Substituted imidazoquinolines and imidazonaphthyridines

Information

  • Patent Grant
  • 8658666
  • Patent Number
    8,658,666
  • Date Filed
    Friday, February 10, 2006
    18 years ago
  • Date Issued
    Tuesday, February 25, 2014
    10 years ago
Abstract
Imidazoquinolines and imidazonaphthyridines with a substituent containing a functional group, e.g., an amide, sulfonamide, urea, or heterocyclyl group, at the 6, 7, 8, or 9-position, pharmaceutical compositions containing the compounds, intermediates, methods of making and methods of use of these compounds as immunomodulators, for inducing cytokine biosynthesis in animals and in the treatment of diseases including viral and neoplastic diseases are disclosed.
Description
BACKGROUND

Certain compounds have been found to be useful as immune response modifiers (IRMs), rendering them useful in the treatment of a variety of disorders. However, there continues to be interest in and a need for compounds that have the ability to modulate the immune response, by induction of cytokine biosynthesis or other mechanisms.


SUMMARY

The present invention provides a new class of compounds that are useful in inducing cytokine biosynthesis in animals. Such compounds are of the following Formula I:




embedded image



wherein RA, RB, R′, and R″ are as defined below.


The compounds of Formula I are useful as immune response modifiers due to their ability to induce cytokine biosynthesis (e.g., induces the synthesis of at least one cytokine) and otherwise modulate the immune response when administered to animals. This makes the compounds useful in the treatment of a variety of conditions such as viral diseases and tumors that are responsive to such changes in the immune response.


The invention further provides pharmaceutical compositions containing an effective amount of a compound of Formula I and methods of inducing cytokine biosynthesis in an animal, treating a viral infection or disease and/or treating a neoplastic disease in an animal by administering an effective amount of a compound of Formula I to the animal.


In addition, methods of synthesizing compounds of Formula I and intermediates useful in the synthesis of these compounds are provided.


As used herein, “a,” “an,” “the,” “at least one,” and “one or more” are used interchangeably.


The terms “comprises” and variations thereof do not have a limiting meaning where these terms appear in the description and claims.


The above summary of the present invention is not intended to describe each disclosed embodiment or every implementation of the present invention. The description that follows more particularly exemplifies illustrative embodiments. In several places throughout the description, guidance is provided through lists of examples, which examples can be used in various combinations. In each instance, the recited list serves only as a representative group and should not be interpreted as an exclusive list.







DETAILED DESCRIPTION OF ILLUSTRATIVE EMBODIMENTS OF THE INVENTION

The present invention provides compounds of the following Formulas I through VII:




embedded image



as well as intermediates of the following Formulas X and XI:




embedded image



wherein RA, RB, R′, R″, R, R1, R2, R3, G, and n are as defined below;


and pharmaceutically acceptable salts thereof.


In one embodiment, the present invention provides a compound of Formula I:




embedded image



wherein:


RA and RB taken together form a fused benzene ring or fused pyridine ring wherein the benzene ring or pyridine ring is substituted by one R3 group, or substituted by one R3 group and one R group;


R3 is selected from the group consisting of:

    • -Z-Y-R4,
    • -Z-Y-X-Y-R4,
    • -Z-R5,
    • -Z-Het,
    • -Z-Het′-R4, and
    • -Z-Het′-Y-R4;


R is selected from the group consisting of alkyl, alkoxy, hydroxy, halogen, and trifluoromethyl;


R′ and R″ are independently selected from the group consisting of hydrogen and non-interfering substitutents;


Z is selected from the group consisting of a bond, alkylene, alkenylene, and alkynylene; wherein alkylene, alkenylene, and alkynylene can be optionally interrupted with 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)—,




embedded image


Het is heterocyclyl which can be unsubstituted or substituted by one or more substituents independently selected from the group consisting of alkyl, alkoxy, haloalkyl, haloalkoxy, halogen, nitro, hydroxy, hydroxyalkyl, cyano, hydroxyalkyleneoxyalkylenyl, amino, alkylamino, dialkylamino, and oxo;


Het′ is heterocyclylene which can be unsubstituted or substituted by one or more substituents independently selected from the group consisting of alkyl, alkoxy, haloalkyl, haloalkoxy, halogen, nitro, hydroxy, hydroxyalkyl, cyano, amino, alkylamino, dialkylamino, and oxo;


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 with arylene, heteroarylene, or heterocyclylene, and optionally interrupted by one or more —O— groups;


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:




embedded image


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, alkyl, alkoxyalkylenyl, hydroxyalkylenyl, arylalkylenyl, and heteroarylalklenyl;


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—, —CH2—, —C(O)—, —S(O)0-2—, and —N(R4)—;


Q is selected from the group consisting of a bond, —C(R6)—, —C(R6)—C(R6)—, —S(O)2—, —C(R6)—N(R8)—W—, —S(O)2—N(R9)—, —C(R6)—O—, and —C(R6)—N(OR5)—;


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;


with the proviso that when R′ is heterocyclyl, then heterocyclyl is attached to the imidazo ring by an atom in heterocyclyl other than a nitrogen atom; and


with the proviso that Z is other than a bond when:

    • R3 is -Z-Y-R4 or -Z-Y-X-Y-R4, and the Y group bonded to Z is —O—, —O—C(R6)—, —OC(O)—O—, —O—C(R6)—N(R8)—,




embedded image



wherein V is —O—C(R6)—,




embedded image




    • R3 is -Z-R5, and R5 is







embedded image



wherein V is —O—C(R6)—; or

    • R3 is -Z-Het, -Z-Het′-R4, or -Z-Het′-Y-R4, and Z is attached to a nitrogen atom in Het or Het′;
    • and with the further proviso that R3 is other than —NH2;


      or a pharmaceutically acceptable salt thereof.


In another embodiment, the present invention provides a compound of Formula II:




embedded image



wherein:


R3 is selected from the group consisting of:

    • -Z-Y-R4,
    • -Z-Y-X-Y-R4,
    • -Z-R5,
    • -Z-Het,
    • -Z-Het′-R4, and
    • -Z-Het′-Y-R4;


R is selected from the group consisting of alkyl, alkoxy, hydroxy, halogen, and trifluoromethyl;


n is 0 or 1;


R1 is selected from the group consisting of:

    • -R4,
    • -X-R4,
    • -X-Y-R4,
    • -X-Y-X-Y-R4, and
    • -X-R5;


R2 is selected from the group consisting of:

    • -R4,
    • -X-R4,
    • -X-Y-R4, and
    • -X-R5;


Z is selected from the group consisting of a bond, alkylene, alkenylene, and alkynylene; wherein alkylene, alkenylene, and alkynylene can be optionally interrupted with 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)—,




embedded image


Het is heterocyclyl which can be unsubstituted or substituted by one or more substituents independently selected from the group consisting of alkyl, alkoxy, haloalkyl, haloalkoxy, halogen, nitro, hydroxy, hydroxyalkyl, cyano, hydroxyalkyleneoxyalkylenyl, amino, allylamino, diallylamino, and oxo;


Het′ is heterocyclylene which can be unsubstituted or substituted by one or more substituents independently selected from the group consisting of alkyl, alkoxy, haloalkyl, haloalkoxy, halogen, nitro, hydroxy, hydroxyalkyl, cyano, amino, alkylamino, dialkylamino, and oxo;


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 with arylene, heteroarylene, or heterocyclylene, and optionally interrupted by one or more —O— groups;


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:




embedded image


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, alkyl, alkoxyalkylenyl, hydroxyalkylenyl, arylalkylenyl, and heteroarylalklenyl;


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—, —CH2—, —C(O)—, —S(O)0-2—, and —N(R4)—;


Q is selected from the group consisting of a bond, —C(R6)—, —C(R6)—C(R6)—, —S(O)2—, —C(R6)—N(R8)—W—, —S(O)2—N(R8)—, —C(R6)—O—, 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;


with the proviso that when R1 is R4, and R4 is heterocyclyl, then heterocyclyl is attached to the imidazo ring by an atom in heterocyclyl other than a nitrogen atom; and


with the proviso that Z is other than a bond when:

    • R3 is -Z-Y-R4 or -Z-Y-X-Y-R4, and the Y group bonded to Z is —O—, —O—C(R6)—, —OC(O)—O—, —O—C(R6)—N(R8)—,




embedded image



wherein V is —O—C(R6)—,




embedded image




    • R3 is -Z-R5, and R5 is







embedded image



wherein V is —O—C(R6)—; or

    • R3 is -Z-Het, -Z-Het′-R4, or -Z-Het′-Y-R4, and Z is attached to a nitrogen atom in Het or Het′;
    • and with the further proviso that R3 is other than —NH2;


      or a pharmaceutically acceptable salt thereof.


In another embodiment, the present invention provides a compound selected from the group consisting Formulas III, IV, V, and VI:




embedded image



wherein:


R3 is selected from the group consisting of:

    • -Z-Y-R4,
    • -Z-Y-X-Y-R4,
    • -Z-R5,
    • -Z-Het,
    • -Z-Het′-R4, and
    • -Z-Het′-Y-R4;


R is selected from the group consisting of alkyl, alkoxy, hydroxy, halogen, and trifluoromethyl;


n is 0 or 1;


R1 is selected from the group consisting of:

    • -R4,
    • -X-R4,
    • -X-Y-R4,
    • -X-Y-X-Y-R4, and
    • -X-R5;


R2 is selected from the group consisting of:

    • -R4,
    • -X-R4,
    • -X-Y-R4, and
    • -X-R5;


Z is selected from the group consisting of a bond, alkylene, alkenylene, and alkynylene; wherein alkylene, alkenylene, and alkynylene can be optionally interrupted with 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)—,




embedded image


Het is heterocyclyl which can be unsubstituted or substituted by one or more substituents independently selected from the group consisting of alkyl, alkoxy, haloalkyl, haloalkoxy, halogen, nitro, hydroxy, hydroxyallyl, cyano, hydroxyalkyleneoxyalkylenyl, amino, alkylamino, diallylamino, and oxo;


Het′ is heterocyclylene which can be unsubstituted or substituted by one or more substituents independently selected from the group consisting of alkyl, alkoxy, haloalkyl, haloalkoxy, halogen, nitro, hydroxy, hydroxyalkyl, cyano, amino, alkylamino, dialkylamino, and oxo;


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 with arylene, heteroarylene, or heterocyclylene, and optionally interrupted by one or more —O— groups;


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:




embedded image


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, alkyl, alkoxyalkylenyl, hydroxyalkylenyl, arylalkylenyl, and heteroarylalklenyl;


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—, —CH2—, —C(O)—, —S(O)0-2—, and —N(R4)—;


Q is selected from the group consisting of a bond, —C(R6)—, —C(R6)—C(R6)—, —S(O)2—, —C(R6)—N(R8)—W—, —S(O)2—N(R8)—, —C(R6)—O—, 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;


with the proviso that Z is other than a bond when:

    • R3 is -Z-Y-R4 or -Z-Y-X-Y-R4, and the Y group bonded to Z is —O—, —O—C(R6)—, —OC(O)—O—, —O—C(R6)—N(R8)—,




embedded image



wherein V is —O—C(R6)—,




embedded image




    • R3 is -Z-R5, and R5 is







embedded image



wherein V is —O—C(R6)—; or

    • R3 is -Z-Het, -Z-Het′-R4, or -Z-Het′-Y-R4, and Z is attached to a nitrogen atom in Het or Het′;


      or a pharmaceutically acceptable salt thereof.


In another embodiment, the present invention provides a compound of Formula VII, which is a prodrug:




embedded image


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-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-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-C1-4 alkylpiperazin-1-yl;


RA and RB are defined as in Formula I above;


R1 and R2 are as defined as in Formula II above;


or a pharmaceutically acceptable salt thereof.


In one embodiment, the present invention provides an intermediate compound of Formula X:




embedded image



wherein:


R3 is selected from the group consisting of:

    • -Z-Y-R4,
    • -Z-Y-X-Y-R4,
    • -Z-R5,
    • -Z-Het,
    • -Z-Het′-R4, and
    • -Z-Het′-Y-R4;


R is selected from the group consisting of alkyl, alkoxy, hydroxy, halogen, and trifluoromethyl;


n is 0 or 1;


R1 is selected from the group consisting of:

    • -R4,
    • -X-R4,
    • -X-Y-R4,
    • -X-Y-X-Y-R4, and
    • -X-R5;


R2 is selected from the group consisting of:

    • -R4,
    • -X-R4,
    • -X-Y-R4, and
    • -X-R5;


Z is selected from the group consisting of a bond, alkylene, alkenylene, and alkynylene; wherein alkylene, alkenylene, and alkynylene can be optionally interrupted with 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)—,




embedded image


Het is heterocyclyl which can be unsubstituted or substituted by one or more substituents independently selected from the group consisting of alkyl, alkoxy, haloalkyl, haloalkoxy, halogen, nitro, hydroxy, hydroxyalkyl, cyano, hydroxyalkyleneoxyalkylenyl, amino, alkylamino, dialkylamino, and oxo;


Het′ is heterocyclylene which can be unsubstituted or substituted by one or more substituents independently selected from the group consisting of alkyl, alkoxy, haloalkyl, haloalkoxy, halogen, nitro, hydroxy, hydroxyalkyl, cyano, amino, alkylamino, dialkylamino, and oxo;


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 with arylene, heteroarylene, or heterocyclylene, and optionally interrupted by one or more —O— groups;


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:




embedded image


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, alkyl, alkoxyalkylenyl, hydroxyalkylenyl, arylalkylenyl, and heteroarylalklenyl;


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—, —CH2—, —C(O)—, —S(O)0-2—, and —N(R4)—;


Q is selected from the group consisting of a bond, —C(R6)—, —C(R6)—C(R6)—, —S(O)2—, —C(R6)—N(R8)—W—, —S(O)2—N(R8)—, —C(R6)—O—, 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;


with the proviso that Z is other than a bond when:

    • R3 is -Z-Y-R4 or -Z-Y-X-Y-R4, and the Y group bonded to Z is —O—, —O—C(R6)—, —OC(O)—O—, —O—C(R6)—N(R8)—,




embedded image



wherein V is —O—C(R6)—,




embedded image




    • R3 is -Z-R5, and R5 is







embedded image



wherein V is —O—C(R6)—; or

    • R3 is -Z-Het, -Z-Het′-R4, or -Z-Het′-Y-R4, and Z is attached to a nitrogen atom in Het or Het′;
    • and with the further proviso that R3 is other than —NH2;


      or a pharmaceutically acceptable salt thereof.


In one embodiment, the present invention provides an intermediate compound of Formula XI:




embedded image



wherein:


R3 is selected from the group consisting of:

    • -Z-Y-R4,
    • -Z-Y-X-Y-R4,
    • -Z-R5,
    • -Z-Het,
    • -Z-Het′-R4, and
    • -Z-Het′-Y-R4;


R is selected from the group consisting of alkyl, alkoxy, hydroxy, halogen, and trifluoromethyl;


n is 0 or 1;


R1 is selected from the group consisting of:

    • -R4,
    • -X-R4,
    • -X-Y-R4,
    • -X-Y-X-Y-R4, and
    • -X-R5;


R2 is selected from the group consisting of:

    • -R4,
    • -X-R4,
    • -X-Y-R4, and
    • -X-R5;


Z is selected from the group consisting of a bond, alkylene, alkenylene, and alkynylene; wherein alkylene, alkenylene, and alkynylene can be optionally interrupted with 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)—,




embedded image


Het is heterocyclyl which can be unsubstituted or substituted by one or more substituents independently selected from the group consisting of alkyl, alkoxy, haloalkyl, haloalkoxy, halogen, nitro, hydroxy, hydroxyalkyl, cyano, hydroxyalkyleneoxyalkylenyl, amino, alkylamino, dialkylamino, and oxo;


Het′ is heterocyclylene which can be unsubstituted or substituted by one or more substituents independently selected from the group consisting of alkyl, alkoxy, haloalkyl, haloalkoxy, halogen, nitro, hydroxy, hydroxyalkyl, cyano, amino, alkylamino, dialkylamino, and oxo;


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 with arylene, heteroarylene, or heterocyclylene, and optionally interrupted by one or more —O— groups;


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:




embedded image


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, alkyl, alkoxyalkylenyl, hydroxyalkylenyl, arylalkylenyl, and heteroarylalklenyl;


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—, —CH2—, —C(O)—, —S(O)0-2—, and —N(R4)—;


Q is selected from the group consisting of a bond, —C(R6)—, —C(R6)—C(R6)—, —S(O)2—, —C(R6)—N(R8)—W—, —S(O)2—N(R8)—, —C(R6)—O—, and —C(R6)—N(OR5)—;


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,


with the proviso that Z is other than a bond when:

    • R3 is -Z-Y-R4 or -Z-Y-X-Y-R4, and the Y group bonded to Z is —O—, —O—C(R6)—, —OC(O)—O—, —O—C(R6)—N(R8)—,




embedded image



wherein V is —O—C(R6)—,




embedded image




    • R3 is -Z-R5, and R5 is







embedded image



wherein V is —O—C(R6)—; or

    • R3 is -Z-Het, -Z-Het′-R4, or -Z-Het′-Y-R4, and Z is attached to a nitrogen atom in Het or Het′;


      or a pharmaceutically acceptable salt thereof.


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 substitutent. For certain embodiments, R′ is hydrogen or a non-interfering substituent. Illustrative non-interfering R′ groups include those described above for R1. For certain embodiments, R″ is hydrogen or a non-interfering substituent. Illustrative non-interfering R″ groups include those described above for R2.


As used herein, the terms “alkyl,” “alkenyl,” “alkynyl” and the prefix “alk-” are inclusive of both straight chain and branched chain groups and of cyclic groups, e.g., cycloalkyl and cycloalkenyl. Unless otherwise specified, these groups contain from 1 to 20 carbon atoms, with alkenyl groups containing from 2 to 20 carbon atoms, and alkynyl groups containing from 2 to 20 carbon atoms. In some embodiments, these groups have a total of up to 10 carbon atoms, up to 8 carbon atoms, up to 6 carbon atoms, or up to 4 carbon atoms. Cyclic groups can be monocyclic or polycyclic and preferably have from 3 to 10 ring carbon atoms. Exemplary cyclic groups include cyclopropyl, cyclopropylmethyl, cyclopentyl, cyclohexyl, adamantyl, and substituted and unsubstituted bornyl, norbornyl, and norbornenyl.


Unless otherwise specified, “alkylene,” “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, 1,1-dioxidotetrahydrothien-3-yl, 2-oxopyrrolidin-1-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 unless otherwise indicated.


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(R6)—N(R8)— each R8 group is independently selected. In another example, when an R2 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, Y, X, RA, RB, R, R1, R2, R3, Q, 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 Formulas I or VII, RA and RB taken together form a fused benzene ring or fused pyridine ring wherein the benzene ring or pyridine ring is substituted by one R3 group, or substituted by one R3 group and one R group.


For certain embodiments of Formulas I or VII, RA and RB are taken together to form a fused benzene ring wherein the benzene ring is substituted by one R3 group, or substituted by one R3 group and one R group. In certain of these embodiments, the fused benzene ring is substituted by one R3 group.


For certain embodiments of Formulas I or VII, RA and RB are taken together to form a fused pyridine ring wherein the pyridine ring is substituted by one R3 group, or substituted by one R3 group and one R group. In certain of these embodiments, the fused pyridine ring is substituted by one R3 group.


For certain embodiments, including any one of the above 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;


with the proviso that when R1 is R4, and R4 is heterocyclyl, then heterocyclyl is attached to the imidazo ring by an atom in heterocyclyl other than a nitrogen atom.


For certain embodiments, including any one of the above embodiments of Formula I, R″ is R2; wherein R2 is selected from the group consisting of:

    • -R4,
    • -X-R4,
    • -X-Y-R4, and
    • -X-R5.


For certain embodiments, the compound selected from the group consisting of Formulas III, IV, V, and VI, or a pharmaceutically acceptable salt thereof is the compound of Formula III:




embedded image



or a pharmaceutically acceptable salt thereof.


For certain embodiments, n is 0 in the above embodiments of Formulas II, III, IV, V, or VI.


For certain embodiments, R is selected from the group consisting of alkyl, alkoxy, hydroxy, halogen, and trifluoromethyl.


For certain embodiments, R′ is hydrogen or a non-interfering substituent.


For certain embodiments, R′ is a non-interfereing substituent.


For certain embodiments, 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, R′ is R1; wherein R1 is selected from the group consisting of alkyl, arylalkylenyl, aryloxyalkylenyl, hydroxyalkyl, dihydroxyalkyl, alkoxyalkylenyl, 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




embedded image


R4 is alkyl, aryl, or heteroaryl; and R5 is




embedded image


For certain embodiments, R″ is hydrogen or a non-interfering substituent.


For certain embodiments, R″ is a non-interfering substituent.


For certain embodiments, R″ is R2; wherein R2 is selected from the group consisting of -R4, -X-R4, -X-Y′-R4, and -X-R5′.


For certain embodiments, R″ is R2; wherein R2 is selected from the group consisting of hydrogen, alkyl, alkoxyalkylenyl, and hydroxyalkylenyl.


For certain embodiments, 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, alkoxyalkylenyl, 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




embedded image



R4 is alkyl, aryl, or heteroaryl; and R5 is




embedded image


For certain embodiments, including any one of the above embodiments wherein R1 is present, R1 is selected from the group consisting of 2-hydroxy-2-methylpropyl, 2-methylpropyl, propyl, ethyl, methyl, 2,3-dihydroxypropyl, 3-isopropoxypropyl, 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 embodiments, including any one of the above embodiments wherein R1 is present except where excluded, R1 is selected from the group consisting of 2-[(methylsulfonyl)amino]ethyl, 2-(1,1-dioxidoisothiazolidin-2-yl)ethyl, 3-(2-oxopyrrolidin-1-yl)propyl, and 2-{[(isopropylamino)carbonyl]amino}ethyl.


For certain embodiments of Formulas III, IV, V, VI, or XI, when R1 is R4, and R4 is heterocyclyl, then heterocyclyl is bonded to the imidazo ring by an atom in heterocyclyl that is other then nitrogen.


For certain embodiments, R2 is selected from the group consisting of -R4, -X-R4, -X-Y-R4, and -X-R5.


For certain embodiments, including any one of the above embodiments wherein R2 is present, R2 is selected from the group consisting of hydrogen, alkyl, alkoxyalkylenyl, and hydroxyalkylenyl. For certain embodiments, R2 is selected from the group consisting of hydrogen, alkyl, and alkoxyalkylenyl. For certain embodiments, R2 is selected from the group consisting of alkyl and alkoxyalkylenyl. For certain embodiments, R2 is selected from the group consisting of hydrogen, methyl, ethyl, propyl, butyl, ethoxymethyl, methoxymethyl, 2-methoxyethyl, hydroxymethyl, and 2-hydroxyethyl. For certain embodiments, R2 is selected from the group consisting of methyl, ethyl, n-propyl, n-butyl, ethoxymethyl, methoxymethyl, and 2-methoxyethyl.


For certain embodiments, R3 is selected from the group consisting of -Z-Y-R4, -Z-Y-X-Y-R4, -Z-R5, -Z-Het, -Z-Het′-R4, and -Z-Het′-Y-R4.


For certain embodiments, including any one of the above embodiments, R3 is


-Z-Y-R4 or -Z-Y-X-Y-R4. For certain of these embodiments, R3 is -Z-Y-R4. In certain of these embodiments, Y is —N(R8)-Q-, and R4 is alkyl, aryl, heteroaryl, or heterocyclyl. In certain of these embodiments, Q is —C(R6)—, —S(O)2—, or —C(R6)—N(R8)—. For certain of these embodiments, Q is —C(R6)—. Alternatively, for certain of these embodiments, Q is —S(O)2—. Alternatively, for certain of these embodiments, Q is —C(R6)—N(R8)—. Alternatively, for certain of these embodiments, Q is —C(R6)—O—. Alternatively, for certain of these embodiments, Q is —S(O)2—N(R8)—. For certain of these embodiments where R3 is -Z-Y-R4 or -Z-Y-X-Y-R4, R3 is -Z-Y-X-Y-R4.


In certain embodiments, including any one of the above embodiments wherein R3 is -Z-Y-R4 or -Z-Y-X-Y-R4, except where excluded, Y is —C(R6)—N(R8)—, and R4 is alkyl, aryl, arylalkylenyl, heteroaryl, heteroarylalkylenyl, or heterocyclyl. For certain of these embodiments, Y in -Y-R4 is —C(R6)—N(R8)—. For certain of these embodiments, R6 is ═O, R8 is C1-4 alkyl, and R4 is C1-4 alkyl. For certain of these embodiments, C1-4 alkyl is methyl.


In certain embodiments, including any one of the above embodiments wherein R3 is -Z-Y-R4 or -Z-Y-X-Y-R4, except where excluded, -Y-R4 is —C(O)—NH2.


In certain embodiments, including any one of the above embodiments wherein R3 is -Z-Y-X-Y-R4, except where excluded, Y in -Z-Y- is —C(O)—, and Y in -Y-R4 is selected from the group consisting of —C(O)—NH—, —C(O)—, and —C(O)—O—. For certain of these embodiments, R4 is alkyl, and when Y in -Y-R4 is —C(O)—NH—, then R4 can also be hydrogen. For certain of these embodiments, X is heterocyclylene or heterocyclylene-alkylene.


In certain embodiments, including any one of the above embodiments wherein R3 is -Z-Y-R4 or -Z-Y-X-Y-R4, except where excluded, Y is —C(O)— and R4 is heterocyclyl.


In certain embodiments, including any one of the above embodiments wherein R3 is -Z-Y-R4, except where excluded, Y is —C(O)—, and R4 is heterocyclyl which is unsubstituted or substituted by one or more substituents independently selected from the group consisting of alkyl, hydroxy, and oxo.


In certain embodiments, including any one of the above embodiments wherein R3 is -Z-Y-R4, except where excluded, Y is —C(O)—NH—, and R4 is heterocyclyl which is unsubstituted or substituted by one or more substituents independently selected from the group consisting of alkyl, hydroxy, and oxo, or R4 is alkyl which is unsubstituted or substituted by one or more substituents independently selected from the group consisting of hydroxy, alkoxy, heteroaryl, and heterocyclyl.


For certain embodiments, including any one of the above embodiments wherein R4 is heterocyclyl, heterocyclyl is selected from the group consisting of tetrahydropyranyl, tetrahydrofuranyl, 1,3-dioxolanyl, pyrrolidinyl, piperidinyl, morpholinyl, thiomorpholinyl, 1,1-dioxothiomorpholinyl, thiazolidinyl, azepanyl, 1,4-oxazepanyl, diazepanyl, dihydroisoquinolin-(1H)-yl, octahydroisoquinolin-(1H)-yl, dihydroquinolin-(2H)-yl, octahydroquinolin-(2H)-yl, dihydro-1H-imidazolyl, 3-azabicyclo[3.2.2]non-3-yl, and piperazinyl. For certain of these embodiments, heterocyclyl is pyrrolidinyl or piperidinyl. Alternatively, for certain of these embodiments, heterocyclyl is 1,1-dioxidotetrahydrothien-3-yl, or 2-oxopyrrolidin-1-yl.


In certain embodiments, including any one of the above embodiments wherein R3 is -Z-Y-R4, except where excluded, Y is selected from the group consisting of —C(O)—O—, —S(O)2—, and —S(O)2—N(R8)—. For certain of these embodiments, Y is —S(O)2—. For certain of these embodiments, Y is —S(O)2—N(R8). For certain of these embodiments, Y is —C(O)—O—. For certain of these embodiments, R4 is alkyl.


In certain embodiments, including any one of the above embodiments wherein R3 is -Z-Y-R4, except where excluded, Y is —O—, and R4 is hydrogen.


In certain embodiments, including any one of the above embodiments wherein R3 is -Z-Y-R4, except where excluded, Y is




embedded image



V is —C(O)—, and R4 is hydroxyalkyl.


For certain embodiments, including any one of the above embodiments not excluding this definition, R3 is -Z-R5. In certain of these embodiments, R5 is




embedded image



In certain of these embodiments, V is —NH—C(O)— or —C(O)—; A is —O—, —CH2—, —S—, or —SO2—; a is 1, 2, or 3; and b is 2.


For certain embodiments wherein R3 is -Z-R5, R5 is —C≡N. In certain of these embodiments, Z is other than a bond. In certain of these embodiments, Z is alkylene. Alternatively, in certain of these embodiments, R3 is —CH═CH—C≡N.


For certain embodiments, including any one of the above embodiments not excluding this definition, R3 is -Z-Het or -Z-Het′-R4. In certain of these embodiments, Het and Het′ are, respectively, selected from the group consisting of the monovalent and divalent forms of tetrahydropyranyl, tetrahydrofuranyl, 1,3-dioxolanyl, pyrrolidinyl, piperidinyl, morpholinyl, thiomorpholinyl, 1,1-dioxothiomorpholinyl, thiazolidinyl, azepanyl, 1,4-oxazepanyl, diazepanyl, dihydroisoquinolin-(1)-yl, octahydroisoquinolin-(1H)-yl, dihydroquinolin-(2H)-yl, octahydroquinolin-(2H)-yl, dihydro-1H-imidazolyl, 3-azabicyclo[3.2.2]non-3-yl, and piperazinyl, each of which is unsubstituted or substituted by one or more substitutents. For certain of these embodiments, Het and Het′ are, respectively, selected from the group consisting of the monovalent and divalent forms of pyrrolidinyl, piperidinyl, and morpholinyl, each of which is unsubstituted or substituted by one or more substitutents. For certain of these embodiments, Het is unsubstituted. For certain of these embodiments, Het′ is unsubstituted. For certain of these embodiments, R4 is heterocyclyl. For certain of these embodiments, Het is substituted by one or more substituents selected from the group consisting of alkyl, hydroxyl, hydroxyalkyl, hydroxyalkyleneoxyalkylenyl, and dialkylamino.


For certain embodiments, including any one of the above embodiments, except where excluded, Z is C1-4 alkylene or C2-4 alkenylene. For certain of these embodiments, Z is C2-4 alkylene. For certain of these embodiments, Z is ethylene.


For certain embodiments, including any one of the above embodiments not excluded by the proviso for Formulas I through VI, and which does not exclude this definition, Z is a bond.


For certain embodiments, including any one of the above embodiments, 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 selected from the group consisting of alkyl, aryl, arylalkylenyl, alkylheteroarylenyl, heteroarylalkylenyl, heteroaryl, and heterocyclyl; wherein alkyl is unsubstituted or substituted by one or more substituents selected from the group consisting of hydroxy, alkoxy, and aryl; wherein aryl and arylalkylenyl are unsubstituted or substituted by one or more substituents selected from the group consisting of alkyl, halogen, cyano, dialkylamino, and alkoxy; and wherein heterocyclyl is unsubstituted or substituted by one or more alkyl substituents.


For certain embodiments, R4 is alkyl, aryl, arylalkylenyl, heteroaryl, heteroarylalkylenyl, or heterocyclyl.


For certain embodiments, R4 is alkyl, aryl, heteroaryl, or heterocyclyl.


For certain embodiments, R4 is alkyl, aryl, or heteroaryl.


For certain embodiments, R4 is alkyl substituted by heterocyclyl. For certain of these embodiments, heterocyclyl is substituted by one or two oxo groups.


For certain embodiments, R4 is heterocyclyl.


For certain embodiments, R4 is hydrogen or alkyl.


For certain embodiments, R4 is alkyl.


For certain embodiments, R4 is C1-4 alkyl.


For certain embodiments, R4 is methyl.


For certain embodiments, R4 is hydrogen.


For certain embodiments, R5 is selected from the group consisting of:




embedded image


For certain embodiments, R5 is selected from the group consisting of:




embedded image


For certain embodiments, R5 is




embedded image


For certain embodiments, R5 is




embedded image



V is —NH—C(O)— or —C(O)—; A is —O—, —CH2—, —S—, or —SO2—; a is 1, 2, or 3; and b is 2.


For certain embodiments, R5 is —C≡N.


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, alkyl, hydroxyalkylenyl, alkoxyalkylenyl, arylalkylenyl, and heteroarylalkylenyl.


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 C1-4 alkyl.


For certain embodiments, R8 is methyl.


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 C3-8 alkylene.


For certain embodiments, R10 is C4-6 alkylene.


For certain embodiments, R10 is C4-5 alkylene.


For certain embodiments, A is selected from the group consisting of —O—, —CH2—, —C(O)—, —S(O)0-2—, and —N(R4)—.


For certain embodiments, A is —O—, —CH2—, —S—, or —S(O)2—.


For certain embodiments, A is —O— or —S(O)2—.


For certain embodiments, A is —S—.


For 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(OC14 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-C1-4 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(R9)—, —C(R6)—O—, and —C(R6)—N(OR9)—.


In certain embodiments, Q is selected from the group consisting of —C(R6)—, —C(R6)—C(R6)—, —S(O)2—, —C(R6)—N(R8)—W—, —S(O)2—N(R5)—, —C(R6)—O—, and —C(R6)—N(OR9)—.


In certain embodiments, Q is other than a bond when R3 is -Z-Y-R4 and Y is —N(R5)-Q-.


In certain embodiments, Q is selected from the group consisting of a bond, —C(R6)—, —S(O)2—, and —C(R6)—N(R8)—.


In certain embodiments, Q is —C(O)—NH—, —C(O)—, or —C(O)—O—.


In certain embodiments, Q is —C(R6)—.


In certain embodiments, Q is —C(O)—.


In certain embodiments, Q is —S(O)2—.


In certain embodiments, Q is —C(R6)—N(R8)—.


In certain embodiments, Q is —C(O)—NH—.


In certain embodiments, Q is —C(R6)—O—.


In certain embodiments, Q is —S(O)2—N(R8)—.


In certain embodiments, Q is a bond.


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 —NH—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 or —C(O)—.


In certain embodiments, W is a bond.


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 with arylene, heteroarylene, or heterocyclylene, and optionally interrupted by one or more —O— groups.


In certain embodiments, X is alkylene.


In certain embodiments, X is heteroarylene-alkylene. For certain of these embodiments, heteroarylene is thiazoldiyl.


In certain embodiments, X is heterocyclylene. For certain of these embodiments, heterocyclylene is piperidin-1,4-diyl or piperazin-1,4-diyl.


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)—,




embedded image



In certain of these embodiments, including any one of the above embodiments wherein Y is present, Y is other than —O—.


In certain embodiments, Y is —N(R8)—C(O)—, —N(R8)—S(O)2—, —N(R8)—C(O)—N(R8)—, or




embedded image


In certain embodiments, Y is —N(R8)-Q-.


In certain embodiments, Y is —N(R8)-Q-, and Q is —C(R6)—, S(O)2—, or —C(R6)—N(R8)—.


In certain embodiments, Y is —N(R8)—C(R6)—.


In certain embodiments, Y is —NH—C(O)—.


In certain embodiments, Y is —NH—S(O)2—.


In certain embodiments, Y is —NH—C(O)—NH—.


In certain embodiments, Y is —NH—C(O)—O—.


In certain embodiments, Y is —NH—S(O)2—N(R8)—.


In certain embodiments, Y is —C(R6)—N(R8)—.


In certain embodiments, Y is —C(O)—NH—.


In certain embodiments, Y is —C(O)—.


In certain embodiments, Y is —C(O)—O—.


In certain embodiments, Y is —C(O)—NH—, —C(O)—, or —C(O)—O—.


In certain embodiments, Z is selected from the group consisting of a bond, alkylene, alkenylene, and alkynylene, wherein alkylene, alkenylene, and alkynylene can be optionally interrupted with one or more —O— groups; with the proviso that Z is other than a bond when:

    • R3 is -Z-Y-R4 or -Z-Y-X-Y-R4, and the Y group bonded to Z is —O—, —O—C(R6)—, —OC(O)—O—, —O—C(R6)—N(R8)—,




embedded image



wherein V is —O—C(R6)—,




embedded image




    • R3 is -Z-R5, and R5 is







embedded image



wherein V is —O—C(R6)—; or

    • R3 is -Z-Het, -Z-Het′-R4, or -Z-Het′-Y-R4, and Z is attached to a nitrogen atom in Het or Het′. In certain of these embodiments, Z is other than a bond further when R5 is —C≡N. In certain of these embodiments, Z is other than a bond further when R3 is -Z-Y-R4, Y is —C(O)— or —C(O)—O—, and R4 is alkyl.


In certain embodiments, Z is a bond except where excluded by the above proviso.


In certain embodiments, Z is C1-4 alkylene or C2-4 alkenylene.


In certain embodiments, Z is C2-4 alkylene.


In certain embodiments, Z is ethylene.


In certain embodiments, Het is heterocyclyl which can be unsubstituted or substituted by one or more substituents independently selected from the group consisting of alkyl, alkoxy, haloalkyl, haloalkoxy, halogen, nitro, hydroxy, hydroxyalkyl, cyano, hydroxyalkyleneoxyalkylenyl, amino, alkylamino, diallylamino, and oxo.


In certain embodiments, Het is selected from the group consisting of tetrahydropyranyl, tetrahydrofuranyl, 1,3-dioxolanyl, pyrrolidinyl, piperidinyl, morpholinyl, thiomorpholinyl, 1,1-dioxothiomorpholinyl, thiazolidinyl, azepanyl, 1,4-oxazepanyl, diazepanyl, dihydroisoquinolin-(1H)-yl, octahydroisoquinolin-(1H)-yl, dihydroquinolin-(2H)-yl, octahydroquinolin-(2H)-yl, dihydro-1H-imidazolyl, 3-azabicyclo[3.2.2]non-3-yl, and piperazinyl, each of which is unsubstituted or substituted by one or more substitutents.


In certain embodiments, Het is substituted by one or more substituents selected from the group consisting of alkyl, hydroxyl, hydroxyalkyl, hydroxyalkyleneoxyalkylenyl, and dialkylamino.


In certain embodiments, Het is unsubstituted.


In certain embodiments, Het′ is heterocyclylene which can be unsubstituted or substituted by one or more substituents independently selected from the group consisting of alkyl, alkoxy, haloalkyl, haloalkoxy, halogen, nitro, hydroxy, hydroxyalkyl, cyano, amino, alkylamino, dialkylamino, and oxo.


In certain embodiments, Het′ is selected from the group consisting of the divalent forms of tetrahydropyranyl, tetrahydrofuranyl, 1,3-dioxolanyl, pyrrolidinyl, piperidinyl, morpholinyl, thiomorpholinyl, 1,1-dioxothiomorpholinyl, thiazolidinyl, azepanyl, 1,4-oxazepanyl, diazepanyl, dihydroisoquinolin-(1H)-yl, octahydroisoquinolin-(1H)-yl, dihydroquinolin-(2H)-yl, octahydroquinolin-(2H)-yl, dihydro-1H-imidazolyl, 3-azabicyclo[3.2.2]non-3-yl, and piperazinyl, each of which is unsubstituted or substituted by one or more substitutents.


In certain embodiments, Het′ is unsubstituted (except by -R4 or -Y-R4).


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 independently 1 to 3.


In certain embodiments, a and b are each 2.


In certain embodiments, a is 1, 2, or 3, and b is 2.


In certain embodiments, n is 0 or 1.


In certain embodiments, n is 0.


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.


For some embodiments, compounds of the invention can be prepared according to Reaction Scheme I, wherein R1, R2, R, and n are as defined above; Hal is —Br or —I; R3a is selected from the group consisting of —C(H)═C(H)—Za—Y—R4, —C(H)═C(H)—Za—Y—X—Y—R4, —C(H)═C(H)—Za—R5, —C(H)═C(H)—Za-Het, —C(H)═C(H)—Za-Het′-R4, —C(H)═C(H)—Za-Het′-Y—R4, —N(R8)—C(R6)—R4, —N(R8)—SO2—R4, and




embedded image



wherein R8, R6, R5, R4, Y, X, Het, and Het′ are as defined above, and Za is selected from the group consisting of a bond and alkylene, wherein alkylene can be optionally interrupted with one or more —O— groups; and R3b is selected from the group consisting of —CH2—CH2—Za—Y—R4, —CH2—CH2—Za—Y—X—Y—R4, —CH2—CH2—Za—R5, —CH2—CH2—Za-Het, —CH2—CH2—Za-Het′-R4, and —CH2—CH2—Za-Het′-Y—R4, wherein R5, R4, Za, Y, X, Het, and Het′ are as defined above. Numerous compounds of Formula XV are known; see, for example, U.S. Patent Application Publication No. US 2004/0147543 and the documents cited therein.


The Heck reaction is used in step (1) of Reaction Scheme I to provide compounds of Formula IIa, wherein R3a is selected from the group consisting of —C(H)═C(H)—Za—Y—R4, —C(H)═C(H)—Za—Y—X—Y—R4, —C(H)═C(H)—Za—R5, —C(H)═C(H)—Za-Het, C(H)═C(H)—Za-Het′-R4, and —C(H)═C(H)—Za-Het′-Y—R4. The Heck reaction is carried out by coupling a 1H-imidazo[4,5-c]quinolin-4-amine of Formula XV with a compound of the Formula H2C═C(H)—Za—Y—R4, H2C═C(H)—Za—Y—X—Y—R4, H2C═C(H)—Za—R5, H2C═C(H)—Za-Het, H2C═C(H)—Za-Het′-R4, or H2C═C(H)—Za-Het′-Y—R4. Several of these vinyl-substituted compounds are commercially available; others can be prepared by known methods. For example, vinyl-substituted compounds are readily prepared from aldehydes, primary alcohols, or primary allyl halides using a variety of conventional methods. The reaction is conveniently carried out by combining the compound of Formula XV and the vinyl-substituted compound in the presence of palladium (II) acetate, triphenylphosphine or tri-ortho-tolylphosphine, and a base such as triethylamine or cesium carbonate in a suitable solvent such as acetonitrile, toluene, or N,N-dimethylformamide (DMF). The reaction can be carried out at an elevated temperature such as 85° C. to 125° C. under an inert atmosphere. The product of Formula IIa, a subgenus of Formulas I and II, or pharmaceutically acceptable salt thereof can be isolated using conventional methods. The Heck reaction can also be carried out on a trifluoromethanesulfonate-substituted 1H-imidazo[4,5-c]quinolin-4-amine, in which Hal in Formula XV is replaced by —O(SO)2—CF3. It is understood by one skilled in the art that certain substrates are not compatible with Heck reaction chemistry; see, R. F. Heck, in Comprehensive Organic Synthesis, Vol. 4 (Eds.: B. M. Trost, I. Fleming), Pergamon Press, Oxford, p. 833, (1991). For example, it is understood by one skilled in the art that for the Heck reaction described above, Za is other than a bond when a Y group bonded to Za is —O—, —O—C(R6)—, —OC(O)—O—, —O—C(R6)—N(R8)—,




embedded image



wherein V is —O—C(R6)—, or




embedded image



or when the R5 bonded to Za is




embedded image



wherein V is —O—C(R6)—; or when Za is attached to a nitrogen atom in Het or Het′.


A copper-mediated coupling reaction can be used to prepare compounds of Formula IIa, wherein R3a is —N(R8)—C(R6)—R4, —N(R8)—SO2-R4, or




embedded image



The reaction is conveniently carried out by combining the 1H-imidazo[4,5-c]quinolin-4-amine of the Formula XV and an amide, sulfonamide, or urea of formula HN(R8)-Q-R4 or




embedded image



in the presence of copper (I) iodide, potassium phosphate, and racemic trans-1,2-diaminocyclohexane in a suitable solvent such as 1,4-dioxane. The reaction can be carried out at an elevated temperature such as 110° C. Many amides, sulfonamides, and ureas are commercially available; others can be made by conventional methods. The compound or a pharmaceutically acceptable salt thereof can be isolated using conventional methods.


The compounds of Formula IIa prepared by the Heck reaction, can undergo reduction of the alkenylene group present in step (2) of Reaction Scheme I to provide compounds of Formula IIb. 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 IIb, a subgenus of Formulas I and II, or a pharmaceutically acceptable salt thereof can be isolated using conventional methods.




embedded image


For some embodiments, compounds of the invention can be prepared according to Reaction Scheme II, wherein R1, R2, R3a, R3b, R, Hal, and n are as defined above. In steps (1) and (2) of Reaction Scheme II, a 1H-imidazo[4,5-c]quinoline of Formula XVI undergoes a metal-mediated coupling reaction to provide a compound of Formula Xa, which may be reduced in step (2) to a compound of Formula Xb. Compounds of Formula Xa and Xb are subgenera of Formula X. Steps (1) and (2) of Reaction Scheme II can be carried out according to the methods described in steps (1) and (2) of Reaction Scheme I. Numerous compounds of Formula XVI are known; see, for example, U.S. Patent Publication Application No. US 2004/0147543 and the documents cited therein.


In steps (3a) and (3b) of Reaction Scheme II, 1H-imidazo[4,5-c]quinolines of Formula Xa and Xb are oxidized to provide 1H-imidazo[4,5-c]quinoline-5N-oxides of Formulas XVII and XVIII, respectively, using a conventional oxidizing agent capable of forming N-oxides. The reaction is conveniently carried out by adding 3-chloroperoxybenzoic acid at room temperature to a solution of a compound of Formula Xa or Xb in a solvent such dichloromethane or chloroform. Alternatively, the oxidation can be carried out by heating a solution of a compound of Formula Xa or Xb in a suitable solvent such as ethyl acetate with peracetic acid at a temperature such as 50° C. and then adding sodium metabisulfite.


In steps (4a) and (4b) of Reaction Scheme II, 1H-imidazo[4,5-c]quinoline-5N-oxides of Formulas XVII and XVIII are aminated to provide 1H-imidazo[4,5-c]quinolin-4-amines of Formulas IIa and IIb, respectively. The amination can be carried out by the activation of an N-oxide of Formula XVII or XVIII 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 followed by p-toluenesulfonyl chloride to a solution of the N-oxide of Formula XVII or XVIII in a suitable solvent such as chloroform or dichloromethane at room temperature. The reaction may also be carried out by adding ammonium hydroxide and p-toluenesulfonyl chloride to the reaction mixture from step (3a) or (3b) without isolating the N-oxide of Formula XVII or XVIII. The product or a pharmaceutically acceptable salt thereof can be isolated using conventional methods.


Alternatively step (4a) or (4b) can be carried out by the reaction of a 1H-imidazo[4,5-c]quinoline-5N-oxide of Formula XVII or XVIII with trichloroacetyl isocyanate followed by hydrolysis of the resulting intermediate to provide a compound of Formula IIa or IIb. The reaction is conveniently carried out in two steps by (i) adding trichloroacetyl isocyanate to a solution of the N-oxide of Formula XVII or XVIII in a solvent such as dichloromethane and stirring at room temperature to provide an isolable amide intermediate. In step (ii), a solution of the intermediate in methanol is treated with a base such as sodium methoxide or ammonium hydroxide at ambient temperature. The product or a pharmaceutically acceptable salt thereof can be isolated using conventional methods.


An R3a or R3b group in a compound of Formula Xa or Xb may contain a —S— functional group, which can be oxidized to —S(O)2— in step (3a) or (3b) of Reaction Scheme II using an excess of the oxidizing agent. Step (4a) or (4b) of Reaction Scheme II may then be carried out to provide a compound of Formula IIa or IIb, wherein R3a or R3b contains a —S(O)2— functional group.




embedded image


For some embodiments, compounds of the invention can be prepared according to Reaction Scheme III, wherein R1, R2, R4, R8, A, R, Za, Hal, a, b, and n are as defined above. In step (1) of Reaction Scheme III, a 1H-imidazo[4,5-c]quinolin-4-amine of Formula XV is coupled with an ester of formula H2C═C(H)—C(O)—Oalkyl to provide an ester-substituted 1H-imidazo[4,5-c]quinolin-4-amine of Formula XIX, a subgenus of Formulas I and II. The reaction can be carried out under the Heck reaction conditions described in step (1) of Reaction Scheme I. Some esters of formula H2C═C(H)—C(O)—O-alkyl, for example methyl acrylate, are commercially available; others can be prepared by known methods. The conditions described in step (2) of Reaction Scheme I may then be used to reduce the double bond in a compound of Formula XIX in step (2) of Reaction Scheme III to provide an ester-substituted 1H-imidazo[4,5-c]quinolin-4-amine of Formula XX, a subgenus of Formulas I and II.


In step (3) of Reaction Scheme III, the ester group of a 1H-imidazo[4,5-c]quinolin-4-amine of Formula XX undergoes base-promoted hydrolysis to the carboxylic acid of Formula XXI. The hydrolysis reaction is conveniently carried out by combining a compound of Formula XX with a base such as potassium hydroxide or sodium hydroxide in a suitable solvent mixture such as an alcohol/water mixture, preferably a methanol/water mixture. The reaction can be carried out at room temperature, and the product of Formula XXI, a subgenus of Formulas I and II, or a pharmaceutically acceptable salt thereof can be isolated by conventional methods.


A carboxylic acid of Formula XXI is converted into an amide of Formula IIg or IIh in step (4) or (4a) of Reaction Scheme III using conventional methods. The reaction is conveniently carried out by treating a solution of a carboxylic acid of Formula XXI with a secondary amine of formula HN(R8)R4 or




embedded image



and a coupling agent, such as 1-[3-(dimethylamino)propyl-3-ethylcarbodiimide hydrochloride, in the presence of 1-hydroxybenzotriazole. The reaction can be carried out at room temperature in a suitable solvent such as DMF, and the product of Formula IIg or IIh, which are subgenera of Formulas I and II, or a pharmaceutically acceptable salt thereof, can be isolated by conventional methods. Numerous secondary amines are commercially available; others can be prepared by known synthetic methods.




embedded image


For some embodiments, compounds of the invention can be prepared according to Reaction Scheme IV, wherein R1, R2, R4, R8, Q, R, Za, Hal, and n are as defined above; and R5a is




embedded image



wherein A, a, b, R7, and R8 are as defined above.


In step (1) of Reaction Scheme IV, a 1H-imidazo[4,5-c]quinoline of Formula XVI undergoes a Heck coupling reaction, according to the method described in step (1) of Reaction Scheme I, with an alkenyl-substituted phthalimide of formula




embedded image



which is commercially available or can be prepared by known methods. The resulting compound of Formula XXIII may then be reduced in step (2) according to the method described in step (2) of Reaction Scheme I. Compounds of Formula XXIII and XXIV are subgenera of Formula X and can be isolated by conventional methods.


In steps (3) and (4) of Reaction Scheme IV, a phthalimide-substituted 1H-imidazo[4,5-c]quinoline of Formula XXIV is first oxidized to a 5N-oxide of Formula XXV, which is then aminated to provide a phthalimide-substituted 1H-imidazo[4,5-c]quinolin-4-amine of Formula XXVI, a subgenus of Formulas I and II. Steps (3) and (4) of Reaction Scheme IV can be carried out according to the methods described in steps (3a) and (4a) of Reaction Scheme II, and the product or a pharmaceutically acceptable salt thereof can be isolated by conventional methods.


In step (5) of Reaction Scheme IV, the phthalimide group of a 1H-imidazo[4,5-c]quinolin-4-amine of Formula XXVI is removed to provide an amino-substituted 1H-imidazo[4,5-c]quinolin-4-amine of Formula XXVII, a subgenus of Formulas I and II. The reaction is conveniently carried out by adding hydrazine or hydrazine hydrate to a solution or suspension of a compound of Formula XXVI in a suitable solvent such as ethanol. The reaction can be carried out at room temperature or at an elevated temperature, such as the reflux temperature of the solvent. The product or a pharmaceutically acceptable salt thereof can be isolated by conventional methods.


In step (6) or (6a) of Reaction Scheme IV, an amino-substituted 1H-imidazo[4,5-c]quinolin-4-amine of Formula XXVII is converted to a 1H-imidazo[4,5-c]quinolinyl compound of Formula XXVIII or XXIX, subgenera of Formulas I and II, using conventional methods. For example, an amino-substituted 1H-imidazo[4,5-c]quinolin-4-amine of Formula XXVII can react with an acid chloride of Formula R4C(O)Cl to provide a compound of Formula XXVIII in which -QR4 is —C(O)—R4. In addition, a 1H-imidazo[4,5-c]quinolin-4-amine of Formula XXVII can react with sulfonyl chloride of Formula R4S(O)2Cl or a sulfonic anhydride of Formula (R4S(O)2)2O to provide a compound of Formula XXVIII in which -QR4 is —S(O)2—R4. Numerous acid chlorides of Formula R4C(O)Cl, sulfonyl chlorides of Formula R4S(O)2Cl, and sulfonic anhydrides of Formula (R4S(O)2)2O are commercially available; others can be readily prepared using known synthetic methods. The reaction is conveniently carried out by adding the acid chloride of Formula R4C(O)Cl, sulfonyl chloride of Formula R4S(O)2Cl, or sulfonic anhydride of Formula (R4S(O)2)2O to a solution of the amino-substituted 1H-imidazo[4,5-c]quinolin-4-amine of Formula XXVII in a suitable solvent such as chloroform, dichloromethane, or 1-methyl-2-pyrrolidinone. Optionally a base such as triethylamine, pyridine, or N,N-diisopropylethylamine, or a combination thereof can be added. The reaction can be carried out at room temperature or initially at a sub-ambient temperature such as 0° C. and then warming to room temperature. The product or a pharmaceutically acceptable salt thereof can be isolated using conventional methods.


Compounds of Formula XXIX where R5a is




embedded image



can be prepared by treating an amino-substituted 1H-imidazo[4,5-c]quinolin-4-amine of Formula XXVII with a chloroalkanesulfonyl chloride of Formula C1-R7S(O)2Cl or a chloroalkanoyl chloride of Formula C1-R7C(O)Cl. The reaction is conveniently carried out by adding the chloroalkanesulfonyl chloride or chloroalkanoyl chloride to a solution of the amino-substituted 1H-imidazo[4,5-c]quinolin-4-amine of Formula XXVII in a suitable solvent such as chloroform at ambient temperature. The isolable intermediate chloroalkanesulfonamide or chloroalkanamide can then be treated with a base such as 1,8-diazabicyclo[5.4.0]undec-7-ene in a suitable solvent such as DMF to effect the cyclization. The product or a pharmaceutically acceptable salt thereof can be isolated using conventional methods.


Ureas of Formula XXVIII, where -QR4 is —C(R6)—NH—W—R4, R6 is ═O, and W is a bond, can be prepared by reacting an amino-substituted 1H-imidazo[4,5-c]quinolin-4-amine of Formula XXVII with isocyanates of Formula R4N═C═O, Numerous isocyanates of Formula R4N═C═O are commercially available; others can be readily prepared using known synthetic methods. The reaction can be conveniently carried out by adding the isocyanate of Formula R4N═C═O to a solution of the amino-substituted 1H-imidazo[4,5-c]quinolin-4-amine of Formula XXVII in a suitable solvent such as dichloromethane or chloroform. Optionally a base such as triethylamine can be added. The reaction can be carried out at room temperature or initially at a sub-ambient temperature such as 0° C. and then warming to room temperature. Alternatively, a compound of Formula XXVII can be treated with an isocyanate of Formula R4(CO)N═C═O, a thioisocyanate of Formula R4N═C═S, a sulfonyl isocyanate of Formula R4S(O)2N═C═O, or a carbamoyl chloride of Formula R4N—(R8)—C(O)Cl, or




embedded image



to provide a compound of Formula XXVIII, wherein -QR4 is —C(R6)—N(R8)—W—, where R6, R8, and W are as defined above, or a compound of Formula XXIX wherein R5a is




embedded image



The product or a pharmaceutically acceptable salt thereof can be isolated using conventional methods.


Sulfamides of Formula XXVIII, where -QR4 is —S(O)2—N(R8)—R4 can be prepared by reacting a compound of Formula XXVII with sulfuryl chloride to generate a sulfamoyl chloride in situ, and then reacting the sulfamoyl chloride with an amine of formula HN(R8)R4. Alternatively, sulfamides of Formula XXVIII can be prepared by reacting a compound of Formula XXVII with a sulfamoyl chloride of formula R4(R8)N—S(O)2Cl. The product or a pharmaceutically acceptable salt thereof can be isolated using conventional methods. Many amines of Formula HN(R8)R4 and some sulfamoyl chlorides of formula R4(R8)N—S(O)2Cl are commercially available; others can be prepared using known synthetic methods.


Compounds of Formula XXVIII, wherein R8 is other than hydrogen can be prepared by reductive alkylation of the amino-substituted 1H-imidazo[4,5-c]quinolin-4-amine of Formula XXVII. The alkylation is conveniently carried out in two parts by (i) adding an aldehyde or ketone to a solution of an amino-substituted 1H-imidazo[4,5-c]quinolin-4-amine of Formula XXVII or a salt thereof in a suitable solvent such as DMF in the presence of a base such as N,N-diisopropylethylamine. In part (ii) the reduction is carried out by adding a suitable reducing agent such as the borane-pyridine complex. Both part (i) and part (ii) can be carried out at ambient temperature, and the product or a pharmaceutically acceptable salt thereof can be isolated using conventional methods. The resulting compound can undergo reaction with an acid chloride, sulfonyl chloride, sulfonic anhydride, isocyanate, or carbamoyl chloride as described above to provide a compound of Formula XXVIII, wherein R8 is other than hydrogen.




embedded image


For some embodiments, compounds of the invention can be prepared according to Reaction Scheme V, wherein R1, R2, Za, R, and n are as defined above; LG is a leaving group such as —Cl, —Br, —I, —O(SO)2CH3, or —O(SO)2CF3; R3c is -Zb-Y-R4, -Zb-Y-X-Y-R4, -Zb-R5, -Zb-Het, -Zb-Het′-R4, and -Zb-Het′-Y-R4, wherein Zb is selected from the group consisting of alkylene, alkenylene, and alkynylene interrupted with one or more —O— groups, and X, Y, R4, R5, Het, and Het′ are as defined above; and R3d is -Het, -Het′-R4, -Het′-Y-R4, —S—R4, —S(O)2—R4, or




embedded image



wherein Y, R4, R7, Q, Het, and Het′ are as defined above.


In step (1) of Reaction Scheme V, an ester-substituted compound of Formula XX is reduced to a hydroxyalkyl-substituted 1H-imidazo[4,5-c]quinolin-4-amine of Formula XXX using conventional methods. For example, the reduction may be carried out at room temperature with lithium borohydride in a suitable solvent such as THF. The product or a pharmaceutically acceptable salt thereof can be isolated by conventional methods.


In step (2a) of Reaction Scheme V, a hydroxy-substituted compound of Formula XXX is converted to an ether substituted 1H-imidazo[4,5-c]quinolin-4-amine of Formula XXXI, which is a subgenus of Formulas I and II, using a Williamson-type ether synthesis. The reaction is effected by treating a compound of Formula XXX with a halide of Formula Halide—R3c in the presence of a base. The reaction can be carried out by combining the halide with a compound of Formula XXX in a suitable solvent such as DMF in the presence of a base such as cesium carbonate. The reaction can be carried out at ambient temperature or at an elevated temperature, for example, 60 to 85° C. Alternatively, the reaction can be carried out by treating a solution of a compound of Formula XXX in a suitable solvent such as DMF with sodium hydride and then adding the halide.


In step (2) of Reaction Scheme V, a hydroxy-substituted compound of Formula XXX is converted to a leaving group in a compound of Formula XXXII using a variety of conventional methods. For example, a hydroxy-substituted compound of Formula XXX can be chlorinated to provide a compound of Formula XXXII wherein LG is —Cl. Conventional chlorinating reagents can be used. The reaction is conveniently carried out by combining a compound of Formula XXX with thionyl chloride in a suitable solvent such as dichloromethane and heating. Alternatively, the reaction may be run neat. A hydroxy-substituted compound of Formula XXX can also be treated with methanesulfonic anhydride to provide a compound of Formula XXXII wherein LG is —O(SO)2CH3.


In step (3) of Reaction Scheme V, the chloro group on a compound of Formula XXXII can be displaced by a thiol under basic conditions to provide a compound of Formula XXXIII wherein R3d is —S—R4. The reaction is conveniently carried out by adding a thiol to a solution of a compound of Formula XXXII in the presence of a base such as potassium tert-butoxide in a suitable solvent such as DMF. The product or a pharmaceutically acceptable salt thereof can be isolated by conventional methods. A compound of Formula XXXIII where R3d is —S—R4 can then be oxidized to a compound of Formula XXXIII where R3d is —S(O)2—R4 using conventional oxidizing agents. The reaction is conveniently carried out by adding peracetic acid to the compound of Formula XXXIII where R3d is —S—R4 in a suitable solvent. The conversion of a compound of Formula XXXII to a compound of Formula XXXIII where R3d is —S(O)2—R4 can conveniently be carried out in one pot without isolating the thioether from the reaction mixture. The product or a pharmaceutically acceptable salt thereof can be isolated by conventional methods.


The chloro group on a compound of Formula XXXII can also be displaced by an amine of Formula




embedded image



several of which are commercially available. Other amines of this formula can be prepared by conventional methods. The reaction is conveniently carried out by combining a compound of Formula XXXII with the amine in the presence of a base such as potassium carbonate and in a suitable solvent such as DMF. Catalytic sodium iodide can optionally be added. The reaction can be carried out at an elevated temperature such as 50° C. or 90-100° C., and the product can be isolated by conventional methods. These reaction conditions can also be used employing a variety of cyclic secondary amines, such as substituted or unsubstituted pyrrolidines, piperidines, or morpholines, to provide compounds of Formula XXXIII wherein R3d is -Het, -Het′-R4, or -Het′-Y-R4, wherein Het or Het′ is attached to the —CH2— group at a nitrogen atom.




embedded image


For some embodiments, compounds of the invention can be prepared according to Reaction Scheme VI wherein R1, R2, R3a, R3b, R, Hal, and n are as defined above. In step (1) of Reaction Scheme VI, an aminopyridine of Formula XXXIV 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 XXXV. The reaction is conveniently carried out by adding a solution of an aminopyridine of Formula XXXIV to a heated mixture of Meldrum's acid and triethyl orthoformate and heating the reaction at an elevated temperature such as 70° C.


In step (2) of Reaction Scheme VI, an imine of Formula XXXV undergoes thermolysis and cyclization to provide a [1,5]naphthyridin-4-ol of Formula XXXVI. 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.


In step (3) of Reaction Scheme VI, the [1,5]naphthyridin-4-ol of Formula XXXVI is nitrated under conventional nitration conditions to provide a 3-nitro[1,5]naphthyridin-4-ol of Formula XXXVII. The reaction is conveniently carried out by heating the [1,5]naphthyridin-4-ol of Formula XXXVI in nitric acid at an elevated temperature such as 90° C.


In step (4) of Reaction Scheme VI, a 3-nitro[1,5]naphthyridin-4-ol of Formula XXXVII is chlorinated using conventional chlorination chemistry to provide a 4-chloro-3-nitro[1,5]naphthyridine of Formula XXXVIII. The reaction is conveniently carried out by treating the 3-nitro[1,5]naphthyridin-4-ol of Formula XXXVII 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.


In step (5) of Reaction Scheme VI, a 4-chloro-3-nitro[1,5]naphthyridine of Formula XXXVIII is treated with an amine of Formula R1—NH2 to provide a 3-nitro[1,5]naphthyridin-4-amine of Formula XXXIX. 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 of Formula XXXIX in a suitable solvent such as dichloromethane or methanol 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.


In step (6) of Reaction Scheme VI, a 3-nitro[1,5]naphthyridin-4-amine of Formula XXXIX is reduced to provide a [1,5]naphthyridine-3,4-diamine of Formula XL. 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, or acetonitrile. The reaction can be carried out at ambient temperature.


Alternatively, the reduction in step (6) can be carried out using a one- or two-phase sodium dithionite reduction. The reaction is conveniently carried out using the conditions described by Park, K. K.; Oh, C. H.; and Joung, W. K.; Tetrahedron Lett., 34, pp. 7445-7446 (1993) by adding sodium dithionite to a compound of Formula XXXIX in a mixture of dichloromethane and water at ambient temperature in the presence of potassium carbonate and ethyl viologen dibromide, ethyl viologen diiodide, or 1,1′-di-n-octyl-4,4′-bipyridinium dibromide.


In step (7) of Reaction Scheme VI, a [1,5]naphthyridine-3,4-diamine of Formula XL is treated with a carboxylic acid equivalent to provide a 1H-imidazo[4,5-c][1,5]naphthyridine of Formula XLI. Suitable carboxylic acid equivalents include orthoesters of Formula R2C(O-alkyl)3, 1,1-dialkoxyalkyl alkanoates of Formula R2C(O-alkyl)2(O—C(O)-alkyl), and acid chlorides of Formula R2C(O)Cl. The selection of the carboxylic acid equivalent is determined by the desired substituent at R2. For example, triethyl orthoformate will provide a compound where R2 is hydrogen, and trimethyl orthovalerate will provide a compound where R2 is a butyl group. The reaction is conveniently carried out by adding the carboxylic acid equivalent to a [1,5]naphthyridine-3,4-diamine of Formula XL in a suitable solvent such as toluene or xylenes. Optionally, catalytic pyridine hydrochloride can be added. The reaction is carried out at a temperature high enough to drive off alcohol or water formed during the reaction. Conveniently, a Dean-Stark trap can be used to collect the volatiles.


Alternatively, step (7) can be carried out in two steps when an acid chloride of Formula R2C(O)Cl is used as the carboxylic acid equivalent. Part (i) of step (7) is conveniently carried out by adding the acid chloride to a solution of a [1,5]naphthyridine-3,4-diamine of Formula XL in a suitable solvent such as dichloromethane or acetonitrile to afford an amide. Optionally, a tertiary amine such as triethylamine, pyridine, or 4-dimethylaminopyridine can be added. The reaction can be carried out at room temperature. The amide product can be isolated and optionally purified using conventional techniques. Part (ii) of step (7) involves heating the amide prepared in part (i) to provide a 1H-imidazo[4,5-c][1,5]naphthyridine of Formula XLI. The reaction is conveniently carried out in a suitable solvent such as toluene at a temperature sufficient to drive off water formed during the reaction. The reaction can also be carried out in a solvent such as ethanol or methanol in the presence of a base such as triethylamine.


In steps (8) and (9) of Reaction Scheme VI, a 1H-imidazo[4,5-c][1,5]naphthyridine of Formula XLI is first oxidized to a 5N-oxide of Formula XLII, which is then aminated to provide a 1H-imidazo[4,5-c][1,5]naphthyridin-4-amine of Formula XLIII. Steps (8) and (9) of Reaction Scheme VI can be carried out according to the methods described in steps (3a) and (4a) of Reaction Scheme II.


In step (10) of Reaction Scheme VI a 1H-imidazo[4,5-c][1,5]naphthyridin-4-amine of Formula XLIII undergoes a metal-mediated coupling reaction to provide a compound of Formula IIIa, which may be reduced, when appropriate, in step (11) to a compound of Formula IIIb. Compounds of Formula IIIa and IIIb are subgenera of Formula III. Steps (10) and (11) of Reaction Scheme VI can be carried out according to the methods described in steps (1) and (2) of Reaction Scheme I, and the products or pharmaceutically acceptable salts thereof can be isolated by conventional methods.


Isomers of the compound of Formula XXXIV or Formula XXXVI can also be synthesized and can be used to prepare compounds of Formulas IV, V, and VI according to the methods shown in Reaction Scheme VI.


For some embodiments, compounds in Reaction Scheme VI can be further elaborated using conventional synthetic methods. For example, an amine of Formula R1—NH2 may be substituted by a hydroxy or second amino group, which may be further functionalized before step (7) of Reaction Scheme VI. Several examples of synthetic elaborations of an R1 group on an imidazo ring compound are known. See, for example, U.S. Patent Publication Application No. US 2004/0147543 and the documents cited therein.


Similar synthetic transformations can be made at R2 if, for example, the acid chloride used in step (7) of Reaction Scheme VI contains a protected hydroxy or amino group. Several acid chlorides of this type, for example acetoxyacetyl chloride, are commercially available. Others can be prepared by known synthetic methods. For examples of synthetic elaborations of an R2 group on an imidazo ring compound, see U.S. Pat. No. 5,389,640 (Gerster et al.).




embedded image


In some embodiments, compounds of the invention can be prepared according to Reaction Scheme VII, wherein R, R1, R2, R4, R8, Q, Hal, and n are as described above.


In step (1) of Reaction Scheme VII, the Heck reaction is used to couple a halogen substituted 1H-imidazo[4,5-c]quinoline of Formula XV with acrylonitrile to provide a 1H-imidazo[4,5-c]quinoline of Formula XLIV. The reaction can be carried out as described in step (1) of Reaction Scheme I.


In step (2) of Reaction Scheme VII, both the nitrile and the alkenylene groups in a 1H-imidazo[4,5-c]quinoline of Formula XLIV are reduced to provide an aminoalkyl substituted 1H-imidazo[4,5-c]quinoline of Formula XLV. The reduction can be carried out by hydrogenation using a conventional heterogeneous hydrogenation catalyst such as palladium on carbon. The reaction can be carried out on a Parr apparatus in a suitable solvent mix, such as methanol and trifluoroacetic acid.


In step (3) of Reaction Scheme VII, the amino group of a 1H-imidazo[4,5-c]quinoline of Formula XLV is further elaborated using the methods described in step (6) of Reaction Scheme IV to provide a 1H-imidazo[4,5-c]quinoline of Formula XLVI, which is a subgenus of Formulas I and II. The product or a pharmaceutically acceptable salt thereof can be isolated using conventional methods.




embedded image


In some embodiments, compounds of the invention can be prepared according to Reaction Scheme VIII, wherein R, R1, R2, and n are as described above.


In step (1) of Reaction Scheme VIII, the alkenylene bond in a 1H-imidazo[4,5-c]quinoline of Formula XLIV is reduced to provide a 1H-imidazo[4,5-c]quinoline of Formula XLVII. The reduction can be carried out as described in step (2) of Reaction Scheme I.


In step (2) of Reaction Scheme VIII, the nitrile group in a 1H-imidazo[4,5-c]quinoline of Formula XLVII is hydrolyzed to provide a 1H-imidazo[4,5-c]quinoline of Formula XLVIII, which is a subgenus of Formulas I and II The reaction can be carried out by treating a solution of a compound of Formula XLVII in a suitable solvent such as methanol with aqueous sodium hydroxide and aqueous hydrogen peroxide. The reaction can be carried out at an elevated temperature, such as for example, 50° C., and the product or a pharmaceutically acceptable salt thereof can be isolated using conventional methods.




embedded image


In some embodiments, compounds of the invention can be prepared according to Reaction Scheme IX, wherein R, R1, R2, R4, R8, Za, and n are as described above.


In step (1) of Reaction Scheme IX, the chloro group of a 1H-imidazo[4,5-c]quinoline of Formula XXXIIa, which is a subgenus of Formula XXXII wherein LG is chloro, is displaced with potassium thioacetate to provide a 1H-imidazo[4,5-c]quinoline of Formula XLIX. The reaction can be carried out at ambient temperature by adding potassium thioacetate to a solution of a compound of Formula XXXIIa in a suitable solvent such as DMF.


In step (2) of Reaction Scheme IX, the thioacetate group of a 1H-imidazo[4,5-c]quinoline of Formula XLIX is hydrolyzed under basic conditions to provide a thiol substituted 1H-imidazo[4,5-c]quinoline of Formula L. The reaction can be carried out by adding a solution of sodium methoxide in methanol to a solution of a compound of Formula XLIX in a suitable solvent such as methanol. The reaction can be carried out at ambient temperature.


In step (3) of Reaction Scheme IX, the thiol group of a 1H-imidazo[4,5-c]quinoline of Formula L is oxidized to provide a sulfonyl chloride substituted 1H-imidazo[4,5-c]quinoline of Formula LI. The reaction can be carried out by adding a solution of sodium chlorate in water to a solution of a compound of Formula L in hydrochloric acid. The reaction can be carried out at a sub-ambient temperature, such as for example, 0° C.


In step (4) of Reaction Scheme IX, a sulfonyl chloride substituted 1H-imidazo[4,5-c]quinoline of Formula LI is treated with an amine to provide a sulfonamide substituted 1H-imidazo[4,5-c]quinoline of Formula LII, which is a subgenus of Formulas I and II. The reaction can be carried out by adding an amine of Formula HN(R4)(R8) to a compound of Formula LI in a suitable solvent such as dichloromethane or pyridine. The reaction can be carried out at ambient temperature.




embedded image


In some embodiments, compounds of the invention can be prepared according to Reaction Scheme X, wherein R, R1, R2, R4, R8, Q, and n are as described above.


In step (1) of Reaction Scheme X, a vinyl substituted 1H-imidazo[4,5-c]quinoline of Formula LIII undergoes ozonolysis to provide an aldehyde substituted 1H-imidazo[4,5-c]quinoline of Formula LIV. The reaction can be carried out by bubbling ozone through a solution of a compound of Formula LIII in a suitable solvent such as dichloromethane and then quenching with triphenylphosphine. The reaction can be carried out at a sub-ambient temperature, such as 0° C. Some vinyl substituted 1H-imidazo[4,5-c]quinolines of Formula LIII are known; others can be prepared using known synthetic methods. See for example, U.S. Patent Application No. 2004/0147543 and the documents cited therein.


In step (2) of Reaction Scheme X, an aldehyde substituted 1H-imidazo[4,5-c]quinoline of Formula LIV is reduced to provide a hydroxy substituted 1H-imidazo[4,5-c]quinoline of Formula LV. The reaction can be carried out by treating a solution of a compound of Formula LIV in a suitable solvent such as THF with sodium borohydride. The reduction can be carried out at ambient temperature or at a sub-ambient temperature, such as 0° C.


In step (3) of Reaction Scheme X, a hydroxy substituted 1H-imidazo[4,5-c]quinoline of Formula LV is chlorinated using conventional methods to provide a chloro substituted 1H-imidazo[4,5-c]quinoline of Formula LVI. The reaction can be carried out by treating a solution of a compound of Formula LV in a suitable solvent such as dichloromethane with thionyl chloride.


In step (4) of Reaction Scheme X, the chloro group of a 1H-imidazo[4,5-c]quinoline of Formula LVI is displaced to provide an azido substituted 1H-imidazo[4,5-c]quinoline of Formula LVII. The reaction can be carried out by treating a solution of a compound of Formula LVI in a suitable solvent such as DMF with sodium azide.


In step (5) of Reaction Scheme X, the azido group of a 1H-imidazo[4,5-c]quinoline of Formula LVII is reduced to provide an aminomethyl substituted 1H-imidazo[4,5-c]quinoline of Formula LVIII. The reduction can be carried out using conventional methods such as, for example, catalytic hydrogenation.


In step (6) of Reaction Scheme X, the amino group of a 1H-imidazo[4,5-c]quinoline of Formula LVIII is further elaborated using the methods described in step (6) of Reaction Scheme IV to provide 1H-imidazo[4,5-c]quinoline of Formula LIX, which is a subgenus of Formulas I and II. The product or a pharmaceutically acceptable salt thereof can be isolated using conventional methods.




embedded image


For certain embodiments, compounds of the invention can be prepared according to Reaction Scheme XI, wherein RA, RB, R1, R2, 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(—NY′)—R′″, —CH(OH)—C(O)—OY′, —CH(OC1-4 alkyl)Y0, —CH2Y1, or —CH(CH3)Y1; 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 Y1 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 XVI 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.




embedded image


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 synthetic routes shown in Reaction Schemes II through V for the preparation of imidazoquinolines can be used to prepare imidazo[1,5]naphthyridines by using a compound of Formula XLIII or XLI in lieu of a compound of Formula XV or XVI. Also, the reduction shown in step (2) of Reaction Scheme III or IV may be eliminated to provide compounds of the invention, wherein Z contains a carbon-carbon double bond. 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 greata; 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
Example 1
2-Ethoxymethyl-1-(3-isopropoxypropyl)-7-[(E)-2-(phenylthio)ethenyl]-1H-imidazo[4,5-c]quinolin-4-amine



embedded image



Part A


7-Bromo-4-chloro-3-nitroquinoline (40 g) was dissolved in dichloromethane (1.4 L) and triethylamine (23.3 mL). 3-Isopropoxypropylamine (19.3 mL) was added dropwise. After 48 hours, the reaction mixture was washed successively with water and saturated aqueous sodium chloride. The organic fraction was dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. (7-Bromo-3-nitroquinolin-4-yl)-(3-isopropoxypropyl)amine was isolated as a tan solid (51.2 g).


Part B


(7-Bromo-3-nitroquinolin-4-yl)-(3-isopropoxypropyl)amine (51 g) was slurried in acetonitrile (750 mL) and added to a Parr flask containing 5% platinum on carbon (5 g). The flask was degassed three times, then charged with hydrogen (30 psi) and shaken for 4 hours with replenishment of the hydrogen as necessary. The platinum catalyst was removed by filtration through a bed of CELITE filter agent. The filtrate was evaporated to afford 7-bromo-N4-(3-isopropoxypropyl)quinoline-3,4-diamine as a yellow oil (45 g).


Part C


7-Bromo-N4-(3-isopropoxypropyl)quinoline-3,4-diamine (45 g) was dissolved in acetonitrile (1.3 L) and triethylamine (19.4 mL). Ethoxyacetyl chloride (18.0 g) was added dropwise to the solution and the reaction was stirred for 16 hours. The solvent was removed under reduced pressure to afford a tan solid. The solid was added to a solution of ethanol (1 L) and triethylamine (77.5 mL) and heated at reflux for 4 hours. The solvent was removed under reduced pressure. Water was added to the solid residue and the crude product was recovered by filtration. Recrystallization from acetonitrile yielded 36.25 g of 7-bromo-2-ethoxymethyl-1-(3-isopropoxypropyl)-1H-imidazo[4,5-c]quinoline as a tan crystalline solid.


Part D


7-Bromo-2-ethoxymethyl-1-(3-isopropoxypropyl)-1H-imidazo[4,5-c]quinoline (20 g) was dissolved in chloroform (400 mL). 3-Chloroperoxybenzoic acid (60% pure, 17.1 g) was added in 2 g portions over a 5 minute period and the reaction was stirred for 1 hour. Ammonium hydroxide (300 mL) was added and the mixture was cooled to 5° C. with an ice/water bath. p-Toluenesulfonyl chloride (9.4 g) was added at the rate of 1 g/min to minimize gas evolution. After stirring for 16 hours, the layers were separated and the aqueous fraction was extracted with chloroform. The combined organic fractions were sequentially washed with 5% aqueous sodium bicarbonate, water and brine; dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by flash column chromatography on silica gel. The polar component of the eluent was chloroform:methanol:ammonium hydroxide 80:18:2 (CMA). The purification was carried out eluting with chloroform:CMA in a gradient from 98:2 to 88:12. The material recovered from the column was recrystallized from acetonitrile to yield 7.0 g of 7-bromo-2-ethoxymethyl-1-(3-isopropoxypropyl)-1H-imidazo[4,5-c]quinolin-4-amine as a tan granular powder.


Part E


7-Bromo-2-ethoxymethyl-1-(3-isopropoxypropyl)-1H-imidazo[4,5-c]quinolin-4-ylamine (1.00 g, 2.46 mmol), tris(2-tolyl)phosphine (16 mg, 0.05 mmol) and a stir bar were added to a pressure vessel. Palladium(II) acetate (6 mg, 0.025 mmol), phenyl vinyl sulfide (0.330 mL, 2.53 mmol) and triethylamine (0.685 mL, 4.92 mmol) were subsequently added, followed by toluene (5 mL). The vessel was sealed with a TEFLON cap and the reaction was heated at 110° C. for 40 hours. The reaction mixture was cooled to ambient temperature and concentrated under reduced pressure. The residue was purified by flash column chromatography (silica gel, eluting with a linear gradient of 1-7% methanol in dichloromethane), followed by recrystallization from acetonitrile to provide 0.35 g of 2-ethoxymethyl-1-(3-isopropoxypropyl)-7-[(E)-2-(phenylthio)ethenyl]-1H-imidazo[4,5-c]quinolin-4-amine hydrobromide as yellow-orange crystals, mp 220-221° C.



1H NMR (300 MHz, DMSO-d6) δ 13.16 (s, 1H), 9.2-8.1 (br s, 2H), 8.29 (d, J=9.1 Hz, 1H), 7.93-7.73 (m, 2H), 7.57-7.32 (m, 6H), 6.90 (d, J=15.6 Hz, 1H), 4.83 (s, 2H), 4.72-4.67 (m, 2H), 3.64-3.51 (m, 5H), 2.15-2.02 (m, 2H), 1.18 (t, J=7.0 Hz, 3H), 1.14 (d, J=6.1 Hz, 6H);


HRMS (ESI) m/z 477.2323 (477.2324 calcd. for C27H32N4O2S, M+H);


Anal. Calcd. for C27H32N4O2S.HBr: C, 58.16; H, 5.97; N, 10.05; Br, 14.33. Found: C, 58.42; H, 6.49; N, 10.06; Br, 14.28.


Example 2
2-Ethoxymethyl-1-(3-isopropoxypropyl)-7-[(E)-2-(phenylsulfonyl)ethenyl]-1H-imidazo[4,5-c]quinolin-4-amine



embedded image



Part A


7-Bromo-2-ethoxymethyl-1-(3-isopropoxypropyl)-1H-imidazo[4,5-c]quinoline (1.00 g, 2.46 mmol), phenyl vinyl sulfone (0.852 g, 5.06 mmol), palladium(II) acetate (12 mg, 0.05 mmol), and tris(2-tolyl)phosphine (32 mg, 0.10 mmol) were added to a pressure tube. Triethylamine (1.37 mL, 9.84 mmol) and acetonitrile (5 mL) were added. The tube was flushed with nitrogen, sealed with a TEFLON plug and heated at 100° C. for 96 hours. The reaction mixture was cooled to ambient temperature and concentrated under reduced pressure. The residue was initially purified by flash column chromatography (silica gel, eluting with a gradient of 50%-100% ethyl acetate in hexanes, followed by a gradient of 2%-6% methanol in ethyl acetate). Recrystallization of the resulting solid from acetonitrile provided 0.600 g 7-(2-benzenesulfonylvinyl)-2-ethoxymethyl-1-(3-isopropoxypropyl)-1H-imidazo[4,5-c]quinoline as a yellow crystalline solid.


Part B


7-(2-Benzenesulfonylvinyl)-2-ethoxymethyl-1-(3-isopropoxypropyl)-1H-imidazo[4,5-c]quinoline (0.600 g, 1.22 mmol) was dissolved in chloroform (12 mL). 3-Chloroperoxybenzoic acid (60% purity, 0.351 g, 1.22 mmol) was added. The reaction was stirred for 30 minutes. Ammonium hydroxide (8 mL) was added and the mixture was stirred for 10 minutes. p-Toluenesulfonyl chloride (0.232 g, 1.22 mmol) was added in one portion and the reaction was stirred for an additional 16 hours. The layers were separated and the aqueous fraction was extracted with chloroform. The combined organic fractions were sequentially washed with water and saturated aqueous sodium chloride, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The residue 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). The polar component of the eluent was chloroform:methanol:ammonium hydroxide 80:18:2 (CMA). The purification was carried out with a silica cartridge eluting with a linear gradient of 1-22% CMA in chloroform. The resulting material was subsequently recrystallized from acetonitrile to yield 0.320 g 2-ethoxymethyl-1-(3-isopropoxypropyl)-7-[(E)-2-(phenylsulfonyl)ethenyl]-1H-imidazo[4,5-c]quinolin-4-amine as green needles, mp 188.5-190.0° C.



1H NMR (300 MHz, DMSO-d6) δ 8.20 (d, J=8.6 Hz, 1H), 7.98-7.92 (m, 3H), 7.81-7.60 (m, 6H), 6.76 (s, 2H), 4.78 (s, 2H), 4.67-4.62 (m, 2H), 3.63-3.44 (m, 5H), 2.13-1.98 (m, 2H), 1.16 (t, J=7.0 Hz, 3H), 1.15 (d, J=6.1 Hz, 6H);


HRMS (ESI) m/z 509.2218 (509.2223 calcd. for C27H32N4O4S, M+H);


Anal. Calcd. for C27H32N4O4S.0.8H2O: C, 62.02; H, 6.47; N, 10.72; S, 6.13. Found: C, 62.03; H, 6.52; N, 10.73; S, 6.12.


Example 3
N-[4-Amino-2-ethoxymethyl-1-(3-isopropoxypropyl)-1H-imidazo[4,5-c]quinolin-7-yl]-2-methylpropanamide



embedded image



Part A


7-Bromo-2-ethoxymethyl-1-(3-isopropoxypropyl)-1H-imidazo[4,5-c]quinoline (0.5 g, 1.23 mmol), potassium phosphate (0.548 g, 2.58 mmol), isobutyramide (0.128 g, 1.48 mmol), copper(I) iodide (46 mg, 0.246 mmol), and trans-(±)-1,2-diaminocyclohexane were added to a 2 dram vial. Dioxane (1.2 mL) and a stir bar were added and the vial was sealed with a TEFLON lined cap. The vial was heated at 110° C. for 16 hours. The reaction was cooled to ambient temperature and then diluted with water and chloroform. The layers were separated and the organic fraction was washed with water and saturated aqueous sodium chloride. The organic fraction was dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. Purification using a HORIZON HPFC system (silica cartridge, eluting with a linear gradient of 2-23% CMA in chloroform) provided 0.402 g of N-[2-ethoxymethyl-1-(3-isopropxypropyl)-1H-imidazo[4,5-c]quinolin-7-yl]-2-methylpropanamide as a yellowish waxy solid.


Part B


N-[2-ethoxymethyl-1-(3-isopropxypropyl)-1H-imidazo[4,5-c]quinolin-7-yl]-2-methylpropanamide (0.400 g, 0.98 mmol) was dissolved in chloroform (10 mL). 3-Chloroperoxybenzoic acid (60% purity, 0.532 g, 1.85 mmol) was added in two portions 30 minutes apart. The reaction was stirred for an additional 30 minutes. Ammonium hydroxide (10 mL) was added and the reaction was stirred for 10 minutes. Benzenesulfonyl chloride (0.236 mL, 1.8 mmol) was added in one portion and the reaction was stirred for 72 hours. The layers were separated and the aqueous fraction was extracted with chloroform. The combined organic fractions were sequentially washed with water and saturated aqueous sodium chloride, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. Purification using a HORIZON HPFC system (silica cartridge, eluting with a linear gradient of 2-25% CMA in chloroform), followed by recrystallization from acetonitrile provided 0.130 g of N-[4-amino-2-ethoxymethyl-1-(3-isopropxypropyl)-1H-imidazo[4,5-c]quinolin-7-yl]-2-methylpropanamide as a beige solid, mp 214-215° C.



1H NMR (300 MHz, DMSO-d6) δ 9.88 (s, 1H), 8.11 (d, J=8.9 Hz, 1H), 7.96 (d, J=2.1 Hz, 1H), 7.51 (dd, J=8.9, 2.1 Hz, 1H), 6.52 (s, 2H), 4.75 (s, 2H), 4.62-4.57 (m, 2H), 3.65-3.48 (m, 5H), 2.73-2.58 (m, 1H), 2.15-2.00 (m, 2H), 1.18-1.12 (m, 15H);



13C NMR (75 MHz, DMSO-d6) δ 175.2, 152.2, 148.3, 145.9, 138.3, 133.1, 120.7, 115.0, 114.0, 110.4, 70.7, 65.3, 64.0, 63.9, 42.8, 35.0, 30.3, 22.0, 19.5, 14.9;


MS (ESI) m/z 428.2657 (428.2662 calcd. for C23H33N5O3, M+H);


Anal. Calcd. for C23H33N5O3.0.25H2O: C, 63.94; H, 7.82; N, 16.21. Found: C, 64.02; H, 8.10; N, 16.25.


Example 4
N-[4-Amino-2-ethoxymethyl-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]ethanesulfonamide



embedded image


The general method described in Part A of Example 3 was followed using 7-bromo-2-ethoxymethyl-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-4-amine (U.S. Patent Application Publication No. US 2004/0147543, Examples 125-135) in lieu of 7-bromo-2-ethoxymethyl-1-(3-isopropoxypropyl)-1H-imidazo[4,5-c]quinoline and ethanesulfonamide in lieu of isobutyramide. Purification using a HORIZON HPFC system (silica cartridge, eluting with a linear gradient of 2-25% CMA in chloroform), followed by recrystallization from acetonitrile afforded 0.108 g of N-[4-amino-2-ethoxymethyl-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]ethanesulfonamide as white crystals, mp 124.0-127.0° C.



1H NMR (300 MHz, DMSO-d6) δ 9.85 (s, 1H), 8.21 (d, J=9.0 Hz, 1H), 7.44 (d, J=2.4 Hz, 1H), 7.11 (dd, J=8.9, 2.3 Hz, 1H), 6.56 (s, 2H), 5.05-4.73 (br s, 1H), 4.86 (s, 2H), 4.62 (s, 2H), 3.50 (q, J=7.0 Hz, 2H), 3.11 (q, J=7.3 Hz, 2H), 1.21 (t, J=7.3 Hz, 3H), 1.17 (br s, 6H), 1.13 (t, J=7.0 Hz, 3H);



13C NMR (125 MHz, DMSO-d6) δ 152.4, 150.4, 146.2, 136.9, 134.2, 125.4, 122.5, 114.7, 113.1, 111.6, 70.6, 65.2, 64.8, 54.7, 44.9, 27.5, 15.0, 8.0;


MS (APCI) m/z 422 (M+H)+;


Anal. Calcd. for C19H27N5O4S.0.25H2O: C, 53.57; H, 6.51; N, 16.44; S, 7.53. Found: C, 53.28; H, 6.72; N, 16.58; S, 7.42.


Example 5
N-{4-[4-Amino-2-ethoxymethyl-1-(3-isopropxypropyl)-1H-imidazo[4,5-c]quinolin-7-yl]butyl}acetamide



embedded image



Part A


7-Bromo-2-ethoxymethyl-1-(3-isopropoxypropyl)-1H-imidazo[4,5-c]quinoline (1.9 g, 4.68 mmol), 2-but-3-enyl-1H-isoindole-1,3(2H)-dione (1.04 g, 5.15 mmol), cesium carbonate (3.05 g, 9.36 mmol), triphenylphosphine (0.246 g, 0.94 mmol), palladium(II) acetate (0.105 g, 0.47 mmol), and N,N-dimethylformamide (DMF) (28 mL) were added to a pressure tube. The vessel was sealed with a TEFLON cap and heated at 140° C. for 2.5 hours. The reaction was cooled to ambient temperature. The reaction was diluted with ethyl acetate and washed multiple times with water. The initial water wash was extracted with ethyl acetate. The organic fractions were combined and washed with water once again. The organic fractions were concentrated under reduced pressure and the residue was purified using a HORIZON HPFC system (silica cartridge, eluting with a linear gradient of 2-20% CMA in chloroform) to yield 1.9 g of 2-{(3E)-4-[2-ethoxymethyl-1-(3-isopropoxypropyl)-1H-imidazo[4,5-c]quinolin-7-yl]but-3-enyl}-1H-isoindole-1,3(2H)-dione as a pale yellow oil.


Part B


2-{(3E)-4-[2-ethoxymethyl-1-(3-isopropoxypropyl)-1H-imidazo[4,5-c]quinolin-7-yl]but-3-enyl}-1H-isoindole-1,3(2H)-dione (1.9 g, 3.6 mmol) was dissolved in ethanol (50 mL) and added to a Parr flask containing 10% palladium on carbon (0.3 g) wetted with ethanol. The flask was degassed three times, charged with hydrogen (50 psi) and shaken for 16 hours. The catalyst was removed by filtration through CELITE filter agent. Concentration of the filtrate under reduced pressure provided 1.9 g of 2-{4-[2-ethoxymethyl-1-(3-isopropoxypropyl)-1H-imidazo[4,5-c]quinolin-7-yl]butyl}-1H-isoindole-1,3(2H)-dione as a pale yellow oil.


Part C


2-{4-[2-(ethoxymethyl)-1-(3-isopropoxypropyl)-H-imidazo[4,5-c]quinolin-7-yl]butyl}-1H-isoindole-1,3(2H)-dione (1.5 g, 2.84 mmol) was dissolved in ethyl acetate (15 mL) and heated to 50° C. 33% Peracetic acid in acetic acid (0.60 mL, 2.84 mmol) was added and the solution was stirred for 3 hours. Another 0.2 mL peracetic acid was added and the reaction was stirred for 1 hour. The mixture was cooled to ambient temperature and quenched with aqueous sodium metabisulfite (0.593 g in 1.2 mL water). The solution was made basic with saturated aqueous sodium carbonate. The mixture was diluted with water and the layers were separated. The aqueous fraction was extracted with ethyl acetate. The organic fractions were combined and concentrated under reduced pressure. The resulting yellow foam was dissolved in dichloromethane (15 mL) and ammonium hydroxide (10 mL). p-Toluenesulfonyl chloride (0.541 g, 2.84 mmol) was added in one portion. The mixture was stirred for 70 hours. The layers were separated and the aqueous fraction was extracted with dichloromethane. The organic fractions were combined and concentrated under reduced pressure to yield 0.525 g of 2-{4-[4-amino-2-ethoxymethyl-1-(3-isopropoxypropyl)-1H-imidazo[4,5-c]quinolin-7-yl]butyl}-1H-isoindole-1,3(2H)-dione as a yellow oil.


Part D


2-{4-[4-amino-2-ethoxymethyl-1-(3-isopropoxypropyl)-1H-imidazo[4,5-c]quinolin-7-yl]butyl}-1H-isoindole-1,3(2H)-dione (1.15 g, 2.1 mmol) was dissolved in ethanol (36 mL. Hydrazine (0.372 mL, 11.4 mmol) was added and the solution was heated at reflux temperature for 1 hour. The resulting white slurry was cooled to ambient temperature and filtered. The solid showed 40% product by NMR. The solid was saved and used in Part E. The filtrate was concentrated under reduced pressure and purified using a HORIZON HPFC system (silica cartridge, eluting with a linear gradient of 2-20% CMA in chloroform) to provide 0.211 g of 7-(4-aminobutyl)-2-ethoxymethyl-1-(3-isopropoxypropyl)-1H-imidazo[4,5-c]quinolin-4-amine as a white solid.


Part E


The crude mixture of 7-(4-aminobutyl)-2-ethoxymethyl-1-(3-isopropoxypropyl)-1H-imidazo[4,5-c]quinolin-4-amine from Part D was slurried in chloroform (7 mL) and triethylamine (0.185 mL, 1.32 mmol). Acetic anhydride (0.061 mL, 1.32 mmol) was added and the mixture was stirred for 16 hours. The resulting solution was concentrated under reduced pressure and the residue was purified using a HORIZON HPFC system (silica cartridge, eluting with a linear gradient of 1-20% CMA in chloroform). The diacylated product was dissolved in methanol (10 mL) and concentrated hydrochloric acid (2 mL). The mixture was heated to reflux for 2 hours and then cooled to ambient temperature. Saturated aqueous sodium carbonate was added to make the reaction basic. The methanol was evaporated under reduced pressure and the residue was extracted with dichloromethane (2×100 mL). The combined organic fractions were dried over sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified using a HORIZON HPFC system (silica cartridge, eluting with a linear gradient of 2-15% CMA in chloroform). Subsequent crystallization from ethyl acetate and hexanes provided 0.192 g of N-{4-[4-amino-2-ethoxymethyl-1-(3-isopropxypropyl)-1H-imidazo[4,5-c]quinolin-7-yl]butyl}acetamide as flocculent white crystals, mp 122-124° C.



1H NMR (300 MHz, DMSO-d6) δ 8.10 (d, J=8.4 Hz, 1H), 7.79 (t, J=4.8 Hz, 1H), 7.42 (d, J=1.1 Hz, 1H), 7.09 (dd, J=8.3, 1.3 Hz, 1H), 6.51 (s, 2H), 4.76 (s, 2H), 4.64-4.59 (m, 2H), 3.64-3.47 (m, 5H), 3.09-3.03 (m, 2H), 2.69 (t, J=7.4 Hz, 2H), 2.15-2.01 (m, 2H), 1.78 (s, 3H), 1.68-1.59 (m, 2H), 1.48-1.36 (m, 2H), 1.16 (t, J=7.0 Hz, 3H), 1.15 (d, J=6.2 Hz, 6H);



13C NMR (125 MHz, DMSO-d6) δ 168.8, 152.0, 148.5, 145.3, 140.7, 133.1, 125.8, 125.3, 122.0, 120.4, 112.6, 70.8, 65.4, 64.1, 63.9, 42.8, 38.3, 34.8, 30.3, 28.8, 28.3, 22.6, 22.0, 14.9;


MS (ESI) m/z 456 (M+H)+;


Anal. Calcd. for C25H37N5O3: C, 65.91; H, 8.19; N, 15.37. Found: C, 65.62; H, 7.94; N, 15.42.


Example 6
N-{4-[4-Amino-2-ethoxymethyl-1-(3-isopropxypropyl)-1H-imidazo[4,5-c]quinolin-7-yl]butyl}-N′-propylurea



embedded image


7-(4-Aminobutyl)-2-ethoxymethyl-1-(3-isopropoxypropyl)-1H-imidazo[4,5-c]quinolin-4-amine (0.250 g, 0.604 mmol) was dissolved in dichloromethane. Propyl isocyanate (0.060 mL, 0.604 mmol) was added and the reaction was stirred for 16 hours. A feathery solid was produced. The dichloromethane was removed under reduced pressure. The residue was diluted (not dissolved) in acetonitrile. This altered the solid to a more granular form. The solid was filtered, washed with acetonitrile, and dried to yield 0.090 g of N-{4-[4-Amino-2-ethoxymethyl-1-(3-isopropxypropyl)-1H-imidazo[4,5-c]quinolin-7-yl]butyl}-N′-propylurea as an off-white solid, mp 134.5-135.5° C. 1H NMR (300 MHz, DMSO-d6) δ 8.10 (d, J=8.4 Hz, 1H), 7.42 (s, 1H), 7.09 (dd, J=8.3, 1.2 Hz, 1H), 6.50 (s, 2H), 5.75 (t, J=5.4 Hz, 1H), 5.71 (t, J=5.7 Hz, 1H), 4.76 (s, 2H), 4.63-4.60 (m, 2H), 3.62-3.54 (m, 3H), 3.49 (t, J=5.5 Hz, 2H), 3.03-2.99 (m, 2H), 2.92 (q, J=6.5 Hz, 2H), 2.68 (t, J=7.5 Hz, 2H), 2.11-2.04 (m, 2H), 1.65-1.59 (m, 2H), 1.44-1.31 (m, 4H), 1.77-1.15 (m, 9H), 0.81 (t, J=7.4 Hz, 3H);



13C NMR (125 MHz, DMSO-d6) δ 158.0, 151.9, 148.5, 145.4, 140.8, 133.1, 125.8, 125.3, 122.1, 120.4, 112.5, 70.8, 65.3, 64.1, 63.9, 42.8, 41.0, 34.9, 30.3, 29.7, 28.3, 23.2, 22.0, 14.9, 11.3;


MS (APCI) m/z 499 (M+H)+;


Anal. Calcd. for C27H42N6O3: C, 65.03; H, 8.49; N, 16.85. Found: C, 64.90; H, 8.38; N, 16.84.


Example 7
2-Ethoxymethyl-1-(3-methoxypropyl)-7-methylsulfonyl-1H-imidazo[4,5-c]quinolin-4-amine



embedded image



Part A


7-Bromo-2-ethoxymethyl-1-(3-methoxypropyl)-1H-imidazo[4,5-c]quinoline (U.S. Patent Application Publication No. US 2004/0147543 Examples 163-175, 1.0 g, 2.64 mmol) and sodium thiomethoxide (0.185 g, 2.64 mmol) were dissolved in N,N-dimethylformamide (25 mL). The reaction mixture was stirred at ambient temperature for 20 minutes and then heated at 55° C. for 16 hours. The reaction was cooled to ambient temperature and the solvent was removed under reduced pressure. The residue was dissolved in dichloromethane and sequentially washed with water and saturated aqueous sodium chloride, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. 2-Ethoxymethyl-1-(3-methoxypropyl)-7-methylthio-1H-imidazo[4,5-c]quinoline was isolated as 1.1 g of a pale yellow oil.


Part B


The crude oil from Part A was dissolved in chloroform (20 mL). 3-Chloroperoxybenzoic acid (60% pure, 1.96 g, 6.84 mmol) was added in one portion. After 30 minutes, an additional 0.5 g of 3-chloroperoxybenzoic acid and 5 mL of chloroform was added. After 20 minutes, the reaction mixture was poured into a separatory funnel and washed with saturated aqueous sodium carbonate. The aqueous fraction was extracted sequentially with dichloromethane, chloroform, and 9:1 chloroform/methanol. The combined organic fractions were sequentially washed with water and saturated aqueous sodium chloride, dried over sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified using a HORIZON HPFC system (silica cartridge, eluting with a linear gradient of 2-30% CMA in chloroform). Fractions containing oxidized product were combined and concentrated. The residue was dissolved dichloromethane (15 mL) and diluted with ammonium hydroxide (10 mL). p-Toluenesulfonyl chloride was added to the mixture and the reaction was stirred for 120 hours. The layers were separated and the aqueous fraction was extracted with dichloromethane. The combined organic fractions were sequentially washed with water and saturated aqueous sodium chloride, dried over sodium sulfate, filtered, and concentrated under reduced pressure. Purification using a HORIZON HPFC system (silica cartridge, eluting with a linear gradient of 1-20% CMA in chloroform), followed by recrystallization from acetonitrile yielded 0.357 g of 2-ethoxymethyl-1-(3-methoxypropyl)-7-methylsulfonyl-1H-imidazo[4,5-c]quinolin-4-amine as white needles, mp 166.5-167.5° C.



1H NMR (300 MHz, DMSO-d6) δ 8.34 (d, J=8.7 Hz, 1H), 8.07 (d, J=2.0 Hz, 1H), 7.70 (dd, J=8.6, 1.9 Hz, 1H), 7.03 (s, 2H), 4.80 (s, 2H), 4.71-4.66 (m, 2H), 3.57 (q, J=7.0 Hz, 2H), 3.47 (t, J=5.6 Hz, 2H), 3.31 (s, 3H), 3.28 (s, 3H), 2.15-2.07 (m, 2H), 1.17 (t, J=7.0 Hz, 3H);



13C NMR (75 MHz, DMSO-d6) δ 153.3, 150.3, 144.6, 138.5, 132.3, 127.8, 124.9, 121.8, 117.8, 117.7, 68.6, 65.5, 64.0, 58.1, 43.7, 43.0, 29.7, 14.9;


MS (ESI) m/z 393.1602 (393.1597 calcd. for C18H24N4O4S, M+H);


Anal. Calcd. for C18H24N4O4S: C, 55.09; H, 6.16; N, 14.28; S, 8.17. Found: C, 55.12; H, 6.02; N, 14.32; S, 8.26.


Example 8
Methyl(2E)-3-[4-amino-2-butyl-1-(2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]prop-2-enoate



embedded image


A thick walled glass tube, equipped with stir bar, was charged with palladium (II) acetate (18 mg, 0.08 mmol), acetonitrile (2 mL), methyl acrylate (69 mg, 0.8 mmol), triethylamine (240 mg, 2.4 mmol), tri-o-tolylphosphine (49 mg, 0.16 mmol) and 7-bromo-2-butyl-1-(2-methylpropyl)-1H-imidazo[4,5-c]quinolin-4-amine (U.S. Patent Application Publication No. US 2004/0147543 Example 1, 300 mg, 0.8 mmol). The reaction mixture was purged with nitrogen and the tube was sealed and heated to 100° C. in an oil bath. The mixture became an amber homogeneous solution after approximately 5 minutes. The reaction was maintained at 100° C. for 32 hours and then concentrated to dryness. The reaction mixture was taken up in water and dichloromethane. Saturated aqueous potassium carbonate solution was added, adjusting to pH 11. The organic layer was separated and concentrated to a golden syrup. The crude product was purified by flash chromatography on silica gel (eluting with 5% methanol/dichloromethane). The resulting tan solid (230 mg) was recrystallized from acetonitrile to give 220 mg of Methyl(2E)-3-[4-amino-2-butyl-1-(2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]prop-2-enoate as a pale gold solid, mp 193-194° C.


MS (APCI) m/z 381.5 (M+H)+.


Example 9
Methyl(2E)-3-[4-amino-1-[4-(1,1-dioxidoisothiazolidin-2-yl)butyl]-2-(ethoxymethyl)-1H-imidazo[4,5-c]quinolin-7-yl]prop-2-enoate



embedded image


A 50-mL thick walled glass vessel was charged with palladium (II) acetate (67 mg, 0.3 mmol), acetonitrile (2 mL), methyl acrylate (0.26 g, 3.0 mmol), triethylamine (0.9 g, 9.0 mmol), tri-o-tolylphosphine (0.18 g, 0.6 mmol) and 7-bromo-1-[4-(1,1-dioxidoisothiazolidin-2-yl)butyl]-2-(ethoxymethyl)-1H-imidazo[4,5-c]quinolin-4-amine (U.S. Patent Application Publication No. US 2004/0147543 Example 152-156, 1.5 g, 3.0 mmol). An additional 13 mL of acetonitrile was added. The reaction mixture was purged with nitrogen then heated to 90° C. The turbid orange mixture was maintained at 90° C. overnight. Anhydrous DMF (30 mL) was added to dissolve the 7-bromoquinoline starting material. The reaction was then heated at 120° C. overnight. The reaction mixture was filtered to remove the catalyst then concentrated to dryness. The desired product was isolated by column chromatography on silica gel (eluting with a gradient of 2%-8% methanol in dichloromethane). The resulting peach solid was taken up in saturated potassium carbonate solution and dichloromethane and stirred overnight at ambient temperature. The organic layer was separated and concentrated to dryness. The residue was recrystallized from acetonitrile to yield 0.65 g of Methyl(2E)-3-[4-amino-1-[4-(1,1-dioxidoisothiazolidin-2-yl)butyl]-2-(ethoxymethyl)-1H-imidazo[4,5-c]quinolin-7-yl]prop-2-enoate as a pale yellow solid, mp 162-163° C.


MS (ACPI) m/z 502.2 (M+H)+.


Anal. Calcd. for C24H31N5O5S: C, 57.47; H, 6.23; N, 13.96. Found: C, 57.21; H, 6.38; N, 14.14.


Example 10
1-{3-[4-Amino-1-(2-methylpropyl)-1H-imidazo[4,5-c]quinolin-8-yl]propyl}pyrrolidin-2-one



embedded image



Part A


A thick walled glass tube, equipped with stir bar, was charged with palladium (II) acetate (67 mg, 0.3 mmol), acetonitrile (15 mL), N-allyl-2-pyrrolidone (650 mg, 5.17 mmol), triethylamine (2.1 ml, 15.65 mmol), tri-o-tolylphosphine (275 mg, 0.9 mmol) and 8-bromo-1-(2-methylpropyl)-1H-imidazo[4,5-c]quinolin-4-amine (U.S. Patent Application Publication No. US 2004/0147543 Example 11, 1.5 g, 4.7 mmol). The reaction mixture was purged with nitrogen, sealed and heated at 120° C. for 24 hours. The reaction mixture was concentrated to dryness. The reaction mixture was slurried in a mixture of 1% aqueous sodium carbonate (75 mL) and chloroform (100 mL). The layers were separated. The aqueous layer was extracted with chloroform (2×25 mL). The combined organic fractions were concentrated to dryness and purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with 0-8% methanol/dichloromethane) to provide 1-{(2E)-3-[4-amino-1-(2-methylpropyl)-1H-imidazo[4,5-c]quinolin-8-yl]prop-2-enyl}pyrrolidin-2-one.


Part B


A glass Parr vessel was charged with 10% palladium on carbon catalyst (0.1 g), methanol (50 mL), ethanol (50 mL) and 1-{(2E)-3-[4-amino-1-(2-methylpropyl)-1H-imidazo[4,5-c]quinolin-8-yl]prop-2-enyl}pyrrolidin-2-one. The vessel was evacuated and charged with hydrogen gas (51 psi, 3.4×105 Pa). The reaction was shaken at ambient temperature overnight (approximately 18 hours). The reaction mixture was filtered to remove the catalyst and concentrated to dryness. The crude product was purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with 0-8% methanol in dichloromethane) followed by recrystallization from acetonitrile to yield 300 mg of 1-{3-[4-amino-1-(2-methylpropyl)-1H-imidazo[4,5-c]quinolin-8-yl]propyl}pyrrolidin-2-one as a white solid, mp 174-175° C.


MS (APCI) m/z=366 (M+H)+;


Anal. Calcd. for C21H27N5O: C, 69.01; H, 7.45; N, 19.16. Found: C, 68.83; H, 7.71; N, 19.42.


Example 11
Methyl(2E)-3-[4-amino-2-(ethoxymethyl)-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]prop-2-enoate



embedded image


A thick walled glass reaction vessel was charged with anhydrous DMF (2 mL), palladium (II) Acetate (0.01 equivalents (eq)), tri-o-tolylphosphine (0.02 eq) and triethylamine (3 eq). To this orange solution was added a solution of 1-[4-amino-7-bromo-2-(ethoxymethyl)-1H-imidazo[4,5-c]quinolin-1-yl]-2-methylpropan-2-ol in 50 mL anhydrous DMF followed by the addition of methyl acrylate (1.0 eq). The reaction mixture was purged with nitrogen, sealed and heated at 120° C. overnight, and slowly cooled to room temperature. The reaction mixture was concentrated to dryness and then slurried in water (50 mL). Saturated potassium carbonate solution (25 mL) was added. The mixture was extracted with dichloromethane (4×50 mL). The combined extracts were concentrated under reduced pressure and purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with 0-25% CMA in chloroform) to provide Methyl(2E)-3-[4-amino-2-(ethoxymethyl)-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]prop-2-enoate as a white solid.


Example 12
Methyl 3-[4-amino-2-(ethoxymethyl)-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]propanoate



embedded image


A glass Parr vessel was charged with 10% palladium on carbon catalyst (0.35 g, 0.1 eq. weight/weight (w/w)), methanol (50 mL) ethanol (50 ml) and Methyl(2E)-3-[4-amino-2-(ethoxymethyl)-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]prop-2-enoate (3.5 g, 8.8 mmol). The vessel was evacuated and charged with hydrogen gas (45 psi, 3.1×105 Pa). The reaction was shaken at ambient temperature overnight (approximately 18 hours). The reaction mixture was filtered to remove the catalyst and concentrated to dryness. The crude product was purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with 0-30% CMA in chloroform) followed by recrystallization from acetonitrile to provide methyl 3-[4-amino-2-(ethoxymethyl)-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]propanoate as a white solid.


Example 13
3-[4-Amino-2-(ethoxymethyl)-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]propanoic acid



embedded image


A 100-mL round bottom flask was charged with methyl 3-[4-amino-2-(ethoxymethyl)-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]propanoate (0.28 g, 0.7 mmol) and Claisen's alkali (4.0 mL), and the reaction was stirred at ambient temperature for one hour and then concentrated to a colorless syrup. Citric acid solution (25 mL of 10% aqueous) was added. A white precipitate formed. The mixture was stirred for 15 minutes, and then the solid was collected by vacuum filtration to yield 0.25 g of 3-[4-amino-2-(ethoxymethyl)-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]propanoic acid.


Example 14
1-[4-Amino-2-(ethoxymethyl)-7-(3-morpholin-4-yl-3-oxopropyl)-1H-imidazo[4,5-c]quinolin-1-yl]-2-methylpropan-2-ol



embedded image


A 20-ml glass vial was charged with anhydrous DMF (1 mL), 3-[4-amino-2-(ethoxymethyl)-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]propanoic acid (165 mg, 0.43 mmol), N-(3-dimethylaminopropyl)-N″-ethylcarbodiimide hydrochloride (100 mg, 0.52 mmol, 1.3 eq), 1-hydroxybenzotriazole (70.3 mg, 0.52 mmol, 1.3 eq), and morpholine (110 mg, 1.29 mmol, 3.0 eq). The reaction was maintained at ambient temperature overnight. The reaction mixture was poured into water (10 mL), and the resulting mixture was extracted with dichloromethane (3×10 mL). The chloroform fractions were combined and concentrated, and the residue was purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with 0-30% CMA in chloroform) followed by recrystallization from acetonitrile to provide 1-[4-amino-2-(ethoxymethyl)-7-(3-morpholin-4-yl-3-oxopropyl)-1H-imidazo[4,5-c]quinolin-1-yl]-2-methylpropan-2-ol as a white solid, mp 118-119° C.


MS (APCI) m/z 456 (M+H)+.


Example 15
Methyl(2E)-3-[4-amino-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]prop-2-enoate



embedded image



Part A


To a suspension of 7-bromo-4-chloro-3-nitroquinoline (U.S. Patent Application Publication No. US 2004/0147543 Example 1, 54.2 g, 0.19 mol) in dichloromethane (1 L) was added triethylamine (100 mL, 0.72 mol., 3.8 eq) in one portion. To the resulting solution was slowly added hydroxyisobutylamine (18.44 g, 0.21 mol, 1.1 eq). The reaction was complete within two hours. The reaction mixture was concentrated to dryness, and the residue was slurried in a solution of 1% aqueous sodium carbonate (600 mL) for two hours. The resulting bright yellow solid was collected by vacuum filtration and dried on the filter funnel overnight to provide 57 g of 1-(7-bromo-3-nitroquinolin-4-ylamino)-2-methylpropan-2-ol.


Part B


A glass Parr vessel was charged with 5% platinum on carbon catalyst (5 g), 1-(7-bromo-3-nitroquinolin-4-ylamino)-2-methylpropan-2-ol (57 g, 0.17 mol), and a 1:1 solution of acetonitrile/toluene (1800 mL). The vessel was evacuated, charged with hydrogen gas (50 psi, 3.4×105 Pa), and shaken for 5 hours. The reaction mixture was then filtered through a 0.2 micron polytetrafluoroethylene (PTFE) membrane filter. The filtrate was concentrated to provide 49 g of 1-[(3-amino-7-bromoquinolin-4-yl)amino]-2-methylpropan-2-ol as a bright yellow-orange solid.


Part C


A 2-L round bottom flask was charged with 1-[(3-amino-7-bromoquinolin-4-yl)amino]-2-methylpropan-2-ol (25.5 g, 82.2 mmol), toluene (1 L), pyridine-HCl (0.1 g, 0.82 mmol, 0.01 eq) and triethylorthoformate (13.4 g, 90.4 mmol, 1.1 eq), and the reaction was heated at reflux for 2 hours. The reaction mixture was concentrated to dryness. The resulting pale yellow solid was slurried in 1% aqueous sodium carbonate solution (250 mL), collected by vacuum filtration, and air-dried on the funnel overnight to provide 25 g of 1-(7-bromo-1H-imidazo[4,5-c]quinolin-1-yl)-2-methylpropan-2-ol.


Part D


A 1-liter round bottom flask was charged with 1-(7-bromo-1H-imidazo[4,5-c]quinolin-1-yl)-2-methylpropan-2-ol (25.0 g, 78.0 mmol), dichloromethane (400 mL) and chloroform (100 mL). To this solution was slowly added in small portions 3-chloroperoxybenxzoic acid (34.0 g of 60% purity, 117 mmol, 1.5 eq). The reaction was maintained at room temperature for 2 hours and then concentrated ammonium hydroxide solution (200 mL) was added. The mixture was vigorously stirred as p-toluenesulfonyl chloride (22.4 g, 117 mmol) was slowly added in small portions. The reaction was complete within 1 hour but was left to stir at room temperature overnight. The reaction mixture was filtered to collect a pale yellow solid. The product was purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with 0-30% CMA in chloroform). The resulting pale yellow solid was slurried in cold acetonitrile, isolated by filtration, and air-dried on the vacuum filter to provide 17.2 g of 1-(4-amino-7-bromo-1H-imidazo[4,5-c]quinolin-1-yl)-2-methylpropan-2-ol. The filtrate was transferred to a separatory funnel and the layers were separated. The aqueous layer was extracted with chloroform (3×100 mL). The chloroform fractions were combined, washed with brine (100 mL), dried over magnesium sulfate and concentrated to dryness to provide an additional 0.8 g of 1-(4-amino-7-bromo-1H-imidazo[4,5-c]quinolin-1-yl)-2-methylpropan-2-ol, mp>250° C.


MS (ESI) m/z 335, m/z 337 (M+H)+;


Anal. Calcd. for C14H15BrN4O: C, 50.17; H, 4.51; N, 16.71. Found: C, 49.96; H, 4.44; N, 16.75.


Part E


A thick walled glass reaction vessel was charged with anhydrous DMF (2 mL), palladium (II) acetate (34 mg, 0.15 mmol, 0.005 eq), tri-o-tolylphosphine (91 mg, 0.3 mmol, 0.01 eq) and triethylamine (12.4 ml, 89.4 mmol, 3 eq). To this orange solution was added a solution of 1-(4-amino-7-bromo-1H-imidazo[4,5-c]quinolin-1-yl)-2-methylpropan-2-ol (10.0 g, 29.8 mmol) in 100 mL anhydrous DMF followed by the addition of methyl acrylate (2.82 g, 32.8 mmol, 1.1 eq). The reaction mixture was purged with nitrogen, sealed and heated at 120° C. overnight. The heat was turned off and the dark orange reaction mixture was slowly cooled to room temperature. The reaction mixture was poured into a solution of water (600 mL), saturated aqueous potassium carbonate (200 mL) and saturated aqueous sodium chloride 100 mL). An orange precipitate crashed out of solution. The solid was collected by vacuum filtration to provide 7.9 g of product. A small amount of product was purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with 20% methanol in dichloromethane:dichloromethane in a gradient from 1-15% over 300 mL, 15-40% over 1700 mL, and 40% for 1000 mL). The resulting material was recrystallized from 75:25 acetonitrile/methanol and dried to provide Methyl(2E)-3-[4-amino-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]prop-2-enoate, mp>250° C.


MS (APCI) m/z 341 (M+H)+;


Anal. Calcd. for C18H20N4O3 0.5 CH3OH: C, 62.35; H, 6.22; N, 15.72. Found: C, 62.24; H, 6.26; N, 16.10.


Example 16
Methyl 3-[4-amino-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]propanoate



embedded image


A Parr vessel was charged 10% palladium on carbon catalyst (3.0 g), and a slurry of Methyl(2E)-3-[4-amino-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]prop-2-enoate (7.5 g, 22.0 mmol) in 1:1 methanol/ethanol solution (200 mL). The vessel was evacuated and charged with hydrogen gas (50 psi, 3.4×105 Pa). The vessel was shaken for 3 days. The reaction was not complete. The reaction was charged with additional catalyst (1 g), placed under hydrogen pressure, and shaken overnight. The reaction was still not complete. The reaction was placed under hydrogen pressure and heated at 50° C. overnight. The reaction was now complete. The catalyst was removed by filtering the reaction mixture through a 0.2 micron PTFE membrane filter. The filtrate was concentrated to dryness. The residue was purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with a CMA:CHCl3 gradient: 0-8% over 200 mL, 3-30% over 3200 mL, and 30% for 2400 mL). The pure fractions were combined and concentrated to give one lot of product. The impure fractions were combined and concentrated, and the residue was slurried in acetonitrile, collected by vacuum filtration, and combined with the first lot to provide 2.7 g of white solid. A small portion of this material was further purified by chromatography using a HORIZON HPFC system (eluting with a 20% methanol/dichloromethane in dichloromethane gradient: 0-40% over 2400 mL). The pure fractions were combined, concentrated under reduced pressure, recrystallized from acetonitrile, filtered, and dried on the vacuum funnel to provide methyl 3-[4-amino-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]propanoate as a white solid, mp 201-205° C.


MS (ESI) m/z 343 (M+H)+;


Anal. Calcd. for C18H22N4O3: C, 63.14; H, 6.48; N, 16.36. Found: C, 63.02; H, 6.56; N, 16.37.


Example 17
Methyl(2E)-3-[4-amino-2-ethyl-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]prop-2-enoate



embedded image


A thick walled glass reaction vessel was charged with anhydrous DMF (2 mL), palladium (II) acetate (19 mg, 0.085 mmol, 0.005 eq), tri-o-tolylphosphine (51 mg, 0.17 mmol, 0.01 eq) and triethylamine (7.25 g, 52.02 mmol, 3 eq). To this orange solution was added a solution of 1-(4-amino-7-bromo-2-ethyl-1H-imidazo[4,5-c]quinolin-1-yl)-2-methylpropan-2-ol (U.S. Patent Application Publication No. US 2004/0147543 Example 142-144, 6.3 g, 17.34 mmol) in 100 mL anhydrous DMF followed by methyl acrylate (1.64 g, 19.08 mmol, 1.1 eq). The reaction mixture was purged with nitrogen, sealed and heated to 120° C. overnight. The heat was turned off and the dark orange reaction mixture was slowly cooled to room temperature. The reaction mixture was poured into a solution of water (600 mL), saturated aqueous potassium carbonate (200 mL) and saturated aqueous sodium chloride (100 mL). The mixture was transferred to a separatory funnel and extracted with chloroform. The chloroform fractions were combined and concentrated to dryness. The product was purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with a CMA:chloroform gradient: 0-5% over 200 mL, 5-30% over 3200 mL, and then 30% for 2400 mL) to provide approximately 4 g of product. A small portion was further purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with a 20% methanol/dichloromethane in dichloromethane gradient: 0-10% over 200 mL, 10-40% over 1650 mL, and then 40% for 500 mL) followed by recrystallization from acetonitrile/methanol to provide Methyl(2E)-3-[4-amino-2-ethyl-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]prop-2-enoate, mp 238-240° C.


MS (ESI) m/z 369 (M+H)+;


Anal. Calcd. for C20H24N4O3 1.0 CH3OH: C, 62.99; H, 7.05; N, 14.00. Found: C, 63.00; H, 7.12; N, 13.67.


Example 18
Methyl(2E)-3-[4-amino-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-8-yl]prop-2-enoate



embedded image



Part A


A 2-liter round bottom flask was charged with 1-(4-amino-1H-imidazo[4,5-c]quinolin-1-yl)-2-methylpropan-2-ol (50.0 g, 0.195 mol.) and DMF (1 L). To this stirred suspension was added a solution of N-bromosuccinimide (41.7 g, 0.234 mol, 1.2 eq) in DMF (200 mL). The reaction was stirred at ambient temperature overnight. The dark red solution was poured into 2% aqueous potassium carbonate solution (8 L). After 30 minutes the resulting mixture was filtered through a layer of CELITE filter agent followed by a rinse of 4 L of deionized water and 1 L of DMF. The aqueous filtrate was divided into five 2.5 L fractions and each was extracted with chloroform (3×600 mL), diluted with DMF (200 mL), and extracted again with chloroform (3×600 mL). The combined organic extracts were dried over magnesium sulfate, filtered and concentrated to dryness. After drying overnight under high vacuum, a reddish brown solid remained (50 g). This material was slurried in concentrated sodium bisulfite (200 mL) for 30 minutes and then filtered, rinsed with water, and dried on vacuum funnel overnight to provide 50 g of product. A small sample was purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with a gradient of chloroform/CMA; 0-8% over 200 mL, 8-20% over 2000 mL, 20-30% over 1000 mL, 30% over 2400 mL) followed by trituration with hot acetonitrile. The resulting bright yellow solid was further purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with a gradient of 20% methanol/dichloromethane in dichloromethane: 0-10% over 200 mL, 10-40% over 2400 mL, and 40% over 1200 mL) followed by recrystallization from acetonitrile/methanol to provide 0.65 g of 1-(4-amino-8-bromo-1H-imidazo[4,5-c]quinolin-1-yl)-2-methylpropan-2-ol as an off-white solid, mp 197-198° C.


MS (APCI) m/z 335, 337 (M+H)+;


Anal. Calcd. for C14H15BrN4O: C, 50.17; H, 4.51; N, 16.71. Found: C, 50.24; H, 4.57; N, 16.62.


Part B


The method described in Example 15 Part E can be used to couple 1-(4-amino-8-bromo-1H-imidazo[4,5-c]quinolin-1-yl)-2-methylpropan-2-ol with methyl acrylate to provide Methyl(2E)-3-[4-amino-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-8-yl]prop-2-enoate.


Examples 19-24

A solution of 7-bromo-2-ethoxymethyl-1-(3-isopropoxypropyl)-1H-imidazo[4,5-c]quinolin-4-amine (42 mg, 0.1 mmol, Example 1 Parts A-D) in 1,4-dioxane (1 mL) was added to a test tube containing copper iodide (8 mg), potassium phosphate (42 mg), and a reagent (0.12 mmol) from the table below and the tube was purged with nitrogen. Trans-1,2-diaminocyclohexane (7 μL) was added to the tube. The tube was purged with nitrogen and then heated with stirring in a sand bath at 110° C. for about 6 days. The reaction mixture was filtered and the filtrate was concentrated by vacuum centrifugation. The compounds were 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 preparative liquid chromatography 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 table below shows the reagent used for each example, the structure of the resulting compound, and the observed accurate mass for the isolated trifluoroacetate salt.














embedded image


















Measured


Example
Reagent
R3
Mass (M + H)





19
Methyl Carbamate


embedded image


416.2328





20
Methanesulfonamide


embedded image


436.2006





21
3-Methylbutanamide


embedded image


442.2820





22
(S)-(+)-2,2- Dimethylcyclopropanecarboxamide


embedded image


454.2854





23
3,4-Difluorobenzamide


embedded image


498.2300





24
Benzenesulfonamide


embedded image


498.2155









Examples 25 and 26

A solution of N-[4-(4-amino-7-bromo-2-ethoxymethyl-1H-imidazo[4,5-c]quinolin-yl)butyl]methanesulfonamide (50 mg, 0.1 mmol, U.S. Patent Application Publication No. US2004/0147543, examples 612-642) in 1,4-dioxane (1 mL) was added to a test tube containing copper iodide (19 mg), potassium phosphate (16 mg), and a reagent (0.2 mmol) from the table below and the tube was purged with nitrogen. Trans-1,2-diaminocyclohexane (18 μL) was added to the tube. The tube was purged with nitrogen, capped, and then heated with stirring in a sand bath at 110° C. for about 16 hours. The reaction mixture was filtered and the filtrate was concentrated by vacuum centrifugation. The compounds were purified using the method described in Examples 19-24. The table below shows the reagent used for each example, the structure of the resulting compound, and the observed accurate mass for the isolated trifluoroacetate salt.














embedded image















Ex-


Measured


ample
Reagent
R3
Mass (M + H)





25
Cyclopropanecarboxamide


embedded image


475.2153





26
1,1-Dimethylurea


embedded image


478.2241









Examples 27-41

A solution of 1-{3-[4-amino-8-(2-aminoethyl)-2-(2-methoxyethyl)-1H-imidazo[4,5-c]quinolin-1-yl]propyl}pyrrolidin-2-one (41 mg, 0.16 mmol, 1.0 eq) in a mixture of N,N-dimethylacetamide (1 mL) and N,N-diisopropylethylamine (34 μL) was added to a test tube containing a reagent from the table below (1.1 eq). The tube was vortexed overnight at ambient temperature and then the reaction was quenched with water (100 μL). The solvent was removed by vacuum centrifugation and the compound was purified using the method described in Examples 19-24. The table below shows the reagent used for each example, the structure of the resulting compound, and the observed accurate mass for the isolated trifluoroacetate salt.














embedded image


















Measured


Example
Reagent
R
Mass (M + H)





27
Acetyl chloride


embedded image


453.2643





28
Cyclopropanecarbonyl chloride


embedded image


479.2805





29
Isobutyryl chloride


embedded image


481.2954





30
Benzoyl chloride


embedded image


515.2795





31
Nicotinoyl chloride hydrochloride


embedded image


516.2712





32
Methanesulfonyl chloride


embedded image


489.2295





33
Isopropylsulfonyl chloride


embedded image


517.2607





34
Dimethylsulfamoyl chloride


embedded image


518.2574





35
Benzenesulfonyl chloride


embedded image


551.2482





36
1-Methylimidazole-4- sulfonyl chloride


embedded image


555.2527





37
Methyl isocyanate


embedded image


468.2766





38
Isopropyl isocyanate


embedded image


496.3044





39
Phenyl isocyanate


embedded image


530.2836





40
4- Morpholinylcarbonyl chloride


embedded image


524.3011





41
4-Methyl-1- Piperazinecarbonyl chloride


embedded image


537.3329









Examples 42-78

A solution of 1-[4-amino-7-(3-aminopropyl)-2-(methoxymethyl)-1H-imidazo[4,5-c]quinolin-1-yl]-2-methylpropan-2-ol (35 mg, 0.1 mmol, 1.0 eq) in a mixture of chloroform (1 mL), N,N-dimethylacetamide (about 80 μL), and N,N-diisopropylethylamine (36 μL) was added to a test tube containing a reagent from the table below (1.1 eq). The tube was vortexed overnight at ambient temperature and then the reaction was quenched with water (50 μL). The solvent was removed by vacuum centrifugation and the compound was purified using the method described in Examples 19-24. The table below shows the reagent used for each example, the structure of the resulting compound, and the observed accurate mass for the isolated trifluoroacetate salt.














embedded image















Ex-


Measured


am-


Mass


ple
Reagent
R
(M + H)





42
Propionyl chloride


embedded image


414.2485





43
Cyclopropanecarbonyl chloride


embedded image


426.2527





44
Butyryl chloride


embedded image


428.2683





45
Isobutyryl chloride


embedded image


428.2637





46
Cyclopentanecarbonyl chloride


embedded image


454.2795





47
Cyclohexanecarbonyl chloride


embedded image


468.2937





48
Dimethylaminoacetyl chloride hydrochloride


embedded image


443.2772





49
3-Methoxybenzoyl chloride


embedded image


492.2614





50
3-Chlorobenzoyl chloride


embedded image


496.2090





51
4-Chlorobenzoyl chloride


embedded image


496.2094





52
Methanesulfonyl chloride


embedded image


436.1988





53
Ethanesulfonyl chloride


embedded image


450.2188





54
1-Propanesulfonyl chloride


embedded image


464.2343





55
Isopropylsulfonyl chloride


embedded image


464.2345





56
Dimethylsulfamoyl chloride


embedded image


465.2298





57
1-Butanesulfonyl chloride


embedded image


478.2488





58
Trifluoromethanesulfonyl chloride


embedded image


490.1765





59
1-Methylimidazole-4- sulfonyl chloride


embedded image


502.2260





60
2,2,2- Trifluoroethanesulfonyl chloride


embedded image


504.1884





61
3- Methoxybenzenesulfonyl chloride


embedded image


528.2272





62
3-Chlorobenzenesulfonyl chloride


embedded image


532.1821





63
4-Chlorobenzenesulfonyl chloride


embedded image


532.1793





64
3-Pyridine sulfonyl chloride hydrochloride


embedded image


499.2130





65
Methyl isocyanate


embedded image


415.2461





66
Ethyl isocyanate


embedded image


429.2572





67
Isopropyl isocyanate


embedded image


443.2747





68
n-Propyl isocyanate


embedded image


443.2764





69
Phenyl isocyanate


embedded image


477.2596





70
Cyclohexyl isocyanate


embedded image


483.3073





71
3-Methoxyphenyl isocyanate


embedded image


507.2736





72
4-Methoxyphenyl isocyanate


embedded image


507.2714





73
3-Chlorophenyl isocyante


embedded image


511.2195





74
4-Chlorophenyl isocyante


embedded image


511.2211





75
1-Pyrrolidinecarbonyl chloride


embedded image


455.2779





76
1-Piperidinecarbonyl chloride


embedded image


469.2917





77
4-Morpholinylcarbonyl chloride


embedded image


471.2733





78
4-Methyl-1- piperazinecarbonyl chloride


embedded image


484.2990









Examples 79-95

A solution of 3-[4-amino-1-(2-hydroxy-2-methylpropyl)-2-(methoxymethyl)-1H-imidazo[4,5-c]quinolin-7-yl]propanoic acid (20 mg, 0.1 mmol, 1 eq) in 1:1 methanol:dichloromethane (2 mL) was placed in a test tube. The solvent was removed by vacuum centrifugation. A solution of 1-hydroxybenzotriazole (29 mg) in pyridine (1 mL) was added to the tube. The tube was sonicated for 15 minutes to provide a uniform suspension. A solution of N-(3-dimethylaminopropyl)-N′-ethylcarbodiimide (350 mg) in pyridine (100 μL) was added to the tube and the mixture was stirred for about 30 minutes. A reagent (2 eq) from the table below was added, the reaction mixture was stirred at ambient temperature overnight, and then the reaction was quenched with water (100 μL). The solvent was removed by vacuum centrifugation and the compound was purified using the method described in Examples 19-24. The table below shows the reagent used for each example, the structure of the resulting compound, and the observed accurate mass for the isolated trifluoroacetate salt.














embedded image


















Measured


Example
Reagent
R
Mass (M + H)





79
Isopropylamine


embedded image


414.2520





80
Cyclopentylamine


embedded image


440.2655





81
Piperidine


embedded image


440.2705





82
(R)-3- Hydroxypyrrolidine


embedded image


442.2455





83
3-Methoxypropylamine


embedded image


444.2607





84
2-Methylpiperidine


embedded image


454.2857





85
Tetrahydrofurfurylamine


embedded image


456.2646





86
3,5-Dimethylpiperidine


embedded image


468.2977





87
Thiophene-2-ethylamine


embedded image


482.2198





88
Nipecotamide


embedded image


483.2740





89
1-Acetylpiperazine


embedded image


483.2754





90
4-Piperidineethanol


embedded image


484.2943





91
1,1- Dioxidotetrahydrothien- 3-ylamine


embedded image


490.2134





92
1-(3-Aminopropyl)-2- pyrrolidinone


embedded image


497.2887





93
Methyl isonipecotate


embedded image


498.2765





94
1-Adamantanamine


embedded image


506.3137





95
Ethyl 2-amino-4- thiazoleacetate


embedded image


541.2277









Example 96
2-(4-Amino-1-isobutyl-1H-imidazo[4,5-c]quinolin-8-yl)ethanol



embedded image



Part A


8-Bromo-1-isobutyl-1H-imidazo[4,5-c]quinolin-4-amine (22.0 g, 68.9 mmol), triethylamine (19.20 mL, 137.8 mmol), dichloro[1,1′-bis(diphenylphosphino)ferrocene]palladium (II) dichloromethane (1.00 g, 1.37 mmol), and potassium vinyltrifluoroborate (10.15 g, 75.81 mmol) were dissolved in n-propanol (20 mL/g). The amber colored solution was heated at reflux temperature for 18 hours and then cooled to ambient temperature. The reaction was concentrated under reduced pressure. The resulting solid was slurried in 1% aqueous sodium carbonate and a fine powder was collected by filtration. The powder was sequentially washed with acetonitrile (5 mL/g) and 10% sodium hydroxide (500 mL). A final purification using a HORIZON HPFC system (silica cartridge, eluting with 4% methanol/dichloromethane) provided 10 g of 1-isobutyl-8-vinyl-1H-imidazo[4,5-c]quinolin-4-amine as an off-white solid, MS (APCI) m/z 267 (M+H)+.


Part B


A 500 mL round bottom flask was charged with 1-isobutyl-8-vinyl-1H-imidazo[4,5-c]quinolin-4-amine (850 mg, 3.2 mmol) and 200 mL of tetrahydrofuran. The flask was purged with nitrogen. A solution of 9-borabicyclo[3.3.1]nonane (0.5M in tetrahydrofuran, 8.5 mL, 4.25 mmol) was added in one portion and the reaction was stirred overnight. Additional 9-borabicyclo[3.3.1]nonane (0.5M in tetrahydrofuran, 12.5 mL, 6.25 mmol) was added and the mixture was stirred for approximately 24 hours. The reaction was cooled to 0° C. with an ice/water bath and 30% H2O2 (2 mL) was added dropwise. After 5 minutes, 10% NaOH (5 mL) was added dropwise. The reaction was stirred for 30 minutes, diluted with water (100 mL) and then extracted with chloroform (3×100 mL). The combined organic fractions were concentrated under reduced pressure and purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with a gradient of 0-30% CMA in chloroform) followed by recrystallization from acetonitrile to provide 580 mg of 2-(4-amino-1-isobutyl-1H-imidazo[4,5-c]quinolin-8-yl)ethanol as an off-white solid, mp 214-216° C.



1H NMR (500 MHz, DMSO-d6) δ 8.15 (s, 1H), 7.82 (m, 1H), 7.54 (d, J=8.4 Hz, 1H), 7.31 (dd, J=8.4, 1.4 Hz, 1H), 6.47 (br s, 2H), 4.68 (t, J=5.2 Hz, 1H), 4.39 (d, J=7.3 Hz, 2H), 3.71-3.68 (m, 2H), 2.87 (t, J=7.0 Hz, 2H), 2.18 (septet, J=6.8 Hz, 1H), 0.93 J=6.6 Hz, 6H);



13C NMR (125 MHz, DMSO-d6) δ 151.5, 143.3, 143.0, 132.1, 131.4, 128.1, 128.0, 125.8, 119.9, 114.5, 62.2, 53.3, 38.8, 28.3, 19.1;


MS (ESI) m/z 285 (M+H)+;


Anal. Calcd. for C16H20N4O: C, 67.58; H, 7.09; N, 19.70. Found: C, 67.51; H, 7.36; N, 19.74.


Example 97
3-[4-Amino-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]-1-(morpholin-4-yl)propan-1-one



embedded image



Part A


Methyl 3-[4-amino-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]propanoate (2.3 g, 6.7 mmol) was stirred in Claisen's alkali (50 mL) for 30 minutes. The solution was concentrated under reduced pressure. The resulting white solid was dissolved in water (200 mL) and washed with chloroform (4×50 mL). Citric acid was added to the aqueous fraction until the pH was 5-6. A fine white precipitate formed that was obtained by filtration to provide 2.5 g of wet 3-[4-amino-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]propanoic acid as a white solid. MS (ESI) m/z 329 (M+H)+.


Part B


3-[4-Amino-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]propanoic acid (500 mg, 1.5 mmol), anhydrous N,N-dimethylformamide (25 mL), and 1-hydroxybenzotriazole (250 mg, 1.8 mmol) were combined and the reaction was stirred for 15 minutes. 1-(3-Dimethylaminopropyl)-3-ethylcarbodiimide (350 mg, 1.8 mmol) was added and the reaction mixture was stirred for an additional 30 minutes. Morpholine (400 mg, 2.3 mmol) was added to the reaction slurry. After five hours 1-hydroxybenzotriazole (250 mg, 1.8 mmol), 1-(3-Dimethylaminopropyl)-3-ethylcarbodiimide (350 mg, 1.8 mmol), and morpholine (400 mg, 2.3 mmol) were added and the mixture was heated at 70° C. for 2 hours. After cooling to ambient temperature, the solvent was removed under reduced pressure and the residue was partitioned between chloroform (100 ml) and 1% aqueous sodium carbonate (50 mL). The fractions were separated and organic fraction was washed 1% aqueous sodium carbonate (2×50 mL). The combined aqueous fractions were back extracted with chloroform (50 mL). The combined organic fractions were dried (MgSO4), filtered, and concentrated under reduced pressure. The resulting off-white solid was purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with a CMA:chloroform gradient; 0-5% CMA over 75 mL, 5-35% over 1500 mL, and 35% for 600 mL) followed by recrystallization from acetonitrile to provide 335 mg of 3-[4-amino-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]-1-(morpholin-4-yl)propan-1-one as an off-white solid, mp 217-218° C.



1H NMR (500 MHz, DMSO-d6) δ 8.20 (d, J=8.4 Hz, 1H), 8.03 (s, 1H), 7.45 (d, J=1.3 Hz, 1H), 7.10 (dd, J=8.4, 1.5 Hz, 1H), 6.50 (br s, 2H), 4.83 (s, 1H), 4.51 (s, 2H), 3.51-3.40 (m, 8H), 2.92 (m, 2H), 2.69 (m, 2H), 1.16 (s, 6H);


MS (APCI) m/z 398 (M+H)+;


Anal. Calcd. for C21H27N5O3: C, 63.46; H, 6.85; N, 17.62. Found: C, 63.29; H, 6.97; N, 17.83.


Example 98
3-[4-Amino-2-ethyl-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]-1-(morpholin-4-yl)propan-1-one



embedded image


3-[4-Amino-2-ethyl-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]propanoic acid (500 mg, 1.4 mmol), chloroform (100 mL) and 1-hydroxybenzotriazole (575 mg, 4.26 mmol) were combined and then stirred for 30 minutes. 1-(3-Dimethylaminopropyl)-3-ethylcarbodiimide (1.00 g, 5.2 mmol) was added and the reaction was stirred for 30 minutes. Morpholine (10 mL) was added in one portion. After 4 hours, 1% aqueous sodium carbonate (100 mL) was added and the mixture was stirred. The fractions were separated and the organic fraction was sequentially washed with 1% aqueous sodium carbonate (50 mL), dried (MgSO4), filtered, and concentrated to a white solid. Recrystallization from acetonitrile provided 225 mg of 3-[4-amino-2-ethyl-1-(2-hydroxy-2-methyl-propyl)-1H-imidazo[4,5-c]quinolin-7-yl]-1-(morpholin-4-yl)propan-1-one as a white solid, mp 237-239° C.



1H NMR (500 MHz, DMSO-d6) δ 8.14 (d, J=8.5 Hz, 1H), 7.42 (d, J=1.5 Hz, 1H), 7.08 (dd, J=8.5, 1.7 Hz, 1H), 6.30 (br s, 2H), 4.76 (s, 1H), 4.51 (br s, 2H), 3.51-3.40 (m, 8H), 3.04 (quartet, J=7.4 Hz, 2H), 2.91 (m, 2H), 2.68 (m, 2H), 1.34 (t, J=7.4 Hz, 3H), 1.16 (br s, 6H);


MS (APCI) m/z 426 (M+H)+;


Anal. Calcd. for C23H31N5O3: C, 64.92; H, 7.34; N, 16.46. Found: C, 64.93; H, 7.31; N, 16.60.


Example 99
1-{4-Amino-7-[2-(methylsulfonyl)ethyl])-1H-imidazo[4,5-c]quinolin-1-yl}-2-methylpropan-2-ol



embedded image



Part A


A thick walled glass tube, equipped with a stir bar, was charged with palladium (II) acetate (15 mg, 0.07 mmol), acetonitrile (50 mL), methyl vinyl sulfone (268 mg, 2.52 mmol), triethylamine (1.00 mL, 7.17 mmol), tri-o-tolylphosphine (45 mg, 0.21 mmol) and 1-(4-amino-7-bromo-1H-imidazo[4,5-c]quinolin-1-yl)-2-methylpropan-2-ol (800 mg, 2.39 mmol). The reaction mixture was purged with nitrogen and the tube was sealed and heated at 120° C. in an oil bath. The reaction was maintained at 120° C. for 12 hours, cooled to ambient temperature, and then concentrated under reduced pressure to yield a dark brown oil. The oil was partitioned between chloroform (75 mL) and a solution comprised of a 1:1 combination of 10% aqueous potassium carbonate and 10% sodium hydroxide. A precipitate formed. The mixture was filtered and the recovered solid was purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with 20% methanol in dichloromethane. The filtrate was also concentrated and the residue purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with a 0-8% gradient of methanol in dichloromethane). The two lots of purified product were combined to provide 380 mg of 1-{4-amino-7-[2-(methylsulfonyl)ethenyl]-1H-imidazo[4,5-c]quinolin-1-yl}-2-methylpropan-2-ol as a pale yellow solid. MS (APCI) m/z 361 (M+H)+.


Part B


A glass Parr vessel was charged with 10% palladium on carbon (0.1 g), methanol (50 mL) and 1-{4-amino-7-[2-(methylsulfonyl)ethenyl]-1H-imidazo[4,5-c]quinolin-1-yl}-2-methylpropan-2-ol (380 mg, 1.05 mmol). The vessel was evacuated and charged with hydrogen gas (40 psi, 2.8×105 Pa). The reaction was shaken at 50° C. for approximately 18 hours. The reaction was cooled to ambient temperature, filtered, and then concentrated under reduced pressure. The resulting crude product was purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with a 0-35% gradient of CMA in chloroform) followed by recrystallization from acetonitrile to provide 176 mg of 1-{4-amino-7-[2-(methylsulfonyl)ethyl]-1H-imidazo[4,5-c]quinolin-1-yl}-2-methylpropan-2-ol as an off-white solid, mp 269-271° C.



1H NMR (500 MHz, DMSO-d6) δ 8.25 (d, J=8.5 Hz, 1H), 8.05 (s, 1H), 7.50 (d, J=1.5 Hz, 1H), 7.15 (dd, J=8.4, 1.7 Hz, 1H), 6.54 (br s, 2H), 4.83 (s, 1H), 4.52 (s, 2H), 3.52-3.49 (m, 2H), 3.13-3.10 (m, 2H), 3.00 (s, 3H), 1.16 (s, 6H);



13C NMR (125 MHz, DMSO-d6) δ 152.1, 145.2, 143.8, 136.3, 132.6, 127.1, 125.3, 121.5, 121.1, 113.6, 69.4, 56.1, 54.3, 40.1, 27.7, 27.0;


MS (APCI) m/z 363 (M+H)+;


Anal. Calcd. for C17H22N4O3S.0.33CH3CN: C, 56.42; H, 6.16; N, 16.13. Found: C, 56.23; H, 6.17; N, 16.16.


Example 100
3-[4-Amino-2-ethyl-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]-N,N-dimethylpropanamide



embedded image



Part A


A thick walled glass tube, equipped with a stir bar, was charged with palladium (II) acetate (50 mg, 0.22 mmol), acetonitrile (50 mL), triethylamine (1.80 mL, 13.0 mmol), tri-o-tolylphosphine (200 mg, 0.65 mmol), N,N-dimethylacrylamide (511 mg, 5.16 mmol) and 1-(7-bromo-2-ethyl-1H-imidazo[4,5-c]quinolin-1-yl)-2-methylpropan-2-ol (1.50 g, 4.30 mmol). The reaction mixture was purged with nitrogen and the tube was sealed and heated at 120° C. in an oil bath. The reaction was maintained at 120° C. for 18 hours and then cooled to ambient temperature. Upon cooling a precipitate formed that was dissolved by the addition of methanol and chloroform. The remaining insoluble material was removed by filtration through a 0.2 micron PTFE membrane filter. The filtrate was concentrated under reduced pressure and purified by chromatography using a HORIZON HPFC system (silica, eluting with a 0-25% gradient of CMA in chloroform). The resulting product was washed with 1% aqueous sodium carbonate and filtered to provide 1.37 g of 3-[2-ethyl-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]-N,N-dimethylprop-2-enamide as an off-white solid, mp 196-198° C.



1H NMR (500 MHz, DMSO-d6) δ 9.15 (s, 1H), 8.61 (d, J=8.9 Hz, 1H), 8.33 (d, J=1.3 Hz, 1H), 8.00 (dd, J=8.9, 1.5 Hz, 1H), 7.66 (d, J=15.4 Hz, 1H), 7.38 (d, J=15.4 Hz, 1H), 4.81 (s, 1H), 4.64 (br s, 2H), 3.21 (s, 3H), 3.10 (m, 2H), 2.96 (s, 3H), 1.39 (t, J=7.4 Hz, 3H), 1.20 (s, 6H);


MS (ESI) m/z 367 (M+H)+;


Anal. Calcd. for C21H26N4O2.H2O: C, 67.99; H, 7.20; N, 15.10. Found: C, 68.00; H, 7.25; N, 15.22.


Part B


A glass Parr vessel was charged with 10% palladium on carbon (0.14 g), methanol (50 mL) and 3-[2-ethyl-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]-N,N-dimethylprop-2-enamide (1.20 g, 3.27 mmol). The vessel was evacuated and charged with hydrogen gas (40 psi, 2.8×105 Pa). The reaction was shaken at 50° C. for approximately 19 hours. After cooling to ambient temperature, the reaction mixture was filtered and the filtrate concentrated to dryness to provide crude 3-[2-ethyl-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]-N,N-dimethylpropanamide as a white semi-solid. MS (APCI) m/z 369 (M+H)+.


Part C


To a stirring solution of crude 3-[2-ethyl-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]-N,N-dimethylpropanamide, (1.2 g, 3.3 mmol) in dichloromethane (75 mL) was added 3-chloroperoxybenzoic acid (60% pure, 941 mg, 3.6 mmol). After 18 hours concentrated ammonium hydroxide (50 mL) was added and the reaction was vigorously stirred for 10 minutes. p-Toluenesulfonyl chloride (690 mg, 3.6 mmol) was then added in one portion and the reaction was stirred for one hour. The fractions were separated and the aqueous fraction was extracted with chloroform (3×50 mL). The combined organic fractions were concentrated under reduced pressure. Purification of the residue by chromatography using a HORIZON HPFC system (silica cartridge, eluting with 0-6% gradient of methanol in dichloromethane) followed by recrystallization from acetonitrile provided 651 mg of 3-[4-amino-2-ethyl-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]-N,N-dimethylpropanamide as an off-white solid, mp 234-237° C.



1H NMR (500 MHz, DMSO-d6) δ 8.14 (d, J=8.5 Hz, 1H), 7.42 (d, J=1.6 Hz, 1H), 7.08 (dd, J=8.5, 1.7 Hz, 1H), 6.30 (br s, 2H), 4.76 (s, 1H), 4.51 (br s, 2H), 3.04 (q, J=7.4 Hz, 2H), 2.94 (s, 3H), 2.89 (m, 2H), 2.83 (s, 3H), 2.66 (m, 2H), 1.34 (t, J=7.4 Hz, 3H), 1.16 (br s, 6H);



13C NMR (75 MHz, DMSO-d6) δ 171.2, 155.5, 151.5, 144.9, 139.0, 133.5, 125.6, 125.1, 121.2, 120.8, 113.4, 70.6, 54.4, 36.5, 34.7, 33.9, 30.5, 27.5, 20.3, 12.0;


MS (ESI) m/z 384 (M+H)+;


Anal. Calcd. for C21H29N5O2: C, 65.77; H, 7.62; N, 18.26. Found: C, 65.76; H, 7.67; N, 18.35.


Example 101
1-{3-[4-Amino-2-(2-methoxyethyl)-8-(3-morpholin-4-yl-3-oxo-propyl)-1H-imidazo[4,5-c]quinolin-1-yl]propyl}pyrrolidin-2-one



embedded image



Part A


A thick walled glass tube, equipped with a stir bar, was charged with palladium (II) acetate (8 mg, 0.04 mmol), acetonitrile (50 mL), triethylamine (0.9 mL, 5.2 mmol), tri-o-tolylphosphine (30 mg, 0.10 mmol), 4-acryloylmorpholine (271 mg, 1.91 mmol) and 1-{3-[8-bromo-2-(2-methoxyethyl)-1H-imidazo[4,5-c]quinolin-1-yl]propyl}pyrrolidin-2-one (0.75 g, 1.7 mmol). The reaction mixture was purged with nitrogen and the tube was sealed and heated at 120° C. in an oil bath. The reaction was maintained at 120° C. for 15 hours and then cooled to ambient temperature. Upon cooling a precipitate formed that was dissolved by the addition of methanol and chloroform. The remaining insoluble material was removed by filtration through a 0.2 micron PTFE membrane filter. The filtrate was concentrated to dryness and purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with a 0-6% gradient of methanol in dichloromethane) to provide an off-white solid.


A glass Parr vessel was charged with 10% palladium on carbon (0.08 g), methanol (50 mL) and the solid. The vessel was evacuated and charged with hydrogen gas (40 psi, 2.8×105 Pa). The mixture was shaken at 50° C. for approximately 19 hours. After cooling to ambient temperature, the reaction mixture was filtered and the filtrate was concentrated to dryness to provide crude 1-{3-[2-(2-methoxyethyl)-8-(3-morpholin-4-yl-3-oxopropyl)-1H-imidazo[4,5-c]quinolin-1-yl]propyl}pyrrolidin-2-one as a white semi-solid. MS (ESI) m/z 494 (M+H)+.


Part B


To a stirring solution of crude 1-{3-[2-(2-methoxyethyl)-8-(3-morpholin-4-yl-3-oxopropyl)-1H-imidazo[4,5-c]quinolin-1-yl]propyl}pyrrolidin-2-one, (0.85 g) in dichloromethane (75 mL) was added 3-chloroperoxybenzoic acid (60% pure, 500 mg, 1.91 mmol). After 18 hours concentrated ammonium hydroxide (50 mL) was added and the reaction was vigorously stirred for 10 minutes. p-Toluenesulfonyl chloride (690 mg, 3.60 mmol) was added in one portion and the mixture was stirred for one hour. The fractions were separated and aqueous fraction was extracted with chloroform (3×50 mL). The combined organic fractions were concentrated under reduced pressure. Purification by chromatography using a HORIZON HPFC system (silica cartridge, eluting with a gradient of 0-30% CMA in chloroform) followed by recrystallization from acetonitrile provided 75 mg of 1-{3-[4-amino-2-(2-methoxyethyl)-8-(3-morpholin-4-yl-3-oxopropyl)-1H-imidazo[4,5-c]quinolin-1-yl]propyl}pyrrolidin-2-one as a pale yellow solid, mp 168-170° C.



1H NMR (300 MHz, DMSO-d6) δ 7.84 (s, 1H), 7.53 (d, J=8.5 Hz, 1H), 7.32 (dd, J=8.5, 1.5 Hz, 1H), 6.35 (br s, 2H), 4.53 (m, 2H), 3.81 (t, J=6.6 Hz, 2H), 3.50-3.33 (m, 12H), 3.29 (s, 3H), 3.18 (t, J=6.7 Hz, 2H), 2.98 (m, 2H), 2.72 (m, 2H), 2.23 (m, 2H), 2.05-1.87 (m, 4H);


MS (APCI) m/z 509 (M+H)+;


Anal. Calcd. for C27H36N6O4: C, 63.76; H, 7.13; N, 16.52. Found: C, 63.55; H, 6.87; N, 16.41.


Example 102
3-{4-Amino-2-(2-methoxyethyl)-1-[3-(2-oxo-pyrrolidin-1-yl)propyl]-1H-imidazo[4,5-c]quinolin-8-yl}-N,N-dimethylpropanamide



embedded image



Part A


A thick walled glass tube, equipped with a stir bar, was charged with palladium (II) acetate (8 mg, 0.04 mmol), acetonitrile (50 mL), triethylamine (0.9 mL, 5.22 mmol), tri-o-tolylphosphine (30 mg, 0.10 mmol), N,N-dimethylacrylamide (189 mg, 1.91 mmol) and 1-{3-[8-bromo-2-(2-methoxyethyl)-1H-imidazo[4,5-c]quinolin-1-yl]propyl}pyrrolidin-2-one (0.75 g, 1.7 mmol). The reaction mixture was purged with nitrogen and the tube was sealed and heated at 120° C. in an oil bath. The reaction was maintained at 120° C. for 18 hours and then cooled to ambient temperature. Upon cooling a precipitate formed that was dissolved by the addition of methanol and chloroform. The remaining insoluble material was removed by filtration through a 0.2 micron PTFE membrane filter. The filtrate was concentrated under reduced pressure and purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with a 0-7% gradient of methanol in dichloromethane) to provide 660 mg of 3-{2-(2-methoxyethyl)-1-[3-(2-oxopyrrolidin-1-yl)propyl]-1H-imidazo[4,5-c]quinolin-8-yl}-N,N-dimethylprop-2-enamide as an off-white solid.


Part B


A glass Parr vessel was charged with 10% palladium on carbon (0.07 g), methanol (50 mL) and 3-{2-(2-methoxyethyl)-1-[3-(2-oxopyrrolidin-1-yl)propyl]-1H-imidazo[4,5-c]quinolin-8-yl}-N,N-dimethylprop-2-enamide (0.66 g, 1.47 mmol). The vessel was evacuated and charged with hydrogen gas (40 psi, 2.8×105 Pa). The mixture was shaken at 50° C. for approximately 72 hours. The reaction mixture was cooled to ambient temperature and filtered. The filtrate was concentrated under reduced pressure to provide 720 mg of crude 3-{2-(2-methoxyethyl)-1-[3-(2-oxopyrrolidin-1-yl)propyl]-1H-imidazo[4,5-c]quinolin-8-yl}-N,N-dimethylpropanamide as a white semi-solid. MS (ESI) m/z 452 (M+H)+.


Part C


To a stirring solution of 3-{2-(2-methoxyethyl)-1-[3-(2-oxopyrrolidin-1-yl)propyl]-1H-imidazo[4,5-c]quinolin-8-yl}-N,N-dimethylpropanamide, (0.66 g) in dichloromethane (75 mL) was added 3-chloroperoxybenzoic acid (60% pure, 423 mg, 1.62 mmol). After 18 hours concentrated ammonium hydroxide (50 mL) was added and the reaction was vigorously stirred for 10 minutes. p-Toluenesulfonyl chloride (311 mg, 1.62 mmol) was then added in one portion. The fractions were separated and aqueous fraction was extracted with chloroform (3×50 mL). The combined organic fractions were concentrated under reduced pressure and purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with a 0-30% gradient of CMA in chloroform). The resulting product was washed with a 1% aqueous sodium carbonate and then recrystallized from acetonitrile to provide 75 mg of 3-{4-amino-2-(2-methoxyethyl)-1-[3-(2-oxopyrrolidin-1-yl)propyl]-1H-imidazo[4,5-c]quinolin-8-yl}-N,N-dimethylpropanamide as an off-white solid, mp 184-186° C.



1H NMR (300 MHz, DMSO-d6) δ 7.84 (d, J=1.0 Hz, 1H), 7.53 (d, J=8.5 Hz, 1H), 7.32 (dd, J=8.5, 1.5 Hz, 1H), 6.35 (br s, 2H), 4.53 (m, 2H), 3.81 (t, J=6.7 Hz, 2H), 3.42-3.34 (m, 4H), 3.30 (s, 3H), 3.18 (t, J=6.7 Hz, 2H), 3.00-2.92 (m, 5H), 2.82 (s, 3H), 2.70 (m, 2H), 2.23 (m, 2H), 2.06-1.86 (m, 4H);


MS (ESI) m/z 467 (M+H)+;


Anal. Calcd for C25H34N6O3: C, 64.36; H, 7.35; N, 18.01. Found: C, 64.47; H, 7.33; N, 18.11.


Example 103
1-{4-Amino-2-ethyl-7-[3-oxo-3-(1,3-thiazolidin-3-yl)propyl]-1H-imidazo[4,5-c]quinolin-1-yl}-2-methylpropan-2-ol



embedded image



Part A


A glass Parr vessel was charged with 10% palladium on carbon (1.0 g), methanol (75 mL), ethanol (75 mL) and methyl 3-[4-amino-2-ethyl-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]prop-2-enoate (approximately 4 g, 10.8 mmol). The vessel was evacuated and charged with hydrogen gas (40 psi, 2.8×105 Pa). The mixture was shaken at 50° C. for approximately 18 hours. The reaction mixture was cooled to ambient temperature and filtered. The filtrate was concentrated under reduced pressure to provide 3.55 g of methyl 3-[4-amino-2-ethyl-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]propanate as an off-white solid. MS (ESI) m/z 371 (M+H)+.


Part B


Methyl 3-[4-amino-2-ethyl-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]propanate was slurried in Claisen's alkali (50 mL) for 30 minutes and then concentrated to dryness. The resulting solid was dissolved in water (200 mL) and citric acid was slowly added until the pH reached 5-6. A fine off-white precipitate formed. Filtration provided 2.83 g of 3-[4-amino-2-ethyl-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]propanoic acid as an off-white solid. MS (ESI) m/z 357 (M+H)+.


Part C


A round bottom flask, equipped with a stir bar, was charged with 3-[4-amino-2-ethyl-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]propanoic acid (500 mg, 1.40 mmol), anhydrous pyridine (50 mL) and 1-hydroxybenzotriazole (379 mg, 2.80 mmol). After 30 minutes of stirring 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide (537 mg, 2.80 mmol) was added to the reaction mixture in two portions. After 15 minutes, thiazolidine (250 mg, 2.80 mmol) was added in one portion. The pale yellow solution was stirred for 18 hours and then concentrated under reduced pressure. The resulting foam was dissolved in dichloromethane and washed with 1% aqueous sodium carbonate (2×15 mL). The combined aqueous fractions were extracted with chloroform (3×30 mL). The combined organic fractions were purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with a 0-25% gradient of CMA in chloroform) followed by recrystallization from acetonitrile to provide 516 mg of 1-{4-amino-2-ethyl-7-[3-oxo-3-(1,3-thiazolidin-3-yl)propyl]-1H-imidazo[4,5-c]quinolin-1-yl}-2-methylpropan-2-ol as a white solid, mp 210-212° C.



1H NMR (300 MHz, DMSO-d6) δ 8.15 (d, J=8.5 Hz, 1H), 7.44 (d, J=1.7 Hz, 1H), 7.09 (dd, J=8.5, 1.8 Hz, 1H), 6.35 (br s, 2H), 4.76 (s, 1H), 4.60-4.44 (m, 4H), 3.72-3.63 (m, 2H), 3.08-2.88 (m, 6H), 2.73 (m, 2H), 1.34 (t, J=7.5 Hz, 3H), 1.17 (br s, 6H);


MS (ESI) m/z 428 (M+H)+;


Anal. Calcd. for C22H29N5O2S: C, 61.80; H, 6.84; N, 16.38. Found: C, 61.55; H, 6.92; N, 16.50.


Example 104
1-{4-Amino-2-(methoxymethyl)-7-[2-(methylsulfonyl)ethyl]-1H-imidazo[4,5-c]quinolin-1-yl}-2-methylpropan-2-ol



embedded image



Part A


A thick walled glass tube, equipped with a stir bar, was charged with palladium (II) acetate (12 mg, 0.05 mmol), acetonitrile (20 mL), N,N-dimethylformamide (15 mL) triethylamine (1.10 mL, 7.92 mmol), tri-o-tolylphosphine (48 mg, 0.16 mmol), methyl vinyl sulfone (294 mg, 2.77 mmol) and 1-[4-amino-7-bromo-2-(methoxymethyl)-1H-imidazo[4,5-c]quinolin-1-yl)-2-methylpropan-2-ol (1.00 g, 2.64 mmol). The reaction mixture was purged with nitrogen. The tube was sealed and heated at 120° C. in an oil bath. The reaction was maintained at 120° C. for 18 hours and then cooled to ambient temperature. The reaction was filtered and the filtrate was concentrated under reduced pressure. The resulting solid was partitioned between chloroform (100 mL) and aqueous 1% sodium carbonate (100 mL). The fractions were separated and the aqueous fraction was extracted with chloroform (5×50 mL). The combined organic fractions were concentrated and purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with 0-6% gradient of methanol in dichloromethane) to provide 750 mg of 1-{4-amino-2-(methoxymethyl)-7-[2-(methylsulfonyl)ethenyl]-1H-imidazo[4,5-c]quinolin-1-yl}-2-methylpropan-2-ol as a yellow semi-solid. MS (ESI) m/z 405 (M+H)+.


Part B


A glass Parr vessel was charged with 10% palladium on carbon (200 mg), methanol (12.5 mL), ethanol (12.5 mL) and 1-{4-amino-2-(methoxymethyl)-7-[2-(methylsulfonyl)ethenyl]-1H-imidazo[4,5-c]quinolin-1-yl}-2-methylpropan-2-ol (750 mg, 1.85 mmol). The vessel was evacuated and charged with hydrogen gas (40 psi, 2.8×105 Pa). The mixture was shaken at 50° C. for approximately 18 hours and then cooled to ambient temperature. The reaction mixture was sequentially filtered through a 0.2 micron PTFE membrane filter, concentrated under reduced pressure, and purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with 0-7% gradient of methanol in dichloromethane) to provide 195 mg of 1-{4-amino-2-(methoxymethyl)-7-[2-(methylsulfonyl)ethyl]-1H-imidazo[4,5-c]quinolin-1-yl}-2-methylpropan-2-ol as a white solid, mp 210-212° C.



1H NMR (500 MHz, DMSO-d6) δ 8.22 (d, J=8.5 Hz, 1H), 7.51 (d, J=1.7 Hz, 1H), 7.15 (dd, J=8.5, 1.8 Hz, 1H), 6.63 (br s, 2H), 5.20-4.50 (m, 5H), 3.51 (m, 2H), 3.30 (s, 3H), 3.12 (m, 2H), 3.00 (s, 3H), 1.16 (br s, 6H);



13C NMR (125 MHz, DMSO-d6) δ 151.9, 150.3, 145.2, 136.4, 134.1, 125.8, 125.3, 121.5, 121.1, 113.6, 70.5, 66.5, 57.6, 54.7, 54.2, 40.1, 27.6, 27.5;


MS (APCI) m/z 407 (M+H)+;


Anal. Calcd. for C19H26N4O4S.CH3CN: C, 56.35; H, 6.53; N, 15.56. Found: C, 56.10; H, 6.55; N, 15.56.


Example 105
1-{4-Amino-2-(hydroxymethyl)-7-[2-(methylsulfonyl)ethyl]-1H-imidazo[4,5-c]quinolin-1-yl}-2-methylpropan-2-ol



embedded image


A stirring solution of 1-{4-amino-2-(methoxymethyl)-7-[2-(methylsulfonyl)ethyl]-1H-imidazo[4,5-c]quinolin-1-yl}-2-methylpropan-2-ol (340 mg, 0.84 mmol) in dichloromethane (40 mL) was sealed with a septum and purged with nitrogen gas. The solution was cooled in an ice/water bath and a 1.0 M solution of boron tribromide in dichloromethane (4.2 mL) was added via syringe. The resulting mixture was stirred for 18 hours while warming to ambient temperature. The mixture was cooled back to 0° C. in an ice/water bath and the second portion of boron tribromide (1.0 M, 4.2 mL) was added. The reaction was stirred for 3 hours while warming to ambient temperature. Methanol (30 mL) was added and the mixture was concentrated to a purple foam. The foam was dissolved in 10% aqueous hydrochloric acid (30 ml) and washed with chloroform (2×25 mL). The aqueous fraction was neutralized by the slow addition of solid potassium carbonate until the pH reached 11. The aqueous fraction was extracted with chloroform (2×50 mL) and allowed to stand overnight. The resulting solid was recovered by filtration to provide 126 mg of 1-{4-amino-2-(hydroxymethyl)-7-[2-(methylsulfonyl)ethyl]-1H-imidazo[4,5-c]quinolin-1-yl}-2-methylpropan-2-ol


as a white crystalline solid, mp 221-223° C.



1H NMR (300 MHz, DMSO-d6) δ 8.21 (d, J=8.5 Hz, 1H), 7.49 (d, J=1.6 Hz, 1H), 7.14 (dd, J=8.5, 1.8 Hz, 1H), 6.51 (br s, 2H), 5.48 (br s, 1H), 5.12-4.77 (m, 3H), 4.69 (br s, 2H), 3.51 (m, 2H), 3.11 (m, 2H), 3.00 (s, 3H), 1.17 (br s, 6H);



13C NMR (125 MHz, DMSO-d6) δ 153.6, 152.0, 145.3, 136.1, 133.9, 125.7, 125.4, 121.4, 121.0, 113.8, 70.5, 56.5, 54.6, 54.3, 40.1, 27.6, 27.5;


MS (APCI) m/z 393 (M+H)+;


Anal. Calcd. for C18H24N4O4S.0.75H2O: C, 53.25; H, 6.33; N, 13.80. Found: C, 53.05; H, 6.51; N, 13.62.


Example 106
1-[4-Amino-2-(methoxymethyl)-7-(3-morpholin-4-yl-3-oxopropyl)-IH-imidazo[4,5-c]quinolin-1-yl]-2-methylpropan-2-ol



embedded image



Part A


A thick walled glass tube, equipped with a stir bar, was charged with palladium (II) acetate (150 mg, 0.66 mmol), acetonitrile (50 mL), N,N-dimethylformamide (20 mL) triethylamine (5.50 mL, 39.54 mmol), tri-o-tolylphosphine (600 mg, 1.98 mmol), methyl acrylate (294 mg, 2.77 mmol) and 1-[4-amino-7-bromo-2-(methoxymethyl)-1H-imidazo[4,5-c]quinolin-1-yl]-2-methylpropan-2-ol (5.00 g, 13.2 mmol). The reaction mixture was purged with nitrogen and the tube was sealed and heated at 120° C. for 18 hours. The reaction was cooled to ambient temperature and then concentrated to dryness under reduced pressure. The resulting solid was partitioned between chloroform (300 mL) and aqueous 1% sodium carbonate (100 mL). The fractions were separated and the aqueous fraction was extracted with chloroform (4×100 mL). The combined organic fractions were concentrated and purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with 0-8% gradient of methanol in dichloromethane) to provide a yellow semi-solid.


The intermediate product was added to a Parr vessel with 10% palladium on carbon (700 mg), methanol (12.5 mL), and ethanol (12.5 mL). The vessel was evacuated and charged with hydrogen gas (40 psi, 2.8×105 Pa). The mixture was shaken at 50° C. for approximately 18 hours followed by cooling to ambient temperature. The mixture was filtered through a 0.2 micron PTFE membrane filter and the filtrate was concentrated under reduced pressure to provide crude methyl 3-[4-amino-1-(2-hydroxy-2-methylpropyl)-2-(methoxymethyl)-1H-imidazo[4,5-c]quinolin-7-yl]propanoate as a white solid. MS (APCI) m/z 387 (M+H)+.


Part B


Methyl 3-[4-amino-1-(2-hydroxy-2-methylpropyl)-2-(methoxymethyl)-1H-imidazo[4,5-c]quinolin-7-yl]propanoate was slurried in Claisen's alkali (50 mL) for 30 minutes and then concentrated to dryness. The resulting solid was dissolved in water (200 mL) and citric acid was slowly added until the pH reached 5-6. A fine off-white precipitate formed. Filtration provided 7.58 g of wet 3-[4-amino-1-(2-hydroxy-2-methylpropyl)-2-(methoxymethyl)-1H-imidazo[4,5-c]quinolin-7-yl]propanoic acid as a white solid. MS (APCI) m/z 373 (M+H)+.


Part C


3-[4-Amino-1-(2-hydroxy-2-methylpropyl)-2-(methoxymethyl)-1H-imidazo[4,5-c]quinolin-7-yl]propanoic acid (1.25 g, 3.30 mmol), anhydrous pyridine (75 mL) and 1-hydroxybenzotriazole (892 mg, 6.60 mmol) were combined and the reaction was stirred for 30 minutes. 1-(3-Dimethylaminopropyl)-3-ethylcarbodiimide (1.27 g, 6.60 mmol) was added and the reaction mixture was stirred for an additional 15 minutes. Morpholine (581 mg, 6.60 mmol) was added in one portion. After stirring for 18 hours the solution was concentrated under reduced pressure. The resulting foam was dissolved in chloroform (50 mL) and washed with 1% aqueous sodium carbonate (2×15 mL). The combined aqueous washes were extracted with chloroform. The combined organic fractions were concentrated under reduced pressure and then purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with a CMA:chloroform gradient; 0-20% CMA over 1500 mL, followed by 20-25% CMA over 500 mL, and finally 25-30% CMA over 500 mL). A final recrystallization from acetonitrile provided 913 mg of 3-[4-amino-2-(methoxymethyl)-7-(3-morpholin-4-yl-3-oxopropyl)-1H-imidazo[4,5-c]quinolin-1-yl]-2-methylpropan-2-ol as an off-white solid, mp 215-217° C.



1H NMR (500 MHz, DMSO-d6) δ 8.15 (d, J=8.5 Hz, 1H), 7.45 (d, J=1.6 Hz, 1H), 7.11 (dd, J=8.5, 1.8 Hz, 1H), 6.55 (br s, 2H), 5.15-4.50 (br s, 2H), 4.89 (s, 1H), 4.63 (br s, 2H), 3.51-3.39 (m, 8H), 3.30 (s, 3H), 2.92 (t, J=7.6 Hz, 2H), 2.69 (t, J=7.6 Hz, 2H), 1.16 (br s, 6H);


MS (ESI) m/z 442 (M+H)+;


Anal. Calcd. for C23H31N5O4: C, 62.57; H, 7.08; N, 15.86. Found: C, 62.42; H, 6.90; N, 15.92.


Example 107
1-[4-Amino-7-[3-(1,1-dioxidothiomorpholin-4-yl)-3-oxopropyl]-2-(methoxymethyl)-1H-imidazo[4,5-c]quinolin-1-yl]-2-methylpropan-2-ol



embedded image


3-[4-Amino-1-(2-hydroxy-2-methylpropyl)-2-(methoxymethyl)-1H-imidazo[4,5-c]quinolin-7-yl]propanoic acid (1.25 g, 3.30 mmol), anhydrous pyridine (75 mL) and 1-hydroxybenzotriazole (892 mg, 6.60 mmol) were combined and the reaction was stirred for 30 minutes. 1-(3-Dimethylaminopropyl)-3-ethylcarbodiimide (1.27 g, 6.60 mmol) was added and the reaction mixture was stirred for an additional 15 minutes. Thiomorpholine 1,1-dioxide (895 mg, 6.60 mmol) was added in one portion. After stirring for 18 hours the solution was concentrated under reduced pressure and the residual foam was dissolved in chloroform (50 mL) and washed with 1% aqueous sodium carbonate (2×25 mL). The combined aqueous washes were extracted with chloroform. The combined organic fractions were concentrated under reduced pressure and purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with a CMA:chloroform gradient; 0-25% CMA over 1500 mL, followed by 25-30% CMA over 500 mL, finally 30% CMA over 1000 mL). The resulting material was subjected to a second chromatography purification using a HORIZON HPFC system (silica cartridge, eluting with CMA:chloroform gradient; 0-25% CMA over 1500 mL, followed by 25-30% CMA over 500 mL) followed by recrystallization from acetonitrile to provide 1.07 g of 1-[4-amino-7-[3-(1,1-dioxidothiomorpholin-4-yl)-3-oxopropyl]-2-(methoxymethyl)-1H-imidazo[4,5-c]quinolin-1-yl]-2-methylpropan-2-ol as a white solid, mp 190-192° C.



1H NMR (500 MHz, DMSO-d6) δ 8.16 (d, J=8.5 Hz, 1H), 7.46 (d, J=1.4 Hz, 1H), 7.11 (dd, J=8.5, 1.6 Hz, 1H), 6.53 (br s, 2H), 5.14-4.50 (br s, 2H), 4.88 (s, 1H), 4.64 (br s, 2H), 3.90-3.83 (m, 4H), 3.30 (s, 3H), 3.13 (m, 2H), 3.07 (m, 2H), 2.93 (t, J=7.5 Hz, 2H), 2.81 (t, J=7.5 Hz, 2H), 1.16 (br s, 6H);


MS (ESI) m/z 490 (M+H)+;


Anal. Calcd. for C23H31N5O5S: C, 56.43; H, 6.38; N, 14.30. Found: C, 56.44; H, 6.33; N, 14.29.


Example 108
3-[4-Amino-1-(2-hydroxy-2-methylpropyl)-2-(methoxymethyl)-1H-imidazo[4,5-c]quinolin-7-yl]-N,N-dimethylpropanamide



embedded image


3-[4-Amino-1-(2-hydroxy-2-methylpropyl)-2-(methoxymethyl)-1H-imidazo[4,5-c]quinolin-7-yl]propanoic acid (1.25 g, 3.30 mmol), anhydrous pyridine (75 mL) and 1-hydroxybenzotriazole (892 mg, 6.60 mmol) were combined and the reaction was stirred for 30 minutes. 1-(3-Dimethylaminopropyl)-3-ethylcarbodiimide (1.27 g, 6.60 mmol) was added and the reaction mixture was stirred for an additional 15 minutes. Dimethylamine hydrochloride (540 mg, 6.60 mmol) was added in one portion and the reaction was stirred for an additional 4 hours. The solution was concentrated under reduced pressure and the resulting oil was dissolved in chloroform (50 mL) and washed with 1% aqueous sodium carbonate (3×25 mL). The combined aqueous washes were extracted with chloroform. The combined organic fractions were purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with a CMA:chloroform gradient; 0-25% CMA over 1500 mL, followed by 25-30% CMA over 1000 mL) followed by recrystallization from acetonitrile to provide 931 mg of 3-[4-amino-1-(2-hydroxy-2-methylpropyl)-2-(methoxymethyl)-1H-imidazo[4,5-c]quinolin-7-yl]-N,N-dimethylpropanamide as a white solid, mp 142-145° C.



1H NMR (500 MHz, DMSO-d6) δ 8.16 (d, J=8.5 Hz, 1H), 7.45 (d, J=1.7 Hz, 1H), 7.11 (dd, J=8.5, 1.7 Hz, 1H), 6.56 (br s, 2H), 5.11-4.52 (br s, 2H), 4.89 (s, 1H), 4.64 (br s, 2H), 3.30 (s, 3H), 2.94 (s, 3H), 2.91 (t, J=7.7 Hz, 2H), 2.83 (s, 3H), 2.67 (t, J=7.7 Hz, 2H), 1.17 (br s, 6H);



13C NMR (125 MHz, DMSO-d6) δ 171.2, 151.9, 150.0, 145.4, 139.7, 134.1, 125.6, 125.1, 121.4, 121.0, 113.2, 70.5, 66.6, 57.5, 54.7, 36.5, 34.7, 33.9, 30.5, 27.5;


MS (APCI) m/z 400 (M+H)+;


Anal. Calcd. for C21H29N5O3.0.5H2O: C, 61.75; H, 7.40; N, 17.14. Found: C, 61.74; H, 7.47; N, 17.25.


Example 109
N-{3-[4-Amino-1-(2-hydroxy-2-methylpropyl)-2-(methoxymethyl)-1H-imidazo[4,5-c]quinolin-7-yl]propyl}methanesulfonamide



embedded image



Part A


A thick walled glass tube, equipped with a stir bar, was charged with palladium (II) acetate (225 mg, 1.00 mmol), acetonitrile (50 mL), N,N-dimethylformamide (20 mL) triethylamine (8.25 mL, 59.3 mmol), tri-o-tolylphosphine (900 mg, 2.97 mmol), acrylonitrile (770 mg, 21.8 mmol) and 1-[4-amino-7-bromo-2-(methoxymethyl)-1H-imidazo[4,5-c]quinolin-1-yl)-2-methylpropan-2-ol (7.50 g, 19.8 mmol). The reaction mixture was purged with nitrogen and the tube was sealed and heated at 120° C. in an oil bath for 18 hours. The reaction was cooled to ambient temperature and then concentrated under reduced pressure. The resulting solid was partitioned between chloroform (300 mL) and aqueous 1% sodium carbonate (100 mL). The fractions were separated and the aqueous fraction was extracted with chloroform (4×100 mL). The combined organic fractions were concentrated under reduced pressure and purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with 0-7% gradient of methanol in dichloromethane) to provide 5.2 g of 3-[4-amino-1-(2-hydroxy-2-methylpropyl)-2-(methoxymethyl)-1H-imidazo[4,5-c]quinolin-7-yl]prop-2-enenitrile as an off-white solid.


MS (ESI) m/z 352 (M+H)+.


Part B


A glass Parr vessel was charged with 10% palladium on carbon (1.0 g), methanol (200 mL), trifluoroacetic acid (5.4 mL, 72.5 mmol) and 3-[4-amino-1-(2-hydroxy-2-methylpropyl)-2-(methoxymethyl)-1H-imidazo[4,5-c]quinolin-7-yl]prop-2-enenitrile (5.1 g, 14.5 mmol). The vessel was evacuated and charged with hydrogen gas (40 psi, 2.8×105 Pa). The mixture was shaken for approximately 18 hours and then filtered through a 0.2 micron PTFE membrane filter and concentrated under reduced pressure. The resulting yellow oil was dissolved in 10% aqueous hydrochloric acid (100 mL) and stirred for 30 minutes. The solution was made basic (pH 10) by the slow addition of potassium carbonate. Potassium hydroxide was added until pH equaled 13. The solution was then concentrated under reduced pressure. The resulting off-white solid was slurried in hot methanol (300 mL) and filtered. The hot methanol slurry procedure was repeated. The combined filtrates were concentrated and purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with a CMA:chloroform gradient; 0-30% CMA) to provide 4.7 g of 1-[4-amino-7-(3-aminopropyl)-2-(methoxymethyl)-1H-imidazo[4,5-c]quinolin-1-yl]-2-methylpropan-2-ol as an off-white foam. MS (APCI) m/z 358 (M+H)+.


Part C


To a solution of 1-[4-amino-7-(3-aminopropyl)-2-(methoxymethyl)-1H-imidazo[4,5-c]quinolin-1-yl]-2-methylpropan-2-ol (1.25 g, 3.50 mmol) and triethylamine (2.4 mL, 17.5 mmol) cooled to 0° C. in a ice water bath was added methanesulfonic anhydride (732 mg, 4.20 mmol). The solution was stirred for approximately 18 hours while warming to ambient temperature. The solution was concentrated under reduced pressure and partitioned between 1% aqueous sodium carbonate (125 mL) and chloroform (100 mL). The aqueous fraction was extracted with chloroform until no product was present in the aqueous layer. The combined organic fractions were concentrated and purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with a CMA:chloroform gradient; 0-25% CMA over 1500 mL, followed by 25-30% CMA over 1000 mL). A final recrystallization from acetonitrile provided 855 mg of N-{3[4-amino-1-(2-hydroxy-2-methylpropyl)-2-(methoxymethyl)-1H-imidazo[4,5-c]quinolin-7-yl]propyl}methanesulfonamide as a white solid, mp 205-207° C.



1H NMR (500 MHz, DMSO-d6) δ 8.17 (d, J=8.5 Hz, 1H), 7.42 (d, J=1.7 Hz, 1H), 7.09 (dd, J=8.5, 1.7 Hz, 1H), 7.05 (t, J=5.6 Hz, 1H), 6.52 (br s, 2H), 5.11-4.57 (br s, 2H), 4.88 (s, 1H), 4.64 (br s, 2H), 3.30 (s, 3H), 2.99 m, 2H), 2.89 (s, 3H), 2.72 (t, J=7.6 Hz, 2H), 1.83 (m, 2H), 1.17 (br s, 6H);


MS (APCI) m/z 435 (M+H)+;


Anal. Calcd. for C20H29N5O4S.0.25H2O: C, 54.59; H, 6.75; N, 15.91. Found: C, 54.47; H, 6.73; N, 16.20.


Example 110
3-[4-Amino-1-(2-hydroxy-2-methylpropyl)-2-(methoxymethyl)-1H-imidazo[4,5-c]quinolin-7-yl]propanenitrile



embedded image


A glass Parr vessel was charged with 10% palladium on carbon catalyst (200 mg), methanol (100 mL), and 3-[4-amino-1-(2-hydroxy-2-methylpropyl)-2-(methoxymethyl)-1H-imidazo[4,5-c]quinolin-7-yl]prop-2-enenitrile (925 mg, 2.63 mmol). The vessel was evacuated and charged with hydrogen gas (40 psi, 2.8×105 Pa). The mixture was shaken at 50° C. for approximately 18 hours. The reaction mixture was cooled to ambient temperature and then filtered through a 0.2 micron PTFE membrane filter. The filtrate was concentrated under reduced pressure and purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with a CMA:chloroform gradient; 0-25% CMA over 1500 mL, followed by 25-30% CMA over 500 mL) to provide 760 mg of 3-[4-amino-1-(2-hydroxy-2-methylpropyl)-2-(methoxymethyl)-1H-imidazo[4,5-c]quinolin-7-yl]propanenitrile as a white solid, mp 191-193° C.



1H NMR (500 MHz, DMSO-d6) δ 8.22 (d, J=8.5 Hz, 1H), 7.52 (d, J=1.7 Hz, 1H), 7.14 (dd, J=8.5, 1.7 Hz, 1H), 6.62 (br s, 2H), 5.11-4.57 (br s, 2H), 4.89 (s, 1H), 4.65 (br s, 2H), 3.30 (s, 3H), 2.99 (t, J=7.0 Hz, 2H), 2.89 (t, J=7.0 Hz, 2H), 1.17 (br s, 6H);



13C NMR (125 MHz, DMSO-d6) δ 152.0, 150.2, 145.3, 137.0, 134.0, 125.8, 125.4, 121.4, 121.0, 120.3, 113.8, 70.5, 66.6, 57.5, 54.7, 30.4, 27.5, 17.9;


MS (APCI) m/z 354 (M+H)+;


Anal. Calcd. for C19H23N5O2.1.0 CH3CN: C, 63.94; H, 6.64; N, 21.31. Found: C, 63.66; H, 6.79; N, 21.12.


Example 111
3-[4-Amino-2-(hydroxymethyl)-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]-N,N-dimethylpropanamide



embedded image


A stirring solution of 3-[4-amino-1-(2-hydroxy-2-methylpropyl)-2-(methoxymethyl)-1H-imidazo[4,5-c]quinolin-7-yl]-N,N-dimethylpropanamide (523 mg, 1.31 mmol) in dichloromethane (125 mL) was sealed with a septum and purged with nitrogen gas. The solution was cooled in an ice/water bath and a 1.0 M solution of boron tribromide in dichloromethane (6.6 mL) was added via syringe. The resulting mixture was stirred for 18 hours while warming to ambient temperature. Methanol (50 mL) was added and the mixture was concentrated under reduced pressure. The resulting solid was dissolved in a 2.0 M solution of ammonia in methanol (75 mL) and the solution was concentrated under reduced pressure to yield a solid. This step was repeated two more times with silica gel (1 tbsp) being added prior to the final concentration. The sample absorbed on silica was purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with a CMA:chloroform gradient; 0-25% CMA over 1200 mL, followed by 25-40% CMA over 1300 mL) to provide 298 mg of 3-[4-Amino-2-(hydroxymethyl)-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]-N,N-dimethylpropanamide as a white crystalline solid, mp 228-230° C.



1H NMR (300 MHz, DMSO-d6) δ 8.15 (d, J=8.5 Hz, 1H), 7.42 (d, J=1.6 Hz, 1H), 7.10 (dd, J=8.5, 1.7 Hz, 1H), 6.43 (br s, 2H), 5.47 (t, J=5.9 Hz, 1H), 4.95 (s, 1H), 4.86 (br s, 2H), 4.68 (br s, 2H), 2.94 (s, 3H), 2.90 (m, 2H), 2.83 (s, 3H), 2.67 (m, 2H), 1.17 (br s, 6H);


MS (ESI) m/z 386 (M+H)+;


Anal. Calcd. for C20H27N5O3.0.5H2O: C, 60.90; H, 7.16; N, 17.75. Found: C, 60.93; H, 7.42; N, 17.85.


Example 112
1-[4-Amino-2-(hydroxymethyl)-7-(3-morpholin-4-yl-3-oxopropyl)-1H-imidazo[4,5-c]quinolin-1-yl]-2-methylpropan-2-ol



embedded image


A stirring solution of 1-[4-amino-2-(methoxymethyl)-7-(3-morpholin-4-yl-3-oxopropyl)-1H-imidazo[4,5-c]quinolin-1-yl]-2-methylpropan-2-ol (460 mg, 1.04 mmol) in chloroform (120 mL) was sealed with a septum and purged with nitrogen gas. The solution was cooled in an ice water bath and a solution of 1.0 M boron tribromide in dichloromethane (6.6 mL) was added via syringe. The reaction was stirred for 18 hours while warming to ambient temperature. In order to complete the reaction three additional portions (3 mL, 10 mL, and 10 mL) of 1.0 M boron tribromide in dichloromethane were added at approximately 18 hour intervals. Methanol (75 mL) was added. The reaction was concentrated to a dark purple liquid and then stirred in 10% aqueous hydrochloric acid (100 mL) for three days. Solid sodium carbonate was slowly added until the pH equaled 10. The solution was concentrated to a white solid. The solid was washed with methanol (2×100 mL) and filtered. The filtrate was absorbed onto silica (2 tbsp) and then purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with a CMA:chloroform gradient; 0-20% CMA over 1400 mL, followed by 20-30% CMA over 1600 mL) to provide 48 mg of 1-[4-amino-2-(hydroxymethyl)-7-(3-morpholin-4-yl-3-oxopropyl)-1H-imidazo[4,5-c]quinolin-1-yl]-2-methylpropan-2-ol as a white crystalline solid, mp 223-225° C.



1H NMR (500 MHz, DMSO-d6) δ 8.15 (d, J=8.5 Hz, 1H), 7.43 (d, J=1.3 Hz, 1H), 7.10 (dd, J=8.5, 1.5 Hz, 1H), 6.44 (br s, 2H), 5.47 (t, J=5.8 Hz, 1H), 4.96 (s, 1H), 4.87 (br s, 2H), 4.68 (br s, 2H), 3.52-3.48 (m, 2H), 3.45-3.38 (m, 6H), 2.92 (t, J=7.6 Hz, 2H), 2.69 (t, J=7.7 Hz, 2H), 1.17 (br s, 6H);


MS (ESI) m/z 428 (M+H)+;


Anal. Calcd. for C22H29N5O4: C, 61.81; H, 6.84; N, 16.38. Found: C, 61.62; H, 6.77; N, 16.26.


Example 113
3-[4-Amino-1-(2-hydroxy-2-methylpropyl)-2-(methoxymethyl)-1H-imidazo[4,5-c]quinolin-7-yl]propanamide



embedded image


3-[4-Amino-1-(2-hydroxy-2-methylpropyl)-2-(methoxymethyl)-1H-imidazo[4,5-c]quinolin-7-yl]propanenitrile (430 mg, 1.30 mmol) in methanol (75 mL) was heated to 50° C. in an oil bath. Aqueous 10% sodium hydroxide (0.33 mmol) and 30% aqueous hydrogen peroxide (516 mg, 4.55 mmol) were added and the reaction was stirred for 18 hours. The mixture was cooled to ambient temperature and then concentrated under reduced pressure. The resulting solid was slurried in water (70 mL). The aqueous mixture was washed with chloroform (35 mL), concentrated to a wet solid, slurried in methanol (100 mL) and filtered. The filtrate was absorbed onto silica (1 tbsp) and purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with CMA:chloroform gradient; 0-20% CMA over 1400 mL, followed by 20-25% CMA over 1200 mL). The material was further purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with a 0-9% gradient of methanol in dichloromethane) to provide 102 mg of 3-[4-amino-1-(2-hydroxy-2-methylpropyl)-2-(methoxymethyl)-1H-imidazo[4,5-c]quinolin-7-yl]propanamide as a white solid, mp 228-230° C.



1H NMR (500 MHz, DMSO-d6) δ 8.15 (d, J=8.4 Hz, 1H), 7.43 (d, J=1.3 Hz, 1H), 7.32 (s, 1H), 7.07 (dd, J=8.4, 1.4 Hz, 1H), 6.77 (s, 1H), 6.49 (br s, 2H), 5.11-4.63 (br s, 2H), 4.87 (s, 1H), 4.63 (br s, 2H), 3.30 (s, 3H), 2.90 (t, J=7.7 Hz, 2H), 2.43 (t, J=7.7 Hz, 2H), 1.16 (br s, 6H);


MS (ESI) m/z 372 (M+H)+;


Anal. Calcd. for C19H25N5O3: C, 61.44; H, 6.78; N, 18.85. Found: C, 61.27; H, 6.64; N, 18.74.


Example 114
1-[4-Amino-7-[3-(1,1-dioxidothiomorpholin-4-yl)-3-oxopropyl]-2-(hydroxymethyl)-1H-imidazo[4,5-c]quinolin-1-yl]-2-methylpropan-2-ol



embedded image


To a stirring solution of 1-[4-amino-7-[3-(1,1-dioxidothiomorpholin-4-yl)-3-oxopropyl]-2-(methoxymethyl)-1H-imidazo[4,5-c]quinolin-1-yl]-2-methylpropan-2-ol (655 mg, 1.34 mmol) in dichloromethane (125 mL), sealed with a septum and purged with nitrogen gas, was added a 1.0 M solution of boron tribromide in dichloromethane (6.6 mL) via syringe. The resulting mixture was stirred for 18 hours. Methanol (50 mL) was added and the solution was concentrated under reduced pressure. The resulting solid was dissolved in a 2.0 M solution of ammonia in methanol (75 mL) and then concentrated under reduced pressure to a solid. This step was repeated two more times with silica gel (1 tbsp) being added prior to the final concentration. The crude product absorbed on silica was purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with a CMA:chloroform gradient; 0-25% CMA over 1200 mL, followed by 25-40% CMA over 1300 mL). The recovered solid was washed with 1% aqueous sodium carbonate and filtered to provide 325 mg of 1-[4-amino-7-[3-(1,1-dioxidothiomorpholin-4-yl)-3-oxopropyl]-2-(hydroxymethyl)-1H-imidazo[4,5-c]quinolin-1-yl]-2-methylpropan-2-ol as an off white semi-solid.



1H NMR (500 MHz, DMSO-d6) δ 8.16 (d, J=8.5 Hz, 1H), 7.45 (d, J=1.4 Hz, 1H), 7.11 (dd, J=8.5, 1.6 Hz, 1H), 6.46 (br s, 2H), 5.48 (br s, 1H), 4.97 (s, 1H), 4.87 (br s, 2H), 4.68 (br s, 2H), 3.91-3.82 (m, 4H), 3.13 (m, 2H), 3.07 (m, 2H), 2.93 (t, J=7.7 Hz, 2H), 2.81 (t, J=7.7 Hz, 2H), 1.17 (br s, 6H);


MS (ESI) m/z 476 (M+H)+;


Anal. Calcd. for C22H29N5O5S.0.5H2O: C, 54.23; H, 6.24; N, 14.45. Found: C, 54.17; H, 6.14; N, 14.68.


Example 115
N-{3-[4-Amino-1-(2-hydroxy-2-methylpropyl)-2-(methoxymethyl)-1H-imidazo[4,5-c]quinolin-7-yl]propyl}acetamide



embedded image


A solution of 1-[4-amino-7-(3-aminopropyl)-2-(methoxymethyl)-1H-imidazo[4,5-c]quinolin-1-yl]-2-methylpropan-2-ol (1.25 g, 3.50 mmol) in triethylamine (2.4 mL, 17.5 mmol) was cooled to 0° C. in an ice water bath and acetic anhydride (393 mg, 3.85 mmol) was added. The solution was stirred for approximately 18 hours while warming to ambient temperature. The solution was concentrated under reduced pressure and the residue was partitioned between 1% aqueous sodium carbonate (125 mL) and chloroform (50 mL). Successive chloroform extractions were performed until no product was present in the aqueous layer. The combined organic fractions were concentrated under reduced pressure and dissolved in methanol (100 mL). To this solution was added solid potassium carbonate (3 tsp) and the reaction was heated to reflux temperature for 2 hours. After cooling to ambient temperature, the mixture was filtered, concentrated and partitioned between water (50 mL) and chloroform (50 mL). Crude product was extracted from the aqueous fraction through successive chloroform extractions until no product remained in the aqueous layer. The combined organic extracts were concentrated and purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with a CMA:chloroform gradient; 0-30% CMA over 1500 mL, followed by 30% CMA over 1000 mL) to provide 578 mg of N-{3-[4-amino-1-(2-hydroxy-2-methylpropyl)-2-(methoxymethyl)-1H-imidazo[4,5-c]quinolin-7-yl]propyl}acetamide as a white semi-solid.



1H NMR (500 MHz, DMSO-d6) δ 8.17 (d, J=8.5 Hz, 1H), 7.86 (t, J=5.1 Hz, 1H), 7.42 (d, J=1.5 Hz, 1H), 7.07 (dd, J=8.4, 1.6 Hz, 1H), 6.53 (br s, 2H), 5.08-4.57 (br s, 2H), 4.88 (s, 1H), 4.64 (br s, 2H), 3.30 (s, 3H), 3.08 (m, 2H), 2.68 (t, J=7.6 Hz, 2H), 1.81 (s, 3H), 1.77 (m, 2H), 1.17 (br s, 6H);


MS (APCI) m/z 400 (M+H)+;


Anal. Calcd. for C21H29N5O3.0.5H2O: C, 61.64; H, 7.41; N, 17.11. Found: C, 61.63; H, 7.13; N, 17.31.


Example 116
N-{3-[4-Amino-2-(hydroxymethyl)-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]propyl}methanesulfonamide



embedded image


A solution of N-{3-[4-amino-1-(2-hydroxy-2-methylpropyl)-2-(methoxymethyl)-1H-imidazo[4,5-c]quinolin-7-yl]propyl}methanesulfonamide (630 mg, 1.45 mmol) in dichloromethane (125 mL) was sealed with a septum and purged with nitrogen gas. A 1.0 M solution of boron tribromide in dichloromethane (7.3 mL) was added via syringe. The resulting mixture was stirred for 18 hours. Methanol (50 mL) was added and the solution was concentrated under reduced pressure. The resulting solid was dissolved in a 2.0 M solution of ammonia in methanol (75 mL) and then concentrated under reduced pressure. This step was repeated two more times with silica gel (1 tbsp) being added prior to final concentration. The crude product absorbed on silica was purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with CMA:chloroform gradient; 0-25% CMA over 1200 mL, followed by 25-40% CMA over 1900 mL). The solid product was washed with water, filtered, and dried to provide 325 mg of N-{3-[4-amino-2-(hydroxymethyl)-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]propyl}methanesulfonamide as a white solid, mp 202-205° C.



1H NMR (500 MHz, DMSO-d6) δ 8.18 (d, J=8.5 Hz, 1H), 7.42 (d, J=1.6 Hz, 1H), 7.09 (dd, J=8.5, 1.7 Hz, 1H), 7.05 (t, J=5.4 Hz, 1H), 6.57 (br s, 2H), 5.51 (br s, 1H), 4.97 (s, 1H), 4.87 (br s, 2H), 4.68 (br s, 2H), 3.00 (m, 2H), 2.89 (s, 3H), 2.72 (t, J=7.2 Hz, 2H), 1.84 (m, 2H), 1.17 (br s, 6H);


MS (APCI) m/z 421 (M+H)+;


Anal. Calcd. for C19H27N5O4S: C, 54.14; H, 6.46; N, 16.61. Found: C, 54.10; H, 6.30; N, 16.66.


Example 117
N-[4-Amino-2-(ethoxymethyl)-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-8-yl]-2-methylpropanamide



embedded image



Part A


Phosphorous oxychloride (5.45 mL, 58.5 mmol) was added to a slurry of 6-bromo-3-nitroquinolin-4-ol (15 g, 55.7 mmol) in N,N-dimethylformamide (100 mL). The reaction mixture was heated to 100° C. for 10 minutes and then cooled to ambient temperature. The solution was poured into ice water (400 mL) and stirred for 20 minutes. The resulting precipitate was filtered, washed with water, and dried. The recovered solid was transferred to a round bottomed flask and tetrahydrofuran (100 mL) was added. A solution of triethylamine (11.6 mL, 83.5 mmol) and 1-amino-2-methylpropan-2-ol (5.21 g, 58.5 mmol) in tetrahydrofuran (20 mL) was added dropwise. The reaction was stirred for 16 hours. The mixture was poured into water and stirred for 15 minutes. The resulting yellow solid was filtered and dried to provide 17.2 g of 1-[(6-bromo-3-nitroquinolin-4-yl)amino]-2-methylpropan-2-ol as a yellow solid.


Part B


1-[(6-Bromo-3-nitroquinolin-4-yl)amino]-2-methylpropan-2-ol (17.2 g, 50.5 mmol), toluene (150 mL) and isopropanol (20 mL) were added to a Parr flask containing 5% platinum on carbon (1.7 g) wetted with toluene. The flask was evacuated three times, charged with hydrogen to 30 psi, and shaken for 72 hours. The reaction mixture was filtered through a pad of CELITE filter agent. The CELITE was washed with several portions of dichloromethane followed by methanol. The filtrate was concentrated to provide crude 1-[(3-amino-6-bromoquinolin-4-yl)amino]-2-methylpropan-2-ol as a brown oil.


Part C


Ethoxyacetyl chloride (6.2 g, 50.5 mmol) was added dropwise to a solution of crude 1-[(3-amino-6-bromoquinolin-4-yl)amino]-2-methylpropan-2-ol, dichloromethane (500 mL), and triethylamine (7 mL, 50.5 mmol). After stirring for 16 hours, water was added and the mixture was stirred for an additional 2 hours. The fractions were separated. The organic fraction was washed with water, concentrated under reduced pressure and redissolved in ethanol (500 mL). Water (150 mL) and potassium carbonate (10.5 g) were added and the mixture was heated at reflux temperature for 3 hours. The ethanol was removed under reduced pressure. The remaining aqueous fraction was extracted with ethyl acetate (2×500 mL). The organic fractions were combined, washed with water followed by saturated aqueous sodium chloride, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. Recrystallization from acetonitrile provided 10.4 g of 1-[8-bromo-2-(ethoxymethyl)-1H-imidazo[4,5-c]quinolin-1-yl]-2-methylpropan-2-ol as a tan powder.


Part D


1-[8-Bromo-2-(ethoxymethyl)-1H-imidazo[4,5-c]quinolin-1-yl]-2-methylpropan-2-ol (0.550 g, 1.45 mmol), isobutyramide (0.150 g, 1.74 mmol), copper(I) iodide (0.055 g, 0.290 mmol), potassium phosphate (0.647 g, 3.05 mmol), and 1,4-dioxane (1.5 mL) were added to a scintillation vial. 1,2-(±)-trans-Diaminocyclohexane (0.035 mL, 0.290 mmol) was added. The vial was flushed with nitrogen, sealed with a Teflon-lined cap and heated at 110° C. for 72 hours. The vial was cooled to ambient temperature and the reaction was diluted with dichloromethane and methanol. Purification by chromatography with a HORIZON HPFC system (silica cartridge, eluting with a linear gradient of 1-15% CMA in chloroform) provided 0.38 g of N-[2-(ethoxymethyl)-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-8-yl]-2-methylpropanamide as a dark semi-solid.


Part E


3-Chloroperoxybenzoic acid (60% pure, 0.283 g, 1.0 mmol) was added to a solution of N-[2-(ethoxymethyl)-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-8-yl]-2-methylpropanamide (0.380 g, 1.0 mmol) in chloroform (10 mL) and the reaction was stirred for 1 hour. The mixture was cooled in an ice bath and ammonium hydroxide (3 mL) was added. After 15 minutes of stirring, p-toluenesulfonyl chloride (0.190 g, 1.0 mmol) was added and the reaction mixture was stirred for 72 hours. The fractions were separated and the aqueous fraction was extracted with chloroform. The combined organic fractions were washed with brine and purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with a linear gradient of 2-20% CMA in chloroform). Recrystallization from acetonitrile provided 0.231 g of N-[4-amino-2-(ethoxymethyl)-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-8-yl]-2-methylpropanamide as an off-white solid, mp 259-261° C.



1H NMR (300 MHz, DMSO-d6) δ 9.88 (s, 1H), 8.86 (d, J=1.7 Hz, 1H), 7.53-7.42 (m, 2H), 6.41 (s, 2H), 5.15-4.73 (m, 2H), 4.90 (s, 1H), 4.63 (s, 2H), 3.51 (q, J=7.0 Hz, 2H), 2.68-2.59 (m, 1H), 1.22 (br s, 6H), 1.14 (d, J=6.9 Hz, 6H), 1.13 (t, J=7.0 Hz, 3H);


MS (ESI) m/z 400.17 (M+H)+;


Anal. Calcd. for C21H29N5O3: C, 63.14; H, 7.32; N, 17.53. Found: C, 62.78; H, 7.20; N, 17.48.


Example 118
N-{2-[4-Amino-2-ethyl-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]ethyl}methanesulfonamide



embedded image



Part A


A thick walled glass tube, equipped with a stir bar, was charged with palladium (II) acetate (64 mg, 0.29 mmol), acetonitrile (50 mL), N-vinylphthalimide (1.19 g, 6.89 mmol), triethylamine (2.4 mL, 17 mmol), tri-o-tolylphosphine (260 mg, 0.86 mmol) and 1-(7-bromo-2-ethyl-1H-imidazo[4,5-c]quinolin-1-yl)-2-methylpropan-2-ol (2.00 g, 5.74 mmol). The reaction mixture was purged with nitrogen and the tube was sealed and heated at 120° C. in an oil bath for 16 hours. The reaction was cooled to ambient temperature and silica gel (20 g) was added followed by concentration under reduced pressure. The sample absorbed on silica was purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with a 0-7% gradient of methanol in dichloromethane). The recovered solid was washed with 1% aqueous sodium carbonate to provide 1.95 g of 2-{2[2-ethyl-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]ethenyl}-1H-isoindole-1,3(2H)-dione as a bright yellow solid. MS (APCI) m/z 441 (M+H)+.


Part B


A glass Parr vessel was charged with 10% palladium on carbon (0.4 g), methanol (75 mL) and 2-{2-[2-ethyl-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]ethenyl}-1H-isoindole-1,3(2H)-dione (1.9 g, 4.3 mmol). The vessel was evacuated and charged with hydrogen gas (40 psi, 2.8×105 Pa). The reaction was shaken at 50° C. for approximately 18 hours and then cooled to ambient temperature. The reaction mixture was filtered followed by concentration under reduced pressure to provide 1.9 g of 2-{2-[2-ethyl-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]ethyl}-1H-isoindole-1,3(2H)-dione as a bright yellow solid. MS (APCI) m/z 443 (M+H)+.


Part C


2-{2-[2-ethyl-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]ethyl}-1H-isoindole-1,3(2H)-dione (1.90 g, 4.29 mmol) in dichloromethane (75 mL) was combined with 3-chloroperoxybenzoic acid (60% pure, 1.24 g, 4.72 mmol) and the reaction was stirred for 18 hours. Concentrated ammonium hydroxide (40 mL) was added and the mixture was vigorously stirred for an additional 10 minutes. p-Toluenesulfonyl chloride (900 mg, 4.72 mmol) was added and the mixture was stirred for an additional 1 hour. The fractions were separated and the aqueous fraction was extracted with chloroform (3×50 mL). The combined organic fractions were sequentially dried (MgSO4), filtered, and concentrated under reduced pressure. Purification by chromatography using a HORIZON HPFC system (silica cartridge, eluting with 0-8% gradient of methanol in dichloromethane) provided 1.05 g of 2-{2-[4-amino-2-ethyl-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]ethyl}-1H-isoindole-1,3(2H)-dione as an off-white foam. MS (APCI) m/z 458 (M+H)+.


Part D:


2-{2-[4-amino-2-ethyl-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]ethyl}-1H-isoindole-1,3(2H)-dione (1.05 g, 2.29 mmol) was dissolved in 2:1 ethanol:tetrahydrofuran (100 mL), combined with hydrazine hydrate (150 mg, 2.98 mmol) and heated to reflux temperature for three hours. Additional hydrazine hydrate (0.25 mL) was added and solution was refluxed for an additional 18 hours. The reaction mixture was cooled to ambient temperature and filtered. The crude solid was combined with triethylamine (2.1 mL, 15.3 mmol) in dichloromethane (75 mL) and the solution was cooled to 0° C. in an ice/water bath. Methanesulfonic anhydride (585 mg, 3.35 mmol) was added and solution was stirred for approximately 18 hours while warming to ambient temperature. The solution was concentrated under reduced pressure and the residue was slurried in 1% aqueous sodium carbonate (125 mL). The aqueous mixture was extracted with chloroform (50 mL) until no product was present in the aqueous fraction. The combined organic fractions were sequentially dried (MgSO4), filtered, concentrated under reduced pressure, and purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with a CMA:chloroform gradient; 0-25% CMA over 1200 mL, followed by 25-30% CMA over 1100 mL). A final recrystallization for acetonitrile provided 328 mg of N-{2-[4-amino-2-ethyl-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]ethyl}methanesulfonamide as an off-white solid, mp 149-152° C.



1H NMR (500 MHz, DMSO-d6) δ 8.18 (d, J=8.5 Hz, 1H), 7.45 (d, J=1.6 Hz, 1H), 7.10 (m, 1H), 7.09 (dd, J=8.5, 1.7 Hz, 1H), 6.37 (br s, 2H), 4.77 (s, 1H), 4.52 (br s, 2H), 3.26 (m, 2H), 3.04 (q, J=7.4 Hz, 2H), 2.87 (t, J=7.4 Hz, 2H), 2.82 (s, 3H), 1.34 (t, J=7.5 Hz, 3H), 1.16 (br s, 6H);


MS (APCI) m/z 406 (M+H)+;


Anal. Calcd. for C19H27N5O3S: C, 56.28; H, 6.71; N, 17.27. Found: C, 56.00; H, 6.87; N, 17.44.


Example 119
1-{3-[4-Amino-8-(2-aminoethyl)-2-(2-methoxyethyl)-1H-imidazo[4,5-c]quinolin-1-yl]propyl}pyrrolidin-2-one



embedded image



Part A


A thick walled glass tube, equipped with a stir bar, was charged with palladium (II) acetate (17 mg, 0.08 mmol), acetonitrile (50 mL), N-vinylphthalimide (716 mg, 4.13 mmol), triethylamine (1.6 mL, 11 mmol), tri-o-tolylphosphine (69 mg, 0.22 mmol) and 1-{3-[8-bromo-2-(2-methoxyethyl)-1H-imidazo[4,5-c]quinolin-1-yl]propyl}pyrrolidin-2-one (1.62 g, 3.76 mmol). The reaction mixture was purged with nitrogen and the tube was sealed and heated at 120° C. in an oil bath for 15 hours. The reaction was cooled to ambient temperature and methanol (50 mL) and chloroform (50 ml) were added. After filtering through a 0.2 micron PTFE membrane, the solution was concentrated under reduced pressure and then purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with a 0-8% gradient of methanol in dichloromethane) to provide 2.0 g of 2-(2-{2-(2-methoxyethyl)-1-[3-(2-oxopyrrolidin-1-yl)propyl]-1H-imidazo[4,5-c]quinolin-8-yl}ethenyl)-1H-isoindole-1,3(2H)-dione as a bright yellow solid.


MS (APCI) m/z 524 (M+H)+.


Part B


A glass Parr vessel was charged with 10% palladium on carbon (0.2 g), methanol (75 mL) and 2-(2-{2-(2-methoxyethyl)-1-[3-(2-oxopyrrolidin-1-yl)propyl]-1H-imidazo[4,5-c]quinolin-8-yl}ethenyl)-1H-isoindole-1,3(2H)-dione (2.0 g, 3.8 mmol). The vessel was evacuated and charged with hydrogen gas (40 psi, 2.8×105 Pa). The reaction was shaken at 50° C. for approximately 18 hours. After cooling to ambient temperature, the reaction mixture was filtered followed by concentration under reduced pressure to provide 2.1 g of 2-(2-{2-(2-methoxyethyl)-1-[3-(2-oxo-pyrrolidin-1-yl)propyl]-1H-imidazo[4,5-c]quinolin-8-yl}ethyl)-1H-isoindole-1,3(2H)-dione as a pale yellow solid. MS (ESI) m/z 526 (M+H)+.


Part C


2-(2-{2-(2-Methoxyethyl)-1-[3-(2-oxo-pyrrolidin-1-yl)propyl]-1H-imidazo[4,5-c]quinolin-8-yl}ethyl)-1H-isoindole-1,3(2H)-dione (2.1 g, 3.8 mmol) in dichloromethane (75 mL) was combined with 3-chloroperoxybenzoic acid (60% pure, 2.0 g, 7.5 mmol) and the reaction was stirred for 18 hours. Concentrated ammonium hydroxide (40 mL) was added and the mixture was vigorously stirred for an additional 10 minutes. p-Toluenesulfonyl chloride (791 mg, 4.12 mmol) was added and the mixture was stirred for an additional 1 hour. The fractions were separated and aqueous fraction was extracted with chloroform (10×35 mL). The combined organic fractions were sequentially dried (MgSO4), filtered, and concentrated to provide 2.2 g of 2-(2-{4-amino-2-(2-methoxyethyl)-1-[3-(2-oxopyrrolidin-1-yl)propyl]-1H-imidazo[4,5-c]quinolin-8-yl}ethyl)-1H-isoindole-1,3(2H)dione as an off-white foam. MS (APCI) m/z 541 (M+H)+.


Part D


2-(2-{4-Amino-2-(2-methoxyethyl)-1-[3-(2-oxopyrrolidin-1-yl)propyl]-1H-imidazo[4,5-c]quinolin-8-yl}ethyl)-1H-isoindole-1,3(2H)dione (2.2 g, 4.1 mmol) was dissolved in 2:1 ethanol:tetrahydrofuran (40 mL), combined with hydrazine hydrate (410 mg, 8.2 mmol) and heated at reflux temperature for three hours. Additional hydrazine hydrate (0.25 mL) was added and the solution was refluxed for an additional 1.5 hours. The reaction was cooled to ambient temperature, filtered and the filtrate concentrated under reduce pressure. Purification by chromatography using a HORIZON HPFC system (silica cartridge, eluting with a 0-40% CMA:chloroform gradient over 3.8 L) provided 650 mg of 1-{3-[4-amino-8-(2-aminoethyl)-2-(2-methoxyethyl)-1H-imidazo[4,5-c]quinolin-1-yl]propyl}pyrrolidin-2-one as an off-white solid. MS (ESI) m/z 411 (M+H)+.


Example 120
2-Ethyl-7-[2-(methylsulfonyl)ethyl])-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-4-amine



embedded image



Part A


A thick walled glass tube, equipped with a stir bar, was charged with palladium (II) acetate (7 mg, 0.03 mmol), acetonitrile (15 mL), N,N-dimethylformamide (15 mL), methyl vinyl sulfone (150 mg, 1.41 mmol), triethylamine (0.53 mL, 3.8 mmol), tri-o-tolylphosphine (23 mg, 0.08 mmol) and 7-bromo-2-ethyl-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-4-amine (500 g, 1.28 mmol). The reaction mixture was purged with nitrogen and the tube was sealed and heated at 120° C. in an oil bath for 16 hours. The reaction mixture was cooled to ambient temperature and then concentrated under reduced pressure. The resulting solid was partitioned between chloroform (100 mL) and 1% aqueous sodium carbonate (100 mL). The fractions were separated and the aqueous fraction was extracted with chloroform (2×50 mL). The combined organic fractions were sequentially dried (MgSO4), filtered, and purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with a CMA:chloroform gradient, 0-15% CMA over 1200 mL followed by 15-20% CMA over 1100 mL). Recrystallization from methanol provided 315 mg of 2-ethyl-7-[2-(methylsulfonyl)ethenyl]-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-4-amine as a pale yellow solid.



1H NMR (500 MHz, DMSO-d6) δ 8.08 (d, J=8.6 Hz, 1H), 7.89 (d, J=1.8 Hz, 1H), 7.62-7.53 (m, 3H), 6.65 (br s, 2H), 4.46 (d, J=7.1 Hz, 2H), 3.82 (m, 2H), 3.15-3.12 (m, 2H), 3.12 (m, 3H), 2.96 (q, J=7.4 Hz, 2H), 2.07 (m, 1H), 1.51-1.40 (m, 4H), 1.38 (t, J=7.4 Hz, 3H);


MS (ESI) m/z 415 (M+H)+.


Part B


A glass Parr vessel was charged with 10% palladium on carbon (0.2 g), methanol (25 mL), ethanol (25 mL) and 2-ethyl-7-[2-(methylsulfonyl)ethenyl]-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-4-amine (315 mg, 0.76 mmol). The vessel was evacuated and charged with hydrogen gas (40 psi, 2.8×105 Pa). The reaction was shaken at 50° C. for approximately 18 hours and then cooled to ambient temperature. The reaction mixture was sequentially filtered, concentrated under reduced pressure, and purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with CMA:chloroform gradient, 0-10% CMA over 1000 mL followed by 10-25% CMA over 1000 mL). A final recrystallization from acetonitrile provided 125 mg of 2-ethyl-7-[2-(methylsulfonyl)ethyl])-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-4-amine as a white crystalline solid, mp 246.5-249.0° C.



1H NMR (500 MHz, DMSO-d6) δ 7.97 (d, J=8.5 Hz, 1H), 7.52 (d, J=1.7 Hz, 1H), 7.21 (dd, J=8.4, 1.8 Hz, 1H), 6.48 (br s, 2H), 4.42 (d, J=7.0 Hz, 2H), 3.83-3.80 (m, 2H), 3.53-3.50 (m, 2H), 3.17-3.11 (m, 4H), 3.02 (s, 3H), 2.94 (q, J=7.4 Hz, 2H), 2.07 (m, 1H), 1.51-1.40 (m, 4H), 1.37 (t, J=7.4 Hz, 3H);



13C NMR (125 MHz, DMSO-d6) δ 154.2, 151.7, 144.9, 136.1, 132.3, 126.2, 125.6, 121.7, 120.2, 113.3, 66.4, 54.3, 49.6, 40.1, 35.6, 29.5, 27.6, 20.0, 11.9;


MS (ESI) m/z 417 (M+H)+;


Anal. Calcd. for C21H28N4O3S: C, 60.55; H, 6.78; N, 13.45. Found: C, 60.63; H, 6.70; N, 13.76.


Example 121
3-[4-Amino-2-ethyl-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-7-yl]propanenitrile



embedded image



Part A


A thick walled glass tube, equipped with a stir bar, was charged with palladium (II) acetate (225 mg, 1.00 mmol), acetonitrile (15 mL), N,N-dimethylformamide (15 mL), acrylonitrile (1.20 g, 22.6 mmol), triethylamine (8.6 mL, 62 mmol), tri-o-tolylphosphine (935 mg, 3.10 mmol) and 7-bromo-2-ethyl-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-4-amine (8.00 g, 20.5 mmol). The reaction mixture was purged with nitrogen and the tube was sealed and heated at 120° C. for approximately 18 hours. The reaction was cooled to ambient temperature and methanol (100 mL) was added. The reaction was filtered and the filtrate concentrated under reduced pressure. The resulting solid was washed with 1% aqueous sodium carbonate (100 mL) followed by washing with water (2×100 mL). Purification by chromatography using a HORIZON HPFC system (silica cartridge, eluting with a CMA:chloroform gradient, 0-15% CMA over 1500 mL followed by 15-25% CMA over 2.4 L) provided 5.2 g of a mixture of cis and trans isomers of 3-[4-amino-2-ethyl-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-7-yl]prop-2-enenitrile as a yellow solid. MS (APCI) m/z 362 (M+H)+.


Part B


A glass Parr vessel was charged with 10% palladium on carbon (0.05 g), methanol (20 mL), and 3-[4-amino-2-ethyl-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-7-yl]prop-2-enenitrile (100 mg, 0.27 mmol). The vessel was evacuated and charged with hydrogen gas (40 psi, 2.8×105 Pa). The reaction was shaken at 50° C. for approximately 18 hours and then cooled to ambient temperature. The reaction mixture was sequentially filtered, concentrated under reduced pressure, and purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with a CMA:chloroform gradient, 0-30% CMA). A final recrystallization from acetonitrile provided 71 mg of 3-[4-amino-2-ethyl-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-7-yl]propanenitrile as a white solid, mp 273-275° C.



1H NMR (500 MHz, DMSO-d6) δ 7.97 (d, J=8.4 Hz, 1H), 7.53 (d, J=1.7 Hz, 1H), 7.20 (dd, J=8.4, 1.8 Hz, 1H), 6.45 (br s, 2H), 4.42 (d, J=7.2 Hz, 2H), 3.82 (m, 2H), 3.15 (m, 2H), 3.00-2.88 (m, 6H), 2.08 (m, 1H), 1.51-1.40 (m, 4H), 1.37 (t, J=7.4 Hz, 3H);



13C NMR (125 MHz, DMSO-d6) δ 154.2, 151.7, 144.8, 136.6, 132.3, 126.2, 125.5, 121.6, 120.3, 120.1, 113.4, 66.4, 49.6, 35.6, 30.4, 29.5, 20.0, 17.9, 11.9;


MS (ESI) m/z 364 (M+H)+;


Anal. Calcd. for C21H25N5O: C, 69.40; H, 6.93; N, 19.27. Found: C, 69.16; H, 6.87; N, 19.25.


Example 122
3-[4-Amino-2-ethyl-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-7-yl]propanamide



embedded image


3-[4-Amino-2-ethyl-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-7-yl]propanenitrile (590 mg, 1.6 mmol), 10% aqueous sodium hydroxide (132 mg, 0.33 mmol), and 30% aqueous hydrogen peroxide (516 mg, 4.55 mmol) were combined in methanol (75 mL) and the reaction was heated at 50° C. for 18 hours. The mixture was concentrated under reduced pressure and then partitioned between chloroform (75 mL) and water (75 mL). The fractions were separated and the aqueous fraction was extracted with chloroform (3×25 mL). The combined organic fractions were sequentially dried (MgSO4), filtered, concentrated to dryness and purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with 2 L of 10% methanol in dichloromethane). A final recrystallization from acetonitrile provided 35 mg of 3-[4-amino-2-ethyl-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-7-yl]propanamide as an off-white solid, mp 197-199° C.



1H NMR (500 MHz, DMSO-d6) δ 7.92 (d, J=8.4 Hz, 1H), 7.44 (d, J=1.4 Hz, 1H), 7.32 (s, 1H), 7.13 (dd, J=8.4, 1.7 Hz, 1H), 6.77 (s, 1H), 6.39 (br s, 2H), 4.40 (d, J=7.0 Hz, 2H), 3.82 (m, 2H), 3.15 (m, 2H), 2.95-2.90 (m, 4H), 2.44 (t, J=7.6 Hz, 2H), 2.08 (m, 1H), 1.51-1.40 (m, 4H), 1.37 (t, J=7.4 Hz, 3H);



13C NMR (125 MHz, DMSO-d6) δ 173.3, 154.1, 151.5, 144.8, 139.4, 132.5, 125.9, 125.0, 121.9, 119.8, 112.8, 66.4, 49.6, 36.3, 35.6, 30.6, 29.5, 20.0, 11.9;


MS (ESI) m/z 382 (M+H)+;


Anal. Calcd. for C21H27N5O2.0.30H2O: C, 65.20; H, 7.19; N, 18.10. Found: C, 65.19; H, 7.14; N, 18.24.


Example 123
7-(3-Aminopropyl)-2-ethyl-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-4-amine



embedded image


A glass Parr vessel was charged with 10% palladium on carbon (1 g), methanol (50 mL), 3-[4-amino-2-ethyl-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-7-yl]prop-2-enenitrile (4.0 g, 11.1 mmol) and trifluoroacetic acid (6.3 g, 55 mmol). The vessel was evacuated, charged with hydrogen gas (40 psi, 2.8×105 Pa) and shaken for approximately 18 hours. The mixture was filtered and concentrated under reduced pressure to a yellow oil. The oil was dissolved in 10% aqueous hydrochloric acid and stirred for 18 hours. Chloroform (100 mL) was added followed by slow addition of potassium carbonate until the pH equaled 11. The fractions were separated and the aqueous fraction was extracted with chloroform (4×50 mL). The combined organic fractions were sequentially dried (MgSO4), filtered, and concentrated under reduced pressure. Recrystallization from acetonitrile provided 3.55 g of the desired product as a white solid. A small amount of this material was further purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with a CMA:chloroform gradient, 0-30% CMA over 1000 mL followed by 30% CMA over 1000 mL). A final recrystallization from acetonitrile provided 7-(3-aminopropyl)-2-ethyl-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-4-amine as a white solid, mp 217-220° C.



1H NMR (500 MHz, DMSO-d6) δ 7.92 (d, J=8.4 Hz, 1H), 7.42 (d, J=1.5 Hz, 1H), 7.12 (dd, J=8.4, 1.7 Hz, 1H), 6.38 (br s, 2H), 4.40 (d, J=7.0 Hz, 2H), 3.82 (m, 2H), 3.16 (m, 2H), 2.93 (q, J=7.4 Hz, 2H), 2.70 (t, J=7.6 Hz, 2H), 2.59 (t, J=6.8 Hz, 2H), 2.09 (m, 1H), 1.71 (m, 2H), 1.51-1.36 (m, 6H), 1.37 (t, J=7.4 Hz, 3H);



13C NMR (125 MHz, DMSO-d6) δ 154.0, 151.5, 144.9, 140.2, 132.5, 125.9, 125.2, 122.0, 119.8, 112.7, 66.4, 49.6, 41.2, 35.6, 34.9, 32.4, 29.5, 20.0, 11.9;


MS (ESI) m/z 368 (M+H)+;


Anal. Calcd. for C21H29N5O.0.25H2O: C, 67.81; H, 7.99; N, 18.83. Found: C, 68.00; H, 8.03; N, 18.75.


Example 124
N-{3-[4-Amino-2-ethyl-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-7-yl]propyl}methanesulfonamide



embedded image


7-(3-Aminopropyl)-2-ethyl-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-4-amine (500 mg, 1.4 mmol), triethylamine (410 mg, 4.1 mmol), and methanesulfonic anhydride (260 mg, 1.5 mmol) were combined in dichloromethane (50 mL) at 0° C. and the reaction was stirred for approximately 18 hours. Aqueous sodium carbonate (1%, 50 mL) was added and the reaction mixture was stirred for one additional hour. The fractions were separated and the aqueous fraction was extracted with chloroform (3×20 mL). The combined organic fractions were sequentially dried (MgSO4), filtered and purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with a CMA:chloroform gradient, 0-20% CMA over 1500 mL followed by 20% CMA over 500 mL). A final recrystallization from acetonitrile provided 165 mg of N-{3-[4-amino-2-ethyl-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-7-yl]propyl}methanesulfonamide as a white solid, mp 168-170° C.



1H NMR (500 MHz, DMSO-d6) δ 7.93 (d, J=8.4 Hz, 1H), 7.44 (d, J=1.6 Hz, 1H), 7.14 (dd, J=8.4, 1.7 Hz, 1H), 7.05 (t, J=5.5 Hz, 1H), 6.40 (br s, 2H), 4.40 (d, J=7.0 Hz, 2H), 3.82 (m, 2H), 3.16 (m, 2H), 3.00 (m, 2H), 2.93 (q, J=7.4 Hz, 2H), 2.89 (s, 3H), 2.73 (t, J=7.6 Hz, 2H), 2.09 (m, 1H), 1.85 (m, 2H), 1.51-1.40 (m, 4H), 1.37 (t, J=7.4 Hz, 3H);



13C NMR (125 MHz, DMSO-d6) δ 154.0, 151.6, 144.9, 139.4, 132.4, 126.0, 125.3, 121.9, 119.9, 112.8, 66.4, 49.6, 42.0, 39.1, 35.6, 32.1, 31.0, 29.5, 20.0, 11.9;


MS (ESI) m/z 446 (M+H)+;


Anal. Calcd. for C22H31N5O3S: C, 59.30; H, 7.01; N, 15.72. Found: C, 59.22; H, 7.29; N, 15.68.


Example 125
N-{3-[4-amino-2-ethyl-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-7-yl]propyl}acetamide



embedded image


7-(3-Aminopropyl)-2-ethyl-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-4-amine (500 mg, 1.4 mmol), triethylamine (410 mg, 4.1 mmol), and acetic anhydride (153 mg, 1.5 mmol) were combined in dichloromethane (50 mL) at 0° C. and the reaction was stirred for approximately 18 hours. Aqueous sodium carbonate (1%, 50 mL) was added and the reaction mixture was stirred for one additional hour. The fractions were separated and the aqueous fraction was extracted with chloroform (3×20 mL). The combined organic fractions were sequentially dried (MgSO4), filtered and purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with a CMA:chloroform gradient, 0-15% CMA over 2000 mL followed by 15-20% CMA over 500 mL). The desired product was further purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with a 0-8% gradient of methanol in dichloromethane). A final recrystallization from acetonitrile provided 142 mg of N-{3-[4-amino-2-ethyl-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-7-yl]propyl}acetamide as a white solid, mp 209-211° C.



1H NMR (500 MHz, DMSO-d6) δ 7.92 (d, J=8.4 Hz, 1H), 7.87 (t, J=5.1 Hz, 1H), 7.44 (d, J=1.5 Hz, 1H), 7.13 (dd, J=8.4, 1.7 Hz, 1H), 6.39 (br s, 2H), 4.40 (d, J=7.0 Hz, 2H), 3.82 (m, 2H), 3.16 (m, 2H), 3.09 (m, 2H), 2.93 (q, J=7.4 Hz, 2H), 2.69 (t, J=7.6 Hz, 2H), 2.09 (m, 1H), 1.81 (s, 3H), 1.77 (m, 2H), 1.51-1.40 (m, 4H), 1.37 (t, J=7.4 Hz, 3H);



13C NMR (125 MHz, DMSO-d6) δ 168.8, 154.0, 151.5, 144.9, 139.6, 132.4, 125.9, 125.3, 121.9, 119.8, 112.8, 66.4, 49.6, 38.1, 35.6, 32.4, 30.6, 29.5, 22.5, 20.0, 11.9;


MS (ESI) m/z 410 (M+H)+;


Anal. Calcd. for C23H31N5O2: C, 67.46; H, 7.63; N, 17.10. Found: C, 67.18; H, 7.37; N, 17.14.


Example 126
N-{3-[4-amino-2-ethyl-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-7-yl]propyl}-N′-isopropylurea



embedded image


7-(3-Aminopropyl)-2-ethyl-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-4-amine (500 mg, 1.4 mmol), triethylamine (410 mg, 4.1 mmol), and isopropyl isocyanate (128 mg, 1.5 mmol) were combined in dichloromethane (50 mL) at 0° C. and the reaction was stirred for approximately 18 hours. Aqueous sodium carbonate (1%, 50 mL) was added and the reaction mixture was stirred for one additional hour. The fractions were separated and the aqueous fraction was extracted with chloroform (3×20 mL). The combined organic fractions were sequentially dried (MgSO4), filtered and purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with a CMA:chloroform gradient, 0-20% CMA over 2000 mL). The desired product was further purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with 10% methanol in dichloromethane over 1500 mL). A final purification by recrystallization from acetonitrile provided 142 mg of N-{3-[4-amino-2-ethyl-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-7-yl]propyl}-N′-isopropylurea as a white solid, mp 213-216° C.



1H NMR (500 MHz, DMSO-d6) δ 7.93 (d, J=8.4 Hz, 1H), 7.43 (d, J=1.6 Hz, 1H), 7.13 (dd, J=8.4, 1.7 Hz, 1H), 6.40 (br s, 2H), 5.76 (t, J=5.6 Hz, 1H), 5.61 (d, J=7.7 Hz, 1H), 4.40 (d, J=7.0 Hz, 2H), 3.82 (m, 2H), 3.65 (m, 1H), 3.16 (m, 2H), 3.02 (m, 2H), 2.93 (q, J=7.4 Hz, 2H), 2.67 (t, J=7.6 Hz, 2H), 2.09 (m, 1H), 1.73 (m, 2H), 1.51-1.40 (m, 4H), 1.37 (t, J=7.4 Hz, 3H), 1.02 (d, J=6.5 Hz, 6H);



13C NMR (125 MHz, DMSO-d6) δ 157.3, 154.1, 151.5, 144.7, 139.7, 132.5, 125.9, 125.1, 122.0, 119.9, 112.7, 66.4, 49.6, 40.7, 38.7, 35.6, 32.4, 31.5, 29.5, 23.1, 20.0, 11.9;


MS (ESI) m/z 453 (M+H)+;


Anal. Calcd. for C25H36N6O2.0.33H2O: C, 65.41; H, 8.06; N, 18.31. Found: C, 65.41; H, 8.26; N, 18.30.


Example 127
1-Isobutyl-8-{[(3-methoxybenzyl)amino]methyl}-1H-imidazo[4,5-c]quinolin-4-amine



embedded image



Part A


A solution of 1-isobutyl-8-vinyl-1H-imidazo[4,5-c]quinolin-4-amine (500 mg, 1.9 mmol) in dichloromethane (30 mL) and methanol (5 mL) was cooled to −78° C. Ozone was bubbled through the solution for 10 minutes. While still cold, the reaction was purged with oxygen for 15 minutes and dimethyl sulfide (0.7 mL, 9.4 mmol) was added. The reaction was warmed to ambient temperature and then concentrated under reduced pressure. The residue was dissolved in dichloromethane (50 mL) and washed with 1% aqueous sodium carbonate (2×15 mL). The combined aqueous fractions were extracted with chloroform (3×20 mL) and sequentially dried (MgSO4), filtered, and purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with a gradient of 0-6% methanol in dichloromethane) to provide 237 mg of 4-amino-1-isobutyl-1H-imidazo[4,5-c]quinoline-8-carbaldehyde as an off-white solid. MS (ESI) m/z 269 (M+H)+.


Part B


4-Amino-1-isobutyl-1H-imidazo[4,5-c]quinoline-8-carbaldehyde (237 mg, 0.66 mmol) and 3-methoxybenzylamine (100 mg, 1.3 mmol) were combined in methanol (25 mL) and dichloromethane (1 mL). The solution was purged with nitrogen gas and a 1M solution of sodium cyanoborohydride in THF (2 mL) was added. The reaction was stirred for 30 minutes. Additional sodium cyanoborohydride in THF (1M, 1 mL) was added and the reaction was stirred for an additional 15 minutes. Aqueous sodium carbonate (1%, 0.25 mL) was added and the reaction was stirred for 7 days. The solvent was removed under reduced pressure and approximately equal portions of chloroform and water were added. The fractions were separated and the organic fraction was sequentially dried (MgSO4), filtered, and purified by chromatography using a HORIZON HPFC system (silica cartridge, eluting with a CMA:chloroform gradient, 0-20% CMA). Recrystallization from acetonitrile provided 25 mg of 1-isobutyl-8-{[(3-methoxybenzyl)amino]methyl}-1H-imidazo[4,5-c]quinolin-4-amine as an off-white solid, mp 125-126° C.



1H NMR (500 MHz, DMSO-d6) δ 8.15 (s, 1H), 7.92 (d, J=1.2 Hz, 1H), 7.57 (d, J=8.5 Hz, 1H), 7.41 (dd, J=8.5, 1.7 Hz, 1H), 7.22 (t, J=7.4 Hz, 1H), 6.95-6.92 (m, 2H), 6.79 (m, 1H), 6.46 (br s, 2H), 4.37 (d, J=7.3 Hz, 2H), 3.82 (s, 2H), 3.73 (s, 3H), 3.71 (s, 2H), 2.69 (br s, 1H), 2.18 (septet, J=6.8 Hz, 1H), 0.90 (d, J=6.6 Hz, 6H);


MS (ESI) m/z 390 (M+H)+;


Anal. Calcd. for C23H27N5O: C, 70.93; H, 6.99; N, 17.98. Found: C, 70.64; H, 6.92; N, 18.01.


Example 128
(2E)-3-[4-Amino-2-ethyl-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-7-yl]-N,N-dimethylprop-2-enamide



embedded image


A thick walled glass vessel, equipped with a stir bar, was charged with a warmed solution of 7-bromo-2-ethyl-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-4-amine (0.58 g, 1.5 mmol) in N,N-dimethylformamide (10 mL). To the solution was added in succession, a solution of palladium acetate (37 mg, 0.15 mmol) and tri-ortho-tolylphosphine (91 mg, 0.3 mmol) in N,N-dimethylformamide (5 mL), triethylamine (3.0 eq. 0.6 mL), and a solution of N,N-dimethylacrylamide (178 mg, 1.8 mmol) in N,N-dimethylformamide (2 mL). The reaction mixture was purged with nitrogen. The vessel was sealed and heated at 120° C. for 18 hours. The reaction was cooled to ambient temperature and then concentrated to dryness under reduced pressure. The resulting solid was dissolved in dichloromethane (100 mL) and washed with saturated aqueous potassium carbonate (50 mL). The fractions were separated and the organic fraction was concentrated to dryness. The resulting off-white solid was purified by chromatography using a HORIZON HPFC system, (silica cartridge, 0-15% CMA/chloroform). A final recrystallization from acetonitrile provided 390 mg of (2E)-3-[4-amino-2-ethyl-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-7-yl]-N,N-dimethylprop-2-enamide as an off-white crystalline solid, mp>260° C.



1H NMR (500 MHz, CDCl3) δ 8.01 (d, J=8.5 Hz, 1H), 7.81 (d, J=1.6 Hz, 1H), 7.68 (dd, J=8.5, 1.9 Hz, 1H), 7.56 (d, J=15.1 Hz, 1H), 7.27 (d, J=15.4 Hz, 1H) 6.52 (s, 2H), 4.47-4.41 (m, 2H), 3.85-3.79 (m, 2H), 3.21-3.13 (m, 5H), 2.98-2.92 (m, 5H) 2.09 (s, 1H), 1.51-1.35 (m, 4H), 1.38 (t, J=7.6 Hz, 3H);


MS (APCI) m/z 408 (M+H)+;


Anal. Calcd. for C23H29N5O2: C, 67.79; H, 7.17; N, 17.19. Found: C, 67.55; H, 7.10; N, 17.05.


Example 129
3-[4-Amino-2-ethyl-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-7-yl]-N,N-dimethylpropanamide



embedded image


A glass Parr bottle was charged with 10% palladium on carbon (0.1 g) wetted with ethanol (10 mL) and a slurry of (2E)-3-[4-amino-2-ethyl-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-7-yl]-N,N-dimethylprop-2-enamide (0.32 g, 0.78 mmol) in methanol (200 mL). The vessel was placed on Parr apparatus, evacuated and charged with hydrogen (55 psi). The mixture was shaken at ambient temperature for 48 hours. The reaction mixture was filtered through a 0.2 micron PTFE membrane filter and the filter was rinsed with methanol (100 mL). The filtrate was concentrated to dryness under reduced pressure. The resulting solid was recrystallized from acetonitrile to provide 220 mg of 3-[4-amino-2-ethyl-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-7-yl]-N,N-dimethylpropanamide as a white crystalline solid, mp 219-220° C.



1H NMR (500 MHz, CDCl3) δ 7.92 (d, J=8.4 Hz, 1H), 7.45 (d, J=1.6 Hz, 1H), 7.16 (dd, J=8.3, 1.6 Hz, 1H), 6.36 (s, 2H), 4.43-4.37 (m, 2H), 3.84-3.78 (m, 2H), 3.19-3.12 (m, 2H), 2.96 (s, 3H), 2.95-2.89 (m, 4H), 2.83 (s, 3H), 2.68 (t, J=8.2 Hz, 2H), 2.08 (s, 1H), 1.51-1.34 (m, 4H), 1.37 (t, J=7.6 Hz, 3H);


MS (APCI) m/z 410 (M+H)+;


Anal. Calcd. for C23H31N5O2H2O: C, 64.61; H, 7.78; N, 16.38. Found: C, 64.63; H, 7.51; N, 16.23.


Example 130
2-Ethyl-7-[(1E)-3-morpholin-4-yl-3-oxoprop-1-enyl]-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-4-amine



embedded image


A thick walled glass vessel, equipped with a stir bar, was charged with a warmed solution of 7-bromo-2-ethyl-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-4-amine (584 mg, 1.5 mmol) in N,N-dimethylformamide (10 mL). To the solution was added in succession, a solution of palladium acetate (37 mg, 0.15 mmol) and tri-ortho-tolylphosphine (91 mg, 0.3 mmol) in N,N-dimethylformamide (5 mL), triethylamine (0.6 mL), and a solution of 4-acryloylmorpholine (254 mg 1.8 mmol) in N,N-dimethylformamide (2 mL). The reaction mixture was purged with nitrogen. The vessel was sealed and heated at 120° C. for 18 hours. The reaction was cooled to ambient temperature and then concentrated to dryness under reduced pressure. The resulting solid was dissolved in dichloromethane (100 mL) and washed with saturated aqueous potassium carbonate (50 mL). The fractions were separated and the organic fraction was concentrated to dryness under reduced pressure. The resulting solid was purified by chromatography using a HORIZON HPFC system (silica cartridge, 0-15% CMA/chloroform). A final recrystallization from acetonitrile provided 345 mg of 2-ethyl-7-[(1E)-3-morpholin-4-yl-3-oxoprop-1-enyl]-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-4-amine as a white crystalline solid, mp>260° C.



1H NMR (500 MHz, CDCl3) δ 8.01 (d, J=8.5 Hz, 1H), 7.83 (d, J=1.6 Hz, 1H), 7.70 (dd, J=8.5, 1.6 Hz, 1H), 7.61 d, J=15.4, 1H), 7.32 (d, J=15.4 Hz, 1H), 6.52 (s, 2H), 4.47-4.42 (m, 2H), 3.84-3.78 (m, 2H), 3.78-3.72 (m, 2H) 3.65-3.55 (m, 6H) 3.19-3.11 (m, 2H), 2.95 (q, J=7.3 Hz, 2H), 2.08 (s, 1H), 1.52-1.36 (m, 4H), 1.38 (t, J=7.6 Hz, 3H);


MS (APCI) m/z 450 (M+H)+.


Example 131
Ethyl (2E)-3-[4-amino-2-ethyl-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-7-yl]prop-2-enoate



embedded image


A thick walled glass vessel, equipped with a stir bar, was charged with a warmed solution of 7-bromo-2-ethyl-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-4-amine (2.5 g, 6.42 mmol) in N,N-dimethylformamide (50 mL). To the solution was added in succession, a solution of palladium acetate (144 mg, 0.642 mmol) and tri-ortho-tolylphosphine (390 mg, 1.28 mmol) in N,N-dimethylformamide (5 mL), triethylamine (2.7 mL) and a solution of ethyl acrylate (0.77 g, 7.7 mmol) in N,N-dimethylformamide (2 mL). The reaction mixture was purged with nitrogen. The vessel was sealed and heated at 120° C. for 18 hours. The reaction was cooled to ambient temperature and then concentrated to dryness under reduced pressure. The resulting solid was dissolved in dichloromethane (150 mL) and washed with saturated aqueous potassium carbonate (100 mL). The fractions were separated and the organic fraction was concentrated to dryness under reduced pressure. The resulting off-white solid was purified by chromatography using a HORIZON HPFC system (silica cartridge, 0-15% CMA/chloroform). A final recrystallization from acetonitrile provided 1.9 g of ethyl (2E)-3-[4-amino-2-ethyl-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-7-yl]prop-2-enoate as an off-white crystalline solid, mp 209-210° C.



1H NMR (500 MHz, CDCl3) δ 8.04 (d, J=8.5 Hz, 1H), 7.84 (d, J=1.6 Hz, 1H), 7.77 (d, J=16.1 Hz, 1H), 7.64 (dd, J=8.5, 1.6 Hz, 1H), 6.67 (d, J=16.1 Hz, 1H), 6.62 (s, 2H), 4.48-4.41 (m, 2H), 4.25-4.18 (m, 2H) 3.85-3.78 (m, 2H) 3.20-3.13 (m, 2H), 2.95 (q, J=7.6 Hz, 2H), 2.12-2.05 (m, 1H), 1.51-1.38 (m, 4H), 1.38 (t, J=7.6 Hz, 3H), 1.28 (t, J=7.3 Hz, 3H);


MS (APCI) m/z 409 (M+H)+;


Anal. Calcd. for C23H28N4O3: C, 67.63; H, 6.91; N, 13.72. Found: C, 67.58; H, 6.71; N, 13.98.


Example 132
Ethyl 3-[4-amino-2-ethyl-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-7-yl]propanoate



embedded image


A glass Parr bottle was charged with 10% palladium on carbon (0.2 g) wetted with ethanol (10 mL) and a slurry of ethyl (2E)-3-[4-amino-2-ethyl-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-7-yl]prop-2-enoate (1.81 g, 4.4 mmol) in ethanol (250 mL). The vessel was placed on a Parr apparatus, evacuated and charged with hydrogen (55 psi). The mixture was shaken at ambient temperature for 48 hours. The reaction mixture was filtered through a 0.2 micron PTFE membrane filter and the filter was rinsed with ethanol (300 ml). The filtrate was concentrated to dryness under reduced pressure. Recrystallization from acetonitrile provided 1.51 g of ethyl 3-[4-amino-2-ethyl-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-7-yl]propanoate as a white crystalline solid, mp 172-173° C.



1H NMR (500 MHz, CDCl3) δ 7.93 (d, J=8.5 Hz, 1H), 7.44 (d, J=1.7 Hz, 1H), 7.14 (dd, J=8.5, 1.7 Hz, 1H), 6.40 (s, 2H), 4.43-4.37 (m, 2H), 4.08-4.02 (m, 2H), 3.84-3.78 (m, 2H), 3.18-3.11 (m, 2H), 2.98-2.90 (m, 4H), 2.69 (t, J=7.6 Hz, 2H), 2.07 (m, 1H), 1.50-1.34 (m, 4H), 1.37 (t, J=7.6 Hz, 3H), 1.15 (t, J=7.3 Hz, 3H);



13C NMR (125 MHz, CDCl3) δ 172.7, 154.6, 152.1, 145.4, 138.9, 132.9, 126.5, 125.7, 122.3, 120.4, 113.5, 67.0, 60.2, 50.1, 36.1, 35.3, 30.7, 30.0, 20.5, 14.5, 12.4;


MS (APCI) m/z 411 (M+H)+;


Anal. Calcd. for C23H30N4O3: C, 67.29; H, 7.37; N, 13.65. Found: C, 67.25; H, 7.53; N, 13.71.


Example 133
4-Amino-2-(ethoxymethyl)-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinoline-7-carbaldehyde



embedded image



Part A


A round bottom flask, equipped with a stir bar, was charged with 1-[4-amino-7-bromo-2-(ethoxymethyl)-1H-imidazo[4,5-c]quinolin-1-yl]-2-methylpropan-2-ol (1.18 g, 3.0 mmol), 1-propanol (30 mL), potassium vinyltrifluoroborate (0.4 g, 3.0 mmol), triethylamine (1.25 mL, 9.0 mmol) and dichloro[1,1′-bis(diphenylphosphino)ferrocene]palladium (II) dichloromethane adduct (Pd(dppf)), (11 mg, 0.015 mmol). The reaction mixture was heated at 80° C. for 66 hours. The reaction mixture was cooled to ambient temperature and then concentrated to dryness under reduced pressure. The solid was dissolved in dichloromethane (100 mL) and washed with saturated aqueous potassium carbonate (50 mL). The fractions were separated and the organic fraction was concentrated to dryness. The off-white solid was purified by chromatography using a HORIZON HPFC system (silica cartridge, 0-15% CMA/chloroform). Recrystallization from acetonitrile provided 0.31 g of 1-[4-amino-2-(ethoxymethyl)-7-vinyl-1H-imidazo[4,5-c]quinolin-1-yl]-2-methylpropan-2-ol as an off-white crystalline solid. MS (APCI) m/z 341 (M+H)+.


Part B


A round bottom flask was charged with 1-[4-amino-2-(ethoxymethyl)-7-vinyl-1H-imidazo[4,5-c]quinolin-1-yl]-2-methylpropan-2-ol (0.26 g, 0.76 mmol) and dichloromethane (100 mL). The resulting solution was cooled in a dry ice/acetone bath. Ozone was bubbled through the reaction mixture for 10 minutes. The reaction mixture was purged with oxygen for 5 minutes followed by a nitrogen purge for an additional 10 minutes. A solution of triphenylphosphine (0.4 g, 1.52 mmol) in dichloromethane (10 mL) was added in one portion. The dry ice/acetone bath was removed and the reaction mixture was allowed to warm to ambient temperature for 18 hours. The reaction mixture was concentrated to dryness under reduced pressure. The resulting solid was purified by chromatography using a HORIZON HPFC system (silica cartridge, 0-15% CMA/chloroform). A final recrystallization from acetonitrile provided 30 mg of 4-amino-2-(ethoxymethyl)-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinoline-7-carbaldehyde as an off-white crystalline solid, mp 203-205° C.



1H NMR (500 MHz, CDCl3) δ 10.09 (s, 1H), 8.47 (d, J=8.5 Hz, 1H), 8.11 (d, J=1.6 Hz, 1H), 7.64 (dd, J=8.5, 1.6 Hz, 1H), 6.85 (s, 2H), 4.92-4.85 (m, 2H), 4.89 (s, 1H), 4.71 (s, 2H) 3.53 (q, J=7.1 Hz, 2H), 1.18 (br s, 6H), 1.14 (t, J=6.9 Hz, 3H);


MS (APCI) m/z 343 (M+H)+;


Anal. Calcd. for C18H22N4O3: C, 63.14; H, 6.48; N, 16.36. Found: C, 63.13; H, 6.46; N, 16.40.


Example 134
3-[4-Amino-2-(ethoxymethyl)-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]propan-1-ol



embedded image


A round bottom flask, equipped with a stir bar, was charged with ethyl 3-[4-amino-2-(ethoxymethyl)-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]propanoate (1.0 g, 2.4 mmol) and anhydrous tetrahydrofuran (20 mL). The reaction mixture was cooled in an ice bath. Lithium aluminum hydride (92 mg, 2.4 mmol) was added in one portion. After 2 hours additional lithium aluminum hydride (92 mg, 2.4 mmol) was added in one portion. The ice bath was removed and the reaction mixture was allowed to warm to ambient temperature. The reaction was quenched by successive dropwise addition of water (1 mL), 10% sodium hydroxide solution (1 mL) and water (3 mL). The reaction mixture was maintained at ambient temperature for 18 hours and then diluted with diethyl ether and water. The fractions were separated and the aqueous fraction was extracted with diethyl ether (3×25 mL). All of the organic fractions were combined and then concentrated to dryness under reduced pressure. Purification by chromatography using a HORIZON HPFC system (silica cartridge, 0-20% CMA/chloroform) provided 0.8 g of 3-[4-amino-2-(ethoxymethyl)-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]propan-1-ol as a white crystalline solid, mp 177-178° C.



1H NMR (500 MHz, CDCl3) δ 8.21 (d, J=8.5 Hz, 1H), 7.45 (s, 1H), 7.12 (d, J=8.5 Hz, 1H), 6.54 (s, 2H), 4.97 (br s, 2H), 4.94 (s, 1H), 4.71 (br s, 2H), 4.54 (t, J=5.2 Hz, 1H), 3.56 (q, J=6.9 Hz, 2H), 3.51 (q, J=6.3 Hz, 2H), 2.76 (t, J=7.7 Hz, 2H), 1.88-1.81 (m, 2H), 1.29-1.20 (m, 6H), 1.19 (t, J=7.1 Hz, 3H)



13C NMR (125 MHz, DMSO-d6) δ 152.3, 150.8, 145.9, 140.8, 134.6, 126.1, 125.6, 121.9, 121.5, 113.6, 71.0 65.6, 65.3, 60.6, 55.2, 34.5, 32.0, 28.1, 15.4 MS (APCI) m/z 373 (M+H)+;


Anal. calcd for C20H28N4O3.0.3H2O: C, 63.57; H, 7.63; N, 14.83. Found: C, 63.71; H, 7.32; N, 14.88.


Example 135
1-[4-Amino-2-(ethoxymethyl)-7-(3-morpholin-4-ylpropyl)-1H-imidazo[4,5-c]quinolin-1-yl]-2-methylpropan-2-ol



embedded image



Part A


A stirred solution of 3-[4-amino-2-(ethoxymethyl)-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]propan-1-ol (0.39 g, 1.05 mmol) in pyridine (10 mL) was cooled in an ice bath. 4-Dimethylaminopyridine (12.8 mg, 0.105 mmol) and methanesulfonic anhydride (204 mg, 1.15 mmol) were added. After stirring for one hour, the reaction was not complete and additional methanesulfonic anhydride was added (204 mg, 1.15 mmol). After another 2 hours of stirring the reaction was still incomplete and additional methanesulfonic anhydride was added (204 mg, 1.15 mmol). The cooling bath was removed and the reaction was stirred for 18 hours. The reaction mixture was concentrated to dryness under reduced pressure. The resulting solid was dissolved in dichloromethane (100 mL) and washed with saturated aqueous potassium carbonate (25 mL). The fractions were separated and the organic fraction was concentrated to provide 0.3 g of 3-[4-amino-2-(ethoxymethyl)-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]propyl methanesulfonate as a pale gold solid. MS (APCI) m/z 451 (M+H)+.


Part B


A round bottom flask, equipped with a stir bar, was charged with 3-[4-amino-2-(ethoxymethyl)-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]propyl methanesulfonate (0.24 g, 0.53 mmol) and acetonitrile (10 mL). Morpholine (0.23 g, 2.7 mmol) was added in one portion. The reaction mixture was heated at 75° C. for 18 hours and then cooled to ambient temperature. After concentrating under reduced pressure, the resulting solid was dissolved in dichloromethane (50 mL) and washed with saturated aqueous potassium carbonate (50 mL). The fractions were separated and the organic fraction was concentrated to dryness. The solid was purified by chromatography using a HORIZON HPFC system (silica cartridge, 0-25% CMA/chloroform). The material was further purified by a second chromatography treatment using a HORIZON HPFC system (silica cartridge, 10% methanol/dichloromethane). A final recrystallization from acetonitrile provided 0.1 g of 1-[4-amino-2-(ethoxymethyl)-7-(3-morpholin-4-ylpropyl)-1H-imidazo[4,5-c]quinolin-1-yl]-2-methylpropan-2-ol as a white crystalline solid, mp 132-133° C.



1H NMR (500 MHz, DMSO-d6) δ 8.16 (d, J=8.5 Hz, 1H), 7.39 (d, J=1.6 Hz, 1H), 7.07 (dd, J=8.4, 1.7 Hz, 1H), 6.48 (br s, 2H), 4.89 (br s, 2H), 4.87 (s, 1H), 4.65 (br s, 2H), 3.59-3.55 (m, 4H), 3.50 (q, J=4.7 Hz, 2H), 2.69 (t, J=7.3 Hz, 2H), 2.36-2.30 (m, 4H) 2.30 (t, J=7.3 Hz, 2H) 1.82-1.75 (m, 2H), 1.25-1.13 (m, 6H), 1.13 (t, J=7.3 Hz, 3H);


MS (APCI) m/z 442 (M+H)+;


Anal. calcd for C24H35N5O3.0.3H2O: C, 64.49; H, 8.03; N, 15.67. Found: C, 64.26; H, 8.16; N, 15.69.


Exemplary Compounds


Certain exemplary compounds, including some of those described above in the Examples, have the following Formulas (IIc, IId, IIe, or IIf) and the following R1 and R2 substituents, wherein each line of the table is matched with Formula IIc, IId, IIe, or IIf to represent a specific embodiment of the invention.
















IIc




embedded image








IId




embedded image








IIe




embedded image








IIf




embedded image















R1
R2







2-methylpropyl
methyl



2-hydroxy-2-methylpropyl
methyl



tetrahydro-2H-pyran-4-ylmethyl
methyl



3-isopropoxypropyl
methyl



2-methylpropyl
ethyl



2-hydroxy-2-methylpropyl
ethyl



tetrahydro-2H-pyran-4-ylmethyl
ethyl



3-isopropoxypropyl
ethyl



2-methylpropyl
n-propyl



2-hydroxy-2-methylpropyl
n-propyl



tetrahydro-2H-pyran-4-ylmethyl
n-propyl



3-isopropoxypropyl
n-propyl



2-methylpropyl
n-butyl



2-hydroxy-2-methylpropyl
n-butyl



tetrahydro-2H-pyran-4-ylmethyl
n-butyl



3-isopropoxypropyl
n-butyl



2-methylpropyl
2-methoxyethyl



2-hydroxy-2-methylpropyl
2-methoxyethyl



tetrahydro-2H-pyran-4-ylmethyl
2-methoxyethyl



3-isopropoxypropyl
2-methoxyethyl



2-methylpropyl
ethoxymethyl



2-hydroxy-2-methylpropyl
ethoxymethyl



tetrahydro-2H-pyran-4-ylmethyl
ethoxymethyl



3-isopropoxypropyl
ethoxymethyl



2-methylpropyl
2-hydroxyethyl



2-hydroxy-2-methylpropyl
2-hydroxyethyl



tetrahydro-2H-pyran-4-ylmethyl
2-hydroxyethyl



3-isopropoxypropyl
2-hydroxyethyl



2-methylpropyl
hydroxymethyl



2-hydroxy-2-methylpropyl
hydroxymethyl



tetrahydro-2H-pyran-4-ylmethyl
hydroxymethyl



3-isopropoxypropyl
hydroxymethyl



2-methylpropyl
methoxymethyl



2-hydroxy-2-methylpropyl
methoxymethyl



tetrahydro-2H-pyran-4-ylmethyl
methoxymethyl



3-isopropoxypropyl
methoxymethyl










Certain exemplary compounds, including some of those described above in the Examples, have the following Formulas (IIi, IIj, IIk, IIm, IIn, IIo, IIp, IIq, IIr, or IIs) and the following R1 and R2 substituents, wherein each line of the table is matched with Formula IIi, IIj, IIk, IIm, IIn, IIo, IIp, IIq, IIr, or IIs to represent a specific embodiment of the invention.
















IIi




embedded image








IIj




embedded image








IIk




embedded image














IIm




embedded image














IIn




embedded image








IIo




embedded image








IIp




embedded image








IIq




embedded image








IIr




embedded image








IIs




embedded image















R1
R2







2-methylpropyl
hydrogen



2-methylpropyl
methyl



2-methylpropyl
ethyl



2-methylpropyl
n-propyl



2-methylpropyl
n-butyl



2-methylpropyl
hydroxymethyl



2-methylpropyl
2-hydroxyethyl



2-methylpropyl
methoxymethyl



2-methylpropyl
ethoxymethyl



2-methylpropyl
2-methoxyethyl



2-hydroxy-2-methylpropyl
hydrogen



2-hydroxy-2-methylpropyl
methyl



2-hydroxy-2-methylpropyl
ethyl



2-hydroxy-2-methylpropyl
n-propyl



2-hydroxy-2-methylpropyl
n-butyl



2-hydroxy-2-methylpropyl
hydroxymethyl



2-hydroxy-2-methylpropyl
2-hydroxyethyl



2-hydroxy-2-methylpropyl
methoxymethyl



2-hydroxy-2-methylpropyl
ethoxymethyl



2-hydroxy-2-methylpropyl
2-methoxyethyl



tetrahydro-2H-pyran-4-ylmethyl
hydrogen



tetrahydro-2H-pyran-4-ylmethyl
methyl



tetrahydro-2H-pyran-4-ylmethyl
ethyl



tetrahydro-2H-pyran-4-ylmethyl
n-propyl



tetrahydro-2H-pyran-4-ylmethyl
n-butyl



tetrahydro-2H-pyran-4-ylmethyl
hydroxymethyl



tetrahydro-2H-pyran-4-ylmethyl
2-hydroxyethyl



tetrahydro-2H-pyran-4-ylmethyl
methoxymethyl



tetrahydro-2H-pyran-4-ylmethyl
ethoxymethyl



tetrahydro-2H-pyran-4-ylmethyl
2-methoxyethyl



2-methyl-2-[(methylsulfonyl)amino]propyl
hydrogen



2-methyl-2-[(methylsulfonyl)amino]propyl
methyl



2-methyl-2-[(methylsulfonyl)amino]propyl
ethyl



2-methyl-2-[(methylsulfonyl)amino]propyl
n-propyl



2-methyl-2-[(methylsulfonyl)amino]propyl
n-butyl



2-methyl-2-[(methylsulfonyl)amino]propyl
hydroxymethyl



2-methyl-2-[(methylsulfonyl)amino]propyl
2-hydroxyethyl



2-methyl-2-[(methylsulfonyl)amino]propyl
methoxymethyl



2-methyl-2-[(methylsulfonyl)amino]propyl
ethoxymethyl



2-methyl-2-[(methylsulfonyl)amino]propyl
2-methoxyethyl



2-[(methylsulfonyl)amino]ethyl
hydrogen



2-[(methylsulfonyl)amino]ethyl
methyl



2-[(methylsulfonyl)amino]ethyl
ethyl



2-[(methylsulfonyl)amino]ethyl
n-propyl



2-[(methylsulfonyl)amino]ethyl
n-butyl



2-[(methylsulfonyl)amino]ethyl
hydroxymethyl



2-[(methylsulfonyl)amino]ethyl
2-hydroxyethyl



2-[(methylsulfonyl)amino]ethyl
methoxymethyl



2-[(methylsulfonyl)amino]ethyl
ethoxymethyl



2-[(methylsulfonyl)amino]ethyl
2-methoxyethyl



4-[(methylsulfonyl)amino]butyl
hydrogen



4-[(methylsulfonyl)amino]butyl
methyl



4-[(methylsulfonyl)amino]butyl
ethyl



4-[(methylsulfonyl)amino]butyl
n-propyl



4-[(methylsulfonyl)amino]butyl
n-butyl



4-[(methylsulfonyl)amino]butyl
hydroxymethyl



4-[(methylsulfonyl)amino]butyl
2-hydroxyethyl



4-[(methylsulfonyl)amino]butyl
methoxymethyl



4-[(methylsulfonyl)amino]butyl
ethoxymethyl



4-[(methylsulfonyl)amino]butyl
2-methoxyethyl



2-{[(isopropylamino)carbonyl]amino}ethyl
hydrogen



2-{[(isopropylamino)carbonyl]amino}ethyl
methyl



2-{[(isopropylamino)carbonyl]amino}ethyl
ethyl



2-{[(isopropylamino)carbonyl]amino}ethyl
n-propyl



2-{[(isopropylamino)carbonyl]amino}ethyl
n-butyl



2-{[(isopropylamino)carbonyl]amino}ethyl
hydroxymethyl



2-{[(isopropylamino)carbonyl]amino}ethyl
2-hydroxyethyl



2-{[(isopropylamino)carbonyl]amino}ethyl
methoxymethyl



2-{[(isopropylamino)carbonyl]amino}ethyl
ethoxymethyl



2-{[(isopropylamino)carbonyl]amino}ethyl
2-methoxyethyl



2-(1,1-dioxidoisothiazolidin-2-yl)ethyl
hydrogen



2-(1,1-dioxidoisothiazolidin-2-yl)ethyl
methyl



2-(1,1-dioxidoisothiazolidin-2-yl)ethyl
ethyl



2-(1,1-dioxidoisothiazolidin-2-yl)ethyl
n-propyl



2-(1,1-dioxidoisothiazolidin-2-yl)ethyl
n-butyl



2-(1,1-dioxidoisothiazolidin-2-yl)ethyl
hydroxymethyl



2-(1,1-dioxidoisothiazolidin-2-yl)ethyl
2-hydroxyethyl



2-(1,1-dioxidoisothiazolidin-2-yl)ethyl
methoxymethyl



2-(1,1-dioxidoisothiazolidin-2-yl)ethyl
ethoxymethyl



2-(1,1-dioxidoisothiazolidin-2-yl)ethyl
2-methoxyethyl










Certain exemplary compounds, including some of those described above in the Examples, have the following Formula IIt and the following R2 and R3 substituents, wherein each line of the table is matched with Formula IIt to represent a specific embodiment of the invention.














IIt




embedded image















R2
R3







hydrogen
2-(acetylamino)ethyl



methyl
2-(acetylamino)ethyl



ethyl
2-(acetylamino)ethyl



n-propyl
2-(acetylamino)ethyl



n-butyl
2-(acetylamino)ethyl



hydroxymethyl
2-(acetylamino)ethyl



2-hydroxyethyl
2-(acetylamino)ethyl



methoxymethyl
2-(acetylamino)ethyl



ethoxymethyl
2-(acetylamino)ethyl



2-methoxyethyl
2-(acetylamino)ethyl



hydrogen
2-[(methylsulfonyl)amino]ethyl



methyl
2-[(methylsulfonyl)amino]ethyl



ethyl
2-[(methylsulfonyl)amino]ethyl



n-propyl
2-[(methylsulfonyl)amino]ethyl



n-butyl
2-[(methylsulfonyl)amino]ethyl



hydroxymethyl
2-[(methylsulfonyl)amino]ethyl



2-hydroxyethyl
2-[(methylsulfonyl)amino]ethyl



methoxymethyl
2-[(methylsulfonyl)amino]ethyl



ethoxymethyl
2-[(methylsulfonyl)amino]ethyl



2-methoxyethyl
2-[(methylsulfonyl)amino]ethyl



hydrogen
2-{[(isopropylamino)carbonyl]amino}ethyl



methyl
2-{[(isopropylamino)carbonyl]amino}ethyl



ethyl
2-{[(isopropylamino)carbonyl]amino}ethyl



n-propyl
2-{[(isopropylamino)carbonyl]amino}ethyl



n-butyl
2-{[(isopropylamino)carbonyl]amino}ethyl



hydroxymethyl
2-{[(isopropylamino)carbonyl]amino}ethyl



2-hydroxyethyl
2-{[(isopropylamino)carbonyl]amino}ethyl



methoxymethyl
2-{[(isopropylamino)carbonyl]amino}ethyl



ethoxymethyl
2-{[(isopropylamino)carbonyl]amino}ethyl



2-methoxyethyl
2-{[(isopropylamino)carbonyl]amino}ethyl



hydrogen
2-(benzoylamino)ethyl



methyl
2-(benzoylamino)ethyl



ethyl
2-(benzoylamino)ethyl



n-propyl
2-(benzoylamino)ethyl



n-butyl
2-(benzoylamino)ethyl



hydroxymethyl
2-(benzoylamino)ethyl



2-hydroxyethyl
2-(benzoylamino)ethyl



methoxymethyl
2-(benzoylamino)ethyl



ethoxymethyl
2-(benzoylamino)ethyl



2-methoxyethyl
2-(benzoylamino)ethyl



hydrogen
2-cyanoethyl



methyl
2-cyanoethyl



ethyl
2-cyanoethyl



n-propyl
2-cyanoethyl



n-butyl
2-cyanoethyl



hydroxymethyl
2-cyanoethyl



2-hydroxyethyl
2-cyanoethyl



methoxymethyl
2-cyanoethyl



ethoxymethyl
2-cyanoethyl



2-methoxyethyl
2-cyanoethyl



hydrogen
2-(aminocarbonyl)ethyl



methyl
2-(aminocarbonyl)ethyl



ethyl
2-(aminocarbonyl)ethyl



n-propyl
2-(aminocarbonyl)ethyl



n-butyl
2-(aminocarbonyl)ethyl



hydroxymethyl
2-(aminocarbonyl)ethyl



2-hydroxyethyl
2-(aminocarbonyl)ethyl



methoxymethyl
2-(aminocarbonyl)ethyl



ethoxymethyl
2-(aminocarbonyl)ethyl



2-methoxyethyl
2-(aminocarbonyl)ethyl



hydrogen
2-[(pyridin-3-ylcarbonyl)amino]ethyl



methyl
2-[(pyridin-3-ylcarbonyl)amino]ethyl



ethyl
2-[(pyridin-3-ylcarbonyl)amino]ethyl



n-propyl
2-[(pyridin-3-ylcarbonyl)amino]ethyl



n-butyl
2-[(pyridin-3-ylcarbonyl)amino]ethyl



hydroxymethyl
2-[(pyridin-3-ylcarbonyl)amino]ethyl



2-hydroxyethyl
2-[(pyridin-3-ylcarbonyl)amino]ethyl



methoxymethyl
2-[(pyridin-3-ylcarbonyl)amino]ethyl



ethoxymethyl
2-[(pyridin-3-ylcarbonyl)amino]ethyl



2-methoxyethyl
2-[(pyridin-3-ylcarbonyl)amino]ethyl










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 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 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 AHC3712) 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 (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.


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 30-0.014 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 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 (μmolar) is the lowest of the tested compound concentrations that induces a response over a fixed cytokine concentration for the tested cytokine (usually 20 pg/mL for IFN-α and 40 pg/mL for TNF-α). The maximal response is the maximal amount of cytokine (pg/ml) produced in the dose-response.


The complete disclosures of the patents, patent documents, and publications cited herein are incorporated by reference in their entirety as if each were individually incorporated. Various modifications and alterations to this invention will become apparent to those skilled in the art without departing from the scope and spirit of this invention. It should be understood that this invention is not intended to be unduly limited by the illustrative embodiments and examples set forth herein and that such examples and embodiments are presented by way of example only with the scope of the invention intended to be limited only by the claims set forth herein as follows.

Claims
  • 1. A compound of the Formula II:
  • 2. The compound or salt of claim 1 wherein n is 0.
  • 3. The compound or salt of claim 1 wherein R3 is -Z-Y-R4.
  • 4. The compound or salt of claim 3 wherein Y is —N(R8)-Q-, and R4 is alkyl, or heterocyclyl.
  • 5. The compound or salt of claim 1 wherein Z is C1-4 alkylene.
  • 6. The compound or salt of claim 1 wherein R3 is at the 7-position.
  • 7. A pharmaceutical composition comprising a therapeutically effective amount of a compound or salt of claim 1 and a pharmaceutically acceptable carrier.
  • 8. The compound or salt of claim 1 wherein R1 is selected from the group consisting of 2-hydroxy-2-methylpropyl, 2-methylpropyl, propyl, ethyl, methyl, 2,3-dihydroxypropyl, 3-isopropoxypropyl, 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, and tetrahydro-2H-pyran-4-ylmethyl.
  • 9. The compound or salt of claim 1 wherein R2 is selected from the group consisting of hydrogen, methyl, ethyl, propyl, butyl, ethoxymethyl, methoxymethyl, 2-methoxyethyl, hydroxymethyl, and 2-hydroxyethyl.
  • 10. The compound or salt of claim 4 wherein Q is —C(R6)—, —S(O)2—, or —C(R6)—N(R8)—.
  • 11. The compound or salt of claim 1 wherein R1 is selected from the group consisting of alkyl, hydroxyalkylenyl, alkoxyalkylenyl, and alkyl substituted by tetrahydropyranyl.
  • 12. The compound or salt of claim 1 wherein R1 is alkyl substituted by —NH—S(O)2-alkyl.
  • 13. The compound or salt of claim 1 wherein R1 is selected from the group consisting of 2-hydroxy-2-methylpropyl, 2-methylpropyl, 4-[(methylsulfonyl)amino]butyl, 2-methyl-2-[(methylsulfonyl)amino]propyl, tetrahydro-2H-pyran-4-ylmethyl, and 3-isopropoxypropyl.
  • 14. The compound or salt of claim 11 wherein R1 is selected from the group consisting of 2-hydroxy-2-methylpropyl, 2-methylpropyl, tetrahydro-2H-pyran-4-ylmethyl, and 3-isopropoxypropyl.
  • 15. A compound selected from the group consisting of: 2-ethyl-7-[2-(methylsulfonyl)ethyl]-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-4-amine;N-{3-[4-amino-2-ethyl-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-7-yl]propyl}methane sulfonamide;N-{3-[4-amino-2-ethyl-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-7-yl]propyl}acetamide;N-{3-[4-amino-2-ethyl-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-7-yl]propyl}-N′-isopropylurea;3-[4-amino-2-ethyl-1-(tetrahydro-2H-pyran-4-ylmethyl)-1H-imidazo[4,5-c]quinolin-7-yl]-N,N-dimethylpropanamide;
  • 16. A compound selected from the group consisting of: 3-[4-amino-2-ethyl-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]-1-(morpholin-4-yl)propan-1-one;1-{4-amino-7-[2-(methylsulfonyl)ethyl]-1H-imidazo[4,5-c]quinolin-1-yl}-2-methylpropan-2-ol;3-[4-amino-2-ethyl-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]-N,N-dimethylpropanamide;1-{4-amino-2-(methoxymethyl)-7-[2-(methylsulfonyl)ethyl]-1H-imidazo[4,5-c]quinolin-1-yl}-2-methylpropan-2-ol;1-{4-amino-2-(hydroxymethyl)-7-[2-(methylsulfonyl)ethyl]-1H-imidazo[4,5-c]quinolin-1-yl}-2-methylpropan-2-ol;1-[4-amino-2-(methoxymethyl)-7-(3-morpholin-4-yl-3-oxopropyl)-1H-imidazo[4,5-c]quinolin-1-yl]-2-methylpropan-2-ol;1-[4-amino-7-[3-(1,1-dioxidothiomorpholin-4-yl)-3-oxopropyl]-2-(methoxymethyl)-1H-imidazo[4,5-c]quinolin-1-yl]-2-methylpropan-2-ol;3-[4-amino-1-(2-hydroxy-2-methylpropyl)-2-(methoxymethyl)-1H-imidazo[4,5-c]quinolin-7-yl]-N,N-dimethylpropanamide;N-{3-[4-amino-1-(2-hydroxy-2-methylpropyl)-2-(methoxymethyl)-1H-imidazo[4,5-c]quinolin-7-yl]propyl}methanesulfonamide;3-[4-amino-2-(hydroxymethyl)-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]-N,N-dimethylpropanamide;1-[4-amino-2-(hydroxymethyl)-7-(3-morpholin-4-yl-3-oxopropyl)-1H-imidazo[4,5-c]quinolin-1-yl]-2-methylpropan-2-ol;3-[4-amino-1-(2-hydroxy-2-methylpropyl)-2-(methoxymethyl)-1H-imidazo[4,5-c]quinolin-7-yl]propanamide;1-[4-amino-7-[3-(1,1-dioxidothiomorpholin-4-yl)-3-oxopropyl]-2-(hydroxymethyl)-1H-imidazo[4,5-c]quinolin-1-yl]-2-methylpropan-2-ol;N-{3-[4-amino-1-(2-hydroxy-2-methylpropyl)-2-(methoxymethyl)-1H-imidazo[4,5-c]quinolin-7-yl]propyl}acetamide;N-{3-[4-amino-2-(hydroxymethyl)-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]propyl}methanesulfonamide;N-{2-[4-amino-2-ethyl-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]ethyl}methanesulfonamide;1-[4-amino-2-(ethoxymethyl)-7-(3-morpholin-4-yl-3-oxopropyl)-1H-imidazo[4,5-c]quinolin-1-yl]-2-methylpropan-2-ol;
  • 17. A compound selected from the group consisting of: 1-{3-[4-amino-2-(2-methoxyethyl)-8-(3-morpholin-4-yl-3-oxo-propyl)-1H-imidazo[4,5-c]quinolin-1-yl]propyl}pyrrolidin-2-one;3-{4-amino-2-(2-methoxyethyl)-1-[3-(2-oxo-pyrrolidin-1-yl)propyl]-1H-imidazo[4,5-c]quinolin-8-yl}-N,N-dimethylpropanamide;N-[4-amino-2-ethoxymethyl-1-(2-hydroxy-2-methylpropyl)-1H-imidazo[4,5-c]quinolin-7-yl]ethanesulfonamide;
  • 18. The compound or salt of claim 4 wherein R8 is hydrogen; Q is —C(O)—, —S(O)2—, or —C(O)—NH—; and R4 is alkyl.
  • 19. The compound or salt of claim 1 wherein R3 is -Z-R5.
  • 20. The compound or salt of claim 19 wherein Z is C2-4 alkylene and R5 is
CROSS REFERENCE TO RELATED APPLICATIONS

This application is a national stage filing under 35 U.S.C. §371 of PCT International application PCT/US2006/004713 designating the United States of America, and filed Feb. 10, 2006. This application claims the benefit under 35 U.S.C. §119(e) of U.S. provisional application Ser. No. 60/652,239, filed Feb. 11, 2005.

PCT Information
Filing Document Filing Date Country Kind 371c Date
PCT/US2006/004713 2/10/2006 WO 00 8/25/2008
Publishing Document Publishing Date Country Kind
WO2006/091394 8/31/2006 WO A
US Referenced Citations (271)
Number Name Date Kind
3314941 Littell et al. Apr 1967 A
3698348 Morgan Oct 1972 A
4689338 Gerster Aug 1987 A
4698348 Gerster Oct 1987 A
4929624 Gerster et al. May 1990 A
4988815 Andre et al. Jan 1991 A
5037986 Gerster Aug 1991 A
5175296 Gerster Dec 1992 A
5238944 Wick et al. Aug 1993 A
5266575 Gerster Nov 1993 A
5268376 Gester Dec 1993 A
5346905 Gerster Sep 1994 A
5352784 Nikolaides et al. Oct 1994 A
5367076 Gerster Nov 1994 A
5389640 Gerster et al. Feb 1995 A
5395937 Nikolaides et al. Mar 1995 A
5444065 Nikolaides et al. Aug 1995 A
5446153 Lindstrom et al. Aug 1995 A
5482936 Lindstrom Jan 1996 A
5494916 Lindstrom et al. Feb 1996 A
5525612 Gerster Jun 1996 A
5605899 Gerster et al. Feb 1997 A
5627281 Nikolaides et al. May 1997 A
5644063 Lindstrom et al. Jul 1997 A
5648516 Nikolaides et al. Jul 1997 A
5693811 Lindstrom Dec 1997 A
5714608 Gerster Feb 1998 A
5741908 Gerster et al. Apr 1998 A
5741909 Gerster et al. Apr 1998 A
5756747 Gerster et al. May 1998 A
5886006 Nikolaides et al. Mar 1999 A
5939090 Beaurline et al. Aug 1999 A
5977366 Gerster et al. Nov 1999 A
6039969 Tomai et al. Mar 2000 A
6069149 Nanba et al. May 2000 A
6083505 Miller et al. Jul 2000 A
6110929 Gerster et al. Aug 2000 A
6194425 Gerster et al. Feb 2001 B1
6200592 Tomai et al. Mar 2001 B1
6245776 Skwierczynski et al. Jun 2001 B1
6331539 Crooks et al. Dec 2001 B1
6348462 Gerster et al. Feb 2002 B1
6365166 Beaurline et al. Apr 2002 B2
6376669 Rice et al. Apr 2002 B1
6440992 Gerster et al. Aug 2002 B1
6451810 Coleman et al. Sep 2002 B1
6486168 Skwierczynski et al. Nov 2002 B1
6514985 Gerster et al. Feb 2003 B1
6518265 Kato et al. Feb 2003 B1
6525064 Dellaria et al. Feb 2003 B1
6541485 Crooks et al. Apr 2003 B1
6545016 Dellaria et al. Apr 2003 B1
6545017 Dellaria et al. Apr 2003 B1
6558951 Tomai et al. May 2003 B1
6573273 Crooks et al. Jun 2003 B1
6610319 Tomai et al. Aug 2003 B2
6627638 Gerster et al. Sep 2003 B2
6627640 Gerster et al. Sep 2003 B2
6630588 Rice et al. Oct 2003 B2
6638944 Mickelson Oct 2003 B2
6656938 Crooks et al. Dec 2003 B2
6660735 Crooks et al. Dec 2003 B2
6660747 Crooks et al. Dec 2003 B2
6664260 Charles et al. Dec 2003 B2
6664264 Dellaria et al. Dec 2003 B2
6664265 Crooks et al. Dec 2003 B2
6667312 Bonk et al. Dec 2003 B2
6670372 Charles et al. Dec 2003 B2
6677347 Crooks et al. Jan 2004 B2
6677348 Heppner et al. Jan 2004 B2
6677349 Griesgraber Jan 2004 B1
6683088 Crooks et al. Jan 2004 B2
6696076 Tomai et al. Feb 2004 B2
6696465 Dellaria et al. Feb 2004 B2
6703402 Gerster et al. Mar 2004 B2
6706728 Hedenstrom et al. Mar 2004 B2
6716988 Dellaria et al. Apr 2004 B2
6720333 Dellaria et al. Apr 2004 B2
6720334 Dellaria et al. Apr 2004 B2
6720422 Dellaria et al. Apr 2004 B2
6743920 Lindstrom et al. Jun 2004 B2
6756382 Coleman et al. Jun 2004 B2
6797718 Dellaria et al. Sep 2004 B2
6800624 Crooks et al. Oct 2004 B2
6809203 Gerster et al. Oct 2004 B2
6818650 Griesgraber Nov 2004 B2
6825350 Crooks et al. Nov 2004 B2
6841678 Merli et al. Jan 2005 B2
6852861 Merli et al. Feb 2005 B2
6878719 Lindstrom et al. Apr 2005 B2
6888000 Crooks et al. May 2005 B2
6894060 Slade May 2005 B2
6897221 Crooks et al. May 2005 B2
6903113 Heppner et al. Jun 2005 B2
6916925 Rice et al. Jul 2005 B1
6921826 Dellaria et al. Jul 2005 B2
6924293 Lindstrom Aug 2005 B2
6943225 Lee et al. Sep 2005 B2
6949649 Bonk et al. Sep 2005 B2
6953804 Dellaria et al. Oct 2005 B2
6969722 Heppner et al. Nov 2005 B2
6989389 Heppner et al. Jan 2006 B2
7030129 Miller et al. Apr 2006 B2
7030131 Crooks et al. Apr 2006 B2
7038053 Lindstrom et al. May 2006 B2
7049439 Crooks et al. May 2006 B2
7078523 Crooks et al. Jul 2006 B2
7091214 Hays et al. Aug 2006 B2
7098221 Heppner et al. Aug 2006 B2
7112677 Griesgraber Sep 2006 B2
7115622 Crooks et al. Oct 2006 B2
7125890 Dellaria et al. Oct 2006 B2
7132429 Griesgraber et al. Nov 2006 B2
7132438 Frenkel et al. Nov 2006 B2
7148232 Gerster et al. Dec 2006 B2
7157453 Crooks et al. Jan 2007 B2
7163947 Griesgraber et al. Jan 2007 B2
7179253 Graham et al. Feb 2007 B2
7199131 Lindstrom Apr 2007 B2
7214675 Griesgraber May 2007 B2
7220758 Dellaria et al. May 2007 B2
7226928 Mitra et al. Jun 2007 B2
7276515 Dellaria et al. Oct 2007 B2
7288550 Dellaria et al. Oct 2007 B2
7301027 Colombo et al. Nov 2007 B2
7375180 Gorden et al. May 2008 B2
7387271 Noelle et al. Jun 2008 B2
7393859 Coleman et al. Jul 2008 B2
7427629 Kedl et al. Sep 2008 B2
7485432 Fink et al. Feb 2009 B2
7544697 Hays et al. Jun 2009 B2
7576068 Averett Aug 2009 B2
7578170 Mayer et al. Aug 2009 B2
7579359 Krepski et al. Aug 2009 B2
7598382 Hays et al. Oct 2009 B2
7612083 Griesgraber Nov 2009 B2
7648997 Kshirsagar et al. Jan 2010 B2
7655672 Statham et al. Feb 2010 B2
7687628 Gutman et al. Mar 2010 B2
7696159 Owens et al. Apr 2010 B2
7699057 Miller et al. Apr 2010 B2
7731967 O'Hagan et al. Jun 2010 B2
7799800 Wightman Sep 2010 B2
7879849 Hays et al. Feb 2011 B2
7884207 Stoermer et al. Feb 2011 B2
7888349 Kshirsagar et al. Feb 2011 B2
7897597 Lindstrom et al. Mar 2011 B2
7897609 Niwas et al. Mar 2011 B2
7897767 Kshirsagar et al. Mar 2011 B2
7902209 Statham et al. Mar 2011 B2
7902210 Statham et al. Mar 2011 B2
7902211 Statham et al. Mar 2011 B2
7902212 Statham et al. Mar 2011 B2
7902213 Statham et al. Mar 2011 B2
7902214 Statham et al. Mar 2011 B2
7902215 Statham et al. Mar 2011 B2
7902216 Statham et al. Mar 2011 B2
7902242 Statham et al. Mar 2011 B2
7902243 Statham et al. Mar 2011 B2
7902244 Statham et al. Mar 2011 B2
7902245 Statham et al. Mar 2011 B2
7902246 Statham et al. Mar 2011 B2
7906506 Griesgraber et al. Mar 2011 B2
7915281 Moser et al. Mar 2011 B2
7939526 Radmer et al. May 2011 B2
7943609 Griesgraber et al. May 2011 B2
7968562 Skwierczynski et al. Jun 2011 B2
7968563 Kshirsagar et al. Jun 2011 B2
7993659 Noelle et al. Aug 2011 B2
8017779 Merrill et al. Sep 2011 B2
8026366 Prince et al. Sep 2011 B2
8034938 Griesgraber et al. Oct 2011 B2
20020055517 Smith May 2002 A1
20020058674 Hedenstrom et al. May 2002 A1
20020107262 Lindstrom Aug 2002 A1
20030133913 Tomai et al. Jul 2003 A1
20030139364 Krieg et al. Jul 2003 A1
20040014779 Gorden et al. Jan 2004 A1
20040132079 Gupta et al. Jul 2004 A1
20040175336 Egging et al. Sep 2004 A1
20040180919 Lee et al. Sep 2004 A1
20040191833 Fink et al. Sep 2004 A1
20040197865 Gupta et al. Oct 2004 A1
20040202720 Wightman et al. Oct 2004 A1
20040214851 Birmachu et al. Oct 2004 A1
20040258698 Wightman et al. Dec 2004 A1
20040265351 Miller et al. Dec 2004 A1
20050048072 Kedl et al. Mar 2005 A1
20050059072 Birmachu et al. Mar 2005 A1
20050070460 Hammerbeck et al. Mar 2005 A1
20050096259 Tomai et al. May 2005 A1
20050106300 Chen et al. May 2005 A1
20050158325 Hammerbeck et al. Jul 2005 A1
20050165043 Miller et al. Jul 2005 A1
20050171072 Tomai et al. Aug 2005 A1
20050239735 Miller et al. Oct 2005 A1
20050245562 Garcia-Echeverria et al. Nov 2005 A1
20060045885 Kedl et al. Mar 2006 A1
20060045886 Kedl Mar 2006 A1
20060051374 Miller et al. Mar 2006 A1
20060088542 Braun Apr 2006 A1
20060142202 Alkan et al. Jun 2006 A1
20060142235 Miller et al. Jun 2006 A1
20060195067 Wolter et al. Aug 2006 A1
20060216333 Miller et al. Sep 2006 A1
20070060754 Lindstrom et al. Mar 2007 A1
20070066639 Kshirsagar et al. Mar 2007 A1
20070072893 Krepski et al. Mar 2007 A1
20070099901 Krepski et al. May 2007 A1
20070123559 Statham et al. May 2007 A1
20070166384 Zarraga et al. Jul 2007 A1
20070167479 Busch et al. Jul 2007 A1
20070213355 Capraro et al. Sep 2007 A1
20070219196 Krepski et al. Sep 2007 A1
20070243215 Miller et al. Oct 2007 A1
20070259881 Dellaria et al. Nov 2007 A1
20070259907 Prince Nov 2007 A1
20070292456 Hammerbeck et al. Dec 2007 A1
20080015184 Kshirsagar et al. Jan 2008 A1
20080039533 Sahouani et al. Feb 2008 A1
20080063714 Sahouani et al. Mar 2008 A1
20080119508 Slade et al. May 2008 A1
20080188513 Skwierczynski et al. Aug 2008 A1
20080193468 Levy et al. Aug 2008 A1
20080193474 Griesgraber et al. Aug 2008 A1
20080207674 Stoesz et al. Aug 2008 A1
20080213308 Valiante et al. Sep 2008 A1
20080262021 Capraro et al. Oct 2008 A1
20080262022 Lee et al. Oct 2008 A1
20080306252 Crooks et al. Dec 2008 A1
20080306266 Martin et al. Dec 2008 A1
20080312434 Lindstrom et al. Dec 2008 A1
20080318998 Prince et al. Dec 2008 A1
20090005371 Rice et al. Jan 2009 A1
20090017076 Miller et al. Jan 2009 A1
20090023720 Kshirsagar et al. Jan 2009 A1
20090023722 Coleman et al. Jan 2009 A1
20090029988 Kshirsagar et al. Jan 2009 A1
20090030030 Bonk et al. Jan 2009 A1
20090030031 Kshirsagar et al. Jan 2009 A1
20090035323 Stoermer et al. Feb 2009 A1
20090062272 Bonk et al. Mar 2009 A1
20090069299 Merrill et al. Mar 2009 A1
20090069314 Kshirsagar et al. Mar 2009 A1
20090075980 Hays et al. Mar 2009 A1
20090099161 Rice et al. Apr 2009 A1
20090105295 Kshirsagar et al. Apr 2009 A1
20090124611 Hays et al. May 2009 A1
20090124652 Ach et al. May 2009 A1
20090163532 Perman et al. Jun 2009 A1
20090163533 Hays et al. Jun 2009 A1
20090176821 Kshirsagar et al. Jul 2009 A1
20090202443 Miller et al. Aug 2009 A1
20090221551 Kshirsagar et al. Sep 2009 A1
20090221556 Kshirsagar et al. Sep 2009 A1
20090240055 Krepski et al. Sep 2009 A1
20090246174 Rook et al. Oct 2009 A1
20090253695 Kshirsagar et al. Oct 2009 A1
20090270443 Stoermer et al. Oct 2009 A1
20090298821 Kshirsagar et al. Dec 2009 A1
20090306388 Zimmerman et al. Dec 2009 A1
20100028381 Gorski et al. Feb 2010 A1
20100056557 Benninghoff et al. Mar 2010 A1
20100096287 Stoesz et al. Apr 2010 A1
20100113565 Gorden et al. May 2010 A1
20100152230 Dellaria et al. Jun 2010 A1
20100158928 Stoermer et al. Jun 2010 A1
20100173906 Griesgraber Jul 2010 A1
20100180902 Miller et al. Jul 2010 A1
20100240693 Lundquist et al. Sep 2010 A1
20110021554 Stoesz et al. Jan 2011 A1
Foreign Referenced Citations (18)
Number Date Country
0385630 Sep 1990 EP
0 394 026 Oct 1990 EP
1 104 764 Jun 2001 EP
9-176116 Jul 1997 JP
9-208584 Aug 1997 JP
11-080156 Mar 1999 JP
11-222432 Aug 1999 JP
2000-247884 Sep 2000 JP
WO 9305042 Mar 1993 WO
WO 9502598 Jan 1995 WO
WO 0236592 May 2002 WO
WO2004058759 Jul 2004 WO
WO 2005003084 Jan 2005 WO
WO 2006028451 Mar 2006 WO
WO2006028451 Mar 2006 WO
WO 2006063072 Jun 2006 WO
WO 2006121528 Nov 2006 WO
WO 2007030775 Mar 2007 WO
Non-Patent Literature Citations (11)
Entry
Federal Registry p. 1766, Federal Register / vol. 76, No. 27 / Wednesday, Feb. 9, 2011.
Wozniak et al., “The Amination of 3-nitro-1, 5-napthyridines by Liquid Ammonia/Potassium Permanganate1,2. A New and Convenient Amination Method.”, Journal of the Royal Netherlands Chemical Society, 102, pp. 511-513, Dec. 12, 1983.
Brennan et al., “Automated Bioassay of Interferons in Micro-test Plates,”, Biotechniques, Jun./Jul. 78, 1983.
Testerman et al., “Cytokine Induction by the Immunomodulators Imiquimod and S-27609.”,Journal of Leukocyte Biology, vol. 58, pp. 365-372, Sep. 1995.
Bachman et al., “Synthesis of Substituted Quinolylamines. Derivatives of 4-Amino-7-Chloroquinoline.”, J. Org. Chem. 15, pp. 1278-1284 (1950).
Jain et al., “Chemical and Pharmacological Investigations of Some ω-Substituted Alkylamino-3-aminopryidines.”, J. Med. Chem., 11, pp. 87-92 (1968).
Baranov et al., “Pyrazoles, Imidazoles, and Other 5-Membered Rings.”, Chem. Abs. 85, 94362, (1976).
Berényi et al., “Ring Transformation of Condensed Dihydro-as-triazines.”, J. Heterocyclic Chem., 18, pp. 1537-1540 (1981).
Chollet et al., “Development of a Topically Active Imiquimod Formulation.”, Pharmaceutical Development and Technology, 4(1), pp. 35-43 (1999).
Izumi et al., “1H-Imidazo[4,5-c]quinoline Derivatives as Novel Potent TNF-α Suppressors: Synthesis and Structure-Activity Relationship of 1-, 2- and 4-Substituted 1H-imidazo[4,5-c]pyridines.”, Bioorganic & Medicinal Chemistry, 11, pp. 2541-2550 (2003).
Torrence and DeClerq, ‘Inducers and Induction of Interferons’ Pharmac Ther, 2, (1977) pp. 1-88.
Related Publications (1)
Number Date Country
20090099161 A1 Apr 2009 US
Provisional Applications (1)
Number Date Country
60652239 Feb 2005 US