated aqueous sodium bicarbonate (20 mL) was added, and the mixture was allowed to warm to room temperature and diluted with dichloromethane (75 mL). The organic layer was separated, dried over magnesium sulfate, filtered, and concentrated under reduced pressure to provide 0.43 g of a white solid. The solid was triturated with hot ethanol (15 mL), allowed to cool to room temperature, isolated by filtration, washed with ethanol (2 x 5 mL), and dried under high vacuum to provide 351 mg of 4-bromo-5 - { [4-fluoro- 1 -(methylsulfonyl)piperidin-4-yl]methyl } - 1 -methyl- 1 H- pyrazole-3-carbonitrile as a white solid. Part H
The reaction conditions and purification methods described in Part J of Example 7 were used to treat the material from Part G. 1 - { [4-Fluoro- 1 -(methylsulfonyl)piperidin-4- yl]methyl}-2-methyl-2H-pyrazolo[3,4-c]quinolin-4-amine (71 mg) was obtained as a tan powder, mp 283-285 °C. 1H NMR (300 MHz, DMSOd6): δ 8.17 (d, J=7.8 Hz, IH), 7.47 (dd, J=I .2, 8.1 Hz, IH), 7.32 (m, IH), 7.17 (m, IH), 6.67 (br s, 2H), 4.11 (s, 3H), 3.76 (dd, J=22.8 Hz, 2H), 3.46 (m, 2H), 2.86 (s, 3H), 2.80 (m, 2H), 1.98-2.28 (m, 2H), 1.86 (m, 2H); MS (APCI) 392 m/z (M+H)+; Anal, calcd for C18H22FN5O2S: C, 55.23; H5 5.66; N, 17.89. Found: C, 54.99; H, 5.55; N5 17.70.
Example 21 l-[(l-Acetyl-4-fluoropiperidin-4-yl)methyl]-2-methyl-2H-pyrazolo[3,4-c]quinolin-4- amine
Figure imgf000083_0001
Part A
Triethylamine (3.4 mL, 3.0 eq.) was added to a stirred suspension of 4-bromo-5- [(4-fluoropiperidin-4-yl)methyl]-l -methyl- lH-pyrazole-3-carbonitrile hydrochloride (2.7 g, 8.0 mmol) in dichloromethane (80 mL), and the mixture was cooled to 0 0C. Acetyl chloride (0.74 mL, 1.3 eq.) was added, and the resulting solution was stirred for 1.5 hours and concentrated under reduced pressure. The residue was dissolved in dichloromethane (300 mL), and the resulting solution was washed with water (100 mL), dried over magnesium sulfate, filtered, and concentrated under reduced pressure to provide 2.58 g of a white foam. The foam was purified by automated flash chromatography (eluting with a gradient of 0-20% CMA in chloroform over 8 column volumes) to provide 0.94 g of 5-[(l- acetyl-4-fluoropiperidin-4-yl)methyl] -4-bromo- 1 -methyl- 1 /f-pyrazole-3 -carbonitrile, which was concentrated from 1 ,2-dimethoxyethane (75 mL) prior to the next step. Part B
The reaction conditions and purification methods described in Part J of Example 7 were used to treat the material from Part A. 1 -[(1 -Acetyl-4-fluoropiperidin-4-yl)methyl]- 2-methyl-2H~pyrazolo[3,4-c]quinolin-4-amine (176 mg) was obtained as a white powder, mp 241-243 °C. 1H NMR (300 MHz, DMSOd6): δ 8.14 (d, J=7.9 Hz, IH), 7.47 (dd, J=I.1, 8.1 Hz, IH), 7.32 (m, IH), 7.16 (m, IH), 6.66 (br s, 2H), 4.30 (br d, J=12.7 Hz,
IH)54.11 (s, 3H), 3.71 (d, J=22.4 Hz, 2H), 3.70 (m, IH), 3.12 (m, IH), 2.61 (m, IH), 2.00 (s, 3H), 1.74-2.14 (m, 4H); MS (ESI) 356 m/z (M+H)+; Anal, calcd for Ci9H22FN5O: C5 64.21; H, 6.24; N, 19.70. Found: C, 64.21; H, 6.04; N, 19.73.
Example 22
4-[(4-Amino-2-methyl-2H-pyrazolo[354-c]quinolin-l-yl)methyl]-4-fluoro-N- isopropylpiperidine- 1 -carboxamide
Figure imgf000084_0001
Part A
Triethylamine (1.5 mL, 1.5 eq.) was added to a stirred suspension of 4-bromo-5- [(4-fluoropiperidin-4-yl)methyl]-l -methyl- li?-pyrazole-3-carbonitrile hydrochloride (2.28 g, 6.75 mmol) in dichloromethane (67 mL), and isopropyl isocyanate (1.34 mL, 2.0 eq.) was added to the resulting solution. The reaction was stirred for 1.5 hours and concentrated under reduced pressure. The residue was dissolved in dichloromethane (300 mL), and the resulting solution was washed with water (100 mL), dried over magnesium sulfate, filtered, and concentrated under reduced pressure to provide 2,45 g of a white foam. The foam was purified by automated flash chromatography (eluting with a gradient of 0-10% CMA in chloroform over 8 column volumes) to provide 1.39 g of 4-[(4-bromo- 5-cyano-2-methyl-2H-pyrazol-3-yl)methyl]-4-fluoro-iV-isopropylpiperidine-l- carboxamide. Part B
The reaction conditions and purification methods described in Part J of Example 7 were used to treat the material from Part A. 4-[(4-Amino-2-methyl-2H-pyrazolo[3,4- c]quinolin-l-yl)methyl]-4-fluoro-Λ/"-isopropylpiperidine-l -carboxamide (0.14 g) was obtained as a white powder, mp 241-244 0C. 1H NMR (300 MHz5 DMSO-d6): δ 8.14 (d, J=7.4 Hz, IH), 7.47 (dd5 J=1.2, 8.1 Hz, IH), 7.32 (m, IH)5 7.16 (m, IH), 6.65 (br s, 2H), 6.17 (d, J=7.6 Hz5 IH), 4.11 (s, 3H), 3.67-3.89 (m, 5H), 2.73 (m, 2H)5 1.64-2.00 (m, 4H), 1.03 (d, J=6.6 Hz5 6H); MS (ESI) m/z 399 (M+H)+; Anal, calcd for C2iH27FN6O0.02 CHCl3: C, 62.98; H, 6.79; N, 20.96. Found: C5 62.59; H5 6.60; N5 20.69.
Example 23 2-Ethyl-l -{ [4-fluoro-l -(methylsulfonyl)piperidin-4-yl]methyl}-2/f-pyrazolo[3;>4- c]quinolin-4-amine
Figure imgf000085_0001
Part A
Starting with tert-bvXy\ 4-(2-oxopropylidene)piperidine-l-carboxylate (25.0 g, 105 mmol) in lieu of l-tetrahydro-4H-pyran-4-ylideneacetone and ethyl hydrazine oxalate
(69.0 g, 460 mmol) in lieu of methyl hydrazine, the general methods of Parts B through F of Example 7 were followed to provide tert-butyl 4-[(5-carbamoyl-2-ethyl-2H-pyrazol-3- yl)methyl]-4-hydroxypiperidine-l-carboxylate (3.25 g, 9.22 mmol), which was treated with triethylamine and trifluoroacetic anhydride according to the method of Part G of Example 7. The crude yellow oil that was obtained was dissolved in methanol (50 rnL), and concentrated ammonium hydroxide (1.84 mL) was added. The reaction was stirred for 1 hour and concentrated under reduced pressure. The residue was partitioned between water (100 mL) and chloroform (100 mL). The aqueous layer was separated and extracted with chloroform (2 x 100 mL). The combined organic fractions were dried over magnesium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by automated flash chromatography (eluting with chloroform for 4 column volumes and then a gradient of 0-20% CMA in chloroform over 6 column volumes) and dried under high vacuum to provide 2.56 g of tert-butyϊ 4-[(5-cyano-2-ethyl-2/7-pyrazol-3- yl)methyl]-4-hydroxypiperidine-l-carboxylate as a yellow oil, which was converted to tert-buty\ 4-[(4-bromo-5-cyano-2-ethyl-2/i-pyrazol-3-yl)methyl]-4-fluoropiperidine-l - carboxylate according to the methods of Parts Η and I of Example 7. Part B
Hydrochloric acid (2.64 mL of 6 M) was added to a suspension of tert-butyl 4-[(4- bromo-5-cyano-2-ethyl-2H-pyrazol-3-yl)methyl]-4-fluoropiperidine-l -carboxylate (3.3 g, 7.95 mmol) in ethanol (80 mL), and the reaction was heated at 80 0C for 2 hours, allowed to cool to room temperature, and concentrated under reduced pressure to provide 4-bromo- l-ethyl-5-[(4-fluoropiperidin-4-yl)methyl-lH-pyrazole-3-carbonitrile hydrochloride as a white solid. Part C
Triethylamine (11.1 mL, 79.5 mmol) and methanesulfonyl chloride (1.24 mL, 15.9 mmol) were sequentially added to a mixture of the material from Part B in dichloromethane (80 mL), and the reaction was stirred for 5 minutes. Brine (40 mL) was added, and the aqueous layer was separated and extracted with dichloromethane (2 x 100 mL). The combined organic fractions were dried over magnesium sulfate, filtered, and concentrated under reduced pressure to provide a brown foam. The foam was purified by automated flash chromatography (eluting with a gradient of 0- 10% CMA in chloroform over 8 column volumes) to provide 1.9 g of 4-bromo-l-ethyl-5-{[4-fluoro-l- (methylsulfonyl)piρeridin-4-yl]methyl}-lH-pyrazole-3-carbonitrile as a tan foam. Part D
The general reaction conditions and purification methods described in Part J of Example 7 were used to treat the material from Part C ( 1.0 g). 2-Ethyl- 1 - { [4-fluoro- 1 - (methylsulfonyl)piperidin-4-yl]methyl}-2H-pyrazolo[3,4-c]quinolin-4-amine (150 mg) was obtained as a light tan solid, mp 269-271 0C. 1HNMR (300 MHz, DMSO-d6): 8.16 (d, J=7.7 Hz, IH), 7.48 (dd, J=8.0, 1.1 Hz, IH)5 7.32 (td, J=7.5, 1.0 Hz, IH), 7.18 (td, J=7.5, 1.2 Hz, IH), 6.64 (s, 2H), 4.44 (q, J=7.2 Hz, 2H), 3.76 (d, J=22.9 Hz, 2H), 3.46 (d, J=I 1.2 Hz, 2H), 2.86 (s, 3H), 2.80 (t, J=12.3 Hz, 2H)5 2.03 (m, 4H), 1.49 (t, J=7.1 Hz,
3H)5 1.03 (d, J=6.6 Hz, 6H); MS (EI) m/z 406 (M+H)+; Anal, calcd for C19H24FN5O2S: C, 56.28; H, 5.97; N, 17.27. Found: C, 55.99; H, 5.71; N, 17.05.
Example 24 l-[(l-Acetyl-4-fluoropiperidin-4-yl)methyl]-2-ethyl-2H"-pyrazolo[334-c]quinolin-4-amine
Figure imgf000087_0001
Part A
Triethylamine (3.35 niL, 24.0 mmol) was added to a stirred suspension of 4- bromo-l-ethyl-5-[(4-fluoropiperidin-4-yl)methyl-lH-pyrazole-3-carbonitrile hydrochloride (2.52 g, 8.0 mmol) in dichloromethane (80 niL), and the resulting solution was cooled to 0 0C. Acetyl chloride (0.74 mL, 10.4 mmol) was added dropwise, and the resulting solution was stirred for 1 hour. Water (50 mL) was added, and then the aqueous layer was separated and extracted with dichloromethane (2 x 100 mL). The combined organic fractions were dried over magnesium sulfate, filtered, and concentrated under reduced pressure to provide a brown foam. The foam was purified by automated flash chromatography (eluting with a gradient of 0-10% CMA in chloroform over 8 column volumes) and dried under high vacuum to provide 1.6 g of 5-[(l-acetyl-4-fluoropiperidin- 4-yl)methyl]-4-bromo~ 1 -ethyl- 1 H-pyrazole-3 -carbonitrile. Part B The reaction conditions described in Part J of Example 7 were used to treat the material from Part A. Following chromatographic purification (eluting with chloroform for 4 column volumes, followed by a gradient of 0-40% CMA in chloroform over 6 column volumes, followed by 40% CMA in chloroform) the resulting oil was recrystallized from acetonitrile. The solid was isolated by filtration, washed with acetonitrile, and dried under high vacuum to provide 121 mg of l-[(l-acetyl-4- fluoropiperidin-4-yl)methyl]-2-ethyl-2iϊ-pyrazolo[354-c]quinolin-4-amine as a light tan solid, mp 228-230 °C. 1H NMR (300 MHz, DMSO-d6): 8.14 (d, J=7.8 Hz, IH), 7.48 (dd, J=8.1, 1.2 Hz, IH), 7.31 (td3 J=7.5, 1.1 Hz, IH)5 7.16 (td, J=7.4, 1.2 Hz, IH), 6.64 (s, 2H), 4.44 (q5 J=7.2 Hz, 2H), 4.30 (d, J=12.2 Hz5 IH)5 3.71 (m, 3H), 3.12 (t, J=12.4 Hz, IH), 2.60 (t, J=12.8 Hz, IH), 2.00 (s, 3H), 1.93 (m, 4H), 1.48 (t, J=7.1 Hz3 3H); MS (EI) m/z 370 (M+H)+; Anal, calcd for C20H24FN5O: C, 65.02; H5 6.55; N5 18.96. Found: C, 65.07; H5 6.32; N, 19.12. Example 25
4-[(4-Amino-2-ethyl-2/i-pyrazolo[3,4-c]quinolin-l-yl)methyl]-4-fluoro-N- isopropylpiperidine-1 -carboxamide
Figure imgf000088_0001
Part A
Triethylamine (2.23 mL, 16.0 mmol) and isopropyl isocyanate (1.01 niL, 10.4 mmol) were sequentially added to a mixture of 4-bromo-l-ethyl-5-[(4-fluoropiperidin-4- yl)methyl-l/7-pyrazole-3-carbonitrile hydrochloride (2.52 g, 8.0 mmol) and dichloromethane (40 mL). The reaction was stirred for 1.5 hours and diluted with dichloromethane (250 mL), washed with water (WO mL), dried over magnesium sulfate, filtered, and concentrated under reduced pressure to provide a brown foam. The foam was purified by automated flash chromatography (eluting with a gradient of 0-10% CMA in chloroform over 8 column volumes). The resulting off-white foam was concentrated from 1,2-dimethoxyethane (50 mL) and dried under high vacuum to provide 1.6 g of 4-[(4- bromo-5-cyano-2-ethyl-2iϊ-pyrazol-3-yl)methyl]-4-fluoro-N-isopropylpiperidine-l- carboxamide. Part B
The reaction conditions described in Part J of Example 7 were used to treat the material from Part A. Following chromatographic purification using the conditions described in Part B of Example 24, the resulting brown solid was recrystallized from ethanol and dried to provide 93 mg of 4-[(4-amino-2-ethyl-2if-pyrazolo[3,4-c]quinolin-l- yl)methyl]-4-fluoro-7V-isopropylpiperidine-l -carboxamide as a white solid, mp 205-207 °C. 1H NMR (300 MHz, DMSOd6): 8.14 (d, J=7.8 Hz, IH), 7.48 (dd, J=8.1, 1.1 Hz, IH)5 7.31 (td, J=7.5, 1.1 Hz, IH), 7.16 (td, J=7.4, 1.2 Hz, IH), 6.63 (s, 2H), 6.17 (d, J=7.6 Hz, IH), 4.44 (q, J=7.2 Hz5 2H), 3.78 (m, 5H), 2.73 (t, J=12.1 Hz, 2H)5 1.81 (m, 4H), 1.47 (t, J=7.1 Hz, 3H)5 1.03 (6, J=6.6 Hz, 6H); MS (EI) m/z 413 (M+H)+; Anal, calcd for C22H29FN6O: C, 64.06; H, 7.09; N, 20.37. Found: C, 63.93; H, 7.17; N, 20.30. Example 26
1 - { [4-Fluoro- 1 -(methylsulfonyl)piperidin-4-yl]niethyl } -2-propyl-2//-pyrazolo [3 ,4- c]quinolin-4-amine
Figure imgf000089_0001
Part A
Starting with tert-butyl 4-(2-oxopropylidene)piρeridine-l-carboxylate (20.0 g, 83.6 mmol) in lieu of l-tetrahydro-4H-pyran-4-ylideneacetone and propyl hydrazine oxalate (34.3 g, 2.5 eq.) in lieu of methyl hydrazine, the general methods of Parts B through F of Example 7 were followed to provide tert-butyl 4-[(5-carbamoyl-2-propyl-2H-pyrazol-3~ yl)methyl]-4-hydroxypiperidine-l-carboxylate (6.20 g, 16.9 mmol), which was treated with triethylamine and trifluoroacetic anhydride according to the method of Part G of Example 7. The crude yellow oil that was obtained was dissolved in methanol (45 mL), and concentrated ammonium hydroxide (3.4 mL) was added. The reaction was stirred for 1 hour and concentrated under reduced pressure. The residue was partitioned between water (150 mL) and chloroform (200 mL). The organic layer was separated and dried over magnesium sulfate, filtered, and concentrated under reduced pressure. The residue was purified twice by automated flash chromatography (eluting first with a gradient of 0- 40% CMA in chloroform over 5 column volumes and second with a gradient of 0-50% ethyl acetate in chloroform over 5 column volumes) to provide 4.52 g of tert-butyl 4-[(5- cyano-2-propyl-2//-ρyrazoly-3-yl)methyl]-4-hydroxypiperidine-l-carboxylate as a viscous semisolid, which was converted to tert-butyl 4-[(4-bromo-5-cyano-2-propyl-2H-pyrazol-3- yl)methyl]-4-fluoropiperidine-l-carboxylate according to the methods of Parts H and I of Example 7. Part B
Hydrochloric acid (3.9 mL of 6 M) was added to a solution of tert-butyl 4-[(4- bromo-5-cyano-2-propyl-27f-pyrazol-3-yl)methyl]-4-fluoropiρeridine-l -carboxylate (3.34 g, 7.78 mmol) in ethanol (40 mL), and the reaction was heated at 70 0C for 1.5 hours, allowed to cool to room temperature, and concentrated under reduced pressure to provide 2.9 g of4-bromo-5-[(4-fluoropiperidin-4-yl)methyl]-l-propyl-l//-pyrazole-3-carbonitrile hydrochloride. The salt was suspended in dichloromethane (40 mL), and triethylamine (4.9 mL, 4.5 eq.) was added. The mixture was stirred for ten minutes to provide a 0.2 M solution of 4-bromo-5-[(4-fluoropiperidin-4-yl)methyl]-l -propyl- lH-pyrazole-3- carbonitrile. Part C
A portion of the solution from Part B (24 mL) was cooled to 0 0C. Methanesulfonyl chloride (0.42 mL, 1.3 eq.) was added dropwise, and the resulting solution was stirred for 1 hour, diluted with dichloromethane (150 mL), and allowed to warm to room temperature. The solution was washed with saturated aqueous sodium bicarbonate (100 mL), dried over magnesium sulfate, filtered, and concentrated under reduced pressure to provide a white foam. The foam was purified by automated flash chromatography (eluting with a gradient of 0-15% CMA in chloroform) to provide 1.5 g of 4-bromo-5- { [4-fluoro- 1 -(methylsulfonyl)ρiperidin-4-yl]methyl} - 1 -propyl- lH-pyrazole- 3-carbonitrile as a white foam. The foam was concentrated from 1,2-dimethoxyethane (40 mL) before it was used in Part D. Part D The reaction conditions and purification methods described in Part J of Example 7 were used to treat the material from Part C. 1 - { [4-Fluoro- 1 -(methylsulfonyl)ρiperidin-4- yl]methyl}-2-ρropyl-2H-pyrazolo[3,4-c]quinolin-4-amine (0.32 g) was obtained as a white powder, mp 268-270 °C, dec. 1H NMR (300 MHz, DMSO-d6): 5 8.16 (d, J=7.9 Hz, IH), 7.48 (dd, J=1.2, 8.1 Hz, IH), 7.32 (m, IH), 7.17 (m, IH), 6.64 (br s, 2H), 4.36 (t, J=7.2 Hz, 2H), 3.76 (d, J=22.3 Hz, 2H), 3.46 (m, 2H), 2.85 (s, 3H), 2.80 (m, 2H), 1.80 (m, 6H), 0.90 (t, J=7.5 Hz, 3H); MS (ESI) m/z 420 (M+H)+; Anal, calcd for C20H26FN5O2S: C, 57.26; H5 6.25; N, 16.69. Found: C, 57.03; H, 6.33; N, 16.71.
Example 27 4-[(4-Amino-2-propyl-2/i-pyrazolo[3,4-c]quinolin-l-yl)methyl]-4-fluoro-iV- isopropylpiperidine- 1 -carboxamide
Figure imgf000091_0001
Part A
Isopropyl isocyanate (0.46 niL, 1.3 eq.) was added dropwise to a portion of the solution from Part B of Example 26 (21 mL) at room temperature. The resulting solution was stirred for 1 hour, diluted with dichloromethane (100 mL), washed with water (70 mL), dried over magnesium sulfate, filtered, and concentrated under reduced pressure to provide 1.48 g of a tan foam. The foam was purified by automated flash chromatography (eluting with a gradient of 0-10% CMA in chloroform over 8 column volumes) to provide 1.05 g of 4-(4-bromo-5-cyano-2-propyl-2#-pyrazol~3-ylmemyl)-4-fluoro-iV- isopropylpiperidine- 1 -carboxamide as a white foam. Part B
The reaction conditions and purification methods described in Part J of Example 7 were used to treat the material from Part A. 4-[(4-Amino-2-propyl-2H-pyrazolo[3,4- c]quinolin-l-yl)methyl]-4-fluoro-A/-isopropylpiperidine-l -carboxamide (105 mg) was obtained as a white powder, mp 227-229 0C, dec. 1H NMR (300 MHz, DMSOd6): δ 8.13 (d, J=7.6 Hz5 IH), 7.47 (dd, J=Ll, 8.1 Hz, IH), 7.31 (m, IH), 7.16 (m, IH), 6.63 (br s, 2H), 6.17 (d, J=7.6 Hz, IH), 4.36 (t, J=7.6 Hz, 2H), 3.68-3.89 (m, 5H), 2.72 (t, J=I 1.9 Hz, 2H), 1.62-1.98 (m, 6H), 1.03 (d, J=6.6 Hz, 6H), 0.09 (t, J=7.6 Hz, 3H); MS (ESI) m/z 427 (M+H)+; Anal, calcd for C23H3)FN6O: C, 64.77; H, 7.33; N, 19.70. Found: C, 64.67; H, 7.23; N, 19.65.
Exemplary Compounds
Certain exemplary compounds, including some of those described above in the Examples, have the following Formulas (IH-I5 IV-I, V-I, or VIII-I) and the following R2, Z, Rj, and m substituents or variables, wherein each line of the table is matched with
Formula III- 1 , IV-I, V-I5 or VIII-I to represent a specific embodiment of the invention.
Figure imgf000092_0001
Figure imgf000092_0002
Figure imgf000093_0001
Figure imgf000094_0001
Figure imgf000095_0001
Figure imgf000096_0002
Certain exemplary compounds, including some of those described above in the Examples, have the following Formulas (III-2. IV-2, V-2, and VIII-2) wherein R2, Q, R4, R1, and m are defined immediately below in the table. Each row of the table is matched . with Formula III-2, IV-2, V-2, or VIII-2 to represent a specific embodiment of the invention.
Figure imgf000096_0001
III-2 IV-2 V-2 VIII-2
Figure imgf000096_0003
Figure imgf000097_0001
Figure imgf000098_0001
Figure imgf000099_0001
Figure imgf000100_0001
Figure imgf000101_0001
Figure imgf000102_0001
Figure imgf000103_0001
Figure imgf000104_0001
Figure imgf000105_0002
Certain exemplary compounds, including some of those described above in the Examples, have the following Formulas (III-3, IV-3, V-3, and VIII-3) wherein R2, Q, Z, R4, and m are defined immediately below in the table. Each row of the table is matched with Formula III-3, IV-3, V-3, or VIII-3 to represent a specific embodiment of the invention.
Figure imgf000105_0001
iπ-3 IV-3 V-3
Figure imgf000106_0001
Figure imgf000106_0002
Figure imgf000107_0001
Figure imgf000108_0001
Figure imgf000109_0001
Figure imgf000110_0001
Figure imgf000111_0001
Figure imgf000112_0001
Certain exemplary compounds, including some of those described above in the Examples, have the following Formulas (III-4. IV-4, V-4, and VIII-4) wherein R2 and m are defined immediately below in the table. Each row of the table is matched with Formula III-4, IV-4, V-4, or VIII-4 to represent a specific embodiment of the invention.
I l l
Figure imgf000113_0001
Figure imgf000113_0002
Compounds of the invention have been found to modulate cytokine biosynthesis by inducing the production of interferon α and/or tumor necrosis factor α in human cells when tested using one of the methods described below.
CYTOKINE INDUCTION IN HUMAN CELLS
An in vitro human blood cell system is used to assess cytokine induction. Activity is based on the measurement of interferon (α) and tumor necrosis factor (α) (IFN-α and TNF-α, respectively) secreted into culture media as described by Testerman et al. in "Cytokine Induction by the Immunomodulators Imiquimod and S-27609," Journal of Leukocyte Biology, 58, 365-372 (September, 1995). Blood Cell Preparation for Culture
Whole blood from healthy human donors is collected by venipuncture into vacutainer tubes or syringes containing EDTA. Peripheral blood mononuclear cells (PBMC) are separated from whole blood by density gradient centrifugation using HISTOPAQUE- 1077 (Sigma, St. Louis, MO) or Ficoll-Paque Plus (Amersham
Biosciences Piscataway, NJ). Blood is diluted 1:1 with Dulbecco's Phosphate Buffered Saline (DPBS) or Hank's Balanced Salts Solution (HBSS). Alternately, whole blood is placed in Accuspin (Sigma) or LeucoSep (Greiner Bio-One, Inc., Longwood, FL) centrifuge frit tubes containing density gradient medium. The PBMC layer is collected and washed twice with DPBS or HBSS and re-suspended at 4 x 106 cells/mL in RPMI complete. The PBMC suspension is added to 96 well flat bottom sterile tissue culture plates containing an equal volume of RPMI complete media containing test compound.
Compound Preparation The compounds are solubilized in dimethyl sulfoxide (DMSO). The DMSO concentration should not exceed a final concentration of 1% for addition to the culture wells. The compounds are generally tested at concentrations ranging from 30-0.014 μM.
Controls include cell samples with media only, cell samples with DMSO only (no compound), and cell samples with reference compound.
Incubation
The solution of test compound is added at 60 μM to the first well containing RPMI complete and serial 3 fold dilutions are made in the wells. The PBMC suspension is then added to the wells in an equal volume, bringing the test compound concentrations to the desired range (usually 30-0.014 μM). The final concentration of PBMC suspension is 2 x
106 cells/mL. The plates are covered with sterile plastic lids, mixed gently and then incubated for 18 hours to 24 hours at 37°C in a 5% carbon dioxide atmosphere.
Separation Following incubation the plates are centrifuged for 10 minutes at 1000 rpm
(approximately 200 x g) at 4°C. The cell-free culture supernatant is removed and transferred to sterile polypropylene tubes. Samples are maintained at -30°C to -70°C until analysis. The samples are analyzed for IFN-α by ELISA and for TNF-α by IGEN/BioVeris Assay.
Interferon (α) and Tumor Necrosis Factor (α) Analysis IFN-α concentration is determined with a human multi-subtype colorimetric sandwich ELISA (Catalog Number 41105) from PBL Biomedical Laboratories, Piscataway, NJ. Results are expressed in pg/mL.
The TNF-α concentration is determined by ORIGEN M-Series Immunoassay and read on an IGEN M-8 analyzer from BioVeris Corporation, formerly known as IGEN International, Gaithersburg, MD. The immunoassay uses a human TNF-α capture and detection antibody pair (Catalog Numbers AHC3419 and AHC3712) from Biosource International, Camarillo, CA. Results are expressed in pg/mL.
Assay Data and Analysis In total, the data output of the assay consists of concentration values of TNF-α and
IFN-α (y-axis) as a function of compound concentration (x-axis).
Analysis of the data has two steps. First, the greater of the mean DMSO (DMSO control wells) or the experimental background (usually 20 pg/mL for IFN-α and 40 pg/mL for TNF-α) is subtracted from each reading. If any negative values result from background subtraction, the reading is reported as " * ", and is noted as not reliably detectable. In subsequent calculations and statistics, " * ", is treated as a zero. Second, all background subtracted values are multiplied by a single adjustment ratio to decrease experiment to experiment variability. The adjustment ratio is the area of the reference compound in the new experiment divided by the expected area of the reference compound based on the past 61 experiments (unadjusted readings). This results in the scaling of the reading (y-axis) for the new data without changing the shape of the dose-response curve. The reference compound used is 2-[4-amino-2-ethoxymethyl-6,7,8,9-tetrahydro-α,α- dimethyl-lH-imidazo[4,5-c]qumolin-l-yl]ethanol hydrate (U.S. Patent No. 5,352,784; Example 91) and the expected area is the sum of the median dose values from the past 61 experiments.
The minimum effective concentration is calculated based on the background- subtracted, reference-adjusted results for a given experiment and compound. The minimum effective concentration (μmolar) is the lowest of the tested compound concentrations that induces a response over a fixed cytokine concentration for the tested cytokine (usually 20 pg/mL for IFN-α and 40 pg/mL for TNF-α). The maximal response is the maximal amount of cytokine (pg/ml) produced in the dose-response.
CYTOKINE INDUCTION IN HUMAN CELLS
(High Throughput Screen)
The CYTOKINE INDUCTION IN HUMAN CELLS test method described above was modified as follows for high throughput screening.
Blood Cell Preparation for Culture
Whole blood from healthy human donors is collected by venipuncture into vacutainer tubes or syringes containing EDTA. Peripheral blood mononuclear cells (PBMC) are separated from whole blood by density gradient centrifugation using HISTOPAQUE- 1077 (Sigma, St. Louis, MO) or Ficoll-Paque Plus (Amersham
Biosciences Piscataway, NJ). Whole blood is placed in Accuspin (Sigma) or LeucoSep (Greiner Bio-One, Inc., Longwood, FL) centrifuge frit tubes containing density gradient medium. The PBMC layer is collected and washed twice with DPBS or HBSS and re- suspended at 4 x 106 cells/mL in RPMI complete (2-fold the final cell density). The PBMC suspension is added to 96-well flat bottom sterile tissue culture plates.
Compound Preparation
The compounds are solubilized in dimethyl sulfoxide (DMSO). The compounds are generally tested at concentrations ranging from 30 - 0.014 μM. Controls include cell samples with media only, cell samples with DMSO only (no compound), and cell samples with a reference compound 2-[4~amino-2-ethoxymethyl-6,7,8,9-tetrahydro-α,α-dimethyl- lH-imidazo[4,5-c]quinolin-l-yl]ethanol hydrate (U.S. Patent No. 5,352,784; Example 91) on each plate. The solution of test compound is added at 7.5 mM to the first well of a dosing plate and serial 3 fold dilutions are made for the 7 subsequent concentrations in DMSO. RPMI Complete media is then added to the test compound dilutions in order to reach a final compound concentration of 2-fold higher (60 - 0.028 μM) than the final tested concentration range. Incubation
Compound solution is then added to the wells containing the PBMC suspension bringing the test compound concentrations to the desired range (usually 30 μM - 0.014 μM) and the DMSO concentration to 0.4%. The final concentration of PBMC suspension is 2x106 cells/mL. The plates are covered with sterile plastic lids, mixed gently and then incubated for 18 to 24 hours at 37°C in a 5% carbon dioxide atmosphere.
Separation Following incubation the plates are centrifuged for 10 minutes at 1000 rpm
(approximately 200 g) at 40C. 4-plex Human Panel MSD MULTI-SPOT 96-well plates are pre-coated with the appropriate capture antibodies by MesoScale Discovery, Inc. (MSD, Gaithersburg, MD). The cell-free culture supernatants are removed and transferred to the MSD plates. Fresh samples are typically tested, although they may be maintained at -30°C to -700C until analysis.
Interferon-α and Tumor Necrosis Factor-α Analysis
MSD MULTI-SPOT plates contain within each well capture antibodies for human TNF-α and human IFN-α that have been pre-coated on specific spots. Each well contains four spots: one human TNF-α capture antibody (MSD) spot, one human IFN- α capture antibody (PBL Biomedical Laboratories, Piscataway, NJ) spot, and two inactive bovine serum albumin spots. The human TNF-α capture and detection antibody pair is from MesoScale Discovery. The human IFN-α multi-subtype antibody (PBL Biomedical Laboratories) captures all IFN-α subtypes except IFN-α F (IFNA21). Standards consist of recombinant human TNF-α (R&D Systems, Minneapolis, MN) and IFN-α (PBL
Biomedical Laboratories). Samples and separate standards are added at the time of analysis to each MSD plate. Two human IFN-α detection antibodies (Cat. Nos. 21112 & 21100, PBL) are used in a two to one ratio (weight: weight) to each other to determine the IFN-α concentrations. The cytokine-specific detection antibodies are labeled with the SULFO-TAG reagent (MSD), After adding the SULFO-TAG labeled detection antibodies to the wells, each well's electrochemoluminescent levels are read using MSD's SECTOR HTS READER. Results are expressed in pg/mL upon calculation with known cytokine standards.
Assay Data and Analysis In total, the data output of the assay consists of concentration values of TNF-α or
IFN-α (y-axis) as a function of compound concentration (x-axis).
A plate-wise scaling is performed within a given experiment aimed at reducing plate-to-plate variability associated within the same experiment. First, the greater of the median DMSO (DMSO control wells) or the experimental background (usually 20 pg/mL for IFN-α and 40 pg/mL for TNF-α) is subtracted from each reading. Negative values that may result from background subtraction are set to zero. Each plate within a given experiment has a reference compound that serves as a control. This control is used to calculate a median expected area under the curve across all plates in the assay. A plate- wise scaling factor is calculated for each plate as a ratio of the area of the reference compound on the particular plate to the median expected area for the entire experiment. The data from each plate are then multiplied by the plate-wise scaling factor for all plates. Only data from plates bearing a scaling factor of between 0.5 and 2.0 (for both cytokines IFN-α, TNF-α) are reported. Data from plates with scaling factors outside the above- mentioned interval are retested until they bear scaling factors inside the above mentioned interval. The above method produces a scaling of the y- values without altering the shape of the curve. The reference compound used is 2-[4-amino-2-ethoxymethyl-6,7,8,9- tetrahydro-α,α-dimethyl-lH-imidazo[4,5-c]quinolin-l-yl]ethanol hydrate (U.S. Patent No. 5,352,784; Example 91). The median expected area is the median area across all plates that are part of a given experiment. A second scaling may also be performed to reduce inter-experiment variability
(across multiple experiments). All background-subtracted values are multiplied by a single adjustment ratio to decrease experiment-to-experiment variability. The adjustment ratio is the area of the reference compound in the new experiment divided by the expected area of the reference compound based on an average of previous experiments (unadjusted readings). This results in the scaling of the reading (y-axis) for the new data without changing the shape of the dose-response curve. The reference compound used is 2-[4- amino-2-ethoxymethyl-6,7,8,9-tetrahydro-a,a-dimethyl-lH-imidazo[4,5-c]quinolin-l- yljethanol hydrate (U.S. Patent No. 5,352,784; Example 91) and the expected area is the sum of the median dose values from an average of previous experiments.
The minimum effective concentration is calculated based on the background- subtracted, reference-adjusted results for a given experiment and compound. The minimum effective concentration (μmolar) is the lowest of the tested compound concentrations that induces a response over a fixed cytokine concentration for the tested cytokine (usually 20 pg/mL for IFN-α and 40 pg/mL for TNF-α). The maximal response is the maximal amount of cytokine (pg/ml) produced in the dose-response.
The complete disclosures of the patents, patent documents, and publications cited herein are incorporated by reference in their entirety as if each were individually incorporated. Various modifications and alterations to this invention will become apparent to those skilled in the art without departing from the scope and spirit of this invention. It should be understood that this invention is not intended to be unduly limited by the illustrative embodiments and examples set forth herein and that such examples and embodiments are presented by way of example only with the scope of the invention intended to be limited only by the claims set forth herein as follows.

Claims

WHAT IS CLAIMED IS:
1. A compound of the Formula I :
Figure imgf000120_0001
I wherein:
Z is selected from the group consisting of:
Figure imgf000120_0002
Figure imgf000121_0001
X is selected from the group consisting of a bond, -C2-3 alkylene-, and -0-C2-3 alkylene-;
Ri is selected from the group consisting of: hydrogen, hydroxy, fluorine, alkoxy,
Figure imgf000121_0002
with the proviso that when Z is a bond, C1-5 alkylene,
, _. . , ,
Figure imgf000121_0003
, and X is a bond, then R1 is other than hydrogen; m is an integer from 1 to 5;
RA and RB are each independently selected from the group consisting of: hydrogen, halogen, alkyl, alkenyl, alkoxy, alkylthio, and -N(Rp)2; or when taken together, RA and RB form a fused aryl ring or heteroaryl ring containing one heteroatom selected from the group consisting of N and S wherein the aryl or heteroaryl ring is unsubstituted or substituted by one or more R groups; or when taken together, RA and RB form a fused 5 to 7 membered saturated ring, optionally containing one heteroatom selected from the group consisting of N and S, and unsubstituted or substituted by one or more R groups;
R is selected from the group consisting of: halogen, hydroxy, alkyl, haloalkyl, alkoxy, and -N(Re)2;
R2 is selected from the group consisting of: hydrogen, alkyl, alkoxyalkylenyl, hydroxyalkylenyl, haloalkylenyl, and R4-C(Re)-O-Ci-4 alkylenyl;
R4 is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, aryl, arylalkylenyl, aryloxyalkylenyl, alkylarylenyl, heteroaryl, heteroarylalkylenyl, heteroaryloxyalkylenyl, alkylheteroarylenyl, and heterocyclyl, wherein the alkyl, alkenyl, alkynyl, aryl, arylalkylenyl, aryloxyalkylenyl, alkylarylenyl, heteroaryl, heteroarylalkylenyl, heteroaryloxyalkylenyl, alkylheteroarylenyl, and heterocyclyl groups can be unsubstituted or substituted by one or more substiruents independently selected from the group consisting of alkyl, alkoxy, hydroxyalkyl, haloalkyl, haloalkoxy, halogen, nitro, hydroxy, mercapto, cyano, aryl, aryloxy, arylalkyleneoxy, heteroaryl, heteroaryloxy, heteroarylalkyleneoxy, heterocyclyl, amino, acetylamino, alkylamino, dialkylamino, (dialkylamino)alkyleneoxy, and in the case of alkyl, alkenyl, alkynyl, and heterocyclyl, oxo;
R6 is selected from the group consisting of =0 and =S;
Rs is selected from the group consisting of hydrogen, alkyl, alkoxyalkylenyl, and arylalkylenyl;
R-9 is selected from the group consisting of hydrogen and alkyl;
Rn is selected from the group consisting of hydrogen, alkyl, halogen, and trifluoroniethyl;
A is selected from the group consisting of -O-, -C(O)-, -S(O)0-2-, -CH2-, and -N(R4)-;
Q is selected from the group consisting of a bond, -C(R6)-, -C(R6)-C(R6)-, -S(O)2-, -C(R6)-N(R8)-W-, -S(O)2-N(R8)-, -C(Re)-O-, and -C(Re)-N(OR9)-;
V is selected from the group consisting Of-C(R6)-, -0-C(R6)-, -N(Rg)-C(R6)-, and -S(O)2-; W is selected from the group consisting of a bond, -C(O)-, and -S(O)2-; and a and b are independently integers from 1 to 6 with the proviso that a + b is < 7; or a pharmaceutically acceptable salt thereof.
2. A compound of the Formula II:
Figure imgf000123_0001
II wherein:
Z is selected from the group consisting of:
Figure imgf000123_0002
Figure imgf000124_0001
X is selected from the group consisting of a bond, -C2-3 alkylene-,
-O-C2-3 alkylene-;
Ri is selected from the group consisting of: hydrogen, hydroxy, fluorine, alkoxy,
Figure imgf000124_0002
Figure imgf000125_0001
with the proviso that when Z is a bond, Q-5 alkylene,
, v . , ,
Figure imgf000125_0002
, and X is a bond, then Ri is other than hydrogen; m is an integer from 1 to 5; , RA1 and RB 1 are each independently selected from the group consisting of: hydrogen, halogen, alkyl, alkenyl, alkoxy, alkylthio, and -N(R9)2; R2 is selected from the group consisting of: hydrogen, alkyl, alkoxyalkylenyl, hydroxyalkylenyl, haloalkylenyl, and R4-C(Re)-O-C1-4 alkylenyl; R4 is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, aryl, arylalkylenyl, aryloxyalkylenyl, alkylarylenyl, heteroaryl, heteroarylalkylenyl, heteroaryloxyalkylenyl, alkylheteroarylenyl, and heterocyclyl, wherein the alkyl, alkenyl, alkynyl, aryl, arylalkylenyl, aryloxyalkylenyl, alkylarylenyl, heteroaryl, heteroarylalkylenyl, heteroaryloxyalkylenyl, alkylheteroarylenyl, and heterocyclyl groups can be unsubstituted or substituted by one or more substituents independently selected from the group consisting of alkyl, alkoxy, hydroxyalkyl, haloalkyl, haloalkoxy, halogen, nitro, hydroxy, mercapto, cyano, aryl, aryloxy, arylalkyleneoxy, heteroaryl, heteroaryloxy, heteroarylalkyleneoxy, heterocyclyl, amino, acetylamino, alkylamino, dialkylamino, (dialkylamino)alkyleneoxy, and in the case of alkyl, alkenyl, alkynyl, and heterocyclyl, oxo;
R6 is selected from the group consisting of =0 and =S;
R-8 is selected from the group consisting of hydrogen, alkyl, alkoxyalkylenyl, and arylalkylenyl; Rg is selected from the group consisting of hydrogen and alkyl;
Rn is selected from the group consisting of hydrogen, alkyl, halogen, and trifluoromethyl;
A is selected from the group consisting of -O-, -C(O)-, -S(0)o-2-, -CH2-, and -N(R4)-; Q is selected from the group consisting of a bond, -C(R6)-, -C(R6)-C(R6)-, -S(O)2-,
-C(R6)-N(R8)-W-, -S(O)2-N(R8)-, -C(Re)-O-, and -C(Re)-N(OR9)-;
V is selected from the group consisting of -C(R6)-, -0-C(R6)-, -N(Rs)-C(R6)-, and -S(O)2-;
W is selected from the group consisting of a bond, -C(O)-, and -S(O)2-; and a and b are independently integers from 1 to 6 with the proviso that a + b is < 7; or a pharmaceutically acceptable salt thereof.
3. A compound of the Formula III:
Figure imgf000126_0001
wherein:
Z is selected from the group consisting of:
Figure imgf000127_0001
X is selected from the group consisting of a bond, -C2-3 alkylene-, -0-C2-3 alkylene-;
R1 is selected from the group consisting of: hydrogen, hydroxy, fluorine, alkoxy, -N(R9)2, -NH-Q-R4,
Figure imgf000128_0001
with the proviso that when Z is a bond, C 1.5 alkylene,
; and x is a bond5
Figure imgf000128_0002
then Ri is other than hydrogen; m is an integer from 1 to 5; R is selected from the group consisting of: halogen, hydroxy, alkyl, haloalkyl, alkoxy, and -N(R9)2; n is an integer from 0 to 4; R2 is selected from the group consisting of: hydrogen, alkyl, alkoxyalkylenyl, hydroxyalkylenyl, haloalkylenyl, and
R4-C(Rs)-O-C1-4 alkylenyl;
R4 is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, aryl, arylalkylenyl, aryloxyalkylenyl, alkylarylenyl, heteroaryl, heteroarylalkylenyl, heteroaryloxyalkylenyl, alkylheteroarylenyl, and heterocyclyl, wherein the alkyl, alkenyl, alkynyl, aryl, arylalkylenyl, aryloxyalkylenyl, alkylarylenyl, heteroaryl, heteroarylalkylenyl, heteroaryloxyalkylenyl, alkylheteroarylenyl, and heterocyclyl groups can be unsubstituted or substituted by one or more substituents independently selected from the group consisting of alkyl, alkoxy, hydroxyalkyl, haloalkyl, haloalkoxy, halogen, nitro, hydroxy, mercapto, cyano, aryl, aryloxy, arylalkyleneoxy, heteroaryl, heteroaryloxy, heteroarylalkyleneoxy, heterocyclyl, amino, acetylamino, alkylamino, dialkylamino, (dialkylamino)alkyleneoxy, and in the case of alkyl, alkenyl, alkynyl, and heterocyclyl, oxo;
R6 is selected from the group consisting of =0 and =S; Rg is selected from the group consisting of hydrogen, alkyl, alkoxyalkylenyl, and arylalkylenyl;
Rg is selected from the group consisting of hydrogen and alkyl;
R11 is selected from the group consisting of hydrogen, alkyl, halogen, and trifluoromethyl; A is selected from the group consisting of -O-, -C(O)-, -S(0)o-2-, -CH2-, and
-N(R4)-;
Q is selected from the group consisting of a bond, -C(R6)-, -C(R6)-C(R6)-, -S(O)2-, -C(Re)-N(Re)-W-, -S(O)2-N(R8)-, -C(Re)-O-, and -C(R6J-N(OR9)-;
V is selected from the group consisting of -C(R6)-, -0-C(R6)-, -N(Rs)-C(R6)-, and -S(O)2-;
W is selected from the group consisting of a bond, -C(O)-, and -S(O)2-; and a and b are independently integers from 1 to 6 with the proviso that a + b is < 7; or a pharmaceutically acceptable salt thereof.
4. A compound of the Formula IV:
Figure imgf000129_0001
IV wherein:
Z is selected from the group consisting of:
Figure imgf000130_0001
X is selected from the group consisting of a bond, -C2-3 alkylene-, -O-C2-3 alkylene-; Ri is selected from the group consisting of: hydrogen, hydroxy, fluorine, alkoxy,
Figure imgf000131_0001
with the proviso that when Z is a bond, Cj-5 alkylene,
s and χ .s a bondj
Figure imgf000131_0002
then Ri is other than hydrogen; m is an integer from 1 to 5; R is selected from the group consisting of: halogen, hydroxy, alkyl, haloalkyl, alkoxy, and -N(Re)2; n is an integer from 0 to 4; R2 is selected from the group consisting of: hydrogen, alkyl, alkoxyalkylenyl, hydroxyalkylenyl, haloalkylenyl, and
Figure imgf000131_0003
R4 is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, aryl, arylalkylenyl, aryloxyalkylenyl, alkylarylenyl, heteroaryl, heteroarylalkylenyl, heteroaryloxyalkylenyl, alkylheteroarylenyl, and heterocyclyl, wherein the alkyl, alkenyl, alkynyl, aryl, arylalkylenyl, aryloxyalkylenyl, alkylarylenyl, heteroaryl, heteroarylalkylenyl, heteroaryloxyalkylenyl, alkylheteroarylenyl, and heterocyclyl groups can be unsubstituted or substituted by one or more substituents independently selected from the group consisting of alkyl, alkoxy, hydroxyalkyl, haloalkyl, haloalkoxy, halogen, nitro, hydroxy, mercapto, cyano, aryl, aryloxy, arylalkyleneoxy, heteroaryl, heteroaryloxy, heteroarylalkyleneoxy, heterocyclyl, amino, acetylamino, alkylamino, dialkylamino, (dialkylamino)alkyleneoxy, and in the case of alkyl, alkenyl, alkynyl, and heterocyclyl, oxo;
R6 is selected from the group consisting of =0 and =S;
R8 is selected from the group consisting of hydrogen, alkyl, alkoxyalkylenyl, and arylalkylenyl; R9 is selected from the group consisting of hydrogen and alkyl;
Rn is selected from the group consisting of hydrogen, alkyl, halogen, and trifluoromethyl;
A is selected from the group consisting of -O-, -C(O)-, -S(0)o-2-, -CH2-, and -N(R4)-; Q is selected from the group consisting of a bond, -C(R6)-, -C(Re)-C(R6)-, -S(O)2-,
-C(R6)-N(R8)-W-, -S(O)2-N(R8)-, -C(R6)-O-, and -C(R6)-N(OR9)-;
V is selected from the group consisting of -C(R6)-, -0-C(R6)-, -N(Rg)-C(R6)-, and -S(O)2-;
W is selected from the group consisting of a bond, -C(O)-, and -S(O)2-; and a and b are independently integers from 1 to 6 with the proviso that a + b is < 7; or a pharmaceutically acceptable salt thereof.
5. A compound selected from the group consisting of the Formulas V, VI, VII, and VIII:
Figure imgf000133_0001
VIII wherein:
Z is selected from the group consisting of:
Figure imgf000133_0002
Figure imgf000134_0001
X is selected from the group consisting of a bond, -C2-3 alkylene-, -0-C2-3 alkylene-;
Ri is selected from the group consisting of: hydrogen, hydroxy, fluorine, alkoxy,
Figure imgf000134_0002
with the proviso that when Z is a bond, C1-5 alkylene,
^ and χ is a bondj
Figure imgf000134_0003
then Ri is other than hydrogen; m is an integer from 1 to 5;
R is selected from the group consisting of: halogen, hydroxy, alkyl, haloalkyl, alkoxy, and
-N(Rg)2; p is an integer from 0 to 3; R2 is selected from the group consisting of: hydrogen, alkyl, alkoxyalkylenyl, hydroxyalkylenyl, haloalkylenyl, and
R4-C(Re)-O-Ci-4 alkylenyl;
R4 is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, aryl, arylalkylenyl, aryloxyalkylenyl, alkylarylenyl, heteroaryl, heteroarylalkylenyl, heteroaryloxyalkylenyl, alkylheteroarylenyl, and heterocyclyl, wherein the alkyl, alkenyl, alkynyl, aryl, arylalkylenyl, aryloxyalkylenyl, alkylarylenyl, heteroaryl, heteroarylalkylenyl, heteroaryloxyalkylenyl, alkylheteroarylenyl, and heterocyclyl groups can be unsubstituted or substituted by one or more substituents independently selected from the group consisting of alkyl, alkoxy, hydroxyalkyl, haloalkyl, haloalkoxy, halogen, nitro, hydroxy, mercapto, cyano, aryl, aryloxy, arylalkyleneoxy, heteroaryl, heteroaryloxy, heteroarylalkyleneoxy, heterocyclyl, amino, acetylamino, alkylamino, dialkylamino,
(dialkylamino)alkyleneoxy, and in the case of alkyl, alkenyl, alkynyl, and heterocyclyl, oxo;
R6 is selected from the group consisting of =0 and =S;
R8 is selected from the group consisting of hydrogen, alkyl, alkoxyalkylenyl, and arylalkylenyl;
R9 is selected from the group consisting of hydrogen and alkyl; Rn is selected from the group consisting of hydrogen, alkyl, halogen, and trifluoromethyl;
A is selected from the group consisting of -O-, -C(O)-, -S(O)0-2-, -CH2-, and -N(R4)-; Q is selected from the group consisting of a bond, -C(R6)-, -C(R6)-C(Rδ)-, -S(O)2-,
-C(Rs)-N(Rg)-W-, -S(O)2-N(R8)-, -C(R6)-O-, and -C(Re)-N(OR8)-;
V is selected from the group consisting of -C(R6)-, -0-C(R6)-, -N(Rs)-C(R6)-, and -S(O)2-;
W is selected from the group consisting of a bond, -C(O)-, and -S(O)2-; and a and b are independently integers from 1 to 6 with the proviso that a + b is < 7; or a pharmaceutically acceptable salt thereof.
6. A compound of the Formula XIII :
Figure imgf000136_0001
XIII wherein:
Z is selected from the group consisting of:
Figure imgf000136_0002
Figure imgf000137_0001
X is selected from the group consisting of a bond, -C2-3 alkylene-, and -0-C2-3 alkylene-; Ri is selected from the group consisting of: hydrogen, hydroxy, fluorine, alkoxy,
Figure imgf000137_0002
Figure imgf000138_0001
with the proviso that when Z is a bond, Ci-5 alkylene,
Figure imgf000138_0002
5 and χ is a bondj then R] is other than hydrogen; m is an integer from 1 to 5;
RA and RB are each independently selected from the group consisting of: hydrogen, halogen, alkyl, alkenyl, alkoxy, alkylthio, and -N(Rs)2; or when taken together, RA and RB form a fused aryl ring or heteroaryl ring containing one heteroatom selected from the group consisting of N and S wherein the aryl or heteroaryl ring is unsubstituted or substituted by one or more R groups; or when taken together, RA and RB form a fused 5 to 7 membered saturated ring, optionally containing one heteroatom selected from the group consisting of N and S, and • unsubstituted or substituted by one or more R groups; R is selected from the group consisting of: halogen, hydroxy, alkyl, haloalkyl, alkoxy, and
-N(R9)2;
R2 is selected from the group consisting of: hydrogen, alkyl, alkoxyalkylenyl, hydroxyalkylenyl, haloalkylenyl, and R4-C(Re)-O-Ci-4 alkylenyl; R4 is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, aryl, arylalkylenyl, aryloxyalkylenyl, alkylarylenyl, heteroaryl, heteroarylalkylenyl, heteroaryloxyalkylenyl, alkylheteroarylenyl, and heterocyclyl, wherein the alkyl, alkenyl, alkynyl, aryl, arylalkylenyl, aryloxyalkylenyl, alkylarylenyl, heteroaryl, heteroarylalkylenyl, heteroaryloxyalkylenyl, alkylheteroarylenyl, and heterocyclyl groups can be unsubstituted or substituted by one or more substituents independently selected from the group consisting of alkyl, alkoxy, hydroxyalkyl, haloalkyl, haloalkoxy, halogen, nitro, hydroxy, mercapto, cyano, aryl, aryloxy, arylalkyleneoxy, heteroaryl, heteroaryloxy, heteroarylalkyleneoxy, heterocyclyl, amino, acetylamino, alkylamino, dialkylamino, (dialkylamino)alkyleneoxy, and in the case of alkyl, alkenyl, alkynyl, and heterocyclyl, oxo;
R6 is selected from the group consisting of =0 and =S;
R8 is selected from the group consisting of hydrogen, alkyl, alkoxyalkylenyl, and arylalkylenyl;
R9 is selected from the group consisting of hydrogen and alkyl; Rn is selected from the group consisting of hydrogen, alkyl, halogen, and trifluoromethyl;
A is selected from the group consisting of -O-, -C(O)-, -S(O)0-2-, -CH2-, and -N(R4)-;
Q is selected from the group consisting of a bond, -C(R6)-, -C(Re)-C(R6)-, -S(O)2-, -C(Re)-N(Rg)-W-, -S(O)2-N(R8)-, -C(Re)-O-, and -C(Rg)-N(OR9)-;
V is selected from the group consisting of -C(R6)-, -0-C(R6)-, -N(Rs)-C(R6)-, and -S(O)2-;
W is selected from the group consisting of a bond, -C(O)-, and -S(O)2-; a and b are independently integers from 1 to 6 with the proviso that a + b is ≤ 7; G1 is selected from the group consisting of:
-C(O)-R', α-aminoacyl, α-aminoacyl-α-aminoacyl,
Figure imgf000140_0001
R' and R" are independently selected from the group consisting of C1-J0 alkyl, C3-7 cycloalkyl, phenyl, and benzyl, each of which may be unsubstituted or substituted by one or more substituents independently selected from the group consisting of halogen, hydroxy, nitro, cyano, carboxy, Cj-6 alkyl, Ci^ alkoxy, aryl, heteroaryl, aryl-Ci-4 alkylenyl, heteroary 1-Ci-4 alky lenyl, halo-Ci-4alkylenyl, halo-Ci-4 alkoxy, -0-C(O)-CH3, -C(O)-O-CH3, -C(O)-NH2, -0-CH2-C(O)-NH2, -NH2, and -S(O)2-NH2, with the proviso that R" can also be hydrogen; α-aminoacyl is an α-aminoacyl group derived from an amino acid selected from the group consisting of racemic, D-, and L-amino acids;
Y' is selected from the group consisting of hydrogen, Ci-6 alkyl, and benzyl; Yo is selected from the group consisting of Cj-6 alkyl, carboxy-Ci-6 alky lenyl, amino-Ci-4 alkylenyl, mono-JV-Ci-6 alkylamino-Ci-4 alkylenyl, and di-_V,-V-Ci-6 alkylamino-C1-4 alkylenyl; and
Y2 is selected from the group consisting of mono-JV-Ci-ealkylamino, di~N, N-Ci-6 alkylamino, morpholin-4-yl, piperidin-1-yl, pyrrolidin-1-yl, and 4-C i -4 alkylpiperazin- 1 -yl ; or a pharmaceutically acceptable salt thereof.
7. A compound of the Formula XIV:
Figure imgf000141_0001
XIV wherein:
Z is selected from the group consisting of:
Figure imgf000141_0002
Figure imgf000142_0001
m is an integer from 1 to 5;
RA and RB are each independently selected from the group consisting of: hydrogen, halogen, alkyl, alkenyl, alkoxy, alkylthio, and -N(Re)2; or when taken together, RA and RB form a fused aryl ring or heteroaryl ring containing one heteroatom selected from the group consisting of N and S wherein the aryl or heteroaryl ring is unsubstituted or substituted by one or more R groups; or when taken together, RA and RB form a fused 5 to 7 membered saturated ring, optionally containing one heteroatom selected from the group consisting of N and S, and unsubstituted or substituted by one or more R groups; R is selected from the group consisting of: halogen, hydroxy, alkyl, haloalkyl, alkoxy, and
-N(Rs)2;
R2 is selected from the group consisting of: hydrogen, alkyl, alkoxyalkylenyl, hydroxyalkylenyl, haloalkylenyl, and R4-C(R6)O-C1-4 alkylenyl; R4 is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, aryl, arylalkylenyl, aryloxyalkylenyl, alkylarylenyl, heteroaryl, heteroarylalkylenyl, heteroaryloxyalkylenyl, alkylheteroarylenyl, and heterocyclyl, wherein the alkyl, alkenyl, alkynyl, aryl, arylalkylenyl, aryloxyalkylenyl, alkylarylenyl, heteroaryl, heteroarylalkylenyl, heteroaryloxyalkylenyl, alkylheteroarylenyl, and heterocyclyl groups can be unsubstituted or substituted by one or more substituents independently selected from the group consisting of alkyl, alkoxy, hydroxyalkyl, haloalkyl, haloalkoxy, halogen, nitro, hydroxy, mercapto, cyano, aryl, aryloxy, arylalkyleneoxy, heteroaryl, heteroaryloxy, heteroarylalkyleneoxy, heterocyclyl, amino, acetylamino, alkylamino, dialkylamino, (dialkylamino)alkyleneoxy, and in the case of alkyl, alkenyl, alkynyl, and heterocyclyl, oxo;
R6 is selected from the group consisting of =0 and =S;
R8 is selected from the group consisting of hydrogen, alkyl, alkoxyalkylenyl, and arylalkylenyl; R9 is selected from the group consisting of hydrogen and alkyl;
Rn is selected from the group consisting of hydrogen, alkyl, halogen, and trifluoromethyl;
Q is selected from the group consisting of a bond, -C(R6)-, -C(R6)-C(R6)-, -S(O)2-, -C(Re)-N(Rs)-W-, -S(O)2-N(R8)-, -C(Rg)-O-, and -C(Re)-N(OR9)-; W is selected from the group consisting of a bond, -C(O)-, and -S(O)2-;
G2 is selected from the group consisting of: -X2-C(O)-R1, α-aminoacyl, α-aminoacyl-α-aminoacyl, -X2-C(O)-O-R',
-C(0)-N(R")R', and -S(O)2-R';
X2 is selected from the group consisting of a bond; -CH2-O-; -CH(CHs)-O-; -C(CHs)2-O-; and, in the case Of -X2-C(O)-O-R1, -CH2-NH-; R' and R" are independently selected from the group consisting of C HO alkyl, C3-7 cycloalkyl, phenyl, and benzyl, each of which may be unsubstituted or substituted by one or more substituents independently selected from the group consisting of halogen, hydroxy, nitro, cyano, carboxy, Ci-6 alkyl, C1-4 alkoxy, aryl, heteroaryl, aryl-Ci-4 alkylenyl, heteroaryl-Ci-4 alkylenyl, halo-Ci^ alkylenyl, halo-C^ alkoxy, -0-C(O)-CH3, -C(O)-O-CH3, -C(O)-NH2, -0-CH2-C(O)-NH2, -NH2, and -S(O)2-NH2, with the proviso that R" can also be hydrogen; and α-aminoacyl is an α-aminoacyl group derived from an amino acid selected from the group consisting of racemic, D-, and L-amino acids; or a pharmaceutically acceptable salt thereof.
8. The compound or salt of claim 2 wherein RA' and RB 1 are independently hydrogen or alkyl.
9. The compound or salt of claim 8 wherein RA1 and RB 1 are both methyl.
10. The compound or salt of claim 3 or claim 4 wherein R is hydroxy.
11. The compound or salt of claim 3 or claim 4 wherein n is O.
12. The compound or salt of claim 5 wherein the compound is selected from the group consisting of the Formulas V and VIII:
Figure imgf000144_0001
V VIII or a pharmaceutically acceptable salt thereof.
13. The compound or salt of claim 5 or claim 12 wherein p is 0.
14. The compound or salt of any one of claims 1 through 13 wherein m is an integer from 1 to 3.
15. The compound or salt of claim 14 wherein m is 1.
16. The compound or salt of any one of claims 1 through 15 wherein Z is selected from the group consisting of a bond and C1-3 alkylene.
17. The compound or salt of any one of claims 1 through 15 wherein Z is selected from the group consisting of:
Figure imgf000145_0001
18. The compound or salt of claim 17 wherein Z is
Figure imgf000145_0002
19. The compound or salt of claim 17 wherein Z is
Figure imgf000146_0001
20. The compound or salt of any one of claims 1 through 15, 17, or 19 wherein Q is selected from the group consisting of -C(O)-, -C(O)-O-, -S(O)2-, and -C(R6)-N(R8)-.
21. The compound or salt of claim 20 wherein R4 is selected from the group consisting of alkyl, aryl, arylalkylenyl, heteroaryl, and heterocyclyl, wherein the aryl group can be unsubstituted or substituted by one or more substituents independently selected from the group consisting of acetylamino, alkyl, alkoxy, cyano, and halogen.
22. The compound or salt of any one of claims 1 through 6, 8 through 13, or 14 through 21 except as dependent on 7 wherein X is a bond.
23. The compound or salt of any one of claims 1 through 6, 8 through 13, 14 through 21 except as dependent on 7, or 22 wherein R1 is selected from the group consisting of hydroxy and methoxy.
24. The compound or salt of any one of claims 1 through 6, 8 through 13, 14 through 21 except as dependent on 7, or 22 wherein Ri is fluoro.
25. The compound or salt of any one of claims 1 through 6, 8 through 13, 14 through 21 except as dependent on 7, or 22 wherein Ri is selected from the group consisting of -NH2, -NH-Q-R4, -C(O)-NH2, and -C(O)-N(Rg)-R4, wherein:
Q is selected from the group consisting of -C(O)-, -S(O)2-, -C(O)-O-, and -C(O)-NH-,
R8 is selected from the group consisting of hydrogen and alkyl, and R4 is selected from the group consisting of alkyl and alkoxyalkylenyl.
26. The compound or salt of any one of claims 1 through 6, 8 through 13, 14 except as dependent on 7, or 15 except as dependent on 7 wherein Z is
x is a bond, and R1 is hydrogen.
Figure imgf000147_0001
27. The compound or salt of claim 13 or claim 11 as dependent on claim 3 wherein Z is
is a bond, and Ri is hydrogen.
Figure imgf000147_0002
28. The compound or salt of any one of claims 1 through 27 wherein R2 is selected from the group consisting of C^ alkyl, Ci-4 alky 1-0-C2-4 alkylenyl, and hydroxyC2-4 alkylenyl.
29. The compound of salt of claim 28 wherein R2 is selected from the group consisting of methyl, ethyl, ^-propyl, «-butyl, 2-methoxyethyl, and 2-hydroxyethyl.
30. A pharmaceutical composition comprising a therapeutically effective amount of a compound or salt of any one of claims 1-29 in combination with a pharmaceutically acceptable carrier.
31. A method of inducing cytokine biosynthesis in an animal comprising administering an effective amount of a compound or salt of any one of claims 1-29 or the pharmaceutical composition of claim 30 to the animal.
32. A method of treating a viral disease in an animal in need thereof comprising administering a therapeutically effective amount of a compound or salt of any one of claims 1-29 or the pharmaceutical composition of claim 30 to the animal.
33. A method of treating a neoplastic disease in an animal in need thereof comprising administering a therapeutically effective amount of a compound or salt of any one of claims 1-29 or the pharmaceutical composition of claim 30 to the animal.
PCT/US2006/012263 2005-04-01 2006-03-31 1-substituted pyrazolo (3,4-c) ring compounds as modulators of cytokine biosynthesis for the treatment of viral infections and neoplastic diseases WO2006107851A1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
EP06749140A EP1863814A1 (en) 2005-04-01 2006-03-31 1-substituted pyrazolo (3,4-c) ring compounds as modulators of cytokine biosynthesis for the treatment of viral infections and neoplastic diseases
AU2006232375A AU2006232375A1 (en) 2005-04-01 2006-03-31 1-substituted pyrazolo (3,4-c) ring compounds as modulators of cytokine biosynthesis for the treatment of viral infections and neoplastic diseases
CA002602590A CA2602590A1 (en) 2005-04-01 2006-03-31 1-substituted pyrazolo (3,4-c) ring compounds as modulators of cytokine biosynthesis for the treatment of viral infections and neoplastic diseases
JP2008504494A JP2008538550A (en) 2005-04-01 2006-03-31 1-Substituted pyrazolo (3,4-c) cyclic compounds as modulators of cytokine biosynthesis for treating viral infections and neoplastic diseases
US11/887,492 US7943636B2 (en) 2005-04-01 2006-03-31 1-substituted pyrazolo (3,4-C) ring compounds as modulators of cytokine biosynthesis for the treatment of viral infections and neoplastic diseases

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US66786905P 2005-04-01 2005-04-01
US60/667,869 2005-04-01
US73303705P 2005-11-03 2005-11-03
US60/733,037 2005-11-03

Publications (1)

Publication Number Publication Date
WO2006107851A1 true WO2006107851A1 (en) 2006-10-12

Family

ID=36763054

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2006/012263 WO2006107851A1 (en) 2005-04-01 2006-03-31 1-substituted pyrazolo (3,4-c) ring compounds as modulators of cytokine biosynthesis for the treatment of viral infections and neoplastic diseases

Country Status (6)

Country Link
US (1) US7943636B2 (en)
EP (1) EP1863814A1 (en)
JP (1) JP2008538550A (en)
AU (1) AU2006232375A1 (en)
CA (1) CA2602590A1 (en)
WO (1) WO2006107851A1 (en)

Cited By (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7879849B2 (en) 2003-10-03 2011-02-01 3M Innovative Properties Company Pyrazolopyridines and analogs thereof
US7897597B2 (en) 2003-08-27 2011-03-01 3M Innovative Properties Company Aryloxy and arylalkyleneoxy substituted imidazoquinolines
US7897767B2 (en) 2003-11-14 2011-03-01 3M Innovative Properties Company Oxime substituted imidazoquinolines
US7906506B2 (en) 2006-07-12 2011-03-15 3M Innovative Properties Company Substituted chiral fused [1,2] imidazo [4,5-c] ring compounds and methods
US7915281B2 (en) 2004-06-18 2011-03-29 3M Innovative Properties Company Isoxazole, dihydroisoxazole, and oxadiazole substituted imidazo ring compounds and method
US7923429B2 (en) 2003-09-05 2011-04-12 3M Innovative Properties Company Treatment for CD5+ B cell lymphoma
US7943609B2 (en) 2004-12-30 2011-05-17 3M Innovative Proprerties Company Chiral fused [1,2]imidazo[4,5-C] ring compounds
US7943636B2 (en) 2005-04-01 2011-05-17 3M Innovative Properties Company 1-substituted pyrazolo (3,4-C) ring compounds as modulators of cytokine biosynthesis for the treatment of viral infections and neoplastic diseases
US7943610B2 (en) 2005-04-01 2011-05-17 3M Innovative Properties Company Pyrazolopyridine-1,4-diamines and analogs thereof
US7968563B2 (en) 2005-02-11 2011-06-28 3M Innovative Properties Company Oxime and hydroxylamine substituted imidazo[4,5-c] ring compounds and methods
US8034938B2 (en) 2004-12-30 2011-10-11 3M Innovative Properties Company Substituted chiral fused [1,2]imidazo[4,5-c] ring compounds
US8598192B2 (en) 2003-11-14 2013-12-03 3M Innovative Properties Company Hydroxylamine substituted imidazoquinolines
US8673932B2 (en) 2003-08-12 2014-03-18 3M Innovative Properties Company Oxime substituted imidazo-containing compounds
US8691837B2 (en) 2003-11-25 2014-04-08 3M Innovative Properties Company Substituted imidazo ring systems and methods
US8735421B2 (en) 2003-12-30 2014-05-27 3M Innovative Properties Company Imidazoquinolinyl sulfonamides
US8802853B2 (en) 2003-12-29 2014-08-12 3M Innovative Properties Company Arylalkenyl and arylalkynyl substituted imidazoquinolines
US8871782B2 (en) 2003-10-03 2014-10-28 3M Innovative Properties Company Alkoxy substituted imidazoquinolines
US8951528B2 (en) 2006-02-22 2015-02-10 3M Innovative Properties Company Immune response modifier conjugates
US9248127B2 (en) 2005-02-04 2016-02-02 3M Innovative Properties Company Aqueous gel formulations containing immune response modifiers
TWI557121B (en) * 2011-04-21 2016-11-11 原真股份有限公司 Novel kinase inhibitors
EP3153180A1 (en) 2011-06-03 2017-04-12 3M Innovative Properties Company Heterobifunctional linkers with polyethylene glycol segments and immune response modifier conjugates made therefrom
US10000482B2 (en) 2012-10-19 2018-06-19 Origenis Gmbh Kinase inhibitors
US10005772B2 (en) 2006-12-22 2018-06-26 3M Innovative Properties Company Immune response modifier compositions and methods

Families Citing this family (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040265351A1 (en) 2003-04-10 2004-12-30 Miller Richard L. Methods and compositions for enhancing immune response
SG149829A1 (en) 2003-10-03 2009-02-27 3M Innovative Properties Co Pyrazolopyridines and analogs thereof
CA2559863A1 (en) 2004-03-24 2005-10-13 3M Innovative Properties Company Amide substituted imidazopyridines, imidazoquinolines, and imidazonaphthyridines
US8017779B2 (en) 2004-06-15 2011-09-13 3M Innovative Properties Company Nitrogen containing heterocyclyl substituted imidazoquinolines and imidazonaphthyridines
US8541438B2 (en) 2004-06-18 2013-09-24 3M Innovative Properties Company Substituted imidazoquinolines, imidazopyridines, and imidazonaphthyridines
US8026366B2 (en) 2004-06-18 2011-09-27 3M Innovative Properties Company Aryloxy and arylalkyleneoxy substituted thiazoloquinolines and thiazolonaphthyridines
US7897609B2 (en) 2004-06-18 2011-03-01 3M Innovative Properties Company Aryl substituted imidazonaphthyridines
WO2007120121A2 (en) 2005-02-09 2007-10-25 Coley Pharmaceutical Group, Inc. Oxime and hydroxylamine substituted thiazolo[4,5-c] ring compounds and methods
AU2006212765B2 (en) 2005-02-09 2012-02-02 3M Innovative Properties Company Alkyloxy substituted thiazoloquinolines and thiazolonaphthyridines
JP2008532933A (en) 2005-02-11 2008-08-21 コーリー ファーマシューティカル グループ,インコーポレイテッド Substituted imidazoquinolines and substituted imidazonaphthyridines
JP2008543725A (en) 2005-02-23 2008-12-04 コーリー ファーマシューティカル グループ,インコーポレイテッド Hydroxyalkyl substituted imidazoquinolines
AU2006216686A1 (en) 2005-02-23 2006-08-31 Coley Pharmaceutical Group, Inc. Method of preferentially inducing the biosynthesis of interferon
WO2006091567A2 (en) 2005-02-23 2006-08-31 Coley Pharmaceutical Group, Inc. Hydroxyalkyl substituted imidazoquinoline compounds and methods
ZA200803029B (en) 2005-09-09 2009-02-25 Coley Pharm Group Inc Amide and carbamate derivatives of alkyl substituted /V-[4-(4-amino-1H-imidazo[4,5-c] quinolin-1-yl)butyl] methane-sulfonamides and methods
US8476292B2 (en) 2005-09-09 2013-07-02 3M Innovative Properties Company Amide and carbamate derivatives of N-{2-[4-amino-2-(ethoxymethyl)-1H-imidazo[4,5-c] quinolin-1-Yl]-1,1-dimethylethyl}methanesulfonamide and methods
EP1948173B1 (en) 2005-11-04 2013-07-17 3M Innovative Properties Company Hydroxy and alkoxy substituted 1h-imidazoquinolines and methods
WO2007106854A2 (en) 2006-03-15 2007-09-20 Coley Pharmaceutical Group, Inc. Hydroxy and alkoxy substituted 1h-imidazonaphthyridines and methods
US8178539B2 (en) 2006-09-06 2012-05-15 3M Innovative Properties Company Substituted 3,4,6,7-tetrahydro-5H-1,2a,4a,8-tetraazacyclopenta[cd]phenalenes and methods
DK2268618T3 (en) * 2008-03-03 2015-08-17 Novartis Ag Compounds and compositions as TLR aktivitetsmodulatorer
MX2011010050A (en) 2009-03-25 2011-12-14 Univ Texas Compositions for stimulation of mammalian innate immune resistance to pathogens.
DK2606047T3 (en) 2010-08-17 2017-03-27 3M Innovative Properties Co COMPOSITIONS AND FORMULATIONS WITH LIPIDIZED IMMUNE RESPONSE-MODIFIING COMPOUND AND PROCEDURES THEREOF
CN103582496B (en) 2011-06-03 2016-05-11 3M创新有限公司 There is the Heterobifunctional connection base of polyethylene glycol segment and the immune response modifier conjugate of being made by it
WO2016044839A2 (en) 2014-09-19 2016-03-24 The Board Of Regents Of The University Of Texas System Compositions and methods for treating viral infections through stimulated innate immunity in combination with antiviral compounds
JP6137225B2 (en) * 2015-03-04 2017-05-31 トヨタ自動車株式会社 Bumper absorber mounting structure
GB201617758D0 (en) * 2016-10-20 2016-12-07 Almac Discovery Limited Pharmaceutical compounds
CA3086439A1 (en) 2017-12-20 2019-06-27 3M Innovative Properties Company Amide substitued imidazo[4,5-c]quinoline compounds with a branched chain linking group for use as an immune response modifier

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4689338A (en) * 1983-11-18 1987-08-25 Riker Laboratories, Inc. 1H-Imidazo[4,5-c]quinolin-4-amines and antiviral use
WO1995002598A1 (en) * 1993-07-15 1995-01-26 Minnesota Mining And Manufacturing Company Fused cycloalkylimidazopyridines
EP1104764A1 (en) * 1998-08-12 2001-06-06 Hokuriku Seiyaku Co., Ltd. 1h-imidazopyridine derivatives
WO2004058759A1 (en) * 2002-12-20 2004-07-15 3M Innovative Properties Company Aryl / hetaryl substituted imidazoquinolines
WO2005079195A2 (en) * 2003-10-03 2005-09-01 3M Innovative Properties Company Pyrazolopyridines and analogs thereof
WO2006028545A2 (en) * 2004-06-18 2006-03-16 3M Innovative Properties Company Substituted imidazoquinolines, imidazopyridines, and imidazonaphthyridines

Family Cites Families (310)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3314941A (en) 1964-06-23 1967-04-18 American Cyanamid Co Novel substituted pyridodiazepins
DE1645976A1 (en) 1966-06-18 1971-01-21 Ajinomoto Kk Process for the preparation of adenosine and 2 ', 3'-O-isopropylidene adenosine
ZA704419B (en) 1969-07-21 1971-04-28 Ici Australia Ltd Injectable aqueous solutions of tetramisole
US3692907A (en) 1970-10-27 1972-09-19 Richardson Merrell Inc Treating viral infections with bis-basic ethers and thioethers of fluorenone and fluorene and pharmaceutical compositons of the same
US3917624A (en) 1972-09-27 1975-11-04 Pfizer Process for producing 2-amino-nicotinonitrile intermediates
US4006237A (en) 1973-10-11 1977-02-01 Beecham Group Limited Tetrahydrocarbostyril derivatives for the prophylaxis of asthma, hayfever and rhinitis
US3891660A (en) 1974-02-07 1975-06-24 Squibb & Sons Inc Derivatives of 1H-imidazo{8 4,5-c{9 pyridine-7-carboxylic acids and esters
US3899508A (en) 1974-04-12 1975-08-12 Lilly Co Eli 5-(2-Aminophenyl)pyrazole-3-carboxylic acids and esters thereof
DE2423389A1 (en) 1974-05-14 1975-12-04 Hoechst Ag PSYCHOTROPIC MEDICINAL PRODUCTS AND THE METHOD OF MANUFACTURING THEREOF
JPS6016438B2 (en) 1976-10-14 1985-04-25 ウェルファイド株式会社 Imidazoquinoline derivatives
US4381344A (en) 1980-04-25 1983-04-26 Burroughs Wellcome Co. Process for producing deoxyribosides using bacterial phosphorylase
DE3204126A1 (en) 1982-02-06 1983-08-11 Bayer Ag, 5090 Leverkusen PYRAZOLOXAZINE, -THIAZINE, -CHINOLINE, METHOD FOR THE PRODUCTION THEREOF AND THEIR USE AS A MEDICINAL PRODUCT
US4758574A (en) 1982-05-03 1988-07-19 Eli Lilly And Company 2-phenylimidazio (4,5-c) pyridines
US4563525A (en) 1983-05-31 1986-01-07 Ici Americas Inc. Process for preparing pyrazolopyridine compounds
CA1271477A (en) 1983-11-18 1990-07-10 John F. Gerster 1h-imidazo[4,5-c]quinolin-4-amines
ZA848968B (en) 1983-11-18 1986-06-25 Riker Laboratories Inc 1h-imidazo(4,5-c)quinolines and 1h-imidazo(4,5-c)quinolin-4-amines
JPS61112075A (en) 1984-11-05 1986-05-30 Shionogi & Co Ltd Thienylpyrazoloquinoline derivative
US4668686A (en) 1985-04-25 1987-05-26 Bristol-Myers Company Imidazoquinoline antithrombrogenic cardiotonic agents
US4593821A (en) 1985-04-25 1986-06-10 Laros Equipment Company, Inc. Belt separator for blow molding parts
US4698346A (en) 1985-05-08 1987-10-06 Usv Pharmaceutical Corporation Thiazolo[5,4-h]quinoline compounds useful as anti-allergy agents
US4826830A (en) 1985-07-31 1989-05-02 Jui Han Topical application of glyciphosphoramide
CA1306260C (en) 1985-10-18 1992-08-11 Shionogi & Co., Ltd. Condensed imidazopyridine derivatives
HU197019B (en) 1985-11-12 1989-02-28 Egyt Gyogyszervegyeszeti Gyar Process for producing thiqzolo (4,5-c) quinoline derivatives and pharmaceuticals comprising same
US4837378A (en) 1986-01-15 1989-06-06 Curatek Pharmaceuticals, Inc. Topical metronidazole formulations and therapeutic uses thereof
JPS6310787A (en) 1986-03-06 1988-01-18 Takeda Chem Ind Ltd Nucleotide analog, production thereof and antiviral agent
US4775674A (en) 1986-05-23 1988-10-04 Bristol-Myers Company Imidazoquinolinylether derivatives useful as phosphodiesterase and blood aggregation inhibitors
CA1287061C (en) 1986-06-27 1991-07-30 Roche Holding Ltd. Pyridine ethanolamine derivatives
US5500228A (en) 1987-05-26 1996-03-19 American Cyanamid Company Phase separation-microencapsulated pharmaceuticals compositions useful for alleviating dental disease
US4880779A (en) 1987-07-31 1989-11-14 Research Corporation Technologies, Inc. Method of prevention or treatment of AIDS by inhibition of human immunodeficiency virus
US4774339A (en) 1987-08-10 1988-09-27 Molecular Probes, Inc. Chemically reactive dipyrrometheneboron difluoride dyes
JPH01180156A (en) 1988-01-12 1989-07-18 Nec Corp Packet switching circuit
US5536743A (en) 1988-01-15 1996-07-16 Curatek Pharmaceuticals Limited Partnership Intravaginal treatment of vaginal infections with buffered metronidazole compositions
US5225183A (en) 1988-12-06 1993-07-06 Riker Laboratories, Inc. Medicinal aerosol formulations
US5736553A (en) 1988-12-15 1998-04-07 Riker Laboratories, Inc. Topical formulations and transdermal delivery systems containing 1-isobutyl-1H-imidazo 4,5-C!quinolin-4-amine
US5238944A (en) 1988-12-15 1993-08-24 Riker Laboratories, Inc. Topical formulations and transdermal delivery systems containing 1-isobutyl-1H-imidazo[4,5-c]quinolin-4-amine
US5756747A (en) 1989-02-27 1998-05-26 Riker Laboratories, Inc. 1H-imidazo 4,5-c!quinolin-4-amines
DE69029212T2 (en) 1989-02-27 1997-05-22 Riker Laboratories Inc 4-Amino-1H-imidazo (4,5-c) quinolines as antiviral agents
US5457183A (en) 1989-03-06 1995-10-10 Board Of Regents, The University Of Texas System Hydroxylated texaphyrins
US4929624A (en) 1989-03-23 1990-05-29 Minnesota Mining And Manufacturing Company Olefinic 1H-imidazo(4,5-c)quinolin-4-amines
US5037986A (en) 1989-03-23 1991-08-06 Minnesota Mining And Manufacturing Company Olefinic 1H-imidazo[4,5-c]quinolin-4-amines
NZ232740A (en) 1989-04-20 1992-06-25 Riker Laboratories Inc Solution for parenteral administration comprising a 1h-imidazo(4,5-c) quinolin-4-amine derivative, an acid and a tonicity adjuster
US4988815A (en) 1989-10-26 1991-01-29 Riker Laboratories, Inc. 3-Amino or 3-nitro quinoline compounds which are intermediates in preparing 1H-imidazo[4,5-c]quinolines
EP0452331A1 (en) 1989-11-02 1991-10-23 PFEIFER &amp; LANGEN Process and device for preventing crust formation in continuously operating sugar-crystallization equipment
US5750134A (en) 1989-11-03 1998-05-12 Riker Laboratories, Inc. Bioadhesive composition and patch
JPH05503517A (en) 1989-12-18 1993-06-10 バージニア・コモンウェルス・ユニバーシティ Sigma receptor ligand and its uses
US5274113A (en) 1991-11-01 1993-12-28 Molecular Probes, Inc. Long wavelength chemically reactive dipyrrometheneboron difluoride dyes and conjugates
US5530114A (en) 1990-04-30 1996-06-25 Isis Pharmaceuticals, Inc. Oligonucleotide modulation of arachidonic acid metabolism
AU653524B2 (en) 1990-06-08 1994-10-06 Roussel-Uclaf New imidazole derivatives, their preparation process, the new intermediates obtained, their use as medicaments and the pharmaceutical compositions containing them
JP2660086B2 (en) 1990-07-03 1997-10-08 明治製菓株式会社 Agent for improving brain and cardiac dysfunction
WO1992006093A1 (en) 1990-10-05 1992-04-16 Minnesota Mining And Manufacturing Company Process for the preparation of imidazo[4,5-c]quinolin-4-amines
MX9203481A (en) 1990-10-18 1992-07-01 Minnesota Mining & Mfg FORMULATIONS.
US5248782A (en) 1990-12-18 1993-09-28 Molecular Probes, Inc. Long wavelength heteroaryl-substituted dipyrrometheneboron difluoride dyes
US5175296A (en) 1991-03-01 1992-12-29 Minnesota Mining And Manufacturing Company Imidazo[4,5-c]quinolin-4-amines and processes for their preparation
HU222250B1 (en) 1991-03-01 2003-05-28 Minnesota Mining And Manufacturing Company Intermediates for producing 1- and 2-substituted 1h-imidazo[4,5-c]quinolin-4-amine derivatives
US5389640A (en) 1991-03-01 1995-02-14 Minnesota Mining And Manufacturing Company 1-substituted, 2-substituted 1H-imidazo[4,5-c]quinolin-4-amines
JPH04327587A (en) 1991-04-26 1992-11-17 Asahi Chem Ind Co Ltd 6'-c-alkyl-3-deazaneplanocin a derivative, its production and use
US5187288A (en) 1991-05-22 1993-02-16 Molecular Probes, Inc. Ethenyl-substituted dipyrrometheneboron difluoride dyes and their synthesis
US5268376A (en) 1991-09-04 1993-12-07 Minnesota Mining And Manufacturing Company 1-substituted 1H-imidazo[4,5-c]quinolin-4-amines
TW300219B (en) 1991-09-14 1997-03-11 Hoechst Ag
PH31245A (en) 1991-10-30 1998-06-18 Janssen Pharmaceutica Nv 1,3-Dihydro-2H-imidazoÄ4,5-BÜ-quinolin-2-one derivatives.
US5266575A (en) 1991-11-06 1993-11-30 Minnesota Mining And Manufacturing Company 2-ethyl 1H-imidazo[4,5-ciquinolin-4-amines
US5378848A (en) 1992-02-12 1995-01-03 Shionogi & Co., Ltd. Condensed imidazopyridine derivatives
IL105325A (en) 1992-04-16 1996-11-14 Minnesota Mining & Mfg Immunogen/vaccine adjuvant composition
EP0641192B1 (en) 1992-05-18 1997-07-23 Minnesota Mining And Manufacturing Company Transmucosal drug delivery device
US5352680A (en) 1992-07-15 1994-10-04 Regents Of The University Of Minnesota Delta opioid receptor antagonists to block opioid agonist tolerance and dependence
US6608201B2 (en) 1992-08-28 2003-08-19 3M Innovative Properties Company Process for preparing 1-substituted, 2-substituted 1H-imidazo[4,5-c]quinolin-4-amines
US5395937A (en) 1993-01-29 1995-03-07 Minnesota Mining And Manufacturing Company Process for preparing quinoline amines
EP0689424B1 (en) 1993-03-17 1998-10-14 Minnesota Mining And Manufacturing Company Aerosol formulation containing an ester-, amide-, or mercaptoester-derived dispersing aid
DE4309969A1 (en) 1993-03-26 1994-09-29 Bayer Ag Substituted hetero-fused imidazoles
DE69314318T2 (en) 1993-04-27 1998-04-09 Agfa Gevaert Nv Method for inserting a water-soluble compound into a hydrophilic layer
US5648516A (en) 1994-07-20 1997-07-15 Minnesota Mining And Manufacturing Company Fused cycloalkylimidazopyridines
ES2149276T3 (en) 1993-07-15 2000-11-01 Minnesota Mining & Mfg IMIDAZO (4,5-C) PIRIDIN-4-AMINAS.
CA2131680C (en) 1993-09-17 2006-11-07 Gerhard Stucky Process for preparing imidazopyridine derivatives
US5837809A (en) 1995-08-11 1998-11-17 Oregon Health Sciences University Mammalian opioid receptor ligand and uses
JPH07163368A (en) 1993-12-15 1995-06-27 Hayashibara Biochem Lab Inc Recombinant dna and transformant containing the same recombinant dna
TW574214B (en) 1994-06-08 2004-02-01 Pfizer Corticotropin releasing factor antagonists
US6207646B1 (en) 1994-07-15 2001-03-27 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
US6239116B1 (en) 1994-07-15 2001-05-29 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
PT772619E (en) 1994-07-15 2006-10-31 Univ Iowa Res Found OLIGONUCLEOTIDOS IMUNOMODULADORES
US5612377A (en) 1994-08-04 1997-03-18 Minnesota Mining And Manufacturing Company Method of inhibiting leukotriene biosynthesis
US5644063A (en) 1994-09-08 1997-07-01 Minnesota Mining And Manufacturing Company Imidazo[4,5-c]pyridin-4-amine intermediates
GB9420168D0 (en) 1994-10-06 1994-11-23 Boots Co Plc Therapeutic agents
US5571819A (en) 1994-11-22 1996-11-05 Sabb; Annmarie L. Imidazopyridines as muscarinic agents
US5482936A (en) 1995-01-12 1996-01-09 Minnesota Mining And Manufacturing Company Imidazo[4,5-C]quinoline amines
US6071949A (en) 1995-03-14 2000-06-06 The United States Of America As Represented By The Department Of Health And Human Services Use of lipoxygenase inhibitors as anti-cancer therapeutic and intervention agents
US5585612A (en) 1995-03-20 1996-12-17 Harp Enterprises, Inc. Method and apparatus for voting
FR2732605B1 (en) 1995-04-07 1997-05-16 Pasteur Merieux Serums Vacc COMPOSITION FOR INDUCING MUCOSAL IMMUNE RESPONSE
US5766789A (en) 1995-09-29 1998-06-16 Energetics Systems Corporation Electrical energy devices
DE69628804T2 (en) 1995-12-08 2003-12-18 Pfizer Substituted heterocyclic derivatives as CRF antagonists
JPH09208584A (en) 1996-01-29 1997-08-12 Terumo Corp Amide derivative, pharmaceutical preparation containing the same, and intermediate for synthesizing the same
US5939047A (en) 1996-04-16 1999-08-17 Jernberg; Gary R. Local delivery of chemotherapeutic agents for treatment of periodontal disease
US5861268A (en) 1996-05-23 1999-01-19 Biomide Investment Limited Partnership Method for induction of tumor cell apoptosis with chemical inhibitors targeted to 12-lipoxygenase
US5741908A (en) 1996-06-21 1998-04-21 Minnesota Mining And Manufacturing Company Process for reparing imidazoquinolinamines
US5693811A (en) 1996-06-21 1997-12-02 Minnesota Mining And Manufacturing Company Process for preparing tetrahdroimidazoquinolinamines
WO1998001448A1 (en) 1996-07-03 1998-01-15 Japan Energy Corporation Novel purine derivatives
US6387938B1 (en) 1996-07-05 2002-05-14 Mochida Pharmaceutical Co., Ltd. Benzimidazole derivatives
KR100518903B1 (en) 1996-10-25 2005-10-06 미네소타 마이닝 앤드 매뉴팩춰링 캄파니 Immune response modifier compounds for treatment of the th2 mediated and related diseases
US5939090A (en) 1996-12-03 1999-08-17 3M Innovative Properties Company Gel formulations for topical drug delivery
US6069149A (en) 1997-01-09 2000-05-30 Terumo Kabushiki Kaisha Amide derivatives and intermediates for the synthesis thereof
US6406705B1 (en) 1997-03-10 2002-06-18 University Of Iowa Research Foundation Use of nucleic acids containing unmethylated CpG dinucleotide as an adjuvant
US6426334B1 (en) 1997-04-30 2002-07-30 Hybridon, Inc. Oligonucleotide mediated specific cytokine induction and reduction of tumor growth in a mammal
US6303347B1 (en) 1997-05-08 2001-10-16 Corixa Corporation Aminoalkyl glucosaminide phosphate compounds and their use as adjuvants and immunoeffectors
US6113918A (en) 1997-05-08 2000-09-05 Ribi Immunochem Research, Inc. Aminoalkyl glucosamine phosphate compounds and their use as adjuvants and immunoeffectors
AU7690898A (en) 1997-05-20 1998-12-11 Ottawa Civic Hospital Loeb Research Institute Vectors and methods for immunization or therapeutic protocols
US6123957A (en) 1997-07-16 2000-09-26 Jernberg; Gary R. Delivery of agents and method for regeneration of periodontal tissues
KR100528112B1 (en) 1997-08-20 2006-03-17 소니 가부시끼 가이샤 Apparatus and method for manufacturing disc-shaped recording medium
US6309623B1 (en) 1997-09-29 2001-10-30 Inhale Therapeutic Systems, Inc. Stabilized preparations for use in metered dose inhalers
DE69817393T2 (en) 1997-11-28 2004-06-17 Sumitomo Pharmaceuticals Co., Ltd. NEW HETEROCYCLIC CONNECTIONS
US6121323A (en) 1997-12-03 2000-09-19 3M Innovative Properties Company Bishydroxyureas
UA67760C2 (en) 1997-12-11 2004-07-15 Міннесота Майнінг Енд Мануфакчурінг Компані Imidazonaphthyridines and use thereof to induce the biosynthesis of cytokines
TW572758B (en) 1997-12-22 2004-01-21 Sumitomo Pharma Type 2 helper T cell-selective immune response inhibitors comprising purine derivatives
JPH11222432A (en) 1998-02-03 1999-08-17 Terumo Corp Preparation for external use containing amide derivative inducing interferon
US6114058A (en) 1998-05-26 2000-09-05 Siemens Westinghouse Power Corporation Iron aluminide alloy container for solid oxide fuel cells
US6110929A (en) 1998-07-28 2000-08-29 3M Innovative Properties Company Oxazolo, thiazolo and selenazolo [4,5-c]-quinolin-4-amines and analogs thereof
US5962636A (en) 1998-08-12 1999-10-05 Amgen Canada Inc. Peptides capable of modulating inflammatory heart disease
US6518280B2 (en) 1998-12-11 2003-02-11 3M Innovative Properties Company Imidazonaphthyridines
EP1495758A3 (en) 1999-01-08 2005-04-13 3M Innovative Properties Company Formulations and methods for treatment of mucosal associated conditions with an immune response modifier
AU776654B2 (en) 1999-01-08 2004-09-16 3M Innovative Properties Company Formulations and methods for treatment of mucosal associated conditions with an immune response modifier
US6486168B1 (en) 1999-01-08 2002-11-26 3M Innovative Properties Company Formulations and methods for treatment of mucosal associated conditions with an immune response modifier
US20020058674A1 (en) 1999-01-08 2002-05-16 Hedenstrom John C. Systems and methods for treating a mucosal surface
US6558951B1 (en) 1999-02-11 2003-05-06 3M Innovative Properties Company Maturation of dendritic cells with immune response modifying compounds
US6294271B1 (en) 1999-02-12 2001-09-25 Shin-Etsu Chemical Co., Ltd. Flip-chip type semiconductor device sealing material and flip-chip type semiconductor device
JP2000247884A (en) 1999-03-01 2000-09-12 Sumitomo Pharmaceut Co Ltd Arachidonic acid-induced skin disease-treating agent
WO2000061765A2 (en) 1999-04-12 2000-10-19 Lexicon Genetics Incorporated Lipoxygenase proteins and polynucleotides encoding the same
US6331539B1 (en) 1999-06-10 2001-12-18 3M Innovative Properties Company Sulfonamide and sulfamide substituted imidazoquinolines
US6451810B1 (en) 1999-06-10 2002-09-17 3M Innovative Properties Company Amide substituted imidazoquinolines
US6756382B2 (en) 1999-06-10 2004-06-29 3M Innovative Properties Company Amide substituted imidazoquinolines
US6573273B1 (en) 1999-06-10 2003-06-03 3M Innovative Properties Company Urea substituted imidazoquinolines
US6541485B1 (en) 1999-06-10 2003-04-01 3M Innovative Properties Company Urea substituted imidazoquinolines
US6315985B1 (en) 1999-06-18 2001-11-13 3M Innovative Properties Company C-17/21 OH 20-ketosteroid solution aerosol products with enhanced chemical stability
AU783745B2 (en) 1999-08-13 2005-12-01 Hybridon, Inc. Modulation of oligonucleotide CpG-mediated immune stimulation by positional modification of nucleosides
US6432989B1 (en) 1999-08-27 2002-08-13 Pfizer Inc Use of CRF antagonists to treat circadian rhythm disorders
CO5271670A1 (en) 1999-10-29 2003-04-30 Pfizer Prod Inc ANTIGONISTS OF THE CORTICITROPINE RELEASE FACTOR AND RELATED COMPOSITIONS
PT1666028E (en) 1999-10-29 2010-06-15 Novartis Ag Dry powder compositions having improved dispersivity
US6376669B1 (en) 1999-11-05 2002-04-23 3M Innovative Properties Company Dye labeled imidazoquinoline compounds
US6916925B1 (en) 1999-11-05 2005-07-12 3M Innovative Properties Co. Dye labeled imidazoquinoline compounds
US6313156B1 (en) 1999-12-23 2001-11-06 Icos Corporation Thiazole compounds as cyclic-AMP-specific phosphodiesterase inhibitors
US20040023870A1 (en) 2000-01-21 2004-02-05 Douglas Dedera Methods of therapy and diagnosis using targeting of cells that express toll-like receptor proteins
GB0001704D0 (en) 2000-01-25 2000-03-15 Glaxo Group Ltd Protein
BR0108303A (en) 2000-02-09 2003-03-05 Hokuriku Pharmaceutical 1h-Imidazopyridine Derivatives
JP2003528068A (en) 2000-03-17 2003-09-24 コリクサ コーポレイション New amphiphilic aldehydes and their use as adjuvants and immune effectors
US20010046968A1 (en) 2000-03-23 2001-11-29 Zagon Ian S. Opioid growth factor modulates angiogenesis
MXPA02009460A (en) 2000-03-30 2003-02-12 Shionogi & Co Novel process for producing fused imidazopyridine derivative and novel crystal form.
US6894060B2 (en) 2000-03-30 2005-05-17 3M Innovative Properties Company Method for the treatment of dermal lesions caused by envenomation
WO2001090323A2 (en) 2000-05-19 2001-11-29 Millennium Pharmaceuticals, Inc. 46638, a putative family member of human lipoxygenase
DE10020465A1 (en) 2000-04-26 2001-11-08 Osram Opto Semiconductors Gmbh Radiation-emitting semiconductor component with luminescence conversion element
DE10029580C1 (en) 2000-06-15 2002-01-10 Ferton Holding Sa Device for removing body stones with an intracorporeal lithotripter
US6387383B1 (en) 2000-08-03 2002-05-14 Dow Pharmaceutical Sciences Topical low-viscosity gel composition
US6900016B1 (en) 2000-09-08 2005-05-31 Applera Corporation Polymorphisms in known genes associated with inflammatory autoimmune disease, methods of detection and uses thereof
JP2005500806A (en) 2000-09-15 2005-01-13 コーリー ファーマシューティカル ゲーエムベーハー Process for high-throughput screening of immune agonist / immunoantagonists based on CpG
US20020055517A1 (en) 2000-09-15 2002-05-09 3M Innovative Properties Company Methods for delaying recurrence of herpes virus symptoms
GB0023008D0 (en) 2000-09-20 2000-11-01 Glaxo Group Ltd Improvements in vaccination
EP1342799B1 (en) 2000-12-07 2007-01-17 Aoyama Seisakusho Co., Ltd. Method for baking steel parts
US6660735B2 (en) 2000-12-08 2003-12-09 3M Innovative Properties Company Urea substituted imidazoquinoline ethers
US6664264B2 (en) 2000-12-08 2003-12-16 3M Innovative Properties Company Thioether substituted imidazoquinolines
WO2002046749A2 (en) 2000-12-08 2002-06-13 3M Innovative Properties Company Screening method for identifying compounds that selectively induce interferon alpha
US6677347B2 (en) 2000-12-08 2004-01-13 3M Innovative Properties Company Sulfonamido ether substituted imidazoquinolines
UA74852C2 (en) 2000-12-08 2006-02-15 3M Innovative Properties Co Urea-substituted imidazoquinoline ethers
US6545016B1 (en) 2000-12-08 2003-04-08 3M Innovative Properties Company Amide substituted imidazopyridines
US6545017B1 (en) 2000-12-08 2003-04-08 3M Innovative Properties Company Urea substituted imidazopyridines
US6677348B2 (en) 2000-12-08 2004-01-13 3M Innovative Properties Company Aryl ether substituted imidazoquinolines
US6660747B2 (en) 2000-12-08 2003-12-09 3M Innovative Properties Company Amido ether substituted imidazoquinolines
US6664260B2 (en) 2000-12-08 2003-12-16 3M Innovative Properties Company Heterocyclic ether substituted imidazoquinolines
US6525064B1 (en) 2000-12-08 2003-02-25 3M Innovative Properties Company Sulfonamido substituted imidazopyridines
UA74593C2 (en) 2000-12-08 2006-01-16 3M Innovative Properties Co Substituted imidazopyridines
US6667312B2 (en) 2000-12-08 2003-12-23 3M Innovative Properties Company Thioether substituted imidazoquinolines
US20020107262A1 (en) 2000-12-08 2002-08-08 3M Innovative Properties Company Substituted imidazopyridines
US6664265B2 (en) 2000-12-08 2003-12-16 3M Innovative Properties Company Amido ether substituted imidazoquinolines
US20020182274A1 (en) 2001-03-21 2002-12-05 Kung-Ming Lu Methods for inhibiting cancer growth, reducing infection and promoting general health with a fermented soy extract
ES2314042T3 (en) 2001-04-17 2009-03-16 Dainippon Sumitomo Pharma Co., Ltd. NEW DERIVATIVES OF ADENINA.
US20020193729A1 (en) 2001-04-20 2002-12-19 Cormier Michel J.N. Microprojection array immunization patch and method
EP1379552B2 (en) 2001-04-20 2014-11-19 The Institute for Systems Biology Toll-like receptor 5 ligands and methods of use
US6627639B2 (en) 2001-04-26 2003-09-30 Wyeth Uses for indoletetrahydropyridine derivatives of 2,3-dihydro-7H-[1,4]dioxino-[2,3-e]indole
US7226928B2 (en) 2001-06-15 2007-06-05 3M Innovative Properties Company Methods for the treatment of periodontal disease
US6756399B2 (en) 2001-06-29 2004-06-29 The United States Of America As Represented By The Department Of Health And Human Services Use of lipoxygenase inhibitors and PPAR ligands as anti-cancer therapeutic and intervention agents
US6987187B2 (en) 2001-07-16 2006-01-17 Shionogi & Co., Ltd. Process for preparation of amidine derivatives
EP1427445A4 (en) 2001-08-30 2006-09-06 3M Innovative Properties Co Methods of maturing plasmacytoid dendritic cells using immune response modifier molecules
US7132438B2 (en) 2001-10-09 2006-11-07 Amgen Inc. Benzimidazole derivatives
CA2462203A1 (en) 2001-10-12 2003-11-20 University Of Iowa Research Foundation Methods and products for enhancing immune responses using imidazoquinoline compounds
PT1719511E (en) 2001-11-16 2009-03-06 Coley Pharm Group Inc N-[4-(4-amino-2-ethyl-1h-imidazo[4,5-c]quinolin-1-yl)butyl]methanesulfonamide, a pharmaceutical composition comprising the same and use thereof
US20050009858A1 (en) 2001-11-17 2005-01-13 Martinez-Colon Maria I Imiquimod therapies
BR0214407A (en) 2001-11-27 2004-10-19 Anadys Pharmaceuticals Inc Compounds, pharmaceutical compositions and method of modulating cytokine immunoactivities
NZ532769A (en) 2001-11-29 2005-12-23 3M Innovative Properties Co Pharmaceutical formulations comprising an immune response modifier
US6677349B1 (en) 2001-12-21 2004-01-13 3M Innovative Properties Company Sulfonamide and sulfamide substituted imidazoquinolines
US6775514B2 (en) 2002-01-11 2004-08-10 Xerox Corporation Substrate size monitoring system for use in copier/printers
US6525028B1 (en) 2002-02-04 2003-02-25 Corixa Corporation Immunoeffector compounds
US20050281813A1 (en) 2002-02-14 2005-12-22 Nuvelo, Inc. Methods of therapy and diagnosis using targeting of cells that express toll-like receptor proteins
US7030129B2 (en) 2002-02-22 2006-04-18 3M Innovative Properties Company Method of reducing and treating UVB-induced immunosuppression
US20030185835A1 (en) 2002-03-19 2003-10-02 Braun Ralph P. Adjuvant for vaccines
US8153141B2 (en) 2002-04-04 2012-04-10 Coley Pharmaceutical Gmbh Immunostimulatory G, U-containing oligoribonucleotides
EP2108370A1 (en) 2002-04-30 2009-10-14 Unigen Pharmaceuticals, Inc. Formulation of a mixture of free-B-ring flavonoids and flavans as a therapeutic agent
GB0211649D0 (en) 2002-05-21 2002-07-03 Novartis Ag Organic compounds
US6743920B2 (en) 2002-05-29 2004-06-01 3M Innovative Properties Company Process for imidazo[4,5-c]pyridin-4-amines
AU2003237386A1 (en) 2002-06-07 2003-12-22 3M Innovative Properties Company Ether substituted imidazopyridines
US6852861B2 (en) 2002-07-23 2005-02-08 Biogal Gyogyszergyar Rt. Preparation of 1H-imidazo [4,5-C] quinolin-4-amines via 1H-imidazo [4,5-C] quinolin-4-phthalimide intermediates
KR20050030958A (en) 2002-07-26 2005-03-31 비오갈 기오기스제르갸르 알티. Preparation of 1h-imidazo[4,5-c]quinolin-4-amines via novel 1h-imidazo[4,5-c]quinolin-4-cyano and 1h-imidazo[4,5-c]quinolin-4-carboxamide intermediates
CA2495570C (en) 2002-08-15 2012-12-04 3M Innovative Properties Company Immunostimulatory compositions and methods of stimulating an immune response
AU2003299082A1 (en) 2002-09-26 2004-04-19 3M Innovative Properties Company 1h-imidazo dimers
CA2502429A1 (en) 2002-10-31 2004-05-21 Amgen Inc. Antiinflammation agents
AU2003294249A1 (en) 2002-11-08 2004-06-03 Trimeris, Inc. Hetero-substituted benzimidazole compounds and antiviral uses thereof
AU2003287324A1 (en) 2002-12-11 2004-06-30 3M Innovative Properties Company Gene expression systems and recombinant cell lines
AU2003287316A1 (en) 2002-12-11 2004-06-30 3M Innovative Properties Company Assays relating to toll-like receptor activity
EP2572715A1 (en) 2002-12-30 2013-03-27 3M Innovative Properties Company Immunostimulatory Combinations
US7375180B2 (en) 2003-02-13 2008-05-20 3M Innovative Properties Company Methods and compositions related to IRM compounds and Toll-like receptor 8
EP1599726A4 (en) 2003-02-27 2009-07-22 3M Innovative Properties Co Selective modulation of tlr-mediated biological activity
EP1601365A4 (en) 2003-03-04 2009-11-11 3M Innovative Properties Co Prophylactic treatment of uv-induced epidermal neoplasia
US7163947B2 (en) 2003-03-07 2007-01-16 3M Innovative Properties Company 1-Amino 1H-imidazoquinolines
BRPI0408125A (en) 2003-03-07 2006-03-01 3M Innovative Properties Co 1-amino 1h-imidazoquinolines
CA2518282C (en) 2003-03-13 2012-11-06 3M Innovative Properties Company Methods of improving skin quality
US7179253B2 (en) 2003-03-13 2007-02-20 3M Innovative Properties Company Method of tattoo removal
AU2004220466A1 (en) 2003-03-13 2004-09-23 3M Innovative Properties Company Methods for diagnosing skin lesions
US20040192585A1 (en) 2003-03-25 2004-09-30 3M Innovative Properties Company Treatment for basal cell carcinoma
WO2004087049A2 (en) 2003-03-25 2004-10-14 3M Innovative Properties Company Selective activation of cellular activities mediated through a common toll-like receptor
ES2423800T3 (en) 2003-03-28 2013-09-24 Novartis Vaccines And Diagnostics, Inc. Use of organic compounds for immunopotentiation
WO2004091500A2 (en) 2003-04-10 2004-10-28 3M Innovative Properties Company Delivery of immune response modifier compounds
US20040265351A1 (en) 2003-04-10 2004-12-30 Miller Richard L. Methods and compositions for enhancing immune response
WO2004096144A2 (en) 2003-04-28 2004-11-11 3M Innovative Properties Company Compositions and methods for induction of opioid receptors
WO2004110991A2 (en) 2003-06-06 2004-12-23 3M Innovative Properties Company PROCESS FOR IMIDAZO[4,5-c]PYRIDIN-4-AMINES
US20050032829A1 (en) 2003-06-06 2005-02-10 3M Innovative Properties Company Process for imidazo[4,5-c]pyridin-4-amines
BRPI0411514A (en) 2003-06-20 2006-08-01 Coley Pharm Gmbh small molecule toll-like receptor antagonists
MY157827A (en) 2003-06-27 2016-07-29 3M Innovative Properties Co Sulfonamide substituted imidazoquinolines
US20050106300A1 (en) 2003-06-30 2005-05-19 Purdue Research Foundation Method for producing a material having an increased solubility in alcohol
JP2007501252A (en) 2003-08-05 2007-01-25 スリーエム イノベイティブ プロパティズ カンパニー Formulation containing immune response modifier
CA2535117A1 (en) 2003-08-12 2005-03-03 3M Innovative Properties Company Oxime substituted imidazo-containing compounds
CA2535338C (en) 2003-08-14 2013-05-28 3M Innovative Properties Company Substituted 1h-imidazo[4,5-c]pyridin-4-amines,1h-imidazo[4,5-c]quinolin -4-amines and 1h-imidazo[4,5-c]naphthyridin-4-amines as immune response modifiers
WO2005018574A2 (en) 2003-08-25 2005-03-03 3M Innovative Properties Company Immunostimulatory combinations and treatments
EP1658076B1 (en) 2003-08-27 2013-03-06 3M Innovative Properties Company Aryloxy and arylalkyleneoxy substituted imidazoquinolines
CA2537763A1 (en) 2003-09-05 2005-03-17 3M Innovative Properties Company Treatment for cd5+ b cell lymphoma
AP2006003542A0 (en) 2003-09-05 2006-04-30 Anadys Pharmaceuticals Inc Administration of TLR7 ligands and prodrugs for treatment of infection by hepatitis C virus.
GB0321615D0 (en) 2003-09-15 2003-10-15 Glaxo Group Ltd Improvements in vaccination
US20050059072A1 (en) 2003-09-17 2005-03-17 3M Innovative Properties Company Selective modulation of TLR gene expression
WO2005033049A2 (en) 2003-10-01 2005-04-14 Taro Pharmaceuticals U.S.A., Inc. METHOD OF PREPARING 4-AMINO-1H-IMIDAZO(4,5-c)QUINOLINES AND ACID ADDITION SALTS THEREOF
EP1673087B1 (en) 2003-10-03 2015-05-13 3M Innovative Properties Company Alkoxy substituted imidazoquinolines
US20090075980A1 (en) 2003-10-03 2009-03-19 Coley Pharmaceutical Group, Inc. Pyrazolopyridines and Analogs Thereof
US7544697B2 (en) 2003-10-03 2009-06-09 Coley Pharmaceutical Group, Inc. Pyrazolopyridines and analogs thereof
CA2543685A1 (en) 2003-10-31 2005-05-12 3M Innovative Properties Company Neutrophil activation by immune response modifier compounds
US20050239733A1 (en) 2003-10-31 2005-10-27 Coley Pharmaceutical Gmbh Sequence requirements for inhibitory oligonucleotides
ITMI20032121A1 (en) 2003-11-04 2005-05-05 Dinamite Dipharma Spa In Forma Abbr Eviata Dipharm PROCEDURE FOR THE PREPARATION OF IMIQUIMOD AND ITS INTERMEDIATES
US8598192B2 (en) 2003-11-14 2013-12-03 3M Innovative Properties Company Hydroxylamine substituted imidazoquinolines
US7897767B2 (en) 2003-11-14 2011-03-01 3M Innovative Properties Company Oxime substituted imidazoquinolines
CU23404A1 (en) 2003-11-19 2009-08-04 Ct Ingenieria Genetica Biotech NEISSERIA MENINGITIDIS CAPSULAR POLYSACARIDS AS MUCOSOPT IMMUNOPOTENTIZERS AND RESULTING FORMULATIONS
AR046845A1 (en) 2003-11-21 2005-12-28 Novartis Ag DERIVATIVES OF 1H-IMIDAZO [4,5-C] QUINOLINE FOR THE TREATMENT OF PROTEIN-KINASE DEPENDENT DISEASES
WO2005054237A1 (en) 2003-11-21 2005-06-16 Novartis Ag 1h-imidazoquinoline derivatives as protein kinase inhibitors
EP1686992A4 (en) 2003-11-25 2009-11-04 3M Innovative Properties Co Hydroxylamine and oxime substituted imidazoquinolines, imidazopyridines, and imidazonaphthyridines
RU2409576C2 (en) 2003-11-25 2011-01-20 3М Инновейтив Пропертиз Компани Systems containing imidazole ring with substitutes, and methods of obtaining said systems
WO2005055932A2 (en) 2003-12-02 2005-06-23 3M Innovative Properties Company Therapeutic combinations and methods including irm compounds
US20050226878A1 (en) 2003-12-02 2005-10-13 3M Innovative Properties Company Therapeutic combinations and methods including IRM compounds
JP2007513170A (en) 2003-12-04 2007-05-24 スリーエム イノベイティブ プロパティズ カンパニー Sulfone substituted imidazo ring ether
US7888349B2 (en) 2003-12-29 2011-02-15 3M Innovative Properties Company Piperazine, [1,4]Diazepane, [1,4]Diazocane, and [1,5]Diazocane fused imidazo ring compounds
EP1701955A1 (en) 2003-12-29 2006-09-20 3M Innovative Properties Company Arylalkenyl and arylalkynyl substituted imidazoquinolines
WO2005065678A1 (en) 2003-12-30 2005-07-21 3M Innovative Properties Company Immunomodulatory combinations
US8735421B2 (en) 2003-12-30 2014-05-27 3M Innovative Properties Company Imidazoquinolinyl sulfonamides
WO2005067500A2 (en) 2003-12-30 2005-07-28 3M Innovative Properties Company Enhancement of immune responses
CA2559863A1 (en) 2004-03-24 2005-10-13 3M Innovative Properties Company Amide substituted imidazopyridines, imidazoquinolines, and imidazonaphthyridines
US20070166384A1 (en) 2004-04-09 2007-07-19 Zarraga Isidro Angelo E Methods , composition and preparations for delivery of immune response modifiers
US20050267145A1 (en) 2004-05-28 2005-12-01 Merrill Bryon A Treatment for lung cancer
US20080015184A1 (en) 2004-06-14 2008-01-17 3M Innovative Properties Company Urea Substituted Imidazopyridines, Imidazoquinolines, and Imidazonaphthyridines
US8017779B2 (en) 2004-06-15 2011-09-13 3M Innovative Properties Company Nitrogen containing heterocyclyl substituted imidazoquinolines and imidazonaphthyridines
US7897609B2 (en) 2004-06-18 2011-03-01 3M Innovative Properties Company Aryl substituted imidazonaphthyridines
WO2006009832A1 (en) 2004-06-18 2006-01-26 3M Innovative Properties Company Substituted imidazo ring systems and methods
WO2006065280A2 (en) 2004-06-18 2006-06-22 3M Innovative Properties Company Isoxazole, dihydroisoxazole, and oxadiazole substituted imidazo ring compounds and methods
US20070259907A1 (en) 2004-06-18 2007-11-08 Prince Ryan B Aryl and arylalkylenyl substituted thiazoloquinolines and thiazolonaphthyridines
US8026366B2 (en) 2004-06-18 2011-09-27 3M Innovative Properties Company Aryloxy and arylalkyleneoxy substituted thiazoloquinolines and thiazolonaphthyridines
PL1789042T3 (en) 2004-09-02 2012-09-28 3M Innovative Properties Co 1-alkoxy 1h-imidazo ring systems and methods
WO2006026760A2 (en) 2004-09-02 2006-03-09 3M Innovative Properties Company 1-amino imidazo-containing compounds and methods
CA2578975A1 (en) 2004-09-02 2006-03-16 3M Innovative Properties Company 2-amino 1h imidazo ring systems and methods
WO2006028451A1 (en) 2004-09-03 2006-03-16 3M Innovative Properties Company 1-amino 1-h-imidazoquinolines
JP4769810B2 (en) 2004-09-14 2011-09-07 ノバルティス ヴァクシンズ アンド ダイアグノスティクス, インコーポレイテッド Imidazoquinoline compounds
EP1819226A4 (en) 2004-12-08 2010-12-29 3M Innovative Properties Co Immunostimulatory combinations and methods
US20110070575A1 (en) 2004-12-08 2011-03-24 Coley Pharmaceutical Group, Inc. Immunomodulatory Compositions, Combinations and Methods
AU2005322843B2 (en) 2004-12-30 2012-03-08 3M Innovative Properties Company Immune response modifier formulations and methods
JP5313502B2 (en) 2004-12-30 2013-10-09 スリーエム イノベイティブ プロパティズ カンパニー Substituted chiral condensed [1,2] imidazo [4,5-c] cyclic compounds
AU2005322898B2 (en) 2004-12-30 2011-11-24 3M Innovative Properties Company Chiral fused (1,2)imidazo(4,5-c) ring compounds
US9248127B2 (en) 2005-02-04 2016-02-02 3M Innovative Properties Company Aqueous gel formulations containing immune response modifiers
AU2006212765B2 (en) 2005-02-09 2012-02-02 3M Innovative Properties Company Alkyloxy substituted thiazoloquinolines and thiazolonaphthyridines
WO2007120121A2 (en) 2005-02-09 2007-10-25 Coley Pharmaceutical Group, Inc. Oxime and hydroxylamine substituted thiazolo[4,5-c] ring compounds and methods
US7968563B2 (en) 2005-02-11 2011-06-28 3M Innovative Properties Company Oxime and hydroxylamine substituted imidazo[4,5-c] ring compounds and methods
WO2006086633A2 (en) 2005-02-11 2006-08-17 Coley Pharmaceutical Group, Inc. Substituted fused [1,2]imidazo[4,5-c] ring compounds and methods
JP2008532933A (en) 2005-02-11 2008-08-21 コーリー ファーマシューティカル グループ,インコーポレイテッド Substituted imidazoquinolines and substituted imidazonaphthyridines
JP2008543725A (en) 2005-02-23 2008-12-04 コーリー ファーマシューティカル グループ,インコーポレイテッド Hydroxyalkyl substituted imidazoquinolines
WO2006091567A2 (en) 2005-02-23 2006-08-31 Coley Pharmaceutical Group, Inc. Hydroxyalkyl substituted imidazoquinoline compounds and methods
AU2006216686A1 (en) 2005-02-23 2006-08-31 Coley Pharmaceutical Group, Inc. Method of preferentially inducing the biosynthesis of interferon
CA2598639A1 (en) 2005-02-23 2006-08-31 Coley Pharmaceutical Group, Inc. Hydroxyalkyl substituted imidazonaphthyridines
WO2006121528A2 (en) 2005-04-01 2006-11-16 Coley Pharmaceutical Group, Inc. Ring closing and related methods and intermediates
AU2006232294A1 (en) 2005-04-01 2006-10-12 Coley Pharmaceutical Group, Inc. Pyrazolo[3,4-c]quinolines, pyrazolo[3,4-c]naphthyridines, analogs thereof, and methods
AU2006232377A1 (en) 2005-04-01 2006-10-12 Coley Pharmaceutical Group, Inc. Pyrazolopyridine-1,4-diamines and analogs thereof
US7943636B2 (en) 2005-04-01 2011-05-17 3M Innovative Properties Company 1-substituted pyrazolo (3,4-C) ring compounds as modulators of cytokine biosynthesis for the treatment of viral infections and neoplastic diseases
GB0510390D0 (en) 2005-05-20 2005-06-29 Novartis Ag Organic compounds
BRPI0615250A2 (en) 2005-09-02 2011-05-10 Pfizer Hydroxy substituted 1h-imidazopyridines, pharmaceutical composition containing same, as well as their uses
US8476292B2 (en) 2005-09-09 2013-07-02 3M Innovative Properties Company Amide and carbamate derivatives of N-{2-[4-amino-2-(ethoxymethyl)-1H-imidazo[4,5-c] quinolin-1-Yl]-1,1-dimethylethyl}methanesulfonamide and methods
ZA200803029B (en) 2005-09-09 2009-02-25 Coley Pharm Group Inc Amide and carbamate derivatives of alkyl substituted /V-[4-(4-amino-1H-imidazo[4,5-c] quinolin-1-yl)butyl] methane-sulfonamides and methods
CA2623541A1 (en) 2005-09-23 2007-03-29 Coley Pharmaceutical Group, Inc. Method for 1h-imidazo[4,5-c]pyridines and analogs thereof
EP1948173B1 (en) 2005-11-04 2013-07-17 3M Innovative Properties Company Hydroxy and alkoxy substituted 1h-imidazoquinolines and methods
WO2007062043A1 (en) 2005-11-23 2007-05-31 Coley Pharmaceutical Group Inc. Method of activating murine toll-like receptor 8
KR20080077982A (en) 2005-12-16 2008-08-26 콜레이 파마시티컬 그룹, 인코포레이티드 Substituted imidazoquinolines, imidazonaphthyridines, and imidazopyridines, compositions, and methods
WO2007079202A2 (en) 2005-12-28 2007-07-12 Coley Pharmaceutical Group, Inc. Treatment for acute lymhoblastic leukemia
WO2007079203A2 (en) 2005-12-28 2007-07-12 3M Innovative Properties Company Treatment for cutaneous t cell lymphoma
WO2007079171A2 (en) 2005-12-28 2007-07-12 Coley Pharmaceutical Group, Inc. Treatment for hodgkin's lymphoma
WO2007079086A1 (en) 2005-12-28 2007-07-12 Coley Pharmaceutical Group, Inc. Pyrazoloalkyl substituted imidazo ring compounds and methods
WO2007079169A2 (en) 2005-12-28 2007-07-12 Coley Pharmaceutical Group, Inc. Treatment for acute myeloid leukemia
WO2007079146A1 (en) 2005-12-28 2007-07-12 Coley Pharmaceutical Group, Inc Treatment for non-hodgkin's lymphoma
WO2007092641A2 (en) 2006-02-10 2007-08-16 Coley Pharmaceutical Group, Inc. Method for substituted 1h-imidazo[4,5-c]pyridines
US8088788B2 (en) 2006-03-15 2012-01-03 3M Innovative Properties Company Substituted fused[1,2] imidazo[4,5-c] ring compounds and methods
WO2007106854A2 (en) 2006-03-15 2007-09-20 Coley Pharmaceutical Group, Inc. Hydroxy and alkoxy substituted 1h-imidazonaphthyridines and methods
WO2007143526A2 (en) 2006-06-05 2007-12-13 Coley Pharmaceutical Group, Inc. Substituted tetrahydroimidazonaphthyridines and methods
WO2008002646A2 (en) 2006-06-27 2008-01-03 Schering-Plough Healthcare Products, Inc. Aerosol lotion formulations
US7906506B2 (en) 2006-07-12 2011-03-15 3M Innovative Properties Company Substituted chiral fused [1,2] imidazo [4,5-c] ring compounds and methods
US8178539B2 (en) 2006-09-06 2012-05-15 3M Innovative Properties Company Substituted 3,4,6,7-tetrahydro-5H-1,2a,4a,8-tetraazacyclopenta[cd]phenalenes and methods
WO2008036312A1 (en) 2006-09-19 2008-03-27 Coley Pharmaceutical Group, Inc. Fungicidal methods using immune response modifier compounds
WO2008045543A1 (en) 2006-10-13 2008-04-17 Coley Pharmaceutical Group, Inc. Substituted 4h-imidazo [4, 5, 1-ij] [1, 6] naphthyridine-9-amines and their pharmaceutical use

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4689338A (en) * 1983-11-18 1987-08-25 Riker Laboratories, Inc. 1H-Imidazo[4,5-c]quinolin-4-amines and antiviral use
WO1995002598A1 (en) * 1993-07-15 1995-01-26 Minnesota Mining And Manufacturing Company Fused cycloalkylimidazopyridines
EP1104764A1 (en) * 1998-08-12 2001-06-06 Hokuriku Seiyaku Co., Ltd. 1h-imidazopyridine derivatives
WO2004058759A1 (en) * 2002-12-20 2004-07-15 3M Innovative Properties Company Aryl / hetaryl substituted imidazoquinolines
WO2005079195A2 (en) * 2003-10-03 2005-09-01 3M Innovative Properties Company Pyrazolopyridines and analogs thereof
WO2006028545A2 (en) * 2004-06-18 2006-03-16 3M Innovative Properties Company Substituted imidazoquinolines, imidazopyridines, and imidazonaphthyridines

Cited By (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8673932B2 (en) 2003-08-12 2014-03-18 3M Innovative Properties Company Oxime substituted imidazo-containing compounds
US7897597B2 (en) 2003-08-27 2011-03-01 3M Innovative Properties Company Aryloxy and arylalkyleneoxy substituted imidazoquinolines
US7923429B2 (en) 2003-09-05 2011-04-12 3M Innovative Properties Company Treatment for CD5+ B cell lymphoma
US8871782B2 (en) 2003-10-03 2014-10-28 3M Innovative Properties Company Alkoxy substituted imidazoquinolines
US7879849B2 (en) 2003-10-03 2011-02-01 3M Innovative Properties Company Pyrazolopyridines and analogs thereof
US8598192B2 (en) 2003-11-14 2013-12-03 3M Innovative Properties Company Hydroxylamine substituted imidazoquinolines
US7897767B2 (en) 2003-11-14 2011-03-01 3M Innovative Properties Company Oxime substituted imidazoquinolines
US8691837B2 (en) 2003-11-25 2014-04-08 3M Innovative Properties Company Substituted imidazo ring systems and methods
US8802853B2 (en) 2003-12-29 2014-08-12 3M Innovative Properties Company Arylalkenyl and arylalkynyl substituted imidazoquinolines
US8735421B2 (en) 2003-12-30 2014-05-27 3M Innovative Properties Company Imidazoquinolinyl sulfonamides
US7915281B2 (en) 2004-06-18 2011-03-29 3M Innovative Properties Company Isoxazole, dihydroisoxazole, and oxadiazole substituted imidazo ring compounds and method
US8034938B2 (en) 2004-12-30 2011-10-11 3M Innovative Properties Company Substituted chiral fused [1,2]imidazo[4,5-c] ring compounds
US7943609B2 (en) 2004-12-30 2011-05-17 3M Innovative Proprerties Company Chiral fused [1,2]imidazo[4,5-C] ring compounds
US9248127B2 (en) 2005-02-04 2016-02-02 3M Innovative Properties Company Aqueous gel formulations containing immune response modifiers
US10071156B2 (en) 2005-02-04 2018-09-11 3M Innovative Properties Company Aqueous gel formulations containing immune response modifiers
US7968563B2 (en) 2005-02-11 2011-06-28 3M Innovative Properties Company Oxime and hydroxylamine substituted imidazo[4,5-c] ring compounds and methods
US7943636B2 (en) 2005-04-01 2011-05-17 3M Innovative Properties Company 1-substituted pyrazolo (3,4-C) ring compounds as modulators of cytokine biosynthesis for the treatment of viral infections and neoplastic diseases
US7943610B2 (en) 2005-04-01 2011-05-17 3M Innovative Properties Company Pyrazolopyridine-1,4-diamines and analogs thereof
US8951528B2 (en) 2006-02-22 2015-02-10 3M Innovative Properties Company Immune response modifier conjugates
EP3085373A1 (en) 2006-02-22 2016-10-26 3M Innovative Properties Company Immune response modifier conjugates
US7906506B2 (en) 2006-07-12 2011-03-15 3M Innovative Properties Company Substituted chiral fused [1,2] imidazo [4,5-c] ring compounds and methods
US10005772B2 (en) 2006-12-22 2018-06-26 3M Innovative Properties Company Immune response modifier compositions and methods
US10144735B2 (en) 2006-12-22 2018-12-04 3M Innovative Properties Company Immune response modifier compositions and methods
TWI557121B (en) * 2011-04-21 2016-11-11 原真股份有限公司 Novel kinase inhibitors
US9499535B2 (en) 2011-04-21 2016-11-22 Origenis Gmbh Kinase inhibitors
EP3153180A1 (en) 2011-06-03 2017-04-12 3M Innovative Properties Company Heterobifunctional linkers with polyethylene glycol segments and immune response modifier conjugates made therefrom
US10000482B2 (en) 2012-10-19 2018-06-19 Origenis Gmbh Kinase inhibitors
US10752624B2 (en) 2012-10-19 2020-08-25 Origenis Gmbh Kinase inhibitors

Also Published As

Publication number Publication date
CA2602590A1 (en) 2006-10-12
EP1863814A1 (en) 2007-12-12
US20090163533A1 (en) 2009-06-25
US7943636B2 (en) 2011-05-17
JP2008538550A (en) 2008-10-30
AU2006232375A1 (en) 2006-10-12

Similar Documents

Publication Publication Date Title
US7943636B2 (en) 1-substituted pyrazolo (3,4-C) ring compounds as modulators of cytokine biosynthesis for the treatment of viral infections and neoplastic diseases
JP5247458B2 (en) Hydroxy and alkoxy substituted 1H-imidazoquinolines and methods
US7943610B2 (en) Pyrazolopyridine-1,4-diamines and analogs thereof
JP5128940B2 (en) Substituted imidazoquinolines, imidazopyridines, and imidazonaphthyridines
US8329721B2 (en) Hydroxy and alkoxy substituted 1H-imidazonaphthyridines and methods
WO2006026760A2 (en) 1-amino imidazo-containing compounds and methods
WO2007079086A1 (en) Pyrazoloalkyl substituted imidazo ring compounds and methods
WO2007143526A2 (en) Substituted tetrahydroimidazonaphthyridines and methods
WO2006074003A2 (en) CHIRAL FUSED [1,2]IMIDAZO[4,5-c] RING COMPOUNDS
EP1784180A2 (en) 2-amino 1h imidazo ring systems and methods
JP2009519955A (en) Substituted imidazoquinolines, imidazonaphthyridines and imidazopyridines, compositions and methods
WO2006065280A2 (en) Isoxazole, dihydroisoxazole, and oxadiazole substituted imidazo ring compounds and methods
EP1924581A1 (en) Hydroxy substituted 1h-imidazopyridines and methods
WO2006038923A2 (en) Aryl substituted imidazonaphthyridines
WO2006009826A1 (en) Aryloxy and arylalkyleneoxy substituted thiazoloquinolines and thiazolonaphthyridines
WO2006086634A2 (en) Oxime and hydroxylamine substituted imidazo[4,5-c] ring compounds and methods
WO2006086633A2 (en) Substituted fused [1,2]imidazo[4,5-c] ring compounds and methods
US9550773B2 (en) Substituted imidazoquinolines, imidazopyridines, and imidazonaphthyridines

Legal Events

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

Ref document number: 2602590

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2006232375

Country of ref document: AU

ENP Entry into the national phase

Ref document number: 2008504494

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2006749140

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2006232375

Country of ref document: AU

Date of ref document: 20060331

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: RU

WWE Wipo information: entry into national phase

Ref document number: 11887492

Country of ref document: US