WO2012166151A1 - Use of dihydropyridophthalazinone inhibitors of poly (adp-ribose) polymerase (parp) in the treatment of myelodysplastic syndrome (mds) and acute myeloid leukaemia (aml) - Google Patents

Use of dihydropyridophthalazinone inhibitors of poly (adp-ribose) polymerase (parp) in the treatment of myelodysplastic syndrome (mds) and acute myeloid leukaemia (aml) Download PDF

Info

Publication number
WO2012166151A1
WO2012166151A1 PCT/US2011/039045 US2011039045W WO2012166151A1 WO 2012166151 A1 WO2012166151 A1 WO 2012166151A1 US 2011039045 W US2011039045 W US 2011039045W WO 2012166151 A1 WO2012166151 A1 WO 2012166151A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
alkyl
msi
microsatellite
compound
Prior art date
Application number
PCT/US2011/039045
Other languages
French (fr)
Inventor
Leonard Post
Ghulam J. MUFTI
Terry J. GAYMES
Original Assignee
Biomarin Pharmaceutical Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Biomarin Pharmaceutical Inc. filed Critical Biomarin Pharmaceutical Inc.
Priority to PCT/US2011/039045 priority Critical patent/WO2012166151A1/en
Publication of WO2012166151A1 publication Critical patent/WO2012166151A1/en

Links

Classifications

    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/50Pyridazines; Hydrogenated pyridazines
    • A61K31/5025Pyridazines; Hydrogenated pyridazines ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6844Nucleic acid amplification reactions
    • C12Q1/6858Allele-specific amplification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/156Polymorphic or mutational markers

Definitions

  • Described herein are methods of using dihydropyridophthalazinone inhibitors of Poly(ADP-Ribose)Polymerase (PARP) to treat or prevent Myelodysplastic Syndrome (MDS) and Acute Myeloid Leukaemia (AML). Also described herein are methods of determining a subject's sensitivity to such PARP inhibitors, based on their Microsatellite Instability (MSI) status.
  • MSI Microsatellite Instability
  • PARP poly(ADP-ribose)polymerases
  • PARP has been implicated in the signaling of DNA damage through its ability to recognize and rapidly bind to DNA single or double strand breaks (D'Amours, et al., Biochem. J., 342, 249-268 ( 1999)). It participates in a variety of DNA-related functions including gene amplification, cell division, differentiation, apoptosis, DNA base excision repair as well as effects on telomere length and chromosome stability (d'Adda di Fagagna, et al., Nature Gen., 23( 1 ), 76-80 ( 1999)).
  • xMyelodysplastic Syndrome MDS
  • AML Acute Myeloid Leukaemia
  • MDS MDS
  • AML Acute Myeloid Leukaemia
  • a study of PARP inhibitors on AML cell lines showed that a proportion ( 15%) of cells derived from the AML patients demonstrated significant cytotoxic responses to PARP inhibitors (Gaymes, et al., Haematologica, 94(5), 638-646 (2009)).
  • Microsatellite Instability can be defined as an elevated rate of uncorrected replication errors in microsatellite sequences ( Han, et al., Science, 97, 226-234 (2006)). Disruption of the mismatch repair machinery results in MSI and loss of mismatch repair and subsequent MSI is observed in 12% of colorectal cancers. MSI in other cancers are less well characterized, but reports show the presence of mismatch repair defects and MSI in haematological maglinancies (Han. et al., Science, 97, 226-234 (2006); Gu, et al., Oncogene, 21 , 5758-5764 (2002)).
  • VISI has the potential to inflict frameshift mutations in genes that confer PARP inhibitor sensitivity.
  • PARP inhibitors and use of PARP inhibitors for treatment of certain cancers are known in the art, as described previously in International Patent Publication No. WO 2010/017055 A2, incorporated herein by reference in its entirety.
  • Methods of DNA repair pathway profiling and analyzing MSI are also known in the art, as described previously in Gaymes et al., Clin, Cancer Res., 1 2, 5104-51 1 1 (2006), incorporated herein by reference in its entirety.
  • Additional PARP inhibitor therapies are sought for certain cancers, e.g.. MDS and AML, which are associated with MSI.
  • a method for screening of candidates for PARP inhibitor therapy through DNA repair pathway profiling and analysis of MSI is also sought.
  • MDS Myelodysplastic Syndrome
  • AML Acute Myeloid Leukemia
  • Y and Z are each independently selected from the group consisting of:
  • a substituent independently selected from the group consisting of hydrogen, alkenyl, alkoxy, alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, alkyl, alkynyl, arylalkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, hydroxyalkylene, oxo, heterocycloalkyl, heterocycloalkylalkyl, alkylcarbonyl, arylcarbonyl, heteroarylcarbonyl alkylsulfonyl, arylsulfonyl,
  • heteroarylsulfonyl (NR A R B )alkylene, (NR A R B )carbonyl, (NR A R B )carbonylalkylene, (NR A R B )sulfonyl, and (NR A R B )sulfonylalkylene;
  • Ri , Ri, and R 3 are each independently selected from the group consisting of hydrogen, halogen, alkenyl, alkoxy, alkoxycarbonyl, alkyl, cycloalkyl, alkynyl, cyano, haloalkoxy, haloalkyl, hydroxyl, hydroxyalkylene, nitro, NR A R B , NR A R B alkylene, and (NR A R B )carbonyl;
  • a and B are each independently selected from the group consisting of hydrogen, Br, CI, F, I, OH, C C f ,aikyl, CrCgcycloalky!, alkoxy, and alkoxyalkyl, wherein C C 6 alkyl, C C s cycloaikyl, aikoxy, and alkoxyalkyl are optionally substituted with at least one substituent selected from the group consisting of OH, N0 2 , CN, Br, CI, F, I, C, -C 6 alkyl, and C 3 -C 3 cycloalkyl, wherein B is not OH;
  • R A . and R B are independently selected from the group consisting of hydrogen, alkyl, cycloalkyl, and alkylcarbonyl; or R A and RB taken together with the atom to which they are attached form a 3- 10 membered heterocycle ring optionally having one to three heteroatoms or hetero functionalities selected from the group consisting of -0-, -NH, -N(C r C 6 -alkyl)-, -NCO(C C ft -alkyl)-,
  • 3- 10 membered heterocycle ring is optionally substituted with one or more substituents
  • R4 and R 5 are each independently selected from the group consisting of hydrogen, alkyl, cycloalkyl, alkoxyalkyl, haloalkyl, hydroxyalkylene, and (NR A RB)alkylene; and
  • each R ⁇ is selected from the group consisting of OH, N0 2 , CN, Br, CI, F, 1.
  • heteroarylalkylthio heterocycloalkoxy, C 2 -C 3 heterocycloalkylthio, heterocyclooxy, heterocyclothio,
  • NR A R B (NR A R B )C, -C 6 alkylene, (NR A R B )carbonyl, (NR A R B )carbonylalkylene, (NR A R B )sulfonyl, and
  • the compound is administered as a single isomer, stereoisomer, or enantiomer, or mixture thereof.
  • the compound is administered as a salt, solvate, chemically protected form or prodrug thereof.
  • the compound is administered with at least one of a
  • the malignant cells of the patient exhibit Microsatellite
  • the malignant cells contain an insertion or deletion in at least one microsatellite.
  • the malignant cells of the patient exhibit MSI in two, three, four, five, six, seven or more microsatellites.
  • the MSI in the at least one microsatellite comprises a plurality of insertions and/or deletions.
  • the MSI in the at least one microsatellite may be detected using primers designed to detect insertion or deletion in the microsatellite.
  • the at least one microsatellite is NR2I, NR22, NR24, Bat25, Bat26.
  • the at least one microsatellite is selected from the group consisting o(NR2l, NR22, Bat25, Bat26, Mono27 and a microsatellite as described herein in the DNA repair genes CTiP, Mrell and ATM.
  • the at least one microsatellite is selected from the group consisting of NR21, NR24 and Bat25.
  • the MSI comprises deletion or insertion of 3 or more base pairs in the microsatellite(s).
  • MSI comprises insertion(s) or deletion(s) of base pairs in microsatellite(s) that lead to inactivation of DNA repair function and/or results in a frameshift mutation in a double stranded DNA repair gene expression product.
  • the DNA repair function is governed by a double stranded DNA repair gene selected from the group consisting oi Mrel l, ATM and CTiP.
  • the method further comprises analyzing for MSI in at least one microsatellite of a patient's malignant cell DNA.
  • the compound is 5-fluoro-8-(4-fluorophenyl)-9-( l -methyl- 1H- l ,2,4-triazol-5-yl)-8,9-dihydro-2H-pyrido[4,3,2-de]phthalazin-3(7H)-one, or a pharmaceutically acceptable salt thereof.
  • a method of treating AML in a patient with AML comprising administering to a patient with AML a therapeutically effective amount of a compound of Formula (I) or stereoisomer or a pharmaceutically acceptable salt thereof.
  • the compound is 5-fluoro-8-(4-fluorophenyl)-9-( 1 -methyl- 1H- 1 ,2,4-triazol-5-yl)-8,9-dihydro-2H- pyrido[4,3,2-de]phthalazin-3(7H)-one, or a stereoisomer or a pharmaceutically acceptable salt thereof.
  • the malignant cells in the patient exhibit MSI in a microsatellite.
  • the malignant cells in the patient exhibit MSI in at least two, three, four, five, six or more microsatellites.
  • the malignant cells in the patient exhibit MSI in a microsatellite.
  • the malignant cells in the patient exhibit MSI in at least two, three, four, five, six or more microsatellites.
  • the method comprises analyzing for MSI in at least one
  • the MSI comprises three or more inserted or deleted base pairs in the at least one microsatellite.
  • the presence of MSI in the at least one microsatellite in the patient's malignant cell DNA identifies the patient's malignancy as likely to be sensitive to therapy Compound of Formula I.
  • the MSI in the at least one microsatellite may be detected using a primer designed to detect insertion or deletion in the microsatellite.
  • the at least one microsatellite is NR21, NR22, NR24, Bat25, Bat26, Mono27 and/or a microsatellite as described herein in the DNA repair genes CTiP, Mrel l, A TM, RacI50, Chkl, BLM, PTEN, A TM and A TR.
  • the at least one microsatellite is NR21, NR22, Ba(25, Bat26, Mono27, or a microsatellite as described herein in the DNA repair genes CTiP, Mrel l md A TM.
  • the at least one microsatellite is selected from the group consisting of NR21, NR24 and Bat25.
  • the microsatellite(s) that contain insertion(s) or deletion(s) of base pairs lead to inactivation of DNA repair function.
  • the DNA repair function is governed by a double stranded DNA repair gene selected from the group consisting of Mrell, A TM and CTiP.
  • the method comprises analyzing for MSI in at least one microsatellite(s) in a patient's malignant cell DNA.
  • the MSI comprises three or more inserted or deleted base pairs in at least one microsatellite in the patient's malignant cell DNA.
  • the presence of MSI in at least one microsatellite of the patient's malignant cell DNA indicates that the patient's malignancy is likely to be sensitive to treatment with a compound of Formula (I).
  • the MSI in the at least one microsatellite may be detected using a primer designed to detect insertion or deletion in the microsatellite.
  • the at least one microsatellite is NR21 , NR22, NR24, Bat25, Bat26, Mono27 and/or a microsatellite as described herein in the DNA repair genes CTiP, Mrel l, A TM, RadSO, Chkl, BLM, PTEN, A TM and A TR.
  • the at least one microsatellite is NR21, NR22, Bat25, Bat26, Mono27, or a microsatellite as described herein in the DNA repair genes CTiP, Mrel l and ATM.
  • the at least one microsatellite is selected from the group consisting of NR21, NR24 and Bat25.
  • the at least one microsatellite contains insertion(s) or deletion(s) of base pairs leads to inactivation of DNA repair function.
  • the DNA repair function is governed by a double stranded DNA repair gene selected from the group consisting of Mrell, A TM and CTiP.
  • the method further comprises cytogenetic analysis of chromosomal abnormalities in the patient's malignant cell DNA.
  • FIG. 1 Response to PARP inhibition in MDS/ AML cell lines. Varying concentrations of Compound 1 were added to MDS/AML cell lines and peripheral blood lymphocytes (PBL) continuously for 12- 14 days. Soft agar clonogenic assays were used to determine cell survival. [0019J FIG. 2. Composite microsatellite analysis of monucleotide repeat NR21 (21 consecutive thymines in 5'UTR of the solute carrier gene, SLC7A8, shown underlined in legend) in cell lines NB4, KG- 1 and P39; comparison to normal PBL cell line.
  • monucleotide repeat NR21 21 consecutive thymines in 5'UTR of the solute carrier gene, SLC7A8, shown underlined in legend
  • FIG. 3 Composite microsatellite analysis of monucleotide repeat Mono27 (27 consecutive intronic adenines in MAP4K3, shown underlined in legend) in cell lines KG- 1 , Molm- 13, Moit 4, NB4 and P39; comparison to normal PBL cell line.
  • FIG. 4 Composite microsatellite analysis of monucleotide repeat CTiP (9 consecutive thymines in exon 1 1 , shown underlined in legend) in MSI-high cell lines Molm-13, KG-1 and P39; comparison to normal PBL cell line. Fluorescent PCR image shown on left, Sanger sequence on right.
  • FIG. 5 Composite microsatellite analysis of monucleotide repeat Mrel l ( 1 1 consecutive intronic thymines, shown underlined in legend) in MSI-high cell lines KG- 1 , NB4, P39 and Molm- 13; comparison to normal PBL cell line. Fluorescent PCR image shown on left, Sanger sequence on right.
  • FIG. 6 Composite microsatellite analysis of monucleotide repeat Mrell (1 1 consecutive intronic thymines, shown underlined in legend) in MSI-stable cell lines HL60, Jurkat, DNA ligase IV " ' (DNL), OCI-AML3 and U937. Fluorescent PCR image shown on left, Sanger sequence on right.
  • FIG. 7 Composite microsatellite analysis of monucleotide repeat A TM( 15 consecutive intronic thymines, shown underlined in legend) in cell lines Jurkat, KG- 1 , DNA ligase IV ' (DNL), NB4, P39, Molm- 13 and OCI-AML3. Fluorescent PCR image shown on left, Sanger sequence on right.
  • FIG. 8. PCR for exon 8 of A TM was performed on the cDNA of cell lines DNL, Jurkat, K562, KG- 1 , Molm- 13, Molt 4, NB4 and P39. The ratio of intensity (RI) of aberrant to wild-type bands was measured using Adobe PhotoshopTM.
  • FIG. 9 PCR for exon 5 of Mrel l was performed on the cDNA of cell lines DNL, Jurkat, K562, KG-1 , Molm-13, Molt 4, NB4 and P39. The ratio of intensity (RI) of aberrant to wild-type bands was measured using Adobe Photoshop 1 M .
  • FIG. 10 Composite microsatellite analysis of CTiP (9 consecutive thymines in exon 1 1 , shown underlined in legend) in 5 patients with high risk MDS. Fluorescent PCR image shown on left, Sanger sequence on right. DETAILED DESCRIPTION OF THE INVENTION
  • MDS Myelodysplastic Syndrome
  • AML Acute Myeloid Leukemia
  • the methods comprise administering to the patient
  • the methods comprise administering to to the patient isomers including single isomers, stereoisomers, enantiomers, diastereoisomers, or mixtures thereof, of compounds having the structure of Formula (I) and Formula (II).
  • Y and Z are each independently selected from the group consisting of:
  • a substituent independently selected from the group consisting of hydrogen, alkenyl, alkoxy, alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, alkyl, alkynyl, arylalkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, hydroxyalkylene, oxo, heterocycloalkyl, heterocycloalkylalkyl, alkylcarbonyl, arylcarbonyl, heteroarylcarbonyl alkylsulfonyl, arylsulfonyl,
  • heteroarylsulfonyl (NR A R s )alkylene.
  • Ri , Ri, and R3 ⁇ 4 are each independently selected from the group consisting of hydrogen, halogen, alkenyl, alkoxy, alkoxycarbonyl, alkyl, cycloalkyl, alkynyl, cyano, haloalko y, haloalkyl, hydroxyl, hydroxyalkylene, nitro, A R B , NR A R B alkylene, and (N R A R B )carbonyl;
  • a and B are each independently selected from hydrogen, Br, CI, F, I, OH, C, -C crampa!ky 1, C 3 -C 8 cycloalkyl, alkoxy, alkoxyalkyl wherein C r C A alkyl, C3-C R cycloalkyl, alkoxy, alkoxyalkyl are optionally substituted with at least one substituent selected from OH, N0 2 , CN, Br, CI, F, I, C r C 6 alkyl, and C 3 - Qcycloalkyl, wherein B is not OH;
  • R A, and R B re independently selected from the group consisting of hydrogen, alkyl, cycloalkyl, and alkylcarbonyl; or R A and R B taken together with the atom to which they are attached form a 3- 10 membered heterocycle ring optionally having one to three heteroatoms or hetero functionalities selected from the group consisting of -0-, -NH, -N(C,-C 6 -alkyl)-, -NCO(C C 6 -alkyl)-, -N(aryl)-, -N(aryl-C
  • R and R 5 are each independently selected from the group consisting of hydrogen, alkyl, cycloalkyl, alkoxyalkyl, haloalkyl, hydroxyalkylene, and (NR A R B )alkylene; and
  • each 3 ⁇ 4 is selected from the group consisting of OH, N0 2 , CN, Br, CI, F, I, C C 6 alkyl, C 3 - C s cycloalkyl, Ci-Cgheterocycloalkyl, C 2 -C 6 alkenyl, alkoxy, alkoxyalkyl, alkoxycarbonyl,
  • alkoxycarbonylalkyl C 2 -C A alkynyl, aryl, arylalkyl, CrCxcycloalkylalkyl, haloalkoxy, haloalkyl, hydroxyalkylene, oxo, heteroaryl, heteroarylalkoxy, heteroaryloxy, heteroarylthio, heteroarylalkylthio, heterocycloalkoxy, C 2 -Qheterocydoalkylthio, heterocyclooxy, heterocyclothio, NR A RB, (NR A R B )C Qalkylene, (NR A R B )carbonyl, (NR A R B )carbonylalkylene, (NR A R B )sulfonyl, and
  • Y is an aryl or heteroaryl group optionally substituted with at least one Re
  • Z is an aryl group optionally substituted with at least one R 6 ;
  • a and B are each independently selected from hydrogen, Br, CI, F, I, OH, C r C 6 alkyl, C 3 -C 8 cycloaikyi, alkoxy, alkoxyalkyl wherein C C 6 alkyl, C 3 -C 8 cycloalkyl, alkoxy, alkoxyalkyl are optionally substituted with at least one substituent selected from OH, N0 2 , CN, Br, CI, F, I, C C 6 alkyl, and C 3 - Qcycloalkyl, wherein B is not OH;
  • R 6 is selected from OH, N0 2 , CN, Br, CI, F, I, C r C 5 alkyl, C 3 -C 8 cycIoalkyl, C 2 -C 8 heterocycloalkyl; CVCeaikenyl, alkoxy, alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C 2 -C 6 alkynyl, aryl, arylalkyl, CrQcycloalkylalkyl, haloalkoxy, haloalkyl, hydroxyalkylene, oxo, heteroaryl, heteroarylalkoxy, heteroaryloxy, heteroarylthio, heteroarylalkylthio, heterocycloalkoxy, CV
  • i is selected from hydrogen. Br, CI, I, or F;
  • R A, and R B are independently selected from the group consisting of hydrogen, C i-C 6 alkyl, C
  • R A and R B taken together with the atom to which they are attached form a 3- 10 membered heterocycle ring optionally having one to three heteroatoms or hetero functionalities selected from the group consisting of -0-, -NH, -N(Ci-C 6 alkyl)-, -NCO(C r C 6 alkyl)-, -NCO(C 3 -C 3 cycloaikyI)-, -N(aryl)-, -N(aryl-C, -C 6 alkyi-)-.
  • the compounds administered in the methods provided herein are compounds of Formula (I) or a therapeutically acceptable salt thereof, wherein R
  • R B are independently selected from the group consisting of hydrogen, alkyl, cycloalkyl, and alkylcarbonyl; or R A and R B taken together with the atom to which they are attached form a 3- 10 membered heterocycle ring optionally having one to three heteroatoms or hetero functionalities selected from the group consisting of -0-,
  • the compounds administered in the methods provided herein are compounds of Formula (I) wherein Y is an aryl group.
  • the aryl group is a phenyl group.
  • the phenyl group is substituted with at least one R, selected from Br, CI, F, or I.
  • R is F.
  • the phenyl group is substituted with at least one R, selected from (NR A R B )C r C 6 alkylene, (NR A R B )carbonyl,
  • R A R B carbonylalkylene, ( R A R n )sulfonyl, and (NR A R B )sulfonylalkylene.
  • R caution is (NR A R B )C i-C ft alkylene.
  • Ci-C 6 alkyl is selected from methylene, ethylene, n-propylene, iso-propylene, n-butylene, iso-butylene, and tert-butylene.
  • C, -C 6 alkyl is methylene.
  • R A and R B are each independently hydrogen, C
  • C r C 6 alkyl is selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl.
  • Cj -Qalkyl is methyl.
  • -C 6 alkyl is ethyl.
  • C 3 -C 8 cycloalkyl is cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl.
  • C 3 -C 8 cycloalkyl is cyclopropyl.
  • R 6 is hydroxyalkylene.
  • hydroxyalkylene is selected from CH 2 OH, CH 2 CH 2 OH, CH 2 CH 2 CH 2 OH, CH(OH)CH 3 , CH(OH)CH 2 CH 3 , CH 2 CH(OH)CH 3 , and CH 2 CH 2 CH 2 CH 2 OH.
  • R A and R B taken together with the nitrogen to which they are attached form a 6 membered heterocycle ring having 1 heteroatom or hetero functionality selected from the group consisting of -0-, -NH, or -N(C r C 6 alkyI).
  • the hetero functionality is -N(C r C 6 alkyl).
  • -C 6 alkyl is selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl. In yet a further embodiment C C 6 alkyl is methyl.
  • Y is a heteroaryl group optionally substituted with at least one R ⁇ ,.
  • the heteroaryl group is selected from furan, pyridine, pyrimidine, pyrazine, imidazole, thiazole, isothiazole, pyrazole, triazole, pyrrole, thiophene, oxazole, isoxazole, 1 ,2,4- oxadiazole, 1 ,3,4-oxadiazole, 1 ,2,4-triazine, indole, benzothiophene, benzoimidazole, benzofuran, pyridazine, 1 ,3,5-triazine, thienothiophene, quinoxaline, quinoline, and isoquinoline.
  • the heteroaryl group is imidazole.
  • imidazole is substituted with Ci-C 6 alkyl selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl.
  • C r C 6 alkyl is methyl.
  • the heteroaryl group is furan.
  • the heteroaryl group is thiazole.
  • the heteroaryl group is 1 ,3,4-oxadiazole.
  • heteroaryl group is substituted with C
  • -C 6 alkyl is methyl.
  • Z is an aryl group.
  • the aryl group is a phenyl group, in yet another embodiment the phenyl group is substituted with at least one R 6 selected from Br, CI, F, or I.
  • R 6 is F.
  • R 6 is CI.
  • the phenyl group is substituted with at least one Rg selected from
  • R 6 is (NR A R B )C,-C 6 alkylene.
  • C C 6 alkyl is selected from methylene, ethylene, n-propylene, iso-propylene, n-butylene, iso-butylene, and tert-butylene.
  • C C 6 alkyl is methylene.
  • R A and R B are each independently hydrogen, C r C ( ,alkyl, or C 3 -C 3 cycloalkyl.
  • C C 6 alkyl is selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl.
  • Ci -C 6 alkyl is methyl.
  • R A and R B taken together with the nitrogen to which they are attached form a 6 membered heterocycle ring having 1 heteroatom or hetero functionality selected from the group consisting of -0-, -NH, or -N(d - C 6 alkyl).
  • the hetero functionality is -N(C
  • C Qalkyl is selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl.
  • -C ft alkyl is methyl.
  • Z is a heteroaryl group optionally substituted with at least one R 6 .
  • the heteroaryl group is selected from furan, pyridine, pyrimidine, pyrazine, imidazole, thiazole, isothiazole, pyrazole, triazole, pyrrole, thiophene, oxazole, isoxazole, 1 ,2,4-oxadiazole, 1.3,4-oxadiazole, 1 ,2,4-triazine, indole, benzothiophene, benzoimidazole, benzofuran, pyridazine, 1 ,3,5-triazine, thienothiophene, quinoxaline, quinoline, and isoquinoline.
  • the heteroaryl group is imidazole.
  • imidazole is substituted with Ci -C 6 alkyl selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl.
  • C C 6 alkyl is methyl.
  • the heteroaryl group is furan.
  • the heteroaryl group is thiazole.
  • the heteroaryl group is 1 ,3,4-oxadiazole.
  • heteroaryl group is substituted with C C 6 alkyl selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl.
  • G -Qalkyl is methyl.
  • R 2 is hydrogen.
  • R is selected from F, CI, Br, and I.
  • R 2 is F.
  • the compounds administered in the methods provided herein are compounds of Formula (1) wherein A is hydrogen.
  • A is C Cealkyl.
  • A is selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, tert- butyl, n-pentyl, and n-hexyl.
  • methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, tert-butyl, n-pentyl, and n-hexyl are optionally substituted with OH, N0 2 , CN, Br, CI, F, and 1.
  • A is methyl, in yet another embodiment, A is selected from F. C'l, Br, and I.
  • A is C 3 -Cscycloalkyl.
  • A is cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl.
  • A is substituted with OH, N0 2 , or CN.
  • B is Ci -C 6 alkyl.
  • B is selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, tert-butyl, n-pentyl, and n-hexyl.
  • methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, tert-butyl, n-pentyl, and n-hexyl are optionally substituted with OH, N0 2 , CN, Br, CI, F, and I.
  • the administered compound is a compound of Formula ( I) wherein B is hydrogen.
  • B is methyl.
  • B is selected from F, CI, Br, and I.
  • B is C 3 - Cscycloalkyl.
  • B is cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl.
  • A is substituted with OH, N0 2 , or CN.
  • the administered compound is a compound of Formula ( I) wherein A is hydrogen and B is selected from Br, CI, F, I, C C 6 alkyl, C 3 -C 8 cycloalkyl, alkoxy, alkoxyalkyl wherein C
  • the administered compound is a compound of Formula (I) wherein B is hydrogen and A is selected from Br, CI, F, I, C C h alkyl, C 3 - Cscycloalkyl, alkoxy, alkoxyalkyl wherein CrQalkyl, C 3 -C 3 cycloalkyl, alkoxy, alkoxyalkyl are optionally substituted with at least one substituent selected from OH, N0 2 , CN.
  • both A and B are hydrogen.
  • both A and B are selected from Br, CI, F, I, C,-C 6 alkyl, C 3 -C 8 cycloalkyl, alkoxy, alkoxyalkyl wherein Ci-C 6 alkyl, C C g cycloalkyl, alkoxy, alkoxyalkyl are optionally substituted with at least one substituent selected from OH, N0 2 , CN, Br, CI, F, I, C
  • the compounds administered in the methods provided herein are compounds of Formula (11) wherein Y is an aryl group.
  • the aryl group is a phenyl group.
  • the phenyl group is substituted with at least one 3 ⁇ 4, selected from Br, CI, F, or I.
  • 3 ⁇ 4 is F.
  • the phenyl group is substituted with at least one 3 ⁇ 4 selected from (NR A R B )C
  • R 6 is (NR A R B )C
  • C C 6 alkyl is selected from methylene, ethylene, n- propylene, iso-propylene, n-butylene, iso-butylene, and tert-butylene.
  • C,- Qalkylene is methylene.
  • R A and R B are each independently hydrogen, C C 5 alkyl, or C 3 -C 8 cycloalkyl.
  • C C 6 alkyl is selected from methyl, ethyl, n- propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl.
  • C r C ⁇ ,alkyl is methyl.
  • -C 6 alkyl is ethyl.
  • C 3 -C 8 cycloalkyl is cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl.
  • C 3 -C 8 cycloalkyl is cyclopropyl.
  • R ⁇ is hydroxyalkylene.
  • hydroxyalkylene is selected from CH 2 OH, CH 2 CH 2 OH, CH 2 CH 2 CH 2 OH, CH(OH)CH 3 , CH(OH)CH 2 CH 3 , CH 2 CH(OH)CH 3 , and CH 2 CH 2 CH 2 CH 2 OH.
  • R A and R B taken together with the nitrogen to which they are attached form a 6 membered heterocycle ring having 1 heteroatom or hetero functionality selected from the group consisting of -0-, -NH, or -N(Ci-C ft alkyl).
  • the hetero functionality is -N(C C 6 alkyl).
  • Ci-C 6 alkyl is selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl.
  • -C 6 alkyl is methyl.
  • Y is a heteroaryl group optionally substituted with at least one Re.
  • the heteroaryl group is selected from furan, pyridine, pyrimidine, pyrazine, imidazole, thiazole, isothiazole, pyrazole, triazole, pyrrole, thiophene, oxazole, isoxazole, 1 ,2,4- oxadiazole, 1 ,3,4-oxadiazole, 1 ,2,4-triazine, indole, benzothiophene, benzoimidazole, benzofuran, pyridazine, 1 ,3,5-triazine, thienothiophene, quinoxaline, quinoline, and isoquinoline.
  • the heteroaryl group is imidazole.
  • imidazole is substituted with C C 6 alkyl selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl.
  • C C 6 alkyl is methyl.
  • the heteroaryl group is furan.
  • the heteroaryl group is thiazole.
  • the heteroaryl group is 1 ,3,4-oxadiazole.
  • heteroaryl group is substituted with CrC e alkyl selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl.
  • -C f ,alkyl is methyl.
  • Z is an aryl group.
  • the aryl group is a phenyl group.
  • the phenyl group is substituted with at least one R 6 selected from Br, CI, F, or I.
  • R 6 is F.
  • R ⁇ is CI.
  • the phenyl group is substituted with at least one R ⁇ , selected from
  • R 6 is (NR A R B )C r C 6 alkylene.
  • Ci-Cealkylene is selected from methylene, ethylene, n-propylene, iso-propylene, n-butylene, iso-butylene, and tert-butylene.
  • -C 6 alkyl is methylene.
  • R A and R B are each independently hydrogen, Ci-C f) alkyl, or C 3 -C 8 cycloalkyl.
  • Ci -C 6 alkyl is selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl.
  • C C(,alkyl is methyl.
  • R A and R B taken together with the nitrogen to which they are attached form a 6 membered heterocycle ring having 1 heteroatom or hetero functionality selected from the group consisting of -0-, -NH, or -N(C r C 6 alkyl).
  • the hetero functionality is -N(Ci-C 6 alkyl).
  • C t - C 6 alkyl is selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl.
  • Q -Ce lkyl is methyl.
  • R 2 is hydrogen.
  • R 2 is selected from F, CI, Br, and I.
  • R 2 is F.
  • the compounds administered in the methods provided herein are compounds of Formula (II) wherein A is hydrogen.
  • A is C
  • A is selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, tert- butyl, n-pentyl, and n-hexyl.
  • methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, tert-butyl, n-pentyl, and n-hexyl are optionally substituted with OH, N0 2 , CN, Br, CI, F, and I.
  • A is methyl.
  • A is selected from F, CI, Br, and I.
  • A is C 3 -C 8 cycloalkyl.
  • A is cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl.
  • A is substituted with OH, N0 2 , or CN.
  • the administered compound is of Formula (II) wherein B is hydrogen.
  • B is Ci-C 6 alkyl.
  • B is selected from methyl, ethyl, n- propyl, iso-propyl, n-butyl, iso-butyl, tert-butyl, n-pentyl, and n-hexyl.
  • methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, tert-butyl, n-pentyl, and n-hexyl are optionally substituted with OH, N0 2 , CN, Br, CI, F, and I.
  • B is methyl.
  • B is selected from F, CI, Br, and I.
  • B is CrCgcycloalkyl.
  • B is cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl.
  • A is substituted with OH, N0 , or CN.
  • the admininistered compound is a compound of Formula (II) wherein A is hydrogen and B is selected from Br, CI, F, I, C r C 6 alkyl, C 3 - C 8 cycloalkyl, alkoxy, alkoxyalkyl wherein Ci-C 6 alkyl, C 3 -C 8 cycloaIkyl, alkoxy, alkoxyalkyl are optionally substituted with at least one substituent selected from OH, NO?, CN, Br, CI, F, I, C C(,alkyl, and C 3 -C 8 cycloalkyl.
  • the administered compound is a compound of Formula (II) w herein B is hydrogen and A is selected from Br, CI, F, I, d -Cealkyl, C 3 -C 3 cycloalkyl, alkoxy, alkoxyalkyl wherein C C ( ,alkyl, C 3 -C 8 cycloalkyl, alkoxy, alkoxyalkyl are optionally substituted with at least one substituent selected from OH, N0 2 , CN, Br, CI, F, I, C r C 6 alkyl, and C C s cycloalkyl.
  • both A and B are hydrogen.
  • both A and B are selected from Br, CI, F, I, C
  • the compound administered in the methods provided herein is a compound selected from:
  • the compound administered in the methods provided herein is a compound selected from:
  • the compound administered in the methods provided herein is a compound selected from:
  • the compound administered in the methods provided herein is a compound selected from:
  • the compound administered in the methods provided herein is a compound selected from:
  • the compound administered in the methods provided herein is of the following formula:
  • the compound administered in the methods provided herein is 5- fluoro-8-(4-fluorophenyl)-9-(l-methyl-lH-l,2,4-triazol-5-yl)-8,9-dihydro-2H-pyrido[4,3,2- de]phthalazin-3(7/ )-one (Compound 1), or a pharmaceutically acceptable salt thereof.
  • the compound administered in the methods provided herein is selected from the group consisting of:
  • the compound administered in the methods provided herein is ( 8S,9R)-5-fluoro-8-(4-fluorophenyl)-M
  • the compounds envisioned to be administered in the methods provided herein are disclosed in International Patent Publication No. WO 2010/017055 A2, which is incorporated by reference in its entirety in the instant application. Additional compounds are disclosed in International Patent Publication No. WO 2010/017055 A2.
  • the methods comprise administering to a subject in need of treatment a therapeutically effective amount of a compound that is a modulator of PARP activity.
  • the methods comprise administering to a subject in need of treatment a therapeutically effective amount of a PARP inhibitor, such as, for example, a compound of Formula (1) or (II).
  • a PARP inhibitor such as, for example, a compound of Formula (1) or (II).
  • the cancer is leukemia.
  • the cancer is Myelodysplastic Syndrome (MDS) or Acute Myeloid Leukemia (AML).
  • the subject is a patient with MDS or AML.
  • the cancer is AML.
  • described herein is a method of treating Myelodysplastic Syndrome (MDS) or Acute Myeloid Leukemia (AML) in a patient with MDS or AML, comprising administering to a patient with MDS or AML a therapeutically effective amount of a compound of Formula (I).
  • MDS Myelodysplastic Syndrome
  • AML Acute Myeloid Leukemia
  • described herein is a method of treating Acute Myeloid Leukemia (AML) in a patient with AML, comprising administering to a patient with AML a therapeutically effective amount of a compound of Formula (I).
  • a compound of Formula (I) or (II) is used for the treatment of MDS or AML in a patient with MDS or AML, or for the preparation of a medicament for the treatment of MDS or AML in a patient with MDS or AML.
  • a compound of Formula (I) or (II) is used for the treatment of AML in a patient with AML, or for the preparation of a medicament for the treatment of AML in a patient with AML.
  • a compound of Formula (I) or (II) is used for the treatment of cancer, or for the preparation of a medicament for the treatment of cancer, which is deficient in HR dependent DNA DSB repair activity.
  • the cancer comprises one or more cancer cells having a reduced or abrogated ability to repair DNA DSB by HR relative to normal cells.
  • the subject is heterozygous for a mutation in a gene encoding a component of the HR dependent DNA DSB repair pathway.
  • the cancer is leukemia.
  • the cancer is Acute Myeloid Leukemia (AML).
  • the cancer is Myelodysplastic Syndrome (MDS).
  • the treatment further comprises administration of ionizing radiation or a chemotherapeutic agent.
  • the HR dependent DNA DSB repair pathway repairs double-strand breaks (DSBs) in DNA via homologous mechanisms to reform a continuous DNA helix.
  • the components of the HR dependent DNA DSB repair pathway include, but are not limited to, ATM (NM 000051 ), ATR (NM_001 184), CTIP (NM_002894), BLM (NM_000057), RAD51 (NM_002875), RAD51 L1 (NM_002877), RAD51C (NM_002876), RAD51 L3 (NM_002878), DMC1 (NM_007068), XRCC2 (NM_005431 ), XRCC3 (NM_005432), RAD52 (NM_002879), RAD54L (NM 003579), RAD54B (NM_012415), BRCA 1 (NMJ)07295), BRCA2 (NM_000059), RAD50 (NM_005732), MRE1 1A (NM 005590) and NBS 1 (NM NM
  • HR dependent DNA DSB repair pathway includes regulatory factors such as EMSY.
  • HR components are also described in Wood, et ai, Science, 291 , 1284- 1289 (2001 ), which is hereby incorporated by reference for such disclosure. . . Khanna and S. P. Jackson, Nat. Genet. 27(3): 247-254 (2001 ); and Hughes-Davies, et ai. Cell, 1 15, pp 523-535 are also incorporated herein by reference for such disclosure.
  • a cancer which is deficient in HR dependent DNA DSB repair includes one or more cancer cells which have a reduced or abrogated ability to repair DNA DSBs through that pathway, relative to normal cells, i.e. the activity of the HR dependent DNA DSB repair pathway are reduced or abolished in the one or more cancer cells.
  • the activity of one or more components of the HR dependent DNA DSB repair pathway is abolished in the one or more cancer cells of an individual having a cancer which is deficient in HR dependent DNA DSB repair.
  • Components of the HR dependent DNA DSB repair pathway include the components listed above.
  • a method of treating a cancer deficient in DNA mismatch repair comprising administering to a subject in need of treatment a therapeutically effective amount of a PARP inhibitor, such as, for example, a compound of Formula (I) or (II).
  • a compound of Formula (I) or (II) is used for the treatment of a cancer deficient in MMR, or for the preparation of a medicament for the treatment of a cancer deficient in MMR, which is deficient in HR dependent DNA DSB repair activity.
  • MMR DNA mismatch repair
  • MSI Microsatellite Instability
  • a heterodimer of MLH 1 and PMS2 coordinates the interplay between the mismatch recognition complex and other proteins necessary for MMR.
  • additional proteins may include at least exonuclease 1 (EXO l ), possibly helicase(s), proliferating cell nuclear antigen (PCNA), single- stranded DNA-binding protein (RPA), and DNA polymerases ⁇ and ⁇ .
  • EXO l exonuclease 1
  • PCNA proliferating cell nuclear antigen
  • RPA single- stranded DNA-binding protein
  • MLH1 may heterodimerize with two additional proteins, MLH3 and PMS l .
  • PMS2 is required for the correction of single-base mismatches
  • PMS2 and MLH3 both contribute to the correction of insertion-deletion loops.
  • Additional homologs of the human MMR proteins are known that are required for functions other than MMR. These proteins include MSH4 and MSH5 that are necessary for
  • HNPCC hereditary nonpolyposis colon cancer
  • MSI occurs in a substantial proportion (2% to 50% of tumors) among non-HNPCC cancers (e.g., cancers of the breast, prostate, and lung).
  • categories MSS, MSI-L, and MSI-H can be distinguished in these cancers in analogy to HNPCC cancers.
  • a method for treating a cancer deficient in mismatch DNA repair pathway In another embodiment is a method for treating a cancer demonstrating microsatellite instability due to reduced or impaired DNA repair pathways. In another embodiment is a method for treating a cancer demonstrating genomic instability due to reduced or impaired DNA repair pathways.
  • BER Base Excision Repair
  • NER Nucleotide Excision Repair
  • the protein sequences for the protein products of these DNA repair genes will be known by one of skill in the art, and are disclosed at publicly-available databases as, e.g., GenPept, RefSeq, Swiss-Prot, PIR, PRE, and PDB, such as those found at the website for the National Center for Biotechnology Information (NCBI) (see
  • the patient's malignant cell DNA exhibits Microsatellite Instability (MSI) in a microsatellite, wherein MSI is explained in the Definition section of the instant application.
  • MSI Microsatellite Instability
  • the patient's malignant cell DNA exhibits MSI in at least two microsatellites.
  • Microsatellites in which MSI is exhibited can include, for example, any microsatellite known to those of skill in the art. Such microsatellites can include, for example, those identified in U.S. Patent No. 6, 150, 100, and U.S. Patent Publication No. 2009/0317815, each of which is incorporated herein by reference in its entirety for all purposes.
  • a microsatellite in which MSI is exhibited is NR21, NR22, NR24, Bat25, Bat26, Mono27 and/or a microsatellite as described herein in the DNA repair genes CTiP, Mrel 1, A TM, Rad50, Chkl, BLM, PTEN, A TM and ATR.
  • the microsatellite is NR21, NR22, Bat25, Bat26, Mono27, or a microsatellite as described herein in the DNA repair genes CTiP, Mrel I and A TM.
  • mutations and polymorphisms associated with cancer are detected at the nucleic acid level by detecting the presence of a variant nucleic acid sequence or at the protein level by detecting the presence of a variant (i.e., a mutant or allelic variant) polypeptide.
  • the methods comprise administering to a subject in need of treatment a therapeutically effective amount of a compound of Formula (I) and Formula (II) in combination with ionizing radiation, one or more chemotherapeutic agents, or a combination thereof.
  • Certain embodiments provide a method of potentiation of cytotoxic cancer therapy of MDS or AML in a subject in recognized need of such treatment comprising administering to the subject a therapeutically acceptable amount of a compound of Formula (1) or (II) or a therapeutically acceptable salt thereof.
  • a method for the treatment of MDS or AML comprising administering to a subject in need of treatment a therapeutically-effective amount of a compound of Formula (I) or (II) in combination with ionizing radiation or one or more
  • the subject is a patient with MDS or AML.
  • the compound described herein is administered simultaneously with ionizing radiation or one or more chemotherapeutic agents. In other embodiments, the compound described herein is administered sequentially with ionizing radiation or one or more chemotherapeutic agents.
  • the one or more chemotherapeutic agents are selected from among alemtuzumab, arsenic trioxide, asparaginase (pegylated or non-), bevacizumab, cetuximab, platinum-based compounds such as cisplatin, cladribine, daunorubicin/doxorubicin/idarubicin, irinotecan, fludarabine, 5-fluorouracil, gemtuzumab, methotrexate, paclitaxel, Taxol®, temozolomide, thioguanine, and classes of drugs including hormones (an antiestrogen, an antiandrogen, or gonadotropin releasing hormone analogues, interferons such as, for example, alpha interferon, nitrogen mustards such as, for example, busulfan, melphalan or mechlorethamine, retinoids such as, for example, tretinoin, topoisomerase
  • hormones an antiestrog
  • the one or more chemotherapeutic agents are selected from: alkylating agents, such as methyl methanesulfonate (MMS), temozolomide and dacarbazine (DTIC); topoisomerase- 1 inhibitors, such as Topotecan, Irinotecan, Rubitecan, Exatecan, Lurtotecan,
  • alkylating agents such as methyl methanesulfonate (MMS), temozolomide and dacarbazine (DTIC)
  • topoisomerase- 1 inhibitors such as Topotecan, Irinotecan, Rubitecan, Exatecan, Lurtotecan
  • Gimetecan Diflomotecan (homocamptothecins), the 7-substituted non-silatecans, the 7-silyl camptothecins and BNP 1350; non-camptothecin topoisomerase-I inhibitors, such as
  • indolocarbazoles and dual topoisomerase-I and II inhibitors, such as the benzophenazines, XR 1 1 76/MLN 576 and benzopyridoindoles.
  • methods of identifying malignant cells having MSI in an MDS or AML patient which comprise analyzing for MSI in one or more microsatellites of a patient's malignant cell DNA, wherein the presence of MSI in at least one microsatellite of the patient's malignant cell DNA identifies the patient as likely to be sensitive to therapy with a compound of Formula I or II.
  • the compound of Formula I or II is 5-fluoro-8-(4-fluorophenyl)- 9-( 1 -methyl- 1 H- 1 ,2,4-triazol-5-y l)-8,9-dihydro-2H-pyrido[4,3,2-de]phthalazin-3(7H)-one, or a stereoisomer thereof.
  • the methods comprise analyzing for MSI in at least one microsatellite of the patient's malignant cell DNA. In certain embodiments, the methods comprise analyzing for MSI in at least two microsatellites of the patient's malignant cell DNA. In certain embodiments, the methods comprise analyzing for MSI in at least three microsatellites of the patient's malignant cell DNA.
  • MSI comprises an insertion or deletion of one or more base pairs in a microsatellite of a patient's malignant cell DNA. Accordingly, in certain embodiments, the MSI comprises one or more deleted base pairs; two or more deleted base pairs; or three or more deleted base pairs in the microsatellite(s) of the patient's malignant cell DNA.
  • low-level MSI is defined as a 1 base pair (bp) deletion or insertion in a microsatellite
  • mid-level MSI (MSI-mid) is defined as ⁇ 2 bp deletion or insertion in a microsatellite
  • high-level MSI MSI-high
  • MSI stable is defined as no deletion or insertion in a microsatellite.
  • the MSI may be MSI-low.
  • the MSI may be MSI-mid.
  • the MSI may be MSI-high.
  • the methods disclosed herein envision that the presence of low-, mid- or high-level MSI in just one microsatellite of the patient's genomic DNA will identify a patient as likely to respond to therapy with a compound of Formula I or II.
  • the presence of MSI (low-, mid- or high-) in additional microsatellites of the patient ' s DNA may also be helpful is such patient identification.
  • the presence of MSI in at least one microsatellite of the patient's malignant cell DNA identifies the patient as likely to respond to therapy with a compound of Formula I or II.
  • the presence of MSI in at least two microsatellites of the patient's malignant cell DNA identifies the patient as likely to respond to therapy with a compound of Formula I or II. In certain embodiments, the presence of MSI in at least three microsatellites of the patient's malignant cell DNA identifies the patient as likely to respond to therapy with a compound of Formula 1 or II. [0077] The presence or absence of MSI within rnicrosatellites in malignant cell DNA may be assessed using methods well known of those of skill in the art.
  • a fluorescent multiplex assay that uses a panel of markers to analyze five nearly monomorphic mononucleotide microsatellite loci (NR21, NR24, Bat25, Bat26 and Mono27) is commercially available from Promega Corp (Madison, WI). See also e.g., Nardon et al., Diagn. Mol. Pathol., 19(4), 236-242 (2010); Bacher et al., Dis. Markers, 20 (4-5), 237-250 (2004); and U.S. Patent Publication No. 2009/0317815.
  • microsatellite as used herein means repeating sequences of 1 -6 base pairs of DNA, and includes those DNA sequences known in the art as simple sequence repeats (SSR), short tandem repeats (STR), or variable number tandem repeats (VNTR).
  • the repeated sequence may consist, e.g., of a sequence of one, two, three, four, five or six nucleotides (mono-, di-, tri-, tetra-, penta-, and hexa-nucleotide repeats, respectively), and can, for instance, be repeated 3 to 100 times. See, e.g., Eckert et al., Mol Carcinog, 48(4), 379-388 (2009).
  • the microsatellite is a mononucleotide repeat sequence, e.g., jj-r- jj- jj (SEQ ID NO. 1 ), referred to as (T) 9 ; TTTTTTTTT (SEQ ID NO. 2), referred to as (T) n ; TTTTTTTTTTTTTTT (SEQ ID NO. 3), referred to as (T), 5 ; TTTTTTTTTTTTTTTTTTTTTTTTTTT (SEQ ID NO. 4), referred to as (T) 2I ; and AAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAA (SEQ ID NO. 5), referred to as (A) 27 .
  • the microsatellite is a dinucleotide repeat sequence, e.g., GTGTGTGTGTGT (SEQ ID NO. 6), referred to as (GT) ft .
  • the microsatellite is a trinucleotide repeat sequence, e.g., CTGCTGCTGCTG (SEQ ID NO. 7), referred to as (CTG) 4 .
  • the microsatellite is a tetranucleotide repeat sequence, e.g., ACTCACTCACTCACTC (SEQ ID NO. 8), referred to as (ACTC) 4 .
  • the microsatellite is a monomorphic, or nearly monomorphic, mononucleotide repeat (e.g., NR21, NR24, Bat25, Bat 26 and Mono27).
  • a microsatellite may contain one or up to six or seven, or more, nucleotides that interrupt the repeat sequence within the microsatellite.
  • microsatellites are typically present in human genomic DNA.
  • the microsatellite is located within a gene, e.g., a DNA repair gene.
  • the microsatellite can, e.g., be located within the following genes: ATM (NM_00005 1), ATR (NM_001 184), CTIP (NM 002894), BLM (NM_000057), RAD51
  • RAD51 L 1 (NM_002877), RAD51 L 1 (NM 002877), RAD51 C (NM_002876), RAD5 1 L3 (NM_002878), DMC l (NM_007068), XRCC2 (NM 00543 1 ), XRCC3 (NM_005432), RAD52 (NM_002879), RAD54L (NM 003579), RAD54B (NM 012415), BRCA 1 (NM 007295), BRCA2 (NM_000059), RAD50 (NM_005732), MRE 1 1 A (NM_005590) or NBS 1 (NM_002485), among others.
  • the microsatellite is located within the intron/exon splice junction of a gene. In certain embodiments, the microsatellite is located within the intron of a gene. In certain embodiments, the microsatellite is located within the exon of a gene. In certain embodiments, the microsatellite is located within one of the control elements of a gene, e.g., the promoter of the gene. In certain embodiments, the gene is a DNA repair gene. In certain
  • the gene is other than a DNA repair gene.
  • the microsatellite is located outside of a gene.
  • Msatellite Instability means an alteration in a microsatellite found in human genomic DNA, e.g., an insertion or deletion of one or more base pairs, as compared to a reference microsatellite.
  • an MSI may be characterized such that the microsatellite length or repeat sequence is different than that, for instance, which was in the genomic DNA when it was inherited, or, for instance, as compared to the microsatellite length or repeat sequence in the general human population.
  • a reference microsatellite is found in the genome of a cell from a human not having MDS or AML, or cells from a population of humans characterized as being cancer free.
  • low-level MSI is defined as 1 base pair (bp) deletion or insertion in a microsatellite
  • mid-level MSI MSI-mid
  • high-level MSI MSI-high
  • MSI stable MSI-stable
  • alkenyl as used herein, means a straight, branched chain, or cyclic (in which case, it would also be known as a "cycloalkenyl") hydrocarbon containing from 2- 10 carbons and containing at least one carbon-carbon double bond formed by the removal of two hydrogens.
  • an alkenyl group is a monoradical or a diradical (i.e., an alkenylene group).
  • alkenyl groups are optionally substituted.
  • alkenyl examples include, but are not limited to, ethenyl, 2-propenyl, 2-methyl-2-propenyl, 3-butenyl, 4-pentenyl, 5-hexenyl, 2-heptenyl, 2-methyl- l -heptenyl, and 3-cecenyl.
  • alkoxy means an alkyl group, as defined herein, appended to the parent molecular moiety through an oxygen atom.
  • alkoxy include, but are not limited to, methoxy, ethoxy, propoxy, 2-propoxy, butoxy, tert-butoxy, pentyloxy, and hexyloxy.
  • alkyl as used herein, means a straight, branched chain, or cyclic (in this case, it would also be known as “cycloalkyl”) hydrocarbon containing from 1 - 10 carbon atoms.
  • alkyl include, but are not limited to, methyl, ethyl, n-propyl, iso-propyl, n-butyl, sec- butyl, tert-butyl, n-pentyl, isopentyl, neopentyl, n-hexyl, 3-methylhexyl, 2,2-dimethylpentyl, 2,3-dimethylhexyl, n-heptyl, n-octyl, n-nonyl, and n-decyl.
  • Ci-C6-alkyl as used herein, means a straight, branched chain, or cyclic (in this case, it would also be known as “cycloalkyl”) hydrocarbon containing from 1-6 carbon atoms.
  • alkyl include, but are not limited to, methyl, ethyl, n-propyl, iso-propyl, cyclopyl, n-butyl, sec-butyl, tert-butyl, cyclobutyl, n-pentyl, isopentyl, neopentyl, cyclopentyl, and n-hexyl.
  • cycloalkyl as used herein, means a monocyclic or polycyclic radical that contains only carbon and hydrogen, and includes those that are saturated, partially unsaturated, or fully unsaturated. Cycloalkyl groups include groups having from 3 to 10 ring atoms. Representative xamples of cyclic include but are not limited to, the following moieties: . In some embodiments, depending on the structure, a cycloalkyl group is a monoradical or a diradical (e.g. , a cycloalkylene group).
  • cycloalkyl groups refers to groups which are optionally substituted with 1 , 2, 3, or 4 substituents selected from alkenyl, alkoxy, alkoxyalkyl, alkoxycarbonyl, alkyl, alkylcarbonyl, alkylcarbonyioxy, alkyithio, alkyithioalkyl, alkynyl, carboxy, cyano, formyl, haloalkoxy, haloalkyl, halogen, hydroxyl, hydroxyalkylene, mercapto, oxo, -NR A RA, and
  • cycloalkylalkyl as used herein, means a cycloalkyl group, as defined herein, appended to the parent molecular moiety through an alkyl group, as defined herein.
  • Representative examples of cycloalkylalkyl include, but are not limited to, cyclopropylmethyl, 2-cyclobutylethyl, cyclopentylmethyl, cyclohexylmethyl, and 4-cycloheptylbutyl.
  • carrier refers to a compound which contains one or more covalently closed ring structures, and that the atoms forming the backbone of the ring are all carbon atoms
  • Carbocycle refers to a ring, wherein each of the atoms forming the ring is a carbon atom.
  • Carbocylic rings include those formed by three, four, five, six, seven, eight, nine, or more than nine carbon atoms. Carbocycles are optionally substituted.
  • alkoxyalkyl means at least one alkoxy group, as defined herein, appended to the parent molecular moiety through an alkyl group, as defined herein.
  • alkoxyalkyl include, but are not limited to, 2-methoxyethyl, 2-ethoxyethyl, tert- butoxyethyl and methoxymethyl.
  • alkoxycarbonyl as used herein, means an alkoxy group, as defined herein, appended to the parent molecular moiety through a carbonyl group, as defined herein.
  • alkoxycarbonyl include, but are not limited to, methoxycarbonyl, ethoxycarbonyl, and tert-butoxy carbonyl.
  • alkoxycarbonylalkyl means an alkoxycarbonyl group, as defined herein, appended to the parent molecular moiety through an alkyl group, as defined herein.
  • alkylearbonyl means an alkyl group, as defined herein, appended to the parent molecular moiety through a carbonyl group, as defined herein.
  • alkylearbonyl include, but are not limited to, acetyl, 1 -oxopropyl, 2,2-dimethyl- l - oxopropyl, 1 -oxoburyl, and 1 -oxopentyl.
  • alkylcarbonyloxy means an alkylearbonyl group, as defined herein, appended to the parent molecular moiety through an oxygen atom.
  • alkylcarbonyloxy include, but are not limited to, acetyloxy, ethylcarbonyloxy, and tert- buty lcarbony loxy .
  • alkylthio or "thioalkoxy” as used herein, means an alkyl group, as defined herein, appended to the parent molecular moiety through a sulfur atom.
  • alkylthio include, but are not limited to, methylthio, ethylthio, butylthio, tert-butylthio, and hexylthio.
  • alkylthioalkyl as used herein, means an alkylthio group, as defined herein, appended to the parent molecular moiety through an alkyl group, as defined herein.
  • alkylthioalkyl include, but are not limited to, methylthiomethyl, 2-(ethylthio)ethyl, butylthiomethyl, and hexylthioethyl.
  • alkynyl as used herein, means a straight, branched chain hydrocarbon containing from 2- 10 carbons and containing at least one carbon-carbon triple bond. In some embodiments, alkynyl groups are optionally substituted. Illustrative examples of alkynyl include, but are not limited to, acetylenyl, 1 -propynyl, 2-propynyl, 3-butynyl, 2-pentynyl, and 1 -butynyl.
  • aromatic refers to a planar ring having a delocalized ⁇ -electron system containing 4n+2 ⁇ electrons, where n is an integer. In some embodiments, aromatic rings are formed by five, six, seven, eight, nine, or more than nine atoms. In other embodiments, aromatics are optionally substituted.
  • the term includes monocyclic or fused-ring polycyclic (i.e., rings which share adjacent pairs of carbon atoms) groups.
  • aryl refers to an aromatic ring wherein each of the atoms forming the ring is a carbon atom. In some embodiments, aryl rings are formed by five, six, seven, eight, nine, or more than nine carbon atoms. Examples of aryl groups include, but are not limited to phenyl, naphthalenyl, phenanthrenyl, anthracenyl, fluorenyl, and indenyl.
  • aryl as used herein means an aryl group that is optionally substituted with one, two, three, four or five substituents independently selected from the group consisting of alkenyl, a!koxy, alkoxyalkyl, aikoxycarbonyl, alkyl, alkylearbonyl,
  • arylalkyl as used herein, means an aryl group, as defined herein, appended to the parent molecular moiety through an alkyl group, as defined herein.
  • arylalkyl examples include, but are not limited to benzyl, 2-phenylethyl, -phenylpropyl, l -methyl-3- pheny [propyl, and 2-naphth-2-ylethyI.
  • carbonyl as used herein, means a -C(O)- group.
  • cyano as used herein, means a -CN group.
  • halo or halogen as used herein, means a -CI, -Br, -I or -F.
  • nitro as used herein, means a -N0 2 group.
  • hydroxy as used herein, means a -OH group.
  • bond refers to a chemical bond between two atoms, or two moieties when the atoms joined by the bond are considered to be part of larger substructure.
  • haloalkyl include alkyl, alkenyl, alkynyl and alkoxy structures in which at least one hydrogen is replaced with a halogen atom.
  • the halogen atoms are all the same as one another, in other embodiments in which two or more hydrogen atoms are replaced with halogen atoms, the halogen atoms are not all the same as one another.
  • fluoroalkyl and fluoroalkoxy include haloalkyl and haloalkoxy groups, respectively, in which the halo is fluorine. In certain embodiments, haloalkyls are optionally substituted.
  • ester refers to a chemical moiety with formula -COOR, where R is selected from among alkyl, cycloalkyl, aryl, heteroaryl (bonded through a ring carbon) and heterocycloalkyl (bonded through a ring carbon), in some embodiments, any hydroxy, or carboxyl side chain on the compounds described herein is esterified.
  • heteroalkyl “heteroalkenyl” and “heteroalkynyl” as used herein, include optionally substituted alkyl, alkenyl and alkynyl radicals in which one or more skeletal chain atoms are selected from an atom other than carbon, e.g., oxygen, nitrogen, sulfur, silicon, phosphorus or combinations thereof.
  • heteroatom refers to an atom other than carbon or hydrogen. Heteroatoms are typically independently selected from among oxygen, sulfur, nitrogen, silicon and phosphorus, but are not limited to these atoms. In embodiments in which two or more heteroatoms are present, the two or more heteroatoms are all the same as one another, or some or all of the two or more heteroatoms are each different from the others.
  • Ring refers to any covalently closed structure. Rings include, for example, carbocycles (e.g. , aryls and cycloalkyls), heterocycles (e.g. , heteroaryls and
  • heterocycloalkyls examples include aromatics (e.g. aryls and heteroaryls), and non-aromatics (e.g. , cycloalkyls and heterocycloalkyls).
  • rings are optionally substituted.
  • rings form part of a ring system.
  • ring system refers to two or more rings, wherein two or more of the rings are fused.
  • fused refers to structures in which two or more rings share one or more bonds.
  • heteroaryl or, alternatively, “heteroaromatic” refers to an aryl group that includes one or more ring heteroatoms selected from nitrogen, oxygen and sulfur.
  • An iV-containing “heteroaromatic” or “heteroaryl” moiety refers to an aromatic group in which at least one of the skeletal atoms of the ring is a nitrogen atom.
  • the polycyclic heteroaryl group is fused or non-fused.
  • Illustrative of heteroaryl groups include, but are not limited to, the following moieties:
  • a heteroaryl group is a monoradical or a diradical (i.e., a heteroaryiene group).
  • heteroaryl means heteroaryl groups that are substituted with 0, 1 , 2, 3, or 4 substituents independently selected from alkenyl, alkoxy, alkoxyalkyl, alkoxycarbonyl, alkyl, alkylcarbonyl, alkylcarbonyloxy, alkylthio, alkylthioalkyl, alynyl, carboxy, cyano, formyl, haloalkoxy, haloalkyl, halogen, hydroxyl, hydroxyalkylene, mercapto, nitro, -NR A R B ,
  • heteroarylalkyl as used herein, means a heteroaryl, as defined herein, appended to the parent molecular moiety through an alkyl group, as defined herein.
  • alkyl group as defined herein.
  • heteroarylalkyl include, but are not limited to, pyridinylmethyl.
  • heterocycloalkyl or “non-aromatic heterocycle” as used herein, refers to a non-aromatic ring wherein one or more atoms forming the ring is a heteroatom.
  • a “heterocycloalkyl” or “non-aromatic heterocycle” group refers to a cycloalkyl group that includes at least one heteroatom selected from nitrogen, oxygen and sulfur.
  • the radicals are fused with an aryl or heteroaryl.
  • heterocycloalkyl rings are formed by three, four, five, six, seven, eight, nine, or more than nine atoms.
  • heterocycloalkyl rings are optionally substituted.
  • heterocycloalkyls contain one or more carbonyl or thiocarbonyl groups such as, for example, oxo- and thio-containing groups.
  • heterocycloalkyls include, but are not limited to, lactams, lactones, cyclic imides, cyclic thioimides, cyclic carbamates, tetrahydrothiopyran, 4H-pyran, tetrahydropyran, piperidine, 1 ,3-dioxin, 1 ,3-dioxane, 1 ,4-dioxin, 1 ,4- dioxane, piperazine, 1 ,3-oxathiane, 1 ,4-oxathiin, 1 ,4-oxathiane, tetrahydro- 1 ,4-thiazine, 2H- 1 ,2- oxazine, maleimide, succinimide, barbituric acid, thiobarbit
  • heterocycloalkyl also includes all ring forms of the carbohydrates, including but not limited to the monosaccharides, the disaccharides and the oligosaccharides.
  • heterocycle refers to heteroaryl and heterocycloalkyl used herein, refers to groups containing one to four heteroatoms each selected from O, S and N, wherein each heterocycle group has from 4 to 10 atoms in its ring system, and with the proviso that the ring of said group does not contain two adjacent O or S atoms.
  • the number of carbon atoms in a heterocycle is indicated (e.g. , Ci -C Intel heterocycle), at least one other atom (the heteroatom) must be present in the ring.
  • Designations such as "C ,-C ⁇ , heterocycle” refer only to the number of carbon atoms in the ring and do not refer to the total number of atoms in the ring. In some embodiments, it is understood that the heterocycle ring has additional heteroatoms in the ring. Designations such as "4-6 membered heterocycle” refer to the total number of atoms that are contained in the ring (i.e., a four, five, or si . membered ring, in which at least one atom is a carbon atom, at least one atom is a heteroatom and the remaining two to four atoms are either carbon atoms or heteroatoms).
  • heterocycles that have two or more heteroatoms, those two or more heteroatoms are the same or different from one another.
  • heterocycles are optionally substituted.
  • binding to a heterocycle is at a heteroatom or via a carbon atom.
  • Heterocycloalkyl groups include groups having only 4 atoms in their ring system, but heteroaryl groups must have at least 5 atoms in their ring system.
  • the heterocycle groups include benzo-fused ring systems.
  • An example of a 4-membered heterocycle group is azetidinyl (derived from azetidine).
  • An example of a 5-membered heterocycle group is thiazolyl.
  • An example of a 6-membered heterocycle group is pyridyl, and an example of a 10-membered heterocycle group is quinolinyl. Examples of
  • heterocycloalkyl groups are pyrrolidinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, tetrahydropyranyl, dihydropyranyl, tetrahydrothiopyranyl, piperidino, morpholino, thiomorpholino, thioxanyl, piperazinyl, azetidinyl, oxetanyl, thietanyl, homopiperidinyl, oxepanyl, thiepanyl, oxazepinyl, diazepinyl, thiazepinyl, 1 ,2,3,6-tetrahydropyridinyl, 2-pyrrolinyl, 3-pyrrolinyl, indolinyl, 2H-pyranyl, 4H-pyranyl, dioxanyl, 1 ,3-dioxolanyl, pyrazolinyl, di
  • heteroaryl groups are pyridinyl, imidazolyl, pyrimidinyl, pyrazolyl, triazolyl, pyrazinyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, oxazolyl, isothiazolyl, pyrrolyl, quinolinyl, isoquinolinyl, indolyl, benzimidazolyl, benzofuranyl, cinnolinyl, indazolyl, indolizinyl, phthalazinyl, pyridazinyl, triazinyl, isoindolyl, pteridinyl, purinyl, oxadiazolyl, thiadiazolyl, furazanyl, benzofurazanyl, benzothiophenyl, be
  • the foregoing groups are C-attached or N- attached where such is possible.
  • a group derived from pyrrole is pyrrol- 1 -yl ( ⁇ -attached) or pyrrol-3-yl (C-attached).
  • a group derived from imidazole is imidazol- l -yl or imidazol-3-yl (both iV-attached) or imidazoI-2-yl, imidazol-4-yl or imidazol-5-yl (all C-attached).
  • a heterocycle group is a inonoradical or a diradical (i.e., a heterocyclene group).
  • heterocycles described herein are substituted with 0, 1 , 2, 3, or 4 substituents independently selected from alkenyl, alkoxy, alkoxyalkyl, alkoxycarbonyl, alkyl, aikylcarbonyl, alkylcarbonyloxy, alkylthio, alkylthioalkyl, alynyl, carboxy, cyano, formyl, haloalkoxy, haloalkyl, halogen, hydroxyl, hydroxyalkylene, mercapto, nitro, -NR A RB, and -(NR A RB)carbonyl.
  • heterocycloalkoxy refers to a heterocycloalkyl group, as defined herein, appended to the parent molecular moiety through an alkoxy group.
  • heterocycloalkylthio refers to a heterocycloalkyl group, as defined herein, appended to the parent molecular moiety through an alkylthio group.
  • heterocyclooxy refers to a heterocycloalkyl group, as defined herein, appended to the parent molecular moiety through an oxygen atom.
  • heterocyclothio refers to a heterocycloalkyl group, as defined herein, appended to the parent molecular moiety through a sulfur atom.
  • heteroarylalkoxy refers to a heteroaryl group, as defined herein, appended to the parent molecular moiety through an alkoxy group.
  • heteroarylalkylthio refers to a heteroaryl group, as defined herein, appended to the parent molecular moiety through an alkylthio group.
  • heteroaryloxy refers to a heteroaryl group, as defined herein, appended to the parent molecular moiety through an oxygen atom.
  • heteroarylthio refers to a heteroaryl group, as defined herein, appended to the parent molecular moiety through a sulfur atom.
  • the term “membered ring” embraces any cyclic structure.
  • the term “membered” is meant to denote the number of skeletal atoms that constitute the ring.
  • non-aromatic 5, 6, 7, 8, 9, 10, 1 1 or 12- bicyclic heterocycle means a heterocycloalkyl, as defined herein, consisting of two carbocyclic rings, fused together at the same carbon atom (forming a spiro structure) or different carbon atoms (in which two rings share one or more bonds), having 5 to 12 atoms in its overall ring system, wherein one or more atoms forming the ring is a heteroatom.
  • Illustrative examples of non-aromatic 5, 6, 7, 8, 9, 10, 1 1 , or 12- bicyclic heterocycle ring include, but are not limited to, 2-azabicyclo[2.2. l ]heptanyl,
  • hydroxyalkylene as used herein, means at least one hydroxy 1 group, as defined herein, is appended to the parent molecular moiety through an alkylene group, as defined herein.
  • hydroxyalkylene include, but not limited to hydroxymethylene,
  • moiety refers to a specific segment or functional group of a molecule. Chemical moieties are often recognized chemical entities embedded in or appended to a molecule.
  • isocyanato refers to a group of formula -NCO.
  • thiocyanato refers to a group of formula -CNS.
  • isothiocyanato refers to a group of formula -NCS.
  • substituent "R" appearing by itself and without a number designation refers to a substituent selected from among from alkyl, cycloalkyl, aryl, heteroaryl (bonded through a ring carbon) and non-aromatic heterocycle (bonded through a ring carbon).
  • substituted means that the referenced group is optionally substituted
  • protecting group refers to a removable group which modifies the reactivity of a functional group, for example, a hydroxyl, ketone or amine, against undesirable reaction during synthetic procedures and to be later removed.
  • hydroxy-protecting groups include, but not limited to, methylthiomethyl, tert-dimethylsilyl, tert-butyldiphenylsilyl, ethers such as
  • ketone protecting groups include, but not limited to, ketals, oximes, O-substituted oximes for example O-benzyl oxime, O-phenylthiornethyl oxime, 1 -isopropoxycyclohexyl oxime, and the like.
  • amine protecting groups include, but are not limited to, tert-butoxycarbonyl (Boc) and carbobenzyloxy (Cbz).
  • stereoisomers exist as stereoisomers, wherein asymmetric or chiral centers are present.
  • Stereoisomers are designated (R) or (S) depending on the configuration of substituents around the chiral carbon atom.
  • the term (R) and (S) used herein are configurations as defined in IUPAC 1974 Recommendations for Section E, Fundamental
  • Stereochemistry Pure Appl. Chem., ( 1976), 45: 13-30, hereby incorporated by reference.
  • Stereoisomers include enantiomers, diastereomers, and mixtures of enantiomers or diastereomers.
  • individual stereoisomers of compounds are prepared synthetically from commercially available starting materials which contain asymmetric or chiral centers or by preparation of racemic mixtures followed by resolution.
  • the methods and formulations described herein include the use of iV-oxides, crystalline forms (also known as polymorphs), or pharmaceutically acceptable salts of compounds described herein, as well as active metabolites of these compounds having the same type of activity.
  • compounds exist as tautomers. All tautomers are included within the scope of the compounds presented herein.
  • the compounds described herein exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like. The solvated forms of the compounds presented herein are also considered to be disclosed herein.
  • the compounds described herein are synthesized using any synthetic techniques including standard synthetic techniques and the synthetic processes described herein. In specific embodiments, the following synthetic processes are utilized.
  • compounds of Formula (I), composing of la to If are prepared in various ways, as outlined in Synthetic Schemes 1 and 2.
  • the variables e.g., R] , R 2 . R 3 , R4, R 5 , Y, and Z
  • R is alkyl
  • Y' is the same or different group defined by Y
  • Z' is the same or different group defined by Z.
  • compounds are synthesized using methodologies analogous to those described below by the use of appropriate alternative starting materials.
  • compounds of Formula (la, and lb) wherein Y is identical to Z are synthesized according to Synthetic Scheme 1.
  • the preparation of the intermediate 3 wherein R5 is hydrogen is achieved by condensation of 4-aminoisobenzofuran- l(3H)-one 1 with an aldehyde 2 in the presence of a base preferably alkaline alkoxides in appropriate solvents such as ethyl acetate or ethyl propionate at either ambient or elevated temperature.
  • Compounds of Formula la wherein R 5 is hydrogen is prepared by treating the intermediate 3 with hydrazine hydrate at ambient or elevated temperature.
  • compounds of Formula ( Ic, and Id) are synthesized according to Synthetic Scheme 2.
  • the intermediate 5 is prepared by condensation of the reagent 1 with an aldehyde 4 in the presence of water absorbent such sodium sulfate or magnesium sulfate at elevated temperature.
  • a subsequent condensation reaction of this intermediate with another aldehyde in the presence of a base preferably alkaline alkoxides in appropriate solvents such as ethyl acetate or ethyl propionate at either ambient or elevated temperature gives the intermediate 6 wherein R 5 is hydrogen.
  • Compounds of Formula Ic wherein R 5 is hydrogen is prepared by treating the intermediate 6 with hydrazine hydrate at ambient or elevated temperature.
  • the term “selective binding compound” refers to a compound that selectively binds to any portion of one or more target proteins.
  • the term “selectively binds” refers to the ability of a selective binding compound to bind to a target protein, such as, for example, PARP, with greater affinity than it binds to a non-target protein.
  • specific binding refers to binding to a target with an affinity that is at least about 10, about 50, about 100, about 250, about 500, about 1000 or more times greater than the affinity for a non-target.
  • target protein refers to a molecule or a portion of a protein capable of being bound by a selective binding compound.
  • a target protein is the enzyme poly(ADP-ribose)polymerase (PARP).
  • treating encompass either or both responsive and prophylaxis measures, e.g. , designed to inhibit, slow or delay the onset of a symptom of a disease or disorder, achieve a full or partial reduction of a symptom or disease state, and/or to alleviate, ameliorate, lessen, or cure a disease or disorder and/or its symptoms.
  • amelioration of the symptoms of a particular disorder by administration of a particular compound or pharmaceutical composition refers to any lessening of severity, delay in onset, slowing of progression, or shortening of duration, whether permanent or temporary, lasting or transient that can be attributed to or associated with administration of the compound or composition.
  • a modulator refers to a compound that alters an activity of a molecule.
  • a modulator includes a compound that causes an increase or a decrease in the magnitude of a certain activ ity of a molecule compared to the magnitude of the activity in the absence of the modulator.
  • a modulator is an inhibitor, which decreases the magnitude of one or more activities of a molecule.
  • an inhibitor completely prevents one or more activities of a molecule.
  • a modulator is an activator, which increases the magnitude of at least one activity of a molecule.
  • the presence of a modulator results in an activity that does not occur in the absence of the modulator.
  • selective modulator refers to a compound that selectively modulates a target activity.
  • PARP refers to the family of the enzyme poly(ADP- ribose)polymerase which includes approximately 18 proteins, particularly poly(ADP- ribose)polymerase- I (PARP- 1 ) and poly(ADP-ribose)polymerase-2 (PARP-2).
  • the term “selective PARP modulator” refers to a compound that selectively modulates at least one activity associated with the enzyme poly(ADP-ribose)polymerase (PARP). In various embodiments, the selective modulator selectively modulates the activity of PARP - 1 , PARP-2, both PARP- 1 and PARP-2 or several members of the family of the enzyme poly(ADP- ribose)polymerase (PARP).
  • the term “method of inhibiting PARP” refers to a method of inhibiting the activity of either one or more of the family of enzyme poly(ADP-ribose)polymerase (PARP).
  • the term “inhibition of PARP” refers to inhibition of the activity of either one or more of the family of enzyme poly(ADP-ribose)polymerase (PARP).
  • modulating the activity of the enzyme poly(ADP- ribose)polymerase refers to a modulating the activity of either one or more of the family of enzyme poly(ADP-ribose)polymerase (PARP).
  • the term "selectively modulates” refers to the ability of a selective modulator to modulate a target activity to a greater extent than it modulates a non-target activity.
  • the target activity is selectively modulated by, for example about 2 fold up to more that about 500 fold, in some embodiments, about 2, 5, 10, 50, 100, 150, 200, 250, 300, 350, 400, 450 or more than 500 fold.
  • cancer refers to an abnormal growth of cells which tend to proliferate in an uncontrolled way and, in some cases, to metastasize (spread).
  • types of cancer include, but are not limited to, solid tumors (such as those of the bladder, bowel, brain, breast, endometrium, heart, kidney, lung, lymphatic tissue (lymphoma), ovary, pancreas or other endocrine organ (thyroid), prostate, skin (melanoma) or hematological tumors (such as the leukemias MDS and AML).
  • carrier refers to relatively nontoxic chemical compounds or agents that facilitate the incorporation of a compound into cells or tissues.
  • co-administration or the like, as used herein, are meant to encompass administration of the selected therapeutic agents to a single patient, and are intended to include treatment regimens in which the agents are administered by the same or different route of administration or at the same or different time.
  • dilute refers to chemical compounds that are used to dilute the compound of interest prior to delivery. Diluents include chemicals used to stabilize compounds because they provide a more stable environment. Salts dissolved in buffered solutions (which also can provide pH control or maintenance) are utilized as diluents in certain embodiments, including, but not limited to a phosphate buffered saline solution.
  • an "effective amount” or “therapeutically effective amount,” as used herein, refer to a sufficient amount of an agent or a compound being administered which will relieve to some extent one or more of the symptoms of the disease or condition being treated. The result includes reduction and/or alleviation of the signs, symptoms, or causes of a disease, or any other desired alteration of a biological system.
  • an "effective amount” for therapeutic uses is the amount of the composition comprising a compound as disclosed herein required to provide a clinically significant decrease in disease symptoms.
  • An appropriate "effective" amount in any individual case is determined using any suitable technique, such as a dose escalation study.
  • the terms “enhance” or “enhancing,” as used herein, means to increase or prolong either in potency or duration a desired effect.
  • the term “enhancing” refers to the ability to increase or prolong, either in potency or duration, the effect of other therapeutic agents on a system.
  • An “enhancing-effective amount,” as used herein, refers to an amount adequate to enhance the effect of another therapeutic agent in a desired system.
  • enzymes refers to unstable or degradable linkages which are degraded by one or more enzymes.
  • PARP-mediated refers to conditions or disorders that are ameliorated by the one or more of the family of enzyme poly(ADP-ribose)polymerase (PARP).
  • a “metabolite” of a compound disclosed herein is a derivative of that compound that is formed when the compound is metabolized.
  • active metabolite refers to a biologically active derivative of a compound that is formed when the compound is metabolized.
  • metabolized refers to the sum of the processes (including, but not limited to, hydrolysis reactions and reactions catalyzed by enzymes) by which a particular substance is changed by an organism. Thus, in certain instances, enzymes produce specific structural alterations to a compound. In some embodiments, metabolites of the compounds disclosed herein are identified either by administration of compounds to a host and analysis of tissue samples from the host, or by incubation of compounds with hepatic cells in vitro and analysis of the resulting compounds.
  • module means to interact with a target either directly or indirectly so as to alter the activity of the target, including, by way of example only, to enhance the activity of the target, to inliibit the activity of the target, to limit the activity of the target, or to extend the activity of the target.
  • nontoxic and non-abrogative materials refers materials that when administered to an individual do not cause substantial, undesirable biological effects and/or do not interact in a deleterious manner with any of the components of the composition in which it is contained.
  • pharmaceutically acceptable salt refers to a formulation of a compound that does not cause significant irritation to an organism to which it is administered and does not abrogate the biological activity and properties of the compound.
  • pharmaceutically acceptable salts are obtained by reacting a compound described herein, with acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, methanesuifonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid and the like.
  • pharmaceutically acceptable salts are obtained by reacting a compound having acidic group described herein with a base to form a salt such as an ammonium salt, an alkali metal salt, such as a sodium or a potassium salt, an alkaline earth metal salt, such as a calcium or a magnesium salt ⁇ a salt of organic bases such as dicyclohexylamine, TV-methyl-D-glucamine,
  • tris(hydroxymethyl)methylamine and salts with amino acids such as arginine, lysine, and the like, or by other methods previously determined.
  • the term "pharmaceutical combination” as used herein, means a product that results from the mixing or combining of more than one active ingredient and includes both fixed and non-fixed combinations of the active ingredients.
  • the term "fixed combination” means that the active ingredients, e.g. a compound described herein and a co-agent, are both administered to a patient simultaneously in the form of a single entity or dosage.
  • the term “non-fixed combination” means that the active ingredients, e.g. a compound described herein and a co-agent, are administered to a patient as separate entities either simultaneously, concurrently or sequentially with no specific intervening time limits, wherein such administration provides effective levels of the two compounds in the body of the patient.
  • cocktail therapy e.g. the administration of three or more active ingredients.
  • composition refers to a mixture of a compound described herein with other chemical components, such as carriers, stabilizers, diluents, dispersing agents, suspending agents, thickening agents, and/or excipients.
  • the pharmaceutical composition facilitates administration of the compound to an organism. Multiple techniques of administering a compound exist in the art including, but not limited to: intravenous, oral, aerosol, parenteral, ophthalmic, pulmonary and topical administration.
  • a prodrug refers to an agent that is converted into the parent drug in vivo. Prodrugs are often useful because, in some situations, they are easier to administer than the parent drug. In certain instances, a prodrug is bioavailable by oral administration whereas the parent is not. In some instances, a prodrug has improved solubility in pharmaceutical compositions over the parent drug.
  • An example, without limitation, of a prodrug is a compound described herein, which is administered as an ester (the "prodrug") to facilitate transmittal across a cell membrane where water solubility is detrimental to mobility but which then is metabolically hydrolyzed to the carbo.xylic acid, the active entity, once inside the cell where water-solubility is beneficial.
  • a further example of a prodrug might be a short peptide (polyaminoacid) bonded to an acid or amino group where the peptide is metabolized to reveal the active moiety.
  • a prodrug upon in vivo administration, is chemically converted to the biologically, pharmaceutically or therapeutically more active form of the compound.
  • a prodrug is enzymatically metabolized by one or more steps or processes to the biologically, pharmaceutically or therapeutically active form of the compound.
  • a pharmaceutically active compound is modified such that the active compound will be regenerated upon in vivo administration.
  • the prodrug is designed to alter the metabolic stability or the transport characteristics of a drug, to mask side effects or toxicity, to improve the flavor of a drug or to alter other characteristics or properties of a drug.
  • subject or “patient” encompasses mammals and non-mammals.
  • mammals include, but are not limited to, any member of the Mammalian class: humans, non-human primates such as chimpanzees, and other apes and monkey species; farm animals such as cattle, horses, sheep, goats, swine; domestic animals such as rabbits, dogs, and cats; laboratory animals including rodents, such as rats, mice and guinea pigs, and the like.
  • non-mammals include, but are not limited to, birds, fish and the like.
  • the mammal is a human.
  • treat include alleviating, abating or ameliorating a disease or condition symptoms, preventing additional symptoms, ameliorating or preventing the underlying metabolic causes of symptoms, inhibiting the disease or condition, e.g. , arresting the development of the disease or condition, relieving the disease or condition, causing regression of the disease or condition, relieving a condition caused by the disease or condition, or stopping the symptoms of the disease or condition either prophylactically and/or therapeutically.
  • compositions that include a compound described herein and a pharmaceutically acceptable diluent(s), excipient(s), and/or carrier(s).
  • the compounds described herein are administered as pharmaceutical
  • compositions in which compounds described herein are mixed with other active ingredients, as in combination therapy are mixed with other active ingredients, as in combination therapy.
  • a pharmaceutical composition refers to a mixture of a compound described herein with other chemical components, such as carriers, stabilizers, diluents, dispersing agents, suspending agents, thickening agents, and/or excipients.
  • a pharmaceutical composition facilitates administration of the compound to an organism.
  • practicing the methods of treatment or use provided herein includes administering or using a pharmaceutical composition comprising a therapeutically effective amount of a compound provided herein.
  • the methods of treatment provided for herein include administering such a pharmaceutical composition to a mammal having a disease or condition to be treated. In one embodiment, the mammal is a human.
  • the therapeutically effective amount varies widely depending on the severity of the disease, the age and relative health of the subject, the potency of the compound used and other factors.
  • the compounds described herein are used singly or in combination with one or more therapeutic agents as components of mixtures.
  • the pharmaceutical compositions provided herein are formulated for intravenous injections.
  • the intravenous injection formulations provided herein are formulated as aqueous solutions, and, in some embodiments, in physiologically compatible buffers such as Hank's solution, Ringer's solution, or physiological saline buffer.
  • the pharmaceutical compositions provided herein are formulated for transmucosal administration.
  • transmucosal formulations include penetrants appropriate to the barrier to be permeated.
  • the pharmaceutical compositions provided herein are formulated for other parenteral injections, appropriate formulations include aqueous or nonaqueous solutions, and in one embodiment, with physiologically compatible buffers or excipients.
  • the pharmaceutical compositions provided herein are formulated for oral administration.
  • the oral formulations provided herein comprise compounds described herein that are formulated with pharmaceutically acceptable carriers or excipients.
  • Such carriers enable the compounds described herein to be formulated as tablets, powders, pills, dragees, capsules, liquids, gels, syrups, elixirs, slurries, suspensions and the like, for oral ingestion by a patient to be treated.
  • pharmaceutical preparations for oral use are obtained by mixing one or more solid excipient with one or more of the compounds described herein, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores.
  • Suitable excipients include, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as: for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methylceliulose, microcrystalline cellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose; or others such as: polyvinylpyrrolidone (PVP or povidone) or calcium phosphate.
  • disintegrating agents are optionally added, such as the cross-linked croscarmellose sodium, polyvinylpyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • a pharmaceutical composition formulated as dragee cores with suitable coatings is provided herein.
  • concentrated sugar solutions are used in forming the suitable coating, and optionally contain gum arabic, talc, polyvinylpyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • dyestuffs and/or pigments are added to tablets, dragees and/or the coatings thereof for, e.g. , identification or to characterize different combinations of active compound doses.
  • pharmaceutical preparations which are used include orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol.
  • the push-fit capsules contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • the active compounds are dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • stabilizers are optionally added.
  • the formulations for oral administration are in dosages suitable for such administration.
  • the pharmaceutical compositions provided herein are formulated for buccal or sublingual administration.
  • buccal or sublingual compositions take the form of tablets, lozenges, or gels formulated in a conventional manner.
  • parenteral injections involve bolus injection or continuous infusion.
  • formulations for injection are presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative.
  • the pharmaceutical composition described herein is in a form suitable for parenteral injection as a sterile suspensions, solutions or emulsions in oily or aqueous vehicles, and optionally contains formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • compositions for parenteral administration include aqueous solutions of the active compounds in water-soluble form.
  • suspensions of the active compounds are prepared as appropriate oily injection suspensions.
  • Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes.
  • aqueous injection suspensions contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran.
  • the suspensions also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
  • the active ingredient is in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen- free water, before use.
  • the compounds described herein are administered topically.
  • the compounds described herein are formulated into a variety of topically administrable compositions, such as solutions, suspensions, lotions, gels, pastes, medicated sticks, balms, creams or ointments.
  • Such pharmaceutical compounds optionally contain solubilizers, stabilizers, tonicity enhancing agents, buffers and/or preservatives.
  • the pharmaceutical compositions provided herein are formulated for transdermal administration of compounds described herein.
  • administration of such compositions employs transdermal delivery devices and transdermal delivery patches.
  • the compositions are lipophilic emulsions or buffered, aqueous solutions, dissolved and/or dispersed in a polymer or an adhesive.
  • patches include those constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents.
  • transdermal delivery of the compounds described herein is accomplished by use of iontophoretic patches and the like.
  • transdermal patches provide controlled delivery of the compounds provided herein, such as, for example, compounds of Formula (I) or (II).
  • the rate of absorption is slowed by using rate-controlling membranes or by trapping the compound within a polymer matrix or gel.
  • absorption enhancers are optionally used to increase absorption.
  • Absorption enhancer and carrier include absorbable pharmaceutically acceptable solvents that assist in passage of the compound through the skin.
  • transdermal devices are in the form of a bandage comprising a backing member, a reservoir containing the compound optionally with carriers, optionally a rate controlling barrier to deliver the compound to the skin of the host at a controlled and predetermined rate over a prolonged period of time, and means to secure the device to the skin.
  • the pharmaceutical compositions provided herein are formulated for administration by inhalation.
  • the compounds described herein are in a form as an aerosol, a mist or a powder.
  • pharmaceutical compositions described herein are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebuliser, with the use of a suitable propellant, e.g. , dichlorodifluoromethane, trichlorofluoromethane,
  • the dosage unit is determined by providing a valve to deliver a metered amount.
  • capsules and cartridges of, such as, by way of example only, gelatin for use in an inhaler or insufflator is formulated containing a powder mix of the compound described herein and a suitable powder base such as lactose or starch.
  • the compounds described herein are formulated in rectal compositions such as enemas, rectal gels, rectal foams, rectal aerosols, suppositories, jelly suppositories, or retention enemas.
  • rectal compositions optionally contain conventional suppository bases such as cocoa butter or other glycerides, as well as synthetic polymers such as polyvinylpyrrolidone, PEG, and the like.
  • a low-melting wax such as, but not limited to, a mixture of fatty acid glycerides, optionally in combination with cocoa butter is first melted.
  • the pharmaceutical compositions are formulated in a conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into pharmaceutically acceptable preparations.
  • proper formulation is dependent upon the route of
  • compositions comprising a compound described herein are manufactured in a conventional manner, such as, by way of example only, by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or compression processes.
  • the pharmaceutical compositions include at least one pharmaceutically acceptable carrier, diluent or excipient and a compound described herein described herein as an active ingredient in free-acid or free-base form, or in a pharmaceutically acceptable salt form.
  • the methods and pharmaceutical compositions described herein include the use of iV-oxides, crystalline forms (also known as polymorphs), as well as active metabolites of these compounds having the same type of activity.
  • compounds described herein exist as tautomers. All tautomers are included within the scope of the compounds presented herein.
  • the solvated and unsolvated forms of the compounds described herein include those that are solvated with pharmaceutically acceptable solvents such as water, ethanol, and the like.
  • the solvated forms of the compounds presented herein are also considered to be disclosed herein.
  • the pharmaceutical compositions described herein include other medicinal or pharmaceutical agents, carriers, adjuvants, such as preserving, stabilizing, wetting or emulsifying agents, solution promoters, salts for regulating the osmotic pressure, and/or buffers.
  • the pharmaceutical compositions described herein also contain other therapeutically valuable substances.
  • compositions containing the compounds described herein include formulating the compounds with one or more inert, pharmaceutically acceptable excipients or carriers to form a solid, semi-solid or liquid.
  • Solid compositions include, but are not limited to, powders, tablets, dispersible granules, capsules, cachets, and suppositories.
  • Liquid compositions include solutions in which a compound is dissolved, emulsions comprising a compound, or a solution containing liposomes, micelles, or nanoparticles comprising a compound as disclosed herein.
  • Semisolid compositions include, but are not limited to, gels, suspensions and creams.
  • compositions are in liquid solutions or suspensions, solid forms suitable for solution or suspension in a liquid prior to use, or as emulsions.
  • These compositions optionally contain minor amounts of nontoxic, auxiliary substances, such as wetting or emulsifying agents, pH buffering agents, and so forth.
  • a composition comprising a compound described herein takes the form of a liquid where the agents are present in solution, in suspension or both.
  • a first portion of the agent is present in solution and a second portion of the agent is present in particulate form, in suspension in a liquid matrix.
  • a liquid composition includes a gel formulation.
  • the liquid composition is aqueous.
  • Useful aqueous suspension optionally contain one or more polymers as suspending agents.
  • Useful polymers include water-soluble polymers such as cellulosic polymers, e.g., hydroxypropyl methylcellulose, and water-insoluble polymers such as cross-linked carboxyl- containing polymers.
  • Useful compositions optionally comprise an mucoadhesive polymer, selected for example from carboxymethylcellulose, carbomer (acrylic acid polymer),
  • poly(methylmethacrylate), polyacrylamide, polycarbophil, acrylic acid/butyl acrylate copolymer, sodium alginate and dextran are examples of poly(methylmethacrylate), polyacrylamide, polycarbophil, acrylic acid/butyl acrylate copolymer, sodium alginate and dextran.
  • compositions optionally include solubilizing agents to aid in the solubility of a compound described herein.
  • solubilizing agent generally includes agents that result in formation of a micellar solution or a true solution of the agent.
  • Solubilizing agents include certain acceptable nonionic surfactants, for example polysorbate 80, and ophthalmically acceptable glycols, polyglycols, e.g. , polyethylene glycol 400, and glycol ethers.
  • compositions optionally include one or more pH adjusting agents or buffering agents, including acids such as acetic, boric, citric, lactic, phosphoric and hydrochloric acids; bases such as sodium hydroxide, sodium phosphate, sodium borate, sodium citrate, sodium acetate, sodium lactate and tris-hydroxymethylaminomethane; and buffers such as citrate/dextrose, sodium bicarbonate and ammonium chloride.
  • acids such as acetic, boric, citric, lactic, phosphoric and hydrochloric acids
  • bases such as sodium hydroxide, sodium phosphate, sodium borate, sodium citrate, sodium acetate, sodium lactate and tris-hydroxymethylaminomethane
  • buffers such as citrate/dextrose, sodium bicarbonate and ammonium chloride.
  • Such acids, bases and buffers are included in an amount required to maintain pH of the composition in an acceptable range.
  • Useful compositions optionally include one or more salts in an amount required to bring osmolality of the composition into an acceptable range.
  • Such salts include those having sodium, potassium or ammonium cations and chloride, citrate, ascorbate, borate, phosphate, bicarbonate, sulfate, thiosulfate or bisulfite anions; suitable salts include sodium chloride, potassium chloride, sodium thiosulfate, sodium bisulfite and ammonium sulfate.
  • compositions optionally include one or more preservatives to inhibit microbial activity.
  • Suitable preservatives include mercury-containing substances such as merfen and thiomersai; stabilized chlorine dioxide; and quaternary ammonium compounds such as benzalkonium chloride, cetyltrimethylammonium bromide and cetylpyridinium chloride.
  • compositions optionally include one or more surfactants to enhance physical stability or for other purposes.
  • Suitable nonionic surfactants include polyoxyethylene fatty acid glycerides and vegetable oils, e.g. , polyoxyethylene (60) hydrogenated castor oil; and
  • compositions optionally one or more antioxidants to enhance chemical stability where required. Suitable antioxidants include, by way of example only, ascorbic acid and sodium metabisulfite.
  • aqueous suspension compositions are packaged in single-dose non-reclosable containers.
  • multiple-dose reclosable containers are used, in which case it is typical to include a preservative in the composition.
  • any delivery system for hydrophobic pharmaceutical compounds is employed.
  • Liposomes and emulsions are examples of delivery vehicles or carriers for hydrophobic drugs.
  • certain organic solvents such as N-methylpyrrolidone are employed.
  • the compounds are delivered using a sustained-release system, such as semipermeable matrices of solid hydrophobic polymers containing the therapeutic agent.
  • sustained-release materials are utilized in the embodiments herein.
  • sustained-release capsules release the compounds for a few weeks up to over 100 days.
  • additional strategies for protein stabilization are employed.
  • the formulations or compositions described herein benefit from and/or optionally comprise antioxidants, metal chelating agents, thiol containing compounds and other general stabilizing agents.
  • stabilizing agents include, but are not limited to: (a) about 0.5% to about 2% w/v glycerol, (b) about 0.1% to about 1 % w/v methionine, (c) about 0.1 % to about 2% w/v monothioglycerol, (d) about 1 mM to about 10 mM EDTA, (e) about 0.01 % to about 2% w/v ascorbic acid, (f) 0.003% to about 0.02% w/v polysorbate 80, (g) 0.001 % to about 0.05% w/v.
  • polysorbate 20 (h) arginine, (i) heparin, (j) dextran sulfate, (k) cyclodextrins, (1) pentosan polysulfate and other heparinoids, (m) divalent cations such as magnesium and zinc; or (n) combinations thereof.
  • the effective amount of the compound is administered by ophthalmic administration;
  • any of the aforementioned embodiments are further embodiments that include single administrations of the effective amount of the compound, including further embodiments in which the compound is administered to the subject (i) once; (ii) multiple times over the span of one day; (iii) continually; or (iv) continuously.
  • any of the aforementioned embodiments are further embodiments that include multiple administrations of the effective amount of the compound, including further embodiments wherein;
  • the method includes a drug holiday, wherein the administration of the compound is temporarily suspended or the dose of the compound being administered is temporarily reduced; at the end of the drug holiday, dosing of the compound is resumed.
  • the length of the drug holiday varies from 2 days to 1 year.
  • the compounds described herein are used in the preparation or manufacture of medicaments for the treatment of diseases or conditions that are mediated by the enzyme poly(ADP-ribose)polymerase (PARP) or in which inhibition of the enzyme poly(ADP- ribose)polymerase (PARP) ameliorates the disease or condition.
  • a method for treating any of the diseases or conditions described herein in a subject in need of such treatment involves administration of pharmaceutical compositions containing at least one compound described herein, or a pharmaceutically acceptable salt, pharmaceutically acceptable N-oxide, pharmaceutically active metabolite, pharmaceutically acceptable prodrug, or pharmaceutically acceptable solvate thereof, in therapeutically effective amounts to said subject.
  • the compositions containing the compound(s) described herein are administered for prophylactic and/or therapeutic treatments.
  • the compositions are administered to a patient already suffering from a disease or condition, in an amount sufficient to cure or at least partially arrest the symptoms of the disease or condition.
  • amounts effective for this use will depend on the severity and course of the disease or condition, previous therapy, the patient's health status, weight, and response to the drugs, and the judgment of the treating physician. In certain instances, it is considered appropriate for the caregiver to determine such therapeutically effective amounts by routine experimentation (including, but not limited to, a dose escalation clinical trial).
  • compositions containing the compounds described herein are administered to a patient susceptible to or otherwise at risk, of a particular disease, disorder or condition.
  • the amount administere is defined to be a "prophylactically effective amount or dose.”
  • the precise amounts of compound administered depend on the patient's state of health, weight, and the like.
  • effective amounts for this use will depend on the severity and course of the disease, disorder or condition, previous therapy, the patient's health status and response to the drugs, and the judgment of the treating physician.
  • a patient's condition does not improve or does not significantly improve following administration of a compound or composition described herein and, upon the doctor's discretion the administration of the compounds is optionally administered chronically, that is, for an extended period of time, including throughout the duration of the patient's life in order to ameliorate or otherwise control or limit the symptoms of the patient's disease or condition.
  • the length of the drug holiday varies between 2 days and 1 year, including by way of example only, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 10 days, 12 days, 15 days, 20 days, 28 days, 35 days, 50 days, 70 days, 100 days, 120 days. 1 0 days, 180 days, 200 days, 250 days, 280 days, 300 days, 320 days, 350 days, or 365 days.
  • the dose reduction during a drug holiday includes a reduction from about 10% to about 100%, including, by way of example only, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100%.
  • a maintenance dose is administered if necessary.
  • the dosage e.g., of the maintenance dose, or the frequency of administration, or both, are reduced, as a function of the symptoms, to a level at which the improved disease, disorder or condition is retained.
  • patients are optionally given intermittent treatment on a long-term basis upon any recurrence of symptoms.
  • the amount of a given agent that corresponds to an effective amount varies depending upon factors such as the particular compound, disease or condition and its severity, the identity (e.g., weight) of the subject or host in need of treatment.
  • the effective amount is, nevertheless, determined according to the particular circumstances surrounding the case, including, e.g., the specific agent that is administered, the route of
  • doses employed for adult human treatment is in the range of about 0.02 to about 5000 mg per day, in a specific embodiment about 1 to about 1500 mg per day.
  • the desired dose is conveniently presented in a single dose or as divided doses administered simultaneously (or over a short period of time) or at appropriate intervals, for example as two, three, four or more sub-doses per day.
  • the pharmaceutical compositions described herein are in a unit dosage form suitable for single administration of precise dosages.
  • the formulation is divided into unit doses containing appropriate quantities of one or more compound.
  • the unit dosage is in the form of a package containing discrete quantities of the formulation.
  • Non-limiting examples are packaged tablets or capsules, and powders in vials or ampoules.
  • aqueous suspension compositions are packaged in single- dose non-reclosable containers.
  • multiple-dose reciosable containers are used, in which case it is typical to include a preservative in the composition.
  • formulations for parenteral injection are, in some embodiments, presented in unit dosage form, which include, but are not limited to ampoules, or in multi-dose containers, with an added preservative.
  • the daily dosages appropriate for the compounds described herein described herein are from about 0.01 to about 2.5 mg/kg per body weight.
  • an indicated daily dosage in the larger subject including, but not limited to, humans, is in the range from about 0.5 mg to about 100 mg, conveniently administered in divided doses, including, but not limited to, up to four times a day or in extended release form.
  • suitable unit dosage forms for oral administration comprise from about 1 to about 50 mg active ingredient.
  • the dosages are altered depending on a number of variables, not limited to the activity of the compound used, the disease or condition to be treated, the mode of administration, the requirements of the individual subject, the severity of the disease or condition being treated, and the judgment of the practitioner.
  • toxicity and therapeutic efficacy of such therapeutic regimens are determined by standard pharmaceutical procedures in cell cultures or experimental animals, including, but not limited to, the determination of the LD 50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population).
  • the dose ratio between the toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio between LD5 0 and ED 50 .
  • compounds exhibiting high therapeutic indices are preferred.
  • the data obtained from cell culture assays and animal studies is used in formulating a range of dosage for use in human.
  • the dosage of such compounds lies within a range of circulating concentrations that include the ED 50 with minimal toxicity.
  • the dosage varies within this range depending upon the dosage form employed and the route of administration utilized.
  • the therapeutic effectiveness of one of the compounds described herein is enhanced by administration of an adjuvant (i.e., in some embodiments, by itself the adjuvant has minimal therapeutic benefit, but in combination with another therapeutic agent, the overall therapeutic benefit to the patient is enhanced).
  • the benefit experienced by a patient is increased by administering one of the compounds described herein with another therapeutic agent (which also includes a therapeutic regimen) that also has therapeutic benefit.
  • the overall benefit experienced by the patient as a result of a combination treatment is additive or synergistic.
  • therapeutically-effective dosages vary when the drugs are used in treatment combinations.
  • therapeutically-effective dosages of drugs and other agents for use in combination treatment regimens is determined in any suitable manner, e.g., through the use of metronomic dosing, i.e., providing more frequent, lower doses in order to minimize toxic side effects.
  • combination treatment regimen described herein encompass treatment regimens in which administration of a PA P inhibitor described herein is initiated prior to, during, or after treatment with a second agent described above, and continues until any time during treatment with the second agent or after termination of treatment with the second agent.
  • Combination treatment further includes periodic treatments that start and stop at various times to assist with the clinical management of the patient.
  • a PARP inhibitor described herein in the combination treatment is administered weekly at the onset of treatment, decreasing to biweekly, and decreasing further as appropriate.
  • compositions and methods for combination therapy are provided herein.
  • the pharmaceutical compositions disclosed herein are used to in a method of treating a PARP mediated disease or condition or a disease or condition that is ameliorated by inhibition of PARP.
  • the pharmaceutical compositions disclosed herein are used to treat vascular disease; septic shock; ischaemic injury; reperfusion injury; neurotoxicity; hemorrhagic shock; inflammatory diseases; multiple sclerosis; secondary effects of diabetes; and acute treatment of cytotoxicity following cardiovascular surgery.
  • the pharmaceutical compositions disclosed herein are used in combination, either simultaneously or sequentially, with ionizing radiation or one or more chemotherapeutic agents.
  • combination therapies described herein are used as part of a specific treatment regimen intended to provide a beneficial effect from the co-action of a PARP inhibitor described herein and a concurrent treatment. It is understood that the dosage regimen to treat, prevent, or ameliorate the condition(s) for which relief is sought, is optionally modified in accordance with a variety of factors.
  • dosages of the co-administered compounds vary depending on the type of co-drug employed, on the specific drug employed, on the disease or condition being treated and so forth.
  • the compound provided herein when co-administered with one or more biologically active agents, is administered either
  • the attending physician will decide on the appropriate sequence of administering protein in combination with the biologically active agent(s).
  • the multiple therapeutic agents are administered in any order or even simultaneously.
  • administration is simultaneous and the multiple therapeutic agents are, optionally, provided in a single, unified form, or in multiple forms (by way of example only, either as a single pill or as two separate pills).
  • one of the therapeutic agents is given in multiple doses, or both are given as multiple doses.
  • administration is not simultaneous and the timing between the multiple doses varies, by way of non-limiting example, from more than zero weeks to less than four weeks.
  • the combination methods, compositions and formulations are not to be limited to the use of only two agents; the use of multiple therapeutic combinations are also envisioned.
  • the compounds described herein are used in combination with procedures that provide additional or synergistic benefit to the patient.
  • patients are expected to find therapeutic and/or prophylactic benefit in the methods described herein, wherein pharmaceutical composition of a compound disclosed herein and/or combinations with other therapeutics are combined with genetic testing to determine whether that individual is a carrier of a mutant gene that is known to be correlated with certain diseases or conditions.
  • the compounds described herein and combination therapies are administered before, during or after the occurrence of a disease or condition.
  • the timing of administering the composition containing a compound varies.
  • the compounds are used as a prophylactic and are administered continuously to subjects with a propensity to develop conditions or diseases in order to prevent the occurrence of the disease or condition.
  • the compounds and compositions are administered to a subject during or as soon as possible after the onset of the symptoms.
  • the administration of the compounds is initiated within the first 48 hours of the onset of the symptoms, within the first 6 hours of the onset of the symptoms, or within 3 hours of the onset of the symptoms.
  • the initial administration is achieved via any route practical, such as, for example, an intravenous injection, a bolus injection, infusion over 5 minutes to about 5 hours, a pill, a capsule, transdermal patch, buccal delivery, and the like, or combination thereof.
  • a compound is administered as soon as is practicable after the onset of a disease or condition is detected or suspected, and for a length of time necessary for the treatment of the disease, such as, from about 1 month to about 3 months.
  • the length of treatment varies for each subject, and the length is determined using any criteria.
  • a compound or a formulation containing the compound is administered for at least 2 weeks, for about 1 month to about 5 years, or for about 1 month to about 3 years.
  • methods for treatment of PARP mediated conditions or diseases include administration to a patient compounds, pharmaceutical compositions, or medicaments described herein in combination with at least one additional agent selected from the group consisting of alemtuzumab, arsenic trioxide, asparaginase (pegylated or non-), bevacizumab, cetuximab, platinum-based compounds such as cisplatin, cladribine, daunorubicin/doxombicin/idarubicin, irinotecan, fludarabine, 5-fluorouracil, gemtuzumab, methotrexate, PaclitaxelTM, taxol, temozolomide, thioguanine, or classes of drugs including hormones (an antiestrogen, an antiandrogen, or gonadotropin releasing hormone analogues, interferons such as alpha interferon, nitrogen mustards such as bus
  • mechlorethamine retinoids such as tretinoin
  • topoisomerase inhibitors such as irinotecan or topotecan
  • tyrosine kinase inhibitors such as gefinitinib or imatinib
  • agents to treat signs or symptoms induced by such therapy including allopurinol, filgrastim, granisetron/ondansetron/palonosetron, and dronabinol.
  • the compounds, pharmaceutical compositions, or medicaments described herein are administered in combination with ionizing radiation, one or more
  • chemotherapeutic agents or a combination thereof.
  • the chemotherapeutic agent is independently selected from the group consisting of alemtuzumab, arsenic trioxide, pegylated asparaginase, non-pegylated
  • the compounds, pharmaceutical compositions, or medicaments described herein are administered in combination with one or more of chemotherapeutic agents where each chemotherapeutic agent is independently an alkylating agent or a topoisomerase-1 inhibitor.
  • the compounds, pharmaceutical compositions, or medicaments described herein are administered in combination with one or more chemotherapeutic agents where each chemotherapeutic agent is independently selected from the group consisting of methyl methanesulfonate, temozolomide, dacarbazine (DTIC), Topotecan, Irinotecan, Rubitecan, Exatecan, Lurtotecan, Gimetecan, Diflomotecan (homocamptothecins), 7-substituted non-silatecans, 7-silyl camptothecins, BNP 1350, and XR 1 1576/MLN 576.
  • chemotherapeutic agents is independently selected from the group consisting of methyl methanesulfonate, temozolomide, dacarbazine (DTIC), Topotecan, Irinotecan, Rubitecan, Exatecan, Lurtotecan, Gimetecan, Diflomotecan (homocamptothecin
  • the compounds, pharmaceutical compositions, or medicaments described herein are administered in combination with one chemotherapeutic agent where the chemotherapeutic agent is irinotecan, cisplatin, or temozolomide.
  • Examples 1 -4 were disclosed in a poster which was presented December 4, 2010, at the 52nd American Society of Hematology Annual Meeting and Exposition in Orlando, Florida.
  • Example 1 PARP Inhibitor Sensitivity in MDS/AML Cell Lines Correlates with the Presence of High Grade Microsatellite Instability (MSI)
  • FIG. 1 shows that MDS/AML cell lines exhibited a varied response to PARP inhibitor, Compound 1.
  • Cell lines, P39, KG- 1 and Molm-13 exhibited significant cytotoxicity to Compound 1
  • MDS/AML cell lines Molt 4, NB4 and normal peripheral blood lymphocytes exhibited relative insensitivity to Compound 1 .
  • MDS/ AML cell lines were obtained from American Type Culture Collection (ATCC).
  • a panel of six mononucleotide repeat sequence markers (NR21, NR22, NR24, Bat25, Mono27 and Bat26) were used to estimate MSI in each cell line.
  • TABLE 1 presents mononucleotide repeat sequence (NR21, NR22, NR24, Bat25, Mono27, and Bat26) versus MDS/AML cell line (PBL, HL60, Jurkat, KG- 1 , Molm- 13, Molt 4, NB4, OCI-AML3, P39, Raji, REH, DS- 1 , U937 and DLD 1 ).
  • Values refer to the size of fluorescent PCR product run in an ABI Genetic Analyzer 3130XL (bp, base pair). Underlined and bolded values were PCR products with > 3 bp deletions or additions.
  • Cell lines were classified as one of four groups: ( 1 ) MSI-stable with no aberrant MSI; (2) MSI-low with one aberrant microsatellite; (3) MSI-mid with > 2 aberrant microsatellite loci; and (4) MSI-high (high grade MSI) with > 3 aberrant microsatellite loci (e.g. , P39, KG- 1 , NB4 and Molm- 13).
  • Double stranded D A repair genes Mrel 1 , A TM, CTiP, CHKl , PTEN, BLM and A TR possess specific microsatellites that when mutated produce frameshift mutations. Using fluorescent PCR and Sanger sequencing, MSI at these loci in a panel of MDS/AML cell lines were tested.
  • TABLE 2 presents the MSI data for double stranded DNA repair genes (Rad50, Mrel I, CTiP, CHKl, BLM, PTEN, A TM and A TR) versus MDS/AML cell line (PBL, KG- 1 , Molm- 13, Molt 4, NB4, P39 and DLD 1 ). Values refer to the size of fluorescent PCR product run in an ABI Genetic Analyzer 3130XL (bp, base pair). Underlined and bolded values were PCR products with > 3 bp deletions or additions.
  • High risk MDS is associated with chromosomal instability, an increased likelihood of AML transformation and poor prognosis.
  • High risk MDS patients were tested using a panel of six mononucleotide repeat sequence markers (NR21, NR22, NR24, Bat25, Mono27 and Bat2 €) by Sanger sequencing and fluorescent PCT MSI analysis, and were further tested for chromosomal instability and mutations in their CTiP DNA repair genes. Cytogenetics were determined using Giemsa staining at metaphase spreads. Single Nucleotide Polymorphisms Analysis (SNPA) was used to determine cryptic chromosomal abnormalities, e.g., uniparental disomy (UPD), not seen using conventional cytogenetic analysis.
  • SNPA Single Nucleotide Polymorphisms Analysis
  • This example provides an exemplary screening of MDS/AML patients for PARP inhibitor therapy.
  • Microsatellite markers in genomic DNA of a sample from a MDS/AML patient may be isolated and compared to microsatellite markers in genomic DNA isolated from normal tissue or cells in the patient, or in a healthy reference individual, in order to detect MSI.
  • microsatellite loci may be amplified and genotyped. Specific procedures used in this example are provided below.
  • the sample may be a histological sample, a biopsy, or a cytological sample such as, e.g., a smear, a swab, a wash, a body fluid containing.
  • the sample may comprise a fixed or preserved specimen, such as a cytological or a histological specimen.
  • the sample may be a tumor biopsy tissue obtained by surgical resection.
  • PCR and Microsatellite Analysis A panel of five nearly monomorphic mononucleotide markers (NR21, NR22, NR24, Bat25, Bat26 and Mono27) may be used for MSI determination of the patient's genomic DNA. The analysis may involve fluorescent polymerase chain reaction (PCR) and/or Sanger sequencing.
  • PCR fluorescent polymerase chain reaction
  • Immunohistochemistry Conventional cytogenetic may be used to analyze for chromosomal abnormalities in the patient's sample.
  • Single Nucleotide Polymorphisms Analysis (SNPA) may be used to analyze for cryptic chromosomal abnormalities in the patient's sample.
  • MSI comprising a > 3 bp deletion or insertion in one or more microsatellite(s) of the patient's malignant DNA, correlating with chromosomal instability, indicates that the patient is likely to respond to treatment with a compound of Formula I or II.
  • Example 6 Phase II Clinical Trial of the Safety and Efficacy of Compounds of
  • This example provides an exemplary phase II trial which purpose is to study the side effects and best dose of a compound of Formula (I) or (II) and to determine how well it works in treating MDS/AML patients with MSI in malignant cells.
  • PARP Poly(ADP-ribose) polymerase
  • o Measurable disease as defined by RECIST criteria and measured by X-ray, CT scan, or MRI
  • DDO Development Office
  • Samples will be analyzed for tumor marker measurements, plasma levels of a compound of Formula (I) or (II) via liquid chromatography/mass spectrometry/mass spectrometry, PARP activity, and PARP protein expression via western blotting immunoassays.
  • Paraffin embedded sections from original diagnostic biopsy are also collected and analyzed for PARP protein expression via immunohistochemical technique.
  • Pleural and ascitic fluid may be collected and analyzed for DNA DS break repair proficiency via immunohistochemical technique.
  • Some patients will also undergo biopsy of tumors and samples will be analyzed for DNA repair enzyme mutation status, PARP activity via validated PARP immunoblotting assay, and chromosomal status using conventional cytogenetic analysis and SNPA analysis
  • PARP Poly(ADP-ribose) polymerase

Abstract

Disclosed herein are compounds of Formula (I) where R1, R2, R3, R4, R5, A, B, Y, and Z are as defined herein for use in the treatment of MDS or AML. Also disclosed are the methods of identifying a patient with MDS or AML who is sensitive to treatment with a PARP inhibitor, in particular a compound of Formula (I).

Description

USE OF DIHYDROPYRIDOPHTHALAZINONE INHIBITORS OF POLY (ADP-RIBOSE)
POLYMERASE
(PARP) IN THE TREATMENT OF MYELODYSPLASTIC SYNDROME (MDS) AND ACUTE
MYELOID
FIELD OF THE INVENTION
[0001 ] Described herein are methods of using dihydropyridophthalazinone inhibitors of Poly(ADP-Ribose)Polymerase (PARP) to treat or prevent Myelodysplastic Syndrome (MDS) and Acute Myeloid Leukaemia (AML). Also described herein are methods of determining a subject's sensitivity to such PARP inhibitors, based on their Microsatellite Instability (MSI) status.
BACKGROUND OF THE INVENTION
[0002] The family of poly(ADP-ribose)polymerases (PARP) includes approximately 18 proteins, which all display a certain level of homology in their catalytic domain but differ in their cellular functions (Ame et al., BioEssays, 26(8), 882-893 (2004)). PARP- 1 and PARP-2 are unique members of the family, in that their catalytic activities are stimulated by the occurrence of DNA strand breaks.
[0003] PARP has been implicated in the signaling of DNA damage through its ability to recognize and rapidly bind to DNA single or double strand breaks (D'Amours, et al., Biochem. J., 342, 249-268 ( 1999)). It participates in a variety of DNA-related functions including gene amplification, cell division, differentiation, apoptosis, DNA base excision repair as well as effects on telomere length and chromosome stability (d'Adda di Fagagna, et al., Nature Gen., 23( 1 ), 76-80 ( 1999)).
|0004] xMyelodysplastic Syndrome (MDS) and Acute Myeloid Leukaemia (AML) are characterized by chromosomal instability, and leukemic cells have inherent defects in double strand DNA repair (Gaymes, et al., Cancer Res. , 62, 2791 -2797 (2002)). Furthermore, a study of PARP inhibitors on AML cell lines showed that a proportion ( 15%) of cells derived from the AML patients demonstrated significant cytotoxic responses to PARP inhibitors (Gaymes, et al., Haematologica, 94(5), 638-646 (2009)).
[0005] Microsatellite Instability (MSI) can be defined as an elevated rate of uncorrected replication errors in microsatellite sequences ( Han, et al., Science, 97, 226-234 (2006)). Disruption of the mismatch repair machinery results in MSI and loss of mismatch repair and subsequent MSI is observed in 12% of colorectal cancers. MSI in other cancers are less well characterized, but reports show the presence of mismatch repair defects and MSI in haematological maglinancies (Han. et al., Science, 97, 226-234 (2006); Gu, et al., Oncogene, 21 , 5758-5764 (2002)). As microsatellite sequences appear in the exons of prominent DSB DNA repair genes, VISI has the potential to inflict frameshift mutations in genes that confer PARP inhibitor sensitivity. [0006] PARP inhibitors and use of PARP inhibitors for treatment of certain cancers are known in the art, as described previously in International Patent Publication No. WO 2010/017055 A2, incorporated herein by reference in its entirety. Methods of DNA repair pathway profiling and analyzing MSI are also known in the art, as described previously in Gaymes et al., Clin, Cancer Res., 1 2, 5104-51 1 1 (2006), incorporated herein by reference in its entirety. Additional PARP inhibitor therapies are sought for certain cancers, e.g.. MDS and AML, which are associated with MSI. A method for screening of candidates for PARP inhibitor therapy through DNA repair pathway profiling and analysis of MSI is also sought.
SUMMARY OF THE INVENTION
[0007] In one aspect, described herein is a method of treating Myelodysplastic Syndrome (MDS) or Acute Myeloid Leukemia (AML) in a patient with MDS or AML, comprising administering to a patient with MDS or AML a therapeutically effective amount of a compound of Formula (I):
Figure imgf000003_0001
Formula (I),
wherein:
Y and Z are each independently selected from the group consisting of:
a) an aryl group optionally substituted with 1 , 2, or 3 R ,;
b) a heteroaryl group optionally substituted with 1 , 2, or 3 and
c) a substituent independently selected from the group consisting of hydrogen, alkenyl, alkoxy, alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, alkyl, alkynyl, arylalkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, hydroxyalkylene, oxo, heterocycloalkyl, heterocycloalkylalkyl, alkylcarbonyl, arylcarbonyl, heteroarylcarbonyl alkylsulfonyl, arylsulfonyl,
heteroarylsulfonyl, (NRARB)alkylene, (NRARB)carbonyl, (NRARB)carbonylalkylene, (NRARB)sulfonyl, and (NRARB)sulfonylalkylene;
Ri , Ri, and R3 are each independently selected from the group consisting of hydrogen, halogen, alkenyl, alkoxy, alkoxycarbonyl, alkyl, cycloalkyl, alkynyl, cyano, haloalkoxy, haloalkyl, hydroxyl, hydroxyalkylene, nitro, NRARB, NRARBalkylene, and (NRARB)carbonyl;
A and B are each independently selected from the group consisting of hydrogen, Br, CI, F, I, OH, C Cf,aikyl, CrCgcycloalky!, alkoxy, and alkoxyalkyl, wherein C C6alkyl, C Cscycloaikyl, aikoxy, and alkoxyalkyl are optionally substituted with at least one substituent selected from the group consisting of OH, N02, CN, Br, CI, F, I, C, -C6alkyl, and C3-C3cycloalkyl, wherein B is not OH;
RA. and RB are independently selected from the group consisting of hydrogen, alkyl, cycloalkyl, and alkylcarbonyl; or RA and RB taken together with the atom to which they are attached form a 3- 10 membered heterocycle ring optionally having one to three heteroatoms or hetero functionalities selected from the group consisting of -0-, -NH, -N(CrC6-alkyl)-, -NCO(C Cft-alkyl)-,
-N(aryl)-, -N(aryl-CrC6-alkyl-)-, -N(substituted-aryl-C, -C6-alkyl-)-, -N(heteroaryl)-, -N(heteroaryl-
CrC6-alkyl-)-, -N(substituted-heteroaryl-C| -C6-alkyl-)-, and -S- or S(0)q- , wherein q is 1 or 2 and the
3- 10 membered heterocycle ring is optionally substituted with one or more substituents;
R4 and R5 are each independently selected from the group consisting of hydrogen, alkyl, cycloalkyl, alkoxyalkyl, haloalkyl, hydroxyalkylene, and (NRARB)alkylene; and
each R<, is selected from the group consisting of OH, N02, CN, Br, CI, F, 1. C C6alkyl, C>
Cgcycloalkyl, C2-C8heterocycloalkyl, C2-C5alkenyl, alkoxy, alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C -C0alkynyl, aryl, arylalkyl, C3-C8cycloalkylalkyl, haloalkoxy, haloalkyl, hydroxyalkylene, oxo, heteroaryl, heteroarylalkoxy, heteroaryloxy, heteroarylthio,
heteroarylalkylthio, heterocycloalkoxy, C2-C3heterocycloalkylthio, heterocyclooxy, heterocyclothio,
NRARB, (NRARB)C, -C6alkylene, (NRARB)carbonyl, (NRARB)carbonylalkylene, (NRARB)sulfonyl, and
(NRARB)sulfonylalkylene.
[0008] In certain embodiments, the compound is administered as a single isomer, stereoisomer, or enantiomer, or mixture thereof.
[0009] In certain embodiments, the compound is administered as a salt, solvate, chemically protected form or prodrug thereof.
[0010] In certain embodiments, the compound is administered with at least one of a
pharmaceutically acceptable diluent, excipient, and carrier.
[0011 ] In certain embodiments, the malignant cells of the patient exhibit Microsatellite
Instability (MSI) in a microsatellite. In certain embodiments, the malignant cells contain an insertion or deletion in at least one microsatellite. In some embodiments, the malignant cells of the patient exhibit MSI in two, three, four, five, six, seven or more microsatellites. In certain embodiments, the MSI in the at least one microsatellite comprises a plurality of insertions and/or deletions. In certain embodiments, the MSI in the at least one microsatellite may be detected using primers designed to detect insertion or deletion in the microsatellite. In some embodiments, the at least one microsatellite is NR2I, NR22, NR24, Bat25, Bat26. Mono27 and/or a microsatellite as described herein in the DNA repair genes CTiP, Mrel l, A TM, Rad50, Chkl , BLM, PTEN, A TM and ATR. In certain embodiments, the at least one microsatellite is selected from the group consisting o(NR2l, NR22, Bat25, Bat26, Mono27 and a microsatellite as described herein in the DNA repair genes CTiP, Mrell and ATM. In certain embodiments, the at least one microsatellite is selected from the group consisting of NR21, NR24 and Bat25. In certain embodiments, the MSI comprises deletion or insertion of 3 or more base pairs in the microsatellite(s). In certain embodiments, MSI comprises insertion(s) or deletion(s) of base pairs in microsatellite(s) that lead to inactivation of DNA repair function and/or results in a frameshift mutation in a double stranded DNA repair gene expression product. In certain embodiments, the DNA repair function is governed by a double stranded DNA repair gene selected from the group consisting oi Mrel l, ATM and CTiP. In certain embodiments, the method further comprises analyzing for MSI in at least one microsatellite of a patient's malignant cell DNA.
[0012] In certain embodiments, the compound is 5-fluoro-8-(4-fluorophenyl)-9-( l -methyl- 1H- l ,2,4-triazol-5-yl)-8,9-dihydro-2H-pyrido[4,3,2-de]phthalazin-3(7H)-one, or a pharmaceutically acceptable salt thereof.
[0013] In certain embodiments, described is a method of treating AML in a patient with AML, comprising administering to a patient with AML a therapeutically effective amount of a compound of Formula (I) or stereoisomer or a pharmaceutically acceptable salt thereof. In certain embodiments, the compound is 5-fluoro-8-(4-fluorophenyl)-9-( 1 -methyl- 1H- 1 ,2,4-triazol-5-yl)-8,9-dihydro-2H- pyrido[4,3,2-de]phthalazin-3(7H)-one, or a stereoisomer or a pharmaceutically acceptable salt thereof.
[0014] In another aspect, described herein is a compound of Formula (I), or a salt, solvate, single isomer, stereoisomer, or enantiomer, or mixture thereof, or chemically protected form or prodrug thereof, for use in the treatment of MDS or AML in a patient with MDS or AML. In certain embodiments, the malignant cells in the patient exhibit MSI in a microsatellite. In certain embodiments, the malignant cells in the patient exhibit MSI in at least two, three, four, five, six or more microsatellites.
[0015] In another aspect, described herein is a use of a compound of Formula (I), or a salt, solvate, single isomer, stereoisomer, or enantiomer, or mixture thereof, or chemically protected form or prodrug thereof, in the formulation of a medicament for the treatment of MDS or AML in a patient with MDS or AML. In certain embodiments, the malignant cells in the patient exhibit MSI in a microsatellite. In certain embodiments, the malignant cells in the patient exhibit MSI in at least two, three, four, five, six or more microsatellites.
[0016] In another aspect, described herein is a method for identifying MSI in an MDS or AML patient. In certain embodiments, the method comprises analyzing for MSI in at least one
microsatellite in a patient's malignant cell DNA. In certain embodiments, the MSI comprises three or more inserted or deleted base pairs in the at least one microsatellite. In certain embodiments, the presence of MSI in the at least one microsatellite in the patient's malignant cell DNA identifies the patient's malignancy as likely to be sensitive to therapy Compound of Formula I. In certain embodiments, the MSI in the at least one microsatellite may be detected using a primer designed to detect insertion or deletion in the microsatellite. In certain embodiments, the at least one microsatellite is NR21, NR22, NR24, Bat25, Bat26, Mono27 and/or a microsatellite as described herein in the DNA repair genes CTiP, Mrel l, A TM, RacI50, Chkl, BLM, PTEN, A TM and A TR. In certain embodiments, the at least one microsatellite is NR21, NR22, Ba(25, Bat26, Mono27, or a microsatellite as described herein in the DNA repair genes CTiP, Mrel l md A TM. In some embodiments, the at least one microsatellite is selected from the group consisting of NR21, NR24 and Bat25. In certain embodiments, the microsatellite(s) that contain insertion(s) or deletion(s) of base pairs lead to inactivation of DNA repair function. In certain embodiments, the DNA repair function is governed by a double stranded DNA repair gene selected from the group consisting of Mrell, A TM and CTiP.
|0017) In another aspect, described herein is a method for identifying an MDS or AML patient likely to respond to treatment with a compound of Formula (I). In certain embodiments, the compound of Formula (I) is 5-fluoro-8-(4-fluorophenyl)-9-( 1 -methyl- 1 H- l , 2,4-triazol-5-yl)-8,9- dihydro-2H-pyrido[4,3,2-de]phthalazin-3(7H)-one. In certain embodiments, the method comprises analyzing for MSI in at least one microsatellite(s) in a patient's malignant cell DNA. In certain embodiments, the MSI comprises three or more inserted or deleted base pairs in at least one microsatellite in the patient's malignant cell DNA. In certain embodiments, the presence of MSI in at least one microsatellite of the patient's malignant cell DNA indicates that the patient's malignancy is likely to be sensitive to treatment with a compound of Formula (I). In certain embodiments, the MSI in the at least one microsatellite may be detected using a primer designed to detect insertion or deletion in the microsatellite. In certain embodiments, the at least one microsatellite is NR21 , NR22, NR24, Bat25, Bat26, Mono27 and/or a microsatellite as described herein in the DNA repair genes CTiP, Mrel l, A TM, RadSO, Chkl, BLM, PTEN, A TM and A TR. In certain embodiments, the at least one microsatellite is NR21, NR22, Bat25, Bat26, Mono27, or a microsatellite as described herein in the DNA repair genes CTiP, Mrel l and ATM. In certain embodiments, the at least one microsatellite is selected from the group consisting of NR21, NR24 and Bat25. In certain embodiments, the at least one microsatellite contains insertion(s) or deletion(s) of base pairs leads to inactivation of DNA repair function. In certain embodiments, the DNA repair function is governed by a double stranded DNA repair gene selected from the group consisting of Mrell, A TM and CTiP. In certain embodiments, the method further comprises cytogenetic analysis of chromosomal abnormalities in the patient's malignant cell DNA.
DESCRIPTION OF THE FIGURES
|0018] FIG. 1. Response to PARP inhibition in MDS/ AML cell lines. Varying concentrations of Compound 1 were added to MDS/AML cell lines and peripheral blood lymphocytes (PBL) continuously for 12- 14 days. Soft agar clonogenic assays were used to determine cell survival. [0019J FIG. 2. Composite microsatellite analysis of monucleotide repeat NR21 (21 consecutive thymines in 5'UTR of the solute carrier gene, SLC7A8, shown underlined in legend) in cell lines NB4, KG- 1 and P39; comparison to normal PBL cell line.
[0020] FIG. 3. Composite microsatellite analysis of monucleotide repeat Mono27 (27 consecutive intronic adenines in MAP4K3, shown underlined in legend) in cell lines KG- 1 , Molm- 13, Moit 4, NB4 and P39; comparison to normal PBL cell line.
[0021 ] FIG. 4. Composite microsatellite analysis of monucleotide repeat CTiP (9 consecutive thymines in exon 1 1 , shown underlined in legend) in MSI-high cell lines Molm-13, KG-1 and P39; comparison to normal PBL cell line. Fluorescent PCR image shown on left, Sanger sequence on right.
[0022] FIG. 5. Composite microsatellite analysis of monucleotide repeat Mrel l ( 1 1 consecutive intronic thymines, shown underlined in legend) in MSI-high cell lines KG- 1 , NB4, P39 and Molm- 13; comparison to normal PBL cell line. Fluorescent PCR image shown on left, Sanger sequence on right.
[0023] FIG. 6. Composite microsatellite analysis of monucleotide repeat Mrell (1 1 consecutive intronic thymines, shown underlined in legend) in MSI-stable cell lines HL60, Jurkat, DNA ligase IV" ' (DNL), OCI-AML3 and U937. Fluorescent PCR image shown on left, Sanger sequence on right.
[0024] FIG. 7. Composite microsatellite analysis of monucleotide repeat A TM( 15 consecutive intronic thymines, shown underlined in legend) in cell lines Jurkat, KG- 1 , DNA ligase IV ' (DNL), NB4, P39, Molm- 13 and OCI-AML3. Fluorescent PCR image shown on left, Sanger sequence on right.
[0025] FIG. 8. PCR for exon 8 of A TM was performed on the cDNA of cell lines DNL, Jurkat, K562, KG- 1 , Molm- 13, Molt 4, NB4 and P39. The ratio of intensity (RI) of aberrant to wild-type bands was measured using Adobe Photoshop™.
10026] FIG. 9. PCR for exon 5 of Mrel l was performed on the cDNA of cell lines DNL, Jurkat, K562, KG-1 , Molm-13, Molt 4, NB4 and P39. The ratio of intensity (RI) of aberrant to wild-type bands was measured using Adobe Photoshop1 M.
[0027] FIG. 10. Composite microsatellite analysis of CTiP (9 consecutive thymines in exon 1 1 , shown underlined in legend) in 5 patients with high risk MDS. Fluorescent PCR image shown on left, Sanger sequence on right. DETAILED DESCRIPTION OF THE INVENTION
Compounds of Formula (I) or Formula (II)
[0028] In certain embodiments, disclosed herein are methods of treating Myelodysplastic Syndrome (MDS) or Acute Myeloid Leukemia (AML) in a patient with MDS or AML, comprising administering to a patient with MDS or AML a therapeutically effective amount of a compound having the structure of Formula (1) or Formula (II).
[0029] In certain embodiments, the methods comprise administering to the patient
phannaceutically acceptable salts, solvates, esters, acids and chemically protected forms or prodrugs thereof of the compounds having the structure of Formula (I) and Formula (11).
|0030] In certain embodiments, the methods comprise administering to to the patient isomers including single isomers, stereoisomers, enantiomers, diastereoisomers, or mixtures thereof, of compounds having the structure of Formula (I) and Formula (II).
[0031 ] Formula (1) is as follows:
Figure imgf000008_0001
Formula (I)
wherein:
Y and Z are each independently selected from the group consisting of:
a) an aryl group optionally substituted with 1 , 2, or 3 ¾ ;
b) a heteroaryl group optionally substituted with 1 , 2, or 3 R6;
c) a substituent independently selected from the group consisting of hydrogen, alkenyl, alkoxy, alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, alkyl, alkynyl, arylalkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, hydroxyalkylene, oxo, heterocycloalkyl, heterocycloalkylalkyl, alkylcarbonyl, arylcarbonyl, heteroarylcarbonyl alkylsulfonyl, arylsulfonyl,
heteroarylsulfonyl, (NRARs)alkylene. (NRARB)carbonyl, (NRARB)carbonylalkylene,
(NRARB)sulfonyl, and (NRAR8)sulfonylalkylene;
Ri , Ri, and R¾ are each independently selected from the group consisting of hydrogen, halogen, alkenyl, alkoxy, alkoxycarbonyl, alkyl, cycloalkyl, alkynyl, cyano, haloalko y, haloalkyl, hydroxyl, hydroxyalkylene, nitro, ARB, NRARBalkylene, and (N RARB)carbonyl;
A and B are each independently selected from hydrogen, Br, CI, F, I, OH, C, -C„a!ky 1, C3-C8cycloalkyl, alkoxy, alkoxyalkyl wherein CrCAalkyl, C3-CRcycloalkyl, alkoxy, alkoxyalkyl are optionally substituted with at least one substituent selected from OH, N02, CN, Br, CI, F, I, CrC6alkyl, and C3- Qcycloalkyl, wherein B is not OH;
RA, and RB re independently selected from the group consisting of hydrogen, alkyl, cycloalkyl, and alkylcarbonyl; or RA and RB taken together with the atom to which they are attached form a 3- 10 membered heterocycle ring optionally having one to three heteroatoms or hetero functionalities selected from the group consisting of -0-, -NH, -N(C,-C6-alkyl)-, -NCO(C C6-alkyl)-, -N(aryl)-, -N(aryl-C|-C6-alkyl-)-, -N(substituted-aryl-C,-C6-alkyl-)-, -N(heteroaryl)-, -N(heteroaryl-C C6- alkyl-)-, -N(substituted-heteroaryl-C|-C6-alkyl-)-, and -S- or S(0)q- , wherein q is 1 or 2 and the 3- 10 membered heterocycle ring is optionally substituted with one or more substituents;
R and R5 are each independently selected from the group consisting of hydrogen, alkyl, cycloalkyl, alkoxyalkyl, haloalkyl, hydroxyalkylene, and (NRARB)alkylene; and
each ¾ is selected from the group consisting of OH, N02, CN, Br, CI, F, I, C C6alkyl, C3- Cscycloalkyl, Ci-Cgheterocycloalkyl, C2-C6alkenyl, alkoxy, alkoxyalkyl, alkoxycarbonyl,
alkoxycarbonylalkyl, C2-CAalkynyl, aryl, arylalkyl, CrCxcycloalkylalkyl, haloalkoxy, haloalkyl, hydroxyalkylene, oxo, heteroaryl, heteroarylalkoxy, heteroaryloxy, heteroarylthio, heteroarylalkylthio, heterocycloalkoxy, C2-Qheterocydoalkylthio, heterocyclooxy, heterocyclothio, NRARB, (NRARB)C Qalkylene, (NRARB)carbonyl, (NRARB)carbonylalkylene, (NRARB)sulfonyl, and
(NRARB)sulfonylalkylene; or
and isomers, salts, solvates, chemically protected forms, and prodrugs thereof.
[0032] Formula (II) is as follows:
Figure imgf000009_0001
Formula (II);
wherein:
Y is an aryl or heteroaryl group optionally substituted with at least one Re;
Z is an aryl group optionally substituted with at least one R6;
A and B are each independently selected from hydrogen, Br, CI, F, I, OH, CrC6alkyl, C3-C8cycloaikyi, alkoxy, alkoxyalkyl wherein C C6alkyl, C3-C8cycloalkyl, alkoxy, alkoxyalkyl are optionally substituted with at least one substituent selected from OH, N02, CN, Br, CI, F, I, C C6alkyl, and C3- Qcycloalkyl, wherein B is not OH;
R6 is selected from OH, N02, CN, Br, CI, F, I, CrC5alkyl, C3-C8cycIoalkyl, C2-C8heterocycloalkyl; CVCeaikenyl, alkoxy, alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C2-C6alkynyl, aryl, arylalkyl, CrQcycloalkylalkyl, haloalkoxy, haloalkyl, hydroxyalkylene, oxo, heteroaryl, heteroarylalkoxy, heteroaryloxy, heteroarylthio, heteroarylalkylthio, heterocycloalkoxy, CV
CHheterocycloalkylthio, heterocyclooxy, heterocyclothio, NRARB, (NRARB)C i-C6alkylene,
(NRARB)carbonyl, (NRARB)carbonylalkylene, (NRARB)sulfonyl, and (NRARB)sulfonylalkylene; i is selected from hydrogen. Br, CI, I, or F;
RA, and RB are independently selected from the group consisting of hydrogen, C i-C6alkyl, C
Cgcycloalkyl, and alkylcarbonyl; or RA and RB taken together with the atom to which they are attached form a 3- 10 membered heterocycle ring optionally having one to three heteroatoms or hetero functionalities selected from the group consisting of -0-, -NH, -N(Ci-C6alkyl)-, -NCO(CrC6alkyl)-, -NCO(C3-C3cycloaikyI)-, -N(aryl)-, -N(aryl-C, -C6alkyi-)-. -N(substituted-aryl-C, -C6alkyi-)-, -N(heteroaryl)-, -N(heteroaryl-CrC6alkyl-)-, -N(substituted-heteroaryl-C|-C6alkyl-)-, and -S- or S(0)q- , wherein q is 1 or 2 and the 3- 10 membered heterocycle ring is optionally substituted with one or more substituents; or a pharmaceutically acceptable salt, solvate or prodrug thereof.
|0033] In certain embodiments the compounds administered in the methods provided herein are compounds of Formula (I) or a therapeutically acceptable salt thereof, wherein R| , Ri, R3 are independently selected from a group consisting of hydrogen, alkyl, and halogen; R4 IS hydrogen and R5 is selected from the group consisting hydrogen, alkyl, cycloalkyl, alkoxyalkyl, haloalkyl, hydroxyalkylene, and (NRARB)alkylene; RA. and RB are independently selected from the group consisting of hydrogen, alkyl, cycloalkyl, and alkylcarbonyl; or RA and RB taken together with the atom to which they are attached form a 3- 10 membered heterocycle ring optionally having one to three heteroatoms or hetero functionalities selected from the group consisting of -0-,
-NH, -N(C, -C6-alkyl)-, -NCO(C,-C6-aIkyl)-, -N(aryl)-, -N(aryl- C, -C6-aIkyI-)-, -N(subsfituted-aryl- C | -C6-alkyl-)-, -N(heteroaryl)-, -N(heteroaryl-C C6-alkyl-)-, -N(substituted-heteroaryl-C C6-alkyl-)-, and -S- or S(0)q- , wherein q is 1 or 2 and the 3- 10 membered heterocycle ring is optionally substituted with one or more substituents.
[0034] In certain embodiments the compounds administered in the methods provided herein are compounds of Formula (I) wherein Y is an aryl group. In another embodiment the aryl group is a phenyl group. In yet another embodiment the phenyl group is substituted with at least one R, selected from Br, CI, F, or I. In one embodiment R, is F. In one embodiment the phenyl group is substituted with at least one R, selected from (NRARB)CrC6alkylene, (NRARB)carbonyl,
(NRARB)carbonylalkylene, ( RARn)sulfonyl, and (NRARB)sulfonylalkylene. In one embodiment R„ is (NRARB)C i-Cftalkylene. In another embodiment Ci-C6alkyl is selected from methylene, ethylene, n-propylene, iso-propylene, n-butylene, iso-butylene, and tert-butylene. In yet another embodiment C, -C6alkyl is methylene. In yet a further embodiment RA and RB are each independently hydrogen, C| -C6alkyl, or C3-C3cycIoalkyl. In one embodiment CrC6alkyl is selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl. In one embodiment Cj -Qalkyl is methyl. In another embodiment C| -C6alkyl is ethyl. In yet another embodiment C3-C8cycloalkyl is cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl. In a further embodiment C3-C8cycloalkyl is cyclopropyl. In yet a further embodiment R6 is hydroxyalkylene. In one embodiment hydroxyalkylene is selected from CH2OH, CH2CH2OH, CH2CH2CH2OH, CH(OH)CH3, CH(OH)CH2CH3, CH2CH(OH)CH3, and CH2CH2CH2CH2OH. In another embodiment RA and RB taken together with the nitrogen to which they are attached form a 6 membered heterocycle ring having 1 heteroatom or hetero functionality selected from the group consisting of -0-, -NH, or -N(CrC6alkyI). In yet another embodiment the hetero functionality is -N(CrC6alkyl). In a further embodiment C| -C6alkyl is selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl. In yet a further embodiment C C6alkyl is methyl. In one embodiment Y is a heteroaryl group optionally substituted with at least one R<,. In another embodiment the heteroaryl group is selected from furan, pyridine, pyrimidine, pyrazine, imidazole, thiazole, isothiazole, pyrazole, triazole, pyrrole, thiophene, oxazole, isoxazole, 1 ,2,4- oxadiazole, 1 ,3,4-oxadiazole, 1 ,2,4-triazine, indole, benzothiophene, benzoimidazole, benzofuran, pyridazine, 1 ,3,5-triazine, thienothiophene, quinoxaline, quinoline, and isoquinoline. In yet another embodiment the heteroaryl group is imidazole. In a further embodiment imidazole is substituted with Ci-C6alkyl selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl. In yet a further embodiment CrC6alkyl is methyl. In one embodiment the heteroaryl group is furan. In another embodiment the heteroaryl group is thiazole. In yet another embodiment the heteroaryl group is 1 ,3,4-oxadiazole. In a further embodiment heteroaryl group is substituted with C| -C6alkyl selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl. In yet a further embodiment C|-C6alkyl is methyl. In one embodiment Z is an aryl group. In another embodiment the aryl group is a phenyl group, in yet another embodiment the phenyl group is substituted with at least one R6 selected from Br, CI, F, or I. In a further embodiment R6 is F. In yet a further embodiment R6 is CI. In one embodiment the phenyl group is substituted with at least one Rg selected from
(NRARB)C rC6alkylene, (NRARB)carbonyl, (NRARB)carbonylalkylene, (NRARB)sulfonyl, and (NRARB)sulfonylalkylene. In another embodiment R6 is (NRARB)C,-C6alkylene. In yet another embodiment C C6alkyl is selected from methylene, ethylene, n-propylene, iso-propylene, n-butylene, iso-butylene, and tert-butylene. In yet a further embodiment C C6alkyl is methylene. In a further embodiment RA and RB are each independently hydrogen, CrC(,alkyl, or C3-C3cycloalkyl. In one embodiment C C6alkyl is selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl. In another embodiment Ci -C6alkyl is methyl. In yet another embodiment RA and RB taken together with the nitrogen to which they are attached form a 6 membered heterocycle ring having 1 heteroatom or hetero functionality selected from the group consisting of -0-, -NH, or -N(d - C6alkyl). In a further embodiment the hetero functionality is -N(C|-Q,alkyl). In one embodiment C Qalkyl is selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl. In yet a further embodiment C| -Cftalkyl is methyl. In one embodiment Z is a heteroaryl group optionally substituted with at least one R6. In another embodiment the heteroaryl group is selected from furan, pyridine, pyrimidine, pyrazine, imidazole, thiazole, isothiazole, pyrazole, triazole, pyrrole, thiophene, oxazole, isoxazole, 1 ,2,4-oxadiazole, 1.3,4-oxadiazole, 1 ,2,4-triazine, indole, benzothiophene, benzoimidazole, benzofuran, pyridazine, 1 ,3,5-triazine, thienothiophene, quinoxaline, quinoline, and isoquinoline. In yet another embodiment the heteroaryl group is imidazole. In a further embodiment imidazole is substituted with Ci -C6alkyl selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl. In yet a further embodiment C C6alkyl is methyl. In one embodiment the heteroaryl group is furan. In another embodiment the heteroaryl group is thiazole. In yet another embodiment the heteroaryl group is 1 ,3,4-oxadiazole. In a further embodiment heteroaryl group is substituted with C C6alkyl selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl. In yet a further embodiment G -Qalkyl is methyl. In another embodiment R2 is hydrogen. In yet another embodiment R; is selected from F, CI, Br, and I. In a further embodiment R2 is F.
10035] In certain embodiments the compounds administered in the methods provided herein are compounds of Formula (1) wherein A is hydrogen. In another embodiment A is C Cealkyl. In a further embodiment, A is selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, tert- butyl, n-pentyl, and n-hexyl. In yet another embodiment, methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, tert-butyl, n-pentyl, and n-hexyl are optionally substituted with OH, N02, CN, Br, CI, F, and 1. In a further embodiment A is methyl, in yet another embodiment, A is selected from F. C'l, Br, and I. In another embodiment, A is C3-Cscycloalkyl. In another embodiment, A is cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl. In one embodiment, A is substituted with OH, N02, or CN. In a further embodiment, B is Ci -C6alkyl. In a further embodiment, B is selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, tert-butyl, n-pentyl, and n-hexyl. In yet another embodiment, methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, tert-butyl, n-pentyl, and n-hexyl are optionally substituted with OH, N02, CN, Br, CI, F, and I. In one embodiment the administered compound is a compound of Formula ( I) wherein B is hydrogen. In a further embodiment B is methyl. In yet another embodiment, B is selected from F, CI, Br, and I. In another embodiment, B is C3- Cscycloalkyl. In another embodiment, B is cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl. In one embodiment, A is substituted with OH, N02, or CN. In a further embodiment, the administered compound is a compound of Formula ( I) wherein A is hydrogen and B is selected from Br, CI, F, I, C C6alkyl, C3-C8cycloalkyl, alkoxy, alkoxyalkyl wherein C | -C6alkyl, CrQcycloalkyl, alkoxy, alkoxyalkyl are optionally substituted with at least one substituent selected from OH, N02, CN, Br, CI. F, I, C] -C6alkyl, and C3-C3cycloalkyl. In another embodiment, the administered compound is a compound of Formula (I) wherein B is hydrogen and A is selected from Br, CI, F, I, C Chalkyl, C3- Cscycloalkyl, alkoxy, alkoxyalkyl wherein CrQalkyl, C3-C3cycloalkyl, alkoxy, alkoxyalkyl are optionally substituted with at least one substituent selected from OH, N02, CN. Br, CI, F, I, Q- C,,alkyl, and C3-C3cycloalkyl. In yet another embodiment, both A and B are hydrogen. In a further embodiment, both A and B are selected from Br, CI, F, I, C,-C6alkyl, C3-C8cycloalkyl, alkoxy, alkoxyalkyl wherein Ci-C6alkyl, C Cgcycloalkyl, alkoxy, alkoxyalkyl are optionally substituted with at least one substituent selected from OH, N02, CN, Br, CI, F, I, C|-C6alkyl, and C3-C3cycloalkyl.
[0036] In certain embodiments the compounds administered in the methods provided herein are compounds of Formula (11) wherein Y is an aryl group. In another embodiment the aryl group is a phenyl group. In yet another embodiment the phenyl group is substituted with at least one ¾, selected from Br, CI, F, or I. In one embodiment ¾ is F. In one embodiment the phenyl group is substituted with at least one ¾ selected from (NRARB)C| -C6alkylene, (NRARB)carbonyl,
(NRARB)carbonylalkylene, (NRARB)sulfonyl, and (NRARB)sulfonylalkylene. In one embodiment R6 is (NRARB)C|-C6alkylene. In another embodiment C C6alkyl is selected from methylene, ethylene, n- propylene, iso-propylene, n-butylene, iso-butylene, and tert-butylene. In yet another embodiment C,- Qalkylene is methylene. In yet a further embodiment RA and RB are each independently hydrogen, C C5alkyl, or C3-C8cycloalkyl. In one embodiment C C6alkyl is selected from methyl, ethyl, n- propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl. In one embodiment CrC<,alkyl is methyl. In another embodiment C|-C6alkyl is ethyl. In yet another embodiment C3-C8cycloalkyl is cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl. In a further embodiment C3-C8cycloalkyl is cyclopropyl. In yet a further embodiment R<, is hydroxyalkylene. In one embodiment hydroxyalkylene is selected from CH2OH, CH2CH2OH, CH2CH2CH2OH, CH(OH)CH3, CH(OH)CH2CH3, CH2CH(OH)CH3, and CH2CH2CH2CH2OH. In another embodiment RA and RB taken together with the nitrogen to which they are attached form a 6 membered heterocycle ring having 1 heteroatom or hetero functionality selected from the group consisting of -0-, -NH, or -N(Ci-Cftalkyl). In yet another embodiment the hetero functionality is -N(C C6alkyl). In a further embodiment Ci-C6alkyl is selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl. In yet a further embodiment C| -C6alkyl is methyl. In one embodiment Y is a heteroaryl group optionally substituted with at least one Re. In another embodiment the heteroaryl group is selected from furan, pyridine, pyrimidine, pyrazine, imidazole, thiazole, isothiazole, pyrazole, triazole, pyrrole, thiophene, oxazole, isoxazole, 1 ,2,4- oxadiazole, 1 ,3,4-oxadiazole, 1 ,2,4-triazine, indole, benzothiophene, benzoimidazole, benzofuran, pyridazine, 1 ,3,5-triazine, thienothiophene, quinoxaline, quinoline, and isoquinoline. In yet another embodiment the heteroaryl group is imidazole. In a further embodiment imidazole is substituted with C C6alkyl selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl. In yet a further embodiment C C6alkyl is methyl. In one embodiment the heteroaryl group is furan. In another embodiment the heteroaryl group is thiazole. In yet another embodiment the heteroaryl group is 1 ,3,4-oxadiazole. In a further embodiment heteroaryl group is substituted with CrCealkyl selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl. In yet a further embodiment C|-Cf,alkyl is methyl. In one embodiment Z is an aryl group. In another embodiment the aryl group is a phenyl group. In yet another embodiment the phenyl group is substituted with at least one R6 selected from Br, CI, F, or I. In a further embodiment R6 is F. In yet a further embodiment R<, is CI. In one embodiment the phenyl group is substituted with at least one R<, selected from
(NRARB)C|-C6alkylene, (NRARB)carbonyl, (NRARB)carbonylalkylene, (NRARB)sulfonyl, and (NRARB)sulfonylalkylene. In another embodiment R6 is (NRARB)CrC6alkylene. In yet another embodiment Ci-Cealkylene is selected from methylene, ethylene, n-propylene, iso-propylene, n-butylene, iso-butylene, and tert-butylene. In yet a further embodiment C| -C6alkyl is methylene. In a further embodiment RA and RB are each independently hydrogen, Ci-Cf)alkyl, or C3-C8cycloalkyl. In one embodiment Ci -C6alkyl is selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl. In another embodiment C C(,alkyl is methyl. In yet another embodiment RA and RB taken together with the nitrogen to which they are attached form a 6 membered heterocycle ring having 1 heteroatom or hetero functionality selected from the group consisting of -0-, -NH, or -N(Cr C6alkyl). In a further embodiment the hetero functionality is -N(Ci-C6alkyl). In one embodiment Ct - C6alkyl is selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl. In yet a further embodiment Q -Ce lkyl is methyl. In another embodiment R2 is hydrogen. In yet another embodiment R2 is selected from F, CI, Br, and I. In a further embodiment R2 is F.
{0037] In certain embodiments the compounds administered in the methods provided herein are compounds of Formula (II) wherein A is hydrogen. In another embodiment A is C|-C6alkyl. In a further embodiment, A is selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, tert- butyl, n-pentyl, and n-hexyl. In yet another embodiment, methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, tert-butyl, n-pentyl, and n-hexyl are optionally substituted with OH, N02, CN, Br, CI, F, and I. In a further embodiment A is methyl. In yet another embodiment, A is selected from F, CI, Br, and I. In another embodiment, A is C3-C8cycloalkyl. In another embodiment, A is cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl. In one embodiment, A is substituted with OH, N02, or CN. In one embodiment, the administered compound is of Formula (II) wherein B is hydrogen. In a further embodiment, B is Ci-C6alkyl. In a further embodiment, B is selected from methyl, ethyl, n- propyl, iso-propyl, n-butyl, iso-butyl, tert-butyl, n-pentyl, and n-hexyl. In yet another embodiment, methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, tert-butyl, n-pentyl, and n-hexyl are optionally substituted with OH, N02, CN, Br, CI, F, and I. In a further embodiment B is methyl. In yet another embodiment, B is selected from F, CI, Br, and I. In another embodiment, B is CrCgcycloalkyl. In another embodiment, B is cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl. In one embodiment, A is substituted with OH, N0 , or CN. In a further embodiment, the admininistered compound is a compound of Formula (II) wherein A is hydrogen and B is selected from Br, CI, F, I, CrC6alkyl, C3- C8cycloalkyl, alkoxy, alkoxyalkyl wherein Ci-C6alkyl, C3-C8cycloaIkyl, alkoxy, alkoxyalkyl are optionally substituted with at least one substituent selected from OH, NO?, CN, Br, CI, F, I, C C(,alkyl, and C3-C8cycloalkyl. In another embodiment, the administered compound is a compound of Formula (II) w herein B is hydrogen and A is selected from Br, CI, F, I, d -Cealkyl, C3-C3cycloalkyl, alkoxy, alkoxyalkyl wherein C C(,alkyl, C3-C8cycloalkyl, alkoxy, alkoxyalkyl are optionally substituted with at least one substituent selected from OH, N02, CN, Br, CI, F, I, CrC6alkyl, and C Cscycloalkyl. In yet another embodiment, both A and B are hydrogen. In a further embodiment, both A and B are selected from Br, CI, F, I, C| -C6alkyl, C C8cycloalkyl, alkoxy, alkoxyaikyl wherein C C(,alkyl, C3-C gcycloalkyl, alkoxy, alkoxyaikyl are optionally substituted with at least one substituent selected from OH, NO,, CN, Br, CI, F, I, C, -C6alkyl, and C3-C8cycloalkyl.
[0038] In certain embodiments the compound administered in the methods provided herein is a compound selected from:
Figure imgf000015_0001
or a pharmaceutically acceptable salt, solvate or prodrug thereof.
[0039] In certain embodiments the compound administered in the methods provided herein is a compound selected from:
Figure imgf000016_0001
or a pharmaceutically acceptable salt, solvate or prodrug thereof. [0040] In certain embodiments the compound administered in the methods provided herein is a compound selected from:
Figure imgf000017_0001
or a pharmaceutically acceptable salt, solvate or prodrug thereof. [0041] In certain embodiments the compound administered in the methods provided herein is a compound selected from:
Figure imgf000018_0001
or a pharmaceutically acceptable salt, solvate or prodrug thereof.
[0042] In certain embodiments the compound administered in the methods provided herein is a compound selected from:
(8S,9R)-5-fluoro-9-( l -methyl- l H-imidazoi-2-y I) -8-phenyl-8,9-dihydro-2//-pyrido[4,3,2- c e]phthalazin-3(7//)-one,
(8R,95)-5-fluoro-9-( l -methyl- lH-imidazol-2-yl) -8-phenyl-8,9-dihydro-2//-pyrido[4,3,2- </e]phthalazin-3(7H)-one,
(8S,9R)-5-fluoro-8-(4-fluorophenyl)-9-( l -methyl- lH-imidazol-2-yl) -8,9-dihydro-2H-pyrido[4,3,2- </e]phthalazin-3(7//)-one,
(8R,95)-5-fluoro-8-(4-fluorophenyl)-9-( l -methyl- lH-imidazol-2-yl)-8,9-dihydro-2H-pyrido[4,3,2- i/e]phthalazin-3(7/ )-one,
(8S,9R)- 8-(4-fluorophenyl)-9-( l -methyl- lH-imidazol-2-yl)-8,9-dihydro-2H-pyrido[4,3.2- cfe]phthalazin-3 ( 7H)-one,
(8R.9S)- 8-(4-fluorophenyl)-9-( l -methyl- lH-imidazol-2-yl)-8,9-dihydro-2H-pyrido[4,3,2- cfe]phthalazin-3 ( 7H)-one,
(85,9fl)-5-fluoro-9-( 1 -methyl- 1 H- 1 ,2,4-triazol-5-yI)-8-pheny l-8,9-dihydro-2H-pyrido[4,3,2- c/e]phthalazin-3(7//)-one,
(8R,9S)-5-fluoro-9-( l -methyI- lH- l ,2.4-triazol-5-yl)-8-phenyl-8,9-dihydro-2H-pyrido[4,3,2- i e]phthalazin-3(7H)-one, (85.9/?)-5-fluoro-8-(4-nuorophenyl)-9-(l -methyl- lH-l,2,4-triazol-5-yl)-8,9-dihydro-2H-pyrido[4,3,2- t/<?]phthalazin-3(7H)-one,
(8R,95)-5-fluoro-8-(4-fluorophenyl)-9-(l-methyl-lH-l,2,4-triazol-5-yl) -8,9-dihydro-2H- pyrido[4,3,2-cfe]phthalazin-3(7 /)-one,
(85,9/?)- 8-(4-fluorophenyl)-9-(l-methyl-lH-l,2,4-triazoI-5-yl) -8,9-dihydro-2/-pyrido[4,3,2- e]phthalazin-3(7/ )-one,
(8Λ.95)- 8-(4-fluorophenyl)-9-(l -methyl- lH-l,2,4-triazol-5-yl) -8,9-dihydro-2#-pyrido[4,3,2- ifc]phthalazin-3(7//)-one,
(8S,9y?)-5-fluoro-8-(4-fluorophenyl)-9-(l-methyl-l/y-l,2,4-triazol-5-yl) -8,9-dihydro-2H- pyrido[4,3,2-t/e]phthalazin-3(7 )-one,
(8?,95)-5-fluoro-8-(4-fluorophenyl)-9-(l-methyl-lH-l,2,4-triazol-5-yl) -8,9-dihydro-2H- pyrido[4,3,2-<sfe]phthalazin-3(7//)-one,
(85,9R)-8-(4-((dimethy lamino)methyl)phenyl)-5-iluoro-9-( 1 -methyl- 1 H- 1 ,2,4-triazol-5-y l)-8,9- dihydro-2H-pyrido[4,3,2- e]phthalazin-3(7//)-one, and
(8J?,9S)-8-(4-((dimethylamino)methyl)phenylV5-fluoro-9-(l-methyl-lH-l,2,4-triazol-5-yl)-8,9- dihydro-2H-pyrido[4,3,2-de]phthalazin-3(7H)-one,
or a pharmaceutically acceptable salt, solvate or prodrug thereof.
[0043] In certain embodiments the compound administered in the methods provided herein is of the following formula:
Figure imgf000019_0001
or a pharmaceutically acceptable salt thereof.
f0044) In certain embodiments, the compound administered in the methods provided herein is 5- fluoro-8-(4-fluorophenyl)-9-(l-methyl-lH-l,2,4-triazol-5-yl)-8,9-dihydro-2H-pyrido[4,3,2- de]phthalazin-3(7/ )-one (Compound 1), or a pharmaceutically acceptable salt thereof.
[0045] In certain embodiments, the compound administered in the methods provided herein is selected from the group consisting of:
(85,9 ?)-5-fluoro-8-(4-fluorophenyI)-9-(l-tnethyl-lH-l,2,4-triazol-5-yl)-8,9-dihydro-2H- pyrido[4,3,2-t(?]phthalazin-3(7 )-one,
(8?.95)-5-fluoro-8-(4-fluorophenyl)-9-(l-methyl-lH-l,2,4-triazol-5-yl)-8,9-dihydro-2H- pyrido[4,3,2-de]phthalazin-3(7H)-one, or a pharmaceutically acceptable salt, solvate or prodrug thereof.
[0046] In certain embodiments, the compound administered in the methods provided herein is ( 8S,9R)-5-fluoro-8-(4-fluorophenyl)-M
de]phthalazin-3(7/ )-one, or a pharmaceutically acceptable salt, solvate or prodrug thereof.
[0047] In certain embodiments, the compounds envisioned to be administered in the methods provided herein are disclosed in International Patent Publication No. WO 2010/017055 A2, which is incorporated by reference in its entirety in the instant application. Additional compounds are disclosed in International Patent Publication No. WO 2010/017055 A2.
Methods of Treatment
[0048] Provided herein are methods of treating cancer comprising administering to a subject in need of treatment a therapeutically effective amount of a compound that is a modulator of PARP activity. In certain embodiments, the methods comprise administering to a subject in need of treatment a therapeutically effective amount of a PARP inhibitor, such as, for example, a compound of Formula (1) or (II). In certain embodiments, the cancer is leukemia. In certain embodiments, the cancer is Myelodysplastic Syndrome (MDS) or Acute Myeloid Leukemia (AML). In certain embodiments, the subject is a patient with MDS or AML. In certain embodiments, the cancer is AML.
[0049] Accordingly, in one aspect, described herein is a method of treating Myelodysplastic Syndrome (MDS) or Acute Myeloid Leukemia (AML) in a patient with MDS or AML, comprising administering to a patient with MDS or AML a therapeutically effective amount of a compound of Formula (I). In certain embodiments, described herein is a method of treating Acute Myeloid Leukemia (AML) in a patient with AML, comprising administering to a patient with AML a therapeutically effective amount of a compound of Formula (I).
[0050] In one embodiment, a compound of Formula (I) or (II) is used for the treatment of MDS or AML in a patient with MDS or AML, or for the preparation of a medicament for the treatment of MDS or AML in a patient with MDS or AML. In one embodiment, a compound of Formula (I) or (II) is used for the treatment of AML in a patient with AML, or for the preparation of a medicament for the treatment of AML in a patient with AML.
[0051] Accordingly, in yet another aspect is a method of treating a cancer deficient in
Homologous Recombination (HR) dependent DNA double strand break (DSB) repair pathway, comprising administering to a subject in need of treatment a therapeutically effective amount of a PARP inhibitor, such as, for example, a compound of Formula (I) or (II). [0052] In one embodiment, a compound of Formula (I) or (II) is used for the treatment of cancer, or for the preparation of a medicament for the treatment of cancer, which is deficient in HR dependent DNA DSB repair activity.
[0053] In one embodiment the cancer comprises one or more cancer cells having a reduced or abrogated ability to repair DNA DSB by HR relative to normal cells. In another embodiment the subject is heterozygous for a mutation in a gene encoding a component of the HR dependent DNA DSB repair pathway. In one embodiment the cancer is leukemia. In one embodiment the cancer is Acute Myeloid Leukemia (AML). In another embodiment the cancer is Myelodysplastic Syndrome (MDS). In another embodiment the treatment further comprises administration of ionizing radiation or a chemotherapeutic agent.
[0054] The HR dependent DNA DSB repair pathway repairs double-strand breaks (DSBs) in DNA via homologous mechanisms to reform a continuous DNA helix. The components of the HR dependent DNA DSB repair pathway include, but are not limited to, ATM (NM 000051 ), ATR (NM_001 184), CTIP (NM_002894), BLM (NM_000057), RAD51 (NM_002875), RAD51 L1 (NM_002877), RAD51C (NM_002876), RAD51 L3 (NM_002878), DMC1 (NM_007068), XRCC2 (NM_005431 ), XRCC3 (NM_005432), RAD52 (NM_002879), RAD54L (NM 003579), RAD54B (NM_012415), BRCA 1 (NMJ)07295), BRCA2 (NM_000059), RAD50 (NM_005732), MRE1 1A (NM 005590) and NBS 1 (NM 002485). Other proteins involved in the HR dependent DNA DSB repair pathway include regulatory factors such as EMSY. HR components are also described in Wood, et ai, Science, 291 , 1284- 1289 (2001 ), which is hereby incorporated by reference for such disclosure. . . Khanna and S. P. Jackson, Nat. Genet. 27(3): 247-254 (2001 ); and Hughes-Davies, et ai. Cell, 1 15, pp 523-535 are also incorporated herein by reference for such disclosure.
[0055] In some embodiments, a cancer which is deficient in HR dependent DNA DSB repair includes one or more cancer cells which have a reduced or abrogated ability to repair DNA DSBs through that pathway, relative to normal cells, i.e. the activity of the HR dependent DNA DSB repair pathway are reduced or abolished in the one or more cancer cells.
[0056] In certain embodiments, the activity of one or more components of the HR dependent DNA DSB repair pathway is abolished in the one or more cancer cells of an individual having a cancer which is deficient in HR dependent DNA DSB repair. Components of the HR dependent DNA DSB repair pathway include the components listed above.
|0057] In yet another aspect is a method of treating a cancer deficient in DNA mismatch repair (MMR), comprising administering to a subject in need of treatment a therapeutically effective amount of a PARP inhibitor, such as, for example, a compound of Formula (I) or (II). [0058] In one embodiment, a compound of Formula (I) or (II) is used for the treatment of a cancer deficient in MMR, or for the preparation of a medicament for the treatment of a cancer deficient in MMR, which is deficient in HR dependent DNA DSB repair activity.
|0059] The primary function of the DNA mismatch repair (MMR) system is to eliminate single- base mismatches and insertion-deletion loops that may arise during DNA replication. Insertion- deletion loops result from gains or losses of short repeat units within microsatellite sequences, also known as Microsatellite Instability (MSI). At least six different MMR proteins are required. For mismatch recognition, the MSH2 protein forms a heterodimer with either MSH6 or MSH3 depending on the type of lesion to be repaired (MSH6 is required for the correction of single-base mispairs, whereas both MSH3 and MSH6 may contribute to the correction of insertion-deletion loops). A heterodimer of MLH 1 and PMS2 coordinates the interplay between the mismatch recognition complex and other proteins necessary for MMR. These additional proteins may include at least exonuclease 1 (EXO l ), possibly helicase(s), proliferating cell nuclear antigen (PCNA), single- stranded DNA-binding protein (RPA), and DNA polymerases δ and ε. In addition to PMS2, MLH1 may heterodimerize with two additional proteins, MLH3 and PMS l . Recent observations indicate that PMS2 is required for the correction of single-base mismatches, and PMS2 and MLH3 both contribute to the correction of insertion-deletion loops. Additional homologs of the human MMR proteins are known that are required for functions other than MMR. These proteins include MSH4 and MSH5 that are necessary for meiotic (and possibly mitotic) recombination but are not presumed to participate in MMR.
|0060] Germline mutations of human MMR genes cause susceptibility to hereditary nonpolyposis colon cancer (HNPCC), one of the most common cancer syndromes in humans. An excess of colon cancer and a defined spectrum of extracolonic cancers, diagnosed at an early age and transmitted as an autosomal dominant trait, constitute the clinical definition of the syndrome. MSI, the hallmark of HNPCC, occurs in approximately 15% to 25% of sporadic tumors of the colorectum and other organs as well. According to international criteria, a high degree of MSI (MSI-high) is defined as instability at two or more of five loci or £30% to 40% of all microsatellite loci studied, whereas instability at fewer loci is referred to as MSI-low (MSI-low). MSI occurs in a substantial proportion (2% to 50% of tumors) among non-HNPCC cancers (e.g., cancers of the breast, prostate, and lung). On the basis of the proportion of unstable markers, categories MSS, MSI-L, and MSI-H can be distinguished in these cancers in analogy to HNPCC cancers. In one embodiment is a method for treating a cancer deficient in mismatch DNA repair pathway. In another embodiment is a method for treating a cancer demonstrating microsatellite instability due to reduced or impaired DNA repair pathways. In another embodiment is a method for treating a cancer demonstrating genomic instability due to reduced or impaired DNA repair pathways. |0061] The role of nuclear PTEN in the maintenance of chromosomal stability has also been demonstrated in both mouse and human systems (Shen et al, Cell 128: 157- 170 (2007)). First, PTEN interacts with centromeres and maintains their stability. It is believed that PTEN does so through its C2 domain, as mutant PTEN without this C2 domain loses the capability to interact with centromeres. Second, PTEN may be necessary for DNA repair because loss of PTEN results in a high frequency of double-strand breaks. PTEN affects double-strand breaks through regulation of Rad51 , a key component for homologous recombination repair of DNA double-strand breaks. It has also been demonstrated that PTEN physically associates with an integral component of centromeres in the nucleus, disruption of which causes centromere breakage and massive chromosomal aberrations (Shen et al, Cell 128: 157- 170 (2007)).
[0062] A list of human DNA repair genes associated with the HR dependent DNA DSB repair and MMR repair pathways described above, and additional DNA repair pathways, such as Base Excision Repair (BER) and Nucleotide Excision Repair (NER) pathways, is disclosed in, e.g., Wood et al., Science, 291 , 1294 (2001 ), updated in Wood et al., Mutation Res., 577, 275 (2005), each of which is hereby incorporated herein by reference in its entirety. The protein sequences for the protein products of these DNA repair genes will be known by one of skill in the art, and are disclosed at publicly-available databases as, e.g., GenPept, RefSeq, Swiss-Prot, PIR, PRE, and PDB, such as those found at the website for the National Center for Biotechnology Information (NCBI) (see
www.ncbi.nlm.nih.gov).
[0063] In certain embodiments, the patient's malignant cell DNA exhibits Microsatellite Instability (MSI) in a microsatellite, wherein MSI is explained in the Definition section of the instant application. In certain embodiments, the patient's malignant cell DNA exhibits MSI in at least two microsatellites.
[0064] Microsatellites in which MSI is exhibited can include, for example, any microsatellite known to those of skill in the art. Such microsatellites can include, for example, those identified in U.S. Patent No. 6, 150, 100, and U.S. Patent Publication No. 2009/0317815, each of which is incorporated herein by reference in its entirety for all purposes. In certain embodiments, a microsatellite in which MSI is exhibited is NR21, NR22, NR24, Bat25, Bat26, Mono27 and/or a microsatellite as described herein in the DNA repair genes CTiP, Mrel 1, A TM, Rad50, Chkl, BLM, PTEN, A TM and ATR. In certain embodiments, the microsatellite is NR21, NR22, Bat25, Bat26, Mono27, or a microsatellite as described herein in the DNA repair genes CTiP, Mrel I and A TM.
|0065] In certain instances, mutations and polymorphisms associated with cancer are detected at the nucleic acid level by detecting the presence of a variant nucleic acid sequence or at the protein level by detecting the presence of a variant (i.e., a mutant or allelic variant) polypeptide. [0066] m certain embodiments, the methods comprise administering to a subject in need of treatment a therapeutically effective amount of a compound of Formula (I) and Formula (II) in combination with ionizing radiation, one or more chemotherapeutic agents, or a combination thereof.
[0067] Certain embodiments provide a method of potentiation of cytotoxic cancer therapy of MDS or AML in a subject in recognized need of such treatment comprising administering to the subject a therapeutically acceptable amount of a compound of Formula (1) or (II) or a therapeutically acceptable salt thereof.
[0068[ m some embodiments, provided herein is a method for the treatment of MDS or AML, comprising administering to a subject in need of treatment a therapeutically-effective amount of a compound of Formula (I) or (II) in combination with ionizing radiation or one or more
chemotherapeutic agents. In some embodiments, the subject is a patient with MDS or AML. In some embodiments, the compound described herein is administered simultaneously with ionizing radiation or one or more chemotherapeutic agents. In other embodiments, the compound described herein is administered sequentially with ionizing radiation or one or more chemotherapeutic agents.
[0069] In certain embodiments, the one or more chemotherapeutic agents are selected from among alemtuzumab, arsenic trioxide, asparaginase (pegylated or non-), bevacizumab, cetuximab, platinum-based compounds such as cisplatin, cladribine, daunorubicin/doxorubicin/idarubicin, irinotecan, fludarabine, 5-fluorouracil, gemtuzumab, methotrexate, paclitaxel, Taxol®, temozolomide, thioguanine, and classes of drugs including hormones (an antiestrogen, an antiandrogen, or gonadotropin releasing hormone analogues, interferons such as, for example, alpha interferon, nitrogen mustards such as, for example, busulfan, melphalan or mechlorethamine, retinoids such as, for example, tretinoin, topoisomerase inhibitors such as, for example, irinotecan or topotecan, tyrosine kinase inhibitors such as, for example, gefmitinib or imatinib, and agents to treat signs or symptoms induced by such therapy including allopurinol, filgrastim, granisetron/ondansetron/palonosetron, and dronabinol.
[0070] In certain embodiments, the one or more chemotherapeutic agents are selected from: alkylating agents, such as methyl methanesulfonate (MMS), temozolomide and dacarbazine (DTIC); topoisomerase- 1 inhibitors, such as Topotecan, Irinotecan, Rubitecan, Exatecan, Lurtotecan,
Gimetecan, Diflomotecan (homocamptothecins), the 7-substituted non-silatecans, the 7-silyl camptothecins and BNP 1350; non-camptothecin topoisomerase-I inhibitors, such as
indolocarbazoles; and dual topoisomerase-I and II inhibitors, such as the benzophenazines, XR 1 1 76/MLN 576 and benzopyridoindoles.
Methods of Identifying an MDS or AML Patient Likely to Respond to Treatment
[0071 ] In one aspect, provided herein are methods of identifying malignant cells having MSI in an MDS or AML patient which comprise analyzing for MSI in one or more microsatellites of a patient's malignant cell DNA, wherein the presence of MSI in at least one microsatellite of the patient's malignant cell DNA identifies the patient as likely to be sensitive to therapy with a compound of Formula I or II.
(0072] In certain embodiments, the compound of Formula I or II is 5-fluoro-8-(4-fluorophenyl)- 9-( 1 -methyl- 1 H- 1 ,2,4-triazol-5-y l)-8,9-dihydro-2H-pyrido[4,3,2-de]phthalazin-3(7H)-one, or a stereoisomer thereof.
[0073] In certain embodiments, the methods comprise analyzing for MSI in at least one microsatellite of the patient's malignant cell DNA. In certain embodiments, the methods comprise analyzing for MSI in at least two microsatellites of the patient's malignant cell DNA. In certain embodiments, the methods comprise analyzing for MSI in at least three microsatellites of the patient's malignant cell DNA.
[0074] As explained in the Definition section of the instant application, MSI comprises an insertion or deletion of one or more base pairs in a microsatellite of a patient's malignant cell DNA. Accordingly, in certain embodiments, the MSI comprises one or more deleted base pairs; two or more deleted base pairs; or three or more deleted base pairs in the microsatellite(s) of the patient's malignant cell DNA.
|0075] As further explained in the Definition section of the instant application, low-level MSI (MSI-low) is defined as a 1 base pair (bp) deletion or insertion in a microsatellite; mid-level MSI (MSI-mid) is defined as ^ 2 bp deletion or insertion in a microsatellite; high-level MSI (MSI-high) is defined as a ^ 3 bp deletion or insertion in a microsatellite; and MSI stable (MSI-stable) is defined as no deletion or insertion in a microsatellite. Accordingly, in certain embodiments, the MSI may be MSI-low. In other embodiments, the MSI may be MSI-mid. In still further embodiments, the MSI may be MSI-high.
|0076] The methods disclosed herein envision that the presence of low-, mid- or high-level MSI in just one microsatellite of the patient's genomic DNA will identify a patient as likely to respond to therapy with a compound of Formula I or II. However, the presence of MSI (low-, mid- or high-) in additional microsatellites of the patient's DNA may also be helpful is such patient identification. Accordingly, in certain embodiments, the presence of MSI in at least one microsatellite of the patient's malignant cell DNA identifies the patient as likely to respond to therapy with a compound of Formula I or II. In certain embodiments, the presence of MSI in at least two microsatellites of the patient's malignant cell DNA identifies the patient as likely to respond to therapy with a compound of Formula I or II. In certain embodiments, the presence of MSI in at least three microsatellites of the patient's malignant cell DNA identifies the patient as likely to respond to therapy with a compound of Formula 1 or II. [0077] The presence or absence of MSI within rnicrosatellites in malignant cell DNA may be assessed using methods well known of those of skill in the art. For instance, a fluorescent multiplex assay that uses a panel of markers to analyze five nearly monomorphic mononucleotide microsatellite loci (NR21, NR24, Bat25, Bat26 and Mono27) is commercially available from Promega Corp (Madison, WI). See also e.g., Nardon et al., Diagn. Mol. Pathol., 19(4), 236-242 (2010); Bacher et al., Dis. Markers, 20 (4-5), 237-250 (2004); and U.S. Patent Publication No. 2009/0317815.
DEFINITIONS
[0078] Unless defined otherwise, all technical and scientific terms used herein have the standard meaning pertaining to the claimed subject matter belongs. In the event that there are a plurality of definitions for terms herein, those in this section prevail. Where reference is made to a URL or other such identifier or address, it understood that such identifiers can change and particular information on the internet can come and go, but equivalent information can be found by searching the internet. Reference thereto evidences the availability and public dissemination of such information.
[0079] It is to be understood that the foregoing general description and the following detailed description are exemplary and explanatory only and are not restrictive of any subject matter claimed. In this application, the use of the singular includes the plural unless specifically stated otherwise. It must be noted that, as used in the specification and the appended claims, the singular forms "a," "an" and "the" include plural referents unless the context clearly dictates otherwise. In this application, the use of "or" means "and/or" unless stated otherwise. Furthermore, use of the term "including" as well as other forms, such as "include", "includes," and "included," is not limiting.
[0080] Unless otherwise indicated, conventional methods of mass spectroscopy, NMR, HPLC, protein chemistry, biochemistry, recombinant DNA techniques and pharmacology are employed. Unless specific definitions are provided, the standard nomenclature employed in connection with, and the standard laboratory procedures and techniques of, analytical chemistry, synthetic organic chemistry, and medicinal and pharmaceutical chemistry are employed. In certain instances, standard techniques are used for chemical syntheses, chemical analyses, pharmaceutical preparation, formulation, and delivery, and treatment of patients. In certain embodiments, standard techniques are used for recombinant DNA, oligonucleotide synthesis, and tissue culture and transformation (e.g. , electroporation, lipofection). In some embodiments, reactions and purification techniques are performed e.g. , using kits of manufacturer's specifications or as commonly accomplished or as described herein.
[0081] As used throughout this application and the appended claims, the following terms have the following meanings:
[0082] The term "microsatellite" as used herein means repeating sequences of 1 -6 base pairs of DNA, and includes those DNA sequences known in the art as simple sequence repeats (SSR), short tandem repeats (STR), or variable number tandem repeats (VNTR). The repeated sequence may consist, e.g., of a sequence of one, two, three, four, five or six nucleotides (mono-, di-, tri-, tetra-, penta-, and hexa-nucleotide repeats, respectively), and can, for instance, be repeated 3 to 100 times. See, e.g., Eckert et al., Mol Carcinog, 48(4), 379-388 (2009). In certain embodiments, the microsatellite is a mononucleotide repeat sequence, e.g., jj-r- jj- jj (SEQ ID NO. 1 ), referred to as (T)9; TTTTTTTTTTT (SEQ ID NO. 2), referred to as (T)n ; TTTTTTTTTTTTTTT (SEQ ID NO. 3), referred to as (T),5; TTTTTTTTTTTTTTTTTTTTT (SEQ ID NO. 4), referred to as (T)2I ; and AAAAAAAAAAAAAAAAAAAAAAAAAAA (SEQ ID NO. 5), referred to as (A)27. In certain embodiments, the microsatellite is a dinucleotide repeat sequence, e.g., GTGTGTGTGTGT (SEQ ID NO. 6), referred to as (GT)ft. In certain embodiments, the microsatellite is a trinucleotide repeat sequence, e.g., CTGCTGCTGCTG (SEQ ID NO. 7), referred to as (CTG)4. In certain embodiments, the microsatellite is a tetranucleotide repeat sequence, e.g., ACTCACTCACTCACTC (SEQ ID NO. 8), referred to as (ACTC)4. In certain embodiments, the microsatellite is a monomorphic, or nearly monomorphic, mononucleotide repeat (e.g., NR21, NR24, Bat25, Bat 26 and Mono27). For example, a microsatellite may contain one or up to six or seven, or more, nucleotides that interrupt the repeat sequence within the microsatellite.
[0083] Those of skill in the art recognize microsatellites are typically present in human genomic DNA. In certain embodiments, the microsatellite is located within a gene, e.g., a DNA repair gene. In certain embodiments, the microsatellite can, e.g., be located within the following genes: ATM (NM_00005 1), ATR (NM_001 184), CTIP (NM 002894), BLM (NM_000057), RAD51
(NM_002875), RAD51 L 1 (NM 002877), RAD51 C (NM_002876), RAD5 1 L3 (NM_002878), DMC l (NM_007068), XRCC2 (NM 00543 1 ), XRCC3 (NM_005432), RAD52 (NM_002879), RAD54L (NM 003579), RAD54B (NM 012415), BRCA 1 (NM 007295), BRCA2 (NM_000059), RAD50 (NM_005732), MRE 1 1 A (NM_005590) or NBS 1 (NM_002485), among others.
[0084] In certain embodiments, the microsatellite is located within the intron/exon splice junction of a gene. In certain embodiments, the microsatellite is located within the intron of a gene. In certain embodiments, the microsatellite is located within the exon of a gene. In certain embodiments, the microsatellite is located within one of the control elements of a gene, e.g., the promoter of the gene. In certain embodiments, the gene is a DNA repair gene. In certain
embodiments, the gene is other than a DNA repair gene. In certain embodiments, the microsatellite is located outside of a gene.
[0085] The term "Microsatellite Instability" (MSI) as used herein, means an alteration in a microsatellite found in human genomic DNA, e.g., an insertion or deletion of one or more base pairs, as compared to a reference microsatellite. For instance, an MSI may be characterized such that the microsatellite length or repeat sequence is different than that, for instance, which was in the genomic DNA when it was inherited, or, for instance, as compared to the microsatellite length or repeat sequence in the general human population. In certain embodiments, a reference microsatellite is found in the genome of a cell from a human not having MDS or AML, or cells from a population of humans characterized as being cancer free. As used herein, low-level MSI (MSI-low) is defined as 1 base pair (bp) deletion or insertion in a microsatellite; mid-level MSI (MSI-mid) is defined as > 2 bp deletion or insertion in a microsatellite; high-level MSI (MSI-high) is defined as a > 3 bp deletion or insertion in a microsatellite; and MSI stable (MSI-stable) is defined as no deletion or insertion in a microsatellite.
[0086] The term "'alkenyl" as used herein, means a straight, branched chain, or cyclic (in which case, it would also be known as a "cycloalkenyl") hydrocarbon containing from 2- 10 carbons and containing at least one carbon-carbon double bond formed by the removal of two hydrogens. In some embodiments, depending on the structure, an alkenyl group is a monoradical or a diradical (i.e., an alkenylene group). In some embodiments, alkenyl groups are optionally substituted. Illustrative examples of alkenyl include, but are not limited to, ethenyl, 2-propenyl, 2-methyl-2-propenyl, 3-butenyl, 4-pentenyl, 5-hexenyl, 2-heptenyl, 2-methyl- l -heptenyl, and 3-cecenyl.
[0087] The term "alkoxy" as used herein, means an alkyl group, as defined herein, appended to the parent molecular moiety through an oxygen atom. Illustrative examples of alkoxy include, but are not limited to, methoxy, ethoxy, propoxy, 2-propoxy, butoxy, tert-butoxy, pentyloxy, and hexyloxy.
|0088] The term "alkyl" as used herein, means a straight, branched chain, or cyclic (in this case, it would also be known as "cycloalkyl") hydrocarbon containing from 1 - 10 carbon atoms. Illustrative examples of alkyl include, but are not limited to, methyl, ethyl, n-propyl, iso-propyl, n-butyl, sec- butyl, tert-butyl, n-pentyl, isopentyl, neopentyl, n-hexyl, 3-methylhexyl, 2,2-dimethylpentyl, 2,3-dimethylhexyl, n-heptyl, n-octyl, n-nonyl, and n-decyl.
[0089] The term "Ci-C6-alkyl" as used herein, means a straight, branched chain, or cyclic (in this case, it would also be known as "cycloalkyl") hydrocarbon containing from 1-6 carbon atoms.
Representative examples of alkyl include, but are not limited to, methyl, ethyl, n-propyl, iso-propyl, cyclopyl, n-butyl, sec-butyl, tert-butyl, cyclobutyl, n-pentyl, isopentyl, neopentyl, cyclopentyl, and n-hexyl.
[0090] The term "cycloalkyl" as used herein, means a monocyclic or polycyclic radical that contains only carbon and hydrogen, and includes those that are saturated, partially unsaturated, or fully unsaturated. Cycloalkyl groups include groups having from 3 to 10 ring atoms. Representative xamples of cyclic include but are not limited to, the following moieties:
Figure imgf000028_0001
Figure imgf000029_0001
. In some embodiments, depending on the structure, a cycloalkyl group is a monoradical or a diradical (e.g. , a cycloalkylene group).
[0091] The term "cycloalkyl groups" as used herein refers to groups which are optionally substituted with 1 , 2, 3, or 4 substituents selected from alkenyl, alkoxy, alkoxyalkyl, alkoxycarbonyl, alkyl, alkylcarbonyl, alkylcarbonyioxy, alkyithio, alkyithioalkyl, alkynyl, carboxy, cyano, formyl, haloalkoxy, haloalkyl, halogen, hydroxyl, hydroxyalkylene, mercapto, oxo, -NRARA, and
(NRARB)carbonyl.
[0092] The term "cycloalkylalkyl" as used herein, means a cycloalkyl group, as defined herein, appended to the parent molecular moiety through an alkyl group, as defined herein. Representative examples of cycloalkylalkyl include, but are not limited to, cyclopropylmethyl, 2-cyclobutylethyl, cyclopentylmethyl, cyclohexylmethyl, and 4-cycloheptylbutyl.
[0093] The term "carbocyclic" as used herein, refers to a compound which contains one or more covalently closed ring structures, and that the atoms forming the backbone of the ring are all carbon atoms
[0094] The term "carbocycle" as used herein, refers to a ring, wherein each of the atoms forming the ring is a carbon atom. Carbocylic rings include those formed by three, four, five, six, seven, eight, nine, or more than nine carbon atoms. Carbocycles are optionally substituted.
[0095] The term "alkoxyalkyl" as used herein, means at least one alkoxy group, as defined herein, appended to the parent molecular moiety through an alkyl group, as defined herein. Illustrative examples of alkoxyalkyl include, but are not limited to, 2-methoxyethyl, 2-ethoxyethyl, tert- butoxyethyl and methoxymethyl.
[0096] The term "alkoxycarbonyl" as used herein, means an alkoxy group, as defined herein, appended to the parent molecular moiety through a carbonyl group, as defined herein. Illustrative examples of alkoxycarbonyl include, but are not limited to, methoxycarbonyl, ethoxycarbonyl, and tert-butoxy carbonyl.
[0097] The term "alkoxycarbonylalkyl" as used herein, means an alkoxycarbonyl group, as defined herein, appended to the parent molecular moiety through an alkyl group, as defined herein. [0098] The term "alkylearbonyl" as used herein, means an alkyl group, as defined herein, appended to the parent molecular moiety through a carbonyl group, as defined herein. Illustrative examples of alkylearbonyl include, but are not limited to, acetyl, 1 -oxopropyl, 2,2-dimethyl- l - oxopropyl, 1 -oxoburyl, and 1 -oxopentyl.
[0099] The term "alkylcarbonyloxy" as used herein, means an alkylearbonyl group, as defined herein, appended to the parent molecular moiety through an oxygen atom. Illustrative examples of alkylcarbonyloxy include, but are not limited to, acetyloxy, ethylcarbonyloxy, and tert- buty lcarbony loxy .
|00100] The term "alkylthio" or "thioalkoxy" as used herein, means an alkyl group, as defined herein, appended to the parent molecular moiety through a sulfur atom. Illustrative examples of alkylthio include, but are not limited to, methylthio, ethylthio, butylthio, tert-butylthio, and hexylthio.
[00101 ] The term "alkylthioalkyl" as used herein, means an alkylthio group, as defined herein, appended to the parent molecular moiety through an alkyl group, as defined herein. Illustrative examples of alkylthioalkyl include, but are not limited to, methylthiomethyl, 2-(ethylthio)ethyl, butylthiomethyl, and hexylthioethyl.
|00102] The term "alkynyl" as used herein, means a straight, branched chain hydrocarbon containing from 2- 10 carbons and containing at least one carbon-carbon triple bond. In some embodiments, alkynyl groups are optionally substituted. Illustrative examples of alkynyl include, but are not limited to, acetylenyl, 1 -propynyl, 2-propynyl, 3-butynyl, 2-pentynyl, and 1 -butynyl.
[00103] The term "aromatic" as used herein, refers to a planar ring having a delocalized π-electron system containing 4n+2 π electrons, where n is an integer. In some embodiments, aromatic rings are formed by five, six, seven, eight, nine, or more than nine atoms. In other embodiments, aromatics are optionally substituted. The term includes monocyclic or fused-ring polycyclic (i.e., rings which share adjacent pairs of carbon atoms) groups.
(00104] The term "aryl" as used herein, refers to an aromatic ring wherein each of the atoms forming the ring is a carbon atom. In some embodiments, aryl rings are formed by five, six, seven, eight, nine, or more than nine carbon atoms. Examples of aryl groups include, but are not limited to phenyl, naphthalenyl, phenanthrenyl, anthracenyl, fluorenyl, and indenyl.
|00105] In some embodiments, the term "aryl" as used herein means an aryl group that is optionally substituted with one, two, three, four or five substituents independently selected from the group consisting of alkenyl, a!koxy, alkoxyalkyl, aikoxycarbonyl, alkyl, alkylearbonyl,
alkylcarbonyloxy, alkylthio, alkylthioalkyl, alkynyl, carbonyl, cyano, formyl, haloalkoxy, haloalkyl, halogen, hydroxyl, hydroxyalkylene, mercapto, nitro, -NRARA, and (NRARB)carbonyl. [00106] The term "arylalkyl" as used herein, means an aryl group, as defined herein, appended to the parent molecular moiety through an alkyl group, as defined herein. Illustrative examples of arylalkyl include, but are not limited to benzyl, 2-phenylethyl, -phenylpropyl, l -methyl-3- pheny [propyl, and 2-naphth-2-ylethyI.
(00107] The term "carbonyl" as used herein, means a -C(O)- group.
[00108] The term "carboxy" as used herein, means a -COOH group.
[00109] The term "cyano" as used herein, means a -CN group.
[00110] The term "formyl" as used herein, means a -C(0)H group.
[001111 The term "halo" or "halogen" as used herein, means a -CI, -Br, -I or -F.
[00112] The term "mercapto" as used herein, means a -SH group.
100113] The term "nitro" as used herein, means a -N02 group.
[00114] The term "hydroxy" as used herein, means a -OH group.
100115] The term "oxo" as used herein, means a =0 group.
100116] The term "bond" or "single bond" as used herein, refers to a chemical bond between two atoms, or two moieties when the atoms joined by the bond are considered to be part of larger substructure.
[00117] The terms "haloalkyl," "haloalkenyl," "haloalkynyl" and "haloalkoxy" as used herein, include alkyl, alkenyl, alkynyl and alkoxy structures in which at least one hydrogen is replaced with a halogen atom. In certain embodiments in which two or more hydrogen atoms are replaced with halogen atoms, the halogen atoms are all the same as one another, in other embodiments in which two or more hydrogen atoms are replaced with halogen atoms, the halogen atoms are not all the same as one another. The terms "fluoroalkyl" and "fluoroalkoxy" include haloalkyl and haloalkoxy groups, respectively, in which the halo is fluorine. In certain embodiments, haloalkyls are optionally substituted.
]001 18] The term "ester" refers to a chemical moiety with formula -COOR, where R is selected from among alkyl, cycloalkyl, aryl, heteroaryl (bonded through a ring carbon) and heterocycloalkyl (bonded through a ring carbon), in some embodiments, any hydroxy, or carboxyl side chain on the compounds described herein is esterified.
[001 19] The terms "heteroalkyl" "heteroalkenyl" and "heteroalkynyl" as used herein, include optionally substituted alkyl, alkenyl and alkynyl radicals in which one or more skeletal chain atoms are selected from an atom other than carbon, e.g., oxygen, nitrogen, sulfur, silicon, phosphorus or combinations thereof. 100120] The term "heteroatom" as used herein refers to an atom other than carbon or hydrogen. Heteroatoms are typically independently selected from among oxygen, sulfur, nitrogen, silicon and phosphorus, but are not limited to these atoms. In embodiments in which two or more heteroatoms are present, the two or more heteroatoms are all the same as one another, or some or all of the two or more heteroatoms are each different from the others.
[00121] The term "ring" as used herein, refers to any covalently closed structure. Rings include, for example, carbocycles (e.g. , aryls and cycloalkyls), heterocycles (e.g. , heteroaryls and
heterocycloalkyls), aromatics (e.g. aryls and heteroaryls), and non-aromatics (e.g. , cycloalkyls and heterocycloalkyls). In some embodiments, rings are optionally substituted. In some embodiments, rings form part of a ring system.
[00122] As used herein, the term "ring system" refers to two or more rings, wherein two or more of the rings are fused. The term "fused" refers to structures in which two or more rings share one or more bonds.
[00123] The terms "heteroaryl" or, alternatively, "heteroaromatic" refers to an aryl group that includes one or more ring heteroatoms selected from nitrogen, oxygen and sulfur. An iV-containing "heteroaromatic" or "heteroaryl" moiety refers to an aromatic group in which at least one of the skeletal atoms of the ring is a nitrogen atom. In some embodiments, the polycyclic heteroaryl group is fused or non-fused. Illustrative of heteroaryl groups include, but are not limited to, the following moieties:
Figure imgf000032_0001
. In some embodiments, depending on the structure, a heteroaryl group is a monoradical or a diradical (i.e., a heteroaryiene group).
100124] The term "heteroaryl" means heteroaryl groups that are substituted with 0, 1 , 2, 3, or 4 substituents independently selected from alkenyl, alkoxy, alkoxyalkyl, alkoxycarbonyl, alkyl, alkylcarbonyl, alkylcarbonyloxy, alkylthio, alkylthioalkyl, alynyl, carboxy, cyano, formyl, haloalkoxy, haloalkyl, halogen, hydroxyl, hydroxyalkylene, mercapto, nitro, -NRARB,
and -(NRARB)carbonyl.
[00125] The term "heteroarylalkyl" as used herein, means a heteroaryl, as defined herein, appended to the parent molecular moiety through an alkyl group, as defined herein. Illustrative examples of heteroarylalkyl include, but are not limited to, pyridinylmethyl.
[00126] The term "heterocycloalkyl" or "non-aromatic heterocycle" as used herein, refers to a non-aromatic ring wherein one or more atoms forming the ring is a heteroatom. A "heterocycloalkyl" or "non-aromatic heterocycle" group refers to a cycloalkyl group that includes at least one heteroatom selected from nitrogen, oxygen and sulfur. In some embodiments, the radicals are fused with an aryl or heteroaryl. In some embodiments, heterocycloalkyl rings are formed by three, four, five, six, seven, eight, nine, or more than nine atoms. In some embodiments, heterocycloalkyl rings are optionally substituted. In certain embodiments, heterocycloalkyls contain one or more carbonyl or thiocarbonyl groups such as, for example, oxo- and thio-containing groups. Examples of heterocycloalkyls include, but are not limited to, lactams, lactones, cyclic imides, cyclic thioimides, cyclic carbamates, tetrahydrothiopyran, 4H-pyran, tetrahydropyran, piperidine, 1 ,3-dioxin, 1 ,3-dioxane, 1 ,4-dioxin, 1 ,4- dioxane, piperazine, 1 ,3-oxathiane, 1 ,4-oxathiin, 1 ,4-oxathiane, tetrahydro- 1 ,4-thiazine, 2H- 1 ,2- oxazine, maleimide, succinimide, barbituric acid, thiobarbituric acid, dioxopiperazine, hydantoin, dihydrouracil, morpholine, trioxane, hexahydro- l ,3,5-triazine, tetrahydrothiophene, tetrahydrofuran, pyrroline, pyrrolidine, pyrrolidone, pyrrolidione, pyrazoline, pyrazolidine, imidazoline, imidazolidine, 1 ,3-dioxole, 1 ,3-dioxolane, 1 ,3-dithiole, 1 ,3-dithiolane, isoxazoline, isoxazolidine, oxazoline, oxazolidine, oxazolidinone, thiazoline, thiazolidine, and 1 ,3-oxathiolane. Illustrative examples of limited to
Figure imgf000033_0001
Figure imgf000034_0001
. The term heterocycloalkyl also includes all ring forms of the carbohydrates, including but not limited to the monosaccharides, the disaccharides and the oligosaccharides.
[00127] The term "heterocycle" refers to heteroaryl and heterocycloalkyl used herein, refers to groups containing one to four heteroatoms each selected from O, S and N, wherein each heterocycle group has from 4 to 10 atoms in its ring system, and with the proviso that the ring of said group does not contain two adjacent O or S atoms. Herein, whenever the number of carbon atoms in a heterocycle is indicated (e.g. , Ci -C„ heterocycle), at least one other atom (the heteroatom) must be present in the ring. Designations such as "C ,-C<, heterocycle" refer only to the number of carbon atoms in the ring and do not refer to the total number of atoms in the ring. In some embodiments, it is understood that the heterocycle ring has additional heteroatoms in the ring. Designations such as "4-6 membered heterocycle" refer to the total number of atoms that are contained in the ring (i.e., a four, five, or si . membered ring, in which at least one atom is a carbon atom, at least one atom is a heteroatom and the remaining two to four atoms are either carbon atoms or heteroatoms). In some embodiments, in heterocycles that have two or more heteroatoms, those two or more heteroatoms are the same or different from one another. In some embodiments, heterocycles are optionally substituted. In some embodiments, binding to a heterocycle is at a heteroatom or via a carbon atom. Heterocycloalkyl groups include groups having only 4 atoms in their ring system, but heteroaryl groups must have at least 5 atoms in their ring system. The heterocycle groups include benzo-fused ring systems. An example of a 4-membered heterocycle group is azetidinyl (derived from azetidine). An example of a 5-membered heterocycle group is thiazolyl. An example of a 6-membered heterocycle group is pyridyl, and an example of a 10-membered heterocycle group is quinolinyl. Examples of
heterocycloalkyl groups are pyrrolidinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, tetrahydropyranyl, dihydropyranyl, tetrahydrothiopyranyl, piperidino, morpholino, thiomorpholino, thioxanyl, piperazinyl, azetidinyl, oxetanyl, thietanyl, homopiperidinyl, oxepanyl, thiepanyl, oxazepinyl, diazepinyl, thiazepinyl, 1 ,2,3,6-tetrahydropyridinyl, 2-pyrrolinyl, 3-pyrrolinyl, indolinyl, 2H-pyranyl, 4H-pyranyl, dioxanyl, 1 ,3-dioxolanyl, pyrazolinyl, dithianyl, dithiolanyl, dihydropyranyl, dihydrothienyl, dihydrofuranyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, 3- azabicyclo[3.1.0]hexanyl, 3-azabicyclo[4. 1.OJheptanyl, 3H-indolyl and quinolizinyl. Examples of heteroaryl groups are pyridinyl, imidazolyl, pyrimidinyl, pyrazolyl, triazolyl, pyrazinyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, oxazolyl, isothiazolyl, pyrrolyl, quinolinyl, isoquinolinyl, indolyl, benzimidazolyl, benzofuranyl, cinnolinyl, indazolyl, indolizinyl, phthalazinyl, pyridazinyl, triazinyl, isoindolyl, pteridinyl, purinyl, oxadiazolyl, thiadiazolyl, furazanyl, benzofurazanyl, benzothiophenyl, benzothiazolyl, benzoxazolyl, quinazolinyl, quinoxalinyl, naphthyridinyl, and furopyridinyl. In some embodiments, the foregoing groups, as derived from the groups listed above, are C-attached or N- attached where such is possible. For instance, in some embodiments, a group derived from pyrrole is pyrrol- 1 -yl (^-attached) or pyrrol-3-yl (C-attached). Further, in some embodiments, a group derived from imidazole is imidazol- l -yl or imidazol-3-yl (both iV-attached) or imidazoI-2-yl, imidazol-4-yl or imidazol-5-yl (all C-attached). The heterocycle groups include benzo-fused ring systems and ring systems substituted with one or two oxo (=0) moieties such as pyrrolidin-2-one. In some embodiments, depending on the structure, a heterocycle group is a inonoradical or a diradical (i.e., a heterocyclene group).
[00128] The heterocycles described herein are substituted with 0, 1 , 2, 3, or 4 substituents independently selected from alkenyl, alkoxy, alkoxyalkyl, alkoxycarbonyl, alkyl, aikylcarbonyl, alkylcarbonyloxy, alkylthio, alkylthioalkyl, alynyl, carboxy, cyano, formyl, haloalkoxy, haloalkyl, halogen, hydroxyl, hydroxyalkylene, mercapto, nitro, -NRARB, and -(NRARB)carbonyl.
[00129] The term "heterocycloalkoxy" refers to a heterocycloalkyl group, as defined herein, appended to the parent molecular moiety through an alkoxy group.
[00130] The term "heterocycloalkylthio" refers to a heterocycloalkyl group, as defined herein, appended to the parent molecular moiety through an alkylthio group.
[00131 ] The term "heterocyclooxy" refers to a heterocycloalkyl group, as defined herein, appended to the parent molecular moiety through an oxygen atom.
|00132] The term "heterocyclothio" refers to a heterocycloalkyl group, as defined herein, appended to the parent molecular moiety through a sulfur atom.
[00133] The term "heteroarylalkoxy" refers to a heteroaryl group, as defined herein, appended to the parent molecular moiety through an alkoxy group.
[00134] The term "heteroarylalkylthio" refers to a heteroaryl group, as defined herein, appended to the parent molecular moiety through an alkylthio group.
[00135] The term "heteroaryloxy" refers to a heteroaryl group, as defined herein, appended to the parent molecular moiety through an oxygen atom.
(00136] The term "heteroarylthio" refers to a heteroaryl group, as defined herein, appended to the parent molecular moiety through a sulfur atom.
[00137] In some embodiments, the term "membered ring" embraces any cyclic structure. The term "membered" is meant to denote the number of skeletal atoms that constitute the ring. Thus, for example, cyclohexyl, pyridine, pyran and thiopyran are 6-membered rings and cyclopcntyl, pyrrole, furan, and thiophene are 5-membered rings. [00138] The term "non-aromatic 5, 6, 7, 8, 9, 10, 1 1 or 12- bicyclic heterocycle" as used herein, means a heterocycloalkyl, as defined herein, consisting of two carbocyclic rings, fused together at the same carbon atom (forming a spiro structure) or different carbon atoms (in which two rings share one or more bonds), having 5 to 12 atoms in its overall ring system, wherein one or more atoms forming the ring is a heteroatom. Illustrative examples of non-aromatic 5, 6, 7, 8, 9, 10, 1 1 , or 12- bicyclic heterocycle ring include, but are not limited to, 2-azabicyclo[2.2. l ]heptanyl,
7-azabicyclo[2.2.1 ]heptanyl, 2-azabicyclo[3.2.0]heptanyl, 3-azabicyclo[3.2.0]heptanyl,
4-azaspiro[2.4]heptanyl, 5-azaspiro[2.4]heptanyl, 2-oxa-5- azabicyclo[2.2.1 ]heptanyl,
4-azaspiro[2.5]octanyl, 5-azaspiro[2.5]octanyl, 5-azaspiro[3.4]octanyl, 6-azaspiro[3.4]octanyl,
4- oxa-7-azaspiro[2.5]octanyl, 2-azabicyclo[2.2.2]octanyl, 1 ,3-diazabicyclo[2.2.2]octanyl,
5- azaspiro[3.5]nonanyl, 6-azaspiro[3.5]nonanyl, 5-oxo-8-azaspiro[3.5]nonanyl,
octahydrocyclopenta[c]pyrrolyl, octahydro- lH-quinolizinyl, 2,3,4,6,7,9a-hexahydro- lH-quinolizinyl, decahydropyrido[l ,2-a]azepinyl, decahydro- lH-pyrido[ l ,2-a]azocinyl, l-azabicyclo[2.2.1 ]heptanyl,
1 - azabicyclo[3.3.1 ]nonanyl, quinuclidinyl, and l -azabicyclo[4.4.0]decanyl.
|00139] The term "hydroxyalkylene" as used herein, means at least one hydroxy 1 group, as defined herein, is appended to the parent molecular moiety through an alkylene group, as defined herein. Illustrative examples of hydroxyalkylene include, but not limited to hydroxymethylene,
2- hydroxy-ethylene, 3-hydroxypropylene and 4-hydroxyheptylene.
[00140] As used herein, the term "moiety" refers to a specific segment or functional group of a molecule. Chemical moieties are often recognized chemical entities embedded in or appended to a molecule.
100141 ] As used herein, the term "sulfonyl" refers to a -S(=0)2-R, where R is selected from the group consisting of alkyl, cycloalkyl, aryl, heteroaryl (bonded through a ring carbon) and heterocycloalkyl (bonded through a ring carbon).
[00142] As used herein, the term "acetyl"' refers to a group of formula -C(=0)CH3.
100143] As used herein, the term "isocyanato" refers to a group of formula -NCO.
[00144] As used herein, the term "thiocyanato" refers to a group of formula -CNS.
[00145] As used herein, the term "isothiocyanato" refers to a group of formula -NCS.
[00146] As used herein, the substituent "R" appearing by itself and without a number designation refers to a substituent selected from among from alkyl, cycloalkyl, aryl, heteroaryl (bonded through a ring carbon) and non-aromatic heterocycle (bonded through a ring carbon).
[00147] The term "substituted" means that the referenced group is optionally substituted
(substituted or unsubstituted) with one or more additional group(s) individually and independently selected from alkyl, cycloalkyl, aryl, heteroaryl, heteroalicyclic, hydroxy, alkoxy, aryloxy, mercapto, alkylthio, arylthio, alkylsulfoxide, arylsulfoxide, alkylsulfone, arylsulfone, cyano, halo, carbonyl, thiocarbonyl, isocyanato, thiocyanato, isothiocyanato, nitro, perhaloalkyl, perfluoroalkyl, silyl, and amino, including mono- and di-substituted amino groups, and the protected derivatives thereof. By way of example an optional substituents is LSRS, wherein each Ls is independently selected from a bond, -0-, -C(=0)-, -S-, -S(=0)-, -S(=0)2-, -NH-, -NHC(O)-, -C(0)NH-, S(=0)2NH-, -NHS(=0)2, - OC(0)NH-, -NHC(0)0-, -(substituted or unsubstituted d-C6 alkyl), or -(substituted or unsubstituted C2-C6 alkenyl); and each Rs is independently selected from H, (substituted or unsubstituted lower alkyl), (substituted or unsubstituted lower cycloalkyl), heteroaryl, or heteroalkyl.
[00148] The term "protecting group" refers to a removable group which modifies the reactivity of a functional group, for example, a hydroxyl, ketone or amine, against undesirable reaction during synthetic procedures and to be later removed. Examples of hydroxy-protecting groups include, but not limited to, methylthiomethyl, tert-dimethylsilyl, tert-butyldiphenylsilyl, ethers such as
methoxymethyl, and esters including acetyl, benzoyl, and the like. Examples of ketone protecting groups include, but not limited to, ketals, oximes, O-substituted oximes for example O-benzyl oxime, O-phenylthiornethyl oxime, 1 -isopropoxycyclohexyl oxime, and the like. Examples of amine protecting groups include, but are not limited to, tert-butoxycarbonyl (Boc) and carbobenzyloxy (Cbz).
[00149] The term "optionally substituted" as defined herein, means the referenced group is substituted with zero, one or more substituents as defined herein.
[00150] In some embodiments, compounds of the described herein exist as stereoisomers, wherein asymmetric or chiral centers are present. Stereoisomers are designated (R) or (S) depending on the configuration of substituents around the chiral carbon atom. The term (R) and (S) used herein are configurations as defined in IUPAC 1974 Recommendations for Section E, Fundamental
Stereochemistry, Pure Appl. Chem., ( 1976), 45: 13-30, hereby incorporated by reference. The embodiments described herein specifically includes the various stereoisomers and mixtures thereof. Stereoisomers include enantiomers, diastereomers, and mixtures of enantiomers or diastereomers. In some embodiments, individual stereoisomers of compounds are prepared synthetically from commercially available starting materials which contain asymmetric or chiral centers or by preparation of racemic mixtures followed by resolution. These methods of resolution are exemplified by ( 1 ) attachment of a mixture of enantiomers to a chiral axillary, separation of the resulting mixture of diastereomers by recrystallization or chromatography and liberation of the optically pure product from the auxiliary or (2) direct separation of the mixture of optical enantiomers on chiral
chromatographic column.
[00151 ] The methods and formulations described herein include the use of iV-oxides, crystalline forms (also known as polymorphs), or pharmaceutically acceptable salts of compounds described herein, as well as active metabolites of these compounds having the same type of activity. In some situations, compounds exist as tautomers. All tautomers are included within the scope of the compounds presented herein. In some embodiments, the compounds described herein exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like. The solvated forms of the compounds presented herein are also considered to be disclosed herein.
[00152] Throughout the specification, groups and substituents thereof are chosen, in certain embodiments, to provide stable moieties and compounds.
Preparation of Compounds Described Herein
100153] In certain embodiments, the compounds described herein are synthesized using any synthetic techniques including standard synthetic techniques and the synthetic processes described herein. In specific embodiments, the following synthetic processes are utilized.
Synthetic Schemes
[00154] In certain embodiments, compounds of Formula (I), composing of la to If, are prepared in various ways, as outlined in Synthetic Schemes 1 and 2. In each scheme, the variables (e.g., R] , R2. R3, R4, R5, Y, and Z) correspond to the same definitions as those recited above while R is alkyl and Y' is the same or different group defined by Y and Z' is the same or different group defined by Z. In some embodiments, compounds are synthesized using methodologies analogous to those described below by the use of appropriate alternative starting materials.
[00155] In certain embodiments, compounds of Formula (la, and lb) wherein Y is identical to Z are synthesized according to Synthetic Scheme 1. Thus, the preparation of the intermediate 3 wherein R5 is hydrogen is achieved by condensation of 4-aminoisobenzofuran- l(3H)-one 1 with an aldehyde 2 in the presence of a base preferably alkaline alkoxides in appropriate solvents such as ethyl acetate or ethyl propionate at either ambient or elevated temperature. Compounds of Formula la wherein R5 is hydrogen is prepared by treating the intermediate 3 with hydrazine hydrate at ambient or elevated temperature. Compounds of Formula la wherein R5 is alkyl or substituted alkyl is prepared from compound of Formula la wherein R5 is hydrogen by reductive amination reaction with R7-CHO wherein R7 is alkyl, substituted alkyl. In some embodiments, the preparation of the compounds in Formula lb is accomplished by further modification of la. Through appropriate functional group transformations on the moiety of Y and Z, one affords the compounds of Formula lb with different entities of Y' and Z' at 2- or 3-positions. Synthetic Scheme 1
Figure imgf000039_0001
[00156] In certain embodiments, compounds of Formula ( Ic, and Id) are synthesized according to Synthetic Scheme 2. For example, the intermediate 5 is prepared by condensation of the reagent 1 with an aldehyde 4 in the presence of water absorbent such sodium sulfate or magnesium sulfate at elevated temperature. A subsequent condensation reaction of this intermediate with another aldehyde in the presence of a base preferably alkaline alkoxides in appropriate solvents such as ethyl acetate or ethyl propionate at either ambient or elevated temperature gives the intermediate 6 wherein R5 is hydrogen. Compounds of Formula Ic wherein R5 is hydrogen is prepared by treating the intermediate 6 with hydrazine hydrate at ambient or elevated temperature. Compounds of Formula Ic wherein R5 is alkyl, substituted alkyl are prepared from compounds of Formula Ic wherein R5 is hydrogen by reductive amination reaction with R7-CHO wherein R7 is alkyl, or substituted alkyl. In some embodiments, the preparation of compounds of Formula Id are accomplished by further modification of Ic. Through appropriate functional group transformations on the moiety of Y and Z. one could afford the compounds of Formula Ic with different entities of Y ' and Z' at 2- or 3-positions.
Synthetic Scheme 2
Figure imgf000039_0002
[00157] Examples 2 - 159 of International Patent Publication No. WO 2010/017055 A2, which is incorporated by reference in its entirety in the instant application, disclose exemplary syntheses of compounds of Formula (1).
Certain Pharmaceutical Terminology
|00158] The term "acceptable" w ith respect to a formulation, composition or ingredient, as used herein, means having no persistent detrimental effect on the general health of the subject being treated.
[0015 J As used herein, the term "selective binding compound" refers to a compound that selectively binds to any portion of one or more target proteins. [00160] As used herein, the term "selectively binds" refers to the ability of a selective binding compound to bind to a target protein, such as, for example, PARP, with greater affinity than it binds to a non-target protein. In certain embodiments, specific binding refers to binding to a target with an affinity that is at least about 10, about 50, about 100, about 250, about 500, about 1000 or more times greater than the affinity for a non-target.
(00161] As used herein, the term "target protein" refers to a molecule or a portion of a protein capable of being bound by a selective binding compound. In certain embodiments, a target protein is the enzyme poly(ADP-ribose)polymerase (PARP).
[00162] As used herein, the terms "treating" or "treatment" encompass either or both responsive and prophylaxis measures, e.g. , designed to inhibit, slow or delay the onset of a symptom of a disease or disorder, achieve a full or partial reduction of a symptom or disease state, and/or to alleviate, ameliorate, lessen, or cure a disease or disorder and/or its symptoms.
[00163] As used herein, amelioration of the symptoms of a particular disorder by administration of a particular compound or pharmaceutical composition refers to any lessening of severity, delay in onset, slowing of progression, or shortening of duration, whether permanent or temporary, lasting or transient that can be attributed to or associated with administration of the compound or composition.
[00164] As used herein, the term "modulator" refers to a compound that alters an activity of a molecule. For example, a modulator includes a compound that causes an increase or a decrease in the magnitude of a certain activ ity of a molecule compared to the magnitude of the activity in the absence of the modulator. In certain embodiments, a modulator is an inhibitor, which decreases the magnitude of one or more activities of a molecule. In certain embodiments, an inhibitor completely prevents one or more activities of a molecule. In certain embodiments, a modulator is an activator, which increases the magnitude of at least one activity of a molecule. In certain embodiments the presence of a modulator results in an activity that does not occur in the absence of the modulator.
[00165] As used herein, the term "selective modulator" refers to a compound that selectively modulates a target activity.
100166] As used herein, the term "PARP" refers to the family of the enzyme poly(ADP- ribose)polymerase which includes approximately 18 proteins, particularly poly(ADP- ribose)polymerase- I (PARP- 1 ) and poly(ADP-ribose)polymerase-2 (PARP-2).
[00167] As used herein, the term "selective PARP modulator" refers to a compound that selectively modulates at least one activity associated with the enzyme poly(ADP-ribose)polymerase (PARP). In various embodiments, the selective modulator selectively modulates the activity of PARP - 1 , PARP-2, both PARP- 1 and PARP-2 or several members of the family of the enzyme poly(ADP- ribose)polymerase (PARP). 100168] As used herein, the term "method of inhibiting PARP" refers to a method of inhibiting the activity of either one or more of the family of enzyme poly(ADP-ribose)polymerase (PARP). As used herein, the term "inhibition of PARP" refers to inhibition of the activity of either one or more of the family of enzyme poly(ADP-ribose)polymerase (PARP).
|00169] As used herein, the term "modulating the activity of the enzyme poly(ADP- ribose)polymerase" refers to a modulating the activity of either one or more of the family of enzyme poly(ADP-ribose)polymerase (PARP).
(00170] As used herein, the term "selectively modulates" refers to the ability of a selective modulator to modulate a target activity to a greater extent than it modulates a non-target activity. In certain embodiments the target activity is selectively modulated by, for example about 2 fold up to more that about 500 fold, in some embodiments, about 2, 5, 10, 50, 100, 150, 200, 250, 300, 350, 400, 450 or more than 500 fold.
|00171] The term "cancer", as used herein refers to an abnormal growth of cells which tend to proliferate in an uncontrolled way and, in some cases, to metastasize (spread). The types of cancer include, but are not limited to, solid tumors (such as those of the bladder, bowel, brain, breast, endometrium, heart, kidney, lung, lymphatic tissue (lymphoma), ovary, pancreas or other endocrine organ (thyroid), prostate, skin (melanoma) or hematological tumors (such as the leukemias MDS and AML).
[00172] The term "carrier," as used herein, refers to relatively nontoxic chemical compounds or agents that facilitate the incorporation of a compound into cells or tissues.
[00173] The terms "co-administration" or the like, as used herein, are meant to encompass administration of the selected therapeutic agents to a single patient, and are intended to include treatment regimens in which the agents are administered by the same or different route of administration or at the same or different time.
[00174] The term "diluent" refers to chemical compounds that are used to dilute the compound of interest prior to delivery. Diluents include chemicals used to stabilize compounds because they provide a more stable environment. Salts dissolved in buffered solutions (which also can provide pH control or maintenance) are utilized as diluents in certain embodiments, including, but not limited to a phosphate buffered saline solution.
[00175] The terms "effective amount" or "therapeutically effective amount," as used herein, refer to a sufficient amount of an agent or a compound being administered which will relieve to some extent one or more of the symptoms of the disease or condition being treated. The result includes reduction and/or alleviation of the signs, symptoms, or causes of a disease, or any other desired alteration of a biological system. For example, an "effective amount" for therapeutic uses is the amount of the composition comprising a compound as disclosed herein required to provide a clinically significant decrease in disease symptoms. An appropriate "effective" amount in any individual case is determined using any suitable technique, such as a dose escalation study.
[00176] The terms "enhance" or "enhancing," as used herein, means to increase or prolong either in potency or duration a desired effect. Thus, in regard to enhancing the effect of therapeutic agents, the term "enhancing" refers to the ability to increase or prolong, either in potency or duration, the effect of other therapeutic agents on a system. An "enhancing-effective amount," as used herein, refers to an amount adequate to enhance the effect of another therapeutic agent in a desired system.
[00177] The term "enzymatically cleavable linker," as used herein refers to unstable or degradable linkages which are degraded by one or more enzymes.
[00178] The term "PARP-mediated", as used herein, refers to conditions or disorders that are ameliorated by the one or more of the family of enzyme poly(ADP-ribose)polymerase (PARP).
[00179] A "metabolite" of a compound disclosed herein is a derivative of that compound that is formed when the compound is metabolized. The term "active metabolite" refers to a biologically active derivative of a compound that is formed when the compound is metabolized. The term
"metabolized," as used herein, refers to the sum of the processes (including, but not limited to, hydrolysis reactions and reactions catalyzed by enzymes) by which a particular substance is changed by an organism. Thus, in certain instances, enzymes produce specific structural alterations to a compound. In some embodiments, metabolites of the compounds disclosed herein are identified either by administration of compounds to a host and analysis of tissue samples from the host, or by incubation of compounds with hepatic cells in vitro and analysis of the resulting compounds.
[00180] The term "modulate," as used herein, means to interact with a target either directly or indirectly so as to alter the activity of the target, including, by way of example only, to enhance the activity of the target, to inliibit the activity of the target, to limit the activity of the target, or to extend the activity of the target.
[00181 ] By "pharmaceutically acceptable" or "therapeutically acceptable", as used herein, refers a material, such as a carrier or diluent, which does not abrogate the biological activity or properties of the compound, and is relatively nontoxic. In certain instances, nontoxic and non-abrogative materials includes materials that when administered to an individual do not cause substantial, undesirable biological effects and/or do not interact in a deleterious manner with any of the components of the composition in which it is contained.
[00182| The term "pharmaceutically acceptable salt" or "therapeutically acceptable salt", refers to a formulation of a compound that does not cause significant irritation to an organism to which it is administered and does not abrogate the biological activity and properties of the compound. In certain instances, pharmaceutically acceptable salts are obtained by reacting a compound described herein, with acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, methanesuifonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid and the like. In some instances, pharmaceutically acceptable salts are obtained by reacting a compound having acidic group described herein with a base to form a salt such as an ammonium salt, an alkali metal salt, such as a sodium or a potassium salt, an alkaline earth metal salt, such as a calcium or a magnesium salt^ a salt of organic bases such as dicyclohexylamine, TV-methyl-D-glucamine,
tris(hydroxymethyl)methylamine, and salts with amino acids such as arginine, lysine, and the like, or by other methods previously determined.
[00183] The term "pharmaceutical combination" as used herein, means a product that results from the mixing or combining of more than one active ingredient and includes both fixed and non-fixed combinations of the active ingredients. The term "fixed combination" means that the active ingredients, e.g. a compound described herein and a co-agent, are both administered to a patient simultaneously in the form of a single entity or dosage. The term "non-fixed combination" means that the active ingredients, e.g. a compound described herein and a co-agent, are administered to a patient as separate entities either simultaneously, concurrently or sequentially with no specific intervening time limits, wherein such administration provides effective levels of the two compounds in the body of the patient. The latter also applies to cocktail therapy, e.g. the administration of three or more active ingredients.
[00184] The term "pharmaceutical composition" refers to a mixture of a compound described herein with other chemical components, such as carriers, stabilizers, diluents, dispersing agents, suspending agents, thickening agents, and/or excipients. The pharmaceutical composition facilitates administration of the compound to an organism. Multiple techniques of administering a compound exist in the art including, but not limited to: intravenous, oral, aerosol, parenteral, ophthalmic, pulmonary and topical administration.
[00185] A "prodrug" refers to an agent that is converted into the parent drug in vivo. Prodrugs are often useful because, in some situations, they are easier to administer than the parent drug. In certain instances, a prodrug is bioavailable by oral administration whereas the parent is not. In some instances, a prodrug has improved solubility in pharmaceutical compositions over the parent drug. An example, without limitation, of a prodrug is a compound described herein, which is administered as an ester (the "prodrug") to facilitate transmittal across a cell membrane where water solubility is detrimental to mobility but which then is metabolically hydrolyzed to the carbo.xylic acid, the active entity, once inside the cell where water-solubility is beneficial. A further example of a prodrug might be a short peptide (polyaminoacid) bonded to an acid or amino group where the peptide is metabolized to reveal the active moiety. In certain embodiments, upon in vivo administration, a prodrug is chemically converted to the biologically, pharmaceutically or therapeutically more active form of the compound. In certain embodiments, a prodrug is enzymatically metabolized by one or more steps or processes to the biologically, pharmaceutically or therapeutically active form of the compound. To produce a prodrug, a pharmaceutically active compound is modified such that the active compound will be regenerated upon in vivo administration. In some embodiments, the prodrug is designed to alter the metabolic stability or the transport characteristics of a drug, to mask side effects or toxicity, to improve the flavor of a drug or to alter other characteristics or properties of a drug.
100186] The term "subject" or "patient" encompasses mammals and non-mammals. Examples of mammals include, but are not limited to, any member of the Mammalian class: humans, non-human primates such as chimpanzees, and other apes and monkey species; farm animals such as cattle, horses, sheep, goats, swine; domestic animals such as rabbits, dogs, and cats; laboratory animals including rodents, such as rats, mice and guinea pigs, and the like. Examples of non-mammals include, but are not limited to, birds, fish and the like. In one embodiment of the methods and compositions provided herein, the mammal is a human.
[00187] The terms "treat," "treating" or "treatment," as used herein, include alleviating, abating or ameliorating a disease or condition symptoms, preventing additional symptoms, ameliorating or preventing the underlying metabolic causes of symptoms, inhibiting the disease or condition, e.g. , arresting the development of the disease or condition, relieving the disease or condition, causing regression of the disease or condition, relieving a condition caused by the disease or condition, or stopping the symptoms of the disease or condition either prophylactically and/or therapeutically.
Pharmaceutical Composition/Formulation
J00188] Provided herein are pharmaceutical compositions that include a compound described herein and a pharmaceutically acceptable diluent(s), excipient(s), and/or carrier(s). In addition, in some embodiments, the compounds described herein are administered as pharmaceutical
compositions in which compounds described herein are mixed with other active ingredients, as in combination therapy.
[00189] A pharmaceutical composition, as used herein, refers to a mixture of a compound described herein with other chemical components, such as carriers, stabilizers, diluents, dispersing agents, suspending agents, thickening agents, and/or excipients. In certain embodiments, a pharmaceutical composition facilitates administration of the compound to an organism. In some embodiments, practicing the methods of treatment or use provided herein, includes administering or using a pharmaceutical composition comprising a therapeutically effective amount of a compound provided herein. In specific embodiments, the methods of treatment provided for herein include administering such a pharmaceutical composition to a mammal having a disease or condition to be treated. In one embodiment, the mammal is a human. In some embodiments, the therapeutically effective amount varies widely depending on the severity of the disease, the age and relative health of the subject, the potency of the compound used and other factors. In various embodiments, the compounds described herein are used singly or in combination with one or more therapeutic agents as components of mixtures.
[00190] In certain embodiments, the pharmaceutical compositions provided herein are formulated for intravenous injections. In certain aspects, the intravenous injection formulations provided herein are formulated as aqueous solutions, and, in some embodiments, in physiologically compatible buffers such as Hank's solution, Ringer's solution, or physiological saline buffer. In certain embodiments, the pharmaceutical compositions provided herein are formulated for transmucosal administration. In some aspects, transmucosal formulations include penetrants appropriate to the barrier to be permeated. In certain embodiments, the pharmaceutical compositions provided herein are formulated for other parenteral injections, appropriate formulations include aqueous or nonaqueous solutions, and in one embodiment, with physiologically compatible buffers or excipients.
[00191] In certain embodiments, the pharmaceutical compositions provided herein are formulated for oral administration. In certain aspects, the oral formulations provided herein comprise compounds described herein that are formulated with pharmaceutically acceptable carriers or excipients. Such carriers enable the compounds described herein to be formulated as tablets, powders, pills, dragees, capsules, liquids, gels, syrups, elixirs, slurries, suspensions and the like, for oral ingestion by a patient to be treated.
(00192] In some embodiments, pharmaceutical preparations for oral use are obtained by mixing one or more solid excipient with one or more of the compounds described herein, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients include, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as: for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methylceliulose, microcrystalline cellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose; or others such as: polyvinylpyrrolidone (PVP or povidone) or calcium phosphate. If desired, disintegrating agents are optionally added, such as the cross-linked croscarmellose sodium, polyvinylpyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
|00193] In certain embodiments, provided herein is a pharmaceutical composition formulated as dragee cores with suitable coatings. In certain embodiments, concentrated sugar solutions are used in forming the suitable coating, and optionally contain gum arabic, talc, polyvinylpyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. In some embodiments, dyestuffs and/or pigments are added to tablets, dragees and/or the coatings thereof for, e.g. , identification or to characterize different combinations of active compound doses.
[00194] In certain embodiments, pharmaceutical preparations which are used include orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. In some embodiments, the push-fit capsules contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In certain embodiments, in soft capsules, the active compounds are dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers are optionally added. In certain embodiments, the formulations for oral administration are in dosages suitable for such administration.
[00195] In certain embodiments, the pharmaceutical compositions provided herein are formulated for buccal or sublingual administration. In certain embodiments, buccal or sublingual compositions take the form of tablets, lozenges, or gels formulated in a conventional manner. In certain embodiments, parenteral injections involve bolus injection or continuous infusion. In some embodiments, formulations for injection are presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative. In some embodiments, the pharmaceutical composition described herein is in a form suitable for parenteral injection as a sterile suspensions, solutions or emulsions in oily or aqueous vehicles, and optionally contains formulatory agents such as suspending, stabilizing and/or dispersing agents. Pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form. In some embodiments, suspensions of the active compounds are prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. In certain embodiments, aqueous injection suspensions contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspensions also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions. In alternative embodiments, the active ingredient is in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen- free water, before use.
[00196] In some embodiments, the compounds described herein are administered topically. In specific embodiments, the compounds described herein are formulated into a variety of topically administrable compositions, such as solutions, suspensions, lotions, gels, pastes, medicated sticks, balms, creams or ointments. Such pharmaceutical compounds optionally contain solubilizers, stabilizers, tonicity enhancing agents, buffers and/or preservatives.
[00197] In certain embodiments, the pharmaceutical compositions provided herein are formulated for transdermal administration of compounds described herein. In some embodiments, administration of such compositions employs transdermal delivery devices and transdermal delivery patches. In certain embodiments, the compositions are lipophilic emulsions or buffered, aqueous solutions, dissolved and/or dispersed in a polymer or an adhesive. Such patches include those constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents. In some embodiments, transdermal delivery of the compounds described herein is accomplished by use of iontophoretic patches and the like. In certain embodiments, transdermal patches provide controlled delivery of the compounds provided herein, such as, for example, compounds of Formula (I) or (II). In certain embodiments, the rate of absorption is slowed by using rate-controlling membranes or by trapping the compound within a polymer matrix or gel. Conversely, absorption enhancers are optionally used to increase absorption. Absorption enhancer and carrier include absorbable pharmaceutically acceptable solvents that assist in passage of the compound through the skin. For example, transdermal devices are in the form of a bandage comprising a backing member, a reservoir containing the compound optionally with carriers, optionally a rate controlling barrier to deliver the compound to the skin of the host at a controlled and predetermined rate over a prolonged period of time, and means to secure the device to the skin.
[00198] In certain embodiments, the pharmaceutical compositions provided herein are formulated for administration by inhalation. In certain embodiments, in such pharmaceutical compositions formulated for inhalation, the compounds described herein are in a form as an aerosol, a mist or a powder. In some embodiments, pharmaceutical compositions described herein are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebuliser, with the use of a suitable propellant, e.g. , dichlorodifluoromethane, trichlorofluoromethane,
dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In certain aspects of a pressurized aerosol, the dosage unit is determined by providing a valve to deliver a metered amount. In certain embodiments, capsules and cartridges of, such as, by way of example only, gelatin for use in an inhaler or insufflator is formulated containing a powder mix of the compound described herein and a suitable powder base such as lactose or starch.
[00199] In some embodiments, the compounds described herein are formulated in rectal compositions such as enemas, rectal gels, rectal foams, rectal aerosols, suppositories, jelly suppositories, or retention enemas. In certain embodiments, rectal compositions optionally contain conventional suppository bases such as cocoa butter or other glycerides, as well as synthetic polymers such as polyvinylpyrrolidone, PEG, and the like. In certain suppository forms of the compositions, a low-melting wax such as, but not limited to, a mixture of fatty acid glycerides, optionally in combination with cocoa butter is first melted.
[00200] In various embodiments provided herein, the pharmaceutical compositions are formulated in a conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into pharmaceutically acceptable preparations. In certain embodiments, proper formulation is dependent upon the route of
administration chosen. In various embodiments, any of the techniques, carriers, and excipients is used as suitable. In some embodiments, pharmaceutical compositions comprising a compound described herein are manufactured in a conventional manner, such as, by way of example only, by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or compression processes.
[00201 J In certain embodiments, the pharmaceutical compositions include at least one pharmaceutically acceptable carrier, diluent or excipient and a compound described herein described herein as an active ingredient in free-acid or free-base form, or in a pharmaceutically acceptable salt form. In addition, the methods and pharmaceutical compositions described herein include the use of iV-oxides, crystalline forms (also known as polymorphs), as well as active metabolites of these compounds having the same type of activity. In some situations, compounds described herein exist as tautomers. All tautomers are included within the scope of the compounds presented herein.
Additionally, included herein are the solvated and unsolvated forms of the compounds described herein. Solvated compounds include those that are solvated with pharmaceutically acceptable solvents such as water, ethanol, and the like. The solvated forms of the compounds presented herein are also considered to be disclosed herein. In some embodiments, the pharmaceutical compositions described herein include other medicinal or pharmaceutical agents, carriers, adjuvants, such as preserving, stabilizing, wetting or emulsifying agents, solution promoters, salts for regulating the osmotic pressure, and/or buffers. In additional embodiments, the pharmaceutical compositions described herein also contain other therapeutically valuable substances.
[00202] Methods for the preparation of compositions containing the compounds described herein include formulating the compounds with one or more inert, pharmaceutically acceptable excipients or carriers to form a solid, semi-solid or liquid. Solid compositions include, but are not limited to, powders, tablets, dispersible granules, capsules, cachets, and suppositories. Liquid compositions include solutions in which a compound is dissolved, emulsions comprising a compound, or a solution containing liposomes, micelles, or nanoparticles comprising a compound as disclosed herein. Semisolid compositions include, but are not limited to, gels, suspensions and creams. In various embodiments, the compositions are in liquid solutions or suspensions, solid forms suitable for solution or suspension in a liquid prior to use, or as emulsions. These compositions optionally contain minor amounts of nontoxic, auxiliary substances, such as wetting or emulsifying agents, pH buffering agents, and so forth.
[00203] In some embodiments, a composition comprising a compound described herein takes the form of a liquid where the agents are present in solution, in suspension or both. In some embodiments, when the composition is administered as a solution or suspension a first portion of the agent is present in solution and a second portion of the agent is present in particulate form, in suspension in a liquid matrix. In some embodiments, a liquid composition includes a gel formulation. In other embodiments, the liquid composition is aqueous.
100204] Useful aqueous suspension optionally contain one or more polymers as suspending agents. Useful polymers include water-soluble polymers such as cellulosic polymers, e.g., hydroxypropyl methylcellulose, and water-insoluble polymers such as cross-linked carboxyl- containing polymers. Useful compositions optionally comprise an mucoadhesive polymer, selected for example from carboxymethylcellulose, carbomer (acrylic acid polymer),
poly(methylmethacrylate), polyacrylamide, polycarbophil, acrylic acid/butyl acrylate copolymer, sodium alginate and dextran.
[00205] Useful compositions optionally include solubilizing agents to aid in the solubility of a compound described herein. The term "solubilizing agent" generally includes agents that result in formation of a micellar solution or a true solution of the agent. Solubilizing agents include certain acceptable nonionic surfactants, for example polysorbate 80, and ophthalmically acceptable glycols, polyglycols, e.g. , polyethylene glycol 400, and glycol ethers.
|00206] Useful compositions optionally include one or more pH adjusting agents or buffering agents, including acids such as acetic, boric, citric, lactic, phosphoric and hydrochloric acids; bases such as sodium hydroxide, sodium phosphate, sodium borate, sodium citrate, sodium acetate, sodium lactate and tris-hydroxymethylaminomethane; and buffers such as citrate/dextrose, sodium bicarbonate and ammonium chloride. Such acids, bases and buffers are included in an amount required to maintain pH of the composition in an acceptable range.
[00207] Useful compositions optionally include one or more salts in an amount required to bring osmolality of the composition into an acceptable range. Such salts include those having sodium, potassium or ammonium cations and chloride, citrate, ascorbate, borate, phosphate, bicarbonate, sulfate, thiosulfate or bisulfite anions; suitable salts include sodium chloride, potassium chloride, sodium thiosulfate, sodium bisulfite and ammonium sulfate.
[00208] Certain useful compositions optionally include one or more preservatives to inhibit microbial activity. Suitable preservatives include mercury-containing substances such as merfen and thiomersai; stabilized chlorine dioxide; and quaternary ammonium compounds such as benzalkonium chloride, cetyltrimethylammonium bromide and cetylpyridinium chloride.
[00209] Some useful compositions optionally include one or more surfactants to enhance physical stability or for other purposes. Suitable nonionic surfactants include polyoxyethylene fatty acid glycerides and vegetable oils, e.g. , polyoxyethylene (60) hydrogenated castor oil; and
polyoxyethylene alkylethers and alkylphenyl ethers, e.g. , octoxynol 10, octoxynol 40. 100210] Certain useful compositions optionally one or more antioxidants to enhance chemical stability where required. Suitable antioxidants include, by way of example only, ascorbic acid and sodium metabisulfite.
[00211] In some embodiments, aqueous suspension compositions are packaged in single-dose non-reclosable containers. In alternative embodiments, multiple-dose reclosable containers are used, in which case it is typical to include a preservative in the composition.
[00212] In various embodiments, any delivery system for hydrophobic pharmaceutical compounds is employed. Liposomes and emulsions are examples of delivery vehicles or carriers for hydrophobic drugs. In certain embodiments, certain organic solvents such as N-methylpyrrolidone are employed. In some embodiments, the compounds are delivered using a sustained-release system, such as semipermeable matrices of solid hydrophobic polymers containing the therapeutic agent. Various sustained-release materials are utilized in the embodiments herein. In certain embodiments, sustained-release capsules release the compounds for a few weeks up to over 100 days. In some embodiments, depending on the chemical nature and the biological stability of the therapeutic reagent, additional strategies for protein stabilization are employed.
[00213] In certain embodiments, the formulations or compositions described herein benefit from and/or optionally comprise antioxidants, metal chelating agents, thiol containing compounds and other general stabilizing agents. Examples of such stabilizing agents, include, but are not limited to: (a) about 0.5% to about 2% w/v glycerol, (b) about 0.1% to about 1 % w/v methionine, (c) about 0.1 % to about 2% w/v monothioglycerol, (d) about 1 mM to about 10 mM EDTA, (e) about 0.01 % to about 2% w/v ascorbic acid, (f) 0.003% to about 0.02% w/v polysorbate 80, (g) 0.001 % to about 0.05% w/v. polysorbate 20, (h) arginine, (i) heparin, (j) dextran sulfate, (k) cyclodextrins, (1) pentosan polysulfate and other heparinoids, (m) divalent cations such as magnesium and zinc; or (n) combinations thereof.
Methods of Dosing and Treatment Regimens
[00214] In any of the aforementioned embodiments are further embodiments in which administration is enteral, parenteral, or both, and wherein:
(a) the effective amount of the compound is systemically administered to the subject;
(b) the effective amount of the compound is administered orally to the subject;
(c) the effective amount of the compound is intravenously administered to the subject;
(d) the effective amount of the compound administered by inhalation;
(e) the effective amount of the compound is administered by nasal administration;
(f) the effective amount of the compound is administered by injection to the subject;
(g) the effective amount of the compound is administered topically (dermal) to the subject;
(h) the effective amount of the compound is administered by ophthalmic administration; and/or
(i) the effective amount of the compound is administered rectally to the subject. [00215] In any of the aforementioned embodiments are further embodiments that include single administrations of the effective amount of the compound, including further embodiments in which the compound is administered to the subject (i) once; (ii) multiple times over the span of one day; (iii) continually; or (iv) continuously.
[00216] In any of the aforementioned embodiments are further embodiments that include multiple administrations of the effective amount of the compound, including further embodiments wherein;
(i) the compound is administered in a single dose;
(ii) the time between multiple administrations is every 6 hours;
(iii) the compound is administered to the subject every 8 hours.
[00217] In further or alternative embodiments, the method includes a drug holiday, wherein the administration of the compound is temporarily suspended or the dose of the compound being administered is temporarily reduced; at the end of the drug holiday, dosing of the compound is resumed. In some embodiments, the length of the drug holiday varies from 2 days to 1 year.
[00218] In certain embodiments, the compounds described herein are used in the preparation or manufacture of medicaments for the treatment of diseases or conditions that are mediated by the enzyme poly(ADP-ribose)polymerase (PARP) or in which inhibition of the enzyme poly(ADP- ribose)polymerase (PARP) ameliorates the disease or condition. In some embodiments, a method for treating any of the diseases or conditions described herein in a subject in need of such treatment, involves administration of pharmaceutical compositions containing at least one compound described herein, or a pharmaceutically acceptable salt, pharmaceutically acceptable N-oxide, pharmaceutically active metabolite, pharmaceutically acceptable prodrug, or pharmaceutically acceptable solvate thereof, in therapeutically effective amounts to said subject.
[00219] In certain embodiments, the compositions containing the compound(s) described herein are administered for prophylactic and/or therapeutic treatments. In certain therapeutic applications, the compositions are administered to a patient already suffering from a disease or condition, in an amount sufficient to cure or at least partially arrest the symptoms of the disease or condition. In some embodiments, amounts effective for this use will depend on the severity and course of the disease or condition, previous therapy, the patient's health status, weight, and response to the drugs, and the judgment of the treating physician. In certain instances, it is considered appropriate for the caregiver to determine such therapeutically effective amounts by routine experimentation (including, but not limited to, a dose escalation clinical trial).
[00220] In certain prophylactic applications, compositions containing the compounds described herein are administered to a patient susceptible to or otherwise at risk, of a particular disease, disorder or condition. In some embodiments, the amount administere is defined to be a "prophylactically effective amount or dose." In certain embodiments of this use, the precise amounts of compound administered depend on the patient's state of health, weight, and the like. In some embodiments, it is considered appropriate for the caregiver to determine such prophylactically effective amounts by routine experimentation (e.g. , a dose escalation clinical trial). In certain embodiments, when used in a patient, effective amounts for this use will depend on the severity and course of the disease, disorder or condition, previous therapy, the patient's health status and response to the drugs, and the judgment of the treating physician.
|00221] In certain instances, a patient's condition does not improve or does not significantly improve following administration of a compound or composition described herein and, upon the doctor's discretion the administration of the compounds is optionally administered chronically, that is, for an extended period of time, including throughout the duration of the patient's life in order to ameliorate or otherwise control or limit the symptoms of the patient's disease or condition.
100222) In certain cases wherein the patient's status does improve or does not substantially improve, upon the doctor's discretion I he administration of the compounds are optionally given continuously; alternatively, the dose of drug being administered is optionally temporarily reduced or temporarily suspended for a certain length of time (i.e., a "drug holiday"). In certain embodiments, the length of the drug holiday varies between 2 days and 1 year, including by way of example only, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 10 days, 12 days, 15 days, 20 days, 28 days, 35 days, 50 days, 70 days, 100 days, 120 days. 1 0 days, 180 days, 200 days, 250 days, 280 days, 300 days, 320 days, 350 days, or 365 days. The dose reduction during a drug holiday includes a reduction from about 10% to about 100%, including, by way of example only, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100%.
[00223] In certain embodiments, once improvement of the patient's conditions has occurred, a maintenance dose is administered if necessary. In some embodiments, the dosage, e.g., of the maintenance dose, or the frequency of administration, or both, are reduced, as a function of the symptoms, to a level at which the improved disease, disorder or condition is retained. In certain embodiments, however, patients are optionally given intermittent treatment on a long-term basis upon any recurrence of symptoms.
[00224] In certain embodiments, the amount of a given agent that corresponds to an effective amount varies depending upon factors such as the particular compound, disease or condition and its severity, the identity (e.g., weight) of the subject or host in need of treatment. In some embodiments, the effective amount is, nevertheless, determined according to the particular circumstances surrounding the case, including, e.g., the specific agent that is administered, the route of
administration, the condition being treated, and the subject or host being treated. In certain embodiments, however, doses employed for adult human treatment is in the range of about 0.02 to about 5000 mg per day, in a specific embodiment about 1 to about 1500 mg per day. In various embodiments, the desired dose is conveniently presented in a single dose or as divided doses administered simultaneously (or over a short period of time) or at appropriate intervals, for example as two, three, four or more sub-doses per day.
[00225] In some embodiments, the pharmaceutical compositions described herein are in a unit dosage form suitable for single administration of precise dosages. In some instances, in unit dosage form, the formulation is divided into unit doses containing appropriate quantities of one or more compound. In certain embodiments, the unit dosage is in the form of a package containing discrete quantities of the formulation. Non-limiting examples are packaged tablets or capsules, and powders in vials or ampoules. In some embodiments, aqueous suspension compositions are packaged in single- dose non-reclosable containers. In alternative embodiments, multiple-dose reciosable containers are used, in which case it is typical to include a preservative in the composition. By way of example only, formulations for parenteral injection are, in some embodiments, presented in unit dosage form, which include, but are not limited to ampoules, or in multi-dose containers, with an added preservative.
[00226] In certain embodiments, the daily dosages appropriate for the compounds described herein described herein are from about 0.01 to about 2.5 mg/kg per body weight. In some embodiments, an indicated daily dosage in the larger subject, including, but not limited to, humans, is in the range from about 0.5 mg to about 100 mg, conveniently administered in divided doses, including, but not limited to, up to four times a day or in extended release form. In certain embodiments, suitable unit dosage forms for oral administration comprise from about 1 to about 50 mg active ingredient. The foregoing ranges are merely suggestive, as the number of variables in regard to an individual treatment regime is large, and considerable excursions from these
recommended values are not uncommon. In certain embodiments, the dosages are altered depending on a number of variables, not limited to the activity of the compound used, the disease or condition to be treated, the mode of administration, the requirements of the individual subject, the severity of the disease or condition being treated, and the judgment of the practitioner.
|00227] In certain embodiments, toxicity and therapeutic efficacy of such therapeutic regimens are determined by standard pharmaceutical procedures in cell cultures or experimental animals, including, but not limited to, the determination of the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between the toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio between LD50 and ED50. In certain embodiments, compounds exhibiting high therapeutic indices are preferred. In some embodiments, the data obtained from cell culture assays and animal studies is used in formulating a range of dosage for use in human. In specific embodiments, the dosage of such compounds lies within a range of circulating concentrations that include the ED50 with minimal toxicity. In certain embodiments, the dosage varies within this range depending upon the dosage form employed and the route of administration utilized.
Combination Treatments
|00228| In certain instances, it is appropriate to administer at least one compound described herein in combination with another therapeutic agent. By way of example only, if one of the side effects experienced by a patient upon receiving one of the compounds herein is inflammation, then, in some embodiments, it is appropriate to administer an anti-inflammatory agent in combination with the initial therapeutic agent. In some embodiments, the therapeutic effectiveness of one of the compounds described herein is enhanced by administration of an adjuvant (i.e., in some embodiments, by itself the adjuvant has minimal therapeutic benefit, but in combination with another therapeutic agent, the overall therapeutic benefit to the patient is enhanced). In certain embodiments, the benefit experienced by a patient is increased by administering one of the compounds described herein with another therapeutic agent (which also includes a therapeutic regimen) that also has therapeutic benefit. In some embodiments, regardless of the disease, disorder or condition being treated, the overall benefit experienced by the patient as a result of a combination treatment is additive or synergistic.
[00229] In certain embodiments, therapeutically-effective dosages vary when the drugs are used in treatment combinations. In some embodiments, therapeutically-effective dosages of drugs and other agents for use in combination treatment regimens is determined in any suitable manner, e.g., through the use of metronomic dosing, i.e., providing more frequent, lower doses in order to minimize toxic side effects. In some embodiments, combination treatment regimen described herein encompass treatment regimens in which administration of a PA P inhibitor described herein is initiated prior to, during, or after treatment with a second agent described above, and continues until any time during treatment with the second agent or after termination of treatment with the second agent. It also includes treatments in which a PARP inhibitor described herein and the second agent being used in combination are administered simultaneously or at different times and/or at decreasing or increasing intervals during the treatment period. Combination treatment further includes periodic treatments that start and stop at various times to assist with the clinical management of the patient. For example, in some embodiments, a PARP inhibitor described herein in the combination treatment is administered weekly at the onset of treatment, decreasing to biweekly, and decreasing further as appropriate.
|00230] In certain embodiments, compositions and methods for combination therapy are provided herein. In accordance with one aspect, the pharmaceutical compositions disclosed herein are used to in a method of treating a PARP mediated disease or condition or a disease or condition that is ameliorated by inhibition of PARP. In accordance with certain aspects, the pharmaceutical compositions disclosed herein are used to treat vascular disease; septic shock; ischaemic injury; reperfusion injury; neurotoxicity; hemorrhagic shock; inflammatory diseases; multiple sclerosis; secondary effects of diabetes; and acute treatment of cytotoxicity following cardiovascular surgery. In a certain aspect, the pharmaceutical compositions disclosed herein are used in combination, either simultaneously or sequentially, with ionizing radiation or one or more chemotherapeutic agents.
(00231 j In certain embodiments, combination therapies described herein are used as part of a specific treatment regimen intended to provide a beneficial effect from the co-action of a PARP inhibitor described herein and a concurrent treatment. It is understood that the dosage regimen to treat, prevent, or ameliorate the condition(s) for which relief is sought, is optionally modified in accordance with a variety of factors.
[00232] In certain combination therapies described herein, dosages of the co-administered compounds vary depending on the type of co-drug employed, on the specific drug employed, on the disease or condition being treated and so forth. In some embodiments, when co-administered with one or more biologically active agents, the compound provided herein is administered either
simultaneously with the biologically active agent(s), or sequentially. In certain aspects wherein the agents are administered sequentially, the attending physician will decide on the appropriate sequence of administering protein in combination with the biologically active agent(s).
[00233] In various embodiments, the multiple therapeutic agents (one of which is one of the compounds described herein) are administered in any order or even simultaneously. In certain instances, administration is simultaneous and the multiple therapeutic agents are, optionally, provided in a single, unified form, or in multiple forms (by way of example only, either as a single pill or as two separate pills). In some embodiments, one of the therapeutic agents is given in multiple doses, or both are given as multiple doses. In some instances, administration is not simultaneous and the timing between the multiple doses varies, by way of non-limiting example, from more than zero weeks to less than four weeks. In addition, the combination methods, compositions and formulations are not to be limited to the use of only two agents; the use of multiple therapeutic combinations are also envisioned.
[00234] In additional embodiments, the compounds described herein are used in combination with procedures that provide additional or synergistic benefit to the patient. By way of example only, patients are expected to find therapeutic and/or prophylactic benefit in the methods described herein, wherein pharmaceutical composition of a compound disclosed herein and/or combinations with other therapeutics are combined with genetic testing to determine whether that individual is a carrier of a mutant gene that is known to be correlated with certain diseases or conditions.
[00235] In certain embodiments, the compounds described herein and combination therapies are administered before, during or after the occurrence of a disease or condition. In certain embodiments, the timing of administering the composition containing a compound varies. Thus, for example, in some embodiments, the compounds are used as a prophylactic and are administered continuously to subjects with a propensity to develop conditions or diseases in order to prevent the occurrence of the disease or condition. In some embodiments, the compounds and compositions are administered to a subject during or as soon as possible after the onset of the symptoms. In certain embodiments, the administration of the compounds is initiated within the first 48 hours of the onset of the symptoms, within the first 6 hours of the onset of the symptoms, or within 3 hours of the onset of the symptoms. The initial administration is achieved via any route practical, such as, for example, an intravenous injection, a bolus injection, infusion over 5 minutes to about 5 hours, a pill, a capsule, transdermal patch, buccal delivery, and the like, or combination thereof. In some embodiments, a compound is administered as soon as is practicable after the onset of a disease or condition is detected or suspected, and for a length of time necessary for the treatment of the disease, such as, from about 1 month to about 3 months. In certain embodiments, the length of treatment varies for each subject, and the length is determined using any criteria. In exemplary embodiments, a compound or a formulation containing the compound is administered for at least 2 weeks, for about 1 month to about 5 years, or for about 1 month to about 3 years.
Other Combination Therapies
|00236] In certain embodiments described herein, methods for treatment of PARP mediated conditions or diseases, such as proliferative disorders, including cancer, include administration to a patient compounds, pharmaceutical compositions, or medicaments described herein in combination with at least one additional agent selected from the group consisting of alemtuzumab, arsenic trioxide, asparaginase (pegylated or non-), bevacizumab, cetuximab, platinum-based compounds such as cisplatin, cladribine, daunorubicin/doxombicin/idarubicin, irinotecan, fludarabine, 5-fluorouracil, gemtuzumab, methotrexate, Paclitaxel™, taxol, temozolomide, thioguanine, or classes of drugs including hormones (an antiestrogen, an antiandrogen, or gonadotropin releasing hormone analogues, interferons such as alpha interferon, nitrogen mustards such as busulfan or melphalan or
mechlorethamine, retinoids such as tretinoin, topoisomerase inhibitors such as irinotecan or topotecan, tyrosine kinase inhibitors such as gefinitinib or imatinib, or agents to treat signs or symptoms induced by such therapy including allopurinol, filgrastim, granisetron/ondansetron/palonosetron, and dronabinol.
[00237] In certain embodiments, the compounds, pharmaceutical compositions, or medicaments described herein are administered in combination with ionizing radiation, one or more
chemotherapeutic agents, or a combination thereof.
[00238] In certain embodiments, the chemotherapeutic agent is independently selected from the group consisting of alemtuzumab, arsenic trioxide, pegylated asparaginase, non-pegylated
asparaginase, bevacizumab, cetuximab, cisplatin, cladribine, daunorubicin/doxorubicin/idarubicin, irinotecan, fludarabine, 5-fluorouracil, gemtuzumab, methotrexate, Paclitaxel™, taxol, temozolomide, thioguanine, an antiestrogen hormone analogue, an antiandrogen hormone analogue, a gonadotropin releasing hormone analogue, alpha interferon, busulfan, melphalan, mechlorethamine, tretinoin, irinotecan, topotecan, gefinitinib, imatinib, allopurinol, Filgrastim,
granisetron/ondansetron/palonosetron, and dronabinol.
[00239] In certain embodiments, the compounds, pharmaceutical compositions, or medicaments described herein are administered in combination with one or more of chemotherapeutic agents where each chemotherapeutic agent is independently an alkylating agent or a topoisomerase-1 inhibitor.
[00240] In certain embodiments, the compounds, pharmaceutical compositions, or medicaments described herein are administered in combination with one or more chemotherapeutic agents where each chemotherapeutic agent is independently selected from the group consisting of methyl methanesulfonate, temozolomide, dacarbazine (DTIC), Topotecan, Irinotecan, Rubitecan, Exatecan, Lurtotecan, Gimetecan, Diflomotecan (homocamptothecins), 7-substituted non-silatecans, 7-silyl camptothecins, BNP 1350, and XR 1 1576/MLN 576.
|00241 ] In certain embodiments, the compounds, pharmaceutical compositions, or medicaments described herein are administered in combination with one chemotherapeutic agent where the chemotherapeutic agent is irinotecan, cisplatin, or temozolomide.
EXAMPLES
100242] The following Examples are intended as an illustration of the various embodiments as defined in the appended claims. All publications, patents, and patent applications cited herein are hereby incorporated by reference for all purposes.
(00243] Examples 1 -4 were disclosed in a poster which was presented December 4, 2010, at the 52nd American Society of Hematology Annual Meeting and Exposition in Orlando, Florida.
Example 1 : PARP Inhibitor Sensitivity in MDS/AML Cell Lines Correlates with the Presence of High Grade Microsatellite Instability (MSI)
[00244] This example demonstrates that MDS/AML cell lines that are sensitive to PARP inhibitor, Compound 1 , exhibit high grade MSI.
[00245] FIG. 1 shows that MDS/AML cell lines exhibited a varied response to PARP inhibitor, Compound 1. Cell lines, P39, KG- 1 and Molm-13 exhibited significant cytotoxicity to Compound 1 , whereas MDS/AML cell lines Molt 4, NB4 and normal peripheral blood lymphocytes exhibited relative insensitivity to Compound 1 . MDS/ AML cell lines were obtained from American Type Culture Collection (ATCC). [00246] A panel of six mononucleotide repeat sequence markers (NR21, NR22, NR24, Bat25, Mono27 and Bat26) were used to estimate MSI in each cell line. Cell lines sensitive to Compound 1 (P39, KG- 1 and Molm- 13) exhibited high grade MSI (> 2 loci with > 3 nucleotide errors) (see FIG. 2 and FIG. 3). For example, P39 showed 5/6 monoallelic variation in MSI loci, while KG-1 and Molm- 13 showed 5/6 mono and bi-allelic variation in MSI loci.
[00247] TABLE 1 presents mononucleotide repeat sequence (NR21, NR22, NR24, Bat25, Mono27, and Bat26) versus MDS/AML cell line (PBL, HL60, Jurkat, KG- 1 , Molm- 13, Molt 4, NB4, OCI-AML3, P39, Raji, REH, DS- 1 , U937 and DLD 1 ). Values refer to the size of fluorescent PCR product run in an ABI Genetic Analyzer 3130XL (bp, base pair). Underlined and bolded values were PCR products with > 3 bp deletions or additions. Cell lines were classified as one of four groups: ( 1 ) MSI-stable with no aberrant MSI; (2) MSI-low with one aberrant microsatellite; (3) MSI-mid with > 2 aberrant microsatellite loci; and (4) MSI-high (high grade MSI) with > 3 aberrant microsatellite loci (e.g. , P39, KG- 1 , NB4 and Molm- 13).
TABLE 1: Mononucleotide repeat sequence versus MDS/AML cell line
Figure imgf000058_0001
Example 2: MSI-high MDS/AML Cell Lines Possess MSI in the Coding Region
Microsatellites of Double Stranded DNA Repair Genes
[002481 This example demonstrates that MSI-high MDS/AML cell lines possess MSI in the coding region of microsatellites of double stranded repair genes Mrel 1 , A TM and CTiP.
[00249] Double stranded D A repair genes Mrel 1 , A TM, CTiP, CHKl , PTEN, BLM and A TR possess specific microsatellites that when mutated produce frameshift mutations. Using fluorescent PCR and Sanger sequencing, MSI at these loci in a panel of MDS/AML cell lines were tested.
[00250] Monoallelic 1 bp deletions were observed in the microsatellites of double stranded DNA repair genes CTiP and Mrel 1 {see FIG. 4 and FIG. 5, respectively; TABLE 2) and a 2 bp deletion was observed in the microsateilite of double stranded DNA repair gene ATM (see FIG. 7) of the MSI- high cell lines P39, KG- 1 , NB4 and Molm- 13. These specific microsateilite deletions were not observed in cell lines that were MSI-stable (see FIG. 6; TABLE 2). No microsateilite deletions were observed for any of the cell lines for CHKl, Rad50, PTEN, BLM and A TR.
[00251] TABLE 2 presents the MSI data for double stranded DNA repair genes (Rad50, Mrel I, CTiP, CHKl, BLM, PTEN, A TM and A TR) versus MDS/AML cell line (PBL, KG- 1 , Molm- 13, Molt 4, NB4, P39 and DLD 1 ). Values refer to the size of fluorescent PCR product run in an ABI Genetic Analyzer 3130XL (bp, base pair). Underlined and bolded values were PCR products with > 3 bp deletions or additions.
TABLE 2: Microsateilite deletions in double stranded DNA repair genes in various MDS/AML cell line
Figure imgf000059_0001
Example 3: Jntronic Mononuceotide Microsateilite Mutations Generate Aberrant Transcript
Splice Products in A TM (497deI22) and Mrell (315d3el88)
[00252] This example demonstrates that intronic mononucleotide microsateilite mutations generate aberrant transcript splice products in double stranded DNA repair genes A TM (497del22) and Mrell (315d3el88) in MSI-high MDS/AML cell lines only. [00253] PCR for exon 8 of A TM and exon 5 of Mrel l was performed on the cDNA of cell lines D L, Jurkat, 562, KG- 1 , Molm- 13, Molt 4, NB4 and P39. The PCR of cDNA from exon 8 of A TM (FIG. 8) and exon 5 of Mrel l (FIG. 9) showed aberrant splice transcripts in MSI-high cell lines P39, Molm- 13 and KG- 1.
Example 4: High Risk MDS Patients' Cells Exhibit MSI Correlating with Chromosomal
Instability
[00254] This example demonstrates that high risk MDS patients exhibit MSI correlating with chromosomal instability.
[00255] High risk MDS is associated with chromosomal instability, an increased likelihood of AML transformation and poor prognosis. High risk MDS patients were tested using a panel of six mononucleotide repeat sequence markers (NR21, NR22, NR24, Bat25, Mono27 and Bat2€) by Sanger sequencing and fluorescent PCT MSI analysis, and were further tested for chromosomal instability and mutations in their CTiP DNA repair genes. Cytogenetics were determined using Giemsa staining at metaphase spreads. Single Nucleotide Polymorphisms Analysis (SNPA) was used to determine cryptic chromosomal abnormalities, e.g., uniparental disomy (UPD), not seen using conventional cytogenetic analysis.
[00256] The results are presented in FIG. 10 and TABLE 3. Of the 63 high risk MDS patients, 13 (21%) exhibited MSI (9 MSI-low and 4 MSI-high). Of these 13 MSI-positive patients, 7 (4 MSI- high, 3 MSI-low) exhibited monosomy 7 and other complex chromosomal abnormalities (Group 1 ); 2 (MSI-low) patients exhibited isolated monosomy 7 (Group 2); and 4 (MSI-low) exhibited normal cytogenetics (Group 3). Significantly, Group 3 with MSI and normal cytogenetics all exhibited widespread uniparental disomy (UPD) and cryptic chromosome changes, as determined by single nucleotide polymorphism analysis (SNPA). By contrast, 12 of the 63 high risk MDS patients (19%) found to be cytogenctically normal and lacking UPD and genomic aberrations by SNPA did not exhibit MSI. Further, 3 patients with MSI-high (Group 1 ) and 1 patient with MSI-low (Group 3) exhibited a monoallelic 1 bp deletion in the CTiP exon coding microsatellite (see FIG. 10), which would result in an abbreviated CTiP gene transcript.
TABLE 3: Incidence of chromosomal instability in high risk MDS patients
Figure imgf000060_0001
5 1 ; NR24 Yes Yes Yes
6 2- NR21+ Bat 25 Yes Yes Yes
7 l ; NR21+ Bat25 Yes Yes Yes
8 i ; Bat25 Yes Yes No
9 \ Bat25 Yes Yes No
10 \ NR24 Yes Yes No
1 1 2; NR21 + Bal 5 Yes Yes No
12 \ ; NR21 No Yes No
13 \ - Bat25 Yes Yes No
[00257] Identification of a cohort of MDS/AML patients with MSI would be helpful for screening of candidates for PARP inhibitor therapy.
Example 5: Screening of MDS/AML Patients for PARP Inhibitor Therapy
[00258] This example provides an exemplary screening of MDS/AML patients for PARP inhibitor therapy.
|00259] Microsatellite markers in genomic DNA of a sample from a MDS/AML patient, e.g., a tumor biopsy sample, may be isolated and compared to microsatellite markers in genomic DNA isolated from normal tissue or cells in the patient, or in a healthy reference individual, in order to detect MSI. Specifically, microsatellite loci may be amplified and genotyped. Specific procedures used in this example are provided below.
[00260] Samples and DNA isolation. The sample may be a histological sample, a biopsy, or a cytological sample such as, e.g., a smear, a swab, a wash, a body fluid containing. The sample may comprise a fixed or preserved specimen, such as a cytological or a histological specimen. The sample may be a tumor biopsy tissue obtained by surgical resection.
[00261 ] PCR and Microsatellite Analysis. A panel of five nearly monomorphic mononucleotide markers (NR21, NR22, NR24, Bat25, Bat26 and Mono27) may be used for MSI determination of the patient's genomic DNA. The analysis may involve fluorescent polymerase chain reaction (PCR) and/or Sanger sequencing.
[00262] Immunohistochemistry: Conventional cytogenetic may be used to analyze for chromosomal abnormalities in the patient's sample. Single Nucleotide Polymorphisms Analysis (SNPA) may be used to analyze for cryptic chromosomal abnormalities in the patient's sample.
100263] Result: MSI comprising a > 3 bp deletion or insertion in one or more microsatellite(s) of the patient's malignant DNA, correlating with chromosomal instability, indicates that the patient is likely to respond to treatment with a compound of Formula I or II. Example 6: Phase II Clinical Trial of the Safety and Efficacy of Compounds of
Formula (I) or (II)
[00264] This example provides an exemplary phase II trial which purpose is to study the side effects and best dose of a compound of Formula (I) or (II) and to determine how well it works in treating MDS/AML patients with MSI in malignant cells.
Objectives-:
Primary:
A. To determine the response rate to a compound of Formula (I) or (II) in MDS/AML patients with or without MSI
B. To evaluate the toxicity of a compound of Formula (I) or (II) in these patients
Secondary:
A. To evaluate the time to progression and overall survival in patients treated with a compound of Formula (I) or (II)
B. To study pharmacokinetics of a compound of Formula (I) or (II) in these patients
C. To evaluate the Poly(ADP-ribose) polymerase (PARP) activity in peripheral blood lymphocytes from these patients
Tertiary:
A. To evaluate PARP expression using quantitative western blotting immuno-assays
B. To investigate pharmacogenomics, including CYP2D6 and CYP3A5, drug transport proteins, as well as polymorphisms in the genes coding for the PARP enzymes themselves
C. To analyze for mutations in their DNA repair genes from original diagnostic biopsies (when possible)
D. To analyze paraffin sections for DNA repair enzyme status using immunohistochemical techniques (from biopsies, when possible)
E To analyze cells obtained from ascitic or pleural fluid (where possible) for primary cell culture for DNA double strand break repair pathway function
F. To perform composite microsatellite analysis of mononucleotide repeat sequences to determine MSI (from biopsies, when possible)
G. To perform conventional cytogenetic analysis to determine chromosomal abnormalities (from biopsies, when possible)
H. To perform Single Nucleotide Polymorphisms Analysis (SNPA) to determine cryptic chromosomal abnormalities (from biopsies, when possible)
F. To analyze for PARP activity and PARP expression (from biopsies, when possible) Patients: Eligible subjects will be men and women 18 years and older
Criteria:
• Disease Characteristics: o Patients with one of the following types of bone marrow disorders as determined by the Laboratory of Pathology or Hematology at the Clinical Center, National Institutes of Health: MDS, AML or chronic myelomonocytic leukemia
o No more than 3 prior chemotherapy regimens for patients with leukemia
o Measurable disease, as defined by RECIST criteria and measured by X-ray, CT scan, or MRI
Patients with bone disease must have other measurable disease for evaluation
Previously irradiated lesions cannot be used for measurable disease
o No known brain metastases
Patient Characteristics;
o WHO performance status 0- 1
o Life expectancy > 12 weeks
o Hemoglobin > 9.0 g/dL
o Absolute neutrophils > 1 ,500/mm3
o Platelets > 100,000/mm3
o Serum bilirubin < 1.5 times upper limit of normal (ULN)
o ALT or AST < 2.5 times ULN (< 5 times ULN if due to tumor)
o Glomerular filtration rate (GFR) > 50 mL/min
o Not pregnant or nursing
o Negative pregnancy test
o Fertile patients must use two highly effective forms of contraception (i.e., oral,
injected, or implanted hormonal contraception, intrauterine device, barrier method of condom plus spermicide, or are surgically sterile) 4 weeks prior to (females), during, and for 6 months after (males and females) completion of study therapy
o Able to cooperate with treatment and follow-up
o No non-malignant systemic disease, including active uncontrolled infection
o No other concurrent malignancy, except adequately treated cone-biopsied carcinoma in situ of the uterine cervix, basal cell or squamous cell carcinoma of the skin, or breast and ovarian carcinoma
Cancer survivors who have undergone potentially curative therapy for a prior malignancy, have no evidence of that disease for 5 years, and are deemed at low risk for recurrence are eligible
o No active or unstable cardiac disease or history of myocardial infarction within the past 6 months
Patients with cardiovascular signs or symptoms should have a MUGA scan or echocardiogram, and those with a left ventricular ejection fraction (LVEF) below the institutional limit of normal should be excluded o No other condition which, in the investigator's opinion, would not make the patient a good candidate for this study
Prior Concurrent Therapy:
• At least 4 weeks since prior radiotherapy (except for palliative reasons), endocrine
therapy, immunotherapy or chemotherapy (6 weeks for nitrosoureas and mitomycin C)
• At least 4 weeks since prior major thoracic and/or abdominal surgery and recovered
• Concurrent radiotherapy for the control of bone pain or skin lesions allowed, but not within 5 days of the last dose of study drug
• Concurrent bisphosphonates allowed provided the dose is stable and treatment was started at least 2 weeks prior to recruitment
• No unresolved toxicities (CTCAE > grade 1) from prior treatments (except for alopecia)
• No concurrent anticancer therapy or investigational drugs
• No concurrent tetracycline antibiotic therapy for prolonged periods (short courses [5-7 days] for treatment of infection are allowed)
• Study Design: This is a dose -escalation study followed by an open label multicenter study.
Patients will be stratified according to disease state (MDS vs AiML vs chronic
myelomonocytic leukemia) and MSI status. Patients will receive a compound of Formula (I) or (II) (at one of several possible dosages) over 30 minutes once daily on days 1 -5. Treatment repeats every 21 days for 12 courses in the absence of disease progression or unacceptable toxicity. Patients who achieve stable or responding disease may receive additional courses of treatment at the discretion of the chief investigator or Drug
Development Office (DDO). Patients will undergo blood sample collection periodically for pharmacokinetic and pharmacodynamic studies. Samples will be analyzed for tumor marker measurements, plasma levels of a compound of Formula (I) or (II) via liquid chromatography/mass spectrometry/mass spectrometry, PARP activity, and PARP protein expression via western blotting immunoassays. Paraffin embedded sections from original diagnostic biopsy are also collected and analyzed for PARP protein expression via immunohistochemical technique. Pleural and ascitic fluid may be collected and analyzed for DNA DS break repair proficiency via immunohistochemical technique. Some patients will also undergo biopsy of tumors and samples will be analyzed for DNA repair enzyme mutation status, PARP activity via validated PARP immunoblotting assay, and chromosomal status using conventional cytogenetic analysis and SNPA analysis
After completion of the study treatment, patients will be followed for 28 days.
Primary Outcome xMeasures:
• Assessment of antitumor activity according to RECIST using tumor size measured
clinically or radiologically with CT scan, MRI, plain x-ray, or other imaging techniques • Safety profile
Secondary Outcome Measures:
• Time to progression and overall survival
• Plasma levels by Liquid Chromatography/Mass Spectrometry/Mass Spectrometry
• Poly(ADP-ribose) polymerase (PARP) activity measured ex vivo using validated assays
• PARP expression using quantitative Western blotting immuno-assays
• Pharmacogenomics including CYP2D6 and CYP3A5, drug transport proteins, as well as polymorphisms in the genes coding for the PARP enzymes themselves
• DNA repair enzyme mutation status, MSI status, PARP activity, and PARP expression in tumor biopsy samples (when possible)
• DNA repair enzyme status using immunohistochemical techniques in paraffin sections from original diagnostic biopsies/operative procedures (when possible)
• Chromosomal status using conventional cytogenetic analysis and SNPA analysis (when possible)
• DNA double strand break repair pathway function in cells obtained from ascitic or pleural fluid (when possible) for primary cell culture
[00265] Other objects, features and advantages of the compounds, methods and compositions described herein will become apparent from the following description. It should be understood, however, that the description and the specific examples, while indicating specific embodiments, are given by way of illustration only, since various changes and modifications within the spirit and scope of the present description will become apparent from this detailed description.
[00266] All references cited herein are incorporated herein by reference in their entirety and for all purposes to the same extent as if each individual publication or patent or patent application was specifically and individually indicated to be incorporated by reference in its entirety for all purposes.

Claims

CLAIMS WHAT IS CLAIMED IS:
1. A method of treating Myelodysplastic Syndrome (MDS) or Acute Myeloid Leukemia (AML) in a patient with MDS or AML, comprising administering to a patient with MDS or AML a therapeutically effective amount of a compound of Formula (1):
Figure imgf000066_0001
Formula (I),
wherein:
Y and Z are each independently selected from the group consisting of:
a) an aryl group optionally substituted with 1 , 2, or 3 R<; ;
b) a heteroaryl group optionally substituted with 1 , 2, or 3 R6; and
c) a substituent independently selected from the group consisting of hydrogen, alkenyl, alkoxy, alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, alkyl, alkynyl, arylalkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, hydroxyalkylene, oxo, heterocycloalkyl, heterocycloalkylalkyl, alkylcarbonyl, arylcarbonyl, heteroarylcarbonyl alkylsulfonyl, arylsulfonyl,
heteroarylsulfonyl, (NRARB)alkylene, (NRARB)carbonyl, (NRARB)carbonylalkylene, (NRARB)sulfonyl, and (NRARB)sulfonylalkylene;
R, , R2, and R3 are each independently selected from the group consisting of hydrogen, halogen, alkenyl, alkoxy, alkoxycarbonyl, alkyl, cycloalkyl, alkynyl, cyano, haloalkoxy, haloalkyl, hydroxyl, hydroxyalkylene, nitro, NRARB, NRARBalkylene, and (NRARB)carbonyl;
A and B are each independently selected from the group consisting of hydrogen, Br, CI, F, 1, OH, Cr C6alkyl,
Figure imgf000066_0002
alkoxy, and alkoxyalkyl, wherein CrC„alkyl, C3-C8cycloalkyl, alkoxy, and alkoxyalkyl are optionally substituted with at least one substituent selected from the group consisting of OH, N02, CN, Br, CI, F, 1, C,-C6alkyl, and C3-Qcycloalkyl, wherein B is not OH;
RA, and RB are independently selected from the group consisting of hydrogen, alkyl, cycloalkyl, and alkylcarbonyl; or RA and RB taken together with the atom to which they are attached form a 3- 10 membered heterocycle ring optionally having one to three heteroatoms or hetero functionalities selected from the group consisting of -0-, -NH, -N(C, -C6-alkyl)-, -NCO(C C5-alkyl)-, -N(aryl)-, -N(aryl-C, -C6-alkyl-)-, -N(substituted-aryl-CrC6-alkyl-)-, -N(heteroaryl)-, -N(heteroaryl-C , -Cft- alkyl-)-, -N(subsiituted-heteroaryi-Ci-C6-a{kyl-)-, and -S- or S(0)q- , wherein q is 1 or 2 and the 3- 10 membered heterocycle ring is optionally substituted with one or more substituents; R4 and R5 are each independently selected from the group consisting of hydrogen, alkyl, cycloalkyl, alkoxyalkyl, haloalkyl, hydroxyalkylene, and (NRARB)alkylene; and
each R<, is selected from the group consisting of OH, NOi, CN, Br, CI, F, I, C| -C„alkyl, C
Qcycioalkyl,
Figure imgf000067_0001
alkoxy, alkoxyalkyl, alkoxycarbony!,
alkoxycarbonylalkyl, C C6alkynyl, aryl, arylalkyl, C3-C8cycloalkylalkyl, haloalkoxy, haloalkyl, hydroxyalkylene, oxo, heteroaryl, heteroarylalkoxy, heteroaryloxy, heteroarylthio, heteroarylalkylthio, heterocycloalkoxy, C:-C8heterocycloalkylthio, heterocyclooxy, heterocyclothio, NRARB, (NRARB)C Qalkylene, (NRARB)carbonyl, (NRARB)carbonylalkylene, (NRARB)sulfonyl, and
(NRARB)sulfonylalkylene; or
a single isomer, stereoisomer, or enantiomer. or mixture thereof;
optionally as a salt, solvate, chemically protected form or prodrug thereof; and additionally optionally with at least one of a pharmaceutically acceptable diluent, excipient, and carrier.
2. The method of claim 1 , wherein a malignant cell's DNA from the patient exhibits Microsatellite Instability (MSI) in at least one microsatellite.
3. The method of claim 2, wherein the MSI comprises an insertion or deletion in the at least one microsatellite.
4. The method of claim 1 , further comprising analyzing for MSI in at least one microsatellite in a malignant cell from the patient, wherein said analyzing comprises using primers to amplify the at least one microsatellite nucleotide sequence.
5. The method of claim 4, wherein the at least one microsatellite is NR21, NR22, NR24, Bat25, Bat26, Mono27, or a microsatellite in a gene selected from the group consisting of Mrel l, A TM and CTiP.
6. The method of claim 5, wherein the at least one microsatellite is selected from the group consisting of NR21, NR24 and Bat25.
7. The method of claim 3, wherein the MSI comprises an insertion or deletion of 3 or more base pairs in the at least one microsatellite.
8. The method of claim 3, where the at least one microsatellite is in a gene coding a double stranded DNA repair protein, and/or wherein the MSI leads to an inactivation of a DNA repair function, and/or wherein the MSI results in a frameshift mutation in expression of a double stranded DNA repair gene.
9. The method of claim 8, wherein the MSI leads to an inactivation of a DNA repair function, wherein the DNA repair function is governed by a double stranded DNA repair gene selected from the group consisting of Mrel 1, A TM and CTiP.
10. The method of claim 1 , wherein the compound is 5-fluoro-8-(4-fluorophenyl)-9-( l - methyl- l/ - l ,2,4-triazol-5-yl)-8,9-dihydro-2H-pyrido[4,3,2-de]phthalazin-3(7 )-one, or a single isomer, stereoisomer, or enantiomer, or mixture thereof, or a pharmaceutically acceptable salt thereof.
11. A compound of Formul
Figure imgf000068_0001
Formula (I),
wherein:
Y and Z are each independently selected from the group consisting of:
a) an aryl group optionally substituted with 1 , 2, or 3 ;
b) a heteroaryl group optionally substituted with 1 , 2, or 3 and
c) a substituent independently selected from the group consisting of hydrogen, alkenyl, alkoxy, alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, alkyl, alkynyl, arylalkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, hydroxyalkylene, oxo, heterocycloalkyl, heterocycloalkylalkyl, alkylcarbonyl, arylcarbonyl, heteroarylcarbonyl alkylsulfonyl, arylsulfonyl,
heteroarylsulfonyl, (NRARs)alkylene, (NRARB)carbonyl, (NRARB)carbonylalkylene,
(NRARB)sulfonyl, and (NRARB)sulfonylalkylene;
Ri , R:, and R3 are each independently selected from the group consisting of hydrogen, halogen, alkenyl, alkoxy, alkoxycarbonyl, alkyl, cycloalkyl, alkynyl, cyano, haloalkoxy, haloalkyl, hydroxyl, hydroxyalkylene, nitro, NRARB, NRARBalkylene, and (NRARB)carbonyl;
A and B are each independently selected from the group consisting of hydrogen, Br, CI, F, 1, OH, C C6alkyl, C3-C8cycloalkyl, alkoxy, and alkoxyalkyl, wherein Ci -C6alkyl, C3-C8cycloalkyl, alkoxy, and alkoxyalkyl are optionally substituted with at least one substituent selected from the group consisting of OH, N02, CN, Br, CI, F, 1, C C6alkyl, and C3-C3cycloalkyl, wherein B is not OH;
RA. and RB are independently selected from the group consisting of hydrogen, alkyl, cycloalkyl, and alkylcarbonyl; or RA and RB taken together with the atom to which they are attached form a 3- 10 membered heterocycle ring optionally having one to three heteroatoms or hetero functionalities selected from the group consisting of -0-, -NH, -N(Ci-C6-alkyl)-, -NCO(C,-C6-alkyl)-, -N(aryl)-, -N(aryl-Ci-C6-alkyl-)-, -N(substituted-aryl-C C6-alkyl-)-, -N(heteroaryl)-, -N(heteroaryl-C C6- aikyl-K -N(substituted-heteroaryl-Ci-C6-aikyl-)-, and -S- or S(0)q- , wherein q is 1 or 2 and the 3- 10 membered heterocycle ring is optionally substituted with one or more substituents;
R4 and R5 are each independently selected from the group consisting of hydrogen, alky , cycloalkyl, alkoxyalkyl, haloalkyl, hydroxyalkylene, and (NRARB)alkylene; and
each Re is selected from the group consisting of OH, N02, CN, Br, CI, F, I, Ci -C6alkyl, C3- Cgcycloalkyl, C2-C8heterocycloalkyl, C2-C6alkenyl, alkoxy, alkoxyalkyl, alkoxycarbonyl,
alkoxycarbonylaikyl, C2-C6alkynyl, aryl, arylalkyl, C3-C8cycloalkylalkyl, haloalkoxy, haloalkyl, hydroxyalkylene, oxo, heteroaryi, heteroarylalkoxy, heteroaryloxy, heteroarylthio, heteroarylalkylthio, heterocycloalkoxy, C2-Cgheterocycloalkylthio, heterocyclooxy, heterocyclothio, NRARB, (NRARB)Cr C6alkylene, (NRARB)carbonyl, (NRARB)carbonylalkylene, (NRARB)sulfonyl, and
( NRARB)sulfonylalkylene; or
a single isomer, stereoisomer, or enantiomer, or mixture thereof;
optionally as a salt, solvate, chemically protected form or prodrug thereof; and additionally optionally with at least one of a pharmaceutically acceptable diluent, excipient, and carrier,
for use in the treatment of MDS or AML in a patient with MDS or AML.
12. The compound of claim 1 1 , wherein a malignant cell from the patient exhibits MSI in a microsateilite.
13. The compound of claim 12, wherein the malignant cell exhibits MSI in at least two microsatellites.
14. A method for identifying an MDS or AML patient likely to respond to PARP inhibitor therapy for treatment of MDS or AML comprising:
analyzing for MSI in at least one microsateilite in a malignant cell from the MDS or AML patient;
wherein the MSI comprises a deletion or insertion of three or more base pairs in the at least one microsateilite; and
wherein the presence of MSI in the at least one microsateilite identifies the patient as likely to respond to PARP inhibitor therapy.
15. The method of claim 14, wherein the PARP inhibitor is 5-fluoro-8-(4-fluorophenyl)- 9-( 1 -methyl- 1 H- 1 ,2,4-triazol-5-y l)-8,9-dihy dro-2H-pyrido[4,3 ,2-de]phthalazin-3( 7H)-one, or a single isomer, stereoisomer, or enantiomer, or mixture thereof, or a pharmaceutically acceptable salt thereof.
16. The method of claim 14, wherein said analyzing comprises using primers to amplify the at least one microsatellite nucleotide sequence.
17. The method of claim 14, wherein the at least one microsatellite is NR2L NR22, NR24, Bat25, Bat26, Mono27 or a microsatellite in a gene selected from the group consisting of Mrel l, A TM and CTiP.
18. The method of claim 17, wherein the at least one microsatellite selected from the group consisting of NR21, NR24 and Bat25.
19. The method of claim 14, wherein the MSI leads to inactivation of a DNA repair function.
20. The method of claim 19, wherein the DNA repair function is that of a double stranded DNA repair gene expression product selected from the group of double stranded DNA repair genes consisting of Mrel l, A TM Άπά CTiP.
21. The method of claim 14, further comprising performing a cytogenetic analysis for chromosomal abnormalities in the malignant cell.
22. The method of claim 14, wherein at least three microsatellitcs are analyzed for MSI.
23. The method of claim 14, wherein the at least one microsatellite is in a double stranded DNA repair gene.
PCT/US2011/039045 2011-06-03 2011-06-03 Use of dihydropyridophthalazinone inhibitors of poly (adp-ribose) polymerase (parp) in the treatment of myelodysplastic syndrome (mds) and acute myeloid leukaemia (aml) WO2012166151A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
PCT/US2011/039045 WO2012166151A1 (en) 2011-06-03 2011-06-03 Use of dihydropyridophthalazinone inhibitors of poly (adp-ribose) polymerase (parp) in the treatment of myelodysplastic syndrome (mds) and acute myeloid leukaemia (aml)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/US2011/039045 WO2012166151A1 (en) 2011-06-03 2011-06-03 Use of dihydropyridophthalazinone inhibitors of poly (adp-ribose) polymerase (parp) in the treatment of myelodysplastic syndrome (mds) and acute myeloid leukaemia (aml)

Publications (1)

Publication Number Publication Date
WO2012166151A1 true WO2012166151A1 (en) 2012-12-06

Family

ID=44627450

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/039045 WO2012166151A1 (en) 2011-06-03 2011-06-03 Use of dihydropyridophthalazinone inhibitors of poly (adp-ribose) polymerase (parp) in the treatment of myelodysplastic syndrome (mds) and acute myeloid leukaemia (aml)

Country Status (1)

Country Link
WO (1) WO2012166151A1 (en)

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8765945B2 (en) 2010-02-08 2014-07-01 Biomarin Pharmaceutical Inc. Processes of synthesizing dihydropyridophthalazinone derivatives
EP2836606A1 (en) * 2012-04-10 2015-02-18 Vib Vzw Novel markers for detecting microsatellite instability in cancer and determining synthetic lethality with inhibition of the dna base excision repair pathway
WO2015040378A1 (en) * 2013-09-17 2015-03-26 King's College London Biomarkers correlated to parp inhibitor treatment success in aml patients
WO2017077326A1 (en) * 2015-11-05 2017-05-11 King's College London Combination of an inhibitor of parp with an inhibitor of gsk-3 or dot1l
US9820985B2 (en) 2008-08-06 2017-11-21 Medivation Technologies Llc Dihydropyridophthalazinone inhibitors of poly(ADP-ribose)polymerase (PARP)
US9957267B2 (en) 2015-07-01 2018-05-01 Crinetics Pharmaceuticals, Inc. Somatostatin modulators and uses thereof
US10189837B2 (en) 2010-10-21 2019-01-29 Medivation Technologies Llc Crystalline (8S,9R)-5-fluoro-8-(4-fluorophenyl)-9-(1-methyl-1H-1,2,4-triazol-5-yl)-8,9-dihydro-2H-pyrido[4,3,2-de]phthalazin-3(7H)-one tosylate salt
US10294529B2 (en) 2012-04-10 2019-05-21 Life Sciences Research Partners Vzw Microsatellite instability markers in detection of cancer
US10493078B2 (en) 2010-02-03 2019-12-03 Medivation Technologies Llc Dihydropyridophthalazinone inhibitors of poly(ADP-ribose)polymerase (PARP) for use in treatment of diseases associated with a PTEN deficiency
US10633707B2 (en) 2012-04-10 2020-04-28 Vib Vzw Markers for detecting microsatellite instability in cancer and determining synthetic lethality with inhibition of the DNA base excision repair pathway
WO2021092299A1 (en) * 2019-11-08 2021-05-14 Life Technologies Corporation Systems and assays for assessing microsatellite instability
US11028068B2 (en) 2017-07-25 2021-06-08 Crinetics Pharmaceuticals, Inc. Somatostatin modulators and uses thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1340819A1 (en) * 2002-02-28 2003-09-03 Institut National De La Sante Et De La Recherche Medicale (Inserm) Microsatellite markers
WO2004105700A2 (en) * 2003-05-28 2004-12-09 Guildford Pharmaceuticals, Inc. Compounds, methods and pharmaceutical compositions for inhibiting parp
WO2010017055A2 (en) * 2008-08-06 2010-02-11 Lead Therapeutics, Inc. Dihydropyridophthalazinone inhibitors of poly(adp-ribose)polymerase (parp)

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1340819A1 (en) * 2002-02-28 2003-09-03 Institut National De La Sante Et De La Recherche Medicale (Inserm) Microsatellite markers
WO2004105700A2 (en) * 2003-05-28 2004-12-09 Guildford Pharmaceuticals, Inc. Compounds, methods and pharmaceutical compositions for inhibiting parp
WO2010017055A2 (en) * 2008-08-06 2010-02-11 Lead Therapeutics, Inc. Dihydropyridophthalazinone inhibitors of poly(adp-ribose)polymerase (parp)

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
GAYMES TERRY J ET AL: "Inhibitors of poly ADP-ribose polymerase (PARP) induce apoptosis of myeloid leukemic cells: potential for therapy of myeloid leukemia and myelodysplastic syndromes", HAEMATOLOGICA, FONDAZIONE FERRATA STORTI, ROME, IT, vol. 94, no. 5, 1 May 2009 (2009-05-01), pages 638 - 646, XP008147848, ISSN: 0390-6078, DOI: 10.3324/HAEMATOL.2008.001933 *
GAYMES TERRY J ET AL: "Microsatellite Instability (MSI) In High Risk Myelodysplastic Syndrome (MDS) and Acute Myeloid Leukaemia (AML) Cells Promotes Frameshift Mutations In DNA Repair Genes: Indications for PARP Inhibitor Therapy", BLOOD; 52ND ANNUAL MEETING OF THE AMERICAN-SOCIETY-OF-HEMATOLOGY (ASH), AMERICAN SOCIETY OF HEMATOLOGY, US; ORLANDO, FL, USA, vol. 116, no. 21, 1 November 2010 (2010-11-01), pages 513, XP008147850, ISSN: 0006-4971 *
HAM MARIA FRANCISCA ET AL: "Impairment of double-strand breaks repair and aberrant splicing of ATM and MRE11 in leukemia-lymphoma cell lines with microsatellite instability", CANCER SCIENCE, vol. 97, no. 3, March 2006 (2006-03-01), pages 226 - 234, XP002668278, ISSN: 1347-9032 *

Cited By (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10780088B2 (en) 2008-08-06 2020-09-22 Medivation Technologies Llc Dihydropyridophthalazinone inhibitors of poly(ADP-ribose)polymerase (PARP)
US10543209B2 (en) 2008-08-06 2020-01-28 Medivation Technologies Llc Dihydropyridophthalazinone inhibitors of poly(ADP-ribose)polymerase (PARP)
US9820985B2 (en) 2008-08-06 2017-11-21 Medivation Technologies Llc Dihydropyridophthalazinone inhibitors of poly(ADP-ribose)polymerase (PARP)
US11364241B2 (en) 2008-08-06 2022-06-21 Medivation Technologies Llc Dihydropyridophthalazinone inhibitors of poly(ADP-ribose)polymerase (PARP)
US10493078B2 (en) 2010-02-03 2019-12-03 Medivation Technologies Llc Dihydropyridophthalazinone inhibitors of poly(ADP-ribose)polymerase (PARP) for use in treatment of diseases associated with a PTEN deficiency
US8765945B2 (en) 2010-02-08 2014-07-01 Biomarin Pharmaceutical Inc. Processes of synthesizing dihydropyridophthalazinone derivatives
US9926303B2 (en) 2010-02-08 2018-03-27 Medivation Technologies Llc Processes of synthesizing dihydropyridophthalazinone derivatives
US10189837B2 (en) 2010-10-21 2019-01-29 Medivation Technologies Llc Crystalline (8S,9R)-5-fluoro-8-(4-fluorophenyl)-9-(1-methyl-1H-1,2,4-triazol-5-yl)-8,9-dihydro-2H-pyrido[4,3,2-de]phthalazin-3(7H)-one tosylate salt
US10294529B2 (en) 2012-04-10 2019-05-21 Life Sciences Research Partners Vzw Microsatellite instability markers in detection of cancer
EP2836606A1 (en) * 2012-04-10 2015-02-18 Vib Vzw Novel markers for detecting microsatellite instability in cancer and determining synthetic lethality with inhibition of the dna base excision repair pathway
EP2836606B1 (en) * 2012-04-10 2021-12-29 Vib Vzw Novel markers for detecting microsatellite instability in cancer and determining synthetic lethality with inhibition of the dna base excision repair pathway
US10633707B2 (en) 2012-04-10 2020-04-28 Vib Vzw Markers for detecting microsatellite instability in cancer and determining synthetic lethality with inhibition of the DNA base excision repair pathway
WO2015040378A1 (en) * 2013-09-17 2015-03-26 King's College London Biomarkers correlated to parp inhibitor treatment success in aml patients
US9957267B2 (en) 2015-07-01 2018-05-01 Crinetics Pharmaceuticals, Inc. Somatostatin modulators and uses thereof
US10799501B2 (en) 2015-11-05 2020-10-13 King's College Hospital Nhs Foundation Trust Combination of an inhibitor of PARP with an inhibitor of GSK-3 or DOT1L
WO2017077326A1 (en) * 2015-11-05 2017-05-11 King's College London Combination of an inhibitor of parp with an inhibitor of gsk-3 or dot1l
US11028068B2 (en) 2017-07-25 2021-06-08 Crinetics Pharmaceuticals, Inc. Somatostatin modulators and uses thereof
WO2021092299A1 (en) * 2019-11-08 2021-05-14 Life Technologies Corporation Systems and assays for assessing microsatellite instability

Similar Documents

Publication Publication Date Title
US10780088B2 (en) Dihydropyridophthalazinone inhibitors of poly(ADP-ribose)polymerase (PARP)
WO2012166151A1 (en) Use of dihydropyridophthalazinone inhibitors of poly (adp-ribose) polymerase (parp) in the treatment of myelodysplastic syndrome (mds) and acute myeloid leukaemia (aml)
US10493078B2 (en) Dihydropyridophthalazinone inhibitors of poly(ADP-ribose)polymerase (PARP) for use in treatment of diseases associated with a PTEN deficiency
WO2011130661A1 (en) Methods of using dihydropyridophthalazinone inhibitors of poly (adp-ribose)polymerase (parp)
US20130053365A1 (en) Dihydropyridophthalazinone inhibitors of poly(adp-ribose)polymerase (parp)
US20110190266A1 (en) 5,6,6a,7,8,9-HEXAHYDRO-2H-PYRIDOPHTHALAZINONE INHIBITORS OF POLY(ADP-RIBOSE)POLYMERASE (PARP)
TW201249840A (en) Methods of using dihydropyridophthalazinone inhibitors of poly (ADP-ribose) polymerase (PARP) in the treatment of myelodysplastic syndrome (MDS) and acute myeloid leukaemia (AML)

Legal Events

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

Ref document number: 11727582

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 11727582

Country of ref document: EP

Kind code of ref document: A1