US20090062328A1 - Oxime and Hydroxylamine Substituted Imidazo[4,5-c] Ring Compounds and Methods - Google Patents

Oxime and Hydroxylamine Substituted Imidazo[4,5-c] Ring Compounds and Methods Download PDF

Info

Publication number
US20090062328A1
US20090062328A1 US11/884,153 US88415306A US2009062328A1 US 20090062328 A1 US20090062328 A1 US 20090062328A1 US 88415306 A US88415306 A US 88415306A US 2009062328 A1 US2009062328 A1 US 2009062328A1
Authority
US
United States
Prior art keywords
group
alkyl
aryl
heteroaryl
alkenyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
US11/884,153
Other versions
US20100069427A9 (en
US7968563B2 (en
Inventor
Tushar A. Kshirsagar
Gregory D. Lundquist, JR.
Joseph F. Dellaria
Matthew R. Radmer
Bernhard M. Zimmermann
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
3M Innovative Properties Co
Coley Pharmaceutical Group Inc
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 US11/884,153 priority Critical patent/US7968563B2/en
Assigned to 3M INNOVATIVE PROPERTIES COMPANY & 3M COMPANY reassignment 3M INNOVATIVE PROPERTIES COMPANY & 3M COMPANY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ZIMMERMANN, BERNHARD M., RADMER, MATTHEW R., DELLARIA, JOSEPH F., JR., KSHIRSAGAR, TUSHAR A., LUNDQUIST, GREGORY D., JR.
Assigned to COLEY PHARMACEUTICAL GROUP, INC. reassignment COLEY PHARMACEUTICAL GROUP, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: 3M COMPANY & 3M INNOVATIVE PROPERTIES COMPANY
Publication of US20090062328A1 publication Critical patent/US20090062328A1/en
Publication of US20100069427A9 publication Critical patent/US20100069427A9/en
Application granted granted Critical
Publication of US7968563B2 publication Critical patent/US7968563B2/en
Expired - Fee Related legal-status Critical Current
Adjusted expiration legal-status Critical

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

  • IRMs immune response modifiers
  • 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:
  • R A , R B , X, Z, R′, and R 2-1 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 F disease in an animal by administering an effective amount of a compound of Formula I to the animal.
  • the present invention provides compounds of the following Formulas I, I, III, IV, V, VI, and VII:
  • R A , R B , R A1 , R B1 , R A2 , R B2 , R, R′, R 1 , R 2-1 , R 3 , m, n, p, G, X, and Z are as defined below; and pharmaceutically acceptable salts thereof.
  • the present invention provides a compound of Formula I:
  • Z is selected from the group consisting of:
  • X is selected from the group consisting of a bond, C 1-4 alkylene and C 2-4 alkenylene;
  • R 2 —, R 2-2 , and R 2-3 are independently selected from the group consisting of:
  • alkyl alkenyl, aryl, arylalkylenyl, heteroaryl, heteroarylalkylenyl, heterocyclyl, and heterocyclylalkylenyl, substituted by one or more substituents selected from the group consisting of:
  • R 2-4 is selected from the group consisting of hydrogen, C 1-4 alkyl, and phenyl;
  • R 2-5 is selected from the group consisting of alkyl, aryl, arylalkylenyl, heteroaryl, and heteroarylalkylenyl, each of which is unsubstituted or substituted by one or more substituents selected from the group consisting of halogen, cyano, nitro, alkoxy, dialkylamino, alkylthio, haloalkyl, haloalkoxy, alkyl, and —N 3 ;
  • Y is selected from the group consisting of:
  • R A and R B are each independently selected from the group consisting of:
  • R A and R B form a fused benzene ring or fused pyridine ring wherein the fused benzene ring or fused pyridine ring is unsubstituted or substituted by one or more R′′′ groups;
  • R A and R B form a fused cyclohexene ring or a fused tetrahydropyridine ring, wherein the fused cyclohexene or tetrahydropyridine ring is unsubstituted or substituted by one or more R groups;
  • R is selected from the group consisting of:
  • R′ is hydrogen or a non-interfering substituent
  • R′′′ is a non-interfering substitutent
  • R 6 is selected from the group consisting of ⁇ O and ⁇ S;
  • R 8 is selected from the group consisting of hydrogen, C 1-10 alkyl, C 2-10 alkenyl, C 1-10 alkoxy-C 1-10 alkylenyl, hydroxy-C 1-10 alkylenyl, heteroaryl-C 1-10 alkylenyl, and aryl-C 1-10 alkylenyl;
  • R 9 is selected from the group consisting of hydrogen and alkyl
  • R 10 is C 3-8 alkylene
  • the present invention provides a compound of Formula II:
  • Z is selected from the group consisting of:
  • X is selected from the group consisting of a bond, C 1-4 alkylene and C 2-4 alkenylene;
  • R 2-1 , R 2-2 , and R 2-3 are independently selected from the group consisting of:
  • alkyl alkenyl, aryl, arylalkylenyl, heteroaryl, heteroarylalkylenyl, heterocyclyl, and heterocyclylalkylenyl, substituted by one or more substituents selected from the group consisting of:
  • R 2-4 is selected from the group consisting of hydrogen, C 1-4 alkyl, and phenyl;
  • R 2-5 is selected from the group consisting of alkyl, aryl, arylalkylenyl, heteroaryl, and heteroarylalkylenyl, each of which is unsubstituted or substituted by one or more substituents selected from the group consisting of halogen, cyano, nitro, alkoxy, dialkylamino, alkylthio, haloalkyl, haloalkoxy, alkyl, and —N 3 ;
  • Y is selected from the group consisting of:
  • R A1 and R B1 are each independently selected from the group consisting of:
  • R 1 is selected from the group consisting of:
  • X′ is selected from the group consisting of alkylene, alkenylene, alkynylene, arylene, heteroarylene, and heterocyclylene, wherein the alkylene, alkenylene, and alkynylene groups can be optionally interrupted or terminated by arylene, heteroarylene or heterocyclylene and optionally interrupted by one or more —O— groups;
  • Y′ is selected from the group consisting of:
  • 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, halogen
  • R 5 is selected from the group consisting of:
  • R 6 is selected from the group consisting of ⁇ O and ⁇ S;
  • R 7 is C 2-7 alkylene
  • R 8 is selected from the group consisting of hydrogen, C 1-10 alkyl, C 2-10 alkenyl, C 1-10 alkoxy-C 1-10 alkylenyl, hydroxy-C 1-10 alkylenyl, heteroaryl-C 1-10 alkylenyl, and aryl-C 1-10 alkylenyl;
  • R 9 is selected from the group consisting of hydrogen and alkyl
  • R 10 is C 3-8 alkylene
  • A is selected from the group consisting of —O—, —C(O)—, —S(O) 0-2 —, —CH 2 —, and —N(-Q-R 4 )—;
  • A′ is selected from the group consisting of —O—, —S(O) 0-2 —, —N(-Q-R 4 )—, and —CH 2 —;
  • Q is selected from the group consisting of a bond, —C(R 6 )—, —C(R 6 )—C(R 6 )—, —S(O) 2 —, —C(R 6 )—N(R 8 )—W—, —S(O)—NR 8 )—, —C(R 6 )—O—, —C(R 6 )—S—, and —C(R 6 )—N(OR 9 )—;
  • V is selected from the group consisting of —C(R 6 )—, —O—C(R 6 )—, —N(R 8 )—C(R 6 )—, 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;
  • the present invention provides a compound of Formula III:
  • Z is selected from the group consisting of:
  • X is selected from the group consisting of a bond, C 1-4 alkylene and C 2-4 alkenylene;
  • R 2-1 , R 2-2 , and R 2-3 are independently selected from the group consisting of:
  • alkyl alkenyl, aryl, arylalkylenyl, heteroaryl, heteroarylalkylenyl, heterocyclyl, and heterocyclylalkylenyl, substituted by one or more substituents selected from the group consisting of:
  • R 2-4 is selected from the group consisting of hydrogen, C 1-4 alkyl, and phenyl;
  • R 2-5 is selected from the group consisting of alkyl, aryl, arylalkylenyl, heteroaryl, and heteroarylalkylenyl, each of which is substituted or substituted by one or more substituents selected from the group consisting of halogen, cyano, nitro, alkoxy, dialkylamino, alkylthio, haloalkyl, haloalkoxy, alkyl, and —N 3 ;
  • Y is selected from the group consisting of:
  • R is selected from the group consisting of:
  • n is an integer from 0 to 4.
  • R 1 is selected from the group consisting of:
  • R 3 is selected from the group consisting of:
  • n 0 or 1;
  • X′ is selected from the group consisting of alkylene, alkenylene, alkynylene, arylene, heteroarylene, and heterocyclylene, wherein the alkylene, alkenylene, and alkynylene groups can be optionally interrupted or terminated by arylene, heteroarylene or heterocyclylene and optionally interrupted by one or more —O— groups;
  • Y′ is selected from the group consisting of:
  • Z′ is a bond or —O—
  • 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, halogen
  • R 5 is selected from the group consisting of:
  • R 6 is selected from the group consisting of ⁇ O and ⁇ S;
  • R 7 is C 2-7 allylene
  • R 8 is selected from the group consisting of hydrogen, C 1-10 alkyl, C 2-10 alkenyl, C 1-10 alkoxy-C 1-10 alkylenyl, hydroxy-C 1-10 alkylenyl, heteroaryl-C 1-10 alkylenyl, and aryl-C 1-10 alkylenyl;
  • R 9 is selected from the group consisting of hydrogen and alkyl
  • R 10 is C 3-8 alkylene
  • A is selected from the group consisting of —O—, —C(O)—, —S(O) 0-2 —, —CH 2 —, and —N(-Q-R 4 )—;
  • A′ is selected from the group consisting of —O—, —S(O) 0-2 —, —N(-Q-R 4 )—, and —CH 2 —;
  • Q is selected from the group consisting of a bond, —C(R 6 )—, —C(R 6 )—C(R 6 )—, —S(O) 2 —, —C(R 6 )—N(R 8 )—W—, —S(O) 2 —N(R 8 )—, —C(R 6 )—O—, —C(R 6 )—S—, and —C(R 6 )—N(OR 9 )—;
  • V is selected from the group consisting of —C(R 6 )—, —O—C(R 6 )—, —N(R 8 )—C(R 6 )—, 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;
  • the present invention provides a compound of Formula IV:
  • Z is selected from the group consisting of:
  • X is selected from the group consisting of a bond, C 1-4 alkylene and C 2-4 alkenylene;
  • R 2-1 , R 2-2 , and R 2-3 are independently selected from the group consisting of:
  • alkyl alkenyl, aryl, arylalkylenyl, heteroaryl, heteroarylalkylenyl, heterocyclyl, and heterocyclylalkylenyl, substituted by one or more substituents selected from the group consisting of:
  • R 2-4 is selected from the group consisting of hydrogen, C 1-4 alkyl, and phenyl;
  • R 2-5 is selected from the group consisting of alkyl, aryl, arylalkylenyl, heteroaryl, and heteroarylalkylenyl, each of which is unsubstituted or substituted by one or more substituents selected from the group consisting of halogen, cyano, nitro, alkoxy, dialkylamino, alkylthio, haloalkyl, haloalkoxy, alkyl, and —N 3 ;
  • Y is selected from the group consisting of:
  • R is selected from the group consisting of:
  • n is an integer from 0 to 4.
  • R 1 is selected from the group consisting of:
  • X′ is selected from the group consisting of alkylene, alkenylene, alkynylene, arylene, heteroarylene, and heterocyclylene, wherein the alkylene, alkenylene, and alkynylene groups can be optionally interrupted or terminated by arylene, heteroarylene or heterocyclylene and optionally interrupted by one or more —O— groups;
  • Y′ is selected from the group consisting of:
  • 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, halogen
  • R 5 is selected from the group consisting of:
  • R 6 is selected from the group consisting of ⁇ O and ⁇ S;
  • R 7 is C 2-7 alkylene
  • R 8 is selected from the group consisting of hydrogen, C 1-10 alkyl, C 2-10 alkenyl, C 1-10 alkoxy-C 1-10 alkylenyl, hydroxy-C 1-10 alkylenyl, heteroaryl-C 10 alkylenyl, and aryl-C 1-10 alkylenyl;
  • R 9 is selected from the group consisting of hydrogen and alkyl
  • R 10 is C 3-8 alkylene
  • A is selected from the group consisting of —O—, —C(O)—, —S(O) 0-2 —, —CH 2 —, and —N(-Q-R 4 )—;
  • A′ is selected from the group consisting of —O—, —S(O) 0-2 —, —N(-Q-R 4 )—, and —CH 2 —;
  • Q is selected from the group consisting of a bond, —C(R 6 )—, —C(R 6 )—C(R 6 )—, —S(O) 2 —, —C(R 6 )—N(R 8 )—W—, —S(O) 2 —N(R 8 )—, —C(R 6 )—O—, —C(R 6 )—S—, and —C(R 6 )—N(OR 9 )—;
  • V is selected from the group consisting of —C(R 6 )—, —O—C(R 6 )—, —N(R 8 )—C(R 6 )—, 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;
  • the present invention provides a compound of Formula V:
  • Z is selected from the group consisting of:
  • X is selected from the group consisting of a bond, C 1-4 alkylene and C 2-4 alkenylene;
  • R 2-1 , R 2-2 , and R 2-3 are independently selected from the group consisting of:
  • alkyl alkenyl, aryl, arylalkylenyl, heteroaryl, heteroarylalkylenyl, heterocyclyl, and heterocyclylalkylenyl, substituted by one or more substituents selected from the group consisting of:
  • R 2-4 is selected from the group consisting of hydrogen, C 1-4 alkyl, and phenyl;
  • R 2-5 is selected from the group consisting of alkyl, aryl, arylalkylenyl, heteroaryl, and heteroarylalkylenyl, each of which is unsubstituted or substituted by one or more substituents selected from the group consisting of halogen, cyano, nitro, alkoxy, dialkylamino, alkylthio, haloalkyl, haloalkoxy, alkyl, and —N 3 ;
  • Y is selected from the group consisting of:
  • R is selected from the group consisting of:
  • p is an integer from 0 to 3;
  • R 1 is selected from the group consisting of:
  • R 3 is selected from the group consisting of:
  • n 0 or 1;
  • X′ is selected from the group consisting of alkylene, alkenylene, alkynylene, arylene, heteroarylene, and heterocyclylene, wherein the alkylene, alkenylene, and alkynylene groups can be optionally interrupted or terminated by arylene, heteroarylene or heterocyclylene and optionally interrupted by one or more —O— groups;
  • Y′ is selected from the group consisting of:
  • Z′ is a bond or —O—
  • R 4 is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, aryl, arylalkylenyl, aryloxyalkylenyl, alkylaryl-enyl, 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, hal
  • R 5 is selected from the group consisting of:
  • R 6 is selected from the group consisting of ⁇ O and ⁇ S;
  • R 7 is C 2-7 alkylene
  • R 8 is selected from the group consisting of hydrogen, C 1-10 allyl, C 2-10 alkenyl, Cl 1-10 alkoxy-C 1-10 alkylenyl, hydroxy-C 1-10 alkylenyl, heteroaryl-C 1-10 alkylenyl, and aryl-C 1-10 alkylenyl;
  • R 9 is selected from the group consisting of hydrogen and alkyl
  • R 10 is C 3-8 alkylene
  • A is selected from the group consisting of —O—, —C(O)—, —S(O) 0-2 —, —CH 2 —, and —N(-Q-R 4 )—;
  • A′ is selected from the group consisting of —O—, —S(O) 0-2 —, —N(-Q-R 4 )—, and —CH 2 —;
  • Q is selected from the group consisting of a bond, —C(R 6 )—, —C(R 6 )—C(R 6 )—, —S(O) 2 —, —C(R 6 )—N(R 8 )—W—, —S(O) 2 —N(R 8 )—, —C(R 6 )—O—, —C(R 6 )—S—, and —C(R 6 )—N(OR 9 )—;
  • V is selected from the group consisting of —C(R 6 )—, —O—C(R 6 )—, —N(R 8 )—C(R 6 )—, 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;
  • the present invention provides a compound of Formula VI:
  • Z is selected from the group consisting of:
  • X is selected from the group consisting of a bond, C 1-4 allylene and C 2-4 alkenylene;
  • R 2-1 , R 2-2 , and R 2-3 are independently selected from the group consisting of:
  • alkyl alkenyl, aryl, arylalkylenyl, heteroaryl, heteroarylalkylenyl, heterocyclyl, and heterocyclylalkylenyl, substituted by one or more substituents selected from the group consisting of:
  • R 2-4 is selected from the group consisting of hydrogen, C 1-4 alkyl, and phenyl;
  • R 2-5 is selected from the group consisting of alkyl, aryl, arylalkylenyl, heteroaryl, and heteroarylalkylenyl, each of which is unsubstituted or substituted by one or more substituents selected from the group consisting of halogen, cyano, nitro, alkoxy, dialkylamino, alkylthio, haloalkyl, haloalkoxy, alkyl, and —N 3 ;
  • Y is selected from the group consisting of:
  • R is selected from the group consisting of:
  • p is an integer from 0 to 3;
  • R 1 is selected from the group consisting of:
  • X′ is selected from the group consisting of alkylene, alkenylene, alkynylene, arylene, heteroarylene, and heterocyclylene, wherein the alkylene, alkenylene, and alkynylene groups can be optionally interrupted or terminated by arylene, heteroarylene or heterocyclylene and optionally interrupted by one or more —O— groups;
  • Y′ is selected from the group consisting of:
  • 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, halogen
  • R 5 is selected from the group consisting of:
  • R 6 is selected from the group consisting of ⁇ O and ⁇ S;
  • R 7 is C 2-7 alkylene
  • R 8 is selected from the group consisting of hydrogen, C 1-10 alkyl, C 2-10 alkenyl, C 1-10 alkoxy-C 1-10 alkylenyl, hydroxy-C 1-10 alkylenyl, heteroaryl-C 1-10 alkylenyl, and aryl-C 1-10 alkylenyl;
  • R 9 is selected from the group consisting of hydrogen and alkyl
  • R 10 is C 3-8 alkylene
  • A is selected from the group consisting of —O—, —C(O)—, —S(O) 0-2 —, —CH 2 —, and —N(-Q-R 4 )—;
  • A′ is selected from the group consisting of —O—, —S(O) 0-2 —, —N(-Q-R 4 )—, and —CH 2 —;
  • Q is selected from the group consisting of a bond, —C(R 6 )—, —C(R 6 )—C(R 6 )—, —S(O) 2 —, —C(R 6 )—N(R 8 )—W—, —S(O) 2 —N(R 8 )—, —C(R 6 )—O—, —C(R 6 )—S—, and —C(R 6 )—N(OR 9 )—;
  • V is selected from the group consisting of —C(R 6 )—, —O—C(R 6 )—, —N(R 8 )—C(R 6 )—, 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;
  • the present invention provides a compound of Formula VII, which is a prodrug:
  • G is selected from the group consisting of:
  • R′′ and R′′′′ are independently selected from the group consisting of C 1-10 alkyl, C 3-7 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, C 1-6 alkyl, C 1-4 alkoxy, aryl, heteroaryl, aryl-C 1-4 alkylenyl, heteroaryl-C 1-4 alkylenyl, halo-CIA alkylenyl, halo-C 1-4 alkoxy, —O—C(O)—CH 3 , —C(O)—O—CH 3 , —C(O)—NH 2 , —O—CH 2 —C(O)—NH 2 , —NH 2 , and —S(O—NH 2 , with the proviso that R′′′′ can also be hydrogen;
  • ⁇ -aminoacyl is an ⁇ -aminoacyl group derived from an ⁇ -amino acid selected from the group consisting of racemic, D-, and L-amino acids;
  • Y 1 is selected from the group consisting of hydrogen, C 1-6 alkyl, and benzyl;
  • Y 0 is selected from the group consisting of C 1-6 alkyl, carboxy-C 1-6 alkylenyl, amino-C 1-4 alkylenyl, mono-N—C 1-6 alkylamino-C 1-4 alkylenyl, and di-N,N—C 1-6 alkylamino-C 1-4 alkylenyl;
  • Y 2 is selected from the group consisting of mono-N—C 1-6 alkylamino, di-N,N—C 1-6 alkylamino, morpholin-4-yl, piperidin-1-yl, pyrrolidin-1-yl, and 4-C 4 alkylpiperazin-1-yl;
  • R A2 and R B2 are each independently selected from the group consisting of:
  • R A2 and R B2 form a fused benzene ring or fused pyridine ring wherein the fused benzene ring or fused pyridine ring is unsubstituted or substituted by one R 3 group, or one R 3 group and one R group, or one, two, three, or four R groups when on the fused benzene ring, or one, two, or three R groups when on the fused pyridine ring;
  • R A2 and R B2 form a fused cyclohexene ring or a fused tetrahydropyridine ring, wherein the fused cyclohexene or tetrahydropyridine ring is unsubstituted or substituted by one or more R groups;
  • X, Z, R 2-1 , R 1 , R, and R 3 are defined as in Formula III above;
  • the present invention provides an intermediate compound of Formula VIII:
  • R 1 is preferrably tetrahydro-2H-pyran-4-ylmethyl as shown in Formula VIIIa:
  • non-interfering means that the ability of the compound or salt, which includes a non-interfering substituent, to modulate the biosynthesis of one or more cytokines is not destroyed by the non-interfering substituent.
  • R′′′ is a non-interfering substituent.
  • Illustrative non-interfering R′ groups include those described above for R 1 .
  • Illustrative non-interfering R′′′ groups include those described above for R and R 3 .
  • alkyl 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, cyclobutyl, cyclobutylmethyl, cyclopentyl, cyclopentylmethyl, cyclohexyl, cyclohexylmethyl, adamantyl, and substituted and unsubstituted bornyl, norbornyl, and norbornenyl.
  • alkylene”, “alkenylene”, and “alkynylene” are the divalent forms of the “alkyl”, “alkenyl”, and “alkynyl” groups defined above.
  • alkylenyl”, “alkenylenyl”, and “alkynylenyl” are use when “alkylene”, “alkenylene”, and “alkynylene”, respectively, are substituted.
  • an arylalkylenyl group comprises an alkylene moiety to which an aryl group is attached.
  • 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.
  • aryl as used herein includes carbocyclic aromatic rings or ring systems. Examples of aryl groups include phenyl, naphthyl, biphenyl, fluorenyl and indenyl.
  • heteroatom refers to the atoms O, S, or N.
  • heteroaryl includes aromatic rings or ring systems that contain at least one ring heteroatom (e.g., O, S, N).
  • 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.
  • 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.
  • 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.
  • 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.
  • 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-(1H)-yl, octahydroisoquinolin-(1H)-yl, dihydroquinolin-(2H)-yl, octahydroquinolin-(2H)-yl, dihydro-1H-imi
  • 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.
  • heterocyclyl contains a nitrogen atom
  • the point of attachment of the heterocyclyl group may be the nitrogen atom
  • arylene is the divalent forms of the “aryl”, “heteroaryl”, and “heterocyclyl” groups defined above.
  • arylenyl is used when “arylene”, “heteroarylene”, and “heterocyclylene”, respectively, are substituted.
  • an alkylarylenyl group comprises an arylene moiety to which an alkyl group is attached.
  • each group is independently selected, whether explicitly stated or not.
  • each R 8 group is independently selected for the formula —N(R 8 )—C(O)—N(R 8 )— each R 8 group is independently selected.
  • each R 4 group is independently selected when an R 1 and an R 3 group both contain an R 4 group.
  • each Y′ group is independently selected, and each R 8 group is independently selected.
  • the invention is inclusive of the compounds described herein in any of their pharmaceutically acceptable forms, including isomers (e.g., diastereomers and enantiomers), salts, solvates, polymorphs, prodrugs, and the like.
  • isomers e.g., diastereomers and enantiomers
  • salts e.g., sodium bicarbonate
  • solvates e.g., sodium bicarbonate
  • polymorphs e.g., sodium bicarbonate
  • prodrugs e.g., sodium bicarbonate
  • the term “compound” includes any or all of such forms, whether explicitly stated or not (although at times, “salts” are explicitly stated).
  • 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.
  • each one of the following variables e.g., Z, X, Y, Y′, R A1 , R B1 , R, R 1 , R 2-1 , R 3 , Q, G, n, and so on
  • each one of the following variables e.g., Z, X, Y, Y′, R A1 , R B1 , R, R 1 , R 2-1 , R 3 , Q, G, n, and so on
  • each of the resulting combinations of variables is an embodiment of the present invention.
  • R′′′ is a non-interfering substituent.
  • the one or more R′′′ groups are one R 3 group, or one R 3 group and one R group, or one, two, three, or four R groups when on the fused benzene ring, or one, two, or three R groups when on the fused pyridine ring; wherein R 3 is selected from the group consisting of -Z′-R 4 , -Z′-X′—R 4 , -Z′-X′—Y′—R 4 , -Z′-X′—Y′—X′—Y′—R 4 , and -Z′-X′—R 5 .
  • R A and R B or R A2 and R B2 are each independently selected from the group consisting of hydrogen, halogen, alkyl, alkenyl, alkoxy, alkylthio, and —N(R 9 ) 2 .
  • R A and R B or R A2 and R B2 are each independently selected from the group consisting of hydrogen and alkyl.
  • R A and R B or R A2 and R B2 are both methyl.
  • R A and R B are taken together to form a fused benzene ring wherein the benzene ring is unsubstituted or substituted by one or more R′′′ groups.
  • the fused benzene ring is substituted by one or two R′′′ groups.
  • the one or two R′′′ groups are one R 3 group, or one R 3 group and one R group.
  • the fused benzene ring is unsubstituted.
  • R A2 and R B2 are taken together to form a fused benzene ring wherein the benzene ring is unsubstituted or substituted by one R 3 group, or one R 3 group and one R group. In certain of these embodiments, the fused benzene ring is unsubstituted.
  • R A and R B are taken together to form a fused pyridine ring wherein the pyridine ring is unsubstituted or substituted by one or more R′′′ groups.
  • the fused pyridine ring is substituted by one or two R′′′ groups.
  • the one or two R′′′ groups are one R 3 group, or one R 3 group and one R group.
  • the fused pyridine ring is
  • the highlighted bond is the position where the ring is fused.
  • the fused pyridine ring is unsubstituted.
  • R A2 and R B2 are taken together to form a fused pyridine ring wherein the pyridine ring is unsubstituted or substituted by one R 3 group, or one R 3 group and one R group.
  • the fused pyridine ring is
  • the highlighted bond is the position where the ring is fused.
  • the fused pyridine ring is unsubstituted.
  • R A and R B or R A2 and R B2 are taken together to form a fused cyclohexene ring wherein the fused cyclohexene ring is unsubstituted or substituted by one or more R groups.
  • the double bond in the cyclohexene ring is the position where the ring is fused. In certain of these embodiments, the fused cyclohexene ring is unsubstituted.
  • R A and R B or R A2 and R B2 are taken together to form a fused tetrahydropyridine ring, wherein the fused tetrahydropyridine ring is unsubstituted or substituted by one or more R groups.
  • the double bond in the tetrahydropyridine ring is the position where the ring is fused.
  • the tetrahydropyridine ring is
  • the highlighted bond indicates the position where the ring is fused.
  • the fused tetrahydropyridine ring is unsubstituted.
  • R′ is hydrogen or a non-interfering substituent.
  • R′ is a non-interfering substituent.
  • R′ is R 1 ; wherein R 1 is selected from the group consisting of —R 4 , —X′—R 4 , —X′—Y′—R 4 , —X′—Y′—X′—Y′—R 4 , and —X′—R 5 .
  • R A1 and R B1 are each independently selected from the group consisting of hydrogen, halogen, alkyl, alkenyl, alkoxy, alkylthio, and —N(R 9 ) 2 .
  • R A1 and R B1 are each independently selected from the group consisting of hydrogen and alkyl.
  • R A1 and R B1 are both methyl.
  • R is halogen or hydroxy.
  • R is bromine
  • n 0.
  • p 0.
  • R 3 is benzyloxy.
  • R 3 is selected from the group consisting of phenyl, pyridin-3-yl, pyridin-4-yl, 5-(hydroxymethyl)pyridin-3-yl, 2-ethoxyphenyl, 3-(morpholine-4-carbonyl)phenyl, and 3-(N,N-dimethylaminocarbonyl)phenyl.
  • Z is selected from the group consisting of —C( ⁇ N—O—R 2-2 )— and —C(R 2-4 )(—N(—OR 2-2 )—Y—R 2-3 )—.
  • Z is —C( ⁇ N—O—R 2-2 )—.
  • Z is —C(—N—O—R 2-2 )—
  • Z is —C(R 2-4 )(—N(—OR 2-2 )—Y—R 2-3 )—.
  • R 2-4 is hydrogen.
  • Y is a bond.
  • R 2-3 is selected from the group consisting of hydrogen and alkyl.
  • Y is selected from the group consisting of —C(O)—, —S(O) 2 —, and —C(O)—NH—.
  • R 2-3 is alkyl.
  • R 2-2 is selected from the group consisting of hydrogen, alkyl, arylalkylenyl, and heteroarylalkylenyl.
  • R 2-2 is hydrogen, C 1-4 alkyl, benzyl, or pyridin-2-ylmethyl.
  • R 2-1 is selected from the group consisting of hydrogen, alkyl, and aryl.
  • R 2-1 is hydrogen, C 1-4 alkyl, or phenyl.
  • X is a bond or C 1-4 alkylene.
  • X is a bond, methylene, or ethylene.
  • R 1 is selected from the group consisting of —R 4 , —X′—R 4 , —X′—Y′—R 4 , —X′—Y′—X′—Y′—R 4 , and —X′—R 5 .
  • R 1 is selected from the group consisting of alkyl, arylalkylenyl, aryloxyalkylenyl, hydroxyalkyl, dihydroxyalkyl, alkylsulfonylalkylenyl, —X′—Y′—R 4 , —X′—R 5 , and heterocyclylalkylenyl; wherein the heterocyclyl of the heterocyclylalkylenyl group is optionally substituted by one or more alkyl groups; wherein X′ is alkylene; Y′ is —N(R 8 )—C(O)—, —N(R 8 )—S(O) 2 —, —N(R 8 )—C(O)—N(R 8 )—, or
  • R 4 is alkyl, aryl, or heteroaryl; and R 5 is
  • R 1 is selected from the group consisting of 2-hydroxy-2-methylpropyl, 2-methylpropyl, propyl, ethyl, methyl, 2,3-dihydroxypropyl, 2-phenoxyethyl, 4-[(methylsulfonyl)amino]butyl, 2-methyl-2-[(methylsulfonyl)amino]propyl, 2-(acetylamino)-2-methylpropyl, 2- ⁇ [(isopropylamino)carbonyl]amino ⁇ -2-methylpropyl, 4- ⁇ [(isopropylamino)carbonyl]amino ⁇ butyl, 4-(1,1-dioxidoisothiazolidin-2-yl)butyl, tetrahydro-2H-pyran-4-ylmethyl, and (2,2-dimethyl-1,3-dioxolan-4-yl)methyl.
  • R 1 is t
  • R 1 is selected from the group consisting of (1-hydroxycyclobutyl)methyl, (1-hydroxycyclopentyl)methyl, and (1-hydroxycyclohexyl)methyl.
  • R 1 is (1-hydroxycyclobutyl)methyl.
  • R is selected from the group consisting of halogen, hydroxy, alkyl, alkenyl, haloalkyl, alkoxy, alkylthio, and —N(R 9 ) 2 .
  • R is selected from the group consisting of alkyl, alkoxy, hydroxy, halogen, and trifluoromethyl.
  • R is halogen or hydroxy
  • R is bromine
  • R 2-1 , R 2-2 , and R 2-3 are independently selected from the group consisting of hydrogen, alkyl, alkenyl, aryl, arylalkylenyl, heteroaryl, heteroarylalkylenyl, heterocyclyl, heterocyclylalkylenyl, and alkyl, alkenyl, aryl, arylalkylenyl, heteroaryl, heteroarylalkylenyl, heterocyclyl, or heterocyclylalkylenyl, substituted by one or more substituents selected from the group consisting of hydroxy, alkyl, haloalkyl, hydroxyalkyl, alkoxy, dialkylamino, —S(O) 0-2 —R 2-5 , —NH—S(O) 2 —R 2-5 , haloalkoxy, halogen, cyano, nitro, —N 3 , aryl, heteroaryl, heterocyclyl, aryl
  • R 2-1 is selected from the group consisting of hydrogen, alkyl, and aryl.
  • R 2-1 is hydrogen, C 1-4 alkyl, or phenyl.
  • R 2-1 is hydrogen
  • R 2-2 is selected from the group consisting of hydrogen, alkyl, arylalkylenyl, and heteroarylalkylenyl.
  • R 2-2 is hydrogen, C 1-4 alkyl, benzyl, or pyridin-2-ylmethyl.
  • R 2-2 is C 1-10 alkyl.
  • R 2-2 is methyl
  • R 2-2 is hydrogen
  • R 2-3 is selected from the group consisting of hydrogen and alkyl.
  • R 2-3 is alkyl
  • R 2-3 is hydrogen or methyl.
  • R 2-4 is selected from the group consisting of hydrogen, C 1-4 alkyl, and phenyl.
  • R 2-4 is hydrogen
  • R 2-5 is selected from the group consisting of alkyl, aryl, arylalkylenyl, heteroaryl, and heteroarylalkylenyl, each of which is unsubstituted or substituted by one or more substituents selected from the group consisting of halogen, cyano, nitro, alkoxy, dialkylamino, alkylthio, haloalkyl, haloalkoxy, alkyl, and —N 3 .
  • R 2-5 is hydrogen or alkyl.
  • R 2-5 is hydrogen or C 1-4 alkyl.
  • R 3 is selected from the group consisting of -Z′-R 4 , -Z′-X′—R 4 , -Z′-X′—Y′—R 4 , -Z′-X′—Y′—X′—Y′—R 4 , and -Z′-X′—R 5 .
  • R 3 is selected from the group consisting of -Z′-R 4 , -Z′-X′—Y′—R 4 , and -Z′-X′—R 5 .
  • R 3 is -Z′-R 4 .
  • R 3 is -Z′-X′—Y′—R 4 .
  • R 3 is -Z′-X′—R 5 .
  • R 3 is selected from the group consisting of phenyl, pyridin-3-yl, pyridin-4-yl, 5-(hydroxymethyl)pyridin-3-yl, 2-ethoxyphenyl, 3-(morpholine-4-carbonyl)phenyl, and 3-(NAN-dimethylaminocarbonyl)phenyl.
  • —R 3 is at the 7-position.
  • 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 4 is alkyl, aryl, or heteroaryl.
  • R 4 is hydrogen or alkyl.
  • R 4 is C 1-4 alkyl.
  • R 5 is selected from the group consisting of:
  • R 5 is selected from the group consisting of:
  • R 5 is
  • R 5 is
  • V is —C(O)—, and A is —O—.
  • R 6 is selected from the group consisting of ⁇ O and —S.
  • R 6 is ⁇ O.
  • R 7 is C 2-7 alkylene.
  • R 7 is C 2-4 alkylene.
  • R 8 is selected from the group consisting of hydrogen, C 1-10 alkyl, C 2-10 alkenyl, C 1-10 alkoxy-C 1-10 alkylenyl, hydroxy-C 1-10 alkylenyl, heteroaryl-C 1-10 alkylenyl, and aryl-C 1-10 alkylenyl.
  • R 8 is selected from the group consisting of hydrogen, C 1-4 alkyl, and C 1-4 alkoxyC 1-4 alkylenyl.
  • R 8 is hydrogen or C 1-4 alkyl.
  • R 8 is hydrogen
  • R 9 is selected from the group consisting of hydrogen and alkyl.
  • R 10 is C 3-48 alkylene.
  • R 10 is C 4-4 alkylene.
  • A is selected from the group consisting of —O—, —C(O)—, —S(O) 0-2 —, —CH 2 —, and —N(-Q-R 4 )—.
  • A is —O—, —CH 2 —, or —S(O) 2 —.
  • A is —O— or —S(O) 2 —.
  • A is —O—.
  • A′ is selected from the group consisting of —O—, —S(O) 0-2 —, —N(-Q-R 4 )—, and —CH 2 —.
  • A′ is selected from the group consisting of —CH 2 —, —S(O) 2 —, and —O—.
  • A′ is —N(-Q-R 4 )—.
  • A′ is —O—.
  • G is selected from the group consisting of —C(O)—R′′, ⁇ -aminoacyl, ⁇ -aminoacyl- ⁇ -aminoacyl, —C(O)—O—R′′, —C(O)—N(R′′′′)R′′, —C( ⁇ NY 1 )—R′′, —CH(OH)—C(O)—OY 1 , —CH(OC 1-4 alkyl)Y 0 , —CH 2 Y 2 , and —CH(CH 3 )Y 2 .
  • R′′ and R′′′′ are independently selected from the group consisting of C 1-10 alkyl, C 3-7 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, C 1-6 alkyl, C 1-4 alkoxy, aryl, heteroaryl, aryl-C 1-4 alkylenyl, heteroaryl-C 1-4 alkylenyl, halo-C 1-4 alkylenyl, halo-C 1-4 alkoxy, —O—C(O)—CH 3 , —C(O)—O—CH 3 , —C(O)—NH 2 , —O—CH 2 —C(O)—NH 2 , —NH 2 , and —S(O) 2 —NH 2 , with the proviso that R′′′′ can
  • G is selected from the group consisting of —C(O)—R′′, ⁇ -aminoacyl, and —C(O)—O—R′′.
  • G is selected from the group consisting of —C(O)—R′′, ⁇ -amino-C 2-11 acyl, and —C(O)—O—R′′.
  • ⁇ -Amino-C 2-11 acyl includes ⁇ -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.
  • R′′ contains one to ten carbon atoms.
  • ⁇ -aminoacyl is an ⁇ -aminoacyl group derived from a naturally occurring ⁇ -amino acid selected from the group consisting of racemic, D-, and L-amino acids.
  • ⁇ -aminoacyl is an ⁇ -aminoacyl group derived from an ⁇ -amino acid found in proteins, wherein the ⁇ -amino acid is selected from the group consisting of racemic, D-, and L-amino acids.
  • Q is selected from the group consisting of a bond, —C(R 6 )—, —C(R 6 )—C(R 6 )—, —S(O) 2 —, —C(R 6 )—N(R 8 )—W—, —S(O) 2 —N(R 8 )—, —C(R 6 )—O—, —C(R 6 )—S—, and —C(R 6 )—N(OR 9 )—.
  • Q is selected from the group consisting of a bond, —C(O)—, —S(O) 2 —, and —C(R 6 )—N(R 8 )—.
  • Q is —C(O)—.
  • V is selected from the group consisting of —C(R 6 )—, —O—C(R 6 )—, —N(R 8 )—C(R 6 )—, and —S(O) 2 —.
  • V is selected from the group consisting of —C(O)— and —N(R 8 )—C(O)—.
  • V is —C(O)—.
  • W is selected from the group consisting of a bond, —C(O)—, and —S(O) 2 —.
  • W is a bond
  • X is selected from the group consisting of a bond, C 1-4 alkylene and C 2-4 alkenylene.
  • X is a bond or C 1-4 alkylene.
  • X is a bond, methylene, or ethylene.
  • X′ is selected from the group consisting of alkylene, alkenylene, alkynylene, arylene, heteroarylene, and heterocyclylene, wherein the alkylene, alkenylene, and alkynylene groups can be optionally interrupted or terminated by arylene, heteroarylene or heterocyclylene and optionally interrupted by one or more —O— groups.
  • X′ is alkylene
  • X′ is phenylene
  • Y is selected from the group consisting of a bond, —C(R 6 )—, —S(O) 2 —, —S(O) 2 —N(R 8 )—,
  • Y is selected from the group consisting of —C(O)—, —S(O) 2 —, —S(O) 2 —N(R 8 )—, —C(O)—O—, and —C(O)—N(R 8 )—.
  • Y is selected from the group consisting of —C(O)—, —S(O) 2 —, and —C(O)—N(H)—.
  • Y is a bond
  • Y′ is selected from the group consisting of —O—, —S(O) 0-2 —, —S(O) 2 —N(R 8 )—, —C(R 6 )—, —C(R 6 )—O—, —O—C(R 6 )—, —O—C(O)—O—, —N(R 8 )-Q-, —C(R 6 )—N(R 8 )—, —O—C(R 6 )—N(R 8 )—, —C(R 6 )—N(OR 9 )—, —O—N(R 8 )-Q-, —O—N ⁇ C(R 4 )—, —C( ⁇ N—O—R 8 )—, —CH(—N(—O—R 8 )-Q-R 4 )—,
  • Y′ is —N(R 8 )—C(O)—, —N(R 8 )—S(O) 2 —, —N(R 8 )—C(O)—N(R 8 )—, or
  • Z′ is a bond or —O—.
  • Z′ is a bond
  • Z′ is —O—.
  • a and b are independently integers from 1 to 6 with the proviso that a+b is ⁇ 7.
  • a and b are each 2.
  • n is an integer form 0 to 4.
  • n is 0 or 1.
  • n 0.
  • p is an integer form 0 to 3.
  • p is 0 or 1.
  • p is 0.
  • n is 0 or 1.
  • n is 0 or 1; with the proviso that when m is 1, then p is 0 or 1.
  • m is 0.
  • n 1
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of a compound or salt of any one of the above embodiments of Formulas I, II, III, IV, V, VI, VII, and VIIIa, and a pharmaceutically acceptable carrier.
  • 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 the above embodiments of Formulas I, II, III, IV, V, VI, VII, and VIIIa, or a pharmaceutical composition comprising an effective amount of a compound or salt of any one of the above embodiments of Formulas I, II, III, IV, V, VI, VII, and VIIIa to the animal.
  • the cytokine is selected from the group consisting of IFN- ⁇ , TNF- ⁇ , IL-6, IL-10, and IL-12.
  • the cytokine is IFN- ⁇ or TNF- ⁇ .
  • the cytokine is IFN- ⁇ .
  • 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 the above embodiments of Formulas I, II, III, IV, V, VI, VII, and VIIIa, or a pharmaceutical composition comprising a therapeutically effective amount of a compound or salt of any one of the above embodiments of Formulas I, II, III, IV, V, VI, VII, and VIIIa to the animal.
  • 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 the above embodiments of Formulas I, II, III, IV, V, VI, VII, and VIIIa, or a pharmaceutical composition comprising a therapeutically effective amount of a compound or salt of any one of the above embodiments of Formulas I, II, III, IV, V, VI, VII, and VIIIa to the animal.
  • 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, Wis., 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.
  • reaction schemes depicted below provide potential routes for synthesizing the compounds of the present invention as well as key intermediates.
  • EXAMPLES section below For more detailed description of the individual reaction steps, see the EXAMPLES section below.
  • Other synthetic routes may be used to synthesize the compounds of the invention.
  • 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.
  • 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.
  • Suitable amino protecting groups include acetyl, trifluoroacetyl, tert-butoxycarbonyl (Boc), benzyloxycarbonyl, and 9-fluorenylmethoxycarbonyl (Fmoc).
  • Suitable hydroxy protecting groups include acetyl and silyl groups such as the tert-butyl dimethylsilyl group.
  • 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.
  • HPLC high performance liquid chromatography
  • column chromatography using common absorbents such as silica gel
  • thin layer chromatography such as silica gel
  • recrystallization i.e., liquid-liquid extraction techniques.
  • Step (1) of Reaction Scheme I a quinoline-3,4-diamine of Formula X is reacted with a carboxylic acid equivalent, which is selected such that it will provide the desired —X—CH 2 -Hal substituent in a 1H-imidazo[4,5-c]quinoline of Formula XI.
  • Suitable carboxylic acid equivalents include ortho esters, acid halides, and imidates or salts thereof.
  • the reaction is conveniently carried out by combining a quinoline-3,4-diamine of Formula X with the imidate in a suitable solvent such 1,2-dichloroethane or chloroform.
  • a suitable solvent such 1,2-dichloroethane or chloroform.
  • the reaction can be carried out at an elevated temperature such as 80° C. or the reflux temperature of the solvent.
  • the product can be isolated by conventional methods.
  • Ethyl chloroacetimidate hydrochloride which can be used to provide a compound of Formula XI in which X is a bond, is a known compound that can be prepared according to the literature procedure: Stillings, M. R. et al., J. Med. Chem., 29, pp. 2280-2284 (1986).
  • the reaction is conveniently carried out by adding the acid halide to a solution of a quinoline-3,4-diamine of Formula X in a suitable solvent such as dichloromethane or 1,2-dichloroethane in the presence of a tertiary amine such as triethylamine.
  • a suitable solvent such as dichloromethane or 1,2-dichloroethane
  • the reaction can be carried out at ambient temperature or at a sub-ambient temperature.
  • the product can be isolated by conventional methods.
  • the reaction with an acid halide of formula Hal-CH 2 —X—C(O)Cl or Hal-CH 2 —X—C(O)Br may be carried out in two parts, which include (i) adding the acid halide to a solution of a quinoline-3,4-diamine of Formula X in a suitable solvent such as dichloromethane or 1,2-dichloroethane optionally in the presence of a tertiary amine such as triethylamine to afford an amide intermediate and (ii) cyclizing to provide a 1H-imidazo[4,5-c]quinoline of Formula XI.
  • the amide intermediate from part (i) can be optionally isolated using conventional techniques.
  • the cyclization in part (ii) may be carried out by heating the amide intermediate from part (i) in a suitable solvent such as toluene.
  • a suitable solvent such as toluene.
  • the cyclization in part (ii) can also be carried out in the presence of a base such as triethylamine.
  • step (2) of Reaction Scheme I a 1H-imidazo[4,5-c]quinoline of Formula XI is oxidized to provide an N-oxide of Formula XII using a conventional oxidizing agent that is capable of forming N-oxides.
  • the reaction can be carried out by treating a solution of a compound of Formula XI in a suitable solvent such as chloroform or dichloromethane with 3-chloroperoxybenzoic acid at room temperature, and the product can be isolated by conventional methods.
  • Step (3) of Reaction Scheme I a 1H-imidazo[4,5-c]quinoline-5N-oxide of Formula XII is aminated to provide a 1H-imidazo[4,5-c]quinolin-4-amine of Formula XIII.
  • Step (3) involves the activation of an N-oxide of Formula XII by conversion to an ester and then reacting the ester with an aminating agent.
  • Suitable activating agents include alkyl- or arylsulfonyl chlorides such as benzenesulfonyl chloride, methanesulfonyl chloride, or p-toluenesulfonyl chloride.
  • Suitable aminating agents include ammonia, in the form of ammonium hydroxide, for example, and ammonium salts such as ammonium carbonate, ammonium bicarbonate, and ammonium phosphate.
  • the reaction is conveniently carried out by adding ammonium hydroxide to a solution of the N-oxide of Formula XII in a suitable solvent such as dichloromethane or chloroform and then adding p-toluenesulfonyl chloride.
  • the reaction can be carried out at room temperature, and the product or a pharmaceutically acceptable salt thereof can be isolated from the reaction mixture using conventional methods.
  • the oxidation and amination can be carried out as a one-pot procedure without isolating the N-oxide of Formula XII by adding 3-chloroperoxybenzoic acid to a solution of a compound of Formula XI in a solvent such as dichloromethane or chloroform and then adding ammonium hydroxide and p-toluenesulfonyl chloride.
  • a solvent such as dichloromethane or chloroform
  • ammonium hydroxide and p-toluenesulfonyl chloride can be isolated by conventional methods.
  • Some compounds of Formula XIII are known, see for example, International Publication Nos. WO2005/048933 and WO2005/048945.
  • step (4) of Reaction Scheme I the Hal group of a 1H-imidazo[4,5-c]quinolin-4-amine of Formula XIII is displaced with a hydroxylamine of formula HN(Y—R 2-3 )OR 2-2 or a salt thereof.
  • the reaction is conveniently carried out by combining a hydroxylamine salt of the formula HN(Y—R 2-3 )OR 2-2 —HCl with a compound of Formula XIII in a suitable solvent, such as N,N-dimethylformamide (DMF), in the presence of a base such as triethylamine.
  • a suitable solvent such as N,N-dimethylformamide (DMF)
  • the reaction can be carried out at room temperature or at an elevated temperature such as 50° C.
  • hydroxylamine salts of the formula HN(Y—R 2-3 )OR 22 —HCl can be obtained commercially.
  • N,O-dimethylhydroxylamine hydrochloride, methoxylamine hydrochloride, and N-methylhydroxylamine hydrochloride are commercially available compounds that can be used to make preferred compounds of Formula XIV, wherein Y is a bond.
  • Other hydroxylamine salts of the formula HN(Y—R 2-3 )OR 2-2 .HCl can be prepared using conventional synthetic methods.
  • the product of Formula XIV, a subgenus of Formulas I and III, or a pharmaceutically acceptable salt thereof can be isolated using conventional methods.
  • Step (1) of Reaction Scheme II a compound of Formula X or a salt thereof is reacted with a carboxylic acid or an equivalent thereof to provide a compound of Formula XV.
  • Suitable equivalents to carboxylic acid include acid anhydrides of Formula O[C(O)—X—CH 2 —O—P] 2 and acid chlorides of Formula Cl—C(O)—X—CH 2 —O—P.
  • the reaction is conveniently carried out by using the conditions described in step (1) of Reaction Scheme I for the reaction with acid chlorides.
  • Some compounds of Formula Cl—C(O)—X—O—P such as acetoxyacetyl chloride, methoxyacetyl chloride, and 2-methoxypropionyl chloride, are commercially available. Others can be prepared by known synthetic methods.
  • step (2) of Reaction Scheme II the protecting group of a 1H-imidazo[4,5-c]quinoline of Formula XV is removed to provide a hydroxyalkyl-substituted 1H-imidazo[4,5-c]quinoline of Formula XVI.
  • the deprotection can be carried out using a variety of methods depending on which P group is present.
  • P is —C(O)—CH 3
  • the reaction is conveniently carried out by adding lithium hydroxide monohydrate to a solution or suspension of the compound of Formula XV in a suitable solvent or solvent system such as tetrahydrofuran:methanol:water.
  • the reaction can be carried out at room temperature, and the product can be isolated by conventional methods.
  • a hydroxyalkyl-substituted 1H-imidazo[4,5-c]quinoline of Formula XVI is oxidized to an aldehyde-substituted 1H-imidazo[4,5-c]quinoline of Formula XVII using one of many conventional methods.
  • the oxidation is conveniently carried out by adding Dess-Martin periodinane, [1,1,1-tris(acetyloxy)-1,1-dihydro-1,2-benziodoxol-3(1H)-one], to a solution or suspension of a hydroxyalkyl-substituted 1H-imidazo[4,5-c]quinoline of Formula XVI in a suitable solvent such as dichloromethane.
  • the reaction can be carried out at room temperature, and the product can be isolated by conventional methods.
  • certain aldehyde-substituted 1H-imidazo[4,5-c]quinolines of Formula XVII in which X is a bond can be prepared from 1H-imidazo[4,5-c]quinolines with a hydrogen at the 2-position, many of which are known; see, for example, U.S. Pat. Nos. 4,689,338 (Gerster) and 5,268,376 (Gerster).
  • the hydrogen at the 2-position of a 1H-imidazo[4,5-c]quinoline may undergo lithiation with butyllithium, and subsequent substitution with DMF provides a compound of Formula XVII in which X is a bond.
  • the reaction is conveniently carried out in a suitable solvent such as THF at a subambient temperature such as ⁇ 78° C.
  • the product or a pharmaceutically acceptable salt thereof can be isolated by conventional methods.
  • step (4) of Reaction Scheme II an aldehyde-substituted 1H-imidazo[4,5-c]quinoline of Formula XVII is converted to an aldoxime of Formula XVIII.
  • the reaction is conveniently carried out by adding a hydroxylamine salt of the formula NH 2 OR 2-2 .HCl, optionally in a suitable solvent such as water, to a solution or suspension of a compound of Formula XVII, in a suitable solvent, such as ethanol or methanol.
  • a base such as aqueous sodium hydroxide can be added.
  • the reaction can be carried out at room temperature or at an elevated temperature such as the reflux temperature of the solvent.
  • Hydroxylamine salts of the formula NH 2 OR 2-2 .HCl can be obtained commercially or they can be prepared using conventional synthetic methods.
  • the product or a pharmaceutically acceptable salt thereof is obtained as a mixture of E and Z isomers and can be isolated using conventional methods.
  • steps (5) and (6) of Reaction Scheme II an aldoxime-substituted 1H-imidazo[4,5-c]quinoline of Formula XVIII is first oxidized to an N-oxide of Formula XIX, which is then aminated to provide a compound of Formula XX, which is a subgenus of Formulas I and III.
  • Steps (5) and (6) of Reaction Scheme II can be carried out according to the methods described in steps (2) and (3) of Reaction Scheme I, and the product can be isolated by conventional methods.
  • step (7) of Reaction Scheme II an aldoxime-substituted 1H-imidazo[4,5-c]quinolin-4-amine of Formula XX is treated with a Grignard reagent of the formula R 2-1 MgHalide to form a hydroxylamine of Formula XXI, a subgenus of Formulas I and III.
  • a Grignard reagent of the formula R 2-1 MgHalide
  • the reaction is conveniently carried out by adding a solution of two equivalents of the Grignard reagent to a solution of the compound of Formula XX in a suitable solvent such as THF.
  • the reaction can be carried out at room temperature, and the product can be isolated using conventional methods.
  • an oxime of Formula XX can be treated with a hydride reducing agent.
  • the reduction is conveniently carried out by treating an oxime of Formula XX with excess sodium cyanoborohydride in a suitable solvent or solvent mixture such as methanol/acetic acid.
  • the reaction can be carried out at ambient temperature.
  • the product or a pharmaceutically acceptable salt thereof can be isolated using conventional methods.
  • step (8) of Reaction Scheme II a hydroxylamine of Formula XXI is converted to a compound of Formula XXII, a subgenus of Formulas I and III.
  • Step (8) is carried out using conventional methods.
  • sulfonamides of Formula XII (Y is —S(O) 2 —) can be prepared by reacting a compound of Formula XXI with a sulfonyl chloride of formula R 2-3 S(O) 2 Cl or a sulfonic anhydride of Formula [R 2-3 S(O) 2 ] 2 O.
  • the reaction can be carried out at room temperature in an inert solvent such as chloroform, dichloromethane, or N,N-dimethylacetamide (DMA) by adding the sulfonyl chloride or sulfonic anhydride to a compound of Formula XI in the presence of a base such as N,N-diisopropylethylamine, triethylamine, or pyridine.
  • an inert solvent such as chloroform, dichloromethane, or N,N-dimethylacetamide (DMA)
  • the product or a pharmaceutically acceptable salt thereof can be isolated using conventional methods.
  • Amides of Formula XXII (Y is —C(O)—) can be prepared from hydroxylamines of Formula XXI using conventional methods.
  • a compound of Formula XXI can be reacted with an acid chloride of formula R 2-3 C(O)Cl.
  • the reaction can be carried out by adding the acid chloride to a solution of a compound of Formula XXI in a suitable solvent such as chloroform or DMA, optionally in the presence of a base such as N,N-diisopropylethylamine, triethylamine, or pyridine, at ambient temperature.
  • a base such as N,N-diisopropylethylamine, triethylamine, or pyridine
  • Ureas and thioureas of Formula XXII (Y is —C(O)—N(R 8 )—, —C(S)—N(R 8 )—, —C(O)—N(R 8 )—S(O) 2 —, —C(O)—N(R 8 )—C(O)—, —C(S)—N(R 8 )—C(O)—, or
  • a compound of Formula VI can be reacted with an isocyanate of formula R 2-3 N ⁇ C ⁇ O.
  • the reaction can be carried out by adding the isocyanate to a solution of a compound of Formula XXI in a suitable solvent such as chloroform or DMA, optionally in the presence of a base such as N,N-diisopropylethylamine, or triethylamine, at room temperature.
  • a compound of Formula XXI can be reacted with a thioisocyanate of formula R 2-3 N ⁇ C ⁇ S, a sulfonyl isocyanate of formula R 2-3 S(O) 2 N ⁇ C ⁇ O or a carbamoyl chloride of formula R 2-3 NC(O)Cl or
  • the product or a pharmaceutically acceptable salt thereof can be isolated using conventional methods.
  • Reaction Scheme II can be modified after step (3) to introduce a R 2-4 group that is other than hydrogen.
  • an aldehyde-substituted 1H-imidazo[4,5-c]quinoline of Formula XVII is treated with a Grignard reagent of the formula R 2-4 MgHalide, which adds to the aldehyde to form a secondary alcohol.
  • a Grignard reagent of the formula R 2-4 MgHalide which adds to the aldehyde to form a secondary alcohol.
  • Several Grignard reagents are commercially available; others can be prepared using known synthetic methods.
  • the reaction is conveniently carried out by adding a solution of the Grignard reagent to a solution of the compound of Formula XVII in a suitable solvent such as THF.
  • the reaction can be carried out at room temperature, and the product can be isolated using conventional methods.
  • the secondary alcohol is then oxidized to a ketone using conventional methods.
  • the reaction is conveniently carried out using Dess-Martin periodinane under the conditions described in step (3) of Reaction Scheme II.
  • the reaction may also be carried out under Swern conditions by adding the secondary alcohol followed by triethylamine to a mixture of oxalyl chloride and dimethylsulfoxide in a suitable solvent such as dichloromethane.
  • the reaction can be carried out at a sub-ambient temperature, and the product can be isolated by conventional methods.
  • Steps (4) through (8) of Reaction Scheme II can then be carried out to provide a hydroxylamine of the invention with an R 2-4 group that is other than hydrogen.
  • D can be —OH, —Cl, Br, or a leaving group prepared using conventional hydroxy activation chemistry, such as employing N-hydroxysuccinimide as an activating agent.
  • Ketals of this formula are readily prepared from esters of formula alkyl-O—C(O)—X—C(O)—R 2-1 using conventional methods.
  • the ketone can be converted to a ketal by heating with ethylene glycol in the presence of pyridinium p-toluenesulfonate in a suitable solvent such as toluene.
  • the carboxyl group can then be activated by first hydrolyzing the ester under basic conditions, for example with sodium hydroxide in water and a lower alcohol, and then reacting with N-hydroxysuccinimide in the presence of 4-methylmorpholine and 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride in a suitable solvent such as dichloromethane.
  • the reaction shown in step (1) of Reaction Scheme III can be carried out according to the methods described for reaction with acid chlorides in step (1) of Reaction Scheme I, or it can be conveniently carried out by heating a quinoline-3,4-diamine of Formula X with a ketal shown above in a suitable solvent such as pyridine.
  • the reaction can be run at the reflux temperature of the solvent, and the product of Formula XXIII can be isolated by conventional methods.
  • step (2) of Reaction Scheme III a 1H-imidazo[4,5-c]quinoline of Formula XXIII is converted to the N-oxide of Formula XXIV using the method described in step (2) of Reaction Scheme I.
  • step (3) of Reaction Scheme III the N-oxide of Formula XXIV is aminated to afford the compound of Formula XXV using one of the methods described in step (3) of Reaction Scheme I.
  • a ketal of Formula XXV is converted to a ketone of Formula XXVI by acid-catalyzed hydrolysis.
  • the reaction is conveniently carried out by adding a strong acid, such as hydrochloric acid, to a ketal of Formula XXV.
  • the reaction may be carried out at room temperature in a suitable solvent such as water, and the product can be isolated by conventional methods.
  • step (5) of Reaction Scheme III a ketone-substituted 1H-imidazo[4,5-c]quinolin-4-amine of Formula XXVI is converted to an oxime of Formula XXVII.
  • the reaction is conveniently carried out as described in step (4) of Reaction Scheme II, and the product or a pharmaceutically acceptable salt thereof can be isolated by conventional methods.
  • Reaction Schemes I through III may be carried out using a [1,5]naphthyridine-3,4-diamine instead of a quinoline-3,4-diamine of Formula X as the starting material to prepare 1H-imidazo[4,5-c][1,5]naphthyridines of the invention.
  • Several [1,5]naphthyridine-3,4-diamines and their preparation are known; see, for example, U.S. Pat. Nos. 6,194,425 (Gerster) and 6,518,280 (Gerster).
  • R 2-1 , and Z are as defined above;
  • X b is selected from the group consisting of a bond and C 1-4 alkylene;
  • R b is selected from the group consisting of hydroxy, alkyl, alkoxy, —N(R 9 ) 2 ;
  • n is 0 to 4;
  • R 1b is a subset of R 1 as defined above that does not include those substituents that one skilled in the art would recognize as being susceptible to reduction under the acidic hydrogenation conditions of the reaction.
  • susceptible groups include, for example, alkenyl, alkynyl, and aryl groups and groups bearing nitro substituents.
  • Compounds of Formula XXVIII can be prepared by the oxidation and amination of a compound of Formula XVI according to steps (2) and (3) of Reaction Scheme I.
  • step (1) of Reaction Scheme IV a compound of Formula XXVIII is reduced to a 6,7,8,9-tetrahydro compound of Formula XXIX.
  • the reaction is conveniently carried out under hetereogeneous hydrogenation conditions by adding platinum (IV) oxide to a solution of the compound of Formula XXVIII in trifluoroacetic acid and placing the reaction under hydrogen pressure.
  • the reaction can be carried out on a Parr apparatus at room temperature.
  • the product or a pharmaceutically acceptable salt thereof can be isolated by conventional methods.
  • Steps (2) through (6) of Reaction Scheme IV can then be used to convert a compound of Formula XXIX to a compound of Formula IVb, a subgenus of Formulas I and IV.
  • Steps (2) through (6) can be carried out, for example, according to steps (3), (4), (7), and (8) of Reaction Scheme II.
  • Compounds of Formula IVb can also be made by treating a compound of Formula XXIX with thionyl chloride under conventional conditions to provide a chloroalkyl-substituted 6,7,8,9-tetrahydro compound, which can then be treated according to step (4) of Reaction Scheme I.
  • the product of Formula IVb or a pharmaceutically acceptable salt thereof can be isolated by conventional methods. 6,7,8,9-Tetrahydro-1H-imidazo[4,5-c][1,5]naphthyridin-4-amines can also be prepared using this Reaction Scheme.
  • Compounds of the invention can be prepared according to Reaction Scheme V, wherein R, R 1 , R 2-1 , X, and Z are as defined above; n is 0 or 1; hal is —Br or —I; and R 3a and R 3b are as defined below.
  • Compounds of Formula XXX, a subgenus of Formulas I and III can be prepared according to the methods described in Reaction Scheme I, II, or III beginning with a compound of Formula X, wherein one of the R groups is —Br, or —I.
  • These halogen-substituted quinolines are known or can be prepared by known methods; see, for example, U.S. Patent Application Publication No. US 2004/0147543 (Hays et al.) and the references cited therein.
  • a halogen-substituted 1H-imidazo[4,5-c]quinolin-4-amine of Formula X) can undergo known palladium-catalyzed coupling reactions such as the Suzuki coupling and the Heck reaction.
  • a halogen-substituted compound of Formula XXX undergoes Suzuki coupling with a boronic acid of Formula R 3a —B(OH) 2 , an anhydride thereof, or a boronic acid ester of Formula R 3a —B(O-alkyl) 2 , wherein R 3a is —R 4b , —X′ a —R 4 , —X′ b —Y′—R 4 , or —X′ b —R 5 ; where X′ a is alkenylene; X′ b is arylene, heteroarylene, and alkenylene interrupted or terminated by arylene or heteroarylene; R 4b is aryl or heteroaryl where the aryl or heteroaryl groups can be unsubstituted or substituted as defined in R 4 above; and R 4 , R 5 , and Y′ are as defined above.
  • the Suzuki coupling is conveniently carried out by combining a compound of Formula XXX with a boronic acid or an ester or anhydride thereof in the presence of palladium (II) acetate, triphenylphosphine, and a base such as sodium carbonate in a suitable solvent such as n-propanol or solvent mixture such as n-propanol/water.
  • the reaction can be carried out at an elevated temperature (e.g., 80-100° C.).
  • Many boronic acids of Formula R 3a —B(OH) 2 , anhydrides thereof, and boronic acid esters of Formula R 3a —B(O-alkyl) 2 are commercially available; others can be readily prepared using known synthetic methods. See, for example, Li, W. et al, J. Org. Chem., 67, 5394-5397 (2002).
  • the product of Formula IIIb, a subgenus of Formulas I and III, or a pharmaceutically acceptable salt thereof can be isolated by
  • the Heck reaction can also be used in step (1) of Reaction Scheme V to provide compounds of Formula IIIb, wherein R 3a is —X′ a —R 4b or —X′ a —Y′—R 4 , wherein X′ a , Y′, R 4b , and R 4 are as defined above.
  • the Heck reaction is carried out by coupling a compound of Formula XXX with a compound of the Formula H 2 C ⁇ C(H)—R 4b or H 2 C ⁇ C(H)—Y′—R 4 .
  • Several of these vinyl-substituted compounds are commercially available; others can be prepared by known methods.
  • the reaction is conveniently carried out by combining the compound of Formula XXX and the vinyl-substituted compound in the presence of palladium (II) acetate, triphenylphosphine or tri-ortho-tolylphosphine, and a base such as triethylamine in a suitable solvent such as acetonitrile or toluene.
  • the reaction can be carried out at an elevated temperature such as 100° C.-120° C. under an inert atmosphere.
  • the product of Formula IIIb or a pharmaceutically acceptable salt thereof can be isolated using conventional methods.
  • Compounds of Formula IIIb wherein R 3a is —X′ c —R 4 , X′ c is alkynylene, and R 4 is as defined above, can also be prepared by palladium catalyzed coupling reactions such as the Stille coupling or Sonogashira coupling. These reactions are carried out by coupling a compound of Formula XXX with a compound of the Formula (alkyl) 3 Sn—C ⁇ C—R 4 , (alkyl) 3 Si—C ⁇ C—R 4 , or H—C ⁇ C—R 4 .
  • Some compounds of Formula IIIb prepared as described above by palladium-mediated coupling reactions wherein R 3a is —X′ a —R 4 , —X′ a —Y′—R 4 , —X′ b2 —Y′—R 4 , —X′ b2 —R 5 , or —X′ c —R 4 , where X′ b2 is alkenylene interrupted or terminated by arylene or heteroarylene, and X′ a , X′ c , Y′, R 4 , and R 5 are as defined above, can undergo reduction of the alkenylene or alkynylene group present in step (2) of Reaction Scheme V to provide compounds of Formula IIIc wherein R 3b is —X′ d —R 4 , —X′ d —Y′—R 4 , —X′ e —Y′—R 4 , or —X′ e —R 5 , where X′ d is alkylene;
  • the reduction can be carried out by hydrogenation using a conventional heterogeneous hydrogenation catalyst such as palladium on carbon.
  • a conventional heterogeneous hydrogenation catalyst such as palladium on carbon.
  • the reaction can conveniently be carried out on a Parr apparatus in a suitable solvent such as ethanol, methanol, or mixtures thereof.
  • a suitable solvent such as ethanol, methanol, or mixtures thereof.
  • the product of Formula IIIc, a subgenus of Formulas I and III, or a pharmaceutically acceptable salt thereof can be isolated using conventional methods.
  • compounds of the invention are prepared according to Reaction Scheme VI, where R 1 , R 2-1 , R 2-2 , R A1 , R B1 , and X are as defined above, and Z a is —C(R 2-4 )(—N(—OR 2-2 )—Y—R 2-3 )— and Ph is phenyl.
  • Steps (1) through (4) of Reaction Scheme VI can be carried out as described in steps (1) through (4) of Reaction Scheme II starting with a compound of Formula XXI.
  • Many tetrazolo[1,5-a]pyridines of Formula XXXI are known; others can be prepared by known methods. See, for example, PCT Publication Nos. WO 2004/110991 (Lindstrom et al.), WO 2004/110992 (Lindstrom et al.), and U.S. Pat. No. 6,797,718 (Dellaria et al.).
  • step (5) of Reaction Scheme VI the tetrazolo ring is removed from a 7H-imidazo[4,5-c]tetrazolo[1,5-a]pyridine of Formula XXII by reaction with triphenylphosphine to form an N-triphenylphosphinyl intermediate of Formula XIII.
  • the reaction with triphenylphosphine can be run in a suitable solvent such as toluene or 1,2-dichlorobenzene under an atmosphere of nitrogen with heating, for example at the reflux temperature.
  • an N-triphenylphosphinyl intermediate of Formula XXXIII is hydrolyzed to provide an oxime-substituted 1H-imidazo[4,5-c]pyridin-4-amine of Formula XXXIV, a subgenus of Formulas I and II.
  • the hydrolysis can be carried out by general methods well known to those skilled in the art, for example, by heating in a lower alkanol or an alkanol/water solution in the presence of an acid such as trifluoroacetic acid, acetic acid, or hydrochloric acid.
  • the product can be isolated from the reaction mixture using conventional methods as the compound of Formula XXXIV or as a pharmaceutically acceptable salt thereof.
  • steps (7) and (8) of Reaction Scheme II may be used to convert a compound of Formula XXIV to a compound of Formula IIb using steps (7) and (8) of Reaction Scheme VI.
  • the product or a pharmaceutically acceptable salt thereof can be isolated by conventional methods.
  • R 1 , R 2-1 , R 2-2 , R, X, and Z a are as defined above; E is carbon (imidazoquinoline ring) or nitrogen (imidazonaphthyridine ring); n is 0 or 1; Bn is benzyl; and R 3c is —O—R 4 , —O—X′—R 4 , —O—X′—Y′—R 4 , —O—X′—Y′—X′—Y′—R 4 , or —O—X′—R 5 , where R 4 , X′, Y′, and R 5 are as defined above.
  • an aniline or aminopyridine of Formula XXXV is treated with the condensation product generated from 2,2-dimethyl-1,3-dioxane-4,6-dione (Meldrum's acid) and triethyl orthoformate to provide an imine of Formula XXXVI.
  • the reaction is conveniently carried out by adding a solution of an aniline or aminopyridine of Formula XXXV to a heated mixture of Meldrum's acid and triethyl orthoformate and heating the reaction at an elevated temperature.
  • the product can be isolated using conventional methods.
  • Many anilines and aminopyridines of Formula XXXV are commercially available; others can be prepared by known synthetic methods.
  • benzyloxypyridines of Formula XXXV can be prepared using the method of Holladay et al., Biorg. Med. Chem. Lett., 8, pp. 2797-2802, (1998).
  • step (2) of Reaction Scheme VII an imine of Formula XVI undergoes thermolysis and cyclization to provide a compound of Formula XXXVII.
  • the reaction is conveniently carried out in a medium such as DOWTHERM A heat transfer fluid at a temperature in the range of 200 to 250° C.
  • the product can be isolated using conventional methods. Isomers of the compound of Formula XV or Formula XXVII, wherein E is nitrogen and at a different position in the ring, can also be synthesized and can be used to prepare compounds of the invention.
  • step (3) of Reaction Scheme VII a compound of Formula XXXVII is nitrated under conventional nitration conditions to provide a compound of Formula XXVIII.
  • the reaction is conveniently carried out by adding nitric acid to the compound of Formula XXXVII in a suitable solvent such as propionic acid and heating the mixture at an elevated temperature.
  • the product can be isolated using conventional methods.
  • a 3-nitro[1,5]naphthyridin-4-ol or 3-nitroquinolin-4-ol of Formula XXVIII is chlorinated using conventional chlorination chemistry to provide a 4-chloro-3-nitro[1,5]naphthyridine or 4-chloro-3-nitroquinoline of Formula XXXIX.
  • the reaction is conveniently carried out by treating the compound of Formula XXXVIII with phosphorous oxychloride in a suitable solvent such as DMF.
  • the reaction can be carried out at ambient temperature or at an elevated temperature such as 100° C., and the product can be isolated using conventional methods.
  • step (5) of Reaction Scheme VII a 4-chloro-3-nitro[1,5]naphthyridine or 4-chloro-3-nitroquinoline of Formula XXI is treated with an amine of Formula R 1 —NH 2 to provide a compound of Formula XL.
  • amines of Formula R 1 —NH 2 are commercially available; others can be prepared by known synthetic methods.
  • the reaction is conveniently carried out by adding the amine of Formula R 1 —NH 2 to a solution of the 4-chloro-3-nitro[1,5]naphthyridine or 4-chloro-3-nitroquinoline of Formula XXXIX in a suitable solvent such as dichloromethane in the presence of a tertiary amine such as triethylamine.
  • a suitable solvent such as dichloromethane
  • the reaction can be carried out at ambient temperature or at a sub-ambient temperature such as, for example, 0° C.
  • the reaction product can be isolated using conventional methods.
  • a compound of Formula XL is reduced to provide a diamine of Formula XLI.
  • the reaction can be carried out by hydrogenation using a heterogeneous hydrogenation catalyst such as platinum on carbon.
  • the hydrogenation is conveniently carried out in a Parr apparatus in a suitable solvent such as toluene, methanol, acetonitrile, or ethyl acetate.
  • the reaction can be carried out at ambient temperature, and the product can be isolated using conventional methods.
  • the reduction in step (6) can be carried out using nickel boride, prepared in situ from sodium borohydride and nickel(II) chloride.
  • the reduction is conveniently carried out by adding a solution of a compound of Formula XL in a suitable solvent or solvent mixture such as dichloromethane/methanol to a mixture of excess sodium borohydride and catalytic nickel(II) chloride in methanol.
  • the reaction can be carried out at ambient temperature.
  • the product can be isolated using conventional methods.
  • Steps (7) through (12) of Reaction Scheme VII are analogous to steps (1) through (6) of Reaction Scheme II and can be carried out using the same methods.
  • step (13) of Reaction Scheme VII the benzyl group in an oxime of Formula XLII is cleaved to provide a hydroxy group.
  • the cleavage is conveniently carried out on a Parr apparatus under hydrogenolysis conditions using a suitable heterogeneous catalyst such as palladium or platinum on carbon in a solvent such as ethanol.
  • the cleavage can be carried out with an acid such as hydrogen bromide in a suitable solvent such as acetic acid at an elevated temperature, such as 65° C.
  • the product of Formula XLIII, prepared by any of these methods, or a pharmaceutically acceptable salt thereof can be isolated using conventional methods.
  • a hydroxy-substituted oxime of Formula XLIII is converted to a compound of Formula XLIV, a subgenus of Formula I, using a Williamson-type ether synthesis.
  • the reaction is effected by treating a hydroxy-substituted compound of Formula XLIII with an aryl, alkyl, or arylalkylenyl halide of Formula Halide-R 4b , Halide-alkylene-R 4 , Halide-alkylene-Y′—R 4 , or Halide-alkylene-R 5 , where R 4b is as defined above, in the presence of a base.
  • alkyl, arylalkylenyl, and aryl halides of these formulas are commercially available, including substituted benzyl bromides and chlorides, substituted or unsubstituted alkyl or arylalkylenyl bromides and chlorides, and substituted fluorobenzenes.
  • Other halides of these formulas can be prepared using conventional synthetic methods. The reaction is conveniently carried out by combining an alkyl, arylalkylenyl, or aryl halide with the hydroxy-substituted compound of Formula XLIII in a solvent such as DMF in the presence of a suitable base such as cesium carbonate.
  • catalytic tetrabutylammonium bromide can be added.
  • the reaction can be carried out at ambient temperature or at an elevated temperature, for example 65° C. or 85° C., depending on the reactivity of the halide reagent.
  • step (14) may be carried out using the Ullmann ether synthesis, in which an alkali metal aryloxide prepared from the hydroxy-substituted compound of Formula XLIII reacts with an aryl halide in the presence of copper salts, to provide a compound of Formula XLIV, where R 3b is —O—R 4b , —O—X′ f —R 4 , or —O—X′ f —Y′—R 4 , wherein X′ f is an arylene or heteroarylene, and R 4b is as defined above.
  • Numerous substituted and unsubstituted aryl halides are commercially available; others can be prepared using conventional methods.
  • the product of Formula XLIV, prepared by either of these methods, or a pharmaceutically acceptable salt thereof can be isolated using conventional methods.
  • steps (7) and (8) of Reaction Scheme II may be used to convert a compound of Formula XLIV to a compound of Formula XLV using steps (15) and (16) of Reaction Scheme VII.
  • the product or pharmaceutically acceptable salt thereof can be isolated by conventional methods.
  • naphthyridines of the invention are prepared from tetrazolo compounds of Formulas XLVI and L according to Reaction Schemes VIII and IX, wherein R 1 , R 2-1 , R 2-2 , R, n, Z a , and X are as defined above.
  • Compounds of Formula XLVI and L and synthetic routes to these compounds are known; see, for example, U.S. Pat. No. 6,194,425 (Gerster).
  • Steps (1) through (4) of Reaction Schemes VIII and IX are analogous to steps (1) through (4) of Reaction Scheme II and can be carried out using the same methods.
  • the tetrazolo group of a compound of Formula XLVII or LI can then be removed in step (5) of Reaction Scheme IX or X to provide a 1H-imidazo[4,5-c]naphthyridin-4-amine of Formula XLVIII or LII, each of which is a subgenus of Formula I.
  • the removal of the tetrazolo group can be carried out as described in steps (5) and (6) of Reaction Scheme VI or by using methods described in U.S. Pat. No. 6,194,425 (Gerster).
  • the product or a pharmaceutically acceptable salt thereof can be isolated by conventional methods.
  • steps (7) and (8) of Reaction Scheme II may be used to convert a compound of Formula XLVIII or LII to a compound of Formula XLIX or LIII using steps (6) and (7) of Reaction Scheme VIII or IX, respectively.
  • the product of Formula XLIX or LIII, each of which is a subgenus of Formula I, or a pharmaceutically acceptable salt thereof can be isolated by conventional methods.
  • step (1) of Reaction Scheme X a 2,4-dichloro-3-nitropyridine of Formula LIV is reacted with an amine of formula R 1 —NH 2 to provide a 2-chloro-3-nitropyridin-4-amine of Formula LV.
  • the reaction can be carried out using the method described in step (5) of Reaction Scheme VII.
  • Some 2,4-dichloro-3-nitropyridines of Formula LIV are known; others can be prepared using known synthetic methods. See, for example, U.S. Pat. No. 6,525,064 (Dellaria) and the references cited therein.
  • step (2) of Reaction Scheme X a 2-chloro-3-nitropyridin-4-amine of Formula LV is reduced to provide a 2-chloropyridine-3,4-diamine of Formula LVI.
  • the reduction can be carried out using the methods described in step (6) of Reaction Scheme VII.
  • step (3) of Reaction Scheme X a 2-chloropyridine-3,4-diamine of Formula LVI is reacted with a carboxylic acid or an equivalent thereof to provide a 4-chloro-1H-imidazo[4,5-c]pyridine of Formula LVII.
  • the reaction can be carried out using the method described in step (1) of Reaction Scheme II.
  • step (4) of Reaction Scheme X the protecting group of a 4-chloro-1H-imidazo[4,5-c]pyridine of Formula LVII is removed to provide a hydroxyalkyl-substituted 4-chloro-1H-imidazo[4,5-c]pyridine of Formula LVIII.
  • the deprotection can be carried out using a variety of methods depending on which P group is present.
  • P is an ethyl group
  • the reaction can be carried out by adding a solution of boron tribromide in a suitable solvent to a solution or suspension of a compound of Formula LVII in a suitable solvent such as dichloromethane.
  • the reaction can be carried out at a sub-ambient temperature such as 0° C.
  • step (5) of Reaction Scheme X a 4-chloro-1H-imidazo[4,5-c]pyridine of Formula LVIII is reacted with 4-methoxybenzylamine to provide an N-(4-methoxybenzyl)-1H-imidazo[4,5-c]pyridin-4-amine of Formula LIX.
  • the reaction can be carried out by combining a compound of Formula LVIII with excess N-(4-methoxybenzyl)amine and excess pyridine hydrochloride in a suitable solvent such as 2,2,2-trifluoroethanol and heating (150° C.) in a microwave reactor.
  • step (6) of Reaction Scheme X the 4-methoxybenzyl group is removed from an N-(4-methoxybenzyl)-1H-imidazo[4,5-c]pyridin-4-amine of Formula LIX to provide a 1H-imidazo[4,5-c]pyridin-4-amine of Formula LX.
  • the reaction can be carried out by treating a compound of Formula LIX with trifluoroacetic acid at ambient temperature.
  • a hydroxyalkyl-substituted 1H-imidazo[4,5-c]pyridin-4-amine of Formula LX is chlorinated to provide a chloroalkyl-substituted 1H-imidazo[4,5-c]pyridin-4-amine of Formula LXI.
  • the reaction can be carried out by treating a solution of a compound of Formula LX in a suitable solvent such as chloroform with thionyl chloride.
  • the reaction can be carried out at an elevated temperature such as the reflux temperature of the solvent.
  • step (8) of Reaction Scheme X the chloro group of a chloroalkyl-substituted 1H-imidazo[4,5-c]pyridin-4-amine of Formula LXI is displaced with a hydroxylamine of formula HN(Y—R 2-3 )OR 2-2 or a salt thereof to provide a 1H-imidazo[4,5-c]pyridin-4-amine of Formula LXII, which is a subgenus of Formulas I and II.
  • the reaction can be carried out using the methods described in step (4) of Reaction Scheme I.
  • step (8a) of Reaction Scheme X the chloro group of a chloroalkyl-substituted 1H-imidazo[4,5-c]pyridin-4-amine of Formula LXI is displaced with a hydroxylamine of formula H 2 NOR 2-2 or a salt thereof to provide a 1H-imidazo[4,5-c]pyridin-4-amine of Formula LXIII, which is a subgenus of Formulas I and II.
  • the reaction can be carried out using the methods described in step (4) of Reaction Scheme I.
  • step (9) of Reaction Scheme X a 1H-imidazo[4,5-c]pyridin-4-amine of Formula LXIII is further derivatized to provide a 1H-imidazo[4,5-c]pyridin-4-amine of Formula LXII, which is a subgenus of Formulas I and II.
  • the reaction can be carried out using the methods described in step (8) of Reaction Scheme II.
  • compounds of the invention can be prepared according to Reaction Scheme XI, wherein R A2 , R B2 , R 1 , R 2-1 , X, Z, and G are as defined above.
  • Compounds of Formula Ia can be prepared according to the methods described above.
  • the amino group of a compound of Formula Ia 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 an amino group with a group such as —C(O)—R′′, ⁇ -aminoacyl, ⁇ -aminoacyl- ⁇ -aminoacyl, —C(O)—O—R′′, —C(O)—N(R′′′′)—R′′, —C(—NY 1 )—R′′, —CH(OH)—C(O)—OY 1 , —CH(OC 1-4 alkyl)Y 0 , —CH 2 Y 2 , or —CH(CH 3 )Y 2 ; wherein R′′ and R′′′′ are each independently C 1-10 alkyl, C 3-7 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, C 1-6 alkyl, C
  • Particularly useful compounds of Formula VII 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 Ia 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.
  • compositions of the invention contain a therapeutically effective amount of a compound or salt described above in combination with a pharmaceutically acceptable carrier.
  • 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.
  • 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 ( ⁇ g/kg) to about 5 mg/kg, of the compound or salt to the subject.
  • 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-1, IL-6, IL-10 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.
  • IFN- ⁇ interferon- ⁇
  • TNF- ⁇ tumor necrosis factor- ⁇
  • IL-12 interleukins
  • 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.
  • 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.
  • 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 H 1 T helper type 1
  • T H 2 T helper type 2
  • 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),
  • 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;
  • 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;
  • 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;
  • atopic diseases such as atopic dermatitis or eczema, eosinophilia, asthma, allergy, allergic rhinitis, and Ommen's syndrome;
  • 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).
  • 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, meningo
  • 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.
  • 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- ⁇ , TNF- ⁇ , IL-1, IL-6, IL-10 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 ⁇ g/kg to about 5 mg/kg.
  • 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 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 .
  • N 4 -(2-Methylpropyl)quinoline-3,4-diamine (U.S. Pat. No. 5,389,640
  • Example 1 41 g, 0.190 mol), dichloromethane (550 mL), triethylamine (40 mL, 0.286 mol), and chloroacetyl chloride (16.7 mL, 0.210 mol) were combined and then stirred at ambient temperature over the weekend.
  • the reaction mixture was diluted with 1,2-dichloroethane (75 mL) and then washed with saturated aqueous sodium bicarbonate (3 ⁇ 400 mL).
  • the aqueous layer was separated and extracted with dichloromethane (2 ⁇ 200 mL). The combined organic fractions were dried over magnesium sulfate, filtered, and concentrated under reduced pressure to provide 32 g of crude product as a tan solid. The crude product was dissolved in dichloromethane (50 mL), and the resulting solution was divided into two portions.
  • Each portion was purified by column chromatography using a HORIZON HPFC system (an automated, modular high-performance flash purification product available from Biotage, Inc, Charlottesville, Va., USA) using a FLASH 65I silica cartridge (also available from Biotage, Inc.) (eluting with ethyl acetate:methanol in a gradient from 98:2 to 85:15) to provide 11.24 g of 2-chloromethyl-1-(2-methylpropyl)-1H-imidazo[4,5-c]quinolin-4-amine as a tan solid.
  • HORIZON HPFC system an automated, modular high-performance flash purification product available from Biotage, Inc, Charlottesville, Va., USA
  • FLASH 65I silica cartridge also available from Biotage, Inc.
  • the organic fraction was concentrated under reduced pressure to afford a white solid, which was purified by preparative high performance liquid chromatography (prep HPLC) using a Waters FractionLynx automated purification system.
  • the prep HPLC fractions were analyzed using a Waters LC/TOF-MS, and the appropriate fractions were centrifuge evaporated to provide the trifluoroacetate salt of the desired compound.
  • Reversed phase prep HPLC was performed with non-linear gradient elution from 5-95% B where A is 0.05% trifluoroacetic acid/water and B is 0.05% trifluoroacetic acid/acetonitrile. Fractions were collected by mass-selective triggering.
  • the prep HPLC purification afforded 1-(2-methylpropyl)-2- ⁇ [methoxy(methyl)amino]methyl ⁇ -1H-imidazo[4,5-c]quinolin-4-amine trifluoroacetate.
  • Methoxylamine hydrochloride (145 mg, 1.73 mmol) was added to a solution of 2-chloromethyl-1-(2-methylpropyl)-1H-imidazo[4,5-c]quinolin-4-amine (0.250 g, 0.866 mmol) in DMF (3 mL). A precipitate formed. Triethylamine (0.263 g, 2.60 mmol) was added, and the reaction was heated with stirring overnight at 50° C., allowed to cool to ambient temperature, and partitioned between ethyl acetate (5 mL) and water (5 mL).
  • the aqueous fraction was extracted twice with ethyl acetate, and the combined organic fractions were dried over magnesium sulfate, filtered, and concentrated under reduced pressure.
  • the crude product was purified by radial chromatography on a silica gel plate (2 mm) (eluting sequentially with 2% and 5% methanol in chloroform).
  • the solid was then purified by prep HPLC as described in Example 1 to provide 1-(2-methylpropyl)-2-[(methoxyamino)methyl]-1H-imidazo[4,5-c]quinolin-4-amine trifluoroacetate.
  • Example 1 The method described in Example 1 was followed using N-methylhydroxylamine hydrochloride (0.058 g, 0.69 mmol) in lieu of N,O-dimethylhydroxylamine hydrochloride.
  • the product was purified by prep HPLC as described in Example 1 to provide 2- ⁇ [hydroxy(methyl)amino]methyl ⁇ -1-(2-methylpropyl)-1H-imidazo[4,5-c]quinolin-amine trifluoroacetate.
  • Methoxylamine hydrochloride (0.659 g, 7.90 mmol) was added to a stirred suspension of 1-(2-methylpropyl)-1H-imidazo[4,5-c]quinoline-2-carbaldehyde (1.00 g, 3.95 mmol) in methanol (10 mL), and the reaction was stirred at room temperature overnight. The methanol was removed under reduced pressure, and the residue was diluted with saturated aqueous sodium bicarbonate (50 mL).
  • Methoxylamine hydrochloride (0.26 g, 3.1 mmol) was added to a stirred solution of 4-[4-amino-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-2-yl]butan-2-one (0.600 g, 1.84 mmol) in pyridine (9 mL), and the reaction was stirred at room temperature for 45 minutes. Water (50 mL) was added. The aqueous layer was separated, extracted with dichloromethane (2 ⁇ 50 mL), adjusted to pH 10 with the addition of saturated aqueous sodium carbonate, and extracted with dichloromethane (2 ⁇ 25 mL).
  • 1-tetrahydro-2H-pyran-4-ylmethanamine 40 g, 0.347 mol was added to a stirred solution of 7-(benzyloxy)-4-chloro-3-nitroquinoline (55 g, 0.176 mol) and triethylamine (50.96 g, 0.503 mol) in N,N-dimethylformamide (DMF) (250 mL) at 0° C. After complete addition, the ice bath was removed. To consume the remaining starting material, additional (1-tetrahydro-2H-pyran-4-ylmethanamine (11 g, 95.50 mmol) was added. The reaction proceeded at ambient temp for 3 days, then was chilled in an ice-water bath.
  • DMF N,N-dimethylformamide
  • a 2 L glass parr vessel was charged with of 7-(benzyloxy)-3-nitro-N-(tetrahydro-2H-pyran-4-ylmethyl)quinolin-4-amine (59.34 g, 0.151 mol), platinum on carbon (5%, 6.6 g) and acetonitrile (600 mL) to provide a black mixture.
  • the vessel was placed on a shaker and pressurized with hydrogen gas ( ⁇ 50 psi, 3.4 ⁇ 10 5 Pa). After shaking for 18 hours at ambient temperature, the mixture was filtered through CELITE filter agent, and the filter cake washed with acetonitrile. The filtrate was concentrated under reduced pressure to provide 7-(benzyloxy)-N 4 -(tetrahydro-2H-pyran-4-ylmethyl)quinoline-3,4-diamine
  • the combined organic fractions were dried over sodium sulfate, filtered, and concentrated under reduced pressure to provide 2.1 g of crude product as a tan solid.
  • the crude product was purified by column chromatography using a HORIZON HPFC system (an automated, modular high-performance flash purification product available from Biotage, Inc, Charlottesville, Va., USA) using a FLASH 40+M silica cartridge (also available from Biotage, Inc.) (eluting with dichloromethane:methanol in a gradient from 100:0 to 90:10) to provide 1.8 g of 7-(benzyloxy)-2-(chloromethyl)-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-4-amine as a tan solid.
  • the aqueous layer was extracted with another portion of dichloromethane (50 mL). The combined organic layers were dried over sodium sulfate, filtered and concentrated under reduced pressure.
  • the crude material was purified by column chromatography using a HORIZON HPFC system equipped with a FLASH 40+M silica cartridge eluting with dichloromethane:methanol in a gradient from 100:0 to 90:10.
  • the resulting product (1.0 g) was recrystallized from acetonitrile, and the crystals were isolated by filtration, washed with acetonitrile, and dried overnight in a vacuum oven at 65° C.
  • the aqueous layer was extracted with another portion of dichloromethane (30 mL). The combined organic layers were dried over sodium sulfate, filtered and concentrated under reduced pressure.
  • the crude material was purified by column chromatography using a HORIZON HPFC system equipped with a FLASH 40+M silica cartridge eluting with dichloromethane:methanol in a gradient from 100:0 to 90:10.
  • the resulting product (0.3 g) was recrystallized from acetonitrile (3 mL), and the crystals were isolated by filtration, washed with acetonitrile, and dried overnight in a vacuum oven at 65° C.
  • the crude material was purified by column chromatography using a HORIZON HPFC system equipped with a FLASH 25+M silica cartridge eluting with dichloromethane:methanol in a gradient from 100:0 to 90:10.
  • the resulting product (0.35 g) was recrystallized from acetonitrile (43 mL), and the crystals were isolated by filtration, washed with acetonitrile, and dried overnight in a vacuum oven at 65° C.
  • Triethylamine (0.747 g, 1.03 mL, 7.38 mmol) and N,O-dimethylhydroxylamine hydrochloride (0.480 g, 4.92 mmol) were added to a stirring suspension of 2-chloromethyl-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-4-amine (0.750 g, 2.46 mmol) in DMF (5 mL).
  • the resulting suspension was heated to 50° C. and stirred for 17 hours.
  • the reaction mixture was cooled to room temperature and poured into water (100 mL). A solid was removed by filtration and discarded.
  • the filtrate was extracted with ethyl acetate (3 ⁇ 100 mL).
  • Triethylamine (1.49 g, 2.1 mL, 14.8 mmol) and methoxylamine hydrochloride (0.822 g, 9.84 mmol) were added to a stirring suspension of 2-chloromethyl-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-4-amine (1.50 g, 4.92 mmol) in DMF (10 mL).
  • the resulting suspension was heated to 50° C. and stirred for 17 hours.
  • the reaction mixture was cooled to room temperature and poured into water (20 mL).
  • the aqueous layer was extracted with ethyl acetate (3 ⁇ 50 mL) and dichloromethane (3 ⁇ 50 mL).
  • Example 6 1-(4-Amino-7-bromo-2- ⁇ [methoxy(methyl)amino]methyl ⁇ -1H-imidazo[4,5-c][1,5]naphthyridin-1-yl)-2-methylpropan-2-ol was prepared according to the methods described in Example 6 parts A-D (using 7-bromo-4-chloro-3-nitro[1,5]naphthyridine and 1-amino-2-methylpropan-2-ol in lieu of 7-(benzyloxy)-4-chloro-3-nitroquinoline and 1-tetrahydro-2H-pyran-4-ylmethanamine, respectively) and Example 7.
  • the product was provided as 0.63 g of a white solid, mp 190-192° C.
  • Certain exemplary compounds including some of those described above in the Examples; have the following Formulas (IIa, IIIa, IVa, or Va) and the following R 1 and R 2 substituents, wherein each line of the table is matched with Formula Ia, IIIa, IVa, or Va to represent a specific embodiment of the invention.
  • Certain exemplary compounds including some of those described above in the Examples, have the following Formulas (IIa, IIIa, IVa, or Va) and the following R 1 and R 2 substituents, wherein each line of the table is matched with Formula IIa, IIIa, IVa, or Va to represent a specific embodiment of the invention.
  • Certain exemplary compounds including some of those described above in the Examples, have the following Formulas (IIId, or Vd) and the following R 1 and R 2 substituents, wherein each line of the table is matched with Formula IIId or Vd 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.
  • cytokine induction An in vitro human blood cell system is used to assess cytokine induction. Activity is based on the measurement of interferon ( ⁇ ) and tumor necrosis factor ( ⁇ ) (IFN- ⁇ and TNF- ⁇ , 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).
  • interferon
  • tumor necrosis factor
  • PBMC Peripheral blood mononuclear cells
  • HISTOPAQUE-1077 Sigma, St. Louis, Mo.
  • Ficoll-Paque Plus Amersham Biosciences Piscataway, N.J.
  • Blood is diluted 1:1 with Dulbecco's Phosphate Buffered Saline (DPBS) or Hank's Balanced Salts Solution (HBSS).
  • DPBS Dulbecco's Phosphate Buffered Saline
  • HBSS Hank's Balanced Salts Solution
  • PBMC whole blood is placed in Accuspin (Sigma) or LeucoSep (Greiner Bio-One, Inc., Longwood, Fla.) centrifuge frit tubes containing density gradient medium.
  • the PBMC layer is collected and washed twice with DPBS or HBSS and re-suspended at 4 ⁇ 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 ⁇ 10 6 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.
  • IFN- ⁇ concentration is determined with a human multi-subtype colorimetric sandwich ELISA (Catalog Number 41105) from PBL Biomedical Laboratories, Piscataway, N.J. Results are expressed in pg/mL.
  • the TNF- ⁇ 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- ⁇ capture and detection antibody pair (Catalog Numbers AHC3419 and AEC3712) from Biosource International, Camarillo, Calif. 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-1H-imidazo[4,5-c]quinolin-1-yl]ethanol hydrate (U.S. Pat. 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.
  • the CYTOKINE INDUCTION IN HUMAN CELLS test method described above was modified as follows for high throughput screening.
  • PBMC Peripheral blood mononuclear cells
  • HISTOPAQUE-1077 Sigma, St. Louis, Mo.
  • Ficoll-Paque Plus Amersham Biosciences Piscataway, N.J.
  • Whole blood is placed in Accuspin (Sigma) or LeucoSep (Greiner Bio-One, Inc., Longwood, Fla.) centrifuge frit tubes containing density gradient medium.
  • the PBMC layer is collected and washed twice with DPBS or HBSS and re-suspended at 4 ⁇ 10 6 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.
  • 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-1H-imidazo[4,5-c]quinolin-1-yl]ethanol hydrate (U.S. Pat. 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 ⁇ M) than the final tested concentration range.
  • 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 2 ⁇ 10 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.
  • MSD MULTI-SPOT plates contain within each well capture antibodies for human TNF- ⁇ and human IFN- ⁇ that have been pre-coated on specific spots.
  • Each well contains four spots: one human TNF- ⁇ capture antibody (MSD) spot, one human IFN- ⁇ a capture antibody (PBL Biomedical Laboratories, Piscataway, N.J.) spot, and two inactive bovine serum albumin spots.
  • the human TNF- ⁇ capture and detection antibody pair is from MesoScale Discovery.
  • the human IFN- ⁇ multi-subtype antibody (PBL Biomedical Laboratories) captures all IFN- ⁇ subtypes except IFN- ⁇ F (IFNA21).
  • Standards consist of recombinant human TNF- ⁇ (R&D Systems, Minneapolis, Minn.) and IFN- ⁇ (PBL Biomedical Laboratories). Samples and separate standards are added at the time of analysis to each MSD plate. Two human IFN- ⁇ detection antibodies (Cat. Nos. 21112 & 21100, PBL) are used in a two to one ratio (weight:weight) to each other to determine the IFN- ⁇ 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.
  • 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-1H-imidazo[4,5-c]quinolin-1-yl]ethanol hydrate (U.S. Pat. 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- ⁇ , ⁇ -dimethyl-1H-imidazo[4,5-c]quinolin-1-yl]ethanol hydrate (U.S. Pat. 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 (1 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

Imidazo[4,5-c] ring compounds, (e.g. imidazo[4,5-c]pyridines, imidazo[4,5-c]quinolines, 6,7,8,9-tetrahydro imidazo[4,5-c]quinolines, imidazo[4,5-c]naphthyridine, and 6,7,8,9-tetrahydro imidazo[4,5-c]naphthyridine compounds) having an oxime or hydroxylamine substituent at the 2-position, pharmaceutical compositions containing the compounds, intermediates, and methods of making and methods of use of these compounds as immunomodulators, for modulating cytokine biosynthesis in animals and in the treatment of diseases including viral and neoplastic diseases are disclosed.

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
  • The present invention claims priority to U.S. Provisional Application Ser. No. 60/652,209, filed Feb. 11, 2005, which is 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 means.
  • 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 US20090062328A1-20090305-C00001
  • wherein RA, RB, X, Z, R′, and R2-1 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 F 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 Formulas I, I, III, IV, V, VI, and VII:
  • Figure US20090062328A1-20090305-C00002
  • as well as certain intermediates of the following Formula VIII:
  • Figure US20090062328A1-20090305-C00003
  • wherein RA, RB, RA1, RB1, RA2, RB2, R, R′, R1, R2-1, R3, m, n, p, G, X, and Z are as defined below; and pharmaceutically acceptable salts thereof.
  • In one embodiment, the present invention provides a compound of Formula I:
  • Figure US20090062328A1-20090305-C00004
  • wherein:
  • Z is selected from the group consisting of:
      • —C(—N—O—R2-2)— and
      • —C(R2-4)(—N(—OR2-2)—Y—R2-3)—;
  • X is selected from the group consisting of a bond, C1-4 alkylene and C2-4 alkenylene;
  • R2—, R2-2, and R2-3 are independently selected from the group consisting of:
      • hydrogen,
      • alkyl,
      • alkenyl,
      • aryl,
      • arylalkylenyl,
      • heteroaryl,
      • heteroarylalkylenyl,
      • heterocyclyl,
      • heterocyclylalkylenyl, and
  • alkyl, alkenyl, aryl, arylalkylenyl, heteroaryl, heteroarylalkylenyl, heterocyclyl, and heterocyclylalkylenyl, substituted by one or more substituents selected from the group consisting of:
      • hydroxy,
      • alkyl,
      • haloalkyl,
      • hydroxyalkyl,
      • alkoxy,
      • dialkylamino,
      • —S(O)0-2—R2-5,
      • —NH—S(O)2—R2-5,
      • haloalkoxy,
      • halogen,
      • cyano,
      • nitro,
      • —N3,
      • aryl,
      • heteroaryl,
      • heterocyclyl,
      • aryloxy,
      • arylalkyleneoxy,
      • —C(O)—O-alkyl,
      • —C(O)—N(R8)2,
      • —N(R8)—C(O)—R2-5,
      • —NH—C(O)—NH—R2-5,
      • —NH—C(O)—NH2
      • —O—(CO)-alkyl, and
      • —C(O)-alkyl;
      • with the proviso that R2-2 is other than alkenyl wherein the carbon atom bonded to —O— is doubly bonded to another carbon atom;
  • R2-4 is selected from the group consisting of hydrogen, C1-4alkyl, and phenyl;
  • R2-5 is selected from the group consisting of alkyl, aryl, arylalkylenyl, heteroaryl, and heteroarylalkylenyl, each of which is unsubstituted or substituted by one or more substituents selected from the group consisting of halogen, cyano, nitro, alkoxy, dialkylamino, alkylthio, haloalkyl, haloalkoxy, alkyl, and —N3;
  • Y is selected from the group consisting of:
      • a bond,
      • —C(R6)—,
      • —S(O)2—,
      • —S(O)2—N(R8)—,
  • Figure US20090062328A1-20090305-C00005
      • —C(O)—O—,
      • —C(R6)—N(R8)—,
      • —C(O)—N(R9)—S(O)2—,
      • —C(R6)—N(R8)—C(O)—,
  • Figure US20090062328A1-20090305-C00006
      • —C(O)—C(O)—,
      • —C(O)—C(O)—O—, and
      • —C(═NH)—N(R8)—;
  • 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 benzene ring or fused pyridine ring wherein the fused benzene ring or fused pyridine ring is unsubstituted or substituted by one or more R′″ groups;
  • or when taken together, RA and RB form a fused cyclohexene ring or a fused tetrahydropyridine ring, wherein the fused cyclohexene or tetrahydropyridine ring is unsubstituted or substituted by one or more R groups;
  • R is selected from the group consisting of:
      • halogen,
      • hydroxy,
      • alkyl,
      • alkenyl,
      • haloalkyl,
      • alkoxy,
      • alkylthio, and
      • —N(R9)2;
  • R′ is hydrogen or a non-interfering substituent;
  • R′″ is a non-interfering substitutent;
  • R6 is selected from the group consisting of ═O and ═S;
  • R8 is selected from the group consisting of hydrogen, C1-10 alkyl, C2-10 alkenyl, C1-10 alkoxy-C1-10 alkylenyl, hydroxy-C1-10 alkylenyl, heteroaryl-C1-10 alkylenyl, and aryl-C1-10alkylenyl;
  • R9 is selected from the group consisting of hydrogen and alkyl; and
  • R10 is C3-8 alkylene;
  • or a pharmaceutically acceptable salt thereof.
  • In another embodiment, the present invention provides a compound of Formula II:
  • Figure US20090062328A1-20090305-C00007
  • wherein:
  • Z is selected from the group consisting of:
      • —C(═N—O—R2-2)— and
      • —C(R2-4)(—N(—OR2-2)—Y—R2-3)—;
  • X is selected from the group consisting of a bond, C1-4 alkylene and C2-4 alkenylene;
  • R2-1, R2-2, and R2-3 are independently selected from the group consisting of:
      • hydrogen,
      • alkyl,
      • alkenyl,
      • aryl,
      • arylalkylenyl,
      • heteroaryl,
      • heteroarylalkylenyl,
      • heterocyclyl,
      • heterocyclylalkylenyl, and
  • alkyl, alkenyl, aryl, arylalkylenyl, heteroaryl, heteroarylalkylenyl, heterocyclyl, and heterocyclylalkylenyl, substituted by one or more substituents selected from the group consisting of:
      • hydroxy,
      • alkyl,
      • haloalkyl,
      • hydroxyalkyl,
      • alkoxy,
      • dialkylamino,
      • —S(O)0-2—R2-5,
      • —NH—S(O)2—R2-5,
      • haloalkoxy,
      • halogen,
      • cyano,
      • nitro,
      • —N3,
      • aryl,
      • heteroaryl,
      • heterocyclyl,
      • aryloxy,
      • arylalkyleneoxy,
      • —C(O)—O-alkyl,
      • —C(O)—N(R8)2,
      • —N(R8)—C(O)—R2-5,
      • —NH—C(O)—NH—R2-5,
      • —NH—C(O)—NH2
      • —O—(CO)-alkyl, and
      • —C(O)-alkyl;
      • with the proviso that R2-2 is other than alkenyl wherein the carbon atom bonded to —O— is doubly bonded to another carbon atom;
  • R2-4 is selected from the group consisting of hydrogen, C1-4alkyl, and phenyl;
  • R2-5 is selected from the group consisting of alkyl, aryl, arylalkylenyl, heteroaryl, and heteroarylalkylenyl, each of which is unsubstituted or substituted by one or more substituents selected from the group consisting of halogen, cyano, nitro, alkoxy, dialkylamino, alkylthio, haloalkyl, haloalkoxy, alkyl, and —N3;
  • Y is selected from the group consisting of:
      • a bond,
      • —C(R6)—,
      • —S(O)2—,
      • —S(O)2—N(R8)—,
  • Figure US20090062328A1-20090305-C00008
      • —C(O)—O—,
      • —C(R6)—N(R8)—,
      • —C(O)—N(R8)—S(O)2—,
      • —C(R6)—N(R8)—C(O)—,
  • Figure US20090062328A1-20090305-C00009
      • —C(O)—C(O)—,
      • —C(O)—C(O)—O—, and
      • —C(═NH)—N(R8)—;
  • RA1 and RB1 are each independently selected from the group consisting of:
      • hydrogen,
      • halogen,
      • alkyl,
      • alkenyl,
      • alkoxy,
      • alkylthio, and
      • —N(R9)2;
  • R1 is selected from the group consisting of:
      • —R4,
      • —X′—R4,
      • —X′—Y′—R4,
      • —X′—Y′—X′—Y′—R4, and
      • —X′—R5;
  • X′ is selected from the group consisting of alkylene, alkenylene, alkynylene, arylene, heteroarylene, and heterocyclylene, wherein the alkylene, alkenylene, and alkynylene groups can be optionally interrupted or terminated by arylene, heteroarylene or heterocyclylene and optionally interrupted by one or more —O— groups;
  • Y′ is selected from the group consisting of:
      • —O—,
      • —S(O)0-2—,
      • —S(O)2—N(R8)—,
      • —C(R6)—,
      • —C(R6)—O—,
      • —O—C(R6)—,
      • —O—C(O)—O—,
      • —N(R8)-Q-,
      • —C(R6)—N(R8)—,
      • —O—C(R6)—N(R8)—,
      • —C(R6)—N(OR9)—,
      • —O—N(R8)-Q-,
      • —O—N═C(R4)—,
      • —C(═N—O—R8)—,
      • —CH(—N(—O—R8)-Q-R4)—,
  • Figure US20090062328A1-20090305-C00010
  • 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, alkylamino, dialkylamino, (dialkylamino)alkyleneoxy, and in the case of alkyl, alkenyl, alkynyl, and heterocyclyl, oxo;
  • R5 is selected from the group consisting of:
  • Figure US20090062328A1-20090305-C00011
  • R6 is selected from the group consisting of ═O and ═S;
  • R7 is C2-7 alkylene;
  • R8 is selected from the group consisting of hydrogen, C1-10 alkyl, C2-10 alkenyl, C1-10 alkoxy-C1-10 alkylenyl, hydroxy-C1-10 alkylenyl, heteroaryl-C1-10 alkylenyl, and aryl-C1-10alkylenyl;
  • R9 is selected from the group consisting of hydrogen and alkyl;
  • R10 is C3-8 alkylene;
  • A is selected from the group consisting of —O—, —C(O)—, —S(O)0-2—, —CH2—, and —N(-Q-R4)—;
  • A′ is selected from the group consisting of —O—, —S(O)0-2—, —N(-Q-R4)—, and —CH2—;
  • 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)—NR8)—, —C(R6)—O—, —C(R6)—S—, and —C(R6)—N(OR9)—;
  • V is selected from the group consisting of —C(R6)—, —O—C(R6)—, —N(R8)—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 Formula III:
  • Figure US20090062328A1-20090305-C00012
  • wherein:
  • Z is selected from the group consisting of:
      • —C(═N—O—R2-2)— and
      • —C(R2-4)(—N(—OR2-2)—Y—R2-3)—;
  • X is selected from the group consisting of a bond, C1-4 alkylene and C2-4 alkenylene;
  • R2-1, R2-2, and R2-3 are independently selected from the group consisting of:
      • hydrogen,
      • alkyl,
      • alkenyl,
      • aryl,
      • arylalkylenyl,
      • heteroaryl,
      • heteroarylalkylenyl,
      • heterocyclyl,
      • heterocyclylalkylenyl, and
  • alkyl, alkenyl, aryl, arylalkylenyl, heteroaryl, heteroarylalkylenyl, heterocyclyl, and heterocyclylalkylenyl, substituted by one or more substituents selected from the group consisting of:
      • hydroxy,
      • alkyl,
      • haloalkyl,
      • hydroxyalkyl,
      • alkoxy,
      • dialkylamino,
      • —S(O)0-2—R2-5,
      • —NH—S(O)2—R2-5,
      • haloalkoxy,
      • halogen,
      • cyano,
      • nitro,
      • —N3,
      • aryl,
      • heteroaryl,
      • heterocyclyl,
      • aryloxy,
      • arylalkyleneoxy,
      • —C(O)—O-alkyl,
      • —C(O)—N(R8)2,
      • —N(R8)—C(O)—R2-5,
      • —NH—C(O)—NH—R2-5,
      • —NH—C(O)—NH2
      • —O—(CO)-alkyl, and
      • —C(O)-alkyl;
      • with the proviso that R2-2 is other than alkenyl wherein the carbon atom bonded to —O— is doubly bonded to another carbon atom;
  • R2-4 is selected from the group consisting of hydrogen, C1-4alkyl, and phenyl;
  • R2-5 is selected from the group consisting of alkyl, aryl, arylalkylenyl, heteroaryl, and heteroarylalkylenyl, each of which is substituted or substituted by one or more substituents selected from the group consisting of halogen, cyano, nitro, alkoxy, dialkylamino, alkylthio, haloalkyl, haloalkoxy, alkyl, and —N3;
  • Y is selected from the group consisting of:
      • a bond,
      • —C(R6)—,
      • —S(O)2—,
      • —S(O)2—N(R8)—,
  • Figure US20090062328A1-20090305-C00013
      • —C(O)—O—,
      • —C(R6)—N(R8)—,
      • —C(O)—N(R8)—S(O)2—,
      • —C(R6)—N(R8)—C(O)—,
  • Figure US20090062328A1-20090305-C00014
      • —C(O)—C(O)—,
      • —C(O)—C(O)—O—, and
      • —C(═NH)—N(R8)—;
  • R is selected from the group consisting of:
      • halogen,
      • hydroxy,
      • alkyl,
      • alkenyl,
      • haloalkyl,
      • alkoxy,
      • alkylthio, and
      • —N(R9)2;
  • n is an integer from 0 to 4;
  • R1 is selected from the group consisting of:
      • —R4,
      • —X′—R4,
      • —X′—Y′—R4,
      • —X′—Y′—X′—Y′—R4, and
      • —X′—R5;
  • R3 is selected from the group consisting of:
      • -Z′-R4,
      • -Z′-X′—R4,
      • -Z′-X′—Y′—R4,
      • -Z′-X′—Y′—X′—Y′—R4, and
      • -Z′-X′—R5;
  • m is 0 or 1, with the proviso that when m is 1 then n is 0 or 1;
  • X′ is selected from the group consisting of alkylene, alkenylene, alkynylene, arylene, heteroarylene, and heterocyclylene, wherein the alkylene, alkenylene, and alkynylene groups can be optionally interrupted or terminated by arylene, heteroarylene or heterocyclylene and optionally interrupted by one or more —O— groups;
  • Y′ is selected from the group consisting of:
      • —O—,
      • —S(O)0-2—,
      • —S(O)2—N(R8)—,
      • —C(R6)—,
      • —C(R6)—O—,
      • —O—C(R6)—,
      • —O—C(O)—O—,
      • —N(R8)-Q-,
      • —C(R6)—N(R8)—,
      • —O—C(R6)—N(R8)—,
      • —C(R6)—N(OR9)—,
      • —O—N(R8)-Q-,
      • —O—N═C(R4)—,
      • —C(═N—O—R8)—,
      • —CH(—N(—O—R8)-Q-R4)—,
  • Figure US20090062328A1-20090305-C00015
  • Z′ is a bond or —O—;
  • 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, alkylamino, dialkylamino, (dialkylamino)alkyleneoxy, and in the case of alkyl, alkenyl, alkynyl, and heterocyclyl, oxo;
  • R5 is selected from the group consisting of:
  • Figure US20090062328A1-20090305-C00016
  • R6 is selected from the group consisting of ═O and ═S;
  • R7 is C2-7 allylene;
  • R8 is selected from the group consisting of hydrogen, C1-10 alkyl, C2-10 alkenyl, C1-10 alkoxy-C1-10 alkylenyl, hydroxy-C1-10 alkylenyl, heteroaryl-C1-10 alkylenyl, and aryl-C1-10 alkylenyl;
  • R9 is selected from the group consisting of hydrogen and alkyl;
  • R10 is C3-8 alkylene;
  • A is selected from the group consisting of —O—, —C(O)—, —S(O)0-2—, —CH2—, and —N(-Q-R4)—;
  • A′ is selected from the group consisting of —O—, —S(O)0-2—, —N(-Q-R4)—, and —CH2—;
  • 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(R6)—O—, —C(R6)—S—, and —C(R6)—N(OR9)—;
  • V is selected from the group consisting of —C(R6)—, —O—C(R6)—, —N(R8)—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 Formula IV:
  • Figure US20090062328A1-20090305-C00017
  • wherein:
  • Z is selected from the group consisting of:
      • —C(═N—O—R2-2)— and
      • —C(R2-4)(—N(—OR2-2)—Y—R2-3)—;
  • X is selected from the group consisting of a bond, C1-4 alkylene and C2-4 alkenylene;
  • R2-1, R2-2, and R2-3 are independently selected from the group consisting of:
      • hydrogen,
      • alkyl,
      • alkenyl,
      • aryl,
      • arylalkylenyl,
      • heteroaryl,
      • heteroarylalkylenyl,
      • heterocyclyl,
      • heterocyclylalkylenyl, and
  • alkyl, alkenyl, aryl, arylalkylenyl, heteroaryl, heteroarylalkylenyl, heterocyclyl, and heterocyclylalkylenyl, substituted by one or more substituents selected from the group consisting of:
      • hydroxy,
      • alkyl,
      • haloalkyl,
      • hydroxyalkyl,
      • alkoxy,
      • dialkylamino,
      • —S(O)0-2—R2-5,
      • —NH—S(O)2—R2-5,
      • haloalkoxy,
      • halogen,
      • cyano,
      • nitro,
      • —N3,
      • aryl,
      • heteroaryl,
      • heterocyclyl,
      • aryloxy,
      • arylalkyleneoxy,
      • —C(O)—O-alkyl,
      • —C(O)—N(R8)2,
      • —N(R8)—C(O)—R2-5,
      • —NH—C(O)—NH—R2-5,
      • NH—C(O)—NH2
      • —O—(CO)-alkyl, and
      • —C(O)-alkyl;
      • with the proviso that R2-2 is other than alkenyl wherein the carbon atom bonded to —O— is doubly bonded to another carbon atom;
  • R2-4 is selected from the group consisting of hydrogen, C1-4alkyl, and phenyl;
  • R2-5 is selected from the group consisting of alkyl, aryl, arylalkylenyl, heteroaryl, and heteroarylalkylenyl, each of which is unsubstituted or substituted by one or more substituents selected from the group consisting of halogen, cyano, nitro, alkoxy, dialkylamino, alkylthio, haloalkyl, haloalkoxy, alkyl, and —N3;
  • Y is selected from the group consisting of:
      • a bond,
      • —C(R6)—,
      • —S(O)2—,
      • —S(O)2—N(R8)—,
  • Figure US20090062328A1-20090305-C00018
      • —C(O)—O—,
      • —C(R6)—N(R8)—,
      • —C(O)—N(R8)—S(O)2—,
      • —C(R6)—N(R8)—C(O)—,
  • Figure US20090062328A1-20090305-C00019
      • —C(O)—C(O)—,
      • —C(O)—C(O)—O—, and
      • —C(═NH)—N(R8)—;
  • R is selected from the group consisting of:
      • halogen,
      • hydroxy,
      • alkyl,
      • alkenyl,
      • haloalkyl,
      • alkoxy,
      • alkylthio, and
      • —N(R9)2;
  • n is an integer from 0 to 4;
  • R1 is selected from the group consisting of:
      • —R4,
      • —X′—R4,
      • —X′Y′—R4,
      • —X′Y′—X′—Y′—R4, and
      • —X′—R5;
  • X′ is selected from the group consisting of alkylene, alkenylene, alkynylene, arylene, heteroarylene, and heterocyclylene, wherein the alkylene, alkenylene, and alkynylene groups can be optionally interrupted or terminated by arylene, heteroarylene or heterocyclylene and optionally interrupted by one or more —O— groups;
  • Y′ is selected from the group consisting of:
      • —O—,
      • —S(O)0-2—,
      • —S(O)2—N(R8)—,
      • —C(R6)—,
      • —C(R6)—O—,
      • —O—C(R6)—,
      • —O—C(O)—O—,
      • —N(R8)-Q-,
      • —C(R6)—N(R8)—,
      • —O—C(R6)—N(R8)—,
      • —C(R6)—N(OR9)—,
      • —O—N(R8)-Q-,
      • —O—N═C(R4)—,
      • —C(═N—O—R8)—,
      • —CH(—N(—O—R8)-Q-R4)—,
  • Figure US20090062328A1-20090305-C00020
  • 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, alkylamino, dialkylamino, (dialkylamino)alkyleneoxy, and in the case of alkyl, alkenyl, alkynyl, and heterocyclyl, oxo;
  • R5 is selected from the group consisting of:
  • Figure US20090062328A1-20090305-C00021
  • R6 is selected from the group consisting of ═O and ═S;
  • R7 is C2-7 alkylene;
  • R8 is selected from the group consisting of hydrogen, C1-10 alkyl, C2-10 alkenyl, C1-10 alkoxy-C1-10 alkylenyl, hydroxy-C1-10 alkylenyl, heteroaryl-C10 alkylenyl, and aryl-C1-10 alkylenyl;
  • R9 is selected from the group consisting of hydrogen and alkyl;
  • R10 is C3-8 alkylene;
  • A is selected from the group consisting of —O—, —C(O)—, —S(O)0-2—, —CH2—, and —N(-Q-R4)—;
  • A′ is selected from the group consisting of —O—, —S(O)0-2—, —N(-Q-R4)—, and —CH2—;
  • 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(R6)—O—, —C(R6)—S—, and —C(R6)—N(OR9)—;
  • V is selected from the group consisting of —C(R6)—, —O—C(R6)—, —N(R8)—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 Formula V:
  • Figure US20090062328A1-20090305-C00022
  • wherein:
  • Z is selected from the group consisting of:
      • —C(═N—O—R2-2)— and
      • —C(R2-4)(—N(—OR2-2)—Y—R2-3)—;
  • X is selected from the group consisting of a bond, C1-4 alkylene and C2-4 alkenylene;
  • R2-1, R2-2, and R2-3 are independently selected from the group consisting of:
      • hydrogen,
      • alkyl,
      • alkenyl,
      • aryl,
      • arylalkylenyl,
      • heteroaryl,
      • heteroarylalkylenyl,
      • heterocyclyl,
      • heterocyclylalkylenyl, and
  • alkyl, alkenyl, aryl, arylalkylenyl, heteroaryl, heteroarylalkylenyl, heterocyclyl, and heterocyclylalkylenyl, substituted by one or more substituents selected from the group consisting of:
      • hydroxy,
      • alkyl,
      • haloalkyl,
      • hydroxyalkyl,
      • alkoxy,
      • dialkylamino,
      • —S(O)0-2—R2-5,
      • —NH—S(O)2—R25,
      • haloalkoxy,
      • halogen,
      • cyano,
      • nitro,
      • —N3,
      • aryl,
      • heteroaryl,
      • heterocyclyl,
      • aryloxy,
      • arylalkyleneoxy,
      • —C(O)—O-alkyl,
      • —C(O)—N(R8)2,
      • —N(R8)—C(O)—R2-5,
      • —NH—C(O)—NH—R2-5,
      • —NH—C(O)—NH2
      • O—(CO)-alkyl, and
      • —C(O)-alkyl;
      • with the proviso that R2-2 is other than alkenyl wherein the carbon atom bonded to —O— is doubly bonded to another carbon atom;
  • R2-4 is selected from the group consisting of hydrogen, C1-4alkyl, and phenyl;
  • R2-5 is selected from the group consisting of alkyl, aryl, arylalkylenyl, heteroaryl, and heteroarylalkylenyl, each of which is unsubstituted or substituted by one or more substituents selected from the group consisting of halogen, cyano, nitro, alkoxy, dialkylamino, alkylthio, haloalkyl, haloalkoxy, alkyl, and —N3;
  • Y is selected from the group consisting of:
      • a bond,
      • —S(O)2—,
      • —S(O)2—N(R8)—,
  • Figure US20090062328A1-20090305-C00023
      • —C(O)—O—,
      • —C(R6)—N(R8)—,
      • —C(O)—N(R8)—S(O)2—,
      • —C(R6)—N(R8)—C(O)—,
  • Figure US20090062328A1-20090305-C00024
      • —C(O)—C(O)—,
      • —C(O)—C(O)—O—, and
      • —C(═NH)—N(R8)—,
  • R is selected from the group consisting of:
      • halogen,
      • hydroxy,
      • alkyl,
      • alkenyl,
      • haloalkyl,
      • alkoxy,
      • alkylthio, and
      • —N(R9)2;
  • p is an integer from 0 to 3;
  • R1 is selected from the group consisting of:
      • —R4,
      • —X′—R4,
      • —X′—Y′—R4,
      • —X′—Y′—X′—Y′—R4, and
      • —X′—R5;
  • R3 is selected from the group consisting of:
      • -Z′-R4,
      • -Z′-X′—R4,
      • -Z′-X′—Y′—X′—Y′—R4, and
      • -Z′-X′—R5;
  • m is 0 or 1, with the proviso that when m is 1 then p is 0 or 1;
  • X′ is selected from the group consisting of alkylene, alkenylene, alkynylene, arylene, heteroarylene, and heterocyclylene, wherein the alkylene, alkenylene, and alkynylene groups can be optionally interrupted or terminated by arylene, heteroarylene or heterocyclylene and optionally interrupted by one or more —O— groups;
  • Y′ is selected from the group consisting of:
      • —O—,
      • —S(O)0-2—,
      • —S(O)2—N(R8)—,
      • —C(R6)—,
      • —C(R6)—O—,
      • —O—C(R6)—,
      • —O—C(O)—O—,
      • —N(R8)-Q-,
      • —C(R6)—N(R8)—,
      • —O—C(R6)—N(R8)—,
      • C(R6)—N(OR9)—,
      • —O—N(R8)-Q-,
      • —O—N═C(R4)—,
      • —C(═N—O—R8)—,
      • —CH(—N(—O—R8)-Q-R4)—,
  • Figure US20090062328A1-20090305-C00025
  • Z′ is a bond or —O—;
  • R4 is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, aryl, arylalkylenyl, aryloxyalkylenyl, alkylaryl-enyl, 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, alkylamino, dialkylamino, (dialkylamino)alkyleneoxy, and in the case of alkyl, alkenyl, alkynyl, and heterocyclyl, oxo;
  • R5 is selected from the group consisting of:
  • Figure US20090062328A1-20090305-C00026
  • R6 is selected from the group consisting of ═O and ═S;
  • R7 is C2-7 alkylene;
  • R8 is selected from the group consisting of hydrogen, C1-10 allyl, C2-10 alkenyl, Cl1-10 alkoxy-C1-10 alkylenyl, hydroxy-C1-10 alkylenyl, heteroaryl-C1-10 alkylenyl, and aryl-C1-10 alkylenyl;
  • R9 is selected from the group consisting of hydrogen and alkyl;
  • R10 is C3-8 alkylene;
  • A is selected from the group consisting of —O—, —C(O)—, —S(O)0-2—, —CH2—, and —N(-Q-R4)—;
  • A′ is selected from the group consisting of —O—, —S(O)0-2—, —N(-Q-R4)—, and —CH2—;
  • 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(R6)—O—, —C(R6)—S—, and —C(R6)—N(OR9)—;
  • V is selected from the group consisting of —C(R6)—, —O—C(R6)—, —N(R8)—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 Formula VI:
  • Figure US20090062328A1-20090305-C00027
  • wherein:
  • Z is selected from the group consisting of:
      • —C(═N—O—R2-2)— and
      • C(R2-4)(—N(—OR2-2)—Y—R2-3)—;
  • X is selected from the group consisting of a bond, C1-4 allylene and C2-4 alkenylene;
  • R2-1, R2-2, and R2-3 are independently selected from the group consisting of:
      • hydrogen,
      • alkyl,
      • alkenyl,
      • aryl,
      • arylalkylenyl,
      • heteroaryl,
      • heteroarylalkylenyl,
      • heterocyclyl,
      • heterocyclylalkylenyl, and
  • alkyl, alkenyl, aryl, arylalkylenyl, heteroaryl, heteroarylalkylenyl, heterocyclyl, and heterocyclylalkylenyl, substituted by one or more substituents selected from the group consisting of:
      • hydroxy,
      • alkyl,
      • haloalkyl,
      • hydroxyalkyl,
      • alkoxy,
      • dialkylamino,
      • —S(O)0-2—R2-5,
      • —NH—S(O)2—R2-5,
      • haloalkoxy,
      • halogen,
      • cyano,
      • nitro,
      • —N3,
      • aryl,
      • heteroaryl,
      • heterocyclyl,
      • aryloxy,
      • arylalkyleneoxy,
      • —C(O)—O-alkyl,
      • —C(O)—N(R8)2,
      • —N(R8)—C(O)—R2-5,
      • —NH—C(O)—NH—R2-5,
      • —NH—C(O)—NH2
      • —O—(CO)-alkyl, and
      • —C(O)-alkyl;
      • with the proviso that R2-2 is other than alkenyl wherein the carbon atom bonded to —O— is doubly bonded to another carbon atom;
  • R2-4 is selected from the group consisting of hydrogen, C1-4alkyl, and phenyl;
  • R2-5 is selected from the group consisting of alkyl, aryl, arylalkylenyl, heteroaryl, and heteroarylalkylenyl, each of which is unsubstituted or substituted by one or more substituents selected from the group consisting of halogen, cyano, nitro, alkoxy, dialkylamino, alkylthio, haloalkyl, haloalkoxy, alkyl, and —N3;
  • Y is selected from the group consisting of:
      • a bond,
      • —C(R)—,
      • —S(O)2—,
      • —S(O)2—N(R8)—,
  • Figure US20090062328A1-20090305-C00028
      • —C(O)—O—,
      • —C(R6)—N(R8)—,
      • —C(O)—N(R8)—S(O)2—,
      • —C(R6)—N(R8)—C(O)—,
  • Figure US20090062328A1-20090305-C00029
      • —C(O)—C(O)—,
      • —C(O)—C(O)—O—, and
      • C(═NH)—N(R8)—;
  • R is selected from the group consisting of:
      • halogen,
      • hydroxy,
      • alkyl,
      • alkenyl,
      • haloalkyl,
      • alkoxy,
      • alkylthio, and
      • —N(R9)2;
  • p is an integer from 0 to 3;
  • R1 is selected from the group consisting of:
      • —R4,
      • —X′—R4,
      • —X′—Y′—R4,
      • —X′—Y′—X′—Y′—R4, and
      • —X′—R5;
  • X′ is selected from the group consisting of alkylene, alkenylene, alkynylene, arylene, heteroarylene, and heterocyclylene, wherein the alkylene, alkenylene, and alkynylene groups can be optionally interrupted or terminated by arylene, heteroarylene or heterocyclylene and optionally interrupted by one or more —O— groups;
  • Y′ is selected from the group consisting of:
      • —O—,
      • —S(O)0-2—,
      • —S(O)2—N(R8)—,
      • —C(R6)—,
      • —C(R6)—O—,
      • —O—C(R6)—,
      • —O—C(O)—O—,
      • —N(R8)-Q-,
      • —C(R6)—N(R8)—,
      • —O—C(R6)—N(R8)—,
      • C(R6)—N(OR9)—,
      • —O—N(R8)-Q-,
      • —O—N═C(R4)—,
      • —C(═N—O—R8)—,
      • —CH(—N(—O—R8)-Q-R4)—,
  • Figure US20090062328A1-20090305-C00030
  • 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, alkylamino, dialkylamino, (dialkylamino)alkyleneoxy, and in the case of alkyl, alkenyl, alkynyl, and heterocyclyl, oxo;
  • R5 is selected from the group consisting of:
  • Figure US20090062328A1-20090305-C00031
  • R6 is selected from the group consisting of ═O and ═S;
  • R7 is C2-7 alkylene;
  • R8 is selected from the group consisting of hydrogen, C1-10 alkyl, C2-10 alkenyl, C1-10 alkoxy-C1-10 alkylenyl, hydroxy-C1-10 alkylenyl, heteroaryl-C1-10 alkylenyl, and aryl-C1-10 alkylenyl;
  • R9 is selected from the group consisting of hydrogen and alkyl;
  • R10 is C3-8 alkylene;
  • A is selected from the group consisting of —O—, —C(O)—, —S(O)0-2—, —CH2—, and —N(-Q-R4)—;
  • A′ is selected from the group consisting of —O—, —S(O)0-2—, —N(-Q-R4)—, and —CH2—;
  • 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(R6)—O—, —C(R6)—S—, and —C(R6)—N(OR9)—;
  • V is selected from the group consisting of —C(R6)—, —O—C(R6)—, —N(R8)—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 Formula VII, which is a prodrug:
  • Figure US20090062328A1-20090305-C00032
  • wherein:
  • G 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(═NY1)—R″,
      • —CH(OH)—C(O)—OY1,
      • —CH(OC1-4 alkyl)Y0,
      • —CH2Y2, and
      • —CH(CH3)Y2;
  • R″ and R″″ are independently selected from the group consisting of C1-10 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, C1-4 alkoxy, aryl, heteroaryl, aryl-C1-4 alkylenyl, heteroaryl-C1-4 alkylenyl, halo-CIA alkylenyl, halo-C1-4 alkoxy, —O—C(O)—CH3, —C(O)—O—CH3, —C(O)—NH2, —O—CH2—C(O)—NH2, —NH2, and —S(O—NH2, with the proviso that R″″ can also be hydrogen;
  • α-aminoacyl is an α-aminoacyl group derived from an α-amino acid selected from the group consisting of racemic, D-, and L-amino acids;
  • Y1 is selected from the group consisting of hydrogen, C1-6 alkyl, and benzyl;
  • Y0 is selected from the group consisting of C1-6 alkyl, carboxy-C1-6 alkylenyl, amino-C1-4 alkylenyl, mono-N—C1-6 alkylamino-C1-4 alkylenyl, and di-N,N—C1-6 alkylamino-C1-4 alkylenyl;
  • Y2 is selected from the group consisting of mono-N—C1-6 alkylamino, di-N,N—C1-6 alkylamino, morpholin-4-yl, piperidin-1-yl, pyrrolidin-1-yl, and 4-C 4 alkylpiperazin-1-yl;
  • RA2 and RB2 are each independently selected from the group consisting of:
      • hydrogen,
      • halogen,
      • alkyl,
      • alkenyl,
      • alkoxy,
      • alkylthio, and
      • —N(R9)2;
  • or when taken together, RA2 and RB2 form a fused benzene ring or fused pyridine ring wherein the fused benzene ring or fused pyridine ring is unsubstituted or substituted by one R3 group, or one R3 group and one R group, or one, two, three, or four R groups when on the fused benzene ring, or one, two, or three R groups when on the fused pyridine ring;
  • or when taken together, RA2 and RB2 form a fused cyclohexene ring or a fused tetrahydropyridine ring, wherein the fused cyclohexene or tetrahydropyridine ring is unsubstituted or substituted by one or more R groups; and
  • X, Z, R2-1, R1, R, and R3 are defined as in Formula III above;
  • or a pharmaceutically acceptable salt thereof.
  • In one embodiment, the present invention provides an intermediate compound of Formula VIII:
  • Figure US20090062328A1-20090305-C00033
  • wherein X, Z, R2-1, R1, R, and n are defined as in Formula III above;
    or a pharmaceutically acceptable salt thereof.
  • In one embodiment of Formula VIII, R1 is preferrably tetrahydro-2H-pyran-4-ylmethyl as shown in Formula VIIIa:
  • Figure US20090062328A1-20090305-C00034
  • which compound or pharmaceutically acceptable salt thereof has been found to induce cytokine biosynthesis as described herein for compounds or salts of Formulas I-VII.
  • Herein, “non-interfering” means that the ability of the compound or salt, which includes a non-interfering substituent, to modulate the biosynthesis of one or more cytokines is not destroyed by the non-interfering substituent. For certain embodiments, R′″ is a non-interfering substituent. Illustrative non-interfering R′ groups include those described above for R1. Illustrative non-interfering R′″ groups include those described above for R and R3.
  • 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, cyclobutyl, cyclobutylmethyl, cyclopentyl, cyclopentylmethyl, cyclohexyl, cyclohexylmethyl, adamantyl, and substituted and unsubstituted bornyl, norbornyl, and norbornenyl.
  • Unless otherwise specified, “alkylene”, “alkenylene”, and “alkynylene” are the divalent forms of the “alkyl”, “alkenyl”, and “alkynyl” groups defined above. The terms, “alkylenyl”, “alkenylenyl”, and “alkynylenyl” are use 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, S, 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-(1H)-yl, octahydroisoquinolin-(1H)-yl, dihydroquinolin-(2H)-yl, octahydroquinolin-(2H)-yl, dihydro-1H-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”, “heteroarylene”, 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.
  • 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(R8)—C(O)—N(R8)— each R8 group is independently selected. In another example, when an R1 and an R3 group both contain an R4 group, each R4 group is independently selected. In a further example, when more than one Y′ group is present and each Y′ group contains one or more R8 groups, then each Y′ group is independently selected, and each R8 group is independently selected.
  • The invention is inclusive of the compounds described herein 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.
  • For any of the compounds presented herein, each one of the following variables (e.g., Z, X, Y, Y′, RA1, RB1, R, R1, R2-1, R3, Q, G, n, 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 of skill in the art. Each of the resulting combinations of variables is an embodiment of the present invention.
  • For certain embodiments of Formula I, R′″ is a non-interfering substituent.
  • For certain embodiments of Formula I, the one or more R′″ groups are one R3 group, or one R3 group and one R group, or one, two, three, or four R groups when on the fused benzene ring, or one, two, or three R groups when on the fused pyridine ring; wherein R3 is selected from the group consisting of -Z′-R4, -Z′-X′—R4, -Z′-X′—Y′—R4, -Z′-X′—Y′—X′—Y′—R4, and -Z′-X′—R5.
  • For certain embodiments of Formula I or VII, RA and RB or RA2 and RB2, respectively, are each independently selected from the group consisting of hydrogen, halogen, alkyl, alkenyl, alkoxy, alkylthio, and —N(R9)2. For certain embodiments, RA and RB or RA2 and RB2 are each independently selected from the group consisting of hydrogen and alkyl. For certain embodiments, RA and RB or RA2 and RB2 are both methyl.
  • For certain embodiments of Formula I, RA and RB are taken together to form a fused benzene ring wherein the benzene ring is unsubstituted or substituted by one or more R′″ groups. In certain of these embodiments, the fused benzene ring is substituted by one or two R′″ groups. In certain of these embodiments, the one or two R′″ groups are one R3 group, or one R3 group and one R group. In certain of these embodiments, the fused benzene ring is unsubstituted.
  • For certain embodiments of Formula VII, RA2 and RB2 are taken together to form a fused benzene ring wherein the benzene ring is unsubstituted or substituted by one R3 group, or one R3 group and one R group. In certain of these embodiments, the fused benzene ring is unsubstituted.
  • For certain embodiments of Formula I, RA and RB are taken together to form a fused pyridine ring wherein the pyridine ring is unsubstituted or substituted by one or more R′″ groups. In certain of these embodiments, the fused pyridine ring is substituted by one or two R′″ groups. In certain of these embodiments, the one or two R′″ groups are one R3 group, or one R3 group and one R group. In certain of these embodiments, the fused pyridine ring is
  • Figure US20090062328A1-20090305-C00035
  • wherein the highlighted bond is the position where the ring is fused. In certain of these embodiments, the fused pyridine ring is unsubstituted.
  • For certain embodiments of Formula VII, RA2 and RB2 are taken together to form a fused pyridine ring wherein the pyridine ring is unsubstituted or substituted by one R3 group, or one R3 group and one R group. In certain of these embodiments, the fused pyridine ring is
  • Figure US20090062328A1-20090305-C00036
  • wherein the highlighted bond is the position where the ring is fused. In certain of these embodiments, the fused pyridine ring is unsubstituted.
  • For certain embodiments of Formula I or VII, RA and RB or RA2 and RB2, respectively, are taken together to form a fused cyclohexene ring wherein the fused cyclohexene ring is unsubstituted or substituted by one or more R groups. The double bond in the cyclohexene ring is the position where the ring is fused. In certain of these embodiments, the fused cyclohexene ring is unsubstituted.
  • For certain embodiments of Formula I or VII, RA and RB or RA2 and RB2, respectively, are taken together to form a fused tetrahydropyridine ring, wherein the fused tetrahydropyridine ring is unsubstituted or substituted by one or more R groups. The double bond in the tetrahydropyridine ring is the position where the ring is fused. In certain of these embodiments, the tetrahydropyridine ring is
  • Figure US20090062328A1-20090305-C00037
  • wherein the highlighted bond indicates the position where the ring is fused. In certain of these embodiments, the fused tetrahydropyridine ring is unsubstituted.
  • For certain embodiments of Formula I, R′ is hydrogen or a non-interfering substituent.
  • For certain embodiments of Formula I, R′ is a non-interfering substituent.
  • For certain embodiments of Formula I, R′ is R1; wherein R1 is selected from the group consisting of —R4, —X′—R4, —X′—Y′—R4, —X′—Y′—X′—Y′—R4, and —X′—R5.
  • For certain embodiments of Formula II, RA1 and RB1 are each independently selected from the group consisting of hydrogen, halogen, alkyl, alkenyl, alkoxy, alkylthio, and —N(R9)2.
  • For certain embodiments of Formula II, RA1 and RB1 are each independently selected from the group consisting of hydrogen and alkyl. For certain of these embodiments, RA1 and RB1 are both methyl.
  • For certain embodiments of Formulas I, III VI, V, VI, VII, or VIII, R is halogen or hydroxy.
  • For certain embodiments of Formulas III, V, or VIII, R is bromine.
  • For certain embodiments of Formulas III, IV or VIII, n is 0.
  • For certain embodiments of Formula V or VI, p is 0.
  • For certain embodiments, including any one of the above embodiments wherein R3 is present, R3 is benzyloxy.
  • For certain embodiments, including any one of the above embodiments wherein R3 is present, except where R3 is benzyloxy, R3 is selected from the group consisting of phenyl, pyridin-3-yl, pyridin-4-yl, 5-(hydroxymethyl)pyridin-3-yl, 2-ethoxyphenyl, 3-(morpholine-4-carbonyl)phenyl, and 3-(N,N-dimethylaminocarbonyl)phenyl.
  • For certain embodiments, including any one of the above embodiments of Formulas III or V wherein this definition is not excluded, m is 0.
  • For certain embodiments, including any one of the above embodiments, Z is selected from the group consisting of —C(═N—O—R2-2)— and —C(R2-4)(—N(—OR2-2)—Y—R2-3)—.
  • For certain embodiments, including any one of the above embodiments, Z is —C(═N—O—R2-2)—.
  • For certain embodiments, including any one of the above embodiments except where Z is —C(—N—O—R2-2)—, Z is —C(R2-4)(—N(—OR2-2)—Y—R2-3)—. For certain of these embodiments, R2-4 is hydrogen. For certain of these embodiments, Y is a bond. For certain of these embodiments, R2-3 is selected from the group consisting of hydrogen and alkyl. Alternatively, Y is selected from the group consisting of —C(O)—, —S(O)2—, and —C(O)—NH—. For certain of these embodiments, R2-3 is alkyl.
  • For certain embodiments, including any one of the above embodiments, R2-2 is selected from the group consisting of hydrogen, alkyl, arylalkylenyl, and heteroarylalkylenyl. For certain of these embodiments, R2-2 is hydrogen, C1-4 alkyl, benzyl, or pyridin-2-ylmethyl.
  • For certain embodiments, including any one of the above embodiments, R2-1 is selected from the group consisting of hydrogen, alkyl, and aryl. For certain of these embodiments, R2-1 is hydrogen, C1-4 alkyl, or phenyl.
  • For certain embodiments, including any one of the above embodiments, X is a bond or C1-4 alkylene. For certain of these embodiments, X is a bond, methylene, or ethylene.
  • For certain embodiments, including any one of the above embodiments wherein R1 is present, R1 is selected from the group consisting of —R4, —X′—R4, —X′—Y′—R4, —X′—Y′—X′—Y′—R4, and —X′—R5.
  • For certain embodiments, including any one of the above embodiments wherein R1 is present, R1 is selected from the group consisting of alkyl, arylalkylenyl, aryloxyalkylenyl, hydroxyalkyl, dihydroxyalkyl, alkylsulfonylalkylenyl, —X′—Y′—R4, —X′—R5, and heterocyclylalkylenyl; wherein the heterocyclyl of the heterocyclylalkylenyl group is optionally substituted by one or more alkyl groups; wherein X′ is alkylene; Y′ is —N(R8)—C(O)—, —N(R8)—S(O)2—, —N(R8)—C(O)—N(R8)—, or
  • Figure US20090062328A1-20090305-C00038
  • R4 is alkyl, aryl, or heteroaryl; and R5 is
  • Figure US20090062328A1-20090305-C00039
  • For certain embodiments, including any one of the above embodiments, R1 is selected from the group consisting of 2-hydroxy-2-methylpropyl, 2-methylpropyl, propyl, ethyl, methyl, 2,3-dihydroxypropyl, 2-phenoxyethyl, 4-[(methylsulfonyl)amino]butyl, 2-methyl-2-[(methylsulfonyl)amino]propyl, 2-(acetylamino)-2-methylpropyl, 2-{[(isopropylamino)carbonyl]amino}-2-methylpropyl, 4-{[(isopropylamino)carbonyl]amino}butyl, 4-(1,1-dioxidoisothiazolidin-2-yl)butyl, tetrahydro-2H-pyran-4-ylmethyl, and (2,2-dimethyl-1,3-dioxolan-4-yl)methyl. For certain of these embodiments, R1 is tetrahydro-2H-pyran-4-ylmethyl.
  • For certain embodiments, including any one of the above embodiments, except where this definition is excluded, R1 is selected from the group consisting of (1-hydroxycyclobutyl)methyl, (1-hydroxycyclopentyl)methyl, and (1-hydroxycyclohexyl)methyl. For certain of these embodiments, R1 is (1-hydroxycyclobutyl)methyl.
  • For certain embodiments, R is selected from the group consisting of halogen, hydroxy, alkyl, alkenyl, haloalkyl, alkoxy, alkylthio, and —N(R9)2.
  • For certain embodiments, R is selected from the group consisting of alkyl, alkoxy, hydroxy, halogen, and trifluoromethyl.
  • For certain embodiments, R is halogen or hydroxy.
  • For certain embodiments, R is bromine.
  • For certain embodiments, R2-1, R2-2, and R2-3 are independently selected from the group consisting of hydrogen, alkyl, alkenyl, aryl, arylalkylenyl, heteroaryl, heteroarylalkylenyl, heterocyclyl, heterocyclylalkylenyl, and alkyl, alkenyl, aryl, arylalkylenyl, heteroaryl, heteroarylalkylenyl, heterocyclyl, or heterocyclylalkylenyl, substituted by one or more substituents selected from the group consisting of hydroxy, alkyl, haloalkyl, hydroxyalkyl, alkoxy, dialkylamino, —S(O)0-2—R2-5, —NH—S(O)2—R2-5, haloalkoxy, halogen, cyano, nitro, —N3, aryl, heteroaryl, heterocyclyl, aryloxy, arylalkyleneoxy, —C(O)—O-alkyl, —C(O)—N(R8)2, —N(R)—C(O)—R2-5, —NH—C(O)—NH—R2-5, —NH—C(O)—NH2, —O—(CO)-alkyl, and —C(O)-alkyl; with the proviso that R2-2 is other than alkenyl wherein the carbon atom bonded to —O— is doubly bonded to another carbon atom;
  • For certain embodiments, R2-1 is selected from the group consisting of hydrogen, alkyl, and aryl.
  • For certain embodiments, R2-1 is hydrogen, C1-4 alkyl, or phenyl.
  • For certain embodiments, R2-1 is hydrogen.
  • For certain embodiments, R2-2 is selected from the group consisting of hydrogen, alkyl, arylalkylenyl, and heteroarylalkylenyl.
  • For certain embodiments, R2-2 is hydrogen, C1-4 alkyl, benzyl, or pyridin-2-ylmethyl.
  • For certain embodiments, R2-2 is C1-10 alkyl.
  • For certain embodiments, R2-2 is methyl.
  • For certain embodiments, R2-2 is hydrogen.
  • For certain embodiments, R2-3 is selected from the group consisting of hydrogen and alkyl.
  • For certain embodiments, R2-3 is alkyl.
  • For certain embodiments, R2-3 is hydrogen or methyl.
  • For certain embodiments, R2-4 is selected from the group consisting of hydrogen, C1-4alkyl, and phenyl.
  • For certain embodiments, R2-4 is hydrogen.
  • For certain embodiments, R2-5 is selected from the group consisting of alkyl, aryl, arylalkylenyl, heteroaryl, and heteroarylalkylenyl, each of which is unsubstituted or substituted by one or more substituents selected from the group consisting of halogen, cyano, nitro, alkoxy, dialkylamino, alkylthio, haloalkyl, haloalkoxy, alkyl, and —N3.
  • For certain embodiments, R2-5 is hydrogen or alkyl.
  • For certain embodiments, R2-5 is hydrogen or C1-4 alkyl.
  • For certain embodiments, R3 is selected from the group consisting of -Z′-R4, -Z′-X′—R4, -Z′-X′—Y′—R4, -Z′-X′—Y′—X′—Y′—R4, and -Z′-X′—R5.
  • For certain embodiments, R3 is selected from the group consisting of -Z′-R4, -Z′-X′—Y′—R4, and -Z′-X′—R5.
  • For certain embodiments, R3 is -Z′-R4.
  • For certain embodiments, R3 is -Z′-X′—Y′—R4.
  • For certain embodiments, R3 is -Z′-X′—R5.
  • For certain embodiments, R3 is selected from the group consisting of phenyl, pyridin-3-yl, pyridin-4-yl, 5-(hydroxymethyl)pyridin-3-yl, 2-ethoxyphenyl, 3-(morpholine-4-carbonyl)phenyl, and 3-(NAN-dimethylaminocarbonyl)phenyl.
  • For certain embodiments, including any of the above embodiments where R3 is present, —R3 is at the 7-position.
  • 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, alkylamino, dialkylamino, (dialkylamino)alkyleneoxy, and in the case of alkyl, alkenyl, alkynyl, and heterocyclyl, oxo.
  • For certain embodiments, R4 is alkyl, aryl, or heteroaryl.
  • For certain embodiments, R4 is hydrogen or alkyl.
  • For certain embodiments, R4 is C1-4 alkyl.
  • For certain embodiments, R5 is selected from the group consisting of:
  • Figure US20090062328A1-20090305-C00040
  • For certain embodiments, R5 is selected from the group consisting of:
  • Figure US20090062328A1-20090305-C00041
  • For certain embodiments, R5 is
  • Figure US20090062328A1-20090305-C00042
  • For certain embodiments, R5 is
  • Figure US20090062328A1-20090305-C00043
  • wherein V is —C(O)—, and A is —O—.
  • For certain embodiments, R6 is selected from the group consisting of ═O and —S.
  • For certain embodiments, R6 is ═O.
  • For certain embodiments, R7 is C2-7 alkylene.
  • For certain embodiments, R7 is C2-4 alkylene.
  • For certain embodiments, R8 is selected from the group consisting of hydrogen, C1-10 alkyl, C2-10 alkenyl, C1-10 alkoxy-C1-10 alkylenyl, hydroxy-C1-10 alkylenyl, heteroaryl-C1-10 alkylenyl, and aryl-C1-10 alkylenyl.
  • For certain embodiments, R8 is selected from the group consisting of hydrogen, C1-4 alkyl, and C1-4 alkoxyC1-4 alkylenyl.
  • For certain embodiments, R8 is hydrogen or C1-4 alkyl.
  • For certain embodiments, R8 is hydrogen.
  • For certain embodiments, R9 is selected from the group consisting of hydrogen and alkyl.
  • For certain embodiments, R10 is C3-48 alkylene.
  • For certain embodiments, R10 is C4-4 alkylene.
  • For certain embodiments, A is selected from the group consisting of —O—, —C(O)—, —S(O)0-2—, —CH2—, and —N(-Q-R4)—.
  • For certain embodiments, A is —O—, —CH2—, or —S(O)2—.
  • For certain embodiments, A is —O— or —S(O)2—.
  • For certain embodiments, A is —O—.
  • For certain embodiments, A′ is selected from the group consisting of —O—, —S(O)0-2—, —N(-Q-R4)—, and —CH2—.
  • In certain embodiments, A′ is selected from the group consisting of —CH2—, —S(O)2—, and —O—.
  • In certain embodiments, A′ is —N(-Q-R4)—.
  • In certain embodiments, A′ is —CH2—.
  • In certain embodiments, A′ is —O—.
  • For certain embodiments, including any one of the above embodiments of Formula VII, G 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(═NY1)—R″, —CH(OH)—C(O)—OY1, —CH(OC1-4 alkyl)Y0, —CH2Y2, and —CH(CH3)Y2. For certain of these embodiments, R″ and R″″ are independently selected from the group consisting of C1-10 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, C1-4 alkoxy, aryl, heteroaryl, aryl-C1-4 alkylenyl, heteroaryl-C1-4 alkylenyl, halo-C1-4 alkylenyl, halo-C1-4 alkoxy, —O—C(O)—CH3, —C(O)—O—CH3, —C(O)—NH2, —O—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 α-amino acid selected from the group consisting of racemic, D-, and L-amino acids; Y1 is selected from the group consisting of hydrogen, C1-6 alkyl, and benzyl; Y0 is selected from the group consisting of C1-6 alkyl, carboxy-C1-6 alkylenyl, amino-C1-4 alkylenyl, mono-N—C1-6 alkylamino-CIA alkylenyl, and di-N,N—C1-6 alkylamino-C1-4 alkylenyl; and Y2 is selected from the group consisting of mono-N—C1-6 alkylamino, di-N,N—C1-6 alkylamino, morpholin-4-yl, piperidin-1-yl, pyrrolidin-1-yl, and 4-C1-4 alkylpiperazin-1-yl.
  • For certain embodiments, including any one of the above embodiments of Formula VII, G is selected from the group consisting of —C(O)—R″, α-aminoacyl, and —C(O)—O—R″.
  • For certain embodiments, including any one of the above embodiments of Formula VII, G is selected from the group consisting of —C(O)—R″, α-amino-C2-11 acyl, and —C(O)—O—R″. α-Amino-C2-11 acyl includes α-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 of these embodiments, R″ contains one to ten carbon atoms.
  • 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 occurring 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 α-amino acid is selected from the group consisting of racemic, D-, and L-amino acids.
  • In certain embodiments, 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(R6)—O—, —C(R6)—S—, and —C(R6)—N(OR9)—.
  • In certain embodiments, Q is selected from the group consisting of a bond, —C(O)—, —S(O)2—, and —C(R6)—N(R8)—.
  • In certain embodiments, Q is —C(O)—.
  • In certain embodiments, V is selected from the group consisting of —C(R6)—, —O—C(R6)—, —N(R8)—C(R6)—, and —S(O)2—.
  • In certain embodiments, V is selected from the group consisting of —C(O)— and —N(R8)—C(O)—.
  • In certain embodiments, V is —C(O)—.
  • In certain embodiments, W is selected from the group consisting of a bond, —C(O)—, and —S(O)2—.
  • In certain embodiments, W is a bond.
  • For certain embodiments, X is selected from the group consisting of a bond, C1-4 alkylene and C2-4 alkenylene.
  • For certain embodiments, X is a bond or C1-4 alkylene.
  • For certain embodiments, X is a bond, methylene, or ethylene.
  • In certain embodiments, X′ is selected from the group consisting of alkylene, alkenylene, alkynylene, arylene, heteroarylene, and heterocyclylene, wherein the alkylene, alkenylene, and alkynylene groups can be optionally interrupted or terminated by arylene, heteroarylene or heterocyclylene and optionally interrupted by one or more —O— groups.
  • In certain embodiments, X′ is alkylene.
  • In certain embodiments, X′ is phenylene.
  • In certain embodiments, Y is selected from the group consisting of a bond, —C(R6)—, —S(O)2—, —S(O)2—N(R8)—,
  • Figure US20090062328A1-20090305-C00044
  • —C(O)—O—, —C(R6)—N(R8)—, —C(O)—N(R8)—S(O)2—, —C(R6)—N(R8)—C(O)—,
  • Figure US20090062328A1-20090305-C00045
  • —C(O)—C(O)—, —C(O)—C(O)—O—, and —C(═NH)—N(R8)—.
  • In certain embodiments, Y is selected from the group consisting of —C(O)—, —S(O)2—, —S(O)2—N(R8)—, —C(O)—O—, and —C(O)—N(R8)—.
  • In certain embodiments, Y is selected from the group consisting of —C(O)—, —S(O)2—, and —C(O)—N(H)—.
  • In certain embodiments, Y is a bond.
  • In certain embodiments, Y′ is selected from the group consisting of —O—, —S(O)0-2—, —S(O)2—N(R8)—, —C(R6)—, —C(R6)—O—, —O—C(R6)—, —O—C(O)—O—, —N(R8)-Q-, —C(R6)—N(R8)—, —O—C(R6)—N(R8)—, —C(R6)—N(OR9)—, —O—N(R8)-Q-, —O—N═C(R4)—, —C(═N—O—R8)—, —CH(—N(—O—R8)-Q-R4)—,
  • Figure US20090062328A1-20090305-C00046
  • —V—NN—C(R6)—;
  • In certain embodiments, Y′ is —N(R8)—C(O)—, —N(R8)—S(O)2—, —N(R8)—C(O)—N(R8)—, or
  • Figure US20090062328A1-20090305-C00047
  • In certain embodiments, Z′ is a bond or —O—.
  • In certain embodiments, Z′ is a bond.
  • In certain embodiments, Z′ is —O—.
  • In certain embodiments, a and b are independently integers from 1 to 6 with the proviso that a+b is ≦7.
  • In certain embodiments, a and b are each 2.
  • In certain embodiments, n is an integer form 0 to 4.
  • In certain embodiments, n is 0 or 1.
  • In certain embodiments, n is 0.
  • In certain embodiments, p is an integer form 0 to 3.
  • In certain embodiments, p is 0 or 1.
  • In certain embodiments, p is 0.
  • In certain embodiments, m is 0 or 1; with the proviso that when m is 1, then n is 0 or 1.
  • In certain embodiments, m is 0 or 1; with the proviso that when m is 1, then p is 0 or 1.
  • In certain embodiments, m is 0.
  • In certain embodiments, m is 1.
  • For certain embodiments, the present invention provides a pharmaceutical composition comprising a therapeutically effective amount of a compound or salt of any one of the above embodiments of Formulas I, II, III, IV, V, VI, VII, and VIIIa, and a pharmaceutically acceptable carrier.
  • For certain 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 the above embodiments of Formulas I, II, III, IV, V, VI, VII, and VIIIa, or a pharmaceutical composition comprising an effective amount of a compound or salt of any one of the above embodiments of Formulas I, II, III, IV, V, VI, VII, and VIIIa to the animal. For certain of these embodiments, the cytokine is selected from the group consisting of IFN-α, TNF-α, IL-6, IL-10, and IL-12. For certain of these embodiments, the cytokine is IFN-α or TNF-α. For certain of these embodiments, the cytokine is IFN-α.
  • For certain 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 the above embodiments of Formulas I, II, III, IV, V, VI, VII, and VIIIa, or a pharmaceutical composition comprising a therapeutically effective amount of a compound or salt of any one of the above embodiments of Formulas I, II, III, IV, V, VI, VII, and VIIIa to the animal.
  • For certain 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 the above embodiments of Formulas I, II, III, IV, V, VI, VII, and VIIIa, or a pharmaceutical composition comprising a therapeutically effective amount of a compound or salt of any one of the above embodiments of Formulas I, II, III, IV, V, VI, VII, and VIIIa 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, Wis., 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, tert-butoxycarbonyl (Boc), benzyloxycarbonyl, and 9-fluorenylmethoxycarbonyl (Fmoc). Suitable hydroxy protecting groups include acetyl and silyl groups such as the tert-butyl 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, USA, 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.
  • Compounds of the invention can be prepared according to Reaction Scheme I where R, R1, R2-2, R2-3, X, Y, and n are as defined above, and Hal is chloro, bromo, or iodo. In step (1) of Reaction Scheme I, a quinoline-3,4-diamine of Formula X is reacted with a carboxylic acid equivalent, which is selected such that it will provide the desired —X—CH2-Hal substituent in a 1H-imidazo[4,5-c]quinoline of Formula XI. Suitable carboxylic acid equivalents include ortho esters, acid halides, and imidates or salts thereof. Many compounds of Formula X are known and can be readily prepared using known synthetic routes; see for example, U.S. Pat. Nos. 4,689,338 (Gerster), 4,929,624 (Gerster et al.), 5,268,376 (Gerster), 5,389,640 (Gerster et al.), 6,331,539 (Crooks et al.), 6,451,810 (Coleman et al.), 6,541,485 (Crooks et al.), 6,660,747 (Crooks et al.), 6,670,372 (Charles et al.), 6,683,088 (Crooks et al.), 6,656,938 (Crooks et al.), 6,664,264 (Dellaria et al.), 6,677,349 (Griesgraber); and U.S. Patent Publication Application No. US 2004/0147543 (Hays et al.).
  • When the carboxylic acid equivalent used in step (1) is an imidate of formula Hal-CH2—X—C(═NH)—O-alkyl or a salt thereof, the reaction is conveniently carried out by combining a quinoline-3,4-diamine of Formula X with the imidate in a suitable solvent such 1,2-dichloroethane or chloroform. The reaction can be carried out at an elevated temperature such as 80° C. or the reflux temperature of the solvent. The product can be isolated by conventional methods. Some imidates of formula Hal-CH2—X—C(═NH)—O-alkyl are known; others can be prepared by known methods. Ethyl chloroacetimidate hydrochloride, which can be used to provide a compound of Formula XI in which X is a bond, is a known compound that can be prepared according to the literature procedure: Stillings, M. R. et al., J. Med. Chem., 29, pp. 2280-2284 (1986).
  • When the carboxylic acid equivalent is an acid halide of formula Hal-CH2—X—C(O)Cl or Hal-CH2—X—C(O)Br, the reaction is conveniently carried out by adding the acid halide to a solution of a quinoline-3,4-diamine of Formula X in a suitable solvent such as dichloromethane or 1,2-dichloroethane in the presence of a tertiary amine such as triethylamine. The reaction can be carried out at ambient temperature or at a sub-ambient temperature. The product can be isolated by conventional methods.
  • The reaction with an acid halide of formula Hal-CH2—X—C(O)Cl or Hal-CH2—X—C(O)Br may be carried out in two parts, which include (i) adding the acid halide to a solution of a quinoline-3,4-diamine of Formula X in a suitable solvent such as dichloromethane or 1,2-dichloroethane optionally in the presence of a tertiary amine such as triethylamine to afford an amide intermediate and (ii) cyclizing to provide a 1H-imidazo[4,5-c]quinoline of Formula XI. The amide intermediate from part (i) can be optionally isolated using conventional techniques. The cyclization in part (ii) may be carried out by heating the amide intermediate from part (i) in a suitable solvent such as toluene. The cyclization in part (ii) can also be carried out in the presence of a base such as triethylamine.
  • In step (2) of Reaction Scheme I a 1H-imidazo[4,5-c]quinoline of Formula XI is oxidized to provide an N-oxide of Formula XII using a conventional oxidizing agent that is capable of forming N-oxides. The reaction can be carried out by treating a solution of a compound of Formula XI in a suitable solvent such as chloroform or dichloromethane with 3-chloroperoxybenzoic acid at room temperature, and the product can be isolated by conventional methods.
  • In step (3) of Reaction Scheme I, a 1H-imidazo[4,5-c]quinoline-5N-oxide of Formula XII is aminated to provide a 1H-imidazo[4,5-c]quinolin-4-amine of Formula XIII. Step (3) involves the activation of an N-oxide of Formula XII by conversion to an ester and then reacting the ester with an aminating agent. Suitable activating agents include alkyl- or arylsulfonyl chlorides such as benzenesulfonyl chloride, methanesulfonyl chloride, or p-toluenesulfonyl chloride. Suitable aminating agents include ammonia, in the form of ammonium hydroxide, for example, and ammonium salts such as ammonium carbonate, ammonium bicarbonate, and ammonium phosphate. The reaction is conveniently carried out by adding ammonium hydroxide to a solution of the N-oxide of Formula XII in a suitable solvent such as dichloromethane or chloroform and then adding p-toluenesulfonyl chloride. The reaction can be carried out at room temperature, and the product or a pharmaceutically acceptable salt thereof can be isolated from the reaction mixture using conventional methods.
  • Alternatively, the oxidation and amination can be carried out as a one-pot procedure without isolating the N-oxide of Formula XII by adding 3-chloroperoxybenzoic acid to a solution of a compound of Formula XI in a solvent such as dichloromethane or chloroform and then adding ammonium hydroxide and p-toluenesulfonyl chloride. The product of Formula XIII or a pharmaceutically acceptable salt thereof can be isolated by conventional methods. Some compounds of Formula XIII are known, see for example, International Publication Nos. WO2005/048933 and WO2005/048945.
  • In step (4) of Reaction Scheme I, the Hal group of a 1H-imidazo[4,5-c]quinolin-4-amine of Formula XIII is displaced with a hydroxylamine of formula HN(Y—R2-3)OR2-2 or a salt thereof. The reaction is conveniently carried out by combining a hydroxylamine salt of the formula HN(Y—R2-3)OR2-2—HCl with a compound of Formula XIII in a suitable solvent, such as N,N-dimethylformamide (DMF), in the presence of a base such as triethylamine. The reaction can be carried out at room temperature or at an elevated temperature such as 50° C. Some hydroxylamine salts of the formula HN(Y—R2-3)OR22—HCl can be obtained commercially. For example N,O-dimethylhydroxylamine hydrochloride, methoxylamine hydrochloride, and N-methylhydroxylamine hydrochloride are commercially available compounds that can be used to make preferred compounds of Formula XIV, wherein Y is a bond. Other hydroxylamine salts of the formula HN(Y—R2-3)OR2-2.HCl can be prepared using conventional synthetic methods. The product of Formula XIV, a subgenus of Formulas I and III, or a pharmaceutically acceptable salt thereof can be isolated using conventional methods.
  • Figure US20090062328A1-20090305-C00048
  • Compounds of the invention can be prepared according to Reaction Scheme II where R, R1, R2-1, R2-2, R2-3, X, Y, and n are as defined above, and P is a hydroxy protecting group. In step (1) of Reaction Scheme II, a compound of Formula X or a salt thereof is reacted with a carboxylic acid or an equivalent thereof to provide a compound of Formula XV. Suitable equivalents to carboxylic acid include acid anhydrides of Formula O[C(O)—X—CH2—O—P]2 and acid chlorides of Formula Cl—C(O)—X—CH2—O—P. The reaction is conveniently carried out by using the conditions described in step (1) of Reaction Scheme I for the reaction with acid chlorides. Some compounds of Formula Cl—C(O)—X—O—P, such as acetoxyacetyl chloride, methoxyacetyl chloride, and 2-methoxypropionyl chloride, are commercially available. Others can be prepared by known synthetic methods.
  • In step (2) of Reaction Scheme II, the protecting group of a 1H-imidazo[4,5-c]quinoline of Formula XV is removed to provide a hydroxyalkyl-substituted 1H-imidazo[4,5-c]quinoline of Formula XVI. The deprotection can be carried out using a variety of methods depending on which P group is present. When P is —C(O)—CH3, the reaction is conveniently carried out by adding lithium hydroxide monohydrate to a solution or suspension of the compound of Formula XV in a suitable solvent or solvent system such as tetrahydrofuran:methanol:water. The reaction can be carried out at room temperature, and the product can be isolated by conventional methods.
  • In step (3) of Reaction Scheme II, a hydroxyalkyl-substituted 1H-imidazo[4,5-c]quinoline of Formula XVI is oxidized to an aldehyde-substituted 1H-imidazo[4,5-c]quinoline of Formula XVII using one of many conventional methods. The oxidation is conveniently carried out by adding Dess-Martin periodinane, [1,1,1-tris(acetyloxy)-1,1-dihydro-1,2-benziodoxol-3(1H)-one], to a solution or suspension of a hydroxyalkyl-substituted 1H-imidazo[4,5-c]quinoline of Formula XVI in a suitable solvent such as dichloromethane. The reaction can be carried out at room temperature, and the product can be isolated by conventional methods.
  • Alternatively, certain aldehyde-substituted 1H-imidazo[4,5-c]quinolines of Formula XVII in which X is a bond can be prepared from 1H-imidazo[4,5-c]quinolines with a hydrogen at the 2-position, many of which are known; see, for example, U.S. Pat. Nos. 4,689,338 (Gerster) and 5,268,376 (Gerster). The hydrogen at the 2-position of a 1H-imidazo[4,5-c]quinoline may undergo lithiation with butyllithium, and subsequent substitution with DMF provides a compound of Formula XVII in which X is a bond. The reaction is conveniently carried out in a suitable solvent such as THF at a subambient temperature such as −78° C. The product or a pharmaceutically acceptable salt thereof can be isolated by conventional methods.
  • In step (4) of Reaction Scheme II, an aldehyde-substituted 1H-imidazo[4,5-c]quinoline of Formula XVII is converted to an aldoxime of Formula XVIII. The reaction is conveniently carried out by adding a hydroxylamine salt of the formula NH2OR2-2.HCl, optionally in a suitable solvent such as water, to a solution or suspension of a compound of Formula XVII, in a suitable solvent, such as ethanol or methanol. Optionally a base such as aqueous sodium hydroxide can be added. The reaction can be carried out at room temperature or at an elevated temperature such as the reflux temperature of the solvent. Hydroxylamine salts of the formula NH2OR2-2.HCl can be obtained commercially or they can be prepared using conventional synthetic methods. The product or a pharmaceutically acceptable salt thereof is obtained as a mixture of E and Z isomers and can be isolated using conventional methods.
  • In steps (5) and (6) of Reaction Scheme II, an aldoxime-substituted 1H-imidazo[4,5-c]quinoline of Formula XVIII is first oxidized to an N-oxide of Formula XIX, which is then aminated to provide a compound of Formula XX, which is a subgenus of Formulas I and III. Steps (5) and (6) of Reaction Scheme II can be carried out according to the methods described in steps (2) and (3) of Reaction Scheme I, and the product can be isolated by conventional methods.
  • In step (7) of Reaction Scheme II, an aldoxime-substituted 1H-imidazo[4,5-c]quinolin-4-amine of Formula XX is treated with a Grignard reagent of the formula R2-1MgHalide to form a hydroxylamine of Formula XXI, a subgenus of Formulas I and III. Several Grignard reagents are commercially available; others can be prepared using known synthetic methods. The reaction is conveniently carried out by adding a solution of two equivalents of the Grignard reagent to a solution of the compound of Formula XX in a suitable solvent such as THF. The reaction can be carried out at room temperature, and the product can be isolated using conventional methods. Alternatively, to prepare a compound of Formula XXI wherein R2-1 is hydrogen, an oxime of Formula XX can be treated with a hydride reducing agent. The reduction is conveniently carried out by treating an oxime of Formula XX with excess sodium cyanoborohydride in a suitable solvent or solvent mixture such as methanol/acetic acid. The reaction can be carried out at ambient temperature. The product or a pharmaceutically acceptable salt thereof can be isolated using conventional methods.
  • In step (8) of Reaction Scheme II, a hydroxylamine of Formula XXI is converted to a compound of Formula XXII, a subgenus of Formulas I and III. Step (8) is carried out using conventional methods. For example, sulfonamides of Formula XII (Y is —S(O)2—) can be prepared by reacting a compound of Formula XXI with a sulfonyl chloride of formula R2-3S(O)2Cl or a sulfonic anhydride of Formula [R2-3S(O)2]2O. The reaction can be carried out at room temperature in an inert solvent such as chloroform, dichloromethane, or N,N-dimethylacetamide (DMA) by adding the sulfonyl chloride or sulfonic anhydride to a compound of Formula XI in the presence of a base such as N,N-diisopropylethylamine, triethylamine, or pyridine.
  • Sulfamides of Formula XXII (Y is —S(O)2—N(R)— or
  • Figure US20090062328A1-20090305-C00049
  • can be prepared by reacting a compound of Formula XXI with sulfuryl chloride to generate a sulfamoyl chloride in situ, and then reacting the sulfamoyl chloride with an amine of formula HN(R8)R2-3, or
  • Figure US20090062328A1-20090305-C00050
  • or by reacting a compound of Formula XXI with a sulfamoyl chloride of formula R2-3(R8)NS(O)2Cl or
  • Figure US20090062328A1-20090305-C00051
  • The product or a pharmaceutically acceptable salt thereof can be isolated using conventional methods. Many sulfonyl chlorides of formula R2-3S(O)2Cl, amines of formulas HN(R8)R2-3, and
  • Figure US20090062328A1-20090305-C00052
  • and some sulfamoyl chlorides of formulas R2-3(R8)NS(O)2Cl and
  • Figure US20090062328A1-20090305-C00053
  • are commercially available; others can be prepared using known synthetic methods.
  • Amides of Formula XXII (Y is —C(O)—) can be prepared from hydroxylamines of Formula XXI using conventional methods. For example, a compound of Formula XXI can be reacted with an acid chloride of formula R2-3C(O)Cl. The reaction can be carried out by adding the acid chloride to a solution of a compound of Formula XXI in a suitable solvent such as chloroform or DMA, optionally in the presence of a base such as N,N-diisopropylethylamine, triethylamine, or pyridine, at ambient temperature. The product or a pharmaceutically acceptable salt thereof can be isolated using conventional methods.
  • Ureas and thioureas of Formula XXII (Y is —C(O)—N(R8)—, —C(S)—N(R8)—, —C(O)—N(R8)—S(O)2—, —C(O)—N(R8)—C(O)—, —C(S)—N(R8)—C(O)—, or
  • Figure US20090062328A1-20090305-C00054
  • can be prepared from hydroxylamines of Formula XXI using conventional methods. For example, a compound of Formula VI can be reacted with an isocyanate of formula R2-3N═C═O. The reaction can be carried out by adding the isocyanate to a solution of a compound of Formula XXI in a suitable solvent such as chloroform or DMA, optionally in the presence of a base such as N,N-diisopropylethylamine, or triethylamine, at room temperature. Alternatively, a compound of Formula XXI can be reacted with a thioisocyanate of formula R2-3N═C═S, a sulfonyl isocyanate of formula R2-3S(O)2N═C═O or a carbamoyl chloride of formula R2-3NC(O)Cl or
  • Figure US20090062328A1-20090305-C00055
  • The product or a pharmaceutically acceptable salt thereof can be isolated using conventional methods.
  • Reaction Scheme II can be modified after step (3) to introduce a R2-4 group that is other than hydrogen. In this modification, an aldehyde-substituted 1H-imidazo[4,5-c]quinoline of Formula XVII is treated with a Grignard reagent of the formula R2-4MgHalide, which adds to the aldehyde to form a secondary alcohol. Several Grignard reagents are commercially available; others can be prepared using known synthetic methods. The reaction is conveniently carried out by adding a solution of the Grignard reagent to a solution of the compound of Formula XVII in a suitable solvent such as THF. The reaction can be carried out at room temperature, and the product can be isolated using conventional methods. The secondary alcohol is then oxidized to a ketone using conventional methods. The reaction is conveniently carried out using Dess-Martin periodinane under the conditions described in step (3) of Reaction Scheme II. The reaction may also be carried out under Swern conditions by adding the secondary alcohol followed by triethylamine to a mixture of oxalyl chloride and dimethylsulfoxide in a suitable solvent such as dichloromethane. The reaction can be carried out at a sub-ambient temperature, and the product can be isolated by conventional methods. Steps (4) through (8) of Reaction Scheme II can then be carried out to provide a hydroxylamine of the invention with an R2-4 group that is other than hydrogen.
  • Figure US20090062328A1-20090305-C00056
  • Compounds of the invention can be prepared according to Reaction Scheme III, wherein R, R1, R2-1, R2-2, X, and n are as defined above. In step (1) of Reaction Scheme III, a quinoline-3,4-diamine of Formula X is reacted with a ketal-substituted carboxylic acid or equivalent thereof of the formula
  • Figure US20090062328A1-20090305-C00057
  • wherein D can be —OH, —Cl, Br, or a leaving group prepared using conventional hydroxy activation chemistry, such as employing N-hydroxysuccinimide as an activating agent. Ketals of this formula are readily prepared from esters of formula alkyl-O—C(O)—X—C(O)—R2-1 using conventional methods. For example, the ketone can be converted to a ketal by heating with ethylene glycol in the presence of pyridinium p-toluenesulfonate in a suitable solvent such as toluene. The carboxyl group can then be activated by first hydrolyzing the ester under basic conditions, for example with sodium hydroxide in water and a lower alcohol, and then reacting with N-hydroxysuccinimide in the presence of 4-methylmorpholine and 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride in a suitable solvent such as dichloromethane. The reaction shown in step (1) of Reaction Scheme III can be carried out according to the methods described for reaction with acid chlorides in step (1) of Reaction Scheme I, or it can be conveniently carried out by heating a quinoline-3,4-diamine of Formula X with a ketal shown above in a suitable solvent such as pyridine. The reaction can be run at the reflux temperature of the solvent, and the product of Formula XXIII can be isolated by conventional methods.
  • In step (2) of Reaction Scheme III, a 1H-imidazo[4,5-c]quinoline of Formula XXIII is converted to the N-oxide of Formula XXIV using the method described in step (2) of Reaction Scheme I.
  • In step (3) of Reaction Scheme III, the N-oxide of Formula XXIV is aminated to afford the compound of Formula XXV using one of the methods described in step (3) of Reaction Scheme I.
  • In step (4) of Reaction Scheme III, a ketal of Formula XXV is converted to a ketone of Formula XXVI by acid-catalyzed hydrolysis. The reaction is conveniently carried out by adding a strong acid, such as hydrochloric acid, to a ketal of Formula XXV. The reaction may be carried out at room temperature in a suitable solvent such as water, and the product can be isolated by conventional methods.
  • In step (5) of Reaction Scheme III, a ketone-substituted 1H-imidazo[4,5-c]quinolin-4-amine of Formula XXVI is converted to an oxime of Formula XXVII. The reaction is conveniently carried out as described in step (4) of Reaction Scheme II, and the product or a pharmaceutically acceptable salt thereof can be isolated by conventional methods.
  • Figure US20090062328A1-20090305-C00058
  • Any of Reaction Schemes I through III may be carried out using a [1,5]naphthyridine-3,4-diamine instead of a quinoline-3,4-diamine of Formula X as the starting material to prepare 1H-imidazo[4,5-c][1,5]naphthyridines of the invention. Several [1,5]naphthyridine-3,4-diamines and their preparation are known; see, for example, U.S. Pat. Nos. 6,194,425 (Gerster) and 6,518,280 (Gerster).
  • Compounds of the invention can be prepared according to Reaction Scheme IV, wherein R2-1, and Z are as defined above; Xb is selected from the group consisting of a bond and C1-4 alkylene; Rb is selected from the group consisting of hydroxy, alkyl, alkoxy, —N(R9)2; n is 0 to 4; and R1b is a subset of R1 as defined above that does not include those substituents that one skilled in the art would recognize as being susceptible to reduction under the acidic hydrogenation conditions of the reaction. These susceptible groups include, for example, alkenyl, alkynyl, and aryl groups and groups bearing nitro substituents. Compounds of Formula XXVIII can be prepared by the oxidation and amination of a compound of Formula XVI according to steps (2) and (3) of Reaction Scheme I. In step (1) of Reaction Scheme IV, a compound of Formula XXVIII is reduced to a 6,7,8,9-tetrahydro compound of Formula XXIX. The reaction is conveniently carried out under hetereogeneous hydrogenation conditions by adding platinum (IV) oxide to a solution of the compound of Formula XXVIII in trifluoroacetic acid and placing the reaction under hydrogen pressure. The reaction can be carried out on a Parr apparatus at room temperature. The product or a pharmaceutically acceptable salt thereof can be isolated by conventional methods. Steps (2) through (6) of Reaction Scheme IV can then be used to convert a compound of Formula XXIX to a compound of Formula IVb, a subgenus of Formulas I and IV. Steps (2) through (6) can be carried out, for example, according to steps (3), (4), (7), and (8) of Reaction Scheme II. Compounds of Formula IVb can also be made by treating a compound of Formula XXIX with thionyl chloride under conventional conditions to provide a chloroalkyl-substituted 6,7,8,9-tetrahydro compound, which can then be treated according to step (4) of Reaction Scheme I. The product of Formula IVb or a pharmaceutically acceptable salt thereof can be isolated by conventional methods. 6,7,8,9-Tetrahydro-1H-imidazo[4,5-c][1,5]naphthyridin-4-amines can also be prepared using this Reaction Scheme.
  • Figure US20090062328A1-20090305-C00059
  • Compounds of the invention can be prepared according to Reaction Scheme V, wherein R, R1, R2-1, X, and Z are as defined above; n is 0 or 1; hal is —Br or —I; and R3a and R3b are as defined below. Compounds of Formula XXX, a subgenus of Formulas I and III, can be prepared according to the methods described in Reaction Scheme I, II, or III beginning with a compound of Formula X, wherein one of the R groups is —Br, or —I. These halogen-substituted quinolines are known or can be prepared by known methods; see, for example, U.S. Patent Application Publication No. US 2004/0147543 (Hays et al.) and the references cited therein.
  • In step (1) of Reaction Scheme V, a halogen-substituted 1H-imidazo[4,5-c]quinolin-4-amine of Formula X) can undergo known palladium-catalyzed coupling reactions such as the Suzuki coupling and the Heck reaction. For example, a halogen-substituted compound of Formula XXX undergoes Suzuki coupling with a boronic acid of Formula R3a—B(OH)2, an anhydride thereof, or a boronic acid ester of Formula R3a—B(O-alkyl)2, wherein R3a is —R4b, —X′a—R4, —X′b—Y′—R4, or —X′b—R5; where X′a is alkenylene; X′b is arylene, heteroarylene, and alkenylene interrupted or terminated by arylene or heteroarylene; R4b is aryl or heteroaryl where the aryl or heteroaryl groups can be unsubstituted or substituted as defined in R4 above; and R4, R5, and Y′ are as defined above. The Suzuki coupling is conveniently carried out by combining a compound of Formula XXX with a boronic acid or an ester or anhydride thereof in the presence of palladium (II) acetate, triphenylphosphine, and a base such as sodium carbonate in a suitable solvent such as n-propanol or solvent mixture such as n-propanol/water. The reaction can be carried out at an elevated temperature (e.g., 80-100° C.). Many boronic acids of Formula R3a—B(OH)2, anhydrides thereof, and boronic acid esters of Formula R3a—B(O-alkyl)2 are commercially available; others can be readily prepared using known synthetic methods. See, for example, Li, W. et al, J. Org. Chem., 67, 5394-5397 (2002). The product of Formula IIIb, a subgenus of Formulas I and III, or a pharmaceutically acceptable salt thereof can be isolated by conventional methods.
  • The Heck reaction can also be used in step (1) of Reaction Scheme V to provide compounds of Formula IIIb, wherein R3a is —X′a—R4b or —X′a—Y′—R4, wherein X′a, Y′, R4b, and R4 are as defined above. The Heck reaction is carried out by coupling a compound of Formula XXX with a compound of the Formula H2C═C(H)—R4b or H2C═C(H)—Y′—R4. Several of these vinyl-substituted compounds are commercially available; others can be prepared by known methods. The reaction is conveniently carried out by combining the compound of Formula XXX and the vinyl-substituted compound in the presence of palladium (II) acetate, triphenylphosphine or tri-ortho-tolylphosphine, and a base such as triethylamine in a suitable solvent such as acetonitrile or toluene. The reaction can be carried out at an elevated temperature such as 100° C.-120° C. under an inert atmosphere. The product of Formula IIIb or a pharmaceutically acceptable salt thereof can be isolated using conventional methods.
  • Compounds of Formula IIIb, wherein R3a is —X′c—R4, X′c is alkynylene, and R4 is as defined above, can also be prepared by palladium catalyzed coupling reactions such as the Stille coupling or Sonogashira coupling. These reactions are carried out by coupling a compound of Formula XXX with a compound of the Formula (alkyl)3Sn—C≡C—R4, (alkyl)3Si—C≡C—R4, or H—C≡C—R4.
  • Some compounds of Formula IIIb prepared as described above by palladium-mediated coupling reactions, wherein R3a is —X′a—R4, —X′a—Y′—R4, —X′b2—Y′—R4, —X′b2—R5, or —X′c—R4, where X′b2 is alkenylene interrupted or terminated by arylene or heteroarylene, and X′a, X′c, Y′, R4, and R5 are as defined above, can undergo reduction of the alkenylene or alkynylene group present in step (2) of Reaction Scheme V to provide compounds of Formula IIIc wherein R3b is —X′d—R4, —X′d—Y′—R4, —X′e—Y′—R4, or —X′e—R5, where X′d is alkylene; X′e is alkylene interrupted or terminated by arylene or heteroarylene; and R4, R5, and Y′ are as defined above. The reduction can be carried out by hydrogenation using a conventional heterogeneous hydrogenation catalyst such as palladium on carbon. The reaction can conveniently be carried out on a Parr apparatus in a suitable solvent such as ethanol, methanol, or mixtures thereof. The product of Formula IIIc, a subgenus of Formulas I and III, or a pharmaceutically acceptable salt thereof can be isolated using conventional methods.
  • Figure US20090062328A1-20090305-C00060
  • For some embodiments, compounds of the invention are prepared according to Reaction Scheme VI, where R1, R2-1, R2-2, RA1, RB1, and X are as defined above, and Za is —C(R2-4)(—N(—OR2-2)—Y—R2-3)— and Ph is phenyl. Steps (1) through (4) of Reaction Scheme VI can be carried out as described in steps (1) through (4) of Reaction Scheme II starting with a compound of Formula XXI. Many tetrazolo[1,5-a]pyridines of Formula XXXI are known; others can be prepared by known methods. See, for example, PCT Publication Nos. WO 2004/110991 (Lindstrom et al.), WO 2004/110992 (Lindstrom et al.), and U.S. Pat. No. 6,797,718 (Dellaria et al.).
  • In step (5) of Reaction Scheme VI, the tetrazolo ring is removed from a 7H-imidazo[4,5-c]tetrazolo[1,5-a]pyridine of Formula XXII by reaction with triphenylphosphine to form an N-triphenylphosphinyl intermediate of Formula XIII. The reaction with triphenylphosphine can be run in a suitable solvent such as toluene or 1,2-dichlorobenzene under an atmosphere of nitrogen with heating, for example at the reflux temperature.
  • In step (6) of Reaction Scheme VI, an N-triphenylphosphinyl intermediate of Formula XXXIII is hydrolyzed to provide an oxime-substituted 1H-imidazo[4,5-c]pyridin-4-amine of Formula XXXIV, a subgenus of Formulas I and II. The hydrolysis can be carried out by general methods well known to those skilled in the art, for example, by heating in a lower alkanol or an alkanol/water solution in the presence of an acid such as trifluoroacetic acid, acetic acid, or hydrochloric acid. The product can be isolated from the reaction mixture using conventional methods as the compound of Formula XXXIV or as a pharmaceutically acceptable salt thereof.
  • When appropriate, the methods shown in steps (7) and (8) of Reaction Scheme II may be used to convert a compound of Formula XXIV to a compound of Formula IIb using steps (7) and (8) of Reaction Scheme VI. The product or a pharmaceutically acceptable salt thereof can be isolated by conventional methods.
  • Figure US20090062328A1-20090305-C00061
  • Compounds of the invention can be prepared according to Reaction Scheme VII where R1, R2-1, R2-2, R, X, and Za are as defined above; E is carbon (imidazoquinoline ring) or nitrogen (imidazonaphthyridine ring); n is 0 or 1; Bn is benzyl; and R3c is —O—R4, —O—X′—R4, —O—X′—Y′—R4, —O—X′—Y′—X′—Y′—R4, or —O—X′—R5, where R4, X′, Y′, and R5 are as defined above. In step (1) of Reaction Scheme VII, an aniline or aminopyridine of Formula XXXV is treated with the condensation product generated from 2,2-dimethyl-1,3-dioxane-4,6-dione (Meldrum's acid) and triethyl orthoformate to provide an imine of Formula XXXVI. The reaction is conveniently carried out by adding a solution of an aniline or aminopyridine of Formula XXXV to a heated mixture of Meldrum's acid and triethyl orthoformate and heating the reaction at an elevated temperature. The product can be isolated using conventional methods. Many anilines and aminopyridines of Formula XXXV are commercially available; others can be prepared by known synthetic methods. For example, benzyloxypyridines of Formula XXXV can be prepared using the method of Holladay et al., Biorg. Med. Chem. Lett., 8, pp. 2797-2802, (1998).
  • In step (2) of Reaction Scheme VII, an imine of Formula XVI undergoes thermolysis and cyclization to provide a compound of Formula XXXVII. The reaction is conveniently carried out in a medium such as DOWTHERM A heat transfer fluid at a temperature in the range of 200 to 250° C. The product can be isolated using conventional methods. Isomers of the compound of Formula XV or Formula XXVII, wherein E is nitrogen and at a different position in the ring, can also be synthesized and can be used to prepare compounds of the invention.
  • In step (3) of Reaction Scheme VII, a compound of Formula XXXVII is nitrated under conventional nitration conditions to provide a compound of Formula XXVIII. The reaction is conveniently carried out by adding nitric acid to the compound of Formula XXXVII in a suitable solvent such as propionic acid and heating the mixture at an elevated temperature. The product can be isolated using conventional methods.
  • In step (4) of Reaction Scheme VII, a 3-nitro[1,5]naphthyridin-4-ol or 3-nitroquinolin-4-ol of Formula XXVIII is chlorinated using conventional chlorination chemistry to provide a 4-chloro-3-nitro[1,5]naphthyridine or 4-chloro-3-nitroquinoline of Formula XXXIX. The reaction is conveniently carried out by treating the compound of Formula XXXVIII with phosphorous oxychloride in a suitable solvent such as DMF. The reaction can be carried out at ambient temperature or at an elevated temperature such as 100° C., and the product can be isolated using conventional methods.
  • In step (5) of Reaction Scheme VII, a 4-chloro-3-nitro[1,5]naphthyridine or 4-chloro-3-nitroquinoline of Formula XXI is treated with an amine of Formula R1—NH2 to provide a compound of Formula XL. Several amines of Formula R1—NH2 are commercially available; others can be prepared by known synthetic methods. The reaction is conveniently carried out by adding the amine of Formula R1—NH2 to a solution of the 4-chloro-3-nitro[1,5]naphthyridine or 4-chloro-3-nitroquinoline of Formula XXXIX in a suitable solvent such as dichloromethane in the presence of a tertiary amine such as triethylamine. The reaction can be carried out at ambient temperature or at a sub-ambient temperature such as, for example, 0° C. The reaction product can be isolated using conventional methods.
  • In step (6) of Reaction Scheme VII, a compound of Formula XL is reduced to provide a diamine of Formula XLI. The reaction can be carried out by hydrogenation using a heterogeneous hydrogenation catalyst such as platinum on carbon. The hydrogenation is conveniently carried out in a Parr apparatus in a suitable solvent such as toluene, methanol, acetonitrile, or ethyl acetate. The reaction can be carried out at ambient temperature, and the product can be isolated using conventional methods.
  • Alternatively, the reduction in step (6) can be carried out using nickel boride, prepared in situ from sodium borohydride and nickel(II) chloride. The reduction is conveniently carried out by adding a solution of a compound of Formula XL in a suitable solvent or solvent mixture such as dichloromethane/methanol to a mixture of excess sodium borohydride and catalytic nickel(II) chloride in methanol. The reaction can be carried out at ambient temperature. The product can be isolated using conventional methods.
  • Steps (7) through (12) of Reaction Scheme VII are analogous to steps (1) through (6) of Reaction Scheme II and can be carried out using the same methods.
  • In step (13) of Reaction Scheme VII, the benzyl group in an oxime of Formula XLII is cleaved to provide a hydroxy group. The cleavage is conveniently carried out on a Parr apparatus under hydrogenolysis conditions using a suitable heterogeneous catalyst such as palladium or platinum on carbon in a solvent such as ethanol. Alternatively, the cleavage can be carried out with an acid such as hydrogen bromide in a suitable solvent such as acetic acid at an elevated temperature, such as 65° C. The product of Formula XLIII, prepared by any of these methods, or a pharmaceutically acceptable salt thereof can be isolated using conventional methods.
  • In step (14) of Reaction Scheme VII, a hydroxy-substituted oxime of Formula XLIII is converted to a compound of Formula XLIV, a subgenus of Formula I, using a Williamson-type ether synthesis. The reaction is effected by treating a hydroxy-substituted compound of Formula XLIII with an aryl, alkyl, or arylalkylenyl halide of Formula Halide-R4b, Halide-alkylene-R4, Halide-alkylene-Y′—R4, or Halide-alkylene-R5, where R4b is as defined above, in the presence of a base. Numerous alkyl, arylalkylenyl, and aryl halides of these formulas are commercially available, including substituted benzyl bromides and chlorides, substituted or unsubstituted alkyl or arylalkylenyl bromides and chlorides, and substituted fluorobenzenes. Other halides of these formulas can be prepared using conventional synthetic methods. The reaction is conveniently carried out by combining an alkyl, arylalkylenyl, or aryl halide with the hydroxy-substituted compound of Formula XLIII in a solvent such as DMF in the presence of a suitable base such as cesium carbonate. Optionally, catalytic tetrabutylammonium bromide can be added. The reaction can be carried out at ambient temperature or at an elevated temperature, for example 65° C. or 85° C., depending on the reactivity of the halide reagent. Alternatively, step (14) may be carried out using the Ullmann ether synthesis, in which an alkali metal aryloxide prepared from the hydroxy-substituted compound of Formula XLIII reacts with an aryl halide in the presence of copper salts, to provide a compound of Formula XLIV, where R3b is —O—R4b, —O—X′f—R4, or —O—X′f—Y′—R4, wherein X′f is an arylene or heteroarylene, and R4b is as defined above. Numerous substituted and unsubstituted aryl halides are commercially available; others can be prepared using conventional methods. The product of Formula XLIV, prepared by either of these methods, or a pharmaceutically acceptable salt thereof can be isolated using conventional methods.
  • When appropriate, the methods shown in steps (7) and (8) of Reaction Scheme II may be used to convert a compound of Formula XLIV to a compound of Formula XLV using steps (15) and (16) of Reaction Scheme VII. The product or pharmaceutically acceptable salt thereof can be isolated by conventional methods.
  • Figure US20090062328A1-20090305-C00062
  • For some embodiments, naphthyridines of the invention are prepared from tetrazolo compounds of Formulas XLVI and L according to Reaction Schemes VIII and IX, wherein R1, R2-1, R2-2, R, n, Za, and X are as defined above. Compounds of Formula XLVI and L and synthetic routes to these compounds are known; see, for example, U.S. Pat. No. 6,194,425 (Gerster).
  • Steps (1) through (4) of Reaction Schemes VIII and IX are analogous to steps (1) through (4) of Reaction Scheme II and can be carried out using the same methods.
  • The tetrazolo group of a compound of Formula XLVII or LI can then be removed in step (5) of Reaction Scheme IX or X to provide a 1H-imidazo[4,5-c]naphthyridin-4-amine of Formula XLVIII or LII, each of which is a subgenus of Formula I. The removal of the tetrazolo group can be carried out as described in steps (5) and (6) of Reaction Scheme VI or by using methods described in U.S. Pat. No. 6,194,425 (Gerster). The product or a pharmaceutically acceptable salt thereof can be isolated by conventional methods.
  • When appropriate, the methods shown in steps (7) and (8) of Reaction Scheme II may be used to convert a compound of Formula XLVIII or LII to a compound of Formula XLIX or LIII using steps (6) and (7) of Reaction Scheme VIII or IX, respectively. The product of Formula XLIX or LIII, each of which is a subgenus of Formula I, or a pharmaceutically acceptable salt thereof can be isolated by conventional methods.
  • Figure US20090062328A1-20090305-C00063
  • Figure US20090062328A1-20090305-C00064
  • Compounds of the invention can be prepared according to Reaction Scheme X where RA1, RB1, R1, R2-2, R2-3, Y, and P are as defined above and PMB is para-methoxybenzyl.
  • In step (1) of Reaction Scheme X, a 2,4-dichloro-3-nitropyridine of Formula LIV is reacted with an amine of formula R1—NH2 to provide a 2-chloro-3-nitropyridin-4-amine of Formula LV. The reaction can be carried out using the method described in step (5) of Reaction Scheme VII. Some 2,4-dichloro-3-nitropyridines of Formula LIV are known; others can be prepared using known synthetic methods. See, for example, U.S. Pat. No. 6,525,064 (Dellaria) and the references cited therein.
  • In step (2) of Reaction Scheme X, a 2-chloro-3-nitropyridin-4-amine of Formula LV is reduced to provide a 2-chloropyridine-3,4-diamine of Formula LVI. The reduction can be carried out using the methods described in step (6) of Reaction Scheme VII.
  • In step (3) of Reaction Scheme X, a 2-chloropyridine-3,4-diamine of Formula LVI is reacted with a carboxylic acid or an equivalent thereof to provide a 4-chloro-1H-imidazo[4,5-c]pyridine of Formula LVII. The reaction can be carried out using the method described in step (1) of Reaction Scheme II.
  • In step (4) of Reaction Scheme X, the protecting group of a 4-chloro-1H-imidazo[4,5-c]pyridine of Formula LVII is removed to provide a hydroxyalkyl-substituted 4-chloro-1H-imidazo[4,5-c]pyridine of Formula LVIII. The deprotection can be carried out using a variety of methods depending on which P group is present. When P is an ethyl group, the reaction can be carried out by adding a solution of boron tribromide in a suitable solvent to a solution or suspension of a compound of Formula LVII in a suitable solvent such as dichloromethane. The reaction can be carried out at a sub-ambient temperature such as 0° C.
  • In step (5) of Reaction Scheme X, a 4-chloro-1H-imidazo[4,5-c]pyridine of Formula LVIII is reacted with 4-methoxybenzylamine to provide an N-(4-methoxybenzyl)-1H-imidazo[4,5-c]pyridin-4-amine of Formula LIX. The reaction can be carried out by combining a compound of Formula LVIII with excess N-(4-methoxybenzyl)amine and excess pyridine hydrochloride in a suitable solvent such as 2,2,2-trifluoroethanol and heating (150° C.) in a microwave reactor.
  • In step (6) of Reaction Scheme X, the 4-methoxybenzyl group is removed from an N-(4-methoxybenzyl)-1H-imidazo[4,5-c]pyridin-4-amine of Formula LIX to provide a 1H-imidazo[4,5-c]pyridin-4-amine of Formula LX. The reaction can be carried out by treating a compound of Formula LIX with trifluoroacetic acid at ambient temperature.
  • In step (7) of Reaction Scheme X, a hydroxyalkyl-substituted 1H-imidazo[4,5-c]pyridin-4-amine of Formula LX is chlorinated to provide a chloroalkyl-substituted 1H-imidazo[4,5-c]pyridin-4-amine of Formula LXI. The reaction can be carried out by treating a solution of a compound of Formula LX in a suitable solvent such as chloroform with thionyl chloride. The reaction can be carried out at an elevated temperature such as the reflux temperature of the solvent.
  • In step (8) of Reaction Scheme X, the chloro group of a chloroalkyl-substituted 1H-imidazo[4,5-c]pyridin-4-amine of Formula LXI is displaced with a hydroxylamine of formula HN(Y—R2-3)OR2-2 or a salt thereof to provide a 1H-imidazo[4,5-c]pyridin-4-amine of Formula LXII, which is a subgenus of Formulas I and II. The reaction can be carried out using the methods described in step (4) of Reaction Scheme I.
  • In step (8a) of Reaction Scheme X, the chloro group of a chloroalkyl-substituted 1H-imidazo[4,5-c]pyridin-4-amine of Formula LXI is displaced with a hydroxylamine of formula H2NOR2-2 or a salt thereof to provide a 1H-imidazo[4,5-c]pyridin-4-amine of Formula LXIII, which is a subgenus of Formulas I and II. The reaction can be carried out using the methods described in step (4) of Reaction Scheme I.
  • In step (9) of Reaction Scheme X, a 1H-imidazo[4,5-c]pyridin-4-amine of Formula LXIII is further derivatized to provide a 1H-imidazo[4,5-c]pyridin-4-amine of Formula LXII, which is a subgenus of Formulas I and II. The reaction can be carried out using the methods described in step (8) of Reaction Scheme II.
  • Figure US20090062328A1-20090305-C00065
  • For certain embodiments, compounds of the invention can be prepared according to Reaction Scheme XI, wherein RA2, RB2, R1, R2-1, X, Z, and G are as defined above. Compounds of Formula Ia can be prepared according to the methods described above. The amino group of a compound of Formula Ia 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 an amino group with a group such as —C(O)—R″, α-aminoacyl, α-aminoacyl-α-aminoacyl, —C(O)—O—R″, —C(O)—N(R″″)—R″, —C(—NY1)—R″, —CH(OH)—C(O)—OY1, —CH(OC1-4 alkyl)Y0, —CH2Y2, or —CH(CH3)Y2; wherein R″ and R″″ are each independently C1-10 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, arylC1-4 alkylenyl, heteroarylC1-4 alkylenyl, haloC1-4 alkylenyl, haloC1-4 alkoxy, —O—C(O)—CH3, —C(O)—O—CH3, —C(O)—NH2, —O—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, C1-6 alkyl, or benzyl; Y0 is C1-6 alkyl, carboxyC1-6 alkylenyl, aminoC1-4 alkylenyl, mono-N—C1-6 alkylaminoC1-4 alkylenyl, or di-N,N—C1-6 alkylaminoC1-4 alkylenyl; and Y2 is mono-N—C1-6 alkylamino, di-N,N-C1-6 alkylamino, morpholin-4-yl, piperidin-1-yl, pyrrolidin-1-yl, or 4-C1-4 alkylpiperazin-1-yl. Particularly useful compounds of Formula VII 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 Ia 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.
  • Figure US20090062328A1-20090305-C00066
  • Compounds of the invention can also be prepared using variations of the synthetic routes shown in Reaction Schemes I through X that would be apparent to one of skill in the art. For example, the order of steps may be changed in Reaction Schemes II, III, and VI through IX to prepare compounds of the invention. Compounds of the invention can also 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 kg0.425×height cm0.725)×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-1, IL-6, IL-10 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 (TH1) 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-1 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-1, IL-6, IL-10 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/m2 to about 5.0 mg/m2, (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. Pat. 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 Preparation of 2-Chloromethyl-1-(2-methylpropyl)-1H-imidazo[4,5-c]quinolin-4-amine Part A
  • N4-(2-Methylpropyl)quinoline-3,4-diamine (U.S. Pat. No. 5,389,640 Example 1, 41 g, 0.190 mol), dichloromethane (550 mL), triethylamine (40 mL, 0.286 mol), and chloroacetyl chloride (16.7 mL, 0.210 mol) were combined and then stirred at ambient temperature over the weekend. The reaction mixture was diluted with 1,2-dichloroethane (75 mL) and then washed with saturated aqueous sodium bicarbonate (3×400 mL). The organic layer was dried over magnesium sulfate, filtered through a layer of CELITE filter aid, and then concentrated under reduced pressure to provide 52.81 g of 2-chloromethyl-1-(2-methylpropyl)-1H-imidazo[4,5-c]quinoline as a brown solid.
  • Part B
  • 3-Chloroperoxybenzoic acid (mCPBA) (32.7 g of 77% pure material, 146 mmol) was added over a period of five minutes to a solution of 2-chloromethyl-1-(2-methylpropyl)-1H-imidazo[4,5-c]quinoline (20.0 g, 73.1 mmol) in chloroform (500 mL); the reaction mixture was stirred at ambient temperature for one hour. Ammonium hydroxide (200 mL) was added, and then p-toluenesulfonyl chloride (16.7 g, 87.7 mmol) was added in portions over a period of 10 minutes. The reaction mixture was stirred at ambient temperature for one hour, and then water (200 mL) was added. The aqueous layer was separated and extracted with dichloromethane (2×200 mL). The combined organic fractions were dried over magnesium sulfate, filtered, and concentrated under reduced pressure to provide 32 g of crude product as a tan solid. The crude product was dissolved in dichloromethane (50 mL), and the resulting solution was divided into two portions. Each portion was purified by column chromatography using a HORIZON HPFC system (an automated, modular high-performance flash purification product available from Biotage, Inc, Charlottesville, Va., USA) using a FLASH 65I silica cartridge (also available from Biotage, Inc.) (eluting with ethyl acetate:methanol in a gradient from 98:2 to 85:15) to provide 11.24 g of 2-chloromethyl-1-(2-methylpropyl)-1H-imidazo[4,5-c]quinolin-4-amine as a tan solid.
  • Example 1 1-(2-Methylpropyl)-2-{[methoxy(methyl)amino]methyl}-1H-imidazo[4,5-c]quinolin-4-amine
  • Figure US20090062328A1-20090305-C00067
  • 2-Chloromethyl-1-(2-methylpropyl)-1H-imidazo[4,5-c]quinolin-4-amine (0.100 g, 0.346 mmol) and triethylamine (0.105 g, 1.04 mmol) were added to a solution of N,O-dimethylhydroxylamine hydrochloride (0.068 g, 0.69 mmol) in. N,N-dimethylformamide (DMF) (2 mL). A precipitate formed after the addition of triethylamine. The reaction was heated with stirring for three days at 50° C., allowed to cool to ambient temperature, and partitioned between ethyl acetate (5 mL) and water (5 mL). The organic fraction was concentrated under reduced pressure to afford a white solid, which was purified by preparative high performance liquid chromatography (prep HPLC) using a Waters FractionLynx automated purification system. The prep HPLC fractions were analyzed using a Waters LC/TOF-MS, and the appropriate fractions were centrifuge evaporated to provide the trifluoroacetate salt of the desired compound. Reversed phase prep HPLC was performed with non-linear gradient elution from 5-95% B where A is 0.05% trifluoroacetic acid/water and B is 0.05% trifluoroacetic acid/acetonitrile. Fractions were collected by mass-selective triggering. The prep HPLC purification afforded 1-(2-methylpropyl)-2-{[methoxy(methyl)amino]methyl}-1H-imidazo[4,5-c]quinolin-4-amine trifluoroacetate.
  • HRMS (ESI) m/z 314.1974 (M+H).
  • Example 2 1-(2-Methylpropyl)-2-[(methoxyamino)methyl]-1H-imidazo[4,5-c]quinolin-4-amine
  • Figure US20090062328A1-20090305-C00068
  • Methoxylamine hydrochloride (145 mg, 1.73 mmol) was added to a solution of 2-chloromethyl-1-(2-methylpropyl)-1H-imidazo[4,5-c]quinolin-4-amine (0.250 g, 0.866 mmol) in DMF (3 mL). A precipitate formed. Triethylamine (0.263 g, 2.60 mmol) was added, and the reaction was heated with stirring overnight at 50° C., allowed to cool to ambient temperature, and partitioned between ethyl acetate (5 mL) and water (5 mL). The aqueous fraction was extracted twice with ethyl acetate, and the combined organic fractions were dried over magnesium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by radial chromatography on a silica gel plate (2 mm) (eluting sequentially with 2% and 5% methanol in chloroform). The solid was then purified by prep HPLC as described in Example 1 to provide 1-(2-methylpropyl)-2-[(methoxyamino)methyl]-1H-imidazo[4,5-c]quinolin-4-amine trifluoroacetate.
  • HRMS (ESI) m/z 300.1839 (M+H).
  • Example 3 2-{[Hydroxy(methyl)amino]methyl}-1-(2-methylpropyl)-1H-imidazo[4,5-c]quinolin-4-amine
  • Figure US20090062328A1-20090305-C00069
  • The method described in Example 1 was followed using N-methylhydroxylamine hydrochloride (0.058 g, 0.69 mmol) in lieu of N,O-dimethylhydroxylamine hydrochloride. The product was purified by prep HPLC as described in Example 1 to provide 2-{[hydroxy(methyl)amino]methyl}-1-(2-methylpropyl)-1H-imidazo[4,5-c]quinolin-amine trifluoroacetate.
  • HRMS (ESI) m/z 300.1826 (M+H).
  • Example 4 4-Amino-1-(2-methylpropyl)-1H-imidazo[4,5-c]quinoline-2-carbaldehyde O-methyloxime
  • Figure US20090062328A1-20090305-C00070
  • Part A
  • A solution of 1-(2-methylpropyl)-1H-imidazo[4,5-c]quinoline (U.S. Pat. No. 5,175,296 Example 1 Part C, 25.64 g, 113.8 mmol) in tetrahydrofuran (THF) (450 mL) was cooled to −78° C. Butyllithium (45.5 mL of a 2.5 M solution in hexanes) was added dropwise over a period of ten minutes, and the resulting solution was stirred for ten minutes. DMF (20.1 mL, 274.4 mmol) was added, and the reaction was allowed to warm to room temperature and stirred for one hour. The THF was then removed under reduced pressure, and the residue was dissolved in ethyl acetate (400 mL). The resulting solution was washed with brine (400 mL), dried over magnesium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography on silica gel (eluting sequentially with 0.5% and 1% methanol in dichloromethane) to provide 10.5 g of 1-(2-methylpropyl)-1H-imidazo[4,5-c]quinoline-2-carbaldehyde as a light brown solid.
  • Part B
  • Methoxylamine hydrochloride (0.659 g, 7.90 mmol) was added to a stirred suspension of 1-(2-methylpropyl)-1H-imidazo[4,5-c]quinoline-2-carbaldehyde (1.00 g, 3.95 mmol) in methanol (10 mL), and the reaction was stirred at room temperature overnight. The methanol was removed under reduced pressure, and the residue was diluted with saturated aqueous sodium bicarbonate (50 mL). The resulting mixture was extracted with dichloromethane (3×30 mL), and the combined extracts were dried over magnesium sulfate, filtered, and concentrated under reduced pressure to provide 1.16 g of 1-(2-methylpropyl)-1H-imidazo[4,5-c]quinoline-2-carbaldehyde O-methyloxime as a pale yellow oil that solidified upon standing.
  • Part C
  • mCPBA (1.75 g of 77% pure material, 7.79 mmol) was added in portions over a period of five minutes to a solution of 1-(2-methylpropyl)-1H-imidazo[4,5-c]quinoline-2-carbaldehyde O-methyloxime (1.1 g, 3.9 mmol) in chloroform (40 mL), and the reaction was stirred at room temperature for one hour. Ammonium hydroxide (20 mL) was added, and the resulting mixture was stirred for five minutes before the addition of p-toluenesulfonyl chloride (0.891 g, 4.68 mmol) in portions. The reaction mixture was stirred at room temperature for 1.5 hours. The aqueous layer was then separated and extracted with dichloromethane (3×30 mL). The combined organic fractions were dried over magnesium sulfate, filtered, and concentrated under reduced pressure to provide 2.2 g of crude product as a brown foamy solid. The crude product was dissolved in dichloromethane (15 mL) and purified by chromatography using a HORIZON HPFC system (FLASH 40M silica cartridge available from Biotage, Inc., eluting with 2% to 7% methanol in ethyl acetate). The resulting product (580 mg) was recrystallized from acetonitrile (10 mL), and the crystals were isolated by filtration, washed with diethyl ether, and dried overnight in a vacuum oven at 65° C. to provide 236 mg of 4-amino-1-(2-methylpropyl)-1H-imidazo[4,5-c]quinoline-2-carbaldehyde O-methyloxime as a brown solid, mp 181-183° C.
  • Anal. Calcd for C16H19N5O: C, 64.63; H, 6.44; N, 23.55. Found: C, 64.37; H, 6.39; N, 23.57.
  • Example 5 4-[4-Amino-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-2-yl]butan-2-one O-methyloxime
  • Figure US20090062328A1-20090305-C00071
  • Part A
  • A mixture of ethyl levulinate (58.35 g, 400.0 mmol), ethylene glycol (75.36 g, 1.21 mol), pyridinium p-toluenesulfonate (100 mg) and toluene (200 mL) was heated at reflux under a Dean Stark trap. The trap was emptied every 15 minutes during the reaction until approximately 200 mL of reaction volatiles had been removed. The remaining toluene was removed under reduced pressure, and the resulting oil was partitioned between ethyl acetate (200 mL) and water (50 mL). The organic layer was separated and washed sequentially with water (2×50 mL), saturated aqueous sodium bicarbonate (50 mL), and brine (50 mL); dried over sodium sulfate, filtered, and concentrated under reduced pressure to provide 68.9 g of ethyl 3-(2-methyl-1,3-dioxolan-2-yl)propanoate as a light yellow oil.
  • Part B
  • A solution of ethyl 3-(2-methyl-1,3-dioxolan-2-yl)propanoate (68.9 g, 366 mmol) in methanol (73 mL) was cooled to approximately 0° C. A warm solution of sodium hydroxide (14.63 g, 366 mmol) in water (73 mL) was added dropwise over a period of three hours. The reaction was then allowed to warm to room temperature and stirred for 17 hours. The methanol was removed under reduced pressure, and the resulting aqueous solution was diluted with water (400 mL) and washed with ethyl acetate (150 mL). The aqueous solution was then cooled to approximately 0° C. and adjusted to pH 2 with the addition of sulfuric acid (180 mL of 1 M). The resulting mixture was extracted with ethyl acetate (2×150 mL), and the combined extracts were washed with brine (50 mL), dried over sodium sulfate, filtered, and concentrated under reduced pressure to provide 38.6 g of 3-(2-methyl-1,3-dioxolan-2-yl)propanoic acid as a light yellow oil.
  • Part C
  • A solution of 3-(2-methyl-1,3-dioxolan-2-yl)propanoic acid (8.95 g, 55.9 mmol) in dichloromethane (111 mL) was cooled to approximately 0° C. N-hydroxysuccinimide (7.10 g, 61.5 mmol), 4-methylmorpholine (6.22 g, 61.5 mmol), and 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (11.8 g, 61.5 mmol) were sequentially added with stirring. The reaction was then allowed to warm to room temperature, stirred for 17 hours, and partitioned between dichloromethane (150 mL) and water (50 mL). The organic layer was separated; sequentially washed with water (50 mL), saturated aqueous sodium bicarbonate (2×50 μL), and brine (50 mL); dried over sodium sulfate; filtered; and concentrated under reduced pressure to provide 13 g of 1-{[3-(2-methyl-1,3-dioxolan-2-yl)propanoyl]oxy}pyrrolidine-2,5-dione as a white solid.
  • Part D
  • A solution of 3-amino-4-(2-hydroxy-2-methylpropylamino)quinoline (U.S. Pat. No. 5,389,640, 6.00 g, 26.0 mmol) and 1-{[3-(2-methyl-1,3-dioxolan-2-yl)propanoyl]oxy}pyrrolidine-2,5-dione (8.01 g, 31.1 mmol) in pyridine (130 mL) was heated at reflux under a Dean-Stark trap for seven hours, allowed to cool to room temperature, and stirred for 48 hours. The volatiles were removed under reduced pressure, and the residue was dissolved in dichloromethane (200 mL). The resulting solution was washed sequentially with saturated aqueous sodium bicarbonate (2×100 mL) and brine (100 mL), dried over sodium sulfate, filtered, and concentrated under reduced pressure. The resulting dark brown oil was purified by column chromatography on silica gel (eluting with dichloromethane and 5% methanol in dichloromethane) to provide 4.5 g of 2-methyl-1-{(2-[2-(2-methyl-1,3-dioxolan-2-yl)ethyl]-1H-imidazo[4,5-c]quinolin-1-yl}propan-2-ol.
  • Part E
  • mCPBA (2.2 g of 60% pure material, 7.7 mmol) was added in portions over a period of five minutes to a stirred solution of 2-methyl-1-{2-[2-(2-methyl-1,3-dioxolan-2-yl)ethyl]-1H-imidazo[4,5-c]quinolin-1-yl}propan-2-ol (2.5 g, 7.0 mmol) in chloroform (47 mL), and the reaction was stirred at room temperature for 20 minutes. The reaction mixture was partitioned between dichloromethane (200 mL) and saturated aqueous sodium carbonate (100 mL). The organic layer was separated and washed with saturated aqueous sodium carbonate (100 mL). The combined aqueous fractions were extracted with dichloromethane (50 mL). The combined organic fractions were washed with brine (50 mL), dried over sodium sulfate, filtered, and concentrated under reduced pressure to provide 2-methyl-1-{2-[2-(2-methyl-1,3-dioxolan-2-yl)ethyl]-5-oxido-1H-imidazo[4,5-c]quinolin-1-yl}propan-2-ol as a tan solid.
  • Part F
  • Ammonium hydroxide (12 mL of 30%) was added to a stirred solution of the material from Part E in dichloromethane (35 mL). p-Toluenesulfonyl chloride (1.34 g, 7.03 mmol) was added in portions over a period of two minutes with vigorous stirring. The reaction mixture was stirred at room temperature for five hours. A solid was present and was isolated by filtration to provide 2.1 g of white powder, which was recrystallized from ethanol (145 mL). The crystals were isolated by filtration, washed with ethanol, and dried for four hours under vacuum at 65° C. to provide 1.7 g of 1-{4-amino-2-[2-(2-methyl-1,3-dioxolan-2-yl)ethyl]-1H-imidazo[4,5-c]quinolin-1-yl}-2-methylpropan-2-ol as a brown solid, mp 231-233° C.
  • Anal. Calcd for C20H26N4O3: C, 64.85; H, 7.07; N, 15.12. Found: C, 64.72; H, 7.40; N, 15.05.
  • Part G
  • Concentrated hydrochloric acid (0.98 mL) was added dropwise to a stirred suspension of 1-{4-amino-2-[2-(2-methyl-1,3-dioxolan-2-yl)ethyl]-1H-imidazo[4,5-c]quinolin-1-yl}-2-methylpropan-2-ol (1.45 g, 3.91 mmol) in water (26 mL), and the resulting solution was stirred at room temperature for two hours and then adjusted to pH 11 with the addition of aqueous sodium hydroxide (20% w/w). A precipitate formed, and the suspension was stirred for several minutes. The precipitate was isolated by filtration and washed with water to provide 1.2 g of 4-[4-amino-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-2-yl]butan-2-one as a white powder.
  • Part H
  • Methoxylamine hydrochloride (0.26 g, 3.1 mmol) was added to a stirred solution of 4-[4-amino-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-2-yl]butan-2-one (0.600 g, 1.84 mmol) in pyridine (9 mL), and the reaction was stirred at room temperature for 45 minutes. Water (50 mL) was added. The aqueous layer was separated, extracted with dichloromethane (2×50 mL), adjusted to pH 10 with the addition of saturated aqueous sodium carbonate, and extracted with dichloromethane (2×25 mL). The combined organic fractions were washed sequentially with saturated aqueous sodium bicarbonate (50 mL) and brine (50 mL), dried over sodium sulfate, filtered, and concentrated under reduced pressure. The resulting white powder was stirred with diethyl ether for one hour, isolated by filtration, and washed with diethyl ether. The resulting solid (0.45 g) was recrystallized from acetonitrile (20 mL), and the crystals were isolated by filtration, washed with acetonitrile, and dried for 15 hours under vacuum at 60° C. to provide 0.40 g of 4-[4-amino-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-2-yl]butan-2-one O-methyloxime as white needles, mp 182-184° C.
  • Anal. Calcd for C19H25N5O2: C, 64.20; H, 7.09; N, 19.70. Found: C, 63.97; H, 7.21; N, 19.84.
  • Example 6 7-(Benzyloxy)-2-[(methoxyamino)methyl]-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-4-amine
  • Figure US20090062328A1-20090305-C00072
  • Part A
  • 1-tetrahydro-2H-pyran-4-ylmethanamine (40 g, 0.347 mol) was added to a stirred solution of 7-(benzyloxy)-4-chloro-3-nitroquinoline (55 g, 0.176 mol) and triethylamine (50.96 g, 0.503 mol) in N,N-dimethylformamide (DMF) (250 mL) at 0° C. After complete addition, the ice bath was removed. To consume the remaining starting material, additional (1-tetrahydro-2H-pyran-4-ylmethanamine (11 g, 95.50 mmol) was added. The reaction proceeded at ambient temp for 3 days, then was chilled in an ice-water bath. Water (250 mL) was added drop wise which caused a solid to precipitate out of solution. After stirring for 1 hour, the solid was collected by filtration, washed with diethyl ether (800 mL), and dried under reduced pressure to afford 7-(benzyloxy)-3-nitro-N-(tetrahydro-2H-pyran-4-ylmethyl)quinolin-4-amine (59.3 g) as a bright yellow solid.
  • Part B
  • A 2 L glass parr vessel was charged with of 7-(benzyloxy)-3-nitro-N-(tetrahydro-2H-pyran-4-ylmethyl)quinolin-4-amine (59.34 g, 0.151 mol), platinum on carbon (5%, 6.6 g) and acetonitrile (600 mL) to provide a black mixture. The vessel was placed on a shaker and pressurized with hydrogen gas (−50 psi, 3.4×105 Pa). After shaking for 18 hours at ambient temperature, the mixture was filtered through CELITE filter agent, and the filter cake washed with acetonitrile. The filtrate was concentrated under reduced pressure to provide 7-(benzyloxy)-N4-(tetrahydro-2H-pyran-4-ylmethyl)quinoline-3,4-diamine
  • (51.2 g) as a viscous oil.
  • Part C
  • To a stirred solution of 7-(benzyloxy)-N4-(tetrahydro-2H-pyran-4-ylmethyl)quinoline-3,4-diamine (51.2 g, 0.141 mol) and triethylamine (39.94 g, 0.395 mol) in dichloromethane (410 mL) at 0° C. was added a solution of chloroacetyl chloride (8.67 g, 76.80 mmol) in dichloromethane (40 mL) drop wise by addition funnel. The reaction was stirred at ambient temperature for 17 hours then heated to reflux for 6 hours. The crude mixture was cooled to ambient temperature and partitioned between water and dichloromethane (200 mL). The layers were separated and the aqueous layer extracted with dichloromethane (2×300 mL). The organic layers were combined, washed with brine (2×300 mL), dried over magnesium sulfate, filtered and concentrated under reduced pressure to provide 7-(benzyloxy)-2-(chloromethyl)-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinoline (61.7 g) as a brown solid.
  • Part D
  • 3-Chloroperoxybenzoic acid (mCPBA) (1.4 g of 77% pure material, 4.74 mmol) was added over a period of five minutes to a solution of 7-(benzyloxy)-2-(chloromethyl)-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinoline (2.0 g, 4.74 mmol) in chloroform (47 mL); the reaction mixture was stirred at ambient temperature for one hour. The reaction solution was partitioned between chloroform (50 mL) and saturated aqueous sodium carbonate (50 mL). The layers were separated and the aqueous layer extracted with chloroform (50 mL). The organic layers were combined, dried over sodium sulfate, filtered, and concentrated under reduced pressure to afford an oil. To the crude N-oxide dissolved in dichloromethane (24 mL) was added ammonium hydroxide (8 mL), and then p-toluenesulfonyl chloride (1.0 g, 3.11 mmol) was added in portions over a period of 2 minutes. The reaction mixture was stirred at ambient temperature for one hour, and then saturated aqueous sodium carbonate (50 mL) and chloroform (50 mL) were added. The aqueous layer was separated and extracted with chloroform (50 mL). The combined organic fractions were dried over sodium sulfate, filtered, and concentrated under reduced pressure to provide 2.1 g of crude product as a tan solid. The crude product was purified by column chromatography using a HORIZON HPFC system (an automated, modular high-performance flash purification product available from Biotage, Inc, Charlottesville, Va., USA) using a FLASH 40+M silica cartridge (also available from Biotage, Inc.) (eluting with dichloromethane:methanol in a gradient from 100:0 to 90:10) to provide 1.8 g of 7-(benzyloxy)-2-(chloromethyl)-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-4-amine as a tan solid.
  • Part E
  • 7-(Benzyloxy)-2-(chloromethyl)-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-4-amine (2.3 g, 5.26 mmol) and triethylamine (1.6 g, 15.78 mmol) were added to a solution of methoxylamine hydrochloride (0.88 g, 10.52 mmol) in N,N-dimethylformamide (DMF) (11 mL). The reaction was heated with stirring for 40 hours at 50° C., allowed to cool to ambient temperature, and partitioned between dichloromethane (50 mL) and water (50 mL). The aqueous layer was extracted with another portion of dichloromethane (50 mL). The combined organic layers were dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude material was purified by column chromatography using a HORIZON HPFC system equipped with a FLASH 40+M silica cartridge eluting with dichloromethane:methanol in a gradient from 100:0 to 90:10. The resulting product (1.0 g) was recrystallized from acetonitrile, and the crystals were isolated by filtration, washed with acetonitrile, and dried overnight in a vacuum oven at 65° C. to provide 0.85 g of 7-(benzyloxy)-2-[(methoxyamino)methyl]-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-4-amine as a yellow, crystalline solid, mp 190-193° C.
  • Anal. Calcd for C25H29N5O3: C, 67.09; H, 6.53; N, 15.65. Found: C, 66.88; H, 6.44; N, 15.55.
  • Example 7 7-(Benzyloxy)-2-{[methoxy(methyl)amino]methyl} 1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-4-amine
  • Figure US20090062328A1-20090305-C00073
  • 7-(Benzyloxy)-2-(chloromethyl)-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-4-amine (0.50 g, 1.14 mmol) and triethylamine (0.34 g, 3.42 mmol) were added to a solution of N, O-dimethylhydroxylamine hydrochloride (0.22 g, 2.29 mmol) in N)N-dimethylformamide (DMF) (2 mL). The resulting suspension was heated with stirring for 40 hours at 50° C., allowed to cool to ambient temperature, and partitioned between dichloromethane (30 mL) and water (20 mL). The aqueous layer was extracted with another portion of dichloromethane (30 mL). The combined organic layers were dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude material was purified by column chromatography using a HORIZON HPFC system equipped with a FLASH 40+M silica cartridge eluting with dichloromethane:methanol in a gradient from 100:0 to 90:10. The resulting product (0.3 g) was recrystallized from acetonitrile (3 mL), and the crystals were isolated by filtration, washed with acetonitrile, and dried overnight in a vacuum oven at 65° C. to provide 0.14 g of 7-(benzyloxy)-2-{[methoxy(methyl)amino]methyl}-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-4-amine as a yellow, crystalline solid, mp 159-161° C.
  • Anal. Calcd for C26H31N5O3.H2O1/4: C, 67.00; H, 6.81; N, 15.03. Found: C, 66.91; H, 6.95; N, 15.08.
  • Example 8 N-{[4-Amino-7-(benzyloxy)-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-2-yl]methyl}-N-methoxymethanesulfonamide
  • Figure US20090062328A1-20090305-C00074
  • To a solution of 7-(benzyloxy)-2-(chloromethyl)-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-4-amine (0.70 g, 1.56 mmol) and triethylamine (0.30 g, 3.12 mmol) in dichloromethane (6 mL) was added methanesulfonyl chloride (0.20 g, 1.72 mmol) dropwise. The brown solution was stirred at ambient temperature for 18 hours, and then concentrated under reduced pressure. The crude material was purified by column chromatography using a HORIZON HPFC system equipped with a FLASH 25+M silica cartridge eluting with dichloromethane:methanol in a gradient from 100:0 to 90:10. The resulting product (0.35 g) was recrystallized from acetonitrile (43 mL), and the crystals were isolated by filtration, washed with acetonitrile, and dried overnight in a vacuum oven at 65° C. to provide 0.19 g of N-{[4-amino-7-(benzyloxy)-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-2-yl]methyl}-N-methoxymethanesulfonamide as a yellow, crystalline solid, mp 195-197° C.
  • Anal. Calcd for C26H31N5O5S: C, 59.41; H, 5.94; N, 13.32. Found: C, 59.78; H, 5.80; N, 13.61.
  • Example 9 1-Isobutyl-2-[(methoxyamino)methyl]-6,7-dimethyl-1H-imidazo[4,5-c]pyridin-4-amine
  • Figure US20090062328A1-20090305-C00075
  • Part A
  • A solution of 2,4-dichloro-5,6-dimethyl-3-nitropyridine (40.0 g, 181 mmol), triethylamine (26.5 mL, 190 mmol), and isobutyl amine (18.9 mL, 190 mmol) in N,N-dimethylformamide (500 mL) was stirred at room temperature over night. The solvent was removed under reduced pressure. The residue was dissolved in ethyl acetate (500 mL) and washed with water (3×80 mL) and brine (40 mL). The aqueous was extracted with ethyl acetate (3×50 mL) and the back-extracts washed with water (3×40 mL) and brine (30 mL). The combined organics were dried over magnesium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by HPFC eluting with a gradient of 10-30% ethyl acetate in hexanes to give 25.8 g of 2-chloro-N-isobutyl-5,6-dimethyl-3-nitropyridin-4-amine as a yellow oil.
  • Part B
  • 2-Chloro-N-isobutyl-5,6-dimethyl-3-nitropyridin-4-amine (25.8 g, 100 mmol) was combined with 5% platinum on carbon (2.58 g) and ethyl acetate (200 mL) in a pressure vessel and hydrogenated at 50 psi (3.4×105 Pa) for 2.5 hours on a Parr apparatus. The reaction mixture was filtered through CELITE filter agent, which was rinsed with ethyl acetate and methanol afterwards. The filtrate was concentrated to give 2-chloro-N4-isobutyl-5,6-dimethylpyridine-3,4-diamine and was used directly in the next step.
  • Part C
  • Under a nitrogen atmosphere, the material from part B was dissolved in dichloromethane (400 mL) and cooled to 0° C. Ethoxyacetyl chloride (14.7 g, 120 mmol) dissolved in dichloromethane (100 mL) was added dropwise through an addition funnel and the solution was stirred at room temperature over night. The solvent was removed under reduced pressure and the white solid used directly in the next step.
  • Part D
  • The material from part C was suspended in ethanol (500 mL), and sodium hydroxide (10.0 g, 250 mmol) was added. The mixture was heated to reflux under a nitrogen atmosphere for four hours. The heat was removed and the solution allowed to stir at room temperature over night. The solvent was removed under reduced pressure and the residue dissolved in dichloromethane (500 mL), washed with water (100 mL) and brine (60 mL), dried over sodium sulfate, filtered and concentrated under reduced pressure to give 29.6 g of 4-chloro-2-(ethoxymethyl)-1-isobutyl-6,7-dimethyl-1H-imidazo[4,5-c]pyridine as a yellow oil.
  • Part E
  • A 500 mL round bottom flask was charged with 4-chloro-2-(ethoxymethyl)-1-isobutyl-6,7-dimethyl-1H-imidazo[4,5-c]pyridine (10.0 g, 33.8 mmol) and dichloromethane (250 mL) under a nitrogen atmosphere. The solution was cooled to 0° C. and boron tribromide (101 mL of 1M in dichloromethane, 101 mmol) was added through an addition funnel over 30 minutes. The reaction mixture was allowed to warm to room temperature and was stirred over night. Methanol was added slowly until no more fizzing occurred, then the solvent was partially removed under reduced pressure. More methanol was added (100 mL) as well as 6N hydrochloric acid (100 mL) and the solution was heated at reflux for 1 hour. The reaction mixture was then allowed to cool to room temperature and stirred over night. The solvent was partially removed under reduced pressure until a solid precipitated, which was isolated by filtration and washed with water, then triturated with ethyl acetate and hexanes to give 6.15 g of (4-chloro-1-isobutyl-6,7-dimethyl-1H-imidazo[4,5-c]pyridin-2-yl)methanol as an off-white solid.
  • Part F
  • A solution of 4-chloro-1-isobutyl-6,7-dimethyl-1H-imidazo[4,5-c]pyridin-2-yl)methanol (1.50 g, 5.60 mmol), 4-methoxybenzylamine (3.66 mL, 28 mmol) and pyridine hydrochloride (1.74 g, 11.2 mmol) in 2,2,2-trifluoroethanol (11.2 mL) was heated to 150° C. in a microwave oven for 2.5 hours. The mixture was allowed to cool to room temperature, then poured into water (75 mL) and stirred for 30 minutes. The precipitate was isolated by filtration and washed with water to give 1.86 g of {1-isobutyl-4-[(4-methoxybenzyl)amino]-6,7-dimethyl-1H-imidazo[4,5-c]pyridin-2-yl}methanol.
  • Part G
  • {1-Isobutyl-4-[(4-methoxybenzyl)amino]-6,7-dimethyl-1H-imidazo[4,5-c]pyridin-2-yl}methanol (3.08 g, 8.36 mmol) was dissolved in trifluoroacetic acid (31 mL) and stirred at room temperature over night. The solvent was removed under reduced pressure and concentrated hydrochloric acid (5 mL) was added. The suspension was stirred for 2 hours, the solid was isolated by filtration and washed with water to give 1.95 g of (4-amino-1-isobutyl-6,7-dimethyl-1H-imidazo[4,5-c]pyridin-2-yl)methanol as a tan powder.
  • Part H
  • A solution of (4-amino-1-isobutyl-6,7-dimethyl-1H-imidazo[4,5-c]pyridin-2-yl)methanol (1.95 g, 7.85 mmol) and thionyl chloride (1.72 mL, 23.6 mmol) in chloroform (80 mL) was heated at reflux for 2 hours. The solvent was removed under reduced pressure and the residue triturated with chloroform to give 1.53 g of 2-(chloromethyl)-1-isobutyl-6,7-dimethyl-1H-imidazo[4,5-c]pyridin-4-amine hydrochloride as an off-white powder.
  • Part I
  • 2-(chloromethyl)-1-isobutyl-6,7-dimethyl-1H-imidazo[4,5-c]pyridin-4-amine hydrochloride (1.53 g, 5.05 mmol) was dissolved in 10 mL of DMF in a pressure tube under a nitrogen atmosphere. Methoxylamine hydrochloride (1.27 g, 15.15 mmol) and triethylamine (4.22 mL, 30.3 mmol) were added, the tube was sealed and the contents heated to 50° C. for two hours, then to 60° C. for five hours. The mixture was then stored in the refrigerator over the weekend. The contents were poured into water (60 mL) and stirred for 20 minutes. The solution was extracted with chloroform (3×70 mL) and the organics were washed with water (4×40 mL) and brine (30 mL), dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by HPFC eluting with a gradient of 0-30% CMA (80/18/2 v/v/v chloroform/methanol/concentrated ammonium hydroxide) in chloroform and then recrystallized from acetonitrile to provide 1-isobutyl-2-[(methoxyamino)methyl]-6,7-dimethyl-1H-imidazo[4,5-c]pyridin-4-amine as a white powder, mp 154.0-156.0° C.
  • Anal. Calcd for C14H23N5O C, 60.62; H, 8.36; N, 25.25. Found: C, 60.63; H, 8.33; N, 25.35.
  • Example 10 1-Isobutyl-2-{[methoxy(methyl)amino]methyl}-6,7-dimethyl-1H-imidazo[4,5-c]pyridin-4-amine
  • Figure US20090062328A1-20090305-C00076
  • 1-Isobutyl-2-{[methoxy(methyl)amino]methyl}-6,7-dimethyl-1H-imidazo[4,5-c]pyridin-4-amine was prepared and purified according to the general methods of Example 9 using N,O-dimethyl hydroxylamine hydrochloride in lieu of methoxylamine hydrochloride in Part I. The pure product was obtained as a white powder, mp 132.0-134.0° C. Anal. Calcd for C15H25N5O C., 61.83; H, 8.65; N, 24.03. Found: C, 61.63; H, 8.88; N, 24.02.
  • Example 11 N-[(4-Amino-1-isobutyl-6,7-dimethyl-1H-imidazo[4,5-c]pyridin-2-yl)methyl]-N-methoxycyclopropanecarboxamide
  • Figure US20090062328A1-20090305-C00077
  • Under a nitrogen atmosphere, 1-isobutyl-2-[(methoxyamino)methyl]-6,7-dimethyl-1H-imidazo[4,5-c]pyridin-4-amine (0.53 g, 1.91 mmol) was dissolved in dichloromethane (19 mL) and cooled to −5° C. Triethylamine (293 uL, 2.10 mmol) was added followed by dropwise addition of cyclopropane carbonyl chloride (173 uL, 1.91 mmol). The solution was allowed to stir at RT for 2 hours, then cooled back to −5° C. More triethylamine (38 uL, 0.27 mmol) and cyclopropane carbonyl chloride (23 uL, 0.25 mmol) were added and the reaction was allowed to stir at 0° C. for 45 minutes. More cyclopropane carbonyl chloride (5 uL, 0.06 mmol) was added and stirring continued for another 30 minutes. Dichloromethane (100 mL) was added and the solution was washed successively with saturated aqueous sodium bicarbonate (30 mL), water (3×25 mL), and brine (20 mL), dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by HPFC eluting with a gradient of 0-35% CMA in chloroform and then recrystallized from ethyl acetate/hexanes to provide 292 mg of pure product as an off-white powder, mp 135.0-137.0° C. Anal. Calcd for C18H27N5O2 C, 62.59; H, 7.88; N, 20.27. Found: C, 62.68; H, 7.82; N, 19.93.
  • Example 12 1-({4-Amino-2-[(methoxyamino)methyl]-1H-imidazo[4,5-c]quinolin-1-yl}methyl)cyclobutanol
  • Figure US20090062328A1-20090305-C00078
  • 1-({4-Amino-2-[(methoxyamino)methyl]-1H-imidazo[4,5-c]quinolin-1-yl}methyl)cyclobutanol was prepared according to the general methods of Part I of Example 9 using 1-{[4-amino-2-(chloromethyl)-1H-imidazo[4,5-c]quinolin-1-yl]methyl}cyclobutanol in lieu of 2-(chloromethyl)-1-isobutyl-6,7-dimethyl-1H-imidazo[4,5-c]pyridin-4-amine hydrochloride. The crude product was purified by HPFC eluting with a gradient of 0-35% CMA in chloroform and then recrystallized from acetonitrile to provide pure product as a white powder, mp 178.0-181.0° C. Anal. Calcd for C17H21N5O2 C, 62.37; H, 6.47; N, 21.39. Found: C, 62.35; H, 6.51; N, 21.26.
  • Example 13 1-[(4-Amino-2-{[methoxy(methyl)amino]methyl}-1H-imidazo[4,5-c]quinolin-1-yl)methyl]cyclobutanol
  • Figure US20090062328A1-20090305-C00079
  • 1-[(4-Amino-2-{[methoxy(methyl)amino]methyl}-1H-imidazo[4,5-c]quinolin-1-yl)methyl]cyclobutanol was prepared according to the general methods of Part I of Example 9 using N,O-dimethyl hydroxylamine hydrochloride in lieu of methoxylamine hydrochloride and using 1-{[4-amino-2-(chloromethyl)-1H-imidazo[4,5-c]quinolin-1-yl]methyl}cyclobutanol in lieu of 2-(chloromethyl)-1-isobutyl-6,7-dimethyl-1H-imidazo[4,5-c]pyridin-4-amine hydrochloride. The crude product was purified by HPFC eluting with a gradient of 0-35% CMA in chloroform and then recrystallized from acetonitrile to provide pure product as a white powder, mp 219.0-221.0° C. Anal. Calcd for C18H23N5O2 C, 63.32; H, 6.79; N, 20.51. Found: C, 63.12; H, 6.56; N, 20.16.
  • Example 14 4-Amino-1-isobutyl-6,7-dimethyl-1H-imidazo[4,5-c]pyridine-2-carbaldehyde O-methyloxime
  • Figure US20090062328A1-20090305-C00080
  • Part A
  • Under a nitrogen atmosphere {1-isobutyl-4-[(4-methoxybenzyl)amino]-6,7-dimethyl-1H-imidazo[4,5-c]pyridin-2-yl}methanol (2.00 g, 5.43 mmol, prepared as in Parts A-F of Example 9) was dissolved in dichloromethane (20 mL) and dimethylsulfoxide (10 mL). Triethylamine (2.27 mL, 16.3 mmol) was added and the solution was cooled to 0° C. Sulfur trioxide pyridine complex (2.60 g, 16.3 mmol) was then added in four portions and the solution was stirred at 0° C. for another 2 hours. The solution was then poured into saturated aqueous ammonium chloride (70 mL) and extracted with diethyl ether (3×80 mL). The organics were washed with water (2×30 mL) and brine (20 mL), dried over magnesium sulfate, filtered, and concentrated under reduced pressure to give 1.84 g of 1-isobutyl-4-[(4-methoxybenzyl)amino]-6,7-dimethyl-1H-imidazo[4,5-c]pyridine-2-carbaldehyde as a yellow residue.
  • Part B
  • The material from part A was dissolved in methanol (50 mL), and methoxylamine hydrochloride (0.63 g, 7.53 mmol) was added. Sodium hydroxide (1.42 mL of a 6N solution, 8.54 mmol) was added dropwise and the solution was stirred at room temperature over night. Water (40 mL) was added and the solution was extracted with chloroform (3×80 mL). The organics were washed with saturated aqueous sodium bicarbonate (2×40 mL) and brine (30 mL), dried over magnesium sulfate, filtered, and concentrated under reduced pressure to give 1.90 g of 1-isobutyl-4-[(4-methoxybenzyl)amino]-6,7-dimethyl-1H-imidazo[4,5-c]pyridine-2-carbaldehyde O-methyloxime as a yellow oil.
  • Part C
  • 1-Isobutyl-4-[(4-methoxybenzyl)amino]-6,7-dimethyl-1H-imidazo[4,5-c]pyridine-2-carbaldehyde O-methyloxime (1.60 g, 4.05 mmol) was dissolved in trifluoroacetic acid (16 mL) and stirred at room temperature over night. The solvent was removed under reduced pressure and concentrated hydrochloric acid (5 mL) was added, The suspension was stirred for 2 hours, then dichloromethane was added (10 mL) and the solution was cooled to 0° C. Sodium hydroxide (6N) was added until the solution was basic. More water (20 mL) was added and the solution was extracted with dichloromethane (2×100 mL). The organics were washed with brine (30 mL), dried over magnesium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by HPFC eluting with a gradient of 0-30% CMA in chloroform and then recrystallized from dichloromethane/hexanes. The solid was triturated with 1N sodium hydroxide to provide 256 mg of pure product as a white powder, mp 179.0-181.0° C. Anal. Calcd for C14H21N5O C, 61.07; H, 7.69; N, 25.43. Found: C, 61.16; H, 7.64; N, 25.69.
  • Example 15 1-(2-Hydroxy-2-methylpropyl)-2-{[methoxy(methyl)amino]methyl}-1H-imidazo[4,5-c]quinolin-4-amine
  • Figure US20090062328A1-20090305-C00081
  • Triethylamine (0.747 g, 1.03 mL, 7.38 mmol) and N,O-dimethylhydroxylamine hydrochloride (0.480 g, 4.92 mmol) were added to a stirring suspension of 2-chloromethyl-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-4-amine (0.750 g, 2.46 mmol) in DMF (5 mL). The resulting suspension was heated to 50° C. and stirred for 17 hours. The reaction mixture was cooled to room temperature and poured into water (100 mL). A solid was removed by filtration and discarded. The filtrate was extracted with ethyl acetate (3×100 mL). The combined organic fractions were dried over magnesium sulfate, filtered and concentrated under reduced pressure. The residue was purified by column chromatography using a HORIZON HPFC system (silica cartridge, eluting with 5-20% methanol in dichloromethane). The resulting oil was crystallized from dichloromethane and isolated by filtration to yield 132 mg of 1-(2-Hydroxy-2-methylpropyl)-2-{[methoxy(methyl)amino]methyl}-1H-imidazo[4,5-c]quinolin-4-amine as a white solid, mp 220-222° C.
  • Anal. calcd for C17H23N5O2: C, 61.99; H, 7.04; N, 21.26. Found: C, 61.97; H, 7.10; N, 21.38.
  • Example 16 1-(2-Hydroxy-2-methylpropyl)-2-{[methoxylamino]methyl}-1H-imidazo[4,5-c]quinolin-4-amine; hydrochloride
  • Figure US20090062328A1-20090305-C00082
  • Triethylamine (1.49 g, 2.1 mL, 14.8 mmol) and methoxylamine hydrochloride (0.822 g, 9.84 mmol) were added to a stirring suspension of 2-chloromethyl-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-4-amine (1.50 g, 4.92 mmol) in DMF (10 mL). The resulting suspension was heated to 50° C. and stirred for 17 hours. The reaction mixture was cooled to room temperature and poured into water (20 mL). The aqueous layer was extracted with ethyl acetate (3×50 mL) and dichloromethane (3×50 mL). A significant amount of the desired product remained in the aqueous layer thus the organic fractions were combined with the aqueous fraction and concentrated under reduced pressure. The residue was purified by column chromatography using a HORIZON HPFC system (silica cartridge, eluting with 10-25% methanol in dichloromethane) and triturated with dichloromethane to provide 760 mg of 1-(2-Hydroxy-2-methylpropyl)-2-{[methoxylamino]methyl)}-1H-imidazo[4,5-c]quinolin-4-amine; hydrochloride as a tan solid, mp 255-257° C.
  • Anal. calcd for C16H21N5O2.HCl.0.5H2O: C, 53.26; H, 6.42; N, 19.41. Found: C, 53.57; H, 6.43; N, 19.34.
  • Example 17 2-[(Methoxyamino)methyl]-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinoline
  • Figure US20090062328A1-20090305-C00083
  • 2-(Chloromethyl)-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinoline (prepared according to the method described in parts A-C of Example 6 using 4-chloro-3-nitroquinoline in lieu of 7-(benzyloxy)-4-chloro-3-nitroquinoline) was converted to 2-[(methoxyamino)methyl]-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinoline using the method detailed in part E of Example 6. The product was provided as 0.11 g of a light yellow glass, Mp 58-63° C.
  • Anal. calcd for C18H22N4O2: C, 66.24; H, 6.79; N, 17.17. Found: C, 66.19; H, 6.60; N, 17.06.
  • Example 18 2-{[Methoxy(methyl)amino]methyl}-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinoline
  • Figure US20090062328A1-20090305-C00084
  • 2-(Chloromethyl)-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinoline (prepared according to the method described in parts A-C of Example 6 using 4-chloro-3-nitroquinoline in lieu of 7-(benzyloxy)-4-chloro-3-nitroquinoline) was converted 2-{[methoxy(methyl)amino]methyl}-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinoline using the method detailed in Example 7. The product was provided as 0.09 g of a white solid, mp 142-144° C.
  • Anal. calcd for C19H24N4O2: C, 67.04; H, 7.11; N, 16.46. Found: C, 67.16; H, 7.17; N, 16.49.
  • Example 19 2-[(Methoxyamino)methyl]-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-4-amine
  • Figure US20090062328A1-20090305-C00085
  • 2-[(Methoxyamino)methyl]-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-4-amine was prepared according to the methods described in parts A-E of Example 6 using 4-chloro-3-nitroquinoline in lieu of 7-(benzyloxy)-4-chloro-3-nitroquinoline. The product was provided as 0.16 g of a yellow solid, mp 169-171° C.
  • Anal. calcd for C18H23N5O2: C, 63.32; H, 6.79; N, 20.51. Found: C, 63.48; H, 6.62; N, 20.69.
  • Example 20 2-{[Methoxy(methyl)amino]methyl}-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-4-amine
  • Figure US20090062328A1-20090305-C00086
  • 2-(Chloromethyl)-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-4-amine (prepared according to the method described in parts A-D of Example 6 using 4-chloro-3-nitroquinoline in lieu of 7-(benzyloxy)-4-chloro-3-nitroquinoline) was converted to 2-{[methoxy(methyl)amino]methyl}-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-4-amine using the method detailed in Example 7. The product was provided as 0.16 g of a yellow solid, mp 223-226° C.
  • Anal. calcd for C19H25N5O2: C, 64.20; H, 7.09; N, 19.70. Found: C, 64.08; H, 7.06; N, 19.74.
  • Example 21 1-{4-Amino-7-bromo-2-[(methoxyamino)methyl]-1H-imidazo[4,5-c][1,5]naphthyridin-1-yl}-2-methylpropan-2-ol
  • Figure US20090062328A1-20090305-C00087
  • 1-{4-Amino-7-bromo-2-[(methoxyamino)methyl]-1H-imidazo[4,5-c][1,5]naphthyridin-1-yl}-2-methylpropan-2-ol was prepared according to the method described in Example 6, parts A-D, using 7-bromo-4-chloro-3-nitro[1,5]naphthyridine and 1-amino-2-methylpropan-2-ol in lieu of 7-(benzyloxy)-4-chloro-3-nitroquinoline and 1-tetrahydro-2H-pyran-4-ylmethanamine, respectively. The product was provided as 0.58 g of a yellow solid, mp 167-168° C.
  • Anal. calcd for C15H19BrN6O2: C, 45.58; H, 4.85; N, 21.26. Found: C, 45.81; H, 4.72; N, 21.45.
  • Example 22 1-{4-Amino-2-[(methoxyamino)methyl]-7-(pyridin-3-yl)-1H-imidazo[4,5-c][1,5]naphthyridin-1-yl}-2-methylpropan-2-ol
  • Figure US20090062328A1-20090305-C00088
  • A stream of nitrogen was bubbled through a stirring suspension of 1-{4-amino-7-bromo-2-[(methoxyamino)methyl]-1H-imidazo[4,5-c][1,5]naphthyridin-1-yl}-2-methylpropan-2-ol (0.590 g, 1.49 mmol),3-pyridineboronic acid (0.220 g, 1.79 mmol), and potassium carbonate (0.681 g, 4.93 mmol) in ethylene glycol dimethyl ether (10 mL) and water (5 mL) in a pressure vessel for 5 minutes. Dichlorobis(triphenylphosphine)palladium(III) (0.031 g, 0.045 mmol) was added and nitrogen was bubbled through for an additional 5 minutes. The pressure vessel was capped and placed in a 110° C. oil bath for 10 minutes. The resulting solution was cooled to ambient temperature and concentrated. The residue was purified by column chromatography using a HORIZON HPFC system (silica cartridge, eluting with 5-25% 1M ammonia/methanol in dichloromethane). The resulting yellow solid was crystallized from acetonitrile and isolated by filtration to yield 167 mg of 1-{4-amino-2-[(methoxyamino)methyl]-7-(pyridin-3-yl)-1H-imidazo[4,5-c][1,5]naphthyridin-1-yl}-2-methylpropan-2-ol as a light yellow solid, mp 195-197° C.
  • Anal. calcd for C20H23N7O2: C, 61.05; H, 5.89; N, 24.92. Found: C, 60.71; H, 5.91; N, 24.66.
  • Example 23 1-(4-Amino-7-bromo-2-{[methoxy(methyl)amino]methyl}-1H-imidazo[4,5-c][1,5]naphthyridin-1-yl)-2-methylpropan-2-ol
  • Figure US20090062328A1-20090305-C00089
  • 1-(4-Amino-7-bromo-2-{[methoxy(methyl)amino]methyl}-1H-imidazo[4,5-c][1,5]naphthyridin-1-yl)-2-methylpropan-2-ol was prepared according to the methods described in Example 6 parts A-D (using 7-bromo-4-chloro-3-nitro[1,5]naphthyridine and 1-amino-2-methylpropan-2-ol in lieu of 7-(benzyloxy)-4-chloro-3-nitroquinoline and 1-tetrahydro-2H-pyran-4-ylmethanamine, respectively) and Example 7. The product was provided as 0.63 g of a white solid, mp 190-192° C.
  • Anal. calcd for C16H21BrN6O2: C, 46.95; H, 5.17; N, 20.53. Found: C, 47.10; H, 4.91; N, 20.70.
  • Example 24 1-(4-Amino-2-{[methoxy(methyl)amino]methyl}-7-(pyridin-3-yl)-1H-imidazo[4,5-c][1,5]naphthyridin-1-yl)-2-methylpropan-2-ol
  • Figure US20090062328A1-20090305-C00090
  • 1-(4-Amino-7-bromo-2-{[methoxy(methyl)amino]methyl}-1H-imidazo[4,5-c][1,5]naphthyridin-1-yl)-2-methylpropan-2-ol was converted to 1-(4-amino-2-{[methoxy(methyl)amino]methyl}-7-(pyridin-3-yl)-1H-imidazo[4,5-c][1,5]naphthyridin-1-yl)-2-methylpropan-2-ol according to the method described in Example 22. The product was provided as 0.23 g of a tan solid, mp 187-189° C.
  • Anal. calcd for C21H25N7O2: C, 61.90; H, 6.18; N, 24.06. Found: C, 62.02; H, 6.14; N, 24.37.
  • Exemplary Compounds
  • Certain exemplary compounds, including some of those described above in the Examples; have the following Formulas (IIa, IIIa, IVa, or Va) and the following R1 and R2 substituents, wherein each line of the table is matched with Formula Ia, IIIa, IVa, or Va to represent a specific embodiment of the invention.
  • IIa
    Figure US20090062328A1-20090305-C00091
    IIIa
    Figure US20090062328A1-20090305-C00092
    IVa
    Figure US20090062328A1-20090305-C00093
    Va
    Figure US20090062328A1-20090305-C00094
    R1 R2
    2-methylpropyl —CH═N—OH
    2-hydroxy-2-methylpropyl —CH═N—OH
    2-methyl-2-[(methylsulfonyl)amino]propyl —CH═N—OH
    4-{(methylsulfonyl)amino]butyl —CH═N—OH
    2-methylpropyl —CH═N—OCH3
    2-hydroxy-2-methylpropyl —CH═N—OCH3
    2-methyl-2-[(methylsulfonyl)amino]propyl —CH═N—OCH3
    4-[(methylsu1fonyl)amino]butyl —CH═N—OCH3
    2-methylpropyl —CH2—NH—OH
    2-hydroxy-2-methylpropyl —CH2—NH—OH
    2-methyl-2-[(methylsulfonyl)amino]propyl —CH2—NH—OH
    4-[(methylsulfonyl)amino]butyl —CH2—NH—OH
    2-methylpropyl —CH2—N(CH3)—OH
    2-hydroxy-2-methylpropyl —CH2—N(CH3)—OH
    2-methyl-2-[(methylsulfonyl)amino]propyl —CH2—N(CH3)—OH
    4-[(methylsulfonyl)amino]butyl —CH2—N(CH3)—OH
    2-methylpropyl —CH2—NH—OCH3
    2-hydroxy-2-methylpropyl —CH2—NH—OCH3
    2-methyl-2-[(methylsulfonyl)amino]propyl —CH2—NH—OCH3
    4-[(methylsulfonyl)amino]butyl —CH2—NH—OCH3
    2-methylpropyl —CH2—N(CH3)—OCH3
    2-hydroxy-2-methylpropyl —CH2—N(CH3)—OCH3
    2-methyl-2-[(methylsulfonyl)amino]propyl —CH2—N(CH3)—OCH3
    4-[(methylsulfonyl)amino]butyl —CH2—N(CH3)—OCH3
    2-methylpropyl —CH2—N(—C(O)—CH3)—OH
    2-hydroxy-2-methylpropyl —CH2—N(—C(O)—CH3)—OH
    2-methyl-2-[(methylsulfonyl)amino]propyl —CH2—N(—C(O)—CH3)—OH
    4-[(methylsulfonyl)amino]butyl —CH2—N(—C(O)—CH3)—OH
    2-methylpropyl —CH2—N(—C(O)—CH3)—OCH3
    2-hydroxy-2-methylpropyl —CH2—N(—C(O)—CH3)—OCH3
    2-methyl-2-[(methylsulfonyl)amino]propyl —CH2—N(—C(O)—CH3)—OCH3
    4-[(methylsulfonyl)amino]butyl —CH2—N(—C(O)—CH3)—OCH3
    2-methylpropyl —CH2—N(—S(O)2—CH3)—OH
    2-hydroxy-2-methylpropyl —CH2—N(—S(O)2—CH3)—OH
    2-methyl-2-[(methylsulfonyl)amino]propyl —CH2—N(—S(O)2—CH3)—OH
    4-[(methylsulfonyl)amino]butyl —CH2—N(—S(O)2—CH3)—OH
    2-methylpropyl —CH2—N(—S(O)2—CH3)—OCH3
    2-hydroxy-2-methylpropyl —CH2—N(—S(O)2—CH3)—OCH3
    2-methyl-2-[(methylsulfonyl)amino]propyl —CH2—N(—S(O)2—CH3)—OCH3
    4-[(methylsulfonyl)amino]butyl —CH2—N(—S(O)2—CH3)—OCH3
    2-methylpropyl —CH2—N(—C(O)—NH—CH3)—OH
    2-hydroxy-2-methylpropyl —CH2—N(—C(O)—NH—CH3)—OH
    2-methyl-2-[(methylsulfonyl)amino]propyl —CH2—N(—C(O)—NH—CH3)—OH
    4-[(methylsulfonyl)amino]butyl —CH2—N(—C(O)—NH—CH3)—OH
    2-methylpropyl —CH2—N(—C(O)—NH—CH3)—OCH3
    2-hydroxy-2-methylpropyl —CH2—N(—C(O)—NH—CH3)—OCH3
    2-methyl-2-[(methylsulfonyl)amino]propyl —CH2—N(—C(O)—NH—CH3)—OCH3
    4-[(methylsulfonyl)amino]butyl —CH2—N(—C(O)—NH—CH3)—OCH3
  • Certain exemplary compounds, including some of those described above in the Examples, have the following Formulas (IIa, IIIa, IVa, or Va) and the following R1 and R2 substituents, wherein each line of the table is matched with Formula IIa, IIIa, IVa, or Va to represent a specific embodiment of the invention.
  • IIa
    Figure US20090062328A1-20090305-C00095
    IIIa
    Figure US20090062328A1-20090305-C00096
    IVa
    Figure US20090062328A1-20090305-C00097
    Va
    Figure US20090062328A1-20090305-C00098
    R1 R2
    (1-hydroxycyclobuyty)methyl —CH═N—OH
    (1-hydroxycyclopentyl)methyl —CH═N—OH
    (1-hydroxycyclohexyl)methyl —CH═N—OH
    tetrahydro-2H-pyran-4-ylmethyl —CH═N—OH
    (1-hydroxycyclobutyl)methyl —CH═N—OCH3
    (1-hydroxycyclopentyl)methyl —CH═N—OCH3
    (1-hydroxycyclohexyl)methyl —CH═N—OCH3
    tetrahydro-2H-pyran-4-ylmethyl —CH═N—OCH3
    (1-hydroxycyclobutyl)methyl —CH2—NH—OH
    (1-hydroxycyclopentyl)methyl —CH2—NH—OH
    (1-hydroxycyclohexyl)methyl —CH2—NH—OH
    tetrahydro-2H-pyran-4-ylmethyl —CH2—NH—OH
    (1-hydroxycyclobutyl)methyl —CH2—N(CH3)—OH
    (1-hydroxycyclopentyl)methyl —CH2—N(CH3)—OH
    (1-hydroxycyclohexyl)methyl —CH2—N(CH3)—OH
    tetrahydro-2H-pyran-4-ylmethyl —CH2—N(CH3)—OH
    (1-hydroxycyclobutyl)methyl —CH2—NH—OCH3
    (1-hydroxycyclopentyl)methyl —CH2—NH—OCH3
    (1-hydroxycyclohexyl)methyl —CH2—NH—OCH3
    tetrahydro-2H-pyran-4-ylmethyl —CH2—NH—OCH3
    (1-hydroxycyclobutyl)methyl —CH2—N(CH3)—OCH3
    (1-hydroxycyclopentyl)methyl —CH2—N(CH3)—OCH3
    (1-hydroxycyclohexyl)methyl —CH2—N(CH3)—OCH3
    tetrahydro-2H-pyran-4-ylmethyl —CH2—N(CH3)—OCH3
    (1-hydroxycyclobutyl)methyl —CH2—N(—C(O)—CH3)—OH
    (1-hydroxycyclopentyl)methyl —CH2—N(—C(O)—CH3)—OH
    (1-hydroxycyclohexyl)methyl —CH2—N(—C(O)—CH3)—OH
    tetrahydro-2H-pyran-4-ylmethyl —CH2—N(—C(O)—CH3)—OH
    (1-hydroxycyclobutyl)methyl —CH2—N(—C(O)—CH3)—OCH3
    (1-hydroxycyclopentyl)methyl —CH2—N(—C(O)—CH3)—OCH3
    (1-hydroxycyclohexyl)methyl —CH2—N(—C(O)—CH3)—OCH3
    tetrahydro-2H-pyran-4-ylmethyl —CH2—N(—C(O)—CH3)—OCH3
    (1-hydroxycyclobutyl)methyl —CH2—N(—S(O)2—CH3)—OH
    (1-hydroxycyclopentyl)methyl —CH2—N(—S(O)2—CH3)—OH
    (1-hydroxycyclohexyl)methyl —CH2—N(—S(O)2—CH3)—OH
    tetrahydro-2H-pyran-4-ylmethyl —CH2—N(—S(O)2—CH3)—OH
    (1-hydroxycyclobutyl)methyl —CH2—N(—S(O)2—CH3)—OCH3
    (1-hydroxycyclopentyl)methyl —CH2—N(—S(O)2—CH3)—OCH3
    (1-hydroxycyclohexyl)methyl —CH2—N(—S(O)2—CH3)—OCH3
    tetrahydro-2H-pyran-4-ylmethyl —CH2—N(—S(O)2—CH3)—OCH3
    (1-hydroxycyclobuytyl)methyl —CH2—N(—C(O)—NH—CH3)—OH
    (1-hydroxycyclopentyl)methyl —CH2—N(—C(O)—NH—CH3)—OH
    (1-hydroxycyclohexyl)methyl —CH2—N(—C(O)—NH—CH3)—OH
    tetrahydro-2H-pyran-4-ylmethyl —CH2—N(—C(O)—NH—CH3)—OH
    (1-hydroxycyclobutyl)methyl —CH2—N(—C(O)—NH—CH3)—OCH3
    (1-hydroxycyclopentyl)methyl —CH2—N(—C(O)—NH—CH3)—OCH3
    (1-hydroxycyclohexyl)methyl —CH2—N(—C(O)—NH—CH3)—OCH3
    tetrahydro-2H-pyran-4-ylmethyl —CH2—N(—C(O)—NH—CH3)—OCH3
  • Certain exemplary compounds, including some of those described above in the Examples, have the following Formulas (IIId, or Vd) and the following R1 and R2 substituents, wherein each line of the table is matched with Formula IIId or Vd to represent a specific embodiment of the invention.
  • IIId
    Figure US20090062328A1-20090305-C00099
    Vd
    Figure US20090062328A1-20090305-C00100
    R1 R2
    2-methylpropyl —CH═N—OH
    2-hydroxy-2-methylpropyl —CH═N—OH
    2-methyl-2-[(methylsulfonyl)amino]propyl —CH═N—OH
    4-[(methylsulfonyl)amino]butyl —CH═N—OH
    (1-hydroxycyclobutyl)methyl —CH═N—OH
    (1-hydroxycyclopentyl)methyl —CH═N—OH
    (1-hydroxycyclohexyl)methyl —CH═N—OH
    tetrahydro-2H-pyran-4-ylmethyl —CH═N—OH
    2-methylpropyl —CH═N—OCH3
    2-hydroxy-2-methylpropyl —CH═N—OCH3
    2-methyl-2-[(methylsulfonyl)amino]propyl —CH═N—OCH3
    4-[(methylsulfonyl)amino]butyl —CH═N—OCH3
    (1-hydroxycyolobutyl)methyl —CH═N—OCH3
    (1-hydroxycyclopentyl)methyl —CH═N—OCH3
    (1-hydroxycyclohexyl)methyl —CH═N—OCH3
    tetrahydro-2H-pyran-4-ylmethyl —CH═N—OCH3
    2-methylpropyl —CH2—NH—OH
    2-hydroxy-2-methylpropyl —CH2—NH—OH
    2-methyl-2-[(methylsulfonyl)amino]propyl —CH2—NH—OH
    4-[(methylsulfonyl)amino]butyl —CH2—NH—OH
    (1-hydroxycyclobutyl)methyl —CH2—NH—OH
    (1-hydroxycyclopentyl)methyl —CH2—NH—OH
    (1-hydroxycyclohexyl)methyl —CH2—NH—OH
    tetrahydro-2H-pyran-4-ylmethyl —CH2—NH—OH
    2-methylpropyl —CH2—N(CH3)—OH
    2-hydroxy-2-methylpropyl —CH2—N(CH3)—OH
    2-methyl-2-[(methylsulfonyl)amino]propyl —CH2—N(CH3)—OH
    4-[(methylsulfonyl)amino]butyl —CH2—N(CH3)—OH
    (1-hydroxycyclobutyl)methyl —CH2—N(CH3)—OH
    (1-hydroxycyclopentyl)methyl —CH2—N(CH3)—OH
    (1-hydroxycyclohexyl)methyl —CH2—N(CH3)—OH
    tetrahydro-2H-pyran-4-ylmethyl —CH2—N(CH3)—OH
    2-methylpropyl —CH2—NH—OCH3
    2-hydroxy-2-methylpropyl —CH2—NH—OCH3
    2-methyl-2-[(methylsulfonyl)amino]propyl —CH2—NH—OCH3
    4-[(methylsulfonyl)amino]butyl —CH2—NH—OCH3
    (1-hydroxycyclobutyl)methyl —CH2—NH—OCH3
    (1-hydroxycyclopentyl)methyl —CH2—NH—OCH3
    (1-hydroxycyclohexyl)methyl —CH2—NH—OCH3
    tetrahydro-2H-pyran-4-ylmethyl —CH2—NH—OCH3
    2-methylpropyl —CH2—N(CH3)—OCH3
    2-hydroxy-2-methylpropyl —CH2—N(CH3)—OCH3
    2-methyl-2-[(methylsulfonyl)amino]propyl —CH2—N(CH3)—OCH3
    4-[(methylsulfonyl)amino]butyl —CH2—N(CH3)—OCH3
    (1-hydroxycyclobutyl)methyl —CH2—N(CH3)—OCH3
    (1-hydroxycyclopentyl)methyl —CH2—N(CH3)—OCH3
    (1-hydroxycyclohexyl)methyl —CH2—N(CH3)—OCH3
    tetrahydro-2H-pyran-4-ylmethyl —CH2—N(CH3)—OCH3
    2-methylpropyl —CH2—N(—C(O)—CH3)—OH
    2-hydroxy-2-methylpropyl —CH2—N(—C(O)—CH3)—OH
    2-methyl-2-[(methylsulfonyl)amino]propyl —CH2—N(—C(O)—CH3)—OH
    4-[(methylsulfonyl)amino]butyl —CH2—N(—C(O)—CH3)—OH
    (1-hydroxycyclobutyl)methyl —CH2—N(—C(O)—CH3)—OH
    (1-hydroxycyclopentyl)methyl —CH2—N(—C(O)—CH3)—OH
    (1-hydroxycyclohexyl)methyl —CH2—N(—C(O)—CH3)—OH
    tehydro-2H-pyran-4-ylmethyl —CH2—N(—C(O)—CH3)—OH
    2-methylpropyl —CH2—N(—C(O)—CH3)—OCH3
    2-hydroxy-2-methylpropyl —CH2—N(—C(O)—CH3)—OCH3
    2-methyl-2-[(methylsulfonyl)amino]propyl —CH2—N(—C(O)—CH3)—OCH3
    4-[(methylsulfonyl)amino]butyl —CH2—N(—C(O)—CH3)—OCH3
    (1-hydroxycyclobutyl)methyl —CH2—N(—C(O)—CH3)—OCH3
    (1-hydxoxycyclopentyl)methyl —CH2—N(—C(O)—CH3)—OCH3
    (1-hydroxycyclohexyl)methyl —CH2—N(—C(O)—CH3)—OCH3
    tetrahydro-2H-pyran-4-ylmethyl —CH2—N(—C(O)—CH3)—OCH3
    2-methylpropyl —CH2—N(—S(O)2—CH3)—OH
    2-hydroxy-2-methylpropyl —CH2—N(—S(O)2—CH3)—OH
    2-methyl-2-[(methylsulfonyl)amino]propyl —CH2—N(—S(O)2—CH3)—OH
    4-[(methylsulfonyl)amino]butyl —CH2—N(—S(O)2—CH3)—OH
    (1-hydroxycyclobutyl)methyl —CH2—N(—S(O)2—CH3)—OH
    (1-hydroxycyclopentyl)methyl —CH2—N(—S(O)2—CH3)—OH
    (1-hydroxycyclohexyl)methyl —CH2—N(—S(O)2—CH3)—OH
    tetrahydro-2H-pyran-4-ylmethyl —CH2—N(—S(O)2—CH3)—OH
    2-methylpropyl —CH2—N(—S(O)2—CH3)—OCH3
    2-hydroxy-2-methylpropyl —CH2—N(—S(O)2—CH3)—OCH3
    2-methyl-2-[(methylsulfonyl)amino]propyl —CH2—N(—S(O)2—CH3)—OCH3
    4-[(methylsulfonyl)amino]butyl —CH2—N(—S(O)2—CH3)—OCH3
    (1-hyclroxycyclobutyl)methyl —CH2—N(—S(O)2—CH3)—OCH3
    (1-hydroxycyclopentyl)methyl —CH2—N(—S(O)2—CH3)—OCH3
    (1-hydroxycyclohexyl)methyl —CH2—N(—S(O)2—CH3)—OCH3
    tetrahydro-2H-pyran-4-ylmethyl —CH2—N(—S(O)2—CH3)—OCH3
    2-methylpropyl —CH2—N(—C(O)—NH—CH3)—OH
    2-hydroxy-2-methylpropyl —CH2—N(—C(O)—NH—CH3)—OH
    2-methyl-2-[(methylsulfonyl)amino]propyl —CH2—N(—C(O)—NH—CH3)—OH
    4-[(methylsulfonyl)amino]butyl —CH2—N(—C(O)—NH—CH3)—OH
    (1-hydroxycyclobutyl)methyl —CH2—N(—C(O)—NH—CH3)—OH
    (1-hydroxycyclopentyl)methyl —CH2—N(—C(O)—NH—CH3)—OH
    (1-hydroxycyclohexyl)methyl —CH2—N(—C(O)—NH—CH3)—OH
    tetrahydro-2H-pyran-4-ylmethyl —CH2—N(—C(O)—NH—CH3)—OH
    2-methylpropyl —CH2—N(—C(O)—NH—CH3)—OCH3
    2-hydroxy-2-methylpropyl —CH2—N(—C(O)—NH—CH3)—OCH3
    2-methyl-2-[(methylsulfonyl)amino]propyl —CH2—N(—C(O)—NH—CH3)—OCH3
    4-[(methylsulfonyl)amino]butyl —CH2—N(—C(O)—NH—CH3)—OCH3
    (1-hydroxycyclobutyl)methyl —CH2—N(—C(O)—NH—CH3)—OCH3
    (1-hydroxycyclopentyl)methyl —CH2—N(—C(O)—NH—CH3)—OCH3
    (1-hydroxycyclohexyl)methyl —CH2—N(—C(O)—NH—CH3)—OCH3
    tetrahydro-2H-pyran-4-ylmethyl —CH2—N(—C(O)—NH—CH3)—OCH3
  • 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, N.J.). 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, Fla.) centrifuge frit tubes containing density gradient medium. The PBMC layer is collected and washed twice with DPBS or HBSS and re-suspended at 4×106 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 2×106 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×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, N.J. 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 AEC3712) from Biosource International, Camarillo, Calif. 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-1H-imidazo[4,5-c]quinolin-1-yl]ethanol hydrate (U.S. Pat. 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, N.J.). Whole blood is placed in Accuspin (Sigma) or LeucoSep (Greiner Bio-One, Inc., Longwood, Fla.) centrifuge frit tubes containing density gradient medium. The PBMC layer is collected and washed twice with DPBS or HBSS and re-suspended at 4×106 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-1H-imidazo[4,5-c]quinolin-1-yl]ethanol hydrate (U.S. Pat. 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 2×106 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 4° C. 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 −70° C. 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 a capture antibody (PBL Biomedical Laboratories, Piscataway, N.J.) 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, Minn.) 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-1H-imidazo[4,5-c]quinolin-1-yl]ethanol hydrate (U.S. Pat. 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-α,α-dimethyl-1H-imidazo[4,5-c]quinolin-1-yl]ethanol hydrate (U.S. Pat. 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 (1 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-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 (28)

1. A compound of the Formula I:
Figure US20090062328A1-20090305-C00101
wherein:
Z is selected from the group consisting of:
—C(═N—O—R2-2)— and
—C(R2-4)(—N(—OR2-2)—Y—R2-3)—;
X is selected from the group consisting of a bond, C1-4 alkylene and C2-4 alkenylene;
R2-1, R2-2, and R2-3 are independently selected from the group consisting of:
hydrogen,
alkyl,
alkenyl,
aryl,
arylalkylenyl,
heteroaryl,
heteroarylalkylenyl,
heterocyclyl,
heterocyclylalkylanyl, and
alkyl, alkenyl, aryl, arylalkylenyl, heteroaryl, hetoroarylalkylenyl, heterocyclyl, and heterocyclylalkylenyl, substituted by one or more substituents selected from the group consisting of:
hydroxy,
alkyl,
haloalkyl,
hydroxyalkyl,
alkoxy,
dialkylamino,
—S(O)0-2R2-5,
—NH—S(O)2—R2-5,
haloalkoxy,
halogen,
cyano,
nitro,
—N3,
aryl,
heteroaryl,
heterocyclyl,
aryloxy,
arylalkyleneoxy,
—C(O)—O-alkyl,
—C(O)—N(R8)2,
—N(R8)—C(O)—R2-5,
—NH—C(O)—NH—R2-5,
—NH—C(O)—NH2,
—O—(CO)-alkyl, and
—C(O)-alkyl;
with the proviso that R2-2 is other than alkenyl wherein the carbon atom bonded to —O— is doubly bonded to another carbon atom;
R2-4 is selected from the group consisting of hydrogen, C1-4alkyl, and phenyl;
R2-5 is selected from the group consisting of alkyl, aryl, arylalkylenyl, heteroaryl, and heteroarylalkylenyl, each of which is unsubstituted or substituted by one or more substituents selected from the group consisting of halogen, cyano, nitro, alkoxy, dialkylamino, alkylthio, haloalkyl, haloalkoxy, alkyl, and —N3;
Y is selected from the group consisting of:
a bond,
—C(R6)—,
—S(O)2—,
—S(O)2—N(R8)—,
Figure US20090062328A1-20090305-C00102
—C(O)—O—,
—C(R6)—N(R8)—,
—C(O)—N(R8)—S(O)2—,
—C(R6)—N(R8)—C(O)—,
Figure US20090062328A1-20090305-C00103
—C(O)—C(O)—,
—C(O)—C(O)—O—, and
—C(═NH)—N(R8)—;
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 benzene ring or fused pyridine ring wherein the fused benzene ring or fused pyridine ring is unsubstituted or substituted by one or more R′″ groups;
or when taken together, RA and RB form a fused cyclohexene ring or a fused tetrahydropyridine ring, wherein the fused cyclohexene or tetrahydropyridine ring is unsubstituted or substituted by one or more R groups;
R is selected from the group consisting of:
halogen,
hydroxy,
alkyl,
alkenyl,
haloalkyl,
alkoxy,
alkylthio, and
N(R9)2;
R′ is hydrogen or a non-interfering substituent;
R′″ is a non interfering substituent;
R6 is selected from the group consisting of —O and ═S;
R8 is selected from the group consisting of hydrogen, C1-10 alkyl, C2-10 alkenyl, C1-10 alkoxy-C1-10 alkylenyl, hydroxy-C1-10 alkylenyl, heteroaryl-C1-10 alkylenyl, and aryl-C1-10 alkylenyl;
R9 is selected from the group consisting of hydrogen and alkyl; and
R10 is C3-8 alkylene;
or a pharmaceutically acceptable salt thereof.
2. The compound or salt of claim 1 wherein the one or more R′″ groups are one R3 group, or one R3 group and one R group, or one, two, three, or four R groups when on the fused benzene ring, or one, two, or three R groups when on the fused pyridine ring; wherein R3 is selected from the group consisting of:
-Z′-R4,
-Z′-X′—R4,
-Z′-X′—Y′—R4,
-Z′-X′—Y′—X′—Y′—R4, and
-Z′-X′—R5;
X′ is selected from the group consisting of alkylene, alkenylene, alkynylene, arylene, heteroarylene, and heterocyclylene, wherein the alkylene, alkenylene, and alkynylene groups can be optionally interrupted or terminated by arylene, heteroarylene or heterocyclylene and optionally interrupted by one or more —O— groups;
Y′ is selected from the group consisting of:
—O—
—S(O)0-2—,
—S(O)2—N(R8)—,
—C(R6)—,
—C(R6)—O—,
—O—C(R6)—,
—O—C(O)—O—,
—N(R8)-Q-,
—C(R6)—N(R8)—,
—O—C(R6)—N(R8)—,
—C(R6)—N(OR9)—,
—O—N(R8)-Q-,
—O—N═C(R4)—,
—C(—N—O—R9)—,
—CH(—N(—O—R8)-Q-R4)—,
Figure US20090062328A1-20090305-C00104
Z′ is a bond or —O—;
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, arylalkleneoxy, heteroaryl, heteroaryloxy, heteroarylalkleneoxy, heterocyclyl, amino, alkylamino, dialkylamino, (dialkylamino)alkyleneoxy, and in the case of alkyl, alkenyl, alkynyl, and heterocyclyl, oxo;
R5 is selected from the group consisting of:
Figure US20090062328A1-20090305-C00105
R6 is selected from the group consisting of ═O and ═S;
R7 is C2-7 alkylene;
R8 is selected from the group consisting of hydrogen, C1-10 alkyl, C2-10 alkenyl, C1-10 alkoxy-C1-10 alkylenyl, hydroxy-C1-10 alkylenyl, heteroaryl-C1-10 alkylenyl, and aryl-C1-10 alkylenyl;
R9 is selected from the group consisting of hydrogen and alkyl;
R10 is C3-8 alkylene;
A is selected from the group consisting of —O—, —C(O)—, —S(O)0-2—, —CH2—, and —N(-Q-R4)—,
A′ is selected from the group consisting of —O—, —S(O)0-2—, N(-Q-R4)—, and —CH2—;
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(R6)—O—, —C(R6)—S—, and —C(R6)—N(OR9)—;
V is selected from the group consisting of —C(R6)—, —O—C(R6)—, N(R8)—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.
3. The compound or salt of claim 1 wherein R′ is R1; wherein R1 is selected from the group consisting of:
—R4,
—X′—R4,
—X′—Y′—R4,
—X′—Y′—X′—Y′—R4, and
—X′—R5;
wherein:
X′ is selected from the group consisting of alkylene, alkenylene, alkynylene, arylene, heteroarylene, and heterocyclylene, wherein the alkylene, alkenylene, and alkynylene groups can be optionally interrupted or terminated by arylene, heteroarylene or heterocyclylene and optionally interrupted by one or more —O— groups;
Y′ is selected from the group consisting of:
—O—,
—S(O)0-2,
—S(O)2—N(R8)—,
—C(R6)—,
—C(R6)—O—,
—O—C(R6)—,
—O—C(O)—O—,
—N(R8)-Q-,
—C(R6)—N(R8)—,
—O—C(R6)—N(R8)—,
—C(R6)—N(OR9)—,
—O—N(R8)-Q-,
—O—N═C(R4)—,
—C(═N—O—R8)—,
—CH(—N(—O—R8)-Q-R4)—,
Figure US20090062328A1-20090305-C00106
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, arylalkleneoxy, heteroaryl, heteroaryloxy, heteroarylalkleneoxy, heterocyclyl, amino, alkylamino, dialkylamino, (dialkylamino)alkyleneoxy, and in the case of alkyl, alkenyl, alkynyl, and heterocyclyl, oxo;
R5 is selected from the group consisting of:
Figure US20090062328A1-20090305-C00107
R6 is selected from the group consisting of ═O and ═S;
R7 is C2-7 alkylene;
R8 is selected from the group consisting of hydrogen, C1-10 alkyl, C2-10 alkenyl, C1-10 alkoxy-C1-10 alkylenyl, hydroxy-C1-10 alkylenyl, heteroaryl-C1-10 alkylenyl, and aryl-C1-10 alkylenyl;
R9 is selected from the group consisting of hydrogen and alkyl;
R10 is C3-8 alkylene;
A is selected from the group consisting of —O—, —C(O)—, —S(O)0-2—, —CH2—, and —N(-Q-R4)—,
A′ is selected from the group consisting of —O—, —S(O)0-2—, N(-Q-R4)—, and —CH2—;
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(R6)—O—, —C(R6)—S—, and —C(R6)—N(OR9)—;
V is selected from the group consisting of —C(R6)—, —O—C(R6)—, N(R8)—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.
4. The compound of claim 1, wherein Formula I is Formula II:
Figure US20090062328A1-20090305-C00108
wherein:
Z is selected from the group consisting of:
—C(═N—O—R2-2)— and
—C(R2-4)(—N(—OR2-2)—Y—R2-3)—;
X is selected from the group consisting of a bond, C1-4 alkylene and C2-4 alkenylene;
R2-1, R2-2, and R2-3 are independently selected from the group consisting of:
hydrogen,
alkyl,
alkenyl,
aryl,
arylalkylenyl,
heteroaryl,
heteroarylalkylenyl,
heterocyclyl,
heterocyclylalkylenyl, and
alkyl, alkenyl, aryl, arylalkylenyl, heteroaryl, heteroarylalkylenyl, heterocyclyl, and heterocyclylalkylenyl, substituted by one or more substituents selected from the group consisting of:
hydroxy,
alkyl,
haloalkyl,
hydroxyalkyl,
alkoxy,
dialkylamino,
—S(O)0-2R2-5,
—NH—S(O)2—R2-5,
haloalkoxy,
halogen,
cyano,
nitro,
—N3,
aryl,
heteroaryl,
heterocyclyl,
aryloxy,
arylalkyleneoxy,
—C(O)—O-alkyl,
—C(O)—N(R8)2,
—N(R8)—C(O)—R2-5,
—NH—C(O)—NH—R2-5,
—NH—C(O)—NH2,
—O—(CO)-alkyl, and
—C(O)-alkyl;
with the proviso that R2-2 is other than alkenyl wherein the carbon atom bonded to —O— is doubly bonded to another carbon atom,
R2-4 is selected from the group consisting of hydrogen, C1-4alkyl, and phenyl;
R2-5 is selected from the group consisting of alkyl, aryl, arylalkylenyl, heteroaryl, and heteroarylalkylenyl, each of which is unsubstituted or substituted by one or more substituents selected from the group consisting of halogen, cyano, nitro, alkoxy, dialkylamino, alkylthio, haloalkyl, haloalkoxy, alkyl, and —N3;
Y is selected from the group consisting of:
a bond,
—C(R6)—,
—S(O)2,
—S(O)2—N(R8)—,
Figure US20090062328A1-20090305-C00109
—C(O)—O—,
—C(R6)—N(R8)—,
—C(O)—N(R8)—S(O)2—,
—C(R6)—N(R8)—C(O)—,
Figure US20090062328A1-20090305-C00110
—C(O)—C(O)—,
—C(O)—C(O)—O—, and
—C(═NH)—N(R8)—,
RA1 and RB1 are each independently selected from the group consisting of:
hydrogen,
halogen,
alkyl,
alkenyl,
alkoxy,
alkylthio, and
N(R9)2;
R1 is selected from the group consisting of:
—R4,
—X′—R4,
—X′—Y′—R4,
—X′—Y′—X′—Y′—R4, and
—X′—R5;
X′ is selected from the group consisting of alkylene, alkenylene, alkynylene, arylene, heteroarylene, and heterocyclylene, wherein the alkylene, alkenylene, and alkynylene groups can be optionally interrupted or terminated by arylene, heteroarylene or heterocyclylene and optionally interrupted by one or more —O— groups;
Y′ is selected from the group consisting of:
—O—,
—S(O)0-2—,
—S(O)2—N(R8)—,
—C(R6)—,
—C(R6)—O—,
—O—C(R6)—
—O—C(O)—O,
—N(R8)-Q-,
—C(R6)—N(R8)—,
—O—C(R6)—N(R8)—,
—C(R6)—N(OR9)—,
—O—N(R8)-Q-,
—O—N═C(R4)—,
—C(═N—O—R8)—,
—CH(—N(—O—R8)-Q-R4)—,
Figure US20090062328A1-20090305-C00111
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, arylalkleneoxy, heteroaryl, heteroaryloxy, heteroarylalkleneoxy, heterocyclyl, amino, alkylamino, dialkylamino, (dialkylamino)alkyleneoxy, and in the case of alkyl, alkenyl, alkynyl, and heterocyclyl, oxo;
R5 is selected from the group consisting of:
Figure US20090062328A1-20090305-C00112
R6 is selected from the group consisting of ═O and ═S;
R7 is C2-7 alkylene;
R8 is selected from the group consisting of hydrogen, C1-10 alkyl, C2-10 alkenyl, C1-10 alkoxy-C1-10 alkylenyl, hydroxy-C1-10alkylenyl, heteroaryl-C1-10alkylenyl, and aryl-C1-10 alkylenyl;
R9 is selected from the group consisting of hydrogen and alkyl;
R10 is C3-8 alkylene;
A is selected from the group consisting of —O—, —C(O)—, —S(O)0-2—, —CH2—, and —N(-Q-R4)—,
A′ is selected from the group consisting of —O—, —S(O)0-2—, N(-Q-R4)—, and —CH2—;
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(R6)—O—, —C(R6)—S—, and —C(R6)—N(OR9)—;
V is selected from the group consisting of —C(R6)—, —O—C(R6)—, N(R8)—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. The compound of claim 1, wherein Formula I is Formula III:
Figure US20090062328A1-20090305-C00113
wherein:
Z is selected from the group consisting of:
—C(═N—O—R2-2)— and
—C(R2-4)(—N(—OR2-2)—Y—R2-3)—;
X is selected from the group consisting of a bond, C1-4 alkylene and C2-4 alkenylene;
R2-1, R2-2, and R2-3 are independently selected from the group consisting of:
hydrogen,
alkyl,
alkenyl,
aryl,
arylalkylenyl,
heteroaryl,
heteroarylalkylenyl,
heterocyclyl,
heterocyclylalkylenyl, and
alkyl, alkenyl, aryl, arylalkylenyl, heteroaryl, heteroarylalkylenyl, heterocyclyl, and heterocyclylalkylenyl, substituted by one or more substituents selected from the group consisting of:
hydroxy,
alkyl,
haloalkyl,
hydroxyalkyl,
alkoxy,
dialkylamino,
—S(O)0-2R2-5,
—NH—S(O)2—R2-5,
haloalkoxy,
halogen,
cyano,
nitro,
—N3,
aryl,
heteroaryl,
heterocyclyl,
aryloxy,
arylalkyleneoxy,
—C(O)—O-alkyl,
—C(O)—N(R8)2,
—N(R8)—C(O)—R2-5,
—NH—C(O)—NH—R2-5,
—NH—C(O)—NH2,
—O—(CO)-alkyl, and
—C(O)-alkyl;
with the proviso that R2-2 is other than alkenyl wherein the carbon atom bonded to —O— is doubly bonded to another carbon atom;
R2-4 is selected from the group consisting of hydrogen, C1-4alkyl, and phenyl;
R2-5 is selected from the group consisting of alkyl, aryl, arylalkylenyl, heteroaryl, and heteroarylalkylenyl, each of which is unsubstituted or substituted by one or more substituents selected from the group consisting of halogen, cyano, nitro, alkoxy, dialkylamino, alkylthio, haloalkyl, haloalkoxy, alkyl, and —N3;
Y is selected from the group consisting of:
a bond,
—C(R6)—,
—S(O)2,
—S(O)2—N(R8)—,
Figure US20090062328A1-20090305-C00114
—C(O)—O—,
—C(R6)—N(R8)—,
—C(O)—N(R8)—S(O)2—,
—C(R6)—N(R8)—C(O)—,
Figure US20090062328A1-20090305-C00115
—C(O)—C(O)—,
—C(O)—C(O)—O—, and
—C(═NH)—N(R8)—,
R is selected from the group consisting of:
halogen,
hydroxy,
alkyl,
alkenyl,
haloalkyl,
alkoxy,
alkylthio, and
—N(R9)2;
n is an integer from 0 to 4;
R1 is selected from the group consisting of:
—R4,
—X′—R4,
—X′—Y′—R4,
—X′—Y′—X′—Y′—R4, and
—X′—R5;
R3 is selected from the group consisting of:
-Z′-R4,
-Z′-X′—R4,
-Z′-X′—Y′—R4,
-Z′-X′—Y′—X′—Y′—R4, and
-Z′-X′—R5;
m is 0 or 1, with the proviso that when m is 1 than n is 0 or 1;
X′ is selected from the group consisting of alkylene, alkenylene, alkynylene, arylene, heteroarylene, and heterocyclylene, wherein the alkylene, alkenylene, and alkynylene groups can be optionally interrupted or terminated by arylene, heteroarylene or heterocyclylene and optionally interrupted by one or more —O— groups;
Y′ is selected from the group consisting of:
—O—,
—S(O)0-2—,
—S(O)2—N(R8)—,
—C(R6)—,
—C(R6)—O—,
—O—C(R6)—
—O—C(O)—O,
—N(R8)-Q-,
—C(R6)—N(R8)—,
—O—C(R6)—N(R8)—,
—C(R6)—N(OR9)—,
—O—N(R8)-Q-,
—O—N═C(R4)—,
—C(═N—O—R8)—,
—CH(—N(—O—R8)-Q-R4)—,
Figure US20090062328A1-20090305-C00116
Z′ is a bond or —O—;
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, arylalkleneoxy, heteroaryl, heteroaryloxy, heteroarylalkleneoxy, heterocyclyl, amino, alkylamino, dialkylamino, (dialkylamino)alkyleneoxy, and in the case of alkyl, alkenyl, alkynyl, and heterocyclyl, oxo;
R5 is selected from the group consisting of:
Figure US20090062328A1-20090305-C00117
R6 is selected from the group consisting of ═O and ═S;
R7 is C2-7 alkylene;
R8 is selected from the group consisting of hydrogen, C1-10 alkyl, C2-10 alkenyl, C1-10 alkoxy-C1-10 alkylenyl, hydroxy-C1-10 alkylenyl, heteroaryl-C1-10 alkylenyl, and aryl-C1-10 alkylenyl;
R9 is selected from the group consisting of hydrogen and alkyl;
R10 is C3-8 alkylene;
A is selected from the group consisting of —O—, —C(O)—, —S(O)0-2—, —CH2—, and —N(-Q-R4)—,
A′ is selected from the group consisting of —O—, —S(O)0-2—, N(-Q-R4)—, and —CH2—;
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(R6)—O—, —C(R6)—S—, and —C(R6)—N(OR9)—;
V is selected from the group consisting of —C(R6)—, —O—C(R6)—, N(R8)—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. (canceled)
7. The compound of claim 1, wherein Formula I is Formula IV:
Figure US20090062328A1-20090305-C00118
wherein:
Z is selected from the group consisting of:
—C(═N—O—R2-2)— and
—C(R2-4)(—N(—OR2-2)—Y—R2-3)—;
X is selected from the group consisting of a bond, C1-4 alkylene and C2-4 alkenylene;
R2-1, R2-2, and R2-3 are independently selected from the group consisting of:
hydrogen,
alkyl,
alkenyl,
aryl,
arylalkylenyl,
heteroaryl,
heteroarylalkylenyl,
heterocyclyl,
heterocyclylalkylenyl, and
alkyl, alkenyl, aryl, arylalkylenyl, heteroaryl, heteroarylalkylenyl, heterocyclyl, and heterocyclylalkylenyl, substituted by one or more substituents selected from the group consisting of:
hydroxy,
alkyl,
haloalkyl,
hydroxyalkyl,
alkoxy,
dialkylamino,
—S(O)0-2R2-5,
—NH—S(O)2—R2-5,
haloalkoxy,
halogen,
cyano,
nitro,
—N3,
aryl,
heteroaryl,
heterocyclyl,
aryloxy,
arylalkyleneoxy,
—C(O)—O-alkyl,
—C(O)—N(R8)2,
—N(R8)—C(O)—R2-5,
—NH—C(O)—NH—R2-5,
—NH—C(O)—NH2,
—O—(CO)-alkyl, and
—C(O)-alkyl;
with the proviso that R2-2 is other than alkenyl wherein the carbon atom bonded to —O— is doubly bonded to another carbon atom,
R2-4 is selected from the group consisting of hydrogen, C1-4alkyl, and phenyl;
R2-5 is selected from the group consisting of alkyl, aryl, arylalkylenyl, heteroaryl, and heteroarylalkylenyl, each of which is unsubstituted or substituted by one or more substituents selected from the group consisting of halogen, cyano, nitro, alkoxy, dialkylamino, alkylthio, haloalkyl, haloalkoxy, alkyl, and —N3;
Y is selected from the group consisting of:
a bond,
—C(R6)—,
—S(O)2,
—S(O)2—N(R8)—,
Figure US20090062328A1-20090305-C00119
—C(O)—O—,
—C(R6)—N(R8)—,
—C(O)—N(R8)—S(O)2—,
—C(R6)—N(R8)—C(O)—,
Figure US20090062328A1-20090305-C00120
—C(O)—C(O)—,
—C(O)—C(O)—O—, and
—C(═NH)—N(R8)—,
R is selected from the group consisting of:
halogen,
hydroxy,
alkyl,
alkenyl,
haloalkyl,
alkoxy,
alkylthio, and
—N(R9)2;
p is an integer from 0 to 3;
R1 is selected from the group consisting of:
—R4,
—X′—R4,
—X′—Y′—R4,
—X′—Y′—X′—Y′—R4, and
—X′—R5;
R3 is selected from the group consisting of:
-Z′-R4,
-Z′-X′—R4,
-Z′-X′—Y′—R4,
-Z′-X′—Y′—X′—Y′—R4, and
-Z′-X′—R5;
m is 0 or 1, with the proviso that when m is 1 then p is 0 or 1;
X′ is selected from the group consisting of alkylene, alkenylene, alkynylene, arylene, heteroarylene, and heterocyclylene, wherein the alkylene, alkenylene, and alkenylene groups can be optionally interrupted or terminated by arylene, heteroarylene or heterocyclylene and optionally interrupted by one or more —O— groups;
Y′ is selected from the group consisting of:
—O—,
—S(O)0-2—,
—S(O)2—N(R8)—,
—C(R6)—,
—C(R6)—O—,
—O—C(R6)—
—O—C(O)—O,
—N(R8)-Q-,
—C(R6)—N(R8)—,
—O—C(R6)—N(R8)—,
—C(R6)—N(OR9)—,
—O—N(R8)-Q-,
—O—N═C(R4)—,
—C(═N—O—R8)—,
—CH(—N(—O—R8)-Q-R4)—,
Figure US20090062328A1-20090305-C00121
Z′ is a bond or —O—;
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, arylalkleneoxy, heteroaryl, heteroaryloxy, heteroarylalkleneoxy, heterocyclyl, amino, alkylamino, dialkylamino, (dialkylamino)alkyleneoxy, and in the case of alkyl, alkenyl, alkynyl, and heterocyclyl, oxo;
R5 is selected from the group consisting of:
Figure US20090062328A1-20090305-C00122
R6 is selected from the group consisting of ═O and ═S;
R7 is C2-7 alkylene;
R8 is selected from the group consisting of hydrogen, C1-10 alkyl, C2-10 alkenyl, C1-10 alkoxy-C1-10 alkylenyl, hydroxy-C1-10 alkylenyl, heteroaryl-C1-10 alkylenyl, and aryl-C1-10 alkylenyl;
R9 is selected from the group consisting of hydrogen and alkyl;
R10 is C3-8 alkylene;
A is selected from the group consisting of —O—, —C(O)—, —S(O)0-2—, —CH2—, and —N(-Q-R4)—,
A′ is selected from the group consisting of —O—, —S(O)0-2—, N(-Q-R4)—, and —CH2—;
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(R6)—O—, —C(R6)—S—, and —C(R6)—N(OR9)—;
V is selected from the group consisting of —C(R6)—, —O—C(R6)—, N(R8)—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.
8. (canceled)
9. A compound of Formula VII:
Figure US20090062328A1-20090305-C00123
wherein:
G 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(═NY1)—R″,
—CH(OH)—C(O)—OY1,
—CH(OC1-4alkyl)Y0,
—CH2Y2, and
—CH(CH3)Y2;
R″ and R″″ are independently selected from the group consisting of C1-10alkyl, C3-7cycloalkyl, 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, C1-6alkyl, C1-4alkoxy, aryl heteroaryl, aryl-C1-4alkylenyl, heteroaryl-C1-4alkylenyl, halo-C1-4alkylenyl, halo-C1-4alkoxy, —O—C(O)—CH3, —C(O)—O—CH3, —C(O)—NH2, —O—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 α-amino acid selected from the group consisting of racemic, D-, and L-amino acids;
Y1 is selected from the group consisting of hydrogen, C1-6alkyl, and benzyl;
Y0 is selected from the group consisting of C1-6alkyl, carboxy-C1-6alkylenyl, amino-C1-4alkylenyl, mono-N—C1-6alkylamino-C1-4alkylenyl, and di-N,N—C1-6alkylamino-C1-4alkylenyl;
Y2 is selected from the group consisting of mono-N—C1-6alkylamino, di-N,N—C1-6alkylamino, morpholin-4-yl, piperidin-1-yl, pyrrolidin-1-yl, and 4-C1-4alkylpiperazin-1-yl;
RA2 and RB2 are each independently selected from the group consisting of;
hydrogen,
halogen,
alkyl,
alkenyl,
alkoxy,
alkylthio, and
—N(R9)2;
or when taken together, RA2 and RB2 form a fused benzene ring or fused pyridine ring wherein the fused benzene ring or fused pyridine ring is unsubstituted or substituted by one R3 group, or one R group and one R group, or one, two, three, or four R groups when on the fused benzene ring, or one, two, or three R groups when on the fused pyridine ring;
or when taken together, RA2 and RB2 form a fused cyclohexene ring or a fused tetrahydropyridine ring, wherein the fused cyclohexene or tetrahydropyridine ring is unsubstituted or substituted by one or more R groups;
Z is selected from the group consisting of:
—C(═N—O—R2-2)— and
—C(R2-4)(—N(—OR2-2)—Y—R2-3)—;
X is selected from the group consisting of a bond, C1-4 alkylene and C2-4 alkenylene;
R2-1, R2-2, and R2-3 are independently selected from the group consisting of:
hydrogen,
alkyl,
alkenyl,
aryl,
arylalkylenyl,
heteroaryl,
heteroarylalkylenyl,
heterocyclyl,
heterocyclylalkylenyl, and
alkyl, alkenyl, aryl, arylalkylenyl, heteroaryl, heteroarylalkylenyl, heterocyclyl, and heterocyclylalkylenyl, substituted by one or more substituents selected from the group consisting of:
hydroxy,
alkyl,
haloalkyl,
hydroxyalkyl,
alkoxy,
dialkylamino,
—S(O)0-2R2-5,
—NH—S(O)2—R2-5,
haloalkoxy,
halogen,
cyano,
nitro,
—N3,
aryl,
heteroaryl,
heterocyclyl,
aryloxy,
arylalkyleneoxy,
—C(O)—O-alkyl,
—C(O)—N(R8)2,
—N(R8)—C(O)—R2-5,
—NH—C(O)—NH—R2-5,
—NH—C(O)—NH2,
—O—(CO)-alkyl, and
—C(O)-alkyl;
with the proviso that R2-2 is other than alkenyl wherein the carbon atom bonded to —O— is doubly bonded to another carbon atom,
R2-4 is selected from the group consisting of hydrogen, C1-4alkyl, and phenyl;
R2-5 is selected from the group consisting of alkyl, aryl, arylalkylenyl, heteroaryl, and heteroarylalkylenyl, each of which is unsubstituted or substituted by one or more substituents selected from the group consisting of halogen, cyano, nitro, alkoxy, dialkylamino, alkylthio, haloalkyl, haloalkoxy, alkyl, and —N3;
Y is selected from the group consisting of:
a bond,
—C(R6)—,
—S(O)2,
—S(O)2—N(R8)—,
Figure US20090062328A1-20090305-C00124
—C(O)—O—,
—C(R6)—N(R8)—,
—C(O)—N(R8)—S(O)2—,
—C(R6)—N(R8)—C(O)—,
Figure US20090062328A1-20090305-C00125
—C(O)—C(O)—,
—C(O)—C(O)—O—, and
—C(═NH)—N(R8)—,
R is selected from the group consisting of:
halogen,
hydroxy,
alkyl,
alkenyl,
haloalkyl,
alkoxy,
alkylthio, and
—N(R9)2;
R1 is selected from the group consisting of:
—R4,
—X′—R4,
—X′—Y′—R4,
—X′—Y′—X′—Y′—R4, and
—X′—R5;
R3 is selected from the group consisting of:
-Z′-R4,
-Z′-X′—R4,
-Z′-X′—Y′—R4,
-Z′-X′—Y′—X′—Y′—R4, and
-Z′-X′—R5;
X′ is selected from the group consisting of alkylene, alkenylene, alkynylene, arylene, heteroarylene, and heterocyclylene, wherein the alkylene, alkenylene, and alkenylene groups can be optionally interrupted or terminated by arylene, heteroarylene or heterocyclylene and optionally interrupted by one or more —O— groups;
Y′ is selected from the group consisting of:
—O—,
—S(O)0-2—,
—S(O)2—N(R8)—,
—C(R6)—,
—C(R6)—O—,
—O—C(R6)—
—O—C(O)—O,
—N(R8)-Q-,
—C(R6)—N(R8)—,
—O—C(R6)—N(R8)—,
—C(R6)—N(OR9)—,
—O—N(R8)-Q-,
—O—N═C(R4)—,
—C(═N—O—R8)—,
—CH(—N(—O—R8)-Q-R4)—,
Figure US20090062328A1-20090305-C00126
Z′ is a bond or —O—;
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, arylalkleneoxy, heteroaryl, heteroaryloxy, heteroarylalkleneoxy, heterocyclyl, amino, alkylamino, dialkylamino, (dialkylamino)alkyleneoxy, and in the case of alkyl, alkenyl, alkynyl, and heterocyclyl, oxo;
R5 is selected from the group consisting of:
Figure US20090062328A1-20090305-C00127
R6 is selected from the group consisting of ═O and ═S;
R7 is C2-7 alkylene;
R8 is selected from the group consisting of hydrogen, C1-10 alkyl, C2-10 alkenyl, C1-10 alkoxy-C1-10 alkylenyl, hydroxy-C1-10 alkylenyl, heteroaryl-C1-10 alkylenyl, and aryl-C1-10 alkylenyl;
R9 is selected from the group consisting of hydrogen and alkyl;
R10 is C3-8 alkylene;
A is selected from the group consisting of —O—, —C(O)—, —S(O)0-2—, —CH2—, and —N(-Q-R4)—,
A′ is selected from the group consisting of —O—, —S(O)0-2—, N(-Q-R4)—, and —CH2—;
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(R6)—O—, —C(R6)—S—, and —C(R6)—N(OR9)—;
V is selected from the group consisting of —C(R6)—, —O—C(R6)—, N(R8)—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.
10. A compound of Formula VIIIa:
Figure US20090062328A1-20090305-C00128
wherein:
Z is selected from the group consisting of:
—C(═N—O—R2-2)— and
—C(R2-4)(—N(—OR2-2)—Y—R2-3)—;
X is selected from the group consisting of a bond, C1-4 alkylene and C2-4 alkenylene;
R2-1, R2-2, and R2-3 are independently selected from the group consisting of:
hydrogen,
alkyl,
alkenyl,
aryl,
arylalkylenyl,
heteroaryl,
heteroarylalkylenyl,
heterocyclyl,
heterocyclylalkylenyl, and
alkyl, alkenyl, aryl, arylalkylenyl, heteroaryl, heteroarylalkylenyl, heterocyclyl, and heterocyclylalkylenyl, substituted by one or more substituents selected from the group consisting of:
hydroxy,
alkyl,
haloalkyl,
hydroxyalkyl,
alkoxy,
dialkylamino,
—S(O)0-2R2-5,
—NH—S(O)2—R2-5,
haloalkoxy,
halogen,
cyano,
nitro,
—N3,
aryl,
heteroaryl,
heterocyclyl,
aryloxy,
arylalkyleneoxy,
—C(O)—O-alkyl,
—C(O)—N(R8)2,
—N(R8)—C(O)—R2-5,
—NH—C(O)—NH—R2-5,
—NH—C(O)—NH2,
—O—(CO)-alkyl, and
—C(O)-alkyl;
with the proviso that R2-2 is other than alkenyl wherein the carbon atom bonded to —O— is doubly bonded to another carbon atom,
R2-4 is selected from the group consisting of hydrogen, C1-4alkyl, and phenyl;
R2-5 is selected from the group consisting of alkyl, aryl, arylalkylenyl, heteroaryl, and heteroarylalkylenyl, each of which is unsubstituted or substituted by one or more substituents selected from the group consisting of halogen, cyano, nitro, alkoxy, dialkylamino, alkylthio, haloalkyl, haloalkoxy, alkyl, and —N3;
Y is selected from the group consisting of:
a bond,
—C(R6)—,
—S(O)2,
—S(O)2—N(R8)—,
Figure US20090062328A1-20090305-C00129
—C(O)—O—,
—C(R6)—N(R8)—,
—C(O)—N(R8)—S(O)2—,
—C(R6)—N(R8)—C(O)—,
Figure US20090062328A1-20090305-C00130
—C(O)—C(O)—,
—C(O)—C(O)—O—, and
—C(═NH)—N(R8)—,
R is selected from the group consisting of:
halogen,
hydroxy,
alkyl,
alkenyl,
haloalkyl,
alkoxy,
alkylthio, and
—N(R9)2;
n is an integer from 0 to 4;
R6 is selected from the group consisting of ═O and ═S;
R8 is selected from the group consisting of hydrogen, C1-10 alkyl, C2-10 alkenyl, C1-10 alkoxy-C1-10 alkylenyl, hydroxy-C1-10alkylenyl, heteroaryl-C1-10alkylenyl, and aryl-C1-10 alkylenyl;
R9 is selected from the group consisting of hydrogen and alkyl;
R10 is C3-8 alkylene;
or a pharmaceutically acceptable salt thereof
11. The compound or salt of claim 4 wherein RA1 and RB1 are each methyl.
12. The compound or salt of claim 5 wherein R is halogen or hydroxy.
13. The compound or salt of claim 5, wherein n is 0.
14. The compound or salt of claim 7 wherein p is 0.
15. The compound or salt of claim 2, wherein R3 is selected from the group consisting of phenyl, pyridin-3-yl, pyridin-4-yl, 5-(hydroxymethyl)pyridin-3-yl, 2-ethoxyphenyl, 3-(morpholine-4-carbonyl)phenyl, and 3-(N,N-dimethylaminocarbonyl)phenyl.
16. The compound or salt of claim 2, wherein R3 is benzyloxy.
17. The compound or salt of claim 5, wherein in is 0.
18. The compound or salt of claim 1 wherein Z is —C(═N—O—R2-2)—.
19-27. (canceled)
28. The compound or salt of claim 1 is a bond or C1-4alkylene.
29. (canceled)
30. The compound or salt of claim 3 wherein R1 is selected from the group consisting of alkyl, arylalkylenyl, aryloxyalkylenyl, hydroxyalkyl, dihydroxyalkyl, alkylsulfonylalkylenyl, —X′—Y′—R4, —X′—R5, and heterocyclylalkylenyl;
wherein the heterocyclyl of the heterocyclylalkylenyl group is optionally substituted by one or more alkyl groups; wherein X′ is alkylene; Y′ is —N(R8)—C(O)—, —N(R8)—S(O)2—, —N(R8)—C(O)—N(R8)—, or
Figure US20090062328A1-20090305-C00131
R4 is alkyl, aryl, or heteroaryl; and R5 is
Figure US20090062328A1-20090305-C00132
31. The compound or salt of claim 30 wherein R1 is selected from the group consisting of 2-hydroxy-2-methylpropyl, 2-methylpropyl, propyl, ethyl, methyl, 2,3-dihydroxypropyl, 2-phenoxyethyl, 4-[(methylsulfonyl)amino]butyl, 2-methyl-2-[(methylsulfonyl)amino]propyl, 2-(acetylamino)-2methylpropyl, 2-{[(isopropylamino)carbonyl]amino}-2-methylpropyl, 4-{[(isopropylamino)carbonyl]amino}butyl, 4-I1,1-dioxidoisothiazolidin-2-yl)butyl, tetrahydro-2H-pyran-4-ylmethyl, and (2,2-dimethyl-1,3-dioxolan-4-yl)methyl.
32. The compound or salt of claim 30 wherein R1 is selected from the group consisting of (1-hydroxycyclobutyl)methyl, (1-hydroxycyclopentyl)methyl, and (1-hydroxycyclohexyl)methyl.
33. A pharmaceutical composition comprising a therapeutically effective amount of a compound or salt of claim 1 and a pharmaceutically acceptable carrier.
34. A method of inducing cytokine biosynthesis in an animal comprising administering an effective amount of a compound or salt of claim 1 to the animal.
35. 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 claim 1 to the animal.
36. 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 claim 1 to the animal.
US11/884,153 2005-02-11 2006-02-10 Oxime and hydroxylamine substituted imidazo[4,5-c] ring compounds and methods Expired - Fee Related US7968563B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/884,153 US7968563B2 (en) 2005-02-11 2006-02-10 Oxime and hydroxylamine substituted imidazo[4,5-c] ring compounds and methods

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US65220905P 2005-02-11 2005-02-11
US11/884,153 US7968563B2 (en) 2005-02-11 2006-02-10 Oxime and hydroxylamine substituted imidazo[4,5-c] ring compounds and methods
PCT/US2006/004737 WO2006086634A2 (en) 2005-02-11 2006-02-10 Oxime and hydroxylamine substituted imidazo[4,5-c] ring compounds and methods

Publications (3)

Publication Number Publication Date
US20090062328A1 true US20090062328A1 (en) 2009-03-05
US20100069427A9 US20100069427A9 (en) 2010-03-18
US7968563B2 US7968563B2 (en) 2011-06-28

Family

ID=36793761

Family Applications (1)

Application Number Title Priority Date Filing Date
US11/884,153 Expired - Fee Related US7968563B2 (en) 2005-02-11 2006-02-10 Oxime and hydroxylamine substituted imidazo[4,5-c] ring compounds and methods

Country Status (6)

Country Link
US (1) US7968563B2 (en)
EP (1) EP1846405A2 (en)
JP (1) JP2008530113A (en)
AU (1) AU2006213746A1 (en)
CA (1) CA2597587A1 (en)
WO (1) WO2006086634A2 (en)

Cited By (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040214851A1 (en) * 2003-04-28 2004-10-28 3M Innovative Properties Company Compositions and methods for induction of opioid receptors
US20070060754A1 (en) * 2003-10-03 2007-03-15 Lindstrom Kyle J Alkoxy substituted imidazoquinolines
US20070066639A1 (en) * 2003-08-12 2007-03-22 Kshirsagar Tushar A Oxime substituted imidazoquinolines
US20070072893A1 (en) * 2003-11-25 2007-03-29 Krepski Larry R Substituted imidazo ring systems and methods
US20070208052A1 (en) * 2004-06-18 2007-09-06 Prince Ryan B Aryloxy and arylalkyleneoxy substituted thiazoloquinolines and thiazolonaphthyridines
US20070213356A1 (en) * 2004-06-15 2007-09-13 Merrill Bryon A Nitrogen-Containing Heterocyclyl Substituted Imidazoquinolines and Imidazonaphthyridines
US20070219196A1 (en) * 2004-03-24 2007-09-20 Krepski Larry R Amide substituted imidazopyridines, imidazoquinolines, and imidazonaphthyridines
US20070219228A1 (en) * 2004-06-18 2007-09-20 Shri Niwas Aryl substituted imidazonaphthyridines
US20070287725A1 (en) * 2004-06-18 2007-12-13 3M Innovative Properties Company Isoxazole, Dihydroisoxazole, And Oxadiazole Substituted Imidazo Ring Compounds And Method
US20080070907A1 (en) * 2006-07-12 2008-03-20 Coley Pharmaceutical Group, Inc. Substituted chiral fused [1,2] imidazo [4,5-C] ring compounds and methods
US20080269192A1 (en) * 2004-12-30 2008-10-30 Coley Pharmaceutical Group, Inc. Chiral Fused [1,2]Imidazo[4,5-C] Ring Compounds
US20080306252A1 (en) * 2000-12-08 2008-12-11 Coley Pharmaceutical Group, Inc. Sulfonamido ether substituted imidazoquinolines
US20080318998A1 (en) * 2005-02-09 2008-12-25 Coley Pharmaceutical Group, Inc. Alkyloxy Substituted Thiazoloquinolines and Thiazolonaphthyridines
US20090018122A1 (en) * 2003-08-27 2009-01-15 Lindstrom Kyle J Aryloxy and Arylalkyleneoxy Substituted Imidazoquinolines
US20090017076A1 (en) * 2003-09-05 2009-01-15 Coley Pharmaceutical Group, Inc. Treatment for cd5+ b cell lymphoma
US20090030030A1 (en) * 2003-12-29 2009-01-29 Bonk Jason D Arylalkenyl and arylalkynyl substituted imidazoquinolines
US20090042925A1 (en) * 2003-11-14 2009-02-12 Coley Pharmaceutical Group, Inc. Oxime substituted imidazoquinolines
US20090075980A1 (en) * 2003-10-03 2009-03-19 Coley Pharmaceutical Group, Inc. Pyrazolopyridines and Analogs Thereof
US20090105295A1 (en) * 2003-11-14 2009-04-23 Coley Pharmaceutical Group, Inc. Hydroxylamine substituted imidazoquinolines
US20090163532A1 (en) * 2005-02-04 2009-06-25 Coley Pharmaceutical Group, Inc. Aqueous Gel Formulations Containing Immune Response Modifiers
US20090221556A1 (en) * 2005-11-04 2009-09-03 Pfizer Inc. Hydroxy and alkoxy substituted 1h-imidazoquinolines and methods
US20090270443A1 (en) * 2004-09-02 2009-10-29 Doris Stoermer 1-amino imidazo-containing compounds and methods
US20090318435A1 (en) * 2003-10-03 2009-12-24 Hays David S Pyrazolopyridines and analogs thereof
US20110070575A1 (en) * 2004-12-08 2011-03-24 Coley Pharmaceutical Group, Inc. Immunomodulatory Compositions, Combinations 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
US8034938B2 (en) 2004-12-30 2011-10-11 3M Innovative Properties Company Substituted chiral fused [1,2]imidazo[4,5-c] ring compounds
US8735421B2 (en) 2003-12-30 2014-05-27 3M Innovative Properties Company Imidazoquinolinyl sulfonamides
US8883174B2 (en) 2009-03-25 2014-11-11 The Board Of Regents, The University Of Texas System Compositions for stimulation of mammalian innate immune resistance to pathogens
US10286065B2 (en) 2014-09-19 2019-05-14 Board Of Regents, The University Of Texas System Compositions and methods for treating viral infections through stimulated innate immunity in combination with antiviral compounds

Families Citing this family (40)

* 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
KR20060120069A (en) 2003-10-03 2006-11-24 쓰리엠 이노베이티브 프로퍼티즈 컴파니 Pyrazolopyridines and analogs thereof
US8541438B2 (en) 2004-06-18 2013-09-24 3M Innovative Properties Company Substituted imidazoquinolines, imidazopyridines, and imidazonaphthyridines
EP1877056A2 (en) 2005-02-09 2008-01-16 Coley Pharmaceutical Group, Inc. Oxime and hydroxylamine substituted thiazoloý4,5-c¨ring compounds and methods
WO2006086634A2 (en) 2005-02-11 2006-08-17 Coley Pharmaceutical Group, Inc. Oxime and hydroxylamine substituted imidazo[4,5-c] ring compounds and methods
US8658666B2 (en) 2005-02-11 2014-02-25 3M Innovative Properties Company Substituted imidazoquinolines and imidazonaphthyridines
CA2598437A1 (en) 2005-02-23 2006-08-31 Coley Pharmaceutical Group, Inc. Method of preferentially inducing the biosynthesis of interferon
US8158794B2 (en) 2005-02-23 2012-04-17 3M Innovative Properties Company Hydroxyalkyl substituted imidazoquinoline compounds and methods
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
US8343993B2 (en) 2005-02-23 2013-01-01 3M Innovative Properties Company Hydroxyalkyl substituted imidazonaphthyridines
CA2621831A1 (en) * 2005-09-09 2007-03-15 Coley Pharmaceutical Group, Inc. 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
WO2007100634A2 (en) 2006-02-22 2007-09-07 3M Innovative Properties Company Immune response modifier conjugates
WO2007106854A2 (en) 2006-03-15 2007-09-20 Coley Pharmaceutical Group, Inc. Hydroxy and alkoxy substituted 1h-imidazonaphthyridines and methods
WO2008030511A2 (en) 2006-09-06 2008-03-13 Coley Pharmaceuticial Group, Inc. Substituted 3,4,6,7-tetrahydro-5h, 1,2a,4a,8-tetraazacyclopenta[cd]phenalenes
US20080149123A1 (en) 2006-12-22 2008-06-26 Mckay William D Particulate material dispensing hairbrush with combination bristles
CN101896474B (en) * 2007-10-09 2016-02-03 希凯迪斯股份有限公司 Prepare the method for ketal and acetal
BR112013000575A2 (en) 2010-08-03 2016-07-05 Segetis Inc methods for making acetals and ketals, and acetals and ketals produced hereby
CN103097386A (en) 2010-08-17 2013-05-08 3M创新有限公司 Lipidated immune response modifier compound compositions, formulations, and methods
US8728486B2 (en) 2011-05-18 2014-05-20 University Of Kansas Toll-like receptor-7 and -8 modulatory 1H imidazoquinoline derived compounds
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
EP2717919B1 (en) 2011-06-03 2016-08-03 3M Innovative Properties Company Heterobifunctional linkers with polyethylene glycol segments and immune response modifier conjugates made therefrom
DK3181567T3 (en) 2012-09-10 2019-06-11 Principia Biopharma Inc Pyrazolopyridamide compounds as kinase inhibitors
RS61101B1 (en) 2013-01-07 2020-12-31 Univ Pennsylvania Compositions and methods for treating cutaneous t cell lymphoma
SG11201606858RA (en) 2014-02-21 2016-09-29 Principia Biopharma Inc Salts and solid form of a btk inhibitor
ES2843323T3 (en) 2014-12-18 2021-07-16 Principia Biopharma Inc Pemphigus treatment
CR20170593A (en) 2015-06-03 2018-05-14 Principia Biopharma Inc THYROSINE-KINASE INHIBITORS
EP3313839A1 (en) 2015-06-24 2018-05-02 Principia Biopharma Inc. Tyrosine kinase inhibitors
WO2017040233A1 (en) 2015-08-31 2017-03-09 3M Innovative Properties Company GUANIDINE SUBSTITUTED IMIDAZO[4,5-c] RING COMPOUNDS
WO2017040234A1 (en) 2015-08-31 2017-03-09 3M Innovative Properties Company IMIDAZO[4,5-c] RING COMPOUNDS CONTAINING SUBSTITUTED GUANIDINE GROUPS
WO2017059280A1 (en) 2015-10-02 2017-04-06 The University Of North Carolina At Chapel Hill Novel pan-tam inhibitors and mer/axl dual inhibitors
CA3028169A1 (en) 2016-06-29 2018-01-04 Principia Biopharma Inc. Modified release formulations of 2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile
WO2018160552A1 (en) 2017-03-01 2018-09-07 3M Innovative Properties Company IMIDAZO[4,5-c] RING COMPOUNDS CONTAINING GUANIDINE SUBSTITUTED BENZAMIDE GROUPS
CN111511740B (en) 2017-12-20 2023-05-16 3M创新有限公司 Amide substituted imidazo [4,5-C ] quinoline compounds with branched linking groups for use as immune response modifiers
MX2020008455A (en) 2018-02-28 2021-10-26 Pfizer Il-15 variants and uses thereof.
SG11202010934SA (en) 2018-05-23 2020-12-30 Pfizer Antibodies specific for gucy2c and uses thereof
KR102602329B1 (en) 2018-05-23 2023-11-16 화이자 인코포레이티드 Antibodies specific for CD3 and their uses
WO2020128893A1 (en) 2018-12-21 2020-06-25 Pfizer Inc. Combination treatments of cancer comprising a tlr agonist
US11702474B2 (en) 2019-12-17 2023-07-18 Pfizer Inc. Antibodies specific for CD47, PD-L1, and uses thereof
BR112023000701A2 (en) 2020-07-17 2023-02-07 Pfizer THERAPEUTIC ANTIBODIES AND THEIR USES

Family Cites Families (315)

* 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
IL73534A (en) 1983-11-18 1990-12-23 Riker Laboratories Inc 1h-imidazo(4,5-c)quinoline-4-amines,their preparation and pharmaceutical compositions containing certain such compounds
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
CA1271477A (en) 1983-11-18 1990-07-10 John F. Gerster 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
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
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
ES2094141T3 (en) 1989-02-27 1997-01-16 Riker Laboratories Inc 1H-IMIDAZO (4,5-C) QUINOLIN-4-AMINAS AS ANTIVIRALS.
US5756747A (en) 1989-02-27 1998-05-26 Riker Laboratories, Inc. 1H-imidazo 4,5-c!quinolin-4-amines
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
US5750134A (en) 1989-11-03 1998-05-12 Riker Laboratories, Inc. Bioadhesive composition and patch
AU658134B2 (en) 1989-12-28 1995-04-06 Virginia Commonwealth University Sigma receptor ligands and the use thereof
US5274113A (en) 1991-11-01 1993-12-28 Molecular Probes, Inc. Long wavelength chemically reactive dipyrrometheneboron difluoride dyes and conjugates
HUT65614A (en) 1990-04-30 1994-07-28 Isis Pharmaceuticals Inc Process for the oligonucleotide modulation of arachidonic acid metabolism
EP0461040A1 (en) 1990-06-08 1991-12-11 Roussel Uclaf Imidazol derivatives, their process for production, intermediates, their application 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
ATE121088T1 (en) 1990-10-05 1995-04-15 Minnesota Mining & Mfg METHOD FOR PRODUCING IMIDAZO(4,5- C>QUINOLINE-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
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
WO1993023011A1 (en) 1992-05-18 1993-11-25 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
JPH0755908B2 (en) 1992-11-24 1995-06-14 財団法人京都パストゥール研究所 Immune function promoter
US5395937A (en) 1993-01-29 1995-03-07 Minnesota Mining And Manufacturing Company Process for preparing quinoline amines
DE69413955T2 (en) 1993-03-17 1999-04-01 Minnesota Mining & Mfg AEROSOL COMPOSITION CONTAINING A DERIVATIVE DERIVATIVE FROM ESTER, AMID OR MERCAPTOESTER
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
US5352784A (en) 1993-07-15 1994-10-04 Minnesota Mining And Manufacturing Company Fused cycloalkylimidazopyridines
JPH09500128A (en) 1993-07-15 1997-01-07 ミネソタ マイニング アンド マニュファクチャリング カンパニー Imidazo [4,5-c] pyridin-4-amine
US5648516A (en) 1994-07-20 1997-07-15 Minnesota Mining And Manufacturing Company Fused cycloalkylimidazopyridines
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
TW530047B (en) 1994-06-08 2003-05-01 Pfizer Corticotropin releasing factor antagonists
CA2560114A1 (en) 1994-07-15 1996-02-01 The University Of Iowa Research Foundation Immunomodulatory oligonucleotides
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
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
ATE243697T1 (en) 1995-12-08 2003-07-15 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
EP0882727B9 (en) 1996-07-03 2005-06-15 Sumitomo Pharmaceuticals Company, Limited Novel purine derivatives
US6387938B1 (en) 1996-07-05 2002-05-14 Mochida Pharmaceutical Co., Ltd. Benzimidazole derivatives
JP4391592B2 (en) 1996-10-25 2009-12-24 スリーエム カンパニー Immune response modifying compounds for the treatment of 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
US6113918A (en) 1997-05-08 2000-09-05 Ribi Immunochem Research, Inc. Aminoalkyl glucosamine phosphate compounds and their use as adjuvants and immunoeffectors
US6303347B1 (en) 1997-05-08 2001-10-16 Corixa Corporation Aminoalkyl glucosaminide phosphate compounds and their use as adjuvants and immunoeffectors
CA2301575C (en) 1997-05-20 2003-12-23 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
US6077349A (en) 1997-08-20 2000-06-20 Sony Corporation Method and apparatus 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
ES2205573T3 (en) 1997-11-28 2004-05-01 Sumitomo Pharmaceuticals Company, Limited NEW HETEROCICLIC COMPOUNDS.
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
JP2000119271A (en) 1998-08-12 2000-04-25 Hokuriku Seiyaku Co Ltd 1h-imidazopyridine derivative
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
CA2361936C (en) 1999-01-08 2009-06-16 3M Innovative Properties Company Formulations comprising imiquimod or other immune response modifiers for treating mucosal conditions
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
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
DE60039290D1 (en) 1999-04-12 2008-08-07 Lexicon Pharmaceuticals Inc LIPOXYGENASE PROTEINS AND POLYNUCLEOTIDES THAT CODE
US6756382B2 (en) 1999-06-10 2004-06-29 3M Innovative Properties Company Amide substituted imidazoquinolines
US6451810B1 (en) 1999-06-10 2002-09-17 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
US6331539B1 (en) 1999-06-10 2001-12-18 3M Innovative Properties Company Sulfonamide and sulfamide 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
IL139197A0 (en) 1999-10-29 2001-11-25 Pfizer Prod Inc Use of corticotropin releasing factor antagonists and related compositions
IL149085A0 (en) 1999-10-29 2002-11-10 Inhale Therapeutic Syst A dry powder composition containing a di or tripeptide
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
PL355866A1 (en) 2000-02-09 2004-05-31 Hokuriku Seiyaku Co, Ltd. 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
US6894060B2 (en) 2000-03-30 2005-05-17 3M Innovative Properties Company Method for the treatment of dermal lesions caused by envenomation
IL151877A0 (en) 2000-03-30 2003-04-10 Shionogi & Co A process for producing imidazopyridine derivatives and a crystal form of an imidazopyridine derivative
US20020137101A1 (en) 2000-05-19 2002-09-26 Meyers Rachel A. 46638, a novel human lipoxygenase family member and uses thereof
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
WO2002022809A2 (en) 2000-09-15 2002-03-21 Coley Pharmaceutical Gmbh PROCESS FOR HIGH THROUGHPUT SCREENING OF CpG-BASED IMMUNO-AGONIST/ANTAGONIST
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
WO2002046479A1 (en) 2000-12-07 2002-06-13 Aoyama Seisakusho Co., Ltd. Method for baking steel part
UA75622C2 (en) 2000-12-08 2006-05-15 3M Innovative Properties Co Aryl ether substituted imidazoquinolines, pharmaceutical composition based thereon
US6545017B1 (en) 2000-12-08 2003-04-08 3M Innovative Properties Company Urea substituted imidazopyridines
US6667312B2 (en) 2000-12-08 2003-12-23 3M Innovative Properties Company Thioether substituted imidazoquinolines
US6664260B2 (en) 2000-12-08 2003-12-16 3M Innovative Properties Company Heterocyclic ether substituted imidazoquinolines
US6664264B2 (en) 2000-12-08 2003-12-16 3M Innovative Properties Company Thioether substituted imidazoquinolines
US6545016B1 (en) 2000-12-08 2003-04-08 3M Innovative Properties Company Amide substituted imidazopyridines
US6664265B2 (en) 2000-12-08 2003-12-16 3M Innovative Properties Company Amido ether substituted imidazoquinolines
CA2430206A1 (en) 2000-12-08 2002-06-13 3M Innovative Properties Company Screening method for identifying compounds that selectively induce interferon alpha
US6677348B2 (en) 2000-12-08 2004-01-13 3M Innovative Properties Company Aryl ether substituted imidazoquinolines
US6525064B1 (en) 2000-12-08 2003-02-25 3M Innovative Properties Company Sulfonamido substituted imidazopyridines
US6660735B2 (en) 2000-12-08 2003-12-09 3M Innovative Properties Company Urea substituted imidazoquinoline ethers
UA74593C2 (en) 2000-12-08 2006-01-16 3M Innovative Properties Co Substituted imidazopyridines
US6660747B2 (en) 2000-12-08 2003-12-09 3M Innovative Properties Company Amido ether substituted imidazoquinolines
US6677347B2 (en) 2000-12-08 2004-01-13 3M Innovative Properties Company Sulfonamido ether substituted imidazoquinolines
US20020107262A1 (en) 2000-12-08 2002-08-08 3M Innovative Properties Company Substituted imidazopyridines
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
JP4331944B2 (en) 2001-04-17 2009-09-16 大日本住友製薬株式会社 New adenine derivatives
US20020193729A1 (en) 2001-04-20 2002-12-19 Cormier Michel J.N. Microprojection array immunization patch and method
DE60141773D1 (en) 2001-04-20 2010-05-20 Inst Systems Biology GREAT SIMILAR RECEPTOR 5-LIGANDS AND USE PROCESS
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
KR20040027878A (en) 2001-07-16 2004-04-01 시오노기 앤드 컴파니, 리미티드 Process for preparation of amidine derivatives
US20030133913A1 (en) 2001-08-30 2003-07-17 3M Innovative Properties Company Methods of maturing plasmacytoid dendritic cells using immune response modifier molecules
JP4445262B2 (en) 2001-10-09 2010-04-07 アムジェン インコーポレイテッド Imidazole derivatives as anti-inflammatory agents
JP2005519990A (en) 2001-10-12 2005-07-07 ユニバーシティ オブ アイオワ リサーチ ファウンデーション Methods and products for enhancing immune responses using imidazoquinoline compounds
ES2318615T3 (en) 2001-11-16 2009-05-01 Coley Pharmaceutical Group, Inc. N- (4- (4-AMINO-2-ETIL-1H-IMIDAZO (4,5-C) QUINOLIN-1-IL) BUTIL) METHANOSULPHONAMIDE, A PHARMACEUTICAL COMPOSITION THAT UNDERSTANDS AND USES ITSELF.
EP1455789A4 (en) 2001-11-17 2008-02-13 Martinez Colon Maria Imiquimod therapies
YU45204A (en) 2001-11-27 2006-08-17 Anadys Pharmaceuticals Inc. 3-betha-d-ribofuranosylthiazolo/4,5-d/ pyrimidine nucleosides and uses thereof
DK1450804T3 (en) 2001-11-29 2009-01-05 3M Innovative Properties Co Pharmaceutical formula rings comprising an immune response modifier
WO2004080430A2 (en) 2003-03-13 2004-09-23 3M Innovative Properties Company Methods of improving skin quality
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
SI1478327T1 (en) 2002-02-22 2015-08-31 Meda Ab Method of reducing and treating uvb-induced immunosuppression
US20030185835A1 (en) 2002-03-19 2003-10-02 Braun Ralph P. Adjuvant for vaccines
ES2543710T3 (en) 2002-04-04 2015-08-21 Zoetis Belgium S.A. Immunostimulatory oligonucleotides containing G and U
JP4723239B2 (en) 2002-04-30 2011-07-13 ユニジェン・インコーポレーテッド Formulation of a mixture of free-B-ring flavonoids and flavans as therapeutic agents
GB0211649D0 (en) 2002-05-21 2002-07-03 Novartis Ag Organic compounds
EP1511746A2 (en) 2002-05-29 2005-03-09 3M Innovative Properties Company Process for imidazo[4,5-c]pyridin-4-amines
EP1513524A4 (en) 2002-06-07 2008-09-03 3M Innovative Properties Co Ether substituted imidazopyridines
CA2493227A1 (en) 2002-07-23 2004-01-29 Biogal Gyogyszergyar Rt. Preparation of 1h-imidazo[4,5-c]quinolin-4-amines via1h-imidazo [4,5-c]quinolin-4-phthalimide intermediates
AU2003254221A1 (en) 2002-07-26 2004-02-16 Biogal Gyogyszergyar Rt. 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
DE60335010D1 (en) 2002-08-15 2010-12-30 3M Innovative Properties Co IMMUNOSTIMULATORY COMPOSITIONS AND METHOD FOR STIMULATING AN IMMUNE RESPONSE
US6818650B2 (en) 2002-09-26 2004-11-16 3M Innovative Properties Company 1H-imidazo dimers
AU2003270701B2 (en) 2002-10-31 2009-11-12 Amgen Inc. Antiinflammation agents
WO2004043913A2 (en) 2002-11-08 2004-05-27 Trimeris, Inc. Hetero-substituted benzimidazole compounds and antiviral uses thereof
WO2004053452A2 (en) 2002-12-11 2004-06-24 3M Innovative Properties Company Assays relating to toll-like receptor activity
WO2004053057A2 (en) 2002-12-11 2004-06-24 3M Innovative Properties Company Gene expression systems and recombinant cell lines
AU2003301052A1 (en) 2002-12-20 2004-07-22 3M Innovative Properties Company Aryl / hetaryl substituted imidazoquinolines
WO2004060319A2 (en) 2002-12-30 2004-07-22 3M Innovative Properties Company Immunostimulatory combinations
WO2004071459A2 (en) 2003-02-13 2004-08-26 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
US8110582B2 (en) 2003-03-04 2012-02-07 3M Innovative Properties Company Prophylactic treatment of UV-induced epidermal neoplasia
TW200505458A (en) 2003-03-07 2005-02-16 3M Innovative Properties Co 1-amino 1h-imidazoquinolines
US7163947B2 (en) 2003-03-07 2007-01-16 3M Innovative Properties Company 1-Amino 1H-imidazoquinolines
AU2004220465A1 (en) 2003-03-13 2004-09-23 3M Innovative Properties Company Method of tattoo removal
WO2004080293A2 (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
US20040191833A1 (en) 2003-03-25 2004-09-30 3M Innovative Properties Company Selective activation of cellular activities mediated through a common toll-like receptor
US7893096B2 (en) 2003-03-28 2011-02-22 Novartis Vaccines And Diagnostics, Inc. Use of small molecule compounds for immunopotentiation
EP1617871A4 (en) 2003-04-10 2010-10-06 3M Innovative Properties Co Delivery of immune response modifier compounds using metal-containing particulate support materials
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
AR044466A1 (en) 2003-06-06 2005-09-14 3M Innovative Properties Co PROCESS FOR THE PREPARATION OF IMIDAZO [4,5-C] PIRIDIN-4-AMINAS
US6943255B2 (en) 2003-06-06 2005-09-13 3M Innovative Properties Company Process for imidazo[4,5-c]pyridin-4-amines
KR20060016817A (en) 2003-06-20 2006-02-22 콜리 파마슈티칼 게엠베하 Small molecule toll-like receptor (tlr) 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
EP1651216A2 (en) 2003-08-05 2006-05-03 3M Innovative Properties Company Infection prophylaxis using immune response modifier compounds
EP1653914A4 (en) 2003-08-12 2008-10-29 3M Innovative Properties Co Oxime substituted imidazo-containing compounds
PL1653959T3 (en) 2003-08-14 2015-10-30 3M Innovative Properties Co Lipid-modified immune response modifiers
CA2551075A1 (en) 2003-08-25 2005-03-03 3M Innovative Properties Company Immunostimulatory combinations and treatments
US7897597B2 (en) 2003-08-27 2011-03-01 3M Innovative Properties Company Aryloxy and arylalkyleneoxy substituted imidazoquinolines
WO2005023190A2 (en) 2003-09-05 2005-03-17 3M Innovative Properties Company Treatment for cd5+ b cell lymphoma
EP1667694B1 (en) 2003-09-05 2010-04-28 Anadys Pharmaceuticals, Inc. Tlr7 ligands for the treatment of hepatitis c
GB0321615D0 (en) 2003-09-15 2003-10-15 Glaxo Group Ltd Improvements in vaccination
WO2005029037A2 (en) 2003-09-17 2005-03-31 3M Innovative Properties Company Selective modulation of tlr gene expression
CA2536530A1 (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
KR20060120069A (en) 2003-10-03 2006-11-24 쓰리엠 이노베이티브 프로퍼티즈 컴파니 Pyrazolopyridines and analogs thereof
BRPI0414856A (en) 2003-10-03 2006-11-21 3M Innovative Properties Co 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
US20050239733A1 (en) 2003-10-31 2005-10-27 Coley Pharmaceutical Gmbh Sequence requirements for inhibitory oligonucleotides
JP2007509987A (en) 2003-10-31 2007-04-19 スリーエム イノベイティブ プロパティズ カンパニー Neutrophil activation by immune response modulator compounds
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
AU2004291101A1 (en) 2003-11-14 2005-06-02 3M Innovative Properties Company Oxime substituted imidazo ring compounds
EP1682544A4 (en) 2003-11-14 2009-05-06 3M Innovative Properties Co Hydroxylamine substituted imidazo ring compounds
CU23404A1 (en) 2003-11-19 2009-08-04 Ct Ingenieria Genetica Biotech NEISSERIA MENINGITIDIS CAPSULAR POLYSACARIDS AS MUCOSOPT IMMUNOPOTENTIZERS AND RESULTING FORMULATIONS
WO2005054237A1 (en) 2003-11-21 2005-06-16 Novartis Ag 1h-imidazoquinoline derivatives as protein kinase inhibitors
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
US8778963B2 (en) 2003-11-25 2014-07-15 3M Innovative Properties Company Hydroxylamine and oxime substituted imidazoquinolines, imidazopyridines, and imidazonaphthyridines
BRPI0416936A (en) * 2003-11-25 2007-01-16 3M Innovative Properties Co substituted imidazo ring systems and methods
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
TW200533352A (en) 2003-12-04 2005-10-16 3M Innovative Properties Co Sulfone substituted imidazo ring ethers
EP1699792A1 (en) 2003-12-29 2006-09-13 3M Innovative Properties Company Piperazine, 1,4¨diazepane, 1,4¨diazocane, and 1,5¨diazocane fused imidazo ring compounds
US8802853B2 (en) 2003-12-29 2014-08-12 3M Innovative Properties Company Arylalkenyl and arylalkynyl substituted imidazoquinolines
AU2004312508A1 (en) 2003-12-30 2005-07-21 3M Innovative Properties Company Imidazoquinolinyl, imidazopyridinyl, and imidazonaphthyridinyl sulfonamides
EP1699398A4 (en) 2003-12-30 2007-10-17 3M Innovative Properties Co Enhancement of immune responses
WO2005065678A1 (en) 2003-12-30 2005-07-21 3M Innovative Properties Company Immunomodulatory combinations
TW200612932A (en) 2004-03-24 2006-05-01 3M Innovative Properties Co Amide substituted imidazopyridines, imidazoquinolines, and imidazonaphthyridines
AU2005244260B2 (en) 2004-04-09 2010-08-05 3M Innovative Properties Company Methods, compositions, 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
WO2006038923A2 (en) 2004-06-18 2006-04-13 3M Innovative Properties Company Aryl substituted imidazonaphthyridines
US8026366B2 (en) 2004-06-18 2011-09-27 3M Innovative Properties Company Aryloxy and arylalkyleneoxy substituted thiazoloquinolines and thiazolonaphthyridines
WO2006065280A2 (en) 2004-06-18 2006-06-22 3M Innovative Properties Company Isoxazole, dihydroisoxazole, and oxadiazole substituted imidazo ring compounds and methods
EP1765348B1 (en) 2004-06-18 2016-08-03 3M Innovative Properties Company Substituted imidazoquinolines, imidazopyridines, and imidazonaphthyridines
WO2006093514A2 (en) 2004-06-18 2006-09-08 3M Innovative Properties Company Aryl and arylalkylenyl substituted thiazoloquinolines and thiazolonaphthyridines
WO2006009832A1 (en) 2004-06-18 2006-01-26 3M Innovative Properties Company Substituted imidazo ring systems and methods
WO2006026760A2 (en) 2004-09-02 2006-03-09 3M Innovative Properties Company 1-amino imidazo-containing compounds and methods
JP2008511683A (en) 2004-09-02 2008-04-17 スリーエム イノベイティブ プロパティズ カンパニー 2-Amino 1H-imidazo ring structure and method
JP5209312B2 (en) 2004-09-02 2013-06-12 スリーエム イノベイティブ プロパティズ カンパニー 1-alkoxy 1H-imidazo ring systems and methods
WO2006028451A1 (en) 2004-09-03 2006-03-16 3M Innovative Properties Company 1-amino 1-h-imidazoquinolines
CA2580343A1 (en) 2004-09-14 2006-03-23 Novartis Vaccines And Diagnostics, Inc. Imidazoquinoline compounds
WO2006063072A2 (en) 2004-12-08 2006-06-15 3M Innovative Properties Company Immunomodulatory compositions, combinations and methods
EP1819226A4 (en) 2004-12-08 2010-12-29 3M Innovative Properties Co Immunostimulatory combinations and methods
WO2006074003A2 (en) 2004-12-30 2006-07-13 3M Innovative Properties Company CHIRAL FUSED [1,2]IMIDAZO[4,5-c] RING COMPOUNDS
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
CA2592573A1 (en) 2004-12-30 2006-07-13 3M Innovative Properties Company Multi-route administration of immune response modifier compounds
CA2597092A1 (en) 2005-02-04 2006-08-10 Coley Pharmaceutical Group, Inc. Aqueous gel formulations containing immune reponse modifiers
EP1877056A2 (en) 2005-02-09 2008-01-16 Coley Pharmaceutical Group, Inc. Oxime and hydroxylamine substituted thiazoloý4,5-c¨ring compounds and methods
AU2006212765B2 (en) 2005-02-09 2012-02-02 3M Innovative Properties Company Alkyloxy substituted thiazoloquinolines and thiazolonaphthyridines
US8658666B2 (en) 2005-02-11 2014-02-25 3M Innovative Properties Company Substituted imidazoquinolines and imidazonaphthyridines
WO2006086634A2 (en) 2005-02-11 2006-08-17 Coley Pharmaceutical Group, Inc. Oxime and hydroxylamine substituted imidazo[4,5-c] ring compounds and methods
US8093390B2 (en) 2005-02-11 2012-01-10 3M Innovative Properties Company Substituted fused [1,2]imidazo[4,5-C] ring compounds and methods
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
US8343993B2 (en) 2005-02-23 2013-01-01 3M Innovative Properties Company Hydroxyalkyl substituted imidazonaphthyridines
US8158794B2 (en) 2005-02-23 2012-04-17 3M Innovative Properties Company Hydroxyalkyl substituted imidazoquinoline compounds and methods
CA2598437A1 (en) 2005-02-23 2006-08-31 Coley Pharmaceutical Group, Inc. Method of preferentially inducing the biosynthesis of interferon
CA2602853A1 (en) 2005-04-01 2006-11-16 Coley Pharmaceutical Group, Inc. Ring closing and related methods and intermediates
US7943610B2 (en) 2005-04-01 2011-05-17 3M Innovative Properties Company Pyrazolopyridine-1,4-diamines and analogs thereof
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
EP1863814A1 (en) 2005-04-01 2007-12-12 Coley Pharmaceutical Group, Inc. 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
EP1924581A1 (en) 2005-09-02 2008-05-28 Pfizer, Inc. Hydroxy substituted 1h-imidazopyridines 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
CA2621831A1 (en) 2005-09-09 2007-03-15 Coley Pharmaceutical Group, Inc. 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
EP1937683A4 (en) 2005-09-23 2010-08-25 Coley Pharm Group Inc METHOD FOR 1H-IMIDAZOÝ4,5-c¨PYRIDINES AND ANALOGS THEREOF
ES2429170T3 (en) 2005-11-04 2013-11-13 3M Innovative Properties Company 1H-Imidazoquinolines substituted with hydroxyl and alkoxy and methods
WO2007062043A1 (en) 2005-11-23 2007-05-31 Coley Pharmaceutical Group Inc. Method of activating murine toll-like receptor 8
AU2006332000A1 (en) 2005-12-16 2007-07-05 Coley Pharmaceutical Group, Inc. Substituted imidazoquinolines, imidazonaphthyridines, and imidazopyridines, compositions, and methods
JP2009522296A (en) 2005-12-28 2009-06-11 スリーエム イノベイティブ プロパティズ カンパニー Treatment of cutaneous T-cell lymphoma
WO2007079202A2 (en) 2005-12-28 2007-07-12 Coley Pharmaceutical Group, Inc. Treatment for acute lymhoblastic leukemia
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
WO2007079086A1 (en) 2005-12-28 2007-07-12 Coley Pharmaceutical Group, Inc. Pyrazoloalkyl substituted imidazo ring compounds and methods
WO2007079171A2 (en) 2005-12-28 2007-07-12 Coley Pharmaceutical Group, Inc. Treatment for hodgkin's lymphoma
WO2007092641A2 (en) 2006-02-10 2007-08-16 Coley Pharmaceutical Group, Inc. Method for substituted 1h-imidazo[4,5-c]pyridines
WO2007106854A2 (en) 2006-03-15 2007-09-20 Coley Pharmaceutical Group, Inc. Hydroxy and alkoxy substituted 1h-imidazonaphthyridines and methods
WO2007106852A2 (en) 2006-03-15 2007-09-20 Coley Pharmaceutical Group, Inc. Substituted fused[1,2]imidazo[4,5-c] ring compounds and methods
WO2007143526A2 (en) 2006-06-05 2007-12-13 Coley Pharmaceutical Group, Inc. Substituted tetrahydroimidazonaphthyridines and methods
CN101511183A (en) 2006-06-27 2009-08-19 先灵-普劳健康护理产品公司 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
WO2008030511A2 (en) 2006-09-06 2008-03-13 Coley Pharmaceuticial Group, Inc. Substituted 3,4,6,7-tetrahydro-5h, 1,2a,4a,8-tetraazacyclopenta[cd]phenalenes
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

Cited By (56)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080306252A1 (en) * 2000-12-08 2008-12-11 Coley Pharmaceutical Group, Inc. Sulfonamido ether substituted imidazoquinolines
US20040214851A1 (en) * 2003-04-28 2004-10-28 3M Innovative Properties Company Compositions and methods for induction of opioid receptors
US8673932B2 (en) * 2003-08-12 2014-03-18 3M Innovative Properties Company Oxime substituted imidazo-containing compounds
US20070066639A1 (en) * 2003-08-12 2007-03-22 Kshirsagar Tushar A Oxime substituted imidazoquinolines
US7897597B2 (en) 2003-08-27 2011-03-01 3M Innovative Properties Company Aryloxy and arylalkyleneoxy substituted imidazoquinolines
US8263594B2 (en) 2003-08-27 2012-09-11 3M Innovative Properties Company Aryloxy and arylalkyleneoxy substituted imidazoquinolines
US20110144099A1 (en) * 2003-08-27 2011-06-16 3M Innovative Properties Company Aryloxy and arylalkyleneoxy substituted imidazoquinolines
US20090018122A1 (en) * 2003-08-27 2009-01-15 Lindstrom Kyle J Aryloxy and Arylalkyleneoxy Substituted Imidazoquinolines
US7923429B2 (en) 2003-09-05 2011-04-12 3M Innovative Properties Company Treatment for CD5+ B cell lymphoma
US20090017076A1 (en) * 2003-09-05 2009-01-15 Coley Pharmaceutical Group, Inc. Treatment for cd5+ b cell lymphoma
US8871782B2 (en) 2003-10-03 2014-10-28 3M Innovative Properties Company Alkoxy substituted imidazoquinolines
US20090075980A1 (en) * 2003-10-03 2009-03-19 Coley Pharmaceutical Group, Inc. Pyrazolopyridines and Analogs Thereof
US7879849B2 (en) 2003-10-03 2011-02-01 3M Innovative Properties Company Pyrazolopyridines and analogs thereof
US20070060754A1 (en) * 2003-10-03 2007-03-15 Lindstrom Kyle J Alkoxy substituted imidazoquinolines
US20090318435A1 (en) * 2003-10-03 2009-12-24 Hays David S 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
US20090105295A1 (en) * 2003-11-14 2009-04-23 Coley Pharmaceutical Group, Inc. Hydroxylamine substituted imidazoquinolines
US20090042925A1 (en) * 2003-11-14 2009-02-12 Coley Pharmaceutical Group, Inc. Oxime substituted imidazoquinolines
US20070072893A1 (en) * 2003-11-25 2007-03-29 Krepski Larry R Substituted imidazo ring systems and methods
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
US20090030030A1 (en) * 2003-12-29 2009-01-29 Bonk Jason D Arylalkenyl and arylalkynyl substituted imidazoquinolines
US8735421B2 (en) 2003-12-30 2014-05-27 3M Innovative Properties Company Imidazoquinolinyl sulfonamides
US8697873B2 (en) 2004-03-24 2014-04-15 3M Innovative Properties Company Amide substituted imidazopyridines, imidazoquinolines, and imidazonaphthyridines
US20070219196A1 (en) * 2004-03-24 2007-09-20 Krepski Larry R Amide substituted imidazopyridines, imidazoquinolines, and imidazonaphthyridines
US20070213356A1 (en) * 2004-06-15 2007-09-13 Merrill Bryon A Nitrogen-Containing Heterocyclyl Substituted Imidazoquinolines and Imidazonaphthyridines
US8017779B2 (en) 2004-06-15 2011-09-13 3M Innovative Properties Company Nitrogen containing heterocyclyl substituted imidazoquinolines and imidazonaphthyridines
US20070219228A1 (en) * 2004-06-18 2007-09-20 Shri Niwas Aryl substituted imidazonaphthyridines
US7915281B2 (en) 2004-06-18 2011-03-29 3M Innovative Properties Company Isoxazole, dihydroisoxazole, and oxadiazole substituted imidazo ring compounds and method
US20070287725A1 (en) * 2004-06-18 2007-12-13 3M Innovative Properties Company Isoxazole, Dihydroisoxazole, And Oxadiazole Substituted Imidazo Ring Compounds And Method
US20070208052A1 (en) * 2004-06-18 2007-09-06 Prince Ryan B Aryloxy and arylalkyleneoxy substituted thiazoloquinolines and thiazolonaphthyridines
US7897609B2 (en) * 2004-06-18 2011-03-01 3M Innovative Properties Company Aryl substituted imidazonaphthyridines
US8026366B2 (en) 2004-06-18 2011-09-27 3M Innovative Properties Company Aryloxy and arylalkyleneoxy substituted thiazoloquinolines and thiazolonaphthyridines
US20090270443A1 (en) * 2004-09-02 2009-10-29 Doris Stoermer 1-amino imidazo-containing compounds and methods
US20110070575A1 (en) * 2004-12-08 2011-03-24 Coley Pharmaceutical Group, Inc. Immunomodulatory Compositions, Combinations and Methods
US7943609B2 (en) 2004-12-30 2011-05-17 3M Innovative Proprerties Company Chiral fused [1,2]imidazo[4,5-C] ring compounds
US8034938B2 (en) 2004-12-30 2011-10-11 3M Innovative Properties Company Substituted chiral fused [1,2]imidazo[4,5-c] ring compounds
US20080269192A1 (en) * 2004-12-30 2008-10-30 Coley Pharmaceutical Group, Inc. Chiral Fused [1,2]Imidazo[4,5-C] Ring Compounds
US20090163532A1 (en) * 2005-02-04 2009-06-25 Coley Pharmaceutical Group, Inc. 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
US9248127B2 (en) 2005-02-04 2016-02-02 3M Innovative Properties Company Aqueous gel formulations containing immune response modifiers
US20080318998A1 (en) * 2005-02-09 2008-12-25 Coley Pharmaceutical Group, Inc. Alkyloxy Substituted Thiazoloquinolines and Thiazolonaphthyridines
US9546184B2 (en) 2005-02-09 2017-01-17 3M Innovative Properties Company Alkyloxy substituted thiazoloquinolines and thiazolonaphthyridines
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
US8377957B2 (en) 2005-11-04 2013-02-19 3M Innovative Properties Company Hydroxy and alkoxy substituted 1H-imidazoquinolines and methods
US20090221556A1 (en) * 2005-11-04 2009-09-03 Pfizer Inc. Hydroxy and alkoxy substituted 1h-imidazoquinolines and methods
US8088790B2 (en) 2005-11-04 2012-01-03 3M Innovative Properties Company Hydroxy and alkoxy substituted 1H-imidazoquinolines and methods
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
US20080070907A1 (en) * 2006-07-12 2008-03-20 Coley Pharmaceutical Group, Inc. Substituted chiral fused [1,2] imidazo [4,5-C] ring compounds and methods
US8883174B2 (en) 2009-03-25 2014-11-11 The Board Of Regents, The University Of Texas System Compositions for stimulation of mammalian innate immune resistance to pathogens
US9186400B2 (en) 2009-03-25 2015-11-17 The Board Of Regents, The University Of Texas System Compositions for stimulation of mammalian innate immune resistance to pathogens
US9504742B2 (en) 2009-03-25 2016-11-29 The Board Of Regents, The University Of Texas System Compositions for stimulation of mammalian innate immune resistance to pathogens
US10722573B2 (en) 2009-03-25 2020-07-28 The Board Of Regents, The University Of Texas System Compositions for stimulation of mammalian innate immune resistance to pathogens
US10286065B2 (en) 2014-09-19 2019-05-14 Board Of Regents, The University Of Texas System Compositions and methods for treating viral infections through stimulated innate immunity in combination with antiviral compounds

Also Published As

Publication number Publication date
US20100069427A9 (en) 2010-03-18
JP2008530113A (en) 2008-08-07
AU2006213746A1 (en) 2006-08-17
CA2597587A1 (en) 2006-08-17
US7968563B2 (en) 2011-06-28
EP1846405A2 (en) 2007-10-24
WO2006086634A2 (en) 2006-08-17
WO2006086634A3 (en) 2007-08-09

Similar Documents

Publication Publication Date Title
US7968563B2 (en) Oxime and hydroxylamine substituted imidazo[4,5-c] ring compounds and methods
US8093390B2 (en) Substituted fused [1,2]imidazo[4,5-C] ring compounds and methods
US8329721B2 (en) Hydroxy and alkoxy substituted 1H-imidazonaphthyridines and methods
US8034938B2 (en) Substituted chiral fused [1,2]imidazo[4,5-c] ring compounds
US8088788B2 (en) Substituted fused[1,2] imidazo[4,5-c] ring compounds and methods
US7906506B2 (en) Substituted chiral fused [1,2] imidazo [4,5-c] ring compounds and methods
US8143270B2 (en) 2-amino 1H-in-imidazo ring systems and methods
US7163947B2 (en) 1-Amino 1H-imidazoquinolines
US8378102B2 (en) Oxime and hydroxylamine substituted thiazolo[4,5-c] ring compounds and methods
US7939526B2 (en) Sulfone substituted imidazo ring ethers
US7943610B2 (en) Pyrazolopyridine-1,4-diamines and analogs thereof
US7943609B2 (en) Chiral fused [1,2]imidazo[4,5-C] ring compounds
US8026366B2 (en) Aryloxy and arylalkyleneoxy substituted thiazoloquinolines and thiazolonaphthyridines
US7897609B2 (en) Aryl substituted imidazonaphthyridines
US7579359B2 (en) 1-alkoxy 1H-imidazo ring systems and methods
US20090105295A1 (en) Hydroxylamine substituted imidazoquinolines
US20090270443A1 (en) 1-amino imidazo-containing compounds and methods
US20080015184A1 (en) Urea Substituted Imidazopyridines, Imidazoquinolines, and Imidazonaphthyridines
US20090163533A1 (en) 1-Substituted Pyrazolo (3,4-C) Ring Compounds as Modulators of Cytokine Biosynthesis for the Treatment of Viral Infections and Neoplastic Diseases
US20070287725A1 (en) Isoxazole, Dihydroisoxazole, And Oxadiazole Substituted Imidazo Ring Compounds And Method

Legal Events

Date Code Title Description
AS Assignment

Owner name: 3M INNOVATIVE PROPERTIES COMPANY & 3M COMPANY, MIN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:KSHIRSAGAR, TUSHAR A.;LUNDQUIST, GREGORY D., JR.;DELLARIA, JOSEPH F., JR.;AND OTHERS;REEL/FRAME:021565/0532;SIGNING DATES FROM 20050308 TO 20080223

Owner name: 3M INNOVATIVE PROPERTIES COMPANY & 3M COMPANY,MINN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:KSHIRSAGAR, TUSHAR A.;LUNDQUIST, GREGORY D., JR.;DELLARIA, JOSEPH F., JR.;AND OTHERS;SIGNING DATES FROM 20050308 TO 20080223;REEL/FRAME:021565/0532

Owner name: 3M INNOVATIVE PROPERTIES COMPANY & 3M COMPANY, MIN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:KSHIRSAGAR, TUSHAR A.;LUNDQUIST, GREGORY D., JR.;DELLARIA, JOSEPH F., JR.;AND OTHERS;SIGNING DATES FROM 20050308 TO 20080223;REEL/FRAME:021565/0532

AS Assignment

Owner name: COLEY PHARMACEUTICAL GROUP, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:3M COMPANY & 3M INNOVATIVE PROPERTIES COMPANY;REEL/FRAME:021595/0388

Effective date: 20070723

Owner name: COLEY PHARMACEUTICAL GROUP, INC.,MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:3M COMPANY & 3M INNOVATIVE PROPERTIES COMPANY;REEL/FRAME:021595/0388

Effective date: 20070723

AS Assignment

Owner name: 3M INNOVATIVE PROPERTIES COMPANY, MINNESOTA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:KSHIRSAGAR, TUSHAR A.;LUNDQUIST, GREGORY D., JR.;DELLARIA, JOSEPH F., JR.;AND OTHERS;SIGNING DATES FROM 20110418 TO 20110427;REEL/FRAME:026201/0672

REMI Maintenance fee reminder mailed
LAPS Lapse for failure to pay maintenance fees
STCH Information on status: patent discontinuation

Free format text: PATENT EXPIRED DUE TO NONPAYMENT OF MAINTENANCE FEES UNDER 37 CFR 1.362

FP Lapsed due to failure to pay maintenance fee

Effective date: 20150628