INDENOISOQUINOLINONE ANALOGS AND METHODS OF USE THEREOF

The present invention relates to Indenoisoquinolinone Analogs, compositions comprising an effective amount of an Indenoisoquinolinone Analog and methods for treating or preventing an inflammatory disease, a reperfusion injury, diabetes mellitus, a diabetic complication, reoxygenation injury resulting from organ transplantation, an ischemic condition, a neurodegenerative disease, renal failure, a vascular disease, a cardiovascular disease, cancer, a complication of prematurity, cardiomyopathy, retinopathy, nephropathy, neuropathy, erectile dysfunction or urinary incontinence, comprising administering to a subject in need thereof an effective amount of an Indenoisoquinolinone Analog.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
1. REFERENCE TO RELATED APPLICATIONS

This application claims the benefit of U.S. Provisional Application No. 60/710,769, filed Aug. 24, 2005, which is incorporated by reference herein in its entirety.

2. FIELD OF THE INVENTION

The present invention relates to Indenoisoquinolinone Analogs, compositions comprising an effective amount of an Indenoisoquinolinone Analog and methods for treating or preventing an inflammatory disease, a reperfusion injury, diabetes mellitus, a diabetic complication, reoxygenation injury resulting from organ transplantation, an ischemic condition, a neurodegenerative disease, renal failure, a vascular disease, a cardiovascular disease, cancer, a complication of prematurity, cardiomyopathy, retinopathy, nephropathy, neuropathy, erectile dysfunction or urinary incontinence, comprising administering to a subject in need thereof an effective amount of an Indenoisoquinolinone Analog.

3. BACKGROUND OF THE INVENTION

Erectile dysfunction (“ED”) is a significant male-health issue. While estimating its prevalence is difficult, estimates range from about 15 million to 30 million sufferers worldwide.

The etiology of erectile dysfunction can be multiple, and can include mechanical trauma to the nerves (such as during prostatectomy), or it can be due to diabetes mellitus, cardiovascular diseases, induced by radiation, certain drugs, or advanced age.

Urinary incontinence affects people of all ages and levels of physical health, both in health care settings and in the community at large. Persons suffering from urinary incontinence can be predisposed to also having urinary-tract infections, pressure ulcers, perineal rashes and urosepsis. Psychosocially, urinary incontinence can be associated with embarrassment, social stigmatization, depression and a risk of institutionalization (Herzo et al., Annu. Rev. Gerontol. Geriatr. 9:74 (1989)).

An inflammatory disease, such as arthritis, colitis, and autoimmune diabetes, typically manifests itself as a disorder distinct from that associated with a reperfusion injury, e.g., stroke and heart attack, and can clinically manifest itself as a different entity. However, there can be common underlying mechanisms between these two types of disorders. Specifically, inflammatory disease and reperfusion injury can induce proinflammatory cytokine and chemokine synthesis which can, in turn, result in production of cytotoxic free radicals such as nitric oxide and superoxide. NO and superoxide can react to form peroxynitrite (ONOO) (Szabó et al., Shock 6:79-88, 1996).

The ONOO-induced cell necrosis observed in inflammatory disease and in reperfusion injury involves the activation of the nuclear enzyme poly (ADP-ribose) polymerase (PARP). Activation of PARP is thought to be an important step in the cell-mediated death observed in inflammation and reperfusion injury (Szabó et al., Trends Pharmacol. Sci. 19:287-98, 1998).

A number of PARP inhibitors have been described in the art. See, e.g., Banasik et al., J. Biol. Chem., 267:1569-75, 1992, and Banasik et al., Mol. Cell. Biochem., 138:185-97, 1994; WO 00/39104; WO 00/39070; WO 99/59975; WO 99/59973; WO 99/11649; WO 99/11645; WO 99/11644; WO 99/11628; WO 99/11623; WO 99/11311; WO 00/42040; Zhang et al., Biochem. Biophys. Res. Commun., 278:590-98, 2000; White et al., J. Med. Chem., 43:4084-4097, 2000; Griffin et al., J. Med. Chem., 41:5247-5256, 1998; Shinkwin et al., Bioorg. Med. Chem., 7:297-308, 1999; and Soriano et at., Nature Medicine, 7:108-113, 2001. Adverse effects associated with administration of PARP inhibitors have been discussed in Milan et al., Science, 223:589-591, 1984.

Indenoisoquinolinone compounds have been previously discussed in the art. For example, cytotoxic non-camptothecin topoisomerase I inhibitors are reported in Cushman et al., J. Med. Chem., 43:3688-3698, 2300 and Cushman et al., J. Med. Chem. 42:446-57, 1999; indeno[1,2-c]isoquinolines are reported as antineoplastic agents in Cushman et al., WO 00/21537; and as neoplasm inhibitors in Hrbata et al., WO 93/05023.

Syntheses of indenoisoquinolinone compounds have been reported. For example, see Wawzonek et al., Org. Prep. Proc. Int., 14:163-8, 1982; Wawzonek et al., Can. J. Chem., 59:2833, 1981; Andoi et al., Bull. Chem. Soc. Japan, 47:1014-17, 1974; Dusemund et al., Arch. Pharm (Weinheim, Ger.), 3 17:381-2, 1984; and Lal et al., Indian J. Chem., Sect. B, 38B:33-39, 1999.

There remains, however, a need in the art for compounds useful for treating or preventing an inflammatory disease, a reperfusion injury, diabetes mellitus, a diabetic complication, reoxygenation injury resulting from organ transplantation, an ischemic condition, a neurodegenerative disease, renal failure, a vascular disease, a cardiovascular disease, cancer, a complication of prematurity, cardiomyopathy, retinopathy, nephropathy, neuropathy, erectile dysfunction or urinary incontinence.

Citation of any reference in Section 3 of this application is not an admission that the reference is prior art.

4. SUMMARY OF THE INVENTION

In one aspect the invention provides a compound of Formula (I)

and pharmaceutically acceptable salts thereof,
wherein

X is —CH2—, —O—, —C(O)—, —NH— or —S—;

R1 is —(CH2)n—N(R2)(R2), —O—(CH2)m—N(R2)(R2), —(CH2)n—OR3 or —O—(CH2)m—OR3;

each R2 is independently —H, —C1-C6 alkyl, —C3-C8 monocyclic cycloalkyl, —C1-C6 alkylene phenyl, phenyl, an N-terminal alpha amino acid residue, an N-terminal alpha amino hydroxymethyl residue, a C1-C6 alkyl ester of an N-terminal alpha amino acid residue, or a nitrogen-containing 3- to 7-membered monocyclic heterocycle, each of which other than hydrogen is independently unsubstituted or substituted with one or more of -halo, —OH, —OP(O)(OH)2, —OS(O)2OH, —C1-C6 alkyl, —O—C1-C6 alkyl, —C3-C8 monocyclic cycloalkyl, a 3- to 7-membered monocyclic heterocycle, —C1-C6 alkyl-substituted 3- to 7- membered monocyclic heterocycle, or —N(Z3)(Z4), where Z3 and Z4 are independently —H, —C1-C5 alkyl, —C1-C5 alkylene-O—C1-C5 alkyl, benzyl, or —C3-C8 monocyclic cycloalkyl, each of which other than hydrogen is independently unsubstituted or substituted with one or more of -halo, —OH, —N(Ra)2, —C1-C5 alkylene-O—C1-C5 alkyl, —NH2, a nitrogen-containing 3-to 7-membered monocyclic heterocycle, or a —C1-C6 alkyl-substituted nitrogen-containing 3- to 7- membered monocyclic heterocycle, wherein each occurrence of Ra is independently —H, benzyl, or —C1-C10 alkyl; or N, Z3 and Z4 are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle which is unsubstituted or substituted with one or more of —C1-C5 alkyl, phenyl, -phenyl-substituted C1-C5 alkyl, -hydroxy-substituted C1-C5 alkyl, -halo, -halo-substituted C1-C5 alkyl, -halo-substituted phenyl, -phenylene-O—C1-C5 alkyl, -cyano-substituted phenyl, —OH, —O—C1-C5 alkyl, —N(Ra)2, —C1-C5 alkylene-N(Ra)2, —C1-C5 alkylene-C(O)O—C1-C5 alkylene-N(Ra)2, —COOH, —C1-C5 alkylene-COOH, —OP(O)(OH)2, —OS(O)2OH, —C1-C5 alkylene-OP(O)(OH)2, —C1-C5 alkylene-OS(O)2OH, —C(O)O—C1-C5 alkyl, —OC(O)—C1-C5 alkyl, —C1-C5 alkylene-C(O)O—C1-C5 alkyl, —C1-C5 alkylene-C(O)NH—C1-C5 alkyl, —C(O)NH2, or —NO2, wherein each occurrence of Ra is independently —H, benzyl, or —C1-C10 alkyl; or N and both Ra groups are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle;

or N and both R2 groups are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle or a nitrogen-containing 7- to 10-membered bicyclic heterocycle, each of which is unsubstituted or substituted with one or more of —C1-C5 alkyl, —C3-C8 monocyclic cycloalkyl, phenyl, a nitrogen-containing 3- to 7-membered monocyclic heterocycle, phenyl-substituted C1-C5 alkyl, -hydroxy-substituted C1-C5 alkyl, -halo, -halo-substituted C1-C5 alkyl, -halo-substituted phenyl, -phenylene-O—C1-C5 alkyl, -cyano-substituted phenyl, —OH, —O—C1-C5 alkyl, —N(Ra)2, —C1-C5 alkylene-N(Ra)2, —C1-C5 alkylene-C(O)O—C1-C5 alkylene-N(Ra)2, —COOH, —C1-C5 alkylene-O—C1-C5 alkyl, —C1-C5 alkylene-C3-C8 monocyclic cycloalkyl, —C1-C5 alkylene-COOH, —OP(O)(OH)2, —OS(O)2OH, —C1-C5 alkylene-OP(O)(OH)2, —C1-C5 alkylene-OS(O)2OH, —C(O)O—C1-C5 alkyl, —OC(O)—C1-C5 alkyl, —C1-C5 alkylene-C(O)O—C1-C5 alkyl, —C1-C5 alkylene-C(O)NH—C1-C5 alkyl, —C(O)NH2, or —NO2, wherein each occurrence of Ra is independently —H, -benzyl, —C1-C10 alkyl; or N and both Ra groups are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle;

R3 is —H, —PO3H2, —P2O6H3, —P3O9H4, —SO3H, —C(O)—C1-C9 alkyl β-D-glucuronyl, or a C-terminal alpha amino acid residue;

n is an integer ranging from 1 to 10; and

m is an integer ranging from 2 to 10.

In another aspect the invention provides a compound of Formula (IIa)

and pharmaceutically acceptable salts thereof,
wherein

X is —CH2—, —O—, —C(O)—, —NH— or —S—;

R1 is —(CH2)n—N(R2)(R2), —O—(CH2)m—N(R2)(R2), or —O—(CH2)m—OR3;

each R2 is independently —H, —C1-C6 alkyl, —C3-C8 monocyclic cycloalkyl, —C1-C6 alkylene phenyl, phenyl, an N-terminal alpha amino acid residue, an N-terminal alpha amino hydroxymethyl residue, a C1-C6 alkyl ester of an N-terminal alpha amino acid residue, or a nitrogen-containing 3- to 7-membered monocyclic heterocycle, each of which other than hydrogen is independently unsubstituted or substituted with one or more of -halo, —OH, —OP(O)(OH)2, —OS(O)2OH, —C1-C6 alkyl, —O—C1-C6 alkyl, —C3-C8 monocyclic cycloalkyl, a 3- to 7-membered monocyclic heterocycle, —C1-C6 alkyl-substituted 3- to 7-membered monocyclic heterocycle, or —N(Z3)(Z4), where Z3 and Z4 are independently —H, —C1-C5 alkyl, —C1-C5 alkylene-O—C1-C5 alkyl, benzyl, or —C3-C8 monocyclic cycloalkyl, each of which other than hydrogen is independently unsubstituted or substituted with one or more of -halo, —OH, —N(Ra)2, —C1-C5 alkylene-O—C1-C5 alkyl, —NH2, a nitrogen-containing 3-to 7-membered monocyclic heterocycle, or a —C1-C6 alkyl-substituted nitrogen-containing 3- to 7-membered monocyclic heterocycle, wherein each occurrence of Ra is independently —H, benzyl, or —C1-C10 alkyl; or N, Z3 and Z4 are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle which is unsubstituted or substituted with one or more of —C1-C5 alkyl, phenyl, -phenyl-substituted C1-C5 alkyl, -hydroxy-substituted C1-C5 alkyl, -halo, -halo-substituted C1-C5 alkyl, -halo-substituted phenyl, -phenylene-O—C1-C5 alkyl, -cyano-substituted phenyl, —OH, —O—C1-C5 alkyl, —N(Ra)2, —C1-C5 alkylene-N(Ra)2, —C1-C5 alkylene-C(O)O—C1-C5 alkylene-N(Ra)2, —COOH, —C1-C5 alkylene-COOH, —OP(O)(OH)2, —OS(O)2OH , —C1-C5 alkylene-OP(O)(OH)2, —C1-C5 alkylene-OS(O)2OH, —C(O)O—C1-C5 alkyl, —OC(O)—C1-C5 alkyl, —C1-C5 alkylene-C(O)O—C1-C5 alkyl, —C1-C5 alkylene-C(O)NH—C1-C5 alkyl, —C(O)NH2, or —NO2, wherein each occurrence of Ra is independently —H, benzyl, or —C1-C10 alkyl; or N and both Ra groups are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle;

or N and both R2 groups are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle or a nitrogen-containing 7- to 10-membered bicyclic heterocycle, each of which is unsubstituted or substituted with one or more of —C1-C5 alkyl, —C3-C8monocyclic cycloalkyl, phenyl, a nitrogen-containing 3- to 7-membered monocyclic heterocycle, phenyl-substituted C1-C5 alkyl, -hydroxy-substituted C1-C5 alkyl, -halo, -halo-substituted C1-C5 alkyl, -halo-substituted phenyl, -phenylene-O—C1-C5 alkyl, -cyano-substituted phenyl, —OH, —O—C1-C5 alkyl, —N(Ra)2, —C1-C5 alkylene-N(Ra)2, —C1-C5 alkylene-C(O)O—C1-C5 alkylene-N(Ra)2, —COOH, —C1-C5 alkylene-O—C1-C5 alkyl, —C1-C5 alkylene-C3-C8 monocyclic cycloalkyl, —C1-C5 alkylene-COOH, —OP(O)(OH)2, —OS(O)2OH, —C1-C5 alkylene-OP(O)(OH)2, —C1-C5 alkylene-OS(O)2OH, —C(O)O—C1-C5 alkyl, —OC(O)—C1-C5 alkyl, —C1-C5 alkylene-C(O)O—C1-C5 alkyl, —C1-C5 alkylene-C(O)NH—C1-C5 alkyl, —C(O)NH2, or —NO2, wherein each occurrence of Ra is independently —H, -benzyl, —C1-C10 alkyl; or N and both Ra groups are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle;

R3 is —H, —PO3H2, —P2O6H3, —P3O9H4, —SO3H, —C(O)—C1-C9 alkyl, β-D-glucuronyl, or a C-terminal alpha amino acid residue;

n is an integer ranging from 1 to 10; and

m is an integer ranging from 2 to 10.

In another aspect the invention provides a compound of Formula (IIb)

and pharmaceutically acceptable salts thereof,
wherein

R1 is —(CH2)n—OR3;

R3 is —H, —PO3H2, —P2O6H3, —P3O9H4, —SO3H, —C(O)—C1-C9 alkyl, β-D-glucuronyl, or a C-terminal alpha amino acid residue; and

n is an integer ranging from 1 to 3.

In another aspect the invention provides a compound of Formula (III)

and pharmaceutically acceptable salts thereof,
wherein

X is —CH2—, —O—, —C(O)—, —NH— or —S—;

R1 is —(CH2)n—N(R2)(R2), —O—(CH2)m—N(R2)(R2), —(CH2)n—OR3 or —O—(CH2)m—OR3;

each R2 is independently —H, —C1-C6 alkyl, —C3-C8 monocyclic cycloalkyl, —C1-C6 alkylene phenyl, phenyl, an N-terminal alpha amino acid residue, an N-terminal alpha amino hydroxymethyl residue, a C1-C6 alkyl ester of an N-terminal alpha amino acid residue, or a nitrogen-containing 3- to 7-membered monocyclic heterocycle, each of which other than hydrogen is independently unsubstituted or substituted with one or more of -halo, —OH, —OP(O)(OH)2, —OS(O)2OH, —C1-C6 alkyl, —O—C1-C6 alkyl, —C3-C8 monocyclic cycloalkyl, a 3- to 7-membered monocyclic heterocycle, —C1-C6 alkyl-substituted 3- to 7-membered monocyclic heterocycle, or —N(Z3)(Z4), where Z3 and Z4 are independently —H, —C1-C5 alkyl, —C1-C5 alkylene-O—C1-C5 alkyl, benzyl, or —C3-C8monocyclic cycloalkyl, each of which other than hydrogen is independently unsubstituted or substituted with one or more of -halo, —OH, —N(Ra)2, —C1-C5 alkylene-O—C1-C5 alkyl, —NH2, a nitrogen-containing 3-to 7-membered monocyclic heterocycle, or a —C1-C6 alkyl-substituted nitrogen-containing 3- to 7-membered monocyclic heterocycle, wherein each occurrence of Ra is independently —H, benzyl, or —C1-C10 alkyl; or N, Z3 and Z4 are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle which is unsubstituted or substituted with one or more of —C1-C5 alkyl, phenyl, -phenyl-substituted C1-C5 alkyl, -hydroxy-substituted C1-C5 alkyl, -halo, -halo-substituted C1-C5 alkyl, -halo-substituted phenyl, -phenylene-O—C1-C5 alkyl, -cyano-substituted phenyl, —OH, —O—C1-C5 alkyl, —N(Ra)2, —C1-C5 alkylene-N(Ra)2, —C1-C5 alkylene-C(O)O—C1-C5 alkylene-N(Ra)2, —COON, —C1-C5 alkylene-COOH, —OP(O)(OH)2, —OS(O)2OH , —C1-C5 alkylene-OP(O)(OH)2, —C1-C5 alkylene-OS(O)2OH, —C(O)O—C1-C5 alkyl, —OC(O)—C1-C5 alkyl, —C1-C5 alkylene-C(O)O—C1-C5 alkyl, —C1-C5 alkylene-C(O)NH—C1-C5 alkyl, —C(O)NH2, or —NO2, wherein each occurrence of Ra is independently —H, benzyl, or —C1-C10 alkyl; or N and both Ra groups are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle;

or N and both R2 groups are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle or a nitrogen-containing 7- to 10-membered bicyclic heterocycle, each of which is unsubstituted or substituted with one or more of —C1-C5 alkyl, —C3-C8 monocyclic cycloalkyl, phenyl, a nitrogen-containing 3- to 7-membered monocyclic heterocycle, phenyl-substituted C1-C5 alkyl, -hydroxy-substituted C1-C5 alkyl, -halo, -halo-substituted C1-C5 alkyl, -halo-substituted phenyl, -phenylene-O—C1-C5 alkyl, -cyano-substituted phenyl, —OH, —O—C1-C5 alkyl, —N(Ra)2, —C1-C5 alkylene-N(Ra)2, —C1-C5 alkylene-C(O)O—C1-C5 alkylene-N(Ra)2, —COOH, —C1-C5 alkylene-O—C1-C5 alkyl, —C1-C5 alkylene-C3-C8 monocyclic cycloalkyl, —C1-C5 alkylene-COOH, —OP(O)(OH)2, —OS(O)2OH, —C1-C5 alkylene-OP(O)(OH)2, —C1-C5 alkylene-OS(O)2OH, —C(O)O—C1-C5 alkyl, —OC(O)—C1-C5 alkyl, —C1-C5 alkylene-C(O)O—C1-C5 alkyl, —C1-C5 alkylene-C(O)NH—C1-C5 alkyl, —C(O)NH2, or —NO2, wherein each occurrence of Ra is independently —H, -benzyl, —C1-C10 alkyl; or N and both Ra groups are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle;

R3 is —H, —PO3H2, —P2O6H3, —P3O9H4, —SO3H, —C(O)—C1-C9 alkyl, β-D-glucuronyl, or a C-terminal alpha amino acid residue;

n is an integer ranging from 1 to 10; and

m is an integer ranging from 2 to 10.

The invention provides compositions comprising an effective amount of a compound of Formula (I), (IIa), (IIb), (III) or a pharmaceutically acceptable salt thereof and a physiologically acceptable carrier or vehicle.

The invention further provides methods for treating or preventing an inflammatory disease, a reperfusion injury, diabetes mellitus, a diabetic complication, reoxygenation injury resulting from organ transplantation, an ischemic condition, a neurodegenerative disease, renal failure, a vascular disease, a cardiovascular disease, cancer, a complication of prematurity, cardiomyopathy, retinopathy, nephropathy, neuropathy, erectile dysfunction or urinary incontinence (each being a “Condition”), comprising administering to a subject in need thereof an effective amount of an Indenoisoquinolinone Analog of Formula (I), (IIa), (IIb), (III) or a pharmaceutically acceptable salt thereof.

The invention provides methods for treating or preventing erectile dysfunction or urinary incontinence, comprising administering to a subject in need thereof an effective amount of a compound of Formula (IIc)

or a pharmaceutically acceptable salt thereof,
wherein

X is —CH2—, —O—, —C(O)—, —NH— or —S—;

R1 is —(CH2)n—N(R2)(R2), —O—(CH2)m—N(R2)(R2), —(CH2)n—OR3 or —O—(CH2)m—OR3;

each R2 is independently —H, —C1-C6 alkyl, —C3-C8 monocyclic cycloalkyl, —C1-C6 alkylene phenyl, phenyl, an N-terminal alpha amino acid residue, an N-terminal alpha amino hydroxymethyl residue, a C1-C6 alkyl ester of an N-terminal alpha amino acid residue, or a nitrogen-containing 3- to 7-membered monocyclic heterocycle, each of which other than hydrogen is independently unsubstituted or substituted with one or more of -halo, —OH, —OP(O)(OH)2, —OS(O)2OH, —C1-C6 alkyl, —O—C1-C6 alkyl, —C3-C8 monocyclic cycloalkyl, a 3- to 7-membered monocyclic heterocycle, —C1-C6 alkyl-substituted 3- to 7-membered monocyclic heterocycle, or —N(Z3)(Z4), where Z3 and Z4 are independently —H, —C1-C5 alkyl, —C1-C5 alkylene-O—C1-C5 alkyl, benzyl, or —C3-C8 monocyclic cycloalkyl, each of which other than hydrogen is independently unsubstituted or substituted with one or more of -halo, —OH, —N(Ra)2, —C1-C5 alkylene-O—C1-C5 alkyl, —NH2, a nitrogen-containing 3-to 7-monocyclic heterocycle, or a —C1-C6 alkyl-substituted nitrogen-containing 3- to 7-membered monocyclic heterocycle, wherein each occurrence of Ra is independently —H, benzyl, or —C1-C10 alkyl; or N, Z3 and Z4 are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle which is unsubstituted or substituted with one or more of —C1-C5 alkyl, phenyl, -phenyl-substituted C1-C5 alkyl, -hydroxy-substituted C1-C5 alkyl, -halo, -halo-substituted C1-C5 alkyl, -halo-substituted phenyl, -phenylene-O—C1-C5 alkyl, -cyano-substituted phenyl, —OH, —O—C1-C5 alkyl, —N(Ra)2, —C1-C5 alkylene-N(Ra)2, —C1-C5 alkylene-C(O)O—C1-C5 alkylene-N(Ra)2, —COOH, —C1-C5 alkylene-COOH, —OP(O)(OH)2, —OS(O)2OH, —C1-C5 alkylene-OP(O)(OH)2, —C1-C5 alkylene-OS(O)2OH, —C(O)O—C1-C5 alkyl, —OC(O)—C1-C5 alkyl, —C1-C5 alkylene-C(O)O—C1-C5 alkyl, —C1-C5 alkylene-C(O)NH—C1-C5 alkyl, —C(O)NH2, or —NO2, wherein each occurrence of Ra is independently —H, benzyl, or —C1-C10 alkyl; or N and both Ra groups are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle;

or N and both R2 groups are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle or a nitrogen-containing 7- to 10-membered bicyclic heterocycle, each of which is unsubstituted or substituted with one or more of —C1-C5 alkyl, —C3-C8 monocyclic cycloalkyl, phenyl, a nitrogen-containing 3- to 7-membered monocyclic heterocycle, phenyl-substituted C1-C5 alkyl, -hydroxy-substituted C1-C5 alkyl, -halo, -halo-substituted C1-C5 alkyl, -halo-substituted phenyl, -phenylene-O—C1-C5 alkyl, -cyano-substituted phenyl, —OH, —O—C1-C5 alkyl, —N(Ra)2, —C1-C5 alkylene-N(Ra)2, —C1-C5 alkylene-C(O)O—C1-C5 alkylene-N(Ra)2, —COOH, —C1-C5 alkylene-O—C1-C5 alkyl, —C1-C5 alkylene-C3-C8 monocyclic cycloalkyl, —C1-C5 alkylene-COOH, —OP(O)(OH)2, —OS(O)2OH, —C1-C5 alkylene-OP(O)(OH)2, —C1-C5 alkylene-OS(O)2OH, —C(O)O—C1-C5 alkyl, —OC(O)—C1-C5 alkyl, —C1-C5 alkylene-C(O)O—C1-C5 alkyl, —C1-C5 alkylene-C(O)NH—C1-C5 alkyl, —C(O)NH2, or —NO2, wherein each occurrence of Ra is independently —H, -benzyl, —C1-C10 alkyl; or N and both Ra groups are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle;

R3 is —H, —PO3H2, —P2O6H3, —P3O9H4, —SO3H, —C(O)—C1-C9 alkyl, β-D-glucuronyl, or a C-terminal alpha amino acid residue;

n is an integer ranging from 1 to 10; and

m is an integer ranging from 2 to 10.

A compound of Formula (I), Formula (IIa), Formula (IIb), Formula (IIc), Formula (III) or a pharmaceutically acceptable salt thereof (an “Indenoisoquinolinone Analog”) is useful for treating or preventing a Condition.

A composition comprising an effective amount of an Indenoisoquinolinone Analog and a physiologically acceptable carrier or vehicle is useful for treating or preventing a Condition.

The details of the invention are set forth in the accompanying description below.

All patents and publications cited in this specification are incorporated by reference in their entirety.

5. DETAILED DESCRIPTION OF THE INVENTION 5.1 Definitions and Abbreviations

The following definitions are used in connection with the Indenoisoquinolinone Analogs:

The term “—C1-C5 alkyl” as used herein, refers to a straight chain or branched non-cyclic hydrocarbon having from 1 to 5 carbon atoms, wherein one of the hydrocarbon's hydrogen atoms has been replaced by a single bond. Representative straight chain —C1-C5 alkyls include -methyl, -ethyl, -n-propyl, -n-butyl and -n-pentyl. Representative branched —C1-C5 alkyls include -isopropyl, -sec-butyl, -isobutyl, -tent-butyl, -isopentyl, -neopentyl, 1-methylbutyl, 2-methylbutyl, 3-methylbutyl, 1,1-dimethylpropyl and 1,2-dimethylpropyl. In one embodiment, the —C1-C5 alkyl is substituted with one or more of -halo, —OH, —N(Ra)2, —C1-C5 alkylene-O—C1-C5 alkyl, —NH2, a nitrogen-containing 3-to 7- membered monocyclic heterocycle, or an —C1-C6 alkyl-substituted nitrogen-containing 3- to 7-membered monocyclic heterocycle. Unless otherwise indicated, the —C1-C5 alkyl is unsubstituted.

The term “-phenyl-substituted C1-C5 alkyl” as used herein, refers to a “—C1-C5 alkyl” wherein one of the hydrocarbon's hydrogen atoms has been replaced by a phenyl group.

The term “—C1-C6 alkyl” as used herein, refers to a straight chain or branched non-cyclic hydrocarbon having from 1 to 6 carbon atoms, wherein one of the hydrocarbon's hydrogen atoms has been replaced by a single bond. Representative straight chain —C1-C6 alkyls include -methyl, -ethyl, -n-propyl, -n-butyl, -n-pentyl and -n-hexyl. Representative branched —C1-C6 alkyls include -isopropyl, -sec-butyl, -isobutyl, -tert-butyl, -isopentyl, -neopentyl, -1-methylbutyl, -isohexyl, -neohexyl, -2-methylbutyl, -3-methylbutyl, -1,1-dimethylpropyl and -1,2-dimethylpropyl. In one embodiment, the —C1-C6 alkyl is substituted with one or more of -halo, —OH, —OP(O)(OH)2, —OS(O)2OH, —C1-C6 alkyl, —O—C1-C6 alkyl, —C3-C8 monocyclic cycloalkyl, a 3- to 7-membered monocyclic heterocycle, —C1-C6 alkyl-substituted 3- to 7- membered monocyclic heterocycle, or —N(Z3)(Z4). Unless otherwise indicated, the —C1-C6 alkyl is unsubstituted.

The term “—C1-C6 alkyl” as used herein, refers to a straight chain or branched non-cyclic hydrocarbon having from 1 to 9 carbon atoms, wherein one of the hydrocarbon's hydrogen atoms has been replaced by a single bond. Representative —C1-C9 alkyls include, methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, -nonyl, isopropyl, isobutyl, sec-butyl and tert-butyl, isopentyl, neopentyl, isohexyl, isoheptyl, isooctyl and isononyl.

The term “—C1-C10 alkyl” as used herein, refers to a straight chain or branched non-cyclic hydrocarbon having from 1 to 10 carbon atoms, wherein one of the hydrocarbon's hydrogen atoms has been replaced by a single bond. Representative —C1-C10 alkyls include, methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, -nonyl, decyl, isopropyl, isobutyl, sec-butyl and tert-butyl, isopentyl, neopentyl, isohexyl, isoheptyl, isooctyl, isononyl and isodecyl.

The term “—C3-C8 monocyclic cycloalkyl” as used herein, refers to a saturated cyclic hydrocarbon having from 3 to 8 carbon atoms. Representative —C3-C8 monocyclic cycloalkyls include -cyclopropyl, -cyclobutyl, -cyclopentyl, -cyclohexyl, -cycloheptyl and -cyclooctyl. In one embodiment, the —C3-C8 monocyclic cycloalkyl is substituted with one or more of -halo, —OH, —OP(O)(OH)2, —OS(O)2OH, —C1-C6 alkyl, —O—C1-C6 alkyl, —C3-C8 monocyclic cycloalkyl, a 3- to 7-membered monocyclic heterocycle, —C1-C6 alkyl-substituted 3- to 7- membered monocyclic heterocycle. In another embodiment, the —C3-C8 monocyclic cycloalkyl is substituted with one or more of —N(Ra)2, —C1-C5 alkylene-O—C1-C5 alkyl, or —NH2. Unless otherwise indicated, the —C3-C8 monocyclic cycloalkyl is unsubstituted.

A “nitrogen containing 3- to 7-membered monocyclic heterocycle” refers to a monocyclic 3- to 7-membered aromatic or non-aromatic monocyclic cycloalkyl group in which one of the cycloalkyl group's ring carbon atoms has been replaced with a nitrogen atom and 0-4 of the cycloalkyl group's remaining ring carbon atoms are independently replaced with a N, O or S atom. The nitrogen containing 3- to 7-membered monocyclic heterocycles can be attached via a nitrogen, sulfur, or carbon atom. Representative examples of nitrogen-containing-3- to 7-membered monocyclic heterocycles include, but are not limited to, piperidinyl, piperazinyl, pyrrolyl, piperidonyl, oxazinyl, thiazinyl, diazinyl, triazinyl, tetrazinyl, imidazolyl, tetrazolyl, pyrrolidinyl, isoxazolyl, pyridinyl, oxazolyl, thiazolyl, pyrazolyl, triazolyl, pyrimidinyl, morpholinyl, furuzanyl, pyrrolinyl, imidazolinyl, imidazolidinyl, pyrazolinyl, isothiazolyl, oxadiazolyl, thiadiazolyl, pyridazinyl, pyrazinyl, pyrazolidinyl, and thiomorpholinyl.

In one embodiment, the nitrogen-containing 3- to 7-membered monocyclic heterocycle is fully saturated or partially saturated.

In another embodiment, the nitrogen-containing 3- to 7-membered monocyclic heterocycle is substituted with one or more of -halo, —OH, —OP(O)(OH)2, —OS(O)2OH, —C1-C6 alkyl, —O—C1-C6 alkyl, —C3-C8 monocyclic cycloalkyl, a 3- to 7-membered monocyclic heterocycle, —C1-C6 alkyl-substituted 3- to 7- membered monocyclic heterocycle, or —N(Z3)(Z4). Unless otherwise indicated, the nitrogen-containing 3- to 7-membered monocyclic heterocycle is unsubstituted.

A “nitrogen-containing 7- to 10-membered bicyclic heterocycle” refers to a 7- to 10-membered bicyclic ring system that contains at least one ring nitrogen atom. The nitrogen-containing 7- to 10-membered bicyclic heterocycles can be attached via a nitrogen, sulfur, or carbon atom. Representative nitrogen-containing 7- to 10-membered bicyclic heterocycles include -quinolinyl, -isoquinolinyl, -chromonyl, -indolyl, -isoindolyl,-indolinyl, indolizinyl, -indazolyl, -purinyl, -4H-quinolizinyl, -isoquinolyl, -quinolyl, -phthalazinyl, -naphthyridinyl, benzimidazolyl, benzthiazolyl, dihydroquinolyl, dihydroisoquinolyl, cinnolyl, phthalazyl, quinazolyl, quinoxalyl, and pteridyl.

In one embodiment, the nitrogen-containing 7- to 10-membered bicyclic heterocycle is saturated or partially saturated.

A “3- to 7-membered monocyclic heterocycle” refers to a monocyclic 3- to 7-membered aromatic or non-aromatic monocyclic cycloalkyl in which 1-4 of the ring carbon atoms have been independently replaced with a N, O or S atom. The 3- to 7-membered monocyclic heterocycles can be attached via a nitrogen, sulfur, or carbon atom. Representative examples of a 3- to 7-membered monocyclic heterocycle group include, but are not limited to, nitrogen-containing 3- to 7-membered monocyclic heterocycles discussed above, tetrahydrofuranyl, dihydrofuranyl, pyranyl, dihydropyranyl, tetrahydropyranyl, thiopyranyl, dihydrothiopyranyl, tetrahydrothiopyranyl, dioxanyl, dithianyl, trithianyl, dioxolanyl, furanyl, and thiophenyl. In one embodiment, the 3- to 7-membered monocyclic heterocycle is a nitrogen-containing 3- to 7-membered monocyclic heterocycle. In another embodiment, the 3- to 7-membered monocyclic heterocycle is saturated or partially saturated.

A “C1-C6 alkyl-substituted 3- to 7-membered monocyclic heterocycle” refers to to a “3- to 7-membered monocyclic heterocycle,” as defined above, wherein one or more of the heterocycle's hydrogen atoms has been replaced by a C1-C6 alkyl, as defined above.

“Halo” is —F, —Cl, —Br or —I.

“Halo-substituted C1-C5 alkyl” refers to a C1-C5 alkyl group, as defined above, wherein one or more of the C1-C5 alkyl group's hydrogen atoms has been replaced with —F, —Cl, —Br or —I. Representative examples of a halo-substituted C1-C5 alkyl include, but are not limited to, —CH2F, —CCl3, —CF3, —CH2Cl, —CH2CH2Br, —CH2CH2I, —CH2CH2CH2F, —CH2CH2CH2Cl, —CH2CH2CH2CH2Br, —CH2CH2CH2CH2I, —CH2CH2CH2CH2CH2Br, —CH2CH2CH2CH2CH2I, —CH2CH(Br)CH3, —CH2CH(Cl)CH2CH3, —CH(F)CH2CH3 and —C(CH3)2(CH2Cl).

“Halo-substituted phenyl” refers to a phenyl group, wherein one or more of the phenyl group's hydrogen atoms has been replaced with —F, Cl, —Br or —I.

“Cyano-substituted phenyl” refers to a phenyl group, wherein one or more of the phenyl group's hydrogen atoms has been replaced with —CN.

A “C1-C6 alkylene phenyl” refers to a C1-C6 alkyl group, as defined above, wherein one of the C1-C6 alkyl group's hydrogen atoms has been replaced with phenyl.

“Hydroxy-substituted C1-C5 alkyl” refers to a C1-C5 alkyl group, as defined above, wherein one or more of the C1-C5 alkyl group's hydrogen atoms has been replaced with —OH. Representative examples of a hydroxy-substituted C1-C5 alkyl include, but are not limited to, —CH2OH, —CH2CH2OH, —CH2CH2CH2OH, —CH2CH2CH2CH2OH, —CH2CH2CH2CH2CH2OH, —CH2CH(OH)CH3, —CH2CH(OH)CH2CH3, —CH(OH)CH2CH3 and —C(CH3)2(CH2OH).

A “C-terminal alpha amino acid residue” refers to an L-, D- or racemic natural or unnatural alpha amino acid, less the hydroxyl group of the alpha amino acid's carboxyl group. Representative examples of a C-terminal alpha amino acid residue include, but are not limited to, CH3—CH(NH2)—C(O)—, HN═C(NH2)—NH—(CH2)3—CH(NH2)—C(O)—, H2N—C(O)—CH2—CH(NH2)—C(O)—, HOOC—CH2—CH(NH2)—C(O)—, HS—CH2—CH(NH2)—C(O)—, H2N—C(O)—(CH2)2—CH(NH2)—C(O)—, HOOC—(CH2)2—CH(NH2)—C(O)—, H2N—CH2—C(O)—, CH3—CH2—CH(CH3)—CH(NH2)—C(O)—, (CH3)2—CH—CH2—CH(NH2)—C(O)—, H2N—(CH2)4—CH(NH2)—C(O)—, CH3—S—(CH2)2—CH(NH2)—C(O)—, HO—CH2—CH(NH2)—C(O)—, CH3—CH(OH)—CH(NH2)—C(O)—, (CH3)2—CH—CH(NH2)—C(O)—,

In one embodiment, the C-terminal alpha amino acid is a natural amino acid.

In another embodiment, the C-terminal alpha amino acid is an unnatural amino acid.

In one embodiment, the C-terminal alpha amino acid is an L amino acid.

An “N-terminal alpha amino acid residue” refers to an L-, D- or racemic natural or unnatural alpha amino acid, less the alpha amino acid's N-terminal amino group. Representative examples of an N-terminal alpha amino acid residue include, but are not limited to, —CH2—COOH, —CH(CH3)COOH, —CH(CH(CH3)2)COOH, —CH(CH2CH(CH3)2)COOH, —CH(CH(CH3)(CH2CH3))COOH, —CH((CH2)4—NH2)COOH, —CH((CH2)3NH(C═NH2)—NH2)COOH, —CH(CH2—OH)COOH, —CH(CH((CH3)(OH)))COOH, —CH(CH2—COOH)COOH, —CH((CH2)2COOH)COOH, —CH((CH2)(CONH2))COOH, —CH((CH2)2(CONH2))COOH, —CH(CH2—SH)COOH, —CH((CH2)2—S—CH3)COOH,

In one embodiment, the N-terminal alpha amino acid residue is substituted with one or more of -halo, -OH, —OP(O)(OH)2, —OS(O)2OH, —C1-C6 alkyl, —O—C1-C6 alkyl, —C3-C8 monocyclic cycloalkyl, a 3- to 7-membered monocyclic heterocycle, -C1-C6 alkyl-substituted 3- to 7- membered monocyclic heterocycle, or —N(Z3)(Z4). Unless otherwise indicated, the N-terminal alpha amino acid residue is unsubstituted.

An “N-terminal alpha amino hydroxymethyl residue” refers to an “N-terminal alpha amino acid residue,” wherein its C-terminal carboxyl group, a side chain's carboxyl group, or both have been replaced with a hydroxymethyl group. In one embodiment, the N-terminal alpha amino hydroxymethyl residue is substituted with one or more of -halo, —OH, —OP(O)(OH)2, —OS(O)2OH, —C1-C6 alkyl, —O—C1-C6 alkyl, —C3-C8 monocyclic cycloalkyl, a 3- to 7-membered monocyclic heterocycle, —C1-C6 alkyl-substituted 3- to 7-membered monocyclic heterocycle, or —N(Z3)(Z4). In another embodiment, the hydroxyl group of the hydroxymethyl residue is substituted with —PO3H2, —P2O6H3, —P3O9H4, —SO3H, —C(O)—C1-C9 alkyl, β-D-glucuronyl, or a C-terminal alpha amino acid residue. Unless otherwise indicated, the N-terminal alpha amino hydroxymethyl residue is unsubstituted.

A “C1-C6 alkyl ester of an N-terminal alpha amino acid residue” refers to an “N-terminal alpha amino acid residue,” wherein the hydrogen atom of the amino acid's C-terminal carboxyl group, a side chain's carboxyl group, or both have been replaced by a —C1-C6 alkyl group. In one embodiment, the C1-C6 alkyl ester of an N-terminal alpha amino acid residue is a methyl, an ethyl, an n-propyl or an iso-propyl ester of an N-terminal alpha amino acid residue. In another embodiment, the C1-C6 alkyl ester of an N-terminal alpha amino acid residue is substituted with one or more of -halo, —OH, —OP(O)(OH)2, —OS(O)2OH, —C1-C6 alkyl, —O—C1-C6 alkyl, —C3-C8 monocyclic cycloalkyl, a 3- to 7-membered monocyclic heterocycle, —C1-C6 alkyl-substituted 3- to 7-membered monocyclic heterocycle, or —N(Z3)(Z4). Unless otherwise indicated, the C1-C6 alkyl ester of an N-terminal alpha amino acid residue is unsubstituted.

In one embodiment, the N-terminal alpha amino acid is a natural amino acid.

In another embodiment, the N-terminal alpha amino acid is an unnatural amino acid.

In one embodiment, the N-terminal alpha amino acid is an L amino acid.

A “β-D-glucuronyl” refers to a group having the formula:

A “subject” is a mammal, e.g., a human, mouse, rat, guinea pig, dog, cat, horse, cow, pig, or non-human primate, such as a monkey, chimpanzee, baboon or rhesus. In one embodiment, the subject is a human.

The phrase “pharmaceutically acceptable salt,” as used herein, is a salt of an acid and a basic nitrogen atom of an Indenoisoquinolinone Analog. Illustrative salts include, but are not limited, to sulfate, citrate, acetate, oxalate, chloride, bromide, iodide, nitrate, bisulfate, phosphate, acid phosphate, isonicotinate, lactate, salicylate, acid citrate, tartrate, oleate, tannate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisinate, fumarate, gluconate, glucaronate, saccharate, formate, benzoate, glutamate, methanesulfonate, ethanesulfonate, benzenesulfonate, p-toluenesulfonate, camphorsulfonate, and pamoate (i.e., 1,1′-methylene-bis-(2-hydroxy-3-naphthoate)) salts. The term “pharmaceutically acceptable salt” also refers to a salt of an Indenoisoquinolinone Analog having an acidic functional group, such as a carboxylic acid functional group, and a base. Suitable bases include, but are not limited to, hydroxides of alkali metals such as sodium, potassium, and lithium; hydroxides of alkaline earth metal such as calcium and magnesium; hydroxides of other metals, such as aluminum and zinc; ammonia, and organic amines, such as unsubstituted or hydroxy-substituted mono-, di-, or tri-alkylamines, dicyclohexylamine; tributyl amine; pyridine; N-methyl, N-ethylamine; diethylamine; triethylamine; mono-, bis-, or tris-(2-OH-lower alkylamines), such as mono-; bis-, or tris-(2-hydroxyethyl)amine, 2-hydroxy-tert-butylamine, or tris-(hydroxymethyl)methylamine, N,N-di-lower alkyl-N-(hydroxyl-lower alkyl)-amines, such as N,N-dimethyl-N-(2-hydroxyethyl)amine or tri-(2-hydroxyethyl)amine; N-methyl-D-glucamine; and amino acids such as arginine, lysine, and the like. The term “pharmaceutically acceptable salt” also includes a hydrate of a compound of the invention.

In one embodiment, the pharmaceutically acceptable salt is a mesylate salt.

In another embodiment, the pharmaceutically acceptable salt is a camphorsulfonate salt.

An “effective amount” when used in connection with an Indenoisoquinolinone Analog is an amount that is effective for treating or preventing a Condition.

An “effective amount” when used in connection with another anticancer agent is an amount that is effective for treating or preventing cancer alone or in combination with an Indenoisoquinolinone Analog. “In combination with” includes administration within the same composition and via separate compositions. In the latter instance, the other anticancer agent is administered during a time when the Indenoisoquinolinone Analog exerts its prophylactic or therapeutic effect, or vice versa.

5.2 The Indenoisoquinolinone Analogs of Formula (I)

The present invention provides Indenoisoquinolinone Analogs according to Formula (I), below:

and pharmaceutically acceptable salts thereof,
wherein:

X and R1 are as defined above for the compounds of Formula (I).

In one embodiment, X is —CH2—. In another embodiment, X is —O—. In another embodiment, X is —C(O)—. In another embodiment, X is —NH—. In another embodiment, X is —S—.

In one embodiment, R3 is —H.

In one embodiment, R1 is —(CH2)n—N(R2)(R2) and X is —CH2—.

In another embodiment, R1 is —(CH2)n—N(R2)(R2) and X is —O—.

In another embodiment, R1 is —(CH2)n—N(R2)(R2) and X is —C(O)—.

In another embodiment, R1 is —(CH2)n—N(R2)(R2) and X is —NH—.

In another embodiment, R1 is —(CH2)n—N(R2)(R2) and X is —S—.

In one embodiment, R1 is —(CH2)n—OR3 and X is —CH2—.

In another embodiment, R1 is —(CH2)n—OR3 and X is —O—.

In another embodiment, R1 is —(CH2)n—OR3 and X is —C(O)—.

In another embodiment, R1 is —(CH2)n—OR3 and X is —NH—.

In another embodiment, R1 is —(CH2)n—OR3 and X is —S—.

In one embodiment, R1 is —O—(CH2)m—N(R2)(R2) and X is —CH2—.

In another embodiment, R1 is —O—(CH2)m—N(R2)(R2) and X is —O—.

In another embodiment, R1 is —O—(CH2)m—N(R2)(R2) and X is —C(O)—.

In another embodiment, R1 is —O—(CH2)m—N(R2)(R2) and X is —NH—.

In another embodiment, R1 is —O—(CH2)m—N(R2)(R2) and X is —S—.

In one embodiment, R1 is —O—(CH2)m—OR3 and X is —CH2—.

In another embodiment, R1 is —O—(CH2)m—OR3 and X is —O—.

In another embodiment, R1 is —O—(CH2)m—OR3 and X is —C(O)—.

In another embodiment, R1 is —O—(CH2)m—OR3 and X is —NH—.

In another embodiment, R1 is —O—(CH2)m—OR3 and X is —S—.

In one embodiment, one R2 is —H, and the other R2 is —C1-C6 alkyl.

In another embodiment, each R2 is —C1-C6 alkyl.

In another embodiment, each R2 is -methyl.

In one embodiment, R3 is —H.

In another embodiment, R3 is —PO3H2.

In another embodiment, R3 is —P2O6H3.

In another embodiment, R3 is —P3O9H4.

In another embodiment, R3 is —SO3H.

In yet another embodiment, R3 is —PO3Na2.

In yet another embodiment, R3 is —P2O6Na3.

In yet another embodiment, R3 is —P3O9Na4.

In yet another embodiment, R3 is —SO3Na.

In another embodiment, R3 is —C(O)—C1-C9 alkyl.

In another embodiment, R3 is β-D-glucuronyl.

In another embodiment, R3 is a C-terminal alpha amino acid residue.

In one embodiment, n is 1.

In another embodiment, n is 2.

In yet another embodiment, n is 3.

In a further embodiment, n is 4, 5, or 6.

In yet a further embodiment, n is 7, 8, or 9.

In still a further embodiment, n is 10.

In one embodiment, m is 2.

In another embodiment, m is 3.

In yet another embodiment, m is 4, 5, or 6.

In a further embodiment, m is 7, 8, or 9.

In yet a further embodiment, m is 10.

In various embodiments, —N(R2)(R2) is:

In other embodiments, —N(R2)(R2) is:

In still other embodiments, —N(R2)(R2) is

    • —N(CH2CH3)(CH3),
    • —N(CH2CH2CH3)(CH3),
    • —N(CH2CH2CH2CH3)(CH3),
    • —NH—CH2CH2CH2CH3, or
    • —NH—CH2CH2—O—CH3.

In some embodiments, —N(R2)(R2) is

In various embodiments, —N(Z3)(Z4) is:

In other embodiments, —N(Z3)(Z4) is:

In still other embodiments, —N(Z3)(Z4) is

    • —N(CH2CH3)(CH3),
    • —N(CH2CH2CH3)(CH3),
    • —N(CH2CH2CH2CH3)(CH3),
    • —NH—CH2CH2CH2CH3, or
    • —NH—CH2CH2—O—CH3.

In some embodiments, —N(Z3)(Z4) is

In one embodiment, X is —CH2—, —O—, —C(O)—, —NH— or —S—;

R1 is —(CH2)n—N(R2)(R2), —O—(CH2)m—N(R2)(R2), —(CH2)n—OR3 or —O—(CH2)m—OR3;

each R2 is independently —H, —C1-C6 alkyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, —C1-C6 alkylene phenyl or -phenyl, each of which other than hydrogen is unsubstituted or substituted with one or more of -halo, —OH, —OP(O)(OH)2, —OS(O)2OH or —N(Z3)(Z4), where Z3 and Z4 are independently —H, —C1-C5 alkyl, or —C1-C5 alkylene-O—C1-C5 alkyl, each of which other than hydrogen is unsubstituted or substituted with one or more of -halo, —OH or —NH2;

or N, Z3 and Z4 are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle which is unsubstituted or substituted with one to three of —C1-C5 alkyl, -phenyl, —C1-C5 alkylene phenyl, -hydroxy-substituted C1-C5 alkyl, -halo, -halo-substituted C1-C5 alkyl, -halo-substituted phenyl, -phenylene-O—C1-C5 alkyl, -cyano-substituted phenyl, —OH, —O—C1-C5 alkyl, —N(Ra)2, —C1-C5 alkylene-N(Ra)2, —C1-C5 alkylene-C(O)O—C1-C5 alkylene-N(Ra)2, —COOH, —C1-C5 alkylene-COOH, —OP(O)(OH)2, —OS(O)2OH , —C1-C5 alkylene-OP((O)(OH)2, —C1-C5 alkylene-OS(O)2OH, —C(O)O—C1-C5 alkyl, OC(O)—C1-C5 alkyl, —C1-C5 alkylene-C(O)O—C1-C5 alkyl, —C1-C5 alkylene-C(O)NH—C1-C5 alkyl, —C(O)NH2, or —NO2, wherein each occurrence of Ra is independently —H, -benzyl, or —C1-C10 alkyl;

or N and both R2 groups are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle which is unsubstituted or substituted with one to three of —C1-C5 alkyl, -phenyl, —C1-C5 alkylene phenyl, -hydroxy-substituted C1-C5 alkyl, -halo, -halo-substituted C1-C5 alkyl, -halo-substituted phenyl, -phenylene-O—C1-C5 alkyl, -cyano-substituted phenyl, —OH, —O—C1-C5 alkyl, —N(Ra)2, —C1-C5 alkylene-N(Ra)2, —C1-C5 alkylene-C(O)O—C1-C5 alkylene-N(Ra)2, —COOH, —C1-C5 alkylene-COOH, —OP(O)(OH)2, —OS(O)2OH , —C1-C5 alkylene-OP(O)(OH)2, —C1-C5 alkylene-OS(O)2OH, —C(O)O—C1-C5 alkyl, —OC(O)—C1-C5 alkyl, —C1-C5 alkylene-C(O)O—C1-C5 alkyl, —C1-C5 alkylene-C(O)NH—C1-C5 alkyl, —C(O)NH2, or —NO2, wherein each occurrence of Ra is independently —H, -benzyl, or —C1-C10 alkyl;

R3 is —H, —PO3H2, —P2O6H3, —P3O9H4, —SO3H, —C(O)—C1-C9 alkyl, β-D-glucuronyl, or a C-terminal alpha amino acid residue;

n is an integer ranging from 1 to 10; and

m is an integer ranging from 2 to 10.

In one embodiment, X is —CH2—, R1 is —CH2—N(R2)(R2), and at least one R2 is methyl.

In another embodiment, X is —CH2—, R1 is —CH2—N(R2)(R2), and at least one R2 is ethyl.

In yet another embodiment, X is —CH2—, R1 is —CH2—N(R2)(R2), and at least one R2 is propyl.

In one embodiment, X is —CH2—, R1 is —CH2—N(R2)(R2), and at least one R2 is —C3-C8 monocyclic cycloalkyl.

In another embodiment, X is —CH2—, R1 is —CH2—N(R2)(R2), and at least one R2 is cyclohexyl.

In yet another embodiment, X is —CH2— and R1 is —CH2—N(R2)(R2), wherein N and both R2 groups are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle.

In one embodiment, X is —CH2— and R1 is —CH2—N(R2)(R2), wherein N and both R2 groups are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle, which is substituted with a hydroxyl.

In another embodiment, X is —CH2— and R1 is —CH2—N(R2)(R2), wherein N and both R2 groups are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle, which is substituted with a nitrogen-containing 3- to 7-membered monocyclic heterocycle.

Illustrative examples of the Indenoisoquinolinone Analogs include the compounds of Formula (I) as set forth below:

(I) Compound n —R1 X (I)(1) 1 —(CH2)n—N(CH3)2 —CH2 (I)(2) 2 —(CH2)n—N(CH3)2 —CH2 (I)(3) 3 —(CH2)n—N(CH3)2 —CH2 (I)(4) 4 —(CH2)n—N(CH3)2 —CH2 (I)(5) 5 —(CH2)n—N(CH3)2 —CH2 (I)(6) 6 —(CH2)n—N(CH3)2 —CH2 (I)(7) 1 —(CH2)n—N(CH3)2 —O— (I)(8) 2 —(CH2)n—N(CH3)2 —O— (I)(9) 3 —(CH2)n—N(CH3)2 —O— (I)(10) 4 —(CH2)n—N(CH3)2 —O— (I)(11) 5 —(CH2)n—N(CH3)2 —O— (I)(12) 6 —(CH2)n—N(CH3)2 —O— (I)(13) 1 —CH2 (I)(14) 2 —CH2 (I)(15) 3 —CH2 (I)(16) 4 —CH2 (I)(17) 5 —CH2 (I)(18) 6 —CH2 (I)(19) 1 —O— (I)(20) 2 —O— (I)(21) 3 —O— (I)(22) 4 —O— (I)(23) 5 —O— (I)(24) 6 —O— (I)(25) 1 —(CH2)n—OH —CH2 (I)(26) 2 —(CH2)n—OH —CH2 (I)(27) 3 —(CH2)n—OH —CH2 (I)(28) 4 —(CH2)n—OH —CH2 (I)(29) 5 —(CH2)n—OH —CH2 (I)(30) 6 —(CH2)n—OH —CH2 (I)(31) 1 —(CH2)n—OH —O— (I)(32) 2 —(CH2)n—OH —O— (I)(33) 3 —(CH2)n—OH —O— (I)(34) 4 —(CH2)n—OH —O— (I)(35) 5 —(CH2)n—OH —O— (I)(36) 6 —(CH2)n—OH —O— Compound m —R1 X (I)(37) 2 —O—(CH2)m—N(CH3)2 —CH2 (I)(38) 3 —O—(CH2)m—N(CH3)2 —CH2 (I)(39) 4 —O—(CH2)m—N(CH3)2 —CH2 (I)(40) 5 —O—(CH2)m—N(CH3)2 —CH2 (I)(41) 6 —O—(CH2)m—N(CH3)2 —CH2 (I)(42) 2 —O—(CH2)m—N(CH3)2 —O— (I)(43) 3 —O—(CH2)m—N(CH3)2 —O— (I)(44) 4 —O—(CH2)m—N(CH3)2 —O— (I)(45) 5 —O—(CH2)m—N(CH3)2 —O— (I)(46) 6 —O—(CH2)m—N(CH3)2 —O— (I)(47) 2 —CH2 (I)(48) 3 —CH2 (I)(49) 4 —CH2 (I)(50) 5 —CH2 (I)(51) 6 —CH2 (I)(52) 2 —O— (I)(53) 3 —O— (I)(54) 4 —O— (I)(55) 5 —O— (I)(56) 6 —O— (I)(57) 2 —O—(CH2)m—OH —CH2 (I)(58) 3 —O—(CH2)m—OH —CH2 (I)(59) 4 —O—(CH2)m—OH —CH2 (I)(60) 5 —O—(CH2)m—OH —CH2 (I)(61) 6 —O—(CH2)m—OH —CH2 (I)(62) 2 —O—(CH2)m—OH —O— (I)(63) 3 —O—(CH2)m—OH —O— (I)(64) 4 —O—(CH2)m—OH —O— (I)(65)(a) 5 —O—(CH2)m—OH —O— (I)(65)(b) 6 —O—(CH2)m—OH —O— Compound —R1 X (I)(66) —CH2—N(CH3)2 —NH— (I)(67) —CH2—N(CH3)2 —S— (I)(68) —CH2—N(CH3)2 —C(O)— (I)(69) —CH2—N(CH2—CH3)2 —CH2 (I)(70) —CH2—N(CH2—CH3)2 —O— (I)(71) —CH2—N(CH2—CH3)2 —NH— (I)(72) —CH2—N(CH2—CH3)2 —S— (I)(73) —CH2—N(CH2—CH3)2 —C(O)— (I)(74) —CH2—N(CH2—CH2—CH3)2 —CH2 (I)(75) —CH2—N(CH2—CH2—CH3)2 —O— (I)(76) —CH2—N(CH2—CH2—CH3)2 —NH— (I)(77) —CH2—N(CH2—CH2—CH3)2 —S— (I)(78) —CH2—N(CH2—CH2—CH3)2 —C(O)— (I)(79) —CH2—N(CH2—CH2OH)2 —CH2 (I)(80) —CH2—N(CH2—CH2OH)2 —O— (I)(81) —CH2—N(CH2—CH2OH)2 —NH— (I)(82) —CH2—N(CH2—CH2OH)2 —S— (I)(83) —CH2—N(CH2—CH2OH)2 —C(O)— (I)(84) —CH2—N(CH2—CH2—N(CH3)2)2 —CH2 (I)(85) —CH2—N(CH2—CH2—N(CH3)2)2 —O— (I)(86) —CH2—N(CH2—CH2—N(CH3)2)2 —NH— (I)(87) —CH2—N(CH2—CH2—N(CH3)2)2 —S— (I)(88) —CH2—N(CH2—CH2—N(CH3)2)2 —C(O)— (I)(89) —CH2—N(CH2—CH2—CH2—N(CH3)2)2 —CH2 (I)(90) —CH2—N(CH2—CH2—CH2—N(CH3)2)2 —O— (I)(91) —CH2—N(CH2—CH2—CH2—N(CH3)2)2 —NH— (I)(92) —CH2—N(CH2—CH2—CH2—N(CH3)2)2 —S— (I)(93) —CH2—N(CH2—CH2—CH2—N(CH3)2)2 —C(O)— (I)(94) —CH2 (I)(95) —O— (I)(96) —NH— (I)(97) —S— (I)(98) —C(O)— (I)(99) —CH2 (I)(100) —O— (I)(101) —NH— (I)(102) —S— (I)(103) —C(O)— (I)(104) —CH2 (I)(105) —O— (I)(106) —NH— (I)(107) —S— (I)(108) —C(O)— (I)(109) —CH2 (I)(110) —O— (I)(111) —NH— (I)(112) —S— (I)(113) —C(O)— (I)(114) —CH2 (I)(115) —O— (I)(116) —NH— (I)(117) —S— (I)(118) —C(O)— (I)(119) —CH2 (I)(120) —O— (I)(121) —NH— (I)(122) —S— (I)(123) —C(O)— (I)(124) —CH2 (I)(125) —O— (I)(126) —NH— (I)(127) —S— (I)(128) —C(O)— (I)(129) —CH2 (I)(130) —S— (I)(131)(a) —C(O)— (I)(131)(b) —CH2 (I)(132) —O— (I)(133) —NH— (I)(134) —S— (I)(135) —C(O)— (I)(136) —CH2 (I)(137) —O— (I)(138) —NH— (I)(139) —S— (I)(140) —C(O)— (I)(141) —CH2 (I)(142) —O— (I)(143) —NH— (I)(144) —S— (I)(145) —C(O)— (I)(146) —CH2 (I)(147) —O— (I)(148) —NH— (I)(149) —S— (I)(150) —C(O)— (I)(151) —CH2 (I)(152) —O— (I)(153) —NH— (I)(154) —S— (I)(155) —C(O)— (I)(156) —CH2 (I)(157) —O— (I)(158) —NH— (I)(159) —S— (I)(160) —C(O)— (I)(161)(a) —CH2 (I)(161)(b) —O— (I)(162) —NH— (I)(163) —S— (I)(164) —C(O)— (I)(165) —CH2 (I)(166) —O— (I)(167) —NH— (I)(168) —S— (I)(169) —C(O)— (I)(170) —CH2 (I)(171) —O— (I)(172) —NH— (I)(173) —S— (I)(174) —C(O)— (I)(175) —CH2 (I)(176) —O— (I)(177) —NH— (I)(178) —S— (I)(179) —C(O)— (I)(180) —CH2 (I)(181) —O— (I)(182) —NH— (I)(183) —S— (I)(184) —C(O)— (I)(185) —CH2 (I)(186) —O— (I)(187) —NH— (I)(188) —S— (I)(189) —C(O)— (I)(190) —CH2 (I)(191) —O— (I)(192) —NH— (I)(193) —S— (I)(194) —C(O)— (I)(195) —CH2 (I)(196) —O— (I)(197) —NH— (I)(198) —S— (I)(199) —C(O)— (I)(200) —CH2 (I)(201) —O— (I)(202) —NH— (I)(203) —S— (I)(204) —C(O)— (I)(205) —CH2—OH —NH— (I)(206) —CH2—OH —S— (I)(207) —CH2—OH —C(O)— (I)(208) —CH2—CH2—OH —NH— (I)(209) —CH2—CH2—OH —S— (I)(210) —CH2—CH2—OH —C(O)— (I)(211) —CH2—CH2—CH2—OH —NH— (I)(212) —CH2—CH2—CH2—OH —S— (L)(213) —CH2—CH2—CH2—OH —C(O)— (I)(214) —CH2 (I)(215) —O— (I)(216) —NH— (I)(217) —S— (I)(218) —C(O)— (I)(219) —CH2—CH2—CH2—OP(O)(OH)2 —CH2 (I)(220) —CH2—CH2—CH2—OP(O)(OH)2 —O— (1)(221) —CH2—CH2—CH2—OP(O)(OH)2 —NH— (I)(222) —CH2—CH2—CH2—OP(O)(OH)2 —S— (I)(223) —CH2—CH2—CH2—OP(O)(OH)2 —C(O)— (I)(224) —CH2—CH2—CH2—OS(O)2OH —CH2 (I)(225) —CH2—CH2—CH2—OS(O)2OH —O— (I)(226) —CH2—CH2—CH2—OS(O)2OH —NH— (I)(227) —CH2—CH2—CH2—OS(O)2OH —S— (I)(228) —CH2—CH2—CH2—OS(O)2OH —C(O)— (I)(229) —CH2 (I)(230) —O— (I)(231) —NH— (I)(232) —S— (I)(233) —C(O)— (I)(234) —CH2 (I)(235) —O— (I)(236) —NH— (I)(237) —S— (I)(238) —C(O)— (I)(239) —(CH2)10—N(CH2—CH2—O—CH3)2 —CH2 (I)(240) —(CH2)10—N(CH2—CH2—O—CH3)2 —O— (I)(241) —(CH2)10—N(CH2—CH2—O—CH3)2 —NH— (I)(242) —(CH2)10—N(CH2—CH2—O—CH3)2 —S— (I)(243) —(CH2)10—N(CH2—CH2—O—CH3)2 —C(O)— (I)(244) —CH2—N(CH2—CH2—O—CH3)2 —CH2 (I)(245) —CH2 (I)(246) —CH2 (I)(247) —CH2 (I)(248) —CH2 (I)(249) —CH2 (I)(250) —CH2 (I)(251) —CH2 (I)(252) —CH2 (I)(253) —CH2 (I)(254) —CH2—NH—CH2—CH2—CH2—N(CH3)2 —CH2 (I)(255) —CH2 (I)(256) —CH2 (I)(257) —CH2 (I)(258) —CH2 (I)(259) —CH2 (I)(260) —CH2 (I)(261) —CH2 (I)(262) —CH2 (I)(263) —CH2 (I)(264) —CH2 (I)(265) —CH2 (I)(266) —CH2 (I)(267) —CH2 (I)(268) —CH2 (I)(269) —CH2 (I)(270) —CH2 (I)(271) —CH2 (I)(272) —CH2 (I)(273) —CH2 (I)(274) —CH2 (I)(275) —CH2—NH—CH3 —CH2 (I)(276) —CH2—NH—CH2—CH3 —CH2 (I)(277) —CH2—NH—CH2—CH2—CH3 —CH2 (I)(278) —CH2 (I)(279) —CH2 (I)(280) —CH2 (I)(281) —CH2 (I)(282) —CH2 (I)(283) —CH2 (I)(284) —CH2 (I)(285) —CH2 (I)(286) —CH2 (I)(287) —CH2 (I)(288) —CH2 (I)(289) —CH2 (I)(290) —CH2 (I)(291) —CH2 (I)(292) —CH2 (I)(293) —CH2 (I)(294) —CH2 (I)(295) —CH2 (I)(296) —CH2 (I)(297) —CH2 (I)(298) —CH2—N(CH2CH3)(CH3) —CH2 (I)(299) —CH2—N(CH2CH2CH3)(CH3) —CH2 (I)(300) —CH2—N(CH2CH2CH2CH3)(CH3) —CH2 (I)(301) —CH2—NH—CH2CH2CH2CH3 —CH2 (I)(302) —CH2—NH—CH2CH2—O—CH3 —CH2 (I)(303) —CH2 (I)(304) —CH2 (I)(305) —CH2 (I)(306) —CH2 (I)(307) —CH2 (I)(308) —CH2 (I)(309) —CH2

and pharmaceutically acceptable salts thereof.

5.3 The Indenoisoquinolinone Analogs of Formula (IIa)

The present invention provides Indenoisoquinolinone Analogs according to Formula (IIa), below:

and pharmaceutically acceptable salts thereof,
wherein:

X and R1 are as defined above for the compounds of Formula (IIa).

In one embodiment, X is —CH2—. In another embodiment, X is —O—. In another embodiment, X is —C(O)—. In another embodiment, X is —NH—. In another embodiment, X is —S—.

In another embodiment, R3 is —H.

In one embodiment, R1 is —(CH2)n—N(R2)(R2) and X is —CH2—.

In another embodiment, R1 is —(CH2)n—N(R2)(R2) and X is —O—.

In another embodiment, R1 is —(CH2)n—N(R2)(R2) and X is —C(O)—.

In another embodiment, R1 is —(CH2)n—N(R2)(R2) and X is —NH—.

In another embodiment, R1 is —(CH2)n—N(R2)(R2) and X is —S—.

In one embodiment, R1 is —O—(CH2)m—N(R2)(R2) and X is —CH2—.

In another embodiment, R1 is —O—(CH2)m—N(R2)(R2) and X is —O—.

In another embodiment, R1 is —O—(CH2)m—N(R2)(R2) and X is —C(O)—.

In another embodiment, R1 is —O—(CH2)m—N(R2)(R2) and X is —NH—.

In another embodiment, R1 is —O—(CH2)m—N(R2)(R2) and X is —S—.

In one embodiment, R1 is —O—(CH2)m—OR3 and X is —CH2—.

In another embodiment, R1 is —O—(CH2)m—OR3 and X is —O—.

In another embodiment, R1 is —O—(CH2)m—OR3 and X is —C(O)—.

In another embodiment, R1 is —O—(CH2)m—OR3 and X is —NH—.

In another embodiment, R1 is —O—(CH2)m—OR3 and X is —S—.

In one embodiment, one R2 is —H, and the other R2 is —C1-C6 alkyl.

In another embodiment, each R2 is —C1-C6 alkyl.

In another embodiment, each R2 is -methyl.

In one embodiment, R3 is —H.

In another embodiment, R3 is —PO3H2.

In another embodiment, R3 is —P2O6H3.

In another embodiment, R3 is —P3O9H4.

In another embodiment, R3 is —SO3H.

In yet another embodiment, R3 is —PO3Na2.

In yet another embodiment, R3 is —P2O6Na3.

In yet another embodiment, R3 is —P3O9Na4.

In yet another embodiment, R3 is —SO3Na.

In another embodiment, R3 is —C(O)—C1-C9 alkyl.

In another embodiment, R3 is β-D-glucuronyl.

In another embodiment, R3 is a C-terminal alpha amino acid residue.

In one embodiment, n is 1.

In another embodiment, n is 2.

In yet another embodiment, n is 3.

In a further embodiment, n is 4, 5, or 6.

In yet a further embodiment, n is 7, 8, or 9.

In still a further embodiment, n is 10.

In one embodiment, m is 2.

In another embodiment, m is 3.

In yet another embodiment, m is 4, 5, or 6.

In a further embodiment, m is 7, 8, or 9.

In yet a further embodiment, m is 10.

In various embodiments, —N(R2)(R2) is:

In other embodiments, —N(R2)(R2) is:

In still other embodiments, —N(R2)(R2) is

    • —N(CH2CH3)(CH3),
    • —N(CH2CH2CH3)(CH3),
    • —N(CH2CH2CH2CH3)(CH3),
    • —NH—CH2CH2CH2CH3, or
    • —NH—CH2CH2—O—CH3.

In some embodiments, —N(R2)(R2) is

In various embodiments, —N(Z3)(Z4) is:

In other embodiments, —N(Z3)(Z4) is:

In still other embodiments, —N(Z3)(Z4) is


—N(CH2CH3)(CH3),


—N(CH2CH2CH3)(CH3),


—N(CH2CH2CH2CH3)(CH3),


—NH—CH2CH2CH2CH3, or


—NH—CH2CH2—O—CH3.

In some embodiments, —N(Z3)(Z4) is

In one embodiment, X is —CH2—, —O—, —C(O)—, —NH— or —S—;

R1 is —(CH2)n—N(R2)(R2), —O—(CH2)m—N(R2)(R2), or —O—(CH2)m—OR3;

each R2 is independently —H, —C1-C6 alkyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, —C1-C6 alkylene phenyl or -phenyl, each of which other than hydrogen is unsubstituted or substituted with one or more of -halo, —OH, —OP(O)(OH)2, —OS(O)2OH or —N(Z3)(Z4), where Z3 and Z4 are independently —H, —C1-C5 alkyl, or —C1-C5 alkylene-O—C1-C5 alkyl, each of which other than hydrogen is unsubstituted or substituted with one or more of -halo, —OH or —NH2;

or N, Z3 and Z4 are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle which is unsubstituted or substituted with one to three of —C1-C5 alkyl, -phenyl, —C1-C5 alkylene phenyl, -hydroxy-substituted C1-C5 alkyl, -halo, -halo-substituted C1-C5 alkyl, -halo-substituted phenyl, -phenylene-O—C1-C5 alkyl, -cyano-substituted phenyl, —OH, —O—C1-C5 alkyl, —N(Ra)2, —C1-C5 alkylene-N(Ra)2, —C1-C5 alkylene-C(O)O—C1-C5 alkylene-N(Ra)2, —COOH, —C1-C5 alkylene-COOH, —OP(O)(OH)2, —OS(O)2OH, —C1-C5 alkylene-OP(O)(OH)2, —C1-C5 alkylene-OS(O)2OH, —C(O)O—C1-C5 alkyl, —OC(O)—C1-C5 alkyl, —C1-C5 alkylene-C(O)O—C1-C5 alkyl, —C1-C5 alkylene-C(O)NH—C1-C5 alkyl, —C(O)NH2, or —NO2, wherein each occurrence of Ra is independently —H, -benzyl, or —C1-C10 alkyl;

or N and both R2 groups are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle which is unsubstituted or substituted with one to three of —C1-C5 alkyl, -phenyl, —C1-C5 alkylene phenyl, -hydroxy-substituted C1-C5 alkyl, -halo, -halo-substituted C1-C5 alkyl, -halo-substituted phenyl, -phenylene-O—C1-C5 alkyl, -cyano-substituted phenyl, —OH, —O—C1-C5 alkyl, —N(Ra)2, —C1-C5 alkylene-N(Ra)2, —C1-C5 alkylene-C(O)O—C1-C5 alkylene-N(Ra)2, —COOH, —C1-C5 alkylene-COOH, —OP(O)(OH)2, —OS(O)2OH, —C1-C5 alkylene-OP(O)(OH)2, —C1-C5 alkylene-OS(O)2OH, —C(O)O—C1-C5 alkyl, —OC(O)—C1-C5 alkyl, —C1-C5 alkylene-C(O)O—C1-C5 alkyl, —C1-C5 alkylene-C(O)NH—C1-C5 alkyl, —C(O)NH2, or —NO2, wherein each occurrence of Ra is independently —H, -benzyl, or —C1-C10 alkyl;

R3 is —H, —PO3H2, —P2O6H3, —P3O9H4, —SO3H, —C(O)—C1-C9 alkyl, β-D-glucuronyl, or a C-terminal alpha amino acid residue;

n is an integer ranging from 1 to 10; and

m is an integer ranging from 2 to 10.

In one embodiment, X is —CH2—, R1 is —CH2—N(R2)(R2), and at least one R2 is methyl.

In another embodiment, X is —CH2—, R1 is —CH2—N(R2)(R2), and at least one R2 is ethyl.

In yet another embodiment, X is —CH2—, R1 is —CH2—N(R2)(R2), and at least one R2 is propyl.

In one embodiment, X is —CH2—, R1 is —CH2—N(R2)(R2), and at least one R2 is —C3-C8 monocyclic cycloalkyl.

In another embodiment, X is —CH2—, R1 is —CH2—N(R2)(R2), and at least one R2 is cyclohexyl.

In yet another embodiment, X is —CH2— and R1 is —CH2—N(R2)(R2), wherein N and both R2 groups are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle.

In one embodiment, X is —CH2— and R1 is —CH2—N(R2)(R2), wherein N and both R2 groups are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle, which is substituted with a hydroxyl.

In another embodiment, X is —CH2— and R1 is —CH2—N(R2)(R2), wherein N and both R2 groups are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle, which is substituted with a nitrogen-containing 3- to 7-membered monocyclic heterocycle.

Illustrative examples of the Indenoisoquinolinone Analogs include the compounds of Formula (IIa) as set forth below:

(IIa) Compound n —R1 X (IIa)(1) 1 —(CH2)n—N(CH3)2 —CH2 (IIa)(2) 2 —(CH2)n—N(CH3)2 —CH2 (IIa)(3) 3 —(CH2)n—N(CH3)2 —CH2 (IIa)(4) 4 —(CH2)n—N(CH3)2 —CH2 (IIa)(5) 5 —(CH2)n—N(CH3)2 —CH2 (IIa)(6) 6 —(CH2)n—N(CH3)2 —CH2 (IIa)(7) 1 —(CH2)n—N(CH3)2 —O— (IIa)(8) 2 —(CH2)n—N(CH3)2 —O— (IIa)(9) 3 —(CH2)n—N(CH3)2 —O— (IIa)(10) 4 —(CH2)n—N(CH3)2 —O— (IIa)(11) 5 —(CH2)n—N(CH3)2 —O— (IIa)(12) 6 —(CH2)n—N(CH3)2 —O— (IIa)(13) 1 —CH2 (IIa)(14) 2 —CH2 (IIa)(15) 3 —CH2 (IIa)(16) 4 —CH2 (IIa)(17) 5 —CH2 (IIa)(18) 6 —CH2 (IIa)(19) 1 —O— (IIa)(20) 2 —O— (IIa)(21) 3 —O— (IIa)(22) 4 —O— (IIa)(23) 5 —O— (IIa)(24) 6 —O— Compound m —R1 X (IIa)(25) 2 —O—(CH2)m—N(CH3)2 —CH2 (IIa)(26) 3 —O—(CH2)m—N(CH3)2 —CH2 (IIa)(27) 4 —O—(CH2)m—N(CH3)2 —CH2 (IIa)(28) 5 —O—(CH2)m—N(CH3)2 —CH2 (IIa)(29) 6 —O—(CH2)m—N(CH3)2 —CH2 (IIa)(30) 2 —O—(CH2)m—N(CH3)2 —O— (IIa)(31) 3 —O—(CH2)m—N(CH3)2 —O— (IIa)(32) 4 —O—(CH2)m—N(CH3)2 —O— (IIa)(33) 5 —O—(CH2)m—N(CH3)2 —O— (IIa)(34) 6 —O—(CH2)m—N(CH3)2 —O— (IIa)(35) 2 —CH2 (IIa)(36) 3 —CH2 (IIa)(37) 4 —CH2 (IIa)(38) 5 —CH2 (IIa)(39) 6 —CH2 (IIa)(40) 2 —O— (IIa)(41) 3 —O— (IIa)(42) 4 —O— (IIa)(43) 5 —O— (IIa)(44) 6 —O— (IIa)(45) 2 —O—(CH2)m—OH —CH2 (IIa)(46) 3 —O—(CH2)m—OH —CH2 (IIa)(47) 4 —O—(CH2)m—OH —CH2 (IIa)(48) 5 —O—(CH2)m—OH —CH2 (IIa)(49) 6 —O—(CH2)m—OH —CH2 (IIa)(50) 2 —O—(CH2)m—OH —O— (IIa)(51) 3 —O—(CH2)m—OH —O— (IIa)(52) 4 —O—(CH2)m—OH —O— (IIa)(53) 5 —O—(CH2)m—OH —O— (IIa)(54) 6 —O—(CH2)m—OH —O— Compound —R1 X (IIa)(55) —CH2—N(CH3)2 —NH— (IIa)(56) —CH2—N(CH3)2 —S— (IIa)(57) —CH2—N(CH3)2 —C(O)— (IIa)(58) —CH2—N(CH2—CH3)2 —CH2 (IIa)(59) —CH2—N(CH2—CH3)2 —O— (IIa)(60) —CH2—N(CH2—CH3)2 —NH— (IIa)(61) —CH2—N(CH2—CH3)2 —S— (IIa)(62) —CH2—N(CH2—CH3)2 —C(O)— (IIa)(63) —CH2—N(CH2—CH2—CH3)2 —CH2 (IIa)(64) —CH2—N(CH2—CH2—CH3)2 —O— (IIa)(65) —CH2—N(CH2—CH2—CH3)2 —NH— (IIa)(66) —CH2—N(CH2—CH2—CH3)2 —S— (IIa)(67) —CH2—N(CH2—CH2—CH3)2 —C(O)— (IIa)(68) —CH2—N(CH2—CH2OH)2 —CH2 (IIa)(69) —CH2—N(CH2—CH2OH)2 —O— (IIa)(70) —CH2—N(CH2—CH2OH)2 —NH— (IIa)(71) —CH2—N(CH2—CH2OH)2 —S— (IIa)(72) —CH2—N(CH2—CH2OH)2 —C(O)— (IIa)(73) —CH2—N(CH2—CH2—N(CH3)2)2 —CH2 (IIa)(74) —CH2—N(CH2—CH2—N(CH3)2)2 —O— (IIa)(75) —CH2—N(CH2—CH2—N(CH3)2)2 —NH— (IIa)(76) —CH2—N(CH2—CH2—N(CH3)2)2 —S— (IIa)(77) —CH2—N(CH2—CH2—N(CH3)2)2 —C(O)— (IIa)(78) —CH2—N(CH2—CH2—CH2—N(CH3)2)2 —CH2 (IIa)(79) —CH2—N(CH2—CH2—CH2—N(CH3)2)2 —O— (IIa)(80) —CH2—N(CH2—CH2—CH2—N(CH3)2)2 —NH— (IIa)(81) —CH2—N(CH2—CH2—CH2—N(CH3)2)2 —S— (IIa)(82) —CH2—N(CH2—CH2—CH2—N(CH3)2)2 —C(O)— (IIa)(83) —CH2 (IIa)(84) —O— (IIa)(85) —NH— (IIa)(86) —S— (IIa)(87) —C(O)— (IIa)(88) —CH2 (IIa)(89) —O— (IIa)(90) —NH— (IIa)(91) —S— (IIa)(92) —C(O)— (IIa)(93) —CH2 (IIa)(94) —O— (IIa)(95) —NH— (IIa)(96) —S— (IIa)(97) —C(O)— (IIa)(98) —CH2 (IIa)(99) —O— (IIa)(100) —NH— (IIa)(101) —S— (IIa)(102) —C(O)— (IIa)(103) —CH2 (IIa)(104) —O— (IIa)(105) —NH— (IIa)(106) —S— (IIa)(107) —C(O)— (IIa)(108) —CH2 (IIa)(109) —O— (IIa)(110) —NH— (IIa)(111) —S— (IIa)(112) —C(O)— (IIa)(113) —CH2 (IIa)(114) —O— (IIa)(115) —NH— (IIa)(116) —S— (IIa)(117) —C(O)— (IIa)(118) —NH— (IIa)(119) —S— (IIa)(120) —C(O)— (IIa)(121) —CH2 (IIa)(122) —O— (IIa)(123) —NH— (IIa)(124) —S— (IIa)(125) —C(O)— (IIa)(126) —CH2 (IIa)(127) —O— (IIa)(128) —NH— (IIa)(129) —S— (IIa)(130) —C(O)— (IIa)(131) —CH2 (IIa)(132) —O— (IIa)(133) —NH— (IIa)(134) —S— (IIa)(135) —C(O)— (IIa)(136) —CH2 (IIa)(137) —O— (IIa)(138) —NH— (IIa)(139) —S— (IIa)(140) —C(O)— (IIa)(141) —CH2 (IIa)(142) —O— (IIa)(143) —NH— (IIa)(144) —S— (IIa)(145) —C(O)— (IIa)(146) —CH2 (IIa)(147) —O— (IIa)(148) —NH— (IIa)(149) —S— (IIa)(150) —C(O)— (IIa)(151) —CH2 (IIa)(152) —O— (IIa)(153) —NH— (IIa)(154) —S— (IIa)(155) —C(O)— (IIa)(156) —CH2 (IIa)(157) —O— (IIa)(158) —NH— (IIa)(159) —S— (IIa)(160) —C(O)— (IIa)(161) —CH2 (IIa)(162) —O— (IIa)(163) —NH— (IIa)(164) —S— (IIa)(165) —C(O)— (IIa)(166) —CH2 (IIa)(167) —O— (IIa)(168) —NH— (IIa)(169) —S— (IIa)(170) —C(O)— (IIa)(171) —CH2 (IIa)(172) —O— (IIa)(173) —NH— (IIa)(174) —S— (IIa)(175) —C(O)— (IIa)(176) —CH2 (IIa)(177) —O— (IIa)(178) —NH— (IIa)(179) —S— (IIa)(180) —C(O)— (IIa)(181) —CH2 (IIa)(182) —O— (IIa)(183) —NH— (IIa)(184) —S— (IIa)(185) —C(O)— (IIa)(186) —CH2 (IIa)(187) —O— (IIa)(188) —NH— (IIa)(189) —S— (IIa)(190) —C(O)— (IIa)(191) —CH2 (IIa)(192) —O— (IIa)(193) —NH— (IIa)(194) —S— (IIa)(195) —C(O)— (IIa)(196) —CH2 (IIa)(197) —O— (IIa)(198) —NH— (IIa)(199) —S— (IIa)(200) —C(O)— (IIa)(201) —CH2 (IIa)(202) —O— (IIa)(203) —NH— (IIa)(204) —S— (IIa)(205) —C(O)— (IIa)(206) —CH2 (IIa)(207) —O— (IIa)(208) —NH— (IIa)(209) —S— (IIa)(210) —C(O)— (IIa)(211) —(CH2)10—N(CH2—CH2—O—CH3)2 —CH2 (IIa)(212) —(CH2)10—N(CH2—CH2—O—CH3)2 —O— (IIa)(213) —(CH2)10—N(CH2—CH2—O—CH3)2 —NH— (IIa)(214) —(CH2)10—N(CH2—CH2—O—CH3)2 —S— (IIa)(215) —(CH2)10—N(CH2—CH2—O—CH3)2 —C(O)— (IIa)(216) —CH2 (IIa)(217) —CH2—NH—CH2—CH2—CH2—N(CH3)2 —CH2 (IIa)(218) —CH2 (IIa)(219) —CH2 (IIa)(220) —CH2 (IIa)(221) —CH2 (IIa)(222) —CH2—N(CH2—CH2—O—CH3)2 —CH2 (IIa)(223) —CH2 (IIa)(224) —CH2 (IIa)(225) —CH2 (IIa)(226) —CH2 (IIa)(227) —CH2 (IIa)(228) —CH2 (IIa)(229) —CH2 (IIa)(230) —CH2 (IIa)(231) —CH2 (IIa)(232) —CH2 (IIa)(233) —CH2 (IIa)(234) —CH2 (IIa)(235) —CH2—NH—CH2—CH3 —CH2 (IIa)(236) —CH2—NH—CH3 —CH2 (IIa)(237) —CH2 (IIa)(238) —CH2 (IIa)(239) —CH2 (IIa)(240) —CH2 (IIa)(241) —CH2 (IIa)(242) —CH2 (IIa)(243) —CH2 (IIa)(244) —CH2 (IIa)(245) —CH2 (IIa)(246) —CH2 (IIa)(247) —CH2 (IIa)(248) —CH2 (IIa)(249) —CH2 (IIa)(250) —CH2 (IIa)(251) —CH2 (IIa)(252) —CH2 (IIa)(253) —CH2 (IIa)(254) —CH2 (IIa)(255) —CH2 (IIa)(256) —CH2 (IIa)(257) —CH2 (IIa)(258) —CH2 (IIa)(259) —CH2 (IIa)(260) —CH2 (IIa)(261) —CH2 (IIa)(262) —CH2 (IIa)(263) —CH2 (IIa)(264) —CH2 (IIa)(265) —CH2 (IIa)(266) —CH2—NH—CH2—CH2—CH3 —CH2 (IIa)(267) —CH2 (IIa)(268) —CH2 (IIa)(269) —CH2 (IIa)(270) —CH2 (IIa)(271) —CH2 (IIa)(272) —CH2 (IIa)(273) —CH2—N(CH2CH3)(CH3) —CH2 (IIa)(274) —CH2—N(CH2CH2CH3)(CH3) —CH2 (IIa)(275) —CH2—N(CH2CH2CH2CH3)(CH3) —CH2 (IIa)(276) —CH2—NH—CH2CH2CH2CH3 —CH2 (IIa)(277) —CH2—NH—CH2CH2—O—CH3 —CH2 (IIa)(278) —CH2 (IIa)(279) —CH2 (IIa)(280) —CH2 (IIa)(281) —CH2 (IIa)(282) —CH2 (IIa)(283) —CH2 (IIa)(284) —CH2

and pharmaceutically acceptable salts thereof.

5.4 The Indenoisoquinolinone Analogs of Formula (IIb)

The present invention provides Indenoisoquinolinone Analogs according to Formula (IIb), below:

and pharmaceutically acceptable salts thereof,
wherein:

R1 is as defined above for the compounds of Formula (IIb).

In one embodiment, R3 is —H.

In another embodiment, R3 is —PO3H2.

In another embodiment, R3 is —P2O6H3.

In another embodiment, R3 is —P3O9H4.

In another embodiment, R3 is —SO3H.

In yet another embodiment, R3 is —PO3Na2.

In yet another embodiment, R3 is —P2O6Na3.

In yet another embodiment, R3 is —P3O9Na4.

In yet another embodiment, R3 is —SO3Na.

In another embodiment, R3—C(O)—C1-C9 alkyl.

In another embodiment, R3 is β-D-glucuronyl.

In another embodiment, R3 is a C-terminal alpha amino acid residue.

In one embodiment, n is 1.

In another embodiment, n is 2.

In yet another embodiment, n is 3.

Illustrative examples of the Indenoisoquinolinone Analogs include the compounds of Formula (IIb) as set forth below:

(IIb) Compound n —R1 (IIb)(1) 1 —(CH2)n—OH (IIb)(2) 2 —(CH2)n—OH (IIb)(3) 3 —(CH2)n—OH Compound —R1 (IIb)(4) —CH2—OP(O)(OH)2 (IIb)(5) —CH2—CH2—OP(O)(OH)2 (IIb)(6) —CH2—CH2—CH2—OP(O)(OH)2 (IIb)(7) —CH2—OS(O)2OH (IIb)(8) —CH2—CH2—OS(O)2OH (IIb)(9) —CH2—CH2—CH2—OS(O)2OH

and pharmaceutically acceptable salts thereof

5.5 The Indenoisoquinolinone Analogs of Formula (IIc)

The present invention provides Indenoisoquinolinone Analogs according to Formula (IIc), below:

and pharmaceutically acceptable salts thereof,
wherein:

X and R1 are as defined above for the compounds of Formula (IIc).

In one embodiment, X is —CH2—. In another embodiment, X is —O—. In another embodiment, X is —C(O)—. In another embodiment, X is —NH—. In another embodiment, X is —S—.

In another embodiment, R3 is —H.

In one embodiment, R1 is —(CH2)n—OR3 and X is —CH2—.

In another embodiment, R1 is —(CH2)n—OR3 and X is —O—.

In another embodiment, R1 is —(CH2)n—OR3 and X is —C(O)—.

In another embodiment, R1 is —(CH2)n—OR3 and X is —NH—.

In another embodiment, R1 is —(CH2)n—OR3 and X is —S—.

In one embodiment, R3 is —H.

In another embodiment, R3 is —PO3H2.

In another embodiment, R3 is —P2O6H3.

In another embodiment, R3 is —P3O9H4.

In another embodiment, R3 is —SO3H.

In yet another embodiment, R3 is —PO3Na2.

In yet another embodiment, R3 is —P2O6Na3.

In yet another embodiment, R3 is —P3O9Na4.

In yet another embodiment, R3 is —SO3Na.

In another embodiment, R3 is —C(O)-C1-C9 alkyl.

In another embodiment, R3 is β-D-glucuronyl.

In another embodiment, R3 is a C-terminal alpha amino acid residue.

In one embodiment, n is 1.

In another embodiment, n is 2.

In yet another embodiment, n is 3.

In a further embodiment, n is 4, 5, or 6.

In yet a further embodiment, n is 7, 8, or 9.

In still a further embodiment, n is 10.

In one embodiment, n is 1 and X is —CH2—.

In another embodiment, n is 1 and X is —S—, —NH— or —C(O)—.

In one embodiment, n is 2 and X is —CH2—.

In another embodiment, n is 2 and X is —S—, —NH— or —C(O)—.

In one embodiment, n is 3 and X is —CH2—.

In another embodiment, n is 3 and X is —S—, —NH— or —C(O)—.

In one embodiment, n is 4 and X is —O—.

In another embodiment, n is 5 and X is —O—.

In yet another embodiment, n is 6 and X is —O—.

In one embodiment, X is —CH2—, —O—, —C(O)—, —NH— or —S—;

R1 is —(CH2)n—N(R2)(R2), —O—(CH2)m—N(R2)(R2); —(CH2)n—OR3 or —O—(CH2)m—OR3;

each R2 is independently —H, —C1-C6 alkyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, —C1-C6 alkylene phenyl or -phenyl, each of which other than hydrogen is unsubstituted or substituted with one or more of -halo, —OH, —OP(O)(OH)2, —OS(O)2OH or —N(Z3)(Z4), where Z3 and Z4 are independently —H, —C1-C5 alkyl, or —C1-C5 alkylene-O—C1-C5 alkyl, each of which other than hydrogen is unsubstituted or substituted with one or more of -halo, —OH or —NH2;

or N, Z3 and Z4 are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle which is unsubstituted or substituted with one to three of —C1-C5 alkyl, -phenyl, —C1-C5 alkylene phenyl, -hydroxy-substituted C1-C5 alkyl, -halo, -halo-substituted C1-C5 alkyl, -halo-substituted phenyl, -phenylene-O—C1-C5 alkyl, -cyano-substituted phenyl, —OH, —O—C1-C5 alkyl, —N(Ra)2, —C1-C5 alkylene-N(Ra)2, —C1-C5 alkylene-C(O)O—C1-C5 alkylene-N(Ra)2, —COOH, —C1-C5 alkylene-COOH, —OP(O)(OH)2, —OS(O)2OH, —C1-C5 alkylene-OP(O)(OH)2, —C1-C5 alkylene-OS(O)2OH, —C(O)O—C1-C5 alkyl, —OC(O)—C1-C5 alkyl, —C1-C5 alkylene-C(O)O—C1-C5 alkyl, —C1-C5 alkylene-C(O)NH—C1-C5 alkyl, —C(O)NH2, or —NO2, wherein each occurrence of Ra is independently —H, -benzyl, or —C1-C10 alkyl;

or N and both R2 groups are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle which is unsubstituted or substituted with one to three of —C1-C5 alkyl, -phenyl, —C1-C5 alkylene phenyl, -hydroxy-substituted C1-C5 alkyl, -halo, -halo-substituted C1-C5 alkyl, -halo-substituted phenyl, -phenylene-O—C1-C5 alkyl, -cyano-substituted phenyl, —OH, —O—C1-C5 alkyl, —N(Ra)2, —C1-C5 alkylene-N(Ra)2, —C1-C5 alkylene-C(O)O—C1-C5 alkylene-N(Ra)2, —COOH, —C1-C5 alkylene-COOH, —OP(O)(OH)2, —OS(O)2OH, —C1-C5 alkylene-OP(O)(OH)2, —C1-C5 alkylene-OS(O)2OH, —C(O)O—C1-C5 alkyl, —OC(O)—C1-C5 alkyl, —C1-C5 alkylene-C(O)O—C1-C5 alkyl, —C1-C5 alkylene-C(O)NH—C1-C5 alkyl, —C(O)NH2, or —NO2, wherein each occurrence of Ra is independently —H, -benzyl, or —C1-C10 alkyl;

R3 is —H, —PO3H2, —P2O6H3, —P3O9H4, —SO3H, —C(O)—C1-C9 alkyl, β-D-glucuronyl, or a C-terminal alpha amino acid residue;

n is an integer ranging from 1 to 10; and

m is an integer ranging from 2 to 10.

In one embodiment, the compound of Formula (IIc) is a compound of Formula (IIa).

In another embodiment, the compound of Formula (IIc) is a compound of Formula (IIb).

Illustrative examples of the Indenoisoquinolinone Analogs include the compounds of Formula (IIc) as set forth below:

(IIc) Compound n —R1 X (IIc)(1) 1 —(CH2)n—OH —CH2 (IIc)(2) 2 —(CH2)n—OH —CH2 (IIc)(3) 3 —(CH2)n—OH —CH2 (IIc)(4) 4 —(CH2)n—OH —CH2 (IIc)(5) 5 —(CH2)n—OH —CH2 (IIc)(6) 6 —(CH2)n—OH —CH2 (IIc)(7) 4 —(CH2)n—OH —O— (IIc)(8) 5 —(CH2)n—OH —O— (IIc)(9) 6 —(CH2)n—OH —O— Compound —R1 X (IIc)(10) —CH2—OH —NH— (IIc)(11) —CH2—OH —S— (IIc)(12) —CH2—OH —C(O)— (IIc)(13) —CH2—CH2—OH —NH— (IIc)(14) —CH2—CH2—OH —S— (IIc)(15) —CH2—CH2—OH —C(O)— (IIc)(16) —CH2—CH2—CH2—OH —NH— (IIc)(17) —CH2—CH2—CH2—OH —S— (IIc)(18) —CH2—CH2—CH2—OH —C(O)— (IIc)(19) —CH2—CH2—CH2—OP(O)(OH)2 —CH2 (IIc)(20) —CH2—CH2—CH2—OP(O)(OH)2 —NH— (IIc)(21) —CH2—CH2—CH2—OP(O)(OH)2 —S— (IIc)(22) —CH2—CH2—CH2—OP(O)(OH)2 —C(O)— (IIc)(23) —CH2—CH2—CH2—OS(O)2OH —CH2 (IIc)(24) —CH2—CH2—CH2—OS(O)2OH —NH— (IIc)(25) —CH2—CH2—CH2—OS(O)2OH —S— (IIc)(26) —CH2—CH2—CH2—OS(O)2OH —C(O)—

illustrative compounds of Formula (IIa) and Formula (IIb) listed above, and pharmaceutically acceptable salts thereof.

5.6 The Indenoisoquinolinone Analogs of Formula (III)

The present invention provides Indenoisoquinolinone Analogs according to Formula (III), below:

and pharmaceutically acceptable salts thereof,
wherein:

X and R1 are as defined above for the compounds of Formula (III).

In one embodiment, X is —O—.

In another embodiment, R3 is —H.

In one embodiment, R1 is —(CH2)n—N(R2)(R2) and X is —CH2—.

In another embodiment, R1 is —(CH2)n—N(R2)(R2) and X is —O—.

In another embodiment, R1 is —(CH2)n—N(R2)(R2) and X is —C(O)—.

In another embodiment, R1 is —(CH2)n—N(R2)(R2) and X is —NH—.

In another embodiment, R1 is —(CH2)n—N(R2)(R2) and X is —S—.

In one embodiment, R1 is —(CH2)n—OR3 and X is —CH2—.

In another embodiment, R1 is —(CH2)n—OR3 and X is —O—.

In another embodiment, R1 is —(CH2)n—OR3 and X is —C(O)—.

In another embodiment, R1 is —(CH2)n—OR3 and X is —NH—.

In another embodiment, R1 is —(CH2)n—OR3 and X is —S—.

In one embodiment, R1 is —O—(CH2)m—N(R2)(R2) and X is —CH2—.

In another embodiment, R1 is —O—(CH2)m—N(R2)(R2) and X is —O—.

In another embodiment, R1 is —O—(CH2)m—N(R2)(R2) and X is —C(O)—.

In another embodiment, R1 is —O—(CH2)m—N(R2)(R2) and X is —NH—.

In another embodiment, R1 is —O—(CH2)m—N(R2)(R2) and X is —S—.

In one embodiment, R1 is —O—(CH2)m—OR3 and X is —CH2—.

In another embodiment, R1 is —O—(CH2)m—OR3 and X is —O—.

In another embodiment, R1 is —O—(CH2)m—OR3 and X is —C(O)—.

In another embodiment, R1 is —O—(CH2)m—OR3 and X is —NH—.

In another embodiment, R1 is —O—(CH2)m—OR3 and X is —S—.

In one embodiment, one R2 is —H, and the other R2 is —C1-C6 alkyl.

In another embodiment, each R2 is —C1-C6 alkyl.

In another embodiment, each R2 is -methyl.

In one embodiment, R3 is —H.

In another embodiment, R3 is —PO3H2.

In another embodiment, R3 is —P2O6H3.

In another embodiment, R3 is —P3O9H4.

In another embodiment, R3 is —SO3H.

In yet another embodiment, R3 is —PO3Na2.

In yet another embodiment, R3 is —P2O6Na3.

In yet another embodiment, R3 is —P3O9Na4.

In yet another embodiment, R3 is —SO3Na.

In another embodiment, R3 is —C(O)—C1-C9 alkyl.

In another embodiment, R3 is β-D-glucuronyl.

In another embodiment, R3 is a C-terminal alpha amino acid residue.

In one embodiment, n is 1.

In another embodiment, n is 2.

In yet another embodiment, n is 3.

In a further embodiment, n is 4, 5, or 6.

In yet a further embodiment, n is 7, 8, or 9.

In still a further embodiment, n is 10.

In one embodiment, m is 2.

In another embodiment, m is 3.

In yet another embodiment, m is 4, 5, or 6.

In a further embodiment, m is 7, 8, or 9.

In yet a further embodiment, m is 10.

In various embodiments, —N(R2)(R2) is:

In other embodiments, —N(R2)(R2) is:

In still other embodiments, —N(R2)(R2) is


—N(CH2CH3)(CH3),


—N(CH2CH2CH3)(CH3),


—N(CH2CH2CH2CH3)(CH3),


—NH—CH2CH2CH2CH3, or


—NH—CH2CH2—O—CH3.

In some embodiments, —N(R2)(R2) is

In various embodiments, —N(Z3)(Z4) is:

In other embodiments, —N(Z3)(Z4) is:

In still other embodiments, —N(Z3)(Z4) is


—N(CH2CH3)(CH3),


—N(CH2CH2CH3)(CH3),


—N(CH2CH2CH2CH3)(CH3),


—NH—CH2CH2CH2CH3, or


—NH—CH2CH2—O—CH3.

In some embodiments, —N(Z3)(Z4) is

In one embodiment, X is —CH2—, —O—, —C(O)—, —NH— or —S—;

R1 is —(CH2)n—N(R2)(R2), —O—(CH2)m—N(R2)(R2), —(CH2)n—OR3 or —O—(CH2)m—OR3;

each R2 is independently —H, —C1-C6 alkyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, —C1-C6 alkylene phenyl or -phenyl, each of which other than hydrogen is unsubstituted or substituted with one or more of -halo, —OH, —OP(O)(OH)2, —OS(O)2OH or —N(Z3)(Z4), where Z3 and Z4 are independently —H, —C1-C5 alkyl, or —C1-C5 alkylene-O—C1-C5 alkyl, each of which other than hydrogen is unsubstituted or substituted with one or more of -halo, —OH or —NH2;

or N, Z3 and Z4 are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle which is unsubstituted or substituted with one to three of —C1-C5 alkyl, -phenyl, —C1-C5 alkylene phenyl, -hydroxy-substituted C1-C5 alkyl, -halo, -halo-substituted C1-C5 alkyl, -halo-substituted phenyl, -phenylene-O—C1-C5 alkyl, -cyano-substituted phenyl, —OH, —O—C1-C5 alkyl, —N(Ra)2, —C1-C5 alkylene-N(Ra)2, —C1-C5 alkylene-C(O)O—C1-C5 alkylene-N(Ra)2, —COOH, —C1-C5 alkylene-COOH, —OP(O)(OH)2, —OS(O)2OH, —C1-C5 alkylene-OP(O)(OH)2, —C1-C5 alkylene-OS(O)2OH, —C(O)O—C1-C5 alkyl, —OC(O)—C1-C5 alkyl, —C1-C5 alkylene-C(O)O—C1-C5 alkyl, —C1-C5 alkylene-C(O)NH—C1-C5 alkyl, —C(O)NH2, or —NO2, wherein each occurrence of Ra is independently —H, -benzyl, or —C1-C10 alkyl;

or N and both R2 groups are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle which is unsubstituted or substituted with one to three of —C1-C5 alkyl, -phenyl, —C1-C5 alkylene phenyl, -hydroxy-substituted C1-C5 alkyl, -halo, -halo-substituted C1-C5 alkyl, -halo-substituted phenyl, -phenylene-O—C1-C5 alkyl, -cyano-substituted phenyl, —OH, —O—C1-C5 alkyl, —N(Ra)2, —C1-C5 alkylene-N(Ra)2, —C1-C5 alkylene-C(O)O—C1-C5 alkylene-N(Ra)2, —COOH, —C1-C5 alkylene-COOH, —OP(O)(OH)2, —OS(O)2OH, —C1-C5 alkylene-OP(O)(OH)2, —C1-C5 alkylene-OS(O)2OH, —C(O)O—C1-C5 alkyl, —OC(O)—C1-C5 alkyl, —C1-C5 alkylene-C(O)O—C1-C5 alkyl, —C1-C5 alkylene-C(O)NH—C1-C5 alkyl, —C(O)NH2, or —NO2, wherein each occurrence of Ra is independently —H, -benzyl, or —C1-C10 alkyl;

R3 is —H, —PO3H2, —P2O6H3, —P3O9H4, —SO3H, —C(O)—C1-C9 alkyl, β-D-glucuronyl, or a C-terminal alpha amino acid residue;

n is an integer ranging from 1 to 10; and

m is an integer ranging from 2 to 10.

In one embodiment, X is —CH2—, R1 is —CH2—N(R2)(R2), and at least one R2 is methyl.

In another embodiment, X is —CH2—, R1 is —CH2—N(R2)(R2), and at least one R2 is ethyl.

In yet another embodiment, X is —CH2—, R1 is —CH2—N(R2)(R2), and at least one R2 is propyl.

In one embodiment, X is —CH2—, R1 is —CH2—N(R2)(R2), and at least one R2 is —C3-C8 monocyclic cycloalkyl.

In another embodiment, X is —CH2—, R1 is —CH2—N(R2)(R2), and at least one R2 is cyclohexyl.

In yet another embodiment, X is —CH2— and R1 is —CH2—N(R2)(R2), wherein N and both R2 groups are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle.

In one embodiment, X is —CH2— and R1 is —CH2—N(R2)(R2), wherein N and both R2 groups are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle, which is substituted with a hydroxyl.

In another embodiment, X is —CH2— and R1 is —CH2—N(R2)(R2), wherein N and both R2 groups are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle, which is substituted with a nitrogen-containing 3- to 7-membered monocyclic heterocycle.

Illustrative examples of the Indenoisoquinolinone Analogs include the compounds of Formula (III) as set forth below:

(III) Compound n —R1 X (III)(1) 1 —(CH2)n—N(CH3)2 —CH2 (III)(2) 2 —(CH2)n—N(CH3)2 —CH2 (III)(3) 3 —(CH2)n—N(CH3)2 —CH2 (III)(4) 4 —(CH2)n—N(CH3)2 —CH2 (III)(5) 5 —(CH2)n—N(CH3)2 —CH2 (III)(6) 6 —(CH2)n—N(CH3)2 —CH2 (III)(7) 1 —(CH2)n—N(CH3)2 —O— (III)(8) 2 —(CH2)n—N(CH3)2 —O— (III)(9) 3 —(CH2)n—N(CH3)2 —O— (III)(10) 4 —(CH2)n—N(CH3)2 —O— (III)(11) 5 —(CH2)n—N(CH3)2 —O— (III)(12) 6 —(CH2)n—N(CH3)2 —O— (III)(13) 1 —CH2 (III)(14) 2 —CH2 (III)(15) 3 —CH2 (III)(16) 4 —CH2 (III)(17) 5 —CH2 (III)(18) 6 —CH2 (III)(19) 1 —O— (III)(20) 2 —O— (III)(21) 3 —O— (III)(22) 4 —O— (III)(23) 5 —O— (III)(24) 6 —O— (III)(25) 1 —(CH2)n—OH —CH2 (III)(26) 2 —(CH2)n—OH —CH2 (III)(27) 3 —(CH2)n—OH —CH2 (III)(28) 4 —(CH2)n—OH —CH2 (III)(29) 5 —(CH2)n—OH —CH2 (III)(30) 6 —(CH2)n—OH —CH2 (III)(31) 1 —(CH2)n—OH —O— (III)(32) 2 —(CH2)n—OH —O— (III)(33) 3 —(CH2)n—OH —O— (III)(34) 4 —(CH2)n—OH —O— (III)(35) 5 —(CH2)n—OH —O— (III)(36) 6 —(CH2)n—OH —O— Compound m —R1 X (III)(37) 2 —O—(CH2)m—N(CH3)2 —CH2 (III)(38) 3 —O—(CH2)m—N(CH3)2 —CH2 (III)(39) 4 —O—(CH2)m—N(CH3)2 —CH2 (III)(40) 5 —O—(CH2)m—N(CH3)2 —CH2 (III)(41) 6 —O—(CH2)m—N(CH3)2 —CH2 (III)(42) 2 —O—(CH2)m—N(CH3)2 —O— (III)(43) 3 —O—(CH2)m—N(CH3)2 —O— (III)(44) 4 —O—(CH2)m—N(CH3)2 —O— (III)(45) 5 —O—(CH2)m—N(CH3)2 —O— (III)(46) 6 —O—(CH2)m—N(CH3)2 —O— (III)(47) 2 —CH2 (III)(48) 3 —CH2 (III)(49) 4 —CH2 (III)(50) 5 —CH2 (III)(51) 6 —CH2 (III)(52) 2 —O— (III)(53) 3 —O— (III)(54) 4 —O— (III)(55) 5 —O— (III)(56) 6 —O— (III)(57) 2 —O—(CH2)mOH —O— (III)(58) 3 —O—(CH2)mOH —CH2 (III)(59) 4 —O—(CH2)mOH —CH2 (III)(60) 5 —O—(CH2)mOH —CH2 (III)(61) 6 —O—(CH2)mOH —CH2 (III)(62) 2 —O—(CH2)mOH —O— (III)(63) 3 —O—(CH2)mOH —O— (III)(64) 4 —O—(CH2)mOH —O— (III)(65)(a) 5 —O—(CH2)mOH —O— (III)(65)(b) 6 —O—(CH2)mOH —O— Compound —R1 X (III)(66) —CH2—N(CH3)2 —NH— (III)(67) —CH2—N(CH3)2 —S— (III)(68) —CH2—N(CH3)2 —C(O)— (III)(69) —CH2—N(CH2—CH3)2 —CH2 (III)(70) —CH2—N(CH2—CH3)2 —O— (III)(71) —CH2—N(CH2—CH3)2 —NH— (III)(72) —CH2—N(CH2—CH3)2 —S— (III)(73) —CH2—N(CH2—CH3)2 —C(O)— (III)(74) —CH2—N(CH2—CH2—CH3)2 —CH2 (III)(75) —CH2—N(CH2—CH2—CH3)2 —O— (III)(76) —CH2—N(CH2—CH2—CH3)2 —NH— (III)(77) —CH2—N(CH2—CH2—CH3)2 —S— (III)(78) —CH2—N(CH2—CH2—CH3)2 —C(O)— (III)(79) —CH2—N(CH2—CH2OH)2 —CH2 (III)(80) —CH2—N(CH2—CH2OH)2 —O— (III)(81) —CH2—N(CH2—CH2OH)2 —NH— (III)(82) —CH2—N(CH2—CH2OH)2 —S— (III)(83) —CH2—N(CH2—CH2OH)2 —C(O)— (III)(84) —CH2—N(CH2—CH2—N(CH3)2)2 —CH2 (III)(85) —CH2—N(CH2—CH2—N(CH3)2)2 —O— (III)(86) —CH2—N(CH2—CH2—N(CH3)2)2 —NH— (III)(87) —CH2—N(CH2—CH2—N(CH3)2)2 —S— (III)(88) —CH2—N(CH2—CH2—N(CH3)2)2 —C(O)— (III)(89) —CH2—N(CH2—CH2—N(CH3)2)2 —CH2 (III)(90) —CH2—N(CH2—CH2—N(CH3)2)2 —O— (III)(91) —CH2—N(CH2—CH2—N(CH3)2)2 —NH— (III)(92) —CH2—N(CH2—CH2—N(CH3)2)2 —S— (III)(93) —CH2—N(CH2—CH2—N(CH3)2)2 —C(O)— (III)(94) —CH2 (III)(95) —O— (III)(96) —NH— (III)(97) —S— (III)(98) —C(O)— (III)(99) —CH2 (III)(100) —O— (III)(101) —NH— (III)(102) —S— (III)(103) —C(O)— (III)(104) —CH2 (III)(105) —O— (III)(106) —NH— (III)(107) —S— (III)(108) —C(O)— (III)(109) —CH2 (III)(110) —O— (III)(111) —NH— (III)(112) —S— (III)(113) —C(O)— (III)(114) —CH2 (III)(115) —O— (III)(116) —NH— (III)(117) —S— (III)(118) —C(O)— (III)(119) —CH2 (III)(120) —O— (III)(121) —NH— (III)(122) —S— (III)(123) —C(O)— (III)(124) —CH2 (III)(125) —O— (III)(126) —NH— (III)(127) —S— (III)(128) —C(O)— (III)(129) —NH— (III)(130) —S— (III)(131)(a) —C(O)— (III)(131)(b) —CH2 (III)(132) —O— (III)(133) —NH— (III)(134) —S— (III)(135) —C(O)— (III)(136) —CH2 (III)(137) —O— (III)(138) —NH— (III)(139) —S— (III)(140) —C(O)— (III)(141) —O— (III)(142) —O— (III)(143) —NH— (III)(144) —S— (III)(145) —C(O)— (III)(146) —CH2 (III)(147) —O— (III)(148) —NH— (III)(149) —S— (III)(150) —C(O)— (III)(151) —CH2 (III)(152) —O— (III)(153) —NH— (III)(154) —S— (III)(155) —C(O)— (III)(156) —CH2 (III)(157) —O— (III)(158) —NH— (III)(159) —S— (III)(160) —C(O)— (III)(161)(a) —CH2 (III)(161)(b) —O— (III)(162) —NH— (III)(163) —S— (III)(164) —C(O)— (III)(165) —CH2 (III)(166) —O— (III)(167) —NH— (III)(168) —S— (III)(169) —C(O)— (III)(170) —CH2 (III)(171) —O— (III)(172) —NH— (III)(173) —S— (III)(174) —C(O)— (III)(175) —CH2 (III)(176) —O— (III)(177) —NH— (III)(178) —S— (III)(179) —C(O)— (III)(180) —CH2 (III)(181) —O— (III)(182) —NH— (III)(183) —S— (III)(184) —C(O)— (III)(185) —CH2 (III)(186) —O— (III)(187) —NH— (III)(188) —S— (III)(189) —C(O)— (III)(190) —CH2 (III)(191) —O— (III)(192) —NH— (III)(193) —S— (III)(194) —C(O)— (III)(195) —CH2 (III)(196) —O— (III)(197) —NH— (III)(198) —S— (III)(199) —C(O)— (III)(200) —CH2 (III)(201) —O— (III)(202) —NH— (III)(203) —S— (III)(204) —C(O)— (III)(205) —CH2—OH —NH— (III)(206) —CH2—OH —S— (III)(207) —CH2—OH —C(O)— (III)(208) —CH2—CH2—OH —NH— (III)(209) —CH2—CH2—OH —S— (III)(210) —CH2—CH2—OH —C(O)— (III)(211) —CH2—CH2—CH2—OH —NH— (III)(212) —CH2—CH2—CH2—OH —S— (III)(213) —CH2—CH2—CH2—OH —S(O)— (III)(214) —CH2 (III)(215) —O— (III)(216) —NH— (III)(217) —S— (III)(218) —C(O)— (III)(219) —CH2—CH2—CH2—OP(O)(OH)2 —CH2 (III)(220) —CH2—CH2—CH2—OP(O)(OH)2 —O— (III)(221) —CH2—CH2—CH2—OP(O)(OH)2 —NH— (III)(222) —CH2—CH2—CH2—OP(O)(OH)2 —S— (III)(223) —CH2—CH2—CH2—OP(O)(OH)2 —C(O)— (III)(224) —CH2—CH2—CH2—OS(O)2OH —CH2 (III)(225) —CH2—CH2—CH2—OS(O)2OH —O— (III)(226) —CH2—CH2—CH2—OS(O)2OH —NH— (III)(227) —CH2—CH2—CH2—OS(O)2OH —S— (III)(228) —CH2—CH2—CH2—OS(O)2OH —C(O)— (III)(229) —CH2 (III)(230) —O— (III)(231) —NH— (III)(232) —S— (III)(233) —C(O)— (III)(234) —CH2 (III)(235) —O— (III)(236) —NH— (III)(237) —S— (III)(238) —C(O)— (III)(239) —(CH2)10—N(CH2—CH2—O—CH3)2 —CH2 (III)(240) —(CH2)10—N(CH2—CH2—O—CH3)2 —O— (III)(241) —(CH2)10—N(CH2—CH2—O—CH3)2 —NH— (III)(242) —(CH2)10—N(CH2—CH2—O—CH3)2 —S— (III)(243) —(CH2)10—N(CH2—CH2—O—CH3)2 —C(O)— (III)(244) —CH2—N(CH2—CH2—O—CH3)2 —CH2 (III)(245) —CH2 (III)(246) —CH2 (III)(247) —CH2 (III)(248) —CH2 (III)(249) —CH2 (III)(250) —CH2 (III)(251) —CH2 (III)(252) —CH2 (III)(253) —CH2 (III)(254) —CH2—NH—CH2—CH2—N(CH3)2 —CH2 (III)(255) —CH2 (III)(256) —CH2 (III)(257) —CH2 (III)(258) —CH2 (III)(259) —CH2 (III)(260) —CH2 (III)(261) —CH2 (III)(262) —CH2 (III)(263) —CH2 (III)(264) —CH2 (III)(265) —CH2 (III)(266) —CH2 (III)(267) —CH2 (III)(268) —CH2 (III)(269) —CH2 (III)(270) —CH2 (III)(271) —CH2 (III)(272) —CH2 (III)(273) —CH2 (III)(274) —CH2 (III)(275) —CH2—NH—CH3 —CH2 (III)(276) —CH2—NH—CH2—CH3 —CH2 (III)(277) —CH2—NH—CH2—CH2—CH3 —CH2 (III)(278) —CH2 (III)(279) —CH2 (III)(280) —CH2 (III)(281) —CH2 (III)(282) —CH2 (III)(283) —CH2 (III)(284) —CH2 (III)(285) —CH2 (III)(286) —CH2 (III)(287) —CH2 (III)(288) —CH2 (III)(289) —CH2 (III)(290) —CH2 (III)(291) —CH2 (III)(292) —CH2 (III)(293) —CH2 (III)(294) —CH2 (III)(295) —CH2 (III)(296) —CH2 (III)(297) —CH2 (III)(298) —CH2—N(CH2CH3)(CH3) —CH2 (III)(299) —CH2—N(CH2CH2CH3)(CH3) —CH2 (III)(300) —CH2—N(CH2CH2CH2CH3)(CH3) —CH2 (III)(301) —CH2—NH—CH2CH2CH2CH3 —CH2 (III)(302) —CH2—NH—CH2CH2—O—CH3 —CH2 (III)(303) —CH2 (III)(304) —CH2 (III)(305) —CH2 (III)(306) —CH2 (III)(307) —CH2 (III)(308) —CH2 (III)(309) —CH2

and pharmaceutically acceptable salts thereof.

5.7 Methods For Making Indenoisoquinolinone Analogs

Methods useful for making the Indenoisoquinolinone Analogs are set forth in the Examples below and generalized in Schemes 1-8.

wherein n and each R2 is defined above for the Indenoisoquinolinone Analogs of Formula (I), (IIa), (IIc), or (III).

As shown in Scheme 1a, a compound of formula 1 can be reacted with methanol in the presence of an acid, for example, sulphuric acid, to provide a compound of formula 2. The compound of formula 2 can then be reacted with cyanide to provide a compound of formula 3. The compound of formula 3 can be brominated using, for example, N-bromosuccinimide, to provide a compound of formula 4. The compound of formula 4 can then be reacted with homophthalic anhydride to provide a compound of formula 5. The compound of formula 5 can then be reduced, for example with lithium aluminum hydride (LAH), to provide a compound of formula 6, which is an Indenoisoquinolinone Analog of Formula (I), (IIc), or (III), wherein R1═—(CH2)n—OH and n is as defined above.

As shown in Scheme 1b, an indenoisoquinolinone compound of formula 6a can be reacted with hydrochloric acid or hydrobromic acid in the presence of formaldehyde to provide a halomethyl compound of formula 7a. The compound of formula 7a can then be reacted with an amine of formula NH(R2)(R2) to provide an Indenoisoquinolinone Analog of Formula (I), (IIa), (IIc), or (III), wherein R1═—(CH2)—N(R2)(R2), wherein each R2 is independently as defined above.

wherein n and R3 are defined above for the Indenoisoquinolinone Analogs of Formula (I), (IIc), or (III).

As shown in Scheme 2a, a compound of formula 2a can be reacted with cyanide to provide a compound of formula 3a. The compound of formula 3a can be brominated using, for example, N-bromosuccinimide, to provide a compound of formula 4a. The compound of formula 4a can then be reacted with homophthalic anhydride to provide a compound of formula 5a, which is an Indenoisoquinolinone Analog of Formula (I), (IIc), or (III), wherein R1═—(CH2)n—OR3 and n and R3 are as defined above for the Indenoisoquinolinone Analogs of Formula (I), (IIc), or (III), or R3 is benzyl, tetrahydropyranyl (THP), or a silyl group such as trimethylsilyl (TMS) or tert-butyldimethylsilyl (TBDMS).

A compound of formula 6 or 5a can be brominated to provide a compound of formula 7 (see, e.g. Scheme 1a). The compound of formula 7 can then be reacted with an amine of formula NH(R2)(R2) to provide an Indenoisoquinolinone Analog of Formula (I), (IIa), (IIc), or (III), wherein R1═—(CH2)n—N(R2)(R2), wherein n and each R2 is as defined above.

wherein n is defined above for the Indenoisoquinolinone Analogs of Formula (I), (IIc), or (III).

As shown in Scheme 2b, a compound of formula 6 can be reacted with SO3-pyridine followed by treatment with an acid, to provide an Indenoisoquinolinone Analog of Formula (I), (IIc), or (III), wherein R1 is —(CH2)n—OS(O)2H.

Alternatively, a compound of formula 6 can be reacted with ClPO(OEt)2 in the presence of a base, such as NEt3, followed by treatment with an acid, to provide an Indenoisoquinolinone Analog of Formula (I), (IIc), or (III), wherein R1 is —(CH2)n—OP(O)(OH)2.

Alternatively, a compound of formula 6 can be reacted with a carboxylic acid in the presence of dicyclodihexylcarbodiimide (DCC) and a base to provide an Indenoisoquinolinone Analog of Formula (I), (IIc), or (III), wherein R1═—(CH2)n—OR3 and R3 is —C(O)—C1-C9 alkyl.

wherein n and each R2 is defined above for the Indenoisoquinolinone Analogs of Formula (I), (IIa), (IIc), or (III).

As shown in Scheme 3, a compound of formula 1 can be converted to its corresponding acid chloride using thionyl chloride, and subsequently reacted with amine of formula NH(R2)(R2) to provide a compound of formula 8. The compound of formula 8 can then be used to obtain an Indenoisoquinolinone Analog of Formula (I), (IIa), (IIc), or (III), wherein R1═—(CH2)n—N(R2)(R2), as set forth in Scheme 1a, above.

wherein n is defined above for the Indenoisoquinolinone Analogs of Formula (I), (IIc), or (III).

Alternatively, as shown in Scheme 4, a compound of formula 9 can be reacted with cyanide to provide a compound of formula 10, which can be converted a compound of formula 11 via a Wittig reaction. The compound of formula 11 can then be reacted with homophthalic anhydride, using procedures of Scheme 1a, to provide a compound of formula 12, which can then be reduced to a compound of formula 6. The compound of formula 6 can be further converted to provide an Indenoisoquinolinone Analog of Formula (I), (IIa), (IIc), or (III), as set forth in Scheme 1a, above. Alternatively, the compound of formula 6 can be further converted to provide an Indenoisoquinolinone Analog of Formula (I), (IIc), or (III), as set forth in Scheme 2b, above.

wherein m, each R2 and R3 is defined above for the Indenoisoquinolinone Analogs of Formula (I), (IIa), (IIc), or (III) and A is a halogen.

As set forth in Scheme 5, a compound of formula 13a is converted to a compound of formula 14 according to procedures outlined in Scheme 1a, for example. The compound of formula 14 is then demethylated, for example using boron tribromide, to provide a compound of formula 15. A compound of formula 15 is then reacted in the presence of base with either A(CH2)mNR2R2 or A(CH2)mOR3, to provide an Indenoisoquinolinone Analog of Formula (I), (IIa), (IIc), or (III), wherein R1═—O—(CH2)m—N(R2)(R2) or R1═—O—(CH2)m—OR3 and m, R2 and R3 are as defined above. As also set forth in Scheme 5, a compound of formula 13b is converted, for example, according to procedures outlined in Scheme 1a, to an Indenoisoquinolinone Analog of Formula (I), (IIa), (IIc), or (III), wherein R1═—O—(CH2)m—OR3 and m and R3 are as defined above. As further set forth in Scheme 5, a compound of formula 13c is converted, for example, according to procedures outlined in Scheme 1a, to an Indenoisoquinolinone Analog of Formula (I), (IIa), (IIc), or (III), wherein R1′—O—(CH2)m—N(R2)(R2) and m and each R2 is as defined above.

wherein n is defined above for the Indenoisoquinolinone Analogs of Formula (I), (IIa), (IIb), (IIc), or (III) and X′ is —OH, —SH, or NHC(O)OMe.

As set forth in Scheme 6, a compound of formula 16 is reacted with a compound of formula 17 in the presence of triethylamine to provide a compound of formula 18, wherein X is O, S, or NC(O)OMe. The compound of formula 18 can then be converted to provide an Indenoisoquinolinone Analog of Formula (I), (IIa), (IIb), (IIc), or (III), using procedures outlined in Scheme 1a and 2b, with the compound of formula 18, wherein X is NC(O)OMe, being deprotected, for example using hydrazine, to provide a compound, wherein X is NH.

wherein n is defined above for the Indenoisoquinolinone Analogs of Formula (I), (IIa), (IIb), (IIc), or (III) and X′ is —OH, —SH, or NHC(O)OMe.

As set forth in Scheme 7, a compound of formula 19 is reacted with a compound of formula 17, to provide a compound of formula 20, wherein X is O, S, or NC(O)OMe. The compound of formula 20 is then converted to provide an Indenoisoquinolinone Analog of Formula (I), (IIa), (IIb), (IIc), or (III), wherein R1═—O—(CH2)m—N(R2)(R2) or R1═—O—(CH2)m—OR3, for example as described in Scheme 5 or 6.

wherein R1 is as defined above for the Indenoisoquinolinone Analogs of Formula (I), (IIa), (IIc), or (III).

As set forth in Scheme 8, compounds of the general formula 21, which can be prepared according to Schemes 1-5, can be oxidized to provide an Indenoisoquinolinone Analog of Formula (I), (IIa), (IIc) or (III), wherein X is C(O). In in one embodiment, the oxidation is achieved using O2 and dimethylsulfoxide. In one aspect, the O2 is that which is present in air. In another embodiment, the oxidation is achieved using Cerium(IV) triflate (Ce(OTf)4). Optional protecting groups can be used (Greene et al., Protective Groups in Organic Synthesis (3rd ed. 1999)). For example, where R1 is —(CH2)n—OH, protecting groups such as —SiEt3, can be used to protect the R1 hydroxyl group during the oxidation and can then be removed, for example with an acid.

5.8 Treatment or Prevention of a Condition

In accordance with the invention, an Indenoisoquinolinone Analog is useful for treatment or prevention of a Condition as set forth below.

5.8.1 Treatment or Prevention of an Inflammatory Disease

The Indenoisoquinolinone Analogs are useful for treating or preventing an inflammatory disease. An inflammatory disease can arise where there is an inflammation of the body tissue. These include local inflammatory responses and systemic inflammation. Examples of inflammatory diseases include, but are not limited to, lupus; pancreatitis; macular degeneration; chronic obstructive pulmonary disease; organ transplant rejection; a chronic inflammatory disease of a joint, including arthritis, rheumatoid arthritis, osteoarthritis and a bone disease associated with increased bone resorption; an inflammatory bowel disease such as ileitis, ulcerative colitis, Barrett's syndrome, and Crohn's disease; an inflammatory lung disease such as asthma, adult respiratory distress syndrome, and chronic obstructive airway disease; an inflammatory disease of the eye including corneal dystrophy, trachoma, onchocerciasis, uveitis, sympathetic ophthalmitis and endophthalmitis; a chronic inflammatory disease of the gum, including gingivitis and periodontitis; tuberculosis; leprosy; an inflammatory disease of the kidney including a uremic complication, glomerulonephritis and nephrosis; an inflammatory disease of the skin including sclerodermatitis, psoriasis and eczema; an inflammatory disease of the central nervous system, including a chronic demyelinating disease of the nervous system, multiple sclerosis, AIDS-related neurodegeneration and Alzheimer's disease, infectious meningitis, encephalomyelitis, Huntington's disease, amyotrophic lateral sclerosis and viral or autoimmune encephalitis; as well as any other disease that can have a significant inflammatory component, including preeclampsia, chronic liver failure, and brain and spinal cord trauma. The inflammatory disease can also be a systemic inflammation of the body, exemplified by gram-positive or gram negative shock, hemorrhagic or anaphylactic shock, or shock induced by cancer chemotherapy in response to pro-inflammatory cytokines, e.g., shock associated with pro-inflammatory cytokines. Such shock can be induced, e.g., by a chemotherapeutic agent that is administered as a treatment for cancer.

In one embodiment, the inflammatory disease is an inflammatory disease of a joint, a chronic inflammatory disease of the gum, an inflammatory bowel disease, an inflammatory lung disease, an inflammatory disease of the central nervous system, an inflammatory disease of the eye, gram-positive shock, gram negative shock, Rickettsial shock, fungal shock, hemorrhagic shock, anaphylactic shock, traumatic shock or chemotherapeutic shock.

5.8.2 Treatment or Prevention of a Reperfusion Injury

The Indenoisoquinolinone Analogs are useful for treating or preventing a reperfusion injury. Reperfusion refers to the process whereby blood-flow in the blood vessels is resumed following ischemia, such as occurs following constriction or obstruction of the vessel. Reperfusion injury can result following a naturally occurring episode, such as a myocardial infarction, stroke, or during a surgical procedure where blood flow in vessels is intentionally or unintentionally blocked. Examples of reperfusion injuries include, but are not limited to, intestinal reperfusion injury, myocardial reperfusion injury, and reperfusion injury resulting from cardiopulmonary bypass surgery, aortic aneurysm repair surgery, carotid endarterectomy surgery, and hemorrhagic shock.

In one embodiment, the reperfusion injury results from cardiopulmonary bypass surgery, aortic aneurysm repair surgery, carotid endarterectomy surgery or hemorrhagic shock.

In another embodiment, the reperfusion injury is stroke or myocardial infarction.

5.8.3 Treatment or Prevention of Reoxygenation Injury Resulting from Organ Transplantation

The Indenoisoquinolinone Analogs are useful for treating or preventing reoxygenation injury resulting from organ transplantation. Examples of reoxygenation injuries include, but are not limited to, transplantation of one or more of the following: heart, lung, liver, kidney, pancreas, intestine and cornea.

In one embodiment, reoxygenation injury resulting from organ transplantation occurs during the organ transplantation.

The Indenoisoquinolinone Analogs are also useful for treating or preventing allograph rejection. Accordingly, the invention provides methods for treating or preventing allograph rejection, comprising administering an effective amount of an Indenoisoquinolinone Analog to a subject in need thereof. In one embodiment, the methods further comprise administering an effective amount of another agent useful for treating or preventing allograph rejection. The other agent includes, but is not limited to, SK-506 and cyclosporine.

5.8.4 Treatment or Prevention of an Ischemic Condition

The Indenoisoquinolinone Analogs are useful for treating or preventing an ischemic condition. Examples of ischemic conditions include, but are not limited to, stable angina, unstable angina, myocardial ischemia, hepatic ischemia, mesenteric artery ischemia, ischemic heart disease, intestinal ischemia, critical limb ischemia, chronic critical limb ischemia, cerebral ischemia, acute cardiac ischemia, and an ischemic disease of the central nervous system, such as stroke or cerebral ischemia.

In one embodiment, the ischemic condition is myocardial ischemia, stable angina, unstable angina, stroke, ischemic heart disease or cerebral ischemia.

5.8.5 Treatment or Prevention of Renal Failure

The Indenoisoquinolinone Analogs are useful for treating or preventing renal failure.

In one embodiment, the renal failure is chronic renal failure.

In another embodiment, the renal failure is acute renal failure.

5.8.6 Treatment or Prevention of a Vascular Disease

The Indenoisoquinolinone Analogs are useful for treating or preventing a vascular disease other than a cardiovascular disease. Examples of vascular diseases include, but are not limited to, hemorrahgic stroke, peripheral arterial occlusion, thromboangitis obliterans, Reynaud's disease and phenomenon, acrocyanosis, erythromelalgia, venous thrombosis, varicose veins, arteriovenous fistula, lymphedema, and lipedema.

In embodiment, the vascular disease is peripheral arterial occlusion, thromboangitis obliterans, Reynaud's disease and phenomenon, acrocyanosis, erythromelalgia, venous thrombosis, varicose veins, arteriovenous fistula, lymphedema or lipedema.

5.8.7 Treatment or Prevention of a Cardiovascular Disease

The Indenoisoquinolinone Analogs are useful for treating or preventing a cardiovascular disease. Examples of cardiovascular diseases include, but are not limited to, congestive heart failure (such as chronic or acute heart failure), atherosclerosis, hypercholesterolemia, circulatory shock, cardiomyopathy, cardiac transplant, myocardial infarction, and a cardiac arrhythmia, such as atrial fibrillation, supraventricular tachycardia, atrial flutter, and paroxysmal atrial tachycardia.

In one embodiment, the cardiovascular disease is chronic heart failure.

In another embodiment, the cardiovascular disease is acute heart failure.

In yet another embodiment, the cardiovascular disease is cardiac arrhythmia.

In still another embodiment, the cardiac arrhythmia is atrial fibrillation, supraventricular tachycardia, atrial flutter or paroxysmal atrial tachycardia.

In one embodiment, the cardiovascular disease is chronic heart failure, atrial fibrillation, supraventricular tachycardia, atrial flutter or paroxysmal atrial tachycardia.

5.8.8 Treatment or Prevention of Diabetes Mellitus or a Diabetic Complication

The Indenoisoquinolinone Analogs are useful for treating or preventing diabetes mellitus or one or more of its complications. Examples of diabetes mellitus include, but are not limited to, Type I diabetes (Insulin Dependent Diabetes Mellitus), Type II diabetes (Non-Insulin Dependent Diabetes Mellitus), gestational diabetes, autoimmune diabetes, insulinopathies, diabetes due to pancreatic disease, diabetes associated with other endocrine diseases (such as Cushing's Syndrome, acromegaly, pheochromocytoma, glucagonoma, primary aldosteronism or somatostatinoma), Type A insulin resistance syndrome, Type B insulin resistance syndrome, lipatrophic diabetes, and diabetes induced by β-cell toxins.

The Indenoisoquinolinone Analogs are useful for treating or preventing a complication of diabetes mellitus. Examples of complications of diabetes mellitus include, but are not limited to, diabetic cataract, glaucoma, retinopathy, nephropathy (such as microalbuminuria or progressive diabetic nephropathy), polyneuropathy, gangrene of the feet, immune-complex vasculitis, systemic lupus erythematosus (SLE), atherosclerotic coronary arterial disease, peripheral arterial disease, nonketotic hyperglycemic-hyperosmolar coma, mononeuropathies, autonomic neuropathy, foot ulcers, joint problems, and a skin or mucous membrane complication (such as an infection, a shin spot, a candidal infection or necrobiosis lipoidica diabeticorumobesity), hyperlipidemia, hypertension, syndrome of insulin resistance, coronary artery disease, retinopathy, neuropathy (such as diabetic neuropathy, polyneuropathy or mononeuropathy), autonomic neuropathy, a foot ulcer, a joint problem, a fungal infection, cardiomyopathy, and a bacterial infection.

In one embodiment diabetes mellitus is Type I diabetes mellitus or Type II diabetes mellitus.

5.8.9 Treatment or Prevention of a Neurodegenerative Disease

The Indenoisoquinolinone Analogs are useful for treating or preventing a neurodegenerative disease. Examples of neurodegenerative diseases include, but are not limited to, Alzheimer's disease, Parkinson's disease and amyotrophic lateral sclerosis.

5.8.10 Treatment or Prevention of Cancer

The Indenoisoquinolinone Analogs are useful for treating or preventing cancer.

Accordingly, the invention provides methods for treating or preventing cancer, comprising administering an effective amount of an Indenoisoquinolinone Analog to a subject in need thereof. In one embodiment, the methods further comprise administering an effective amount of another anticancer agent.

Examples of cancers include, but are not limited to, the cancers disclosed below in Table 1 and metastases thereof.

TABLE 1 Solid tumors, including but not limited to: fibrosarcoma myxosarcoma liposarcoma chondrosarcoma osteogenic sarcoma chordoma angiosarcoma endotheliosarcoma lymphangiosarcoma lymphangioendotheliosarcoma synovioma mesothelioma Ewing's tumor leiomyosarcoma rhabdomyosarcoma colon cancer colorectal cancer kidney cancer pancreatic cancer bone cancer breast cancer ovarian cancer prostate cancer esophageal cancer stomach cancer oral cancer nasal cancer throat cancer squamous cell carcinoma basal cell carcinoma adenocarcinoma sweat gland carcinoma sebaceous gland carcinoma papillary carcinoma papillary adenocarcinomas cystadenocarcinoma medullary carcinoma bronchogenic carcinoma renal cell carcinoma hepatoma bile duct carcinoma choriocarcinoma seminoma embryonal carcinoma Wilms' tumor cervical cancer uterine cancer testicular cancer small cell lung carcinoma bladder carcinoma lung cancer epithelial carcinoma skin cancer melanoma metastatic melanoma neuroblastoma retinoblastoma blood-borne cancers, including but not limited to: acute lymphoblastic leukemia (“ALL”) acute lymphoblastic B-cell leukemia acute lymphoblastic T-cell leukemia acute myeloblastic leukemia (“AML”) acute promyelocytic leukemia (“APL”) acute monoblastic leukemia acute erythroleukemic leukemia acute megakaryoblastic leukemia acute myelomonocytic leukemia acute nonlymphocyctic leukemia acute undifferentiated leukemia chronic myelocytic leukemia (“CML”) chronic lymphocytic leukemia (“CLL”) hairy cell leukemia multiple myeloma acute and chronic leukemias: lymphoblastic myelogenous lymphocytic myelocytic leukemias Lymphomas: Hodgkin's disease non-Hodgkin's Lymphoma Multiple myeloma Waldenström's macroglobulinemia Heavy chain disease Polycythemia vera Central nervous system lymphomas CNS and Brain cancers: glioma pilocytic astrocytoma astrocytoma anaplastic astrocytoma glioblastoma multiforme medulloblastoma craniopharyngioma ependymoma pinealoma hemangioblastoma acoustic neuroma oligodendroglioma meningioma vestibular schwannoma adenoma metastatic brain tumor meningioma spinal tumor medulloblastoma

In one embodiment, the cancer is lung cancer, breast cancer, colorectal cancer, prostate cancer, a leukemia, a lymphoma, non-Hodgkin's lymphoma, skin cancer, a brain cancer, a cancer of the central nervous system, ovarian cancer, uterine cancer, stomach cancer, pancreatic cancer, esophageal cancer, kidney cancer, liver cancer, or a head and neck cancer.

In another embodiment, the cancer is metastatic cancer.

In yet another embodiment, the cancer is brain cancer or melanoma.

In one embodiment, the brain cancer is metastatic brain cancer or a glioma.

In one embodiment, the glioma is pilocytic astrocytoma, astrocytoma, anaplastic astrocytoma or glioblastoma multiforme.

In one embodiment, the cancer is homologous-recombination deficient, such as BRCA-1 or BRCA-2 deficient, or is deficient in one or more proteins of the Fanconi family. In one embodiment, the deficiency is caused by a genetic mutation. In another embodiment, the phenotype resulting from the deficiency is caused by abnormally low expression of BRCA-1 or BRCA-2 protein. In another embodiment, the phenotype resulting from the deficiency is caused by abnormally low expression of one or more proteins of the Fanconi family.

In still another embodiment, the subject in need of treatment has previously undergone or is presently undergoing treatment for cancer. The treatment includes, but is not limited to, chemotherapy, radiation therapy, surgery or immunotherapy, such as administration of a cancer vaccine.

The Indenoisoquinolinone Analogs are also useful for treating or preventing a cancer caused by a virus. Such viruses include human papilloma virus, which can lead to cervical cancer (see, e.g., Hernandez-Avila et al., Archives of Medical Research (1997) 28:265-271); Epstein-Barr virus (EBV), which can lead to lymphoma (see, e.g., Herrmann et al., J Pathol (2003) 199(2):140-5); hepatitis B or C virus, which can lead to liver carcinoma (see, e.g., El-Serag, J Clin Gastroenterol (2002) 35(5 Suppl 2):S72-8); human T cell leukemia virus (HTLV)-I, which can lead to T-cell leukemia (see e.g., Mortreux et al., Leukemia (2003) 17(1):26-38); human herpesvirus-8 infection, which can lead to Kaposi's sarcoma (see, e.g., Kadow et al., Curr Opin Investig Drugs (2002) 3(11):1574-9); and Human Immune deficiency Virus (HIV) infection, which can lead to cancer as a consequence of immunodeficiency (see, e.g., Dal Maso et al., Lancet Oncol (2003) 4(2):110-9).

5.8.10.1 Prophylactic Methods for Cancer

The Indenoisoquinolinone Analogs are also useful for preventing cancer, or preventing progression of a cancer, including but not limited to the cancers listed in Table 1. Such prophylactic use includes that in which non-neoplastic cell growth such as hyperplasia, metaplasia, or most specifically, dysplasia has occurred.

Alternatively or in addition to the presence of abnormal cell growth characterized as hyperplasia, metaplasia, or dysplasia, the presence of one or more characteristics of a transformed phenotype, or of a malignant phenotype, displayed in vivo or displayed in vitro by a cell sample from a subject, can indicate the desirability of prophylactic or therapeutic administration of an Indenoisoquinolinone Analog. Such characteristics of a transformed phenotype include morphology changes, looser substratum attachment, loss of contact inhibition, loss of anchorage dependence, protease release, increased sugar transport, decreased serum requirement, expression of fetal antigens, disappearance of the 250,000 dalton cell surface protein, etc. (see also id., at pp. 84-90 for characteristics associated with a transformed or malignant phenotype).

In a specific embodiment, leukoplakia, a benign-appearing hyperplastic or dysplastic lesion of the epithelium, or Bowen's disease, a carcinoma in situ, is treatable or preventable according to the present methods.

In another embodiment, fibrocystic disease (cystic hyperplasia, mammary dysplasia, specifically adenosis (benign epithelial hyperplasia)) is treatable or preventable according to the present methods.

In other embodiments, a subject that has one or more of the following predisposing factors for malignancy can be treated by administration of an effective amount of an Indenoisoquinolinone Analog: a chromosomal translocation associated with a malignancy (e.g., the Philadelphia chromosome for chronic myelogenous leukemia; t(14;18) for follicular lymphoma); familial polyposis or Gardner's syndrome; benign monoclonal gammopathy; a first degree kinship with persons having a cancer or precancerous disease showing a Mendelian (genetic) inheritance pattern (e.g., familial polyposis of the colon, Gardner's syndrome, hereditary exostosis, polyendocrine adenomatosis, medullary thyroid carcinoma with amyloid production and pheochromocytoma, Peutz-Jeghers syndrome, neurofibromatosis of Von Recklinghausen, retinoblastoma, carotid body tumor, cutaneous melanocarcinoma, intraocular melanocarcinoma, xeroderma pigmentosum, ataxia telangiectasia, Chediak-Higashi syndrome, albinism, Fanconi's aplastic anemia, and Bloom's syndrome); and exposure to carcinogens (e.g., smoking, second-hand smoke exposure, and inhalation of or contacting with certain chemicals).

5.8.10.2 Combination Chemotherapy for the Treatment of Cancer

In one aspect, the present methods for treating or preventing cancer can further comprise the administration of another anticancer agent.

In one embodiment, the present invention provides methods for treating or preventing cancer, comprising the administration of an effective amount of an Indenoisoquinolinone Analog and another anticancer agent to a subject in need thereof.

The Indenoisoquinolinone Analog and another anticancer agent can be administered concurrently. In this embodiment, the Indenoisoquinolinone Analog and another anticancer agent can be administered within the same composition, or can be administered from different compositions, via the same or different routes of administration.

In another embodiment, the Indenoisoquinolinone Analog is administered during a time when the other anticancer agent exerts its prophylactic or therapeutic effect, or vice versa.

In another embodiment, the Indenoisoquinolinone Analog or other anticancer agent are administered in doses commonly employed when such agents are used as monotherapy for the treatment of cancer.

In one embodiment, the Indenoisoquinolinone Analog or other anticancer agent are administered in doses that are lower than the doses commonly employed when such agents are used as monotherapy for the treatment of cancer.

In another embodiment, the Indenoisoquinolinone Analog and other anticancer agent act synergistically and are administered in doses that are lower than the doses commonly employed when such agents are used as monotherapy for the treatment of cancer.

The dosage of the Indenoisoquinolinone Analog or other anticancer agent administered as well as the dosing schedule can depend on various parameters, including, but not limited to, the cancer being treated, the subject's general health, and the administering physician's discretion.

An Indenoisoquinolinone Analog can be administered prior to (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks before), concurrently with, or subsequent to (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks after) the administration of the other anticancer agent, to a subject in need thereof. In various embodiments an Indenoisoquinolinone Analog and the other anticancer agent are administered 1 minute apart, 10 minutes apart, 30 minutes apart, less than 1 hour apart, 1 hour apart, 1 hour to 2 hours apart, 2 hours to 3 hours apart, 3 hours to 4 hours apart, 4 hours to 5 hours apart, 5 hours to 6 hours apart, 6 hours to 7 hours apart, 7 hours to 8 hours apart, 8 hours to 9 hours apart, 9 hours to 10 hours apart, 10 hours to 11 hours apart, 11 hours to 12 hours apart, no more than 24 hours apart or no more than 48 hours apart. In one embodiment, an Indenoisoquinolinone Analog and the other anticancer agent are administered within 3 hours. In another embodiment, an Indenoisoquinolinone Analog and the other anticancer agent are administered at 1 minute to 24 hours apart.

In one embodiment, an effective amount of an Indenoisoquinolinone Analog and an effective amount of other anticancer agent are present in the same composition. In one embodiment, this composition is useful for oral administration. In another embodiment, this composition is useful for intravenous administration.

In one embodiment, the compositions comprise an amount of an Indenoisoquinolinone Analog and the other anticancer agent which together are effective to treat or prevent cancer.

In another embodiment, the compositions comprise an effective amount of temozolomide, procarbazine, dacarbazine, interleukin-2, irinotecan, or doxorubicin, a physiologically acceptable carrier or vehicle, and an effective amount of an Indenoisoquinolinone Analog.

In one embodiment, the amount of an Indenoisoquinolinone Analog and the other anticancer agent is at least about 0.01% of the combined combination chemotherapy agents by weight of the composition. When intended for oral administration, this amount can be varied from about 0.1% to about 80% by weight of the composition. Some oral compositions can comprise from about 4% to about 50% of combined amount of an Indenoisoquinolinone Analog and the other anticancer agent by weight of the composition. Other compositions of the present invention are prepared so that a parenteral dosage unit contains from about 0.01% to about 2% by weight of the composition.

Cancers that can be treated or prevented by administering an Indenoisoquinolinone Analog and the other anticancer agent include, but are not limited to, the list of cancers set forth above in Table 1.

In one embodiment, the cancer is brain cancer.

In specific embodiments, the brain cancer is pilocytic astrocytoma, astrocytoma, anaplastic astrocytoma, glioblastoma multiforme or a metastatic brain tumor.

In one embodiment, the cancer is melanoma.

In a specific embodiment, the melanoma is metastatic melanoma.

The Indenoisoquinolinone Analog and other anticancer agent can act additively or synergistically. A synergistic combination of an Indenoisoquinolinone Analog and the other anticancer agent, might allow the use of lower dosages of one or both of these agents and/or less frequent administration of the agents to a subject with cancer. The ability to utilize lower dosages of one or both of the Indenoisoquinolinone Analog and other anticancer agent and/or to administer the agents less frequently can reduce any toxicity associated with the administration of the agents to a subject without reducing the efficacy of the agents in the treatment of cancer. In addition, a synergistic effect might result in the improved efficacy of these agents in the treatment of cancer and/or the reduction of any adverse or unwanted side effects associated with the use of either agent alone.

In one embodiment, the administration of an effective amount of an Indenoisoquinolinone Analog and an effective amount of another anticancer agent inhibits the resistance of a cancer to the other anticancer agent. In one embodiment, the cancer is a tumor.

Suitable other anticancer agents useful in the methods and compositions of the present invention include, but are not limited to temozolomide, a topoisomerase I inhibitor, procarbazine, dacarbazine, gemcitabine, capecitabine, methotrexate, taxol, taxotere, mercaptopurine, thioguanine, hydroxyurea, cytarabine, cyclophosphamide, ifosfamide, nitrosoureas, cisplatin, carboplatin, mitomycin, dacarbazine, procarbizine, etoposide, teniposide, campathecins, bleomycin, doxorubicin, idarubicin, daunorubicin, dactinomycin, plicamycin, mitoxantrone, L-asparaginase, doxorubicin, epirubicin, 5-fluorouracil, taxanes such as docetaxel and paclitaxel, leucovorin, levamisole, irinotecan, estramustine, etoposide, nitrogen mustards, BCNU, nitrosoureas such as carmustine and lomustine, vinca alkaloids such as vinblastine, vincristine and vinorelbine, platinum complexes such as cisplatin, carboplatin and oxaliplatin, imatinib mesylate, hexamethylmelamine, topotecan, tyrosine kinase inhibitors, tyrphostins herbimycin A, genistein, erbstatin, and lavendustin A.

In one embodiment, the other anticancer agent is, but is not limited to, a drug listed in Table 2.

TABLE 2 Alkylating agents Nitrogen mustards: Cyclophosphamide Ifosfamide Trofosfamide Chlorambucil Nitrosoureas: Carmustine (BCNU) Lomustine (CCNU) Alkylsulphonates: Busulfan Treosulfan Triazenes: Dacarbazine Procarbazine Temozolomide Platinum containing complexes: Cisplatin Carboplatin Aroplatin Oxaliplatin Plant Alkaloids Vinca alkaloids: Vincristine Vinblastine Vindesine Vinorelbine Taxoids: Paclitaxel Docetaxel DNA Topoisomerase Inhibitors Epipodophyllins: Etoposide Teniposide Topotecan 9-aminocamptothecin Camptothecin Crisnatol Mitomycins: Mitomycin C Anti-metabolites Anti-folates: DHFR inhibitors: Methotrexate Trimetrexate IMP dehydrogenase Inhibitors: Mycophenolic acid Tiazofurin Ribavirin EICAR Ribonuclotide reductase Hydroxyurea Inhibitors: Deferoxamine Pyrimidine Analogs: Uracil Analogs: 5-Fluorouracil Fluoxuridine Doxifluridine Ralitrexed Cytosine Analogs: Cytarabine (ara C) Cytosine arabinoside Fludarabine Gemcitabine Capecitabine Purine Analogs: Mercaptopurine Thioguanine DNA Antimetabolites: 3-HP 2′-deoxy-5-fluorouridine 5-HP alpha-TGDR aphidicolin glycinate ara-C 5-aza-2′-deoxycytidine beta-TGDR cyclocytidine guanazole inosine glycodialdehyde macebecin II Pyrazoloimidazole Hormonal therapies: Receptor antagonists: Anti-estrogen: Tamoxifen Raloxifene Megestrol LHRH agonists: Goscrclin Leuprolide acetate Anti-androgens: Flutamide Bicalutamide Retinoids/Deltoids Cis-retinoic acid Vitamin A derivative: All-trans retinoic acid (ATRA-IV) Vitamin D3 Analogs: EB 1089 CB 1093 KH 1060 Photodynamic therapies: Vertoporfin (BPD-MA) Phthalocyanine Photosensitizer Pc4 Demethoxy-hypocrellin A (2BA-2-DMHA) Cytokines: Interferon-α Interferon-β Interferon-γ Tumor necrosis factor Interleukin-2 Angiogenesis Inhibitors: Angiostatin (plasminogen fragment) antiangiogenic antithrombin III Angiozyme ABT-627 Bay 12-9566 Benefin Bevacizumab BMS-275291 cartilage-derived inhibitor (CDI) CAI CD59 complement fragment CEP-7055 Col 3 Combretastatin A-4 Endostatin (collagen XVIII fragment) Fibronectin fragment Gro-beta Halofuginone Heparinases Heparin hexasaccharide fragment HMV833 Human chorionic gonadotropin (hCG) IM-862 Interferon alpha/beta/gamma Interferon inducible protein (IP- 10) Interleukin-12 Kringle 5 (plasminogen fragment) Marimastat Metalloproteinase inhibitors (TIMPs) 2-Methoxyestradiol MMI 270 (CGS 27023A) MoAb IMC-1C11 Neovastat NM-3 Panzem PI-88 Placental ribonuclease inhibitor Plasminogen activator inhibitor Platelet factor-4 (PF4) Prinomastat Prolactin 16 kD fragment Proliferin-related protein (PRP) PTK 787/ZK 222594 Retinoids Solimastat Squalamine SS 3304 SU 5416 SU6668 SU11248 Tetrahydrocortisol-S Tetrathiomolybdate Thalidomide Thrombospondin-1 (TSP-1) TNP-470 Transforming growth factor-beta (TGF-β) Vasculostatin Vasostatin (calreticulin fragment) ZD6126 ZD 6474 farnesyl transferase inhibitors (FTI) Bisphosphonates Antimitotic agents: Allocolchicine Halichondrin B Colchicine colchicine derivative dolstatin 10 Maytansine Rhizoxin Thiocolchicine trityl cysteine Others: Isoprenylation inhibitors: Dopaminergic neurotoxins: 1-methyl-4-phenylpyridinium ion Cell cycle inhibitors: Staurosporine Actinomycins: Actinomycin D Dactinomycin Bleomycins: Bleomycin A2 Bleomycin B2 Peplomycin Anthracyclines: Daunorubicin Doxorubicin (adriamycin) Idarubicin Epirubicin Pirarubicin Zorubicin Mitoxantrone MDR inhibitors: Verapamil Ca2+ATPase inhibitors: Thapsigargin

Other additional anticancer agents that are useful in the compositions and methods of the present invention include, but are not limited to: acivicin; aclarubicin; acodazole hydrochloride; acronine; adozelesin; aldesleukin; altretamine; ambomycin; ametantrone acetate; aminoglutethimide; amsacrine; anastrozole; anthramycin; asparaginase; asperlin; azacitidine; azetepa; azotomycin; batimastat; benzodepa; bicalutamide; bisantrene hydrochloride; bisnafide dimesylate; bizelesin; bleomycin sulfate; brequinar sodium; bropirimine; busulfan; cactinomycin; calusterone; caracemide; carbetimer; carboplatin; carmustine; carubicin hydrochloride; carzelesin; cedefingol; chlorambucil; cirolemycin; cisplatin; cladribine; crisnatol mesylate; cyclophosphamide; cytarabine; dacarbazine; dactinomycin; daunorubicin hydrochloride; decitabine; dexormaplatin; dezaguanine; dezaguanine mesylate; diaziquone; docetaxel; doxorubicin; doxorubicin hydrochloride; droloxifene; droloxifene citrate; dromostanolone propionate; duazomycin; edatrexate; eflornithine hydrochloride; elsamitrucin; enloplatin; enpromate; epipropidine; epirubicin hydrochloride; erbulozole; esorubicin hydrochloride; estramustine; estramustine phosphate sodium; etanidazole; etoposide; etoposide phosphate; etoprine; fadrozole hydrochloride; fazarabine; fenretinide; floxuridine; fludarabine phosphate; fluorouracil; flurocitabine; fosquidone; fostriecin sodium; gemcitabine hydrochloride; hydroxyurea; idarubicin hydrochloride; ifosfamide; ilmofosine; interleukin-2 (including recombinant interleukin-2, or rIL2), interferon alfa-2α; interferon alfa-2β; interferon alfa-n1; interferon alfa-n3; interferon beta-Iα; interferon gamma-I3; iproplatin; irinotecan hydrochloride; lanreotide acetate; letrozole; leuprolide acetate; liarozole hydrochloride; lometrexol sodium; lomustine; losoxantrone hydrochloride; masoprocol; maytansine; mechlorethamine hydrochloride; megestrol acetate; melengestrol acetate; melphalan; menogaril; mercaptopurine; methotrexate; methotrexate sodium; metoprine; meturedepa; mitindomide; mitocarcin; mitocromin; mitogillin; mitomalcin; mitomycin; mitosper; mitotane; mitoxantrone hydrochloride; mycophenolic acid; nocodazole; nogalamycin; ormaplatin; oxisuran; paclitaxel; pegaspargase; peliomycin; pentamustine; peplomycin sulfate; perfosfamide; pipobroman; piposulfan; piroxantrone hydrochloride; plicamycin; plomestane; porfimer sodium; porfiromycin; prednimustine; procarbazine hydrochloride; puromycin; puromycin hydrochloride; pyrazofurin; riboprine; rogletimide; safingol; safingol hydrochloride; semustine; simtrazene; sparfosate sodium; sparsomycin; spirogermanium hydrochloride; spiromustine; spiroplatin; streptonigrin; streptozocin; sulofenur; talisomycin; tecogalan sodium; tegafur; teloxantrone hydrochloride; temoporfin; teniposide; teroxirone; testolactone; thiamiprine; thioguanine; thiotepa; tiazofurin; tirapazamine; toremifene citrate; trestolone acetate; triciribine phosphate; trimetrexate; trimetrexate glucuronate; triptorelin; tubulozole hydrochloride; uracil mustard; uredepa; vapreotide; verteporfin; vinblastine sulfate; vincristine sulfate; vindesine; vindesine sulfate; vinepidine sulfate; vinglycinate sulfate; vinleurosine sulfate; vinorelbine tartrate; vinrosidine sulfate; vinzolidine sulfate; vorozole; zeniplatin; zinostatin; zorubicin hydrochloride.

Further anticancer drugs that are useful in the methods and compositions of the invention include, but are not limited to: 20-epi-1,25dihydroxyvitamin D3; 5-ethynyluracil; abiraterone; aclarubicin; acylfulvene; adecypenol; adozelesin; aldesleukin; ALL-TK antagonists; altretamine; ambamustine; amidox; amifostine; aminolevulinic acid; amrubicin; amsacrine; anagrelide; anastrozole; andrographolide; angiogenesis inhibitors; antagonist D; antagonist G; antarelix; anti-dorsalizing morphogenetic protein-1; antiandrogen, prostatic carcinoma; antiestrogen; antineoplaston; antisense oligonucleotides; aphidicolin glycinate; apoptosis gene modulators; apoptosis regulators; apurinic acid; ara-CDP-DL-PTBA; arginine deaminase; asulacrine; atamestane; atrimustine; axinastatin 1; axinastatin 2; axinastatin 3; azasetron; azatoxin; azatyrosine; baccatin III derivatives; balanol; batimastat; BCR/ABL antagonists; benzochlorins; benzoylstaurosporine; beta Lactam Derivatives; beta-alethine; betaclamycin B; betulinic acid; bFGF inhibitor; bicalutamide; bisantrene; bisaziridinylsperrnine; bisnafide; bistratene A; bizeiesin; breflate; bropirimine; budotitane; buthionine sulfoximine; calcipotriol; calphostin C; camptothecin derivatives; canarypox IL-2; carboxamide-amino-triazole; carboxyamidotriazole; CaRest M3; CARN 700; cartilage derived inhibitor; carzelesin; casein kinase inhibitors (ICOS); castanospermine; cecropin B; cetrorelix; chlorins; chloroquinoxaline sulfonamide; cicaprost; cis-porphyrin; cladribine; clomifene Analogues; clotrimazole; collismycin A; collismycin B; combretastatin A4; combretastatin Analogue; conagenin; crambescidin 816; crisnatol; cryptophycin 8; cryptophycin A derivatives; curacin A; cyclopentanthraquinones; cycloplatam; cypemycin; cytarabine ocfosfate; cytolytic factor; cytostatin; dacliximab; decitabine; dehydrodidemnin B; deslorelin; dexamethasone; dexifosfamide; dexrazoxane; dexverapamil; diaziquone; didemnin B; didox; diethylnorspermine; dihydro-5-acytidine; dihydrotaxol; dioxamycin; diphenyl spiromustine; docetaxel; docosanol; dolasetron; doxifluridine; droloxifene; dronabinol; duocarmycin SA; ebselen; ecomustine; edelfosine; edrecolomab; eflornithine; elemene; emitefur; epirubicin; epristeride; estramustine Analogue; estrogen agonists; estrogen antagonists; etanidazole; etoposide phosphate; exemestane; fadrozole; fazarabine; fenretinide; filgrastim; finasteride; flavopiridol; flezelastine; fluasterone; fludarabine; fluorodaunorunicin hydrochloride; forfenimex; formestane; fostriecin; fotemustine; gadolinium texaphyrin; gallium nitrate; galocitabine; ganirelix; gelatinase inhibitors; gemcitabine; glutathione inhibitors; hepsulfam; heregulin; hexamethylene bisacetamide; hypericin; ibandronic acid; idarubicin; idoxifene; idramantone; ilmofosine; ilomastat; imidazoacridones; imiquimod; immunostimulant peptides; insulin-like growth factor-1 receptor inhibitor; interferon agonists; interferons; interleukins; iobenguane; iododoxorubicin; ipomeanol, 4-; iroplact; irsogladine; isobengazole; isohomohalicondrin B; itasetron; jasplakinolide; kahalalide F; lamellarin-N triacetate; lanreotide; leinamycin; lenograstim; lentinan sulfate; leptolstatin; letrozole; leukemia inhibiting factor; leukocyte alpha interferon; leuprolide+estrogen+progesterone; leuprorelin; levamisole; liarozole; linear polyamine Analogue; lipophilic disaccharide peptide; lipophilic platinum complexes; lissoclinamide 7; lobaplatin; lombricine; lometrexol; lonidamine; losoxantrone; lovastatin; loxoribine; lurtotecan; lutetium texaphyrin; lysofylline; lytic peptides; maitansine; mannostatin A; marimastat; masoprocol; maspin; matrilysin inhibitors; matrix metalloproteinase inhibitors; menogaril; merbarone; meterelin; methioninase; metoclopramide; MIF inhibitor; mifepristone; miltefosine; mirimostim; mismatched double stranded RNA; mitoguazone; mitolactol; mitomycin Analogues; mitonafide; mitotoxin fibroblast growth factor-saporin; mitoxantrone; mofarotene; molgramostim; monoclonal antibody, human chorionic gonadotrophin; monophosphoryl lipid A+myobacterium cell wall sk; mopidamol; multiple drug resistance gene inhibitor; multiple tumor suppressor 1-based therapy; mustard anticancer agents; mycaperoxide B; mycobacterial cell wall extract; myriaporone; N-acetyldinaline; N-substituted benzamides; nafarelin; nagrestip; naloxone+pentazocine; napavin; naphterpin; nartograstim; nedaplatin; nemorubicin; neridronic acid; neutral endopeptidase; nilutamide; nisamycin; nitric oxide modulators; nitroxide antioxidant; nitrullyn; O6-benzylguanine; octreotide; okicenone; oligonucleotides; onapristone; ondansetron; ondansetron; oracin; oral cytokine inducer; ormaplatin; osaterone; oxaliplatin; oxaunomycin; paclitaxel; paclitaxel Analogues; paclitaxel derivatives; palauamine; palmitoylrhizoxin; pamidronic acid; panaxytriol; panomifene; parabactin; pazelliptine; pegaspargase; peldesine; pentosan polysulfate sodium; pentostatin; pentrozole; perflubron; perfosfamide; perillyl alcohol; phenazinomycin; phenylacetate; phosphatase inhibitors; picibanil; pilocarpine hydrochloride; pirarubicin; piritrexim; placetin A; placetin B; plasminogen activator inhibitor; platinum complex; platinum complexes; platinum-triamine complex; porfimer sodium; porfiromycin; prednisone; propyl bis-acridone; prostaglandin J2; proteasome inhibitors; protein A-based immune modulator; protein kinase C inhibitor; protein kinase C inhibitors, microalgal; protein tyrosine phosphatase inhibitors; purine nucleoside phosphorylase inhibitors; purpurins; pyrazoloacridine; pyridoxylated hemoglobin polyoxyethylene conjugate; raf antagonists; raltitrexed; ramosetron; ras famesyl protein transferase inhibitors; ras inhibitors; ras-GAP inhibitor; retelliptine demethylated; rhenium Re 186 etidronate; rhizoxin; ribozymes; RII retinamide; rogletimide; rohitukine; romurtide; roquinimex; rubiginone B1; ruboxyl; safingol; saintopin; SarCNU; sarcophytol A; sargramostim; Sdi 1 mimetics; semustine; senescence derived inhibitor 1; sense oligonucleotides; signal transduction inhibitors; signal transduction modulators; single chain antigen binding protein; sizofiran; sobuzoxane; sodium borocaptate; sodium phenylacetate; solverol; somatomedin binding protein; sonermin; sparfosic acid; spicamycin D; spiromustine; splenopentin; spongistatin 1; squalamine; stem cell inhibitor; stem-cell division inhibitors; stipiamide; stromelysin inhibitors; sulfinosine; superactive vasoactive intestinal peptide antagonist; suradista; suramin; swainsonine; synthetic glycosaminoglycans; tallimustine; tamoxifen methiodide; tauromustine; tazarotene; tecogalan sodium; tegafur; tellurapyrylium; telomerase inhibitors; temoporfin; temozolomide; teniposide; tetrachlorodecaoxide; tetrazomine; thaliblastine; thiocoraline; thrombopoietin; thrombopoietin mimetic; thymalfasin; thymopoietin receptor agonist; thymotrinan; thyroid stimulating hormone; tin ethyl etiopurpurin; tirapazamine; titanocene bichloride; topsentin; toremifene; totipotent stem cell factor; translation inhibitors; tretinoin; triacetyluridine; triciribine; trimetrexate; triptorelin; tropisetron; turosteride; tyrosine kinase inhibitors; tyrphostins; UBC inhibitors; ubenimex; urogenital sinus-derived growth inhibitory factor; urokinase receptor antagonists; vapreotide; variolin B; vector system, erythrocyte gene therapy; velaresol; veramine; verdins; verteporfin; vinorelbine; vinxaltine; vitaxin; vorozole; zanoterone; zeniplatin; zilascorb; and zinostatin stimalamer.

In one another embodiment, the other anticancer agent is interferon-α.

In another embodiment, the other anticancer agent is interleukin-2.

In one embodiment, the other anticancer agent is an alkylating agent, such as a nitrogen mustard, a nitrosourea, an alkylsulfonate, a triazene, or a platinum-containing agent.

In one embodiment, the other anticancer agent is a triazene alkylating agent.

In another embodiment, the other anticancer agent is temozolomide, procarbazine, dacarbazine, interleukin-2, irinotecan, doxorubicin, or a combination thereof.

In a specific embodiment, the other anticancer agent is temozolomide.

Temozolomide can be administered to a subject at dosages ranging from about 60 mg/m2 (of a subject's body surface area) to about 250 mg/m2 and from about 100 mg/m2 to about 200 mg/m2. In specific embodiments, the dosages of temozolomide are about 10 mg/m2, about 1 mg/m2, about 5 mg/m2, about 10 mg/m2, about 20 mg/m2, about 30 mg/m2, about 40 mg/m2, about 50 mg/m2, about 60 mg/m2, about 70 mg/m2, about 80 mg/m2, about 90 mg/m2, about 100 mg/m2, about 110 mg/m2, about 120 mg/m2, about 130 mg/m2, about 140 mg/m2, about 150 mg/m2, about 160 mg/m2, about 170 mg/m2, about 180 mg/m2, about 190 mg/m2, about 200 mg/m2, about 210 mg/m2, about 220 mg/m2, about 230 mg/m2, about 240 mg/m2, or about 250 mg/m2.

In a specific embodiment, temozolomide is administered orally.

In one embodiment, temozolomide is administered orally to a subject at a dose ranging from about 150 mg/m 2 to about 200 mg/m2.

In another embodiment, temozolomide is administered orally to a subject once per day for five consecutive days at a dose ranging from about 150 mg/m2 to about 200 mg/m2.

In a specific embodiment, temozolomide is administered orally to a subject once per day for five consecutive days at a dose ranging from about 150 mg/m2 to about 200 mg/m2 on days 1-5, then again orally once per day for five consecutive days on days 28-32 at a dose ranging from about 150 mg/m2 to about 200 mg/m2, then again orally once per day for five consecutive days on days 55-59 at a dose ranging from about 150 mg/m2 to about 200 mg/m2.

In another embodiment, temozolomide is administered orally to a subject once per day for a week, two weeks, three weeks, a month or longer at the foregoing daily dosage.

In a specific embodiment, the other anticancer agent is procarbazine.

Procarbazine can be administered to a subject at dosages ranging from about 50 mg/m2 (of a subject's body surface area) to about 100 mg/m2 and from about 60 mg/m2 to about 100 mg/m2. In specific embodiments, the dosages of procarbazine are about 10 mg/m2, about 1 mg/m2, about 5 mg/m2, about 10 mg/m2, about 20 mg/m2, about 30 mg/m2, about 40 mg/m2, about 50 mg/m2, about 60 mg/m2, about 70 mg/m2, about 80 mg/m2, about 90 mg/m2, about 100 mg/m2, about 110 mg/m2, about 120 mg/m2, about 130 mg/m2, about 140 mg/m2, about 150 mg/m2, about 160 mg/m2, about 170 mg/m2, about 180 mg/m2, about 190 mg/m2, about 200 mg/m2, about 210 mg/m2, about 220 mg/m2, about 230 mg/m2, about 240 mg/m2, about 250 mg/m2, about 260 mg/m2, about 270 mg/m2, about 280 mg/m2, about 290 mg/m2, about 300 mg/m2, about 310 mg/m2, about 320 mg/m2, about 330 mg/m2, about 340 mg/m2, about 350 mg/m2, about 360 mg/m2, about 370 mg/m2, about 380 mg/m2, about 390 mg/m2, about 400 mg/m2, about 410 mg/m2, about 420 mg/m2, about 430 mg/m2, about 440 mg/m2, about 450 mg/m2, about 460 mg/m2, about 470 mg/m2, about 480 mg/m2, about 490 mg/m2, or about 500 mg/m2.

In a specific embodiment, procarbazine is administered intravenously.

In one embodiment, procarbazine is administered intravenously to a subject at a dose ranging from about 50 mg/m2 to about 100 mg/m2.

In another embodiment, procarbazine is administered intravenously to a subject once per day for five consecutive days at a dose ranging from about 50 mg/m2 to about 100 mg/m2.

In a specific embodiment, procarbazine is administered intravenously to a subject once per day for five consecutive days at a dose ranging from about 50 mg/m2 to about 100 mg/m2 on days 1-5, then again intravenously once per day for five consecutive days on days 28-32 at a dose ranging from about 50 mg/m2 to about 100 mg/m2, then again intravenously once per day for five consecutive days on days 55-59 at a dose ranging from about 50 mg/m2 to about 100 mg/m2.

In another embodiment, procarbazine is administered once intravenously to a subject at a dose ranging from about 50 mg/m2 to about 100 mg/m2.

In another embodiment, procarbazine is administered intravenously to a subject once per day for a week, two weeks, three weeks, a month or longer at the foregoing daily dosage.

In a specific embodiment, the other anticancer agent is dacarbazine.

Dacarbazine can be administered to a subject at dosages ranging from about 60 mg/m2 (of a subject's body surface area) to about 250 mg/m2 and from about 150 mg/m2 to about 250 mg/m2. In specific embodiments, the dosages of dacarbazine are about 10 mg/m2, about 1 mg/m2, about 5 mg/m2, about 10 mg/m2, about 20 mg/m2, about 30 mg/m2, about 40 mg/m2, about 50 mg/m2, about 60 mg/m2, about 70 mg/m2, about 80 mg/m2, about 90 mg/m2, about 100 mg/m2, about 110 mg/m2, about 120 mg/m2, about 130 mg/m2, about 140 mg/m2, about 150 mg/m2, about 160 mg/m2, about 170 mg/m2, about 180 mg/m2, about 190 mg/m2, about 200 mg/m2, about 210 mg/m2, about 220 mg/m2, about 230 mg/m2, about 240 mg/m2, about 250 mg/m2, about 260 mg/m2, about 270 mg/m2, about 280 mg/m2, about 290 mg/m2, about 300 mg/m2, about 310 mg/m2, about 320 mg/m2, about 330 mg/m2, about 340 mg/m2, about 350 mg/m2, about 360 mg/m2, about 370 mg/m2, about 380 mg/m2, about 390 mg/m2, about 400 mg/m2, about 410 mg/m2, about 420 mg/m2, about 430 mg/m2, about 440 mg/m2, about 450 mg/m2, about 460 mg/m2, about 470 mg/m2, about 480 mg/m2, about 490 mg/m2, or about 500 mg/m2.

In a specific embodiment, dacarbazine is administered intravenously.

In one embodiment, dacarbazine is administered intravenously to a subject at a dose ranging from about 150 mg/m2 to about 250 mg/m2.

In another embodiment, dacarbazine is administered intravenously to a subject once per day for five consecutive days at a dose ranging from about 150 mg/m2 to about 250 mg/m2.

In a specific embodiment, dacarbazine is administered intravenously to a subject once per day for five consecutive days at a dose ranging from about 150 mg/m2 to about 250 mg/m2 on days 1-5, then again intravenously once per day for five consecutive days on days 28-32 at a dose ranging from about 150 mg/m2 to about 250 mg/m2, then again intravenously once per day for five consecutive days on days 55-59 at a dose ranging from about 150 mg/m2 to about 250 mg/m2.

In one embodiment, dacarbazine is administered once intravenously to a subject at a dose ranging from about 150 m g/m2 to about 250 mg/m2.

In another embodiment, dacarbazine is administered intravenously to a subject once per day for a week, two weeks, three weeks, a month or longer at the foregoing daily dosage.

In a specific embodiment, the other anticancer agent is doxorubicin.

Doxorubicin can be administered to a subject at dosages ranging from about 50 mg/m2 (of a subject's body surface area) to about 100 mg/m2 and from about 60 mg/m2 to about 100 mg/m2. In specific embodiments, the dosages of doxorubicin are about 10 mg/m2, about 1 mg/m2, about 5 mg/m2, about 10 mg/m2, about 20 mg/m2, about 30 mg/m2, about 40 mg/m2, about 50 mg/m2, about 60 mg/m2, about 70 mg/m2, about 80 mg/m2, about 90 mg/m2, about 100 mg/m2, about 110 mg/m2, about 120 mg/m2, about 130 mg/m2, about 140 mg/m2, about 150 mg/m2, about 160 mg/m2, about 170 mg/m2, about 180 mg/m2, about 190 mg/m2, about 200 mg/m2, about 210 mg/m2, about 220 mg/m2, about 230 mg/m2, about 240 mg/m2, about 250 mg/m2, about 260 mg/m2, about 270 mg/m2, about 280 mg/m2, about 290 mg/m2, about 300 mg/m2, about 310 mg/m2, about 320 mg/m2, about 330 mg/m2, about 340 mg/m2, about 350 mg/m2, about 360 mg/m2, about 370 mg/m2, about 380 mg/m2, about 390 mg/m2, about 400 mg/m2, about 410 mg/m2, about 420 mg/m2, about 430 mg/m2, about 440 mg/m2, about 450 mg/m2, about 460 mg/m2, about 470 mg/m2, about 480 mg/m2, about 490 mg/m2, or about 500 mg/m2.

In a specific embodiment, doxorubicin is administered intravenously.

In one embodiment, doxorubicin is administered intravenously to a subject at a dose ranging from about 50 mg/m2 to about 100 mg/m2.

In another embodiment, doxorubicin is administered intravenously to a subject once per day for five consecutive days at a dose ranging from about 50 mg/m2 to about 100 mg/m2.

In a specific embodiment, doxorubicin is administered intravenously to a subject once per day for five consecutive days at a dose ranging from about 50 mg/m2 to about 100 mg/m2 on days 1-5, then again intravenously once per day for five consecutive days on days 28-32 at a dose ranging from about 50 mg/m2 to about 100 mg/m2, then again intravenously once per day for five consecutive days on days 55-59 at a dose ranging from about 50 mg/m2 to about 100 mg/m2.

In another embodiment, doxorubicin is administered once intravenously to a subject at a dose ranging from about 50 mg/m2 to about 100 mg/m2.

In another embodiment, doxorubicin is administered intravenously to a subject once per day for a week, two weeks, three weeks, a month or longer at the foregoing daily dosage.

In one embodiment, the other anticancer agent is a Topoisomerase I inhibitor, such as etoposide, teniposide, topotecan, irinotecan, 9-aminocamptothecin, camptothecin, or crisnatol.

In a specific embodiment, the other anticancer agent is irinotecan.

Irinotecan can be administered to a subject at dosages ranging from about 50 mg,/m2 (of a subject's body surface area) to about 150 mg/m2 and from about 75 mg/m2 to about 150 mg/m2. In specific embodiments, the dosages of irinotecan are about 10 mg/m2, about 1 mg/m2, about 5 mg/m2, about 10 mg/m2, about 20 mg/m2, about 30 mg/m2, about 40 mg/m2, about 50 mg/m2, about 60 mg/m2, about 70 mg/m2, about 80 mg/m2, about 90 mg/m2, about 100 mg/m2, about 110 mg/m2, about 120 mg/m2, about 130 mg/m2, about 140 mg/m2, about 150 mg/m2, about 160 mg/m2, about 170 mg/m2, about 180 mg/m2, about 190 mg/m2, about 200 mg/m2, about 210 mg/m2, about 220 mg/m2, about 230 mg/m2, about 240 mg/m2, about 250 mg/m2, about 260 mg/m2, about 270 mg/m2, about 280 mg/m2, about 290 mg/m2, about 300 mg/m2, about 310 mg/m2, about 320 mg/m2, about 330 mg/m2, about 340 mg/m2, about 350 mg/m2, about 360 mg/m2, about 370 mg/m2, about 380 mg/m2, about 390 mg/m2, about 400 mg/m2, about 410 mg/m2, about 420 mg/m2, about 430 mg/m2, about 440 mg/m2, about 450 mg/m2, about 460 mg/m2, about 470 mg/m2, about 480 mg/m2, about 490 mg/m2, or about 500 mg/m2.

In a specific embodiment, irinotecan is administered intravenously.

In one embodiment, irinotecan is administered intravenously to a subject at a dose ranging from about 50 mg/m2 to about 150 mg/m2.

In another embodiment, irinotecan is administered intravenously to a subject once per day for five consecutive days at a dose ranging from about 50 mg/m2 to about 150 mg/m2.

In a specific embodiment, irinotecan is administered intravenously to a subject once per day for five consecutive days at a dose ranging from about 50 mg/m2 to about 150 mg/m2 on days 1-5, then again intravenously once per day for five consecutive days on days 28-32 at a dose ranging from about 50 mg/m2 to about 150 mg/m2, then again intravenously once per day for five consecutive days on days 55-59 at a dose ranging from about 50 mg/m2 to about 150 mg/m2.

In another embodiment, irinotecan is administered intravenously to a subject once per day for a week, two weeks, three weeks, a month or longer at the foregoing daily dosage.

In one embodiment, the other anticancer agent is O-6-benzylguanine.

In another embodiment, the other anticancer agent is O-6-benzylguanine and temozolomide.

In another embodiment, the other anticancer agent is O-6-benzylguanine and procarbazine.

In still another embodiment, the other anticancer agent is O-6-benzylguanine and dacarbazine.

5.8.10.3 Multi-Therapy for Cancer

The Indenoisoquinolinone Analogs can be administered to a subject that has undergone or is currently undergoing one or more additional anticancer therapies including, but not limited to, surgery, radiation therapy, or immunotherapy, such as cancer vaccines.

In one embodiment, the invention provides methods for treating or preventing cancer comprising administering to a subject in need thereof an effective amount of an Indenoisoquinolinone Analog to treat or prevent cancer and another anticancer therapy including, but not limited to, surgery, radiation therapy, or immunotherapy, such as a cancer vaccine.

In one embodiment, the other anticancer therapy is radiation therapy.

In another embodiment, the other anticancer therapy is surgery.

In still another embodiment, the other anticancer therapy is immunotherapy.

In a specific embodiment, the present methods for treating or preventing cancer comprise administering an effective amount of an Indenoisoquinolinone Analog and radiation therapy. The radiation therapy can be administered concurrently with, prior to, or subsequent to the Indenoisoquinolinone Analog, in one embodiment at least an hour, five hours, 12 hours, a day, a week, a month, in another embodiment several months (e.g., up to three months), prior or subsequent to administration of the Indenoisoquinolinone Analog.

Where the other anticancer therapy is radiation therapy, any radiation therapy protocol can be administered depending upon the type of cancer to be treated. For example, but not by way of limitation, X-ray radiation can be administered; specifically, high-energy megavoltage (radiation of greater that 1 MeV energy) can be administered for deep tumors, and electron beam and orthovoltage X-ray radiation can be administered for skin cancers. Gamma-ray emitting radioisotopes, such as radioactive isotopes of radium, cobalt and other elements, can also be administered.

Additionally, the invention provides methods of treatment of cancer comprising administering an Indenoisoquinolinone Analog as an alternative to chemotherapy or radiation therapy where the chemotherapy or the radiation therapy results in a negative side effect in the subject being treated. The subject being treated can, optionally, be treated with another anticancer therapy such as surgery, radiation therapy, or immunotherapy.

The Indenoisoquinolinone Analogs can also be administered in vitro or ex vivo, such as for the treatment of certain cancers, including, but not limited to leukemias and lymphomas, such treatment involving autologous stem cell transplants. This can involve a process in which the subject's autologous hematopoietic stem cells are harvested and purged of all cancer cells, the subject's remaining bone-marrow cell population is then eradicated via the administration of an Indenoisoquinolinone Analog and/or radiation, and the resultant stem cells are infused back into the subject. Supportive care can be subsequently provided while bone marrow function is restored and the subject recovers.

5.8.11 Treatment or Prevention of Erectile Dysfunction

The Indenoisoquinolinone Analogs are useful for treating or preventing erectile dysfunction. Erectile dysfunction includes an inability to achieve or maintain a full erection, specifically that which is sufficient to achieve or maintain sexual intercourse. The inability can be a total inability, an inconsistent ability, or a tendency to maintain only a brief erection. Erectile dysfunction includes idiopathic erectile dysfunction, as well as that which can result, for example, from trauma, including mechanical trauma, specifically that resulting from surgery, to the nerves (such as during prostatectomy); diabetes mellitus; a cardiovascular disease, including atherosclerosis; radiation; or certain drugs. The erectile dysfunction can also be age-related.

In one embodiment, the erectile dysfunction results from prostate surgery.

In a further embodiment, the erectile dysfunction results from prostate nerve injury.

5.8.12 Treatment or Prevention of Urinary Incontinence

The Indenoisoquinolinone Analogs are also useful for treating or preventing urinary incontinence. Urinary incontinence can result, for example, from trauma, including mechanical trauma, specifically during childbirth or that resulting from surgery, to the nerves (such as during prostatectomy or gynecological surgery); diabetes mellitus; a cardiovascular disease, including atherosclerosis; radiation; or certain drugs. The urinary incontinence can also be age-related.

In one embodiment, the subject in need of urinary incontinence treatment or prevention is male.

In one embodiment, the subject in need of urinary incontinence treatment or prevention is female.

5.8.13 Treatment or Prevention of a Complication of Prematurity

The Indenoisoquinolinone Analogs are also useful for treating or preventing a complication of prematurity. Examples of complications of prematurity include, but are not limited to, retinopathy, hyaline-membrane disease and necrotizing enterocolitis.

5.8.14 Treatment or Prevention of Cardiomyopathy

The Indenoisoquinolinone Analogs are also useful for treating or preventing cardiomyopathy.

5.8.15 Treatment or Prevention of Retinopathy

The Indenoisoquinolinone Analogs are also useful for treating or retinopathy.

5.8.16 Treatment or Prevention of Nephropathy

The Indenoisoquinolinone Analogs are also useful for treating or preventing nephropathy.

5.8.17 Treatment or Prevention of Neuropathy

The Indenoisoquinolinone Analogs are also useful for treating or preventing neuropathy.

5.9 Therapeutic/Prophylactic Administration and Compositions of the Invention

Due to their activity, the Indenoisoquinolinone Analogs are advantageously useful in veterinary and human medicine. As described above, the Indenoisoquinolinone Analogs are useful for treating or preventing a Condition in a subject in need thereof.

The Indenoisoquinolinone Analogs can be administered in amounts that are effective to treat or prevent a Condition in a subject in need thereof.

When administered to a subject, the Indenoisoquinolinone Analogs can be administered as a component of a composition that comprises a physiologically acceptable carrier or vehicle. The present compositions, which comprise an Indenoisoquinolinone Analog, can be administered orally. The Indenoisoquinolinone Analogs can also be administered by any other convenient route, for example, by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral, rectal, or intestinal mucosa) and can be administered together with another biologically active agent. Administration can be systemic or local. Various delivery systems are known, e.g., encapsulation in liposomes, microparticles, microcapsules and capsules.

Methods of administration include, but are not limited to, intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, oral, sublingual, intracerebral, intravaginal, transdermal, rectal, by inhalation, or topical, specifically to the ears, nose, eyes, or skin. In some instances, administration will result in the release of an Indenoisoquinolinone Analog into the bloodstream.

In one embodiment, the Indenoisoquinolinone Analogs are administered orally.

In other embodiments, it can be desirable to administer the Indenoisoquinolinone Analogs locally. This can be achieved, for example, and not by way of limitation, by local infusion during surgery, topical application, e.g., in conjunction with a wound dressing after surgery, by injection, by means of a catheter, by means of a suppository or enema, or by means of an implant, said implant being of a porous, non-porous, or gelatinous material, including membranes, such as sialastic membranes, or fibers.

In certain embodiments, it can be desirable to introduce the Indenoisoquinolinone Analogs into the central nervous system or gastrointestinal tract by any suitable route, including intraventricular, intrathecal, and epidural injection, and enema. Intraventricular injection can be facilitated by an intraventricular catheter, for example, attached to a reservoir, such as an Ommaya reservoir.

Pulmonary administration can also be employed, e.g., by use of an inhaler of nebulizer, and formulation with an aerosolizing agent, or via perfusion in a fluorocarbon oar, synthetic pulmonary surfactant. In certain embodiments, the Indenoisoquinolinone Analogs can be formulated as a suppository, with traditional binders and excipients such as triglycerides.

In another embodiment Indenoisoquinolinone Analogs can be delivered in a vesicle, specifically a liposome (see Langer, Science 249:1527-1533 (1990) and Treat or prevent et al., Liposomes in Therapy of Infectious Disease and Cancer 317-327 and 353-365 (1989)).

In yet another embodiment, the Indenoisoquinolinone Analogs can be delivered in a controlled-release system or sustained-release system (see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol. 2, pp. 115-138 (1984)). Other controlled or sustained-release systems discussed in the review by Langer, Science 249:1527-1533 (1990) can be used. In one embodiment a pump can be used (Langer, Science 249:1527-1533 (1990); Sefton, CRC Crit. Ref Biomed. Eng. 14:201 (1987); Buchwald et al., Surgery 88:507 (1980); and Saudek et al., N. Engl. J Med. 321:574 (1989)). In another embodiment polymeric materials can be used (see Medical Applications of Controlled Release (Langer and Wise eds., 1974); Controlled Drug Bioavailability, Drug Product Design and Performance (Smolen and Ball eds., 1984); Ranger and Peppas, J. Macromol. Sci. Rev. Macromol. Chem. 2:61 (1983); Levy et al., Science 228:190 (1935); During et al., Ann. Neural. 25:351 (1989); and Howard et al., J. Neurosurg. 71:105 (1989)).

In yet another embodiment a controlled- or sustained-release system can be placed in proximity of a target of the Indenoisoquinolinone Analogs, e.g., the spinal column, brain, skin, lung, or gastrointestinal tract, thus requiring only a fraction of the systemic dose.

The present compositions can optionally comprise a suitable amount of a pharmaceutically acceptable excipient so as to provide the form for proper administration to the subject.

Such pharmaceutical excipients can be liquids, such as water and oils, including those of petroleum, animal, vegetable, or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. The pharmaceutical excipients can be saline, gum acacia, gelatin, starch paste, talc, keratin, colloidal silica, urea and the like. In addition, auxiliary, stabilizing, thickening, lubricating, and coloring agents can be used. In one embodiment, the pharmaceutically acceptable excipients are sterile when administered to a subject. Water is a useful excipient when the Indenoisoquinolinone Analog is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid excipients, specifically for injectable solutions. Suitable pharmaceutical excipients also include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like. The present compositions, if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents.

The present compositions can take the form of solutions, suspensions, emulsion, tablets, pills, pellets, capsules, capsules containing liquids, powders, sustained-release formulations, suppositories, emulsions. aerosols, sprays, suspensions, or any other form suitable for use. In one embodiment, the composition is in the form of a capsule (see e.g. U.S. Pat. No. 5,698,155). Other examples of suitable pharmaceutical excipients are described in Remington's Pharmaceutical Sciences 1447-1676 (Alfonso R. Gennaro eds., 19th ed. 1995), incorporated herein by reference.

In one embodiment, the Indenoisoquinolinone Analog is formulated in accordance with routine procedures as a composition adapted for oral administration to human beings. Compositions for oral delivery can be in the form of tablets, lozenges, aqueous or oily suspensions, granules, powders, emulsions, capsules, syrups, or elixirs for example. Orally administered compositions can contain one or more agents, for example, sweetening agents such as fructose, aspartame or saccharin; flavoring agents such as peppermint, oil of wintergreen, or cherry; coloring agents; and preserving agents, to provide a pharmaceutically palatable preparation. Moreover, where in tablet or pill form, the compositions can be coated to delay disintegration and absorption in the gastrointestinal tract thereby providing a sustained action over an extended period of time. Selectively permeable membranes surrounding an osmotically active driving an Indenoisoquinolinone Analog are also suitable for orally administered compositions. In these latter platforms, fluid from the environment surrounding the capsule is imbibed by the driving compound, which swells to displace the agent or agent composition through an aperture. These delivery platforms can provide an essentially zero order delivery profile as opposed to the spiked profiles of immediate release formulations. A time-delay material such as glycerol monostearate or glycerol stearate can also be useful. Oral compositions can include standard excipients such as mannitol, lactose, starch, magnesium stearate, sodium saccharin, cellulose, and magnesium carbonate. In one embodiment, the excipients are of pharmaceutical grade.

In another embodiment, the Indenoisoquinolinone Analogs can be formulated for intravenous administration. Typically, compositions for intravenous administration comprise sterile isotonic aqueous buffer. Where necessary, the compositions can also include a solubilizing agent. Compositions for intravenous administration can optionally include a local anesthetic such as lignocaine to lessen pain at the site of the injection. Generally, the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized-powder or water-free concentrate in a hermetically sealed container such as an ampule or sachette indicating the quantity of active agent. Where the Indenoisoquinolinone Analogs are to be administered by infusion, they can be dispensed, for example, with an infusion bottle containing sterile pharmaceutical grade water or saline. Where the Indenoisoquinolinone Analogs are administered by injection, an ampule of sterile water for injection or saline can be provided so that the ingredients can be mixed prior to administration.

Indenoisoquinolinone Analogs can be administered by controlled-release or sustained-release means or by delivery devices that are well known to those of ordinary skill in the art. Examples include, but are not limited to, those described in U.S. Pat. Nos. 3,845,770; 3,916,899; 3,536,809; 3,598,123; 4,008,719; 5,674,533; 5,059,595; 5,591,767; 5,120,548; 5,073,543; 5,639,476; 5,354,556; and 5,733,556, each of which is incorporated herein by reference in its entirety. Such dosage forms can be useful for providing controlled- or sustained-release of one or more active ingredients using, for example, hydropropylmethyl cellulose, other polymer matrices, gels, permeable membranes, osmotic systems, multilayer coatings, microparticles, liposomes, microspheres, or a combination thereof to provide the desired release profile in varying proportions. Suitable controlled- or sustained-release formulations known to those skilled in the art, including those described herein, can be readily selected for use with the active ingredients of the invention. The invention thus encompasses single unit dosage forms suitable for oral administration such as, but not limited to, tablets, capsules, gelcaps, and caplets that are adapted for controlled- or sustained-release.

In one embodiment a controlled- or sustained-release composition comprises a minimal amount of an Indenoisoquinolinone Analog to treat or prevent the Condition over a period of time. Advantages of controlled- or sustained-release compositions include extended activity of the drug, reduced dosage frequency, and increased subject compliance. In addition, controlled- or sustained-release compositions can favorably affect the time of onset of action or other characteristics, such as blood levels of the Indenoisoquinolinone Analog, and can thus reduce the occurrence of adverse side effects.

Controlled- or sustained-release compositions can initially release an amount of an Indenoisoquinolinone Analog that promptly produces the desired therapeutic or prophylactic effect, and gradually and continually release other amounts of the Indenoisoquinolinone Analog to maintain this level of therapeutic or prophylactic effect over an extended period of time. To maintain a constant level of the Indenoisoquinolinone Analog in the body, the Indenoisoquinolinone Analog can be released from the dosage form at a rate that will replace the amount of Indenoisoquinolinone Analog being metabolized and excreted from the body. Controlled- or sustained-release of an active ingredient can be stimulated by various conditions, including but not limited to, changes in pH, changes in temperature, concentration or availability of enzymes, concentration or availability of water, or other physiological conditions or compounds.

The amount of the Indenoisoquinolinone Analog that is effective in the treatment or prevention of a Condition can be determined by standard clinical techniques. In addition, in vitro or in vivo assays can optionally be employed to help identify optimal dosage ranges. The precise dose to be employed can also depend on the route of administration, and the seriousness of the condition being treated and can be decided according to the judgment of the practitioner and each subject's circumstances in view of, e.g., published clinical studies. Suitable effective dosage amounts, however, range from about 10 micrograms to about 5 grams about every 4 hours, although they are typically about 500 mg or less per every 4 hours. In one embodiment, the effective dosage is about 0.01 mg, 0.5 mg, about 1 mg, about 50 mg, about 100 mg, about 200 mg, about 300 mg, about 400 mg, about 500 mg, about 600 mg, about 700 mg, about 800 mg, about 900 mg, about 1 g, about 1.2 g, about 1.4 g, about 1.6 g, about 1.8 g, about 2.0 g, about 2.2 g, about 2.4 g, about 2.6 g, about 2.8 g, about 3.0 g, about 3.2 g, about 3.4 g, about 3.6 g, about 3.8 g, about 4.0 g, about 4.2 g, about 4.4 g, about 4.6 g, about 4.8 g, and about 5.0 g, every 4 hours. Equivalent dosages can be administered over various time periods including, but not limited to, about every 2 hours, about every 6 hours, about every 8 hours, about every 12 hours, about every 24 hours, about every 36 hours, about every 48 hours, about every 72 hours, about every week, about every two weeks, about every three weeks, about every month, and about every two months. The effective dosage amounts described herein refer to total amounts administered; that is, if more than one Indenoisoquinolinone Analog is administered, the effective dosage amounts correspond to the total amount administered.

Compositions can be prepared according to conventional mixing, granulating or coating methods, respectively, and the present compositions can contain, in one embodiment, from about 0.1% to about 99%; and in another embodiment from about 1% to about 70% of the Indenoisoquinolinone Analog by weight or volume.

The dosage regimen utilizing the Indenoisoquinolinone Analog can be selected in accordance with a variety of factors including type, species, age, weight, sex and medical condition of the subject; the severity of the condition to be treated; the route of administration; the renal or hepatic function of the subject; and the specific Indenoisoquinolinone Analog employed. A person skilled in the art can readily determine the effective amount of the drug useful for treating or preventing the Condition.

An Indenoisoquinolinone Analog can be administered in a single daily dose, or the total daily dosage can be administered in divided doses of two, three or four times daily. Furthermore, an Indenoisoquinolinone Analog can be administered in intranasal form via topical use of suitable intranasal vehicles, or via transdermal routes, using those forms of transdermal skin patches well known to those of ordinary skill in that art. To be administered in the form of a transdermal delivery system, the dosage administration can be continuous rather than intermittent throughout the dosage regimen. Other illustrative topical preparations include creams, ointments, lotions, aerosol sprays and gels, wherein the concentration of Indenoisoquinolinone Analog ranges from about 0.1% to about 15%, w/w or w/v.

The Indenoisoquinolinone Analogs can be assayed in vitro or in vivo for the desired therapeutic or prophylactic activity prior to use in humans. Animal model systems can be used to demonstrate safety and efficacy.

The present methods for treating or preventing a Condition in a subject in need thereof can further comprise administering another prophylactic or therapeutic agent to the subject being administered an Indenoisoquinolinone Analog. In one embodiment, the other prophylactic or therapeutic agent is administered in an effective amount. The other prophylactic or therapeutic agent includes, but is not limited to, an anti-inflammatory agent, an anti-renal failure agent, an anti-diabetic agent, and anti-cardiovasculare disease agent, an antiemetic agent, a hematopoietic colony stimulating factor, an anxiolytic agent, and an analgesic agent.

In one embodiment, the other prophylactic or therapeutic agent is an agent useful for reducing any potential side effect of an Indenoisoquinolinone Analog. Such potential side effects include, but are not limited to, nausea, vomiting, headache, low white blood cell count, low red blood cell count, low platelet count, headache, fever, lethargy, a muscle ache, general pain, bone pain, pain at an injection site, diarrhea, neuropathy, pruritis, a mouth sore, alopecia, anxiety or depression.

In one embodiment, the Indenoisoquinolinone Analog can be administered prior to, concurrently with, or after an anti-inflammatory agent, or on the same day, or within 1 hour, 2 hours, 12 hours, 24 hours, 48 hours or 72 hours of each other.

In another embodiment, the Indenoisoquinolinone Analog can be administered prior to, concurrently with, or after an anti-renal failure agent, or on the same day, or within 1 hour, 2 hours, 12 hours, 24 hours, 48 hours or 72 hours of each other.

In still another embodiment, the Indenoisoquinolinone Analog can be administered prior to, concurrently with, or after an anti-diabetic agent, or on the same day, or within 1 hour, 2 hours, 12 hours, 24 hours, 48 hours or 72 hours of each other.

In yet another embodiment, the Indenoisoquinolinone Analog can be administered prior to, concurrently with, or after an anti-cardiovascular disease agent, or on the same day, or within 1 hour, 2 hours, 12 hours, 24 hours, 48 hours or 72 hours of each other.

In a further embodiment, the Indenoisoquinolinone Analog can be administered prior to, concurrently with, or after an antiemetic agent, or on the same day, or within 1 hour, 2 hours, 12 hours, 24 hours, 48 hours or 72 hours of each other.

In another embodiment, the Indenoisoquinolinone Analog can be administered prior to, concurrently with, or after a hematopoietic colony stimulating factor, or on the same day, or within 1 hour, 2 hours, 12 hours, 24 hours, 48 hours, 72 hours, 1 week, 2 weeks, 3 weeks or 4 weeks of each other.

In still embodiment, the Indenoisoquinolinone Analog can be administered prior to, concurrently with, or after an opioid or non-opioid analgesic agent, or on the same day, or within 1 hour, 2 hours, 12 hours, 24 hours, 48 hours or 72 hours of each other.

In yet another embodiment, the Indenoisoquinolinone Analog can be administered prior to, concurrently with, or after an anxiolytic agent, or on the same day, or within 1 hour, 2 hours, 12 hours, 24 hours, 48 hours or 72 hours of each other.

Effective amounts of the other prophylactic or therapeutic agents are well known to those skilled in the art. However, it is well within the skilled artisan's purview to determine the other prophylactic or therapeutic agent's optimal effective amount range. In one embodiment of the invention, where another prophylactic or therapeutic agent is administered to a subject, the effective amount of the Indenoisoquinolinone Analog is less than its effective amount would be where the other prophylactic or therapeutic agent is not administered. In this case, without being bound by theory, it is believed that Indenoisoquinolinone Analogs and the other prophylactic or therapeutic agent act synergistically to treat or prevent a Condition.

In one embodiment, the other therapeutic or prophylactic agent is an anti-inflammatory agent. Anti-inflammatory agents useful in the methods of the present invention include but are not limited to adrenocorticosteroids, such as cortisol, cortisone, fludrocortisone, prednisone, prednisolone, 6α-methylprednisolone, triamcinolone, betamethasone, and dexamethasone; and non-steroidal anti-inflammatory agents (NSAIDs), such as aspirin, acetaminophen, indomethacin, sulindac, tolmetin, diclofenac, ketorolac, ibuprofen, naproxen, flurbiprofen, ketoprofen, fenoprofen, oxaprozin, mefenamic acid, meclofenamic acid, piroxicam, meloxicam, nabumetone, rofecoxib, celecoxib, etodolac, and nimesulide.

In one embodiment, the other therapeutic or prophylactic agent is an anti-renal failure agent. Anti-renal failure agents useful in the methods of the present invention include include but are not limited to ACE (angiotensin-converting enzyme) inhibitors, such as captopril, enalaprilat, lisinopril, benazepril, fosinopril, trandolapril, quinapril, and ramipril; diuretics, such as mannitol, glycerin, furosemide, toresemide, tripamide, chlorothiazide, methyclothiazide, indapamide, amiloride, and spironolactone; and fibric acid agents, such as clofibrate, gemfibrozil, fenofibrate, ciprofibrate, and bezafibrate.

In one embodiment, the other therapeutic or prophylactic agent is an anti-diabetic agent. Anti-diabetic agents useful in the methods of the present invention include include but are not limited to glucagons; somatostatin; diazoxide; sulfonylureas, such as tolbutamide, acetohexamide, tolazamide, chloropropamide, glybenclamide, glipizide, gliclazide, and glimepiride; insulin secretagogues, such as repaglinide, and nateglinide; biguanides, such as metformin and phenformin; thiazolidinediones, such as pioglitazone, rosiglitazone, and troglitazone; and α-glucosidase inhibitors, such as acarbose and miglitol.

In one embodiment, the other therapeutic or prophylactic agent is an anti-cardiovascular agent. Anti-cardiovascular disease agents useful in the methods of the present invention include include but are not limited to carnitine; thiamine; and muscarinic receptor antagonists, such as atropine, scopolamine, homatropine, tropicamide, pirenzipine, ipratropium, tiotropium, and tolterodine.

In one embodiment, the other therapeutic or prophylactic agent is an antiemetic agent. Antiemetic agents useful in the methods of the present invention include include, but are not limited to, metoclopromide, domperidone, prochlorperazine, promethazine, chlorpromazine, trimethobenzamide, ondansetron, granisetron, hydroxyzine, acetylleucine monoethanolamine, alizapride, azasetron, benzquinamide, bietanautine, bromopride, buclizine, clebopride, cyclizine, dimenhydrinate, diphenidol, dolasetron, meclizine, methallatal, metopimazine, nabilone, oxyperndyl, pipamazine, scopolamine, sulpiride, tetrahydrocannabinol, thiethylperazine, thioproperazine, tropisetron, and mixtures thereof.

In one embodiment, the other therapeutic or prophylactic agent is a hematopoietic colony stimulating factor. Hematopoietic colony stimulating factors useful in the methods of the present invention include, but are not limited to, filgrastim, sargramostim, molgramostim and epoietin alfa.

In one embodiment, the other therapeutic or prophylactic agent is an opioid analgesic agent. Opioid analgesic agents useful in the methods of the present invention include, but are not limited to, morphine, heroin, hydromorphone, hydrocodone, oxymorphone, oxycodone, metopon, apomorphine, normorphine, etorphine, buprenorphine, meperidine, lopermide, anileridine, ethoheptazine, piminidine, betaprodine, diphenoxylate, fentanil, sufentanil, alfentanil, remifentanil, levorphanol, dextromethorphan, phenazocine, pentazocine, cyclazocine, methadone, isomethadone and propoxyphene.

In one embodiment, the other therapeutic or prophylactic agent is a non-opioid analgesic agent. Non-opioid analgesic agents useful in the methods of the present invention include, but are not limited to, aspirin, celecoxib, rofecoxib, diclofenac, diflusinal, etodolac, fenoprofen, flurbiprofen, ibuprofen, ketoprofen, indomethacin, ketorolac, meclofenamate, mefanamic acid, nabumetone, naproxen, piroxicam and sulindac.

In one embodiment, the other therapeutic or prophylactic agent is an anxiolytic agent. Anxiolytic agents useful in the methods of the present invention include, but are not limited to, buspirone, and benzodiazepines such as diazepam, lorazepam, oxazapam, chlorazepate, clonazepam, chlordiazepoxide and alprazolam.

5.10 Kits

The invention encompasses kits that can simplify the administration of an Indenoisoquinolinone Analog to a subject.

A typical kit of the invention comprises a unit dosage form of an Indenoisoquinolinone Analog. In one embodiment, the unit dosage faun is a container, which can be sterile, containing an effective amount of an Indenoisoquinolinone Analog and a physiologically acceptable carrier or vehicle. The kit can further comprise a label or printed instructions instructing the use of the Indenoisoquinolinone Analog to treat or prevent a Condition. The kit can also further comprise a unit dosage form of another prophylactic or therapeutic agent, for example, a container containing an effective amount of the other prophylactic or therapeutic agent. In one embodiment, the kit comprises a container containing an effective amount of an Indenoisoquinolinone Analog and an effective amount of another prophylactic or therapeutic agent. Examples of other prophylactic or therapeutic agents include, but are not limited to, those listed above.

The invention is further described in the following examples, which do not limit the scope of the invention described in the claims.

6. Examples Example 1 Illustrative Indenoisoquinolinone Analogs

General Methods:

1H-NMR spectra were obtained using Varian 300 MHz spectrophotometer and chemical shifts (δ) are reported in parts per million (ppm). 1H-NMR spectra were obtained using DMSO-d6 or CDCl3 as solvents. Analytical thin layer chromatography (TLC) was performed using TLC plates pre-coated with silica gel 60 F-254. Compounds were visualized with short wavelength UV light. All final compounds were characterized on the basis of 1H-NMR or mass-spectrometry (MS) data. Homophthalic anhydride, 2-methyl-3-bromobenzoic acid, benzonitrile, and 3-methyl-4-bromobenzoic acid were obtained commercially (Sigma-Aldrich Corp., St. Louis, Mo.).

Compound (I)(25)

Methyl3-bromo-2-methylbenzoate

2 mL of concentrated sulfuric acid were added to a solution of 3-bromo-2-methylbenzoic acid (20 g, 93 mmol) in methanol (150 mL). The resultant mixture was refluxed for 6 hours. Methanol was removed under reduced pressure, and the residue was dissolved in a mixture of chloroform (100 mL) and saturated solution of Na2CO3 (100 mL). The organic layer was separated and washed with brine, dried over Na2SO4 and concentrated to provide methyl3-bromo-2-methylbenzoate (21.1 g). 1H NMR spectrum (in DMSO-d6): δ 7.79 (d, J=8.0 Hz, 1H); 7.69 (d, J=7.9 Hz, 1H); 7.23 (t, 1H); 3.82 (s, 3H).

Methyl3-cyano-2-methylbenzoate

To a flask was added methyl3-bromo-2-methylbenzoate (21 g, 91.7 mmol), potassium hexacyanoferrate (II) trihydrate (9.7 g, 22.9 mmol), Na2CO3 (9.7 g, 91.7 mmol) and Pd(OAc)2 (102 mg) in N,N-dimethylacetamide (100 mL). The flask's atmosphere was evacuated and replaced with nitrogen, and the reaction mixture heated at 120° C. for 10 hours. The reaction mixture was cooled to room temperature and diluted with ethyl acetate (150 mL). The resultant slurry was filtered and washed thoroughly with ethyl acetate (2×50 mL). The combined filtrate was washed with brine and water, dried over Na2SO4 and evaporated under reduced pressure. The residue obtained after the concentration was purified using flash chromatography (ethyl acetate-hexanes (1:9) as the eluent) to provide methyl3-cyano-2-methylbenzoate (11.2 g, 70% yield). 1H NMR spectrum (in DMSO-d6): δ 8.03 (d, J=8.1 Hz, 1H); 7.98 (d, J=7.9 Hz, 1H); 7.49 (t, 1H); 3.84 (s, 3H); 2.65 (s, 3H).

Methyl2-bromomethyl-3-cyanobenzoate

A suspension of methyl3-cyano-2-methylbenzoate (10.6 g, 60.5 mmol), NBS (16.2 g, 90.7 mmol) and azobisisobutyronitrile (AIBN) (100 mg) in tetrachloromethane (200 mL) was refluxed under nitrogen atmosphere for 6 hours. Alternatively, the bromination step was carried out using NBS in benzene, using benzoyl peroxide as a catalyst. TLC analysis showed the complete consumption of methyl3-cyano-2-methylbenzoate (TLC solvent: 10% ethyl acetate in hexane). The reaction mixture was filtered and washed with chloroform (3×20 mL). The combined filtrate and washings were concentrated under reduced pressure. The residue was washed through a short column of silica gel using 10% ethyl acetate in hexanes as an eluent. The eluent was concentrated and the resultant product dried under vacuum overnight to provide 15.1 g of methyl2-bromomethyl-3-cyanobenzoate (98% yield). 1H NMR spectrum (in DMSO-d6): δ 8.15 (d, J=8.0 Hz, 1H); 8.12 (d, J=7.9 Hz, 1H); 7.67 (t, 1H); 5.04 (s, 2H); 3.89 (s, 3H).

Methyl5,6-dihydro-5-oxo-11-H-indeno[1,2-c]isoquinoline-10-carboxylate

A mixture of methyl2-bromomethyl-3-cyanobenzoate (14.1 g, 55.5 mmol) and homophtalic anhydride (22.5 g, 138.8 mmol) in acetonitrile (150 mL) was stirred at room temperature. A solution of triethylamine (23.2 mL) in acetonitrile (100 mL) was added dropwise over the period of 1 hour. After the completion of addition, the resultant suspension was refluxed for 4 hours. The reaction mixture was then cooled to room temperature and filtered. The resultant solid was washed thoroughly with acetonitrile (100 mL) and ethanol (2×100 mL) and dried in vacuum oven at 50° C. to provide methyl5,6-dihydro-5-oxo-11-H-indeno[1,2-c]isoquinoline-10-carboxylate was obtained in 81% yield (13.1 g). 1H NMR spectrum (in DMSO-d6): δ 12.21 (s, 1H); 8.20 (m, 2H); 7.83 (d, J=7.5 Hz, 1H); 7.71 (m, 2H); 7.47 (m, 2H); 4.07 (s, 2H); 3.89 (s, 3H).

Compound (I)(25):

Fine powder of methyl5,6-dihydro-5-oxo-11-H-indeno[1,2-c]isoquinoline-10-carboxylate (11.3 g, 28.8 mmol) was suspended in THF (150 mL) and stirred at room temperature. A 2M solution of LAH in THF (39 mL) was added slowly to the stirred suspension. After the complete addition, the mixture was stirred at room temperature for 6 hours, then cooled to 0° C. The reaction mixture was quenched with dropwise addition of ethyl acetate (50 mL). During the quenching the reaction temperature was kept below 50° C. The reaction mixture was poured into 1N HCl solution (200 mL) and stirred for 1 hour. The resultant solid was collected by filtration, washed with 1N HCl solution (20 mL) and ethyl acetate, and dried in a vacuum oven overnight to provide Compound (I)(25) in 93% yield (9.5 g). 1H NMR spectrum (in DMSO-d6): δ 12.28 (s, 1H); 8.23 (d, J=8.1 Hz, 1H); 7.89 (m, 1H); 7.73 (m, 2H); 7.45 (m, 1H); 7.36 (m, 2H); 5.22 (d, J=5.1 Hz, 1H), 4.68 (d, 2H); 3.85 (s, 2H).

Compound (I)(1)

10-Bromomethyl-5,6-dihydro-5-oxo-11-H-indeno[1,2-c]isoquinoline

Fine powder of Compound (I)(25) (10-hydroxymethyl-5,6-dihydro-5-oxo-11-H-indeno[1,2-c]isoquinoline, 7.3 g, 27.7 mmol) was suspended in THF (150 mL). A solution of PBr3 (83 mL, 1M in dichloromethane) was added dropwise at room temperature. After the complete addition, the resultant mixture was stirred overnight and quenched by addition of water (150 mL). The suspension was stirred for 1 hour and filtered. The resultant solid was washed with water (2×100 mL) and ethyl acetate (2×100 mL) and dried in a vacuum oven to provide 10-Bromomethyl-5,6-dihydro-5-oxo-11-H-indeno[1,2-c]isoquinoline in 81% yield. 1H NMR spectrum (in DMSO-d6): δ 12.31 (s, 1H); 8.23 d, J=7.5 Hz, 1H); 7.95 (m, 1H); 7.75 (m, 2H); 7.45 (m, 2H); 4.87 (s, 2H); 3.97 (s, 2H).

Compound (I)(1):

A suspension of 10-bromomethyl-5,6-dihydro-5-oxo-11-H-indeno[1,2-c]isoquinoline (0.425 g, 1.3 mmol) in 33% ethanolic solution of dimethylamine was refluxed overnight. The reaction mixture was stirred at room temperature for 30 minutes and filtered. The resultant solid was washed with ethanol (2×20 mL) and dried in a vacuum oven to provide Compound (I)(1) in 92% yield (0.350 g). 1H NMR spectrum (in DMSO-d6): δ 12.25 (s, 1H); 8.23 (d, J=8.1 Hz, 1H); 7.91 (d, J=8.1 Hz, 1H); 7.73 (m, 2H); 7.44 (m, 1H); 7.36 (t, J=7.5 Hz, 1H); 7.27 (d, J=7.5 Hz, 1H); 3.88 (s, 2H); 3.53 (s, 2H); 2.19 (s, 6H).

Compound (I)(13) was prepared according to the method used to make Compound (I)(1) above, except that morpholine was used in place of dimethylamine, and the reaction solvent was ethanol, DMF, or DMSO.

Compound (I)(69) was prepared according to the method used to make Compound (I)(1) above, except that diethylamine was used in place of dimethylamine, and the reaction solvent was ethanol, DMF, or DMSO.

Compound (I)(74) was prepared according to the method used to make Compound (I)(1) above, except that di-n-propylamine was used in place of dimethylamine, and the reaction solvent was ethanol, DMF, or DMSO.

Compound (I)(79) was prepared according to the method used to make Compound (I)(1) above, except that diethanolamine was used in place of dimethylamine, and the reaction solvent was ethanol, DMF, or DMSO.

Compound (I)(109) was prepared according to the method used to make Compound (I)(1) above, except that 3-hydroxypyrrolidine was used in place of dimethylamine, and the reaction solvent was ethanol, DMF, or DMSO.

Compound (I)(146) was prepared according to the method used to make Compound (I)(1) above, except that 2-N-morpholinoethylamine was used in place of dimethylamine, and the reaction solvent was ethanol, DMF, or DMSO.

Compound (I)(180) was prepared acoording to the method used to make Compound (I)(1) above, except that 4-(4-fluorophenyl)-1,2,3,6-tetrahydropyridine was used in place of dimethylamine, and the reaction solvent was ethanol, DMF, or DMSO.

Methanesulfonate Salt of Compound (I)(1):

Compound (I)(1) (0.490 g, 1.69 mmol) was suspended in ethanol. To the stirred suspension was added methanesulfonic acid (0.164 mL, 2.53 mmol). The mixture became clear and the methanesulfonate salt started to precipitate from the solution after several minutes. The mixture was stirred at room temperature for 2 hours, filtered and washed with ethanol (2×20 mL). The filtered solid was dissolved in water (10 mL) and filtered. The filtrate was lyophilized to provide the methanesulfonate salt of Compound (I)(1) in 90% yield (0.587 g). 1HNMR spectrum (in DMSO-d6): δ 12.37 (s, 1H); 9.59 (s, 1H); 8.25 (d, J=8.1 Hz, 1H); 8.07 (d, J=7.4 Hz, 1H); 7.79 (t, J=7.4 Hz, 1H); 7.70 (d, J=7.5 Hz, 1H); 7.48 (m, 3H); 4.44 (s, 2H); 4.05 (s, 2H); 2.82 (s, 6H); 2.28 (s, 3H).

Using similar methodology, the hydrochloride salt of Compound (I)(1) was prepared.

Compound (IIc)(1)

Methyl4-cyano-3-methylbenzoate

Methyl4-cyano-3-methylbenzoate was obtained in 86% yield according to the procedure used to prepare methyl3-cyano-2-methylbenzoate, except that methyl4-bromo-3-methylbenzoate was used in place of methyl3-bromo-2-methylbenzoate. 1H NMR spectrum (in DMSO-d6): δ 7.96 (s, 1H); 7.85 (m, 2H); 3.85 (s, 3H); 2.46 (s, 3H).

Methyl3-bromomethyl-4-cyanobenzoate

Methyl3-bromomethyl-4-cyanobenzoate was obtained in 60% yield according to the procedure used to prepare methyl2-bromomethyl-3-cyanobenzoate, except that methyl4-cyano-3-methylbenzoate was used in place of methyl3-cyano-2-methylbenzoate. 1H NMR spectrum (in DMSO-d6): δ 8.26 (s, 1H); 8.02 (s, 2H); 4.89 (s, 2H); 3.88 (s, 3H).

Methyl5,6-dihydro-5-oxo-11-H-indeno[1,2-c]isoquinoline-9-carboxylate

Methyl5,6-dihydro-5-oxo-11-H-indeno[1,2-c]isoquinoline-9-carboxylate was obtained in 65% yield according to the procedure used to prepare methyl5,6-dihydro-5-oxo-11-H-indeno[1,2-c]isoquinoline-10-carboxylate, except that methyl3-bromomethyl-4-cyanobenzoate was used in place of methyl2-bromomethyl-3-cyanobenzoate. 1H NMR spectrum (in DMSO-d6): δ 12.35 (s, 1H); 8.25 (d, J=8.1 Hz, 1H); 8.11 (s, 1H); 8.06 (d, J=7.8 Hz, 1H); 7.98 (d, J=8.1 Hz, 1H); 7.75 (m, 2H); 7.49 (m, 1H); 3.95 (s, 2H); 3.85 (s, 3H).

Compound (IIc)(1):

Compound (IIc)(1) was obtained in 96% yield according to the procedure used to prepare Compound (I)(25), except that methyl5,6-dihydro-5-oxo-11-H-indeno[1,2-c]isoquinoline-9-carboxylate was used in place of methyl5,6-dihydro-5-oxo-11-H-indeno[1,2-c]isoquinoline-10-carboxylate. 1H NMR spectrum (in DMSO-d6): δ 12.26 (s, 1H); 8.22 (d, J=8.1 Hz, 1H); 7.91 (s, J=8.1 Hz, 1H); 7.70 (m, 2H); 7.54 (s, 1H); 7.42 (m, 1H); 7.30 (d, J=8.1 Hz, 1H); 5.21 (d, 1H); 4.55 (d, J=5.4 Hz, 2H); 3.89 (s, 2H).

Compound (IIa)(1)

9-Bromomethyl-5,6-dihydro-5-oxo-11-H-indeno[1,2-c]isoquinoline

9-Bromomethyl-5,6-dihydro-5-oxo-11-H-indeno[1,2-c]isoquinoline was obtained in 65% yield according to the procedure used to prepare 10-bromomethyl-5,6-dihydro-5-oxo-11-H-indeno[1,2-c]isoquinoline, except that 9-bromomethyl-5,6-dihydro-5-oxo-11-H-indeno[1,2-c]isoquinoline was used in place of 10-bromomethyl-5,6-dihydro-5-oxo-11-H-indeno[1,2-c]isoquinoline. 1H NMR spectrum (in DMSO-d6): δ 12.24 (s, 1H); 8.23 (d, J=7.5 Hz, 1H); 7.94 (d, J=7.8 Hz, 1H); 7.72 (m, 2H); 7.65 (s, 1H); 7.45 (m, 2H); 4.79 (s, 2H); 3.89 (s, 2H).

Compound (IIa)(1)

Compound (IIa)(1) was obtained in 95% yield according to the procedure for preparing Compound (I)(1), except that 9-bromomethyl-5,6-dihydro-5-oxo-11-H-indeno[1,2-c]isoquinoline was used in place of 10-bromomethyl-5,6-dihydro-5-oxo-11-H-indeno[1,2-c]isoquinoline. 1H NMR spectrum (in DMSO-d6): δ 12.22 (s, 1H); 8.23 (d, J=7.8 Hz, 1H); 7.91 (d, J=7.8 Hz, 1H); 7.71 (m, 2H); 7.51 (s, 1H); 7.43 (m, 1H); 7.27 (d, J=7.8 Hz, 1H); 3.86 (s, 2H); 3.43 (s, 2H); 2.15 (s, 6H).

Compound (IIa)(63) was prepared according to the method used to make Compound (IIa)(1) above, except that di-n-propylamine was used in place of dimethylamine, and the reaction solvent was ethanol, DMF, or DMSO.

Compound (IIa)(68) was prepared according to the method used to make Compound (IIa)(1) above, except that diethanolamine was used in place of dimethylamine, and the reaction solvent was ethanol, DMF, or DMSO.

Compound (IIa)(136) was prepared according to the method used to make Compound (IIa)(1) above, except that 2-N-morpholinoethylamine was used in place of dimethylamine, and the reaction solvent was ethanol, DMF, or DMSO.

Methanesulfonate Salt of Compound (IIa)(1):

The methanesulfonate salt of Compound (IIa)(1) was obtained in 95% yield according to the procedure used to prepare the methanesulfonate salt of Compound (I)(1), except that Compound (IIa)(1) was used in place of Compound (I)(1).

1H NMR data for illustrative Indenoisoquinolinone Analogs are presented below in Table 3.

TABLE 3 (I)(258) δ 1.41 (t, J = 9.9 Hz, 2H), 1.68-1.73 (m, 2H), 2.10 (t, J = 9.3 Hz, 2H), 2.68-2.73 (m, 2H), 3.60 (s, 2H), 3.47 (m, 1H), 3.90 (s, 2H), 4.55 (d, J = 3.6 Hz, 1H), 7.28-7.49 (m, 3H), 7.75-7.77 (m, 2H), 7.91 (d, J = 6.6 Hz, 1H), 8.25 (d, J = 7.8 Hz, 1H), 12.30 (s, 1H) (I)(260) δ 1.41 (t, J = 9.9 Hz, 2H), 1.68-1.73 (m, 2H), 2.10 (t, J = 9.3 Hz, 2H), 2.68-2.73 (m, 2H), 3.60 (s, 2H), 3.47 (m, 1H), 3.90 (s, 2H), 4.55 (d, J = 3.6 Hz, 1H), 7.28-7.49 (m, 3H), 7.75-7.77 (m, 2H), 7.91 (d, J = 6.6 Hz, 1H), 8.25 (d, J = 7.8 Hz, 1H), 12.30 (s, 1H) (I)(263) δ 1.39-1.48 (m, 4H), 1.65-1.74 (m, 4H), 3.04-3.08 (m, 1H), 3.84 (s, 1H), 3.91 (s, 1H), 7.35-7.36 (m, 2H), 7.43-7.48 (m, 1H), 7.75-7.76 (M, 2H), 7.87-7.90 (d, J = 8.1 and 3.9 Hz, 1H), 12.2 (bs, 1H) (I)(265) δ 0.93-0.97 (t, J = 6.6 Hz, 3H), 1.11-1.33 (m, 4H), 1,56 (d, J = 8.1 Hz, 1H), 1.73-1.83 (m, 6H), 2.50-2.52 (m, 2H), 3.77 (s, 1H), 3.90 (s, 1H), 7.32-7.48 (3H), 7.74 (s, 1H), 8.87-8.89 (dd, J = 1.8 and 6 Hz, 1H), 8.25 (d, J = 7.8 Hz, 1H), 12.27 (s, 1H) (I)(271) δ 12.24 (s, 1H); 8.23 (d, J = 8.1 Hz, 1H); 7.84 (d, J = 6.9 Hz, 1H); 7.724 (m, 2H); 7.43 (m, 2H); 7.34 (t, J = 7.5 Hz, 1H); 3.89 (s, 2H); 3.84 (s, 2H); 3.02 (t, J = 6.6 Hz, 1H); 1.72 (m, 4H); 1.51 (d, J = 10.8 Hz, 1H); 1.32 (m, 2H); 1.11 (m, 2H); 1.02 (d, J = 6.6 Hz, 6H) (I)(272) δ 12.25 (s, 1H); 8.23 (d, J = 8.1 Hz, 1H); 7.86 (m, 1H); 7.75 (m, 2H); 7.44 (m, 2H); 7.34 (t, J = 3.0 Hz, 1H); 4.43 (d, J = 3.3 Hz, 1H); 3.88 (s, 4H); 2.39 (m, 1H); 1.91 (m, 2H); 1.77 (m, 2H); 1.10 (m, 4H) (I)(273) δ 12.23 (s, 1H); 8.23 (d, J = 8.1 Hz, 1H); 7.87 (dd, J = 2.7 Hz, 5.7 Hz, 1H); 7.73 (m, 2H); 7.44 (m, 1H); 7.34 (m, 2H); 4.21 (t, J = 5.3 Hz, 1H); 3.90 (s, 2H); 3.80 (s, 2H); 3.33 (m, 2H); 2.54 (t, J = 6.9 Hz, 2H); 2.40 (m, 1H); 1.81 (d, J = 11.4 Hz, 2H); 1.73 (d, J = 13.2 Hz, 2H); 1.54 (d, J = 7.2 Hz, 1H); 1.27 (m, 2H); 1.10 (m, 3H) (I)(276) δ 12.24 (s, 1H); 8.25 (d, J = 7.8 Hz, 1H); 8.01 (d, J = 6.9 Hz, 1H); 7.74 (m, 2H); 7.47 (m, 13H); 4.24 (s, 2H); 4.02 (s, 2H); 3.02 (q, J = 6.9 Hz, 2H); 1.21 (t, J = 6.9 Hz, 2H) (I)(277) δ 12.25 (s, 1H); 8.22 (d, J = 7.8 Hz, 1H); 7.86 (t, J = 4.2 Hz, 1H); 7.73 (m, 2H); 7.44 (m, 1H); 7.33 (d, J = 4.2 Hz, 2H); 3.88 (s, 2H); 3.84 (s, 2H); 2.49 (overlap, 2H); 1.45 (q, J = 7.2 Hz, 2H); 0.86 (t, J = 7.2 Hz, 3H) (I)(278) δ 12.24 (s, 1H); 8.23 (d, J = 7.5 Hz, 1H); 7.86 (m, 1H); 7.76 (m, 2H); 7.45 (m, 1H); 7.35 (m, 2H); 3.90 (s, 2H); 3.86 (s, 2H); 2.73 (m, 1H); 1.03 (d, J = 6.2 Hz, 6H) (I)(298) δ 1.32 (t, J = 6.9 Hz, 3H), 2.33 (s, 3H), 2.77 (d, J = 4.8 Hz, 3H), 3.19-3.26 (m, 2H), 4.09 (s, 2H), 4.37-4.44 (dd, J = 6.9 and 13.5 Hz, 1H), 4.55-4.60 (dd, J = 4.5 and 13.5 Hz, 1H), 7.49-7.55 (m, 3H), 7.74-7.84 (m, 2H), 8.08-8.11 (m, 1H), 8.28 (d, J = 7.8 Hz, 1H), 9.55 (s, 1H), 12.39 (s, 1H) (I)(299) δ 0.93 (t, J = 6.9 Hz, 3H), 1.79 (d, J = 6.6 Hz, 2H), 22.33 (s, 3H), 2.77 (d, J = 4.2 Hz, 3H), 3.14-3.17 (m, 2H), 4.10 (s, 2H), 4.34-4.41 (m, 1H), 4.56-4.61 (m, 1H), 7.49-7.56 (m, 3H), 7.74-7.84 (m, 2H), 8.08 (d, J = 6.6 Hz, 1H), 8.28 (d, J = 8.4 Hz, 1H), 9.77 (s, 1H), 12.39 (s, 1H) (I)(300) δ 12.35 (s, 1H); 8.24 (d, J = 8.1 Hz, 1H); 7.92 (t, J = 4.1 Hz, 1H); 7.75 (m, 2H); 7.46 (m, 1H); 7.38 (m, 2H); 3.97 (s, 2H); 3.93 (s, 2H); 2.66 (t, J = 6.2 Hz, 2H); 1.48 (m, 2H); 1.33 (m, 2H); 0.86 (t, J = 7.0 Hz, 3H) (I)(302) δ 12.27 (s, 1H); 8.25 (d, J = 7.8 Hz, 1H); 7.94 (dd, J = 3.3 Hz, 5.2 Hz, 1H); 7.75 (m, 2H); 7.48 (m, 1H); 7.40 (m, 2H); 4.04 (s, 2H); 3.95 (s, 2H); 3.49 (t, J = 5.3 Hz, 2H); 3.27 (s, 3H); 2.88 (t, J = 5.3 Hz, 2H) (IIa)(93) δ 12.27 (s, 1H); 8.22 (d, J = 7.8 Hz, 1H); 7.90 (d, J = 7.8 Hz, 1H); 7.70 (m, 2H); 7.52 (s, 1H); 7.43 (m, 1H); 7.29 (d, J = 7.8 Hz, 1H); 3.86 (s, 2H); 3.63 (s, 2H); 2.43 (m, 4H); 1.68 (m, 4H) (IIa)(108) δ 12.27 (s, 1H); 8.22 (d, J = 8.1 Hz, 1H); 7.91 (d, J = 7.8 Hz, 1H); 7.71 (m, 2H); 7.50 (s, 1H); 7.43 (m, 1H); 7.28 (d, J = 7.8 Hz, 1H); 3.87 (s, 2H); 3.48 (s, 2H); 2.32 (m, 4H); 1.48 (m, 4H); 1.38 (m, 2H) (IIa)(113) δ 12.29 (s, 1H); 8.21 (d, J = 7.8 Hz, 1H); 7.91 (d, J = 7.8 Hz, 1H); 7.70 (m, 2H); 7.50 (s, 1H); 7.43 (m, 1H); 7.28 (d, J = 7.8 Hz, 1H); 3.86 (s, 2H); 3.49 (s, 2H); 2.24 (m, 8H); 2.12 (s, 3H) (IIa)(131) δ 12.25 (s, 1H); 8.21 (d, J = 7.8 Hz, 1H); 7.89 (d, J = 7.5 Hz, 1H); 7.70 (m, 2H); 7.50 (s, 1H); 7.43 (m, 1H); 7.27 (d, J = 7.5 Hz, 1H); 3.85 (s, 2H); 3.57 (s, 2H); 2.40 (m, 1H); 2.08 (s, 3H); 1.78 (t, J = 12.6 Hz, 4H); 1.52 (1H, d, J = 11.8 Hz, 1H); 1.21 (m, 5H) (IIa)(136) δ 12.23 (s, 1H); 8.22 (d, J = 8.1 Hz, 1H); 7.90 (d, J = 7.8 Hz, 1H); 7.70 (m, 2H); 7.54 (s, 1H); 7.44 (m, 1H); 7.31 (d, J = 7.8 Hz, 1H); 3.86 (s, 2H); 3.76 (s, 2H); 3.53 (t, J = 4.2 Hz, 4H); 2.58 (t, J = 6.3 Hz, 2H); 2.37 (t, J = 6.3 Hz, 2H); 2.30 (t, J = 4.2 Hz, 4H) (IIa)(141) δ 12.23 (s, 1H); 8.21 (d, J = 7.8 Hz, 1H); 7.90 (d, J = 7.8 Hz, 1H); 7.69 (m, 2H); 7.52 (s, 1H); 7.41 (m, 1H); 7.30 (d, J = 7.8 Hz, 1H); 3.83 (s, 2H); 3.71 (s, 2H); 3.50 (m, 4H); 2.27 (m, 6H); 1.55 (t, J = 6.6 Hz, 2H) (IIa)(156) δ 12.23 (s, 1H); 8.21 (d, J = 7.8 Hz, 1H); 7.89 (d, J = 7.8 Hz, 1H); 7.70 (m, 2H); 7.54 (m, 2H); 7.43 (t, J = 6.0 Hz, 1H); 7.30 (d, J = 7.8 Hz, 1H); 7.11 (s, 1H); 6.83 (s, 1H); 4.00 (t, J = 6.6 Hz, 2H); 3.84 (s, 2H); 3.70 (s, 2H); 2,41 (t, J = 6.6 Hz, 2H); 1.82 (t, J = 6.6 Hz, 2H) (IIa)(216) δ 12.23 (s, 1H); 8.22 (d, J = 7.8 Hz, 1H); 7.91 (d, J = 7.8 Hz, 1H); 7.72 (m, 2H); 7.50 (s, 1H); 7.43 (t, J = 5.9 Hz, 1H); 7.28 (d, J = 7.8 Hz, 1H); 3.86 (s, 2H); 3.49 (s, 2H); 2.87 (m, 5H); 2.70 (m, 2H); 1.92 (t, J = 11.4 Hz, 2H); 1.66 (m, 2H); 1.47 (m, 8H) (IIa)(218) δ 12.23 (s, 1H); 8.21 (d, J = 8.1 Hz, 1H); 7.90 (d, J = 8.1 Hz, 1H); 7.70 (m, 2H); 7.50 (s, 1H); 7.43 (t, J = 6.0 Hz, 1H); 7.28 (d, J = 8.1 Hz, 1H); 3.86 (s, 2H); 3.49 (s, 2H); 2.76 (d, J = 10.8 Hz, 2H); 2.49 (m, 4H); 1.96 (t, J = 10.5 Hz, 3H); 1.76 (d, J = 11.1 Hz, 2H); 1.63 (m, 4H); 1.47 (m, 2H) (IIa)(222) δ 12.28 (s, 1H); 8.20 (d, J = 8.1 Hz, 1H); 7.89 (d, J = 7.5 Hz, 1H); 7.71 (m, 2H); 7.52 (s, 1H); 7.42 (m, 1H); 7.30 (d, J = 7.5 Hz, 1H); 3.85 (s, 2H); 3.68 (s, 2H); 3.88 (m, 4H); 3.18 (s, 6H); 2.62 (m, 4H) (IIa)(223) δ 12.27 (s, 1H); 8.21 (d, J = 7.8 Hz, 1H); 7.90 (d, J = 7.5 Hz, 1H); 7.70 (m, 2H); 7.54 (s, 1H); 7.43 (m, 1H); 7.31 (d, J = 7.5 Hz, 1H); 4.45 (s, 1H); 4.12 (d, J = 12.9 Hz, 1H); 3.86 (s, 2H); 3.44 (s, 2H); 3.32 (overlap, 1H); 2.79 (m, 1H); 2.60 (m, 1H); 2.18 (m, 1H) (IIa)(224) δ 12.25 (s, 1H); 8.22 (d, J = 7.8 Hz, 1H); 7.92 (d, J = 7.5 Hz, 1H); 7.72 (m, 2H); 7.57 (s, 1H); 7.44 (m, 1H); 7.34 (d, J = 8.1 Hz, 1H); 3.87 (s, 2H); 3.83 (s, 2H); 2.65 (t, J = 6.3 Hz, 2H); 2.39 (t, J = 6.3 Hz, 2H); 2.33 (m, 4H); 1.45 (m, 4H); 1.35 (m, 2H) (IIa)(234) δ 12.24 (s, 1H); 8.22 (d, J = 8.1 Hz, 1H); 7.91 (d, J = 7.8 Hz, 1H); 7.70 (m, 2H); 7.50 (s, 1H); 7.43 (m, 1H); 7.28 (d, J = 7.5 Hz, 1H); 3.87 (s, 2H); 3.50 (s, 2H); 2.37 (m, 10H); 0.95 (t, J = 6.9 Hz, 3H)

Example 2 Determination of the Effect of Indenoisoquinolinone Analogs on in Vitro PARP Activity

The ability of an illustrative Indenoisoquinolinone Analog to inhibit PARP and prevent peroxynitrite induced cytotoxicity can be demonstrated using methods described in Virag et al., Br. J. Pharmacol. 1999, 126(3):769-77; and Immunology 1998, 94(3):345-55.

The potency of inhibition of purified PARP enzyme can be subsequently determined for selected Indenoisoquinolinone Analogs, and the potency is compared with that of 3-aminobenzamide, a prototypical benchmark PARP inhibitor. The assay is performed in 96 well ELISA plates according to instructions provided with a commercially available PARP inhibition assay kit (for example, from Trevigen, Gaithersburg, Md.).

Example 3 Determination of the Effect of Indenoisoquinolinone Analogs in an in Vitro Model of Cell Death

Using an in vitro, oxidant-stimulated thymocyte assay (described, in detail, in Virag et al., Immunology 94(3):345-55, 1998), an illustrative Indenoisoquinolinone Analog can be tested for its ability to prevent the oxidant-induced suppression of the viability of the cells and as such, this assay represents an in vitro model of reperfusion related cell death in ischemic organs.

Example 4 Determination of the Effect of Indenoisoquinolinone Analogs on in Vivo Models of Inflammatory Diseases

The effect of an illustrative Indenoisoquinolinone Analog can be determined using a systemic inflammatory model induced by bacterial lipopolysaccharide (LPS), which is reported to be responsible for causing reperfusion injurys and inflammatory diseases such as septic shock and systemic inflammatory response syndrome in animals (see Parrillo, N. Engl. J. Med., 328:1471-1478 (1993) and Lamping, J. Clin. Invest. 101:2065-2071 (1998).

Example 5 Determination of the Effect of Indenoisoquinolinone Analogs on in Vivo Models of Reperfusion Injury

The efficacy of an illustrative Indenoisoquinolinone Analog in a mouse model of ischemic and reperfused gut can be determined according to the method described in Liaudet et al., Shock 2000, 14(2):134-41.

In another set of experiments, the effect of an illustrative Indenoisoquinolinone Analog in a rat model of middle cerebral artery occlusion/reperfusion can be assayed as described in Abdelkarim et al., Int. J. Mol. Med. 2001, 7(3):255-60.

Example 6 Determination of the Effect of Indenoisoquinolinone Analogs in an in Vivo Model of Diabetes Mellitus

PARP inhibitors and PARP deficiency are known to reduce the development of diabetes mellitus and the incidence of diabetic complications. The anti-diabetic effect of an illustrative Indenoisoquinolinone Analog can be determined using a single high-dose streptozotocin model of diabetes mellitus, which can be used as conducted as described in Mabley et al., Br. J. Pharmacol. 2001, 133(6):909-9; and Soriano et al., Nat. Med. 2001, 7(1):108-13. Briefly, 160 mg/kg streptozotocin is injected to mice treated with vehicle (control) or with an illustrative Indenoisoquinolinone Analog intraperitoneally (3 mg/kg) and 3 days later blood sugar levels are determined using a blood glucose meter.

Example 7 Determination of the Effect of Indenoisoquinolinone Analogs in an in Vivo Model of Erectile Dysfunction

Experiments are conducted in male Sprague-Dawley rats according to previously published methods for forceps-induced nerve crush injury and erectile function measurements (Rehman, J., et al., Urology 51:640-644, 1998; Sezen, S. F., et al., Int. J. Impot. Res. 14:506-12, 2002). Subjects are anesthetized with Phenobarbital. The prostate of the subjects is exposed and the cavernosal nerve is clipped on either side with a forceps to induce mechanical injury (crush). This rat model mimics the nerve injury that develops during human male prostatectomy, leading to nerve injury and subsequent erectile dysfunction. Subjects are studied 2 weeks after the injury. Two groups of subjects are used, one group treated with vehicle and one group treated with an illustrative Indenoisoquinolinone Analog. The illustrative Indenoisoquinolinone Analog is injected at 30 mg/kg i.v. immediately before the crush injury, and on the following day at the same dose. Thereafter, for 12 days, subjects are treated with 60 mg/kg of theillustrative Indenoisoquinolinone Analog intraperitoneally. At 2 weeks, subjects are re-anesthetized and measured for mean arterial blood pressure (MAP) and intracavernosal pressure (ICP). Cavernosal nerve stimulation is conducted at 5 and 7.5 V using a square pulse stimulator for 30 msec. ICP is determined as the area under curve (mmHg×sec). In addition, IPC/MAP ratios are determined.

Example 8 Effect of Indenoisoquinolinone Analogs on PARP Activity in Cultured Macrophages, Using a Whole-Cell Based Assay

Demonstration of Indenoisoquinolinone Analogs' ability to inhibit PARP and prevent peroxynitrite induced cytotoxicity was shown using methods described in Virag et al., Br. J. Pharmacol. 1999, 126(3):769-77; and Immunology 1998, 94(3):345-55. Human alveolar epithelial cells (A549), an adenocarcinoma cell line with the alveolar type 2 phenotype were cultured in RPMI 1650 medium supplemented with 10% fetal bovine serum. Cells were grown to 80% confluence in 12-well plates and pretreated with various concentrations (10 nM-300 nM) of an Indenoisoquinolinone Analog for 15 minutes. Hydrogen peroxide, a prototypical oxidant that induces DNA single strand breakage, was used to induce PARP activation. Hydrogen peroxide was diluted in culture media and added to the cells in a bolus of 20 μl. After 20 minutes of incubation, the medium was aspirated and 0.5 mL of assay buffer (56 mM HEPES pH 7.5, 28 mM KCl, 28 mM NaCl, 2 mM MgCl2, 0.01% w/v digitonin and 0.5 μCi/ml 3H-NAD+) was added to cells. Following incubation in the assay buffer (15 minutes at 37° C.), PARP activity was measured. Cells were scraped from the plate, transferred to a 1.5 mL eppendorf tube and then added 200 μl of ice cold 50% w/v TCA. After 4 hours incubation at 4° C., samples were then spun (10 minutes at 10,000 g) and pellets washed twice with ice cold 5% w/v TCA and solubilized overnight in 250 μl 2% w/v SDS/0.1 N NaOH at 37° C. The contents of the tubes were added to 6.5 mL ScintiSafe Plus scintillation liquid (Fisher Scientific) and radioactivity was determined using a liquid scintillation counter (Wallac, Gaithersburg, Md.). The results shown in Table 4 demonstrate that the illustrative Indenoisoquinolinone Analogs significantly and dose-dependently inhibit the activation of PARP in the macrophage assay. EC50 values were determined from a dose-response curve. IC50 values were determined using a commercially available in vitro PARP-1 inhibition assay kit (Trevigen, Gaithersburg, Md., USA).

TABLE 4 Compound EC50 value IC50 value (I)(1)  10 nM (I)(13)  85 nM (I)(25) 290 nM (I)(69)  10 nM (I)(74)  15 nM 300 nM (I)(79) 275 nM (I)(109)  20 nM (I)(146)  90 nM (I)(180) 300 nM (IIa)(1)  75 nM (IIa)(63) 300 nM (IIa)(68) 285 nM (IIa)(136)  12 nM (IIc)(1) 100 nM — = not determined

Example 9 Effect of Indenoisoquinolinone Analogs on PARP Activity in Cultured Macrophages, Using a Whole-Cell Based Assay

Demonstration of Indenoisoquinolinone Analogs' ability to inhibit PARP and prevent peroxynitrite induced cytotoxicity was shown using methods described in Virag et al., Br. J. Pharmacol. 1999, 126(3):769-77; and Immunology 1998, 94(3):345-55. The murine RAW macrophages (ATCC, American Type Culture Collection, Manassas, Va.) were grown in RPMI 1650 (Invitrogen Life Technologies; Carlsbad, Calif.) medium supplemented with 10% heat-inactivated FBS, 2 mM L-glutamine, and 100 U/ml of penicillin and streptomycin. Cells, between passages 5 and 15, were seeded at a density of 250,000 cells/well in 12 well plates and allowed to grow 48 hours before use. Growth medium was changed on the day of use. Cells were treated with an Indenoisoquinolinone Analog diluted in a growth medium supplemented with 10% fetal bovine serum (FBS) for 1 hour prior to the addition of hydrogen peroxide (0.5 mM) for a further 25 minutes. For the measurement of PARP activity, media was removed and replaced with 0.5 ml of the assay buffer (56 mM HEPES-pH 7.5, 28 mM KCl, 28 mM NaCl, 2 mM MgCl2, 0.01% digitonin, and 0.5 μCi/ml of 3H-NAD+) for 20 minutes. After aspirating the assay buffer, cells were lysed and transferred to eppendorf tubes containing 250 μl of 50% ice-cold trichloroacetic acid (TCA), which were then placed at 4° C. for 4 hours. Samples were centrifuged at 10,000 g for 10 minutes and supernatant removed. The pellets were washed twice with 500 μl of ice-cold 5% w/v TCA. The pellets were then solubilized in 250 μl of NaOH (0.1 M) containing 2% SDS overnight at 37° C. and the PARP activity was then determined by measuring the radioactivity incorporated using a Wallac scintillation counter. The solubilized protein (250 μl) was mixed with 5 ml of scintillation fluid (ScintiSafe Plus, Fisher Scientific) before being counted for 3 minutes. EC50 values were determined from a dose-response curve.

TABLE 5 Compound EC50 value [nM] (I)(1) 10 (I)(69) <10 (I)(74) 15 (I)(99) 15 (I)(104) 12 (I)(119) <10 (I)(124) 35 (I)(136) 50 (I)(141) 3 (I)(246) 30 (I)(248) 15 (I)(253) 40 (I)(255) 28 (I)(256) 50 (I)(259) 20 (I)(260) <10 (I)(261) 10 (I)(262) <10 (I)(263) <10 (I)(264) <10 (I)(265) <10 (I)(266) <10 (I)(267) 14 (I)(268) 10 (I)(271) 15 (I)(272) 20 (I)(273) <10 (I)(274) 30 (I)(275) 8 (I)(276) 5 (I)(277) <5 (I)(278) <5 (I)(279) 50 (I)(280) 20 (I)(281) 40 (I)(282) 45 (I)(283) 9 (I)(298) 8 (I)(299) 3 (I)(301) 5.5 (I)(302) 8 (IIa)(93) 20 (IIa)(108) 25 (IIa)(121) 30 (IIa)(131) 25 (IIa)(136) 12 (IIa)(141) <10 (IIa)(156) 30 (IIa)(176) <10 (IIa)(216) 30 (IIa)(218) 35 (IIa)(221) 30 (IIa)(222) 20 (IIa)(223) 20 (IIa)(224) 20 (IIa)(225) 40 (IIa)(229) 30 (IIa)(230) 50 (IIa)(231) 45 (IIa)(234) 30 (IIa)(238) 40 (IIa)(240) 25 (IIa)(241) 50 (IIa)(242) 15 (IIa)(243) 15 (IIa)(244) 50 (IIa)(278) 45

The present invention is not to be limited in scope by the specific embodiments disclosed in the examples, which are intended as illustrations of a few aspects of the invention and any embodiments that are functionally equivalent are within the scope of this invention. Indeed, various modifications of the invention in addition to those shown and described herein will become apparant to those skilled in the art and are intended to fall within the scope of the appended claims.

A number of references have been cited, the entire disclosures of which have been incorporated herein in their entirety.

Claims

1. A compound having the formula: or a pharmaceutically acceptable salt thereof wherein

X is —CH2—, —O—, —C(O)—, —NH— or —S—;
R1 is —(CH2)n—N(R2)(R2), —O—(CH2)m—N(R2)(R2), —(CH2)n—OR3 or —O—(CH2)m,—OR3;
each R2 is independently —H, —C1-C6 alkyl, —C3-C8 monocyclic cycloalkyl, —C1-C6 alkylene phenyl, phenyl, an N-terminal alpha amino acid residue, an N-terminal alpha amino hydroxymethyl residue, a C1-C6 alkyl ester of an N-terminal alpha amino acid residue, or a nitrogen-containing 3- to 7-membered monocyclic heterocycle, each of which other than hydrogen is independently unsubstituted or substituted with one or more of -halo, —OH, —OP(O)(OH)2, —OS(O)2OH, —C1-C6 alkyl, —O—C1-C6 alkyl, —C3-C8 monocyclic, a 3- to 7-membered monocyclic heterocycle, —C1-C6 alkyl-substituted 3- to 7-membered monocyclic heterocycle, or —N(Z3)(Z4), where Z3 and Z4 are independently —H, —C1-C5 alkyl, —C1-C5 alkylene-O—C1-C5 alkyl, benzyl, or —C3-C8 monocyclic cycloalkyl, each of which other than hydrogen is independently unsubstituted or substituted with one or more of -halo, —OH, —N(Ra)2, —C1-C5 alkylene-O—C1-C5 alkyl, —NH2, a nitrogen-containing 3-to 7-membered monocyclic heterocycle, or an —C1-C6 alkyl-substituted nitrogen-containing 3- to 7-membered monocyclic heterocycle, wherein each occurrence of Ra is independently —H, benzyl, or —C1-C10 alkyl;
or N, Z3 and Z4 are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle which is unsubstituted or substituted with one or more of —C1-C5 alkyl, phenyl, -phenyl-substituted C1-C5 alkyl, -hydroxy-substituted C1-C5 alkyl, -halo, -halo-substituted C1-C5 alkyl, -halo-substituted phenyl, -phenylene-O—C1-C5 alkyl, -cyanosubstituted phenyl, —OH, —O—C1-C5 alkyl, —N(Ra)2, —C1-C5 alkylene-N(Ra)2, —C1-C5 alkylene-C(O)O—C1-C5 alkylene-N(Ra)2, —COOH, —C1-C3 alkylene-COOH, —OP(O)(OH)2, —OS(O)2OH, —C1-C5 alkylene-OP(O)(OH)2, —C1-C5 alkylene-OS(O)2OH, —C(O)O—C1-C5 alkyl, —OC(O)—C1-C5 alkyl, —C1-C5 alkylene-C(O)O—C1-C5 alkyl, —C1-C5 alkylene-C(O)NH—C1-C5 alkyl, —C(O)NH2, or —CO2, wherein each occurrence of Ra is independently —H, benzyl, or —C1-C10 alkyl;
or N and both Ra groups are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle;
or N and both R2 groups are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle or a nitrogen-containing 7- to 10-membered bicyclic heterocycle, each of which is unsubstituted or substituted with one or more of —C1-C5 alkyl, —C3-C8 monocyclic cycloalkyl, phenyl, a nitrogen-containing 3- to 7-membered monocyclic heterocycle, phenyl-substituted C1-C5 alkyl, -hydroxy-substituted C1-C5 alkyl, -halo, -halo-substituted C1-C5 alkyl, -halo-substituted phenyl, -phenylene-O—C1-C5 alkyl, -cyanosubstituted phenyl, —OH, —O—C1-C5 alkyl, —N(Ra)2, —C1-C5 alkylene-N(Ra)2, —C1-C5 alkylene-C(O)O—C1-C5 alkylene-N(Ra)2, —COOH, —C1-C5 alkylene-O—C1-C5 alkyl, —C1-C5 alkylene-C3-C8 monocyclic cycloalkyl, —C1-C5 alkylene-COOH, —OP(O)(OH)2, —OS(O)2OH, —C1-C5 alkylene-OP(O)(OH)2, —C1-C5 alkylene-OS(O)2OH, —C(O)O—C1-C5 alkyl, —OC(O)—C1-C5 alkyl, —C1-C5 alkylene-C(O)O—C1-C5 alkyl, —C1-C5 alkylene-C(O)NH—C1-C5 alkyl, —C(O)NH2, or —NO2, wherein each occurrence of Ra is independently —H, -benzyl, —C1-C10 alkyl;
or N and both Ra groups are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle;
R3 is —H, —PO3H2, —P2O6H3, —P3O9H4, —SO3H, —C(O)—C1-C9 alkyl, β-D-glucuronyl, or a C-terminal alpha amino acid residue;
n is an integer ranging from 1 to 10; and
m is an integer ranging from 2 to 10.

2-7. (canceled)

8. A compound having the formula: or a pharmaceutically acceptable salt thereof wherein

X is —CH2—, —O—, —C(O)—, —NH— or —S—;
R1 is —(CH2)n—N(R2)(R2), —O—(CF2)m—N(R2)(R2), or —O—(CH2)m—OR3;
each R2 is independently —H, —C1-C6 alkyl, —C3-C8 monocyclic cycloalkyl, —C1-C6 alkylene phenyl, phenyl, an N-terminal alpha amino acid residue, an N-terminal alpha amino hydroxymethyl residue, a C1-C6 alkyl ester of an N-terminal alpha amino acid residue, or a nitrogen-containing 3- to 7-membered monocyclic heterocycle, each of which other than hydrogen is independently unsubstituted or substituted with one or more of -halo, —OH, —OP(O)(OH)2, —OS(O)2OH, —C1-C6 alkyl, —O—C1-C6 alkyl, —C3-C8 monocyclic cycloalkyl, a 3- to 7-membered monocyclic heterocycle, —C1-C6 alkyl-substituted 3- to 7-membered monocyclic heterocycle, or —N(Z3)(Z4), where Z3 and Z4 are independently —C1-C5 alkyl, —C1-C5 alkylene-O—C1-C5 alkyl, benzyl, or —C3-C8 monocyclic cycloalkyl, each of which other than hydrogen is independently unsubstituted or substituted with one or more of -halo, —OH, —N(Ra)2, —C1-C5 alkylene-O—C1-C5 alkyl, —NH2, a nitrogen-containing 3-to 7-membered monocyclic heterocycle, or an —C1-C6 alkyl-substituted nitrogen-containing 3- to 7-membered monocyclic heterocycle, wherein each occurrence of Ra is independently —H, benzyl, or —C1-C10 alkyl;
or N, Z3 and Z4 are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle which is unsubstituted or substituted with one or more of —C1-C5 alkyl, phenyl, -phenyl-substituted C1-C5 alkyl, -hydroxy-substituted C1-C5 alkyl, -halo, -halo-substituted C1-C5 alkyl, -halo-substituted phenyl, -phenylene-O—C1-C5 alkyl, -cyanosubstituted phenyl, —OH, —O—C1-C5 alkyl, —N(Ra)2, —C1-C5 alkylene-N(Ra)2, —C1-C5 alkylene-C(O)O—C1-C5 alkylene-N(Ra)2, —COON, —C1-C5 alkylene-COOH, —OP(O)(OH)2, —OS(O)2OH, —C1-C5 alkylene-OP(O)(OH)2, —C1-C5 alkylene-OS(O)2OH, —C(O)O—C1-C5 alkyl, —OC(O)—C1-C5 alkyl, —C1-C5 alkylene-C(O)O—C1-C5 alkyl, —C1-C5 alkylene-C(O)NH—C1-C5 alkyl, —C(O)NH2, or —NO2, wherein each occurrence of Ra is independently —H, benzyl, or —C1-C10 alkyl;
or N and both Ra groups are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle;
or N and both R2 groups are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle or a nitrogen-containing 7- to 10-membered bicyclic heterocycle, each of which is unsubstituted or substituted with one or more of —C1-C5 alkyl, —C3-C8 monocyclic cycloalkyl, phenyl, a nitrogen-containing 3- to 7-membered monocyclic heterocycle, phenyl-substituted C1-C5 alkyl, -hydroxy-substituted C1-C5 alkyl, -halo, -halo-substituted C1-C5 alkyl, -halo-substituted phenyl, -phenylene-O—C1-C5 alkyl, -cyanosubstituted phenyl, —OH, —O—C1-C5 alkyl, —N(Ra)2, —C1-C5 alkylene-N(Ra)2, —C1-C5 alkylene-C(O)O—C1-C5 alkylene-N(Ra)2, —COOH, —C1-C5 alkylene-O—C1-C5 alkyl, —C1-C5 alkylene-C3-C8 monocyclic cycloalkyl, —C1-C5 alkylene-COOH, —OP(O)(OH)2, —OS(O)2OH, —C1-C5 alkylene-OP(O)(OH)2, —C1-C5 alkylene-OS(O)2OH, —C(O)O—C1-C5 alkyl, —OC(O)—C1-C5 alkyl, —C1-C5 alkylene-C(O)O—C1-C5 alkyl, —C1-C5 alkylene-C(O)NH-C1-C5 alkyl, —C(O)NH2, or —NO2, wherein each occurrence of Ra is independently —H, -benzyl, —C1-C10 alkyl;
or N and both Ra groups are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle;
R3 is —H, —PO3H2, —P2O6H3, —P3O9H4, —SO3H, —C(O)—C1-C9 alkyl, β-D-glucuronyl, or a C-terminal alpha amino acid residue;
n is an integer ranging from 1 to 10; and
m is an integer ranging from 2 to 10.

9-14. (canceled)

15. A compound having the formula: or a pharmaceutically acceptable salt thereof wherein

R1 is —(CH2)n—OR3;
R3 is —H, —PO3H2, —P2O6H3, —P3O9H4, —SO3H, —C(O)—C1-C9 alkyl, β-D-glucuronyl, or a C-terminal alpha amino acid residue; and
n is an integer ranging from 1 to 3.

16-17. (canceled)

18. A compound having the formula: or a pharmaceutically acceptable salt thereof wherein

X is —CH2—, —O—, —C(O)—, —NH— or —S—;
R1 is —(CH2)n—N(R2)(R2), —O—(CH2)m—N(R2)(R2), —(CH2)n—OR3 or —O—(CH2)m—OR3;
each R2 is independently —H, —C1-C6 alkyl, —C3-C8 monocyclic cycloalkyl, —C1-C6 alkylene phenyl, phenyl, an N-terminal alpha amino acid residue, an N-terminal alpha amino hydroxymethyl residue, a C1-C6 alkyl ester of an N-terminal alpha amino acid residue, or a nitrogen-containing 3- to 7-membered monocyclic heterocycle, each of which other than hydrogen is independently unsubstituted or substituted with one or more of -halo, —OH, —OP(O)(OH)2, —OS(O)2OH, —C1-C6 alkyl, —O—C1-C6 alkyl, —C3-C8 monocyclic cycloalkyl, a 3- to 7-membered monocyclic heterocycle, —C1-C6 alkyl-substituted 3- to 7-membered monocyclic heterocycle, or —N(Z3)(Z4), where Z3 and Z4 are independently —H, —C1-C5 alkyl, —C1-C5 alkylene-O—C1-C5 alkyl, benzyl, or —C3-C8 monocyclic cycloalkyl, each of which other than hydrogen is independently unsubstituted or substituted with one or more of -halo, —OH, —N(Ra)2, —C1-C5 alkylene-O—C1-C5 alkyl, —NH2, a nitrogen-containing 3-to 7-membered monocyclic heterocycle, or an —C1-C6 alkyl-substituted nitrogen-containing 3- to 7-membered monocyclic heterocycle, wherein each occurrence of Ra is independently —H, benzyl, or —C1-C10 alkyl; or N, Z3 and Z4 are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle which is unsubstituted or substituted with one or more of-C1-C5 alkyl, phenyl, -phenyl-substituted C1-C5 alkyl, -hydroxy-substituted C1-C5 alkyl, -halo, -halo-substituted C1-C5 alkyl, -halo-substituted phenyl, -phenylene-O—C1-C5 alkyl, -cyano-substituted phenyl, —OH, —O—C1-C5 alkyl, —N(Ra)2, —C1-C5 alkylene-N(Ra)2, —C1-C5 alkylene-N(Ra)2, —COOH, —C1-C5 alkylene-COOH, —OP(O)(OH)2, —OS(O)2OH, —C1-C5 alkylene-OP(O)(OH)2, —C1-C5 alkylene-OS(O)2OH, —C(O)O—C1-C5 alkyl, —OC(O)—C1-C5 alkyl, —C—C5 alkylene-C(O)O—C1-C5 alkyl, —C1-C5 alkylene-C(O)NH—C1-C5 alkyl, —C(O)NH2, or —NO2, wherein each occurrence of Ra is independently —H, benzyl, or —C1-C10 alkyl;
or N and both Ra groups are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle;
or N and both R2 groups are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle or a nitrogen-containing 7- to 10-membered bicyclic heterocycle, each of which is unsubstituted or substituted with one or more of —C1-C5 alkyl, —C3-C8 monocyclic cycloalkyl, phenyl, a nitrogen-containing 3- to 7-membered monocyclic heterocycle, phenyl-substituted C1-C5 alkyl, -hydroxy-substituted C1-C5 alkyl, -halo, -halo-substituted C1-Cs alkyl, -halo-substituted phenyl, -phenylene-O—C1-C5 alkyl, -cyanosubstituted phenyl, —OH, —O—C1-C5 alkyl, —N(Ra)2, —C1-C5 alkylene-N(Ra)2, —C1-C5 alkylene-C(O)O—C1-C5 alkylene-N(Ra)2, —COOH, —C1-C5 alkylene-O—C1-C5 alkyl, —C1-C5 alkylene-C3-C8 monocyclic cycloalkyl, —C1-C5 alkylene-COOH, —OP(O)(OH)2, —OS(O)2OH, —C1-C5 alkylene-OP(O)(OH)2, —C1-C5 alkylene-OS(O)2OH, —C(O)O—C1-C5 alkyl, —OC(O)—C1-C5 alkylene-C(O)O—C1-C5 alkyl, —C1-C5 alkylene-C(O)NH—C1-C5 alkyl, —C(O)NH2, or —NO2, wherein each occurrence of Ra is independently —H, -benzyl, —C1-C10 alkyl;
or N and both Ra groups are taken together to form a nitrogen-containing 3- to 7-membered monocyclic heterocycle;
R3 is —H, —PO3H2, —P2O6H3, —P3O9H4, —SO3H, —C(O)—C1-C9 alkyl, β-D-glucuronyl, or a C-terminal alpha amino acid residue; and
n is an integer ranging from 1 to 10; and
m is an integer ranging from 2 to 10.

19-24. (canceled)

25. A composition comprising a physiologically acceptable carrier or vehicle and an effective amount of a compound or a pharmaceutically acceptable salt of a compound of claim 1, claim 8, claim 15 or claim 18.

26-28. (canceled)

29. A method for treating cancer, renal failure, reperfusion injury, inflammatory disease, diabetes mellitus, an ischemic condition, reoxygenation injury resulting from organ transplantation, neurodegenerative disease, vascular disease, diabetic complication, cardiovascular disease, erectile dysfunction, urinary incontinence, a complication of prematurity, cardiomyopathy, retinopathy, nephropathy, neuropathy, comprising administering an effective amount of a compound or a pharmaceutically acceptable salt of a compound of claim 1, claim 8, claim 15 or claim 18 to a subject in need thereof.

30-108. (canceled)

Patent History
Publication number: 20100179140
Type: Application
Filed: Dec 11, 2009
Publication Date: Jul 15, 2010
Applicant: Inotek Pharmaceuticals Corporation (Beverly, MA)
Inventors: Prakash JAGTAP (N. Andover, MA), Duy-Phong Pham-Huu (Beverly, MA)
Application Number: 12/636,032
Classifications
Current U.S. Class: Polycyclo Ring System Having The Additional Hetero Ring As One Of The Cyclos (514/232.8); Tetracyclo Ring System Having The Six-membered Hetero Ring As One Of The Cyclos (546/61); Tetracyclo Ring System Having The Six-membered Hetero Ring As One Of The Cyclos (514/284); The Additional Six-membered Hetero Ring Is One Of The Cyclos In A Polycyclo Ring System (544/125)
International Classification: A61K 31/5377 (20060101); C07D 217/24 (20060101); A61K 31/473 (20060101); C07D 413/10 (20060101); C07D 401/10 (20060101); C07D 413/12 (20060101); A61P 35/00 (20060101); A61P 29/00 (20060101); A61P 3/10 (20060101); A61P 9/10 (20060101); A61P 13/12 (20060101); A61P 25/28 (20060101); A61P 9/00 (20060101); A61P 15/10 (20060101); A61P 27/02 (20060101);