CA2517358A1 - Isoquinoline derivatives and methods of use thereof - Google Patents

Isoquinoline derivatives and methods of use thereof Download PDF

Info

Publication number
CA2517358A1
CA2517358A1 CA002517358A CA2517358A CA2517358A1 CA 2517358 A1 CA2517358 A1 CA 2517358A1 CA 002517358 A CA002517358 A CA 002517358A CA 2517358 A CA2517358 A CA 2517358A CA 2517358 A1 CA2517358 A1 CA 2517358A1
Authority
CA
Canada
Prior art keywords
alkyl
compound
formula
disease
pharmaceutically acceptable
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA002517358A
Other languages
French (fr)
Inventor
Prakash Jagtap
Erkan Baloglu
John H. Van Duzer
Csaba Szabo
Andrew L. Salzman
Aloka Roy
William Williams
Alexander Nivorozhkin
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Rocket Pharmaceuticals Inc
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Publication of CA2517358A1 publication Critical patent/CA2517358A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D221/00Heterocyclic compounds containing six-membered rings having one nitrogen atom as the only ring hetero atom, not provided for by groups C07D211/00 - C07D219/00
    • C07D221/02Heterocyclic compounds containing six-membered rings having one nitrogen atom as the only ring hetero atom, not provided for by groups C07D211/00 - C07D219/00 condensed with carbocyclic rings or ring systems
    • C07D221/04Ortho- or peri-condensed ring systems
    • C07D221/18Ring systems of four or more rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4738Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4745Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having nitrogen as a ring hetero atom, e.g. phenantrolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/02Stomatological preparations, e.g. drugs for caries, aphtae, periodontitis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems

Abstract

The invention relates to Isoquinoline Derivatives; compositions comprising an effective amount of an Isoquinoline Derivative; and methods for treating or preventing an inflammatory disease or a reperfusion disease comprising the administration of an effective amount of an Isoquinoline Derivative.

Description

ISOOUINOLINE DERIVATIVES AND METHODS OF USE THEREOF
1. FIELD OF THE INVENTION
The invention relates to Isoquinoline Derivatives; compositions comprising an effective amount of an Isoquinoline Derivative; and methods for treating or preventing an inflammatory disease or a reperfusion disease comprising the administration of an effective amount of an Isoquinoline Derivative.
2,. I~ACII~~-I~OUI'~TD OF TIME II'~T C1E~~TTIOI~V
Inflammatory diseases, such as arthritis, colitis, and autoimmune diabetes, typically manifest themselves as disorders distinct from those associated with reperfusion diseases, e.g., stroke and heart attack, and can clinically manifest themselves as different entities. However, there can be common underlying mechanisms between these two types of disorders. In particular, inflammatory disease and reperfusion disease 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-) (Szabo et al., Shock 6:79-88, 1996).
The ONOO~-induced cell necrosis observed in inflammatory disease and in reperfusion disease involves the activation of the nuclear enzyme poly (ADP-ribose) synthetase (PARS). Activation of PARS is thought to be an important step in the cell-mediated death observed in inflammation and reperfusion disease (Szabo et al., Trends Pharmacol. Sci. 19:287-98, 1998).
A number of PARS 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 PARS
inhibitors have been discussed in Milan et al., Science, 223:589-591, 1984.
Isoquinoline 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., W~ 93/05023.
Syntheses of isoquinoline compounds have been reported. Fox example, see Wawzonek et al., ~rg. 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 (WelTlhelnl, (per.), 3 17:381-2, 1984; and Lal et al., I11d1aI1 J.
Chem., Sect. B, 388:33-39, 1999.
There remains, however, a need in the art for compounds useful for treating or preventing inflammatory diseases or reperfusion diseases.
Citation of any reference in Section 2 of this application is not an admission that the reference is prior art.
3. SUMMARY OF THE INVENTION
The invention is based in part on the discovery of novel substituted tetracyclic benzamide derivatives and their demonstrated effects in the treatment or prevention of inflammation, cell death and in treating shock and reperfusion diseases.
Accordingly, in one aspect the invention includes a compound of Formula I, Formula Ia, Formula Ib, Formula II, Formula III, Formula 13, Formula 22, Formula 37 or Formula 40, or a pharmaceutically acceptable salt or hydrate thereof (an "Isoquinoline Derivative") as set forth below in the Detailed Description of the Invention.
Also provided by the invention is a method for treating or preventing an inflammatory disease or a reperfusion disease in a subject, comprising administering to a subject in need of such treatment or prevention an effective amount of an Isoquinoline Derivative.
In a further aspect, the invention also includes methods for making an Isoquinoline Derivative of Formula Ia, Formula Ib, Formula II, Formula III, Formula 13, Formula 22, Formula 37 or Formula 40.
_2_ R~
R
Re (II) (III) (I, Ia and Ib) Rg '1U

Rg Rg Rio ' The Isoquinoline Derivatives can be used to treat or prevent a variety of conditions and diseases, including, but not limited to, an inflammatory disease or a reperfusion disease.
10 The invention also includes pharmaceutical compositions that comprise an effective amount of an Tsoquinoline Derivative and a pharmaceutically acceptable carrier.
The compositions are useful for treating or preventing an inflammatory disease or a reperfusion disease. The invention includes an Isoquinoline Derivative when provided as a pharmaceutically acceptable prodrug, a hydrated salt, such as a pharmaceutically acceptable salt, or mixtures thereof.
The details of the invention are set forth in the accompanying description below.
Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, illustrative methods and materials are now described. Other features, objects, and advantages of the invention will be apparent from the description and from the claims. In the specification and the appended claims, the singular forms also include the plural unless the context clearly dictates otherwise. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary shill in the art to which this invention belongs. All patents and publications cited in this specification are incorporated by reference.
4. DETAILED DESCRIPTION OF THE INVENTION
The present invention provides Isoquinoline Derivatives according to Formula I, Formula Ia, Formula Ib, Foumula II, Formula III, Formula 13, Formula 37 and Formula 40 as set forth below:

(I) and pharmaceutically acceptable salts and hydrates thereof, wherein:
RS is NH or S;
RG is -H or C1-C~ alkyl;
X is -C(~)-, -CH2-, -CH(halo)-, -CH(~H)-(CHI)"-, -CH(~H)-arylene-, -~-, -NH-, -S-, -CH(NRl IR12)- or -N(S~~Y)-, wherein Y is -OH, -NH2 or -allcylheterocycle and n is an integer ranging from 0-5;
~5 Ru and R12 are independently -hydrogen or -C1-C~ alkyl, or N, Rl1 and R12 are taken together to form a heterocyclic amine;
Rl is -hydrogen, -halo, -C1-Clo alkyl, -alkylhalo, -C2-Clo alkenyl, -C3-C$
carbocycle, -aryl, -NH2, -alkylamino, -C(O)OH, -C(O)O(C1-Cs alkyl), NO2 or -A-B;
A is -SOS-, -S~~NH-9 -NHC~-, -NHCONH-, -C~-, -C ~ ~-, -CONH-, -CON(Ci-C4 alkyl)-, -NH-, -CHI-, -S- or -C(S)-;
B is -C1-Clo alkyl, -Ca-Clo alkenyl, -heterocycle, -C3-C8 carbocycle, -aryl, -NZ1Z~, -(C1-Cs alkylene)-NZ1,Z2, -alkylamino, -aminodialkyl, -alkylheterocycle, -arylamido, -C(O)OH, -C(O)O-(C1-C~ alkyl), -C(O)O-phenyl or -C(NH)NH~, each of which is unsubstituted or substituted with one or more of -O-(C1-CS alkyl), -halo, -alkylhalo, -alkanol, -alkylamino, -hydroxy, -NO2, -NHS, -CN, -aminoalkyl, -aminodialkyl, -heterocyclic amine, -C1-Clo alkyl, -C~-C1o alkenyl, -C2-Clo alkynyl, -aryl, -benzyl, -alkylamido, -alkylcarboxy, -C(O)OH, -Cl-CS alkylene-C(O)O-(C1-CS alkyl) or -alkylene-OC(O)-(C1-CS alkyl);
R2, R3, R4, R7, R8, R9 and Rlo are independently -hydrogen, -halo, -hydroxy, -O-(C1-CS alkyl), -C1-Clo alkyl, -alkylhalo, -C2-Clo, alkenyl, -C3-C$
carbocycle, -aryl, -NH2, -alkylamino, -C(O)OH, -C(O)O(C1-CS alkyl), -OC(O)(C~-CS alkyl), NO~ or -A-B;
and at least one of R1, RZ, R3, R~, R7, R8, R9 or Rlo is other than hydrogen;
A is -SOZ-, -SOZNH-, -NHCO-, -NHCONH-, -O-, -CO-, -OC(O)-, -C(O)O-, -CONH-, -CON(C1-C4 alkyl)-, -NH-, -CHI-, -S- or -C(S)-;
B is -C1-Clo alkyl, -C2-Clo alkenyl, -heterocycle, -C3-C$ carbocycle, -aryl, -NZ1Z2, -(C1-CS alkylene)-NZ1Z2, -alkylamino, -aminodialkyl, -alkylheterocycle, -arylamido, C(O)OH, -C(O)O-(C 1-CS alkyl), -C(O)O-phenyl or -C(NH)NH~, each of which is unsubstituted or substituted with one or more of -O-(C1-CS alkyl), -halo, -alkylhalo, -alkanol, -alkylamino, -hydroxy, -N02, -NH2, -CN, -aminoalkyl, -aminodialkyl, -heterocyclic amine, -C1-C1o alkyl, -CZ-Clo alkenyl, -C2-Clo alkynyl, -aryl, -benzyl, -alkylamido, -alkylcarboxy, -C(O)OH, -Cl-CS alkylene-C(O)O-(C1-CS alkyl) or -alkylene-OC(O)-(C1-CS alkyl); and Zl and ZZ are independently -H or -C1-Clo alkyl, which is unsubstituted or substituted with ogle or more of -halo, -OH or -N(Z3)(Z~), where Z3 and Z4 are independently, -H or -C1-CS alkyl, which is unsubstituted or substituted with one or more of -halo, -hydroxy or -NHS; or N, Z~ and Za. aa°e taken together to form a heterocyclic amine; or N, Zl and Z2 are taken together to form a heterocyclic amine.
In one embodiment, X is -C(O)-, -CHZ-, -CH(halo)-, -CH(OH)-(CH2)n , -CH(OH)-arylene-, -O-, -NH-, -S- or -CH(NRllRlz)-, wherein n is an integer ranging from 0-5.
In another embodiment, B is -Cl-Clo alkyl, -Cz-Clo alkenyl, -heterocycle, -C3-carbocycle, -aryl, -N~l~z, -alkylamino, -aminodialkyl, -alkylheterocycle, -arylamido, -C(O)OII, -C(O)O-(C1-C5 alkyl) or -C(O)O-phenyl, each of which is unsubstituted or substituted with one or more of -O-(Cl-C5 alkyl), -halo, -alkylhalo, -alkanol, -alkylamino, -hydroxy, -NOz, -NHz, -aminoalkyl, -aminodialkyl, -heterocyclic amine, -Cl-Cto alkyl, -Cz-Clo alkcnyl, -Cz-Clo alkynyl, -aryl, -benzyl, -alkylamido, -alkylcarboxy, -CI-Cs alkylene-C(O)O-C1-CS alkyl or -C1-C5 alkylene-OC(~)-C,-CS alkyl.
In another embodiment, R1-R4 are hydrogen.
In a further embodiment, at least one of RI, Rz, R3, R~, R7, R8, R~ and Rlo is other than hydrogen.
The invention also relates to a compounds of formula (Ia):
Re (Ia) and pharmaceutically acceptable salts and hydrates thereof, wherein:
RS is NH or S;
R~ is -H or C1-C4 alkyl;
X is -C(O)-, -CHz-, -CH(halo)-, -CH(OH)-(CHz)"-, -CH(OH)-arylene-, -O-, -NH-, -S-, -CH(NRllRiz)- or -N(SOzY)-, wherein Y is -OH, -NHz or -alkylheterocycle and n is an integer ranging from 0-5;
R11 and Rlz are independently -hydrogen or -C1-C~ alkyl, or N, RIl and Rlz are ?5 taken together to form a heterocyclic amine;
Rl is -hydrogen, -halo, -Cl-Clo alkyl, -alkylhalo, -Cz-Clo alkenyl, -C3-C$

carbocycle, -aryl, -NH2, -alkylamino, -C(O)OH, -C(O)O(Cl-CS alkyl), NOZ or -A'-B';
A' is -S02-, -SOZNH-, -NHCO-, -NHCONH-, -CO-, -C(O)O-, -CONH-, -CON(CI-C~ alkyl)-, -NH-, -CH2-, -S- or -C(S)-;
B' is -C1-CIO alkyl, -C2-CIO alkenyl, -heterocycle, -C3-C8 carbocycle, -aryl, -NZ1Z~, -(CI-C~ allcylene)-NZ1Z~,, -alkyla~xiino, -aminodialkyl, -alkylheterocycle, -arylamido, -C(O)OH, -C(O)O-(CI-CS alkyl), -C(O)O-phenyl or -C(NH)NH~, each of which is unsubstituted or substituted with one or more of -O-(CI-CS alkyl), -halo, -alkylhalo, -alkanol, -alkylamino, -hydroxy, -N02, -NHS, -CN, -aminoalkyl, -aminodialkyl, -heterocyclic amine, -CI-CIO alkyl, -C~-CIO alkenyl, -CZ-CIO
alkynyl, -aryl, -benzyl, -alkylamido, -alkylcarboxy, -C(O)OH, -CI-CS alkylene-C(O)O-(CI-CS
alkyl) or -CI-CS alkylene-OC(Oj-(CI-CS alkyl);
R2, R3, Rø, R7, R8, R9 and RIO are independently -hydrogen, -halo, -hydroxy, -O-(C1-CS alkyl), -CI-ClO alkyl, -alkylhalo, -C2-CIO alkenyl, -C3-C$
carbocycle, -aryl, -NHa -alkylamino, -C(O)OH, -C(O)O(C1-CS alkyl), -OC(O)(CI-CS alkyl), N02 or -A-B;
and at least one of Rl, R~, R3, R4, R7, R8, R9 or RIO is other than hydrogen;
A is -SOZ-, -S02NH-, -NHCO-, -NHCONH-, -O-, -CO-, -OC(O)-, -C(O)O-, -CONH-, -CON(CI-C4 alkyl)-, -NH-, -CHI-, -S- or -C(S)-;
B is -CI-CIO alkyl, -CZ-CIO alkenyl, -heterocycle, -C3-C8 carbocycle, -aryl, -NZIZ2, -(CI-CS alkylene)-NZIZ2, -alkylamino, -aminodialkyl, -alkylheterocycle, -arylamido, -C(O)OH, -C(O)O-(CI-CS alkyl), -C(O)O-phenyl or -C(NH)NH2, each of which is unsubstituted or substituted with one or more of -O-(C1-CS alkyl), -halo, -alkylhalo, -alkanol, -alkylamino, -hydroxy, -NOZ, -NHS, -CN, -aminoalkyl, -aminodialkyl, -heterocyclic amine, -CI-ClO alkyl, -C~-CIO alkenyl, -C2-CIO alkynyl, -aryl, -benzyl, -alkylamido, -alkylcarboxy, -C(O)OH, -C1-CS alkylene-C(O)O-(C1-CS alkyl) or -CI-CS
alkylene-OC(O)-(CI-CS alkyl); and Zl, and ZZ are independently -H or -C1-CIO alkyl, which is unsubstituted or substituted with one or more of -halo, -OH or -N(Z3)(Z4), where Z3 and Z~ are independently, -H or -CI-CS alkyl, which is unsubstituted or substituted with one or more of -halo, -hydroxy or -NHS; or N, Z3 and Z4 are taken together to form a heterocyclic amine; or N, ZI and Z2 are taken together to form a heterocyclic amine.
In one embodiment, ~ is -C(O)-, -CHI-, -CH(halo)-, -CH(OH)-(CH~)", -CH(OH)-arylene-, -O-, -NH-, -S- or -CH(NRIIRIa)-, wherein n is an integer ranging from 0-5.
_g_ In another embodiment, B is -C1-C1o alkyl, -C2-C1o alkenyl, -heterocycle, -C3-C$
carbocycle, -aryl, -NZ1Z2, -alkylamino, -aminodialkyl, -alkylheterocycle, -arylamido, -C(O)OH, -C(O)O-(Cl-CS alkyl) or -C(O)O-phenyl, each of which is unsubstituted or substituted with one or more of -O-(C1-CS alkyl), -halo, -alkylhalo, -alkanol, -alkylamino, -hydroxy, -NO2, -1VH~, -aminoalkyl, -aminodialkyl, -haterocyclic amine, -CI-CIO alkyl, -C~-Cio alkenyl, -C~-Cio alkynyl, -aryl, -ben~yl, -alkylamido, -alkylcarboxy, -Cl-Cs alkylene-C(O)~-C1-CS alkyl or -C1-CS alkylene-OC(~)-C1-C$ alkyl.
lia another embodiment, R1-R~ are hydrogen.
In a further embodiment, at least one of R~, R2, R3, R~, R7, R8, R9 and Rlo is other than hydrogen.
Iu one embodiment, h is -S02-.
In other illustrative embodiments RS and X in a compound of formula Ia are as set forth below:

NH -C(O)-NH -CH(halo)-NH -CH(OH)CHZ)n-NH -CH(arylene)(OH)-NH -~-NH -NH-NH -S-NH -CH(NRllRia)-NH -N(SO~Y)-S _C(~)-S -CH~-S -CH(halo)_ S -CH(OH)(CH2)ri RS X

S -CH)(arylene)(OH)-S _~_ S -NH-S _S_ S -CH(NR"R' z)_ S _N(SOZY)_ The invention also relates to compounds of Formula Ib:
R<

R$
(Ib) and pharmaceutically acceptable salts and hydrates thereof, when ein:
RS is O, NH or S;
R6 is -H or C~-C4 alkyl;
X is -C(O)-, -CHI-, -CH(halo)-, -CH(OH)-(CHI)"-, -CH(OH)-arylene-, -O-, -NH-, -S-, -CH(NRllRia)- or -N(S02Y)-, wherein Y is -OH, -NH2 or -alkylheterocycle and n is an integer ranging from 0-5;
Rll and R12 are independently -hydrogen or -C1-C9 alkyl, or N, R11 and R12 are taken together to form a heterocyclic amine;
Rl is -hydrogen, -halo, -C1-Clo alkyl, -alkylhalo, -C~-Clo alkenyl, -C3-C8 carbocycle, -aryl, -NH2, -alkylamino, -C(O)OH, -C(O)O(C1-CS alkyl), NO~ or -A'-B';
A' is -SO2-, -SOZNH-, -NHC~-, -NHCONH-, -C~-, -C(~)~-, -CONH-, -CON(C1-C4 alkyl)-, -NH-, -CH2-, -S- or -C(S)-;
B' is -C1-Clo alkyl, -C2-Clo alkenyl, -heterocycle, -C3-C$ carbocycle, -aryl, -NZ17~, -(Cl-CS alkylene)-NZ1Z~, -alkylamino, -aminodialkyl, -alkylheterocycle, -arylamido, -C(O)OH, -C(O)O-(CI-CS alkyl), -C(O)O-phenyl or -C(NH)NH2, each of which is unsubstituted or substituted with one or more of -O-(CI-CS alkyl), -halo, -alkylhalo, -alkanol, -alkylamino, -hydroxy, -NO2,, -NH?, -CN, -aminoalkyl, -aminodialkyl, -heterocyclic amine, -CI-CIO alkyl, -CZ-CIO alkenyl, -C~-CIO
alkynyl, -aryl, -benzyl, -alkylamido, -alkylcarboxy, -C(OjOH, -CI-CS alkylene-C(O)O-(CI-CS
alkyl) or -CI-CS alkylene-OC(O)-(CI-CS alkyl);
R~, R3, R4, R7, R8, R9 and RIO are independently -hydrogen, -halo, -hydroxy, -O-(CI-CS alkyl), -C1-CIO alkyl, -alkylhalo, -C~-CIO alkenyl, -C3-C8 carbocycle, -aryl, -NHS, -alkylamino, -C(O)OH, -C(O)O(CI-CS alkyl), -OC(O)(CI-CS alkyl), NOZ or -h-B;
and at least one of Rl, R~, R3, R4, R7, R8, R~ or RIO is other than hydrogen;
A is -SO~-, -SOZNH-, -NHCO-, -NHCONH-, -O-, -CO-, -OC(O)-, -C(O)O-, -CONH-, -CON(CI-C~ alkyl)-, -NH-, -CHZ, -S- or -C(S)-;
B is -CI-CIO alkyl, -CZ-ClO alkenyl, -heterocycle, -C3-C8 carbocycle, -aryl, -NZIZ2, -(CI-CS alkylene)-NZIZ2, -alkylamino, -aminodialkyl, -alkylheterocycle, -arylamido, C(O)OH, -C(O)O-(CI-CS alkyl), -C(O)O-phenyl or -C(NH)NH2, each of which is unsubstituted or substituted with one or more of -O-(C1-CS alkyl), -halo, -alkylhalo, -alkanol, -alkylamino, -hydroxy, -N02, -NH2, -CN, -aminoalkyl, -aminodialkyl, -heterocyclic amine, -CI-CIO alkyl, -C2-ClO alkenyl, -CZ-CIO alkynyl, -aryl, -benzyl, -alkylamido, -alkylcarboxy, -C(O)OH, -CI-CS allcylene-C(O)O-(C1-CS alkyl) or -alkylene-OC(O)-(CI-CS alkyl); and ZI and Z2 are independently -H or -CI-CIO alkyl, which is unsubstituted or substituted with one or more of -halo, -OH or -N(Z3)(Z4), where Z3 and Z4 are independently, -H or -C1-CS alkyl, which is unsubstituted or substituted with one or more of -halo, -hydroxy or -NH2; or N, Z3 and Z4 are taken together to form a heterocyclic amine; or N, Zl and ZZ are taken together to form a heterocyclic amine.
In one embodiment, X is -C(O)-, -CHa-, -CH(halo)-, -CH(OH)-(CHZ)"-, -CH(OH)-arylene-, -O-, -NH-, -S- or -CH(NRIIRIa)-, wherein n is an integer ranging from 0-5.
In another embodiment, ~ is -N(SO2Y)-.
W another embodiment, B is -CI-CIO alkyl, -C~-CIO alkenyl, -heterocycle, -C3-C8 carbocycle, -aryl, -NZIZZ, -alkylamino, -aminodialkyl, -alkylheterocycle, -arylamido, -C(O)OH, -C(O)O-(C1-CS alkyl) or -C(O)O-phenyl, each of which is unsubstituted or substituted with one or more of -O-(C1-CS alkyl), -halo, -alkylhalo, -alkanol, -alkylamino, -hydroxy, -N02, -NH2, -aminoalkyl, -aminodialkyl, -heterocyclic amine, -Cl-Clo alkyl, -C2-Clo alkenyl, -C2-Clo alkynyl, -aryl, -benzyl, -alkylamido, -alkylcarboxy, -C1-CS alkylene-C(O)O-CI-C~ alkyl or -Cl-CS alkylene-OC(O)-Cl-Cs alkyl.
In another embodiment, R1-R~ are hydrogen.
In a further embodiment, at least one of RI, R2, R3, R~, R7, Rg, R9 and Rio is other than hydrogen.
In one embodiment, A is -SO~- or -SO~NH~-.
In another embodiment, RS is NH.
In another embodiment, RS is S.
In yet another embodiment, RS is O.
In illustrative embodiments RS and X in a compound of formula Ib are as set forth below:

O -CH(halo)-O -CH(OH)(CH~)ri O -CH(arylene)(OH)-O -O-O -NH-O -S-O -CH(NR"Ri 2)-O -N(S02Y)_ Illustrative Compounds of Formula Ib are set forth below:

C0ll1p011nd~7 I~g I~9 X10 22a -H -H -H -H

22b -H -OMe -H -H

22c -H -H -OMe -H

22d -H -H -H -OMe 22e -H -Me -H -H

22f -H -COOH -H -H

22g -H -H -COOH -H

23a -H -OH -H -H

23b -H -H -OH -H

23c -H -H -H -OH

25a -H -H -(CH2)40H -H

25b -H -H -(CHZ)SOH -H

25c -H -H -(CH2)60H -H

25d -H -H -(CH2)4COOH -H

25e -H -H -(CH~)SCOOH -H

26a -H -C(O)NH(CH~)3-IV-moipholine-H -H

26b -H -C(O)NH(CHZ)?-COON -H -H

26c -H -C(O)NH(CH2)3-N (1,3- -H -H

imidazole) 26d -H -C(O)NH(CH2)2-NMe2 -H -H

and pharmaceutically acceptable salts and hydrates thereof.
Additional Illustrative Compounds of Formula I~ are set forth below:
Compound X R9 34 -N(S03H)- -S03H

35a -N(SO~NH2)- -S02NH~

35b -N[S02NH(CHZ)3 -S02NH(CH~)3 (N-morpholine)]- (N moipholine) ' 40a -S- -H

and pharmaceutically acceptable salts and hydrates thereof.
The invention also relates to compounds of Formula II:

(II) and pharmaceutically acceptable salts and hydrates thereof, wherein:
R6 is -H or C1-C4 alkyl;
RI is -hydrogen, -halo, -CI-Clo alkylp -alkylhalo, -C~-Clo alkenylp -C~-C8 carbocycle, -aryl, -NHS, -alkylamino, -C(O)OHp -C(O)O(C1-CS alkyl), NO? or -A'-B';
A' is -SO~-, -SO2NH-, -NHCO-, -NHCONH-, -CO-, -C(O)O-, -CONH-, -CON(C1-C4 alkylj-, -NH-, -CH2-, -S- or -C(S)-;
B' is -C~-Clo alkyl, -C~-C1o alkenyl, -heterocycle, -C3-C8 carbocycle, -aryl, -NHS, alkylamino, -aminodialkyl, -alkylheterocycle, -arylamido, -C(O)OH, -C(O)O-(Cl-CS
alkyl), -C(O)O-phenyl or -NZ1Z~;
R2, R3, R~, R7, R8, Rg and Rlo are independently -hydrogen, -halo, -hydroxy, -O-(Cl-CS alkyl), -C1-Clo alkyl, -alkylhalo, -CZ-Clo alkenyl, -C3-C8 carbocycle, -aryl, -NH2, -alkylamino, -C(O)OH, -C(O)O(C1-CS alkyl), -OC(O)(C1-CS alkyl), NOz or -A-B;
wherein at least one of R1, R4 and Rl° is other than hydrogen;
A is -SOZ-, -S02NH-, -NHCO-, -NHCONH-, -O-, -CO-, -OC(O)-, -C(O)O-, -CONH-, -CON(Ci-C4 alkyl)-, -NH-, -CHZ-, -S- or -C(S)-;
B is -C1-Clo alkyl, -C2-Clo alkenyl, -heterocycle, -C3-C8 carbocycle, -aryl, -NH2, -alkylamino, -aminodialkyl, -alkylheterocycle, -arylamido, -C(O)OH, -C(O)O-(C1-CS
alkyl), -C(O)O-phenyl or -NZ1Z2; and Zl and Z2 are independently -H or -C1-Clo alkyl, which is unsubstituted or substituted with one or more of -halo, -OH or -N(Z3)(Z4), where Z3 and Z4 are independently, -H or -C1-C5 alkyl, which is unsubstituted or substituted with one or more of -halo, -hydroxy or -NH2; or N, Z3 and Z4 are taken together to form a heterocyclic amine; or N, Zl and Z2 are taken together to form a heterocyclic amine.
In one embodiment, B is a heterocyclic amine.
In another embodiment, B is arylalkyl.
In still another embodiment, Rl is -hydrogen, -halo, -C1-Cio alkyl, -allylhalo, ~0 -C2-Clo alkenyl, -C3-C~ carbocycle, -aryl, -NHS, -alkylamino, -C(O)OH, -C(O)O(C1-CS alkyl), NO~ or -A-B;
IS - 2,-p - 7NH-p -NHC~-p -NH -p - -, - -9 - ONH-9 -CON(C1-C4 alkyl)-, -NH-, -CHI, -S- or -C(S)-;

B is -C1-Clo alkyl, -CZ-Clo alkenyl, -heterocycle, -C3-C8 carbocycle, -aryl, -NH2, -alkylamino, -aminodialkyl, -alkyiheterocycle, -arylamido, -C(O)OH, -C(O)O-(C1-CS alkyl), -C(O)O-phenyl or -NZ1Z2.
In a further embodiment, at least one of Rl, R~, R3, R4, R7, R8, Rg and Rio is not hydrogen.
The invention also relates to compounds of Formula III:
Rg (III) and pharmaceutically acceptable salts and hydrates thereof, wherein:
X is -CHI- or -O-;
R2 and R3 are independently -hydrogen, -halo, -alkylhalo, -hydroxy, -O-(C1-C5 alkyl), -C1-C3 alkyl, -N02, -NH2, -CONH2, -C(O)OH, -OC(O)-C1-CS alkyl or -C(O)O-C1-CS alkyl;
R8 and R9 are independently -hydrogen or -A-B;
A is -SOZ-, -S02NH- or -NHCO-; and B is -C1-C3 alkyl, -NZ1Z2, -heterocycle or -alkylamino, each unsubstituted or substituted with one or more of -alkanol, -alkylamino, -aminoallcyl, -aminodialkyl or -heterocycle, each unsubstituted or substituted with -C1-Clo alkyl or -alkanol; and Zl and Z2 are independently -hydrogen or -C1-C$ alkyl, which is unsubstituted or substituted with one or more of -hydroxy or -NZ3Z4, where Z3 and Z4 are independently -H or -Cl-C~ alkyl, which is unsubstituted or substituted with one or more of -hydroxy or -NHS, or N, Z3 arid Z4 are taken together to a heterocyclic amine, or N, Zl and Z~ are 2~ taken together to form a heterocyclic amine.
In one embodiment, -X- is -CHZ-.

In another embodiment, -X- is -O-.
In one embodiment, Rg is hydrogen and R9 is -A-B.
In another embodiment, R$ is -A-B and R9 is hydrogen.
In one embodiment, either R8 is hydrogen and R~ is -A-B, or R8 is -A-B and R~
is hydrogen.
In one embodiment, R3, Rs and R9 are hydrogen and R' is -A-B, wherein A is -NHC(O)-.
In another embodiment, R2, R8 and R~ are hydrogen and R3 is -A-B, wherein A is -NHC(O)-.
In still another embodiment, R2, R3 and R8 are hydrogen and R9 is -A-B, wherein A is -SO?- or -S02NH-.
In a further embodiment, at least one of R~, R3, R8 and R9 is not hydrogen.
The invention further relates to compounds of Formula 13:
RB

and pharmaceutically acceptable salts and hydrates thereof, wherein:
Rl, R2, R3, Rø, R7, R8, R9 and Rlo are independently -hydrogen, -halo, -hydroxy, -O-(C1-CS alkyl), -C1-Clo alkyl, -allcylhalo, -C2-Clo alkenyl, -C3-C8 carbocycle, -aryl, -NHS, -alkylamino, -C(O)OH, -C(O)O(CI-CS alkyl), -OC(O)(CI-CS allcyl), NOZ or -A-B;
AisSO~-,- ~ -,- -a- -a- -,- -~- -,--CONH-, -CON(CI-C~. alkyl)-, -NH-, -CHI-, -S- or -C(S)-;
?5 B is -C1-CIO alkyl, -C~-CIO alkenyl, -heterocycle, -C~-Cs carbocycle, -aryl, -NZ1Z~, -(C1-CS allcylene)-NZ1Z2, -alkylamino, -aminodialkyl, -alkylheterocycle, -arylamido, -C(O)OH, -C(O)O-(C1-C5 alkyl), -C(O)O-phenyl or -C(NH)NH2, each of which is unsubstituted or substituted with one or more of -O-(C1-CS alkyl), -halo, -alkylhalo, -alkanol, -alkylamino, -hydroxy, -NO2, -NH2, -CN, -aminoalkyl, -aminodialkyl, -heterocyclic amine, -C1-Clo alkyl, -C2-Clo alkenyl, -C?-Clo alkynyl, -aryl, -benzyl, -alkylamido, -alkylcarboxy, -C(O)OH, -C1-CS alkylene-C(O)O-(C1-C~ alkyl) or -C~-Cs alkylene-OC(O)-(C1-CS alkyl); and Zl and ~, are independently -H or -C1-Clo alkyl, which is unsubstituted or substituted with one or more of -halo, -OH or -N(Z3)(~4), where ~3 and ~~ are independently, -H or -C1-CS alkyl, which is unsubstituted or substituted with one or more of -halo, -hydroxy or -NHS; or N, ~3 and Zø are taken together to form a heterocyclic amine; or N, Zl and ~~ are taken together to form a heterocyclic amine.
In one embodiment, R9 is -A-B, wherein -A- is -S02- or -S02NH-.
In mother embodiment, Rl-R~ are each hydrogen. ..-In another embodiment, Rl-R4 are each hydrogen.
In a further embodiment, at least one of Rl, R2, R3, R~, R7, R8, R9 and Rlo is other than hydrogen.
The invention further still relates to compounds of Formula 22:
Rg and phaumaceutically acceptable salts and hydrates thereof, wherein:
Rl-R4 and R7-Rlo are as defined above for Formula g3.
In one embodiment, R9 is -A-B, wherein -A- is -SO?- or -SO~NH-.
In another embodiment, Rl-R4 are each hydrogen.

In a further embodiment, at least one of Rl, R2, R3, R4, R7, R8, R9 and Rlo is other than hydrogen.
The invention further still relates to compounds of Formula 37:
R
R

and pharmaceutically acceptable salts and hydrates thereof, v~ r~erein:
Rl-R4 and R7-Rlo are as defined above for Formula 13.
In one embodiment, Rl-R4 are each hydrogen.
In a further embodiment, at least one of R1, R2, R3, R~., R7, Rg, R9 and Rlo is other than hydrogen.
The invention also relates to compounds of Formula 40:

and pharmaceutically acceptable salts and hydrates thereof, wherein:
RL-R4 and R7-Rio are as defined above for Formula 13.
In one embodiment, RI-R4 are each hydrogen.
In a further embodiment, at least one of R1, RZ, R3, R4, R7, R8, R9 and Rln is other than hydrogen.
4.1 DEFINITIONS
The following definitions are used in connection with the Isoquinoline Derivatives:
"C1-C~ alkyl" refers to a straight or branched chain saturated hydrocarbon containing 1-3 carbon atoms. Examples of a C1-C3 alkyl group include, but are not limited to, methyl, ethyl, propyl and isopropyl.
"C1-C~. alkyl" refers to a straight or branched chain saturated hydrocarbon containing 1-4 carbon atoms. Examples of a C1-C4 alkyl group include, but are not limited to, methyl, ethyl, propyl, butyl, isopropyl, isobutyl, sec-butyl and ter-t-butyl.
"C1-CS alkyl" refers to a straight or branched chain saturated hydrocarbon containing 1-4 carbon atoms. Examples of a C1-CS alkyl group include, but are not limited to, methyl, ethyl, propyl, butyl, pentyl, isopropyl, isobutyl, sec-butyl and tart-butyl, isopentyl and neopentyl.
"C1-C8 alkyl" refers to a straight or branched chain saturated hydrocarbon containing 1-8 carbon atoms. Examples of a C1-C8 alkyl group include, but are not limited to, methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, isopropyl, isobutyl, see-butyl and tey~t-butyl, isopentyl, neopentyl, isohexyl, isoheptyl and isooctyl.
"CI-C~ alkyl" refers to a straight or branched chain saturated hydrocarbon containing 1-9 carbon atoms. Examples of a CI-C9 alkyl group include, but are not limited to, methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, nonyl, isopropyl, isobutyl, sec-butyl and tar°t-butyl, isopentyl, neopentyl, isohexyl, isoheptyl, isooctyl and isononyl.
"Cl-Clo alkyl" refers to a straight or branched chain saturated hydrocarbon containing 1-10 carbon atoms. Examples of a CI-Clo alkyl group include, but are not limited to, methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl, isopropyl, isobutyl, sec-butyl and tart-butyl, isopentyl, neopentyl, isohexyl, isoheptyl, isooctyl, isononyl and isodecyl.
'6C~-Cio alkenyl" refers to a straight or branched chain unsaturated hydrocarbon containing 2-10 carbon atoms and at least one double bond. Examples of a C~-Clo alkenyl group~include, but are not limited to, ethylene, propylene, 1-butylene, 2-butylene, isobutylene, see-butylene, 1-pentane, 2-pentane, isopentene, 1-hexane, 2-hexane, 3-hexene, isohexene, 1-heptene, 2-heptene, 3-heptene, 1-octane, 2-octane, 3-octane, 4-octene, 1-nonene, 2-nonene, 3-nonene, 4-nonene, 1-decene, 2-decene, 3-decene, 4-decene and 5-decene.
"C2-Clo alkynyl" refers to a straight or branched chain unsaturated hydrocarbon containing 2-10 carbon atoms and at least one triple bond. Examples of a CZ-Clo alkynyl group include, but are not limited to, acetylene, propyne, 1-butyne, 2-butyne, isobutyne, sec-butyne, 1-pentyne, 2-pentyne, isopentyne, 1-hexyne, 2-hexyne, 3-hexyne, isohexyne, 1-heptyne, 2-heptyne, 3-heptyne, 1-octyne, 2-octyne, 3-octyne, 4-octyne, 1-nonyne, 2-nonyne, 3-nonyne, 4-nonyne, 1-decyne, 2-decyne, 3-decyne, 4-decyne and 5-decyne.
"C1-C4 alkylene" refers to a C1-C4 alkyl group in which one of the C1-C4 alkyl group's hydrogen atoms has been replaced with a bond. Examples of a Cl-C4 alkylene include -CHI-, -CH~CH2-, -CH2CH~CH2- and -CHzCH2CH2CHz-.
"Ci-CS alkylene" refers to a CI-CS alkyl group in which one of the C1-CS alkyl group's hydrogen atoms has been replaced with a bond. Examples of a C1-C4 alkylene include -CHZ-, -CHI CHI-, -CH2CH~CH2- and examples of a C1-C4 alkylene include -CH2-, -CH2CH2-, -CH2CH2CH2-, -CH2CH2CH2CH2- and -CH2CH~CH2CH2CH2-.
"Alkylhalo" refers to a C1-CS alkyl group, as defined above, wherein one or more of the C1-CS alkyl group's hydrogen atoms has been replaced with -F, -Cl, -Br or -I.
Representative examples of an alkylhalo group include, belt are not limited to, -CHEF, -CC13, -CF3, -CH2Cl, -CH?CH~Br, -CH~CH2I, -CH~CHZCH~F, -CH?CH2CH2Cl, - 2 ? 2 2 1, - Z ~? 2 ? , - 2 ~ 3 2CH?Bl, -CH2CH2CHZCH2CHZI, -CH~CH(Br)CH3, -CH2CH(Cl)CH~CH3, -CH(F)CH2CH3 and -C(CH3)2(CHZCl).

"Alkylamino" refers to a C1-C4 alkyl group, as defined above, wherein one or more of the C1-C4 alkyl group's hydrogen atoms has been replaced with -NH2.
Representative examples of an alkyl amino group include, but are not limited to, -CH~NH~, -CH?CH~NH2, -CH2CHZCH?NH2, -CHZCH2CH?CH2NH2, -CH2CH(NH2)CH3, -CH~CH(NH~)CH~CH3, -CH(NH~)CH~CH3 and -C(CH~)~(CH~NHZ).
''Anunoalkyl" refers to an -NH group, the nitrogen atom of said group being attached to a C1-C4 alkyl group, as defined above. Representative examples of an aminoalkyl group include, but are not limited to, -NHCH3, -NHCH~CH3, -NHCHZCHZCH3, -NHCH2CH~CH2CH3, -NHCH(CH3)2, -NHCH~CH(CH3)~, -NHCH(CH3)CH~CH3 and -NH-C(CH3)3.
"Aminodialkyl" refers to a nitrogen atom which has attached to it two C1-C4 alkyl groups, as defined above. Representative examples of a aminodialkyl group include, but are not limited to, -N(CH3)2, -N(CHZCH3)(CH3), -N(CH2CH3)2, -N(CH2CHZCH3)2, -N(CH~CH~CH~CH;)?, -N(CH(CH3)~)~, -l~(CH(CH3)~)(CH3), -N(CH~CH(CH,)~)~, -NH(CH(CH3)CH~CH3)2, -N(C(CH3)s)a and -N(C(CH3)3)(CH3).
"Aryl" refers to a phenyl or pyridyl group. Examples of an aryl group include, but are not limited to, phenyl, N-pyridyl, 2-pyridyl, 3-pyridyl and 4-pyridyl. An aryl group can be unsubstituted or substituted with one or more of the following groups: -Ct-CS
alkyl, halo, -alkylhalo, hydroxy, -O-C1-CS alkyl, -NH2, -aminoalkyl, -aminodialkyl, -COOH, -C(O)O-(C1-CS alkyl), -OC(O)-(C1-CS alkyl), -N-amidoalkyl, -C(O)NH~, -carboxamidoalkyl, or -N02.
"Arylalkyl" refers to an aryl group, as defined above, wherein one of the aryl group's hydrogen atoms has been replaced with a C1-CS alkyl group, as defined above.
Representative examples of an arylalkyl group include, but are not limited to, 2-methylphenyl, 3-methylphenyl, 4-methylphenyl, 2-ethylphenyl, 3-ethylphenyl, 4-ethylphenyl, 2-propylphenyl, 3-propylphenyl, 4-propylphenyl, 2-butylphenyl, 3-butylphenyl, 4-butylphenyl, 2-pentylphenyl, 3-pentylphenyl, 4-pentylphenyl, 2-isopropylphenyl, 3-isopropylphenyl, 4-isopropylphenyl, 2-isobutylphenyl, 3-isobutylphenyl, 4-isobutylphenyl, 2-sec-butylphenyl, 3-sec-butylphenyl, 4-sec-butylphenyl, 2-t-butylphenyl, 3-t-butylphenyl and 4-t-butylphenyl.
"Arylamido" refers to an aryl group, as defined above, wherein one of the aryl group's hydrogen atoms has been replaced with one or more -C(O)NH~ groups.
Representative examples of an arylamido group include 2-(H2N(O)C)-phenyl, 3-(HZN(O)C)-phenyl, 4-(H2N(O)C)-phenyl, 2-(H2N(O)C)-pyridyl, 3-(H2N(O)C)-pyridyl and 4-(H2N(O)C)-pyridyl.
"Alkylheterocycle" refers to a Cl-CS alkyl group, as defined above, wherein one of the Cl-CS alkyl group's hydrogen atoms has been replaced with a heterocycle.
Representative examples of an alkylheterocycle group include, but are not limited to, -CH~CH2-morpholine, -CH~CH?-piperidine, -CH~CH~CH~-morpholine and -CH2CH?CHZ-imida~ole.
"Alkylamido" refers to a CI-CS alkyl group, as defined above, wherein one of the C1-CS alkyl group's hydrogen atoms has been replaced with a -C(O)NH~ group.
Representative examples of an alkylamido group include, but are not limited to, -CH~C(O)NH~, -CH~CH~C(O)NH~, -CH2CH2CH~C(O)NH~, -CHZCH~CH~CHZC(O)NH~, -CH2CH~CH~CH2CH~C(O)NHZ, -CH2CH(C(O)NHZ)CH3, -CH2CH(C(O)NH~)CH~CH3, -CH(C(O)NH~)CH2CH3 and -C(CH3)ZCH2C(O)NHZ.
"Alkanol" refers to a C1-CS alkyl group, as defined above, wherein one of the C1-Cs alkyl group's hydrogen atoms has been replaced with a hydroxyl group.
Representative examples of an alkanol group include, but are not limited to, -CH~OH, -CH2CH20H, -CHZCH2CH20H, -CH~CH2CHZCHzOH, -CH2CHZCH2CHZCHZOH, -CH~CH(OH)CH3, -CH~CH(OH)CH~CH~, -CH(OH)CH~CH3 and -C(CH3)2CH~OH.
"Alkylcarboxy" refers to a C1-CS alkyl group, as defined above, wherein one of the C1-CS alkyl group's hydrogen atoms has been replaced with a -COOH group.
Representative examples of an alkylcarboxy group include, but are not limited to, -CHZCOOH, -CH2CH2COOH, -CH~CH2CHZCOOH, -CHZCH2CHZCH2COOH, -CH~CH(COOH)CH3, -CH~CH2CH~CH2CHZCOOH, -CH2CH(COOH)CH2CH3, -CH(COOH)CH?CH3 and -C(CH3)2CH2COOH.
"N-amidoallcyl" refers to a -NHC(O)- group in which the carbonyl carbon atom of said group is attached to a Cr-CS alkyl group, as defined above.
Representative examples of a N-amidoalkyl group include, but are not limited to, -NHC(O)CH3, -NHC(O)CH~CH3, -NHC(O)CH2CH2CH3, -NHC(O)CH2CH2CH~CH3, -NHC(O)CH2CHZCH2CH2CH3, -NHC(O)CH(CH3)Z, -NHC(O)CHZCH(CH3)Z, -NHC(O)CH(CH3)CH~CH3, -NHC(O)C(CH3)3 arid -NHC(O)CH2C(CH3)3.
"Carboxamidoalkyl" refers to a -C(O)NH- group in which the nitrogen atom of said group is attached to a CI-CS alkyl group, as defined above.
Representative examples of a carboxamidoalkyl group include, but are not limited to, -C(O)NHCH3, -C(O)NHCH2CH3, -C(O)NHCH2CHaCH3, -C(O)NHCH2CH2CHZCH3 C(O)NHCH2CH2CH2CH2CH3, -C(O)NHCH(CH3)Z, -C(O)NHCH2CH(CH3)2, -C(O)NHCH(CH3)CHZCH3, -C(O)NHC(CH3)3 and -C(O)NHCHZC(CH3)3.
An "Arylene" group is a phenyl group in which one of the phenyl group's hydrogen atoms has been replaced with a bond. An arylene group can bein an ortho, meta, or para configuration and can be unsubstituted or independently substituted with one or more of the following groups: -Cl-Cs alkyl, halo, -alkyihalo, hydroxy, -O-C1-Cs alkyl, -NH2, -aminoalkyl, -aminodialkyl, -COO1=I, -C(O)O-(C1-Cs alkyl), -OC(O)-(C1-Cs alkyl), -N-amidoalkyl, -C(O)NH2, -carboxamidoalkyl or -NO?.
A "C3-Cg carbocycle" is a non-aromatic, Saturated, monocyclic hydrocarbon ring containing 3-& carbon atoms. Representative examples of a C3-C8 carbocycle include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl. A C3-C$ carbocycle can be unsubstituted or independently substituted with one or more of the following groups: -CI-Cs alkyl, halo, -alkylhalo, hydroxy, -O-C1-Cs alkyl, -NH2, -aminoalkyl, -aminodialkyl, -COOH, -C(O)O-(C1-Cs alkyl), -OC(O)-(C1-Cs alkyl), -N-amsdoalkyl, -C(O)NH~, -carboxyamsdoalkyl or -NO2. ,.
"Heterocycle" refers to a 5- to 10-membered aromatic or non-aromatic carbocycle in which 1-4 of the ring carbon atoms have been independently replaced with a N, O or S
atom. Representative examples of a heterocycle group include, but are not limited to, piperidinyl, piperazinyl, morpholinyl, pyrrolyl, oxazinyl, thiazinyl, diazinyl, triazinyl, tetrazinyl, imidazolyl, benzimidazolyl, tetrazolyl, indolyl, isoquinolinyl, quinolinyl, quinazolinyl, pyrrolidinyl, purinyl, isoxazolyl, benzisoxazolyl, furanyl, furazanyl, pyridinyl, oxazolyl, benzoxazolyl, thiazolyl, benzthiazolyl, thiophenyl, pyrazolyl, triazolyl, benzodiazolyl, benzotriazolyl, pyrimidinyl, isoindolyl and indazolyl. A
heterocycle group can be unsubstituted or substituted with one or more of the following groups: -C1-Cs allcyl, halo, -allcylhalo, hydroxy, -O-C1-Cs alkyl, -NH2, -aminoalkyl, -aminodialkyl, -COOH, -C(O)O-(C1-Cs alkyl), -OC(O)-(C1-Cs alkyl), -N-amidoalkyl, -C(O)NHZ, -carboxamidoalkyl or -N02.
A "Heterocyclic amine" is a heterocycle, defined above, having 1-4 ring nitrogen atoms. Representative examples of heterocyclic amines include, but ar a not limited to, piperidinyl, piperazinyl, pyrrolyl, oxazinyl, thiazinyl, diazinyl, triazinyl, tetrazinyl, imidazolyl, benzimidazolyl, tetrazolyl, indolyl, isoquinolinyl, quinolinyl, quinazolinyl, pyrrolidinyl, purinyl, isoxazolyl, benzisoxazolyl, pyridinyl, oxazolyl, benzoxazolyl, thiazolyl, benzthiazolyl, pyrazolyl, triazolyl, benzodiazolyl, benzotriazolyl, pyrimidinyl, isoindolyl, indazolyl and morpholinyl; each of which can be unsubstituted or substituted with one or more of -N-(C1-Cs alkyl), -C(O)-(C1-CS alkyl), -N-C(O)(Cl-C~
alkyl), _ 2q. _ -O-(Cl-CS alkyl), -halo, -alkylhalo, -alkanol, -alkylamino, -hydroxy, -NOZ, -NHZ, -aminoalkyl, -aminodialkyl, -heterocyclic amine, -C1-Cio alkyl, -C2-Clo alkenyl, -C1-Cio alkynyl, -aryl, -benzyl, -alkylamido, -alkylcarboxy, -COOH, -CI-CS
alkylene-OC(O)-C1-CS alkyl, -CI-CS alkylene-C(O)O-C1-CS alkyl, or a heterocycle or C3-C8 carbocycle which can be unsubstituted or substituted with one or more of -C1-Clo alkyl, -O-(C1-CS alkyl), -halo, -alkylhalo, -alkanol, -alkylamino, -hydroxy, -NO2 or -NH2.
"Halo" is -F, -Cl, -13r or -I.
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.
The invention also includes pharmaceutical compositions comprising an effective amount of an Isoquinoline Derivative and a pharmaceutically acceptable carrier. The invention includes an Isoquinoline Derivative when provided as a pharmaceutically acceptable prodrug, hydrated salt, such as a pharmaceutically acceptable salt, or mixtures thereof.
, Representative "pharmaceutically acceptable salts" include, e.g., water-soluble and water-insoluble salts, such as the acetate, amsonate (4,4-diaminostilbe.ne-2, 2 -disulfonate), benzenesulfonate, benzonate, bicarbonate, bisulfate, bitartrate, borate, bronude, butyrate, calcium edetate, camsylate, carbonate, chloride, citrate, clavulariate, dihydrochloride, edetate, edisylate, estolate, esylate, fiunarate, gluceptate, gluconate, glutamate, glycollylarsanilate, hexafluorophosphate, hexylresorcinate, hydrabamine, hydrobromide, hydrochloride, hydroxynaphthoate, iodide, isothionate, lactate, lactobionate, laurate, malate, maleate, mandelate, mesylate, methylbromide, methylnitrate, methylsulfate, mucate, napsylate, nitrate, N-methylglucamine ammonium salt, 3-hydroxy-2-naphthoate, oleate, oxalate, palmitate, pamoate (l,l-methene-bis-2-hydroxy-3-naphthoate, einbonate), pantothenate, phosphate/diphosphate, picrate, polygalacturonate, propionate, p-toluenesulfonate, salicylate, stearate, subacetate, succinate, sulfate, sulfosaliculate, summate, tannate, tartrate, teoclate, tosylate, triethiodide, and valerate salts.
An "effective amount" when used in connection an Isoquinoline Derivative is an amount effective for: (a) treating or preventing an inflammatory disease or a reperfusion disease or (b) inhibiting PARS in an irz vivo or an irz vitr~ cell.
The following abbreviations are used herein and have the indicated definitions:
AcOH is acetic acid, CEP is Cecal Ligation and Puncture, DMEM is Dulbecco's Modified Eagle Medium, DMF is N,N-dimethylformamide, DMSO is dimethylsulfoxide, EtOAc is ethyl acetate, EtOH is ethanol, HEPES is 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid, HPLC is high pressure liquid chromatography, LPS is lipopolysaccharide, MeCN is acetonitrile, MeOH is methanol, MS is mass spectrometry, Ms is mesyl (methanesulfonyl), NEt3 is triethylamine, NMR is nuclear magnetic resonance, PBS is phosphate-buffered saline (pH 7.4~), PARS is poly(ADP-ribosejsynthetase, Py is pyridine, SDS is dodecyl sulfate (sodium salt), STS
is streptozotocin, TCA is tricholoroacetic acid, Tf is triflyl (trifluoromethanesulfonyl), TFA
is trifluoroacetic acid, THF is tetrahydrofuran; TLC is thinlayer chromatography, TNF is tumor necrosis factor, TRIS is Tris(hydroxymethyl)aminomethane and Ts is tosyl (p-toluenesulfonyl).
Methods for using Isoquiyzoliyae Derivatives The invention also includes methods for inhibiting PARS in a cell. PARS, which is also kr~owr~ as poly(ADP-ribose)syr~tl~~ctase, PARP ((poly(ADP-ribose) polymeras;., EC
2.4.99) and ADP-ribosyltransferase (ADPRT, EC 2.4.2.30), is a nuclear enzyme that catalyzes a transfer of the ADP ribose moiety of NAD+ to an acceptor protein.
In one embodiment, the method comprises contacting a cell with an Isoquinoline Derivative in an amount sufficient to inhibit PARS in the cell. In general, any cell having, or capable of having, PARS activity or capable of expressing PARS can be used.
The cell can be provided in any form. For example, the cell can be provided i~2 vitro, ex vivo, or in vivo. PARS activity can be measured using any method known in the art, e.g., methods as described in Banasik et al., J. Biol. Chem. 267:1569-75 (1991).
Illustrative examples of cells capable of expressing PARS include, but are not limited to, muscle, bone, gum, nerve, brain, liver, kidney, pancreas, lung, heart, bladder, stomach, colon, rectal, small intestine, skin, esophageal, eye, larynx, uterine, ovarian, prostate, tendon, bone maiTOw, blood, lymph, testicular, vaginal and neoplastic cells.
Also provided in the invention is a method for inhibiting, preventing, or treating inflammation or an inflammatory disease in a subject. The inflammation can be associated with an inflammatory disease. Inflammatory diseases can arise where there is an inflammation of the body tissue. These include local inflammatory responses and systemic inflammation. Examples of such diseases include: organ transplant rejection;
reoxygenation injury resulting from organ transplantation (see Carupp et al., J. 1121. Cell Cardzol. 31:297-303 (1999)) including, but not limited to, transplantation of the following organs: heart, lung, liver and kidney; chronic inflammatory diseases of the joints, including arthritis, rheumatoid arthritis, osteoarthritis and bone diseases associated with increased bone resorption; inflammatory bowel diseases such as ileitis, ulcerative colitis, Barren's syndrome, and Crohn's disease; inflammatory lung diseases such as asthma, adult respiratory distress syndrome, and chronic obstructive airway disease;
inflammatory diseases of the eye including corneal dystrophy, trachoma, onchocerciasis, uveitis, sympathetic ophthalmitis and endophthalmitis; chronic inflammatory diseases of the gum, including gingivitis and periodontitis; tuberculosis; leprosy; inflammatory diseases of the kidney including uremic complications, glomerulonepluritis and nephrosis;
inflammatory diseases of the skin including sclerodermatitis, psoriasis and eczema;
inflammatory diseases of the central nervous system, including chronic demyelinating diseases of the nervous system, multiple sclerosis, AIDS-related neurodegeneration and Alzheimer s disease, infectious meningitis, encephalomyelitis, Parkinson's disease, Huntington's disease, am;~otrnphic lateral sclerosis and viral or autoinimune encephalitis;
a~~r_oimmune diseases including Type I and Type II diabetes mellitus; diabetic complications, including, but not limited to, diabetic cataract, glaucoma, retinopathy, nephropathy, such as nucroaluminuria and progressive diabetic nephropathy, polyneuropathy, gangrene of the feet, atherosclerotic coronary arterial disease, peripheral arterial disease, nonketotic hyperglycemiehyperosmolar 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 diabeticorum; immune-complex vasculitis, systemic lupus erythematosus (SLE); inflammatory diseases of the heart such as cardiomyopathy, ischemic heart disease hypercholesterolemia, and atherosclerosis; as well as various other diseases that can have significant inflammatory components, including preeclampsia; chronic liver failure, brain and spinal cord trauma, and cancer. 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.~., shock associated with pro-inflammatory cytokines. Such shock can be induced, e.,g., by a chemotherapeutic agent that is adminstered as a treatment for cancer.
In one embodiment, a reoxygenation injury resulting from organ transplantation occurs during the organ transplantation.
The invention also includes methods for treating, preventing, or otherwise inhibiting reperfusion disease in a subject in need of treatment, prevention, or inhibition thereof. The method comprises administering an Isoquinoline Derivative in an amount sufficient to treat, prevent or inhibit repei~fusion disease in the subject.
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 disease can result following a naturally occurring episode, such as a myocardial infarction, stroke, or during a surgical procedure where blood fhow in vessehs is intentionally or unintentionally blocked.
In some embodiments, the subject is administered an effective amount of an Isoquinoline Derivative.
The invention also includes pharmaceuticah compositions useful for treating or preventing an inflammatory disease or a reperfusion disease, or for inhibiting PARE
activity, or more than one of these activities. The compositions can be suitable for internal use and comprise an effective amount of an Isoquinoline Derivative and a ph2rp-y~e~tj~~lly accP.ptahle carrier. The Isoquinoline Derivatives are especial_hy usefzsl ix~
that they demonstrate very low peripheral toxicity or, no peripheral toxicity.
The Isoquinoline Derivatives can be administered in amounts that are sufficient to treat or prevent an inflammatory disease or a reperfusion disease and/or prevent the development thereof in subjects.
Administration of the Isoquinoline Derivatives can be accomplished via any mode of administration for therapeutic agents. These modes include systemic or local administration such as oral, nasal, parenteral, transdermah, subcutaneous, vaginal, buccal, rectah or topical administration modes.
Depending on the intended mode of administration, the compositions can be in solid, semi-solid or liquid dosage form, such as, for example, injectables, tablets, suppositories, pihls, time-release capsules, ehixirs, tinctures, emulsions, syrups, powders, liquids, suspensions, or the like, preferably in unit dosages and consistent with conventional pharmaceutical practices. Likewise, they can also be administered in intravenous (both bolus and infusion), intraperitoneal, subcutaneous or intramuscular form, all using forms well known to those skilled in the pharmaceutical arts.
Illustrative pharmaceutical compositions are tabhets and gelatin capsules comprising an Isoquinohine Derivative and a pharmaceutically acceptable carrier, such as a) a dihuent, e.g., lactose, dextrose, sucrose, mannitol, sorbitoh, cellulose, sodium, saccharin, glucose and/or glycine; b) a lubricant, e.g., silica, talcum, stearic acid, its magnesium or calcium salt, sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and/or polyethylene glycol; for tablets also; c) a binder, e.g., magnesium aluminum silicate, starch paste, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose, magnesium carbonate, natural sugars such as glucose or beta-lactose, com sweeteners, natural and synthetic gums such as acacia, tragacanth or sodium alginate, waxes and/or polyvinylpyrrolidonc, if desired; d:) a disintegrant, e.g., starches, agar, methyl cellulose, bentonite, xanthan gum, algiic acid or its sodium salt, or effervescent mixtures; and/or e) absorbent, colorant, flavorant and sweetener.
Liquid, particularly injectable, compositions can, for example, be prepared by dissolution, dispersion, etc. For example, the Isoquinoline Derivative is dissolved in or mixed with a phamnaceutically acceptable solvent such as, for example, water, saline, aqueous dextrose, glycerol, ethanol, and the like, to thereby form an injectable isotonic solution or suspension.
T_h_P Tsoquinoline Derivatives can be. also fo_rmulate.d as a suppositn_ry that can be prepared from fatty emulsions or suspensions; using polyalkylene glycols such as propylene glycol, as the carrier.
The Isoquinoline Derivatives can also be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamellar vesicles and multilamellar vesicles. Liposomes can be formed from a variety of phospholipids, containing cholesterol, stearylamine or phosphatidylcholines. In some embodiments, a film of lipid components is hydrated with an aqueous solution of drug to a form lipid layer encapsulating the drug, as described in United States Patent No.
5,262,564.
Isoquinoline Derivatives can also be delivered by the use of monoclonal antibodies as individual caiTiers to which the Isoquinoline Derivative molecules are coupled. The Isoquinoline Derivatives can also be coupled with soluble polymers as targetable drug caiTiers. Such polymers can include polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamide-phenol, polyhydroxyethylaspanamidephenol, or polyethyleneoxidepolylysine substituted with palmitoyl residues. Furthermore, the Isoquinoline Derivatives can be coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for example, polylactic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates and cross-linked or amphipathic block copolymers of hydrogels.
Parental injectable administration is generally used for subcutaneous, intramuscular or intravenous injections and infusions. Injectables can be prepared in conventional forms, either as liquid solutions or suspensions or solid forms suitable for dissolving in liquid prior to injection.
~ne embodiment, for parenteral administration employs the implantation of a slow-release or sustained-released system, according to IJ.S. Pat. IVo.
3,710,795, incorporated herein by reference.
The compositions can be sterilized or contain non-toxic amounts of adjuvants, such as preserving, stabilizing, wetting or emulsifying agents, solution promoters, salts for regulating the osmotic pressure pH buffering agents, and other substances, including, but not limited to, sodium acetate or triethanolamine oleate. In addition, they can also contain other therapeutically valuable substances.
Compositions can be prepared according to conventional mixing, granulating or coating methods, respectively, and the present pharmaceutical compositions can contain from about 0.1 % to abort 99 %, preferably from about 1 % to about 70 % of the Isoquinoline Derivative by weight or volume.
The dosage regimen utilizing the Isoquinoline Derivative is, selected in accordance with a variety of factors including type, species, age, weight, sex and medical condition of the patient; the severity of the condition to be treated; the route of administration; the renal or hepatic function of the patient; and the particular Isoquinoline Derivative employed. A physician or veterinarian of ordinary skill in the art can readily determine and prescribe the effective amount of the drug required to prevent, counter or arrest the progress of the condition.
Effective dosage amounts of the present invention, when used for the indicated effects, range from about 0.05 to about 1000 mg of Isoquinoline Derivative per day.
Compositions for izz vivo or irz vitro use can contain about 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0, 50.0, 100.0, 250.0, 500.0 or 1000.0 mg of Isoquinoline Derivative. In one embodiment, the compositions are in the form of a tablet that can be scored.
Effective plasma levels of the Isoquinoline Derivatives can range from about 0.002 mg to about 50 mg per kg of body weight per day.
Isoquinoline Derivatives 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, Isoquinoline Derivatives 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 Isoquinoline Derivative ranges from about 0.1 % to about 15 °Io, w/w or w/v.
Methods for frlaki~ag the Is~e~uiriealiaze I~erivativ~s Examples of synthetic pathways useful for making Isoquinoline Derivatives are set forth in the Examples below and generalized in schemes ~-10.
lVIethods useful for making Isoquinoline Derivatives of formula (I~) wherein X
is -CHI- and 125 is O are illustrated below in Scheme 1.

Scheme 1 MeOH, ~ 2. NaBH4, EtOH, 24 h~ r~~ i H or RMgX, THF, rt H
1 2 3a-c a: R=H;
3. TFA, Et3SiH b: R=Me;
3. TFA, Et3SiH c: R=m-MeOC6H4 4. PBr3, TFA or CICOCH2CI, Py i°C
7. HNRiR2, NEt3, CHZCIz ° H
or NaHCOc/EtOAc, HNRiR2 9. Fuming HN03, AcOH, 0 C; °
or nitrofluoroborate, MeCN, -40 C 4a-b S. MeS03H, 0°C, 1-2 hr a: X=OCOCHzCI;
b: X=B!
OZN
5. Amine, MeOH
r 10.
Ammonium formate, Pd-C, DMF

11.
CICOCH2CI, EtOAc, sat.
NaHC03 12.
NHRiR2, DMSO, rt O
O Sa-a RHxC"NH
\ a: A=NMez N H b: A=NEt2 c: A=4-Me-piperazine-1-yl d: A=piperidine-1-yl e: A=morpholine-4-yl l0a-b L
a: R=morpholine-4-yl b: R=NMe2 wherein compounds 8a-8af are as follows:
Sa-of a.R=4-Methyl-piperazine-1-yl q. R= N(CH2CH2NMez)z b.R=4-CH2C02Me-piperazine-1-ylr. R=-N(CHZCH2OH)2 c.R=4-CH2COZOH-piperazine-1-yls. R=-NHCHZCHaCN

d.R=imidazole-1-yl t. R=-NHC(NH)NH2 e.R=L-prolinol u. R=-NH[4-(1,2,4-triazole)1 f.R=morpholine-4-yl v. R= NH[4-(N-morpholine)phenyl]

g.R=NHCHZCHZNMe2 w. R=-NHCH2CH2(4-N-benzylpiperidine) h.R=NHCH2CH2-piperidine-1-yl x. R= NHCHzCH2(2-thienyl) i.R=NHCHzCHZN-(pyridine-2-yl)y. R=-NH[i-(4-azabenzimidazole)]

].R=NHCH2CH2-morpholine-4-yl z. R=-NH[i-(4-(2'-pyridyl)piperazine)]

k.R=NHCHZCH~-(2-N-Me-tetrahydropyrrolidine-1-aa. R=-NHCHZCH2N[CH2CH20H]2 yl) ab. R= NH[1-(4-benzlpiperazine)]

I.R=NHCH2CH2CH2-morpholine-4-ylac. R=-NH2 m.R=NHCHzCH2CH2-(tetrahydropyrrolidine-1-yl)ad. R=-NHCHZCHzPh n.R=NHCH2CHzCH2-imidazole-1-ylae. R=-NHCH2CH2[4-OMe(phenyl)]

o.R=NHCHZCHzCH2-(4-methylpiperazine-1-yi)af. R= NHC(O)(N-morpholine) p.R=N(CH2CH2NEtz)z 5,6-dihydro-5,11-diketo-11H-isoquinoline (2) was prepared by reacting compound 1 (Aldrich Chemical, Milwaukee, WI) with ammonia in methanol.
(~) 11-hydroxy-5,6-dihydro-5-oxo-11H-indeno[1,2-c]isoquinoline (3a) was prepared by reacting 2 with NaBH4 in ethanol.
(~) 11-hydroxy-11-methyl-5,6-dihydro-5-oxo-11H isoquinoline (3b) was prepared by reacting 2 with MeMgI.
(~) 11-hydroxy-11-(m-methoxyphenyl)-5,6-dihydro-5-oxo-11H-indeno[1,2-c]isoquinoline (3c) was prepared from 2 using m-Me0-C~H4MgI.
(~) 11-N,N-dimethylamino-5,6-dihydro-5-oxo-11H-indeno[1,2-c]isoquinoline (Sa) was prepared from 3a using chloroacetylchloride followed by reacting with dimethylamine. Similarly prepared are: (~) 11-N,N-diethylamino-5,6-dihydro-5-oxo-11H-indeno[1,2-c]isoquinoline (5b), (~) 11-N (piperidino-1-yl)-5,6-dihydro-5-oxo-11H-indeno[1,2-c]isoquinoline (5d), (~) 11-N (4-methylpiperazino-1-y1)-5,6-dihydro-5-oxol IH-indeno[1,2-c]isoquinoline (Sc), (~) 11-N (moipholino-4-yl)-5,6-dihydro-oxollH-isoquinoline (Se). (+) 11-N (morpholino-4-yl)-5,6-dihydro-5-oxo-11H-indeno[1,2-c]isoquinoline (Se) was also prepared from (~) 11-bromo-5,6-dihydro-5-oxo-11H-indeno[1,2-c]isoquinoline (4b).
5,6-I~ihydro-5-oxo-llFl-indeno-[1,2-c]isoquinoline (6) is prepared by reduction of 5,6-dihydro-5,11-dil~eto-11H-isoquinoline (2,) or (~) 11-hydroxy-5,6-dihydro5-oxo-11 H-isoquinoline (3n) using CF~C~~H/triethylsilane. 9-Chlorosulphonyl-5,6-dihydro-5-oxo-1 1H-indeno-[1,2-c]isoquinoline (7) was prepared by chlorosulfonation of 5,6-dihydro-5-oxo-11H-indeno-[1,2-c]isoquinoline (6). 9-[N-(4-methylpiperazine-lyl)sulphonyl]-5,6-dihydro-5-oxo-11H-indeno-[1,2-c]isoquinoline (8~) was prepared from 9-chlorosulphonyl-5,6-dihydro-5-oxo-11H-indeno-[1,2-c]isoquinoline (7), and N-methylpiperazine. Similarly prepared are: 9-[N-(4-carbomethoxymethyienepiperazino-lyl)sulphoriyl]-5,6-dihydro-5-oxo-11 H-indeno-[ 1,2-c]isoquinoline (8b), 9-[N 4-(2-hydroxyethylpiperazino-1-yl)-sulphonyl]-5,6-dihydro-5-oxo-11H-indeno-[1,2-clisoquinoline (8c), 9-[N-(imidazolo-1-yl)sulphonyl]-5,6-ciihy~lro-5-oxo-1 IH-isoquinoline (8d), 9-[N (2-hydroxyprolinyl)sulphonyl]-5,6-dihydro-5-oxo-11H-indeno[1,2-c]isoquinoline (8e), 9-[N moipholinesulphonyl]-5,6-dihydro-5-oxo-11H-indeno[l,2-c]isoquinoline (8f7, 9-[N (2-[N,N
dimethylamino]ethyl)aminosulphonyl]-5,6-dihydro-5-oxo-11H-indeno[1,2-c]isoquinoline (8g), 9-[N-(2-[piperidino-1-yl]ethyl)-aminosulphonyl]-5,6-dihydro-5-oxo-11H-indeno[1,2-c]isoquinoline (8h), 9-[N
(2-(pyridino-2-yl)-ethyl)-aminosulphonyl]-5,6-dihydro-5-oxo-11H-indeno[1,2c]isoquinoline (8i), 9-[N-(2 -[morpholino-4-yl]ethyl) -aminosulphonyl]-5,6-dihydro-5-oxo-11H-indeno[1,2-c]isoquinoline (8j), 9-[N-(2-[N methyltetrahydropyrroiidino-1-yl]ethyl) aminosulphonyl]-5,6-dihydro-5-oxo-11H-indeno-[1,2-c]isoquinoline (8k), 9-[N-(3-[morpholino-4-yl]
propyl)-aminosulphonyl]-5,6-dihydro-5-oxo-11H-indeno-[1,2c]isoquinoline (81), 9-[N (3-[tetrahydropyrrolodino-1-yl]propyl)aminosulphonyl]-5,6-dihydro-5-oxo-11H-indeno-[1,2-c]isoquinoline (8m), 9-[N (3-[imidazolo-1-yl]propyl)aminosulphonyl]-5,6-dihydro-5-oxo-11H-indeno-[1,2-c]isoquinoline (8n), 9-[N [3-(4-methylpiperazino-1-yl]propyl)-aminosulphonyl]-5,6-dihydro-5-oxo-11H-indeno-[ 1,2c]isoquinoline (8~), 9-[N,N di-(2-[N,N-diethylamino]ethyl)-aminosulphonyl]-5,6-dihydro-5-oxo-11H-indeno-[1,2-c]isoquinoline (gyp), 9-[N,N di-(2-[N,N
dimethylamino]ethyl)aminosulphonyl]-5,6-dihydro-5-oxo-11H-indeno-[1,2-c]isoquinoline (~q), and 9-[Id l~ di(2-[N,N dihydroxyethylamino] ethyl)-aminosulphonyl]-5,6-dihydro-5-oxo-11H-indeno-[1,2c]isoquinoline (8r).

Compounds 8s-8af can be prepared using the methods described above for making compounds of 8a-8r, using appropriate amine intermediates.
Scheme 2 illustrates a method useful for making terminal carboxylic acid compounds of formulas Bag-8a~. This method comprises reacting sulfonyl chloride 7 with the alkyl ester of an amino acid in the presence of a base, preferably triethyamine, to provide an intermediate terminal carboxylic acid alkyl ester, which is then hydrolyzed using a base such as sodium hydroxide to provide the corresponding teuminal carboxylic acid.
scheme 2 3')COOR" (4't), N
or ~)~COOR" (42), N
2. NaOH
7 Bag-ao ag.R=-NHCHzCOOH

ah.R=-NH(CHz)zCOOH

ai.R=-NH(CHz)sCOOH

aj.R=-NH(CHz)4COOH

ak.R=-NH(CHz)SCOOH

al.R=-NHCH(CHzCOOH)COOH

am.R=-NHCH((CHz)zCOOH)COOH

an.R=-NHCH((CHz)aNHz)COOH

ao.R=-NHCH(CHzOH)COOH

wherein:
R' is -alkylcarboxy, -alkylamino or -alkanol;
R" is -C1-C6 alkyl; and n is an integer ranging from 1 to 6.
Caeneral Procedure for tnakizy 9-sulfonamide carboxylic acid derivatives PT~pezf"czt1011 ~f 9-SafLfOr~cznldelca cer~b~.~~ldc aeZe~ ester To a 0.5 M solution of an ester of formula 41 or 42 in CH~C12 is added compound 7 ( 1.0 eq) and the resulting mixture is stitTed for 5 minutes. Triethylamine (about 5 eq) is then added and the resulting reaction is stirred at room temperature and monitored using TLC or HPLC until complete. The reaction mixture is filtered, the solid is washed using MeOH to provide the intermediate 9-sulfonamido carboxylic acid ester which can be used without further purification.
Ester- I~yds~~lysis To an approximately 0.5 M solution of a 9-sulfonamide carboxylic acid ester in ethanol is added about 3.0 N aqueous sodium hydroxide (about 5.0 eq) and the resulting reaction is refluxed if necessary and monitored using TLC or HPLC until completion.
The reaction nuxture is neutralized to about pH 7.0 using about 1.0 N HCl and the neutralized reaction mixture is extracted twice using EtOAc. The combined EtOAc layers are washed sequentially with water and saturated aqueous sodium chloride, then dried ov..r s,:~diuyn sulfate and concentrated in vacuo to afford a crude residue vahich i~
purified using flash column chromatography to provide the desired 9-sulfonamide carboxylic acid compound.
Acid hydrolysis with neat TFA can be useful where the sulfonamide has a t-butyl ester group.
In another embodiment, illustrated below in Scheme 3, Isoquinoline Derivatives of general formula 13 can be made by a method comprising contacting a compound of formula 11 and a compound of formula 12 in the presence of a base for a time and at a temperature sufficient to make a compound of formula 13.

Scheme 3 Ra wherein:
Rl-R4 and R7-Rlo are as defined above for formula (I); and Rb is -Cl, -Br, -I, -OMs, -OTs or -OTf.
In one embodiment, Rb is -Br.
In another embodiment, Rv and R~ are both -Br.
In one embodiment, about 0.1 to about 10 equivalents of a compound of Formula 12 are used per about 1 equivalent of a compound of Formula 11.
In another embodiment, about 0.5 to about 5 equivalents of a compound of Formula 12 are used per about 1 equivalent of a compound of Formula 11.
In still another embodiment, about 1 to about 2 equivalents of a compound of Formula 12 are used per about 1 equivalent of a compound of Formula 11.
In one embodiment, about 1 to about 10 equivalents of base are used per about equivalent of a compound of Formula 11.
In another embodiment, about 3 to about 7 equivalents of base are used per about 1 equivalent of a compound of Formula 11.
In a yet another embodiment, about 5 to about 6 equivalents of base are used per about 1 equivalent of a compound of Formula 11.
Suitable bases for use in the method of Scheme 3 are organic bases such as triethylamine, diisopropylamine, diisopropylethylamine, pyridine, lutidine and imida~ole;
and inorganic bases such as all~ali metal carbonates, including sodium carbonate, potassium carbonate and cesium carbonate.
In one embodiment, the base is triethylamine.

In another embodiment, the base is potassium carbonate.
The method of Scheme 3 can be carried out in the presence of a solvent, such as acetonitrile, methylene chloride, chloroform, THF, DMF, DMSO, ethyl acetate, acetone, benzene, diethyl ether, water or mixtures thereof.
In one embodiment, the solvent is acetonitrile.
In another embodiment, the solvent is DMF.
In still another embodiment, where the solvent is not water, the solvent is substantially anhydrous, i.e., comprises less than about 1 % water.
In one embodiment, the method of Scheme 3 is carried out for a time of about 0.5 hours to about 48 hours.
In another embodiment, the method of Scheme 3 is cauried out for a time of about 3 hours to about 36 hours.
In still another embodiment, the method of Scheme 3 is carried out for a time of abOLlt 8 1?OL~rS tt3 abrJLlt ~4 hoLLrS.
In yet another embodiment, the method of Scheme 3 is carried out for a time of about 15 hours to about 20 hours.
In a further embodiment, the method of Scheme 3 is carried out at a temperature of about 0°C to about 200°C.
In another embodiment, the method of Scheme 3 is carried out at a temperature of about 25°C to about 150°C.
In yet another embodiment, the method of Scheme 3 is carried out at a temperature of about 50°C to about 100°C.
General Procedure For The Preparation of Compounds of Formula 13 To a solution of a homophthalic anhydride of formula 11 (about 1 equivalent) in a suitable solvent, such as acetonitrile, is added a compound of Formula 12 (about 1 to about 2 eq) followed by a suitable base, such as triethylamine (about 1 to about 5 eq).
The resulting reaction is reaction is allowed to stir for about 1 hour, at which time a colored precipitate appears. The reaction is then heated at reflux for about 20 hours, cooled to room temperature and filter ed. The collected solid is washed using acetonitrile and dried under vacuum to provide a compound of Formula 13.
Scheme 4 o ci p i. KNOg, HZS04 ~NH POCl3, 145 °C ~ ~ N 2. HCl (50%), heat ~ ~ ANN
/ ~ ~ OzN ~ /
O p O

1. pyridine Hl3r perbromide, Ammonium fur mate, AcOH MeOH, Pd-C, 100 °C
2. dil. HCI, heat NH \ ~NH
/ NzN / /
Br 1. CICOCH,C1, EtOAc, sat. NaHC03 2. Me,NH, DMSO
O
O ~ ~NH
Me2N~N / /
H

The amide derivative 2-dimethylamino-N-(5-oxo-5,11-dihydro-6H-indeno[1,2c]isoquinoiin-2-yl)-acetamide (17) was prepared from 5-chloro-11H-indeno [1,2c]isoquinoline (14). Compound 14 was subjected to nitration to provide nitro compound 15, which was reduced using ammonium formate to provide amine 16, which was derivatized to acetamide 17, and followed by amination of the chloroacetamide intermediate. 2-bromo5,6-dihydro-5-oxo-11H-indeno[1,2-c]isoquinoline (18) was prepared by bromination of Compound 14.
Scheme 5 illustrates methods useful for making oxygen-substituted Isoquinoline Derivatives of formula (I), where RS and X are oxygen.

Scheme 5 Ri OH
R2 \ COpRa R10 \ CN 1. KN03, HZSOQ f R ~ / COpRa + / 2. HCI (50%), heat R9 ~ ~R7 R4 Rb R
a O O
NH
NH BBr~
/ / ---~ / /
CHZCh 10CH3 ~~~OH
22b-d 23a-c R'X HCl ---~~~u R' O O
NH
NH R,~NH \
/ / -- / /
O
22f, g ~ ~ C02H O ~ \ C02NHR"
wherein:
RI-Rs are as defined above for formula (I);
each occurrence of Ra is independently CI-C3 alkyl;
Rb is -Cl, -Br, -I, -OMs, -OTs or -OTf;
R' is -CI-CIO alkyl, alkanol or alkylcarboxy; and R" is -CI-CIO alkyl, aryl, heterocycle, alkanol or alkylcarboxy.
In one embodiment, Ra is methyl.
In another embodiment, Rb is -Br In another embodiment, illustrated above in Scheme 5, Isoquinoline Compounds of formula 22 can be made by a method comprising contacting a compound of formula 20 and a compound of formula 21 in the presence of a base for a time and at a temperature sufficient to make a compound of formula 22.
In one embodiment, about 0.1 to about 10 equivalents of a compound of Formula 20 are used per about 1 equivalent of a compound of Formula 21.
In a~iother embodiment, about 0.5 to about 5 equivalents of a compound of Formula 20 are used per about 1 equivalent of a compound of Formula 21.
In still another embodiment, about 1 to about 2 equivalents of a compound of Formula 20 are used per about 1 equivalent of a compound of Formula 21.
In one embodiment, about 1 to about 10 equivalents of base are used per about equivalent of a compound of Formula 21.
In another embodiment, about 3 to about 7 equivalents of base are used per about 1 equivalent of a compound of Formula 21.
In a yet another embodiment, about 5 to about 6 equivalents of base are used per about 1 equivalent of a compound of Formula 21.
Suitable bases for use in the method are organic bases such as triethylamine, diisopropylamine, diisopropylethylamine, pyridine, lutidine and imidazole; and inorganic bagec cph as alkali metal carbonates s~2ch as sodium carbonate, potassium carbonate an.~1 cesium carbonate.
In one embodiment, the base is potassium carbonate.
In another embodiment, the base is triethylamine.
The method can be caiTied ou.t in the presence of a solvent, such as acetonitrile, methylene chloride, chloroform, THF, DMF, DMSO, ethyl acetate, acetone, benzene, diethyl ether, water or mixtures thereof.
In one embodiment, the solvent is DMF.
In another embodiment, the solvent is acetonitrile.
In still another embodiment, the solvent is substantially anhydrous, i.e., comprises less than about 1 % water.
In one embodiment, the method is carried out for a time of about 1 hour to about hours.
In another embodiment, the method is carried out for a time of about 18 hours to about 72 hours.
In yet another embodiment, the method is carried out for a time of about 24 hours to about 48 hours.
In one. embodiment, the method is carried out at a teanperature of about 25°C to about 200°C.
In another embodiment, the method is carried out at a temperature of about 50°C

to about 150°C.
In still another embodiment, the method is carried out at a temperature of about 75°C to about 125°C.
Scheme 6 illustrates methods useful for making nitrogen-substituted Isoquinoline Derivatives of the invention.
~chexrm 6 NO
\ NaNO~ \ Na~S?O,~
Ph ~ ~ Ph --,.-N AcOH s N
H H

NHS NHCO2Et \ ~ cico2Ec \ ~ o Ph I Ph pyridine ~ H Ph O

Ac~O

HY
Y
HZO a. Y = -NH=
b. Y =-NH(CH=)i (,morpholin-4-yl) -42.-In an alternate embodiment, illustrated below in Scheme 7, nitrogen-substituted Isoquinoline Derivatives of general formula 37 can be made by a method comprising contacting a compound of formula 36 and a compound of formula 11 or fol-mula 20 in the S presence of a base for a time and at a temperature sufficient to make a colxlpound of formula 37.
scheme 7 Ri Rp ~ COpRa Rb R
base RQ
O ORa 2~
20a: Ra=CH3 Ru=Br when ein:
c(o)R~

base Rl-R4 and R7-Rlo are as defined above for formula (I);
IO each occurrence of Ra is independently C1-C3 allcyl;
Rb 15 -Cl, -fir, -I, -~Ms, -OTs or -~Tf, and RC, is C1-C3 alkyl.
In one embodiment, Ra is methyl.
In another embodiment, Rb is -Br.
15 In a further embodiment, Ra is methyl and Rb is -Br.
In still another embodiment, R~ is methyl.

In one embodiment, about 0.1 to about 10 equivalents of a compound of Formula 11 are used per about 1 equivalent of a compound of Formula 36.
In another embodiment, about 0.5 to about 5 equivalents of a compound of Fonnula 11 are used per about 1 equivalent of a compound of Formula 36.
In still another embodiment, about I to about ? equivalents of a compound of Formula 11 are used per about 1 equivalent of a compound of Formula 36.
In one embodiment, about 0.1 to about 10 equivalents of a compound of Formula 20 are used per about 1 equivalent of a compound of Formula 36.
In another embodiment, about 0.5 to about 5 equivalents of a compound of Formula 20 are used per about 1 equivalent of a compound of Formula 36.
In still another embodiment, about 1 to about 2 equivalents of a compound of Formula 20 are used per about 1 equivalent of a compound of Formula 36.
In one embodiment, about 1 to about 10 equivalents of base are used per about enL,i~mlP.~,r of a cnmpour,r_l of Forml2la 36.
In another embodiment, about 3 to about 7 equivalents of base are used per about 1 equivalent of a compound of Formula 11.
In a yet another embodiment, about 5 to about 6 equivalents of base are used per about 1 equivalent of a compound of Formula 11.
Suitable bases for use in the method of Scheme 7 are organic bases such as triethylamine, diisopropylamine, diisopropylethylamine, pyridine, lutidine and imidazole;
and inorganic bases such as alkali metal carbonates such as sodium carbonate, potassium carbonate and cesium carbonate.
In one embodiment, the base is potassium carbonate.
In another embodiment, the base is triethylamine.
The method of Scheme 7 can be carried out in the presence of a solvent, such as acetonitrile, methylene chloride, chloroform, THF, DMF, DMSO, ethyl acetate, acetone, benzene, diethyl ether, water or mixtures thereof.
In one embodiment, the solvent is DMF.
In another embodiment, the solvent is acetonitrile.
In still another embodiment, the solvent is substantially anhydrous, i.e., comprises less than about 1 % water.
In one embodiment, the method of Scheme 7 is carried out for a time of about 1 hour to about 96 hours.
In another embodiment, the method of Scheme 7 is cam-ied out for a time of about 18 hours to about 72 hours.
In yet another embodiment, the method of Scheme 7 is carried out for a time of about 24 hours to about 48 hours.
In one embodiment, the method of Scheme 7 is carried out at a temperature of about 25°C to about 200°C.
In another embodiment, the method of Scheme 7 is carried out at a temperature of about 50°C to about 150°C.
In still another embodiment, the method of Scheme 7 is carried out at a temperature of about 75°C to about 125°C.
General Procedur a For The Prepay anon of Compounds of For mule 3'7 Front a Itot~T.ophtdtala.te:
To a solution of a homophthalate of Foumula 20 (about 1 eq) and an N-acylanthranilonitrile of Formula 36 (about 1 to about 2 eq) in a solvent such as DMF, under inert atmosphere, is added a base (about 5 eq), such as potassium carbonate and the reaction is allowed to stir for about 48 hours at about 100°C, then cooled to room temperature. The reaction mixture is then poured into about 1 N sodium hydroxide and the resulting solution is extracted with EtOAc. The EtOAc layer is washed sequentially with about 1 N HCl, saturated aqueous sodium chloride, dried over sodium sulfate, filtered and concentrated itt vacuo. The resulting residue is dissolved using warming in toluene and the resulting solution is cooled to room temperature and precipitated using hexanes. The solid precipitate is filtered, washed using hexanes and dried in a vacuum oven at 50°C for 72 h to provide a Compound of Formula 36.
The synthesis of phenyl amide 36, which is a useful intermediate in Scheme 7, is described below in Scheme 8. In this procedure, the amine group of a cyanoaniline compound of formula 38 is acylated using an acyl chloride or an anhydride in the presence of an acid.

Scheme 8 Re R~ O R~
acid 2 Rs Ran 3~ 36 wherein:
R7-R are as defined above for formula (I); and R° is C1-C3 alkyl.
Suitable acids for use in the method of Scheme 8 include, but are not limited to, S11.1f~lrlC a~I~ and phosphoric acid.
In one embodiment, the acid is sulfuric acid.
In another embodiment, R° is methyl.
The method of Scheme 8 can be carried out in the presence of a solvent, including, but not limited to, acetonitrile, methylene chloride, chloroform, THF, DMF, DMSO, ethyl acetate, acetone, benzene, diethyl ether or mixtures thereof.
General Procedure For Makin~pound of Formula 36 To a solution of a compound of Formula 38 (about 1 eq) in acetic anhydride (about 6 eq) at 90°C is added 1 drop of sulfuric acid (catalytic) and the resulting reaction is stil-red at about 90°C for about 2 h, and is then allowed to sit at room temperature for about 12 h. The reaction mixture is poured onto ice and the resulting solution is stirred for about 2 h, after which time the solution is neutralized to about pH 7.0 using 1 N
sodium hydroxide. The resulting precipitate is filtered, washed using water (about 4x) and dried under vacuum for about 72 h to provide a compound of Formula 36.
In another embodiment, illustrated below in Scheme 9, sulfur substituted Isoquinoline Derivatives of formula 40 can be made by a method comprising contacting a compound of formula 39 and a compound of formula 11a or formula 2~ in the presence of a base for a time and at a temperature sufficient to make a compound of formula 40.

Scheme 9 R~
R2 ~ C02Ra Rt R
bas0 O~ ORa 11a R~
R

base R1-R4 and R7-Rlo are as defined above for formula (I);
each occurrence of Ra is independently C1-C3 alkyl;
5 Rb is -Cl, -Br, -I, -OMs, -OTs or -OTf; and Rd is -H or -Br.
In one embodiment, Ra is methyl.
In another embodiment, Rb is -Br.
10 In still another embodiment, Ra is methyl and Rb is -Br.
In yet another embodiment, Rd is -H.
In a further embodiment, Rd is -Br.
In one embodiment, about 0.1 to about 10 equivalents of a compound of Formula lla are used per about 1 equivalent of a compound of Formula ~9.
15 In another embodiment, about 0.5 to about 5 equivalents of a compound of Formula 11a are used per about I equivalent of a compound of Formula ~9.
In still another embodiment, about 1 to about 2 equivalents of a compound of Formula lla are used per about I equivalent of a compound of Formula 39.
In one embodiment, about 0.1 to about 10 equivalents of a compound of Formula 11a are used per about 1 equivalent of a compound of Formula 39.
In another embodiment, about 0.5 to about 5 equivalents of a compound of Formula lla are used per about I equivalent of a compound of Formula 39.
In yet another embodiment, about 1 to about 2 equivalents of a compound of Formula lla are used per about 1 equivalent of a compound of Formula 39.
In one embodiment, about 0.1 to about 10 equivalents of a compound of Formula 20 are used per about 1 equivalent of a compound of Formula 39.
In another embodiment, about 0.5 to about 5 equivalents of a compound of Formula 20 are used per about 1 equivalent of a compound of Formula 39.
In yet another embodiment, about 1 to about 2 equivalents of a compound of Formula 20 are used per about 1 equivalent of a compound of Formula 39.
In one embodiment, about 1 to about 1_0 equivalents of base arP nzsed per about 1 equivalent of a compound of Formula 39.
W another embodiment, about 3 to about 7 equivalents of base are used per about 1 equivalent of a compound of Formula 39.
In a yet another embodiment, about 5 to about 6 equivalents of base are used per about 1 equivalent of a compound of Formula 39.
Suitable bases for use in the method of Scheme 9 are organic bases, such as triethylamine, diisopropylamine, diisopropylethylamine, pyridine, lutidine and imidazole;
and inorganic bases such as all~ali metal carbonates, including sodium carbonate, potassium carbonate and cesium carbonate.
In one embodiment, the base is potassium carbonate.
In another embodiment, the base is triethylamine.
The method of Scheme 9 can be carried out in the presence of a solvent, such as acetonitrile, methylene chloride, chloroform, THF, DMF, DMSO, ethyl acetate, acetone, benzene, diethyl ether, water or mixtures thereof.
In one embodiment, the solvent is DMF.
In another embodiment, the solvent is acetonitrile.
In one embodiment, the method of Scheme 9 is carried out for a time of about 1 hour to about 120 hours.
In another embodiment, the method of Scheme 9 is carried out for a time of about 24 hours to about 96 hours.

In yet another embodiment, the method of Scheme 9 is carried out for a time of about 60 hours to about 80 hours.
In one embodiment, the method of Scheme 9 is carried out at a temperature of about 0°C to about 200°C.
In another embodiment, the method of Scheme 9 is carried out at a temperature of about 25°C to about 150°C.
In still another embodiment, the method of Scheme 9 is cauried out at a temperature of about 50°C to about 100°C.
(seneral Procedure for the Preparation Compounds of Formula 40 Fr-oni c~ howoplatlac~lic anlzyclr-ide:
A solution of a mercaptobenzonitrile of Formula 39 (about 1.0 eq) and a homophthalic anhydride of Formula 11a (about 2.0 eq) in a suitable solvent such as acetonitrile under inert atmosphere is warmed with stirring until all reactants are in solution. A suitable base such as triethylamine (about 1 to about 5 eq) is added and the reaction is allowed to stir at about 90°C for about 72 hours, then cooled to room temperature. The reaction mixture is filtered, and the collected solid is washed using methanol, then dried in a vacuum oven at about 50°C to provide a compound of Formula 40.
From a IaomoplZtlzalate:
A solution of a mercaptobenzonitrile of Formula 39 (about 1.0 eq) and a homophthalate of Formula 20 (about 2.0 eq) in a suitable solvent such as acetonitrile under inert atmosphere is warmed with stirring until all reactants are in solution. A
suitable base such as triethylamine (about 1 to about 5 eq) is added and the reaction is allowed to stir at about 90°C for about 72 hours, then cooled to room temperature. The reaction mixture is filtered, and the collected solid is washed using methanol, then dried in a vacuum oven at about 50°C to provide a compound of Formula 40.
Scheme 10 below illustrates a method useful for mal~ing the Isoquinoline Derivatives of Formula (I) where X is -N(S~~~')- and Rl-R9 and ~' are as defined above for the Isoquinoline Derivatives of Formula (I).

Scheme 10 R4 Y~2S R R~ R5 R3 \ CO~~H3 ~2 R3 \
R2 ~ / (~~~(~H3 ~ 2 ~ / / H R6 R1 Br NaH, THF R
R N R
41 ~'~z~ R9 Rs Isoquinoline Derivatives of Formula (Ij where x is -N(S~~Y)-isoquinoiine uerivatives of Formula (i) where X is -N(S~02Y)- can be maue using a one pot couplinglcyclization process by reacting a bromo intermediate of formula 41 with an aromatic nitrite of formula 42 in the presence of sodium hydride. The intermediates of formulas 41 and 42 can be made from commercially available starting materials using techniques well known to those skilled in the art of organic synthesis.
The invention is further described in the following examples, which do not limit the scope of the invention described in the claims. The following examples illustrate the synthesis of illustrative Isoquinoline Derivatives and demonstrate their usefulness for treating or preventing an inflammatory disease or reperfusion disease.
S. EXAMPLES
Example 1: Preparation of Illustrative Isoquinoline Derivatives a) General Methods Proton NMl2 spectra were obtained using a Varian 300 NHz spectrophotometer and chemical shift values (8) are reported in parts per million (ppm). TLC was performed using TLC plates precoated with silica gel 60 F-254, and preparative TLC was performed using precoated Whatman 60A TLC plates. All intermediates and final compounds were NC ~ R7 I

characterized on the basis of'H NMR and MS data.
b) Preparation of 5,6-dihydro-5,11-diketo-11H-indeno[1,2-c]isoquinoline (2):

A stirred suspension of 1 (55 g, 0.22 mol) in NH3/MeOH (7.0 N, 700 mL) was refluxed for 24 h. The reaction mixture was then allowed to cool to room temperature and was filtered and washed with MeOH to provide 46 g of the orange colored above-titled product in 84 % yield. 'H NMR (DMSO-d~): 8 7.48-7.61 (m, 4H), 7.80-7.88 (m, 1H), 7.86 (d, J= 8.7 Hz, 1H), 8.22 (d, J= 8.4 Hz, 1H), 8.44 (d, J= 7.5 Hz, 1H), 13.05 (s, 1H); '3C NMR (DMSO-D6): ~ 106.33, 121.63, 122.94, 1.23.27, 124.80, 1.28.4.5, 132.17, 133.60, 134.03, 134.08, 134.8, 134.81, 137.09, 156.41, 163.76,190.57; MS (ES-): m/z 246.2 (M-1); Anal. Calcd for Cl6HgN0~: C, 77.72; H, 3.67; N, 5.67; Found: C, 77.54;
H, 3.69, N, 5.69.
c) Preparation of (~) 11-hydroxy-5,6-dihydro-5-oxo-11H-indeno[1,2-c]isoquinoline (3a):
h 3a To a stirred suspension of 2 (2.5 g, 0.01 mol) in EtOH (25 mL) was added NaEH4 (3.75 g, 0.1 mol) at room temperature in small portions over 30 nun. The reaction mixture was stirred for an additional 2 h and then cooled to 0°C. It was then triturated with 10 % HCl (10 % soln.). The resulting solid precipitated was filtered and washed with water and MeOH to provide 3a (2.326 g, 92 %). 1H NMR (DMSO-d6): 8 5.58 (d, J = 8.1 Hz, 1H), 5.78 (d, J =8.7 Hz, 1H), 7.33 -7.89 (m, 6H), 7.95 (d, J = 7.8 Hz, 1H, 8.22 (d, J= 7.8 Hz, 1H), 12.29 (s, 1H); 13C NMR (DMSO-d6): S 77.44, 118.81, 120.15, 124.28, 125.04, 125.67, 126.34, 128.46, 128.64, 128.95, 133.27, 135.62, 136.12, 139.93, 148.559 163.69.; MS (~ES+): m/z 250.1 (M+1); Anal. Calcd for C6H11N~2: C, 77.10; H, 4.45; N, 5.62. Found: C, 77.01; H, 4.57, N, 5.59.
Similarly, by reacting 2 with MeMgI and m-MeO-C6H4MgEr, respectively, compounds (~) 11-hydroxy-11-methyl-5,6-dihydro-5-oxo-11H-indeno[1,2-c]isoquinoline (3b) and (~) 11-hydroxy-11-(2-methoxyphenyl)-5,6-dihydro-5-oxo-11H-indeno[1,2c]
isoquinoline (3c) were prepared.
d) Preparation of 11-substituted 5,6-dihydro-5-oxo-11F1-indeno[1,2-c]isoquinolines (5a-e):
H
w~ "
ci 4b Sa-a 5 a: R = NMe2 5b: R = NEt2 5c: R = -piperidine-1-yl 5d: R = -N-methyl-piperazin-4-yl 5e: R = -moipholin-1-yl To a stin-ed suspension of 3a (0.5 g, 2 mmol) in pyridine (10 mL) was added chloroacetyl chloride (0.81 g, 0.006 mol) at 0°C. The reaction mixture was allowed to warm to room temperature and allowed to stir for 24 h. The reaction nuxture was then poured on ice and extracted with EtOAc. The organic layer was separated, dried and concentrated to pi°ovide crude compound ~.a, which was treated further with dimethylamine and stirred at room temperature for 24 h. The reaction mixture was poured on ice, and treated with 10 % HCl The resulting mixture was then basified using saturated aqueous NaHC03 and the resulting solid was filtered to provide the desired product Sa. 1H NMR (DMSO-D6): b 2.31 (s, 6H), 5.00 (s, 1H), 7.28-7.45 (m, 3H), 7.68-7.73 (m, 2H), 7.95 (d,J = 6.9 Hz, 1H), 8.10 (d, J = 7.8 Hz, 1H), 8.21 (d, J = 8.1 Hz, 1H), 12.26 (s, 1H); 13C NMR (DMSO-D6): S 68.09, 116.28, 120.52, 124.58, 125.74, 126.27, 126.34, 127.68, 128.64, 133.02, 136.27, 144.45, 163.80; MS (ES+): m/~
277.2 (M+1 ).
The following compounds were also prepared by reacting ~.a as above with diethylamine, piperidine, N-methylpiperidine and morpholine, respectively: (~) diethylamino-5,6-dihydro-5-oxo-11H indeno[1,2-c]isoquinoline (5b), (~) 11-piperizin-5,6-dihydro-5-oxo-11H-indeno[1,2-c]isoquinoline (5c), (~) 11-(IV-methylpiperazin)-5,6-dihydro-5-oxo-11H-indeno[1,2-c]isoquinoline (5d), and (~) 11-morpholino-5,6-dihydro-5-oxo-11H-indeno[1,2-c]isoquinoline (5e).
e) Preparation of (~) 11-morpholino-5,6-dihydro-5-oxo-11H-indeno[1,2-c]isoquinoline (Se):
4b Se To a stirred suspension of 3a (0.6 g, 2.4 mmol) in trifluoroacetic acid (5 mL) was added phosphorus tribromide (1.0 M soln. in CH~C12, 3 mL) at room temperature, and the reaction mixture was stiiTed for 8 h. The reaction mixture was poured on ice and the resulting solid was filtered to provide bromo compound 4b (0.61 g, 76 °Io). 1H NMR
(DMSO-d6): b 7.35-7.50 (m, 3H), 7.61 (d, J= 6.6 Hz, 1H), 7.73 -7.82 (m, 2H), 7.94 (d, J
= 6.6 Hz, 1H), 8.23 (d, J= 7.8 Hz, 1H, 12.41 (s, 1H);'3NMR (DMSO-d~): ~ 52.06, 79.35, 114.43, 120.56, 123.58, 125.27, 125.50, 126.68, 128.55, 128.86, 129.66, 133.73, 135.91, 136.61, 141.39, 143.95, 163.74.

Compound 4b (0.5 g) was suspended in MeOH (10 mL) and treated with excess morpholine (about 5.0 eq) at room temperature and stirred at 60 °C for 3 h. The reaction mixture was poured on ice, and diluted with ethyl acetate (40 mL). The organic layer was separated and extracted in dil. HCl ( 10 % sole.), the aqueous layer was then basified with sat. aq. NaHCO3 and the resulting solid precipitated was filtered and dried to provide compound 5e (0.46 g, 90 %~). 1H NMR (DMSO-d6): ci 2.56 (m, 4H), 3.4-9 (m, 4H), 5.04 (s, 1H), 7.31-7.45 (m, 3H), 7.65 -7.76 (m, 2H), 7.96 (d, J= 7.2 Hz, 1H), 8.20-8.24 (m, 2H), 12.29 (s, 1H); 13C NMR (DMSO-D6): ~ 49.36, 67.62, 68.11, 115.20, 120.60, 124.47, 125.84, 126.34-, 126.4-1, 127.76, 128.30, 128.72, 133.09, 136.30, 135.96,140.35, 144.44, 163.67.
f) Preparation of 5,6-dihydro-5-oxo-11I~-indeno[1,2-c]isoquinoline (6):

Method I: To a stirred solution of the alcohol 3a (0.35 g, 1.4 mmol) in trifluoroacetic acid (10 mL) was added at room temperature triethylsilane (0.812 g, 7 mmol) and the reaction mixture was stiiTed for 24 h. Trifluoroacetic acid was evaporated iv vacuo and EtOAc was added to the resulting crude product. The resulting solid was filtered and washed with H20 and EtOAc to provide the above-titled compound 6 (0.285 g, 87 %). 1H NMR (DMSO-D~): 8 3.89 (s, 2H), 7.30 -7.47 (m, 3H), 7.59 (d, J=
6.9 Hz, 1H), 7.72 -7.74 (m, 2H), 7.98 (d, J= 7.8 Hz, 1H), 8.23 (d, J= 8.4 Hz, 1H), 12.31 (s, 1H);
13C NMR(DMSO-d6): b 33.51, 116.50, 120.19, 124.01, 125.51, 125.55, 126.42, 127.50, 127.68, 128.56, 133.45, 136.39, 137.53, 140.18, 143.80, 163.46; MS (ES): m/z 232.1 (M-1); Anal. Calcd for C1~H11N0: C, 82.38; H, 4.75; N, 6.00. Found: C, 81.79;
H, 4.45, N, 5.99.
Method II: To a stirred suspension of 2 (40 g, 0.16 mol) in trifluoroacetic acid (2.5 L) was added triethylsilane (94 g, 0.8 mol) in small portions at room temperature and the reaction mixture was stirred for 96 h, during which time the reaction progress was monitored using TLC (eluent - 5 % MeOH/CH2C12). The reaction mixture was slowly poured on ice, filtered, washed with copious amounts of H20 and MeOH and dried in vacuo to provide the above-titled compound 6 (33.1 g, 88 %), whose spectral data were identical to those of a sample of compound 6 that was obtained using Method I.
g) l~rep~x°nti0n 0f 9-chl~~ ~~ulf~nyl-5~~a-dihydx~-5-~~~-1 1H-grad~n~[1~2-c]i~~quin~line (7):

Compound 6 (40 g, 0.17 mol) was added in small portions to chlorosulfonic acid (112 mL, 1.71 mol) at 0°C and the reaction mixture was allowed to warm to room temperature and allowed to stir for 2 h. The reaction mixture was slowly poured on ice and the resulting yellow solid was filtered, washed thoroughly with water and EtOAc and dried i.ra vacuo to provide the above-titled product 7 (52 g, 92 %). 1H NMR
(DMSO-dG):
8 3.91 (s, 2H), 7.43 -7.48 (W , 1H), 7.60 (d, J= 7.2 Hz, 1H), 7.74 -7.76 (m, 2H), 7.79 (s, 1H), 7.90 (d, J= 7.5 Hz, IH), 8.23 (d, J= 7.8 Hz, 1H), Anal. Calcd. for Cl6HiaC1N04S:
C, 54.94; H, 3.46; N, 4.00. Found: C, 55.28; H, 3.43, N, 3.68, Karl-Fisher, 2.95.

h) Preparation of 9-sulphonamido derivatives of 5,6-dihydro-5-oxo-11H-indeno[1,2-c]isoquinolines (8a-af):
8a-of a.R=4-Methyl-piperazine-1-yl q. R=-N(CH2CH2NMez)z b.R=4-CHZCOZMe-piperazine-1-ylr. R=-N(CHZCHZOH)z c.R=4-CHzc;OzOH-piperazinP-1-yls. R=-NHCHzCHzCN

d.R=imidazole-1-yl t. R=-NHC(NH)NHz e.R=L-prolinol u. R=-NH[4-(1,2,4-triazole)]

f.R=morpholine-4-yl v. R=-NH[4-(N-morpholine)phenyl]

g.R=NHCHzCH2NMez w. R=-NHCHZCHz(4-N-benzylpiperidine) h.R=NHCHzCHz-piperidine-1-yl x. R=-NHCHzCHz(2-thienyl) i.R=NHCI-IzC;HzN-(pyridine-2-yl)y. R=-NH[i-(4-azabenzimidazole)]

j.R=NHCHzCHz-morpholine-4-yi z. R-= NH[i-(4-(2'-pyridyl)piperazine)]

k.R=NHCH2CHz-(2-N-Me-tetrahydropyrrolidine-1-aa. R= NHCHZCHZN[CHzCH20H]z yi) ab. R=-NH[1-(4-benzlpiperazine)]

I.R=NHCH2CH2CHz-morpholine-4-ylac. R=-NHz m.R=NHCHzCHzCHz-(tetrahydropyrrolidine-1-yl)ad. R=-NHCHzCHzPh n.R=NHCHzCH2CHz-imidazole-1-ylae. R=-NHCH2CHz[4-OMe(phenyl)]

o.R=NHCHZCHzCHz-(4-methylpiperazine-1-yl)af. R= NHC(O)(N-morpholine) p.R=N(CH2CHzNEtz)z Method I: To a stirred suspension of 3-(4-morpholino)-1-propylamine (17.28 g, 0.12 mol) in EtOAc was added sat. aq. NaHCO3 (300 mL), and the mixture was allowed to stir for 15 min. Compound 7 (4.0 g, 0.012 mol) was then introduced in small portions at room temperature. The reaction mixture was stirred for 24 h; filtered and washed with H20, EtOAc and MeOH; refluxed in MeOH for 30 min; filtered while still warm;
and washed with MeOH to provide compound 81 as a free base (2.33 g, 44 %). 1H
NMR(DMSO-d6): ~ 1.47-1.52 (m, 2H), 2.16-2.21 (m, 4H), 2.47-2.48 (m, 2I3), 3.44-3.48 (m, 2H), 3.23 (m, 4H), 4.02 (s, 2H), 7.49 -7.58 (m,1H), 7.78-7.82 (m, 3H), 7.97 (s, 1H), 8.14 (d, J= 7.8 Hz, 1H), 8.26 (d, ,l= 7.8 Hz, 1H), 9.59 (s, 1H), 12.42 (s, 1H).
The free bases of ~d, 8g, ~h, 8j, 81, 8m-8r were also prepared by Method I, but substituting 3-(4-morpholino)-1-propylamine with imidazole, 2-dimethylamino-ethylamine, 2-(N piperidinyl)-ethylamine, 2-(N morpholinyl)-ethylamine, 3-(N moipholinyl)propylamine, 3-(N tetrahydropyrrolidinyl)-propylamine, 3-(N imidazolyl)-propylamine, 3(N (4-methylpiperazinyl)-propylamine, di-(2-(diethylamino)-ethyl)amine, di-(2(dimethylamino)-ethyl)amine and di-(2-hydroxyethyl)amine, respectively.
Method II: To a stirred suspension of 3-(4-morpholino)-1-propylamine (4.250 g) in CH~Ch (100 mLj was added 7 (:1.950 g, 5.89 mnol) and the resulting mixture was stirred for 5 minutes. Subsequently, triethylamine (3 mL) was added and the reaction mixture was stiiTed for 24 h at room temperature. After this time the precipitate was collected and washed with MeOH (2 x100 mL) and the crude solid product transferred to a round bottom flask. This material was diluted with MeOH (200 mL), heated to reflux for 30 min. and filtered while still warm. The resulting filtercake was washed with MeOH (200 mL) to provide the desired product as the free base of 81 ( 1.460 g, 56 % j.
The free bases of compounds 8a-r were prepared using Method II, but substituting 3(4-moipholino)-1-propylamine with about an equivalent amount of imidazole, 2-dimet_hylamino-ethylam~ne, 2-(N piperidinyl)-ethylam~ne, 2-(N moipholinyl)-ethylamine, 3(N-morpholinyl)-propylamine, 3-(N tetrahydropynolidinyl)-propylamine, 3-(N imidazolyl)propylamine, 3-(N-(4-methylpiperazinyl) propylamine, di-(2-(diethylamino)-ethyl)amine, di(2-(dimethylamino)-ethyl)amine and di-(2-hydroxyethyl)amine, respectively.
k) Preparation of the mesylate salt of 81:
Free base 81 (l.Og) was added to methanesulfonic acid (10 mL) at 0°C
and the resulting mixture was allowed to warm to room temperature and then stirred for 2 h. The reaction mixture was then poured into cold MeOH (100 mL, between -10°C
and 0°C) and the precipitated solid was filtered, washed with MeOH (100 mL) and dried LTl vacLlo. The dried solid was then dissolved in water (100 mL), filtered and lyophilized to provide the methanesulfonate monohydrate salt 81. (1.020 g, 84 %). 1H NMR (DMSO-d~): 8 1~.75-1.85 (m, 2H), 2.35 (s, 3H), 2.78-2.84 (m, 2H), 2.96-3.12 (m, 4H), 3.36 (d, J =
12.3 Hz, 2H), 3.61 (t, J = 11.4 Hz, 2H), 3.94 (d, J = 12.9 Hz, 2H), 4.03 (s, 2H), 7.49 -7.55 (m, 1 H), 7.76-7.84 (m, 3H), 7.99 (d, J = 0.9 Hz, 1H), 8.15 (d, J = 8.4 Hz, 1H), 8.25 (d, J = 8.4~ Hz, 1H), 9.59 (s, 1H), 12.42 (s, 1H); 13C NMR (DMSO-dG): b 24.27, 33.86, 51.89, 54.51, 64.02, 119.70, 120.39, 123.53, 126.09, 126.45, 128.63, 133.66, 135.80, 138.71, 141.21, 144..57, 163.299 Anal. Calcd for CL~H3iN30~S~: C> 52.06; H, 5.4.6; N, 7.59, Karl-Fisher, 3.36. Found: C, 51.85; H, 5.35, N, 7.30, Karl-Fisher, 4.32.

Similarly, HCl, H2S04, CH3COOH, and succinic acid salts of 81 were prepared by substituting methanesulfonic acid with about an equivalent amount of HCl, H2SO4 and CH3COOH, respectively.
1) Preparation of 5?6-dihydro-~-o~~o-11H-indeno[1%2-a]i~oquinoline (13a) 13a To a solution of homophthalic anhydride (324 mg, 2.0 mmol) in acetonitrile (15 mL) was added 2-cyanobenzyl bromide (431 mg, 2.0 mmol, 1.0 eq) and triethylamine (5 ml~). The reaction was stirred under inert atmosphere at room temperature for minutes, after which time a yellow precipitate appeared. The reaction mixture was then heated at reflex for 18 h and the resulting white precipitate was filtered, washed using acetonitrile (3 x 8 mL) and dried under vacuum to provide Compound 13a as a white crystalline solid. Yield = 150 mg (32 %).
m) Preparation of a-Bromodimethylhomophthalate (20a) I?imethylhomophthalate (19a) (83.1 g) was dissolved in dichloromethane (2 L) and N-bromosuccinimide (121 g, 1.7 eq) was added. The resulting suspension was iiTadiated for 18 h with a 500 Watt quartz-halogen lamp, which brought the reaction mixture to reflex. The reaction mixture was then washed sequentially with saturated aqueous sodium bicarbonate (4 L), saturated aqueous sodium bisulfite (2 L), and saturated aqueous sodium chloride (2 L). The organic phase was dried using sodium sulfate with a small amount of silica added to remove polar impurities. The organic phase was filtered and concentrated ira vcreuo to provide Compound 20a as a dark orange oil.
Yield = 120.3 g ( 100 ~~).

n) Preparation of 8-Methoxy-6H 11-oxa-6-aza-benzo[a]fluoren-5-one (22a) a-Bromodimethylhomophthalate (20a) (1.16 g) and 2-hydroxy-5 -methoxybenzonitrile (0.6 g, 4 mmol, 1 eq) were dissolved by wainning in acetonitrile (6 mL). Triethylamine (5.6 mL, 10 eq) was then added and the reaction was heated at reflex for 48 h under inert atmosphere, then cooled to room temperature. The reaction mixture was diluted with saturated sodium bicarbonate (40 mL) and the resulting suspension was allowed to stir for 2 h, and was then filtered. The filtercake was washed sequentially with 1 N HCl (2 x 50 mL), acetonitrile (2 x 50 mL) and dichloromethane (50 mL), then dried in a vacuum oven at 50°C for three days to provide Compound 2,2a as an white solid.
Yield = 0.81 g (76 %).
o) Preparation of 8-Hydroxy-6H-11-oxa-6-aza-benzo[a]fluoren-5-one (23a) 8-Methoxy-6H-11-oxa-6-aza-benzo[a.]fluoren-5-one (22a) (5.0 g) was cooled using an ice bath, and boron tribromide (1 M in methylene chloride, 95 mL, 95 mmol, 5 eq) added in a steady stream under nitrogen. The reaction was heated at reflex under inert atmosphere for two hours, then cooled to room temperature and poured into water ( 150 mL). The resulting suspension was allowed to stir for 1. h, filtered, and the solids were washed with water (2 x 200 mL). The solids were then diluted with 5 N
sodium hydroxide (600 mL) using heating. The resulting solution was cooled to 0°C using an ice bath and the solution was acidified to pH 1 using conc. HCI. The resulting precipitate was vacuum filtered, and the solids washed sequentially with water (3 x 300 mL) and diethyl ether (300 mL) then dried overnight using a vacuum oven at 50°C
to provide Compound 23a as a gray solid. Yield 4.74 g (100 %).
p) Preparation of 3-Nitroso-2-Phenyindole (28) A solution of 2-phenylindole (27) (25 gm, 0.129 mol) in acetic acid (250 mL) was cooled to 18°C and a solution of sodium nitrite (8 g, 0.115 mol) in water (10 mL) was added dropwise while keeping the temperature of the reaction at ca.
20°C. The resulting reaction was stirred for 30 min at room temperature then diluted with ice water (250 mL).
The reaction mixtm°e was was filtered and the solid was washed with water then recrystallized using methanol to provide Compound 28. Yield = 27.5 gm (96.4 %). ES-MS: 223.22 (M++lj; NMIZ (DMS~-d6): ~ 7.0 (m,lH), 7.1 (m, 1H), 7.22 (m, 1H), 7.32 (m, 2H), 7.40 (m, 1H), 7.48 (m, 2H), 7.60 (m,lH).

q) Preparation of 3-Amino-2-Phenylindole (29) To a solution of 3-nitroso-2-phenyl indole (28) (25 gm, 0.129 mol) in ethanol (450 ml) was added 2N sodium hydroxide (300 mL, 5.0 eq) followed by sodium dithionite (38 g). The reaction was heated at reflex for 5 h, then filtered. The solid was washed with water and dried under vacuum to provide Compound 29 as a yellov~r solid. Yield = 15 g (72.1 %). ES-MS: 209.25 (M++ 1); NMR (DMSO-d6): b 7.0 (m, 1H), 7.1 (m, 1H), 7.22 (m, 1H), 7.32 (m, 2H), 7.40 (m, 1H), 7.48 (m, 2H), 7.60 (m, 1H).
r) Preparation of 2-Phenylindole-3-ethylcarlaarnate (30) To a 0 'C solution of 3-amino-2-phenylindole (29) (1.7 g, 8.17 mmol) in dichloromethane (150 ml) was added triethylamine (5 mL, 4..5 eq) followed by ethyl chloroformate (1 mL). The reaction was allowed to stir for 15 h, after which time the reaction mixture was diluted with water and transferred to a separatory funnel. The dichloromethane (50 mL), washed with water (2 x 50 mL), brine (50 mL) and dried over sodium sulfate. The solvent was removed and dried under vacuum to provide Compound .
30 as a black solid (1.6 gm, 72.7 %). ES-MS: 281.25 (M++1); NMR (DMSO-d6): ~
1.30 (t,3H), 4.1.2 (t, 2H), 7.0 (m, 1H), 7.1 (m, 1H), 7.22 (m,2H), 7.32 (m, 2H), 7.40(m,lH), 7.48 (m, 2H), 7.60 (m, 1H).
s) Preparation of 6H,11H-Indolo[3,2-c~Isoquinoline-5-one (31).
A solution of 2-Phenylindole-3-aminoethylcarbamate (30) (1.4 g, S mmol) in diphenyl ether (10 ml) was heated at reflex for 4 h, then cooled to room temperature. The reaction mixture was filtered and the solid was washed sequentially using warm hexane and warm dichloromethane and dried under vacuum to provide Compound 31 as a gray solid. Yield = 1.6 g (72.7 %). ES-MS: 235.25 (M++1); NMR (DMSO-d~): 8 7.1 (t, 1H), 7.25 (t, 1H), 7.50 (m, 2H), 7.82 (t, 1H), 8.0 (d, I H), 8.14(d, 1H), 8.32 (t,l H), 11.7(s, IH), 12.2 (s,lH).
t) Preparation of 6H,11H-Indolo[392-c)I~oquinoline-5-one-9,11-diacetate (32).
To a 0°C solution of 6H,11H-Indolo[3,2-c]Isoquinoh ne-5-one (31) (117 mg, 0.5 mmol) in dichloromethane ( 10 mL) was added triethylamine (2 mL, 30 eq) followed by acetic anhydride (1.8 mL, 35 eq). The reaction was stirred at room temperature for 48 h, then poured over ice and extracted with dichloromethane (100 mL). The dichloromethane layer was washed sequentially using water (2 x 20 mL) and brine (25 mL), then dried using sodium sulfate and concentrated ih vacuo. The resulting solid residue was dried under vacuum to provide Compound 32 as a brown solid. Yield = 180 mg, 83.7 %. ES-MS: 430.57 (M++1).
u) Preparation of 6H,11H-Indolo[3,2-c]Isoqxainoline-5-one-9,11-di~ulfonylchloride (33).
Compound 31 ( 117 mg, 0.5 mmol) was added to chlorosulfonic acid (2 mL,, 60 eq) and the resulting reaction mixture was allowed to stir at room temperature for 4 hours, after which time the reaction mixture was poured over ice. The resulting precipitate was filtered, washed sequentially with water and ethyl acetate and dried under vacuum to provide Compound 33 as a light-yellow solid. Yield = 180 mg (83.7 %). ES-MS:
430.57 (M++1); NMR (DMSO-d~): 8 7.1 (t, 1H), 7.25 (t, 1H), 7.50 (m, 2H), 7.82 (t, 1H), 8.0 (d, 1H), 8.14(d, 1H), 8.32 (t, 1H), 11.7(s, 1H), 12.2 (s, 1H).
v) Preparation of 6H,11H-Indolo[3,2-c]Isoquinoline-5-one-9,11-disulfonamide (35a).
To a solution of 33 (215 mg, 0.5 mmol) in methanol (10 mL) at 0°C was added a % solution of ammonia in methanol (10 mL). The reaction mixture was allowed to stir at room temperature for 15 hours and was then filtered. The resulting solid was washed with methanol and the dried under vacuum to provide Compound 35a as a yellow solid.
20 Yield = 140 mg , 71.4 %). ES-MS: 392.81 (M++1).
w) Preparation of N-acetylantliranilonitrile (36a) NHC(O)CH3 36a To a solution of anthranilonitrile (4.0 g, 32 mmol) in acetic anhydride ( 18 mL, 5.5 eq) at 90°C was added 1 drop of sulfuric acid and the resulting reaction was stirred at 90°C for 2 h, then allowed to sit at room temperatuaw for 12 h. The reaction mixture was poured onto ice (ca. 200 mL) and the resulting solution was stirred for 2 h, after which time the solution was neutralized to pH 7.0 using 5 N sodium hydroxide. The resulting precipitate was filtered, washed using water (4 x 50 mL) and dried under vacuum for 72 h to provide Compound 36a as a white crystalline solid. Yield = 1.07 g (16 %).
Fx~para~i~n 0f 6~aglue-ind01~[3,2-c]ls0quan~hn-5-One (37a) 37a Front. a-Bromodimetlzylhofr~ophthalate a-Bromodimethylhomophthalate (20a) (603 mg, 2.1 mmol) and N-acetylanthranilonitrile (36a) (370 mg, 1.1 eq) were dissolved in DMF (5 mL) under inert atmosphere. Potassium carbonate (1.45 g, 5.0 eq) was added and the reaction was stirred for 48 h at 100°C, then cooled to room temperature. The reaction mixture was poured into 1 N sodium hydroxide and the resulting mixture was extracted with EtOAc (50 mL).
The EtOAc layer was washed sequentially with 1N HCl (50 mL), saturated aqueous sodium chloride (50 mL), dried over sodium sulfate, filtered and concentrated iya vacicc~.
The resulting residue was dissolved by warming in toluene (70 mL) and the solution was cooled to room temperature and upon addition of hexanes (200 mL), a solid precipitate appeared. The solid precipitate was filtered, washed using hexanes (50 mL) and dried in a vacuum oven at 50°C for 72 h to provide Compound 37a as a yellow powder. Yield =
33 mg (6.7 %).

y) Preparation of 6H,11H this-6-aza-benzo[a]fluorene-5-one (40a) O
4.Oa F~-orra hon2ophthali.c an.7aydi°ide:
A solution of 2-mercaptobenzonitrile (39a) (1.35 g, 10 mmol) and homophthalic anr~ydride (11a) (1.~ g, 10.0 mmol, 1.0 eq) in acetonitrile (150 mL) under inert . .
atmosphere was warmed with stirring until all reactants were in solution.
Triethylamine (6.9 mL, 50 mmol, 5.0 eq) was added and the reaction was heated at reflux for 72 hours, then cooled to room temperature. After cooling, the reaction mixture was filtered, and the collected solid was washed using methanol (3 x 50 mL), then dried in a vacuum oven at 50°C to provide Compound 40a as a white solid. Yield = 225 mg (9 070).
From a-b~o~r2odimethyl7~of~zoplztlzalate:
A solution of 2-mercaptobenzonitrile (39a) (1.35 g, 10 mmol) and a-bromodimethylhomophthalate (20a) (2.87 g, 10.0 mmol, 1.0 eq) in acetonitrile (150 mL) under inert atmosphere was warmed with stirring until all reactants were in solution.
Triethylamine (6.9 mL, 50 mmol, 5.0 eq) was added and the reaction was heated at reflux for 72 hours, then cooled to room temperature. After cooling, the reaction mixture was filtered, and the collected solid was washed using methanol (3 x 50 mL), then dried in a vacuum oven at 50°C to provide Compound 40a as a white solid. Yield =
250 mg (10 ~'/o).
E~arnple 2. lEffect of fLllustrative lsoquinoline Derivatives on PAff~~
aetivity ia1 eultured anacrophages, using a whole-sell based assay and a purified enzyme assay.
Demonstration of illustrative Isoquinoline Derivatives' ability to inhibit PARS
and prevent peroxynitrite induced cytotoxicity was shown using methods described in Virag et al., Br. J. Pharrrzacol. 1999, 126(3):769-77; and Immunology 1998, 94(3):345-55. RAW mouse macrophages were cultured in DMEM medium with high glucose and supplemented with 10 % fetal bovine serum. Cells were used at 80 % confluence in 12-well plates. Cells were pretreated with various concentrations (100 nM -1 gM) of an Isoquinoline Derivative for 10 min. Peroxynitrite, a prototypical oxidant which induces DNA single strand breakage, was used to induce PARS activation. Peroxynitrite was diluted in phosphate buffered saline (PBS) (ppI 11.0) and added to the cells in a bolus of 50 pl. Cells were then incubated for 20 min. Peroxynitrite was decomposed by incubation for 30 min at pH 7.0, used as a control, and failed to influence the parameter studied. After the 20 min incubation, the cells were spun, the medium was aspirated and the cells were resuspended in 0.5 ml assay buffer (56 mM HEPES pH 7.5, 28 mM
KCI, 28 mM NaCI, 2 mM MgCl2, 0.01 % w/v digitonin and 0.125 ~,M NAD+ and 0.5 pCi/ml 3H-NAD+). Following an incubation in assay buffer, (10 min at 37°C), PARS activity ~~~as measured as follows: 200 ~1 ice cold 50 % u~lv TCA was added and the samples, were incubated for 4 hours at 4°C. Samples were then spun (10 min @
10,000 g) and pellets washed twice with ice cold 5 % w/v TCA and solubilized overnight in 250 ~,l 2 %
w/v SDSJ0.1 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 detemnined using a liquid scintillation counter (Wallac, Gaithersburg, MD). The results shown in Table 1 demonstrate that the illustrative Isoquinoline Derivatives significantly and dose-dependently inhibit the activation of PARS in the macrophage assay.
Table 1. Inhibitory effect of various novel substituted isoquinolines on PARE
activation in cultured murine macrophages.
Compound No. % PARS inhibition% PARS inhibition% PARE inhibition at 1 ~,M at 300 nM at 100 nM

3 a 67 NT 8 3b 25 0 NT

3c 21 9 NT

4.b 88 NT 51 5a 55 NT 10 Compound No. % PARS inhibition% PARS inhibition% PARS inhibition at 1 ~.M at 300 nM at 100 nM

5b 33 NT 0 Sc 24 NT 0 5d 48 NT 0 5e 21 NT 0 8a NT 47 NT

8c NT 27 NT

8d NT 82 77 8e NT 68 NT

8g NT 55 34 8h NT 76 56 8j NT 76 34 8k NT 38 24 8m NT 50 NT

8n NT 82 74 8o NT 55 ' 48 8p NT 45 27 8q NT 28 20 8r NT 28 20 8s 54. NT 30 8t 29 NT 17 8u NT NT 59 Compound No. % PARS inhibition% PARS inhibition% PARS inhibition at 1 ~M at 300 nM at 100 nM

8w NT NT 69 8x NT NT 54 8~ NT NT 67 8aa NT NT 64 8ab NT NT 49 Bag 59 NT 35 Bah 63 NT 67 8 ai 90 NT 69 8ak NT 22k 8=~

8a1 84 NT 49 Sam NT NT ~ 65v 8 an 40=~ NT 40 '~

Sao 60 NT 40 l0a NT 59 55 10b NT 17 17 22a 81 NT 51 22b NT 20'r 12~' 22c 83 66 62 22d 13 ~' NT NT

22e 53 56 38 22f 27 23 NT

22b 27 23 NT

23a 84 79 34 Compound No. ~ PARS inhibition% PARS inhibition7o PARS inhibition at 1 ~.M at 300 nM at 100 nM

23b 58 57 53 23c 63 66 63 25a 51 57 53 25b 40 29 25 25c 58 34 23 25d 67 66 53 25e 58 63 40 26a 90 74 51 26b 51 ~~ 29 * 21 a' 34 NT 33 ~' 14*
35a 75 55 14 35b 42 51 25 NT - Not Tested tested in purified enzyme assay The potency of inhibition on purified PARS enzyme was subsequently determined for selected Isoquinoline Derivatives, and the potency was compared with that of 3-aminobenzamide, a prototypical benchmark PARS inhibitor. The assay was performed in 96 well ELISA plates according to instructions provided with a commercially available PARS inhibition assay lit (Trevigen, Gaithersburg, MD). Briefly, wells were coated with 1 mg/mL histone (50 ~,1/well) at 4°C overnight. Plates were then washed four times with . PBS and then blocked by adding 50 ~,1 Strep-Diluent (supplied with the ki.t). After incubation (lh, room temperature), the plates were washed four times with PBS.
Appropriate dilutions of PARS inhibitors were combined with 2x PARS cocktail (1.95 mI~I NAD+, 50 ~.M
biotinylated NAD+ in 50 mM TRIS pIi 8.0, 25 mM MgCl2) and high specific activity PARE enzyme (both were supplied with the kit) in a volume of 50 ~,1. The reaction was allowed to proceed for 30 min at room temperature. After 4 washes in PBS, incorporated biotin was detected by peroxidase-conjugated streptavidin (1:500 dilution) and TACS
Sapphire substrate. The assay confirmed the results of the macrophage-based PARS
assay. For example, the PARS inhibitor 81 exerted 50 % inhibition of PARS
activity in this assay at 3 nM, and thus was approximately 50,000 times more potent than the reference compound 3-aminobenzannide.
E~arnple ~: Effects 0f illu~txative Tl~Oquin~line Derivatives in vai i0us rn~del~ 0f inflaxnrnat0ry disease and xeperfusi0n disease a: Effects of illustrative Isoquinoline Derivatives on i~z vitz-~ cell disease models In additional izz vitra studies in isolated thymocytes, cells were exposed to peroxynitrite or hydrogen peroxide (toxic oxidant species) to induce cytotoxicity. In this system the toxicity is, at least in part, related to activation of the nuclear enzyme PARE.
In this oxidant-stimulated thymocyte assay (described, in detail, in Virag et al., Irzzzzzuzzology 94(3):345-55, 1998), the compounds tested prevented the oxidant-induced suppression of the viability of the cells and did so at the low nanomolar concentration range. An example of this response (Compound 81) is shown in Table 2. This assay represents an in vitro model of cells dying because of exposure to pro-oxidant species, as it occurs in during the reperfusion of ischerilic organs.
Table 2. Reduction of peroxynitrite induced cytotoxicity by 30 nM - 3 ~,M of the Isoquinoline Derivative 81.
+81 +81 +81 +81 +81 Control 30 nM 100 nM 300 nM 1 p,M 3 pM

Cytotoxicity98 % 74 % 39 % 2 % 0 % 0 %

b: Effect of illustrative Isoquinoline Derivatives on iu vivo models of inflammatory diseases In order to substantiate the efficacy of the compounds in inflammatory diseases, the effect of illustrative Isoquinoline Derivatives was demonstrated in a systemic inflammatory model induced by bacterial lipopolysaccharide (LPS), which is reported to be responsible for causing reperfusion diseases and inflammatory diseases such as septic shock and systemic inflammatory response syndrome in animals (see Parrillo, N.
Ezzgl. J.
Med., 328:1471-1478 (1993) and Lumping, J. Clip. Izzvest. 101:2065-2071 (1998). In a series of experiments, mice were pretreated with intraperitoneal injection of 0.1 and 1 mg/kg of compounds 81, 8p and 8j, and LPS at 10 mg/kg was injected i.p., and TNF-alpha was measured in the plasma at 90 minutes. As shown in Table 3, all compounds substantially reduced TNF production, indicative of the compounds' anti-inflammatory activity.
Table 3. Reduction of LPS induced TNF production by 0.1-1 mg/kg intraperitoneal injection of the PARE inhibitor compounds 81, 8p and 8j in mice iaz vivo 8j (0.1 8j ( 8p (0.18p ( 81 (0.1 81 ( 1.0 1.0 1.0 vehicle mg/kg) mg/kg) mg/kg) mg/kg) mg/kg) mg/kg) TNF 3831.6 5038.8 4470.0 5090.8 3714.6 3509.8 6994.0 (ng/ml) 385.2 377.1 184.4 203.7 300.9 311.5 904.4 All compounds markedly suppressed LPS induced TNF production when compared to control.
At bigh doses, LPS causes multiple organ dysfunction resembling of septic shock, and ultimately death (in part because of the early release of TNF-alpha).
Similarly, in a model induced by cecal ligation and puncture (CLP), the live bacteria that derive from the intestinal flora induce systemic inflammation and shock. Agents that inhibit inflammatory mediator production, PARS activation, and cell death in this model prevent mortality induced by LPS or CLP. In experiments with Balb/c mice, injection of mglkg LPS intraperitoneally caused death in 50 °~o of the animals over 24 h, whereas treatment of the animals with 3 mg/kg/day of compound 81 reduced the endotoxin-induced mortality to 10 % under the same experimental conditions. In response to CLP
induced shock, compound 81 (3 mg/kg/day) caused a reduction in the mortality from 100 °lo death to 60 % death over 24 h.
The data demonstrating the reduction of TNF production by illustrative Isoquinoline Derivatives in animals subjected to an inflammation model, coupled with the fact that TNF production is an important trigger of inflammation in various inflammatory diseases (such as, for example, colitis, arthritis and neuroinflammation and shock) indicate that the Isoquinoline Derivatives have therapeutic effects in various systemic and local inflammatory diseases, including the rejection of transplanted organs, which entails both an inflannnatory disease component and a reperfusion disease component and, accordingly, are useful for treating or preventing an inflammatory disease or a reperfusion disease.
c: Effect of illustrative Isoquinoline Derivatives on ira vivo models of reperfusion disease In order to substantiate the efficacy of the Isoquinoline Derivatives in ischemia-reperfusion conditions, the effect of an illustrative Isoquinoline Derivative in a m~use model of ischemic and reperfused gut was tested. The superior mesenteric artery was occluded for 45 min, followed by a reperfusion for 1 h. Following the end of the reperfusion, gut permeability was measured with the FD4 method in evened gut sacks (Liaudet et al., Sh~ck 2000, 14(2):134-41). Ischemia-reperfusion increased the I O permeability of the gut from 11 ~ 4 to 216 ~ 27 ml/miz~/cm', indicative of severe damage of the reperfused gut. Treatment with Compound 81 (3 mg/kg i.v., injected 10 min prior to initiation of reperfusion) reduced the increase in the permeability of the gut by approximately 73 %, indicating a marked maintenance of the gut function. The ischemia-reperfusion studies in the gut were associated with a 80 % mortality over 12.
hours, whereas only 15 % mortality was noted in the animals treated with 81.
In another set of experiments, the effect of Compound 81 in a rat model of middle cerebral artery occlusion/reperfusion was assayed as described in ~bdellcarim et al., Int zl Mol Med. 2001, 7(3):255-60. Occlusion lasted for 2 hours, followed by reperfusion for 24 hours. Infarct size was quantified with tetrazolium staining. Compound 81 was administered at 3 mg/kg/day in 3 divided intraperitoneally injected doses, the first dose being administered 10 min prior to the initiation of reperfusion. There was an approximately 80 % reduction in the degree of cortical necrosis and neuronal death in the animals administered with 81, when compared to vehicle-treated controls. This protection also translated into functional benefit, such as neurological improvements in the PARS
inhibitor treated group.
These data indicate that the Isoquinoline Derivatives have therapeutic effects in various systemic and local conditions of reperfusion diseases, including the rejection of transplanted organs, which entails both an inflammatory disease component and a reperfusion disease component and, accordingly, are useful for treating or preventing an inflammatory disease or a repe.rfusion disease.
d: Effect of illustrative Isoquinoline Derivatives in a diabetes model PARS inhibitors and PARE deficiency are known to reduce the development of diabetes and the incidence of diabetic complications (Mabley et al., Br J
Plzarsyaacol.
2001, 133(6):909-9; and Soriano et al., Nat Med. 2001, 7(1):108-13). In order to substantiate the efficacy of the Isoquinoline Derivatives in a diabetes model, a single high-dose streptozotocin model of diabetes was conducted as previously described.
Briefly, 160 mg/kg stropto~otooin was injected to mice treated with vehicle or with illustrative Isoquinoline Derivatives intraperitoneally (3 mglpg) and 3 days later blood sugar levels were determined using a blood glucose meter. The data shown in Table 4 demonstrate that the illustrative Isoquinoline Derivatives attenuate the streptozotocin-induced onset of diabetes as they reduce the hyperglycemia.
Table 4. Reduction of streptozotocin (STZ) induced hyperglycemia by 3 mg/kg intraperitoneal injection of the PARE inhibitor compounds 81, 8p and 8j in mice ira vavo Basal STZ + Vehicle STZ + 8j STZ + 8p 81 Glucose (mg/ml) Accordingly, the Isoquinoline Derivatives are useful for treating or preventintr diabetes or a diabetic complication.
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 spilled 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 (23)

1. A compound of the formula or a pharmaceutically acceptable hydrate or salt thereof, wherein:
R5 is O, NH or S;
R6 is -H or C1-C4 alkyl;
Y is -OH, -NH2 or alkylheterocycle;
R1 is -hydrogen, -halo, -C1-C10 alkyl, -alkylhalo, -C2-C10 alkenyl, -C3-C8 carbocycle, -aryl, -NH2, -alkylamino, -C(O)OH, -C(O)O(C1-C5 alkyl), NO2 or -A'-B';
A' is -SO2-, -SO2NH-, -NHCO-, -NHCONH-, -CO-, -C(O)O-, -CONH-, -CON(C1-C4 alkyl)-, -NH-, -CH2-, -S- or -C(S)-;
B' is -C1-C10 alkyl, -C2 C10 alkenyl, -heterocycle, -C3-C8 carbocycle, -aryl, -NZ1Z2, -(C1-C5 alkylene)-NZ1Z2, -alkylamino, -aminodialkyl, -alkylheterocycle, -arylamido, -C(O)OH, -C(O)O-(C1-C5 alkyl), -C(O)O-phenyl or -C(NH)NH2, each of which is unsubstituted or substituted with one or more of -O-(C1-C5 alkyl), -halo, -alkylhalo, -alkanol, -alkylamino, -hydroxy, -NO2, -NH2, -CN, -aminoalkyl, -aminodialkyl, -heterocyclic amine, -C1-C10 alkyl, -C2-C10) alkenyl, -C2-C10 alkynyl, -aryl, -benzyl, -alkylamido, -alkylcarboxy, -C(O)OH, -C1-C5 alkylene-C(O)O-(C1-alkyl) or -C1-C5 alkylene-OC(O)(C1-C5 alkyl);
R2, R3 R4, R7, R8, R9 and R10 are independently -hydrogen, -halo, -hydroxy, -O-(C1-C5 alkyl), -C1-C10 alkyl, -alkylhalo, -C2-C10 alkenyl, -C3-C8 carbocycle, -aryl, -NH2, -alkylamino, -C(O)OH, -C(O)O(C1-C5 alkyl), -OC(O)(C1-C5 alkyl), NO2 or -A-B;
and at least one of R1, R2, R3, R4, R7, R8, R9 or R10 is other than hydrogen;

A is -SO2-, -SO2NH-, -NHCO-, -NHCONH-, -O-, -CO-, -OC(O)-, -C(O)O-, -CONH-, -CON(C1-C4 alkyl)-, -NH-, -CH2-, -S- or -C(S)-;
B is -C1-C10 alkyl, -C2-C10 alkenyl, -heterocycle, -C3-C8 carbocycle, -aryl, -NZ1Z2, -(C1-C5 alkylene)-NZ1Z2, -alkylamino, -aminodialkyl, -alkylheterocycle, -arylamido, -C(O)OH, -C(O)O-(C1-C5 alkyl), -C(O)O-phenyl or -C(NH)NH2, each of which is unsubstituted or substituted with one or more of -O-(C1-C5 alkyl), -halo, -alkylhalo, -alkanol, -alkylamino, -hydroxy, -NO2, -NH2, -CN, -aminoalkyl, -aminodialkyl, -heterocyclic amine, -C1-C10 alkyl, -C2-C10 alkenyl, -C2-C10 alkynyl, -aryl, -benzyl, -alkylamido, -alkylcarboxy, -C(O)OH, -C1-C5 alkylene-C(O)O-(C1-C5 alkyl) or -alkylene-OC(O)-(C1-C5 alkyl); and Z1 and Z2 are independently -H or -C1-C10 alkyl, which is unsubstituted or substituted with one or more of -halo, -OH or -N(Z3)(Z4), where Z3 and Z4 are independently, -H or -C1-C5 alkyl, which is unsubstituted or substituted with one or more of -halo, -hydroxy or -NH2; or N, Z3 and Z4 are taken together to form a heterocyclic amine; or N, Z1 and Z2 are taken together to form a heterocyclic amine.
2. The compound of claim 1 wherein R5 is O, and R6 is hydrogen.
3. The compound of claim 1, wherein either R8 or R9, but not both, is -A-B.
4. The compound of claim 3 wherein R1, R2, R3, R4, R7, and R10 are hydrogen.
5. The compound of claim 1, wherein R9 is -A-B.
6. The compound of claim 1, wherein R9 is -A-B and R8 is hydrogen.
7. The compound of claim 1, wherein A is -SO2-, -SO2NH- or -NHCO-.
8. The compound of claim 1, wherein R9 is -A-B and B is -C1-C10 alkyl, -heterocycle, -C3-C8 carbocycle, -aryl, -alkylamino or -alkylheterocycle.
9. The compound of claim 1, wherein A is -SO2- and B is -NZ1Z2.
10. The compound of claim 9, wherein B is a heterocycle, which is unsubstituted or substituted with methyl, ethyl or -alkanol.
11. The compound of claim 9, wherein Z1 is hydrogen; Z2 is -C2-C3 alkyl, which is substituted with -N(Z3)(Z4); and Z3 and Z4 are independently methyl or ethyl, or, taken together, NZ3Z4 form a heterocyclic amine.
12. A method for inhibiting poly(ADP)-ribose synthase activity in a cell, the method comprising contacting the cell with a compound or pharmaceutically acceptable hydrate or salt of a compound of claim 1 in an amount effective to inhibit poly (ADP)-ribose synthase in the cell.
13. The method of claim 12 wherein the cell is an in vivo cell.
14. A method for treating or preventing an inflammatory disease comprising administering to a subject in need thereof a compound or a pharmaceutically acceptable hydrate or salt of a compound of claim 1 in an amount effective to treat an inflammatory disease.
15. The method of claim 14, wherein 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, hemorrhagic shock, anaphylactic shock, traumatic shock or chemotherapeutic shock.
16. A method for treating a reperfusion disease, comprising administering to a subject in need thereof a compound or pharmaceutically acceptable hydrate or salt of a compound of claim 1 in an amount effective to treat a reperfusion disease.
17. The method of claim 16 wherein the reperfusion disease is stroke or myocardial infarction.
18. A method for treating diabetes or a diabetic complication, comprising administering to a subject in need thereof a compound or pharmaceutically acceptable hydrate or salt of a compound of claim 1 in an amount effective to treat diabetes or a diabetic complication.
19. A method for treating reoxygenation injury resulting from organ transplantation, comprising administering to a subject in need thereof a compound or pharmaceutically acceptable hydrate or salt of a compound of claim 1 in an amount effective to treat the reoxygenation injury.
20. A method for treating Parkinson's disease, comprising administering to a subject in need thereof a compound or pharmaceutically acceptable hydrate or salt of a compound of claim 1 in an amount effective to treat Parkinson's disease.
21. A method for treating Huntington's disease, comprising administering to a subject in need thereof a compound or pharmaceutically acceptable hydrate or salt of a compound of claim 1 in an amount effective to treat Huntington's disease.
22. A method for treating amyotrophic lateral sclerosis, the method comprising administering to a subject in need thereof a compound or pharmaceutically acceptable hydrate or salt of a compound of claim 1 in an amount effective to treat amyotrophic lateral sclerosis.
23. A composition comprising an effective amount of a compound or pharmaceutically acceptable salt or hydrate of the compound of claim 1 and a pharmaceutically acceptable carrier or vehicle.
CA002517358A 2003-02-28 2004-02-26 Isoquinoline derivatives and methods of use thereof Abandoned CA2517358A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US10/376,746 US6956035B2 (en) 2001-08-31 2003-02-28 Isoquinoline derivatives and methods of use thereof
US10/376,746 2003-02-28
PCT/US2004/005849 WO2004078712A2 (en) 2003-02-28 2004-02-26 Isoquinoline derivatives and methods of use thereof

Publications (1)

Publication Number Publication Date
CA2517358A1 true CA2517358A1 (en) 2004-09-16

Family

ID=32961227

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002517358A Abandoned CA2517358A1 (en) 2003-02-28 2004-02-26 Isoquinoline derivatives and methods of use thereof

Country Status (11)

Country Link
US (3) US6956035B2 (en)
EP (1) EP1603568A4 (en)
JP (1) JP2006520817A (en)
KR (1) KR20050118168A (en)
CN (2) CN100400515C (en)
AU (1) AU2004218023A1 (en)
BR (1) BRPI0407922A (en)
CA (1) CA2517358A1 (en)
PL (1) PL378294A1 (en)
RU (2) RU2005130159A (en)
WO (1) WO2004078712A2 (en)

Families Citing this family (50)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030096833A1 (en) * 2001-08-31 2003-05-22 Jagtap Prakash G. Substituted ideno[1,2-c]isoquinoline derivatives and methods of use thereof
US6956035B2 (en) * 2001-08-31 2005-10-18 Inotek Pharmaceuticals Corporation Isoquinoline derivatives and methods of use thereof
JP2007501857A (en) * 2003-02-28 2007-02-01 イノテック ファーマシューティカルズ コーポレーション Tetracyclic benzamide derivatives and methods of use thereof
US10425538B2 (en) * 2003-10-06 2019-09-24 Yellowpages.Com Llc Methods and apparatuses for advertisements on mobile devices for communication connections
US20050228007A1 (en) * 2004-02-26 2005-10-13 Prakash Jagtap Isoquinoline derivatives and methods of use thereof
RU2006134024A (en) * 2004-02-26 2008-04-10 Инотек Фармасьютикалз Корпорейшн (Us) TETRACYCLIC DERIVATIVES OF LACTAM AND THEIR USE
EP1784186A4 (en) * 2004-06-16 2008-05-14 Inotek Pharmaceuticals Corp Methods for treating or preventing erectile dysfunction or urinary incontinence
AU2006219023A1 (en) * 2005-02-25 2006-09-08 Inotek Pharmaceuticals Corporation Tetracyclic Amino and Carboxamido Compounds and methods of use thereof
WO2006093666A2 (en) * 2005-02-25 2006-09-08 Inotek Pharmaceuticals Corporation Isoqunoline compounds and methods of use thereof
JP2008531563A (en) * 2005-02-25 2008-08-14 イノテック ファーマシューティカルズ コーポレイション Tetracyclic sulfonamide compounds and methods of use thereof
WO2007008926A1 (en) * 2005-07-11 2007-01-18 Aerie Pharmaceuticals, Inc. Isoquinoline compounds
RU2008110955A (en) * 2005-08-24 2009-09-27 Инотек Фармасьютикалз Корпорейшн (Us) INDENOISOCHINOLINONE ANALOGUES AND WAYS OF THEIR APPLICATION
CA2664335C (en) 2006-09-20 2014-12-02 Boehringer Ingelheim International Gmbh Rho kinase inhibitors
US8455513B2 (en) 2007-01-10 2013-06-04 Aerie Pharmaceuticals, Inc. 6-aminoisoquinoline compounds
CA2677046A1 (en) * 2007-02-28 2008-09-04 Inotek Pharmaceuticals Corporation Indenoisoquinolinone analogs and methods of use thereof
US8455514B2 (en) * 2008-01-17 2013-06-04 Aerie Pharmaceuticals, Inc. 6-and 7-amino isoquinoline compounds and methods for making and using the same
US8450344B2 (en) 2008-07-25 2013-05-28 Aerie Pharmaceuticals, Inc. Beta- and gamma-amino-isoquinoline amide compounds and substituted benzamide compounds
CA2929545C (en) 2009-05-01 2019-04-09 Aerie Pharmaceuticals, Inc. Dual mechanism inhibitors for the treatment of disease
EP2569307A2 (en) 2010-05-10 2013-03-20 Radikal Therapeutics Inc. Lipoic acid and nitroxide derivatives and uses thereof
WO2013004652A1 (en) 2011-07-04 2013-01-10 Bayer Intellectual Property Gmbh Use of substituted isoquinolinones, isoquinolindiones, isoquinolintriones and dihydroisoquinolinones or in each case salts thereof as active agents against abiotic stress in plants
TWI519515B (en) 2011-12-21 2016-02-01 諾維拉治療公司 Hepatitis b antiviral agents
JP5929229B2 (en) * 2012-01-24 2016-06-01 東洋紡株式会社 Alkaline phosphatase
EA027280B1 (en) 2012-08-28 2017-07-31 Янссен Сайенсиз Айрлэнд Юси Sulfamoyl-arylamides and the use thereof as medicaments for the treatment of hepatitis b
CA2899706C (en) 2013-02-28 2021-10-19 Janssen Sciences Ireland Uc Sulfamoyl-arylamides and the use thereof as medicaments for the treatment of hepatitis b
US8993771B2 (en) 2013-03-12 2015-03-31 Novira Therapeutics, Inc. Hepatitis B antiviral agents
EP3461484B9 (en) 2013-03-15 2021-06-09 Aerie Pharmaceuticals, Inc. Dimesylate salts of 4-(3-amino-1-(isoquinolin-6-ylamino)-1-oxopropan-2-yl)benzyl, their combinations with prostaglandins and the use thereof in the treatment of ocular disorders
US9895349B2 (en) 2013-04-03 2018-02-20 Janssen Sciences Ireland Us N-phenyl-carboxamide derivatives and the use thereof as medicaments for the treatment of hepatitis B
JO3603B1 (en) 2013-05-17 2020-07-05 Janssen Sciences Ireland Uc Sulphamoylpyrrolamide derivatives and the use thereof as medicaments for the treatment of hepatitis b
BR112015028538A2 (en) 2013-05-17 2017-07-25 Janssen Sciences Ireland Uc sulfamoylthiophenamide derivatives and their use as medicines for the treatment of hepatitis b
NO3024819T3 (en) 2013-07-25 2018-07-21
CA2923712C (en) 2013-10-23 2021-11-02 Janssen Sciences Ireland Uc Carboxamide derivatives and the use thereof as medicaments for the treatment of hepatitis b
US10392349B2 (en) 2014-01-16 2019-08-27 Novira Therapeutics, Inc. Azepane derivatives and methods of treating hepatitis B infections
US9169212B2 (en) 2014-01-16 2015-10-27 Novira Therapeutics, Inc. Azepane derivatives and methods of treating hepatitis B infections
US9181288B2 (en) 2014-01-16 2015-11-10 Novira Therapeutics, Inc. Azepane derivatives and methods of treating hepatitis B infections
RU2702109C1 (en) 2014-02-05 2019-10-04 Новира Терапьютикс, Инк. Combined therapy for treating hepatitis b infections
CN110483484A (en) 2014-02-06 2019-11-22 爱尔兰詹森科学公司 Sulfamoyl pyrrole amides derivative and its purposes for being used to treat hepatitis B as drug
US9400280B2 (en) 2014-03-27 2016-07-26 Novira Therapeutics, Inc. Piperidine derivatives and methods of treating hepatitis B infections
CN107847762A (en) 2015-03-19 2018-03-27 诺维拉治疗公司 Azacyclooctane and azacyclo- nonane derivatives and the method for treating hepatitis B infection
US10875876B2 (en) 2015-07-02 2020-12-29 Janssen Sciences Ireland Uc Cyclized sulfamoylarylamide derivatives and the use thereof as medicaments for the treatment of hepatitis B
EP3356328A1 (en) 2015-09-29 2018-08-08 Novira Therapeutics, Inc. Crystalline forms of a hepatitis b antiviral agent
US9643927B1 (en) 2015-11-17 2017-05-09 Aerie Pharmaceuticals, Inc. Process for the preparation of kinase inhibitors and intermediates thereof
JP6832946B2 (en) 2015-11-17 2021-02-24 アエリエ ファーマシューティカルズ インコーポレイテッド How to prepare kinase inhibitors and their intermediates
EP3442524A2 (en) 2016-04-15 2019-02-20 Novira Therapeutics Inc. Combinations and methods comprising a capsid assembly inhibitor
BR112019003945A2 (en) 2016-08-31 2019-05-21 Aerie Pharmaceuticals, Inc. ophthalmic compositions
EP3567044A4 (en) * 2017-01-06 2020-06-10 Korea University Research and Business Foundation, Sejong Campus Novel quinolinone derivative and pharmaceutical composition for preventing or treating allergic disease such as asthma or atopy comprising same as active ingredient
CN110506037A (en) 2017-03-31 2019-11-26 爱瑞制药公司 Aryl cyclopropyl-amino-isoquinolin amide compound
US10973801B2 (en) 2018-03-14 2021-04-13 Janssen Sciences Ireland Unlimited Company Capsid assembly modulator dosing regimen
AU2019337703B2 (en) 2018-09-14 2023-02-02 Aerie Pharmaceuticals, Inc. Aryl cyclopropyl-amino-isoquinolinyl amide compounds
US11096931B2 (en) 2019-02-22 2021-08-24 Janssen Sciences Ireland Unlimited Company Amide derivatives useful in the treatment of HBV infection or HBV-induced diseases
US11491148B2 (en) 2019-05-06 2022-11-08 Janssen Sciences Ireland Unlimited Company Amide derivatives useful in the treatment of HBV infection or HBV-induced diseases

Family Cites Families (41)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US96833A (en) * 1869-11-16 Improvement in barrel-filling- apparatus with whistling indicators
US3710795A (en) * 1970-09-29 1973-01-16 Alza Corp Drug-delivery device with stretched, rate-controlling membrane
IT1054655B (en) * 1975-08-27 1981-11-30 Lepetit Spa CONDENSED DERIVATIVES OF L ISOKINOLINE
US4263304A (en) * 1978-06-05 1981-04-21 Sumitomo Chemical Company, Limited 7 H-indolo[2,3-c]isoquinolines
GB2025932B (en) 1978-06-13 1982-10-27 Sumitomo Chemical Co Indoloisoquinolines and processes for producing them
JPS57172923A (en) 1981-04-17 1982-10-25 Toray Ind Inc Production of polybutylene terephthalate polymer fine particle
GB8828806D0 (en) * 1988-12-09 1989-01-18 Beecham Group Plc Novel compounds
US5260316A (en) * 1991-07-30 1993-11-09 Ciba-Geigy Corporation Isoquinolyl substituted hydroxylamine derivatives
US5597831A (en) * 1991-08-29 1997-01-28 Vufb A.S 6-[X-(2-hydroxyethyl) aminoalkyl]-5,11-dioxo-5,6-dihydro-11-H-indeno[1,2-c]isoquinolines and their use as antineoplastic agents
CZ281473B6 (en) 1991-08-29 1996-10-16 Vúfb, A.S. 2-HYDROXYETHYLAMINO SUBSTITUTED DERIVATIVES OF 5,11-DIOXO-5,6-DIHYDRO-11H-INDENO£1,2-c|ISOQUINOLINE, PROCESS PROCESS OF THEIR PREPARATION, AND PHARMACEUTICAL COMPOSITION CONTAINING THEREOF
US5262564A (en) * 1992-10-30 1993-11-16 Octamer, Inc. Sulfinic acid adducts of organo nitroso compounds useful as retroviral inactivating agents anti-retroviral agents and anti-tumor agents
TW440562B (en) * 1994-05-20 2001-06-16 Taiho Pharmaceutical Co Ltd Condensed-indan derivative and pharmaceutically acceptable salts thereof
AU8784698A (en) 1997-08-15 1999-03-08 Johns Hopkins University, The Method of using selective parp inhibitors to prevent or treat neurotoxicity
US6250301B1 (en) 1997-08-28 2001-06-26 Hortal Harm B.V. Vaporizer for inhalation and method for extraction of active ingredients from a crude natural product or other matrix
US6346536B1 (en) * 1997-09-03 2002-02-12 Guilford Pharmaceuticals Inc. Poly(ADP-ribose) polymerase inhibitors and method for treating neural or cardiovascular tissue damage using the same
US20020028813A1 (en) 1997-09-03 2002-03-07 Paul F. Jackson Thioalkyl compounds, methods, and compositions for inhibiting parp activity
US6197785B1 (en) 1997-09-03 2001-03-06 Guilford Pharmaceuticals Inc. Alkoxy-substituted compounds, methods, and compositions for inhibiting PARP activity
US6635642B1 (en) * 1997-09-03 2003-10-21 Guilford Pharmaceuticals Inc. PARP inhibitors, pharmaceutical compositions comprising same, and methods of using same
US6514983B1 (en) 1997-09-03 2003-02-04 Guilford Pharmaceuticals Inc. Compounds, methods and pharmaceutical compositions for treating neural or cardiovascular tissue damage
WO1999011644A1 (en) 1997-09-03 1999-03-11 Guilford Pharmaceuticals Inc. Di-n-heterocyclic compounds, methods, and compositions for inhibiting parp activity
JP2002515488A (en) 1998-05-15 2002-05-28 ギルフォード ファーマシューティカルズ インコーポレイテッド Carboxamide compounds, compositions, and methods for inhibiting PARP activity
AU9298798A (en) 1998-05-15 1999-12-06 Guilford Pharmaceuticals Inc. Fused tricyclic compounds which inhibit parp activity
DE69940523D1 (en) 1998-10-14 2009-04-16 Purdue Research Foundation NEW INDENOISOQUINOLINE AS ANTINEOPLASTIC AGENT
US7781596B1 (en) 1998-11-03 2010-08-24 Abbott Laboratories Substituted 2-phenylbenzimidazoles, the production thereof and their use
US6387902B1 (en) 1998-12-31 2002-05-14 Guilford Pharmaceuticals, Inc. Phenazine compounds, methods and pharmaceutical compositions for inhibiting PARP
US6201020B1 (en) 1998-12-31 2001-03-13 Guilford Pharmaceuticals, Inc. Ortho-diphenol compounds, methods and pharmaceutical compositions for inhibiting parp
EP1140936B1 (en) 1999-01-11 2004-03-17 Agouron Pharmaceuticals, Inc. Tricyclic inhibitors of poly(adp-ribose) polymerases
US6346535B1 (en) * 1999-01-29 2002-02-12 American Cyanamid Company Fungicidal mixtures
US6465448B1 (en) 1999-08-13 2002-10-15 Case Western Reserve University Methoxyamine potentiation of temozolomide anti-cancer activity
WO2001090077A1 (en) 2000-05-19 2001-11-29 Guilford Pharmaceuticals, Inc. Sulfonamide and carbamide derivatives of 6(5h)phenanthridinones and their uses
GB0017508D0 (en) 2000-07-17 2000-08-30 Novartis Ag Antimicrobials
US6790199B1 (en) 2000-09-28 2004-09-14 North American Medical Products, Inc. Needle protective assembly for multi-draw needle
ITMI20002358A1 (en) * 2000-10-31 2002-05-01 Flavio Moroni TIENO DERIVATIVES, 2, 3-C | ISOCHINOLIN-3-ONE AS INHIBITORS OF POLY (DP-RIBOSE) POLYMERASE
US6956035B2 (en) 2001-08-31 2005-10-18 Inotek Pharmaceuticals Corporation Isoquinoline derivatives and methods of use thereof
US20030096833A1 (en) * 2001-08-31 2003-05-22 Jagtap Prakash G. Substituted ideno[1,2-c]isoquinoline derivatives and methods of use thereof
JP2003267888A (en) 2002-03-15 2003-09-25 Seishi Yoneda Cardiac arrest fluid
WO2004014862A1 (en) 2002-08-09 2004-02-19 Rutgers, The State University Nitro and amino substituted heterocycles as topoisomerase i targeting agents
JP2007501857A (en) * 2003-02-28 2007-02-01 イノテック ファーマシューティカルズ コーポレーション Tetracyclic benzamide derivatives and methods of use thereof
GB0317466D0 (en) 2003-07-25 2003-08-27 Univ Sheffield Use
SG150548A1 (en) 2003-12-01 2009-03-30 Kudos Pharm Ltd Dna damage repair inhibitors for treatment of cancer
US20050228007A1 (en) * 2004-02-26 2005-10-13 Prakash Jagtap Isoquinoline derivatives and methods of use thereof

Also Published As

Publication number Publication date
EP1603568A4 (en) 2009-02-25
US20040039009A1 (en) 2004-02-26
US7393955B2 (en) 2008-07-01
CN100400515C (en) 2008-07-09
RU2005130159A (en) 2006-03-27
US20050282848A1 (en) 2005-12-22
WO2004078712A3 (en) 2005-02-24
CN101270087A (en) 2008-09-24
US20050049270A1 (en) 2005-03-03
EP1603568A2 (en) 2005-12-14
CN1780625A (en) 2006-05-31
PL378294A1 (en) 2006-03-20
BRPI0407922A (en) 2006-03-01
US6956035B2 (en) 2005-10-18
US7268143B2 (en) 2007-09-11
JP2006520817A (en) 2006-09-14
RU2009144059A (en) 2011-06-10
WO2004078712A2 (en) 2004-09-16
AU2004218023A1 (en) 2004-09-16
KR20050118168A (en) 2005-12-15

Similar Documents

Publication Publication Date Title
CA2517358A1 (en) Isoquinoline derivatives and methods of use thereof
EP1420785B1 (en) Substituted indeno [1,2-c]isoquinoline derivatives and methods of use thereof
AU2002329920A1 (en) Substituted indeno[1,2-c]isoquinoline derivatives and methods of use thereof
MXPA06009701A (en) Isoquinoline derivatives and methods of use thereof.
US6277990B1 (en) Substituted phenanthridinones and methods of use thereof
EP1784186A2 (en) Methods for treating or preventing erectile dysfunction or urinary incontinence
AU713107B2 (en) New pyrido-thiopyranoindazoles with antitumor activity
WO1998049172A1 (en) Regioisomeric benzothiopyranopyridines having antitumor activity
ITMI971850A1 (en) PYRAZOL-ACRIDINE DERIVATIVES HAVING ANTI-CANCER ACTIVITY
ITMI971851A1 (en) AMINO-ACRIDINE DERIVATIVES CARBOXYMIDES WITH ANTI-CANCER ACTIVITY

Legal Events

Date Code Title Description
EEER Examination request
FZDE Discontinued