WO2013028495A1 - Dihydropyridophthalazinone inhibitors of poly (adp-ribose) polymerase (parp) for the treatment of multiple myeloma - Google Patents

Dihydropyridophthalazinone inhibitors of poly (adp-ribose) polymerase (parp) for the treatment of multiple myeloma Download PDF

Info

Publication number
WO2013028495A1
WO2013028495A1 PCT/US2012/051297 US2012051297W WO2013028495A1 WO 2013028495 A1 WO2013028495 A1 WO 2013028495A1 US 2012051297 W US2012051297 W US 2012051297W WO 2013028495 A1 WO2013028495 A1 WO 2013028495A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
pharmaceutical composition
methyl
peaks
alkyl
Prior art date
Application number
PCT/US2012/051297
Other languages
French (fr)
Inventor
Ying Feng
Andres A. GUTIERREZ
Yuqiao Shen
Evelyn W. WANG
Augustus O. Okhamafe
Christopher P. Price
Tianwei CHOU
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.
Publication of WO2013028495A1 publication Critical patent/WO2013028495A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/2027Organic macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyvinyl pyrrolidone, poly(meth)acrylates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • A61K9/2054Cellulose; Cellulose derivatives, e.g. hydroxypropyl methylcellulose
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/06Peri-condensed systems

Definitions

  • compositions comprising (85 , ,9R)-5-fluoro-8-(4-fluorophenyl)-9-( 1 -methyl- 1 ,2,4-triazol-5-yl)-8,9-dihydro-2 /-pyrido[4,3,2-de]phthalazin-3(7H)-one tosylate salt, unit dosage forms thereof, and methods of their preparation.
  • PARP poly(ADP-ribose)polymerases
  • 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.
  • the mammalian enzyme PARP- 1 is a multidomain protein. PARP has been implicated in the signal ing of DNA damage through its ability to recognize and rapidly bind to DNA single or double strand breaks (D'Amours, et al., Biochem.
  • PARP has an essential role in facilitating DNA repair, control l ing RNA transcription, mediating cell death, and regulating immune response. Studies on the mechanism by which PARP- 1 modulates DNA repair and other processes identifies its importance in the formation of poly(ADP-ribose) chains within the cellular nucleus.
  • the DNA-bound, activated PARP- 1 utilizes NAD+ to synthesize poly(ADP-ribose) on a variety of nuclear target proteins, including topoisomerases, histones and PARP itself.
  • PARP inhibitors are used for the treatment of cancer which is deficient in Homologous Recombination (HR) dependent DNA double strand break (DSB) repair activity.
  • HR Homologous Recombination
  • DSB DNA double strand break
  • the HR dependent DNA DSB repair pathway repairs double-strand breaks (DSBs) in DNA via homologous mechanisms to reform a continuous DNA helix.
  • Compound A The high potency of (8S,97?)-5-Fluoro-8-(4-fluorophenyl)-9-( l -methyl- 1 /- 1 , 2,4-triazol-5- yl)-8,9-dihydro-2H-pyrido[4,3,2-deJphthalazin-3(7H)-one (referred to herein as "Compound A") as a PARP inhibitor as well as its physiochemical properties can pose difficulties in formulating drug delivery forms of Compound A, specifically, forms which can be manufactured on a large scale in which
  • Compound A is stable, for example, under ambient conditions, and from which a consistent amount of Compound A wil l be delivered to the patient. Improved pharmaceutical compositions ofCompound A and unit dosage forms thereof are desired.
  • One aspect is a method of treating multiple myeloma with a Compound according to
  • Y and Z are each independently selected from the group consisting of:
  • a heteroaryl group optionally substituted with 1 , 2, or 3 R 6 ; c) a substituent independently selected from the group consisting of hydrogen, alkenyl, alkoxy, alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, alkyl, alkynyl, arylalkyl, cycloalkyi, cycloalkylalkyl, haloalkyl, hydroxyalkylene, oxo, heterocycloalkyl, heterocycloalkylalkyl, alkylcarbonyl, arylcarbonyl.
  • R[, R 2 , and R 3 are each independently selected from the group consisting of hydrogen, halogen, alkenyl, alkoxy, alkoxycarbonyl, alkyl, cycloalkyi, alkynyl, cyano, haloalkoxy, haloalkyl, hydroxyl, hydroxyalkylene, nitro, -NR A R B , -(C i-C 6 -alkylene)-NR A R B , and -C(0)(NR A R B );
  • a and B are each independently selected from hydrogen, Br, CI, F, I, OH, Q-C ⁇ alkyl, C 3 -C 8 cycloaIkyl, alkoxy, alkoxyalkyl wherein C
  • R A and R B are independently selected from the group consisting of hydrogen, alkyl, cycloalkyi, and
  • 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 functional ities selected from the group consisting of -0-, -NH-, -N(C C 6 -alkyl)-, -NC(0)(Ci-C 6 -alkyl)-, -N(aryl)-, -N(aryl-C
  • R.i and R 5 are each independently selected from the group consisting of hydrogen, alkyl, cycloalkyi, alkoxyalkyl, haloalkyl, hydroxyalkylene, and -(C
  • each R ⁇ i is independently selected from OH, N0 2 , CN, Br, CI, F, I, C r C 5 alkyl, Cs-Qeycloalkyl,
  • Another aspect is a method of treating multiple myeloma with a pharmaceutical composition comprising a compound of Formula (I) or a single isomer, stereoisomer, or enantiomer or mixture thereof, optionally as a pharmaceutically acceptable salt, pharmaceutically acceptable solvate, or pharmaceutically acceptable prodrug thereof and a pharmaceutically acceptable carrier, excipient, binder or diluent thereof.
  • a pharmaceutical combination which comprises a therapeutical ty-effective amount of a compound of Formula (I) or a pharmaceutical composition thereof and a proteasome inhibitor.
  • Another aspect is the use of a compound of Formula (I) for the treatment of multiple myeloma.
  • Another aspect is the use of a pharmaceutical combination comprising a therapeutical ly- effective amount of a compound of Formula (I) or a pharmaceutical composition thereof and a proteasome inhibitor for the treatment of multiple myeloma in a subject in need thereof.
  • Another aspect is an article of manufacture, comprising packaging material, a compound of Formula (I), optionally a proteasome inhibitor, and a label, wherein the compound is for treatment, prevention or amelioration of multiple myeloma, wherein the compound is packaged within the packaging material, and wherein the label indicates that the compound, or pharmaceutically acceptable salt, pharmaceutically acceptable N-oxide, pharmaceutically active metabolite, pharmaceutically acceptable prodrug, or pharmaceutically acceptable solvate thereof, or a pharmaceutical composition comprising such a compound is used for treatment, prevention or amelioration multiple myeloma.
  • composition comprising
  • Compound A tosylate salt in solid form and one or more excipients are included in solid form and one or more excipients.
  • a pharmaceutical composition comprising Compound A tosylate salt and silicified microcrystalline cellulose, wherein the pharmaceutical composition is prepared using crystalline Compound A tosylate salt.
  • a unit dosage form comprising pharmaceutical composition, the pharmaceutical composition comprising Compound A tosylate salt in solid form and one or more pharmaceutically acceptable excipients, wherein the weight/weight percent of Compound A tosylate salt in the pharmaceutical composition is between about 0.01 % to about 1.000 %.
  • compositions comprising Compound A tosylate salt and one or more excipients.
  • compositions produced by the methods described herein for producing a pharmaceutical composition.
  • kits for treating a cancer or symptom thereof comprising administering to a cancer patient an effective amount of a pharmaceutical composition comprising Compound A tosylate salt as described herein.
  • Fig.l provides an overlay of five examples of powder X-ray powder diffraction (XRPD) patterns of crystall ine (8S,9i?)-5-fluoro-8-(4-fIuorophenyl)-9-( l -methyl- 2,4-triazol-5-yl)-8,9- dihydro-2 -pyrido[4,3 ,2-de]phthaIazin-3(7/7)-one tosylate salt.
  • Fig.2a provides a differential scanning calorimetric (DSC) graph of a crystalline (85,9/?)-
  • Fig.2b provides a thermogravimetric analysis (TGA) graph of a crystalline (85,9?)-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-
  • Fig.3a provides a DSC graph of a crystalline (85,9?)-5-fluoro-8-(4-fluorophenyl)-9-(l- methyl-l /-l,2,4-triazol-5-yl)-8,9-dihydro-2//-pyrido[4,3,2-de]phthalazin-3(7/)-one tosylate salt polymorph, 5, from DCM-ACN preparation.
  • Fig.3b provides a TGA graph of a crystalline (85,9/?)-5-fluoro-8-(4-fluorophenyl)-9-( 1- methyl-l//-l,2,4-triazol-5-yl)-8,9-dihydro-2/-pyrido[4,3,2-de]phthalazin-3(7//)-one tosylate salt polymorph, 5, from DCM-ACN preparation.
  • Fig.4a provides a DSC graph of a crystalline (85,9/?)-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 tosylate salt polymorph, 6, from acetone preparation.
  • Fig.4b provides a TGA graph of a crystalline (85,9/?)-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 tosylate salt polymorph, 6, from acetone preparation.
  • Fig.5a provides a DSC graph of a crystalline (85,9?)-5-fluoro-8-(4-fluorophenyl)-9-(l- methyl-l/f-l,2,4-triazol-5-yl)-8,9-dihydro-2 -pyrido[4,3,2-de]phthalazin-3(7 /)-one tosylate salt polymorph, 7, from THF preparation.
  • Fig.5b provides a TGA graph of a crystalline (85,9/?)-5-fluoro-8-(4-fluorophenyl)-9-(l- methyl-li-l,2,4-triazol-5-yl)-8,9-dihydro-2i : /-pyrido[4,3,2-de]phthalazin-3(7 f)-one tosylate salt polymorph, 7, from THF preparation.
  • Fig.6 provides an X-ray powder diffraction (XRPD) spectrogram of a crystalline
  • Fig.7a provides a DSC graph of a crystalline (85,9?)-5-fIuoro-8-(4-fluorophenyl)-9-( 1 - methyl-lJ-l,2,4-triazol-5-yI)-8,9-dihydro-2//-pyrido[4,3,2-de]phthalazin-3(7H)-one tosylate salt polymorph.
  • Fig.7b provides a TGA graph of a crystalline (85,9?)-5-fIuoro-8-(4-fluorophenyl)-9-(l- methyl- 1 H- 1 ,2,4-triazol-5-yl)-8,9-dihydro-2H-pyrido[4,3,2-de]phthalazin-3(7H)-one tosylate salt polymorph.
  • Fig.8 depicts an X-ray powder diffraction (XRPD) spectrogram of a crystalline (85,9/?)-
  • Fig. 9 provides a dynamic vapor sorption isotherm plot of a crystalline (8S,9R)-5-fluoro-
  • Figs. 10a and 10b depict dose response curves and IC 50 in H929 Myeloma Cells at 5, 7, and 10 days.
  • Fig. 11 depicts dose response curves and 1C 50 in MM. I S Myeloma Cells at 7 and 10 days.
  • Fig. 12 provides a solid state 13 C NMR spectrum for a (8S,9.ft)-5-fIuoro-8-(4- fluorophenyl)-9-( l -methyl- lH- l ,2,4-triazol-5-yl)-8,9-dihydro-2 /-pyrido[4,3,2-de]phthalazin-3(7fl)-one tosylate salt polymorph.
  • the subject is a human.
  • compounds provided herein have the structure of Formu la (I) and chemically protected forms and pharmaceutically acceptable salts, solvates, esters, acids and prodrugs thereof.
  • compounds having the structure of Formula (I) that are inhibitors of the enzyme poly(ADP-ribose)polymerase (PARP).
  • PARP poly(ADP-ribose)polymerase
  • R T , R 2 , Rj are independently selected from a group consisting of hydrogen, alky I, and halogen; R» is hydrogen and R 5 is selected from the group consisting hydrogen, a Iky I, cycloalkyl, alkoxyalkyl, haloalkyl, hydroxyalkylene, and -(Cj-C6-alkylene)(NR A R B ); RA, and R B re independently selected from the group consisting of hydrogen, alky i, cycloalkyl, and alkylcarbonyl; or R A and R B taken together with the atom to which they are attached form a 3- 1 0 membered heterocycle ring optionally having one to three heteroatoms or hetero functional ities selected from the group consisting of -0-, -NH, -N(C ,-C 6 -alkylK -NC(0)(C,-C 6 -alky
  • a substituent independently selected from the group consisting of hydrogen, alkenyl, alkoxy, alkoxyalkyi, alkoxycarbonyl, alkoxycarbonylalkyl, alkyl, alkynyl, arylalkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, hydroxyalkylene, oxo, heterocycloalkyl, heterocy c I oa I ky I a I ky I , alkylcarbonyl, arylcarbonyl, heteroarylcarbonyl alkyisulfonyl, arylsulfonyl, heteroarylsulfonyl, -(C r C 6 -alkylene)(NR A R B ), -C(0)( R A R B ), -(C,-C 6 -aIkylene)C(0)(NR A R B ), -S(0) 2 (NR A R B ), and -(Ci-C
  • each R ⁇ is selected from OH, N0 2 , CN, Br, CI, F, I, C ,-C 6 alkyl, C 3 ⁇ C 8 cycloalkyl, C 2 -C 8 heterocyc loalkyl;
  • R b R 2 , and R 3 are independently selected from a group consisting of hydrogen, alkyl, and halogen; R4 and R 5 are hydrogen; R A and 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-O-, -NH, -N(C rQ-alkyl)-, - NC(0)(C r C 6 -alkyl)-, -N(aryl)-, -N(aryl-C,-C 5 -alkyl-)-, -N(substituted-aryl-C C 6 -alkyl-)-, -N(heteroaryl)
  • R R 2 , R restroom R. and R 5 are hydrogen; R A and 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-, -NH, -N(C f -C 6 - alkyl)-, -NC(0)(C ,-C 6 -alkyl)-, -N(aryl)-, -N(aryl- C C 6 -alkyl-)-, -N(substituted-aryl- C r C 6 -alkyl-)-, - N(heteroaryl)-, -N(heteroaryI- Ci-C 6 -
  • R 3 ⁇ 4 R3 are each independently selected from a group consisting of hydrogen, alkyl, and halogen; 4 is hydrogen and R 5 is selected from the group consisting hydrogen, alkyl, cycloalkyi, alkoxyalkyl, haloalkyl, hydroxyalkylene, and -(C
  • , R 2 , R3 are each independently selected from a group consisting of hydrogen, alkyl, and halogen; R 4 and R 5 are hydrogen.
  • R R 2 , R 3 , R4 are each hydrogen and R 3 is alkyl.
  • [0048J in one embodiment is a compound of Formula (I) wherein Rj, R 2 , and Rj are each hydrogen.
  • R 5 is hydrogen or an alkyl group.
  • R 5 is hydrogen.
  • R 5 is C r C 6 alkyl.
  • R 5 is CH 3 .
  • R 5 is CH 2 CH 3 .
  • R4 is hydrogen or an alkyl group.
  • R4 is hydrogen.
  • the Compound of Formula ( 1) is that where R ? is selected from the group consisting of hydrogen, halogen, alkenyl, alkoxy, alkoxycarbonyl, alkyl, cycloalkyi, alkynyl, cyano, haloalkoxy, haloalkyl, hydroxyl, hydroxyalkylene, nitro, -N R A R B , -(Ci-C6alkylene)-NR A R B , and -C(0)(NR A R B ).
  • R 3 is hydrogen.
  • is selected from the group consisting of hydrogen, halogen, alkenyl, alkoxy, alkoxycarbonyl, alkyl, cycloalkyi, alkynyl, cyano, haloalkoxy, haloalkyl, hydroxyl, hydroxyalkylene, nitro, -NR A R B , -(C
  • Rj is hydrogen
  • Embodiment A Another emdodiment is a method of treating multiple myeloma with a
  • Y is an aryl or heteroaryl group optional ly substituted with at least one R(,;
  • 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 5 alkyl, C ⁇ -Cscycloalkyl, alkoxy, and alkoxyalkyi wherein C
  • Re is selected from OH, NO2, CN, Br, CI, F, I, C r C 6 alkyl, C 3 -Cgcycloalkyl, Ci-Cgheterocyc loalky 1;
  • R 2 is selected from hydrogen, Br, CI, I, or F;
  • R A and R are independently selected from the group consisting of hydrogen, C r C 6 alkyl, C 3 - Cgcycloalkyl, 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-, -N H, -N(C C 6 alkyl)-, -NC(0)(C i -C 6 a I ky I )-, -NC(0)(C 3 -Cgcycloalkyl)-, -N(aryl)-, -N(aryl-C r C 6 alkyl-)-, -N(substituted-aryl-C,-C 6 alkyl-)-, - N(heteroaryl)-, -N(heteroaryl-C r C 6 alkyl-)-, -N(substituted-he
  • a substituent independently selected from the group consisting of hydrogen, alkoxyalkyl, alkoxycarbonylalkyl, alkyl, arylalkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, oxo, heterocycloalkyl, heterocycloalkylalkyl, alkylcarbonyl, arylearbonyl, heteroarylcarbonyl, -(C,-C 6 aikylene)-(NR A R B ), -C(0)(NR A R B ), and -(C r C 6 alkylene)-C(0)(NR A R B ).
  • the Compound of Formula (I) or (II) is that where R 2 is selected from the group consisting of hydrogen, halogen, alkenyl, alkoxy, alkoxycarbonyl, alkyl, cycloalkyl, alkynyl, cyano, ha!oalkoxy, haloalkyl, hydroxy], hydroxyalkylene, nitro, -NR A R B , -(C r C 6 alkylene)-NR A R B , and -C(OXNR A R B ).
  • R 2 is a halogen selected from F, CI, Br, and I.
  • R 2 is fluorine.
  • R 2 is hydrogen.
  • Z is an aryl group optionally substituted with 1 , 2, or 3 R ⁇ ; wherein each R > is selected from OH, NO?, CN, Br, CI, F, I, Q- Qalkyl, Q-Cscycloalkyl, C 2 -C 8 heterocycloalkyl; C 2 -C coordinationkenyl, alkoxy, alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C 2 -C 6 alkynyl, aryl, arylalkyl, C 3 -C 8 cycloalkylalkyl, haloalkoxy, haloalkyl, hydroxyalkylene, oxo, heteroaryl, heteroarylalkoxy, heteroaryloxy, heteroarylthio, heteroarylalkylthio, heterocycloalkoxy, C 2 -C 8 heteiOcycloal
  • C 6 alkyleneMN R A RB -C(0)(NR A R B ), -(C , -Qalky lene)-C(0)( R A R B ), -S(0) 2 (NR A R B ), and -(C r C 6 alkylene)-S(0)2(NR A RB).
  • Z is an optionally substituted phenyl group.
  • Z is a phenyl group.
  • the phenyl group is optionally substituted with at least one R ⁇ .
  • Re is -(Q -Qalky Iene)-(NR A R B ).
  • R trash is -CH 2 (NR A R B ).
  • R A is -CH 2 (NR A R B ) wherein -NR A R B is azetidine, pyrrolidine, piperidine or morpholine.
  • R A is H or alkyl. In another embodiment, R A is C r C 6 alkyl. In yet another embodiment, R A is -CH : ,. In another embodiment, R B is H or alkyl. In one embodiment, R B is C r C 6 alkyl. In yet another embodiment, R B is -CH 3 . In a further embodiment, R 6 is -CH 2 NHCH 3 . In yet a further embod iment. Re is -CH2NCH 3 CH 3 . In one embodiment, R 6 is -(CO)heterocycIoalkyl(CO)alkyl.
  • R réelle is -(CO)heterocyc loa 1 ky l(CO)alky 1 wherein the heterocyeloalkyl group has at least one heteroatom selected from O, N, and S. In another embodiment, the heterocyeloalkyl group has two N atoms. In a further embodiment, R 6 is
  • alkyl is selected from methyl, ethyl, n-propyl, iso-propyl, cyclopropyl, n-butyl, iso-butyl, and t-butyl.
  • the alkyl group is cyclopropyl.
  • the alkyl group is iso-propyl. In one embodiment, R 6 is . In another
  • R is
  • the heteroaryl group is selected from pyridine, pyrimidine, pyrazine, pyrazole, oxazole, thiazole, isoxazole, isothiazole, 1 ,3,4 -oxadiazole, pyridazine, 1 ,3,5-trazine, 1 ,2,4-triazine, quinoxaline, benzimidazole, benzotriazole, purine, I / -[ l ,2,3]triazolo[4,5- i/]pyrimidine, triazole, imidazole, thiophene, furan, isobenzofuran, pyrrole, indolizine, isoindole, indole, indazole, isoquinoline, quinoline, phthalazine, naphthyridine,
  • R 3 are independently selected from a group consisting of hydrogen, alkyl, and halogen;
  • R. t is hydrogen and R 5 is selected from the group consisting hydrogen, alkyl, cycloalkyi, alkoxyalkyi, haloalkyi, hydroxyalkylene, and -(Ci-C6-alkylene)(NR A R B );
  • R A and R B are independently selected from the group consisting of hydrogen, alkyl, cycloalkyi, 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 -NH, -N(C,-C 6 -alkyl)-, -NC(0)(C,-C 6 -alkyl)-, -N(ary
  • 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, and I.
  • Rs is F
  • the phenyl group is substituted with at least one R6 selected from -(C r C 6 -alkylene)(NR A R B ), -C(0)(NR A R B )carbonyl, -(C r C 6 -alkylene)C(0)(NR A R B ), -S(0) 2 (NR A R B ), and -(C,-C 6 - alkyIene)S(0) 2 (NR A R B ).
  • R « is -(C r C 6 -alkylene)(NR A R B ).
  • C C 6 alkyl is selected from methylene, ethylene, n-propylene, iso-propylene, n-butylene, iso-butylene, and tert-butylene.
  • CrQalkyl is methylene.
  • R A and R B are each independently hydrogen, Ci-C ( ,alkyl, or C3-C 8 cycloalkyl.
  • CrQalkyl is selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl.
  • Q -Qalkyl is methyl.
  • Q-Qalkyl is ethyl.
  • Q- Qcycloaikyl is cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl.
  • Q- Qcyeloalkyl is cyclopropyl.
  • R ⁇ > is hydroxyalkylene. In one embodiment hydroxyalkylene is selected from -CH 2 OH, -CH 2 CH 2 OH, -CH 2 CH,CH 2 OH, -CH(OH)CH 3 ,
  • 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
  • the hetero functionality is -N(C r C 6 alkyl).
  • Q-Qalkyl is selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl.
  • Q-Qalkyl 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 Q- Qalkyl selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl.
  • Q-Qalkyl 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 Q-Qalky selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-buty l.
  • Q-Qalkyl is methyl.
  • Z 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, and I.
  • R,- is F.
  • R render is CI.
  • the phenyl group is substituted with at least one R harm selected from -(Q-Q-alkylene)(NR A R B ), -C(0)(NR A R B ), -(Q - C 6 -alkylene)C(0)(NR A R B ), -S(0) 2 (NR A R B ), and -(Ci-C 6 -alkylene)S(0) 2 (NR A R B ).
  • R 6 is -(Q-Q-aIkylene)(>JR A R B ).
  • C C (J alkyi is selected from methylene, ethylene, n-propylene, iso-propylene, n-butylene, iso-butylene, and tert-butylene.
  • C r C 6 alkyl is methylene.
  • R A and R B are each independently hydrogen, Q-Qalkyl, or Q-Qcycloalkyl.
  • Q-Qalkyl is selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl.
  • -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, and -N(CrQalkyl).
  • the hetero functionality is -N(C Qalkyl).
  • C r C fatiguealkyI is selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl.
  • Q-Qalkyl is methyl.
  • Z is a heteroaryl group optional ly 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, benzoth iophene, benzoimidazole. benzofuran, pyridazine, 1 ,3,5-triazine, thienothiophene, quinoxaline, quinoline, and isoquinoline.
  • the heteroaryl group is imidazole.
  • imidazole is substituted with Q-Qalkyl selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso- butyl, and tert-butyl .
  • Q-Qalkyl 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 Q- Qalkyl selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl.
  • Q- Qalkyl selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl.
  • C Qal yl is methyl.
  • R 2 is hydrogen.
  • R 2 is selected from F, CI, Br, and I.
  • R 2 is F.
  • A is a compound of Formula (I) or (II) wherein A is hydrogen.
  • A is Q-Qalkyl.
  • A is selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, tert-butyl, n-pentyi, and n-hexyl.
  • methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, tert-butyl, n-pentyl, and n-hexyl are optionally substituted w ith OH, N0 2 , CN, Br, CI, F, and I.
  • A is methyl .
  • A is selected from F, CI, Br, and I.
  • A is CrCgcycloalkyl.
  • A is cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl.
  • A is substituted with OH, N0 2 , or CN.
  • B is C
  • 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 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 Q-Qcycloalk l. In another embodiment, B is cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl. In one embodiment, A is substituted with OH, N0 2 , or CN.
  • B is hydrogen and A is selected from Br, CI, F, I, Q-Qalkyl.
  • both A and B are hydrogen.
  • both A and B are selected from Br, CI, F, I, C
  • Y is an aryl group optionally substituted with 1 , 2, or 3 R 6 ; wherein each 3 ⁇ 4 is selected from OH, N0 2 , CN, Br, CI, F, I, C C 6 alkyl, C 3 -C 8 cyeloalkyl, and C 2 -C 3 heterocycloalkyl, C ⁇ -G,alken l.
  • Y is an optionally substituted phenyl group.
  • Y is a phenyl group.
  • the phenyl group is optionally substituted with at least one R ⁇ , selected from OH, N0 2 , CN, Br, CI, F, 1, C r Qalkyl, C 3 -C 8 cycloalkyl, C 2 -C 8 heterocycloalkyl, C 2 -Qalkenyl, alkoxy, alkoxyalkyi, alkoxycarbonyl, alkoxycarbonylalkyl, C -C 6 alkynyl, aryl, arylalkyl, C 3 -C 8 cycloalkylalkyl, haloalkoxy, haloalkyl, hydroxyalkylene, oxo, heteroaryl, heteroarylalkoxy, heteroaryloxy, heteroarylthio, heteroarylalkylthio, heterocycloalkoxy, C 2 -Qheterocycloalkylthio, heterocyclooxy, heterocyclothio, -NR A R B , -(Q
  • 3 ⁇ 4 is -CH 2 (NR A R B ).
  • R A is H or alkyl.
  • R A is C r C 6 alkyI.
  • R A is -CH 3 .
  • R B is H or alkyl.
  • R B is C r C 6 alkyl.
  • R B is -Cl h.
  • R A is -CH 2 NHCH 3 .
  • R ⁇ - is -(CO)heterocycloalkyl(CO)alkyl.
  • R ⁇ is -(CO)heterocycloalkyl(CO)alkyl wherein the heterocycloalkyl group has at least one heteroatom selected from O, N, and S. In another embodiment, the heterocycloalkyl group has two N atoms.
  • R ⁇ is -(CO)heteroeycloalkyl(CO)alkyI wherein alkyl is selected from methyl, ethyl, n- propyl, iso-propyl, cycloprop l, n-butyl, iso-butyl, and t-butyl.
  • the alkyl group is er embodiment, the alkyl group is iso-propyl.
  • R 6 is
  • heteroaryl group is selected from pyridine, pyrimidine, pyrazine, pyrazole, oxazole, thiazole, isoxazole, isothiazole, 1 ,3,4 -oxadiazole, pyridazine, 1,3,5-trazine, 1 ,2,4-triazine, quinoxaline, benzirnidazole, benzotriazole, purine, lH-[ l,2,3]triazoio[4,5- i ]pyrimidine, triazole, imidazole, thiophene, furan, isobenzofuran, pyrrole, indolizine, isoindole, indole, indazole, isoquinoline, quinoline, phthalazine, naphthyridine
  • Y is pyridine. In another embodiment, Y is optionally substituted pyridine. In one embodiment, Y is imidazole. In another embodiment, Y is optionally substituted imidazole. In one embodiment, Y is triazole. in another embodiment, Y is optionally substituted triazole.
  • the Compound of Formula (I) or (II) is that where Y is a substituent independently selected from the group consisting of hydrogen, alkenyl, alkoxy, alkoxyalkyl, alkoxycarbonyl. alkoxycarbonylalkyl, alkyl, alkynyl, arylalkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, hydroxyalkylene, oxo, heterocycloalkyi, heterocycloalkylalkyl, alkylcarbonyl, arylcarbonyl,
  • Y is alkyl. In another embodiment, Y is C[-C 6 alkyl. In a further embodiment, Y is selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl or tert-butyl. In another embodiment, Y is iso-propyl.
  • heterocycloalkyi group is selected from 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, ind
  • the heterocycloalkyi group is selected from pyrrolidinyl, imidazolidinyl, piperidinyl, piperazinyl, pyrazolidinyl, tetrahydrofuranyl, tetrahydroth iopheny 1, 1 ,3- oxathiolanyl, indolinyl, isoindolinyl, morpholinyl, and pyrazol inyl.
  • the heterocycloalkyi group is piperidinyl.
  • R R 2 , 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, - R A R B . -(Ci-C 6 alkylene)-NR A B , and -C(0)(NR A RB);
  • R A, and 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 lieteroatoms or hetero functionalities selected from the group consisting of -O-.
  • R4 and R 3 are each independently selected from the group consisting of hydrogen, alkyl, cycloalkyl, alkoxyalkyi, haloalkyl, hydroxyalkylene, and -(
  • a and B are each independently selected from hydrogen, Br, CI, F, I, OH, C
  • Y is selected from the group consisting of:
  • an aryl group optionally substituted with 1 , 2, or 3 substituents R $ ; is selected from OH, N0 2 , CN, Br, CI, F, I, C C 6 alkyl, C 3 -C 8 cycloalkyl, C 2 -C 8 heterocycloalkyl; C2-C 6 alkenyl, alkoxy, alkoxyalkyi, alkoxycarbonyl, alkoxycarbonylalkyl, C 2 -C 6 alkynyl, aryl, arylalkyl, C 3 - Cgcycloalkylalkyl, haloalkoxy, haloalkyl, hydroxyalkylene, oxo, heteroaryl, heteroarylalkoxy, heteroaryloxy, heteroarylthio, heteroarylalkylthio, heterocycloalkoxy, C 2 -C 8 heterocycloalkylthio, heterocyclooxy, heterocyclothio, -NR A R B ,
  • a substituent independently selected from the group consisting of hydrogen, alkenyl, alkoxy, alkoxyalkyi, alkoxycarbonyl, alkoxycarbonylalkyl, alkyl, alkynyl, arylalkyl.
  • n is an integer from 0-4; or a pharmaceutically acceptable salt, solvate or prodrug thereof.
  • Y is an aryl group.
  • Y is a heteroaryl group.
  • the aryl group is a phenyl group.
  • the phenyl group is substituted with at least one R ⁇ , selected from Br, C I, F, and I.
  • R 6 is F.
  • In one embodiment is a compound of Formula (IA), (IA 1 ), or (IA2) wherein the phenyl group is substituted with at least one R 6 selected from -(C,-C 6 alkylene)(NR A R B ), -C(0)(NR A R B ), -(C r C 6 alkylene)C(0)(NR A R B ), -S(0) 2 (NR A R B ), and -(Ci-C 6 alkylene)S(0)2(NR A R B ).
  • R 6 is -(C C 6 alkylene)(NR A R B ).
  • -C 6 alkylene is selected from methylene, ethylene, n-propylene, iso-propylene, n-butylene, iso-butylene, and tert-butylene.
  • -C ( ,aikylene is methylene.
  • R A and R B are each independently hydrogen, C r C 6 alkyl, or C3-C 8 cycloalkyl.
  • CrQalkyl is selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert- butyl.
  • Ci-C 6 alkyl is methyl.
  • -C 6 alkyl is ethyl.
  • Cj-Cgcycloalkyl is cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl.
  • C 3 -C 8 cycloalkyl is cyclopropyl.
  • Re is hydroxyalkylene.
  • hydroxyalkylene is selected from -CH 2 OH, -CH 2 CH 2 OH, -CH 2 CH,CH 2 OH, -CH(OH)CH : literally -CH(OH)CH 2 CH ? , -CH 2 CH(OH)CH 3 . and -CH 2 CH 2 CH,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 alkyl).
  • the hetero functionality is -N(C r C 6 alkyl)-.
  • C C 6 alkyl is selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-biityl.
  • C r C 6 alkyI is methyl.
  • Y is a heteroaryl group optionally substituted with at least one Rg.
  • the heteroaryl group is selected from furan, pyridine, pyrimidine, pyrazine, imidazole, thiazole, isothiazole, pyrazole, triazole, pyrrole, thiophenc, oxazole, isoxazole, 1 ,2,4-oxadiazole, 1 ,3,4-oxadiazole, 1,2,4-triazine, indole, benzothiophene, benzo im idazole, benzofuran, pyridazine, 1 ,3.5-triazine, thienothiophene, quinoxaline, quinoline, and isoquinoline.
  • the heteroaryl group is imidazole.
  • imidazole is substituted with C r C6alkyl selected from methyl, ethyl, n-propyl, iso-propyl, n- butyl, iso-butyl, and tert-butyl.
  • Ci -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 Ci-C 6 alkyl selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert- butyl.
  • Ci-C 6 alkyl selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert- butyl.
  • C C s alkyl is methyl.
  • [0071] in one embodiment is a compound of Formula (IA), (IA 1), or (IA2) wherein A and B are each independently selected from hydrogen, Br, CI, F, I, OH, C
  • the Compound of Formula (IA), (IA L ), or (IA2) is that wherein A and B are hydrogen.
  • Y is a heteroaryl group 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.
  • Y is a heteroaryl group selected from furan, pyridine, pyrimidine, pyrazine, imidazole, thiazole, isothiazole, pyrazole, triazole, pyrrole, thiophene, oxazole, iso
  • Y is an imidazole group.
  • the imidazole group is substituted with a CpQalkyl group.
  • the Q - C 6 alkyl group is methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl.
  • Ci-C 6 alkyl is methyl.
  • Y is a substituted imidazole group and Z is selected from an aryl group or a heteroaryl group.
  • Z is an aryl group.
  • the aryl group is a phenyl group.
  • the aryl group is a phenyl group substituted by a halogen.
  • Z is a heteroaryl group.
  • the heteroaryl group is 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.
  • the im idazole group is substituted with a C C 6 alkyl group.
  • the C C 6 alkyl group is methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl.
  • C r C 6 alkyI is methyl.
  • Y is a triazole group.
  • the triazole group is substituted with a C C ( ,alkyl group.
  • the - alk l group is methyl, ethyl, n-propyl, iso-propyl, n- but l, iso-butyl, and tert-butyl.
  • CrC 6 alkyl is methyl.
  • Y is a substituted triazole group and Z is selected from an aryl group or a heteroaryl group.
  • Z is an aryl group.
  • the aryl group is a phenyl group.
  • the aryl group is a phenyl group substituted by a halogen.
  • Z is a heteroaryl group.
  • the heteroaryl group is furan.
  • the heteroaryl group is triazole.
  • the triazole group is substituted with a C r C 6 alkyl group.
  • -C 6 alkyl group is methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl.
  • C C ( ,alkyl is methyl.
  • Y 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, and I.
  • R A is F.
  • the phenyl group is substituted with at least one R cargo selected from -(C r
  • C 6 alkylene)S(0) 2 (NR A R B ).
  • R « is -(C r C 6 alkylene)(NR A R B ).
  • -C,,alkylene is selected from methylene, ethylene, n-propylene, iso-propylene, n-butylene, iso-butylene, and tert-butylene.
  • C :-C penaltyalkylene is methylene.
  • R A and R B are each independently hydrogen, Ci-C 6 alkyl, or C 3 -CgcycloaIkyl.
  • Ci-Qalkyl is selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl.
  • Ci -C 6 alkyl is methyl.
  • C C 6 alkyl is ethyl.
  • C 3 - Cgcycloalkyl is cyclopropyl, eye lo butyl, cyclopentyl, and cyc!ohexyl.
  • C ⁇ - Cgcycloalkyl is cyclopropyl.
  • R A is hydroxyalkylene. I n one embodiment hydroxyalkylene is selected from -CH 2 OH, -CH 2 CH 2 OH, -CH 2 CH 2 CH 2 OH, -CH(OH)CH : Struktur
  • the hetero functionality is -N(C r C 6 alkyl)-.
  • Cj-Ceaikyl is selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl.
  • Ci-C 6 alkyl is methyl.
  • In one embodiment is a compound of Formula (I), (IIA), (IA 1 ), (IA2), or (II) wherein Z 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, and I.
  • R 3 ⁇ 4 is F.
  • R tile is CI.
  • the phenyl group is substituted with at least one R ⁇ ; selected from -(C,-C 6 alkylene)(NR A R B ), -C(0)(NR A R B ), -(C r C 6 alkylene)C(0)(NR A R B ), -S(0) 2 (NR A R B ), and (NR A R B )suifonylalkylene.
  • R 6 is -(C C 6 aIkylene)(NR A R B ).
  • C r C 6 alkylene is selected from methylene, ethylene, n-propylene, iso-propylene, n-butyiene, iso-butylene, and tert-butyiene.
  • C r C 6 aikyIene is methylene.
  • R A and RB are each independently hydrogen, Ci-C 6 aiky l, or C 3 -C 8 cycloalkyl.
  • -C 6 alkyl is selected from methyl, ethyl, n-propyi, iso-propyl, n-butyl, iso-butyl, and tert- butyl.
  • C r 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(C r C 6 alkyl)-.
  • the hetero functionality is— (C
  • Ci-C f talkyl is selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl.
  • C r Qalkyl is methyl.
  • NR A R B is -CH 2 (NR A R B ) wherein NR A R B is azetidine, pyrrolidine, piperidine or morpholine.
  • R 2 is hydrogen.
  • R 2 is selected from F, CI, Br, and I.
  • R 2 is F.
  • a and B are hydrogen.
  • a and B are independently selected from hydrogen and Q - Qalkyl.
  • Z is aryl and Y is independently selected from the group consisting of
  • phenyl group optionally substituted with 1 , 2, or 3 R ⁇ , ;
  • a substituent independently selected from the group consisting of hydrogen, alkoxyalkyl, alkoxycarbonylalkyl, alkyl, arylalkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, oxo, heterocycloalkyl, heterocycloalkylalkyl, aikylcarbonyl, arylcarbonyl, heteroarylcarbonyl, -(C ,-C 6 alkylene)(NR A R B ), and -C(0)(NR A R B ).
  • Z is phenyl and Y is independently selected from the group consisting of
  • a substituent independently selected from the group consisting of hydrogen, alkoxyalkyl, alkoxycarbonylalkyi, alkyl, arylalkyl, cycloalkyl, cycloalkyialkyl, haloalkyl, oxo, heterocycloalkyl, heterocycloalkylalkyi, alkylcarbonyl, aryicarbonyl, heteroarylcarbonyi, -(C r C 6 alkylene)(NR A R B ), and -C(0)(NR A R B ).
  • Z is phenyl substituted with 1 , 2, or 3 R ⁇ , and Y is independently selected from the group consisting of a) phenyl group optionally substituted with 1 , 2, or 3 R 6 ;
  • a substituent independently selected from the group consisting of hydrogen, alkoxyalkyl, a 1 koxy carbony I a 1 ky I , alkyl, arylalkyl, cycloalkyl, cycloalkyialkyl, haloalkyl, oxo, heterocycloalkyi, heterocycloalkylalkyi, alkylcarbonyl, aryicarbonyl, heteroarylcarbonyi, -(C C 6 alkyiene)(NR A R B ), and -C(0)(NR A R B ).
  • the Compound of Formula (I), (IA), (IA 1 ), (IA2), or (II) is that where 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-butyi, 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 OH.
  • A is cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl.
  • A is substituted with OH, N0 2 , or CN.
  • A is hydrogen.
  • B is hydrogen.
  • B is C C 6 lkyI.
  • B is selected from methyl, ethyl, n-propyi, iso- propyl, n-butyl, iso-butyl, tert-butyl, n-pentyl, and n-hexyl.
  • methyi, ethyl, n- propyl, iso-propyl, n-butyi, iso-butyl, tert-butyl, n-pentyl, and n-hexyl are optionally substituted with OH, O2, CN, Br, CI, F, and I.
  • B is methyl.
  • B is selected from F, CI, Br, and I.
  • B is Cj-Cgcycloalkyl.
  • B is cyclopropy l, cyclobutyl, cyclopentyl, or cyclohexyl.
  • A is substituted with OH, N0 2 , or CN.
  • Ci-C 6 alkyl, and Cj-Cgcyctoalkyl is a compound of Formula (II) wherein B is hydrogen and A is selected from Br, CI. F, I, C t - Qalkyl, C 3 -C 8 cycloalkyl, alkoxy, and alkoxyalkyl wherein C r C 6 alkyl, Cj-Cgcycloalkyl, alkoxy, and alkoxyalkyl are optionally substituted with at least one substituent selected from OH, NO?, CN, Br, C I, F, I, C t -C 6 alkyl, and Cj-Cgcycloalkyl.
  • both A and B are hydrogen.
  • both A and B are selected from Br, CI, F, I, OH, C r C 6 alkyl, C 3 -CgcycioaikyI, alkoxy, and alkoxyalkyl wherein CpQalkyl, C-rQcycloaikyl, alkoxy, and alkoxyalkyl are optionally substituted with at least one substituent selected from OH, N0 2 , CN, Br, CI, F, I, C r C 5 alkyl, and Q-Qcycloalkyl wherein B is not OH.
  • enantiomers such as enantiomers, diastereomers, and mixtures of enantiomers or diastereomers.
  • diastereomers such as enantiomers, diastereomers, and mixtures of enantiomers or diastereomers.
  • mixtures of enantiomers or diastereomers are also described herein.
  • Y is an aryl or heteroaryl group optionally substituted with at least one R ⁇ ,
  • Z is an aryl group optionally substituted with at least one R «;
  • a and B are each independently selected from hydrogen, Br, CI, F, I, OH, C
  • R ⁇ is selected from OH, NO,, CN, Br, CI, F, I, C,-C ( ,alkyl.
  • R is selected from hydrogen, Br, Cl, I, or F;
  • R A and R B are independently selected from the group consisting of hydrogen, Ci-C 6 alkyl, C 3 - Qcycloalkyl, 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
  • the Q-Cealkyl is methyi.
  • the phenyl group is substituted with a halogen.
  • the halogen is F.
  • the halogen is selected from Br, CI, and I.
  • the phenyl group of Z is substituted with a halogen, selected from F, Br, CI, and I.
  • the phenyl group of Z is substituted with F. In yet a further embodiment the phenyl group of Z is substituted with -C
  • the compound optionally as a single stereoisomer or mixture of stereoisomers thereof and additionally optionally as a pharmaceutically acceptable salt thereof, is selected from
  • the tosylate salt is in a crystalline form.
  • the (85,9i?)-5-fluoro-8-(4-fluorophenyl)-9-( l -methyl-lH- U2,4- triazol-5-yl)-8,9-dihydro-2H-pyrido[4,3,2-de]phthalazin-3(7/i)-one tosylate salt provided herein is in a crystalline form.
  • the crystalline form is unsolvated.
  • the crystalline form is a solvate.
  • a crystalline solvate form may be a hydrate.
  • the (8S,9i?)-5-fluoro-8-(4-fIuorophenyl)-9-( 1 -methyl- 1 //- 1 ,2,4-triazol-5-y l)-8,9-dihydro- 2/ -pyrido[4,3,2-de]phthalazin-3(7 /)-one tosylate salt provided herein is in an amorphous form.
  • prov ided herein is a solid form (e.g., a crystalline form, an amorphous form, or a mixture of forms) of (8.S ' .9/?)-5-nuoro-8-(4-nuorophenyD-9-( 1 -methyl- 1 /-/- 1 .2.4-triazol-5-yl)-8.9-dihydro-2/A pyrido[4,3,2-de]phthalazin-3(7i ⁇ )-one or a salt or solvate thereof ⁇ e.g., a salt provided herein elsewhere).
  • the (8S,9R)-5-fluoro-8-(4-fIuorophenyl)-9-( l -methyl- 1H- 1 ,2,4- triazol-5-yl)-8,9-dihydro-2 f-pyrido[4,3,2-de]phthalazin-3(7/i)-one tosylate crystalline salt form provided herein is substantially pure.
  • the crystalline tosylate salt purity is of at least about 90%, at least about 95%, at least about 97%, at least about 98%, at least about 99%, at least about 99.2%, at least about 99.5%, at least about 99.6%, at least about 99.7% or at least about 99.8% by- weight of a single crystalline form, the remainder of the total weight which may be other crystal line or amorphous forms and/or other compounds.
  • the crystalline tosylate salt is essentially a single-component crystalline form or a single polymorph.
  • the crystalline tosylate salt is a multiple-component crystalline form comprising a first crystal line form and at least one other crystalline and/or amorphous form of (8S,9R)-5-fliioro-8-(4-fluorophenyl)-9-( l -methyl- lH- l ,2,4-triazol- 5-yl)-8,9-dihydro-2//-pyrido[4,3,2-de]phthalazin-3(7i/)-one.
  • the crystalline form is substantially free of an amorphous form of (85,9R)-5-fIuoro-8-(4-fluorophenyl)-9-( l -methyl- 1H- l ,2,4-triazol-5-yl)-8,9-dihydro-2 /-pyrido[4,3,2-de]phthalazin-3(7 /)-one.
  • an XRPD peak is expressed in 2 ⁇ angle degrees, it should be understood that copper Kal radiation is used.
  • the 2 ⁇ angle degrees value provided herein may vary to an extent of about ⁇ 0.2 ° ⁇ , while still describing the same XRPD peak.
  • the peak positions can vary depending on factors such as signal-to-noise ratio, peak width, temperature, spinning speed, decoupling efficiency, magic angle setting, data processing procedures and parameters, and software peak picking algorithm.
  • peak position is relative to the chemical shift referencing procedure.
  • chemical shift reference standards may be used and wi ll not necessarily give the same results. This may lead to peak positions that are different by several ppm.
  • typical ly al l of the peaks wil l have a systematic change in position in the same direction if a different reference standard was used or if the analyst used a different value for the reference peak position of the same standard.
  • the ppm values in the l C solid state NMR provided herein may vary to an extent of about ⁇ 0.2 ppm, while still describing the same peak.
  • a crystalline salt of (85,9 J R)-5-fluoro-8-(4-fluorophenyl)-9-( l - methyl-1 //- l ,2,4-triazol-5-yl)-8,9-dihydro-2i -pyrido[4,3,2-de]phthalazin-3(7H)-one having an XRPD pattern comprising one or more (e.g.
  • the crystalline salt is a tosylate salt having one or more (e.g., one, two, three, four, five, six, seven, eight, nine, ten, or greater than ten; or at least three, at least four, at least five, at least six, or at least seven) XRPD peaks selected from peaks with 2 ⁇ angle degrees according to any one of Tables 9, 11, 13, 15, 17 and 25.
  • the crystalline tosylate salt has an XRPD pattern substantially as provided in Figures 1, 6, or 8. [0092J In certain embodiments, the crystalline tosylate salt provided herein has an XRPD pattern comprising one or more (e.g., one, two, three, four, five, six, seven, eight, nine, ten, or greater than ten; or at least three, at least four, at least five, at least six, or at least seven) XRPD peaks selected from peaks with 2 ⁇ angle degrees or d values (A) as provided in Table 15.
  • the crystalline tosylate salt has XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1.79, 5.86, 4.90, 4.42, 4.35, 3.93, and 3.70.
  • the crystalline tosylate salt has XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1.79; 5.86; 4.90; 4.65; 4.42; 4.35; 4.13; 3.93; and 3.70.
  • the XRPD pattern of the crystalline tosylate salt comprises one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1 .79; 5.86; 4.98; 4,90; 4.79; 4.65; 4.42; 4.35; 4.13; 3.93; 3.70; and 3.58.
  • peaks e.g., at least three, at least four, at least five, at least six, or at least seven peaks
  • the XRPD pattern of the crystalline tosylate salt comprises one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) comprise 1 1 .79; 7.07; 6.13; 5.86; 5.10; 4.98; 4.90; 4.79; 4.65 ; 4.42; 4.35; 4.13; 4.08; 3.93; 3.85; 3.70; 3.58; 3.3 1 ; and 2.99.
  • peaks with d values comprise 1 1 .79; 7.07; 6.13; 5.86; 5.10; 4.98; 4.90; 4.79; 4.65 ; 4.42; 4.35; 4.13; 4.08; 3.93; 3.85; 3.70; 3.58; 3.3 1 ; and 2.99.
  • the crystal line tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2 ⁇ angle degrees of about 7.49, 15.10, 18.10, 20.06, 20.40, 22.61 , and 24.01.
  • peaks e.g., at least three, at least four, at least five, at least six, or at least seven peaks
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2 ⁇ angle degrees of about 7.49, 15.10, 1 8.10, 19.08, 20.06, 20.40, 21.49, 22.61 , and 24.01.
  • peaks e.g., at least three, at least four, at least five, at least six, or at least seven peaks
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 20 angle degrees of about 7.49, 15.10, 17.78, 18.10, 1 8.49, 19.08, 20.06, 20.40, 21 .49, 22.61, 24.01 , and 24.84.
  • peaks e.g., at least three, at least four, at least five, at least six, or at least seven peaks
  • the crystal line tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 20 angle degrees of about 7.49, 12.50, 14.44, 15.10, 17.38, 17.78, 18.10, 18.49, 19.08, 20.06, 20.40, 21.49, 21 .76, 22.61 , 23.05, 24.01, 24.84, 26.93, and 29.82,
  • peaks e.g., at least three, at least four, at least five, at least six, or at least seven peaks
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2 ⁇ angle degrees or d values (A) as provided in Table 15.
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1.8, 5.9, 4.9, 4.42, 4.35, 3.9, and 3.7.
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1.8; 5.9; 4.9; 4,7; 4.42; 4.35; 4.1 ; 3.9; and 3.7.
  • peaks e.g., at least three, at least four, at least five, at least six, or at least seven peaks
  • the XRPD pattern comprises one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) comprise about 1 1.8; 5.9; 5.0; 4.9; 4.8; 4.7; 4.42; 4.35; 4.1 ; 3.9; 3.70; and 3.58.
  • peaks e.g., at least three, at least four, at least five, at least six, or at least seven peaks
  • peaks with d values (A) comprise about 1 1.8; 5.9; 5.0; 4.9; 4.8; 4.7; 4.42; 4.35; 4.1 ; 3.9; 3.70; and 3.58.
  • the XRPD pattern comprises one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) comprise about 1 1.8; 7.1 ; 6.1 ; 5.9; 5.1 ; 5.0; 4.9; 4.8; 4.7; 4.42; 4.35; 4.1 ; 4.1 ; 3.9; 3.9; 3.7; 3.6; 3.3; and 3.0.
  • peaks with d values (A) comprise about 1 1.8; 7.1 ; 6.1 ; 5.9; 5.1 ; 5.0; 4.9; 4.8; 4.7; 4.42; 4.35; 4.1 ; 4.1 ; 3.9; 3.9; 3.7; 3.6; 3.3; and 3.0.
  • the crystal line tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2 ⁇ angle degrees of about 7.5, 15.1 , 18.1, 20.1 , 20.4, 22.6, and 24.0.
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 20 angle degrees of about 7.5, 15.1 , 18.1 , 19. 1 , 20.1 , 20.4, 21 .5, 22.6, and 24.0.
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2 ⁇ angle degrees of about 7.5, 1 5.1 , 17.8, 18. 1 , 1 8.5, 19. 1 , 20.1 , 20.4, 21.5, 22.6, 24.0, and 24.8.
  • peaks e.g., at least three, at least four, at least five, at least six, or at least seven peaks
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 20 angle degrees of about 7.5, 12.5, 14.4, 15.1 , 17.4, 17.8, 18.1 , 18.5, 19.1 , 20.1 , 20.4, 21.5, 21 .8, 22.6, 23.1 , 24.0, 24.8, 26.9, and 29.8.
  • peaks e.g., at least three, at least four, at least five, at least six, or at least seven peaks
  • the crystalline tosylate salt comprises XRPD peaks at 20 angle degrees or d values (A) as provided in Table 9.
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 20 angle degrees of about 7.42, 15.01 , 17.70, 18.01 , 18.47, 18.98, 19.98, 20.33 , 21.41 , 22.58, 23.95, and 24.76.
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2 ⁇ angle degrees of about 7.42, 15.01 , 18.01 , 19.98, 20.33, 22.58, and 23.95.
  • peaks e.g., at least three, at least four, at least five, at least six, or at least seven peaks
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2 ⁇ angle degrees of about 7.42, 15.01 , 17.70, 18.01, 19.98, 20.33, 21.41 , 22.58, 23.95, and 24.76.
  • peaks e.g., at least three, at least four, at least five, at least six, or at least seven peaks
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1.90, 5.90, 5.01 , 4.92, 4.44, 4.37, 4.15, 3.93, 3.71 , and 3.59.
  • peaks e.g., at least three, at least four, at least five, at least six, or at least seven peaks
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1.90, 5.90, 5.01 , 4.92, 4.80, 4.67, 4.44, 4.37, 4.1 5, 3.93, 3.71 , and 3.59.
  • peaks e.g., at least three, at least four, at least five, at least six, or at least seven peaks
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1 .90, 5.90, 4.92, 4.44, 4.37, 3.93, and 3.71.
  • the crystalline tosyiate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2 ⁇ angle degrees or d values (A) as provided in Table 9.
  • the crystal line tosyiate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 20 angle degrees of about 7,4, 15.0, 17.7, 1 8.0, 18.5, 19.0, 20.0, 20.3, 21.4, 22.6, 24.0, and 24.8.
  • the crystalline tosyiate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six. or at least seven peaks) selected from peaks with 2 ⁇ angle degrees of about 7.4, 15.0, 18.0, 20.0, 20.3, 22.6, and 24.0.
  • the crystalline tosyiate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 20 angle degrees of about 7.4; 15.0; 17.7; 18.0; 20.0; 20.3; 21 .4; 22.6; 24.0; and 24.8.
  • peaks e.g., at least three, at least four, at least five, at least six, or at least seven peaks
  • the crystalline tosyiate salt has an XRPD partem comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1 .9, 5.9, 5.0, 4.9, 4.44, 4.37, 4. 1 , 3.9. 3.7, and 3.6.
  • the crystalline tosyiate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1.9, 5.9, 5.0, 4.9, 4.8, 4.7, 4.44, 4.37, 4. 1 , 3.9, 3.7, and 3.6.
  • the crystalline tosyiate salt comprises XRPD peaks at d values (A) of about 1 1.9, 5.9, 4.9, 4.44, 4.37, 3.9, and 3.7.
  • the crystal line tosyiate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 20 angle degrees or d values (A) as provided in Table 11.
  • peaks e.g., at least three, at least four, at least five, at least six, or at least seven peaks
  • the crystalline tosyiate salt has an XRPD partem comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2 ⁇ angle degrees of about 7.42, 15.02, 17.38, 17.74, 18.03, 18.54, 19.02, 20.08, 20.39, 2 1 .44, 22.63, 24.00, and 24.83.
  • peaks e.g., at least three, at least four, at least five, at least six, or at least seven peaks
  • the crystal line tosyiate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 20 angle degrees of about 7.42, 15.02, 18.03, 20.08, 20.39, 22.63, and 24.00.
  • peaks e.g., at least three, at least four, at least five, at least six, or at least seven peaks
  • the crystalline tosyiate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1.91 , 5.89, 5.10, 5.00, 4.92, 4.78, 4.66, 4.42, 4.35, 4.14, 3.93, 3.71 , and 3.58.
  • peaks e.g., at least three, at least four, at least five, at least six, or at least seven peaks
  • the crystalline tosyiate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1.91 , 5.89, 4.92, 4.42, 4.35, 3.93, and 3.71 .
  • the crystalline tosyiate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 20 angle degrees or d values (A) as provided in Table 11.
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2 ⁇ angle degrees of about 7.4, 15.0, 17.3, 17.7, 1 8.0, 18.5, 19.0, 20. 1 , 20.4, 21.4, 22.6, 24.0, and 24.8.
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2 ⁇ angle degrees of about 7.4, 1 5.0, 18.0, 20.1 , 20.4, 22.6, and 24.0.
  • peaks e.g., at least three, at least four, at least five, at least six, or at least seven peaks
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1.9, 5.9, 5.1 , 5.0, 4.9, 4.8, 4.7, 4.42, 4.35, 4.1 , 3.9, 3.7, and 3.6.
  • peaks e.g., at least three, at least four, at least five, at least six, or at least seven peaks
  • the crystal line tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1.9, 5.9, 4.9, 4.42, 4.35, 3.9, and 3.7.
  • peaks e.g., at least three, at least four, at least five, at least six, or at least seven peaks
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2 ⁇ angle degrees or d values (A) as provided in Table 13.
  • peaks e.g., at least three, at least four, at least five, at least six, or at least seven peaks
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 20 angle degrees of about 7.46, 12.47, 14.45, 15.09, 17.40, 17.74, 18.1 1 , 18.53, 19.05, 20.09, 20.43, 21.46, 22.63, 23.10, 24.03, 24.85, and 26.96.
  • peaks e.g., at least three, at least four, at least five, at least six, or at least seven peaks
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 20 angle degrees of about 7.46, 14.45, 15.09, 17.74, 18. 1 1 , 1 8.53, 19.05, 20.09, 20.43, 21 .46, 22.63, 24.03, 24.85, and 26.96.
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g.
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1.84, 7.09, 6.13, 5.87, 5.09, 5.00, 4.89, 4.78, 4,66, 4.42, 4.34, 4.13, 4.08, 3.93, 3.85, 3.70, 3.58, 3.30, 2,99, and 2.86.
  • d values A
  • the crystal line tosylate salt has an XRPD pattern comprising one or more peaks (e.g. , at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1 .84, 6.13, 5.87, 5.09, 5.00, 4.89, 4.78, 4.42, 4.34, 4.13, 3.93, 3.70, 3.58, and 3.30.
  • peaks e.g. , at least three, at least four, at least five, at least six, or at least seven peaks
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1 .84, 5.87, 4.89, 4.42, 4.34. 3.93, and 3.70.
  • peaks e.g., at least three, at least four, at least five, at least six, or at least seven peaks
  • the crystalline tos late salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2 ⁇ angle degrees or d values (A) as provided in Table 13.
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2 ⁇ angle degrees of about 7.5.
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2 ⁇ angle degrees of about 7.5, 14.5, 15.1 , 17.7, 18.1 , 18.5, 19.0, 20.1 , 20.4, 21.5, 22.6, 24.0, 24.9, and 27.0.
  • peaks e.g., at least three, at least four, at least five, at least six, or at least seven peaks
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2 ⁇ angle degrees of about 7.5, 15.1, 1 8.1 , 20.1 , 20.4, 22.6, and 24.0.
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1.8, 7.1 , 6. 1 , 5.9, 5.
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1.8, 6. 1 , 5.9, 5.0, 4.9, 4.8, 4.7, 4.4, 4.3, 4.1 , 3.9, 3.7, 3.6, and 3.3.
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1.8, 5.9, 4.9, 4.4, 4.3, 3.9, and 3.7.
  • peaks e.g., at least three, at least four, at least five, at least six, or at least seven peaks
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2 ⁇ angle degrees or d values (A) as provided in Table 17.
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2 ⁇ angle degrees of about 7.50, 12.5 1 , 14.48, 15.12, 17.38, 17.78, 18.
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 20 angle degrees of about 7.50, 15. 12, 1 7.38, 17.78, 1 8.17, 1 8.58, 19.
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2 ⁇ angle degrees of about 7.50, 15.12, 18.17, 20.09, 20.54, 22.65, and 24.08.
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1 .78, 7.07, 6.1 1 , 5.85, 5. 10, 4.98, 4.88, 4.77, 4.64, 4.42, 4.32, 4.12, 4.06, 3.92, 3.83, 3.69, 3.57, 3.30, 2.98, 2.93, 2.78, 2.75, 2.39.
  • peaks e.g., at least three, at least four, at least five, at least six, or at least seven peaks
  • d values A
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1 .78, 5.85, 5.10, 4.98, 4.88, 4.77, 4.64, 4.42, 4.32, 4. 12, 4.06, 3.92, 3.83, 3.69, 3.57, and 3.30.
  • the crystalline tos late salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1.78, 5.85, 4.88, 4.42, 4.32, 3.92, and 3.69.
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2 ⁇ angle degrees or d values (A) as provided in Table 17.
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2 ⁇ angle degrees of about
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 20 angle degrees of about 7.5, 15.1 , 17.4, 17.8, 18.2, 18.6, 19. 1 , 20.1 ,
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2 ⁇ angle degrees of about 7.5, 15.1 , 18.2, 20.1 , 20.5, 22.6, and 24.1.
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1.8, 7.1 , 6.1 , 5.9, 5.1, 5.0, 4.9, 4.8, 4.6, 4.4, 4.3, 4.12, 4.06, 3.9, 3.8, 3.7,
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1 .8, 5.9, 5.1 , 5.0, 4.9, 4.8, 4.6, 4.4, 4.3, 4.12, 4.06, 3.9, 3.8, 3.7, 3.6, and 3.3.
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1.8, 5.9, 4.9, 4.4, 4.3, 3.9, and 3.7.
  • peaks e.g., at least three, at least four, at least five, at least six, or at least seven peaks
  • the crystalline tosylate salt has an XRPD pattern substantially as provided in Figure 6.
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2 ⁇ angle degrees as provided in Table 25.
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with ⁇ about 0.2 2 ⁇ angle degrees of about 7.51 , 14.47, 15.
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with ⁇ about 0.2 20 angle degrees of about 7.51 , 14.47, 15. 14, 20.09, 21.48, and 24.05.
  • peaks e.g., at least three, at least four, at least five, at least six, or at least seven peaks
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with ⁇ about 0.2 2 ⁇ angle degrees of about 7.51 , 15.14, 18.12, 20.09, 20.46, 22.65, and 24.05.
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with ⁇ about 0.2 2 ⁇ angle degrees of about 7.5 1 , 20.09, and 24.05.
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with ⁇ about 0.2 2 ⁇ angle degrees of about 7.5, 14.5, 15.1 , 17.4, 18.1 , 18.5, 19.1 , 20.1 , 20.46, 21.48, 2 1.8, 24.1 , 24.8, and 29.8.
  • peaks e.g., at least three, at least four, at least five, at least six, or at least seven peaks
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with ⁇ about 0.2 2 ⁇ angle degrees of about 7.5, 14.5, 15.1 , 20.1 , 21.5, and 24.1.
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with ⁇ about 0.2 2 ⁇ angle degrees of about 7.5, 15.1, 18.1 , 20.1 , 20.5, 22.6, and 24.1.
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with ⁇ about 0.2 2 ⁇ angle degrees of about 7.5, 20.1 , and 24.1.
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) as provided in Table 25.
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1 .8, 6.1 , 5.9, 5.1, 4.9, 4.8, 4.6, 4.4, 4.3, 4.1 , 4.1 , 3.7, 3.6, and 3.0.
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1 .8, 6.1 , 5.9, 4.4, 4.1 , and 3.7.
  • the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1.8, 5.9, 4.9, 4.4, 4.3, 3.9, and 3.7.
  • the crystal line tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1.8, 4.4, and 3.7.
  • peaks e.g., at least three, at least four, at least five, at least six, or at least seven peaks
  • the crystal line tosylate salt exhibits a "C NMR spectrum corresponding substantially to the spectrum in Figure 12 or exhibits a spectrum with peaks corresponding substantially to those in Table 27.
  • the crystalline tosylate salt exhibits a l3 C NMR spectrum with one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from about ⁇ 0.2 ppm at about 166.9, 164.3, 162.2, 160.6, 1 5 1 .8. 149.4, 143.2,
  • the crystalline tosylate salt exhibits a l3 C NMR spectrum with peaks about ⁇ 0.2 ppm at about 151.8, 149.4, 143.2, 136.0, 13 1.8, 123.9, 1 16.8, 1 15.1 , 1 12.2, 105.2,
  • the crystalline tosylate salt exhibits a , C NMR spectrum with peaks about ⁇ 0.2 ppm at about 143.2, 136.0, 131.8, 123.9, 1 12.2. 105.2, 100.3, 58.5, 45.3, 37.4, and 23.9.
  • the crystalline tosylate salt exhibits a l 3 C NMR spectrum with peaks about ⁇ 0.2 ppm at about 143.2, 136.0, 13 1.8, 123.9, 1 12.2, 105.2, and 100.3.
  • the crystalline salt has an XRPD peaks at 2 ⁇ angle degrees of about 7.4, 15.1 , 1 7.4, 17.8, 18.1 , 18.5, 19.1 , 20.1 , 20.4, 21.5, 22.6, 24.0, 24.8, and 27.0. In some or any embodiments, the crystalline salt has an XRPD peaks at 2 ⁇ angle degrees of about 7.4, 15.1, 18.1 , 19.1 , 20.1, 20.4, 21.5, 22.6, and 24.0. In some or any embodiments, the crystalline salt has an XRPD peaks at 2 ⁇ angle degrees of about 7.4, 15.1, 20.1 , 20.4, 22.6, 24.0, and 24.8. In some or any embodiments, the crystalline salt has an XRPD peaks at 20 angle degrees of about 7.4, 15.1 , 18.1 , 20. 1 , 20.4, 22.6, and 24.0.
  • the crystal line salt has an XRPD pattern comprising peaks at d values (A) of about 1 1.9, 5.9, 5.1 , 5.0, 4.9, 4.8, 4.6, 4.4, 4.3, 4.1 , 3.9, 3.7, 3.6, and 3.3.
  • the crystalline salt has an XRPD pattern comprising peaks at d values (A) of about 1 1 .9, 5.9, 4.9, 4.6, 4.4, 4.3, 4.1 , 3.9, and 3.7.
  • the crystalline salt has an XRPD pattern comprising peaks at d values (A) of about 1 1 .9, 5.9, 4.4, 4.3, 3.9, 3.7, and 3.6. In some or any embodiments, the crystalline salt has an XRPD peaks at d values (A) of about 1 1.9, 5.9, 4.9, 4.4, 4.3, 3.9, and 3.7.
  • crystalline (8S,9/?)-5-fluoro-8-(4-fluorophenyl)-9-( l- methyl- l //- 1 ,2,4-triazol-5-yl)-8,9-dihydro-2H-pyrido[4,3,2-de]phthalazin-3(7 /)-one tosylate is a crystalline polymorph exhibiting at least one of
  • an X-ray diffraction pattern comprising peak reflectances at 2 ⁇ angle degrees ⁇ 0.2 20 angle degrees of 7.4, 1 5. 1 , 18.1 , 20.1 , 20.4, 22.6, and 24.0.
  • the crystalline tosylate salt exhibits a single endothermal peak on differential scanning calorimetry between room temperature and about 350 ° C, where the single endothermal peak maximum occurs between about 320 °C to about 335 °C. In certain embodiments, the single endothermal peak maximum occurs at between about 330 " C to about 335 ° C. In some embodiments,
  • the single endothermal peak maximum occurs at about 333 °C to about 334 ° C.
  • 9-( I -methyl- l H- l ,2,4-triazol-5-yl)-8,9-dihydro-2H-pyrido[4,3,2-de]phthaIazin-3(7H)-one as provided herein has no observable endotherm from about 25 "C to about 250 °C as determined by DSC.
  • a crystalline tosylate salt of (8S,9i?)-5-fluoro-8-(4-fluoropheny I)-9-( 1 -methyl- 1 H- 1 ,2,4- triazol-5-yI)-8,9-dihydro-2 -pyrido[4,3,2-de]phthalazin-3(7//)-one as provided herein has a DSC thermogram comprising an endotherm with a maximum at between about 320 ' C to about 335 'C, between 330 ° C to about 335 * C, or between about 333 * C to about 334 * C.
  • a crystalline tosylate salt of (8S,9i?)-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 as provided herein does not exhibit significant weight change (e.g. , less than about 0.05 wt %, less than about 0.1 wt %, less than about 0.15 wt %, or less than about 0.2 wt %) from about 0% to about 95% relative humidity.
  • 9-(l -methyl-lH- l ,2,4 riazol-5-yl)-8,9-dihydro-2H-pyrido[4,3,2-de]phthalazin-3(7H)-one as provided herein has a TGA thermogram corresponding substantially to the TGA thermograph of Figure 2b, 3b, 4b, 5b, or 7b.
  • a crystalline tosylate salt of (8S,9i?)-5-fluoro-8-(4-fluorophenyl)-9-( l - methyl- lH- l ,2,4-triazol-5-yl)-8,9-dihydro-2/ -pyrido[4,3,2-de]phthalazin-3(7H)-one as provided herein has a TGA thermogram that does not exhibit significant weight loss (e.g., less than about 0.05 wt %, less than about 0.1 wt %, less than about 0.5 wt %, less than about 1 wt %, less than about 5 wt %, less than about 10 wt %, less than about 1 5 wt %, less than about 20 wt %, or less than about 25 wt %) when heated from about room temperature to a temperature of about 200 "C, about 210 °C, about 220 °C, about 230 °C, about
  • the crystall ine tosylate salt provided herein has a weight loss of no greater than about 1%, no greater than about 0.5%, or no greater than about 0.1 % in a thermogravimetric thermogram between about 25 ° C to about 200 ' C.
  • a crystalline tosylate salt as provided herein is anhydrous.
  • a crystalline tosylate salt as provided herein is anhydrous.
  • a crystalline tosylate salt of (8S,9R)-5- fluoro-8- ⁇ 4-tl uorophenyl)-9-( I -methyl- 1 H- 1 .2,4-triazol-5-yl)-8,9-dihydro-2//-pyrido[4,3.2-de]phthalazin- 3(7//)-one as provided herein is non-hygroscopic.
  • compositions comprise or consist essentially of substantially pure (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, in another embodiment in a crystalline form thereof as provided herein.
  • a pharmaceutical composition comprising a tosylate salt of (85,9R)-5-fluoro-8-(4-fluorophenyl)-9-( l -methyl-l H- l ,2,4-triazol-5-yl)-8,9-dihydro-2H- pyrido[4,3,2-de]phthalazin-3(7 )-one, including a crystalline form thereof as described herein, and one or more pharmaceutically acceptable excipient(s) and/or carrier(s).
  • excipient to a large extent, depends on factors, such as the particular mode of administration, the effect of the excipient on the solubility and stability of the active ingredient, and the nature of the dosage form.
  • compositions formulated such that Compound A tosylate salt is in a stable form, for instance when stored in an open container for up to 4 weeks or more under conditions such as at 40 °C and 75% relative humidity (RH).
  • pharmaceutical compositions provided herein are made with Compound A tosylate salt in a crystalline form.
  • compositions comprising a Compound of Formula I or Compound A tosylate salt and one or more pharmaceutically acceptable excipients.
  • the pharmaceutical composition is in a solid form, for example, in granules or in a powder, tablet, or capsule.
  • the pharmaceutical composition provided herein is suitable for oral administration to a subject. Components of the pharmaceutical compositions provided herein are - described below.
  • the pharmaceutical composition is prepared using Compound A tosylate salt consisting of a crystalline solid form.
  • the pharmaceutical composition provided herein comprises crystalline Compound A tosylate salt as described herein, in another example as defined in any of the Compound A embodiments herein.
  • the pharmaceutical composition is prepared with, and/or comprises crystalline Compound A tosylate salt which is unsolvated.
  • the crystalline form is a solvate, for example, a hydrate.
  • the weight weight percent of Compound A tosylate salt in the pharmaceutical composition provided herein is between about 0.01 % to about 5.0 %, between about 0.1%> and about 2.5%>, between about 1 % and about 2.5%», between about 1 % and about 3%, between about 2% and about 4%, between about 2% and about 3%, between about 3%o and about 5%, between about 0. 1 % and about 2%, between about 0.01 % and about 1.0 %, between about 0.
  • the percentages above refer to weight/weight percentages of Compound A tosylate salt in the pharmaceutical composition, excluding, for instance, any coating where the pharmaceutical composition is in a tablet form, or capsule shell where the pharmaceutical composition is encapsulated in a capsule.
  • composition provided herein comprises
  • a pharmaceutical composition as provided herein comprises a solid form Compound A tosylate salt that is substantially pure as defined herein.
  • solid form is a crystalline form, it can be substantially free of other crystalline polymorphs or amorphous forms of Compound A tosylate salt.
  • the solid Compound A tosylate salt in the pharmaceutical composition is stable, as defined herein.
  • the crystalline Compound A tosylate salt used in the preparation of the pharmaceutical composition is stable as defined, for instance, by no detectable change in its crystalline solid form (e.g., polymorphic change and/or chemical change such as, for example, due to degradation) when stored for 18 months or more at, for example, 25 ° C or ambient conditions or, for 4 weeks or more at 40 ° C and 75% RH.
  • the crystalline tosylate salt is essentially a single-component crystalline form or a single polymorph.
  • a low dose formulation of Compound A tosylate salt that is, where the weight/weight percent of Compound A tosylate salt in the pharmaceutical composition is such that a low dose, as defined herein, is delivered to a subject with an indication as provided herein, when milligrams (e.g., ranging in amounts from about 5 mg to about 50 mg) or tens of milligrams (e.g., ranging in amounts from about 30 mg to about 1000 mg or from about 30 mg to about 1500 mg) of the pharmaceutical composition are administered to the subject.
  • milligrams e.g., ranging in amounts from about 5 mg to about 50 mg
  • tens of milligrams e.g., ranging in amounts from about 30 mg to about 1000 mg or from about 30 mg to about 1500 mg
  • the pharmaceutical composition comprises Compound A tosylate salt and one or more excipients, wherein the Compound A tosylate salt is in a powder form and/or the Compound A tosylate salt consists of crystalline Compound A tosylate salt.
  • the one or more pharmaceutically acceptable excipients in the pharmaceutical composition provided herein can, for instance, include any excipient known to those skil led in the art or combination thereof. Further a person of ordinary ski ll in the art would understand that while a particular excipient is described as a binder, for example, the excipient may also have other properties, such as of a disintegrant or diluent, for example.
  • the one or more excipients comprise one or more binders, diluents, carriers, disintegrants, glidants, lubricants, fillers or a combination thereof.
  • the one or more excipients comprise a binder, for example, hydroxypropy l cellulose.
  • Suitable diluents or carriers include, but are not limited to, si licified microcrystalline cel lulose, microcrystalline cellulose, dibasic calcium phosphate, partially pregelatinized starch, lactose, sucrose, mannitol and mixtures thereof.
  • a diluent or carrier is preferrably present from about 75% to about 99.5% by weight of the final composition.
  • Suitable disintegrants include, but are not limited to, silicified microcrystalline cellulose, microcrystalline cellulose, crosearmellose sodium, starch, crospovidone, sodium starch glycolate, partially pregelatinized starch and m ixtures thereof.
  • a disintegrant can, for example, be present from about 1% to about 10% by weight of the final composition.
  • Suitable binders include, but are not limited to, hydroxypropyl cellulose, hypromellose, low substituted hydroxypropyl cellulose, povidone, copovidone, pregelatinized starch, partially pregelatinzed starch, and mixtures thereof.
  • a binder can, for example, be present from about 0.1% to about 10% by weight of the final composition.
  • Suitable glidants include, but are not limited to, colloidal silicon dioxide and talc.
  • Suitable lubricants include, but are not limited to, magnesium stearate, calcium stearate, sodium lauryl sulfate, stearic acid and sodium stearyl fumarte.
  • a lubricant can, for example, be present from about 0.1% to about 5% by weight of the final composition.
  • an excipient is a lubricant (e.g.. sodium stearyl fumarate, magnesium stearate, stearic acid), a binder (e.g., hydroxypropyl cellulose, low substituted HPC, hypromellose, maltodextrin, gelatin, polyethylene glycol, pregelatinized starch, partially pregelatinized starch), a glidant (e.g., talc, fumed silica, colloidal silicon dioxide, magnesium carbonate), a diluent (e.g.
  • a given material need not be limited to being one type of excipient.
  • a disintegrant e.g. , crospovidone, croscarmellose sodium, sodium starch glycol ate, alginic acid, starch, partially pregelatinized starch
  • a flavoring agent e.g., a sweetener, a coloring agent
  • a preservative e.g., an antioxidant, antimicrobial or other types of preservative including those such as vitamin A, vitamin E, vitamin C, cysteine, citric acid; methyl paraben, propyl paraben
  • a surfactant emulsifier, a sorbent, or a combination of any of the above. It will be understood that a given material need not be limited to being one type of excipient.
  • Suitable lubricants include, but are not limited to, calcium stearate; magnesium stearate; sodium stearyl fumarate; mineral oil; l ight mineral oil; glycerin; sorbitol; mann itol; glycols, such as glycerol behenate and polyethylene glycol (e.g., PEG400 and PEG6000); stearic acid; sodium lauryl sulfate; talc; hydrogenated vegetable oil, including peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, corn oil, and soybean oil; zinc stearate; ethy l oleate; ethyl laureate; agar; starch; lycopodium; silica (si licone dioxide) or silica gels, such as Aerosil® 200 (W.R. Grace Co., Baltimore, MD) and CAB- O-SIL® (Cabot Co. of Boston, MA); and mixtures thereof.
  • the pharmaceutical
  • Binders impart cohesiveness to a tablet to ensure the tablet remains intact after compression.
  • Suitable binders include, but are not limited to, starches, such as corn starch, potato starch, partially pregelatinized starch (e.g., Starch 1500) and pregelatinized starch; gelatin; sugars, such as sucrose, glucose, dextrose, molasses, and lactose; natural and synthetic gums, such as acacia, alginic acid, alginates, extract of Irish moss, panwar gum, ghatti gum, mucilage of isabgol husks,
  • carboxymethylcellulose methylcellu lose, polyvinylpyrrolidone (PVP), copovidone, larch arabogalactan, powdered tragacanth, and guar gum; magnesium aluminum silicate (e.g., Veegum*); celluloses, such as ethyl cel lulose, cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethyl cellulose, methyl cellulose, hydroxyethylcellulose (HEC), hydroxypropy (cellulose (HPC), hypromellose;
  • m icrocrystalline celluloses such as Avicel ® PH- 101 , Avicel* PH- 103, Avicel ® RC-58 1 . Avicel * PH- 105 (FMC Corp., Marcus Hook, PA); and mixtures thereof.
  • Suitable diluents include, but are not limited to, dibasic calcium phosphate, calcium sulfate, lactose, sorbitol, sucrose, inositol, cellulose, kaolin, mannitol, sodium chloride, dry starch, and powdered sugar.
  • Certain diluents, such as mannitol, xylitol, lactose, sorbitol, sucrose, and inositol when present in sufficient quantity, can impart properties to some compressed tablets that permit disintegration in the mouth by chewing. Such compressed tablets can be used as chewable tablets.
  • Suitable fillers include, but are not limited to, talc, calcium carbonate, macrocrystalline cellulose, powdered cellulose, dextrates, kaolin, mannitol, silicic acid, sorbitol, starch, pre-gelatinized starch, and mixtures thereof.
  • Suitable disintegrants include, but are not limited to, agar; bentonite; celluloses, such as methylcellulose and carboxymethylcel lulose; wood products; natural sponge; cation-exchange resins; alginic acid; gums, such as guar gum; citrus pulp; cross-linked celluloses, such as croscarmellose; cross- linked polymers, such as crospovidone; cross-linked starches; calcium carbonate; microcrystalline cellulose; modified cr ss-linked starch derivatives such as sodium starch glycolate; polacrilin potassium; starches, such as corn starch, potato starch, tapioca starch, and partially pregelatinized starch; clays, such as Veegum ® and VAN GEL ® ; aligns; and mixtures thereof.
  • the amount of a disintegrant in the pharmaceutical compositions provided herein varies upon the type of formulation, and is readily discernible to those of ordinary skil l in the art.
  • the pharmaceutical compositions provided herein may contain from about 0.5 to about 15% or from about 1 to about 10% by weight of a disintegrant.
  • Suitable gl idants include colloidal silicon dioxide, CAB-O-SIL ® (Cabot Co. of Boston,
  • Coloring agents include any of the approved, certified, water soluble FD&C dyes, and water insoluble FD&C dyes suspended on alumina hydrate, and color lakes and mixtures thereof.
  • a color lake is the combination by adsorption of a water-soluble dye to a hydrous oxide of a heavy metal, resulting in an insoluble form of the dye.
  • Flavoring agents include natural flavors extracted from plants, such as fruits, and synthetic blends of compounds which produce a pleasant taste sensation, such as peppermint and methyl salicylate.
  • Sweetening agents include sucrose, lactose, mannitol, syrups, glycerin, and artificial sweeteners, such as saccharin and aspartame.
  • Suitable emulsifying agents include gelatin, acacia, tragacanth, bentonite, and surfactants, such as polyoxyethylene sorbitan monooleate (e.g., Tween* 20), poloxamers (e.g., Pluronic ® F68), polyoxyethylene sorbitan monooleate 80 (e.g., T ween 8 80), and triethanolamine oleate.
  • surfactants such as polyoxyethylene sorbitan monooleate (e.g., Tween* 20), poloxamers (e.g., Pluronic ® F68), polyoxyethylene sorbitan monooleate 80 (e.g., T ween 8 80), and triethanolamine oleate.
  • Suspending and dispersing agents include sodium
  • carboxymethylcellulose pectin, tragacanth, Veegum 8 , acacia, sodium carbomethylcel lulose, hypromellose, polyvinylpyrrolidone, and lauroyl polyoxylglycerides (e.g., Gelucire ® 44/14).
  • Preservatives include glycerin, methyl and propylparaben, benzoic add, sodium benzoate and alcohol.
  • Wetting agents include propylene glycol monostearate, sorbitan monooleate. diethylene glycol monolaurate, and polyoxyethylene lauryl ether.
  • Solvents include glycerin, sorbitol, ethyl alcohol, and syrup. Examples of non-aqueous liquids uti l ized in emulsions include mineral oil and cottonseed oil.
  • Organic acids include citric and tartaric acid.
  • Sources of carbon dioxide include sodium bicarbonate and sodium carbonate.
  • the excipient is silicified microcrystalline cellulose (SMCC), for example.
  • SMCC silicified microcrystalline cellulose
  • Prosolv SMCC 50, Prosolv SMCC 90, or Prosolv SMCC HD90 is silicified microcrystalline cellulose
  • the excipient comprises SMCC and HPC.
  • a pharmaceutical composition provided herein comprises from about 0.5 % to about 10 % HPC, from about 0.8 % to about 5 % HPC, or from about 0.9 % to about 3 % HPC.
  • the pharmaceutical composition comprises from about 50 % to about 99.5 % SMCC, from about 75 % to about 99.5 % SMCC, or about 90 % to about 98 % SMCC.
  • the percentages herein refer to weight/weight percentages of the given excipient in the pharmaceutical composition, excluding, for instance, any coating if in a tablet form, or shell of a capsule, if in a capsule.
  • the pharmaceutical composition provided herein consists essentially of Compound A tosylate salt and SMCC.
  • the pharmaceutical composition consists essentially of Compound A tosylate salt, SMCC and HPC.
  • the pharmaceutical composition consists essentially of Compound A tosylate salt, SMCC, optionally, HPC, and if the pharmaceutical composition is in a tablet form, a tablet coating.
  • the pharmaceutical composition consists essentially of Compound A tosylate salt, SMCC, optionally, HPC, and if the pharmaceutical composition is in a capsule, a capsule shell.
  • a pharmaceutical composition provided herein can be a dry blend formulation, for example, prepared according to the dry blend methods described herein.
  • a pharmaceutical composition provided herein can be a prepared using a wet granulation method, for example, in accordance with the wet granulation methods described herein. Dry blend methods and wet granulation methods are described herein.
  • compositions comprising Compound A tosylate salt and one or more pharmaceutically acceptable excipient(s), where the pharmaceutical composition can be any of those provided in this application.
  • the methods provided include, for example, a dry blend method, and, as another example, a wet granulation method.
  • the method for preparing a pharmaceutical composition is a dry blend method wherein crystalline Compound A tosylate salt is geometrically blended with silicified microcrystalline cel lulose.
  • a dry blend method comprises mixing crystalline Compound A tosylate salt and a first fraction of silicified microcrystalline cel lulose to produce a first mixture; mixing the first mixture and a second fraction of silicified microcrystalline cellulose to produce a second mixture, which is then mixed with a third fraction of silicified microcrystalline cellulose to produce a pharmaceutical composition comprising a weight/weight percent of Compound A tosylate salt between about 0.01 % to about 4.000 %, or as described elsewhere herein.
  • An exemplary dry blend method for preparing a pharmaceutical composition comprises geometric blending Compound A tosylate salt with SMCC.
  • the method comprises (a) mixing crystal line Compound A tosylate salt and a first fraction of SMCC to produce a first mixture; (b) mixing the first mixture and a second fraction of SMCC; and (c) mixing the mixture produced in step (b) and a third fraction of SMCC to produce the pharmaceutical composition.
  • the mixture produced in step (b) is geometrical ly blended with SMCC prior to being mixed in step (c).
  • a fourth fraction of SMCC is added to the mixture, which is mixed to produce a mixture for use in step (c).
  • a fifth fraction of SMCC is added to the mixture and mixed to produce a mixture for use in step (c).
  • Any number of additional fractions e.g., a sixth fraction, a seventh fraction, an eighth fraction, a ninth fraction, a tenth fraction, etc. can be added and mixed to produce a mixture for use in step (c).
  • fractions of SMCC added in any step of the method can be identical amounts, or can be different amounts, or can be the same in some cases and different in others (e.g., the first and third fractions can be the same, while the second fraction is different from the first and third fractions).
  • an excipient other than SMCC is added during one of the steps (for example, step (a), step (b) or step (c)) of the dry blending method and mixed together with the Compound A tosylate salt and SMCC.
  • excipients can, for example, be any of those known in the art and/or provided herein.
  • the weight/weight percent of Compound A tosylate salt in the pharmaceutical composition prepared according to the dry blend method provided herein can, for instance, be within the ranges described herein.
  • the dry blend method for preparing a pharmaceutical composition comprises:
  • step (b) mixing the first mixture and a second fraction of silicified microcrystalline cellulose; and c) mixing the mixture produced in step (b) and a third fraction of silicified microcrystal line cellulose to produce a pharmaceutical composition comprising a weight/weight percent of Compound A tosylate salt between about 0.01 % to about 4.000 %.
  • the method for a pharmaceutical composition is a wet granulation method wherein Compound A tosylate salt is subject to high shear granulation using si licified microcrystalline cellulose as a carrier.
  • one embodiment comprises dissolving crystalline Compound A tosylate salt and a binder in a solvent and mixing the resulting solution in a high shear granulator with silicified microcrystalline cellulose, and then dr ing the granules to produce a to produce a pharmaceutical composition comprising, for example, between about 0.01 % to about 4.000 %
  • An exemplary wet granulation method comprises dissolving crystall ine Compound A tosylate salt and a binder in a first volume of solvent to produce a solution; mixing the solution in a high shear granulator with SMCC to produce granulated material; and drying the granulated material to produce a to produce a pharmaceutical composition.
  • the binder is hypromellose or
  • HPC HPC.
  • the binder is HPC.
  • the weight/weight percent of HPC in the pharmaceutical composition is about 0.5 % to about 2.0 %.
  • the solvent is acetone, ethanol, ethyl acetate or water, or a co-solvent selected from acetone/water, ethanol/water and ethyl acetate/water. In certain embodiments, the solvent is a co-solvent of acetone/water.
  • the weight/weight ratio of acetone to water is 9: 1.
  • the first volume comprises about 95 % to about 98 %
  • the weight/weight percent of crystalline Compound A tosylate salt in the pharmaceutical composition prepared according to the wet granulation method provided herein can, for instance, be within the ranges described herein.
  • the pharmaceutical composition prepared according to the wet granulation method provided herein comprises between about 0.10 % to about 1.0 % Compound A tosylate salt and about 0.5 % to about 2.0 % binder.
  • the wet granulation method for preparing a pharmaceutical composition comprises:
  • step (a) dissolving crystal line Compound A tosylate salt and a binder in a first volume of solvent; b) mixing the solution of step (a) in a high shear granulator with silicified macrocrystall ine cellulose; and
  • Unit dosage forms can, for instance, comprise Compound A tosylate salt in solid form and one or more pharmaceutically acceptable excipients.
  • the unit dosages form provided herein is a capsule. In other embodiments, it is a tablet.
  • the unit dosage form contains a mass of, for instance, about 25 ⁇ g, about 50 ⁇ ag, about 250 g or about 1 mg Compound A, wherein said mass excludes the tosylate portion of the salt.
  • unit dosage forms typically comprise a pharmaceutical composition comprising Compound A tosylate salt and one or more pharmaceutically acceptable excipients, where the pharmaceutical composition can be any of those as prov ided herein.
  • Unit dosage forms as provided herein can, for example, be suitable for oral
  • Unit dosage forms can, for example, be a capsule. Other examples include those where the unit dosage form is a tablet.
  • a unit dose form provided herein is in a tablet, wherein the pharmaceutical composition is prepared by a wet granulation method.
  • the amount of Compound A tosylate salt in a unit dosage form can, for example, be a low dose, such as defined herein.
  • the amount of Compound A tosylate salt in the unit dosage form is in a range from about 1 ⁇ g to about 8000 ⁇ g, from about 5 ⁇ to about 1500 g, from about 20 to about 1250 ⁇ g, from about 25 ⁇ to about 1000 pg, from about 25 g to about 1500 ⁇ g.
  • the amount of Compound A tosylate salt in the unit dosage form is about 10 g, about 20 g, about 25 ug. about 30 g, about 50 g, about 75 ⁇ g, about 100 ⁇ , about 150 ⁇ , about 200 ⁇ g, about 250 ⁇ g, about 1000 ⁇ g, about 1250 ⁇ , or about 1 500 ⁇ g.
  • Unit dosage forms provided herein may be provided in capsules, including, for instance, soft or hard capsules.
  • Capsules can, for example, be prepared according to methods known to the skilled artisan using materials known to the skilled artisan.
  • Material for the capsules can include, for example, gelatin, pullulan, methylcellulose, starch, calcium alginate or hypromellose.
  • the capsule shel l is hypromellose.
  • Encapsulating the pharmaceutical compositions provided herein can, for instance, be performed using the Xcleodose automated encapsulator system (Capsugel, Peapack, New Jersey, USA).
  • the hard gelatin capsule also known as the dry-filled capsule (DFC)
  • the soft elastic capsule is a soft, globular shell, such as a gelatin shel l, which is plasticized by the addition of glycerin, sorbitol, or a similar polyol.
  • the soft gelatin shel ls may contain a preservative to prevent the growth of microorganisms. Suitable preservatives are those as described herein, including methyl- and propyl-parabens, and sorbic acid.
  • the capsules may also be coated as known by those of ski l l in the art in order to modify or sustain dissolution of the active ingredient.
  • Tablets can, for instance, be formed according to methods known to the skilled artisan.
  • Tablets may be provided as compressed tablets, tablet triturates, chewable lozenges, rapidly dissolving tablets, multiple compressed tablets, or enteric-coating tablets, sugar-coated, or film-coated tablets.
  • Multiple compressed tablets are compressed tablets made by more than one compression cycle, including layered tablets, and press-coated or dry-coated tablets.
  • Enteric-coated tablets are compressed tablets coated with substances that resist the action of stomach acid but dissolve or disintegrate in the intestine, thus protecting the active ingredients from the acidic environment of the stomach.
  • Enteric-coatings include, but are not limited to, fatty acids, fats, phenyl salicylate, waxes, shellac, ammoniated shellac, and cellulose acetate phthalates.
  • Sugar-coated tablets are compressed tablets surrounded by a sugar coating, which can be beneficial in covering up objectionable tastes or odors and in protecting the tablets from oxidation.
  • Film-coated tablets are compressed tablets that are covered with a thin layer or film of a water-soluble material.
  • Film coating imparts the same general characteristics as sugar coating.
  • Film coatings include polymeric coating materials, plasticizers, surfactants, and colorants.
  • Suitable polymeric coating materials include, but are not limited to, hydroxyethylcellulose, sodium carboxymethylcellulose, hypromellose, polyvyl, hydroxypropyl cellulose, hydroxyethyl methyl cellulose, polyethylene glycol 4000, and cellulose acetate phthalate.
  • Suitable plasticizers include, but are not limited to glycerol, propylene glycol, polyethylene glycol, dibutyl sebacete, citrate esters, and triacetin.
  • Suitable surfactants include, but are not limited to polysorbate 80 and sodium I aur I sulfate.
  • Suitable colorants include various iron oxides, FD&C blue no.2, and titanium dioxide.
  • Ingredients in the film coating composition are dispersed in a suitable solvent, preferrably water, and applied onto capsules or tablets using a conventional spray
  • the tablet dosage forms may be prepared from Compound A tosylate salt in solid form, for instance, in a crystalline form, in combination with one or more excipients described herein, including binders, disintegrants, controlled-release polymers, lubricants, diluents, and/or colorants. Flavoring and sweetening agents are especially useful in the formation of chewable tablets and lozenges
  • Tablets can, for example, be made according to techniques known to the skilled artisan.
  • Tablet presses are commercially available, for instance, from Korsch AG (Berlin, Germany), Oystar Manesty (Merseyside, England), ikusui Seisakusho Ltd. (Kyoto, Japan), Elizabeth-Hata International (North Huntingdon, PA, USA), Gea Pharma Systems Courtoy (Wommelgem, Belgium ), Ima Kill ian (Scarletallee, Germany), among others. Tablet coating equipment are commerically available, for instance, from O'Hara (Ontario, Canada), Oystar Manesty (Merseyside, England) and Vector Corporation (Marion, IA, USA).
  • the unit dosage forms may be provided in high-density polyethylene bottles provided with a desiccant capsule or pouch; or in foil-on-foi l blister packaging, or in blister packaging comprising apolymer film.
  • Such indications that can be treated include, for example, leukemia (including acute myelogenous leukemia, chronic lymphocytic leukemia), myelodysplastic syndrome, colon cancer, EBV-associated tumors (including Burkitt's lymphoma, nasopharyngeal carcinoma, lymphomas in AIDS patients, smooth muscle tumors in AIDS patients, Hodgkin's disease, non-Hodgkin's lymphoma, lymphoproliferative disease in immunosuppressed patients, leiomyosarcomas in immunosuppressed patients, EBV+ gastric cancer, EBV+ breast cancer, T-cell lymphoma), endometrial cancers (including carcinomas, sarcomas, endometrioid serous adenocarcinoma, endometrioid clear cel l adenocarcinoma, and adenosquamous carcinoma, or mixed histology, any grade), gastrointestinal stromal tumor, gliomas, gli
  • compositions including unit dosage forms comprising low dosages of Compound A tosylate salt, are provided, which methods, without intending to be bound by any theory or mechanism, can minimize loss of Compound A and can be used to consistently achieve a desired proportion of Compound A relative to inactive ingredients.
  • the pharmaceutical composition to be administered to the subject is a pharmaceutical composition as provided herein, for example, comprising Compound A tosylate salt in solid (e.g., crystal line) form and one or more pharmaceutically acceptable excipients.
  • the pharmaceutical composition can, for instance, be in a powder form, or in a unit dosage form (e.g., capsule or tablet).
  • the pharmaceutical composition may be formulated to be suitable for oral administration to the subject.
  • a pharmaceutical composition as described herein, for the treatment of a disease or conidition, or symptom thereof, in a subject.
  • a method of treating cancer, or symptom thereof, comprising administering to a subject with cancer a therapeutically effective amount of a pharmaceutical composition, as described herein, is prov ided.
  • Certain embodiments provide a method of potentiation of cytotoxic cancer therapy in a subject in recognized need of such treatment comprising administering to the subject a therapeutically acceptable amount of a pharmaceutical composition, as described herein,.
  • a method for the treatment of cancer comprising administering to a subject in need of treatment a therapeutical ly effective amount of a pharmaceutical composition, as described herein, in combination with ionizing radiation or one or more chemotherapeutic agents.
  • the pharmaceutical composition described herein is administered simultaneously with ionizing radiation or one or more chemotherapeutic agents.
  • the pharmaceutical composition described herein is administered sequentially with ionizing radiation or one or more chemotherapeutic agents. Ion izing radiation and chemotherapeutic agents are known to those skilled in the art.
  • the therapeutic agent(s) is an alkylating agent, such as methyl methanesulfonate (MMS), temozolomide and dacarbazine (DTIC); a topoisomerase-1 inhibitor such as Topotecan, Irinotecan. Rubitccan. Exatecan, Lurtotecan. Gimetecan, Diflomotecan
  • alkylating agent such as methyl methanesulfonate (MMS), temozolomide and dacarbazine (DTIC); a topoisomerase-1 inhibitor such as Topotecan, Irinotecan. Rubitccan. Exatecan, Lurtotecan. Gimetecan, Diflomotecan
  • cetuximab platinum-based compounds such as cisplatin, carboplatin, oxaliplatin, and triplatin tetranitrate; c ladribine;. daunorubicin; doxorubicin; idarubicin; fludarabine; 5-fluorouracil; gemtuzumab;
  • methotrexate Pacl itaxelTM; taxol; temozolomide; thioguanine; hormone therapies such as an antiestrogen, an antiandrogen, and gonadotropin releasing hormone analogues; interferons such as alpha interferon; nitrogen mustards such as busulfan, melphalan, and mechlorethamine; retinoids such as tretinoin; tyrosine kinase inhibitors such as gefinitinib and imatinib; a proteasome inhibitor such as bortezomib; or agents to treat signs or symptoms induced by such therapy including allopurinol, filgrastim,
  • a method for the treatment of a cancer comprising administering to a subject in need of treatment a pharmaceutical composition as described herein in combination with a topoisomerase inhibitor.
  • the topoisomerase inhibitor is SN38, irinotecan, or NK.012.
  • the topoisomerase inhibitor is irinotecan.
  • the cancer is breast cancer.
  • a method for the treatment of a cancer comprising administering to a subject in need of treatment a pharmaceutical composition as described herein in combination with a platin.
  • the plat in is cisplatin, carboplatin, oxaliplatin, or triplatin tetranitrate.
  • the topoisomerase inhibitor is cisplatin.
  • the cancer is breast cancer.
  • a method for the treatment of a cancer comprising administering to a subject in need of treatment a pharmaceutical composition as described herein in combination with temozolomide.
  • the cancer is colorectal cancer.
  • provided herein is a method of treatment of a cancer deficient in
  • the cancer includes one or more cancer cells having a reduced or abrogated abil ity to repair DNA DSB by HR relative to normal cells.
  • the cancer cells have a BRCA 1 or BRCA2 deficient phenotype.
  • the cancer cells are deficient in BRCA 1 or BRCA2.
  • the methods provided herein involve treatment of an individual who is heterozygous for a mutation in a gene encoding a component of the HR dependent DNA DSB repair pathway.
  • the individual is heterozygous for a mutation in BRCA 1 and/or BRCA2.
  • the cancer to be treated comprises phosphatase and tensin homolog (PTEN) deficient cel ls (e.g., cel ls in which PTEN is mutated or its expression is minimal or absent).
  • PTEN tensin homolog
  • the cancer to be treated comprises cel ls with a PTEN gene mutation.
  • Exemplary cancers associated with PTEN deficiency can include, for example, glioblastoma, endometrial cancer, prostate cancer, lung cancer and breast cancer.
  • the cancer to be treated is one resulting from an activation mutation of the Wnt signaling pathway ("Wnt mediated cancers”)- It will be understood that by
  • activation mutation of the Wnt signal ing pathway it is meant to include, for example, oncogene mutations in genes leading to accumulation of ⁇ -catenin in cancer cells, gain-of-function mutations in the CTNNB 1 gene (which encodes ⁇ -catenin), mutations in the APC tumor suppressor gene or mutations in the AXIN2 gene. It is believed, for instance, and without intending to be limited to any theory or mechanism, that useful treatment of a Wnt mediated cancer may be had by way of inhibiting tankyrase ("TNK.S”), an enzyme having poly(ADP-ribose) polymerase activity.
  • TNK.S tankyrase
  • Exemplary Wnt mediated cancers that can be treated according to the methods provided herein include, for instance, bladder cancer, breast fibromatoses, cervical cancer, colorectal cancer, colon carcinoma, Desmoid tumor, esophageal adenocarcinoma, familial adenomatous polyposis, fundic gland polyps, gastric carcinoma, gastric adenoma, gastrointestinal carcinoid tumor, hepatoblastoma, hepatocellular carcinoma, juvenile nasopharyngeal angiofibroma, non-Hodgkin lymphoma, lung adenocarcinoma, medulloblastoma, melanoma, ovarian carcinoma, pancreatic cancer (including, for example, non-ductal solid
  • pseudopapil lary and non-ductal acinal cell carcinoma pancreoblastoma, pilomatricomas, prostate cancer, small intestinal adenocarcinoma, synovial sarcoma, thyroid carcinoma, uterine cervical cancer, uterine endometrial cancer and Wilm's tumor.
  • the cancer is bladder cancer, breast cancer (including metastatic, BRCA-positive, and BRCA-negative), cervical cancer, colon cancer, colorectal cancer, EBV-associated tumors (including Burkitt's lymphoma, nasopharyngeal carcinoma, lymphomas in AIDS patients, smooth muscle tumors in AIDS patients, Hodgkin's disease, non-Hodgkin's lymphoma, lymphoproliferative disease in immunosuppressed patients, leiomyosarcomas in immunosuppressed patients, EBV+ gastric cancer, E V r breast cancer, T-cell lymphoma), endometrial cancers (including carcinomas and sarcomas), gastrointestinal stromal tumor, gliomas, glioblastoma (including, for instance, gl ioblastoma multiforme and anaplastic astrocytoma), head and neck cancer,
  • EBV-associated tumors including Burkitt's lymphoma, nasopharynge
  • a method of treating a disease or condition, or symptom thereof, associated with a PTEN deficiency comprising administering to a subject with the disease or condition associated with a PTEN deficiency a therapeutically effective amount of a pharmaceutical composition, as described herein.
  • exemplary diseases and conditions associated with a PTEN deficiency include, for example, Cowden syndrome, Bannayan-Riley-Ruvalcaba syndrome, Lhermitte-Duclos disease, Proteus syndrome, Proteus-like syndrome or hamatome tumor syndrome.
  • Certain embodiments provide a method of treating inflammatory diseases including, but not limited to, arthritis, gout, inflammatory bowel disease, CNS inflammation, multiple sclerosis, allergic encephalitis, sepsis, septic shock, hemorrhagic shock, pulmonary fibrosis, and uveitis in a subject in recognized need of such treatment comprising administering to the subject a therapeutically acceptable amount of the pharmaceutical composition.
  • inflammatory diseases including, but not limited to, arthritis, gout, inflammatory bowel disease, CNS inflammation, multiple sclerosis, allergic encephalitis, sepsis, septic shock, hemorrhagic shock, pulmonary fibrosis, and uveitis
  • Certain embodiments provide a method of treating immunological diseases or disorders such a rheumatoid arthritis and septic shock in a subject in recognized need of such treatment comprising administering to the subject a therapeutically acceptable amount of the pharmaceutical composition.
  • Ceri in embodiments provide a method of treating degenerative diseases including, but not limited to, diabetes and Parkinson's disease in a subject in recognized need of such treatment comprising administering to the subject a therapeutically acceptable amount of the pharmaceutical composition.
  • the precise amount of Compound A tosylate salt administered to the subject in the pharmaceutical composition depends on the patient's state of health, weight, and the l ike. In some embodiments, it is considered appropriate for the caregiver to determine such therapeutical ly 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.
  • the pharmaceutical composition is administered to the subject in a regimen of about 1 week, about 2 weeks, about 3 weeks, about 1 month, about 2 months or about 6 months.
  • the tosylate salt is administered daily in the regimen.
  • the tosylate salt is administered for 2, 3 or 4 days per week, interspersed or fol lowed by days of the weekly regimen where the tosylate salt is not administered.
  • a patient ' s condition does not improve or does not significantly improve following administration of the pharmaceutical composition, and, upon the doctor's discretion, the administration of the compound 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 administration of the active ingredient is 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").
  • the length of the drug holiday v aries 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, 150 days, 180 days, 200 days, 250 days, 280 days, 300 days, 320 days, 350 days, or 365 days.
  • the amount of Compound A tosylate salt that corresponds to an effective amount of the pharmaceutical composition 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 administration, the condition being treated, and the subject or host being treated.
  • doses employed for adult human treatment is in the range of about 1 to about 8000 ⁇ g per day, in a specific embodiment about 10 to about 5000 ⁇ g per day.
  • the amount of Compound A tosylate salt to be administered is between about 50 ⁇ g to about 5000 between about 50 ⁇ to about 1 500 ⁇ g, between about 50 g to about 1000 ⁇ g, between about 50 ⁇ g to about 500 iig, between about 50 g to about 250 g, between about 50 g to about 200 ⁇ g, between about 50 ⁇ g to about 150 ⁇ g, between about 50 ⁇ g to about 100 ⁇ g, between about 25 ⁇ g to about 2500 ⁇ g, between about 25 ⁇ g to about 1000 g, between about 25 ug to about 250 ⁇ , between about 25 ⁇ g to about 150 ⁇ g, or between about 25 ⁇ g to about 75 ⁇ g.
  • 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 daily dosages of Compound A tosylate salt in the pharmaceutical composition appropriate for the methods described herein are from about 0.01 to about 100 ⁇ /kg per body weight. It wi l l be understood that these amounts refer specifical ly to the mass of Compound A, excluding the mass contributed by the tosylate portion of Compound A tosylate salt, or contributed by any excipient in the pharmaceutical composition.
  • an indicated daily dosage in the larger subject including, but not limited to, humans, is in the range from about of about 25 to about 8000 ⁇ , 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 those described herein.
  • 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.
  • the pharmaceutical composition is as described herein and in another example comprises one or more pharmaceutical ly acceptable carrier, excipient(s), binder and/or diluent.
  • the compound of Formula I is ( 8.S',9/?)-5 - fl uoro-8 -( 4- nuorophenyl)-9-( l -methyl- 1 //- l ,2,4-triazol-5-y
  • a method of treating multiple myeloma comprising administering to a subject in need thereof a pharmaceutical combination which comprises a
  • proteasome inhibitor as defined in the Summary of the Invention or in any of the embodiments described herein (or a single isomer, stereoisomer, or enantiomer or mixture thereof, optionally as a pharmaceutically acceptable salt, pharmaceutically acceptable solvate, or pharmaceutically acceptable prodrug thereof) and a proteasome inhibitor.
  • the proteasome inhibitor is bortezomib, disulfiram, epigallocatechin-3-gallate, salinosporamide A (marizomib), carfilzomib, ONX 0912 (oprozomib), CEP-
  • the proteasome inhibitor is bortezomib.
  • the compound of Formula I is (8S,9R)-5-fluoro-8-(4-fluorophenyl)-9-( l -methyl- I H- l , 2,4-triazol-5-yl)-8,9-dihydro-2/ - pyrido[4,3,2-ife]phthalazin-3(7H)-one or a pharmaceutically acceptable salt thereof.
  • the Compound of Formula (IA), (IA l ), (IA2), (II), (IIA), (IIB), (IIC), (IID) as defined in the Summary of the Invention or in any of the embodiments described herein (or a single isomer, stereoisomer, or enantiomer or mixture thereof, optionally as a pharmaceutically acceptable salt, pharmaceutically acceptable solvate, or pharmaceutically acceptable prodrug thereof) is used as a pharmaceutical composition, as described herein, additionally comprising one or more pharmaceutical ly acceptable carrier, excipient(s), binder and/or diluent.
  • a method of treating multiple myeloma with a pharmaceutical composition comprising 1 ) a compound, optionally as a pharmaceutically acceptable salt thereof, of Formula (I), (IA), (IA l ), (IA2), (II), (IIA), (IIB), (IIC), or (IID) as defined in the Summary of the Invention or in any of the above embodiments (or a single isomer, stereoisomer, or enantiomer or mixture thereof, optionally as a pharmaceutically acceptable salt, pharmaceutically acceptable solvate, or pharmaceutically acceptable prodrug thereof) and 2) one or more pharmaceutically acceptable carriers), excipient(s), binder or diluent(s) thereof.
  • a pharmaceutically acceptable salt thereof of Formula (I), (IA), (IA l ), (IA2), (II), (IIA), (IIB), (IIC), or (IID) as defined in the Summary of the Invention or in any of the above embodiments (or a single iso
  • the compound of Formula I is (8S,9i?)-5- fluoro-8-(4-fluorophenyl)-9-( 1-methyl- 1 H- 1 ,2,4-triazol-5-yl)-8,9-dihydro-2H-pyrido[4,3,2- ⁇ ]phthalazin- 3(7H)-one or a pharmaceutically acceptable salt thereof.
  • a method of treating multiple myeloma comprising administering to a subject in need of treatment a therapeutically effective amount of a compound of Formula (I), (IA), (IA1), (IA2), (II), (IIA), (I IB), (HQ, or (IID) as defined in the Summary of the Invention or in any of the above embodiments (or a single isomer, stereoisomer, or enantiomer or mixture thereof, optionally as a pharmaceutically acceptable salt, pharmaceutically acceptable solvate, or pharmaceutically acceptable prodrug thereof).
  • the compound of Formula I is (8.
  • the disease is mediated by poly(ADP-ribose)polymerase.
  • the compound of Formula I is (8S',9R)-5-fluoro-8-(4-fIuorophenyl)-9-( I -methyl- 1 //- l,2,4-triazol-5-y l)-8,9- dihydro-2if-pyrido[4,3,2- e]phthalazin-3(7/i)-one or a pharmaceutically acceptable salt thereof.
  • the compound of Formula I is (8S,9jR)-5-fluoro-8-(4-fluorophenyl)-9-( l -methyl- l /-l,2,4-triazol-5-yl)-8,9-dihydro-2H-py i e] phthal azin-3 (7H)-one or a pharmaceutically acceptable salt thereof.
  • an article of manufacture comprising packaging material, a compound of Formula (I), (IA), (IA 1 ), (IA2), (II). (IIA), (IIB). (IIC), or (IID) as defined in the Summary of the Invention or in any of the above embodiments (or a single isomer, stereoisomer, or enantiomer or mixture thereof, optionally as a pharmaceutically acceptable salt, pharmaceutically acceptable solvate, or pharmaceutically acceptable prodrug thereof), optionally a proteasome inhibitor, and a label, wherein the compound is effective for modulating the activity of the enzyme poly(ADP-ribose)po!ymerase, or for treatment, prevention or amelioration of one or more symptoms of multiple myeloma, wherein the compound is packaged within the packaging material, and wherein the label indicates that the compound, or pharmaceutically acceptable salt, pharmaceutically acceptable N-oxide, pharmaceutically active metabolite, pharmaceutically acceptable prodrug, or pharmaceutically acceptable solvate
  • the proteasome inhibitor when present, is bortezomib.
  • the compound of Formula I is (85,9R)-5- tluoro-8-(4-fluorophenyl)-9-( 1 -methyl- 1 //- 1.2,4-triazol-5-yl)-8.9-dihydrcv2//-pyrido[4,3,2 /£'lphthalazin- 3(7H)-one or a pharmaceutically acceptable salt thereof.
  • Certain embodiments provide a use of a compound of Formula (I), (IA), (IA 1 ), (IA2),
  • the compound of Formula I is (8S,9R)-5-fluoro-8-(4- fluorophenyl)-9-( l -methyl-lH-l ,2,4-triazol-5-yI)-8,9-dihydro-2H-pyridot4,3,2-Je]phthalazin-3(7H)-one or a pharmaceutically acceptable salt thereof.
  • a pharmaceutical composition comprising a compound, pharmaceutically acceptable salt, pharmaceutically acceptable N-oxide, pharmaceutically active metabol ite, pharmaceutically acceptable prodrug, or pharmaceutically acceptable solvate of any of the compounds disclosed herein.
  • the pharmaceutical compositions further comprises one or more pharmaceutically acceptable excipient(s).
  • the pharmaceutical composition further comprises a second pharmaceutically active ingredient.
  • the effective amount of the compound is administered by ophthalmic administration;
  • the effective amount of the compound is adm inistered rectal ly to the subject.
  • 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; (i i) 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: (i) the compound is administered in a single dose;
  • 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.
  • 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, etho y, propoxy, 2-propoxy, butoxy, tert-butoxy, pentyloxy, and hexyloxy.
  • alky means a straight, branched chain, or cyclic (in this case, it would also be known as "cycloalkyl”) hydrocarbon containing from I - 10 carbon atoms.
  • alkyl include, but are not l imited 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-dirnethylhexyl, n-heptyl, n-octyl, n-nonyl, and n-decyl.
  • Ci-Q-alkyl as used herein, means a straight, branched chain, or cycl ic (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 monocycl ic 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 examples of
  • a cycloalkyl group is a monoradical or a di successionl (e.g., a cyc loalkylene group).
  • cycloalkyl groups refers to groups which are optionally substituted with 1 , 2, 3, or 4 substituents selected from alkenyl, alkoxy, alkoxyalkyi, alkoxycarbonyl, alkyl, alkylcarbonyl, alkylcarbonyloxy, alkylthio, alkylthioalkyl, alkynyl, carboxy, cyano, formyl, haloalkoxy, haloalkyl, halogen, hydroxy I, hydroxyalkylene, mercapto, oxo, -NR ⁇ R D . and
  • cycloalkylalkyl 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-cyclobutylethyI, cyclopentylmethyl, cyclohexylmethyl, and 4-cyc lohepty Ibuty I .
  • 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.
  • alkoxyalkyi means at least one alkoxy group, as defined herein, appended to the parent molecular moiety through an alkyl group, as defined herein.
  • alkoxyalkyi 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 as used herein, means an alkoxycarbonyl group, as defined herein, appended to the parent molecular moiety through an alkyl group, as defined herein.
  • alkylcarbonyl as used herein, means an alkyl group, as defined herein, appended to the parent molecular moiety through a carbonyl group, as defined herein.
  • alkylcarbonyl include, but are not limited to, acetyl, 1 -oxopropyl, 2,2-d imethy 1- 1 -oxopropy I, 1-oxobutyl, and 1 -oxopentyl.
  • alkylcarbonyloxy means an alkylcarbonyl 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-butylcarbonyloxy.
  • 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.
  • alkylthioalkyi means an alkylthio group, as defined herein, appended to the parent molecular moiety through an alkyl group, as defined herein.
  • alkylthioalkyi 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.
  • alkynyl groups are optionally substituted.
  • 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, naphthalcnyl, 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, alkoxy, alkoxyalkyl, alkoxycarbonyl, alkyl, alkylcarbonyl, alkylcarbonyloxy, alkylthio, alkylthioalkyi, alkynyl, carbonyl, cyano, formyl, haloalkoxy, haloalkyl, halogen, hydroxyl, hydroxyalkylene, mercapto, nitro, -N RC D, and -C(0)(NR C RD)-
  • arylalkyi 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 arylalkyi include, but are not limited to benzyl, 2-phenylethyl, -phenylpropyl, l -methyl-3-phenylpropyl, and 2- naphth-2-ylethyl.
  • halo or halogen as used herein, means a -CI, -Br, -I or -F.
  • mercapto as used herein, means a -SH 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 and haloalkoxy include alkyl 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 al l 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 haloaikyl 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 as used herein, includes optional ly substituted alkyl radical 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
  • a heteroaryl group is a monoradical or a diradical (i.e., a heteroarylene 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, haloaikyl, halogen, hydroxy!, hydroxyalkylene, mercapto, nitro, -N RCRD, and -C(0)(NRCRD).
  • 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 refers to a non-aromatic ring wherein one or more atoms forming the ring is a heteroatom.
  • a “heterocycloalkyl” or “non-aromatic heterocycie” 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.
  • heterocycloalkyis contain one or more carbonyl or thiocarbonyl groups such as, for example, oxo- and thio-containing groups.
  • heterocycloalkyis 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-oxathi in, 1 ,4-oxathiane, tetrahydro- l ,4-thiazine, 2H- l ,2-oxazine , male
  • heterocycloalkyl also includes all ring forms of the carbohydrates, including but not limited to the monosaccharides, the disaccharides and the oligosaccharides.
  • heterorocycie refers to heteroaryl and heterocycloalkyl used herein, refers to groups containing one to four heteroatoms each selected from O, S and N, wherein each heterocycie 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.
  • a heterocycie e.g., Ci-C 6 heterocycie
  • at least one other atom the heteroatom
  • C Cs heterocycie 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 heterocycie ring has additional heteroatoms in the ring. Designations such as “4-6 membered heterocycie” refer to the total number of atoms that are contained in the ring (i.e., a four, five, or six 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.
  • heterocycloalkyl groups are pyrrol idinyl, tetrahydrofuranyl, dihydrofuranyl, tctrahydrothienyl, tetrahydropyranyl, dihydropyran l, tetrahydrothiopyranyl, piperidino.
  • heteroaryl groups are pyridinyl, imidazolyl, pyrimidinyl, pyrazolyl, triazolyl, pyrazinyl, tetrazolyl, fury I, 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.
  • the foregoing groups are C-attached or -attached where such is possible.
  • a group derived from pyrrole is pyrrol- 1 - I (N-attached) or pyrrol-3-yl (C-attached).
  • a group derived from imidazole is imidazol-l -yl or imidazol-3-yl (both ⁇ '-attached) or imidazol-2-yl, imidazol-4-yl or imidazol-5-yl (all C-attached).
  • a heterocycle group is a monoradical or a diradical (i.e., a heterocyclene group).
  • heterocyc les described herein 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, -N R C R 3 ⁇ 4 and -C(0)(NRCRD)- [00241 ]
  • the term "heterocycloalkoxy” refers to a -OR group where R is -alky l-heterocyc loal ky I group, where the alkyl and heterocycloalkyl arc as defined herein, appended to the parent molecular moiety through an alkoxy group.
  • heterocycloalkylthio refers to a, -SR group where R is -alkyl-heterocycloalkyl group where the alkyl and heterocycloalkyl are 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 an -OR group where the R is heteroarylalkyl, as defined herein.
  • heteroarylalkylthio refers to an -SR group where R is heteroarylalkyl group, as defined herein.
  • 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 molecu lar moiety through a sulfur atom.
  • the term "membered ring” embraces any cyclic structure.
  • cyclohexyl, pyridine, pyran and thiopyran are 6-membered rings and cyclopentyi, pyrrole, furan, and thiophene are 5-membered rings.
  • 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.
  • non-aromatic 5, 6, 7, 8, 9, 10, 1 1, or 12- bicyclic heterocycle ring include, but are not l imited to, 2- azabicyclo[2.2.1 ]heptanyl, 7- azabicyclo[2.2.1 ]heptanyl, 2- azabicyclo[3.2.0]heptanyl, 3-azabicyclo[3.2.0]heptanyl, 4- azasp iro[2.4 ] heptan I, 5- azaspiro[2.4]heptanyl, 2-oxa-5- azabicyclo[2.2.
  • hydroxyalkylene means at least one hydroxy I group, as defined herein, is appended to the parent molecular moiety through an alkylene group, as defined herein.
  • hydroxyalkylene examples include, but not limited to hydroxy methylene, 2-hydroxy- ethylene, 3-hydroxypropylene and 4-hydroxyheptylene.
  • N R C N R D means two group, R C and R D , which are appended to the parent molecular moiety through a nitrogen atom.
  • Rc and R D are each independently hydrogen, alky I, and alky lcarbonyl.
  • Illustrative examples of RC D include, but are not limited to, amino, methylamino, acetylamino, and acetylmethylamino.
  • 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, cycloalkyi, aryl, heteroaryl (bonded through a ring carbon) and non-aromatic heterocycle (bonded through a ring carbon),
  • substituted or “substituent(s)” means that the referenced group is optionally substituted (substituted or unsubstituted) with one or more additional group(s) individually and independently selected from alkyl, cycloalkyi, aryl, heteroaryl, heteroalicyclic, hydroxy, alkoxy, aryloxy, mercapto, alkylthio, arylthio, alkylsulfoxide, arylsulfoxide, alkylsulfone, arylsulfone, cyano, halo, carbonyl, thiocarbonyi, isocyanato, thiocyanato, isothiocyanato, nitro, perhaioalkyi, perfluoroalkyl, silyl, and amino, including mono- and di-substituted amino groups, and the protected derivatives thereof.
  • DNA-damaging agent as used herein is melphalan, cyclophosphamide, bendamustine, or radiation.
  • proteasome inhibitor means an inhibitor which blocks the action of proteasomes, e.g. p53 protein, and include, but are not limited to, bortezomib, disulfiram, epigallocatechin-3-gallate, salinosporamide A (marizomib), carfilzomib. ONX 0912 (oprozomib), C EP- 1 8770 (delanzomib), and MLN9708 (ixazomib citrate); or a pharmaceutically acceptable salt thereof.
  • 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 methoxymethyl, and esters including acetyl, benzoyl, and the like.
  • ketone protecting groups include, but not limited to, ketals, oximes, O-substituted oximes for example O-benzyl oxime, O-phenylthiomethyl oxime, 1 -isopropoxycyclohex l oxime, and the like.
  • amine protecting groups include, but are not limited to, tert-butoxycarbonyl (Boc) and carbobenzyloxy (Cbz).
  • protected-hydroxy refers to a hydroxy group protected with a hydroxy protecting group, as defined above.
  • Compound A tosylate salt refers to a tosylate salt of Compound A, comprising a cation of Compound A and an anion from -toluenesulfonic acid, where the anion is referred to herein as "tosylate.”
  • a tosylate salt of (8S,9 ⁇ )-5-tluoro-8-(4-fluorophenyl)-9-( 1 - methyl-1 H- 1 ,2,4-triazol-5-yl)-8,9-dihydro-2 /-pyrido[4,3,2-(ie]phthalazin-3(7//)-one comprises a cation of (85,9y?)-5-fluoro-8-(4-f1uorophenyl)-9-( l -methyl- l H- l ,2,4-triazol-5-yl)-8,9-dihydro-2H-pyrido[4,3 cfe]phthalazin-3(7H)-one (e.g., in one embodiment, protonated at one atomic position, or in other embodiments, protonated at more than one atom ic position) and an anion of p-toluenesulfonic acid, where the
  • the terms "about” and “approximately,” when used in connection with a numeric value or range of values which is provided to describe a particular solid form e.g. , a specific temperature or temperature range, such as, for example, that describing a melting, dehydration, desolvation or glass transition; a mass change, such as, for example, a mass change as a function of temperature or humidity; a solvent or water content, in terms of, for example, mass or a percentage; or a peak position, such as, for example, in analysis by, for example, ' " 'C NMR, DSC, TGA and XRPD; indicate that the value or range of values may deviate to an extent deemed reasonable to one of ordinary skil l in the art while still describing the particular solid form.
  • the terms "about” and “approximately,” when used in this context, indicate that the numeric value or range of values may vary by 5%, 4%, 3%, 2%, 1 %, 0.9%, 0.8%, 0.7%, 0.6%, 0.5%, 0.4%, 0.3%, 0.2% or 0.1 % of the recited value or range of values while still describing the particular solid form.
  • the term "mixing" can refer to any process by which two or more ingredients are mixed together to produce a mixture.
  • the two or more ingredients can be mixed by being stirred by hand or by being stirred by a mechanical stirrer, including magnetic stirrer; or can be mixed by a hand-operated mixer or by a machine-operated mixer; or can be mixed by being blended in a blender, tumbled in a tumbler, or shaken in a hand-operated shaker or by a machine-operated shaker; and so forth, such that a mixture of the two ingredients is produced.
  • the terms “geometric blending” and “geometric mixing” refers to a sequence of steps where in the first step one ingredient (e.g., an active pharmaceutical ingredient) is mixed with an amount of a second ingredient (e.g., a diluent), and then in the second step more of the second ingredient (e.g. , a diluent) is added and mixed with mixture from the first step.
  • additional steps each comprising adding more of the second ingredient (e.g.
  • diluent and mixing it with to the mixture of the first and second ingredients can be performed, where such additional steps can be performed up to 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 1 7, 1 8, 19, 20 or more times.
  • low dose refers to a range of about 1 ⁇ g to about 8000 ⁇ g, about 1 ⁇ g to about 1500 ⁇ g, about 1 g to about 1000 ⁇ , about 1 ⁇ to about 750 g, about 1 ⁇ g to about 500 ⁇ g, about 1 ⁇ g to about 250 ⁇ g, about 1 ⁇ g to about 150 g, about 1 ⁇ g to about 100 ⁇ g, about 1 ⁇ g to about 75 ⁇ g, about 5 g to about 1 500 g, about 5 ⁇ g to about 30 ug, about 10 ug to about 85 about 15 ⁇ g to about 85 ⁇ g, about 20 ⁇ g to about 1250 ⁇ -g, about 20 ⁇ g to about 1 12 ⁇ g, about 20 ⁇ g to about 60 ⁇ £, about 25 ⁇ to about 1000 ⁇ g, about 25 ⁇ g to about 250 ⁇ g, about 20 ⁇ to about 85 ⁇ g, about 25 g to about 85 ⁇ g, about 25 g to about 85 ⁇ g, about 25
  • unit dose means a single dose which is capable of being administered to a patient, and which can be readily handled and packaged, remaining as a physically and chemically stable unit dose, comprising a pharmaceutical composition as provided herein, for instance, having Compound A tosylate salt and one or more pharmaceutically acceptable excipients.
  • stable used in reference to Compound A tosylate salt, including when in a pharmaceutical composition or unit dosage form, means no detectable loss of potency when stored for 18 months or more at, for example, 25 ° C or ambient conditions or, for 4- 12 weeks or more at 40 ° C and 75% RH.
  • the term "stable" used in reference to crystalline Compound A tosylate salt, including when in a pharmaceutical composition or unit dosage form, means no detectable change (e.g., polymorphic change and/or chemical change such as, for example, due to degradation), in its crystalline form when stored for 18 months or more at, for example, 25 * C or ambient conditions or, for 4- 12 weeks or more at 40 ' C and 75% RH.
  • amorphous or “amorphous form” is intended to mean that the substance, component, or product in question is not substantially crystalline as determined, for instance, by XRPD or where the substance, component, or product in question, for example is not birefringent when viewed microscopically.
  • a sample comprising an amorphous form of a substance may be substantially free of other amorphous forms and/or crystalline forms.
  • crystalline form or “crystal form” refers to a crystalline solid form of a chemical compound, including, but not limited to, a single-component or multiple-component crystal form, e.g., a polymorph of a compound; or a solvate, a hydrate, a clathrate, a cocrystal, a salt of a compound, or a polymorph thereof.
  • crystal forms and related terms herein refers to the various crystalline modifications of a given substance, including, but not limited to, polymorphs, solvates, hydrates, co-crystals and other molecular complexes, as well as salts, solvates of salts, hydrates of salts, other molecular complexes of salts, and polymorphs thereof. Crystal forms of a substance can be obtained by a number of methods, as known in the art.
  • Such methods include, but are not limited to, melt recrystal lization, melt cooling, solvent recrystallization, recrystallization in confined spaces such as, e.g., in nanopores or capillaries, recrystal lization on surfaces or templates such as, e.g., on polymers, recrystallization in the presence of additives, such as, e.g., co-crystal counter-molecules, desolvation, dehydration, rapid evaporation, rapid cooling, slow cooling, vapor diffusion, sublimation, grinding and solvent-drop grinding.
  • additives such as, e.g., co-crystal counter-molecules, desolvation, dehydration, rapid evaporation, rapid cooling, slow cooling, vapor diffusion, sublimation, grinding and solvent-drop grinding.
  • 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).
  • 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 activity 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 1 8 proteins, particularly poly(ADP-ribose)polymerase- l (PARP-1 ) and poly(ADP-ribose)polymerase-2 (PARP-2).
  • selective PARP modulator refers to a compound that selectively modulates at least one activity associated with the enzyme poly(ADP-ribose)polymerase (PARP).
  • PARP poly(ADP-ribose)polymerase
  • 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).
  • 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).
  • PARP poly(ADP-ribose)polymerase
  • 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.
  • target activity refers to a biological activity capable of being modulated by a selective modulator.
  • Certain exemplary target activities include, but are not limited to, binding affinity, signal transduction, enzymatic activity, tumor growth, inflammation or inflammation- related processes, and amelioration of one or more symptoms associated with a disease or condition.
  • the term "inhibitor” refers to a compound, the presence of which results in a decrease in the magnitude of a biological activity of a protein. In certain embodiments, the presence of an antagonist results in complete inhibition of a biological activity of a protein, such as, for example, the enzyme poly(ADP-ribose)poiymerase (PARP).
  • PARP poly(ADP-ribose)poiymerase
  • the IC 50 refers to an amount, concentration or dosage of a particular test compound that achieves a 50% inhibition of a maximal response, such as modulation of PARP, in an assay that measures such response.
  • EC 50 refers to a dosage, concentration or amount of a particular test compound that elicits a dose-dependent response at 50% of maximal expression of a particular response that is induced, provoked or potentiated by the particular test compound.
  • 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).
  • 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 prevent the development of and/or 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.
  • terapéuticaally effective amount also refers to the amount of a compound that is sufficient to elicit the biological or medical response being sought by a researcher, veterinarian, medical doctor, or clinician, for example, to inhibit PARP activity in vivo, to inhibit cancer cel l growth and/or proliferation, and/or to decrease cancer cell numbers.
  • 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 abil ity 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.
  • 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.
  • 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 inhibit the activity of the target, to limit the activity of the target, or to extend the activity of the target.
  • the term "pharmaceutically acceptable” refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem complications commensurate with a reasonable benefit/risk ratio.
  • a “pharmaceutically acceptable excipient,” as used herein, refers to a pharmaceutically acceptable material, composition, or vehicle, such as a liquid or solid filler, diluent, solvent, or encapsulating material.
  • each component is "pharmaceutically acceptable" in the sense of being compatible with the other ingredients of a pharmaceutical formulation, and suitable for use in contact with the tissue or organ of humans and an imals without excessive toxicity, irritation, allergic response, immunogenicity, or other problems or complications, commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically acceptable in the sense of being compatible with the other ingredients of a pharmaceutical formulation, and suitable for use in contact with the tissue or organ of humans and an imals without excessive toxicity, irritation, allergic response, immunogenicity, or other problems or complications, commensurate with a reasonable benefit/risk ratio.
  • an excipient can, for example, be an antiadherent, a binder, a buffer, a carrier, a disintegrant, an emulsifying agent, a filler, a diluent, a flavoring agent, a coloring agent, a lubricant, a glidant, a preservative, an antioxidant, a sorbent and/or a sweetner.
  • 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.
  • pharmaceutical ly acceptable salts are obtained by reacting a compound described herein, with acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, methanesulfonic 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, N-methyl-D-glucamine, tris(hydroxymethyl)methylamine, and salts with amino acids such as arginine, lysine, and the like, or by other methods previously determined.
  • 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, N-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.
  • polymorph refers to one of two or more crystal forms that comprise the same molecule, molecules or ions.
  • Different polymorphs may have different physical properties such as, for example, melting temperatures, heats of fusion, solubilities, dissolution rates, and/or vibrational spectra as a result of the arrangement or conformation of the molecules or ions in the crystal lattice.
  • the differences in physical properties exhibited by polymorphs may affect pharmaceutical parameters, such as storage stability, compressibility, density (important in formulation and product manufacturing), and dissolution rate (an important factor in bioavailability).
  • Differences in stability can result from changes in chemical reactiv ity (e.g., differential oxidation, such that a dosage form discolors more rapidly when comprised of one polymorph than when comprised of another polymorph), mechanical changes (e.g., tablets crumble on storage as a kinetica!ly favored polymorph converts to
  • thermodynarnically more stable polymorph or both (e.g., tablets of one polymorph are more susceptible to breakdown at high humidity).
  • some polymorphic transitions may result in lack of potency or, at the other extreme, toxicity.
  • the physical properties of a crystalline form may be important in processing; for example, one polymorph might be more l ikely to form solvates or might be difficult to filter and wash free of impurities (e.g., particle shape and size distribution might be different between polymorphs).
  • 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 bioavaiiable 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 faci litate transmittal across a cell membrane where water solubility is detrimental to mobility but which then is metabolicaily hydrolyzed to the carboxylic 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.
  • stereomerically pure means a composition that comprises one stereoisomer of a compound and is substantially free of other stereoisomers of that compound.
  • stereomerically pure 8.S ⁇ 9/?)-5-fluoro-8-(4- fluorophenyl)-9-( 1 -methyl- l H-l ,2,4-triazol-5-yl)-8,9-dihydro-2/ -pyrido[4,3,2- ⁇ :/e]phthalazin-3(7H)-one is provided herein that is substantially free of the other stereoisomers including, for example, (8R,9S)-5- fluoro-8-(4-fluorophenyI)-9-( l -methyl- l H- l ,2,4-triazol-5-yl)-8,9-dihydro-2H-pyrido[4,3,2-cfe]phthalazin- 3(7//)-one.
  • a stereomerically pure compound comprises greater than about 80 percent by weight of one stereoisomer of the compound and less than about 20 percent by weight of other stereoisomers of the compound, greater than about 90 percent by weight of one stereoisomer of the compound and less than about 10 percent by weight of the other stereoisomers of the compound, greater than about 95 percent by weight of one stereoisomer of the compound and less than about 5 percent by weight of the other stereoisomers of the compound, greater than about 97 percent by weight of one stereoisomer of the compound and less than about 3 percent by weight of the other stereoisomers, or greater than about 99 percent by weight of one stereoisomer of the compound and less than about I percent by weight of the other stereoisomers of the compound.
  • term "stereomerically pure" (8S,9R)-5-fluoro-8-(4-fluoropheny l)-9-( 1 -methyl- i H- 1 ,2,4-triazol-5-yl)-8,9- dihydro-2H-pyrido[4,3,2-i e]phthalazin-3(7H)-one means that the compound is made up of approximately 100% by weight of this particular stereoisomer. The above percentages are based on the total amount of combined stereoisomers of the compound.
  • a crystalline or amorphous form that is "pure,” i.e. , substantially free of other crystalline or amorphous forms contains less than about 10 percent by weight of one or more other crystalline or amorphous form, less than about 5 percent by weight of one or more other crystalline or amorphous form, less than about 3 percent by weight of one or more other crystalline or amorphous form, less than about 1 percent by weight of one or more other crystalline or amorphous form, or less than about 0.5 percent by weight of one or more other crystalline or amorphous form.
  • -'substantially pure (85,9R)-5-fluoro-8-(4-fluorophenyl)-9-( I -methyl- 1/7- 1 , 2,4-triazol-5-yl)-8,9- dihydro-2//-pyrido[4.3.2-i/t']phthalazin-3(7//)-one or a salt or solvate thereof can mean free of other chemical compounds, for example, unreacted precursors and side products that might be present in process for preparing (8S,9/i)-5-fluoro-8-(4-fluorophenyl)-9-( l -methyl- 2,4-triazol-5-yl)-8,9- dihydro-2H-pyrido[4,3,2-i/e]phthalazin-3(7H)-one.
  • a "substantially pure" solid form ⁇ e.g., crystalline form or amorphous form) of (8S,9R)-5-fluoro-8-(4-fluorophenyl)-9-( t - methyl- ⁇ H- 1 ,2,4-triazol-5-yl)-8,9-dihydro-2H-pyrido[4,3,2-i t?]phthalazin-3(7H)-one or a salt or solvate thereof
  • substantially pure (8S.9 ⁇ )-5-fluoro-8-(4-fluorophenyl)-9-( l -methyl- l H- 1 , 2,4-triazol-5-yl)-8,9-dihydro- 2H-pyrido[4,3,2-c/e]phthalazin-3(7//)-one may comprise, in certain embodiments, less than about 10%, 5%, 3%, 2%, 1 %, 0.75%, 0.5%, 0.25%, or 0.1% by weight of one or more other crystal forms and amorphous forms of the compound and/or other chemical compounds.
  • a solid form that is substantially pure is substantially free of one or more other particular crystal forms, amorphous forms, and/or other chemical compounds.
  • 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 l ike.
  • 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 cond ition, 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.
  • the terms “treat,” “treating,” and “treatment” are meant to include alleviating or abrogating a disorder, disease, or condition, or one or more of the symptoms associated with the disorder, disease, or condition; or to slowing the progression, spread or worsening of a disease, disorder or condition or of one or more symptoms thereof.
  • the beneficial effects that a subject derives from a therapeutic agent do not result in a complete cure of the disease, disorder or condition.
  • 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, amel iorate, 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.
  • 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 lUPAC 1 974 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.
  • individual stereoisomers of compounds are prepared synthetical ly 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
  • the methods and formulations described herein include the use of JV-oxides, cry stal line 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.
  • AH 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.
  • compositions are formulated in any manner, including using one or more physiologically acceptable carriers comprising excipients and/or auxiliaries which facilitate processing of the active compounds into pharmaceutical preparations.
  • physiologically acceptable carriers comprising excipients and/or auxiliaries which facilitate processing of the active compounds into pharmaceutical preparations.
  • proper formulation is dependent upon the route of administration chosen.
  • any techniques, carriers, and excipients are used as suitable.
  • compositions that include a compound described herein and one or more 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.
  • 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.
  • 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 other excipients.
  • Such carriers enable the compounds described herein to be formulated as tablets, powders, pi lls, 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 tragaeanth, methylcellulose, microcrystalline cellulose, hydroxypropy Imethylcel lulose, 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, optional ly, stabilizers.
  • the active compounds are dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • stabil izers are optional ly added.
  • the formulations for oral administration are in dosages suitable for such administration.
  • the pharmaceutical compositions provided herein are formu lated 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.
  • Pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form.
  • suspensions of the active compounds are prepared as appropriate oi ly 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 l iposomes.
  • 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, in some embodiments, 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), (IA) 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 del iver the compound to the skin of the host at a control led 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 admin istration 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 propel lan t, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • the dosage unit is determined by prov iding a valve to del iver 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.
  • 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 administration chosen.
  • any of the techniques, carriers, and excipients is used as suitable.
  • 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.
  • 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 jV-oxides, cr stalline forms (also known as polymorphs), as wel l 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. 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.
  • 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
  • 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.
  • Semi-solid 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 methylcellulo.se. and water-insoluble polymers such as cross-linked carboxyl-containing polymers.
  • Useful compositions optionally comprise an mucoadhesive polymer, selected for example from
  • carboxymethylcellulose carboxymethylcellulose.
  • carbomer acrylic acid polymer
  • polyacrylamide poly(methylmethacrylate)
  • polycarbophil acrylic acid/butyl acrylate copolymer
  • sodium alginate 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.
  • 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.
  • 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 me r fen and thiomersal; stabilized chlorine dioxide; and quaternary ammon ium compounds such as benzalkonium chloride, cetyltrimethylammonium bromide and cetylpyridinium chloride.
  • Some usefu l 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.
  • 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 iV-methylpyrrolidone are employed.
  • the compounds are delivered using a sustained-release system, such as
  • 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 stabi lizing 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, (I) pentosan polysulfate and other heparinoids, (m) divalent cations such as magnesium and zinc; or (n) combinations thereof.
  • 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.
  • PARP poly(ADP-ribose)polymerase
  • 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 activ e metabol ite,
  • 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 administered is defined to be a "prophyiactically effective amount or dose.”
  • 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 prophyiactically effective amounts by routine
  • experimentation e.g., a dose escalation clinical trial.
  • 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 fol lowing administration of a compound or composition described herein and, upon the doctor's d iscretion the adm inistration 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 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").
  • 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, 150 days, 1 80 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 admin istration, 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 administration, the condition being treated, and the subject or host being treated.
  • 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 reclosable 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.
  • Un it dosage forms provided herein comprise Compound A in a solid tosylate salt form.
  • unit dosage forms referred to herein are intended to mean the amount of Compound A in the unit dosage form, not taking into account the mass contributed by the tosylate portion of the salt.
  • a "25 ⁇ g capsules” will be understood to mean, unless indicated otherwise in the context in which it is used, that the capsule contains about 36.325 fig Compound A tosylate salt.
  • a conversion factor of 1.453 mg Compound A tosylate salt per 1.000 mg Compound A can be used to convert the amount of Compound A indicated in a given unit dosage form to the amount of Compound A tosylate salt contained in the unit dosage form.
  • amounts of (85,9/i)-5-fliioro-8-(4-fIuorophenyl)-9-( l -methyl- lf/- l ,2,4-triazol-5-yl)-8,9-dihydro-2//-pyrido[4,3,2-de]phthalazin-3(7//)-one tosylate salt in unit dosage forms can, for example, be from about 5 g to about 1500 ⁇ g, from about 20 g to about 1250 ⁇ , from about 25 ⁇ g to about 1000 ⁇ g, or from about 25 ⁇ g to about 250 g.
  • de]phthalazin-3(7H)-one tosylate salt in a unit dosage form comprises (where the following do not including mass contributed by the tosylate portion of the salt) about 5 ⁇ g to about 30 ⁇ g, from about 20 ⁇ to about 60 ⁇ g, from about 50 ⁇ g to about 100 ⁇ g, from about 120 ⁇ g to about 250 ⁇ , from about 20 ⁇ g to about 1 12 ⁇ g, or from about 25 g to about 250 ug (8£,9i?)-5-fluoro-8-(4-fluorophenyI)-9-( 1 -methyl- l / - l ,2,4-triazol-5-yl)-8,9-dihydro-2//-pyrido[4,3,2-de]phthalazin-3(7 )-one.
  • pyrido[4,3,2-de]phthalazin-3(7 /)-one is about 10 ⁇ g, about 20 g, about 25 ⁇ g, about 30 ⁇ , about 50 ⁇ g, about 75 ⁇ g, about 100 ⁇ 3 ⁇ 4 about 150 g, about 200 ⁇ g, about 250 ⁇ g, or about 1000 ⁇ g.
  • the pharmaceutical composition as provided herein, is formulated for oral administration to a subject.
  • Pharmaceutical compositions suitable for oral administration can be presented as discrete dosage forms, such as, but are not limited to, tablets, chewable tablets, caplets, capsules, and liquids (e.g. , flavored syrups).
  • Such dosage forms contain predetermined amounts of active ingredients, and may be prepared by methods of pharmacy wel l known to those skilled in the art. See generally, Remingto 's Pharmaceutical Sciences, 1 8th ed., Mack Publishing, Easton PA ( 1990).
  • oral dosage forms provided herein may be prepared by combining (8.9,9/?)-
  • tablets can be coated by standard aqueous or nonaqueous techniques.
  • dosage forms can be prepared by any of the methods of pharmacy.
  • pharmaceutical compositions and dosage forms are prepared by uniformly and intimately admixing the active ingredient with l iquid carriers, finely divided solid carriers, or both, and then shaping the product into the desired presentation if necessary.
  • Disintegrants or lubricants can be used in pharmaceutical compositions and dosage forms of the invention. Production of pharmaceutical compositions or dosage forms in accordance with the present invention may require, in addition to the therapeutic drug ingredients, exeipients or additives including, but not limited to, diluents, binders, lubricants, disintegrants, colorants, flavors, sweetening agents and the like or mixtures thereof.
  • dosage forms e.g., tablets, capsules, caplets, troches and the like
  • dosage forms include, for example, hard gelatin capsules, caplets, sugar-coated tablets, enteric-coated tablets (for example to delay action), multiple compressed tablets, prolonged-action tablets, tablets for solution, effervescent tablets, buccal and sublingual tablets, troches and the like.
  • the dose form or dosage formulation may be formed by methods well known in the art. See, e.g. , Remington 's Pharmaceutical Sciences, 16th and 18th eds., Mack Publishing Co., Easton, Pa. ( 1980 and 1990). See also U.S. Pharmacopeia XXI, U.S.
  • parenteral dosage forms can be administered to patients by various routes including, but not l imited to, subcutaneous, intravenous (including bolus injection), intramuscular, and intraarterial. Because their administration typically bypasses patients' natural defenses against contaminants, parenteral dosage forms are in some embodiments sterile or capable of being sterilized prior to administration to a patient. Examples of parenteral dosage forms include, but are not limited to, solutions ready for injection, dry products ready to be dissolved or suspended in a pharmaceutically acceptable vehicle for injection, suspensions ready for injection, and emulsions. Suitable vehicles that can be used to provide parenteral dosage forms provided herein are well known to those ski lled in the art.
  • the pharmaceutical composition as provided herein can be formulated in a transdermal, topical or mucosal dosage form.
  • Transdermal, topical, and mucosal dosage forms provided herein include, but are not limited to, ophthalmic solutions, sprays, aerosols, creams, lotions, ointments, gels, solutions, emulsions, suspensions, or other forms known to one of skill in the art.
  • Suitable exeipients e.g., carriers and diluents
  • other materials that can be used to provide transdermal, topical, and mucosal dosage forms encompassed by this disclosure are wel l known to those skilled in the pharmaceutical arts, and depend on the particular tissue to which a given pharmaceutical composition or dosage form will be appl ied.
  • 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 ED 50 (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 LD $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. Kits/Articles of Manufacture
  • kits and articles of manufacture are also described herein.
  • such kits comprise a carrier, package, or container that is compartmentalized to receive one or more containers such as vials, tubes, and the like, each of the container(s) comprising one of the separate elements to be used in a method described herein.
  • Suitable containers include, for example, bottles, vials, syringes, and test tubes.
  • the containers are formed from a variety of materials such as glass or plastic.
  • the articles of manufacture provided herein contain packaging materials.
  • Packaging materials for use in packaging pharmaceutical products include, but are not limited to, blister packs, bottles, tubes, inhalers, pumps, bags, vials, containers, syringes, bottles, and any packaging material suitable for a selected formulation and intended mode of administration and treatment.
  • the container(s) described herein comprise one or more compounds described herein, optionally in a composition or in combination with another agent as disclosed herein.
  • the containers) optionally have a sterile access port (for example in some embodiments the container is an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • kits optionally comprise a compound with an identifying description or label or instructions relating to its use in the methods described herein.
  • a kit will comprises one or more additional containers, each with one or more of various materials (such as reagents, optionally in concentrated form, and/or devices) desirable from a commercial and user standpoint for use of a compound described herein.
  • materials include, but are not l imited to, buffers, diluents, filters, needles, syringes; carrier, package, container, vial and/or tube labels listing contents and/or instructions for use, and package inserts with instructions for use.
  • a set of instructions is optionally included.
  • a label is on or associated with the container.
  • a label is on a container when letters, numbers or other characters form ing the label are attached, molded or etched into the container itself; a label is associated with a container when it is present within a receptacle or carrier that also holds the container, e.g., as a package insert.
  • a label indicates that the contents are to be used for a specific therapeutic application.
  • the label indicates directions for use of the contents, such as in the methods described herein.
  • the pharmaceutical compositions are presented in a pack or dispenser device wh ich contains one or more unit dosage forms containing a compound provided herein.
  • the pack contains a metal or plastic foil, such as a bl ister pack.
  • the pack or dispenser device is optionally accompanied by instructions for administration.
  • the pack or dispenser is accompanied with a notice associated with the container in form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals, which notice is reflective of approval by the agency of the form of the drug for human or veterinary administration. In certain embodiments, such notice is, for example, the labeling approved by the U.S. Food and Drug Administration for prescription drugs, or the approved product insert.
  • compositions containing a compound provided herein are formulated in a compatible pharmaceutical carrier and are placed in an appropriate container labeled for treatment of an indicated condition.
  • 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 util ized.
  • the synthetic methods described in WO 2010/017055, WO/201 1/097602, and WO2012054698 can be used to prepare a compound used in the methods disclosed herein, each of which is incorporated herein by reference in its entirety.
  • Techniques for characterizing crystal forms and amorphous forms include, but are not limited to, TGA, DSC, XRPD, single crystal X-ray diffractometry, vibrational spectroscopy, e.g., IR and Raman spectroscopy, solid-state NMR, optical microscopy, hot stage optical microscopy, SEM, electron crystallography and quantitative analysis, PSA, surface area analysis, solubility studies and dissolution studies.
  • Example l a Parenteral Composition
  • a parenteral pharmaceutical composition suitable for administration by injection 100 mg of a water-soluble salt of a compound described herein is dissolved in DM SO and then mixed with 10 mL of 0.9% sterile saline. The mixture is incorporated into a dosage unit form suitable for administration by injection.
  • Example l b Oral Composition
  • a pharmaceutical composition for oral delivery 100 mg of a compound described herein is mixed with 750 mg of starch. The mixture is incorporated into an oral dosage unit for, such as a hard gelatin capsule, which is suitable for oral administration.
  • Example l c Sublingual (Hard Lozenge) Composition
  • a pharmaceutical composition for buccal delivery such as a hard lozenge
  • the mixture is gently blended and poured into a mold to form a lozenge suitable for buccal administration.
  • Example I d Inhalation Composition
  • a pharmaceutical composition for inhalation delivery 20 mg of a compound described herein is mixed with 50 mg of anhydrous citric acid and 100 mL of 0.9% sodium chloride solution. The mixture is incorporated into an inhalation delivery unit, such as a nebulizer, which is suitable for inhalation administration.
  • an inhalation delivery unit such as a nebulizer
  • a pharmaceutical composition for rectal delivery 100 mg of a compound described herein is mixed with 2.5 g of methylcel luose ( 1500 mPa), 100 mg of methylparapen, 5 g of glycerin and 100 mL of purified water. The resulting gel mixture is then incorporated into rectal delivery units, such as syringes, which are suitable for rectal administration.
  • a pharmaceutical topical gel composition 100 mg of a compound described herein is mixed with 1 .75 g of hydroxypropy I cellulose, 10 mL of propylene glycol, 10 mL of isoprop l myristate and 100 mL of purified alcohol USP. The resu lting gel mixture is then incorporated into containers, such as tubes, which are suitable for topical administration.
  • ophthalm ic solution composition 100 mg of a compound described herein is mixed with 0.9 g of NaCl in 100 mL of purified water and filtered using a 0.2 micron niter. The resulting isotonic solution is then incorporated into ophthalm ic delivery units, such as eye drop containers, which are suitable for ophthalmic administration.
  • reaction mixture was heated from ambient to reflux temperatures (85 - 95 °C) for 80 - 90 minutes, and the mixture was refluxed for another 70 - 90 minutes.
  • the reaction mixture was monitored by HPLC, indicating compound (2) was reduced to ⁇ 5 %.
  • the resulting slurry was cooled down to 5 -
  • Example 161 A An alternative workup procedure to that illustrated in Example 161 A follows. Instead of evaporating the reaction mixture to dryness, it was condensed to 2 volumes, followed by solvent exchange with 12 volumes of THF, and then 12 volumes of heptane. The slurry mixture was concentrated to 2 volumes and filtered to give the product.
  • Example 162A An alternative workup procedure to that illustrated in Example 162A follows. After the completion of the reaction, the mixture was extracted with isopropyl acetate (20 volumes x 4) without water dilution. The product was isolated by solvent exchange of isopropyl acetate with heptanes followed by re-slurry with TBE and filtration. As such, 3 kilograms of methyl 5-fluoro-2-(2-( l -methyl- l H- l ,2,4- triazol-5-yl)acetyl)-3-nitrobenzoate (4) afforded 2.822 kilograms of the title compound (5) (yield 81 %).
  • the separation can also be achieved on a Simulated Moving Bed (SMB) unit with a CHIRALPA ®1C column and acetonitrile as a mobile phase.
  • SMB Simulated Moving Bed
  • the retention times for the two enantiomers are 3.3 and 4.1 minutes, respectively.
  • the productivity can be greater than 6 kg Feed/day/kg CSP.
  • Crystalline Compound A tosylate salt can, for instance, be prepared using crystallization techniques known in the art, including, but not limited to, melt cooling, rapid melt cooling, freeze drying, spray drying, roller drying, lyophilization, quench cooling the melt, rapid solvent evaporation, slow solvent evaporation, solvent recrystallization, slurry reerystal lization.
  • melt crystallization, desolvation, sublimation, recrystallization in confined spaces e.g., in nanopores or capillaries
  • recrystallization on surfaces or templates e.g., on polymers
  • recrystallization in the presence of additives e.g., co-crystal counter-molecules
  • dehydration rapid cool ing, slow cooling, vapor diffusion, grinding, cryo-grinding, solvent-drop grinding, microwave-induced precipitation, ultrasonication-induced precipitation, laser- induced precipitation, and precipitation from a supercritical fluid.
  • Salt forms of (8S,9R)-5-fluoro-8-(4-fluorophenyl)-9-( l -methyl-l H- l ,2,4-triazoi-5-yl)-8,9- dihydro-2H-pyrido[4,3,2-Je]phthalazin-3(7 )-one were prepared by treating the compound under a given condition with an acid. Salts forms, nos.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Provided is a methods of treating multiple myeloma with inhibitors of poly(ADP-ribose)polymerase according to Formula (I):

Description

DIHYDROPYMDOPHTHALAZINONE INHIBITORS OF POLY(ADP-RTBOSE)POLYMERASE (PARP) FOR THE TREATMENT OF MULTIPLE MYELOMA
CROSS REFERENCE TO RELATED APPLICATIONS
[0001] The present application claims the benefit of priority of U.S. Provisional Application
No. 61/525,645 filed on August 19, 201 1 and U.S. Provisional Application No. 61/558,719 filed on November 1 1 , 201 1 , the contents of which are hereby incorporated by reference in their entireties.
FIELD OF THE INVENTION
[00021 Described herein are methods of using compounds for the treatment of mu ltiple myeloma as well as pharmaceutical compositions comprising (85,,9R)-5-fluoro-8-(4-fluorophenyl)-9-( 1 -methyl- 1 ,2,4-triazol-5-yl)-8,9-dihydro-2 /-pyrido[4,3,2-de]phthalazin-3(7H)-one tosylate salt, unit dosage forms thereof, and methods of their preparation.
BACKGROUND OF THE INVENTION
[0003] The family of poly(ADP-ribose)polymerases (PARP) includes approximately 18 proteins, wh ich all display a certain level of homology in their catalytic domain but differ in their cellular functions (Ame et al., BioEssa s., 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. The mammalian enzyme PARP- 1 is a multidomain protein. PARP has been implicated in the signal ing 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 ( 1 999); and Virag et al. Pharmacological Reviews, vol. 54, no. 3, 375-429 (2002)). It participates in a variety of DNA-related functions including gene ampl ification, 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] PARP has an essential role in facilitating DNA repair, control l ing RNA transcription, mediating cell death, and regulating immune response. Studies on the mechanism by which PARP- 1 modulates DNA repair and other processes identifies its importance in the formation of poly(ADP-ribose) chains within the cellular nucleus. The DNA-bound, activated PARP- 1 utilizes NAD+ to synthesize poly(ADP-ribose) on a variety of nuclear target proteins, including topoisomerases, histones and PARP itself. (Althaus, F. R. and Richter, C, ADP-Ribosylation of Proteins: Enzymology and Biological Significance, Springer-Verlag, Berlin ( 1987); and Rhun, et al, Biochem. Biophys. Res. Commun., 245, 1 - 10 ( 1998)). PARP inhibitors are used for the treatment of cancer which is deficient in Homologous Recombination (HR) dependent DNA double strand break (DSB) repair activity. The HR dependent DNA DSB repair pathway repairs double-strand breaks (DSBs) in DNA via homologous mechanisms to reform a continuous DNA helix.
[0005] While the use of a PARP inhibitor as a single agent for the treatment of multiple myeloma has been suggested as early as 1 99 (WO9901 162), results have not substantiated the suggestion. Combinations of other anticancer agents, e.g. proteasome inhibitors, with a PARP inhibitor have been studied and found to act synergistically (L. Nong et, al., Bortezomib-Mediated Down Regulation of FANCD2 Facilitates Synergism in Combination with PARP Inhibition (Olaparib), Poster P-281 at the 13th International Myeloma Workshop in Paris, France May 3-6, 201 1 ). However, as yet, there is no report of a PARP inhibitor as a single agent for the treatment of multiple myeloma. In fact, even though there have been reports that a PARP inhibitor has significantly reduced PAR polymers in multiple myeloma cells, it had no effect on the viability of the cells (P. Neri et. al., "PARP Inhibitor as Novel Therapeutic Approach in Multiple Myeloma, Poster P-301 " at the 13th International Myeloma Workshop in Paris, France May 3- 6, 201 1 ). Applicants herein provide PARP inhibitors which are useful as single agents, as well as in combination with other agents, for the treatment of multiple myeloma.
[0006] The high potency of (8S,97?)-5-Fluoro-8-(4-fluorophenyl)-9-( l -methyl- 1 /- 1 , 2,4-triazol-5- yl)-8,9-dihydro-2H-pyrido[4,3,2-deJphthalazin-3(7H)-one (referred to herein as "Compound A") as a PARP inhibitor as well as its physiochemical properties can pose difficulties in formulating drug delivery forms of Compound A, specifically, forms which can be manufactured on a large scale in which
Compound A is stable, for example, under ambient conditions, and from which a consistent amount of Compound A wil l be delivered to the patient. Improved pharmaceutical compositions ofCompound A and unit dosage forms thereof are desired.
SUMMARY OF THE INVENTION
[0007] The following only summarizes certain aspects of the invention and is not intended to be limiting in nature. These aspects and other aspects and embodiments are described more ful ly below. All publications, patents, and patent applications cited in this specification are hereby incorporated by reference in their entirety for all purposes. In the event of a discrepancy between the express disclosure of this specification and the references incorporated by reference, the express disclosure of this specification shall control.
[0008] One aspect is a method of treating multiple myeloma with a Compound according to
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 R6; c) a substituent independently selected from the group consisting of hydrogen, alkenyl, alkoxy, alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, alkyl, alkynyl, arylalkyl, cycloalkyi, cycloalkylalkyl, haloalkyl, hydroxyalkylene, oxo, heterocycloalkyl, heterocycloalkylalkyl, alkylcarbonyl, arylcarbonyl. heteroarylcarbonyl alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, -(Ci -C6-alkylene)-NRARB, -C(0)(NRARB), -(C C6-alkylene)-C(0)NRARB, -S(0)2(NRARB), and -(CrC6-alkylene)-S(0)2(NRARB);
R[, R2, and R3 are each independently selected from the group consisting of hydrogen, halogen, alkenyl, alkoxy, alkoxycarbonyl, alkyl, cycloalkyi, alkynyl, cyano, haloalkoxy, haloalkyl, hydroxyl, hydroxyalkylene, nitro, -NRARB, -(C i-C6-alkylene)-NRARB, and -C(0)(NRARB);
A and B are each independently selected from hydrogen, Br, CI, F, I, OH, Q-C^alkyl, C3-C8cycloaIkyl, alkoxy, alkoxyalkyl wherein C|-C6alkyl, Ca-Cgcycloalkyl, alkoxy, and alkoxyalkyl are optionally substituted with at least one substituent selected from OH, NO?, CN, Br, CI, F, I, C C6alkyl, and QrCgcycloalkyl, wherein B is not OH;
RA and RB are independently selected from the group consisting of hydrogen, alkyl, cycloalkyi, 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 functional ities selected from the group consisting of -0-, -NH-, -N(C C6-alkyl)-, -NC(0)(Ci-C6-alkyl)-, -N(aryl)-, -N(aryl-C |-C6-alkyl-)-, -N(substituted-aryl-CrC6-alkyl-)-, -N(heteroaryl)-, -N(heteroaryl-C C6-alkyl- )-, -N(substituted-heteroaryl-CrC6-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.i and R5 are each independently selected from the group consisting of hydrogen, alkyl, cycloalkyi, alkoxyalkyl, haloalkyl, hydroxyalkylene, and -(C|-C6-alkylene)-NRARB;
each R<i is independently selected from OH, N02, CN, Br, CI, F, I, CrC5alkyl, Cs-Qeycloalkyl,
Figure imgf000004_0001
-C6alkenyl, alkoxy, alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C2-C6alkynyl, aryl, arylalkyl, QrQcycloalkylalkyl, haloalkoxy, haloalkyl, hydroxyalkylene, oxo, heteroaryl, heteroarylalkoxy, heteroaryloxy, heteroarylthio, heteroarylalkylthio, heterocycloalkoxy, Cj-Cgheterocycloalkylthio, heterocyclooxy, heterocyclothio, -NRARB, -(C 1 -C6-alky lene)-N RARB, -C(0)(NRARB), -(Ci -C6-aIkylene)-C(0)NRARB, -S(0)2(NRARB), and -(C,-C6-alkylene)- S(0)2(NRARB);
a single isomer, stereoisomer, or enantiomer or mixture thereof, optionally as a salt, solvate, chemically protected form or prodrug thereof.
[0009] Another aspect is a method of treating multiple myeloma with a pharmaceutical composition comprising a compound of Formula (I) or a single isomer, stereoisomer, or enantiomer or mixture thereof, optionally as a pharmaceutically acceptable salt, pharmaceutically acceptable solvate, or pharmaceutically acceptable prodrug thereof and a pharmaceutically acceptable carrier, excipient, binder or diluent thereof. [0010] Another aspect is a method of treating multiple myeloma comprising administering to a subject in need thereof a pharmaceutical combination which comprises a therapeutical ty-effective amount of a compound of Formula (I) or a pharmaceutical composition thereof and a proteasome inhibitor.
[0011] Another aspect is the use of a compound of Formula (I) for the treatment of multiple myeloma.
[0012] Another aspect is the use of a pharmaceutical combination comprising a therapeutical ly- effective amount of a compound of Formula (I) or a pharmaceutical composition thereof and a proteasome inhibitor for the treatment of multiple myeloma in a subject in need thereof.
[0013] Another aspect is an article of manufacture, comprising packaging material, a compound of Formula (I), optionally a proteasome inhibitor, and a label, wherein the compound is for treatment, prevention or amelioration of multiple myeloma, wherein the compound is packaged within the packaging material, and wherein the label indicates that the compound, or pharmaceutically acceptable salt, pharmaceutically acceptable N-oxide, pharmaceutically active metabolite, pharmaceutically acceptable prodrug, or pharmaceutically acceptable solvate thereof, or a pharmaceutical composition comprising such a compound is used for treatment, prevention or amelioration multiple myeloma.
[0014] In another aspect, a pharmaceutical composition is provided herein comprising
Compound A tosylate salt in solid form and one or more excipients.
[0015] In another aspect, a pharmaceutical composition is provided comprising Compound A tosylate salt and silicified microcrystalline cellulose, wherein the pharmaceutical composition is prepared using crystalline Compound A tosylate salt.
[0016] In another aspect, a unit dosage form is provided comprising pharmaceutical composition, the pharmaceutical composition comprising Compound A tosylate salt in solid form and one or more pharmaceutically acceptable excipients, wherein the weight/weight percent of Compound A tosylate salt in the pharmaceutical composition is between about 0.01 % to about 1.000 %.
[0017] In yet another aspect, provided here are methods for preparing a pharmaceutical composition comprising Compound A tosylate salt and one or more excipients.
[0018] In another aspect, provided herein are pharmaceutical compositions produced by the methods described herein for producing a pharmaceutical composition.
[0019] In yet another aspect, provided herein are methods of treating a cancer or symptom thereof comprising administering to a cancer patient an effective amount of a pharmaceutical composition comprising Compound A tosylate salt as described herein.
BRIEF DESCRIPTION OF THE DRAWINGS
[0020] Fig.l provides an overlay of five examples of powder X-ray powder diffraction (XRPD) patterns of crystall ine (8S,9i?)-5-fluoro-8-(4-fIuorophenyl)-9-( l -methyl- 2,4-triazol-5-yl)-8,9- dihydro-2 -pyrido[4,3 ,2-de]phthaIazin-3(7/7)-one tosylate salt. [0021) Fig.2a provides a differential scanning calorimetric (DSC) graph of a crystalline (85,9/?)-
5-fluoro-8-(4-fIuorophenyl)-9-(l -methyl- 1 H-l,2,4-triazol-5-yl)-8,9-dihydro-2H-pyrido[4,3,2- de]phthaiazin-3(7 /)-one tosylate salt polymorph, 8, from acetone-THF preparation.
[0022] Fig.2b provides a thermogravimetric analysis (TGA) graph of a crystalline (85,9?)-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 tosylate salt polymorph, 8, from acetone-THF preparation.
[0023] Fig.3a provides a DSC graph of a crystalline (85,9?)-5-fluoro-8-(4-fluorophenyl)-9-(l- methyl-l /-l,2,4-triazol-5-yl)-8,9-dihydro-2//-pyrido[4,3,2-de]phthalazin-3(7/)-one tosylate salt polymorph, 5, from DCM-ACN preparation.
[0024] Fig.3b provides a TGA graph of a crystalline (85,9/?)-5-fluoro-8-(4-fluorophenyl)-9-( 1- methyl-l//-l,2,4-triazol-5-yl)-8,9-dihydro-2/-pyrido[4,3,2-de]phthalazin-3(7//)-one tosylate salt polymorph, 5, from DCM-ACN preparation.
[0025] Fig.4a provides a DSC graph of a crystalline (85,9/?)-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 tosylate salt polymorph, 6, from acetone preparation.
[0026] Fig.4b provides a TGA graph of a crystalline (85,9/?)-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 tosylate salt polymorph, 6, from acetone preparation.
[0027] Fig.5a provides a DSC graph of a crystalline (85,9?)-5-fluoro-8-(4-fluorophenyl)-9-(l- methyl-l/f-l,2,4-triazol-5-yl)-8,9-dihydro-2 -pyrido[4,3,2-de]phthalazin-3(7 /)-one tosylate salt polymorph, 7, from THF preparation.
[0028] Fig.5b provides a TGA graph of a crystalline (85,9/?)-5-fluoro-8-(4-fluorophenyl)-9-(l- methyl-li-l,2,4-triazol-5-yl)-8,9-dihydro-2i:/-pyrido[4,3,2-de]phthalazin-3(7 f)-one tosylate salt polymorph, 7, from THF preparation.
[0029] Fig.6 provides an X-ray powder diffraction (XRPD) spectrogram of a crystalline
(85,9?)-5-fluoro-8-(4-fluorophenyl)-9-(l-methy
de]phthalazin-3(7//)-one tosylate salt polymorph.
[0030] Fig.7a provides a DSC graph of a crystalline (85,9?)-5-fIuoro-8-(4-fluorophenyl)-9-( 1 - methyl-lJ-l,2,4-triazol-5-yI)-8,9-dihydro-2//-pyrido[4,3,2-de]phthalazin-3(7H)-one tosylate salt polymorph.
[0031] Fig.7b provides a TGA graph of a crystalline (85,9?)-5-fIuoro-8-(4-fluorophenyl)-9-(l- methyl- 1 H- 1 ,2,4-triazol-5-yl)-8,9-dihydro-2H-pyrido[4,3,2-de]phthalazin-3(7H)-one tosylate salt polymorph.
[0032] Fig.8 depicts an X-ray powder diffraction (XRPD) spectrogram of a crystalline (85,9/?)-
5-fluoro-8-(4-fluorophenyl)-9-(l-methyl-lH-l,2,4-triazol-5-yI)-8,9-dihydro-2H-pyrido[4,3,2- de]phthaIazin-3(7H)-one tosylate salt polymorph, 8. from acetone-THF preparation. 10033] Fig. 9 provides a dynamic vapor sorption isotherm plot of a crystalline (8S,9R)-5-fluoro-
8-(4-fluorophenyl)-9-( 1 -methyl- 1 H- 1 ,2,4-triazol-5-yl)-8,9-dihydro-2H-pyrido[4,3,2-de]phtlialazin-3(7H)- one tosylate salt polymorph, 8, from acetone-THF preparation.
[0034] Figs. 10a and 10b depict dose response curves and IC50 in H929 Myeloma Cells at 5, 7, and 10 days.
[0035] Fig. 11 depicts dose response curves and 1C50 in MM. I S Myeloma Cells at 7 and 10 days.
[0036] Fig. 12 provides a solid state 13C NMR spectrum for a (8S,9.ft)-5-fIuoro-8-(4- fluorophenyl)-9-( l -methyl- lH- l ,2,4-triazol-5-yl)-8,9-dihydro-2 /-pyrido[4,3,2-de]phthalazin-3(7fl)-one tosylate salt polymorph.
DETAILED DESCRIPTION OF THE INVENTION
[0037] Any combination of the groups described herein for the various variables is contemplated herein, as well as any combination of embodiments.
[0038] In some or any embodiments the subject is a human.
[0039] In some embodiments, compounds provided herein have the structure of Formu la (I) and chemically protected forms and pharmaceutically acceptable salts, solvates, esters, acids and prodrugs thereof. Provided herein are compounds having the structure of Formula (I) that are inhibitors of the enzyme poly(ADP-ribose)polymerase (PARP).
[0040] The following paragraphs present a number of embodiments of compounds that can be used in the methods disclosed herein. In each instance the embodiment includes both the recited compounds as wel l as a single stereoisomer or mixture of stereoisomers thereof, as well as a
pharmaceutically acceptable salt thereof.
[0041] In certain embodiments are provided compounds of Formula (I) or a therapeutically acceptable salt thereof wherein RT, R2, Rj are independently selected from a group consisting of hydrogen, alky I, and halogen; R» is hydrogen and R5 is selected from the group consisting hydrogen, a Iky I, cycloalkyl, alkoxyalkyl, haloalkyl, hydroxyalkylene, and -(Cj-C6-alkylene)(NRARB); RA, and RB re independently selected from the group consisting of hydrogen, alky i, cycloalkyl, and alkylcarbonyl; or RA and RB taken together with the atom to which they are attached form a 3- 1 0 membered heterocycle ring optionally having one to three heteroatoms or hetero functional ities selected from the group consisting of -0-, -NH, -N(C ,-C6-alkylK -NC(0)(C,-C6-alkyl)-, -N(aryl)-, -N(aryl- C C6-alkyl-)-, -N(substituted-aryl- Ci-Ce-alkyl-)-, -N(heteroaryl)-, -N(lieteroaryl- C|-C5-alkyl-)-, -N(substituted-heteroaryl- d-Q-a!kyl-)-, - S- and -S(0)q- , wherein q is 1 or 2 and the 3- 1 0 membered heterocycle ring is optionally substituted with one or more substituents; A and B are each independently selected from hydrogen, Br, CI, F, 1, OH, Cr C5alkyl, CrCgcycloalkyl, alkoxy, and alkoxyalkyl wherein C rC6alkyl, Cj-Cgcycloalkyl, alkoxy, and alkoxyalkyl are optionally substituted with at least one substituent selected from OH, N02, CN, Br, CI, F, I, CrQalkyl, and C3-Cgcycloalkyl, wherein B is not OH; Y and Z are each independently selected from the group consisting of:
a) an aryl group optionally substituted with 1 , 2, or 3 R¾; b) a heteroaryl group optionally substituted with 1 , 2, or 3 ¾;
c) a substituent independently selected from the group consisting of hydrogen, alkenyl, alkoxy, alkoxyalkyi, alkoxycarbonyl, alkoxycarbonylalkyl, alkyl, alkynyl, arylalkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, hydroxyalkylene, oxo, heterocycloalkyl, heterocy c I oa I ky I a I ky I , alkylcarbonyl, arylcarbonyl, heteroarylcarbonyl alkyisulfonyl, arylsulfonyl, heteroarylsulfonyl, -(C rC6-alkylene)(NRARB), -C(0)( RARB), -(C,-C6-aIkylene)C(0)(NRARB), -S(0)2(NRARB), and -(Ci-C6-alkylene)S(0)2 NRARB); or a pharmaceutically acceptable salt, solvate or prodrug thereof; and
each R<, is selected from OH, N02, CN, Br, CI, F, I, C ,-C6alkyl, C3~C8cycloalkyl, C2-C8heterocyc loalkyl;
C2-C6alkenyl, alkoxy, alkoxyalkyi, alkoxycarbonyl, alkoxycarbonylalkyl, C2-C6alkynyl, aryl, arylalkyl, C3-C8cycloaIkylalkyI, haloalkoxy, haloalkyl, hydroxyalkylene, oxo, heteroaryl, heteroarylalkoxy, heteroaryloxy, heteroarylthio, heteroarylalkylthio, heterocycloalkoxy, C2- Csheterocycloalkylthio, heteroeyclooxy, heterocyclothio, -NRARB, -(CrC6-alkylene)(NRARB), -C(0)fNRARB), -(C,-C6-alkylene)C(0)(NRARB), -S(0)2(NRARB), and -(C rC6- alkylene)S(0)2(NRARB).
[0042] In certain embodiments are provided compounds of Formula (I) or a therapeutically acceptable salt thereof wherein Rb R2, and R3 are independently selected from a group consisting of hydrogen, alkyl, and halogen; R4 and R5 are hydrogen; 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-O-, -NH, -N(C rQ-alkyl)-, - NC(0)(CrC6-alkyl)-, -N(aryl)-, -N(aryl-C,-C5-alkyl-)-, -N(substituted-aryl-C C6-alkyl-)-, -N(heteroaryl)- , -N(heteroaryl-CrC6-alkyl-)-, -N(substituted-heteroaryl-CrC6-alkyl-)-, -S- and -S(0)q- , wherein q is 1 or 2 and the 3- 10 membered heterocycle ring is optionally substituted with one or more substituentsf A and B are each independently selected from hydrogen, Br, CI, F, 1, CrC6alkyl, C3-C8cycloalkyl, alkoxy, alkoxyalkyi wherein CrC6alkyl, C3-C8cycloaIkyl, alkoxy, alkoxyalkyi are optionally substituted with at least one substituent selected from OH, N02, CN, Br, CI, F, I, C C6alkyl, and Cj-Cgcycloalkyl; 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 R^;
c) a substituent independently selected from the group consisting of hydrogen, alkenyl. alkoxy, alkoxyalkyi, alkoxycarbonyl, alkoxycarbonylalkyl, alkyl, alkynyl, arylalkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, hydroxyalkylene, oxo, heterocycloalkyl, heterocycloalkylalkyl, alkylcarbonyl, arylcarbonyl, heteroarylcarbonyl alkyisul fonyl, arylsulfonyl, heteroarylsulfonyl, -(C,-C6-alkylene)(N RARB), -C(0)(NRARB), -(C , -C6-alky lene)C(0)( RARB), -S(0)2(NRARB), and -(C , -C6-al ky lene)S(0)2(N RARB); and each R„ is selected from OH, N02, CN, Br, CI, F, I, C,-C6alkyl, C3-C8cycloalkyl, C2-C8heterocycloalkyl; C2-C6alkenyl, alkoxy, alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C2-C6alkynyl, aryl, arylalkyl, C3-CgcycloalkyIalkyl, haloalkoxy, haloalkyl, hydroxyalkylene, oxo, heteroaryl, heteroarylalkoxy, heteroaryloxy, heteroarylthio, heteroarylalkylthio, heterocycloalkoxy, C2~ Cgheterocycloalkylthio, heterocyclooxy, heterocyclothio, -NRARB, -(C i -C6-al ky leneX RARB),
-C(0)(NRARB), -(CrC6-alkylene)C(0)(NRARB), -S(0)2(NRARB), and -(C,-C6- atkylene)S(0)2(NRARB);
or a single isomer, stereoisomer, or enantiomer or mixture thereof optionally as a pharmaceutically acceptable salt, solvate or prodrug thereof.
[0043] In certain embodiments are provided compounds of Formula (I) or a therapeutically acceptable salt thereof wherein R R2, R„ R.( and R5 are hydrogen; 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(Cf -C6- alkyl)-, -NC(0)(C ,-C6-alkyl)-, -N(aryl)-, -N(aryl- C C6-alkyl-)-, -N(substituted-aryl- C rC6-alkyl-)-, - N(heteroaryl)-, -N(heteroaryI- Ci-C6-alkyl-)-, -N(substituted-heteroaryl- C rC6-alkyI-)-, -S- and -S(0)q- , wherein q is 1 or 2 and the 3- 10 membered heterocycle ring is optionally substituted with one or more substituents; A and B are each independently selected from hydrogen, Br, CI, F, I, OH, CrC6alkyl, C3- Cgcycloalkyl, alkoxy, and alkoxyalkyl wherein C|-C6alkyl, C3-C8cycloaIkyl, alkoxy, and alkoxyalkyl are optional ly substituted with at least one substituent selected from OH, N02, CN, Br, CI, F, I, CrC0alkyl, and C Cgcycloalkyl, wherein B is not OH; Y and Z are each independently selected from the group consisting of:
a) an aryl group optional ly substituted with I , 2, or 3 R6;
b) a heteroaryl group optional ly substituted with 1 , 2, or 3 R^;
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, heteroary lcarbony 1 alkylsulfonyl, arylsulfonyl, heteroary I su I fony 1 , -(C C6-alkylene)(NRARB), -C(0)( RARB), -(C , -C6-al ky lene)C(0)(N RARB), -S(0)2(NRARB), and -(Ci-C6-aIkylene)S(0)2(NRARB); and
is selected from OH, N02, CN, Br, CI, F, I, CrC6alkyl, C3-C8cycloalkyl, C2-C8heterocycloalkyl; C2- C6alkenyl, alkoxy, alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C2-C6alkynyl, aryl, arylalkyl, C -CgcycloalkylaIkyl, haloalkoxy, haloalkyl, hydroxyalkylene, oxo, heteroaryl, heteroarylalkoxy, heteroaryloxy, heteroarylthio, heteroarylalkylthio, heterocycloalkoxy, C2-C8heterocycloalkylthio, heterocyclooxy, heterocyclothio, -NRARB, -(CrC6-alkylene)(NRARB), -C(0){NRARB), -(Ct-C6- aIkylene)C(0)(NRARB), S(0)2(NRARB), and -(C,-C6-alkylene)S(0)2(NRARB); or a single isomer, stereoisomer, or enantiomer or mixture thereof optionally as a pharmaceutically acceptable salt, solvate or prodrug thereof.
[0044] In one embodiment is a compound of Formula (1) wherein ¾, R¾ R3 are each independently selected from a group consisting of hydrogen, alkyl, and halogen; 4 is hydrogen and R5 is selected from the group consisting hydrogen, alkyl, cycloalkyi, alkoxyalkyl, haloalkyl, hydroxyalkylene, and -(C |-C6-aIkylene)-(NRARB).
[0045] In another embodiment is a compound of Formu la (I) wherein R|, R2, R3 are each independently selected from a group consisting of hydrogen, alkyl, and halogen; R4 and R5 are hydrogen.
[0046] In a further embodiment is a compound of Formula (I) wherein R R2, R3, R4 are each hydrogen and R3 is alkyl.
[0047] In yet another embodiment is a compound of Formula (I) wherein Ri, R?, R3, are each hydrogen; and R5 is methyl.
[0048J In one embodiment is a compound of Formula (I) wherein Rj, R2, and Rj are each hydrogen.
[0049] In one embodiment is a compound of Formula (I) wherein R5 is hydrogen or an alkyl group. In another embodiment, R5 is hydrogen. In a further embodiment, R5 is CrC6 alkyl. In yet a further embodiment, R5 is CH3. In another embodiment, R5 is CH2CH3.
[0050] In another embodiment is a compound of Formula (1) wherein R4 is hydrogen or an alkyl group. In yet another embodiment, R4 is hydrogen.
[0051 ] In another embodiment, the Compound of Formula ( 1) is that where R? is selected from the group consisting of hydrogen, halogen, alkenyl, alkoxy, alkoxycarbonyl, alkyl, cycloalkyi, alkynyl, cyano, haloalkoxy, haloalkyl, hydroxyl, hydroxyalkylene, nitro, -N RARB, -(Ci-C6alkylene)-NRARB, and -C(0)(NRARB). In a further embodiment, R3 is hydrogen. In some embodiments, R| is selected from the group consisting of hydrogen, halogen, alkenyl, alkoxy, alkoxycarbonyl, alkyl, cycloalkyi, alkynyl, cyano, haloalkoxy, haloalkyl, hydroxyl, hydroxyalkylene, nitro, -NRARB, -(C| -C6alkylene)-NRARB, and
-C(0)(NRARB). In a further embodiment, Rj is hydrogen.
[0052] Embodiment A: Another emdodiment is a method of treating multiple myeloma with a
Compound according to Formula Formula (11) is as follows:
Figure imgf000010_0001
Formula (II);
wherein:
Y is an aryl or heteroaryl group optional ly substituted with at least one R(,;
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, CrC5alkyl, C^-Cscycloalkyl, alkoxy, and alkoxyalkyi wherein C|-C6alkyl, C3-Cgcycloalkyl, alkoxy, and alkoxyalkyi are optionally substituted with at least one substituent selected from OH, NO2, CN, Br, CI, F, I, CrC6alkyl, and C3-CgcycIoalkyl, wherein B is not OH;
Re is selected from OH, NO2, CN, Br, CI, F, I, CrC6alkyl, C3-Cgcycloalkyl, Ci-Cgheterocyc loalky 1;
C?-C6alkenyl, alkoxy, alkoxyalkyi, alkoxycarbonyl, alkoxycarbonylalkyl, C?-C6alkynyl, aryl, arylalkyl, C3-C8cycloalkylalkyl, haloalkoxy, haloalkyl, hydroxyalkylene, oxo, heteroaryl, heteroarylalkoxy, heteroaryloxy, heteroarylthio, heteroarylalkylthio, heterocycloalkoxy, C2- Cgheterocyc I oalky 1th i o, heterocyclooxy, heterocyclothio, - RARB, -(C 1 -C6alky lene)(N RARB), -C(0)(NRARB), -(C,-C6alkylene)-C(0)(NRARB), -S(0)2(NRARB), and -(C,- C6alkyIene)-S(0)2(NRARB);
R2 is selected from hydrogen, Br, CI, I, or F;
RA and R are independently selected from the group consisting of hydrogen, CrC6alkyl, C3- 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-, -N H, -N(C C6alkyl)-, -NC(0)(C i -C6a I ky I )-, -NC(0)(C3-Cgcycloalkyl)-, -N(aryl)-, -N(aryl-CrC6alkyl-)-, -N(substituted-aryl-C,-C6alkyl-)-, - N(heteroaryl)-, -N(heteroaryl-CrC6alkyl-)-, -N(substituted-heteroaryl-Ci-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
or a single isomer, stereoisomer, or enantiomer or mixture thereof optionally as a pharmaceutically acceptable salt, solvate or prodrug thereof.
[0053] In another embodiment is a compound of Formula (I) or (II) wherein Y and Z are each independently selected from the group consisting of:
a) a phenyl group optional ly substituted with 1 , 2, or 3 R6;
b) a pyridyl group optionally substituted with 1 , 2, or 3 R¾; and
c) a substituent independently selected from the group consisting of hydrogen, alkoxyalkyi, alkoxycarbonylalkyl, alkyl, arylalkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, oxo, heterocycloalkyl, heterocycloalkylalkyl, alkylcarbonyl, arylcarbonyl, heteroarylcarbonyl, -(C , -Qaiky lene)-( RARB), -C(0)(NRARB), and -(C,-C6alkylene)-C(0)(NRARB),
|0054] In a further embodiment is a compound of Formula (I) or (II) wherein Y and Z are each independently selected from the group consisting of
a) a phenyl group optionally substituted with 1, 2, or 3 ¾;
b) an imidazole group optionally substituted with 1 , 2, or 3 R6; and
c) a substituent independently selected from the group consisting of hydrogen, alkoxyalkyi, alkoxycarbonylalkyl, alkyl, arylalkyl, cycloalkyl. cycloalkylalkyl, haloalkyl, oxo, heterocycloalkyl, heterocycloalkylalkyl, alkylcarbonyl, arylearbonyl, heteroarylcarbonyl, -(C,-C6alkylene)-(NRARB), -C(0)(NRARB), and -(CrC6alkylene)-C(0)(NRARB).
[0055] In a further embodiment is a compound of Formula (1) or (II) wherein Y and Z are each independently selected from the group consisting of
a) a phenyl group optional ly substituted with 1 , 2, or 3 R6;
b) a triazole group optionally substituted with I , 2, or 3 R6; and
c) a substituent independently selected from the group consisting of hydrogen, alkoxyalkyl, alkoxycarbonylalkyl, alkyl, arylalkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, oxo, heterocycloalkyl, heterocycloalkylalkyl, alkylcarbonyl, arylearbonyl, heteroarylcarbonyl, -(C,-C6aikylene)-(NRARB), -C(0)(NRARB), and -(CrC6alkylene)-C(0)(NRARB).
[0056] In a further embodiment is a compound of Formula (I) or (II) wherein Y and Z are each independently selected from the group consisting of
a) a phenyl group optionally substituted with 1 , 2, or 3 R6;
d) an imidazole group optionally substituted with I , 2, or 3 R¾; and
b) a triazole group optional ly substituted with 1 , 2, or 3 Re.
[0057] In one embodiment, the Compound of Formula (I) or (II) is that where R2 is selected from the group consisting of hydrogen, halogen, alkenyl, alkoxy, alkoxycarbonyl, alkyl, cycloalkyl, alkynyl, cyano, ha!oalkoxy, haloalkyl, hydroxy], hydroxyalkylene, nitro, -NRARB, -(CrC6alkylene)-NRARB, and -C(OXNRARB). In a further embodiment R2 is a halogen selected from F, CI, Br, and I. In yet a further embodiment, R2 is fluorine. In one embodiment, R2 is hydrogen.
[0058] Also disclosed herein are compounds of Formula (I) or (II) wherein Z is an aryl group optionally substituted with 1 , 2, or 3 R^; wherein each R> is selected from OH, NO?, CN, Br, CI, F, I, Q- Qalkyl, Q-Cscycloalkyl, C2-C8heterocycloalkyl; C2-C„alkenyl, alkoxy, alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C2-C6alkynyl, aryl, arylalkyl, C3-C8cycloalkylalkyl, haloalkoxy, haloalkyl, hydroxyalkylene, oxo, heteroaryl, heteroarylalkoxy, heteroaryloxy, heteroarylthio, heteroarylalkylthio, heterocycloalkoxy, C2-C8heteiOcycloalkylthio, heterocyclooxy, heterocyclothio, -NRARB, -(Cr
C6alkyleneMN RARB), -C(0)(NRARB), -(C , -Qalky lene)-C(0)( RARB), -S(0)2(NRARB), and -(Cr C6alkylene)-S(0)2(NRARB). In one embodiment is a compound of Formula (I) or (II) wherein Z is an optionally substituted phenyl group. In one embodiment, Z is a phenyl group. In another embodiment, the phenyl group is optionally substituted with at least one R<. selected from OH, N02, CN, Br, CI, F, I, C Qalkyl, Cs-Cgcycloalkyl, G>-C8heterocycIoaIkyl; C2-Qalkenyl, alkoxy, alkoxyalkyl, alkoxycarbonyl, alkoxycarbony lalkyl, C2-C6alkynyl, aryl, arylalkyl, Ci-Cscycloalkylalkyl, haloalkoxy, haloalkyl, hydroxyalkylene, oxo, heteroaryl, heteroarylalkoxy, heteroaryloxy, heteroarylthio, heteroarylalkylthio, heterocycloalkoxy, C2-C8heterocycloalkylthio, heterocyclooxy, heterocyclothio, -NRARB, -(Cr
Qalkylene)- (N RARB), -C(0)(NRARB), -(C C6alkylene)-C(0)(NRARa), -S(0)2( RARB), and -(Cr C6alkyIene)-S(0)2(NRARB). In another embodiment, Re is -(Q -Qalky Iene)-(NRARB). In a further embodiment, R„ is -CH2(NRARB). In a further embodiment, RA is -CH2(NRARB) wherein -NRARB is azetidine, pyrrolidine, piperidine or morpholine. In yet a further embodiment, RA is H or alkyl. In another embodiment, RA is CrC6alkyl. In yet another embodiment, RA is -CH:,. In another embodiment, RB is H or alkyl. In one embodiment, RB is CrC6alkyl. In yet another embodiment, RB is -CH3. In a further embodiment, R6 is -CH2NHCH3. In yet a further embod iment. Re is -CH2NCH3CH3. In one embodiment, R6 is -(CO)heterocycIoalkyl(CO)alkyl. In one embodiment R„ is -(CO)heterocyc loa 1 ky l(CO)alky 1 wherein the heterocyeloalkyl group has at least one heteroatom selected from O, N, and S. In another embodiment, the heterocyeloalkyl group has two N atoms. In a further embodiment, R6 is
-(CO)heterocycloalkyl(CO)alkyl wherein alkyl is selected from methyl, ethyl, n-propyl, iso-propyl, cyclopropyl, n-butyl, iso-butyl, and t-butyl. In one embodiment, the alkyl group is cyclopropyl. In another
embodiment, the alkyl group is iso-propyl. In one embodiment, R6 is
Figure imgf000013_0001
. In another
embodiment, R is
Figure imgf000013_0002
[0059] Presented herein are compounds of Formula (I) or (II) wherein Z is an optionally substituted heteroaryl group. In one embodiment, the heteroaryl group is selected from pyridine, pyrimidine, pyrazine, pyrazole, oxazole, thiazole, isoxazole, isothiazole, 1 ,3,4 -oxadiazole, pyridazine, 1 ,3,5-trazine, 1 ,2,4-triazine, quinoxaline, benzimidazole, benzotriazole, purine, I / -[ l ,2,3]triazolo[4,5- i/]pyrimidine, triazole, imidazole, thiophene, furan, isobenzofuran, pyrrole, indolizine, isoindole, indole, indazole, isoquinoline, quinoline, phthalazine, naphthyridine, quinazoline, cinnoline, and pteridine. In one embodiment, Z is pyridine. In another embodiment, Z is optionally substituted pyridine.
[0060] In certain embodiments are provided compounds of Formu la (I) or a therapeutically acceptable salt thereof wherein Ri, R2. R3 are independently selected from a group consisting of hydrogen, alkyl, and halogen; R.t is hydrogen and R5 is selected from the group consisting hydrogen, alkyl, cycloalkyi, alkoxyalkyi, haloalkyi, hydroxyalkylene, and -(Ci-C6-alkylene)(NRARB); RA and RB are independently selected from the group consisting of hydrogen, alkyl, cycloalkyi, 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 -NH, -N(C,-C6-alkyl)-, -NC(0)(C,-C6-alkyl)-, -N(aryl)-, -N(aryl- C-Q-aikyl-)-, -N(substituted-aryl- C,-C6-alkyl-)-, -N(heteroaryl)-, -N( heteroaryl- C,-C6-alkyl-)-, -N(substituted-heteroaryl- CrC6-alkyI-)-, - S- and -S(0)q- , wherein q is 1 or 2 and the 3- 10 membered heterocycle ring is optionally substituted with one or more substituents.
[0061 ] In one embodiment is a compound of Formula (I) or (II) 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, and I. In one embodiment Rs is F, In one embodiment the phenyl group is substituted with at least one R6 selected from -(C rC6-alkylene)(NRARB), -C(0)(NRARB)carbonyl, -(CrC6-alkylene)C(0)(NRARB), -S(0)2(NRARB), and -(C,-C6- alkyIene)S(0)2(NRARB). In one embodiment R« is -(CrC6-alkylene)(NRARB). 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 CrQalkyl is methylene. In yet a further embodiment RA and RB are each independently hydrogen, Ci-C(,alkyl, or C3-C8cycloalkyl. In one embodiment CrQalkyl is selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl. In one embodiment Q -Qalkyl is methyl. In another embodiment Q-Qalkyl is ethyl. In yet another embodiment Q- Qcycloaikyl is cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl. In a further embodiment Q- Qcyeloalkyl is cyclopropyl. In yet a further embodiment R<> is hydroxyalkylene. In one embodiment hydroxyalkylene is selected from -CH2OH, -CH2CH2OH, -CH2CH,CH2OH, -CH(OH)CH3,
-CH(OH)CH2CHj, -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(C|-C6alkyl)-. In yet another embodiment the hetero functionality is -N(CrC6alkyl). In a further embodiment Q-Qalkyl is selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl. In yet a further embodiment Q-Qalkyl 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 Q- Qalkyl selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl. In yet a further embodiment Q-Qalkyl 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 Q-Qalky selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-buty l. In yet a further embodiment Q-Qalkyl 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 ¾ selected from Br, CI, F, and I. In a further embodiment R,-, 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 -(Q-Q-alkylene)(NRARB), -C(0)(NRARB), -(Q - C6-alkylene)C(0)(NRARB), -S(0)2(NRARB), and -(Ci-C6-alkylene)S(0)2(NRARB). In another embodiment R6 is -(Q-Q-aIkylene)(>JRARB). In yet another embodiment C C(Jalkyi is selected from methylene, ethylene, n-propylene, iso-propylene, n-butylene, iso-butylene, and tert-butylene. In yet a further embodiment CrC6alkyl is methylene. In a further embodiment RA and RB are each independently hydrogen, Q-Qalkyl, or Q-Qcycloalkyl. In one embodiment Q-Qalkyl is selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl. In another embodiment C|-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, and -N(CrQalkyl). In a further embodiment the hetero functionality is -N(C Qalkyl). In one embodiment C rC„alkyI is selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl. In yet a further embodiment Q-Qalkyl is methyl. In one embodiment Z is a heteroaryl group optional ly 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, benzoth iophene, 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 Q-Qalkyl selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso- butyl, and tert-butyl . In yet a further embodiment Q-Qalkyl 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 Q- Qalkyl selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl. In yet a further embodiment C Qal yl 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.
[0062] In one embodiment is a compound of Formula (I) or (II) wherein A is hydrogen. In another embodiment A is Q-Qalkyl. In a further embodiment, A is selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, tert-butyl, n-pentyi, 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 w ith 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 CrCgcycloalkyl. 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 C|-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 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 Q-Qcycloalk l. 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, is a compound of Formula (I) wherein A is hydrogen and B is selected from Br, CI, F, I, Q-Qalkyl, Q-Qcycloalkyl, alkoxy, alkoxyalkyl wherein Q-Qalkyl, Q-Qcycloalkyl, alkoxy, alkoxyalkyl are optionally substituted with at least one substituent selected from OH, N02, CN, Br, CI, F, I, CrQalkyl, and CrCgcycloalkyl. In another embodiment, is a compound of Formula (I) wherein B is hydrogen and A is selected from Br, CI, F, I, Q-Qalkyl. C6-Qcycloalkyl, alkoxy, alkoxyalkyi wherein d-Qalkyl, C3-Cgcycloalkyl, alkoxy, alkoxyalkyi are optionally substituted with at least one substituent selected from OH, N02, CN, Br, CI, F, I, CrC6alkyl, and C3-C8cycloalkyl. 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-Qcycloalkyl, alkoxy, and alkoxyalkyi wherein Ci-Cealkyl, C3- Cscycloalkyl, alkoxy, and alkoxyalkyi are optionally substituted with at least one substituent selected from OH, N02, CN, Br, CI, F, I, C,-C6alkyl, and C3-C8cycloalkyl,
[0063] Also disclosed herein are compounds of Formula (I) or (II) wherein Y is an aryl group optional ly substituted with 1 , 2, or 3 R6; wherein each ¾ is selected from OH, N02, CN, Br, CI, F, I, C C6alkyl, C3-C8cyeloalkyl, and C2-C3heterocycloalkyl, C\-G,alken l. alkoxy, alkoxyalkyi, alkoxycarbonvl, alkoxycarbonylalkyl, C2-C6alkynyl, aryl, arylalkyl, C3-C8cycloalkylalkyl, haloalkoxy, haloalkyl, hydroxyalkylene, oxo, heteroaryl, heteroarylalkoxy, heteroaryloxy, heteroarylthio, heteroarylalkylthio, heterocycloalkoxy, C2-C8heterocycloalkylthio, heterocyclooxy, heterocyclothio, -NRARB. -(C
Qalkylene)- (NRARB), -C(0)(NRARB), -(C,-C6alkylene)-C(0)(NRARB), -S(0)2(NRARB). and -(C ,- C6alkylene)-S(0)2(NRARB). In one embodiment is a compound of Formula (I) or (II) wherein Y is an optionally substituted phenyl group. In one embodiment, Y is a phenyl group. In another embodiment, the phenyl group is optionally substituted with at least one R<, selected from OH, N02, CN, Br, CI, F, 1, Cr Qalkyl, C3-C8cycloalkyl, C2-C8heterocycloalkyl, C2-Qalkenyl, alkoxy, alkoxyalkyi, alkoxycarbonyl, alkoxycarbonylalkyl, C -C6alkynyl, aryl, arylalkyl, C3-C8cycloalkylalkyl, haloalkoxy, haloalkyl, hydroxyalkylene, oxo, heteroaryl, heteroarylalkoxy, heteroaryloxy, heteroarylthio, heteroarylalkylthio, heterocycloalkoxy, C2-Qheterocycloalkylthio, heterocyclooxy, heterocyclothio, -NRARB, -(Q- C6alkylene)-(NRARB), (NRARB)carbonyl, -(C rC6alkylene)-C(0)(NRARB), -S(0)2(NRARB), and -(Cr C6alkylene)-S(0)2(NRARB). In a further embodiment, ¾ is -CH2(NRARB). In yet a further embodiment, RA is H or alkyl. In another embodiment, RA is CrC6alkyI. In yet another embodiment, RA is -CH3. In another embodiment. RB is H or alkyl. In one embodiment, RB is CrC6alkyl. In yet another embodiment, RB is -Cl h. In a further embodiment, RA is -CH2NHCH3. In yet a further embodiment, R(, is
-CH2NCH3CH3. In one embodiment, R<-, is -(CO)heterocycloalkyl(CO)alkyl. In one embodiment R<, is -(CO)heterocycloalkyl(CO)alkyl wherein the heterocycloalkyl group has at least one heteroatom selected from O, N, and S. In another embodiment, the heterocycloalkyl group has two N atoms. In a further embodiment, R<, is -(CO)heteroeycloalkyl(CO)alkyI wherein alkyl is selected from methyl, ethyl, n- propyl, iso-propyl, cycloprop l, n-butyl, iso-butyl, and t-butyl. In one embodiment, the alkyl group is er embodiment, the alkyl group is iso-propyl. In one embodiment, R6 is
Figure imgf000016_0001
In another embodiment,
[G064J Presented herein are compounds of Formula (I) or (II) wherein Y is an optionally substituted heteroaryl group. In one embodiment, the heteroaryl group is selected from pyridine, pyrimidine, pyrazine, pyrazole, oxazole, thiazole, isoxazole, isothiazole, 1 ,3,4 -oxadiazole, pyridazine, 1,3,5-trazine, 1 ,2,4-triazine, quinoxaline, benzirnidazole, benzotriazole, purine, lH-[ l,2,3]triazoio[4,5- i ]pyrimidine, triazole, imidazole, thiophene, furan, isobenzofuran, pyrrole, indolizine, isoindole, indole, indazole, isoquinoline, quinoline, phthalazine, naphthyridine, quinazoline, cinnoline, and pteridine. In one embodiment, Y is pyridine. In another embodiment, Y is optionally substituted pyridine. In one embodiment, Y is imidazole. In another embodiment, Y is optionally substituted imidazole. In one embodiment, Y is triazole. in another embodiment, Y is optionally substituted triazole.
[0065] In one embodiment, the Compound of Formula (I) or (II) is that where Y is a substituent independently selected from the group consisting of hydrogen, alkenyl, alkoxy, alkoxyalkyl, alkoxycarbonyl. alkoxycarbonylalkyl, alkyl, alkynyl, arylalkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, hydroxyalkylene, oxo, heterocycloalkyi, heterocycloalkylalkyl, alkylcarbonyl, arylcarbonyl,
heteroarylcarbonyl alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, -(C|-Qalkylene)-(NRARB),
-C(0)(NRARB), -(C rC6alkylene)-C(0)(NRARB), -S(0)2(NRARB), and -(C ,-C6alky lene)-S(0)2(N RARB). In one embodiment, Y is alkyl. In another embodiment, Y is C[-C6 alkyl. In a further embodiment, Y is selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl or tert-butyl. In another embodiment, Y is iso-propyl.
[0066] Also disclosed herein are compounds of Formu la (I) or (II) wherein Y is an optionally substituted heterocycloalkyi group. In one embodiment, the heterocycloalkyi group is selected from 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, pyrazol idinyl, imidazolinyl, imidazolidinyl, 3-azabicyclo[3.1.OJhexanyl, 3-azabicyclo[4.1 .OJheptanyl, 3H- indolyl and quinolizinyl. In another embodiment the heterocycloalkyi group is selected from pyrrolidinyl, imidazolidinyl, piperidinyl, piperazinyl, pyrazolidinyl, tetrahydrofuranyl, tetrahydroth iopheny 1, 1 ,3- oxathiolanyl, indolinyl, isoindolinyl, morpholinyl, and pyrazol inyl. In another embodiment, the heterocycloalkyi group is piperidinyl.
[0067] In another aspect is a compound of Formula (IA):
Figure imgf000017_0001
Formula (IA)
or a single isomer, stereoisomer, or enantiomer or mixture thereof optionally as a pharmaceutically acceptable salt, solvate or prodrug thereof wherein 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, - RARB. -(Ci-C6alkylene)-NRA B, and -C(0)(NRARB);
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 lieteroatoms or hetero functionalities selected from the group consisting of -O-. - H, -N(C,-C6-alkyl)-, -NC(0)(C,-Q-alkyl)-, -N(aryl)-, -N(aryl-CrC6- alkyl-)-, -N(substituted-ary -C C6-alkyl-)-, -N(heteroaryl)-, -N(heteroaryl-C,-C6-alkyl-)-, -N(substituted- heteroary 1-C i -C6-alky 1-)-, -S- and -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 R3 are each independently selected from the group consisting of hydrogen, alkyl, cycloalkyl, alkoxyalkyi, haloalkyl, hydroxyalkylene, and -(Q- C6alkylene)- (NRARB);
A and B are each independently selected from hydrogen, Br, CI, F, I, OH, C| -C6alkyl, C.rQcycloalkyl, alkoxy, and alkoxyalkyi wherein CrC6alkyl, C3-C8cycloalkyl, alkoxy, and alkoxyalkyi are optionally substituted with at least one substituent selected from OH, N02, CN, Br, CI, F, I, CpCfjalkyl, and C3- Cgcycloalkyl, wherein B is not OH;
Y is selected from the group consisting of:
a) an aryl group optionally substituted with 1 , 2, or 3 substituents R$; is selected from OH, N02, CN, Br, CI, F, I, C C6alkyl, C3-C8cycloalkyl, C2-C8heterocycloalkyl; C2-C6alkenyl, alkoxy, alkoxyalkyi, alkoxycarbonyl, alkoxycarbonylalkyl, C2-C6alkynyl, aryl, arylalkyl, C3- Cgcycloalkylalkyl, haloalkoxy, haloalkyl, hydroxyalkylene, oxo, heteroaryl, heteroarylalkoxy, heteroaryloxy, heteroarylthio, heteroarylalkylthio, heterocycloalkoxy, C2-C8heterocycloalkylthio, heterocyclooxy, heterocyclothio, -NRARB, -(C,-C6alkylene)-(NRARB), -C(0)(NRARB), -(Cr C6alkylene)-C(0)(NRARB), -S(0)2(NRARB), and -(C rC6alkylene)-S(0)2(NRARB);
b) a heteroaryl group optionally substituted with 1 , 2, or 3 substituents R6; R« is selected
independently from the group consisting of alkenyl, alkoxy, alkoxyalkyi, alkoxycarbonyl, alkoxycarbonylalkyl, alkyl, alkynyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, cyano, haloalkoxy, haloalkyl, halogen, hydroxyl, hydroxyalkylene, nitro, oxo, heteroaryl,
heteroarylalkoxy, heteroar loxy, heteroarylthio, heteroarylalkylthio, heterocycloalkyl, heterocycloalkoxy, heterocycloalkylthio, heterocyclooxy, heterocyclothio, -NRARB, -(C(- C6alkylene)-(NRARB), -C(0)( RARB), -(C C6alkylene)-C(0)( RARB), -S(0)2(NRARB), and -(C r C6alkyIene)-S(0)2(NRARB);
c) a substituent independently selected from the group consisting of hydrogen, alkenyl, alkoxy, alkoxyalkyi, alkoxycarbonyl, alkoxycarbonylalkyl, alkyl, alkynyl, arylalkyl. cycloalkyl, cycloalkylalkyl, haloalkyl, hydroxyalkylene, oxo, heterocycloalkyl, heterocycloalkylalkyl, alkylcarbonyl, arylcarbonyl, heteroarylcarbonyl alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl. -(erC6aikylene)-(NRARB), -C(0)(NRARB), -(C,-C6alkylene)-C(0)(NRARB), -S(0)2(NRAR8), and -(Ci -C6alky lene)-S(0)2(NRARB); and
n is an integer from 0-4; or a pharmaceutically acceptable salt, solvate or prodrug thereof.
[0068] In another ) having the structure:
Figure imgf000019_0001
or a single isomer, stereoisomer, or enantiomer or mixture thereof optionally as a pharmaceutically acceptable salt, solvate or prodrug thereof.
[0069] In one embodiment is a compound of Formula (IA), (IA 1 ), or (IA2) wherein Y is an aryl group. In another embodiment, Y is a heteroaryl group. In a further embodiment, the aryl group is a phenyl group. In yet a further embodiment is a compound of Formula (IA), (IA 1 ), or (IA2) wherein the phenyl group is substituted with at least one Re. In yet another embodiment the phenyl group is substituted with at least one R<, selected from Br, C I, F, and I. In one embodiment R6 is F. In one embodiment is a compound of Formula (IA), (IA 1 ), or (IA2) wherein the phenyl group is substituted with at least one R6 selected from -(C,-C6alkylene)(NRARB), -C(0)(NRARB), -(C rC6alkylene)C(0)(NRARB), -S(0)2(NRARB), and -(Ci-C6alkylene)S(0)2(NRARB). In one embodiment R6 is -(C C6alkylene)(NRARB). In another embodiment C|-C6alkylene is selected from methylene, ethylene, n-propylene, iso-propylene, n-butylene, iso-butylene, and tert-butylene. In yet another embodiment C|-C(,aikylene is methylene. In yet a further embodiment RA and RB are each independently hydrogen, CrC6alkyl, or C3-C8cycloalkyl. In one embodiment CrQalkyl is selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert- butyl. In one embodiment Ci-C6alkyl is methyl. In another embodiment C|-C6alkyl is ethyl. In one embodiment is a compound of Formula (IA) , (IA 1 ), or (IA2) wherein Cj-Cgcycloalkyl is cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl. In a further embodiment C3-C8cycloalkyl is cyclopropyl. In one embodiment is a compound of Formula (IA), (IA 1 ), or (IA2) wherein Re is hydroxyalkylene. In one embodiment hydroxyalkylene is selected from -CH2OH, -CH2CH2OH, -CH2CH,CH2OH, -CH(OH)CH:„ -CH(OH)CH2CH?, -CH2CH(OH)CH3. and -CH2CH2CH,CH2OH. 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(CrC6alkyl). 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-biityl. In yet a further embodiment CrC6alkyI is methyl.
[0070] In one embodiment is a compound of Formula (IA), (IA 1 ), (IA2), or (II) wherein Y is a heteroaryl group optionally substituted with at least one Rg. In another embodiment the heteroaryl group is selected from furan, pyridine, pyrimidine, pyrazine, imidazole, thiazole, isothiazole, pyrazole, triazole, pyrrole, thiophenc, oxazole, isoxazole, 1 ,2,4-oxadiazole, 1 ,3,4-oxadiazole, 1,2,4-triazine, indole, benzothiophene, benzo im idazole, 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 CrC6alkyl selected from methyl, ethyl, n-propyl, iso-propyl, n- butyl, iso-butyl, and tert-butyl. In yet a further embodiment Ci -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 Ci-C6alkyl selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert- butyl. In yet a further embodiment C Csalkyl is methyl.
[0071] In one embodiment is a compound of Formula (IA), (IA 1), or (IA2) wherein A and B are each independently selected from hydrogen, Br, CI, F, I, OH, C|-Qalkyl, C C8cycloalkyl, alkoxy, alkoxyalkyl wherein Ci-C6alkyl, CrC8cycloalkyl, alkoxy, alkoxyalkyl are optionally substituted with at least one substituent selected from OH, N02, CN, Br, CI, F, I, C rC6alkyl, and C3-C8cycloalkyl and B is not OH. In another embod iment the Compound of Formula (IA), (IA L ), or (IA2) is that wherein A and B are hydrogen.
[0072] In one embodiment is a compound of Formula (I), (IA), (IA 1 ), ( 1A2), and (II) wherein Y is a heteroaryl group 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 another embodiment, Y is an imidazole group. In yet another embodiment, the imidazole group is substituted with a CpQalkyl group. In another embodiment, the Q - C6alkyl group is methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl. In a further embodiment Ci-C6alkyl is methyl. In another embodiment is a compound of Formula (I), (IA), (IA I ), ( 1 A2), and (II) wherein Y is a substituted imidazole group and Z is selected from an aryl group or a heteroaryl group. In a further embodiment, Z is an aryl group. In yet a further embodiment, the aryl group is a phenyl group. In yet a further embodiment, the aryl group is a phenyl group substituted by a halogen. In yet a further embodiment Z is a heteroaryl group. In another embodiment, the heteroaryl group is 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 a further embodiment, the heteroaryl group is imidazole. In another embodiment, the im idazole group is substituted with a C C6alkyl group. In another embodiment, the C C6alkyl group is methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl. In a further embodiment CrC6alkyI is methyl.
[0073] In another embodiment is a compound of Formula (I), (IA), (IA I ), ( 1 A2), and (II) wherein Y is a triazole group. In yet another embodiment, the triazole group is substituted with a C C(,alkyl group. In another embodiment, the - alk l group is methyl, ethyl, n-propyl, iso-propyl, n- but l, iso-butyl, and tert-butyl. In a further embodiment CrC6alkyl is methyl. In another embodiment is a compound of Formula (I), (IA), (IA l ), ( 1 A2), and (II) wherein Y is a substituted triazole group and Z is selected from an aryl group or a heteroaryl group. In a further embodiment, Z is an aryl group. In yet a further embodiment, the aryl group is a phenyl group. In yet a further embodiment, the aryl group is a phenyl group substituted by a halogen. In yet a further embodiment Z is a heteroaryl group. In another embodiment, the heteroaryl group is furan. pyridine, pyrimidine, pyrazine, imidazole, thiazole, isothiazole, pyrazo le, 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 a further embodiment, the heteroaryl group is triazole. In another embodiment, the triazole group is substituted with a CrC6alkyl group. In another embodiment, the C |-C6alkyl group is methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl. In a further embodiment C C(,alkyl is methyl.
d selected from
Figure imgf000021_0001
or a single isomer, stereoisomer, or enantiomer or mixture thereof optionally as a pharmaceutically acceptable salt, solvate or prodrug thereof.
[0075] In one embodiment is a compound of Formula (I), (ΠΑ), (IA l ), (IA2), or (II) 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 R6 selected from Br, CI, F, and I. In one embodiment RA is F. In one embodiment the phenyl group is substituted with at least one R„ selected from -(Cr
C6alkylene)(NRARB), -C(0)(NRARB), -(CrC6alkylene)C(0)(NRARB), -S(0)2(NRARB), and -(Cr
C6alkylene)S(0)2(NRARB). In one embodiment R« is -(CrC6alkylene)(NRARB). In another embodiment C| -C,,alkylene is selected from methylene, ethylene, n-propylene, iso-propylene, n-butylene, iso-butylene, and tert-butylene. In yet another embodiment C :-C„alkylene is methylene. In yet a further embod iment RA and RB are each independently hydrogen, Ci-C6alkyl, or C3-CgcycloaIkyl. In one embodiment Ci-Qalkyl is selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl. In one embodiment Ci -C6alkyl is methyl. In another embodiment C C6alkyl is ethyl. In yet another embodiment C3- Cgcycloalkyl is cyclopropyl, eye lo butyl, cyclopentyl, and cyc!ohexyl. In a further embodiment C\- Cgcycloalkyl is cyclopropyl. In yet a further embodiment RA is hydroxyalkylene. I n one embodiment hydroxyalkylene is selected from -CH2OH, -CH2CH2OH, -CH2CH2CH2OH, -CH(OH)CH:
-CH(OH)CH,C¾, -CH2CH(OH)CH-, and -CH2CH2CH2CH2OH. In another embodiment RA and RB taken together with the nitrogen to wh ich they are attached form a 6 membered heterocycle ring having 1 heteroatom or hetero functionality selected from the group consisting of -O-, -NH-, or -N(C C6alkyl)-. In yet another embodiment the hetero functionality is -N(CrC6alkyl)-. In a further embodiment Cj-Ceaikyl is selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl. In yet a further embodiment Ci-C6alkyl is methyl.
[0076] In one embodiment is a compound of Formula (I), (IIA), (IA 1 ), (IA2), or (II) wherein 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 ¾ selected from Br, CI, F, and I. In a further embodiment R¾ 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 -(C,-C6alkylene)(NRARB), -C(0)(NRARB), -(CrC6alkylene)C(0)(NRARB), -S(0)2(NRARB), and (NRARB)suifonylalkylene. In another embodiment R6 is -(C C6aIkylene)(NRARB). In yet another embodiment C rC6alkylene is selected from methylene, ethylene, n-propylene, iso-propylene, n-butyiene, iso-butylene, and tert-butyiene. In yet a further embodiment CrC6aikyIene is methylene. In a further embodiment RA and RB are each independently hydrogen, Ci-C6aiky l, or C3-C8cycloalkyl. In one embodiment C|-C6alkyl is selected from methyl, ethyl, n-propyi, iso-propyl, n-butyl, iso-butyl, and tert- butyl. In another embodiment CrC6alkyl 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(C rC6alkyl)-. In a further embodiment the hetero functionality is— (C|-C6alkyl). In one embodiment Ci-Cftalkyl is selected from methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl. In yet a further embodiment Cr Qalkyl is methyl. In a further embodiment, is -CH2(NRARB) wherein NRARB is azetidine, pyrrolidine, piperidine or morpholine. 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.
10077] In one embodiment is a compound of Formula (I), (IA), (IA1 ), (IA2), or (II) wherein A and B are hydrogen. In another embodiment A and B are independently selected from hydrogen and Q - Qalkyl.
(0078] In a further embodiment is a compound of Formu la (I), (IA), (IA 1 ), (IA2), or (II) wherein
Z is aryl and Y is independently selected from the group consisting of
a) phenyl group optionally substituted with 1 , 2, or 3 R<, ;
b) a imidazole group optionally substituted with 1 , 2, or 3 Re;
c) a triazole group optionally substituted with 1 , 2, or 3 R(>; and
d) a substituent independently selected from the group consisting of hydrogen, alkoxyalkyl, alkoxycarbonylalkyl, alkyl, arylalkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, oxo, heterocycloalkyl, heterocycloalkylalkyl, aikylcarbonyl, arylcarbonyl, heteroarylcarbonyl, -(C ,-C6alkylene)(NRARB), and -C(0)(NRARB).
[0079) In a further embodiment is a compound of Formula (I), (IA), (IA 1 ), (IA2), or (II) wherein
Z is phenyl and Y is independently selected from the group consisting of
a) phenyl group optionally substituted with 1 , 2, or 3 R6 ;
b) a imidazole group optionally substituted with 1 , 2, or 3 R¾; c) a triazole group optionally substituted with 1 , 2, or 3 ¾; and
d) a substituent independently selected from the group consisting of hydrogen, alkoxyalkyl, alkoxycarbonylalkyi, alkyl, arylalkyl, cycloalkyl, cycloalkyialkyl, haloalkyl, oxo, heterocycloalkyl, heterocycloalkylalkyi, alkylcarbonyl, aryicarbonyl, heteroarylcarbonyi, -(CrC6alkylene)(NRARB), and -C(0)(NRARB).
10080] In a further embodiment is a compound of Formula (I), (IA), (IA 1 ), (IA2), or (II) wherein
Z is phenyl substituted with 1 , 2, or 3 R<, and Y is independently selected from the group consisting of a) phenyl group optionally substituted with 1 , 2, or 3 R6 ;
b) a imidazole group optional ly substituted with 1 , 2. or 3 R„;
c) a triazole group optionally substituted with 1 , 2, or 3 R6; and
d) a substituent independently selected from the group consisting of hydrogen, alkoxyalkyl, a 1 koxy carbony I a 1 ky I , alkyl, arylalkyl, cycloalkyl, cycloalkyialkyl, haloalkyl, oxo, heterocycloalkyi, heterocycloalkylalkyi, alkylcarbonyl, aryicarbonyl, heteroarylcarbonyi, -(C C6alkyiene)(NRARB), and -C(0)(NRARB).
[00811 In a further embodiment, the Compound of Formula (I), (IA), (IA 1 ), (IA2), or (II) is that where 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-butyi, 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 a further embodiment, A is OH. 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, A is hydrogen. In a further embodiment, B is hydrogen. In a further embodiment, B is C C6 lkyI. In a further embodiment, B is selected from methyl, ethyl, n-propyi, iso- propyl, n-butyl, iso-butyl, tert-butyl, n-pentyl, and n-hexyl. In yet another embodiment, methyi, ethyl, n- propyl, iso-propyl, n-butyi, iso-butyl, tert-butyl, n-pentyl, and n-hexyl are optionally substituted with OH, O2, 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 Cj-Cgcycloalkyl. In another embodiment, B is cyclopropy l, cyclobutyl, cyclopentyl, or cyclohexyl. In one embodiment, A is substituted with OH, N02, or CN. In a further embodiment, is a compound of Formula (I), (IA), (IA 1 ), (IA2), or (II) wherein A is hydrogen and B is selected from Br, CI, F, I, Ct-C6alkyl, Cj-Cgcycloalkyl, alkoxy, and alkoxyalkyl wherein Ci-C6alkyl, C3-C8cycloalkyl, alkoxy, and alkoxyalkyl are optionally substituted with at least one substituent selected from OH, N02, CN, Br, CI, F, I. Ci-C6alkyl, and Cj-Cgcyctoalkyl. In another embodiment, is a compound of Formula (II) wherein B is hydrogen and A is selected from Br, CI. F, I, Ct- Qalkyl, C3-C8cycloalkyl, alkoxy, and alkoxyalkyl wherein C rC6alkyl, Cj-Cgcycloalkyl, alkoxy, and alkoxyalkyl are optionally substituted with at least one substituent selected from OH, NO?, CN, Br, C I, F, I, Ct-C6alkyl, and Cj-Cgcycloalkyl. 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, OH, C rC6alkyl, C3-CgcycioaikyI, alkoxy, and alkoxyalkyl wherein CpQalkyl, C-rQcycloaikyl, alkoxy, and alkoxyalkyl are optionally substituted with at least one substituent selected from OH, N02, CN, Br, CI, F, I, CrC5alkyl, and Q-Qcycloalkyl wherein B is not OH.
[0082] Also described herein are stereoisomers of compounds of Formula (I), (IA), or (II), such as enantiomers, diastereomers, and mixtures of enantiomers or diastereomers. In one embodiment is a
Figure imgf000024_0001
HA IIS IIC IID
wherein:
Y is an aryl or heteroaryl group optionally substituted with at least one R^,;
Z is an aryl group optionally substituted with at least one R«;
A and B are each independently selected from hydrogen, Br, CI, F, I, OH, C|-C6alkyl, C^-Cscycioaikyl, alkoxy, and alkoxyalkyl wherein Ci-Cealkyl, CrC8cycloalkyl, alkoxy, and alkoxyalkyl are optionally substituted with at least one substituent selected from OH, N02, CN, Br, Cl, F, I, d-Qalkyl, and C3- Cgcycloalkyl, wherein B is not OH;
R<, is selected from OH, NO,, CN, Br, CI, F, I, C,-C(,alkyl. C3-C8cycIoalkyl, C2-C8heterocycloaikyl; C2-C6alkenyl, alkoxy, alkoxyalkyl, alkoxycarbon l, alkoxycarbonylalkyl, C2-C6alkynyl, aryl, arylalkyl, Ci-Cgcycloalkylalkyl, haloalkoxy, haloalkyi, hydroxyalkylene, oxo, heteroaryl, heteroarylalkoxy, heteroaryloxy, heteroarylthio, heteroaryialkylthio, heterocycloalkoxy, C2-C8heterocycloalkylthio, heterocyclooxy, heterocyclothio, -NRARB, -(C|-C6aikylene)(NRARB), -C(0)(NRARB), -(C,- C6alkylene)C(0)(NRARB), -S(0)2(NRARB), and -(C C6alkylene)S(0)2(NRAR8);
R is selected from hydrogen, Br, Cl, I, or F;
RA and RB are independently selected from the group consisting of hydrogen, Ci-C6alkyl, C3- Qcycloalkyl, 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|-C()aIkyl)-, -NC(0)(C 1 -C6alky 1)-, - NC(0)(C3-C8cycloalkyl)-, -N(aryl)-, -N(aryl-CrC6alkyl-)-, -N(substituted-ar l-CrC6alkyl-)-, - N(heteroary!)-, -N(heteroary 1-C ) -Qalky I-)-, -N(substituted-lieteroaryi-CrC6alkyl-)-, -S- and -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.
[0083] In one embodiment is the stereoisomer of the compound of Formula (HA), (IIB), (IIC), or
(I ID), having the substituents shown above, wherein R2 is fluorine. In another embodiment is the compound of Formula (IIA), ( I IB), (IIC), or (IID), having the substituents shown above, wherein Y is an imidazole group. In another embodiment is the compound of Formula (II) shown above, having the substituents shown above, wherein the imidazole group of Y is substituted with a C|-C6alkyl group. In a further embodiment, the CpQalkyl is methyl. In yet another embodiment is a compound of Formula (IIA), (IIB), (IIC), or (IID), having the substituents shown above, wherein Y is a triazole group. In another embodiment is the compound of Formula (IIA), (IIB), (IIC), or (IID), having the substituents shown above, wherein the triazole group of Y is substituted with a d-C6alkyl group. In a further embodiment, the Q-Cealkyl is methyi. In yet a further embodiment is the compound of Formula (IIA), (IIB), (IIC), or (IID), having the substituents above, wherein the Y group is an aryl group. In a further embodiment is the compound of Formula (II) shown above, having the substituents shown above, wherein the aryl group of Y is a phenyl group. In a further embodiment, the phenyl group is substituted with a halogen. In yet a further embodiment, the halogen is F. In yet another embodiment the halogen is selected from Br, CI, and I. In yet another embodiment, is the compound of Formula (IIA), (IIB), (IIC), or (IID), having the substituents shown above, wherein Z is an aryl group. In yet another embodiment is the compound of Formula (II) shown above, having the substituents shown above, wherein the aryl group of Z is a phenyl group. In a further embodiment, the phenyl group of Z is substituted with a halogen, selected from F, Br, CI, and I. In yet another embodiment, the phenyl group of Z is substituted with F. In yet a further embodiment the phenyl group of Z is substituted with -C|-C6alkylene(NRARB). In yet a further embodiment, the C rC6alkylene group is methylene. In yet another embodiment NRARB is azetidine.
[0084] In another embodiment, the compound, optionally as a single stereoisomer or mixture of stereoisomers thereof and additionally optionally as a pharmaceutically acceptable salt thereof, is selected from
Figure imgf000025_0001
W 201
Figure imgf000026_0001
Figure imgf000027_0001
Figure imgf000028_0001
Figure imgf000029_0001
Figure imgf000030_0001
Figure imgf000031_0001
Figure imgf000032_0001
Figure imgf000033_0001
W
Figure imgf000034_0001
Figure imgf000035_0001
Figure imgf000036_0001
Figure imgf000037_0001
Cmpd
Structure Name
No.
H
8,9-di(pyridin-3-y!)-8,9-dihydro-2H-pyrido[4,3,2-
86
c/e|phthalazin-3(7//)-one
H
8,9-di(pyridin-2-yI)-8,9-dihydro-2H-pyrido[4,3,2-
87
i/e]phthalazin-3(7H)-one
H
9-isopropyl-8-phenyl-8,9-dihydro-2j¾r-pyrido[4,3,2-
88
c/e]phthalazin-3(7/f)-one
9-(4-((methylamino)methyl)phenyl)-8-pheny 1-8,9-
89
dihydro-2H-pyrido[4,3,2-£fe]phthalazin-3(7 )-one
9-(4-((dimethylamino)methyl)phenyl)-8-phenyl-8,9-
90
dihydro-2 /-pyrido[4,3,2-i/e]phthalazin-3(7 f)-one
H
9-(3-((methylamino)methyl)phenyl)-8-phenyl-8,9-
91
dihydro-2/- -pyrido[4.3.2-i/e ]phthalazin-3(7//)-one
H
8-(4-((meth lamino)methyl)phenyl)-9-pheny 1-8,9-
92
dihydro-2H-pyrido[4,3,2-i/e]phthalazin-3(7H)-one
H H
Figure imgf000039_0001
Figure imgf000040_0001
Figure imgf000041_0001
Figure imgf000042_0001
Figure imgf000043_0001
Figure imgf000044_0001
Figure imgf000045_0001
Figure imgf000046_0001
Figure imgf000047_0001
Figure imgf000048_0001
Figure imgf000049_0001
Figure imgf000050_0001
Figure imgf000051_0001
Figure imgf000052_0001
Figure imgf000053_0001
Figure imgf000054_0001
Cmpd
Structure Name
No.
H
ll oHjj Γ 9-(4-fluorophenyl)-9-hydroxy-8-( 1 -methyl- 1 //-imidazol-
203 2-yl)-8,9-dihydro-2 /-pyrido[4,3,2-^]phthalazin-3(7/i)- one
[0085] In another embodiment is a compound selected from:
(8S,9R)-5-fluoro-8-(4-fluorophenyl)-9-(l-methyI-lH-l,2,4-triazol-5-yI)-8,9-dihydro-2 -pyrido[4,3,2- de] phthalazin-3 ( 7//)-one;
(8/?,9>S>5-nuoro-8-(4-nuorophenyi)-9-{ 1 -methyl- 1 //- 1 ,2.4-triazol-5-yl)-8.9-dihydro-2//-pyrido[4.3.2- <sfe]phthalazin-3(7//)-one;
(&S,9R)- 8-(4-fluorophenyl)-9-(l -methyl-l -imidazol-2-yl)-8,9-dihydro-2//-pyrido[4,3,2-<:/e]phthalazin- 3(7#)-one;
(8/?,95> 8-(4-fluorophenyl)-9-(l -methyl-l -imidazol-2-yl)-8,9-dihydro-2/-pyrido[4,3,2-c/e]phthalazin- 3(7#)-one;
(8S,9R> 8-(4-fluorophenyl)-9-( 1 -methyl-lH-l,2,4-triazol-5-yl)-8,9-dihydro-2H-pyrido[4,3,2- c/e]phthalazin-3(7H)-one;
(8R.9S)- 8-(4-fluorophenyl)-9-(l -methyl-l//-l,2,4-tnazol-5-yl)-8,9-dihydro-2/f-pyrido[4,3,2- <r/e]phthalazin-3(7/f)-one;
(8S*,9R 8-(4-(azetidin-l-ylmethyl)phenyl)-9-(4-fluorophenyl)-8,9-dihydro-2H-pyrido[4,3,2- de]phthalazin-3(7H)-one;
(SR.9S)- 8-(4-(azetidin-l-ylmethyl)phenyl)-9-(4-fluoropheny!)-8,9-dihydro-2H-pyrido[4J,2- de]phthalazin-3(7H)-one;
(8S',9i?)-5-fluoro-9-(l-methyl-l//-imidazol-2-yl)-8-phenyl-8,9-dihydro-2i-pyrido[4,3,2-£/e]phthalazin- 3(7H)-one;
(8/?,95)-5-fluoro-9-(l -methyl- l f-imidazol-2-yl)-8-phenyI-8,9-dihydro-2 -pyrido[4,3,2-ifeJphthalazin- 3(7#)-one;
(8,">,9Λ)-5-ί1υοΓο-9-( 1 -methyl- 1 H- 1 ,2,4-triazol-5-y l)-8-phenyl-8,9-dihydro-2H-pyrido[4,3,2-<;fe]phthalazin- 3(7 /)-one;
(8R,9S)-5-fluoro-9-(l-methyl-l /-l,2,4-triazol-5-yl)-8-phenyl-8,9-dihydro-2 /-pyrido[4,3,2-rfe]phthalazi
3(7//)-one;
(8/?,9^8-(4-fluorophenyi)-9-(l-meth l-l /-imidazol-2-yl)-8,9-dihydrcv2//-pyr
3(7//)-one;
(85,9^)-8-(4-fluorophenyl)-9-(l -methyl- lH-imidazol-2-yl)-8,9-dihydro-2H-pyrido[4,3,2-i/e]phthalazin- 3(7//)-one;
(8&95 5-fluoro-8-(4-fluorophenyl)-9-(l -methyl- lH-imidazol-2-y)-8,9-dihydro-2H-pyrido[4,3 ,2- ife]phthalazin-3(7i/)-one; (8S,9.K)-5-fluoro-8-(4-fluorophenyl)-9-( l-methyl- lH-^
o¾?]phthalazin-3(7H)-one;
(8R,95)-8-(4-fIuorophenyl)-9-( l -methyl- lH-l ,2,4-triazol-5-yl)-8,9-dihydro-2/J-pyrido[4,3,2- t/i?]phthalazin-3(7H)-one;
(85'!9R)-8-(4-fluoropheny l)-9-( 1 -methyl- 1 H- 1 ,2,4-triazol-5-yl)-8,9-dihydro-2H-pyrido[4,3,2- tfe]phthalazin-3(7H)-one;
(8i?,9S)-8-(4-((dimethylamino)me
2H-pyrido[4,3,2-i e]phthalazin-3(7H)-one;
(85,9R)-8-(4-((dimethylamino)methyl)phenyl)-5-fluoro-9-( l -methyl-lH- l ,2,4-triazol-5-yl)-8,9-dihydro- 2//-pyrido[4,3,2-c/e]phthalazin-3(7i -one;
(8R,9S)-8-(4-(azetidin- l -ylmethyl)phenyl)-9-(4-fluorophenyl)-8,9-dihydro-2/ -pyrido[4,3,2- </t'lphthalazin-3( 7 )-one; and
(85,,9R)-8-(4-(azetidin- l -ylmethyl)phenyl)-9-(4-fluorophenyl)-8,9-dihydro-2 -pyrido[4,3 ,2- e]phthalazin-3(7//)-one; or
a pharmaceutically acceptable salt, solvate or prodrug thereof.
Compound A Polymorph Embodiments
[0086] In one embodiment, provided herein is a tosylate salt of (8S,9i?)-5-fIuoro-8-(4- fluorophenyl)-9-( l -methyl- l H- l ,2,4-triazol-5-yl)-8,9-dihydro-2H-pyrido[4,3,2-de]phthalazin-3(7H)-one. In some embodiments the tosylate salt is in a crystalline form. In some embodiments, provided herein are solid forms of a tosylate salt of (85,9i?)-5-fluoro-8-(4-fluorophenyl)-9-( l -methyl-l H- l ,2,4-triazol-5-yl)- 8,9-dihydro-2//-pyrido[4,3,2-de]phthalazin-3(7H)-one, including crystalline forms, amorphous forms, or mixtures thereof.
[0087] In some embodiments, the (85,9i?)-5-fluoro-8-(4-fluorophenyl)-9-( l -methyl-lH- U2,4- triazol-5-yl)-8,9-dihydro-2H-pyrido[4,3,2-de]phthalazin-3(7/i)-one tosylate salt provided herein is in a crystalline form. In some embodiments, the crystalline form is unsolvated. In other embodiments, the crystalline form is a solvate. For instance, a crystalline solvate form may be a hydrate. In other embodiments, the (8S,9i?)-5-fluoro-8-(4-fIuorophenyl)-9-( 1 -methyl- 1 //- 1 ,2,4-triazol-5-y l)-8,9-dihydro- 2/ -pyrido[4,3,2-de]phthalazin-3(7 /)-one tosylate salt provided herein is in an amorphous form. In other embodiments, prov ided herein is a solid form (e.g., a crystalline form, an amorphous form, or a mixture of forms) of (8.S'.9/?)-5-nuoro-8-(4-nuorophenyD-9-( 1 -methyl- 1 /-/- 1 .2.4-triazol-5-yl)-8.9-dihydro-2/A pyrido[4,3,2-de]phthalazin-3(7i¥)-one or a salt or solvate thereof {e.g., a salt provided herein elsewhere). In one embodiment, provided herein is a crystalline form of (8S,9/?)-5-fluoro-8-(4-fluorophenyl)-9-( l - methy I- 1 /- 1.2,4-triazol-5-y l)-8.9-dihydro-2 -pyridof4,3,2-de jphthalazin-3(7/ )-one or a salt or solvate thereof. In one embodiment, provided herein is an amorphous form of (8.Y,9 ?)-5-fluoro-8-(4- fluoropheny l)-9-( 1 -methy I- 1 H- 1 ,2,4-triazot-5^
or a salt or solvate thereof. [0088] In certain embodiments, the (8S,9R)-5-fluoro-8-(4-fIuorophenyl)-9-( l -methyl- 1H- 1 ,2,4- triazol-5-yl)-8,9-dihydro-2 f-pyrido[4,3,2-de]phthalazin-3(7/i)-one tosylate crystalline salt form provided herein is substantially pure. For instance, in various embodiments, the crystalline tosylate salt purity is of at least about 90%, at least about 95%, at least about 97%, at least about 98%, at least about 99%, at least about 99.2%, at least about 99.5%, at least about 99.6%, at least about 99.7% or at least about 99.8% by- weight of a single crystalline form, the remainder of the total weight which may be other crystal line or amorphous forms and/or other compounds. In one embodiment, the crystalline tosylate salt is essentially a single-component crystalline form or a single polymorph. In another embodiment, the crystalline tosylate salt is a multiple-component crystalline form comprising a first crystal line form and at least one other crystalline and/or amorphous form of (8S,9R)-5-fliioro-8-(4-fluorophenyl)-9-( l -methyl- lH- l ,2,4-triazol- 5-yl)-8,9-dihydro-2//-pyrido[4,3,2-de]phthalazin-3(7i/)-one. In some or any embodiments, the crystalline form is substantially free of an amorphous form of (85,9R)-5-fIuoro-8-(4-fluorophenyl)-9-( l -methyl- 1H- l ,2,4-triazol-5-yl)-8,9-dihydro-2 /-pyrido[4,3,2-de]phthalazin-3(7 /)-one.
[0089] Unless otherwise specified, when an XRPD peak is expressed in 2Θ angle degrees, it should be understood that copper Kal radiation is used. In some embodiments, the 2Θ angle degrees value provided herein may vary to an extent of about ± 0.2 °θ, while still describing the same XRPD peak.
[0090] In the l 3C solid state NMR, the peak positions can vary depending on factors such as signal-to-noise ratio, peak width, temperature, spinning speed, decoupling efficiency, magic angle setting, data processing procedures and parameters, and software peak picking algorithm. In addition, peak position is relative to the chemical shift referencing procedure. Several different chemical shift reference standards may be used and wi ll not necessarily give the same results. This may lead to peak positions that are different by several ppm. However, typical ly al l of the peaks wil l have a systematic change in position in the same direction if a different reference standard was used or if the analyst used a different value for the reference peak position of the same standard. In some embodiments, the ppm values in the l C solid state NMR provided herein may vary to an extent of about ± 0.2 ppm, while still describing the same peak.
[0091] In certain embodiments, a crystalline salt of (85,9JR)-5-fluoro-8-(4-fluorophenyl)-9-( l - methyl-1 //- l ,2,4-triazol-5-yl)-8,9-dihydro-2i -pyrido[4,3,2-de]phthalazin-3(7H)-one is provided having an XRPD pattern comprising one or more (e.g. one, two, three, four, five, six, seven, eight, nine, ten, or greater than ten; or at least three, at least four, at least five, at least six, or at least seven) characteristic peaks selected from peaks expressed in d-values (A) according to any one of Tables 9, 11, 13, 15, 17 and 25. In another embodiment, the crystalline salt is a tosylate salt having one or more (e.g., one, two, three, four, five, six, seven, eight, nine, ten, or greater than ten; or at least three, at least four, at least five, at least six, or at least seven) XRPD peaks selected from peaks with 2Θ angle degrees according to any one of Tables 9, 11, 13, 15, 17 and 25. In certain embodiments, the crystalline tosylate salt has an XRPD pattern substantially as provided in Figures 1, 6, or 8. [0092J In certain embodiments, the crystalline tosylate salt provided herein has an XRPD pattern comprising one or more (e.g., one, two, three, four, five, six, seven, eight, nine, ten, or greater than ten; or at least three, at least four, at least five, at least six, or at least seven) XRPD peaks selected from peaks with 2Θ angle degrees or d values (A) as provided in Table 15. In some embodiments, the crystalline tosylate salt has XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1.79, 5.86, 4.90, 4.42, 4.35, 3.93, and 3.70. In some embodiments, the crystalline tosylate salt has XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1.79; 5.86; 4.90; 4.65; 4.42; 4.35; 4.13; 3.93; and 3.70. Within certain embodiments, the XRPD pattern of the crystalline tosylate salt comprises one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1 .79; 5.86; 4.98; 4,90; 4.79; 4.65; 4.42; 4.35; 4.13; 3.93; 3.70; and 3.58. In certain embodiments, the XRPD pattern of the crystalline tosylate salt comprises one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) comprise 1 1 .79; 7.07; 6.13; 5.86; 5.10; 4.98; 4.90; 4.79; 4.65 ; 4.42; 4.35; 4.13; 4.08; 3.93; 3.85; 3.70; 3.58; 3.3 1 ; and 2.99. In some embodiments, the crystal line tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2Θ angle degrees of about 7.49, 15.10, 18.10, 20.06, 20.40, 22.61 , and 24.01. In some embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2Θ angle degrees of about 7.49, 15.10, 1 8.10, 19.08, 20.06, 20.40, 21.49, 22.61 , and 24.01. In some embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 20 angle degrees of about 7.49, 15.10, 17.78, 18.10, 1 8.49, 19.08, 20.06, 20.40, 21 .49, 22.61, 24.01 , and 24.84. In some embodiments, the crystal line tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 20 angle degrees of about 7.49, 12.50, 14.44, 15.10, 17.38, 17.78, 18.10, 18.49, 19.08, 20.06, 20.40, 21.49, 21 .76, 22.61 , 23.05, 24.01, 24.84, 26.93, and 29.82,
(0093) In certain embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2Θ angle degrees or d values (A) as provided in Table 15. In some embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1.8, 5.9, 4.9, 4.42, 4.35, 3.9, and 3.7. In some embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1.8; 5.9; 4.9; 4,7; 4.42; 4.35; 4.1 ; 3.9; and 3.7. Within certain embodiments, the XRPD pattern comprises one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) comprise about 1 1.8; 5.9; 5.0; 4.9; 4.8; 4.7; 4.42; 4.35; 4.1 ; 3.9; 3.70; and 3.58. In certain embodiments, the XRPD pattern comprises one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) comprise about 1 1.8; 7.1 ; 6.1 ; 5.9; 5.1 ; 5.0; 4.9; 4.8; 4.7; 4.42; 4.35; 4.1 ; 4.1 ; 3.9; 3.9; 3.7; 3.6; 3.3; and 3.0. In some embodiments, the crystal line tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2Θ angle degrees of about 7.5, 15.1 , 18.1, 20.1 , 20.4, 22.6, and 24.0. In some embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 20 angle degrees of about 7.5, 15.1 , 18.1 , 19. 1 , 20.1 , 20.4, 21 .5, 22.6, and 24.0. In some embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2Θ angle degrees of about 7.5, 1 5.1 , 17.8, 18. 1 , 1 8.5, 19. 1 , 20.1 , 20.4, 21.5, 22.6, 24.0, and 24.8. In some embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 20 angle degrees of about 7.5, 12.5, 14.4, 15.1 , 17.4, 17.8, 18.1 , 18.5, 19.1 , 20.1 , 20.4, 21.5, 21 .8, 22.6, 23.1 , 24.0, 24.8, 26.9, and 29.8.
[0094] In certain embodiments, the crystalline tosylate salt comprises XRPD peaks at 20 angle degrees or d values (A) as provided in Table 9. In some embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 20 angle degrees of about 7.42, 15.01 , 17.70, 18.01 , 18.47, 18.98, 19.98, 20.33 , 21.41 , 22.58, 23.95, and 24.76. In some embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2Θ angle degrees of about 7.42, 15.01 , 18.01 , 19.98, 20.33, 22.58, and 23.95. In some embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2Θ angle degrees of about 7.42, 15.01 , 17.70, 18.01, 19.98, 20.33, 21.41 , 22.58, 23.95, and 24.76. In some embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1.90, 5.90, 5.01 , 4.92, 4.44, 4.37, 4.15, 3.93, 3.71 , and 3.59. In some embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1.90, 5.90, 5.01 , 4.92, 4.80, 4.67, 4.44, 4.37, 4.1 5, 3.93, 3.71 , and 3.59. In some embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1 .90, 5.90, 4.92, 4.44, 4.37, 3.93, and 3.71. [009SJ In certain embodiments, the crystalline tosyiate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2Θ angle degrees or d values (A) as provided in Table 9. In some embodiments, the crystal line tosyiate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 20 angle degrees of about 7,4, 15.0, 17.7, 1 8.0, 18.5, 19.0, 20.0, 20.3, 21.4, 22.6, 24.0, and 24.8. In some embodiments, the crystalline tosyiate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six. or at least seven peaks) selected from peaks with 2Θ angle degrees of about 7.4, 15.0, 18.0, 20.0, 20.3, 22.6, and 24.0. In some embodiments, the crystalline tosyiate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 20 angle degrees of about 7.4; 15.0; 17.7; 18.0; 20.0; 20.3; 21 .4; 22.6; 24.0; and 24.8. In some embodiments, the crystalline tosyiate salt has an XRPD partem comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1 .9, 5.9, 5.0, 4.9, 4.44, 4.37, 4. 1 , 3.9. 3.7, and 3.6. In some embodiments, the crystalline tosyiate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1.9, 5.9, 5.0, 4.9, 4.8, 4.7, 4.44, 4.37, 4. 1 , 3.9, 3.7, and 3.6. In some embodiments, the crystalline tosyiate salt comprises XRPD peaks at d values (A) of about 1 1.9, 5.9, 4.9, 4.44, 4.37, 3.9, and 3.7.
[0096] In other embodiments, the crystal line tosyiate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 20 angle degrees or d values (A) as provided in Table 11. In other embodiments, the crystalline tosyiate salt has an XRPD partem comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2Θ angle degrees of about 7.42, 15.02, 17.38, 17.74, 18.03, 18.54, 19.02, 20.08, 20.39, 2 1 .44, 22.63, 24.00, and 24.83. In other embodiments, the crystal line tosyiate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 20 angle degrees of about 7.42, 15.02, 18.03, 20.08, 20.39, 22.63, and 24.00. In other embodiments, the crystalline tosyiate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1.91 , 5.89, 5.10, 5.00, 4.92, 4.78, 4.66, 4.42, 4.35, 4.14, 3.93, 3.71 , and 3.58. In other embodiments, the crystalline tosyiate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1.91 , 5.89, 4.92, 4.42, 4.35, 3.93, and 3.71 .
[0097] In other embodiments, the crystalline tosyiate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 20 angle degrees or d values (A) as provided in Table 11. In other embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2Θ angle degrees of about 7.4, 15.0, 17.3, 17.7, 1 8.0, 18.5, 19.0, 20. 1 , 20.4, 21.4, 22.6, 24.0, and 24.8. In other embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2Θ angle degrees of about 7.4, 1 5.0, 18.0, 20.1 , 20.4, 22.6, and 24.0. In other embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1.9, 5.9, 5.1 , 5.0, 4.9, 4.8, 4.7, 4.42, 4.35, 4.1 , 3.9, 3.7, and 3.6. In other embodiments, the crystal line tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1.9, 5.9, 4.9, 4.42, 4.35, 3.9, and 3.7.
[0098] In some embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2Θ angle degrees or d values (A) as provided in Table 13. In other embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 20 angle degrees of about 7.46, 12.47, 14.45, 15.09, 17.40, 17.74, 18.1 1 , 18.53, 19.05, 20.09, 20.43, 21.46, 22.63, 23.10, 24.03, 24.85, and 26.96. In other embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 20 angle degrees of about 7.46, 14.45, 15.09, 17.74, 18. 1 1 , 1 8.53, 19.05, 20.09, 20.43, 21 .46, 22.63, 24.03, 24.85, and 26.96. In other embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g. , at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2Θ angle degrees of about 7.46, 15.09, 18.1 1 , 20.09, 20.43, 22.63, and 24.03. In other embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1.84, 7.09, 6.13, 5.87, 5.09, 5.00, 4.89, 4.78, 4,66, 4.42, 4.34, 4.13, 4.08, 3.93, 3.85, 3.70, 3.58, 3.30, 2,99, and 2.86. In other embodiments, the crystal line tosylate salt has an XRPD pattern comprising one or more peaks (e.g. , at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1 .84, 6.13, 5.87, 5.09, 5.00, 4.89, 4.78, 4.42, 4.34, 4.13, 3.93, 3.70, 3.58, and 3.30. In other embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1 .84, 5.87, 4.89, 4.42, 4.34. 3.93, and 3.70.
[0099] In some embodiments, the crystalline tos late salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2Θ angle degrees or d values (A) as provided in Table 13. In other embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2Θ angle degrees of about 7.5. 12.5, 14.5, 15.1 , 17.4, 17.7, 18.1 , 18.5, 19.0, 20.1 , 20.4, 21.5, 22.6, 23.1 , 24.0, 24.9, and 27.0. In other embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2Θ angle degrees of about 7.5, 14.5, 15.1 , 17.7, 18.1 , 18.5, 19.0, 20.1 , 20.4, 21.5, 22.6, 24.0, 24.9, and 27.0. In other embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2Θ angle degrees of about 7.5, 15.1, 1 8.1 , 20.1 , 20.4, 22.6, and 24.0. In other embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1.8, 7.1 , 6. 1 , 5.9, 5. 1 , 5.0, 4.9, 4.8, 4,7, 4.4, 4.3, 4.13, 4.08, 3.9, 3.8, 3.7, 3.6, 3.3, 3.0, and 2.9. In other embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1.8, 6. 1 , 5.9, 5.0, 4.9, 4.8, 4.7, 4.4, 4.3, 4.1 , 3.9, 3.7, 3.6, and 3.3. In other embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1.8, 5.9, 4.9, 4.4, 4.3, 3.9, and 3.7.
[00100] In some embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2Θ angle degrees or d values (A) as provided in Table 17. In other embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2Θ angle degrees of about 7.50, 12.5 1 , 14.48, 15.12, 17.38, 17.78, 18. 17, 18.58, 19.1 1 , 20.09, 20.54, 21.54, 21 .86, 22.65, 23.19, 24.08, 24.86, 26.98, 29.97, 30.44, 30.84, 32.07, 32.49, and 37.56. In other embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 20 angle degrees of about 7.50, 15. 12, 1 7.38, 17.78, 1 8.17, 1 8.58, 19. 1 1 , 20.09, 20.54, 21 .54, 21.86, 22.65, 23.19, 24.08, 24.86, and 26.98. In other embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2Θ angle degrees of about 7.50, 15.12, 18.17, 20.09, 20.54, 22.65, and 24.08. In other embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1 .78, 7.07, 6.1 1 , 5.85, 5. 10, 4.98, 4.88, 4.77, 4.64, 4.42, 4.32, 4.12, 4.06, 3.92, 3.83, 3.69, 3.57, 3.30, 2.98, 2.93, 2.78, 2.75, 2.39. In other embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1 .78, 5.85, 5.10, 4.98, 4.88, 4.77, 4.64, 4.42, 4.32, 4. 12, 4.06, 3.92, 3.83, 3.69, 3.57, and 3.30. In other embodiments, the crystalline tos late salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1.78, 5.85, 4.88, 4.42, 4.32, 3.92, and 3.69.
[00101] In some embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2Θ angle degrees or d values (A) as provided in Table 17. In other embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2Θ angle degrees of about
7.5, 12.5, 14.5, 15.1 , 17.4, 17.8, 18.2, 18.6, 19.1 , 20.1 , 20.5, 21.5, 21.9, 22.6, 23.2, 24.1 , 24.9, 27.0, 30.0,
30.4, 30.8, 32.1 , 32.5, and 37.6. In other embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 20 angle degrees of about 7.5, 15.1 , 17.4, 17.8, 18.2, 18.6, 19. 1 , 20.1 ,
20.5, 21.5, 21.9, 22.6, 23.2, 24. 1 , 24.9, and 27.0. In other embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2Θ angle degrees of about 7.5, 15.1 , 18.2, 20.1 , 20.5, 22.6, and 24.1. In other embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1.8, 7.1 , 6.1 , 5.9, 5.1, 5.0, 4.9, 4.8, 4.6, 4.4, 4.3, 4.12, 4.06, 3.9, 3.8, 3.7,
3.6, 3.3, 3.0, 2.9, 2.78, 2.75, 2.4. In other embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1 .8, 5.9, 5.1 , 5.0, 4.9, 4.8, 4.6, 4.4, 4.3, 4.12, 4.06, 3.9, 3.8, 3.7, 3.6, and 3.3. In other embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1.8, 5.9, 4.9, 4.4, 4.3, 3.9, and 3.7.
[00102] In some embodiments, the crystalline tosylate salt has an XRPD pattern substantially as provided in Figure 6. In some embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with 2Θ angle degrees as provided in Table 25. In some embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with ± about 0.2 2Θ angle degrees of about 7.51 , 14.47, 15. 14, 17.41 , 18.12, 18.53, 1 .07, 20.09, 20.46, 21 .48, 21 .81 , 24.05, 24.83, and 29.81. In some embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with ± about 0.2 20 angle degrees of about 7.51 , 14.47, 15. 14, 20.09, 21.48, and 24.05. In some embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with ± about 0.2 2Θ angle degrees of about 7.51 , 15.14, 18.12, 20.09, 20.46, 22.65, and 24.05. In some embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with ± about 0.2 2Θ angle degrees of about 7.5 1 , 20.09, and 24.05. In some embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with ± about 0.2 2Θ angle degrees of about 7.5, 14.5, 15.1 , 17.4, 18.1 , 18.5, 19.1 , 20.1 , 20.46, 21.48, 2 1.8, 24.1 , 24.8, and 29.8. In some embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with ± about 0.2 2Θ angle degrees of about 7.5, 14.5, 15.1 , 20.1 , 21.5, and 24.1. In some embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with ± about 0.2 2Θ angle degrees of about 7.5, 15.1, 18.1 , 20.1 , 20.5, 22.6, and 24.1. In some embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with ± about 0.2 2Θ angle degrees of about 7.5, 20.1 , and 24.1. In some embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) as provided in Table 25. In other embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1 .8, 6.1 , 5.9, 5.1, 4.9, 4.8, 4.6, 4.4, 4.3, 4.1 , 4.1 , 3.7, 3.6, and 3.0. In other embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1 .8, 6.1 , 5.9, 4.4, 4.1 , and 3.7. In other embodiments, the crystalline tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1.8, 5.9, 4.9, 4.4, 4.3, 3.9, and 3.7. In other embodiments, the crystal line tosylate salt has an XRPD pattern comprising one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from peaks with d values (A) of about 1 1.8, 4.4, and 3.7.
[00103] In some or any embodiments, the crystal line tosylate salt exhibits a "C NMR spectrum corresponding substantially to the spectrum in Figure 12 or exhibits a spectrum with peaks corresponding substantially to those in Table 27. In some or any embodiments, the crystalline tosylate salt exhibits a l3C NMR spectrum with one or more peaks (e.g., at least three, at least four, at least five, at least six, or at least seven peaks) selected from about ± 0.2 ppm at about 166.9, 164.3, 162.2, 160.6, 1 5 1 .8. 149.4, 143.2,
140.2, 139. 1 , 136.0, 13 1.8, 129.4, 128.6, 127.7, 1 23.9. 1 16.8, 1 15.1 , 1 12.2, 105.2, 100.3, 58.5, 45.3, 37.4, and 23.9. In some or any embodiments, the crystalline tosylate salt exhibits a l3C NMR spectrum with peaks about ± 0.2 ppm at about 151.8, 149.4, 143.2, 136.0, 13 1.8, 123.9, 1 16.8, 1 15.1 , 1 12.2, 105.2,
100.3, 58.5, 45.3, 37.4, and 23.9. In some or any embodiments, the crystalline tosylate salt exhibits a , C NMR spectrum with peaks about ± 0.2 ppm at about 143.2, 136.0, 131.8, 123.9, 1 12.2. 105.2, 100.3, 58.5, 45.3, 37.4, and 23.9. In some or any embodiments, the crystalline tosylate salt exhibits a l 3C NMR spectrum with peaks about ± 0.2 ppm at about 143.2, 136.0, 13 1.8, 123.9, 1 12.2, 105.2, and 100.3.
[00104] In some or any embodiments, the crystalline salt has an XRPD peaks at 2Θ angle degrees of about 7.4, 15.1 , 1 7.4, 17.8, 18.1 , 18.5, 19.1 , 20.1 , 20.4, 21.5, 22.6, 24.0, 24.8, and 27.0. In some or any embodiments, the crystalline salt has an XRPD peaks at 2Θ angle degrees of about 7.4, 15.1, 18.1 , 19.1 , 20.1, 20.4, 21.5, 22.6, and 24.0. In some or any embodiments, the crystalline salt has an XRPD peaks at 2Θ angle degrees of about 7.4, 15.1, 20.1 , 20.4, 22.6, 24.0, and 24.8. In some or any embodiments, the crystalline salt has an XRPD peaks at 20 angle degrees of about 7.4, 15.1 , 18.1 , 20. 1 , 20.4, 22.6, and 24.0.
[00105] In some or any embodiments, the crystal line salt has an XRPD pattern comprising peaks at d values (A) of about 1 1.9, 5.9, 5.1 , 5.0, 4.9, 4.8, 4.6, 4.4, 4.3, 4.1 , 3.9, 3.7, 3.6, and 3.3. In some or any embodiments, the crystalline salt has an XRPD pattern comprising peaks at d values (A) of about 1 1 .9, 5.9, 4.9, 4.6, 4.4, 4.3, 4.1 , 3.9, and 3.7. In some or any embodiments, the crystalline salt has an XRPD pattern comprising peaks at d values (A) of about 1 1 .9, 5.9, 4.4, 4.3, 3.9, 3.7, and 3.6. In some or any embodiments, the crystalline salt has an XRPD peaks at d values (A) of about 1 1.9, 5.9, 4.9, 4.4, 4.3, 3.9, and 3.7.
[00106] In some or any embodiments, crystalline (8S,9/?)-5-fluoro-8-(4-fluorophenyl)-9-( l- methyl- l //- 1 ,2,4-triazol-5-yl)-8,9-dihydro-2H-pyrido[4,3,2-de]phthalazin-3(7 /)-one tosylate is a crystalline polymorph exhibiting at least one of
a solid state i C NMR spectrum with peaks at 143.2, 136.0, 13 1.8, 123.9, 1 12.2, 105.2, and 100.3 ppm ± 0.2 ppm;
an X-ray diffraction partem comprising characteristic peaks expressed in d-values (A): 1 1.9, 5.9, 4.9, 4.4, 4.3, 3.9, and 3.7; and
an X-ray diffraction pattern comprising peak reflectances at 2Θ angle degrees ± 0.2 20 angle degrees of 7.4, 1 5. 1 , 18.1 , 20.1 , 20.4, 22.6, and 24.0.
[00107] In some embodiments, the crystalline tosylate salt exhibits a single endothermal peak on differential scanning calorimetry between room temperature and about 350 °C, where the single endothermal peak maximum occurs between about 320 °C to about 335 °C. In certain embodiments, the single endothermal peak maximum occurs at between about 330 "C to about 335 °C. In some
embodiments, the single endothermal peak maximum occurs at about 333 °C to about 334 °C.
[00108J In certain embodiments, a crystalline tosylate salt of (85,9i?)-5-fluoro-8-(4-fluorophenyl)-
9-( I -methyl- l H- l ,2,4-triazol-5-yl)-8,9-dihydro-2H-pyrido[4,3,2-de]phthaIazin-3(7H)-one as provided herein has no observable endotherm from about 25 "C to about 250 °C as determined by DSC. In certain embodiments, a crystalline tosylate salt of (8S,9i?)-5-fluoro-8-(4-fluoropheny I)-9-( 1 -methyl- 1 H- 1 ,2,4- triazol-5-yI)-8,9-dihydro-2 -pyrido[4,3,2-de]phthalazin-3(7//)-one as provided herein has a DSC thermogram comprising an endotherm with a maximum at between about 320 'C to about 335 'C, between 330 °C to about 335 *C, or between about 333 *C to about 334 *C. In certain embodiments, a crystalline tosylate salt of (8S,9i?)-5-fluoro-8-(4-fIuorophenyl)-9-( 1 -methyl- 1 H- 1 ,2,4-triazol-5-y I)-8,9- W
dihydro-2H-pyrido[4,3,2-de]phthalazin-3(7//)-one as provided herein has a DSC thermogram
corresponding substantially to the DSC thermograph of Figure 2a, 3a, 4a, 5a, or 7a.
[00109] In certain embodiments, a crystalline tosylate salt of (8S,9R)-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 as provided herein has a DVS isotherm plot corresponding substantially to the DVS isotherm plot of Figure 9. In certain embodiments, a crystalline tosylate salt of (8S,9i?)-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 as provided herein does not exhibit significant weight change (e.g. , less than about 0.05 wt %, less than about 0.1 wt %, less than about 0.15 wt %, or less than about 0.2 wt %) from about 0% to about 95% relative humidity.
[00110] In certain embodiments, a crystalline tosylate salt of (8<S',9R)-5-fluoro-8-(4-fluorophenyl)-
9-(l -methyl-lH- l ,2,4 riazol-5-yl)-8,9-dihydro-2H-pyrido[4,3,2-de]phthalazin-3(7H)-one as provided herein has a TGA thermogram corresponding substantially to the TGA thermograph of Figure 2b, 3b, 4b, 5b, or 7b. In certain embodiments, a crystalline tosylate salt of (8S,9i?)-5-fluoro-8-(4-fluorophenyl)-9-( l - methyl- lH- l ,2,4-triazol-5-yl)-8,9-dihydro-2/ -pyrido[4,3,2-de]phthalazin-3(7H)-one as provided herein has a TGA thermogram that does not exhibit significant weight loss (e.g., less than about 0.05 wt %, less than about 0.1 wt %, less than about 0.5 wt %, less than about 1 wt %, less than about 5 wt %, less than about 10 wt %, less than about 1 5 wt %, less than about 20 wt %, or less than about 25 wt %) when heated from about room temperature to a temperature of about 200 "C, about 210 °C, about 220 °C, about 230 °C, about 240 °C, about 250 *C, about 260 °C, about 270 °C, about 280 °C, about 290 °C, about 300 'C, about 3 10 °C, about 320 °C, or greater than about 320 "C. In various embodiments, the crystall ine tosylate salt provided herein has a weight loss of no greater than about 1%, no greater than about 0.5%, or no greater than about 0.1 % in a thermogravimetric thermogram between about 25 °C to about 200 'C.
[00111] In certain embodiments, a crystalline tosylate salt of (85',9i?)-5-fluoro-8-(4-fluorophenyl)-
9-(l -methyl- lH- l ,2,4 riazol-5-yl)-8,9-dihydro-2 i-pyrido[4,3,2-de]phthalazin-3(7H)-one as provided herein is unsolvated. In certain embodiments, a crystalline tosylate salt of (8S,9 ?)-5-fluoro-8-(4- fliK ophenyl)-9-( l -methyl- 1 /7- 1 .^
as provided herein is anhydrous. In certain embodiments, a crystalline tosylate salt
Figure imgf000066_0001
(4-nuoropheny] )-9-( 1 -meth l- l /f- 1.2J-m
one as provided herein is thermally stable. In certain embodiments, a crystalline tosylate salt of (8S,9R)-5- fluoro-8-{4-tl uorophenyl)-9-( I -methyl- 1 H- 1 .2,4-triazol-5-yl)-8,9-dihydro-2//-pyrido[4,3.2-de]phthalazin- 3(7//)-one as provided herein is non-hygroscopic.
[00112] In yet another embodiment, compositions are provided that comprise or consist essentially of substantially pure (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, in another embodiment in a crystalline form thereof as provided herein.
[00113] In some embodiments, a pharmaceutical composition is provided comprising a tosylate salt of (85,9R)-5-fluoro-8-(4-fluorophenyl)-9-( l -methyl-l H- l ,2,4-triazol-5-yl)-8,9-dihydro-2H- pyrido[4,3,2-de]phthalazin-3(7 )-one, including a crystalline form thereof as described herein, and one or more pharmaceutically acceptable excipient(s) and/or carrier(s). The choice of excipient, to a large extent, depends on factors, such as the particular mode of administration, the effect of the excipient on the solubility and stability of the active ingredient, and the nature of the dosage form.
Pharmaceutical Composition Embodiments
[00114] Provided herein are pharmaceutical compositions formulated such that Compound A tosylate salt is in a stable form, for instance when stored in an open container for up to 4 weeks or more under conditions such as at 40 °C and 75% relative humidity (RH). In one example, pharmaceutical compositions provided herein are made with Compound A tosylate salt in a crystalline form.
[00115] Provided herein are pharmaceutical compositions comprising a Compound of Formula I or Compound A tosylate salt and one or more pharmaceutically acceptable excipients. In some embodiments, the pharmaceutical composition is in a solid form, for example, in granules or in a powder, tablet, or capsule. In certain embodiments, the pharmaceutical composition provided herein is suitable for oral administration to a subject. Components of the pharmaceutical compositions provided herein are - described below. In certain embodiments, the pharmaceutical composition is prepared using Compound A tosylate salt consisting of a crystalline solid form.
[00116] In some or any embodiments, the pharmaceutical composition provided herein comprises crystalline Compound A tosylate salt as described herein, in another example as defined in any of the Compound A embodiments herein. In certain embodiments of the pharmaceutical composition provided herein, the pharmaceutical composition is prepared with, and/or comprises crystalline Compound A tosylate salt which is unsolvated. In other embodiments, the crystalline form is a solvate, for example, a hydrate.
[00117] In some embodiments, the weight weight percent of Compound A tosylate salt in the pharmaceutical composition provided herein is between about 0.01 % to about 5.0 %, between about 0.1%> and about 2.5%>, between about 1 % and about 2.5%», between about 1 % and about 3%, between about 2% and about 4%, between about 2% and about 3%, between about 3%o and about 5%, between about 0. 1 % and about 2%, between about 0.01 % and about 1.0 %, between about 0. 1 % and about 1.0 %, between about 0.1 %» and about 0.80 %, between about 0.20 % and about 0.750 %, between about 0.290 % and about 0.726 %, or about 0.2 %, about 0.290 %, about 0.5 %, about 0.726 %, about 1 %, about 1 .25%, about 1.5%,, about 1.75%, about 2%, about 2.25%, about 2.5%>, about 2.75%, about 3%, or about 3.25%. In certain embodiments, the percentages above refer to weight/weight percentages of Compound A tosylate salt in the pharmaceutical composition, excluding, for instance, any coating where the pharmaceutical composition is in a tablet form, or capsule shell where the pharmaceutical composition is encapsulated in a capsule.
[00118] In certain embod iments, a pharmaceutical composition provided herein comprises
Compound A tosylate salt which is stereomerically pure as defined herein. In some embodiments, a pharmaceutical composition as provided herein comprises a solid form Compound A tosylate salt that is substantially pure as defined herein. For instance, where solid form is a crystalline form, it can be substantially free of other crystalline polymorphs or amorphous forms of Compound A tosylate salt. In certain embodiments, the solid Compound A tosylate salt in the pharmaceutical composition is stable, as defined herein. In other embodiments, the crystalline Compound A tosylate salt used in the preparation of the pharmaceutical composition is stable as defined, for instance, by no detectable change in its crystalline solid form (e.g., polymorphic change and/or chemical change such as, for example, due to degradation) when stored for 18 months or more at, for example, 25 °C or ambient conditions or, for 4 weeks or more at 40 °C and 75% RH. In one embodiment, the crystalline tosylate salt is essentially a single-component crystalline form or a single polymorph.
[00119] In certain embodiments of the pharmaceutical composition provided herein is a low dose formulation of Compound A tosylate salt, that is, where the weight/weight percent of Compound A tosylate salt in the pharmaceutical composition is such that a low dose, as defined herein, is delivered to a subject with an indication as provided herein, when milligrams (e.g., ranging in amounts from about 5 mg to about 50 mg) or tens of milligrams (e.g., ranging in amounts from about 30 mg to about 1000 mg or from about 30 mg to about 1500 mg) of the pharmaceutical composition are administered to the subject. 100120] In certain embodiments, the pharmaceutical composition comprises Compound A tosylate salt and one or more excipients, wherein the Compound A tosylate salt is in a powder form and/or the Compound A tosylate salt consists of crystalline Compound A tosylate salt.
Excipients
[00121] The one or more pharmaceutically acceptable excipients in the pharmaceutical composition provided herein can, for instance, include any excipient known to those skil led in the art or combination thereof. Further a person of ordinary ski ll in the art would understand that while a particular excipient is described as a binder, for example, the excipient may also have other properties, such as of a disintegrant or diluent, for example.
[00122] In certain embodiments, the one or more excipients comprise one or more binders, diluents, carriers, disintegrants, glidants, lubricants, fillers or a combination thereof. In some embodiments the one or more excipients comprise a binder, for example, hydroxypropy l cellulose.
Suitable diluents or carriers include, but are not limited to, si licified microcrystalline cel lulose, microcrystalline cellulose, dibasic calcium phosphate, partially pregelatinized starch, lactose, sucrose, mannitol and mixtures thereof. A diluent or carrier is preferrably present from about 75% to about 99.5% by weight of the final composition. Suitable disintegrants include, but are not limited to, silicified microcrystalline cellulose, microcrystalline cellulose, crosearmellose sodium, starch, crospovidone, sodium starch glycolate, partially pregelatinized starch and m ixtures thereof. A disintegrant can, for example, be present from about 1% to about 10% by weight of the final composition. Suitable binders include, but are not limited to, hydroxypropyl cellulose, hypromellose, low substituted hydroxypropyl cellulose, povidone, copovidone, pregelatinized starch, partially pregelatinzed starch, and mixtures thereof. A binder can, for example, be present from about 0.1% to about 10% by weight of the final composition. Suitable glidants include, but are not limited to, colloidal silicon dioxide and talc. Suitable lubricants include, but are not limited to, magnesium stearate, calcium stearate, sodium lauryl sulfate, stearic acid and sodium stearyl fumarte. A lubricant can, for example, be present from about 0.1% to about 5% by weight of the final composition.
[00123] in certain embodiments, an excipient is a lubricant (e.g.. sodium stearyl fumarate, magnesium stearate, stearic acid), a binder (e.g., hydroxypropyl cellulose, low substituted HPC, hypromellose, maltodextrin, gelatin, polyethylene glycol, pregelatinized starch, partially pregelatinized starch), a glidant (e.g., talc, fumed silica, colloidal silicon dioxide, magnesium carbonate), a diluent (e.g. ,microcrystalline cellulose, dibasic calcium phosphate, lactose, sucrose, sorbitol, starch, partially pregelatinized starch), a filler, a disintegrant (e.g. , crospovidone, croscarmellose sodium, sodium starch glycol ate, alginic acid, starch, partially pregelatinized starch), a flavoring agent, a sweetener, a coloring agent, a preservative (e.g., an antioxidant, antimicrobial or other types of preservative including those such as vitamin A, vitamin E, vitamin C, cysteine, citric acid; methyl paraben, propyl paraben), a surfactant, an emulsifier, a sorbent, or a combination of any of the above. It will be understood that a given material need not be limited to being one type of excipient.
[00124] Suitable lubricants include, but are not limited to, calcium stearate; magnesium stearate; sodium stearyl fumarate; mineral oil; l ight mineral oil; glycerin; sorbitol; mann itol; glycols, such as glycerol behenate and polyethylene glycol (e.g., PEG400 and PEG6000); stearic acid; sodium lauryl sulfate; talc; hydrogenated vegetable oil, including peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, corn oil, and soybean oil; zinc stearate; ethy l oleate; ethyl laureate; agar; starch; lycopodium; silica (si licone dioxide) or silica gels, such as Aerosil® 200 (W.R. Grace Co., Baltimore, MD) and CAB- O-SIL® (Cabot Co. of Boston, MA); and mixtures thereof. The pharmaceutical compositions provided herein may contain about 0.1 to about 5% by weight of a lubricant.
[00125] Binders impart cohesiveness to a tablet to ensure the tablet remains intact after compression. Suitable binders include, but are not limited to, starches, such as corn starch, potato starch, partially pregelatinized starch (e.g., Starch 1500) and pregelatinized starch; gelatin; sugars, such as sucrose, glucose, dextrose, molasses, and lactose; natural and synthetic gums, such as acacia, alginic acid, alginates, extract of Irish moss, panwar gum, ghatti gum, mucilage of isabgol husks,
carboxymethylcellulose, methylcellu lose, polyvinylpyrrolidone (PVP), copovidone, larch arabogalactan, powdered tragacanth, and guar gum; magnesium aluminum silicate (e.g., Veegum*); celluloses, such as ethyl cel lulose, cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethyl cellulose, methyl cellulose, hydroxyethylcellulose (HEC), hydroxypropy (cellulose (HPC), hypromellose;
m icrocrystalline celluloses, such as Avicel® PH- 101 , Avicel* PH- 103, Avicel® RC-58 1 . Avicel * PH- 105 (FMC Corp., Marcus Hook, PA); and mixtures thereof.
[00126] Suitable diluents include, but are not limited to, dibasic calcium phosphate, calcium sulfate, lactose, sorbitol, sucrose, inositol, cellulose, kaolin, mannitol, sodium chloride, dry starch, and powdered sugar. Certain diluents, such as mannitol, xylitol, lactose, sorbitol, sucrose, and inositol, when present in sufficient quantity, can impart properties to some compressed tablets that permit disintegration in the mouth by chewing. Such compressed tablets can be used as chewable tablets.
[00127] Suitable fillers include, but are not limited to, talc, calcium carbonate, macrocrystalline cellulose, powdered cellulose, dextrates, kaolin, mannitol, silicic acid, sorbitol, starch, pre-gelatinized starch, and mixtures thereof.
[00128] Suitable disintegrants include, but are not limited to, agar; bentonite; celluloses, such as methylcellulose and carboxymethylcel lulose; wood products; natural sponge; cation-exchange resins; alginic acid; gums, such as guar gum; citrus pulp; cross-linked celluloses, such as croscarmellose; cross- linked polymers, such as crospovidone; cross-linked starches; calcium carbonate; microcrystalline cellulose; modified cr ss-linked starch derivatives such as sodium starch glycolate; polacrilin potassium; starches, such as corn starch, potato starch, tapioca starch, and partially pregelatinized starch; clays, such as Veegum® and VAN GEL®; aligns; and mixtures thereof. The amount of a disintegrant in the pharmaceutical compositions provided herein varies upon the type of formulation, and is readily discernible to those of ordinary skil l in the art. The pharmaceutical compositions provided herein may contain from about 0.5 to about 15% or from about 1 to about 10% by weight of a disintegrant.
[00129] Suitable gl idants include colloidal silicon dioxide, CAB-O-SIL® (Cabot Co. of Boston,
MA), and asbestos-free talc. Coloring agents include any of the approved, certified, water soluble FD&C dyes, and water insoluble FD&C dyes suspended on alumina hydrate, and color lakes and mixtures thereof. A color lake is the combination by adsorption of a water-soluble dye to a hydrous oxide of a heavy metal, resulting in an insoluble form of the dye. Flavoring agents include natural flavors extracted from plants, such as fruits, and synthetic blends of compounds which produce a pleasant taste sensation, such as peppermint and methyl salicylate. Sweetening agents include sucrose, lactose, mannitol, syrups, glycerin, and artificial sweeteners, such as saccharin and aspartame. Suitable emulsifying agents include gelatin, acacia, tragacanth, bentonite, and surfactants, such as polyoxyethylene sorbitan monooleate (e.g., Tween* 20), poloxamers (e.g., Pluronic® F68), polyoxyethylene sorbitan monooleate 80 (e.g., T ween 8 80), and triethanolamine oleate. Suspending and dispersing agents include sodium
carboxymethylcellulose, pectin, tragacanth, Veegum8, acacia, sodium carbomethylcel lulose, hypromellose, polyvinylpyrrolidone, and lauroyl polyoxylglycerides (e.g., Gelucire® 44/14).
Preservatives include glycerin, methyl and propylparaben, benzoic add, sodium benzoate and alcohol. Wetting agents include propylene glycol monostearate, sorbitan monooleate. diethylene glycol monolaurate, and polyoxyethylene lauryl ether. Solvents include glycerin, sorbitol, ethyl alcohol, and syrup. Examples of non-aqueous liquids uti l ized in emulsions include mineral oil and cottonseed oil. Organic acids include citric and tartaric acid. Sources of carbon dioxide include sodium bicarbonate and sodium carbonate.
[00130] In some embodiments, the excipient is silicified microcrystalline cellulose (SMCC), for example. Prosolv SMCC 50, Prosolv SMCC 90, or Prosolv SMCC HD90.
[00131] In some embodiments, the excipient comprises SMCC and HPC. [00132] In some embodiments, a pharmaceutical composition provided herein comprises from about 0.5 % to about 10 % HPC, from about 0.8 % to about 5 % HPC, or from about 0.9 % to about 3 % HPC. In certain embodiments, the pharmaceutical composition comprises from about 50 % to about 99.5 % SMCC, from about 75 % to about 99.5 % SMCC, or about 90 % to about 98 % SMCC.
[00133] The percentages herein refer to weight/weight percentages of the given excipient in the pharmaceutical composition, excluding, for instance, any coating if in a tablet form, or shell of a capsule, if in a capsule.
[00134] In certain embodiments, the pharmaceutical composition provided herein consists essentially of Compound A tosylate salt and SMCC. In some embodiments, the pharmaceutical composition consists essentially of Compound A tosylate salt, SMCC and HPC. In some embodiments, the pharmaceutical composition consists essentially of Compound A tosylate salt, SMCC, optionally, HPC, and if the pharmaceutical composition is in a tablet form, a tablet coating. In some embodiments, the pharmaceutical composition consists essentially of Compound A tosylate salt, SMCC, optionally, HPC, and if the pharmaceutical composition is in a capsule, a capsule shell.
Formulating a Pharmaceutical Composition
[00135] In certain embodiments, a pharmaceutical composition provided herein can be a dry blend formulation, for example, prepared according to the dry blend methods described herein In certain embodiments, a pharmaceutical composition provided herein can be a prepared using a wet granulation method, for example, in accordance with the wet granulation methods described herein. Dry blend methods and wet granulation methods are described herein.
[00136] In another aspect, provided herein are methods for preparing a pharmaceutical composition comprising Compound A tosylate salt and one or more pharmaceutically acceptable excipient(s), where the pharmaceutical composition can be any of those provided in this application. The methods provided include, for example, a dry blend method, and, as another example, a wet granulation method.
[00137] In certain embodiments, the method for preparing a pharmaceutical composition is a dry blend method wherein crystalline Compound A tosylate salt is geometrically blended with silicified microcrystalline cel lulose. For instance, in one embodiment a dry blend method comprises mixing crystalline Compound A tosylate salt and a first fraction of silicified microcrystalline cel lulose to produce a first mixture; mixing the first mixture and a second fraction of silicified microcrystalline cellulose to produce a second mixture, which is then mixed with a third fraction of silicified microcrystalline cellulose to produce a pharmaceutical composition comprising a weight/weight percent of Compound A tosylate salt between about 0.01 % to about 4.000 %, or as described elsewhere herein.
[00138] An exemplary dry blend method for preparing a pharmaceutical composition comprises geometric blending Compound A tosylate salt with SMCC. For instance, in certain embodiments, the method comprises (a) mixing crystal line Compound A tosylate salt and a first fraction of SMCC to produce a first mixture; (b) mixing the first mixture and a second fraction of SMCC; and (c) mixing the mixture produced in step (b) and a third fraction of SMCC to produce the pharmaceutical composition. In some embodiments, the mixture produced in step (b) is geometrical ly blended with SMCC prior to being mixed in step (c). For example, in certain embodiments, after mixing to produce the mixture of first mixture and second fraction of SMCC, a fourth fraction of SMCC is added to the mixture, which is mixed to produce a mixture for use in step (c). In some embodiments, a fifth fraction of SMCC is added to the mixture and mixed to produce a mixture for use in step (c). Any number of additional fractions (e.g., a sixth fraction, a seventh fraction, an eighth fraction, a ninth fraction, a tenth fraction, etc.) of SMCC can be added and mixed to produce a mixture for use in step (c). The fractions of SMCC added in any step of the method (first fraction, second fraction, etc.) can be identical amounts, or can be different amounts, or can be the same in some cases and different in others (e.g., the first and third fractions can be the same, while the second fraction is different from the first and third fractions).
[00139) In some embodiments of the dry blending method provided herein, an excipient other than SMCC is added during one of the steps (for example, step (a), step (b) or step (c)) of the dry blending method and mixed together with the Compound A tosylate salt and SMCC. Such other excipients can, for example, be any of those known in the art and/or provided herein.
[00140] The weight/weight percent of Compound A tosylate salt in the pharmaceutical composition prepared according to the dry blend method provided herein can, for instance, be within the ranges described herein. In some embodiments, the dry blend method for preparing a pharmaceutical composition comprises:
a) mixing crystalline Compound A tosylate salt and a first fraction of silicified microcrystal line cellu lose to produce a first mixture;
b) mixing the first mixture and a second fraction of silicified microcrystalline cellulose; and c) mixing the mixture produced in step (b) and a third fraction of silicified microcrystal line cellulose to produce a pharmaceutical composition comprising a weight/weight percent of Compound A tosylate salt between about 0.01 % to about 4.000 %.
[00141] In other embodiments, the method for a pharmaceutical composition is a wet granulation method wherein Compound A tosylate salt is subject to high shear granulation using si licified microcrystalline cellulose as a carrier. For instance, one embodiment comprises dissolving crystalline Compound A tosylate salt and a binder in a solvent and mixing the resulting solution in a high shear granulator with silicified microcrystalline cellulose, and then dr ing the granules to produce a to produce a pharmaceutical composition comprising, for example, between about 0.01 % to about 4.000 %
Compound A tosylate salt and about 0.5 % to about 2.0 % binder.
[00142] An exemplary wet granulation method comprises dissolving crystall ine Compound A tosylate salt and a binder in a first volume of solvent to produce a solution; mixing the solution in a high shear granulator with SMCC to produce granulated material; and drying the granulated material to produce a to produce a pharmaceutical composition. [00143J In certain embodiments of the wet granulation method, the binder is hypromellose or
HPC. In some embodiments, the binder is HPC. In some embodiments, the weight/weight percent of HPC in the pharmaceutical composition is about 0.5 % to about 2.0 %.
(00144] In certain embodiments of the wet granulation method, the solvent is acetone, ethanol, ethyl acetate or water, or a co-solvent selected from acetone/water, ethanol/water and ethyl acetate/water. In certain embodiments, the solvent is a co-solvent of acetone/water.
(00145J In some embodiments where the solvent is acetone/water, the weight/weight ratio of acetone to water is 9: 1.
[00146] In some embodiments, the first volume comprises about 95 % to about 98 %
(weight/weight) of the solution of step (a) of the wet granulation method provided above.
[00147] The weight/weight percent of crystalline Compound A tosylate salt in the pharmaceutical composition prepared according to the wet granulation method provided herein can, for instance, be within the ranges described herein. In some embodiments, the pharmaceutical composition prepared according to the wet granulation method provided herein comprises between about 0.10 % to about 1.0 % Compound A tosylate salt and about 0.5 % to about 2.0 % binder.
[00148] In some embodiments, the wet granulation method for preparing a pharmaceutical composition comprises:
a) dissolving crystal line Compound A tosylate salt and a binder in a first volume of solvent; b) mixing the solution of step (a) in a high shear granulator with silicified macrocrystall ine cellulose; and
c) drying the granules to produce a to produce a pharmaceutical composition comprising between about 0.01 % to about 4.000 % Compound A tosylate salt and about 0.5 % to about 2.0 % binder. Unit Dosage Forms
[00149] In another aspect, provided herein are unit dosage forms. Unit dosage forms can, for instance, comprise Compound A tosylate salt in solid form and one or more pharmaceutically acceptable excipients. In certain embodiments, the unit dosages form provided herein is a capsule. In other embodiments, it is a tablet. In some embodiments the unit dosage form contains a mass of, for instance, about 25 μg, about 50 \ag, about 250 g or about 1 mg Compound A, wherein said mass excludes the tosylate portion of the salt.
[00150] In another aspect, provided herein are unit dosage forms. The unit dosage forms typically comprise a pharmaceutical composition comprising Compound A tosylate salt and one or more pharmaceutically acceptable excipients, where the pharmaceutical composition can be any of those as prov ided herein. Unit dosage forms as provided herein can, for example, be suitable for oral
administration to a subject. Unit dosage forms can, for example, be a capsule. Other examples include those where the unit dosage form is a tablet.
[00151 J In certain embodiments, a unit dose form provided herein is in a tablet, wherein the pharmaceutical composition is prepared by a wet granulation method. [00152J The amount of Compound A tosylate salt in a unit dosage form can, for example, be a low dose, such as defined herein. In certain embodiments, the amount of Compound A tosylate salt in the unit dosage form, where the following amounts refer to the amount of Compound A and do not include the mass contributed by the tosylate portion of the salt, is in a range from about 1 μg to about 8000 μg, from about 5 μ§ to about 1500 g, from about 20 to about 1250 μg, from about 25 μ to about 1000 pg, from about 25 g to about 1500 μg. from about 50 μ% to about 1250 g, from about 25 g to about 250 μg, from about 5 μg to about 30 μg, from about 20 μg to about 60 μg, from about 50 μg to about 100 μg, from about 120 g to about 250 μg, from about 20 μg to about 1 12 μg, or from about 25 μ§ to about 250 μg. In certain embodiments, the amount of Compound A tosylate salt in the unit dosage form is about 10 g, about 20 g, about 25 ug. about 30 g, about 50 g, about 75 μg, about 100 μ , about 150 μ§, about 200 μg, about 250 μg, about 1000 μg, about 1250 μ , or about 1 500 μg.
[00153] Unit dosage forms provided herein may be provided in capsules, including, for instance, soft or hard capsules. Capsules can, for example, be prepared according to methods known to the skilled artisan using materials known to the skilled artisan. Material for the capsules can include, for example, gelatin, pullulan, methylcellulose, starch, calcium alginate or hypromellose. In certain embodiments, the capsule shel l is hypromellose. Encapsulating the pharmaceutical compositions provided herein can, for instance, be performed using the Xcleodose automated encapsulator system (Capsugel, Peapack, New Jersey, USA).
[00154] The hard gelatin capsule, also known as the dry-filled capsule (DFC), consists of two sections, one slipping over the other, thus completely enclosing the active ingredient. The soft elastic capsule (SEC) is a soft, globular shell, such as a gelatin shel l, which is plasticized by the addition of glycerin, sorbitol, or a similar polyol. The soft gelatin shel ls may contain a preservative to prevent the growth of microorganisms. Suitable preservatives are those as described herein, including methyl- and propyl-parabens, and sorbic acid.
[00155] The capsules may also be coated as known by those of ski l l in the art in order to modify or sustain dissolution of the active ingredient.
[00156] Tablets can, for instance, be formed according to methods known to the skilled artisan.
Tablets may be provided as compressed tablets, tablet triturates, chewable lozenges, rapidly dissolving tablets, multiple compressed tablets, or enteric-coating tablets, sugar-coated, or film-coated tablets.
Multiple compressed tablets are compressed tablets made by more than one compression cycle, including layered tablets, and press-coated or dry-coated tablets.
[00157] Enteric-coated tablets are compressed tablets coated with substances that resist the action of stomach acid but dissolve or disintegrate in the intestine, thus protecting the active ingredients from the acidic environment of the stomach. Enteric-coatings include, but are not limited to, fatty acids, fats, phenyl salicylate, waxes, shellac, ammoniated shellac, and cellulose acetate phthalates. Sugar-coated tablets are compressed tablets surrounded by a sugar coating, which can be beneficial in covering up objectionable tastes or odors and in protecting the tablets from oxidation. Film-coated tablets are compressed tablets that are covered with a thin layer or film of a water-soluble material.
[00158] Film coating imparts the same general characteristics as sugar coating. Film coatings include polymeric coating materials, plasticizers, surfactants, and colorants. Suitable polymeric coating materials include, but are not limited to, hydroxyethylcellulose, sodium carboxymethylcellulose, hypromellose, polyvyl, hydroxypropyl cellulose, hydroxyethyl methyl cellulose, polyethylene glycol 4000, and cellulose acetate phthalate. Suitable plasticizers include, but are not limited to glycerol, propylene glycol, polyethylene glycol, dibutyl sebacete, citrate esters, and triacetin. Suitable surfactants include, but are not limited to polysorbate 80 and sodium I aur I sulfate. Suitable colorants include various iron oxides, FD&C blue no.2, and titanium dioxide. Ingredients in the film coating composition are dispersed in a suitable solvent, preferrably water, and applied onto capsules or tablets using a conventional spray coating techniques or pan coating.
[00159] The tablet dosage forms may be prepared from Compound A tosylate salt in solid form, for instance, in a crystalline form, in combination with one or more excipients described herein, including binders, disintegrants, controlled-release polymers, lubricants, diluents, and/or colorants. Flavoring and sweetening agents are especially useful in the formation of chewable tablets and lozenges
[00160] Tablets can, for example, be made according to techniques known to the skilled artisan.
Tablet presses are commercially available, for instance, from Korsch AG (Berlin, Germany), Oystar Manesty (Merseyside, England), ikusui Seisakusho Ltd. (Kyoto, Japan), Elizabeth-Hata International (North Huntingdon, PA, USA), Gea Pharma Systems Courtoy (Wommelgem, Belgium ), Ima Kill ian (Scarletallee, Germany), among others. Tablet coating equipment are commerically available, for instance, from O'Hara (Ontario, Canada), Oystar Manesty (Merseyside, England) and Vector Corporation (Marion, IA, USA).
[00161] The unit dosage forms may be provided in high-density polyethylene bottles provided with a desiccant capsule or pouch; or in foil-on-foi l blister packaging, or in blister packaging comprising apolymer film.
Methods of Treatment Using the Pharmaceutical Compositions Described Herein
[00162] Without intending to be limited to any particular theory or mechanism, it is bel ieved that, owing to its high potency as a PARP inhibitor, administration of low doses (e.g., as defined herein) of Compound A can be effective to treat indications such as those discussed herein. Such indications that can be treated include, for example, leukemia (including acute myelogenous leukemia, chronic lymphocytic leukemia), myelodysplastic syndrome, colon cancer, EBV-associated tumors (including Burkitt's lymphoma, nasopharyngeal carcinoma, lymphomas in AIDS patients, smooth muscle tumors in AIDS patients, Hodgkin's disease, non-Hodgkin's lymphoma, lymphoproliferative disease in immunosuppressed patients, leiomyosarcomas in immunosuppressed patients, EBV+ gastric cancer, EBV+ breast cancer, T-cell lymphoma), endometrial cancers (including carcinomas, sarcomas, endometrioid serous adenocarcinoma, endometrioid clear cel l adenocarcinoma, and adenosquamous carcinoma, or mixed histology, any grade), gastrointestinal stromal tumor, gliomas, glioblastoma, lymphoma (including mantle cell lymphoma), melanoma, breast (including metastatic, BRCA-positive, and BRCA-negative), ovarian (including advanced, high grade serous, platinum-sensitive, platinum- resistant, platinum-refractory, and BRCA-negative), cervical, pancreatic (including BRCA-negative), peritoneal, prostate (including BRCA-negative, metastatic, and castration resistant), hereditary nonpolyposis colon cancer (HNPCC), lung cancer (including non small cell lung carcinoma, small cell lung carcinoma), colorectal carcinomas, uterine carcinosarcoma, solid tumors (such as those of the bladder, bowel, brain, breast, endometrium, heart, kidney, lung, cancer of non-pancreatic endocrine organ (thyroid), and head and neck), and hematological tumors. Unit dosage forms comprising low doses of Compound A tosylate salt are provided such that effective amounts of the Compound A tosylate salt can be easily administered to patients having an indication to be treated. Methods of preparing
pharmaceutical compositions, including unit dosage forms comprising low dosages of Compound A tosylate salt, are provided, which methods, without intending to be bound by any theory or mechanism, can minimize loss of Compound A and can be used to consistently achieve a desired proportion of Compound A relative to inactive ingredients.
[00163] In another aspect, provided herein are methods of treatment wherein a subject is administered with a pharmaceutical composition as explained herein. In the fol lowing, it will be understood that the pharmaceutical composition to be administered to the subject is a pharmaceutical composition as provided herein, for example, comprising Compound A tosylate salt in solid (e.g., crystal line) form and one or more pharmaceutically acceptable excipients. The pharmaceutical composition can, for instance, be in a powder form, or in a unit dosage form (e.g., capsule or tablet). The pharmaceutical composition may be formulated to be suitable for oral administration to the subject.
[001641 In some or any embodiments, provided herein is a use of a pharmaceutical composition, as described herein, for the treatment of a disease or conidition, or symptom thereof, in a subject.
[00165] In certain embodiments, a method of treating cancer, or symptom thereof, comprising administering to a subject with cancer a therapeutically effective amount of a pharmaceutical composition, as described herein, is prov ided.
[00166] Certain embodiments provide a method of potentiation of cytotoxic cancer therapy in a subject in recognized need of such treatment comprising administering to the subject a therapeutically acceptable amount of a pharmaceutical composition, as described herein,.
[00167] In some embodiments, provided herein is a method for the treatment of cancer, comprising administering to a subject in need of treatment a therapeutical ly effective amount of a pharmaceutical composition, as described herein, in combination with ionizing radiation or one or more chemotherapeutic agents. In some embodiments, the pharmaceutical composition described herein is administered simultaneously with ionizing radiation or one or more chemotherapeutic agents. In other embod iments, the pharmaceutical composition described herein is administered sequentially with ionizing radiation or one or more chemotherapeutic agents. Ion izing radiation and chemotherapeutic agents are known to those skilled in the art.
[00168] In some or any embodiments, the therapeutic agent(s) is an alkylating agent, such as methyl methanesulfonate (MMS), temozolomide and dacarbazine (DTIC); a topoisomerase-1 inhibitor such as Topotecan, Irinotecan. Rubitccan. Exatecan, Lurtotecan. Gimetecan, Diflomotecan
(homocamptothecins), 7-substituted non-si latecans, the 7-si lyl camptothecins (BNP 1350), SN38, N 012 (a SN-38-releasing nanodevice constructed by covalently attaching SN-38 to the block copolymer PEG- PGIu, followed by self-assembly of amphiph ilic block copolymers in aqueous media), and XR
1 1576/MLN 576; alemtuzumab; arsenic trioxide; asparaginase (pegylated or non-); bevacizumab;
cetuximab; platinum-based compounds such as cisplatin, carboplatin, oxaliplatin, and triplatin tetranitrate; c ladribine;. daunorubicin; doxorubicin; idarubicin; fludarabine; 5-fluorouracil; gemtuzumab;
methotrexate; Pacl itaxel™; taxol; temozolomide; thioguanine; hormone therapies such as an antiestrogen, an antiandrogen, and gonadotropin releasing hormone analogues; interferons such as alpha interferon; nitrogen mustards such as busulfan, melphalan, and mechlorethamine; retinoids such as tretinoin; tyrosine kinase inhibitors such as gefinitinib and imatinib; a proteasome inhibitor such as bortezomib; or agents to treat signs or symptoms induced by such therapy including allopurinol, filgrastim,
granisetron/ondansetron/palonosetron, and dronabinol.
[00169] In some or any embodiments, provided is a method for the treatment of a cancer, comprising administering to a subject in need of treatment a pharmaceutical composition as described herein in combination with a topoisomerase inhibitor. In some or any embodiments, the topoisomerase inhibitor is SN38, irinotecan, or NK.012. In some or any embodiments, the topoisomerase inhibitor is irinotecan. In some or any embodiments, the cancer is breast cancer.
[00170] In some or any embodiments, provided is a method for the treatment of a cancer, comprising administering to a subject in need of treatment a pharmaceutical composition as described herein in combination with a platin. In some or any embodiments, the plat in is cisplatin, carboplatin, oxaliplatin, or triplatin tetranitrate. In some or any embodiments, the topoisomerase inhibitor is cisplatin. In some or any embodiments, the cancer is breast cancer.
[00171 ] In some or any embodiments, provided is a method for the treatment of a cancer, comprising administering to a subject in need of treatment a pharmaceutical composition as described herein in combination with temozolomide. In some or any embodiments, the cancer is colorectal cancer.
[0 172] In some embodiments, provided herein is a method of treatment of a cancer deficient in
Homologous Recombination (HR) dependent DNA double strand break (DSB) repair pathway, which includes administering to a subject in need of treatment a therapeutically effective amount of a pharmaceutical composition, as described herein. In certain embodiments, the cancer includes one or more cancer cells having a reduced or abrogated abil ity to repair DNA DSB by HR relative to normal cells. In some embodiments, the cancer cells have a BRCA 1 or BRCA2 deficient phenotype. In some embod iments, the cancer cells are deficient in BRCA 1 or BRCA2. In some embodiments, the methods provided herein involve treatment of an individual who is heterozygous for a mutation in a gene encoding a component of the HR dependent DNA DSB repair pathway. In certain embodiment, the individual is heterozygous for a mutation in BRCA 1 and/or BRCA2.
100173] In certain embodiments, the cancer to be treated comprises phosphatase and tensin homolog (PTEN) deficient cel ls (e.g., cel ls in which PTEN is mutated or its expression is minimal or absent). In certain embodiments, the cancer to be treated comprises cel ls with a PTEN gene mutation. Exemplary cancers associated with PTEN deficiency can include, for example, glioblastoma, endometrial cancer, prostate cancer, lung cancer and breast cancer.
[00174] In certain embodiments, the cancer to be treated is one resulting from an activation mutation of the Wnt signaling pathway ("Wnt mediated cancers")- It will be understood that by
"activation mutation of the Wnt signal ing pathway" it is meant to include, for example, oncogene mutations in genes leading to accumulation of β-catenin in cancer cells, gain-of-function mutations in the CTNNB 1 gene (which encodes β-catenin), mutations in the APC tumor suppressor gene or mutations in the AXIN2 gene. It is believed, for instance, and without intending to be limited to any theory or mechanism, that useful treatment of a Wnt mediated cancer may be had by way of inhibiting tankyrase ("TNK.S"), an enzyme having poly(ADP-ribose) polymerase activity. Exemplary Wnt mediated cancers that can be treated according to the methods provided herein include, for instance, bladder cancer, breast fibromatoses, cervical cancer, colorectal cancer, colon carcinoma, Desmoid tumor, esophageal adenocarcinoma, familial adenomatous polyposis, fundic gland polyps, gastric carcinoma, gastric adenoma, gastrointestinal carcinoid tumor, hepatoblastoma, hepatocellular carcinoma, juvenile nasopharyngeal angiofibroma, non-Hodgkin lymphoma, lung adenocarcinoma, medulloblastoma, melanoma, ovarian carcinoma, pancreatic cancer (including, for example, non-ductal solid
pseudopapil lary and non-ductal acinal cell carcinoma) pancreoblastoma, pilomatricomas, prostate cancer, small intestinal adenocarcinoma, synovial sarcoma, thyroid carcinoma, uterine cervical cancer, uterine endometrial cancer and Wilm's tumor.
[00175] In some embodiments of the method of treatment of a cancer, or symptom thereof, provided herein, the cancer is bladder cancer, breast cancer (including metastatic, BRCA-positive, and BRCA-negative), cervical cancer, colon cancer, colorectal cancer, EBV-associated tumors (including Burkitt's lymphoma, nasopharyngeal carcinoma, lymphomas in AIDS patients, smooth muscle tumors in AIDS patients, Hodgkin's disease, non-Hodgkin's lymphoma, lymphoproliferative disease in immunosuppressed patients, leiomyosarcomas in immunosuppressed patients, EBV+ gastric cancer, E V r breast cancer, T-cell lymphoma), endometrial cancers (including carcinomas and sarcomas), gastrointestinal stromal tumor, gliomas, glioblastoma (including, for instance, gl ioblastoma multiforme and anaplastic astrocytoma), head and neck cancer, hepatocellular carcinoma, hereditary nonpolyposis colon cancer (HNPCC), kidney cancer, leukem ia (including acute myelogenous leukemia, chronic lymphocytic leukemia), lung cancer (including non small cell lung carcinoma, small cel l lung carcinoma), lymphoma (including mantle cell lymphoma), medulloblastoma, melanoma, meningioma, myelodysplastic syndrome, ovarian cancer (including advanced, high grade serous, platinum-sensitive, platinum-resistant, platinum-refractory, and BRCA-negative), pancreatic cancer (including BRCA- negative), peritoneal cancer, prostate cancer (including BRCA-negative, metastatic, and castration resistant), renal cancer, thyroid cancer, uterine carcinosarcoma, or uterine cancer.
[00176] In certain embodiments, provided herein is a method of treating a disease or condition, or symptom thereof, associated with a PTEN deficiency comprising administering to a subject with the disease or condition associated with a PTEN deficiency a therapeutically effective amount of a pharmaceutical composition, as described herein. Exemplary diseases and conditions associated with a PTEN deficiency include, for example, Cowden syndrome, Bannayan-Riley-Ruvalcaba syndrome, Lhermitte-Duclos disease, Proteus syndrome, Proteus-like syndrome or hamatome tumor syndrome.
[00177] Certain embodiments provide a method of treating inflammatory diseases including, but not limited to, arthritis, gout, inflammatory bowel disease, CNS inflammation, multiple sclerosis, allergic encephalitis, sepsis, septic shock, hemorrhagic shock, pulmonary fibrosis, and uveitis in a subject in recognized need of such treatment comprising administering to the subject a therapeutically acceptable amount of the pharmaceutical composition.
[00178] Certain embodiments provide a method of treating immunological diseases or disorders such a rheumatoid arthritis and septic shock in a subject in recognized need of such treatment comprising administering to the subject a therapeutically acceptable amount of the pharmaceutical composition.
[00179] Ceri in embodiments provide a method of treating degenerative diseases including, but not limited to, diabetes and Parkinson's disease in a subject in recognized need of such treatment comprising administering to the subject a therapeutically acceptable amount of the pharmaceutical composition.
[00180] In certain embodiments of this use, the precise amount of Compound A tosylate salt administered to the subject in the pharmaceutical composition depends on the patient's state of health, weight, and the l ike. In some embodiments, it is considered appropriate for the caregiver to determine such therapeutical ly 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.
[00181] In certain embodiments, the pharmaceutical composition is administered to the subject in a regimen of about 1 week, about 2 weeks, about 3 weeks, about 1 month, about 2 months or about 6 months. In s me embodiments the tosylate salt is administered daily in the regimen. In other embodiments, the tosylate salt is administered for 2, 3 or 4 days per week, interspersed or fol lowed by days of the weekly regimen where the tosylate salt is not administered.
[00182] In certain instances, a patient's condition does not improve or does not significantly improve following administration of the pharmaceutical composition, and, upon the doctor's discretion, the administration of the compound 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.
[00183] In certain cases wherein the patient's status does improve or does not substantially improve, upon the doctor's discretion, the administration of the active ingredient is 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 v aries 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, 150 days, 180 days, 200 days, 250 days, 280 days, 300 days, 320 days, 350 days, or 365 days.
[00184] In certain embodiments, the amount of Compound A tosylate salt that corresponds to an effective amount of the pharmaceutical composition 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 1 to about 8000 μg per day, in a specific embodiment about 10 to about 5000 μg per day. In certain embodiments, the amount of Compound A tosylate salt to be administered is between about 50 μg to about 5000
Figure imgf000080_0001
between about 50 μ to about 1 500 μg, between about 50 g to about 1000 μg, between about 50 μg to about 500 iig, between about 50 g to about 250 g, between about 50 g to about 200 μg, between about 50 μg to about 150 μg, between about 50 μg to about 100 μg, between about 25 μg to about 2500 μg, between about 25 μg to about 1000 g, between about 25 ug to about 250 μο, between about 25 μg to about 150 μg, or between about 25 μg to about 75 μg. 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.
[00185] In certain embodiments, the daily dosages of Compound A tosylate salt in the pharmaceutical composition appropriate for the methods described herein are from about 0.01 to about 100 μ /kg per body weight. It wi l l be understood that these amounts refer specifical ly to the mass of Compound A, excluding the mass contributed by the tosylate portion of Compound A tosylate salt, or contributed by any excipient in the pharmaceutical composition. In some embodiments, an indicated daily dosage in the larger subject, including, but not limited to, humans, is in the range from about of about 25 to about 8000 μ§, 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 those described herein. 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.
Methods of Treating Multiple Myeloma
[00186] In some embodiments, a compound of Formula (I), (LA), (IA I ), (IA2), (II), (IIA), (IIB),
(IIC) , or (IID) as defined in the Summary of the Inventionor in any of the embodiments described herein (or a single isomer, stereoisomer, or enantiomer or mixture thereof, optionally as a pharmaceutical ly acceptable salt, pharmaceutically acceptable solvate, or pharmaceutically acceptable prodrug thereof) is used in the treatment of multiple myeloma, which includes administering to a subject in need of treatment a therapeutical ly-effective amount of the compound, and in one embodiment in the form of a
pharmaceutical composition. In another embodiment the pharmaceutical composition is as described herein and in another example comprises one or more pharmaceutical ly acceptable carrier, excipient(s), binder and/or diluent. In another embodiment the compound of Formula I is ( 8.S',9/?)-5 - fl uoro-8 -( 4- nuorophenyl)-9-( l -methyl- 1 //- l ,2,4-triazol-5-y
or a pharmaceutically acceptable salt thereof.
[00187] In some embodiments, disclosed is a method of treating multiple myeloma comprising administering to a subject in need thereof a pharmaceutical combination which comprises a
therapeutical ly-effective amount of a compound of Formula (I), (IA), (IA 1 ), (IA2), (II), (IIA), (IIB), (IIC),
(IID) as defined in the Summary of the Invention or in any of the embodiments described herein (or a single isomer, stereoisomer, or enantiomer or mixture thereof, optionally as a pharmaceutically acceptable salt, pharmaceutically acceptable solvate, or pharmaceutically acceptable prodrug thereof) and a proteasome inhibitor. In one embodiment the proteasome inhibitor is bortezomib, disulfiram, epigallocatechin-3-gallate, salinosporamide A (marizomib), carfilzomib, ONX 0912 (oprozomib), CEP-
1 8770 (delanzomib), or MLN9708 (ixazomib citrate); or a pharmaceutically acceptable salt thereof. In another embodiment, the proteasome inhibitor is bortezomib. In another embodiment the compound of Formula I is (8S,9R)-5-fluoro-8-(4-fluorophenyl)-9-( l -methyl- I H- l , 2,4-triazol-5-yl)-8,9-dihydro-2/ - pyrido[4,3,2-ife]phthalazin-3(7H)-one or a pharmaceutically acceptable salt thereof. In another embodiment, the Compound of Formula (IA), (IA l ), (IA2), (II), (IIA), (IIB), (IIC), (IID) as defined in the Summary of the Invention or in any of the embodiments described herein (or a single isomer, stereoisomer, or enantiomer or mixture thereof, optionally as a pharmaceutically acceptable salt, pharmaceutically acceptable solvate, or pharmaceutically acceptable prodrug thereof) is used as a pharmaceutical composition, as described herein, additionally comprising one or more pharmaceutical ly acceptable carrier, excipient(s), binder and/or diluent.
[00188] In another embodiment, disclosed is a method of treating multiple myeloma with a pharmaceutical composition comprising 1 ) a compound, optionally as a pharmaceutically acceptable salt thereof, of Formula (I), (IA), (IA l ), (IA2), (II), (IIA), (IIB), (IIC), or (IID) as defined in the Summary of the Invention or in any of the above embodiments (or a single isomer, stereoisomer, or enantiomer or mixture thereof, optionally as a pharmaceutically acceptable salt, pharmaceutically acceptable solvate, or pharmaceutically acceptable prodrug thereof) and 2) one or more pharmaceutically acceptable carriers), excipient(s), binder or diluent(s) thereof. In another embodiment the compound of Formula I is (8S,9i?)-5- fluoro-8-(4-fluorophenyl)-9-( 1-methyl- 1 H- 1 ,2,4-triazol-5-yl)-8,9-dihydro-2H-pyrido[4,3,2-^]phthalazin- 3(7H)-one or a pharmaceutically acceptable salt thereof.
[00189] In another embodiment, disclosed is a method of treating multiple myeloma comprising administering to a subject in need of treatment a therapeutically effective amount of a compound of Formula (I), (IA), (IA1), (IA2), (II), (IIA), (I IB), (HQ, or (IID) as defined in the Summary of the Invention or in any of the above embodiments (or a single isomer, stereoisomer, or enantiomer or mixture thereof, optionally as a pharmaceutically acceptable salt, pharmaceutically acceptable solvate, or pharmaceutically acceptable prodrug thereof). In another embodiment the compound of Formula I is (8. ,9ii)-5-fluoro-8-(4-fluorophenyl)-9-( l -methyl- [H- l ,2,4-triazol-5-yl)-8,9-dihydro-2H-pyndo[4,3,2- i/e]phthalazin-3(7H)-one or a pharmaceutically acceptable salt thereof.
[00190] In another embodiment is the use of a compound of Formula (I), (IA), (IA 1 ), (IA2), (II),
(IIA), (IIB), (IIC), or (IID) as defined in the Summary of the Invention or in any of the above embodiments (or a single isomer, stereoisomer, or enantiomer or mixture thereof, optionally as a pharmaceutically acceptable salt, pharmaceutically acceptable solvate, or pharmaceutical ly acceptable prodrug thereof) in the formulation of a medicament for the treatment of multiple myeloma. In another embodiment, the disease is mediated by poly(ADP-ribose)polymerase. In another embodiment the compound of Formula I is (8S',9R)-5-fluoro-8-(4-fIuorophenyl)-9-( I -methyl- 1 //- l,2,4-triazol-5-y l)-8,9- dihydro-2if-pyrido[4,3,2- e]phthalazin-3(7/i)-one or a pharmaceutically acceptable salt thereof.
[00191] In another embodiment, disclosed is the use of a compound of Formula (I), (IA), (IA 1 ),
(IA2), (II), (IIA), (IIB), (IIC), or (IID) as defined in the Summary of the Invention or in any of the above embodiments (or a single isomer, stereoisomer, or enantiomer or mixture thereof, optionally as a pharmaceutically acceptable salt, pharmaceutically acceptable solvate, or pharmaceutically acceptable prodrug thereof) for the treatment of multiple myeloma. In another embodiment the compound of Formula I is (8S,9jR)-5-fluoro-8-(4-fluorophenyl)-9-( l -methyl- l /-l,2,4-triazol-5-yl)-8,9-dihydro-2H-py i e] phthal azin-3 (7H)-one or a pharmaceutically acceptable salt thereof.
[00192] In another aspect is an article of manufacture, comprising packaging material, a compound of Formula (I), (IA), (IA 1 ), (IA2), (II). (IIA), (IIB). (IIC), or (IID) as defined in the Summary of the Invention or in any of the above embodiments (or a single isomer, stereoisomer, or enantiomer or mixture thereof, optionally as a pharmaceutically acceptable salt, pharmaceutically acceptable solvate, or pharmaceutically acceptable prodrug thereof), optionally a proteasome inhibitor, and a label, wherein the compound is effective for modulating the activity of the enzyme poly(ADP-ribose)po!ymerase, or for treatment, prevention or amelioration of one or more symptoms of multiple myeloma, wherein the compound is packaged within the packaging material, and wherein the label indicates that the compound, or pharmaceutically acceptable salt, pharmaceutically acceptable N-oxide, pharmaceutically active metabolite, pharmaceutically acceptable prodrug, or pharmaceutically acceptable solvate thereof, or a pharmaceutical composition comprising such a compound is used for treatment, prevention or amelioration of one or more symptoms of multiple myeloma. In some or any embodiments the proteasome inhibitor, when present, is bortezomib. In another embodiment the compound of Formula I is (85,9R)-5- tluoro-8-(4-fluorophenyl)-9-( 1 -methyl- 1 //- 1.2,4-triazol-5-yl)-8.9-dihydrcv2//-pyrido[4,3,2 /£'lphthalazin- 3(7H)-one or a pharmaceutically acceptable salt thereof.
[00193] Certain embodiments provide a use of a compound of Formula (I), (IA), (IA 1 ), (IA2),
(II), (HA), (IIB), (IIC), or (IID) as defined in the Summary of the Invention or in any of the above embodiments (or a single isomer, stereoisomer, or enantiomer or mixture thereof, optionally as a pharmaceutically acceptable salt, pharmaceutically acceptable solvate, or pharmaceutically acceptable prodrug thereof), to prepare a medicament for treating multiple myeloma in a subject in recognized need of such treatment. In another embodiment the compound of Formula I is (8S,9R)-5-fluoro-8-(4- fluorophenyl)-9-( l -methyl-lH-l ,2,4-triazol-5-yI)-8,9-dihydro-2H-pyridot4,3,2-Je]phthalazin-3(7H)-one or a pharmaceutically acceptable salt thereof.
[00194] In one embodiment, disclosed herein is a pharmaceutical composition comprising a compound, pharmaceutically acceptable salt, pharmaceutically acceptable N-oxide, pharmaceutically active metabol ite, pharmaceutically acceptable prodrug, or pharmaceutically acceptable solvate of any of the compounds disclosed herein. In some embodiments, the pharmaceutical compositions further comprises one or more pharmaceutically acceptable excipient(s). In certain embodiments, the pharmaceutical composition further comprises a second pharmaceutically active ingredient.
[00195] 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 admin istered 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 adm inistered rectal ly to the subject.
[00196] 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; (i i) multiple times over the span of one day; (iii) continually; or (iv) continuously.
[00197} 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.
[00198] 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.
[00199J 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.
DEFINITIONS
Abbreviations
Figure imgf000084_0001
Abbreviation Meaning
PSA particle size analysis
PTEN phosphatase and tensin homolog
PVP polyvinylpyrrolidone
FLH relative humidity
RSD relative standard deviation
SEC soft elastic capsule
SEM scanning electron microscopy
SMCC silicified microcrystalline cellulose
TGA thermogravimetric analysis
THF tetrahydrofuran
TsOH p-toluenesulfonic acid
UV ultraviolet
w/w weight/weight
XRPD X-ray powder diffraction
[00200] To facilitate understanding of the disclosure set forth herein, a number of terms are defined herein. Generally, the nomenclature used herein and the laboratory procedures in organic chemistry, medicinal chemistry, and pharmacology described herein are those well known and commonly employed in the art. 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.
[00201J 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.
[00202] 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. [00203) As used throughout this application and the appended claims, the following terms have the following meanings:
[00204] 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.
[00205] 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, etho y, propoxy, 2-propoxy, butoxy, tert-butoxy, pentyloxy, and hexyloxy.
[00206] The term "'alky!" as used herein, means a straight, branched chain, or cyclic (in this case, it would also be known as "cycloalkyl") hydrocarbon containing from I - 10 carbon atoms. Illustrative examples of alkyl include, but are not l imited 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-dirnethylhexyl, n-heptyl, n-octyl, n-nonyl, and n-decyl.
[00207] The term "Ci-Q-alkyl" as used herein, means a straight, branched chain, or cycl ic (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.
[00208] The term "cycloalkyl" as used herein, means a monocycl ic 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 examples of
Figure imgf000086_0001
Figure imgf000087_0001
, In some embodiments, depending on the structure, a cycloalkyl group is a monoradical or a di adical (e.g., a cyc loalkylene group).
[00209] 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, alkoxyalkyi, alkoxycarbonyl, alkyl, alkylcarbonyl, alkylcarbonyloxy, alkylthio, alkylthioalkyl, alkynyl, carboxy, cyano, formyl, haloalkoxy, haloalkyl, halogen, hydroxy I, hydroxyalkylene, mercapto, oxo, -NR< RD. and
(NRcRo)carbonyl.
[00210] 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-cyclobutylethyI, cyclopentylmethyl, cyclohexylmethyl, and 4-cyc lohepty Ibuty I .
[00211] 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
[00212] 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.
[00213] The term "alkoxyalkyi" 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 alkoxyalkyi include, but are not limited to, 2-methoxyethyl, 2-ethoxyethyl, tert-butoxyethyl and methoxymethyl.
[00214] 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.
[00215] 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.
[00216] The term "alkylcarbonyl" 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 alkylcarbonyl include, but are not limited to, acetyl, 1 -oxopropyl, 2,2-d imethy 1- 1 -oxopropy I, 1-oxobutyl, and 1 -oxopentyl.
[00217] The term "alkylcarbonyloxy" as used herein, means an alkylcarbonyl 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-butylcarbonyloxy.
[00218] 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. [00219] The term "alkylthioalkyi" 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 alkylthioalkyi include, but are not limited to, methylthiomethyl, 2-(ethylthio)ethyl, butylthiomethyl, and hexylthioethyl.
[00220] 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.
[00221] 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.
100222] 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, naphthalcnyl, phenanthrenyl, anthracenyl, fluorenyl, and indenyl.
[00223] 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, alkoxy, alkoxyalkyl, alkoxycarbonyl, alkyl, alkylcarbonyl, alkylcarbonyloxy, alkylthio, alkylthioalkyi, alkynyl, carbonyl, cyano, formyl, haloalkoxy, haloalkyl, halogen, hydroxyl, hydroxyalkylene, mercapto, nitro, -N RC D, and -C(0)(NRCRD)-
[00224] The term "arylalkyi" 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 arylalkyi include, but are not limited to benzyl, 2-phenylethyl, -phenylpropyl, l -methyl-3-phenylpropyl, and 2- naphth-2-ylethyl.
[00225] The term "halo" or "halogen" as used herein, means a -CI, -Br, -I or -F.
[00226] The term "mercapto" as used herein, means a -SH group.
[00227] The term "oxo" as used herein, means a =0 group.
[00228] 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.
[00229] The terms "haloalkyl" and "haloalkoxy" as used herein, include alkyl 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 al l 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 haloaikyl and haloalkoxy groups, respectively, in which the halo is fluorine. In certain embodiments, haloalkyls are optionally substituted.
[00230] 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.
[002311 The term "heteroalkyl" as used herein, includes optional ly substituted alkyl radical 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.
[00232] 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.
[00233] 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.
[00234] 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.
[00235] 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
Figure imgf000089_0001
depending on the structure, a heteroaryl group is a monoradical or a diradical (i.e., a heteroarylene group).
[00236] 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, haloaikyl, halogen, hydroxy!, hydroxyalkylene, mercapto, nitro, -N RCRD, and -C(0)(NRCRD). [00237] 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.
[00238] The term "heterocycloalkyl"' or "non-aromatic heterocycie" 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 heterocycie" 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, heterocycloalkyis contain one or more carbonyl or thiocarbonyl groups such as, for example, oxo- and thio-containing groups. Examples of heterocycloalkyis 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-oxathi in, 1 ,4-oxathiane, tetrahydro- l ,4-thiazine, 2H- l ,2-oxazine , maleimide, succinimide, barbituric acid, thiobarbituric acid, dioxopiperazine, hydantoin, dihydrouracil, morpholine, trioxane, hexahydro- 1 ,3,5-triazine, tetrahydrothiophene, tetrahydrofuran, pyrroline, pyrrolidine, pyrrolidone, pyrrolidine, 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. Il lustrative examples of heterocycloalkyl groups, also referred to as non-aromatic heterocycles, include, but are not limited to
Figure imgf000090_0001
The term heterocycloalkyl also includes all ring forms of the carbohydrates, including but not limited to the monosaccharides, the disaccharides and the oligosaccharides.
[00239] The term "heterocycie" refers to heteroaryl and heterocycloalkyl used herein, refers to groups containing one to four heteroatoms each selected from O, S and N, wherein each heterocycie 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 heterocycie is indicated (e.g., Ci-C6 heterocycie), at least one other atom (the heteroatom) must be present in the ring.
Designations such as "C Cs heterocycie" 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 heterocycie ring has additional heteroatoms in the ring. Designations such as "4-6 membered heterocycie" refer to the total number of atoms that are contained in the ring (i.e., a four, five, or six 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 pyrrol idinyl, tetrahydrofuranyl, dihydrofuranyl, tctrahydrothienyl, tetrahydropyranyl, dihydropyran l, tetrahydrothiopyranyl, piperidino. morpholino, thiomorpholino, thioxanyl, piperazinyl, azetidinyl, oxetanyl, thietanyl, homopiperidinyl, oxepanyl, thiepanyl, oxazepinyl, diazepinyl, thiazepinyl, 1 ,2,3,6-tetrahydropyridinyl, 2-pyrrolinyl, 3-pyrrol inyl, indolinyl, 2H-pyranyl, 4H-pyranyl, dioxanyl, 1 ,3-dioxolanyl, pyrazolinyl, dithianyl, dithiolanyl, dihydropyranyl, dihydrothienyl, dihydrofuranyl, pyrazol idinyl, imidazolinyl, imidazolidinyl,
3-azabicyclo[3.1.0]hexanyl, 3-azabicyclo[4. L0]heptanyl, 3H-indolyl and quinolizinyl. Examples of heteroaryl groups are pyridinyl, imidazolyl, pyrimidinyl, pyrazolyl, triazolyl, pyrazinyl, tetrazolyl, fury I, 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 -attached where such is possible. For instance, in some embodiments, a group derived from pyrrole is pyrrol- 1 - I (N-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 Λ'-attached) or imidazol-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 monoradical or a diradical (i.e., a heterocyclene group).
[ 00240] The heterocyc les described herein 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, -N RCR¾ and -C(0)(NRCRD)- [00241 ] The term "heterocycloalkoxy" refers to a -OR group where R is -alky l-heterocyc loal ky I group, where the alkyl and heterocycloalkyl arc as defined herein, appended to the parent molecular moiety through an alkoxy group. [00242] The term "heterocycloalkylthio" refers to a, -SR group where R is -alkyl-heterocycloalkyl group where the alkyl and heterocycloalkyl are as defined herein, appended to the parent molecular moiety through an alkylthio group.
[00243] The term "heterocyclooxy" refers to a heterocycloalkyl group, as defined herein, appended to the parent molecular moiety through an oxygen atom.
[00244] The term ''heterocyclothio" refers to a heterocycloalkyl group, as defined herein, appended to the parent molecular moiety through a sulfur atom.
[00245J The term "heteroarylalkoxy" refers to an -OR group where the R is heteroarylalkyl, as defined herein.
[00246] The term "heteroarylalkylthio" refers to an -SR group where R is heteroarylalkyl group, as defined herein.
[00247] The term "heteroaryloxy" refers to a heteroaryl group, as defined herein, appended to the parent molecular moiety through an oxygen atom.
[00248] The term "heteroarylthio" refers to a heteroaryl group, as defined herein, appended to the parent molecu lar moiety through a sulfur atom.
[00249] 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 cyclopentyi, pyrrole, furan, and thiophene are 5-membered rings.
[00250] 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 l imited to, 2- azabicyclo[2.2.1 ]heptanyl, 7- azabicyclo[2.2.1 ]heptanyl, 2- azabicyclo[3.2.0]heptanyl, 3-azabicyclo[3.2.0]heptanyl, 4- azasp iro[2.4 ] heptan I, 5- azaspiro[2.4]heptanyl, 2-oxa-5- azabicyclo[2.2. l ]heptanyl, 4- azaspiro[2.5]octanyl, 5- azasp i ro[2.5 ]octany 1 , 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 jnonanyl, 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- l H-quinolizinyl, decahydropyrido[ 1 ,2-a]azepinyl, decahydro- 1 //- pyridof l ,2-a]azocinyl, 1 -azabicyclo[2.2.1 Jheptanyl, 1 -azabicyclo[3.3.1 Jnonanyl, quinuclidinyl, and 1 - azabicyclo[4.4.0]decanyl.
[00251] The term hydroxyalkylene" as used herein, means at least one hydroxy I group, as defined herein, is appended to the parent molecular moiety through an alkylene group, as defined herein.
I llustrative examples of hydroxyalkylene include, but not limited to hydroxy methylene, 2-hydroxy- ethylene, 3-hydroxypropylene and 4-hydroxyheptylene. [00252] The term "N RCN RD" as used herein, means two group, RC and RD, which are appended to the parent molecular moiety through a nitrogen atom. Rc and RD are each independently hydrogen, alky I, and alky lcarbonyl. Illustrative examples of RC D include, but are not limited to, amino, methylamino, acetylamino, and acetylmethylamino.
[002531 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.
[00254] As used herein, the term "sulfonyl" refers to a -S(=0)2-R, where R is selected from the group consisting of alkyl, cycloalkyi, aryl, heteroaryl (bonded through a ring carbon) and heterocycloalkyi (bonded through a ring carbon).
[00255] As used herein, the term "isocyanato" refers to a group of formula -NCO.
[00256] As used herein, the term "thiocyanato" refers to a group of formula -CNS.
[00257] As used herein, the term "isothiocyanato" refers to a group of formula -NCS.
[00258] As used herein, the substituent "R" appearing by itself and without a number designation refers to a substituent selected from among from alkyl, cycloalkyi, aryl, heteroaryl (bonded through a ring carbon) and non-aromatic heterocycle (bonded through a ring carbon),
[00259] The term "substituted" or "substituent(s)" means that the referenced group is optionally substituted (substituted or unsubstituted) with one or more additional group(s) individually and independently selected from alkyl, cycloalkyi, aryl, heteroaryl, heteroalicyclic, hydroxy, alkoxy, aryloxy, mercapto, alkylthio, arylthio, alkylsulfoxide, arylsulfoxide, alkylsulfone, arylsulfone, cyano, halo, carbonyl, thiocarbonyi, isocyanato, thiocyanato, isothiocyanato, nitro, perhaioalkyi, 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(=OK -S(=0)2-, -NH-, -NHC(O)-, -C(0)NH-, S(=0)2NH-, -NHS(=0)2, -OC(0)NH-, - NHC(0)0-, -(substituted or unsubstituted C C6 alkyl), or -(substituted or unsubstituted C2-C6 alkenyl); and each R5 is independently selected from H, (substituted or unsubstituted lower alkyl), (substituted or unsubstituted lower cycloalkyi), heteroaryl, or heteroalkyl.
[00260] "DNA-damaging agent" as used herein is melphalan, cyclophosphamide, bendamustine, or radiation.
[00261 ] "Proteasome inhibitor" means an inhibitor which blocks the action of proteasomes, e.g. p53 protein, and include, but are not limited to, bortezomib, disulfiram, epigallocatechin-3-gallate, salinosporamide A (marizomib), carfilzomib. ONX 0912 (oprozomib), C EP- 1 8770 (delanzomib), and MLN9708 (ixazomib citrate); or a pharmaceutically acceptable salt thereof.
[00262] 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-phenylthiomethyl oxime, 1 -isopropoxycyclohex l oxime, and the like. Examples of amine protecting groups include, but are not limited to, tert-butoxycarbonyl (Boc) and carbobenzyloxy (Cbz).
[00263] The term "optional ly substituted" as defined herein, means the referenced group is substituted with zero, one or more substituents as defined herein.
[00264] The term "protected-hydroxy" refers to a hydroxy group protected with a hydroxy protecting group, as defined above.
[00265] As used herein, a reference to "(8S,9R)-5-fluoro-8-(4-fluoropheny l)-9-( I -methyl- 1 H- l ,2,4-triazol-5-yl)-8,9-dihydro-2//-pyri )-one" or to its formula,
Figure imgf000094_0001
Compound A
will, unless otherwise noted or made clear in the context in which the reference is used, be a reference to the free base form of the compound. The term "Compound A tosylate salt" refers to a tosylate salt of Compound A, comprising a cation of Compound A and an anion from -toluenesulfonic acid, where the anion is referred to herein as "tosylate."
[00266] It will be understood that a tosylate salt of (8S,9^)-5-tluoro-8-(4-fluorophenyl)-9-( 1 - methyl-1 H- 1 ,2,4-triazol-5-yl)-8,9-dihydro-2 /-pyrido[4,3,2-(ie]phthalazin-3(7//)-one comprises a cation of (85,9y?)-5-fluoro-8-(4-f1uorophenyl)-9-( l -methyl- l H- l ,2,4-triazol-5-yl)-8,9-dihydro-2H-pyrido[4,3 cfe]phthalazin-3(7H)-one (e.g., in one embodiment, protonated at one atomic position, or in other embodiments, protonated at more than one atom ic position) and an anion of p-toluenesulfonic acid, where the anion is referred to herein as "tosylate." In certain embodiments, solid forms of (8S,9R)-5-fluoro-8-(4- fluorophenyl)-9-( l -methyl- 1H- 1 , 2,4-triazol-5-yl)-8,9-dihydro-2H-pyrido[4,3,2-rfe]phthalazin-3(7//)-one tosylate salt will comprise a cation to anion molar ratio of about 1 : 1. In certain embodiments, the cation to anion molar ratio in the solid salt will be about 1 : 1 ,33, about 1 : 1 .5 or about 1 :2.
[00267] As used herein, and unless otherwise specified, the terms "about" and "approximately," when used in connection with doses, amounts, or weight percent of ingredients of a composition or a dosage form, mean a dose, amount, or weight percent that is recognized by those of ordinary skil l in the art to provide a pharmacological effect equivalent to that obtained from the specified dose, amount, or weight percent. Specifically, the terms "about" and "approximately," when used in this context, contemplate a dose, amount, or weight percent within 15%, within 10%, within 5%, within 4%, within 3%, within 2%, within 1 %, or within 0.5% of the specified dose, amount, or weight percent. [00268] As used herein, and unless otherwise specified, the terms "about" and "approximately," when used in connection with a numeric value or range of values which is provided to describe a particular solid form, e.g. , a specific temperature or temperature range, such as, for example, that describing a melting, dehydration, desolvation or glass transition; a mass change, such as, for example, a mass change as a function of temperature or humidity; a solvent or water content, in terms of, for example, mass or a percentage; or a peak position, such as, for example, in analysis by, for example, '"'C NMR, DSC, TGA and XRPD; indicate that the value or range of values may deviate to an extent deemed reasonable to one of ordinary skil l in the art while still describing the particular solid form. Specifically, the terms "about" and "approximately," when used in this context, indicate that the numeric value or range of values may vary by 5%, 4%, 3%, 2%, 1 %, 0.9%, 0.8%, 0.7%, 0.6%, 0.5%, 0.4%, 0.3%, 0.2% or 0.1 % of the recited value or range of values while still describing the particular solid form.
[00269] As used herein, the term "mixing" can refer to any process by which two or more ingredients are mixed together to produce a mixture. For example, the two or more ingredients can be mixed by being stirred by hand or by being stirred by a mechanical stirrer, including magnetic stirrer; or can be mixed by a hand-operated mixer or by a machine-operated mixer; or can be mixed by being blended in a blender, tumbled in a tumbler, or shaken in a hand-operated shaker or by a machine-operated shaker; and so forth, such that a mixture of the two ingredients is produced.
[00270] As used herein, the terms "geometric blending" and "geometric mixing" refers to a sequence of steps where in the first step one ingredient (e.g., an active pharmaceutical ingredient) is mixed with an amount of a second ingredient (e.g., a diluent), and then in the second step more of the second ingredient (e.g. , a diluent) is added and mixed with mixture from the first step. Optionally, additional steps each comprising adding more of the second ingredient (e.g. , diluent) and mixing it with to the mixture of the first and second ingredients can be performed, where such additional steps can be performed up to 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 1 7, 1 8, 19, 20 or more times.
[00271 [ The term "low dose" as used herein in conjunction with Compound A tosylate salt, means, unless otherwise indicated, an amount of Compound A tosylate salt equal to or less than
2 mil ligrams. In some embodiments, "low dose" refers to a range of about 1 μg to about 8000 μg, about 1 μg to about 1500 μg, about 1 g to about 1000 μ§, about 1 μα to about 750 g, about 1 μg to about 500 μg, about 1 μg to about 250 μg, about 1 μg to about 150 g, about 1 μg to about 100 μg, about 1 μg to about 75 μg, about 5 g to about 1 500 g, about 5 μg to about 30 ug, about 10 ug to about 85
Figure imgf000095_0001
about 15 μg to about 85 μg, about 20 μg to about 1250 μ-g, about 20 μg to about 1 12 μg, about 20 μg to about 60 μ£, about 25 μ§ to about 1000 μg, about 25 μg to about 250 μg, about 20 μ§ to about 85 μg, about 25 g to about 85 μg, about 50 μ§ to about 100 μg or about 120 μ§ to about 250 g Compound A tosylate salt.
[00272] As used herein, "unit dose" means a single dose which is capable of being administered to a patient, and which can be readily handled and packaged, remaining as a physically and chemically stable unit dose, comprising a pharmaceutical composition as provided herein, for instance, having Compound A tosylate salt and one or more pharmaceutically acceptable excipients. [00273] In certain embodiments, the term "stable" used in reference to Compound A tosylate salt, including when in a pharmaceutical composition or unit dosage form, means no detectable loss of potency when stored for 18 months or more at, for example, 25 °C or ambient conditions or, for 4- 12 weeks or more at 40 °C and 75% RH. In some embodiments, the term "stable" used in reference to crystalline Compound A tosylate salt, including when in a pharmaceutical composition or unit dosage form, means no detectable change (e.g., polymorphic change and/or chemical change such as, for example, due to degradation), in its crystalline form when stored for 18 months or more at, for example, 25 *C or ambient conditions or, for 4- 12 weeks or more at 40 'C and 75% RH.
[00274] The term "amorphous" or "amorphous form" is intended to mean that the substance, component, or product in question is not substantially crystalline as determined, for instance, by XRPD or where the substance, component, or product in question, for example is not birefringent when viewed microscopically. In certain embodiments, a sample comprising an amorphous form of a substance may be substantially free of other amorphous forms and/or crystalline forms.
[00275] The term "crystalline form" or "crystal form" refers to a crystalline solid form of a chemical compound, including, but not limited to, a single-component or multiple-component crystal form, e.g., a polymorph of a compound; or a solvate, a hydrate, a clathrate, a cocrystal, a salt of a compound, or a polymorph thereof. The term "crystal forms" and related terms herein refers to the various crystalline modifications of a given substance, including, but not limited to, polymorphs, solvates, hydrates, co-crystals and other molecular complexes, as well as salts, solvates of salts, hydrates of salts, other molecular complexes of salts, and polymorphs thereof. Crystal forms of a substance can be obtained by a number of methods, as known in the art. Such methods include, but are not limited to, melt recrystal lization, melt cooling, solvent recrystallization, recrystallization in confined spaces such as, e.g., in nanopores or capillaries, recrystal lization on surfaces or templates such as, e.g., on polymers, recrystallization in the presence of additives, such as, e.g., co-crystal counter-molecules, desolvation, dehydration, rapid evaporation, rapid cooling, slow cooling, vapor diffusion, sublimation, grinding and solvent-drop grinding.
Certain Pharmaceutical Terminology
[00276] The term "acceptable" with 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.
[00277] As used herein, the term "selective binding compound" refers to a compound that selectively binds to any portion of one or more target proteins.
[00278] 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. [00279] 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).
[00280] 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 activity 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.
[00281] As used herein, the term "selective modulator" refers to a compound that selectively modulates a target activity.
[00282] As used herein, the term "PARP " refers to the family of the enzyme poly(ADP- ribose)polymerase which includes approximately 1 8 proteins, particularly poly(ADP-ribose)polymerase- l (PARP-1 ) and poly(ADP-ribose)polymerase-2 (PARP-2).
[00283] 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).
[00284] 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).
[00285] 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.
[00286] As used herein, the term "target activity" refers to a biological activity capable of being modulated by a selective modulator. Certain exemplary target activities include, but are not limited to, binding affinity, signal transduction, enzymatic activity, tumor growth, inflammation or inflammation- related processes, and amelioration of one or more symptoms associated with a disease or condition.
[00287] As used herein, the term "inhibitor" refers to a compound, the presence of which results in a decrease in the magnitude of a biological activity of a protein. In certain embodiments, the presence of an antagonist results in complete inhibition of a biological activity of a protein, such as, for example, the enzyme poly(ADP-ribose)poiymerase (PARP). [00288] As used herein, the IC50 refers to an amount, concentration or dosage of a particular test compound that achieves a 50% inhibition of a maximal response, such as modulation of PARP, in an assay that measures such response.
[00289] As used herein, EC50 refers to a dosage, concentration or amount of a particular test compound that elicits a dose-dependent response at 50% of maximal expression of a particular response that is induced, provoked or potentiated by the particular test compound.
[00290] 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).
[00291] 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.
[00292] 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.
[00293] 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.
[00294] 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 prevent the development of and/or 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. The term "therapeutically effective amount" also refers to the amount of a compound that is sufficient to elicit the biological or medical response being sought by a researcher, veterinarian, medical doctor, or clinician, for example, to inhibit PARP activity in vivo, to inhibit cancer cel l growth and/or proliferation, and/or to decrease cancer cell numbers.
[00295] 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 abil ity 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.
[00296] 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).
[00297] The terms "kit" and "article of manufacture" are used as synonyms.
[00298] 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.
[00299] 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 inhibit the activity of the target, to limit the activity of the target, or to extend the activity of the target.
[00300] As used herein, the term "pharmaceutically acceptable" refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem complications commensurate with a reasonable benefit/risk ratio. A "pharmaceutically acceptable excipient," as used herein, refers to a pharmaceutically acceptable material, composition, or vehicle, such as a liquid or solid filler, diluent, solvent, or encapsulating material. In one embodiment, each component is "pharmaceutically acceptable" in the sense of being compatible with the other ingredients of a pharmaceutical formulation, and suitable for use in contact with the tissue or organ of humans and an imals without excessive toxicity, irritation, allergic response, immunogenicity, or other problems or complications, commensurate with a reasonable benefit/risk ratio. See, e.g., Remington: The Science and Practice of Pharmacy, 21st ed.; Lippincott Williams & Wilkins: Philadelphia, PA, 2005; Handbook of Pharmaceutical Excipients, 6th ed. ; Rowe et ah, Eds.; The Pharmaceutical Press and the American Pharmaceutical Association: 2009; Handbook of Pharmaceutical Additives, 3rd ed. ; Ash and Ash Eds.; Gower Publishing Company: 2007; Pharmaceutical Preformttlation and Formulation, 2nd ed. ; Gibson Ed.; CRC Press LLC: Boca Raton, FL, 2009. As wi ll be understood by those skilled in the art, an excipient can, for example, be an antiadherent, a binder, a buffer, a carrier, a disintegrant, an emulsifying agent, a filler, a diluent, a flavoring agent, a coloring agent, a lubricant, a glidant, a preservative, an antioxidant, a sorbent and/or a sweetner. [00301] 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, pharmaceutical ly acceptable salts are obtained by reacting a compound described herein, with acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, methanesulfonic 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, N-methyl-D-glucamine, tris(hydroxymethyl)methylamine, and salts with amino acids such as arginine, lysine, and the like, or by other methods previously determined.
[00302J 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.
[00303J 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.
[00304] The term "'polymorph" or "polymorphic form" refers to one of two or more crystal forms that comprise the same molecule, molecules or ions. Different polymorphs may have different physical properties such as, for example, melting temperatures, heats of fusion, solubilities, dissolution rates, and/or vibrational spectra as a result of the arrangement or conformation of the molecules or ions in the crystal lattice. The differences in physical properties exhibited by polymorphs may affect pharmaceutical parameters, such as storage stability, compressibility, density (important in formulation and product manufacturing), and dissolution rate (an important factor in bioavailability). Differences in stability can result from changes in chemical reactiv ity (e.g., differential oxidation, such that a dosage form discolors more rapidly when comprised of one polymorph than when comprised of another polymorph), mechanical changes (e.g., tablets crumble on storage as a kinetica!ly favored polymorph converts to
thermodynarnically more stable polymorph), or both (e.g., tablets of one polymorph are more susceptible to breakdown at high humidity). As a result of solubi lity/dissolution differences, in the extreme case, some polymorphic transitions may result in lack of potency or, at the other extreme, toxicity. In addition, the physical properties of a crystalline form may be important in processing; for example, one polymorph might be more l ikely to form solvates or might be difficult to filter and wash free of impurities (e.g., particle shape and size distribution might be different between polymorphs).
[00305] 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 bioavaiiable 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 faci litate transmittal across a cell membrane where water solubility is detrimental to mobility but which then is metabolicaily hydrolyzed to the carboxylic 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.
[00306] As used herein and unless otherwise indicated, the term "stereomerically pure" means a composition that comprises one stereoisomer of a compound and is substantially free of other stereoisomers of that compound. In certain embodiments, stereomerically pure ( 8.S\9/?)-5-fluoro-8-(4- fluorophenyl)-9-( 1 -methyl- l H-l ,2,4-triazol-5-yl)-8,9-dihydro-2/ -pyrido[4,3,2-<:/e]phthalazin-3(7H)-one is provided herein that is substantially free of the other stereoisomers including, for example, (8R,9S)-5- fluoro-8-(4-fluorophenyI)-9-( l -methyl- l H- l ,2,4-triazol-5-yl)-8,9-dihydro-2H-pyrido[4,3,2-cfe]phthalazin- 3(7//)-one. In certain embodiments, a stereomerically pure compound comprises greater than about 80 percent by weight of one stereoisomer of the compound and less than about 20 percent by weight of other stereoisomers of the compound, greater than about 90 percent by weight of one stereoisomer of the compound and less than about 10 percent by weight of the other stereoisomers of the compound, greater than about 95 percent by weight of one stereoisomer of the compound and less than about 5 percent by weight of the other stereoisomers of the compound, greater than about 97 percent by weight of one stereoisomer of the compound and less than about 3 percent by weight of the other stereoisomers, or greater than about 99 percent by weight of one stereoisomer of the compound and less than about I percent by weight of the other stereoisomers of the compound. In certain embodiments, term "stereomerically pure" (8S,9R)-5-fluoro-8-(4-fluoropheny l)-9-( 1 -methyl- i H- 1 ,2,4-triazol-5-yl)-8,9- dihydro-2H-pyrido[4,3,2-i e]phthalazin-3(7H)-one means that the compound is made up of approximately 100% by weight of this particular stereoisomer. The above percentages are based on the total amount of combined stereoisomers of the compound.
[00307] As used herein, a crystalline or amorphous form that is "pure," i.e. , substantially free of other crystalline or amorphous forms, contains less than about 10 percent by weight of one or more other crystalline or amorphous form, less than about 5 percent by weight of one or more other crystalline or amorphous form, less than about 3 percent by weight of one or more other crystalline or amorphous form, less than about 1 percent by weight of one or more other crystalline or amorphous form, or less than about 0.5 percent by weight of one or more other crystalline or amorphous form. In certain contexts, as used herein, -'substantially pure" (85,9R)-5-fluoro-8-(4-fluorophenyl)-9-( I -methyl- 1/7- 1 , 2,4-triazol-5-yl)-8,9- dihydro-2//-pyrido[4.3.2-i/t']phthalazin-3(7//)-one or a salt or solvate thereof can mean free of other chemical compounds, for example, unreacted precursors and side products that might be present in process for preparing (8S,9/i)-5-fluoro-8-(4-fluorophenyl)-9-( l -methyl- 2,4-triazol-5-yl)-8,9- dihydro-2H-pyrido[4,3,2-i/e]phthalazin-3(7H)-one. In other contexts, as used herein, a "substantially pure" solid form {e.g., crystalline form or amorphous form) of (8S,9R)-5-fluoro-8-(4-fluorophenyl)-9-( t - methyl- \ H- 1 ,2,4-triazol-5-yl)-8,9-dihydro-2H-pyrido[4,3,2-i t?]phthalazin-3(7H)-one or a salt or solvate thereof can mean free of other solid forms of (85,9R)-5-fluoro-8-(4-fluorophenyl)-9-( l -methyl- \H- 1 ,2,4- triazol-5-yl)-8,9-dihydro-2//-pyrido[4,3,2-(ie]phthalazin-3(7H)-one or salts or solvates thereof. As such, "substantially pure" (8S.9^)-5-fluoro-8-(4-fluorophenyl)-9-( l -methyl- l H- 1 , 2,4-triazol-5-yl)-8,9-dihydro- 2H-pyrido[4,3,2-c/e]phthalazin-3(7//)-one may comprise, in certain embodiments, less than about 10%, 5%, 3%, 2%, 1 %, 0.75%, 0.5%, 0.25%, or 0.1% by weight of one or more other crystal forms and amorphous forms of the compound and/or other chemical compounds. In certain embodiments, a solid form that is substantially pure is substantially free of one or more other particular crystal forms, amorphous forms, and/or other chemical compounds.
[00308J 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 l ike. In one embodiment of the methods and compositions provided herein, the mammal is a human.
[00309} 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 cond ition, 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.
[00310] Alternatively the terms "treat," "treating," and "treatment" are meant to include alleviating or abrogating a disorder, disease, or condition, or one or more of the symptoms associated with the disorder, disease, or condition; or to slowing the progression, spread or worsening of a disease, disorder or condition or of one or more symptoms thereof. Often, the beneficial effects that a subject derives from a therapeutic agent do not result in a complete cure of the disease, disorder or condition.
[00311] Alternatively, 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, amel iorate, lessen, or cure a disease or disorder and/or its symptoms.
[00312] 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.
[00313] 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 lUPAC 1 974 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 synthetical ly 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 chiral auxiliary or (2) direct separation of the mixture of optical enantiomers on chiral chromatographic column.
[00314] The methods and formulations described herein include the use of JV-oxides, cry stal line 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. AH 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.
[00315] Throughout the specification, groups and substituents thereof are chosen, in certain embodiments, to provide stable moieties and compounds. Pharmaceutical Composition/F ormulation
(00316] In certain embodiments, pharmaceutical compositions are formulated in any manner, including using one or more physiologically acceptable carriers comprising excipients and/or auxiliaries which facilitate processing of the active compounds into pharmaceutical preparations. In some embodiments, proper formulation is dependent upon the route of administration chosen. In various embodiments, any techniques, carriers, and excipients are used as suitable.
[00317] Provided herein are pharmaceutical compositions that include a compound described herein and one or more 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. (00318] 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.
[00319] 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.
[00320] 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 other excipients. Such carriers enable the compounds described herein to be formulated as tablets, powders, pi lls, dragees, capsules, liquids, gels, syrups, elixirs, slurries, suspensions and the like, for oral ingestion by a patient to be treated. [003211 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 tragaeanth, methylcellulose, microcrystalline cellulose, hydroxypropy Imethylcel lulose, 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.
[00322] 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.
[00323] 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, optional ly, 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, stabil izers are optional ly added. In certain embodiments, the formulations for oral administration are in dosages suitable for such administration.
[00324] In certain embodiments, the pharmaceutical compositions provided herein are formu lated 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 oi ly 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 l iposomes. 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.
[00325J 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.
[00326] 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), (IA) 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 del iver the compound to the skin of the host at a control led and predetermined rate over a prolonged period of time, and means to secure the device to the skin.
[00327] In certain embodiments, the pharmaceutical compositions provided herein are formulated for admin istration 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 propel lan t, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In certain aspects of a pressurized aerosol, the dosage unit is determined by prov iding a valve to del iver 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. [00328J 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.
[00329] 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.
[00330] 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 jV-oxides, cr stalline forms (also known as polymorphs), as wel l 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 regu lating the osmotic pressure, and/or buffers. In additional embodiments, the pharmaceutical compositions described herein also contain other therapeutically valuable substances.
[00331] 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. Semi-solid 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.
[00332] 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.
[00333J 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 methylcellulo.se. 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.
[00334] 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.
[00335] 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.
[00336] 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.
[00337] Certain useful compositions optionally include one or more preservatives to inhibit microbial activity. Suitable preservatives include mercury-containing substances such as me r fen and thiomersal; stabilized chlorine dioxide; and quaternary ammon ium compounds such as benzalkonium chloride, cetyltrimethylammonium bromide and cetylpyridinium chloride.
[00338] Some usefu l 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.
[00339] 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.
[00340] 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.
[00341] 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 iV-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.
[00342] 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 stabi lizing 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, (I) pentosan polysulfate and other heparinoids, (m) divalent cations such as magnesium and zinc; or (n) combinations thereof.
Methods of Dosing and Treatment Regimens
[00343] 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 activ e metabol ite,
pharmaceutically acceptable prodrug, or pharmaceutical ly acceptable solvate thereof, in therapeutical ly effective amounts to said subject.
[00344] 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).
[00345] 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 administered is defined to be a "prophyiactically 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 prophyiactically 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.
(00346] In certain instances, a patient's condition does not improve or does not significantly improve fol lowing administration of a compound or composition described herein and, upon the doctor's d iscretion the adm inistration 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.
[00347] In certain cases wherein the patient's status does improve or does not substantially improve, upon the doctor's discretion the 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, 150 days, 1 80 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%.
[00348] 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 admin istration, 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.
[00349J 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.
100350) 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 reclosable 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.
[00351] 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.
[00352] Un it dosage forms provided herein comprise Compound A in a solid tosylate salt form.
Unless the context dictates otherwise, unit dosage forms referred to herein, for instance, "25 μg capsules," "50 ng capsules," "250 μ§ capsules," 'Ί mg capsules." and the like, are intended to mean the amount of Compound A in the unit dosage form, not taking into account the mass contributed by the tosylate portion of the salt. For example, a "25 μg capsules" will be understood to mean, unless indicated otherwise in the context in which it is used, that the capsule contains about 36.325 fig Compound A tosylate salt. A conversion factor of 1.453 mg Compound A tosylate salt per 1.000 mg Compound A can be used to convert the amount of Compound A indicated in a given unit dosage form to the amount of Compound A tosylate salt contained in the unit dosage form.
[00353] In another example amounts of (85,9/i)-5-fliioro-8-(4-fIuorophenyl)-9-( l -methyl- lf/- l ,2,4-triazol-5-yl)-8,9-dihydro-2//-pyrido[4,3,2-de]phthalazin-3(7//)-one tosylate salt in unit dosage forms can, for example, be from about 5 g to about 1500 μg, from about 20 g to about 1250 μ§, from about 25 μg to about 1000 μg, or from about 25 μg to about 250 g. In some embodiments, the amount of (8-V,9/0-5-fluoro-8-(4-nuorophen^
de]phthalazin-3(7H)-one tosylate salt in a unit dosage form comprises (where the following do not including mass contributed by the tosylate portion of the salt) about 5 μg to about 30 μg, from about 20 μ to about 60 μg, from about 50 μg to about 100 μg, from about 120 μg to about 250 μ§, from about 20 μg to about 1 12 μg, or from about 25 g to about 250 ug (8£,9i?)-5-fluoro-8-(4-fluorophenyI)-9-( 1 -methyl- l / - l ,2,4-triazol-5-yl)-8,9-dihydro-2//-pyrido[4,3,2-de]phthalazin-3(7 )-one. In certain embodiments, the amount of (8S,9i?)-5-fluoro-8-(4-fIuoiOphenyl)-9-( l -m^^
pyrido[4,3,2-de]phthalazin-3(7 /)-one is about 10 μg, about 20 g, about 25 μg, about 30 μ , about 50 μg, about 75 μg, about 100 μ¾ about 150 g, about 200 μg, about 250 μg, or about 1000 μg.
[00354] Oral Dosage Forms. In certain embodiment, the pharmaceutical composition, as provided herein, is formulated for oral administration to a subject. Pharmaceutical compositions suitable for oral administration can be presented as discrete dosage forms, such as, but are not limited to, tablets, chewable tablets, caplets, capsules, and liquids (e.g. , flavored syrups). Such dosage forms contain predetermined amounts of active ingredients, and may be prepared by methods of pharmacy wel l known to those skilled in the art. See generally, Remingto 's Pharmaceutical Sciences, 1 8th ed., Mack Publishing, Easton PA ( 1990).
[00355] For instance, oral dosage forms provided herein may be prepared by combining (8.9,9/?)-
5-fluoro-8-(4-fluorophenyl)-9-( l -methyl- 1//- 1 , 2,4-triazol-5-yl)-8,9-dihydro-2H-pyrido[4,3,2- de] phtha lazin-3 ( 7i/)-one tosylate salt, in one embodiment in a crystalline form thereof as provided herein (hereinafter, the "active ingredient"), an intimate admixture with at least one excipient according to conventional pharmaceutical compounding techniques. Excipients can take a wide variety of forms depending on the form of preparation desired for administration. Because of their ease of administration, tablets and capsules represent the most advantageous oral dosage unit forms, in which case solid excipients are employed. If desired, tablets can be coated by standard aqueous or nonaqueous techniques. Such dosage forms can be prepared by any of the methods of pharmacy. In general, pharmaceutical compositions and dosage forms are prepared by uniformly and intimately admixing the active ingredient with l iquid carriers, finely divided solid carriers, or both, and then shaping the product into the desired presentation if necessary. Disintegrants or lubricants can be used in pharmaceutical compositions and dosage forms of the invention. Production of pharmaceutical compositions or dosage forms in accordance with the present invention may require, in addition to the therapeutic drug ingredients, exeipients or additives including, but not limited to, diluents, binders, lubricants, disintegrants, colorants, flavors, sweetening agents and the like or mixtures thereof. By the incorporation of these and other additives, a variety of dosage forms (e.g., tablets, capsules, caplets, troches and the like) may be made. These include, for example, hard gelatin capsules, caplets, sugar-coated tablets, enteric-coated tablets (for example to delay action), multiple compressed tablets, prolonged-action tablets, tablets for solution, effervescent tablets, buccal and sublingual tablets, troches and the like. The dose form or dosage formulation may be formed by methods well known in the art. See, e.g. , Remington 's Pharmaceutical Sciences, 16th and 18th eds., Mack Publishing Co., Easton, Pa. ( 1980 and 1990). See also U.S. Pharmacopeia XXI, U.S.
Pharmacopeia Convention, Inc., Rockville, Md. ( 1985).
[00356] Other Dosage Forms. In certain embodiments, the pharmaceucal composition as provided herein can be formulated in parenteral dosage forms. Parenteral dosage forms can be administered to patients by various routes including, but not l imited to, subcutaneous, intravenous (including bolus injection), intramuscular, and intraarterial. Because their administration typically bypasses patients' natural defenses against contaminants, parenteral dosage forms are in some embodiments sterile or capable of being sterilized prior to administration to a patient. Examples of parenteral dosage forms include, but are not limited to, solutions ready for injection, dry products ready to be dissolved or suspended in a pharmaceutically acceptable vehicle for injection, suspensions ready for injection, and emulsions. Suitable vehicles that can be used to provide parenteral dosage forms provided herein are well known to those ski lled in the art.
[00357] In yet other embodiments, the pharmaceutical composition as provided herein can be formulated in a transdermal, topical or mucosal dosage form. Transdermal, topical, and mucosal dosage forms provided herein include, but are not limited to, ophthalmic solutions, sprays, aerosols, creams, lotions, ointments, gels, solutions, emulsions, suspensions, or other forms known to one of skill in the art.
th ih
See, e.g., Remington 's Pharmaceutical Sciences, 16 and 18 eds., Mack Publishing, Easton PA ( 1980 &
ill
1990); and Introduction to Pharmaceutical Dosage Forms, 4 ed., Lea & Febiger, Philadelphia ( 1985). Suitable exeipients (e.g., carriers and diluents) and other materials that can be used to provide transdermal, topical, and mucosal dosage forms encompassed by this disclosure are wel l known to those skilled in the pharmaceutical arts, and depend on the particular tissue to which a given pharmaceutical composition or dosage form will be appl ied.
[00358] 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 LD$0 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. Kits/Articles of Manufacture
[00359] For use in the therapeutic applications described herein, kits and articles of manufacture are also described herein. In various embodiments, such kits comprise a carrier, package, or container that is compartmentalized to receive one or more containers such as vials, tubes, and the like, each of the container(s) comprising one of the separate elements to be used in a method described herein. Suitable containers include, for example, bottles, vials, syringes, and test tubes. In some embodiments, the containers are formed from a variety of materials such as glass or plastic.
|00360] In some embodiments, the articles of manufacture provided herein contain packaging materials. Packaging materials for use in packaging pharmaceutical products include, but are not limited to, blister packs, bottles, tubes, inhalers, pumps, bags, vials, containers, syringes, bottles, and any packaging material suitable for a selected formulation and intended mode of administration and treatment.
[00361] In some embodiments, the container(s) described herein comprise one or more compounds described herein, optionally in a composition or in combination with another agent as disclosed herein. The containers) optionally have a sterile access port (for example in some embodiments the container is an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle). Such kits optionally comprise a compound with an identifying description or label or instructions relating to its use in the methods described herein.
[00362] In some embodiments, a kit will comprises one or more additional containers, each with one or more of various materials (such as reagents, optionally in concentrated form, and/or devices) desirable from a commercial and user standpoint for use of a compound described herein. Non-limiting examples of such materials include, but are not l imited to, buffers, diluents, filters, needles, syringes; carrier, package, container, vial and/or tube labels listing contents and/or instructions for use, and package inserts with instructions for use. A set of instructions is optionally included.
[00363] In certain embodiments, a label is on or associated with the container. In some embodiments, a label is on a container when letters, numbers or other characters form ing the label are attached, molded or etched into the container itself; a label is associated with a container when it is present within a receptacle or carrier that also holds the container, e.g., as a package insert. In certain embodiments, a label indicates that the contents are to be used for a specific therapeutic application. In some embodiments, the label indicates directions for use of the contents, such as in the methods described herein.
[00364] In certain embodiments, the pharmaceutical compositions are presented in a pack or dispenser device wh ich contains one or more unit dosage forms containing a compound provided herein. In some embodiments, the pack contains a metal or plastic foil, such as a bl ister pack. The pack or dispenser device is optionally accompanied by instructions for administration. In some embodiments, the pack or dispenser is accompanied with a notice associated with the container in form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals, which notice is reflective of approval by the agency of the form of the drug for human or veterinary administration. In certain embodiments, such notice is, for example, the labeling approved by the U.S. Food and Drug Administration for prescription drugs, or the approved product insert. In some embodiments, compositions containing a compound provided herein are formulated in a compatible pharmaceutical carrier and are placed in an appropriate container labeled for treatment of an indicated condition.
Preparation of Compounds described herein
[00365] 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 util ized. In particular, the synthetic methods described in WO 2010/017055, WO/201 1/097602, and WO2012054698 can be used to prepare a compound used in the methods disclosed herein, each of which is incorporated herein by reference in its entirety.
[00366] Techniques for characterizing crystal forms and amorphous forms include, but are not limited to, TGA, DSC, XRPD, single crystal X-ray diffractometry, vibrational spectroscopy, e.g., IR and Raman spectroscopy, solid-state NMR, optical microscopy, hot stage optical microscopy, SEM, electron crystallography and quantitative analysis, PSA, surface area analysis, solubility studies and dissolution studies.
EXAMPLES
[00367] The following Examples are intended as an illustration of the various embodiments as defined in the appended claims. In some embodiments, the compounds are prepared by a variety of synthetic routes. All publications, patents, and patent applications cited herein are hereby incorporated by reference for all purposes.
EXAMPLE 1
Example l a: Parenteral Composition
[00368] To prepare a parenteral pharmaceutical composition suitable for administration by injection, 100 mg of a water-soluble salt of a compound described herein is dissolved in DM SO and then mixed with 10 mL of 0.9% sterile saline. The mixture is incorporated into a dosage unit form suitable for administration by injection.
Example l b: Oral Composition
[00369] To prepare a pharmaceutical composition for oral delivery, 100 mg of a compound described herein is mixed with 750 mg of starch. The mixture is incorporated into an oral dosage unit for, such as a hard gelatin capsule, which is suitable for oral administration.
Example l c: Sublingual (Hard Lozenge) Composition [00370] To prepare a pharmaceutical composition for buccal delivery, such as a hard lozenge, mix 100 mg of a compound described herein, with 420 mg of powdered sugar mixed, with 1 ,6 mL of light corn syrup, 2.4 mL distilled water, and 0.42 mL mint extract. The mixture is gently blended and poured into a mold to form a lozenge suitable for buccal administration.
Example I d: Inhalation Composition
[00371] To prepare a pharmaceutical composition for inhalation delivery, 20 mg of a compound described herein is mixed with 50 mg of anhydrous citric acid and 100 mL of 0.9% sodium chloride solution. The mixture is incorporated into an inhalation delivery unit, such as a nebulizer, which is suitable for inhalation administration.
Example l e: Rectal Gel Composition
[00372] To prepare a pharmaceutical composition for rectal delivery, 100 mg of a compound described herein is mixed with 2.5 g of methylcel luose ( 1500 mPa), 100 mg of methylparapen, 5 g of glycerin and 100 mL of purified water. The resulting gel mixture is then incorporated into rectal delivery units, such as syringes, which are suitable for rectal administration.
Example I f: Topical Gel Composition
[00373] To prepare a pharmaceutical topical gel composition, 100 mg of a compound described herein is mixed with 1 .75 g of hydroxypropy I cellulose, 10 mL of propylene glycol, 10 mL of isoprop l myristate and 100 mL of purified alcohol USP. The resu lting gel mixture is then incorporated into containers, such as tubes, which are suitable for topical administration.
Example l g: Ophthalmic Solution Composition
[00374] To prepare a pharmaceutical ophthalm ic solution composition, 100 mg of a compound described herein is mixed with 0.9 g of NaCl in 100 mL of purified water and filtered using a 0.2 micron niter. The resulting isotonic solution is then incorporated into ophthalm ic delivery units, such as eye drop containers, which are suitable for ophthalmic administration.
Example 2
5-fluoro-8-(4-fluorophenyl)-9-( l -methyl- l ^
de]phthalazin-3(7H)-one
Example 2A
Ethyl 7-fluoro-2-(4-fluorophenyl)-3-( l -methyl- 1 H- L2,4-triazol-5-yl)-4-oxo-l , 2,3 ,4-tetrahydroquinoIine-
5-carboxylate
[00375] To a solution of (£)-6-fluoro-4-(4-fluorobenzylideneamino)isobenzofuran- l (3 /)-one (4 g, 14.6 mmol) and 1 -methyl- 1 H- 1 ,2,4-triazole-5-carbaldehyde (4. 1 g, 36.9 mmol) in ethyl propionate (220 mL) was added EtONa ((sodium 940 mg, 40.9 mmol), in 70 mL ethano l) at 37 °C, then the mixture was stirred at 40 °C for 6 hr. The resulting mixture was evaporated under reduced pressure and extracted with ethyl acetate ( 100 mL χ 4).The extract was concentrated to dryness to give a crude product, which was purified by column chromatography (silica gel, dich!oromethane: methanol = 200: 1 to 100: 1 ) to obtain a green solid ( 1 .02 g, yield 14%). LC-MS (ESI) m/z: 413 (M+ l f. Example 2B
5-Fluoro-8-(4-fluorophenyl)-9-( 1 -methyl- 1 H- 1 ,2,4-triazoI-5-yl)-8,9-dihydro-2H-pyrido[4,3,2- de]phthalazin-3( 7H)-one
[00376] To a solution of ethyl 7-fluoro-2-(4-fluorophenyI)-3-( l-methyl- l H-l ,2,4-triazol-5-yl)-4- oxo- 1 ,2,3,4-tetrahydroquinoline-5-carboxyIate (840 mg, 2.04 mmol) in methanol (2 mL) was added hydrazine monohydrate ( 1 mL), and the m ixture was stirred under 25 °C for 10 hr. Then the mixture was filtered to obtain a white solid (650 mg, yield 84%). LC-MS (ESI) m/z: 381 (M+ l )+. Ή-NMR (400 MHz, DMSC ,) δ (ppm): 3.66 (s, 3H), 4.97-5.04 (m, 2H), 6.91 -6.94 (dd, J\ = 1 1 .2 Hz, J2 = 2.4 Hz, 1 H), 7.06- 7.09 (dd, ·/,= 8.8 Hz, ./2 = 2.4 Hz, 1 H), 7. 14-7.18 (m, 3H), 7.47-7.5 1 (m, 2H), 7.72 (s, 1 H), 7.80 (s, 1 H), 12.35 (s, 1 H)
Examples on Chiral Resolution
Example 3
Enantiomers of (8i?,95 -5-fluoro-8-(4-fluorophenyl)-9-( l-methyl- lH- l ,2,4-triazol-5-yl)-8,9-dihydro-2//- pyrido[4,3 ,2-i/e] phthalazin-3 (7 )-one and (8 9i?)-5-fluoro-8-(4-fluorophenyl)-9-( 1 -methyl- 1 //- 1 ,2,4- triazol-5-yl)-8,9-dihydro-2H-pyrido[4,3,2-ife]phthalazin-3(7 )-one
[003771 5-Fluoro-8-(4-fluorophenyl)-9-( l -methyl- l i- l,2,4-triazol-5-yl)-8,9-dihydro-2//- pyrido[4,3,2-de]phthalazin-3(7H)-one was dissolved in DMF and chiral resolution was performed using super-fluid chromatography (SFC) with IA chiral column and methanol (20%) and CO? (80%) as the eluents.
Example 4
(Z)-6-Fluoro-3-(( 1 -methyl- 1 / - 1 ,2,4-triazol-5 -y I )methy lene)-4-n itroisobenzofuran- 1 (3 )-one (3)
Figure imgf000117_0001
[00378] To a 80 L jacketed glass reactor equipped with a chil ler, mechanical stirrer,
thermocouple, and nitrogen inlet/outlet, at 15 - 25 °C, anhydrous 2-methyl-tetrahydrofuran (22.7 kg), 6- fluoro-4-nitroisobenzofuran- l (3 )-one (2) (2.4 kg, 12.2 mol, 1.00 eq.), and 2-methyl-2H- l ,2,4-triazole-3- carbaldehyde (49.6 - 52.6 % concentration in dichloromethane by GC, 3.59 - 3.38 kg, 16.0 mol, 1.3 1 eq.) were charged consecutively. Triethylamine ( 1.50 kg, 14.8 mol, 1 ,21 eq.) was then charged into the above reaction mixture. The reaction mixture was stirred for another 10 minutes. Acetic anhydride (9.09 - 9.10 kg, 89.0 - 89.1 mol, 7.30 eq.) was charged into the above reaction mixture at room temperature for 20 -
30 minutes. The reaction mixture was heated from ambient to reflux temperatures (85 - 95 °C) for 80 - 90 minutes, and the mixture was refluxed for another 70 - 90 minutes. The reaction mixture was monitored by HPLC, indicating compound (2) was reduced to < 5 %. The resulting slurry was cooled down to 5 -
15 °C for 150 - 250 minutes. The slurry was aged at 5 - 1 5 °C for another 80 - 90 minutes. The slurry was filtered, and the wet cake was washed with ethyl acetate (2L χ 3). The wet cake was dried under vacuum at 40 - 50 °C for 8 hours to give 2.65 - 2.76 kg of (Z)-6-fluoro-3-(( l -methyl- lH-1 , 2,4-triazol-3- yl)methylene)-4-nitroisobenzofuran- l (3H)-one (3) as a yellow solid (2.66 kg, yield: 75.3 %, purity: 98.6 - 98.8 % by HPLC). LC-MS (ESI) m z: 291 (M+ l )+. Ή-NMR (400 MHz, DMSO-d6) δ (ppm): 3.94 (s, 3H), 7. 15 (s, 1 H), 8. 10 (s, 1 H), 8.40-8.42 (dd, J, = 6.4 Hz, J2 = 2.4 Hz, 1 H), 8.58-8.61 (dd. .7, - 8.8 Hz, ./, = 2.4 Hz, 1 H).
Example 5
Methy 1 5 -fluoro-2-(2-( 1 - methy 1- 1 H- 1 ,2,4-triazol-5 -y l)acety l)-3 -n itrobenzoate (4)
Figure imgf000118_0001
Example 5A
[00379] (Z)-6-Fluoro-3-(( 1 -methyl- 1 H- 1.2.4-triazol-3 -yl)methylene)-4-nitroisobenzofuran- 1 (3//)- one (3) ( 177 g, 0.6 mol, 1.0 eq.), and HC1 (2 N in methanol, 3 L, 6 mol, 10 eq.) were charged into a 5 L 3-neck flask equipped with mechanical stirrer, thermometer, and nitrogen inlet outiet. The reaction mixture was stirred at room temperature for 25 hours. The reaction mixture was monitored by HPLC, indicating 0.8 % compound (3) remained. The reaction mixture was concentrated under vacuum at 40 °C to dryness, and methyl 5-fluoro-2-(2-( l -methyl- lH- l ,2,4-triazole-3-yl)acetyl)-3-nitrobenzoate hydrochloride (4) was obtained as a yel low solid (201 g, yield: 93.4 %). It was used for the next step without further purification. LC-MS (ESI) m/z: 323 (M+ l )+ ' H-NMR (400 MHz, DMSC s) δ (ppm): 3.89 (s, 3H), 3.92 (s, 3 H), 4.60 (s, 2H), 7.85 (s, 1 H), 8.25-8.28 (dd, ./, = 8.4 Hz, J2 = 2.8 Hz, 2H), 8.52- 8.54 (dd, J, ·= 8.4 Hz. = 2.8 Hz, 2H).
Example 5B
[00380] An alternative workup procedure to that illustrated in Example 161 A follows. Instead of evaporating the reaction mixture to dryness, it was condensed to 2 volumes, followed by solvent exchange with 12 volumes of THF, and then 12 volumes of heptane. The slurry mixture was concentrated to 2 volumes and filtered to give the product. As such, 1.8 kilograms of (Z)-6-fluoro-3-(( l -methyl- lH- l,2.4- triazol-3-yl)methylene)-4-nitroisobenzofuran- l (3 )-one (3) gave 2.15 kilograms (yield 96.4 %) of the product methyl 5-fluoro-2-(2-( l -methyl- 1/ - l ,2,4-triazole-3-yl)acetyl)-3-nitrobenzoate hydrochloride (4).
Example 6
Methyl 7-tluoro-2-(4-fluorophenyl)-3-( l -methyl- l H- 1 , 2,4-triazol-5-yl)-4-oxo- 1 ,2,3 ,4- tetrahydroquinoline-5-carboxylate (5)
Figure imgf000119_0001
Example 6A
[00381] To a suspension of methyl 5-fluoro-2-(2-( 1 -methy I- 1 H- 1 ,2,4-triazo l-5-y l)acety I )-3- nitrobenzoate (4) (5 g, 15 ,5 mmol, leq.) and 4-fluorobenzaldehyde (3.6 g, 29 mmol, 1 .87 eq.) in a mixture of solvents tetrahydrofuran (30 mL) and MeOH (5 mL) was added titanium(lll) chloride (20 % w/w solution in 2N Hydrochloric acid) (80 mL, 6 eq.) dropwise with stirring at room temperature. The reaction mixture was allowed to stir at 3O~50°C for 2 hours. The mixture was then diluted with water ( 160 mL), and the resulting solution was extracted with ethyl acetate ( 100 mL x 4). The combined organic layers were washed with saturated NaHC(¾ (50 mL x 3) and aqueous NaHS(¾ ( 100 mL x 3), dried by Na2S04, and concentrated to dryness. This afforded a crude solid, which was washed with petroleum ether ( 120 mL) to obtain the title compound as a yellow solid (5.9 g, yield: 95 %, purity: 97 %). LC-MS (ESI) m/z: 399 (M+ l )\ Ή-NMR (400 MHz, CDCh) 5 (ppm): 3.58 (s, 31 1). 3.87 (s, 3H), 4.16-4. 19 (d, .12=1 .2 Hz, 1 H), 4.88 (s. 1 H), 5.37-5.40 (d, J2=13.2 Hz, 1 H), 6.47-6.53 (m, 2H) , 6.97-7.01 (m, 2H), 7.37-7.41 (m, 2H), 7.80 (s, 1 H).
Example 6B
[00382] An alternative workup procedure to that illustrated in Example 162A follows. After the completion of the reaction, the mixture was extracted with isopropyl acetate (20 volumes x 4) without water dilution. The product was isolated by solvent exchange of isopropyl acetate with heptanes followed by re-slurry with TBE and filtration. As such, 3 kilograms of methyl 5-fluoro-2-(2-( l -methyl- l H- l ,2,4- triazol-5-yl)acetyl)-3-nitrobenzoate (4) afforded 2.822 kilograms of the title compound (5) (yield 81 %).
Example 6C
[00383] To a stirred solution of methyl 5-fluoro-2-(2-( 1 -methyl- 1 H- 1 ,2,4-triazol-5-yl)acetyl)-
3-nitrobenzoate (4) (580 mg, 2 mmol) and 4-fluorobenzaldehyde (488 mg. 4 mmol) in methanol (0.75 mL) and tetrahydrofuran (4.5 mL) was added concentrated HC 1 solution (w/w 37 %, 6 mL), then reductive powdered Fe (672 mg, 12 mmol) was added slowly to the reaction system. After the addition was complete, the resulting mixture was heated to 60 °C and kept at this temperature for 3 hours. After the disappearance of the starting material (4) as monitored by LC-MS, the reaction mixture was partitioned between ethyl acetate (30 mL) and water (30 mL) and the aqueous phase was extracted with ethyl acetate (20 mL x 3). The combined organic phase was dried with Na2S0 , concentrated in vacuo and purified by column chromatography (ethyl acetate: petroleum ether = 1 : 1 ) to give the title compound (5) as a pale yellow solid (300 mg, yield 40 %). LC-MS (ESI) m/z: 399 (M+ l )'. Ή-NMR (400 MHz, CDC1,)5 (ppm): 3.58 (s, 3H), 3.87 (s, 3H), 4.1 7 (d, 1 H), 4.87 (s, 1 H), 5.38 (d, 1 H), 6.50 (dd, 2H), 6.99 (dd, 2H), 7.38 (dd, 2H), 7.80 (s, 1 H). Example 6D
[00384] To a stirred solution of methyl 5-fluoro-2-(2-( 1 -methyl- 1 H- l , 2,4-triazol-5-yl)acetyl)-3- nitrobenzoate (4) (580 mg, 2 mmol) and 4-fluorobenzaIdehyde (488 mg, 4 mmol) in methanol (0.75 mL) and tetrahydrofuran (4.5 mL) was added SnC (2.28 g, 12 mmol) and concentrated HC1 (w/w 37 %, 6 mL), the resulting mixture was reacted at 45 °C for 3 hours, until LC-MS indicating the disappearance of the starting material (4) and about 50 % formation of the product. The mixture was then partitioned between ethyl acetate (30 mL) and water (30 mL) and the aqueous phase was extracted with ethyl acetate (20 mL x 3). The combined organic phase was dried with Na2SO,, concentrated in vacuo and purified by column chromatography (ethyl acetate: petroleum ether = 1 : 1 ) to give the title compound (5) as a pale yellow solid ( 10 mg, yield 1.3 %). LC-MS (ESI) m/z: 399 (M+l f . Ή-NMR (400 MHz, CDC13)5 (ppm): 3.58 (s, 3H), 3.87 (s, 3H), 4.17 (d, I H), 4.87 (s, I H), 5.38 (d, I H), 6.50 (dd, 2H), 6.99 (dd, 2H), 7.38 (dd, 2H), 7.80 (s, I H).
Example 6E
[00385] A solution of methyl 5-ttuoro-2-(2-( l -methyl- l / -l,2,4-triazol-5-yl)acetyl)-3- nitrobenzoate (4) (580 mg, 2 mmol) and 4-fluorobenzaldehyde (488 mg, 4 mmol) in methanol (20 mL) and acetic acid ( 1 mL) was stirred at room temperature for 24 hours under hydrogen ( 1 barr) in the presence of a catalytic amount of 10 % Pd/C (212 mg, 0.2 mmol). After the reaction was complete, the catalyst was removed by filtration through a pad of Celite, the solvent was removed in vacuo, and the residue was purified by column chromatography (ethyl acetate: petroleum ether = 1 : 1 ) to give the title compound (5) as a pale yellow solid (63 mg, yield 8 %). LC-MS (ESI) m/z: 399 (M+l)+ . ' l lNMR (400 MHz, DMSO-d6)6 (ppm): 3.56 (s, 3H), 3.86 (s, 3H), 7.02 (dd, 2H), 7.21 (dd, 2H), 7.90 (s, I H), 8.08 (s, 1 H), 8.26 (dd, 1 H), 8.56 (dd, 1 H).
Example 7
5-Fluoro-8-(4-fluorophenyI)-9-( l -methyl-l - l ,2,4-triazol-5-yl)-8,9-dihydro-2//-pyrido[4,3,2-
Figure imgf000120_0001
[00386] Methyl 7-fluoro-2-(4-fluorophenyl 3-( l -methyl- l H-l ,2.4-triazol-5-yl)-4-oxo- l ,2,3,4- tetrahydroquinoline-5-carboxylate (5) ( 150 g, 0.38 mol, 1.0 eq.) and methanol ( 1.7 L) were charged into a 3 L 3 -neck flask equipped with a mechanical stirrer, thermometer, and nitrogen inlet/outlet. The resulted suspension was stirred at room temperature for 15 minutes. Hydrazine hydrate (85 % of purity, 78.1 g, 1 .33 moi, 3.5 eq.) was charged dropwise into the above reaction mixture within 30 minutes at ambient temperature. The reaction mixture was stirred at room temperature overnight. The react ion was monitored by HPLC, showing about 2 % of compound (5) left. The obtained slurry was fi ltered. The wet cake was suspended in methanol (2 L) and stirred at room temperature for 3 hours. The above slurry was filtered, and the wet cake was washed with methanol (0.5 L). The wet cake was then dried in vacuum at 45 - 55 °C for 12 hours. This afforded the title compound as a pale yellow solid ( 1 12 g. yield: 78. 1 %, purity: 95.98 % by HPLC). LC-MS (ESI) m/z: 38 1 (M+ l )+. ' H-NMR (400 MHZ, DMSO-<¾) δ (ppm): 3 .66 (s, 3H), 4.97-5.04 (m, 21 1). 6. 1 -6.94 (dd, J, = 2.4, J2 = 1 1 .2 Hz, 1 H), 7.06-7.09 (dd. J, = 2.4, J2 = 8.8 Hz, 1 H), 7. 14-7. 1 8 (m, 3 H), 7.47-7.5 1 (m, 2H), 7.72 (s, 1 H), 7.80 (s, 1 H), 12.35 (s, 1 H).
Example 8
5-Amino-7-fluoro-4-(( 1 -methyl- 1 H- 1 ,2,4-triazol-5-yl)methyl)phthalazin- 1 (2H)-one
Figure imgf000121_0001
[00387] To a sol ution of 6-fluoro-3-(( 1 -methyl- 1 H- 1 ,2,4-triazol-3-yl)methylene)-4-nitroiso- benzofuran- 1 (3//)-one (3) (4.0 g, 135 mmol) in THF ( 100 mL) was added hydrazine monohydrate (85 %) (6 mL) at room temperature under nitrogen atmosphere. The mixture was stirred for 2 hours, then acetic acid (6 mL) was added and the mixture was heated to and kept at 60 °C for 18 hours. The resulting mixture was di luted with water ( 100 mL) and extracted with ethyl acetate ( 100 mL x 3). The organic layer was dried over anhydrous Na2S04 and evaporated to dryness to afford the title compound as a yellow solid ( 1.6 g, yield 42 %). LC-MS (ESI) m/z: 275(M+ 1 )+.
Exam le 9
(£)-7-fluoro.5-(4-fluorobenzylideneamino)-4-(( l -methyl- 1 H- l ,2,4-triazol-5-yl)methyl)phthalazin- l (2J¥)- one
Figure imgf000121_0002
(8)
[00388J To a suspended of 5-amino-7-fluoro-4-(( 1 -methyl- 1 H- 1 ,2,4-triazol-3-yl)methy 1) phthalazin- 1 (2H)-om (7) ( 1 .6 g, 5.8 mmol) in acetonitrile (50 mL) was added 4-fluorobenzaIdehyde (2.2 g, 17.5 mmol). The mixture was stirred under reflux under nitrogen for 48 hours. The precipitate was fi ltered and washed with a mixture of solvents (ethyl acetate/hexane, 1 : 1 , 10 mL). After drying in vacuum, it afforded the title compound as a yellow solid ( 1 .2 g, yield 52 %). LC-MS (ESI) m/z: 38 1 (M+ 1 )+. Example 10
-8 4-fluoropheny l)-9-( 1 -meth l- 1 ^
Figure imgf000122_0001
(8) (1)
[00389] To a suspension of (£,)-7-fluoro-5-(4-fluorobenzylideneamino)-4-(( l -methyl-lH- l ,2,4- triazol-5-yl)methyl)phthalazin- l (2H)-one (8) (2.0 a, 5.3 mmol) in THF (80 mL) was added cesium carbonate (3.4 g, 1 0.6 mmol). The reaction mixture was stirred at 55 °C for 4 hours and cooled down to room temperature. The mixture was diluted with water (50 ml) and extracted with ethyl acetate (50 mL x 3). The combined organic layers were dried over anhydrous Na2SC>4 and evaporated to dryness to afford the title compound as a white solid ( l .6 g, yield 80 %). LC- S (ESI) m/z: 381(M+ 1 )+. Ή-NMR (400 MHz, DMSO-t/6) δ (ppm): 3.66 (s, 3 H), 4.97-5.04 (m, 2H), 6.91 -6.94 (dd, J, = 2.4, J2 = 1 1.2 Hz, 1 H), 7.06-7.09 (dd, Jx = 2.4, ,/2 = 8.8 Hz, 1 H), 7.14-7.18 (m, 3 H), 7.47-7.51 (m, 2H), 7.72 (s, 1 H), 7.80 (s, 1 H), 12.35 (s, 1 H).
Example 11
(£)-Methyl 5-fluoro-2-(3-(4-rluorophenyl)-2-( l-methyl- l - l ,2,4-triazol-5-yl)acryloyl)-3-nitrobenzoate
Figure imgf000122_0002
(4) (9)
[00390] To a stirred solution of methyl 5-fl uoro-2-(2-( 1 -methyl- 1 H- 1 ,2,4-triazol-5-y l)acety l)-3 - nitrobenzoate (4) (580mg, 2 mmol) and 4-fluorobenzaldehyde (488 mg, 4 mmol) in dimethylsulfoxide (2 mL) was added L-proline (230 mg, 2 mmol). The resulting mixture was kept with stirring at 45 °C for 48 hours. The reaction system was then partitioned between ethyl acetate (50 mL) and water (30 mL). and the organic phase was washed with water (20 mL x 3), dried with Na2S04, concentrated in vacuo, and purified by column chromatography (ethyl acetate: petroleum ether = 1 :3) to give the title compound (9) as a pale yellow foam (340 mg, yield 40 %). LC-MS (ESI) m/z: 429 (M+l f. Ή-NMR (400 MHz, DMSO-ds); δ (ppm): 3.56 (s, 3H), 3.86 (s, 3H), 7.02 (dd, 2H), 7.21 (dd, 2H), 7.90 (s, I H), 8.08 (s, 1 H), 8.26 (dd, I H), 8.56 (dd, I H). Example 12
Methyl 7-fIuoro-2-(4-fIuoropheny I)- 1 -hydroxy-3-( 1 -methyl- 1 H- 1 ,2,4-triazol-5-y l)-4-oxo- 1 ,2,3,4- tetrah droquinoline-5-carbox late ( 10)
Figure imgf000123_0001
[00391 } To a solution of ( )-Methyl S-fluoro-2-(3-(4-fluorophenyl)-2-( l-methy!-lH- l ,2,4-triazol-
5-yl)acryloyl)-3-nitrobenzoate (9) (200 mg. 0.467 mmol) in methanol (20 mL) was added 10 % Pd/C (24 mg). After the addition, the mixture was stirred under H2 ( 1 atm) at room temperature for 0.5 h. The reaction system was then filtered and evaporated under reduced pressure. The residue was purified by chromatography (ethyl acetate: petroleum ether = 1 : 1 ) to give the title compound ( 10) ( 1 10 mg, yield 57 %) as an off-white foam. LC-MS (ESI) m/z: 415 (M+H)+. Ή-NMR (400 MHz, DMSO-d6)5 (ppm): 3.53 (s, 3H), 3.73 (s, 3 H), 5.08 (d, 2H), 5.27 (d, 2H), 6.95 (dd, I H), 7.08 (dd, 2H), 7. 15 (dd, I H), 7.42 (dd, 2H), 7.77 (s, I H), 9.92 (s, I H).
Example 13
Methyl 7-fluoro-2-(4-fluorophenyl)-3-( 1 -methyl- lH- l ,2,4-triazol-5-yI)-4-oxo- 1 ,2,3 ,4- 5 )
Figure imgf000123_0002
( 10) (5)
[00392] To a stirred solution of methyl 7-fluoro-2-(4-fl uoropheny I )- 1 -hydroxy-3 -( 1 -methyl- 1 H-
1 ,2,4-triazol-5-y l)-4-oxo- 1 ,2,3,4-tetrahydroquinoline-5-carboxylate ( 10) (41.4 mg, 0.1 mmol) in methanol (5 mL) was added concentrated HCI solution (w/w 37 %, 1 mL) and reductive powdered Fe (56 mg, 1 mmol). The reaction mixture was refluxed for 3 hours. After the disappearance of compound ( 10) as monitored by LC-MS, the reaction system was partitioned between ethyl acetate (20 mL) and water (20 mL) and then the aqueous phase was extracted with ethyl acetate ( 10 mL x 3). The combined organic phase was dried with Na2SC<4, concentrated in vacuo and purified by column chromatography (ethyl acetate: petroleum ether = 1 : 1 ) to give the title compound (5) as a pale yellow solid ( 12 mg, yield 30 %). LC-MS (ESI) m/z; 399 (M+ l f. Ή-NMR (400 MHz, CDC13)5 (ppm): 3.58 (s, 3H), 3.87 (s, 3 H), 4. 17 (d, 1 H), 4.87 (s, 1 H), 5.38 (d, 1 H), 6.50 (dd, 2H), 6.99 (dd, 2H), 7.38 (dd, 2H), 7.80 (s, 1 H).
Exam le 14 Methyl 7-fluoro-2-(4-fluorophenyl)-3-( l -methyl- lH-l ,2,4-triazol-5-yl)-4-oxo- l ,2,3,4-
Figure imgf000124_0001
[00393] To a solution of (£)- ethyl 5-fluoro-2-(3-(4-fluorophenyl)-2-( l -methyl- \ H- 1 , 2,4-triazol-
5-yl)acryloyl)-3-nitrobenzoate (9) (214 mg, 0.5 mmol) in methanol (5 mL) was added concentrated HCI solution (w/w 37 %, 1 mL), then reductive Fe powder ( 140 mg, 2.5 mmol) was added slowly to the reaction system. After the addition was complete the resulting mixture was refluxed for 24 hours. The reaction mixture was then fi ltered, concentrated, neutralized with saturated NaHC03 (20 mL), and extracted with ethyl acetate ( 10 mL x 3). The residue was purified by chromatography (ethyl acetate: petroleum ether = 1 : 1 ) to give the title compound (5) (30 mg, yield 15 %) as an off-white foam. LC-MS (ESI) m/z: 399 (M+Hf.' H-NMR (400 MHz, DMSO-d,,) δ (ppm): 3.56 (s, 3H), 3.86 (s, 3H), 7.02 (dd, 2H), 7.2 1 (dd, 2H), 7.90 (s, 1 H), 8.08 (s, 1 H), 8.26 (dd, 1 H), 8.56 (dd, 1 H).
Example 15
(8R,95 -5-fluoro-8-(4-f1uorophenyl)-9-( l -methyl-lH-l ,2,4-triazoI-5-yl)-8,9-dihydro-2H-pyrido[4,3,2- ife]phthalazin-3(7#)-one ( 1 a) and (8S,9R)-5-fluoro-8-(4-fiuoropheny l)-9-( 1 -methyl- 1 H- 1 ,2,4-triazol-5-
Figure imgf000124_0002
( 1 ) ( la) ( l b)
100394] A chiral resolution of 5-fluoro-8-(4-fluorophenyl)-9-( l-methyl- l - l ,2,4-triazol-5-yl)-8,9- dihydro-2H-pyrido[4,3,2-ife]phthalazin-3(7//)-one (1) (52.5 g) was carried out on a super-fluid chromatography (SFC) unit using a CHIRALPAK 1A column and C(¾/ methano l/di ethy lam ine (80/30/0.1 ) as a mobi le phase. This afforded two enantiomers with retention times of 7.9 minute (23.6 g. recovery 90 %, > 98 % ee) and 9.5 minute (20.4 g, recovery 78 %, > 98 % ee) as analyzed with a CHIRALPAK IA 0.46 cm x 15 cm column and C02/methanol/diethylamtne (80/30/0. 1 ) as a mobile phase at a flow rate of 2 g/minute.
Example 16
(2R,3R)-methyI 7-fluoro-2-(4-fluorophenyl)-3-( 1 -methyl- l//- l ,2,4-triazol-5-yl)-4-oxo- 1 ,2,3,4- tetrahydroquinoline-5-carboxylate (6a) and (2S,3S)-methyl 7-fluoro-2-(4-fluorophenyl)-3-( l -methyl- 1 H- l ,2,4-triazol-5-yl)-4-oxo- l ,2.3,4-tetrahydroquinoline-5-carboxylate (6b)
Figure imgf000125_0001
Example 16A
[00395] The chiral resolution of compound (5) was carried out on a SFC unit with a
CHIRALPA ®IC 3 cm (I.D.) x 25 cm, 5 μπι column, using C02/MeOH (80/20) as a mobi le phase at a flow rate of 65 gf minute while maintaining the column temperature at 35 °C and with a detection UV wavelength of 254 nm. As such, a racemate of compound (5) (5 g) in methanol solution was resolved, which resulted in two enantiomers with a retention times of 2.35 minute (2.2 g, 88 % recovery, >98 % ee) and 4.25 minute (2.3 g, 92 % recovery, >98 % ee), respectively when analyzed using CHIRALPAK®IC 0.46 cm x 1 5 cm column and CO?/MeOH(80/20) as a mobile phase at a flow rate of 2 mL/ minute.
Example 16B
[00396] The chiral resolution of compound (5) was carried out on a SFC unit with a
C H IRA LP A K S IC 5 cm (I.D.) x 25 cm, 5 μιτι column, using C02/MeOH (75/25) as a mobile phase at a Flow rate of 200 mL/ minute while maintaining the column temperature at 40 °C and with a detection UV wavelength of 255 nm. As such, a racemate of compound (5) ( 1.25 kg) in methanol solution was resolved, which resulted in two enantiomers in about 83 % yield and 97.4 % purity.
Example 16C
[00397] Alternatively, the separation can also be achieved on a Simulated Moving Bed (SMB) unit with a CHIRALPA ®1C column and acetonitrile as a mobile phase. The retention times for the two enantiomers are 3.3 and 4.1 minutes, respectively. In certain embodiments, the productivity can be greater than 6 kg Feed/day/kg CSP.
Example 17
(8i?,9S)-5-fluoro-8-(4-fluorophenyl)-9-{ l -methyl- 1^
c/e]phthalazin-3(7/ )-one ( 1 a) and (8S,9i?)-5-fIuoro-8-(4-fluorophenyl)-9-( 1 -methyl- 1 H- 1 ,2,4-triazol-5- ( l b) "
Figure imgf000125_0002
(6a) ( la)
Figure imgf000126_0001
(6b) ( l b)
Example 17A
[00398] To a solution of (2R,3J¾)-methyl 7-fluoro-2-(4-fluoropheny l)-3-( 1 -methyl- 1 H- I ,2,4- triazol-5-yl)-4-oxo- 1 ,2,3,4-tetrahydroquinoline-5-carboxylate (6a) or (2S,3S)-methyl 7-fluoro-2-(4- fluorophenyl)-3-( l -methyl-lH-l ,2,4-triazol-5-yl)-4-oxo- l ,2,3,4-tetrahydroqumoline-5-carboxyIate (6b) (400 mg, 1 .0 mmol) in ethanol (8.0 mL) was added hydrazine monohydrate (85 %, 2.0 mL), and the solution stirred at room temperature for 2 hours. The resulting solution was then concentrated to a volume of 2 mL and filtered, and the resultant cake washed with ethanol ( 1 mL). After drying in vacuum at 50°C, this afforded the title compound as a white solid (209 mg, yield 55 %). LC- S (ESI) m/z: 381 (M+ l)f. Ή- N M R (400 MHz, DMSO-d6): δ (ppm): 3.681 (s, 3H), 4.99-5.06 (m, 21 1), 6.92-6.96 (m, 1 H), 7.08-7.1 1 (m, 1 H), 7. 16-7.21 (t, J = 8.8 Hz, 2H), 7.49-7.53 (m, 2H), 7.75 (s, 1 H), 7.83 (s, 1 H), 12.35 (s, 1 H).
Example 17B
100399] To a solution of (2R,3#)-methyl 7-fluoro-2-(4-fluoropheny l)-3 -( 1 -methyl- 1 H- 1 ,2,4- triazol-5-yl)-4-oxo- l ,2,3,4-tetrahydroquinoline-5-carboxylate (6a) or (2S,35)-methyl 7-fluoro-2-(4- fluoropheny l)-3-( 1 -methyl- 1 H- 1 ,2,4-triazol-5-yl)-4-oxo- 1 ,2,3 ,4-tetrahydroquinoline-5-carboxy late (6b) (446 g) in acetoi. itrile ( 10 volume) was added hydrazine monohydrate (2.9 eq.), and the se+ution stirred at room temperature for 2 hours. The resulting solution was then concentrated to a volume of 2 mL and filtered. The crude product was re-slurried with water (3~5 volumes) at 15~16 °C. After drying in vacuum at 50 °C, this affords the title compound as a white solid (329 g, yield 77%, 99.93% purity). LC- MS (ESI) m/z: 381(M+ 1 )+; Ή-NMR (400 MHz, DMSO-d6) δ (ppm): 3.681 (s, 3H), 4.99-5.06 (m, 2H), 6.92-6.96 (m, 1 H), 7.08-7.1 1 (m, 1 H), 7. 16-7.21 (t, J = 8.8 Hz, 2H), 7.49-7.53 (m, 2H), 7.75 (s, 1 H), 7.83 (s, l H), 12.35 (s, 1 H).
Preparation and Comparison of (8S, 9R)-5-fluoro-8-(4-fluorophenyl)-9-(l-methyl-l Η-Λ 2, 4-triazol-5-yl)-
8.9-dihydro-2H^yrido{4, 3,2-deIphthalazin-3(7H)-one salt and free base forms
[00400] Crystalline Compound A tosylate salt can, for instance, be prepared using crystallization techniques known in the art, including, but not limited to, melt cooling, rapid melt cooling, freeze drying, spray drying, roller drying, lyophilization, quench cooling the melt, rapid solvent evaporation, slow solvent evaporation, solvent recrystallization, slurry reerystal lization. melt crystallization, desolvation, sublimation, recrystallization in confined spaces (e.g., in nanopores or capillaries), recrystallization on surfaces or templates (e.g., on polymers), recrystallization in the presence of additives (e.g., co-crystal counter-molecules), dehydration, rapid cool ing, slow cooling, vapor diffusion, grinding, cryo-grinding, solvent-drop grinding, microwave-induced precipitation, ultrasonication-induced precipitation, laser- induced precipitation, and precipitation from a supercritical fluid.
[00401] The following example provides the preparation and characterization of (SS^^-fluoro-S-
(4-nuorophenyl)-9-( l-methyl-lH- l ,2,4-triazol-5-yl)-8,9-dihydro-2H-pyrido[4,3,2-rfe]phthalazin-3(7H)- one salt and free base polymorphs. Properties of a tosylate salt are shown to be superior to those of other salt and free base forms of (8S,9R)-5-fluoro-8-(4-fluorophenyl)-9-( l -meth> -lH-l ,2,4-triazol-5-yl)-8,9- dihydro-2H-pyrido[4,3,2-cfe]phthalazin-3(7//)-one. These improved properties include, but are not limited to, presence of a single crystalline form, no solvation, high melting point, non-hygroscopic ity, and/or thermal stability.
[00402] Salt forms of (8S,9R)-5-fluoro-8-(4-fluorophenyl)-9-( l -methyl-l H- l ,2,4-triazoi-5-yl)-8,9- dihydro-2H-pyrido[4,3,2-Je]phthalazin-3(7 )-one were prepared by treating the compound under a given condition with an acid. Salts forms, nos. 1-18, are identified in Table 1 below, where the acid used to treat (8S,9/?)-5-fiuoro-8-(4-fluorophenyl)-9-( l -methyl- lH- l ,2,4-triazol-5-yl)-8,9-dihydro-2H-pyrido[4,3,2- i/t']phthalazin-3(7//)-one are provided in the column under "Acid," and the conditions used during treatment are provided in the column under "'Condition."
[00403] Polymorphs of free base (8S,9R)-5-Fluoro-8-(4-fluoropheny l)-9-( 1 -methy 1- 1 H- 1 ,2,4- triazol-5-yl)-8,9-ciihydro-2//-pyrido[4,3,2-i/i']phthalazin-3( 7/ )-oiie were prepared from solutions prepared with different solvents. Characterizations of free base forms, nos. 19-24, are summarized in Table 2 and paragraphs referring to Table 2.
[00404] Microscopy and other standard laboratory measurements were employed to characterize solid forms, melting temperatures, solvation and/or hydration, and other physiochemical properties (including, but not limited to, solubility, P a, and Log P), of the salt forms and free base polymorphs.
TABLE 1: Salt forms of (8S,9R)-5-fluoro-8-(4-fluorophenyl)-9-( l -methyl- lH- 1 , 2,4-triazol-5-yl)-8,9- dihydro-2 -pyrido[4,3,2-t e]phthalazin-3(7 /)-one
Figure imgf000127_0001
Yield,
No. Acid Condition Product
%
Isethionic acid in
12 THF, 40 °C, 10 min, standing overnight White solid 98 ethanol (0.62 M)
Isethionic acid in THF (dilute), 60 °C, 10 min, precipitated
13 White solid 80 ethanol (0.62 M) quickly, standing overnight
14 Fumaric acid MeOH, 30 °C, 1.5 h No salt formed
15 Fumaric acid THF, 40 °C, 40 min No salt formed
MeOH, 32 °C, 0.5 h , precipitated out
16 83% H3PO4 White powder 40 through adding water
Acetone, 30-40 °C, 1 .5 h, precipitated out
17 83% H3PO4 White powder 40 through adding water
Acetone-MeOH, 52 °C, 20 min, standing at
18 83% H3PO4 r.t. overnight, precipitated from Acetone- 64
MeOH
TABLE 2: Solvents in preparing polymorphs of (85,9R)-5-fluoro-8-(4- fluorophenyl)-9-( 1 -methyl- 1 H- 1 ,2,4-triazol-5-yl)-8,9-dihydro- 2H-pyrido[4,3,2-fife]phthalazin-3(7H)-one
Figure imgf000128_0002
[004051 HCl Salt, 1: Microscopy: largely crystalline. Results based on microscopy, XRPD (Table
3), DSC (Table 4) and TGA (Table 5) suggest that this HCl salt is in mixed crystalline and amorphous forms and possibly solvated.
TABLE 3: X-Ray Powder Diffraction : Selected Peaks of 1 *
* Peaks with Relative Intensity of less than 20% are not reported
Relative
Angle, value,
Intensity,
°2Θ A /o
6.083 14.5174 76
259 7.21408 40.8
471 4 79966 22
21 609 10925 29
22.068 02473 24
22.392 96718 23.4
22.952 87167 100
>5.765 45499 29.8
26.389 37471 27.9
Figure imgf000128_0001
Figure imgf000129_0001
TABLE 5: Results of Th rmoaravimetric Analysis of 1
Ramp 10.00 °C/min to 300.
Figure imgf000129_0002
{00406] Mesylate Salt. 3: Microscopy: mixed crystalline and amorphous. Melting point: 179- 184
°C by melting point apparatus. Results based on microscopy, XRPD (Table 6), DSC (Table 7) and TGA (Table 8) suggest that this mesylate salt is in mixed forms and possibly solvated.
TABLE 6: X-Ray Powder Diffraction: Selected Peaks of 3*
* Peaks with Relative Intensity of less than 20% are not reported
Figure imgf000129_0003
TABLE 7: Results of Differential Scanning Calorimetry of 3
Ramp 10.00 °C/min to 300.00 °C
Figure imgf000129_0004
Ramp 10.00 "C/min to 300.00 *C
Starting Temperature = 25.64 *C
Figure imgf000129_0005
[00407] Tosylate Salt. 4. from methanol-acetonitrile: (8S,9R)-5-fluoro-8-(4-fluorophenyt)-9-( l - meth l- l H- 1 ,2,4-triazol-5-yl)-8,9-d^ ( 190 mg, 0.5 mmol) was dissolved in MeOH (2 mL) and CH3CN (2 mL) at 50 °C. TsOH ( 190 mg, 1 mmol), dissolved in a mixture of MeOH (2 mL) and CH3CN (2 mL), was then added. After 30 seconds, a white solid precipitated from the solution and the solution was allowed to cool to 25 °C. Stirring was continued at 25 °C for approximately 2 h. The white solid was collected by filtration, washed with CTLC (2 mL), and dried under vacuum at 45 °C for 3 days. Three preparations were made with yields of 47%, 56% and 56%, with similar results as determined by microscopy, XRPD. DSC and TGA (ramp 10.00 *C/min to 300.00 °C). Results provided herein are from one of these preparations. Microscopy: crystalline. Results based on microscopy, XRPD (Table 9) and DSC (Table 10) suggest that this tosylate salt is substantially a single crystal line form.
TABLE 9: X-Rav Powder Diffraction: Selected Peaks of 4*
* Peaks with Relative Intensity of less than 8% are not reported
Figure imgf000130_0001
TABLE 10: Results of Differential Scanning Calorimetry of 4
Ram 10.00 'C/min to 400.00 °C
Figure imgf000130_0002
[00408] Tosvlate Sal 5, from dich loromethane-aceton itri le: (8 \9/i)-5-fluoro-8-i4-fluoropheny 1)-
9-( l -methyl- lH- l ,254-triazol-5-yl)-8,9-dihydro-2H-pyrido[4,3,2-ife]phthalazin-3(7H)-one ( 120 mg, 0.3 16 mmol) was suspended in CFLCL (5 mL) and CH3CN (5 mL) at 40 °C, then TsOH (66 mg, 0.348 mmol) was added, after adding, the solution was clear. After 5 seconds, a white solid was precipitated from the solution, continued to stir at 25 °C for 1 h, filtered to obtain the white crystal sol id, the solid was washed by Ci hCN (2 mL), dried under vacuum at 45 °C for 3 days. Microscopy: birefringent, crystalline. Results based on microscopy, XRPD (Table 11), DSC (Table 12a and Figure 3a), and TGA (Table 12b and
Figure 3b) suggest that this tosylate salt is substantially a single crystall ine form. TABLE 11 : X-Rav Diffraction: Selected Peaks of 5*
* Peaks with Relative Intensity of less than 10% are not reported
Figure imgf000131_0001
TABLE 12a: Results of Differential Scanning Calorimetry of 5
Ram 10.00 °C/min to 400.00 "C
Figure imgf000131_0002
TABLE 12b: Results of TGA of 5
Ramp 10.00 °C/min to 400.00 °C
Figure imgf000131_0003
J00409] Tosylate Salt. 6. from acetone: (8S,9/?)-5-fluoro-8-(4-fluorophenyl)-9-( 1 -methyl- 1//- l ,2,4-triazol-5-yl)-8,9-dihydro-2H-pyrido[4,3,2-^]phthalazin-3(7/J)-one ( 120 mg, 0.3 16 mmol) was suspended in acetone ( 15 mL) at 25 °C. After heating to 60 °C, the mixture became clear and the temperature was reduced to 45 C. TsOH (70 mg, 0.35 mmol) was then added, and after 30 seconds, a white solid was precipitated from the solution which was allowed to cool to 25 °C. Stirring was continued at 25 °C for 1 h. The white crystal solid was collected by filtration, washed with acetone (8 mL), and dried under vacuum at 45 °C for 3 days. Microscopy: biretringent, crystalline. Results based on microscopy, XRPD (Table 13), DSC (Table 14a and Figure 4a), and TGA (Table 14b and Figure 4b) suggest that this tosylate salt is substantially a single crystalline form.
TABLE 13: X-Rav Powder Diffraction: Selected Peaks of 6*
* Peaks with Relative Intensity of less than 10% are not reported
Relative
Angle, d value,
Intensity,
"2Θ A
%
7.461 1 1 .83921 100 Relative
Angle, d value,
A Intensity,
°2Θ
%
12.474 7.09046 10.2
14.447 6.12594 23.7
1 5.092 5.86557 27.3
17.4 5.09252 19.6
17.741 4.99533 17.9
18. 1 1 4.89433 29.4
18.529 4.78464 15.8
19.045 4.65619 22.1
20.092 4.41595 1 0
20.425 4.34457 38.1
21.464 4.13667 22.7
21.772 4.0787 22.4
22.626 3.92672 22.4
23. 101 3.84709 12.9
24.03 3.70042 78.7
24.851 3.5799 17.2
26.96 3.30456 12
29.82 2.99375 13.6
31 .304 2.8551 1 10.5
TABLE 14a: Results of Differential Scanning Calorimetry of 6
Ramp 10.00 'C/min to 400.00 °C
Figure imgf000132_0001
TABLE 14b: Results of TGA of 6
Figure imgf000132_0002
[00410] Tosylate Salt. 7. from THF: ( 8.y.9/?)-5-tluoro-8-(4-nuorophenvD-9-( 1 -inethvl- l //- 1 .2.4- triazol-5-yl)-8,9-dihydro-2 -pyrido[4,3,2-cfe]phthalazin-3(7 )-one ( 120 mg, 0.316 mmol) was suspended i THF (6 mL) at 25 °C. After heating to reflux, the mixture became clear and the temperature was reduced to 45 °C. TsOH (66 mg, 0.35 mmol) was then added, and after 1.5 min, a white solid was precipitated from the solution which was allowed to cool to 25 °C. Stirring was continued at 25 °C for 30 min. The white crystal solid was collected by filtration, washed with CH2CI2 ( 10 mL), and dried under vacuum at 45 °C for 3 days. Microscopy: birefringent, crystalline. Results based on microscopy, XRPD (Table 15), DSC (Table 16a and Figure 5a), and TGA (Table 16b and Figure 5b) suggest that this rosy late salt is substantially a single crystalline form. TABLE 15: X-Ray Powder Diffraction: Selected Peaks of 7*
* Peaks with Relative Intensity of less than 10% are not reported
Figure imgf000133_0001
TABLE 16a: Results of Differential Scanning Calorimetry of 7
Ramp 10.00 "C/rnin to 400.00 °C
Figure imgf000133_0002
TABLE 16b: Resu Its of TGA of 7
Ramp 10.00 "C/min to 400.00 'C
Figure imgf000133_0003
[00411J Tosvlate Salt, 8. from acetone-THF: (85,9i?)-5-fluoro-8-(4-fluorophenyl)-9-( l -methyl- lH- l ,2,4 riazol-5-yl)-8,9-dihydro-2H-pyrido[4,3,2-c/e]phthaIazin-3(7i -one (400 mg, 1 .05 mmol) was suspended in a mixture of acetone (27 mL) and THF ( 13 mL) at 25 °C, after heating to reflux, the reaction m ixture became clear. Then TsOH (220 mg, 1 .16 mmol) was added, after 30 seconds, a white solid was precipitated from the solution, continued stirring at 25 °C for 30 inin. filtered to obtain the white crystal solid, which was washed by a mixture of acetone ( 10 mL) and 1 ,4-dioxane (4 mL), dried under vacuum at 45 °C for 3 days. Microscopy: birefringent, crystalline. Results based on microscopy, XRPD (Table 17 and Figure 8), DSC (Table 18a and Figure 2a), and TGA (Table 18b and Figure 2b) suggest that this tosylate salt is substantially a single crystalline form. DVA was performed on this polymorph and showed a 0.1574% weight gain from 0%-95% RH (Figure 8).
TABLE 17: X-Rav Powder Diffraction: Selected Peaks of 8*
* Peaks with Relative Intensity of less than 2% are not reported
Figure imgf000134_0001
TABLE 18a: Results of Differential Scanning Calorimetry of 8
Ram 10.00 °C/min to 400.00 'C
Figure imgf000134_0002
TABLE 18b: Results of TGA of 8
Ram 10.00 °C/min to 400.00 °C
Figure imgf000134_0003
[00412] The XRPD peaks from preparations of crystalline tosylate salt forms 4, 5, 6, 7, and 8 are similar. Overlay of XRPD graphs for 5, 6, 7, and 8 are shown in Figure 1. Each tosylate salt form 4, 5, 6, 7, and 8 had a single endothermic peak above 300 °C as determined by DSC, whereas other salt and free base forms all had endothermic peaks well below 300 °C. These results indicate a single relatively stable polymorph exists for the tosylate salt of (8S,9i?)-5-fluoro-8-(4-fluorophenyl)-9-( l -methyl- ΪΗ- 1 ,2,4- triazol-5-yl)-8,9-dihydro-2/f-pyrido[4,3,2-£/e]phthalazin-3(7H)-one.
[00413] Isethionic Salt, 11. 12 and 13, from THF: Microscopy: crystall ine. DSC results were not consistent between 11, 12 and 13, although all three preparation had at least 2, sometimes 3, endothermic peaks, including a broad endothermic peak below 150 "C and a sharp endothermic in the vicinity of 272 °C to 282 "C. Unlike the tosylate salts described above, which did not show any weight change below 200 "C by TGA, isethionic salt 11 had a 5.72 % weight loss at 134.8 °C, and isethionic salt 12 showed a 7.034 % weight loss at 139.06 °C. Results obtained on the isethionic salts suggest that this salt form exists in more than one form.
[00414] Phosphate Salt, 16, 17 and 18: Microscopy, XRPD and DSC results were consistent with mixed crystalline and amorphous forms being present in each of the three phosphate salt preparations.
[00415] Free base, 19. from acetone: Microscopy, XRPD and DSC results were consistent with a mixture of forms.
TABLE 19: X-Rav Powder Diffraction: Selected Peaks of free base 19*
* Peaks with Re /alive Intensity of less than 30% are not reported
Figure imgf000135_0001
TABLE 20: Results of Differential Scanning Calorimetry of free base 19
Ramp 10.00 Cm in to 300.00 °C
Figure imgf000135_0002
TABLE 21: Results of Thermogravimetric Analysis of free base 9
Ramp 10.00 'C/min to 300.00 °C
Starting Temperature = 25.64 *C
Temperature, Weight Change,
C %
120 1 .054
161 4.256
174 5.977 Temperature, Weight Change,
°c %
185 7.82
238 8.874
[00416] Free base, 20, from MeOH/EtOAc: Microscopy showed birefringent crystalline form;
XRPD and DSC results were consistent with a mixture of forms.
TABLE 22: X-Rav Powder Diffraction: Selected Peaks of free base 20*
* Peaks with Relative intensity of less than 14% are not reported
Figure imgf000136_0001
TABLE 23: Results of Differential Scanning Calorimetry of free base 20
Figure imgf000136_0002
TABLE 24: Results of Thermogravimetric Analysis of free base 20
Ramp 10.00 *C/min to 300.00 °C
Starting Temnerature = 25.64 °C
Figure imgf000136_0003
[00417] Free base form 21, from acetonitrile: A mixture of crystalline and amorphous forms was observed. DSC: broad endotherm with maximum near 133.05 *C, exotherm maximum near 209.21 *C and sharp endotherm with maximum at 253.85 *C. TGA: weight loss, at 142.46 °C was 3.7%, and at 256.48 "C was 4.259%. The d values in angstroms (A) for peaks, greater than 1 % relative intensity, resolved by XRPD were as follows: 16.58486; 1 1 .49904; 6.33021 ; and 6.01 178.
[00418] Free base form 22. from methanol: A mixture of cry stalline and amorphous forms with properties similar to that of free base form 20 was observed.
[00419] Free base form 23. from isopropanol: The material produced was amorphous. [00420] Free base form 24, from DMS/ethanol: A mixture of crystalline and amorphous forms was observed. DSC: endotherm peaks: 52.20 °Cmax ( 12.54 J/g); 202.63 °Cmax ( 120.9 J/g); 249.34 °Cmax (40.65 J/g). TGA: ambient to 120 *C, 0.9388% weight loss; at 223.8 *C, 17.47% weight loss.
|004211 These results demonstrate that crystalline forms of (8S,9R)-5-fluoro-8-(4-fluorophenyl)-9-
( 1 -methyl-1 /- l,2,4 riazol-5-yl)-8,9-dihydro-2H-pyrido[4,3,2-i e]phthalazin-3(7H)-one free base can be prepared from certain solvents, however these preparations were found not to be single crystal line forms having stability matching that of a crystalline tosylate salt form provided herein.
Exemplary Preparation of f8S.9R)-5-fluoro-8-(4-fluorophenyl)-9-(l-methyl-l - 2,4-triazol-5-vl)-8,9- dihvdro-2H-pyridof4,3,2-de]p thalazin-3f7H)-one tosylate salt
[00422) The following exemplifies a process suitable for GMP large scale production of (8S.9R)-
5-fluoro-8-(4-fliiorophenyl)-9-( 1 -methyl- 1 //- 1 ,2.4-triazol-5-y l)-8.9-dihydro-2H-pyrido[4,3.2- cfe]phthalazin-3(7H)-one mono-tosylate salt as a single crystalline form.
[00423] Variation 1 : To a solution of (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-i/e]phthalazin-3(7H)-one ( 12.4 g) in THF (40 vols) was slowly added a solution of TsOH ( 1 .05 equiv.) in THF (5 vols) at 50 °C~54 °C. The mixture was stirred for additional 30 minutes at this temperature. The mixture was then concentrated down to 3-5 vols by distil lation at 30 °C-40 °C under reduced pressure (vacuum:-0.07MPa~ O.OSMPa). Further removal of THF solvent was achieved by adding acetone (20 vols) and then distil ling down to 3~5 vols under reduced pressure, which was repeated three times. The mixture was cooled down to 5 °C and filtered under nitrogen protection. The solid was dried at 60 °C for 17 hours. It gave the title compound as a white crystal solid ( 16.0 g, 89% yield). LC-MS (ESI) m/z: 381 (M+ l . ' H-NMR (400 MHz, DMSO-d6) δ (ppm): 2.29 (s, 3H), 3.67 (s, 3H), 4.97-5.06 (m, 2H), 6.91-6.94 (dd, j'=2, J = 1 0.8 Hz, 1 H), 7.06-7.19 (m, 5H), 7. 19-7.5 1 (m, 4H), 7.74 (s, 1 H), 7.87 (s, 1 H), 10.32 (brs, 1 H), 12.36 (s, 1 H).
[00424] Variation 2: To a solution of ( 8S,9/ -5 -fluoro-8-(4-fluorophenyl)-9-( 1 -methyl- 1 //- 1 ,2,4- triazol-5-yl)-8,9-dihydro-2/f-pyrido[4,3,2-i/i?]phthalazin-3(7H)-one (0.24 kg) in THF (40 vols) was slowly added a solution of TsOH ( 1.05 equiv.) in THF (5 vols) at 50 °C~54 °C. The mixture was stirred for additional 30 minutes at this temperature. The mixture was then concentrated down to 3-5 vols by distil lation at 30 °C-40 °C under reduced pressure (vacuum:-0.07MPa— 0.08MPa). Further removal of THF solvent was achieved by adding acetone (20 vols) and then distilling down to 3~5 vols under reduced pressure, which was repeated three times. After removal of THF solvent and distilling, the mixture was re- slurried with 12 vols of acetone for 14- 15 hours at 50 °C~54 °C. The mixture was then cooled down to 5 °C and filtered under nitrogen protection. The solid was dried at 60 °C for 17 hours. It gave the title compound as a white crystal solid (0.3 1 kg, 91.7% yield, 99.65% purity).
[00425] When subject to H- MR. the one-to-one integration ratio of N-methyl at 3.67 ppm from the free-base and the methyl at 2.29 ppm from toluene sulfonic acid suggests a mono-tosylate salt formation. [00426] When subjected to DVS at 25 °C, the tosylate salt produced from using the steps described in Variation 2 showed negligible weight gain (less than 1 %) of the tosylate salt as humidity was increased from 0% to 95% which suggests that it is not hygroscopic. (See Table 28.)
[004271 The XRPD pattern in Figure 6 was collected with a PANalytical X'Pert PRO
MPD PW3040 diffraetometer using an incident beam of Cu radiation produced using an Optix long, tine- focus source. An elliptically graded multilayer mirror was used to focus Cu a X-rays (1.54059
A) through the specimen and onto the detector. Prior to the analysis, a silicon specimen (NIST SRM 640d) was analyzed to verify the observed position of the Si 1 1 1 peak is consistent with the NIST- certified position. A specimen of the sample was sandwiched between . un-thiek films and analyzed in transmission geometry. A beam-stop, short antiscatter extension, antiscatter knife edge were used to minimize the background generated by air. So Her slits (0.02 * 0.02 radians) for the incident and diffracted beams were used to minimize broadening from axial divergence. Diffraction patterns were col lected using a scanning position-sensitive detector (X'Celerator) located 240 mm from the specimen and Data Collector software v. 2.2b. The XRPD data acquisition parameters are: transmission mode, X-ray tube settings of 45 kV and 40 mA, 1.00-39.99 °2Θ scan range, 0.017 °2Θ step size, 1939 seconds collection time, 1 .2°/minute scan speed, ½° divergence sl it, and 1.0 second sample revolution time. The entire l ist of peaks identified in the XRPD pattern, or a subset thereof, may be sufficient to characterize the polymorph(s) obtained. Results provided for XRPD (Table 25) are for a tosylate salt prepared using the steps described in Variation 2. The data in Table 25 are from the XRPD spectrogram provided in Figure 6.
[00428] DSC analyses were performed on the tosylate salts prepared using variation 1 and variation 2 above and simi lar results were observed.
[00429] A DSC thermogram for tosylate salt prepared using the steps described in Variation 2 was acquired using a ettler Toledo Differential Scanning Calorimeter 1 . The temperature was ramped from 25 °C to 400 "C at 10 ° C/minute. In the DSC figure, exothermic events are plotted in the upward direction. The data in Table 26 are from the DSC graph provided in Figure 7a. The thermogram shows a very sharp onset of melting, and an exotherm after the completion of melting indicates that melting occurs with decomposition. The flat baseline prior to reaching the melting onset indicates no thermal transitions before melting, suggesting no solv ate formation and no transformation of one crystalline form to another.
[00430] A TGA thermogram for tosylate salt prepared using the steps described in Variation 2 was acquired using a Mettler Toledo Thermogravi metric Analyzer/ Differential Scanning Calorimeter 1. The temperature was ramped from 25 °C to 400 *C at 10 * C/minute. The TGA graph is prov ided in Figure 7b. The trace shows a stable baseline up to the start of melting and decomposition. This indicates that there is little if any residual solvent or absorbed water present in the crystal line product, and that the product is stable to heat, prior to the onset of decomposition at the melting point.
[00431 ] The solid-state 13C cross polarization magic angle spinning (CP/MAS) NMR spectrum for the tosylate salt prepared using the steps described in Variation 2 was acquired at 25 °C on a Varian mmINOVA-AQQ spectrometer (Larmor frequencies: 13C = 100.543 MHz, Ή = 399.787 MHz). The sample was packed into a 4 mm PENCIL type zirconia rotor and rotated at 12 kHz at the magic angle. The spectrum was acquired with phase modulated SPINAL-64 high power Ή decoupling during the acquisition time using a Ή pu lse width of 2.6 β (90°), a ramped amplitude cross polarization contact time of 5 ins, a 3 ms acquisition time, a 20 second delay between scans, a spectral width of ~45 kHz with 2799 data points, and with 400 co-added scans. The free induction decay (FID) was processed using Varian/Agilent VNMR 6.1C software with 65536 points and an exponential line broadening factor of 10 Hz to improve the signal-to-noise ratio. The first three data points of the FID were back predicted using the VNMR linear prediction algorithm to produce a flat baseline. The chemical shifts of the spectral peaks were external ly referenced to the carbonyl carbon resonance of glycine at 176.5 ppm. The entire list of peaks identified in the 13C NMR spectrum, or a subset thereof, may be sufficient to characterize the polymorph(s) obtained. The data in Table 27 are from the spectrum provided in Figure 12.
TABLE 25: XRPD: Selected Peaks of Tosylate Salt prepared from Large Scale Process *
* Peaks with Relative Intensity of less than 2% are not reported
Relative
Angle, d value,
Intensity,
20 (±0.2 2Θ) A
%
7.22 12.242 ± 0.348 6
7.51 1 1.780 ± 0.322 46
9.49 9.3 16 ± 0.200 2
1 1.27 7.855 ± 0.142 2
12.35 7.166 ± 0.1 17 8
12.52 7.071 ± 0.1 14 7
13.82 6.407 ± 0.094 1
14.47 6. 120 ± 0.085 1 8
15.14 5.851 ± 0.078 15
17.41 5.092 ± 0.059 9
17.62 5.035 ± 0.057 3
17.78 4.988 ± 0.056 5
18.12 4.897 ± 0.054 10
18.53 4.787 ± 0.052 6
19.07 4.654 ± 0.049 9
20.09 4.420 ± 0.044 1 00
20.46 4.342 ± 0.042 13
21.48 4.138 ± 0.038 1 7
21.81 4.075 ± 0.037 12
22.26 3.994 ± 0.036 6
22.65 3.927 ± 0.035 5
23.10 3.851 ± 0.033 8
24.05 3.701 ± 0.03 1 53
24.25 3.670 ± 0.030 6
24.83 3.585 ± 0.029 6
25.25 3.527 ± 0.028 5
26.37 3.380 ± 0.025 2
26.77 3.330 ± 0.025
26.96 3.308 ± 0.024 3
27.84 3.204 ± 0.023 3 Relative
Angle, d value,
Intensity,
°2Θ (±0.2 2Θ) A %
28.64 3.1 16 ± 0.021 1
28.78 3.102 ± 0.021 2
29.18 3.061 ± 0.021 1
29.81 2.997 ± 0.020 8
TABLE 26: Results of DSC of Tosylate Salt prepared from Large Scale Proces
Ramp 10.00 'C/min from 25.00 to 400 00 *C
Figure imgf000140_0001
TABLE 27: Results of Solid State l 3C NMR of Tosylate Salt prepared from Large Scale Process
Figure imgf000140_0002
TABLE 28: Results of DVS Isotherm of Tosylate Salt prepared from Large Scale Process
Target % Change in Mass (%)
P/Po Sorption Desorption Hysteresis
0.0 0.0002 -0.0015
5.0 0.0214 0.0203 -0.001 1
10.0 0.0301 0.0326 0.0025
15.0 0.0375 0.0421 0.0045
20.0 1 0.0529 0.05 13 -0.0016
25.0 ! 0.0617 0.0649 0.0033 Target % Change in Mass (%)
P Po Sorption Desorption Hysteresis
30.0 0.0698 0.0791 0.0092
35.0 0.0812 0.0947 0.0134
40.0 0.1043 0.1084 0.0042
45.0 0.1233 0.1266 0.0033
50.0 0.1451 0.1456 0.0005
55.0 0.1641 0.1668 0.0027
60.0 0.1 842 0.1895 0.0053
65.0 0.2127 0.2192 0.0065
70.0 0.2450 0.2535 0.0085
75.0 0.2798 0.2919 0.0121
80.0 0.3253 0.3402 0.0149
85.0 0.3922 0.41 13 0.0190
90.0 0.4983 0.5230 0.0247
95.0 0.7473 0.7473
Stability, Solubility and Dissolution o f Compound A Tosylate Salt
[00432] When tested in aqueous buffers, the solubility of crystalline Compound A tosy late salt was determined to vary from 43
Figure imgf000141_0001
(pH 0.8) to 17 μ$*/ηιΙ (pH 9.6) at 25 °C. A stability study over period of 20 days, indicated that Compound A tosylate salt is most stable in aqueous solution at 25 "C from pH 1 -8, as opposed to solutions with more basic pH. The intrinsic dissolution rate CIDR") of crystalline Compound A tosylate salt was measured using from a rotating disk with constant surface area to predict absorption rate in vivo. Its IDR in stimulated gastric fluid (without enzyme) was 39
/ΰτη2/ηιίη. In fasting simulated intestinal fluid the IDR was 36 μ§Λ;πι2/πιίη.
Excipient Compatibility
[00433] To test for excipient compatibility, samples of crystal line Compound A tosylate salt in the presence of a given excipient (i.e., binary mixtures) were stored in an open container at 40 °C (ambient humidity) or at 40 °C/75% RH, and tested after being stored for 2 weeks or 4 weeks. Excipients tested included diluents (e.g., si I ici fled microcrystalline cellulose, dibasic calcium phosphate anhydrous, lactose), disintegrants (e.g., crospovidone, croscarmellose sodium, sodium starch glycolate), binders (e.g., povidone, hydroxypropyl cellulose, hypromellose, maltodextrin), lubricants (e.g., sodium stearyl fumarate, magnesium stearate, stearic acid), glidants (e.g. , talc, col loidal si licon dioxide), and capsule shell materials (e.g., gelatin, hypromellose), among others. No significant increase in total related substances (TRS) or decrease in potency was typically observed in the binary mixtures when stored at the 40 °C/75%RH storage condition for up to 4 weeks. Results showed that crystalline Compound A tosylate salt may be less compatible with, for instance, dibasic calcium phosphate, corn starch or povidone than certain other excipients. This example indicates that excipients are not equal in their compatibility with crystalline Compound A tosylate salt.
Exemplary Dry Blend Method No. 1
[00434] The fol lowing exemplary dry blend method was used to prepare Compound A tosylate salt as a pharmaceutical composition suitable for administration to a patient. [004351 The method involves geometric dry blending of PROSOLV SMCC 50 (total amount in final product was 992.735 g and crystalline Compound A tosylate salt (7.265 g). Ingredients are sieved through a dedicated 250 μιτι (60 mesh) screen prior to being weighed and used. Alternatively,
PROSOLV® SMCC HD90 can be used and crystalline Compound A tosylate salt comprises in one example about 0. 1 % wt/wt to about 0.6% wt wt of the formulation.
[00436] Step 1 : Pre-Blending Using Mortar and Pestle. Add to the mortar, and in the following order, 7.200 g Prosolv* SMCC 50, 7.265 g Compound A tosylate salt and 7.200 g PROSOLV® SMCC 50. Triturate until homogenous (typically 13- 16 min).
[00437] Add 65.00 g Prosolv* SMCC 50 to the mixture, and triturate until homogenous (typically
1 1 - 13 min).
[00438] Add 65.00 g Prosolv* SMCC 50 to the mixture, and triturate until homogenous (typically
1 1 - 13 min).
[00439] Add 100.00 g Prosolv* SMCC 50 to the mixture, and triturate until homogenous
(typically 1 1 - 13 min). The resulting mixture is the "pre-blend mixture" which contains Compound A tosylate salt.
[00440] Step 2: Mixing in Flo-Bin* Blender. Add the fol lowing materials to a 5 L bowl of the bin blender (F: 417.07 g Prosolv* SMCC 50, followed by the pre-blend mixture.
[00441 ] "Rinse" the mortar/pestle by adding 66.00 g Prosolv* SMCC 50 to the mortar and triturate for 3-6 min, and transfer to the bin blender.
[00442] Add 66.00 g Prosolv* SMCC 50 to the mortar and triturate for 3-6 min, and transfer to the bin blender.
[00443] Add 66.00 g Prosolv* SMCC 50 to the mortar and triturate for 3-6 min, and transfer to the bin blender.
[00444] Add 66.00 g Prosolv* SMCC 50 to the mortar and triturate for 3-6 min, and transfer to the bin blender.
[00445] Add 67.26 g Prosolv® SMCC 50 to the mortar and triturate for 3-6 min, and transfer to the bin blender.
[00446] Mix the contents in the bin blender at 20 RPM for 10 min, stop the blend and manually mix the blend using a customized spatula of 45 cm. and repeat this step twice for total bin blender mixing time of is 30 min. Sieve the blended mixture through a dedicated 60 mesh screen and transfer the sieved material into the bin blender bowl and mix at 20 RPM, as before, for 30 min. Transfer the contents of the blend into an appropriate container to yield the exemplary pharmaceutical composition (0.726% wt wt Compound A tosylate salt).
Exemplary Dry Blend Method No. 2
[00447] A modified version of exemplary dry blend method no. 1 was performed, where the modifications were to perform the method on a smaller scale using a Turbula* blender, and to replace a small amount of the Prosolv* SMCC 50 added directly to the blender with an equal amount of colorant (FD&C Blue No. 2). With this modified method, a pharmaceutical composition was prepared having 0.726% wt/wt Compound A tosylate salt and 0.200% wt vvt FD&C Blue No. 2. Alternatively,
PROSOLV® SMCC HD90 can be used and crystal line Compound A tosylate salt comprises in one example about 0.1 % wt/wt to about 0.6% wt wt of the formulation.
Exemplary Dry Blend Method No. 3
[00448] The following prophetic example provides a modified version of exemplary dry blend method no. 1 which can be used to prepare Compound A tosylate salt as a pharmaceutical composition. Alternatively, PROSOLV® SMCC HD90 can be used and crystalline Compound A tosylate salt comprises in one example about 0.1% wt/wt to about 0.6% wt/wt of the formulation.
[00449J For each 1 kg of dry blend formulation to be prepared, 7.2 grams of SMCC are added to an agate mortar, followed by 7.265 g of crystalline Compound A tosylate salt and another 7.2 g of SMCC. These powders are triturated with an agate pestle for 13- 16 min, making sure that no clumps or agglomerates are present. Following this, 65 g of SMCC is added to the mortar, and trituration is repeated for 1 1- 13 min. A 5-1 stainless steel bin blender is charged with 417 g of SMCC, followed by the dry blend from the mortar. The remaining SMCC to make the 1 kg batch is divided into three portions (each about 132 g); each portion is triturated in the mortar for five min and transferred to the bin blender to rinse residual drug substance that may be present from the mortar/pestle into the blender.
Exemplary Dry Blend Formulations
[00450] The fol lowing prophetic example provides formulations suitable for filling into capsules or for being compressed into tablets.
[00451] Exemplary dry blend formulation (A-D): Follow the procedures for preparing exemplary dry blend method no. 1 described above. After all the SMCC is blended into the formulation, add disintegrant to the powder mixture and blend the mixture for 10 min. Add magnesium stearate and mix for another 2 to 5 min to yield the final blend. Transfer the final blend to a capsule filing machine for encapsulation or a tablet press for tablet compression.
Example A: Composition of 25 μg capsule or tablet prepared by dry blending
Amount per
capsule or tablet
Ingredient %, vv/w (mg)
Compound A tosylate salt 0.7265 0.036
SMCC 94.774 4.739
Crospovidone 4.000 0.200
Magnesium stearate 0.500 0.025
Total 100.000 5.000 Example B: Composition of 50 capsule or tablet prepared by dry blending
Amount per
Ingredient %, w/w tablet (mg)
Compound A tosylate salt 0.7265 0.073
SMCC 94.774 9.477
Crospovidone 4.000 0.400
Magnesium stearate 0.500 0.050
Total 100.000 10.000
Example C: Composition of 250 μ capsule or tablet prepared by dry blending
Amount
per tablet
Ingredients %, w/w (mg)
Compound A tosylate salt 0.7265 0.363
SMCC 94.774 47.387
Crospovidone 4.000 2.000
Magnesium stearate 0.500 0.250
Total 100.000 50.000
Example D: Composition of 1 mg capsule or tablet prepared by dry blending
Amount
per tablet
Ingredients %, w/w (mg)
Compound A tosylate salt 0.7265 1.453
SMCC 94.774 189.547
Crospovidone 4.000 8.000
Magnesium stearate 0.500 1.000
Total 100.000 200.000
Exemplary Unit Dosage Forms in Capsules
[00452] The following unit dosage forms were prepared in capsules using the XCELODOSE® automated encapsulator system (Capsugel, Peapack, New Jersey, USA) with the pharmaceutical composition prepared according to exemplary dry blend method no. 1 described above.
[00453] 25 μ¾ Capsule. Add 5 mg of the dry blend pharmaceutical composition as described above (0.726% wt/'wt Compound A tosylate salt and Prosolv® SMCC 50) to an appropriate sized hypromellose capsule. The amount of Compound A tosylate salt in the capsule is 36.325 g, which, using the conversion factor of 1.453 mg Compound A tosylate salt per 1.000 mg of Compound A, provides about 25 μg of Compound A.
[00454] 50 μ¾ Capsule. Add 10 mg of the dry blend pharmaceutical composition as described above (0.726% wt wt Compound A tosylate salt and Prosolv* SMCC 50) to an appropriate sized hypromellose capsule. [00455] 250 μζ Capsule. Add 50 mg of the dry blend pharmaceutical composition as described above (0.726% wt/wt Compound A tosylate salt and Prosolv* SMCC 50) to an appropriate sized hypromel lose capsule.
[00456] 1 mg Capsule. Add 200 mg of the dry blend pharmaceutical composition as described above (0.726% wt wt Compound A tosylate salt and Prosolv® SMCC 50) to an appropriate sized hypromellose capsule.
Comparison of Excipient Compatibility in Dry Blend Formulations
[00457] This example provides a comparison of blend uniformity of three formulation prototypes each prepared with crystalline Compound A tosyalte salt and one of three excipients (starch 1500, mannitol and SMCC). The formulation prototypes were prepared using a modified version of exemplary dry blend method no. 2, described above, which employs geometric mixing.
[00458] Of the three excipients evaluated, the formulation prototype prepared using SMCC exhibited the best blend uniformity in terms of relative standard deviation ("%RSD"), mean drug content and acceptance value ("AV"). Results are provided in Table 29. In general, formulations with better blend uniformity have a lower % RSD, higher drug content and lower AV. The requirements for dosage and blend uniformity are met if the AV of 10 samples randomly selected throughout the blend or encapsulation process is less than or equal to 15% (see, e.g., USP <905>, Ph. Eur. 2.9.1 ).
TABLE 29: Effect of Diluents on Formulation Blend Uniformity
Figure imgf000145_0001
Content Uniformity of Unit Dosage Forms Prepared with SMCC
[00459] Two 1 -kilogram scale blends were prepared with Compound A tosylate salt and SMCC according to exemplary dry blend method no. 1 described herein and were filled into Size 0 hypromellose capsules (VCAPS* PLUS, Capsugel, USA) using an Xcelodose® unit. Four capsule strengths (25 μg, 50 μg, 250 μg, and 1 ing) were filled. Ten capsules from each batch were collected randomly throughout the encapsulation process and assayed for drug content. The capsules yielded acceptable content uniformity as shown in Table 30.
TABLE 30: Content Uniformity of Two 1 Kg Scale-up Batches
Figure imgf000146_0001
ND = Not Determined
[00460] This example demonstrates preparation of unit dosage forms having content uniform ity well within a range accepted in the art (e.g., between 85%- 1 15%).
Exemplary High Shear Wet Granulation Method
[00461] The following exemplary high shear wet granulation method was used to prepare pharmaceutical composition (0.29% wt wt Compound A tosylate salt) suitable for administration to a patient.
[00462] Dissolve 2.530 g HPC (Klucel* EF) in 91.129 g of a 9: 1 (w/w) acetone: w ater mixture, and add 0.735 g Compound A tosylate salt to the mixture while mixing on a magnetic stirrer. The solution is added to a GLATT high shear granulator using 249.74 g PROSOLV* SMCC 50 as the substrate with solution addition speed of 20 g/min and impeller speed of 700 rpm. An additional amount (20 g) of mixture is then added to the granulator. Granules are then dried in a vacuum dryer at a temperature of 75 *C. Dried granules are sieved through a 60 mesh screen to obtain the exemplary pharmaceutical composition.
Exemplary High Shear Wet Granulation Formulations in Capsules
[00463] The following unit dosage forms were prepared in capsules using the XCLEODOSE* automated encapsulator system (Capsugel, Peapack, New Jersey, USA). [00464| 25 μ¾ Capsule. Add 12.5 mg of the high shear wet granulation pharmaceutical composition as described above (0.290% wt wt Compound A tosylate salt) to an appropriate sized hypromellose capsule. The amount of Compound A tosylate salt in the capsule is 36.3 μg, which, using the conversion factor of 1.453 mg Compound A tosylate salt per 1.000 mg of Compound A, provides about 25 Ltg of Compound A.
[00465] 50 μζ Capsule. Add 25 mg of the high shear wet granulation pharmaceutical composition as described above to an appropriate sized hypromellose capsule.
[00466] 250 pg Capsule. Add 125 mg of the high shear wet granulation pharmaceutical composition as described above to an appropriate sized hypromellose capsule.
[00467] 500 II u Capsule. Add 250 mg of the high shear wet granulation pharmaceutical composition as described above to an appropriate sized hypromellose capsule.
[00468] 1 mg Capsule. Add 500 mg of the high shear wet granulation pharmaceutical composition as described above to an appropriate sized hypromellose capsule.
[00469] Randomly selected samples were found to have a mean drug content in a range from 90% to 1 10% and an AV less than 15%. These unit dosage forms meet USP and Ph.Eur. requirements for dosage form content uniformity.
Exemplary Tableting and Coating Method
[00470] This prophetic example provides a preparation of tablets using granules prepared from the exemplary high shear wet granulation method described above.
[00471] Add disintegrant to the dried granules and mix in a suitable blender for 10 min. Add magnesium stearate or sodium stearyl fumarate to the blend and mix for an additional 2 to 5 min to yield the final blend. Transfer the final blend to a suitable tableting machine and compress into tablets with suitable size toolings. Transfer tablets into a tablet coating machine. Under constant rotation and heated air flow, the film coating suspension is continuously sprayed onto the tablet cores using spray nozzle(s) where the solvent evaporates by applying heated air flow in contact with the tablet surface. Continue the process until the desired amount of coating is applied.
[00472] The following are examples of prophetic tablet formulations (E-M) prepared by high shear wet granulation followed by direct compression.
Example E. Composition of 25 pg tablet prepared by
high shear granulation followed by direct compression
%, Amount per
Ingredients w/w tablet (mg)
Compound A tosylate salt 0.29 0.036
SMCC 94.21 1 1.776
Hydroxy-propyl cellulose 1.00 0.125
Crospovidone 4.00 0.500
Magnesium stearate 0.50 0.063
Acetonerwater (9: 1 )* qs
Total 100.00 12.500
Removed by drying Example F. Composition of 50 g tablet prepared by high shear granulation followed by direct compression
%, Amount per
Ingredients w/w tablet (mg)
Compound A tosylate salt 0.29 0.073
SMCC 94.21 23.552
Hydroxypropyl cellulose 1.00 0.250
Crospovidone 4.00 1.000
Magnesium stearate 0.50 0.125
Aeetone:water (9:1)* qs
Total 100.00 25.000
* Removed by drying
Example G. Composition of 500 tablet prepared by high shear granulation followed by direct compression
%, Amount per
Ingredients w/w tablet (mg)
Compound A tosylate salt 0.29 0.727
SMCC 94.21 235.524
Hydroxypropyl cellulose 1.00 2.500
Crospovidone 4.00 10.000
Magnesium stearate 0.50 1.250
Acetone: water (9: 1)* qs
Total 100.00 250.000
* Removed by drying
Example H. Composition of 1 mg tablet prepared high shear granulation followed by direct compression
%, Amount per
Ingredients w/w tablet (mg)
Compound A tosylate salt 0.29 1.453
SMCC 94.21 471.047
Hydroxypropyl cellulose 1.00 5.000
Crospovidone 4.00 20.000
Magnesium stearate 0.50 2.500
Acetone: ater (9: 1)* ¾L
Total 100.00 500.000
* Removed by drying Example J. Composition of 25 ig tablet prepared by high shear granulation followed by direct compression
%, Amount per
Ingredients w/w tablet (mg)
Compound A tosylate salt 0.29 0.036
SMCC 93.21 11.651
Hydroxypropyl cellulose 1.00 0.125
Crospovidone 4.00 0.500
Sodium stearyl tumarate 1.50 0. 188
Acetone: water (9: 1 )* qs —
Total 100.00 12.500
* Removed by drying
Example K. Composition f 50 ig tablet prepared by high shear granulation followed by direct compression
%, Amount per
Ingredients w/w tablet (mg)
Compound A tosylate salt 0.29 0.073
SMCC 93.21 23.303
Hydroxypropyl cellulose 1.00 0.250
Crospovidone 4.00 1.000
Sodium stearyl tumarate 1.50 0.375
Acetone: water (9: 1 )* qs —
Total 100.00 25.000
Removed by drying
Example L. Composition of 500 μ tablet prepared by high shear granulation followed by direct compression
%, Amount per
Ingredients w/w tablet (mg)
Compound A tosylate salt 0.29 0.727
SMCC 93.21 235.025
Hydroxypropyl cellulose 1.00 2.500
Crospovidone 4.00 10.000
Sodium stearyl tumarate 1.50 3.75
Acetone: water (9: 1)* qs
Total 100.00 250.000
* Removed by drying
Example M. Composition of 1 mg tablet prepared
by high shear granulation followed by direct compression
%, Amount per
Ingredients w/w tablet (mg)
Compound A tosylate salt 0.29 1.453
SMCC 93.21 466.05
Hydroxypropyl cellulose 1.00 5.000
Crospovidone 4.00 20.000
Sodium stearyl fumarate 1.50 7.500
Acetone: water (9: 1)* qs —
Total 100.00 500.000
* Removed by drying
Biological Studies
Biochemical PARP 1 Assay
[00473] Inhibitory effects of test compounds against human PARP 1 enzyme were assessed using
Trevigen's Universal Chemiluminescent PARP Assay Kit (Trevigen CAT#4676-096-K) following the manufacturer's recommended protocol . One of ordinary skill in the art would know of alternatives methods with which to measure PARP activity.
[00474] Immediately prior to performing the assay, the fol lowing reagents were prepared: A) 20*
PARP Assay Buffer was diluted to 1 * with dH20; B) I Ox PARP Cocktail, which contains a mixture of NAD and biotinylated NAD, was diluted by the addition of lOx Activated DNA and 1 * PARP Assay Buffer. Both the PARP Cocktai l and Activated DNA are l x after the dilution; C) all test compounds were initially dissolved in DMSO, and subsequently serial diluted with l PARP Assay Buffer; D) recombinant human PARP 1 enzyme was diluted with 1 χ PARP Assay Buffer to generate 0.5 unit/ 15 μΐ; E) I Ox Strep- Diluent was diluted to 1 χ with I PBS/0. 1 % Triton X- 100; F) Just before use, dilute Strep-HRP 500-fold with l Strep-Di luent.
[00475] The chemiluminescent assays for PARP activity were performed in white 96-well plates that are pre-coated with histones. Briefly, strip wells were removed from the wrapper, 50 μΙ/well of I X PARP Buffer was added to rehydrate the histones and incubation was allowed for 30 minutes at room temperature. Removal of the I X PARP Buffer from the wells was accomplished by tapping the strip wells on paper towel . Serial dilutions of the test compounds were added to duplicate wells in 10 μΐ/well volume. Final assay concentrations of test compounds were typically between 1 and 0.0001 μΜ. Subsequently, recombinant human PARP 1 enzyme was added to 0.5 unit of PARP 1 enzyme/well in 1 5 μΐ/well volume. Combined volume of enzyme and inhibitor was 25 μΐ. Incubate the enzyme/inhibitor mixtures for 10 minutes at room temperature. To start the reaction, 25 μΐ/well of the I X PARP Cocktail was added to all the wel ls. Controls included background wells with l x Assay Buffer alone (no PARP) and wells with no inhibitor for determining the maximum or 100% PARP activit value. In all cases the final reaction volume was 50 μΐ. [00476] The reactions were allowed to proceed for 1 hour at room temperature. The plate was then washed 4 times with 200 μΙ/well I X PBS/0.1 % Triton X-100, using EL.xSO Automated Strip Washer (BIO-TEK). After washing, all wells were incubated for 60 minutes with 50 μΐ/well Strep-HRP, diluted 1 :500 with 1 * Strep- Diluent. The plate was washed 4 times with 200 μΐ/well I X PBS/0.1% Triton X- 100 using ELx50 Automated Strip Washer (BIO-TEK). After washing, dry the wells by tapping plate onto paper towels. Mix equal volumes of PeroxyGIow™ A and B together and add 100 μΙ per well. The light output was immediately determined in a plate reader ( En V ision, by Perkin Elmer) set up for measuring chein ilum inescence.
[00477] The % enzyme activity for each compound is then calculated using the following equation:
Act i vi ty Ctrl - X
" ■ x 100%
Act ivi tv Ctrl - Negative Ctrl
[00478] IC50 values (the concentration at which 50% of the enzy me activity is inhibited) of each test compound were calculated using GraphPad Prism 5 software.
[00479] Al l of the compounds tested had or are expected to have enzymatic PARP inhibitory activity. Of the compounds tested, over 100 compounds had a PARP inhibitory activity in the enzymatic assay of less than 50 nM, with approximately 60 of these compounds having an inhibitory activity of less than 5 nM.
Cellular Assays
[00480] A very potent PARP 1/2 selective inhibitor Compound A ((SS^/ -S-fluoro-S-^- fluoropheny l)-9-( 1 -methyl- 1 -U2,4-triazol-5-yl)-8.9-dihvdro-2H-pyridoi4,3,2-de1phthalazin-3(7H)-one PARP1 IC50 = 0.57nM) was used to assess the sensitivity of human multiple myeloma (MM) cell lines, H929 and MM. I S, to PARP inhibition. Myeloma cell line H929 was cultured in RPMI1640 medium containing 10% FBS. O.OSm β-mercaptoethanol, l OOU/ml Penicillin and 100μ /ml Streptomycin; mveloma cel l line MM. I S was cultured in RPMI 1 640 medium containing 10% FBS, l OOU/ml Penicillin and 1 00μ§/ηι1 Streptomycin. All cell lines were maintained in 37°C incubator with 5% C02 till ready for assay. H929 cells were seeded in 96- we 11 plates at 1000 cells/well (for 5, 7 and 10-day assay) or 2500 cells/well (for 5-day assay) in 96- we 11 plates, MM. 1 S cells were seeded in 96-wel l plates at 8000 cells/well (for 7-day assay) or 5000 cells/well (for 10-day assay) in 96-wel l plates. Cel ls were incubated for overnight at 37 °C before treated with their corresponding growth medi containing Compound A in 0.1 % DMSO at various concentrations ranging from 0.025 nM to 2000 nM or as indicated in the figures. 0. 1 % DMSO was used as mock treatment or control. After 5- 10 days incubation, cell survival was measured by Cel lTiter Glo (Promega) and cell survival fraction was calculated relative to control.
GraphPad Prism5 software was used to plot data and calculate 1C50 values.
[00481] In a 7-day cytotoxicity assay, Compound A showed cell killing of 1 1929 and MM.l S (IC50 of 20nM and 45nM, respectively). Prolonged incubation to 10 days with Compound A in H929 and MM. I S cells further decreased IC50s to low nM range, indicating certain MM cells may be highly sensitive to Compound A treatment.

Claims

WHAT IS CLAIMED IS:
1. A method of treating multiple myeloma comprising administering a Compound according to Formula (I):
Figure imgf000152_0001
Formula (I)
wherein:
Y and Z are each independently selected from the group consisting of:
d) an aryl group optionally substituted with 1 , 2, or 3 Re;
e) a heteroaryl group optionally substituted with 1 , 2, or 3 Re;
f) a substituent independently selected from the group consisting of hydrogen, alkenyl, alkoxy, alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, alkyl, alkynyl, arylalkyl, cycloalkyi, cycloalkylalkyl, haloalkyl, hydroxyalkylene, oxo, heterocycloalkyl, heterocycloalkylalkyl, alkylcarbonyl, arylcarbonyl, heteroarylcarbonyl alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, -(CRC6-alkylene)-NRARB, -C(0)(NRARB), -(C, -C6-aIkylene)-C(0)NRARB, -S(0)2(NRARB), and -(CL -C6-alkylene)-S(0)2(NRARB);
Ri, R2, and Rj are each independently selected from the group consisting of hydrogen, halogen, alkenyl, alkoxy, alkoxycarbonyl, alkyl, cycloalkyi, alkynyl, cyano, haloalkoxy, haloalkyl, hydroxyl, hydroxyalkylene, nitro, -NRARB, -(C,-C6-alkylene)-NRARB, and -C(0)(NRARB);
A and B are each independently selected from hydrogen, Br, CI, F, I, OH, C C6alkyl, C3-C3cycloalkyl, alkoxy, alkoxyalkyl wherein CrC6alkyl, C3-C8cycloalkyl, alkoxy, and alkoxyalkyl are optionally substituted with at least one substituent selected from OH, N02, CN, Br, CI, F, I, C C6alkyl, and Cj-Cgcycloalkyl, wherein B is not OH;
R,Y and RB are independently selected from the group consisting of hydrogen, alkyl, cycloalkyi, 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-aIkyl)-, -NC(0)(C i -C6-alky I)-, -N(ary!)-, -N(aryl-C|-C6-alkyl-)-, -N(substituted-aryl-C C6-alkyl-)-, -N(heteroaryl)-, -N(heteroaryl-C C6-alkyI- )-, -N(substituted-heteroaryl-Ci-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, cycloalkyi, alkoxyalkyl, haloalkyl, hydroxyalkylene, and -(CrC6-alkylene)-NRARB;
each R6 is independently selected from OH, N02, CN, Br, CI, F, I, CrC6alkyl, C-Cgcycloalkyl,
C)-C8heterocycloalkyl; C2-C(,alkenyl, alkoxy, alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C2-C6alkynyl, aryl, arylalkyl, C3-C8cycloalkylalkyl, haloalkoxy, haloalkyl, hydroxyalkylene, oxo, heteroaryl, heteroarylalkoxy, heteroaryloxy, heteroarylthio, heteroarylalkylthio, heterocycloaikoxy, Co-Qheterocycloalkylthio, heterocyclooxy, heterocyclothio, -NRARB, -(Ci-C6-alkylene)-NRARB, -C(0)(NRARB), -(CrC6-alkylene)-C(0)NRARB, -S(0):(NRARB ), and -(C,~C6-alkylene)- S(0)2(NRARB);
a single isomer, stereoisomer, or enantiomer, or mixture thereof, optionally as a pharmaceutically
acceptable salt, solvate, hydrate and/or chemically protected form thereof.
2. The method of Claim 1 where the Compound of Formula (I) is according to Formula (II)
Figure imgf000153_0001
Formula (II)
where A and B are hydrogen.
3. The method of Claim 1 or 2 where Y and Z are independently a phenyl group optionally substituted with 1 , 2, or 3 R,,: an imidazole group optionally substituted with 1 , 2, or 3 R/,; or a triazole group optionally substituted with 1 , 2, or 3 Re.
4. The method of Claim 1 , 2, or 3 where Y is an imidazole group optionally substituted with 1 R<¾; and Z is a phenyl group substituted with 1 R<,.
5. The method of Claim 1 , 2, or 3 where Y is a triazole group optionally substituted with 1 R^; and Z is a phenyl group substituted with 1 R<;.
6. The method of Claim 1 , 2, or 3 where where Y and Z are each a phenyl group independently optionally substituted with 1 R,,.
7. The method of Claim 1 , 2, 3, or 4 where Y is imidazole group substituted with C] -C6alkyl .
8. The method of Claim 1 , 2, 3, or 5 where Y is triazole group substituted with Ci-C6alkyl.
9. The method of Claim 1 , 2, 3, or 5 where Y is triazole group substituted with CrC6alkyl and Z is phenyl substituted with halo.
10. The method of Claim 1 , 2, 3, 4, 5, or 6 where Z is phenyl substituted with halo.
1 1 . The method of Claim 1 , 2, 3, 4, 5, 6, 7, 8. 9, or 10 where R2 is hydrogen or halo.
12. The method of any of Claims 1 - 1 1 where the compound, optionally as a single stereoisomer or mixture of stereoisomers thereof and additionally optionally as a pharmaceutically acceptable salt thereof, is selected from
8,9-bis(4-((dimethylamino)methyl)phenyl)-8,9-dihydro-2/ -pyrido[4,3,2-de]phthalazin-3(7/jr)-one;
8-(4-((dimethylamino)methyl)phenyl)-9-phenyl-8,9-dihydro-2/f-pyrido[4,3,2-de]phthalazin-3(7 /)-one;
8- (4-fluorophenyl)-9-( I -methyl- -imidazol-2-yl)-8,9-dihydro-2H-pyrido[4,3,2-de]phthalazin-3(7//)-one;
9- ( l -methyl- lH-imidazol-2-yl)-8-phenyl-8,9-dihydro-2H-pyrido[4,3,2-de]phthalazin-3(7 )-one;
5-fluoro-9-( l -methyl- H-imidazol-2-yl)-8-phenyl-8,9-dihydro-2H-pyrido[4,3,2-de]phthalazin-3(7 ^-one; 5-fluoro-8-(4-fIuorophenyl)-9-( l -methyl- lH-imidazol-2-yl)-8,9-dihydro-2H-pyrido[4,3,2-de]phthalazin- 3(7H -one; ^
Figure imgf000154_0001
Figure imgf000155_0001
Figure imgf000156_0001
Figure imgf000157_0001
Figure imgf000158_0001
Figure imgf000159_0001
( )-one;. ,
13. The method of any of Claims 1 - 1 1 where the compound or a pharmaceutically acceptable salt thereof is selected from:
(8S,9R)-5-fluoro-8-(4-fluorophenyl)-9-( l -m^
.fe]phthaIazin-3(7H)-one;
(8/?, 9¾-5-fluoro-8-(4-fluorophenyl)-9-( l -methyl- lH- l ,2,4-triazol-5-yl)-8,9-dihydro-2//-pyrido[4,3,2- c/_]phthalazin-3(7H)-one;
(85,9/?)- 8-(4-fluorophenyl)-9-( 1 -methyl- lH-itnidazol-2-yl)-8,9-dihydro-2H-pyrido[4,3,2-cfe]phthalazin-
3(7H)-o ;
(8R,9S)- 8-(4-fluorophenyl)-9-( l -methyl- lH-imidazoI-2-yl)-8,9-dihydro-2H-pyrido[4,3,2-c/e]phthalazin- 3(7#)-one;
(85,9/?)- 8-(4-fluorophenyl)-9-( l -methyl- lH- l ,2,4-triazol-5-yl)-8,9-dihydro-2H-pyrido[4,3,2- J.]phthalazin-3(7H)-one;
(8R.95)- 8-(4-fluorophenyl)-9-( 1 -methyl- 1 H- 1 ,2,4-triazol-5-yl)-8,9-dihydro-2H-pyrido[4,3,2- c/_jphthalazin-3(7H)-one;
(85,97?)- 8-(4-(azetidin- l -ylmethyI)phenyI)-9-(4-fluorophenyi)-8,9-dihydro-2H-pyrido[4,3,2- de]phthalazin-3(7H)-one;
(BR, 9S)- 8-(4-(azetidin-l -ylmethyl)phenyl)-9-(4-fluorophenyl 8,9-dihydro-2H-pyrido[4,3,2- de]phthaIazin-3(7H)-one; (8S,9R)-5-fluoro-9-(l-methyl-lH-imidazol-2-yl)-8-ph^
3(lH)-one;
(8R,9S)-5-fluoro-9-(l -methyl- lH-imidazol-2-yl)-8-phen^
3(7 /)-one;
(85,9 )-5-fluoro-9-(l-methyl-lH-l,2,4-tnazo^
3(7H)-one;
(8/?,9.V)-5-tluoro-9-( 1 -methyl- 1//- 1.2.4-tnazol-5-yl)-8 ihenyl-8.9-dihydro-2 -pyrido[4.3.2-i/e]phthalazin- 3(7fl)-one;
(8R,9¾-8-(4-fluorophenyl)-9-(l-methyl-l -imidazol-2-yl)-8,9-dihydro-2H-pyrido[4,3,2-i/e]phthalazin- 3(7H)-one;
(85,9R)-8-(4-fluorophenyl)-9-( 1 -methyl- 1 H-imidazol-2-yl)-8,9-dihydro-2H-pyrido[4 ,2- e]phthalazin- 3(7//)-one;
(8R,9S)-5-fluoro-8-(4-fluorophenyl)-9-(l -methyl- IH-imidazol-2 -yl)-8,9-dihydro-2H~pyrido[4,3,2- t/i?]phthalazin-3(7 )-one;
(8S,9R)-5-fluoro-8-(4-fluorophenyl)-9-(l -methyl- lH-imidazol-2-yl)-8,9-dihydro-2H-pyrido[4,3,2- ife]phthalazin-3(7H)-one;
(8R,9S 8-(4-fluorophenyl)-9-(l -methyl- IH-], 2,4-triazol-5~yI)-8,9-dihydro-2/f-pyrido[4,3,2- ie]phthalazin-3(7H)-one;
(85,9R)-8-(4-fluorophenyl)-9-(l-methyl-lH-K2,4-triazol-5-yI)-8,9-dihydro-2H-pyridot4,3,2- i/ ]phthalazin-3(7/ )-one;
(8R,9¾-8-(4-((dimethylamiiio)methyt)phenyl)-5-fluoro-9-(l-methyl-lH-l,2,4-triazol-5-yl)-8,9-dihydro-
2H-pyrido[4,3,2-£/e]phthalazin-3(7H)-one;
(85',9?)-8-(4-((dimethyIamino)methyl)phenyl)-5-fluoro-9-(l -methyl- lH-l,2,4-triazol-5-yl)-8,9-dihydro- 2 /-pyrido[4,3,2- e]phthalazin-3(7if)-one;
(8R,95)-8-(4-(azetidin-I-ylmethyl)phenyl)-9-(4-fluorophenyI)-8,9-dihydro-2H-pyridof4,3,2- i/e]phthalazin-3(7H)-one; and
(8>S,,9i?)-8-(4-(azetidin-l-ylmethyl)phenyl)-9-(4-fluorophenyl)-8,9-dihydro-2H-pyrido[4,3,2- i/f|phthalazin-3(7/ )-one.
14. The method of any of Claims 1-3, 5, and 8-13 where the compound is (8S,9/i)-5-tluoro-8-(4- fluorophenyl)-9-(l-methyl-lH-l,2,4-triazol-5-y^
15. The method of any of Claims 1-14 where the pharmaceutically acceptable salt is tosylate.
16. A method of treating multiple myeloma comprising administering a pharmaceutical composition comprising a compound of any of Claims 1-15 and a pharmaceutically acceptable carrier(s), excipient(s), binder(s) or diluent(s) thereof.
17. The method of Claim 16 where the compound is in solid form and where the weight/weight percent of the Compound as a tosylate salt in the pharmaceutical composition is between about 0.01 % to about 4.000 %.
18. The method of Claim 16 or 17 where one or more pharmaceutically acceptable excipients comprise silicified microcrystaliine cellulose.
19. The method of Claim 16, 17, or 18 where the one or more pharmaceutically acceptable excipients consists essentially of silicified microcrystaliine cellulose.
20. The method of Claim 16, 17, or 18 where the one or more pharmaceutically acceptable excipients consist essentially of silicified microcrystaliine cellulose, hydroxypropyl cellulose, crospovidone and magnesium stearate.
21. The method of any of Claims 16-20 where the weight/weight percent of the compound as a tosylate salt in the pharmaceutical composition is between about about 0.1 % to about 0.8 %.
22. The method of any of Claims 16-21 where the pharmaceutical composition is encapsulated in a capsule shell.
23. The method of any of Claims 16-22 where the pharmaceutical composition comprises a mass of about 25 μ§, about 50 jtg. about 250 μg or about 1 mg of the Compound as a tosylate salt excluding the tosylate portion of the salt.
24. The method of any of Claims 16-2 1 where the pharmaceutical composition is a tablet.
25. A method of treating multiple myeloma by administering a compound of any of Claims 1 - 15 or administering a pharmaceutical composition according to any of Claims 16-24 sequentially or simultaneously in combination with a proteasome inhibitor.
26. The method of Claim 25 where the proteasome inhibitor is bortezomib.
27. A method of treating multiple myeloma by administering a compound of any of Claims 1 - 15 or administering a pharmaceutical composition according to any of Claims 16-24 sequentially or simultaneously in combination with a DNA-damaging agent.
28. The method of Claim 27 where the DNA-damaging agent is melphalan, cyclophosphamide, bendamustine, or radiation.
29. A pharmaceutical composition comprising Compound A tosylate salt in solid form and one or more pharmaceutically acceptable excipients wherein the weight/weight percent of Compound A tosylate salt in the pharmaceutical composition is between about 0.01 % to about 5.000 %.
30. The pharmaceutical composition of Claim 29, wherein the one or more pharmaceutically acceptable excipients comprise silicified microcrystaliine cellulose.
3 1. The pharmaceutical composition of Claim 29 or 30, wherein the one or more pharmaceutically acceptable excipients consists essentially of silicified microcrystaliine cellulose.
3 1 . The pharmaceutical composition of Claim 29, 30, or 31 , wherein the one or more
pharmaceutically acceptable excipients consist essentially of silicified microcry staliine cellulose, hydroxypropyl cellulose, crospovidone and magnesium stearate or sodium stearyl turn a rate.
32. The pharmaceutical composition of any of Claims 29-3 1 , wherein the Compound A tosylate salt is in a crystalline solid form exhibiting at least one of a solid state "C NMR spectrum with peaks at 143.2, 136.0, 13 1.8, 123.9, 1 12.2, 105.2, and 100.3 ppm ± 0.2 ppm;
a differential scanning calorimetry thermogram having an endotherm with a maximum at between about 320 °C and about 335 °C:
a thermogravimetric analysis thermogram indicative of an unsolvated material;
a dynamic vapor sorption isotherm plot which does not exhibit a significant weight change from 0 to 95% relative humidity;
an X-ray powder diffraction pattern comprising characteristic peaks expressed in d-values (A) of about 1 1.9, 5.9, 4.9, 4.4, 4.3, 3.9, and 3.7; and
an X-ray powder diffraction pattern comprising peaks at 2Θ angle degrees ± 0.2 2Θ angle degrees of 7.4, 15.1, 1 8.1 , 20.1 , 20.4, 22.6, and 24.0.
33. The pharmaceutical composition of Claim 32, wherein the Compound A tosylate salt is in a crystalline solid form exhibiting at least one of
a solid state 13C NMR spectrum with peaks at 143.2, 136.0, 13 1.8, 123.9, 1 12.2, 105.2, and 100.3 ppm ± 0.2 ppm;
an X-ray powder diffraction pattern comprising characteristic peaks expressed in d-values (A) of about 1 1.9, 5.9, 4.9, 4.4, 4.3, 3.9, and 3.7; and
an X-ray powder diffraction pattern comprising peaks at 2Θ angle degrees ± 0.2 20 angle degrees of 7.4, 15.1 , 18.1 , 20.1 , 20.4, 22.6, and 24.0.
34. The pharmaceutical composition of Claim 33, where the Compound A tosylate salt is in a crystalline form exhibiting an X-Ray Powder Diffraction pattern comprising characteristic peaks expressed in d-values (A) of about 1 1 .9, 5.9, 4.9, 4.4, 4.3, 3.9, and 3.7.
35. The pharmaceutical composition of Claim 33, where the Compound A tosylate salt is in a crystalline form exhibiting an X-Ray Powder Diffraction pattern comprising characteristic peaks expressed in 2Θ angle degrees ± 0.2 2Θ angle degrees of 7.4, 15.1 , 18.1 , 20. 1 , 20.4, 22.6, and 24.0.
36. The pharmaceutical composition of Claim 33, where the Compound A tosylate salt is in a crystalline form exhibiting a solid state l3C NMR spectrum with peaks at 143.2, 136.0, 131.8, 123.9, 1 12.2, 105.2, and 100.3 ppm ± 0.2 ppm.
37. The pharmaceutical composition of any of Claims 29-36, wherein the weight/weight percent of Compound A tosylate salt in the pharmaceutical composition is between about about 0. 1 % to about 0.8 %.
38. A pharmaceutical composition comprising Compound A tosylate salt and silicified
microcrystalline cellulose, wherein the pharmaceutical composition is prepared using crystalline Compound A tosylate salt.
39. A unit dosage form comprising pharmaceutical composition, the pharmaceutical composition comprising Compound A tosylate salt in solid form and one or more pharmaceutically acceptable excipients, wherein the weight/weight percent of Compound A tosylate salt in the pharmaceutical composition is between about 0.01 % to about 3.000 %.
40. The unit dosage form of Claim 39, wherein the pharmaceutical composition is encapsulated in a capsule shell.
41. The unit dosage form of Claim 39 or 40, wherein the pharmaceutical composition comprises a mass of about 25 μg, about 50 ^ig, about 250 μg or about 1 mg Compound A tosylate salt excluding the tosylate portion of the salt.
42. The unit dosage form of Claim 39, wherein the unit dosage form is a tablet.
43. The unit dosage form of Claim 42, further comprising a tablet coating.
44. A dry blend method for preparing a pharmaceutical composition comprising:
a) mixing crystalline Compound A tosylate salt and a first fraction of silicified microcrystalline cellulose to produce a first mixture;
b) mixing the first mixture and a second fraction of silicified microcrystalline cellulose; and c) mixing the mixture produced in step (b) and a third fraction of silicified microcrystalline cellulose to produce a pharmaceutical composition comprising a weight/weight percent of Compound A tosylate salt between about 0.01 % to about 5.000 %.
45. The method of Claim 44, wherein the mixture produced in step (b) is geometrically blended with microcrystalline cellulose prior to being mixed in step (c).
46. A pharmaceutical composition produced by method according to Claim 44 or 45.
47. A wet granulation method for preparing a pharmaceutical composition comprising:
a) dissolving crystalline Compound A tosylate salt and a binder in a first volume of solvent; b) mixing the solution of step (a) in a high shear granulator with silicified microcrystalline cellulose; and
c) drying the granules to produce a to produce a pharmaceutical composition comprising between about 0.01 % to about 5.000 % Compound A tosylate salt and about 0.5 % to about 2.0 % binder.
48. The wet granulation method of Claim 47, wherein the first volume comprises about 95 % to about 98 % (weight/weight) of the solution of step (a).
49. The wet granulation method of Claim 47 or 48, wherein the solvent is acetone, ethanol, ethyl acetate or water or a co-solvent of acetone/water, ethanol/water or ethyl acetate/water.
50. The wet granulation method of Claim 47, 48, or 49, wherein the solvent is a co-solvent of acetone and water, wherein the weight ratio of acetone to water is 9: 1.
5 1. The wet granulation method of Claim 47, 48, 49, or 50, wherein the binder is hypromel lose or HPC.
52. A pharmaceutical composition produced by method according to Claim 47, 48, 49, 50 or 51.
53. A method of treating a cancer or symptom thereof in a patient comprising administering to a cancer patient a pharmaceutical composition or unit dosage form according to any one of Claims 29-43, 46, and 52 wherein the amount of Compound A tosylate salt administered to the cancer patient per day is between about 1 μ§ to about 8 mg in a dosage regiment effective to treat the cancer or symptom thereof.
54. The method of Claim 53, wherein the cancer is selected from bladder cancer, breast cancer, cervical cancer, colon cancer, colorectal cancer, acute myelogenous leukemia, mantle cell lymphoma, chronic lymphocytic leukemia, Burkitt's lymphoma, nasopharyngeal carcinoma, EBV+ gastric cancer, endometrial cancer, gastrointestinal stromal tumor, glioblastoma, head and neck cancer, hepatocellular carcinoma, kidney cancer, leukemia, lung cancer, lymphoma, medulloblastoma, melanoma, meningioma, ovarian cancer, pancreatic cancer, prostate cancer, renal cancer, small cell lung carcinoma, thyroid cancer, multiple myeloma, endometrial cancer, peritoneal cancer, or uterine cancer.
55. The method of Claim 53 or 54 where the cancer is breast cancer, prostate cancer, ovarian cancer, colon cancer, colorectal cancer or pancreatic cancer.
56. The method of Claim 53 or 54 where the cancer is multiple myeloma.
PCT/US2012/051297 2011-08-19 2012-08-17 Dihydropyridophthalazinone inhibitors of poly (adp-ribose) polymerase (parp) for the treatment of multiple myeloma WO2013028495A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201161525645P 2011-08-19 2011-08-19
US61/525,645 2011-08-19
US201161558719P 2011-11-11 2011-11-11
US61/558,719 2011-11-11

Publications (1)

Publication Number Publication Date
WO2013028495A1 true WO2013028495A1 (en) 2013-02-28

Family

ID=47746777

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2012/051297 WO2013028495A1 (en) 2011-08-19 2012-08-17 Dihydropyridophthalazinone inhibitors of poly (adp-ribose) polymerase (parp) for the treatment of multiple myeloma

Country Status (1)

Country Link
WO (1) WO2013028495A1 (en)

Cited By (16)

* 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
WO2014164749A1 (en) 2013-03-13 2014-10-09 Forma Therapeutics, Inc. Novel compounds and compositions for inhibition of fasn
JP2017523243A (en) * 2014-07-31 2017-08-17 メディヴェイション テクノロジーズ, インコーポレーテッド (2S, 3S) -Methyl 7-fluoro-2- (4-fluorophenyl) -3- (1-methyl-1H-1,2,4-triazol-5-yl) -4-oxo-1,2, Co-former salts of 3,4-tetrahydroquinoline-5-carboxylate and methods for producing them
US9820985B2 (en) 2008-08-06 2017-11-21 Medivation Technologies Llc Dihydropyridophthalazinone inhibitors of poly(ADP-ribose)polymerase (PARP)
WO2018153279A1 (en) * 2017-02-25 2018-08-30 Suzhou Kintor Pharmaceuticals, Inc. Dihydropyridophthalazinone compounds as inhibitors of poly (adp-ribose) polymerase (parp) for treatment of diseases and method of use thereof
WO2018205938A1 (en) * 2017-05-08 2018-11-15 广州丹康医药生物有限公司 Parp inhibitor, pharmaceutical composition, preparation method and use 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
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
US10793554B2 (en) 2018-10-29 2020-10-06 Forma Therapeutics, Inc. Solid forms of 4-(2-fluoro-4-(1-methyl-1H-benzo[d]imidazol-5-yl)benzoyl)piperazin-1-yl)(1-hydroxycyclopropyl)methanone
CN111868060A (en) * 2018-03-14 2020-10-30 上海艾力斯医药科技股份有限公司 Dihydropyridophthalazinone derivative, preparation method and application thereof
US10875848B2 (en) 2018-10-10 2020-12-29 Forma Therapeutics, Inc. Inhibiting fatty acid synthase (FASN)
JP2021523186A (en) * 2018-05-14 2021-09-02 ニューベイション・バイオ・インコーポレイテッドNuvation Bio Inc. Anti-cancer nuclear hormone receptor targeting compound
US11826430B2 (en) 2019-05-14 2023-11-28 Nuvation Bio Inc. Anti-cancer nuclear hormone receptor-targeting compounds
US11834458B2 (en) 2021-03-23 2023-12-05 Nuvation Bio Inc. Anti-cancer nuclear hormone receptor-targeting compounds
WO2023239789A1 (en) * 2022-06-08 2023-12-14 Valo Health, Inc. Tricyclic phthalazinone parp inhibitors and methods of use
US11952349B2 (en) 2019-11-13 2024-04-09 Nuvation Bio Inc. Anti-cancer nuclear hormone receptor-targeting compounds

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080058325A1 (en) * 2003-05-28 2008-03-06 Mgi Gp, Inc. Compounds, methods and pharmaceutical compositions for inhibiting parp
US20090098084A1 (en) * 2007-10-03 2009-04-16 Mgi Gp, Inc. Parp inhibitor compounds, compositions and methods of use
US20100035883A1 (en) * 2008-08-06 2010-02-11 Lead Therapeutics, Inc. Dihydropyridophthalazinone inhibitors of poly(adp-ribose)polymerase (parp)
US20110190288A1 (en) * 2010-02-03 2011-08-04 Daniel Chu Dihydropyridophthalazinone inhibitors of poly(adp-ribose)polymerase (parp) for use in treatment of diseases associated with a pten deficiency
US20120129865A1 (en) * 2010-10-21 2012-05-24 Biomarin Pharmaceutical Inc. 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

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080058325A1 (en) * 2003-05-28 2008-03-06 Mgi Gp, Inc. Compounds, methods and pharmaceutical compositions for inhibiting parp
US20090098084A1 (en) * 2007-10-03 2009-04-16 Mgi Gp, Inc. Parp inhibitor compounds, compositions and methods of use
US20100035883A1 (en) * 2008-08-06 2010-02-11 Lead Therapeutics, Inc. Dihydropyridophthalazinone inhibitors of poly(adp-ribose)polymerase (parp)
US8012976B2 (en) * 2008-08-06 2011-09-06 Biomarin Pharmaceutical Inc. Dihydropyridophthalazinone inhibitors of poly(ADP-ribose)polymerase (PARP)
US20110237581A1 (en) * 2008-08-06 2011-09-29 Bing Wang Dihydropyridophthalazinone inhibitors of poly(adp-ribose)polymerase (parp)
US20110190288A1 (en) * 2010-02-03 2011-08-04 Daniel Chu Dihydropyridophthalazinone inhibitors of poly(adp-ribose)polymerase (parp) for use in treatment of diseases associated with a pten deficiency
US20120129865A1 (en) * 2010-10-21 2012-05-24 Biomarin Pharmaceutical Inc. 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

Cited By (41)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11364241B2 (en) 2008-08-06 2022-06-21 Medivation Technologies Llc Dihydropyridophthalazinone inhibitors of poly(ADP-ribose)polymerase (PARP)
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)
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
US9926303B2 (en) 2010-02-08 2018-03-27 Medivation Technologies Llc Processes of synthesizing dihydropyridophthalazinone derivatives
US8765945B2 (en) 2010-02-08 2014-07-01 Biomarin Pharmaceutical Inc. 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
US10800750B2 (en) 2013-03-13 2020-10-13 Forma Therapeutics, Inc. Compounds and compositions for inhibition of FASN
US10995078B2 (en) 2013-03-13 2021-05-04 Forma Therapeutics, Inc. Compounds and compositions for inhibition of FASN
KR20150143479A (en) * 2013-03-13 2015-12-23 포르마 세라퓨틱스 인크. Novel compounds and compositions for inhibition of fasn
WO2014164749A1 (en) 2013-03-13 2014-10-09 Forma Therapeutics, Inc. Novel compounds and compositions for inhibition of fasn
RU2686323C2 (en) * 2013-03-13 2019-04-25 Форма Терапьютикс, Инк. Novel compounds and compositions for inhibition of fasn
US10399951B2 (en) 2013-03-13 2019-09-03 Forma Therapeutics, Inc. Compounds and compositions for inhibition of FASN
US10450286B2 (en) 2013-03-13 2019-10-22 Forma Therapeutics, Inc. Compounds and compositions for inhibition of FASN
JP2019048833A (en) * 2013-03-13 2019-03-28 フォーマ セラピューティクス,インコーポレイテッド Novel compound and composition for inhibiting fasn
US10457655B2 (en) 2013-03-13 2019-10-29 Forma Therapeutics, Inc. Compounds and compositions for inhibition of FASN
US10472342B2 (en) 2013-03-13 2019-11-12 Forma Therapeutics, Inc. Compounds and compositions for inhibition of FASN
EP2968316A4 (en) * 2013-03-13 2016-09-21 Forma Therapeutics Inc Novel compounds and compositions for inhibition of fasn
KR102194745B1 (en) 2013-03-13 2020-12-24 포르마 세라퓨틱스 인크. Novel compounds and compositions for inhibition of fasn
JP2016517412A (en) * 2013-03-13 2016-06-16 フォーマ セラピューティクス,インコーポレイテッド Novel compounds and compositions for inhibiting FASN
RU2737434C2 (en) * 2013-03-13 2020-11-30 Форма Терапьютикс, Инк. Novel compounds and fasn inhibition compositions
JP2017523243A (en) * 2014-07-31 2017-08-17 メディヴェイション テクノロジーズ, インコーポレーテッド (2S, 3S) -Methyl 7-fluoro-2- (4-fluorophenyl) -3- (1-methyl-1H-1,2,4-triazol-5-yl) -4-oxo-1,2, Co-former salts of 3,4-tetrahydroquinoline-5-carboxylate and methods for producing them
JP2020125303A (en) * 2014-07-31 2020-08-20 メディヴェイション テクノロジーズ, エルエルシー Coformer salts of (2s,3s)-methyl 7-fluoro-2-(4-fluorophenyl)-3-(1-methyl-1h-1,2,4-triazol-5-yl)-4-oxo-1,2,3,4-tetrahydroquinoline-5-carboxylate and methods of preparing them
CN110382468A (en) * 2017-02-25 2019-10-25 苏州开拓药业股份有限公司 Dihydro pyrido benzodiazine ketone compound and application thereof with poly- (ADP- ribose) polymerase (PARP) inhibitory activity
CN110382468B (en) * 2017-02-25 2023-03-24 苏州开拓药业股份有限公司 Dihydropyridophthalazinone compounds having poly (ADP-ribose) polymerase (PARP) inhibitory activity and uses thereof
WO2018153279A1 (en) * 2017-02-25 2018-08-30 Suzhou Kintor Pharmaceuticals, Inc. Dihydropyridophthalazinone compounds as inhibitors of poly (adp-ribose) polymerase (parp) for treatment of diseases and method of use thereof
WO2018205938A1 (en) * 2017-05-08 2018-11-15 广州丹康医药生物有限公司 Parp inhibitor, pharmaceutical composition, preparation method and use thereof
CN110650961A (en) * 2017-05-08 2020-01-03 广州丹康医药生物有限公司 PARP inhibitor, pharmaceutical composition, preparation method and application thereof
CN111868060B (en) * 2018-03-14 2023-06-09 上海艾力斯医药科技股份有限公司 Dihydropyridino phthalazinone derivatives, preparation method and application thereof
CN111868060A (en) * 2018-03-14 2020-10-30 上海艾力斯医药科技股份有限公司 Dihydropyridophthalazinone derivative, preparation method and application thereof
EP3793608A4 (en) * 2018-05-14 2022-04-20 Nuvation Bio Inc. Anti-cancer nuclear hormone receptor-targeting compounds
JP2021523186A (en) * 2018-05-14 2021-09-02 ニューベイション・バイオ・インコーポレイテッドNuvation Bio Inc. Anti-cancer nuclear hormone receptor targeting compound
US11299484B2 (en) 2018-10-10 2022-04-12 Forma Therapeutics, Inc. Inhibiting fatty acid synthase (FASN)
US10875848B2 (en) 2018-10-10 2020-12-29 Forma Therapeutics, Inc. Inhibiting fatty acid synthase (FASN)
US11267805B2 (en) 2018-10-29 2022-03-08 Forma Therapeutics, Inc. Solid forms of (4-(2-fluoro-4-(1-methyl-1H-benzo[d]imidazol-5-yl)benzoyl) piperazine-1-yl)(1-hydroxycyclopropyl)methanone
US10793554B2 (en) 2018-10-29 2020-10-06 Forma Therapeutics, Inc. Solid forms of 4-(2-fluoro-4-(1-methyl-1H-benzo[d]imidazol-5-yl)benzoyl)piperazin-1-yl)(1-hydroxycyclopropyl)methanone
US11826430B2 (en) 2019-05-14 2023-11-28 Nuvation Bio Inc. Anti-cancer nuclear hormone receptor-targeting compounds
US11952349B2 (en) 2019-11-13 2024-04-09 Nuvation Bio Inc. Anti-cancer nuclear hormone receptor-targeting compounds
US11834458B2 (en) 2021-03-23 2023-12-05 Nuvation Bio Inc. Anti-cancer nuclear hormone receptor-targeting compounds
WO2023239789A1 (en) * 2022-06-08 2023-12-14 Valo Health, Inc. Tricyclic phthalazinone parp inhibitors and methods of use

Similar Documents

Publication Publication Date Title
WO2013028495A1 (en) Dihydropyridophthalazinone inhibitors of poly (adp-ribose) polymerase (parp) for the treatment of multiple myeloma
JP6946194B2 (en) Solid form of compounds that regulate kinases
TWI448464B (en) Dihydropyridophthalazinone inhibitors of poly(adp-ribose)polymerase (parp)
CA2787844C (en) Dihydropyridophthalazinone inhibitors of poly(adp-ribose) polymerase (parp) for use in treatment of diseases associated with a pten deficiency
TW201011024A (en) Compounds and methods for kinase modulation, and indications therefor
KR20220049518A (en) Pyrazolo[3,4-B]pyrazine SHP2 phosphatase inhibitor
JP6689856B2 (en) 6,7-Dihydropyrazolo [1,5-a] pyrazin-4 (5H) -one compounds and their use as negative allosteric modulators of the MGLUR2 receptor
JP5897566B2 (en) Cyclic N, N&#39;-diarylthiourea and N, N&#39;-diarylurea-androgen receptor antagonists, anticancer agents, methods for their preparation and uses
HUE029275T2 (en) Phthalazinone ketone derivative, preparation method thereof, and pharmaceutical use thereof
WO2011130661A1 (en) Methods of using dihydropyridophthalazinone inhibitors of poly (adp-ribose)polymerase (parp)
TW200900396A (en) Phthalazinone derivatives
US20130053365A1 (en) Dihydropyridophthalazinone inhibitors of poly(adp-ribose)polymerase (parp)
WO2018234978A1 (en) Substituted 5-cyanoindole compounds and uses thereof
TW200936140A (en) Substituted arylamide oxazepinopyrimidone derivatives
JP2023071922A (en) 7h-pyrrolo[2,3-d]pyrimidine jak inhibitor
TWI815820B (en) Solid forms of 2-(5-(4-(2-morpholinoethoxy)phenyl)pyridin-2-yl)-n-benzylacetamide
WO2021060453A1 (en) Crosslinked optically active secondary amine derivative
WO2023078451A1 (en) Compound used as cdk7 kinase inhibitor and use thereof
WO2011095057A1 (en) Benzimidazole derivatives and pharmaceutical compositions and uses thereof
JP2017529356A (en) Crystal forms of tyrosine kinase inhibitors and salts thereof
US20110190266A1 (en) 5,6,6a,7,8,9-HEXAHYDRO-2H-PYRIDOPHTHALAZINONE INHIBITORS OF POLY(ADP-RIBOSE)POLYMERASE (PARP)
US20220098201A1 (en) The monohydrate of rogaratinib hydrochloride and solid states thereof
ES2650797T3 (en) Crystal forms of (S) -4-amino-N- (1- (4-chlorophenyl) -3-hydroxypropyl) -1- (7H-pyrrolo [2,3-d] pyrimidin-4-yl) piperidine-4- carboxamide
KR20170095243A (en) Heterocyclyl linked imidazopyridazine derivatives as pi3kbeta inhibitors
WO2023239846A1 (en) Heterocyclic compounds as pi3kα inhibitors

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: 12825335

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: 12825335

Country of ref document: EP

Kind code of ref document: A1