CA2806324A1 - Phthalazinone ketone derivative, preparation method thereof, and pharmaceutical use thereof - Google Patents

Phthalazinone ketone derivative, preparation method thereof, and pharmaceutical use thereof Download PDF

Info

Publication number
CA2806324A1
CA2806324A1 CA2806324A CA2806324A CA2806324A1 CA 2806324 A1 CA2806324 A1 CA 2806324A1 CA 2806324 A CA2806324 A CA 2806324A CA 2806324 A CA2806324 A CA 2806324A CA 2806324 A1 CA2806324 A1 CA 2806324A1
Authority
CA
Canada
Prior art keywords
group
compounds
formula
pyrazine
pharmaceutically acceptable
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
CA2806324A
Other languages
French (fr)
Other versions
CA2806324C (en
Inventor
Pengcho Tang
Xin Li
Xiangqin Li
Yang Chen
Bin Wang
Zhe ZHU
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Jiangsu Hengrui Medicine Co Ltd
Original Assignee
Jiangsu Hansoh Pharmaceutical Co Ltd
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 Jiangsu Hansoh Pharmaceutical Co Ltd filed Critical Jiangsu Hansoh Pharmaceutical Co Ltd
Publication of CA2806324A1 publication Critical patent/CA2806324A1/en
Application granted granted Critical
Publication of CA2806324C publication Critical patent/CA2806324C/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/50Pyridazines; Hydrogenated pyridazines
    • A61K31/502Pyridazines; Hydrogenated pyridazines ortho- or peri-condensed with carbocyclic ring systems, e.g. cinnoline, phthalazine
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41881,3-Diazoles condensed with other heterocyclic ring systems, e.g. biotin, sorbinil
    • 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
    • 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
    • A61K31/4985Pyrazines or piperazines ortho- or peri-condensed with heterocyclic ring systems
    • 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Epidemiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

A phthalazinone ketone derivative as represented by formula (I), a preparation method thereof, a pharmaceutical composition containing the derivative, a use thereof as a poly (ADP-ribose) polymerase (PARP) inhibitor, and a cancer treatment method thereof.
(see formula I)

Description

PHTHALAZINONE KETONE DERIVATIVE, PREPARATION

METHOD THEREOF, AND PHARMACEUTICAL USE THEREOF

FIELD OF THE INVENTION

The present invention relates to a novel phthalazinone ketone derivative as represented by formula (I), the preparation methods thereof, the pharmaceutical composition containing the derivative, and the use thereof as a therapeutic agent, especially as the poly (ADP-ribose) polymerase (PARP) inhibitor.

BACKGROUND OF THE INVENTION

The chemotherapy and radiation therapy are two common methods to treat cancer.
Both treatments can induce single-stranded and/or double-stranded DNA break to produce cytotoxicity, then the targeted tumor cells will die due to chromosomal damage.
An important result in response to DNA damage signal is the signal of the cell cycle in regulation site is activated, the purpose of which is to protect cells from mitosis in the case of DNA damage thereby preventing cell damage. In most cases, the tumor cells exhibit the defects of regulation signal in the cell cycle and have high proliferation rate.
So it can be predicated that the tumor cells have specific DNA repair mechanisms, which can respond quickly to repair chromosome damage relevant to proliferation regulation, thereby saving them from cytotoxic effects of some treatment and keep alive.
In the clinical application, the effective concentration of the chemotherapeutical drug or therapeutic radiation intensity can fight these DNA repair mechanism to ensure the killing effect on the target tumor cells. However, the tumor cells can develop tolerance for treatment by enhancing its DNA damage repair mechanisms, and survive from the lethal DNA damage. In order to overcome the tolerance, it is usually necessary to increase the dosage of the therapeutic drug or radiation intensity. This approach will produce adverse effects on the normal tissue nearby the lesions, and then make the treatment course complicated by severe adverse reactions, thereby increasing the risk of treatment. At the same time, the ever-increasing tolerance will reduce the therapeutic effect, so it can be concluded that the cytotoxic of the DNA damage agents can be improved in the way of tumor cell-specificity by controling the repair mechanism promoted by the siginal of DNA damage.
PARPs (Poly (ADP-ribose) polymerases), characterized by poly ADP-ribosylation activity, are constituted by the superfamily of 18 nucleus enzymes and cytoplasmic enzymes. Such poly ADP-ribosylation effect can adjust the activity of the targeted protein and the interaction between proteins, and regulate other many fundamental biological processes, including DNA repair and cell death. In addition, genomic stability is also associated with the poly ADP-ribosylation (see D' Amours et al.
Biochem. J, 1999, 342, 249).

The activity of PARP-1 accounts for about 80% of the total cellular PARP
activity.
PARP-1, together with PARP-2, which is most similar to PARP-1, are the members having the DNA damage repair capacity in the PARP family. As a sensor and a signaling protein of DNA damage, PARP-1 can detect the DNA damage sites quickly and bond to them directly, and then induce the aggregation of various proteins required for DNA
repair, thereby enabling the DNA damage to be repaired. When the cells lack PARP-1, PARP-2 can realize the repair of the DNA damage instead of PARP-1.
Studies have shown that, compared with normal cells, the expression of PARPs protein in solid tumors is generally enhanced. In addition, the tumors (such as breast cancer and ovarian cancer), whose DNA repair related gene is missing (such as or BRCA-2), show extremely sensitive to PARP-1 inhibitors. This suggests the potential uses of PARP inhibitors as a single agent in the treatment of a tumor, which can be called triple negative breast cancer (see Plummer, E. R. Curr. Opin.
Pharmacol. 2006, 6, 364; Ratnam, et al; Clin. Cancer Res. 2007, 13, 1383). At the same time, because DNA
damage repair mechanism is the main mechanism of tumor cells response to the tolerance produced by chemotherapeutic drugs and ionizing radiation treatment, PARP-1 is considered to be an effective target to explore the new methods of cancer therapy.
PARP inhibitors are early developed and designed using nicotinamide of NAD-f, which can be used as PARP catalytic substrate, as a template to develop its analogs. As competitive inhibitors of NAD+, these inhibitors compete with NAD+ for PARP
catalytic sites, thereby preventing the synthesis of the poly (ADP-ribose) chain. PARP
without poly (ADP-ribosylation) modification can't be dissociated from the DNA

damage sites, which will lead other proteins involved in the repair into the damage site, thereby preventing performance of the repair process. Therefore, in the effect of the cytotoxic drugs or radiation, PARP inhibitor will eventually kill tumor cells with DNA
damage.
In addition, the NAD+, which is consumed as the PARP catalytic substrate, is the essential factor in the ATP synthesis process of the cells. Under the high level of PARP
activity, intracellular NAD+ levels will significantly decrease, thereby affecting the intracellular ATP level. Due to lack of intracellular ATP content, the cells can't achieve programmed ATP-dependent cell death process, and can only turn to necrosis, a special apoptosis process. During the necrosis, a lot of inflammatory cytokines will be released, thereby producing toxic effects on other organs and tissues (Horvath EM et al . Drug News Perspect, 2007, 20, 171-181). Therefore, PARP inhibitors can also be used for the treatment of a variety of diseases related to this mechanism, including neurodegenerative diseases (such as Alzheimer's disease, Huntington's disease, Parkinson's disease), diabetes, concurrent diseases in the ischemia or ischemia-reperfusion process, such as myocardial infarction and acute renal failure, circulatory system diseases, such as septic shock and inflammatory diseases, such as chronic rheumatism, etc (see Tentori L, et al. Pharmacol. Res., 2002, 45, 73-85; Horvath
2 =

EM et al. Drug News Perspect., 2007, 20, 171.; Faro R, et al. Ann. Thorac.
Surg., 2002, 73, 575.; Kumaran D, et al. Brain Res., 2008,192, 178.) Currently, a series of patent application have been disclosed on phthalazinone ketone PARP inhibitor, including W02002036576, W02004080976 and W02006021801.
Although there are a series of PARP inhibitors for tumor treatment have been disclosed, it's still need to develop new compounds with better efficacy and pharmacokinetics results. After continuous efforts, the present invention designs a series of compounds of formula (I), and finds that the compound having such structure exhibit an excellent effect and function.
SUMMARY OF THE INVENTION
The present invention is directed to provide phthalazinone ketone derivatives of formula (I) and tautomers, enantiomer, diastereomers, racemates, and pharmaceutically acceptable salts thereof, as well as their metabolites, metabolic precursor or prodrugs thereof:
cA I Nr0 R3 R2 ( I ) wherein:
A and B are taken together with the attached carbon atoms to form cycloalkyl, heterocyclyl, aryl or heteroaryl, wherein said cycloalkyl, heterocyclyl, aryl or heteroaryl is each independently and optionally substituted with one or more groups selected from the group consisting of alkyl, halogen, hydroxyl, alkoxyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -C(0)0R5, -0C(0)R5, -0(CH2),C(0)0R5, -C(0)R5, -NHC(0)R5, -NR6R7, -0C(0)NR6R7 and -C(0)NR6R7;
RI, R2, R3 or R4 is each independently selected from the group consisting of hydrogen, halogen, alkyl, cyano and alkoxyl, wherein said alkyl or alkoxyl is each independently and optionally substituted with one or more groups selected from the group consisting of halogen, hydroxyl, alkyl and alkoxyl;
D, E, or G is each independently selected from the group consisting of nitrogen atom and C(R8);
R5 is selected from the group consisting of hydrogen, alkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein said alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl is each independently and optionally substituted with one or more groups selected from the group consisting of alkyl, halogen, hydroxyl, alkoxyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, carboxyl and alkoxycarbonyl;
3 R6 or R7 is each independently selected from the group consisting of hydrogen, alkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein said alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl is each independently and optionally substituted with one or more groups selected from the group consisting of alkyl, halogen, hydroxyl, alkoxyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, carboxyl and alkoxycarbonyl;
or, R6 and R7 are taken together with the attached N atom to form heterocyclyl, wherein said heterocyclyl contains one or more N, 0 or S(0),õ heteratoms, and said heterocyclyl is optionally substituted with one or more groups selected from the group consisting of alkyl, halogen, hydroxyl, alkoxyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, carboxyl and alkoxycarbonyl;
R8 is selected from the group consisting of hydrogen, alkyl, halogen, hydroxyl, cyano, alkoxyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, benzyl, -C(0)0R5, -0C(0)R5, -0(CH2)11C(0)0R5, -(CH2)õNR6R7, -C(0)R5, -NHC(0)R5, -NR6R7, -0C(0)NR6R7 and -C(0)NR6R7, wherein said alkyl, alkoxyl, cycloalkyl, heterocyclyl, aryl, heteroaryl or benzyl is each independently and optionally substituted with one or more groups selected from the group consisting of alkyl, halogen, hydroxyl, alkoxyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, oxo, -C(0)0R5, -0C(0)R5, -0(CH2)õC(0)0R5, -C(0)R5, -NHC(0 )R5, -NR6R7, -0C(0)NR6R7 and -C(0)NR6R7;
m is selected from the group consisting of 0, 1 and 2; and n is selected from the group consisting of 0, 1 and 2.
A preferable embodiment of the invention, a compound of formula (I) or its pharmaceutically acceptable salt, wherein A and B are taken together with the attached carbon atoms to form aryl, preferably said aryl is phenyl.
Preferably, the compound of formula (I) or its pharmaceutically acceptable salt, wherein R' is hydrogen.
Preferably, the compound of formula (I) or its pharmaceutically acceptable salt, wherein RI is halogen, preferably fluorine atom.
Preferably, the compound of formula (I) or its pharmaceutically acceptable salt, wherein RI is halogen, preferably fluorine atom.
Preferably, the compound of formula (I) or its pharmaceutically acceptable salt, wherein RI, R2, R3 or R4 is each independently selected from hydrogen atom.
Preferably, the compound of formula (I) or its pharmaceutically acceptable salt, wherein R8 is selected from the group consisting of hydrogen, alkyl, halogen, cyano, -C(0)0R5, -(CH2)õNR6R7 and -C(0)NR6R7, wherein said alkyl is optionally substituted with one or more halogen atoms.
Preferably, the compound of formula (I) or its pharmaceutically acceptable salt, wherein R8 is trifluoromethyl.
The compound of formula (I) may contain asymmetric carbon atoms, therefore it can exist in the form of optically pure diastereomer, diastereomeric mixture, diastereomeric racemate, a mixture of diastereomeric racemate or as a meso-compound.
The present invention includes all these forms. Diastereomeric mixture, diastereomeric
4 racemate or the mixture of diastereomeric racemate can be isolated by conventional methods, such as column chromatography, thin layer chromatography and high performance liquid chromatography.

The equivalent can be understood by an ordinary person skilled in the art that the compound of formula (I) may also have tautomers. The tautomeric forms of the compound (I) include, but not limited to the structure represented by the following formula (II):
OH

rA N
I
N

R3 (110 Ri ( II ) The compounds of the invention include, but not limited to the following:

Example Structure and Name No.

,)JH

4[[4-fluoro-343-(trifluoromethyl)-6,8-dihydro-5H41,2,4]triazolo[4,3-a]
pyrazine-7-carbonyl]-phenyl]methy1]-2H-phthalazin-1-one ,r1H

F

44[3-(3,4-dihydro-1H-pyrrolo[1,2-a]pyrazine-2-carbony1)-4-fluoro-phenyl] methyl] -2H-phthal azin- 1 -one N

methyl 7- [2-fluoro-5 - [(4-oxo-3H-phthalazin- 1 -yl)methyl]benzoy1]-3 -(trifluoromethyl)-6,8-dihydro-5H-imidazo[1,5-a]pyrazine-l-carboxylate
5 . CA 02806324 2013-01-21 0 ril " o al yr,,Ni F

4-[[3-(6,8-dihydro-5H-imidazo[1,2-a]pyrazine-7-carbony1)-4-fluoro-phenyl]methyl]-2H-phthalazin-1-one NH

le" 0
6 5 .. F 2\1...N

F

4-[[4-fluoro-3-[3-(trifluoromethyl)-6,8-dihydro-5H-imidazo[1,5-cdpyrazine
-7-carbonyl]phenylimethy1]-2H-phthalazin-1-one 10 ,N'" 0 OH

aii y N IN

F

4-[[4-fluoro-3-[1-(hydroxymethyl)-3-(trifluoromethyl)-6,8-dihydro-5H-imidazo[1,5-c]pyrazine-7-carbonyl]phenylimethyl]-2H-phthalazin-1-one NH
1.1 0 0 H

F

N-ethy1-7-[2-fluoro-5-[(4-oxo-3H-phthalazin-1-y1)methyl]benzoy11-3-(trifluoromethyl)-6,8-dihydro-5H-imidazo[1,5-a]pyrazine-1-carboxamide 0 0.--OH

F L"---N-i
8 F4-F
F

7-[2-fluoro-5-[(4-oxo-3H-phthalazin-1-yl)methyl]benzoyl]-3-(trifluoromethyl)-6,8-dihydro-5H-imidazo[1,5-c]pyrazine-1-carboxylic acid NH
40 , ri o rli/
9 N-i .1111P. F
F -F
F

4-[[4-fluoro-3-[1-(methylaminomethyl)-3-(trifluoromethyl)-6,8-dihydro-5H-imidazo[1,5-a]pyrazine-7-carbonyliphenyl]methyl]-2H-phthalazin-1-0 ne NH

N
F
FF

7-[2-fluoro-5-[(4-oxo-3H-phthalazin-1-yl)methyl]benzoyl]-3-(trifluoromethyl)-6,8-dihydro-5H-imidazo[1,5-c]pyrazine-1-carboxamide r\rijF1 Br 4- [[3-[1-bromo-3-(trifluoromethyl)-6,8-dihydro-5H-imidazo [1,5-a]
pyrazine-7-carbonyl]-4-fluoro-phenyl]methy1]-2H-phthalazin-1-one NH

o /FF
12 F \F

44[4-fluoro-342-(trifluoromethyl)-6,8-dihydro-5H-imidazo[1,2-a]pyrazine -7-carbonyl]phenyl]methy1]-2H-phthalazin-1-one NH
= 0 0 /
y N

F F

7- [2-fluoro-5- [(4-oxo-3H-phthalazin-1-yOmethyl]benzoy1]-N-methyl-3-(trifluoromethyl)-6,8-dihydro-5H-imidazo[1,5-a]pyrazine-1-carboxamide NH

. . CA 02806324 2013-01-21 ethyl 7-[2-fluoro-5-[(4-oxo-3H-phthalazin-1-yOmethyl]benzoyl]-6,8-dihydro-5H-imidazo[1,2-c]pyrazine-3-carboxylate o NH

* 0 FF
F

44[343-(trifluoromethyl)-6,8-dihydro-5H41,2,4]triazolo[4,3-c]pyrazine-7-carbonyl]phenyllmethyl]-2H-phthalazin-1-one NH
S" 0 16 0 F1......,õ-N- N

44[3-(6,8-dihydro-5H41,2,4]triazolo[1,5-a]pyrazine-7-carbony1)-4-fluoro-phenyl]methy11-2H-phthalazin-1-one o NH

* 0 17 a N Th--%N=N
FL---.!/

41[3-(6,8-dihydro-5H41,2,4]triazolo[4,3-alpyrazine-7-carbony1)-4-fluoro-phenyl]methyl]-2H-phthalazin-1-one o 1101 ,r F F F

44[4-fluoro-341-(pyrrolidine-1-carbony1)-3-(trifluoromethyl)-6,8-dihydro-5H-imidazo[1,5-c]pyrazine-7-carbonyl]phenyllmethyl]-2H-phthalazin-1-one o NH

19 00 F l'\1--1\11 \F

4[[4-fluoro-342-(trifluoromethyl)-6,8-dihydro-5H41,2,41triazolo[1,5-a]
pyrazine-7-carbonyl]phenyl]methy1]-2H-phthalazin-1-one 0 r'0 Ni-%-tNN\----j 4"--F
F F

4-[[4-fluoro-3-[1-(morpholine-4-carbony1)-3-(trifluoromethyl)-6,8-dihydro-5H-imidazo [1,5-c]pyrazine-7-carbonyl]phenyl]methy1]-2H-phthalazin-1-one 1.1 o N

FX-F

N-methyl-7- [3- [(4-oxo-3H-phthalazin-1-yl)methyl]benzoyl] -3 -(trifluoromethyl)-6,8-dihydro-5H-imidazo [1,5-c]pyrazine-l-carboxamide o /
o F

7- [2-fluoro-5- [(4-oxo-3H-phthalazin-1-yl)methyl]benzoyl]-N,N-dimethyl-3-(trifluoromethyl)-6,8-dihydro-5H-imidazo[1,5-a]pyrazine-1-carboxamide NH

N%-r\I=N

F

4[[343-(difluoromethyl)-6,8-dihydro-5H41,2,4]triazolo [4,3 -c]pyrazine-7-carbony1]-4-fluoro-phenyl]methy11-2H-phthalazin-1-one NH

r:1 0 r-4 NH

N

F

A"--F
F F

N-(cyclopropylmethyl)-7- [2-fluoro-5- [(4-oxo-3H-phthalazin-1-yOmethyl]

benzoy1]-3-(trifluoromethyl)-6,8-dihydro-5H-imidazo[1,5-a]pyrazine-1-carboxamide NH

F

FA-F

7- [2-fluoro -5 - [(4-oxo-3H-phthalazin-1-yOmethyl]benzoyl] -3-(trifluoromethyl)-6,8-dihydro-5H-imidazo [1,5 -alpyrazine-1 -c arbonitrile =NH 0 26 =NF

[[4-fluoro-343 -(2,2,2-trifluoroethyl)-6,8-dihydro-5H-[1,2 ,4]triazolo [4,3 -a]pyrazine-7-carbonyliphenyl]methy11-2H-phthalazin-1 -one or pharmaceutically acceptable salts thereof.

This invention relates to a preparation process of the compound of formula (I) or a pharmaceutically acceptable salt thereof, comprising the steps of:

(¨A
NH
I I
N

Ra HN

( IA ) (IB) optionally hydrolyzing the compound of formula (IA) to a carboxylic acid, reacting the carboxylic acid with the compound of formula (TB) or a salt thereof in the presence of a condensing agent such as benzotriazole-N, N, N', N'-tetramethyl urea hexafluorophosphate under alkaline condition to obtain the compound of formula (I);

wherein:

Ra is selected from the group consisting of hydroxyl, halogen and alkoxyl;

A, B, D, E, G and R' to R4 are defined as those in formula (I).

In another aspect, this present invention relates to the use of the compounds of formula (I) or the pharmaceutically acceptable salt thereof in the preparation of the PARP inhibitors.

In another aspect, this present invention relates to a method for inhibiting PARP, comprising administrating the subject in need thereof a therapeutically effective amount of the compounds of formula (I) or the pharmaceutically acceptable salt thereof.

In another aspect, this present invention relates to the use of the compounds of
10 formula (I) or the pharmaceutically acceptable salt thereof in the preparation of an adjuvant in the treatment of cancer or a medicament causing tumor cells sensitive to ionizing radiation or chemotherapy.
In another aspect, this present invention relates to the compounds of formula (I) or the pharmaceutically acceptable salt thereof, for use as an adjuvant in the treatment of cancer or causing tumor cells sensitive to ionizing radiation or chemotherapy.
In another aspect, this present invention relates to the compounds of formula (I) or the pharmaceutically acceptable salt thereof, for use as PARP inhibitors.
In another aspect, this present invention relates to the use of the compounds of formula (I) or the pharmaceutically acceptable salt thereof in the preparation of a medicament for the treatment of cancer, wherein said cancer is selected from the group consisting of breast cancer, ovarian cancer, pancreatic cancer, prostate cancer, liver cancer and colon cancer, wherein said medicament is further co-administered with a therapeutically effective amount of drug selected from the group consisting of Temozolomide, Adriamycin, Taxol, Cisplatin, Carboplatin, Dacarbazine, Topotecan, Irinotecan, Gemcitabine and Bevacizumab.
In another aspect, this present invention relates to the methods for treating cancer, comprising administrating the subject in need thereof a therapeutically effective amount of the compounds of formula (I) or the pharmaceutically acceptable salt thereof, wherein said cancer is selected from the group consisting of breast cancer, ovarian cancer, pancreatic cancer, prostate cancer, liver cancer and colon cancer, wherein said compound of formula (I) or the pharmaceutically acceptable salt thereof is further co-administered with a therapeutically effective amount of drug selected from the group consisting of Temozolomide, Adriamycin, Taxol, Cisplatin, Carboplatin, Dacarbazine, Topotecan, Irinotecan, Gemcitabine and Bevacizumab.
In another aspect, this present invention relates to the compounds of formula (I) or the pharmaceutically acceptable salt thereof, for use as a medicament for the treatment of cancer, wherein said cancer is selected from the group consisting of breast cancer, ovarian cancer, pancreatic cancer, prostate cancer, liver cancer and colon cancer, wherein said medicament is further co-administered with a therapeutically effective amount of drug selected from the group consisting of Temozolomide, Adriamycin, Taxol, Cisplatin, Carboplatin, Dacarbazine, Topotecan, Irinotecan, Gemcitabine and Bevacizumab.
Furthermore, the present invention also relates to a pharmaceutical composition, comprising a therapeutically effective amount of the compounds of formula (I) or the pharmaceutically acceptable salt thereof according to the present invention, and the pharmaceutically acceptable carrier or excipient. And the present invention relates to the said pharmaceutical composition, for use as a PARP inhibitor, or as an adjuvant in the treatment of cancer or a medicament causing tumor cells sensitive to ionizing radiation or chemotherapy, or as a medicament for the treatment of cancer. And the present invention relates to the use of the said pharmaceutical composition in the preparation of
11 a PARP inhibitor. And the present invention relates to the use of the said pharmaceutical composition in the preparation of an adjuvant in the treatment of cancer or a medicament causing tumor cells sensitive to ionizing radiation or chemotherapy. And the present invention relates to the use of the said pharmaceutical composition in the preparation of a medicament for the treatment of cancer, wherein said cancer is selected from the group consisting of breast cancer, ovarian cancer, pancreatic cancer, prostate cancer, liver cancer and colon cancer, wherein said pharmaceutical composition is further co-administered with the therapeutically effective amount of drug selected from the group consisting of Temozolomide, Adriamycin, Taxol, Cisplatin, Carboplatin, Dacarbazine, Topotecan, Irinotecan, Gemcitabine and Bevacizumab.

DETAILED DESCRIPTION OF THE INVENTION

Unless otherwise stated, the terms used in the specification and claims have the meanings described below.
"Alkyl" refers to a saturated aliphatic hydrocarbon group including C 1 -C20 straight chain and branched chain groups. Preferably an alkyl group is an alkyl having 1 to 12 carbon atoms. Representative examples include, but are not limite d to methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-p entyl, 1, 1- dime thyl propyl, 1,2- dime thyl propyl, 2,2- dimethyl propyl, 1-ethyl propy 1, 2-methylbutyl, 3 -methy lbutyl, n-hexyl, 1 -ethyl-2 -methy lpropyl, 1,1,2 -trimethy lpro pyl, 1,1-dimethylbutyl, 1,2-dimethylbutyl, 2,2-dimethylbutyl, 1,3-dimethylbutyl, 2-et hylbutyl. 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2,3-dimethylbutyl, n-hept yl, 2-methylhexyl, 3-methylhexyl, 4-methylhexyl, 5-methylhexyl, 2,3-dimethylpenty 1, 2,4-dimethylpentyl, 2,2-dimethylpentyl, 3,3-dimethylpentyl, 2-ethylpentyl, 3-ethyl pentyl, n-octyl, 2,3-dimethylhexyl, 2,4-dimethylhexyl, 2,5-dimethylhexyl, 2,2-dimet hylhexyl, 3,3-dimethylhexyl, 4,4-dimethylhexyl, 2-ethylhexyl, 3-ethylhexyl, 4-ethylh exyl, 2-methyl-2-ethylpentyl, 2-methyl-3-ethylpentyl, n-nonyl, 2-methyl-2-ethylhexyl, 2-methyl-3-ethylhexyl, 2,2-diethylpentyl, n-decyl, 3,3-diethylhexyl, 2,2-diethylhexy 1, and the isomers of branched chain thereof More preferably an alkyl group is a lower alkyl having 1 to 6 carbon atoms. Representative examples includes, but are not limited to methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1,1-dimethylpropyl, 1,2-dimethylpropyl, 2,2-dimethylpropyl, 1-ethy lpropyl, 2-methylbutyl, 3 -methy lb utyl, n-hexyl, 1 -ethyl-2 -methy lpropyl, 1 ,1,2-tri methylpropyl, 1,1-dimethylbutyl, 1,2-dimethylbutyl, 2,2-dimethylbutyl, 1,3-dimethyl butyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2,3-dimethylbu tyl and etc. The alkyl group may be substituted or unsubstituted. When substitut ed, the substituent group(s) may be substituted at any available connection point, preferably the substituent group(s) is one or more groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkyxoyl, alkylsulfo, alkyla mino, halogen, thiol, hydroxyl, nitro, cyano, cycloalkyl, heterocyclic alkyl, aryl, h eteroaryl, cycloalkyoxyl, heterocylic alkyoxyl, cycloalkylthio, heterocylic alkylthio,
12 OXO, -C(0)0R5, -0C(0)R5, -0(CH2)riC(0)0R5, -C(0)R5, -NHC(0)R5, -NR6R7, -0 C(0)NR6R7 and -C(0)NR6R7.

"Cycloalkyl" refers to saturated and/or partially unsaturated monocyclic or po lycyclic hydrocarbon group and have 3 to 20 carbon atoms, preferably 3 to 12 c arbon atoms, more preferably 3 to 10 carbon atoms. Representative examples of monocyclic cycloalkyl include, but are not limited to cyclopropyl, cyclobutyl, cyc lopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cy cloheptatrienyl, cyclooctyl and etc. Polycyclic cycloalkyl includes the cycloalkyl h aving spiro ring, fused ring and bridged ring.

"Spiro Cycloalkyl" refers to 5 to 20 membered polycyclic group with rings connected through one common carbon atom (called as spiro atom), wherein on e or more rings may contain one or more double bonds, but none of the rings has a completely conjugated pi-electron system. Preferably a spiro cycloalkyl is 6 to 14 membered, more preferably 7 to 10 membered. According to the number of the common spiro atom, spiro cycloalkyl is divided into mono-spirocyclic rin g, di-spirocyclic ring or poly-spirocyclic ring, preferably refers to mono-spirocycli c ring or di-spirocyclic ring. More preferably spiro cycloalkyl is 4-membered/4-m embered, 4-membered/5-membered, 4-membered/6-membered, 5-membered/5-memb ered, or 5-membered/6-membered monocyclic spiro ring. Representative examples of spiro cycloalkyl include, but are not limited to the following groups:

LIFI(Czand "Fused Cycloalkyl" refers to 5 to 20 membered polycyclic hydrocarbon gro ups, wherein each ring in the system shares an adjacent pair of carbon atoms wi th other ring, wherein one or more rings may contain one or more double bonds, but none of the rings has a completely conjugated pi-electron system.
Preferably a fused cycloalkyl group is 6 to 14 membered, more preferably 7 to 10 membe red. According to the number of membered ring, fused cycloalkyl is divided into bicyclic ring, tricyclic ring, tetracyclic ring or polycyclic ring fused cycloalkyl, preferably refers to fused bicyclic ring or tricyclic ring fused cycloalkyl.
More pr eferably fused cycloalkyl is 5-membered/5-membered, or 5-membered/6-membered bicyclic ring fused cycloalkyl. Representative examples of fused cycloalkyl inclu de, but are not limited to the following groups:

it and.

"Bridged Cycloalkyl" refers to 5 to 20 membered polycyclic hydrocarbon group, wherein every two rings in the system share with two disconnected carbon atoms. The
13 = CA 02806324 2013-01-21 said rings could have one or more double bonds but have no completely conjugated pi-electron system. Preferably a bridged cycloalkyl is 6 to 14 membered, more preferably 7 to 10 membered. According to the number of membered ring, bridged cycloalkyl is divided into bridged bicyclic ring, tricyclic ring, tetracyclic ring or polycyclic ring, preferably refers to bicyclic ring, tricyclic ring or tetracyclic ring bridged cycloalkyl, more preferably bicyclic ring or tricyclic ring bridged cycloalkyl.
Representative examples of bridged cycloalkyl include, but are not limited to the following groups:

A 4,, IV* and 1;1-t-The said cycloalkyl can be fused to the ring of aryl, heteroaryl or heterocyclic alkyl, wherein the ring connected with parent structure is cycloalkyl. Representative examples include, but are not limited to indanylacetic, tetrahydronaphthalene, benzocydoheptyl and so on. The said cycloalkyl may be optionally substituted or unsubstituted.
When substituted, the substituent group(s) is preferably one or more groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkyxoyl, alkylsulfo, alkylamino, halogen, thiol, hydroxyl, nitro, cyano, cycloalkyl, heterocyclic alkyl, aryl, heteroaryl, cycloalkyoxyl, heterocylic alkyoxyl, cycloalkylthio, heterocylic alkylthio, oxo, -C(0)0R5, -0C(0)R5, -0(CH2)õC(0)0R5, -C(0)R5, -NHC(0)R5, -NR6R7, -0C(0)NR6R7 and -C(0)NR6R7.
"Alkenyl" refers to an alkyl defined as above that have at least two carbon atoms and at least one carbon-carbon double bond. For example, vinyl, 1-propenyl, 2-propenyl, 1-, 2- or 3-butenyl and etc. The alkenyl group may be substituted or unsubstituted. When substituted, the substituent group(s) is preferably one or more group(s) independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkyxoyl, alkylsulfo, alkylamino, halogen, thiol, hydroxyl, nitro, cyano, cycloalkyl, heterocyclic alkyl, aryl, heteroaryl, cycloalkyoxyl, heterocylic alkyoxyl, cycloalkylthio, heterocylic alkylthio, -C(0)0R5, -0C(0)R5, -0(CH2)C(0)0R5, -C(0)R5, -NHC(0)R5, -NR6R7, -0C(0)NR6R7 and -C(0)NR6R7.
"Alkynyl" refers to an alkyl defined as above that have at least two carbon atoms and at least one carbon-carbon triple bond. For example, ethynyl, 1-propynyl, 2-propynyl, 1-, 2- or 3-butynyl and etc. The alkynyl group may be substituted or unsubstituted. When substituted, the substituent group(s) is preferably one or more group(s) independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkyxoyl, alkylsulfo, alkylamino, halogen, thiol, hydroxyl, nitro, cyano, cycloalkyl, heterocyclic alkyl, aryl, heteroaryl, cycloalkyoxyl, heterocylic alkyoxyl, cycloalkylthio,
14 heterocylic alkylthio, -C(0)0R5, -0C(0)R5, -0(CH2)C(0)0R5, -C(0)R5, -NHC(0)R5, -NR6R7, -0C(0)NR6R7 and -C(0)NR6R7.
"Heterocycly1" refers to 3 to 20 membered saturated and/or partially unsaturated monocyclic or polycyclic hydrocarbon group having one or more heteroatoms selected from the group consisting of N, 0, or S(0)m (wherein m is 0, 1 or 2) as ring atoms, but excluding -0-0-, -0-S- or -S-S- in the ring, the remaining ring atoms being C.

Preferably, heterocyclyl is 3 to 12 membered having 1 to 4 said heteroatoms;
more preferably 3 to 10 membered. Representative examples of monocyclic heterocyclyl include, but are not limited to pyrrolidyl, piperidyl, piperazinyl, morpholinyl, sulfo-morpholinyl, homopiperazinyl and so on. Polycyclic heterocyclyl includes the heterocyclyl having spiro ring, fused ring and bridged ring. "Spiro heterocyclyl" refers to 5 to 20 membered polycyclic heterocyclyl with rings connected through one common carbon atom (called as spiro atom), wherein said rings have one or more heteroatoms selected from the group consisting of N, 0, and S(0)p (wherein p is 0,1 or 2) as ring atoms, the remaining ring atoms being C, wherein one or more rings may contain one or more double bonds, but none of the rings has a completely conjugated pi-electron system. Preferably an spiro heterocyclyl is 6 to 14 membered, more preferably 7 to 10 membered. According to the number of common spiro atom, spiro heterocyclyl is divided into mono-spiro heterocyclyl, di-sipro heterocyclyl or poly-spiro heterocyclyl, preferably refers to mono-spiro heterocyclyl and di-sipro heterocyclyl. More preferably spiro heterocyclyl is 4-membered/4-membered, 4-membered/5-membered, 4-membered/6-membered, 5-membered/5-membered, or 5-membered/6-membered mono-spiro heterocyclyl. Representative examples of spiro heterocyclyl include, but are not limited to the following groups:

¨0-271 0 0 S 01¨ and N
"Fused I leterocycly1" refers to 5 to 20 membered polycyclic heterocyclyl group, wherein each ring in the system shares an adjacent pair of carbon atoms with other ring, wherein one or more rings may contain one or more double bonds, but none of the rings has a completely conjugated pi-electron system, and wherein said rings have one or more heteroatoms selected from the group consisting of N, 0, and S(0)p (wherein p is 0, 1 or 2) as ring atoms, the remaining ring atoms being C. Preferably an fused heterocyclyl is 6 to 14 membered, more preferably 7 to 10 membered. According to the number of membered ring, fused heterocyclyl is divided into bicyclic ring, tricyclic ring, tetracyclic ring or polycyclic ring fused heterocyclyl, preferably refers to bicyclic ring or tricyclic ring fused heterocyclyl. More preferably fused heterocyclyl is 5-membered/5-membered, or 5-membered/6-membered bicyclic ring fused heterocyclyl.
Representative examples of fused heterocyclyl include, but are not limited to the
15 following groups:

p a CcriN'14 Ni34 and "Bridged Heterocycly1" refers to 5 to 14 membered polycyclic heterocyclic alkyl group, wherein every two rings in the system share with two disconnected atoms, the said rings could have one or more double bonds but have no completely conjugated pi-electron system, and the said rings have one or more heteroatoms selected from the group consisting of N, 0, and S (0),, (wherein m is 0, 1 or 2) as ring atoms, the remaining ring atoms being C. Preferably an bridged heterocyclyl is 6 to 14 membered, more preferably 7 to 10 membered. According to the number of membered ring, bridged heterocyclyl is divided into bicyclic ring, tricyclic ring, tetracyclic ring or polycyclic ring bridged heterocyclyl, preferably refers to bicyclic ring, tricyclic ring or tetracyclic ring bridged heterocyclyl, more preferably bicyclic ring or tricyclic ring bridged heterocyclyl. Representative examples of bridged heterocyclyl include, but are not limited to the following groups:
rCy )17-, -i(AAN43' 7:L\N 4vvl, and 1\d--2).
The said ring of heterocyclyl can be fused to the ring of aryl, heteroaryl or cycloalkyl, wherein the ring connected with parent structure is heterocyclyl.
Representative examples include, but are not limited to the following groups:

N and \ S , etc.

The heterocyclyl may be optionally substituted or unsubstituted. When substituted, the substituent group(s) is preferably one or more group(s) independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkyxoyl, alkylsulfo, alkylamino, halogen, thiol, hydroxyl, nitro, cyano, cycloalkyl, heterocyclic alkyl, aryl, heteroaryl, cycloalkyoxyl, heterocylic alkyoxyl, cycloalkylthio, heterocylic alkylthio, oxo, -C(0)0R5, -0C(0)R5, -0(CH2)C(0)0R5, -C(0)R5, -NHC(0)R5, -NR6R7, -0C(0)NR6R7 and -C(0)NR6R7.
"Aryl" refers to refers to a 6 to 14 membered all-carbon monocyclic ring or a polycyclic fused ring (a "fused" ring system means that each ring in the system shares an adjacent pair of carbon atoms with other ring in the system) group, and has a
16 = CA 02806324 2013-01-21 completely conjugated pi-electron system. Preferably aryl is 6 to 10 membered, such as phenyl and naphthyl. The said aryl can be fused to the ring of heteroaryl, heterocyclyl or cycloalkyl, wherein the ring connected with parent structure is aryl.
Representative examples include, but are not limited to the following groups:
1111 0 NõN io oN (r1 = 0 0 0 uwo 0 N,N\ 110 N S 0 0 and The aryl group may be substituted or unsubstituted. When substituted, the substituent group(s) is preferably one or more groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkyxoyl, alkylsulfo, alkylamino, halogen, thiol, hydroxyl, nitro, cyano, cycloalkyl, heterocyclic alkyl, aryl, heteroaryl, cycloalkyoxyl, heterocylic alkyoxyl, cycloalkylthio, heterocylic alkylthio, -C(0)0R5, -0C(0)R5, -0(CH2)11C(0)0R5, -C(0)R5, -NHC(0)R5, -NR6R7, -0C(0)NR6R7 and -C(0)NR6R7.
"Heteroaryl" refers to a heteroaryl system having 1 to 4 heteroatoms selected from the group consisting of 0, S and N as ring atoms and having 5 to 14 annular atoms.
Preferably heteroaryl is 5- to 10- membered. More preferably heteroaryl is 5-or 6-membered. The examples of heteroaryl groups include furyl, thienyl, pyridyl, pyrrolyl, N-alkyl pyrrolyl, pyrimidinyl, pyrazinyl, imidazolyl, tetrazolyl, and the like. The said heteroaryl can be fused with the ring of aryl, heterocyclyl or cycloalkyl, wherein the ring connected with parent structure is heteroaryl. Representative examples include, but are not limited to the following groups, N

N NH
NO S and The heteroaryl group may be substituted or unsubstituted. When substituted, the substituent group(s) is preferably one or more groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkyxoyl, alkylsulfo, alkylamino, halogen, thiol, hydroxyl, nitro, cyano, cycloalkyl, heterocyclic alkyl, aryl, heteroaryl, cycloalkyoxyl, heterocylic alkyoxyl, cycloalkylthio, heterocylic alkylthio, -C(0)0R5, -0C(0)R5, -0(CH2),C(0)0R5, -C(0)R5, -NHC(0)R5, -NR6R7, -0C(0)NR6R7 and -C(0)NR6R7.
"Alkoxyl" refers to both an -0-(alkyl) and an -0-(unsubstituted cycloalkyl) group, wherein the alkyl is defined as above. Representative examples include, but are not limited to, methoxy, ethoxy, propoxy, butoxy, cyclopropyloxy, cyclobutyloxy, cyclopentyloxy, cyclohexyloxy, and the like. The alkoxyl may be optionally substituted
17 or unsubstituted. When substituted, the substituent is preferably one or more groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkyxoyl, alkylsulfo, alkylamino, halogen, thiol, hydroxyl, nitro, cyano, cycloalkyl, heterocyclic alkyl, aryl, heteroaryl, cycloalkyoxyl, heterocylic alkyoxyl, cycloalkylthio, heterocylic alkylthio, -C(0)0R5, -0C(0)R5, -0(CH2)C(0)0R5, -C(0)R5, -NHC(0)R5, -NR6R7, -0C(0)NR6R7 and -C(0)NR6R7.
"Hydroxy" refers to an -OH group.
"Halogen" refers to fluoro, chloro, bromo or iodo atom.
"Amino" refers to a -NH2 group.
"Cyano" refers to a -CN group.
"Nitro" refers to a -NO2 group.
"Benzyl" refers to a -CH2-(penyl) group.
"Oxo" refers to an =0 group.
"Carboxyl" refers to a -C(0)0H group.
"Alkoxycarbonyl" refers to a -C(0)0(alkyl) or (cycloalkyl) group, wherein the alkyl and cycloalkyl are defined as above.
"Optional" or "optionally" means that the event or circumstance described subsequently may, but not need to occur, and the description includes the instances of the event or circumstance may or may not occur. For example, "the heterocyclic group optionally substituted by an alkyl" means that an alkyl group may be, but not need to be present, and the description includes the case of the heterocyclic group being substituted with an alkyl and the heterocyclic group being not substituted with an alkyl.
"Substituted" refers to one or more hydrogen atoms in the group, preferably up to 5, more preferably 1 to 3 hydrogen atoms independently substituted with a corresponding number of substituents. It goes without saying that the substituents exist in their only possible chemical position. The person skilled in the art are able to determine the substitution is possible or impossible without paying excessive efforts by experiment or theory. For example, the combination of amino or hydroxyl group having free hydrogen and carbon atoms having unsaturated bonds (such as olefinic) may be unstable.
A "pharmaceutical composition" refers to a mixture of one or more of the compounds described in the present invention or physiologically/pharmaceutically acceptable salts or prodrugs thereof and other chemical components such as physiologically/pharmaceutically acceptable carriers and excipients. The purpose of a pharmaceutical composition is to facilitate administration of a compound to an organism, which is conducive to the absorption of the active ingredient and thus displaying biologically activity.
m, n and R5 to R7 are defined as those in the compounds of formula (I).

SYNTHESIS METHOD OF THE COMPOUND IN THE PRESENT INVENTION
In order to complete the purpose of the invention, the present invention applies the
18 following technical solution:
A preparation method of the compounds of formula (I) of the invention or pharmaceutically acceptable salt thereof, comprising the steps of:

cA NH

Ra HN
R3 Ri R2 N¨G
( IA ) (IB) optionally hydrolyzing the compound of formula (IA) to a carboxylic acid, then reacting the carboxylic acid with the compound of formula (IB) or salt thereof in the presence of a condensing reagent such as benzotriazole-N, N, N', N'-tetramethyl urea hexafluorophosphate under alkaline condition to obtain the compound of formula (I);
wherein:
Ra is selected from hydroxyl, halogen or alkoxyl;
A, B, D, E, G and RI to R4 are defined as those in the formula (I).
The above condensation reaction is carried out between an acid compound and an amine compound in the presence of a condensing agent under basic condition, wherein the condensing agent is selected from the group consisting of N, N'-dicyclohexylcarbodiimide, N, N'-Diisopropylcarbodiimide and 0-(benzotriazol-1-y1)-N, N, N', N'-tetramethyluronium tetrafluoroborate (TBTU), preferably 0-(benzotriazol-1-y1)-N, N, N', N'-tetramethyluronium tetrafluoroborate (TBTU); alkaline condition is provided by an organic or inorganic base, wherein the organic base is selected from the group consisting of diisopropyl ethylamine, pyridine, triethylamine, hexahydropyridine, N-methyl-piperazine, 4-dimethylamino pyridine, etc., preferably diisopropyl ethylamine; wherein the solvent used is selected from the group consisting of toluene, benzene, dichloromethane, tetrahydrofuran, chloroform, N, N-dimethyl formamide, or the mixture of the solvents above, preferably N, N-dimethyl formamide; the reaction temperature is controlled between -80 C and 100 C, preferably between 0 C and 60 C; the reaction time is usually controlled between I
minute and 72 hours, preferably between 15 minutes and 24 hours.

PREFERRED EMBODIMENTS

The following examples serve to illustrate the invention, but the examples should not be considered as limiting the scope of the invention.
Examples The compound's structure was indentified by NMR and/or MS. NMR chemical shifts (8) were given in le(ppm). NMR is determined by a Bruker AVANCE-400 machine. The solvents were deuterated-dimethyl sulfoxide (DMSO-d6),
19 deuterated-chloroform (CDC13) and deuterated-methanol (CD30D) with tetramethylsilane (TMS) as an internal standard.
MS was determined by a FINNIGAN LCQAd (ESI) mass spectrometer (manufacturer: Thermo, type: Finnigan LCQ advantage MAX).
HPLC was determined on an Agilent 1200DAD high pressure liquid chromatography spectrometer (Sunfire C18 150 x4.6 mm chromatographic column) and a Waters 2695-2996 high pressure liquid chromatography spectrometer (Gimini 150 x 4.6 mm chromatographic column).
IC50 was determined by a NovoStar ELIASA (BMG Co., German);
The thin-layer silica gel used Yantai Huanghai HSGF254 or Qingdao GF254 silica gel plate. The dimension of the plates used in TLC was 0.15 mm to 0.2 mm, and the dimension of the plates used in thin-layer chromatography for product purification was 0.4 mm to 0.5 mm.
Column chromatography generally used Yantai Huanghai 200 to 300 mesh silica gel as carrier.
The known starting material of the invention can be prepared by the conventional synthesis method in the prior art, or be purchased from ABCR GmbH & Co. KG, Acros Organics, Aldrich Chemical Company, Accela ChemBio Inc or Dan i chemical Company, etc.
Unless otherwise stated in the examples, the following reactions were placed under argon atmosphere or nitrogen atmosphere.
The term "argon atmosphere" or "nitrogen atmosphere" refers to that a reaction flask is equipped with a balloon having 1 L of argon or nitrogen.
In hydrogenation reactions, the reaction system was generally vacuumed and filled with hydrogen, and the above operation was repeated for three times.
Microwave reactions were performed with a CEM Discover-S 908860 microwave reactor.
Unless otherwise stated in the examples, the solution used in following reactions refers to an aqueous solution.
Unless otherwise stated in the examples, the reaction temperature in the following reaction was room temperature.
Room temperature was the most proper reaction temperature, which was 20 C to 30 C.
The reaction process was monitored by thin layer chromatography (TLC), the system of developing solvent included: A: dichloromethane and methanol system, B:
n-hexane and ethyl acetate system, C: petroleum ether and ethyl acetate system, D:
acetone. The ratio of the volume of the solvent was adjusted according to the polarity of the compounds.
The elution system of purificaiton the compounds by the column chromatography and thin layer chromatography included: A: dichloromethane and methanol system, B:
n-hexane and ethyl acetate system, the ratio of the volume of the solvent was adjusted
20 = = CA 02806324 2013-01-21 according to the polarity of the compounds, and sometimes a little alkaline reagent such as triethylamine or an acidic reagent such as acetic acid was also can be added.

Example 1 4- [ [4-fluoro-3 -(trifluoromethyl)-6,8-dihydro-5H41,2,4]triazolo [4,3-a]pyrazine-7-carbonyliphenylimethyl]-2H-phthalazin-1-one 140 ,r a try--N,N
F F F

0 HNTh%NN ,r 0 1401 r 0 + ---4" N-M----NN

la lb 1 F 0 2-Fluoro-5-[(4-oxo-3H-phthalazin-1-yl)methyl]benzoic acid la (150 mg, 0.50 mmol, prepared according to a known method disclosed by "patent application W02004080976") was dissolved in 2 mL of N, N-dimethylformamide, followed by addition of 0-(1-benzotriazoly1)-N, N, N', N'-tetramethyluronium hexafluorophosphate (284 mg, 0.75 mmol), 3-(trifluoromethyl)-5,6,7,8-tetrahydro-[1,2,41triazolo[4,3-a]

pyrazine hydrochloride lb (138 mg, 0.60 mmol, prepared according to a known method disclosed by "patent application W02004080958") and N, N-diisopropylethylamine (0.2 mL. 1 mmol). After stirring for 12 hours, the reaction mixture was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A to obtain 4-[[4-fluoro-3-[3-(trifluoromethyl)-6,8-dihydro-5H41,2,4]triazolo [4,3-c]pyrazine-7-carbonyliphenyl]methyl] -2H-phthalazin-1 -one 1 (25 mg, yield 10.6%) as a white solid.

MS miz (ESI): 473.2 [M+1 11-1 NMR (400 MHz, CDC13): 6 10.04 (br. s, 1H), 8.48 (d, 1H), 7.80 (m, 3H), 7.55 (m, 1H), 7.40 (m, 1H), 7.15 (m, 1H), 4.29 (s, 2H), 4.23 (m, 2H), 3.74 (m, 2H), 3.20 (m, 2H) Example 2 4- [ [3-(3 ,4-dihydro-1H-pyrrolo [1,2-a]pyrazine-2-carbony1)-4-fluoro-phenyl]methy1]-2H-phthalazin-1-one 0111 rsp N o F NCN-r-)
21 NNy step 1 N step 2 HN / 4. el NH 0 step 3 0 y--Nor/

2a 2b 2c la 2 Step 1 2-pyrrol-1-yl-ethanamine Pyrrole 2a (12 g, 17.90 mmol) was dissolved in 150 mL of acetonitrile, followed by addition of 2-chloroethylamine hydrochloride (24.60 g, 21.20 mmol), sodium hydroxide (0.50 g, 4 mmol) and tetrabutyl ammonium hydrogen sulfate (2.40 g, 7 mmol).
After stirring for 4 hours under reflux condition, the reaction mixture was heated to 50 C and reacted for 12 hours. The reaction mixture was concentrated under reduced pressure to obtain 2-pyrrol-1-yl-ethanamine 2b (8 g, yield 41.0%) as a light yellow oil.
Step 2 1,2,3 ,4-tetrahydropyrrolo [1,2-c]pyrazine 2-1334rol-1-yl-ethanamine 2b (2 g, 18 mmol) was dissolved in 40 mL of ethanol, followed by addition of formaldehyde solution (40%, 1.5 mL, 18 mmol) and a slow dropwise addition of 1 mL of trifluoroacetic acid. The reaction mixture was heated to 50 C for 15 minutes, then cooled to room temperature and stirred for 12 hours. The reaction mixture was concentrated under reduced pressure, added with 50 mL of ethyl acetate, washed with saturated sodium bicarbonate solution (50 mL), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure to obtain 1,2,3,4-tetrahydropyrrolo[1,2-c]pyrazine 2c (1.60 g, yield 72.7%) as a light yellow oil.
Step 3 4- [ [3-(3 ,4-dihydro-1H-pyrrolo [1,2-c]pyrazine-2-carbony1)-4-fluoro-phenyl]methy11-2H-phthalazin-1 -one 2-Fluoro-5-[(4-oxo-3H-phthalazin- 1 -yl)methyl]benzoic acid la (300 mg, 1 mmol) was dissolved in 3 mL of N, N-dimethylformamide, followed by addition of 0-(1-benzotriazoly1)-N, N, N', N'-tetramethyluronium hexafluorophosphate (568 mg, 1.50 mmol), 1,2,3,4-tetrahydropyrrolo[1,2-c]pyrazine 2c (210 mg, 1.50 mmol) and N, N-diisopropylethylamine (350 1.iL, 2 mmol). After stirring for 12 hours, the reaction mixture was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A to obtain 44[3-(3,4-dihydro-pyrrolo[1,2-alpyrazine-2-carbony1)-4-fluoro-phenyl]methyl]-2H-phthalazin-1-one 2 (15 mg, yield 3.7%) as a white solid.
MS m/z (ESI): 403.1 [M+11 1H NMR (400 MHz, CDC13): 6 10.19 (br. s, 1H), 8.51 (d, 111), 7.82 (m, 3H), 7.41 (m, 2H), 7.13 (m, I H), 6.65 (m, 1H), 6.24 (m, 1H), 5.81 (m, 111), 4.97 (s, 1H), 4.59 (s, 111), 4.33 (s, 2H), 4.13 (m, 1H), 4.00 (m, 1H), 3.71 (m, 1H), 2.85 (m, 1H)
22 = = CA 02806324 Example 3 methyl 7- [2-fluoro-5- [(4-oxo-3H-phthalazin-1-y pmethyl]benzoyl] -3 -(trifluoromethy 1)-6,8-dihydro-5H-imidazo [1,5-c]pyrazine-1-carboxylate r;INH

1401 \
F F

0 0 N il ¨3step 1- OH step 2 F

3a 3b la F

Step 1 methy13-(trifluoromethyl)-5,6,7,8-tetrahydroimidazo [1,5 -c]pyrazine-1-carboxylate 07- tert-buty101-methy13-(trifluoromethyl)-6, 8- dihy dro-5H-imi dazo [1,5-a]pyrazin e-1,7-dicarboxylate 3a (600 mg, 1.72 mmol, prepared according to a known method disclosed by "patent application W02009082881") was dissolved in 20 mL of a solution of hydrogen chloride in 1,4-dioxane (2 M). After stirring for 5 hours, the reaction mixture was concentrated under reduced pressure and added with 50 mL of dichloromethane. Saturated sodium bicarbonate solution was added dropwise to the reaction mixture until the pH is 8. The organic phase was separated, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure to obtain crude methyl 3-(trifluoromethyl)-5,6,7,8-tetrahydroimidazo[1,5-a]

pyrazine- 1 -carboxylate 3b (430 mg) as a white solid. The product was used directly in the next reaction without purification.

Step 2 methyl 7- [2-fluoro-5- [(4-oxo-3H-phthalazin-1-yOmethyl]benzoyl]-3-(trifluoromethyl)-6,8-dihydro-5H-imidazo [1,5-c]pyrazine-1-carboxylate 2-Fluoro-5[(4-oxo-3H-phthalazin-1-yl)methyl]benzoic acid la (300 mg, 1 mmol) was dissolved in 2 mL of N, N-dimethylformamide, followed by addition of 0-(1-benzotriazoly1)-N, N, N', N'-tetramethyluronium hexafluorophosphate (568 mg, 1.50 mmol), crude methyl 3-(trifluoromethyl)-5,6,7,8-tetrahydroimidazo[1,5-a]pyrazine -1-carboxylate 3b (300 mg, 1.50 mmo) and N, N-diisopropylethylamine (0.4 mL, 2 mmol). After stirring for 12 hours, the reaction mixture was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A to obtain methyl 7-[2-fluoro-5-[(4-oxo-3H-phthalazin-1-yOmethyl]benzoyl]-3-(trifluoromethyl)-6,8-dihydro-5H-imidazo[1,5-a]pyrazine-1-carboxylate 3 (120 mg, yield 23.0%) as a light yellow solid.

MS m/z (ESI): 530.1 [M+l]

11-1 NMR (400 MHz, CDC13): 6 10.48 (br. s, 1H), 8.52 (d, 1H), 7.87 (m, 3H), 7.43 (m, 2H), 7.30 (m, 1H), 5.02 (m, 2H), 4.34 (s, 2h), 4.17 (m, 2H), 3.99 (m, 2H), 3.00 (s, 3H)
23 Example 4 4- [ [3 -(6,8-dihydro-5H-imidazo [1,2-a]pyrazine-7-carbony1)-4-fluoro-phenyl] methyl] -2H-phthalazin-1-one =,2 0Naf21/ 0 N NH2 NH 0 -1Ø ,1\111F1 step 1 step 2 HrlN io= step 3 OHF
4a 4b 4c la 4 Step 1 imidazo [1,2-c]pyrazine Pyrazin-2-amine 4a (5 g, 52 mmol) was dissolved in a 40% 2-chloroacetaldehyde solution (15 mL, 78 mmol), followed by addition of sodium bicarbonate (6.60 g, mmol). After stirring for 48 hours at 100 C, the reaction mixture was cooled to room temperature, added with 100 mL of a saturated potassium carbonate solution, and extracted with dichloromethane (100 mL X 3). The organic phase was combined, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure to obtain imidazo[1,2-a]pyrazine 4b (3 g, yield 50.0%) as a brown solid.
MS m/z (ESI): 120.1 [M+l]
Step 2 5,6,7,8-tetrahydroimidazo[1,2-a]pyrazine Imidazo[1,2-a]pyrazine 4b (500 mg, 4.20 mmol) was dissolved in 5 mL of 2-methoxyethanol, followed by addition of platinum dioxide (100 mg, 0.36 mmol), and the reactor was purged with hydrogen for three times. After stirring for 12 hours, the reaction mixture was filtered. The filtrate was concentrated under reduced pressure to obtain 5,6,7,8-tetrahydroimidazo[1,2-cdpyrazine 4c (200 mg, yield 38.7%) as a yellow oil.
MS rniz (ESI): 124.1 [M+1]
Step 3 44 [3 -(6,8-dihydro-5H-imidazo [1,2-a]pyrazine-7-carbony1)-4- fluoro-phenylimethyl] -2H-phthalazin-l-one 2-Fluoro-5-[(4-oxo-3H-phthalazin-1-y1)methyl]benzoic acid la (323 mg, 1.08 mmol) was dissolved in 5 mL of N, N-dimethylformamide, followed by addition of 0-(1-benzotriazoly1)-N, N, N', N'-tetramethyluronium hexafluorophosphate (614 mg, 1.63 mmol), 5,6,7,8-tetrahydroimidazo[1,2-a]pyrazine 4c (200 mg, 1.63 mmol) and N, N-diisopropylethylamine (0.4 mL, 2.16 mmol). After stirring for 12 hours, the reaction mixture was concentrated under reduced pressure and the resulting residue was purified
24 .

by thin layer chromatography with elution system A to obtain 4-[[3-(6,8-dihydro-5H-imidazo[1,2-a]pyrazine-7-carbony1)-4-fluoro-phenyllmethyl]-2H-phthalazin-1-one (10 mg, yield 2.3%) as a white solid.
MS m/z (ESI): 404.1 [M+1]
5 11-1 NMR (400 MHz, CDC13): 6 10.07 (hr. s, 1H), 8.53 (d, 1H), 7.96 (m, 1H), 7.83 (m, 3H), 7.51 (m, 1H), 7.30 (m, 2H), 6.01 (t, 1H), 4.73 (d, 2H), 4.35 (s, 2H), 1.60 (m, 2H), 1.34 (m, 2H) Example 5 10 44 [4-fluoro-343-(trifluoromethyl)-6,8-dihydro-5H-imidazo [1,5-cdpyrazine-7-c arbonyl]phenyl]methy1]-2H-phthalazin-1-one0 =NH --rj 0 F NCrN FE F
4 01 0 NH 0 _v. 1101 r\NIH0 0 2HCI 5a 40 OH F

2-Fluoro-54(4-oxo-3H-phthalazin-1-yOmethyl]benzoic acid la (500 mg, 1.68 15 mmol) was dissolved in 5 mL of N, N-dimethylformamide, followed by addition of 0-(1-benzotriazoly1)-N, N, N', N'-tetramethyluronium hexafluorophosphate (955 mg, 2.52 mmol), 3-trifluoromethy1-5,6,7,8-tetraahydroimidazo[1,5-a]pyrazine hydrochloride 5a (457 mg, 2 mmol, prepared according to a known method disclosed by "patent application W02009082881") and N, N-diisopropylethylamine (0.6 mL, 3.36 mmol).
20 After stirring for 12 hours, the reaction mixture was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A to obtain 41[4-fluoro-343-(trifluoromethyl)-6,8-dihydro-5H-imidazo[1,5-a]pyrazine-7-carbonyl]phenyl]methy1]-2H-phthalazin-1-one 5 (400 mg, yield 50.5%) as a white solid.
25 MS miz (ESI): 472.1 [M+1 11-1 NMR (400 MHz, CDC13): 6 10.81 (br. s, 1H), 8.49 (m, 1H), 7.79 (m, 3H), 7.42 (m, 2H), 7.08 (m, 1H), 5.00 (m, 1H), 4.64 (m, 1H), 4.32 (m, 2H), 4.16 (m, 3H), 3.75 (m, 1H), 3.49 (s, 1H) Example 6 4- [ [4-fluoro-3 - [1 -(hydroxymethyl)-3 -(trifluoromethyl)-6,8-dihy dro-5H-imidazo [1,5 -c]pyrazine-7-carbony l]phenylimethyl]-2H-phthalazin-l-one . .. CA 02806324 2013-01-21 o 100 .fr 0 OH

MX

FF F

0 0 OH * NH 0 SI NI NH
\
OH
HN-r-Z- step 1 HN-----1 N 0 step 2 * 0 N --,--(-N
OH F
F F F

3b 6a la Step 1 [3 -(trifluoromethyl)-5 ,6,7,8-tetrahydroimidazo [1,5-c]pyrazin-1 -yl]methanol Methyl 3-(trifluoromethyl)-5,6,7,8-tetrahydroimidazo[1,5-a]pyrazine-1-carboxylate 3b (315 mg, 1.26 mmol) was dissolved in 10 mL of ethanol, followed by addition of sodium borohydride (240 mg, 6.33 mmol). After stirring for 12 hours, the reaction mixture was added dropwise with 2 M hydrochloric acid until no gas was generated in the reaction mixture. The reaction mixture was concentrated under reduced pressure to obtain the crude [3-(trifluoromethyl)-5,6,7,8-tetrahydroimidazo[1,5-a]

pyrazin- 1 -ylimethanol 6a (230 mg) as a white solid. The product was used directly in the next reaction without purification.

Step 2 4- [ [4-fluoro-3 - [1-(hy droxymethyl)-3 -(trifluoromethyl)-6,8-dihydro-5H-imidazo [1,5 -a]pyrazine-7-carbonyl]phenyl]methyl] -2H-phthalazin-1-one 2-Fluoro-5[(4-oxo-3H-phthalazin-1-yl)methylibenzoic acid la (372 mg, 1.25 mmol) was dissolved in 5 mL of N, N-dimethylformamide, followed by addition of N-hydroxybenzotriazole (85 mg, 0.63 mmol), [3-(trifluoromethyl)-5,6,7,8-tetrahydroimidazo [1,5-c]pyrazin-1-yllmethanol 6a (277 mg, 1.25 mmol), 1-ethyl-(3-dimethyl-aminopropyl) carbodiimide hydrochloride (359 mg, 1.88 mmol) and triethylamine (0.3 mL, 2.5 mmol). After stirring for 12 hours, the reaction mixture was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A to obtain 41[4-fluoro-341-(hydroxymethyl)-3-(trifluoromethyl)-6,8-dihydro-5H-imidazo [1,5-c]pyrazine-7-carbonyllphenyl]methy1]-2H-phthalazin- 1 -one 6 (400 mg, yield 64.0%) as a white solid.

MS miz (ESI): 502.2 [M+1]

1H NMR (400 MHz, CDC13): 6 10.81 (br. s, 1H), 8.47 (s, 1H), 7.83-7.75 (m, 3H), 7.42-7.36 (m, 2H), 7.14-7.12 (m, 1H), 5.31 (s, 1H), 5.04 (s, 1H), 4.69 (d, 1H), 4.50 (s, 1H), 4.32-4.25 (m, 4H), 4.16-4.10 (m, 1H), 2.05 (s, 1H) Example 7 N-ethy1-7-[2-fluoro-5-[(4-oxo-3H-phthalazin-1-yOmethyl]benzoyd -3 -(trifluoromethyl)-6,8-dihydro-5H-imidazo [1,5-c]pyrazine-1-carboxamide
26 /110 N,J

F

step 1 1\1---ri * OH step 2 110 F F F F

3b 7a la 7 Step 1 N-ethyl-3-(trifluoromethyl)-5,6,7,8-tetrahydroimidazo[1,5-a]pyrazine-1-carboxamide Methyl 3-(trifluoromethyl)-5,6,7,8-tetrahydroimidazo[1,5-a]pyrazine-1-carboxylate 3h (1 g, 4 mmol) was dissolved in 40 mL of ethylamine solution (60%). After stirring at 50 C for 12 hours, the reaction mixture was concentrated under reduced pressure to obtain the crude N-ethyl-3 -(trifluoromethyl)-5,6,7,8-tetrahy droimidazo [1,5 -a]pyrazine-1-carboxamide 7a (1.15 g) as a white solid. The product was used directly in the next reaction without purification.

MS m/z (ESI): 263.1 [M+1]

Step 2 N-ethyl- 7- [2-fluoro-5- [(4-oxo-3H-phthalazin-1-yl)methyl]benzoyl]-3 -(trifluoromethyl)-6,8-dihydro-5H-imidazo [1,5-a]pyrazine-1-carboxamide 2-Fluoro-5-[(4-oxo-3H-phthalazin-1-yl)methyl]benzoic acid la (250 mg, 0.84 mmol) was dissolved in 20 mL of N, N-dimethylformamide, followed by addition of 0-(1-benzotriazoly1)-N, N, N', N'-tetramethyluronium hexafluorophosphate (480 mg, 1.26 mmol), crude N-ethy1-3-(trifluoromethyl)-5,6,7,8-tetrahydroimidazo[1,5-alpyrazine-1-carboxamide 7a (242 mg, 0.92 mmol) and N, N-diisopropylethylamine (0.3 mL, 1.68 mmol). After stirring for 12 hours, the reaction mixture was added with 50 mL of H20, and extracted with dichloromethane (50 mL
X

3). The organic phase was combined, concentrated under reduced pressure, added with100 mL of ethyl acetate, washed successively with saturated sodium bicarbonate solution (40 mL), saturated sodium chloride solution (40 mL), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A

to obtain N-ethyl-7- [2-fluoro-5- [(4-oxo-3H-phthalazin-1-yl)methyl]benzoyl]-3-(trifluoromethyl)-6,8-dihydro-5H-imidazo[1,5-a]pyrazine-1-carboxamide 7 (200 mg, yield 43.9%) as a white solid.

MS m/z (ESI): 543.2 [M+11 1H NMR (400 MHz, CDC13): 6 11.38 (hr. s, 1H), 8.47 (m, 1H), 7.84 (m, 3H), 7.37 (m, 2H), 7.19 (m, 1H), 5.10 (s, 2H), 4.30 (s, 2H), 4.29 (m, 4H), 3.47 (m, 2H), 1.27 (m, 3H)
27 Example 8 7[2-fluoro-5- [(4-oxo-3H-phthalazin-1 -yl)methyl]benzoy11-3 -(trifluoromethyl)-6,8-dihydro-5H-imidazo [1,5 -a]pyrazine-l-carboxylic acid .zH 0 0 Ir)----="
F F

, NH -N NH
0 o OH
o LI\

Methyl 7- [2-fluoro-5 - [(4-oxo-3H-phthalazin-1-yl)methyl]benzoyl] -3-(trifluoromethy 1)-6,8- dihydro-5H-imidazo [1,5-a]pyrazine-1-carboxy late 3 (30 mg, 0.057 mmol) was dissolved in 1.5 mL of a mixed solvent of tetrahydrofuran, methanol and water (V/V/V = 1:1:1), followed by addition of sodium hydroxide (10 mg, 0.25 mmol). After stirring for 12 hours, the reaction mixture was added dropwise with concentrated hydrochloric acid until the pH was 2. The reaction mixture was extracted with dichloromethane (15 mL X 2). The organic phase was combined, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A to obtain 7-[2-fluoro-5-[(4-oxo-3H-phthalazin-1-yl)methyl] benzoy1]-3 -(trifluoromethyl)-6,8-dihydro-5H-imidazo [1,5 -a]pyrazine-1-carboxylic acid 8 (10 mg, yield 34.4%) as a light yellow solid.
MS m/z (ESI): 516.5 [M+1]
1H NMR (400 MHz, CD30D): .5 8.36 (d, 1H), 7.93 (d, 1H), 7.83 (m, 2H), 7.60 (d, 1H), 7.29 (m, 1H), 6.97 (t, 1H), 4.32 (s, 2H), 3.41 (m, 6H) Example 9 4- [ [4- fluoro-3 -[1 -(methylaminomethyl)-3 -(trifluoromethyl)-6,8-dihy dro-imidazo[1, 5-a]pyrazine-7- carbonyl]phenyl]methy1]-2H-phthalazin- 1-one , insA;--/N rtl F F
28 = CA 02806324 2013-01-21 NH NH NH
N 0 OH 101 0 CI , N 0y N step 1 1.1 _r\--N step 2 F F
6 9a 9 Step 1 4- [[3 -[1 -(chloromethyl)-3 -(trifluoromethyl)-6,8-dihydro-5H-imidazo [1,5 -a]pyrazine-7-carbony1]-4-fluoro-phenyl]methy1]-2H-phthalazin-1 -one 4- [ [4-F luoro-3 -[1 -(hy droxymethyl)-3 -(trifluoromethyl)-6,8-dihy dro-5H-imidazo [1, 5-a]pyrazine-7-carbonyl]phenyl]methy1]-2H-phthalazin-1-one 6 (200 mg, 0.40 mmol) was dissolved in 5 mL of thionyl chloride. The reaction mixture was heated to reflux for 4 hours. The reaction mixture was concentrated under reduced pressure, added with 10 mL of H20, extracted with dichloromethane (10 mL X 3). The organic phase was combined, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure to obtain 4-[[3-[1-(chloromethyl)-3-(trifluoromethyl)-6,8-dihydro-5H-imidazo [1,5 -a]pyrazine-7-carbonyl]-4-fluoro-phenyl]

methy1]-2H-phthalazin-1-one 9a (200 mg, yield 96.6%) as a yellow solid.

MS m/z (ESI): 520.1 [M+1]
Step 2 4- [[4-fluoro-3- [1 -(methylaminomethyl)-3 -(trifluoromethyl)-6,8-dihy dro-5H-imidazo [1,5-a] pyrazine-7-carbonyl] phenylimethy1]-2H-phthalazin-l-one 4- [ [3- [1 -(Chloromethyl)-3-(trifluoromethy 1)-6,8-dihydro-5H-imidazo [1,5-a]

pyrazine-7-c arbonyl] -4-fluoro-phenyl]methyl] -2H-phthalazin-1 -one 9a (372 mg, 1.25 mmol) was dissolved in 5 mL of acetonitrile, followed by addition of 0.6 mL of a 2 M

solution of methylamine in tetrahydrofuran and potassium carbonate (159 mg,1.15 mmol). The reaction mixture was heated to reflux for 6 hours. The reaction mixture was filtered. The filtrate was concentrated under reduced pressure and was purified by thin layer chromatography with elution system A to obtain 4-[[4-fluoro-3-[1 -(methylaminomethyl)-3 -(trifluoromethyl)-6,8-dihy dro-5H-imidazo [1,5 -a]pyrazine-7-carbonyl]phenyl]methy1]-2H-phthalazin- 1-one 9 (20 mg, yield 10.1%) as a yellow solid.

MS m/z (ESI): 515.2 [M+1]

NMR (400 MHz, CDC13): 6 11.87 (br. s, 1H), 8.35-8.42 (m, 1H), 7.72-7.81 (m, 3H), 7.35-7.43 (m, 1H), 6.96-7.06 (m, 1H), 5.01-5.02 (m, 1H), 3.99-4.28 (m, 6H), 3.71-3.72 (m, 1H), 3.47 (s, 1H), 2.74 (d, 3H), 2.03-2.05 (m, 1H) Example 10 7- [2-fluoro-5- [(4-oxo-3H-phthalazin-1-yl)methyl]benzoyl]-3-(trifluoromethyl)-6,8-dihydro-5H-imidazo [1,5 -a] pyrazine-l-c arboxamide
29 . .

o 101 ,rsrijH 0 o 41111. F L,N i F

0r`INFI 0 HN..."...)-): NW-- ....; N 2 0 _H
Nz ........ N
A... ste p 2 IP" F NI_ F F
3b 10a la Step 1 3 -(trifluoromethyl)-5 ,6,7,8-tetrahydroimidazo [1,5-a]pyrazine-1-carboxamide 5 Methyl 3 -(trifluoromethyl)-5 ,6,7,8-tetrahydroimidazo [1,5-a]pyrazine-1-carboxylate 3b (250 mg, 1 mmol) and 10 mL of ammonium hydroxide were added in a 20 mL sealed tube. The reaction mixture was heated to 100 C and reacted for 3 hours.

The reaction mixture was concentrated under reduced pressure to obtain crude 3 -(trifluoromethyl)-5 ,6,7,8-tetrahydroimidazo [1,5-a]pyrazine-1-carboxamide 10a (240 mg) as a white solid. The product was used directly in the next reaction without purification.

MS m/z (ESI): 235.1 [M+1]

Step 2 7- [2-fluoro-5- [(4-oxo-3H-phthalazin-1-yl)methyl]benzoyl] -3 -(trifluoromethyl)-6,8-dihydro-5H-imidazo [1,5 -a]pyrazine-1- carboxamide 2-Fluoro-5-[(4-oxo-3H-phthalazin-1 -yl)methyl]benzoic acid la (150 mg, 0.50 mmol) was dissolved in 10 mL of N, N-dimethylformamide, followed by addition of 0-(1-benzotriazoly1)-N, N, N', N'-tetramethyluronium hexafluorophosphate (285 mg, 0.75 mmol), crude 3 -(trifluoromethyl)-5 ,6,7,8-tetrahydroimidazo [1,5-a]pyrazine-1-carboxamide 10a (130 mg, 0.55 mmol) and N, N-diisopropylethylamine (0.2 mL, mmol). After stirring for 12 hours, the reaction mixture was added with 50 mL
of H20, extracted with dichloromethane (60 mL x3). The organic phase was combined, concentrated under reduced pressure, added with 100 mL of ethyl acetate, washed successively with H20 (40 mL) and saturated sodium chloride solution (40 mL), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A to obtain 742-fluoro-5-[(4-oxo-3H-phthalazin-1 -yl)methyl]benzoy1]-3 -(trifluoromethyl)-6,8-dihydro-5H-imidazo [1,5-a]pyrazine-1-carboxamide 10 (50 mg, yield 20.0%) as a white solid.

MS rn/z (ESI): 515.1 [M+1]

11-1 NMR (400 MHz, CDC13): 6 8.49 (m, 1H), 7.85 (m, 3H), 7.33 (m, 2H), 7.15 (m, 1H), 5.07 (s, 2H), 4.30 (s, 2H), 4.23 (m, 4H) Example 11
30 =

4- [ [3- [1-bromo-3 -(trifluoromethyl)-6,8-dihydro-5H-imidazo [1,5-a]pyrazine-carbony l] -4-fluoro-phenyl]methyl] -2H-phthalazin-1 -one N

Br F
F-7¨F

41 ....NH

Br Br *I NH

Br HN---'1,---N;N
.-1,N^-r-NN
-N''''"=(--KN
op step 1 step 2 step 3 HCI
OH
t F F
F la: s F
2HCI F5a 11a F
11b F
11c F

Step 1 tert-butyl 3-(trifluoromethyl)-6,8-dihydro-5H-imidazo [1,5-c]pyrazine-7-carboxylate 3 -(Trifluoromethyl)-5 ,6,7,8-tetrahydroimidazo [1,5 -a]pyrazine hydrochloride 5a (2.20 g, 8.30 mmol) was dissolved in 20 mL of dichloromethane, followed by addition of triethylamine (4.6 mL, 33.20 mmol) and di-tert-butyl dicarbonate (2.70 g, 12.50 mmol). After stirring for 12 hours, the reaction mixture was added with 50 mL
of H20, extracted with dichloromethane (50 mL x3). The organic phase was combined, washed successively with saturated ammonium chloride solution (40 mL) and saturated sodium chloride solution (40 mL), dried over anhydrous sodium sulfate and filtered.
The filtrate was concentrated under reduced pressure to obtain tert-butyl 3-(trifluoromethyl)-6,8-dihydro-5H-imidazo[1,5-c]pyrazine-7-carboxylate ha (2.20 g, yield 91.7%) as a light brown solid.
MS m/z (ESI): 292.1 [M+11 Step 2 tert-butyll-bromo-3 -(trifluoromethyl)-6,8-dihy dro-5H-imidazo [1,5-a]
pyrazine-7-carboxylate Tert-butyl 3 -(trifluoromethyl)-6,8-dihydro-5H-imidazo [1,5 -c]pyrazine-7-carboxylate ha (370 mg, 1.27 mmol) was dissolved in 30 mL of tetrahydrofuran, followed by addition of N-bromosuccinimide (453 mg, 2.54 mmol) under -78 C.
After stirring for 1 hour, the reaction mixture was heated to room temperature and reacted for 12 hours. The reaction mixture was added with 50 mL of H20, extracted with ethyl acetate (60 mL x3). The organic phase was combined, washed with saturated sodium chloride solution (40 mL), dried over anhydrous sodium sulfate and filtered.
The filtrate was concentrated under reduced pressure to obtain crude tert-butyl 1-bromo-3 -(trifluoromethyl)-6,8-dihydro-5H-imidazo [1,5 -a]pyrazine-7-carboxy late llb (510 mg) as a light yellow oil. The product was used directly in the next reaction without purification.
MS m/z (ESI): 372.0 [M+l]
Step 3 1 -bromo-3 -(trifluoromethyl)-5,6,7,8-tetrahydroimidazo [1,5 -c]pyrazine hydrochloride
31 = =

Crude tert-butyl 1 -bromo-3 -(trifluoromethyl)-6,8-dihydro-5H-imidazo [1,5-a]
pyrazine-7-carboxylate lib (470 mg, 1.27 mmol) was dissolved in 50 mL of a 2 M

solution of hydrogen chloride in 1,4-dioxane. After stirring for 4 hours, the reaction mixture was concentrated under reduced pressure to obtain 1-bromo-3 -5 (trifluoromethyl)-5,6,7,8-tetrahydroimidazo[1,5-c]pyrazine hydrochloride 11c (220 mg, yield 56.5%) as a light yellow oil.

Step 4 4-[[3-[1-bromo-3-(trifluoromethyl)-6,8-dihydro-5H-imidazo[1,5-c]pyrazine-7-carbonyl]-4-fluoro-phenyl]methyl]-2H-phthalazin-1-one 10 2-Fluoro-5-[(4-oxo-3H-phthalazin- 1 -yl)methyl]benzoic acid la (210 mg, 0.70 mmol) was dissolved in 30 mL of N, N-dimethylformamide, followed by addition of 0-(1-benzotriazoly1)-N, N, N', N'-tetramethyluronium hexafluorophosphate (360 mg, 0.95 mmol), 1-bromo-3-(trifluoromethyl)-5,6,7,8-tetrahydroimidazo[1,5-a]pyrazine hydrochloride 11c (214 mg, 0.70 mmol) and N, N-diisopropylethylamine (0.4 mL, 2.10 15 mmol). After stirring for 12 hours, the reaction mixture was added with 50 mL of H20, extracted with dichloromethane (80 mL x3). The organic phase was combined, concentrated under reduced pressure, added with 100 mL of ethyl acetate, washed successively with saturated sodium carbonate solution (40 mL), H20 (40 mL) and saturated sodium chloride solution (40 mL), dried over anhydrous sodium sulfate and 20 filtered. The filtrate was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A to obtain 41[341- bromo-3-(trifluoromethyl)-6,8-dihydro-5H-imidazo [1,5-c]pyrazine-7-carbonyl]
-4-fluoro-phenylimethy11-2H-phthalazin-1 -one 11(185 mg, yield 48.0%) as a white solid.
25 MS m/z (ESI): 552.0 [M+1 11-1 NMR (400 MHz, CDC13): 6 8.48 (m, 1H), 7.73 (m, 3H), 7.31 (m, 2H), 7.11 (m, 1H), 4.89 (s, 2H), 4.49 (s, 214), 4.48 (m, 4H) Example 12 30 44 [4-fluoro-342-(trifluoromethyl)-6,8-dihydro-5H-imidazo [1,2-c]pyrazine-7-carbonyl]
phenylimethyl]-2H-phthalazin-1-one op _NH

F

14:-.747) (FFF step 2Htsl-Thfr.) (FFF

4a 12a A step 12b 12c la is OH step 3 101 12 F
(FF F

Step 1 2-(trifluoromethyl)imidazo [1,2-c]pyrazine Pyrazin-2-amine 4a (5.25 g, 55.20 mmol) was dissolved in 120 mL of ethanol,
32 , =

followed by addition of 3-bromo-1,1,1-trifluoro-propan-2-one 12a (5.7 mL, 55.20 mmol). The reaction mixture was heated to reflux for 16 hours. The reaction mixture was concentrated under reduced pressure, added with 100 mL of ethyl acetate and 100 mL of saturated sodium bicarbonate solution and separated. The aqueous phase was 5 extracted with ethyl acetate (50 mL x3). The organic phase was combined, washed with saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and the resulting residue was purified by silica gel column chromatography with elution system B to obtain 2-(trifluoromethyl)imidazo[1,2-c]pyrazine 12b (2.40 g, yield 22.8%) as a yellow solid.
10 MS m/z (ESI): 188.0 [M+11 Step 2 2-(trifluoromethyl)-5,6,7,8-tetrahydroimidazo [1,2-a] pyrazine 2-(Trifluoromethyl)imidazo[1,2-a]pyrazine 12b (2.40 g, 12.55 mmol) was dissolved in 100 mL of methanol, followed by addition of Pd-C (10%, 480 mg), and the 15 reactor was purged with hydrogen for three times. After stirring for 12 hours, the reaction mixture was filtered and the filter cake was washed with methanol.
The filtrate was concentrated under reduced pressure to obtain 2-(trifluoromethyl)-5,6,7,8-tetrahydroimidazo[1,2-a]pyrazine 12c (2.30 g, yield 95.8%) as a yellow oil.
Step 3 20 44[4-fluoro-3[2-(trifluoromethyl)-6,8-dihydro-5H-imidazo[1,2-a]pyrazine-7-carbonyl]
phenylimethyl] -2H-phthalazin-1 -one 2-Fluoro-5-[(4-oxo-3H-phthalazin-1-yOmethyl]benzoic acid la (500 mg, 1.68 mmol) was dissolved in 10 mL of N, N-dimethylformamide, followed by addition of 0-(1-benzotriazoly1)-N, N, N', N'-tetramethyluronium hexafluorophosphate (830 mg, 25 2.52 mmol), 2-(trifluoromethyl)-5,6,7,8-tetrahydroimidazo[1,2-cdpyrazine 12c (384 mg, 2 mmol) and N, N-diisopropylethylamine (1 mL, 5 mmol). After stirring for 12 hours, the resulting residue was purified by silica gel column chromatography with elution system A to obtain 4-[[4-fluoro-3-[2-(trifluoromethyl)-6,8-dihydro-5H-imidazo [1,2-c]pyrazine-7-carbonyllphenyl]methy1]-2H-phthalazin-1 -one 12 (200 mg, 30 yield 25.0%) as a white solid.
MS m/z (ESI): 472.1[M+1]
11-1 NMR (400 MHz, CDC13): 8 10.29 (br. s, 1H), 8.47 (m, 1H), 7.80 (m, 3H), 7.37 (m, 2H), 7.25 (m, 1H), 6.50 (m, 1H), 4.67 (s, 2H), 4.28 (m, 2H), 4.14 (m, 2H), 3.73 (m, 2H) Example 13 7- [2-fluoro-5-[(4-oxo-3H-phthalazin-1-yOmethylibenzoyll-N-methyl-3-(trifluoromethyl )-6,8-dihydro-5H-imidazo [1,5 -a]pyrazine-1 -carboxamide 0 , N: 0 F F F
33 0 0 0 / NH 101 NH 0 rt 0 0 /
step N (10 OH step 2 y N
F F F

3b 13a la 13 F F

Step 1 N-methyl-3 -(trifluoromethyl)-5,6,7,8-tetrahy droimidazo [1,5 -a] pyrazine-1 -carboxamide Methyl 3 -(trifluoromethyl)-5,6,7,8-tetrahy droimidazo [1,5-a] pyrazine-l-c arboxylate 3b (500 mg, 2 mmol) was dissolved in 8 mL of methylamine solution (20% to 30%) was added in a 20 mL sealed tube. After stirring at 60 C for 6 hours, the reaction mixture was concentrated under reduced pressure to obtian the crude N-methyl-3-(trifluoromethyl)-5 ,6,7,8-tetrahydroimidazo [1,5 -a]pyrazine-1 -carboxamide 13a (498 mg) as a white solid. The product was used directly in the next reaction without purification.

MS m/z (ESI): 249.1 [M+1 Step 2 7- [2-fluoro-5- [(4-oxo-3H-phthalazin-1-yOmethyllbenzoyl]-N-methyl-3-(trifluoromethyl)-6,8-dihydro-5H-imidazo[1,5-a]pyrazine-1-carboxamide 2-Fluoro-5-[(4-oxo-3H-phthalazin-1-yl)methyl]benzoic acid la (598 mg, 2 mmol) was dissolved in 10 mL of N, N-dimethylformamide, followed by addition of 1-hydroxybenzotriazole (135 mg, 1 mmol), crude N-methy1-3-(trifluoromethyl)-5,6,7,8-tetrahydroimidazo [1,5 -a]pyrazine-l-carboxamide 13a (498 mg, 2 mmol), 1-ethyl-(3-dimethylaminopropyl) carbodiimide hydrochloride (573 mg, 3 mmol) and N, N-diisopropylethylamine (774 mg, 6 mmol). After stirring for 12 hours, the reaction mixture was concentrated under reduced pressure, added with 30 mL of H20, extracted with ethyl acetate (50 mL x3). The organic phase was combined, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A to obtain 712-fluoro-5- [(4-oxo-3H-phthalazin-l-yl)methyl]benzoy11-N-methy1-(trifluoromethyl)-6,8-dihydro-5H-imidazo [1,5-a]pyrazine-1-carboxamide 13 (650 mg, yield 61.0%) as a white solid.

MS m/z (ESI): 529.1 [M+1]

1H NMR (400 MHz, CD30D): 6 8.36-8.34 (t, 1H), 7.96-7.94 (d, 1H), 7.86-7.81 (m, 2H), 7.50-7.45 (m, 2H), 7.22-7.15 (dd, 1H), 5.23 (s, 1H), 4.95 (s, 1H), 4.39 (d, 2H), 4.32 (d, 1H), 4.21 (s, 1H), 4.14 (s, 1H), 3.76 (s, 1H), 2.85 (d, 311) Example 14 ethyl 7- [2-fluoro-5- [(4-oxo-3H-phthalazin-1 -yOmethyl]benzoyl]-6,8-dihydro-5H-imidazo [1,2 -c]pyrazine-3-carboxylate
34 =/11-1 F

NH

re step 1 0 0 step 2 0 0 IS OH step 3 140 0 4a 14a 14b la 14 0 Step 1 ethyl imidazo[1,2-cdpyrazine-3-carboxylate Pyrazin-2-amine 4a (1 g, 10 mmol) was dissolved in 50 mL of ethylene glycol dimethyl ether, followed by addition of 50 mL of methanol and 3-bromo-2-oxo-propionate (2.30 g, 12 mmol). After stirring for 4 hours at room temperature, the reaction mixture was cooled to 0 C and stirred for 30 minutes until a solid precipitated. The reaction mixture was filtered, and the filter cake was washed with ether (10 mL x3). The solid was dissolved in 50 mL of anhydrous ethanol and the solution was refluxed for 4 hours. The reaction mixture was concentrated under reduced pressure, added with 100 mL of dichloromethane, washed successively with saturated sodium carbonate solution (40 mL) and saturated sodium chloride solution (40 mL), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure to obtain ethyl imidazo[1,2-a]pyrazine-3-carboxylate 14a (0.55 g, yield 28.9%) as a brown solid.

MS m/z (ESI): 192.1 [M+1.]

Step 2 ethyl 5,6,7,8-tetrahydroimidazo [1,2-a]pyrazine-3-carboxylate Ethyl imidazo[1,2-a]pyrazine-3-carboxylate 14a (550 mg, 2.76 mmol) was dissolved in 30 mL of methanol, followed by addition of Pd-C (10%, 100 mg), and the reactor was purged with hydrogen for three times. After stirring for 3 hours, the reaction mixture was filtered and the filtrate was concentrated under reduced pressure to obtain ethyl 5,6,7,8-tetrahydroimidazo[1,2-a]pyrazine-3-carboxylate 14b (480 mg, yield 87.6%) as a yellow oil.

MS m/z (ESI): 196.1 [M+1 Step 3 ethyl 7- [2-fluoro-5- [(4-oxo-3H-phthalazin-l-yOmethyl]benzoy1]-6,8-dihydro-5H-imidazo [1,2 -a]pyrazine-3 -c arboxy late 2-Fluoro-5-[(4-oxo-3H-phthalazin-1-y1)methyl]benzoic acid la (300 mg, 1 mmol) was dissolved in 20 mL of N, N-dimethylformamide, followed by addition of
35 0-(1-benzotriazoly1)-N, N, N', N'-tetramethyluronium hexafluorophosphate (570 mg, 1.50 mmol), ethyl 5,6,7,8-tetrahydroimidazo[1,2-a]pyrazine-3-carboxylate 14b (200 mg, 1 mmol) and N, N-diisopropylethylamine (0.3 mL, 2 mmol). After stirring for 12 hours, the reaction mixture was added with 50 mL of H20, extracted with dichloromethane (80 mL x3). The organic phase was combined, concentrated under reduced pressure, added with 100 mL of ethyl acetate, washed successively with saturated sodium carbonate solution (40 mL), H20 (40 mL), saturated sodium chloride solution (40 mL), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A to obtain ethyl 742-fluoro-5-[(4-oxo-3H-phthalazin-1-yl)methyl]
benzoy1]-6,8-dihydro-5H-imidazo[1,2-c]pyrazine-3-carboxylate 14 (280 mg, yield 58.6%) as a white solid.
MS m/z (ESI): 476.1 [M+l]
11-1 NMR (400 MHz, CDC13): 6 10.53 (br. s, 1H), 8.46 (m, 1H), 7.76 (m, 3H), 7.59 (s, 1H), 7.36 (m, 2H), 7.08 (m, Hi), 4.69 (s, 211), 4.37 (m, 2H), 4.31 (s, 2H), 4.27 (m, 4H), 1.26 (t, 3H) Example 15 44 [343 -(trifluoromethy 1)-6,8-dihydro-5H41,2,4]triazolo [4,3 -a]pyrazine-7-carbonyl]phenyllmethy11-2H-phthalazin-l-one r0 N

sof11-1 N 0 + \r'rN N 0 N N
OH HCI F F
15a 41111.v. lb 15 31(4-0x6-3H-phthalazin-1-yl)methylThenzoic acid 15a (300 mg, 1.07 mmol, prepared according to a known method disclosed by "patent application W02004080976") was dissolved in 10 mL of N, N-dimethylformamide, followed by addition of 0-(1-benzotriazoly1)-N, N, N', N'-tetramethyluronium hexafluorophosphate (730 mg, 1.93 mmol), 3 -(trifluoromethyl)-5,6,7,8-tetrahydro-[1,2,4]triazolo [4,3-a]
pyrazine hydrochloride lb (269 mg, 1.40 mmol) and N, N-diisopropylethylamine (0.9 mL, 5.30 mmol). After stirring for 12 hours, the reaction mixture was added with 15 mL
of 1120, extracted with ethyl acetate (20 mLx3). The organic phase was combined, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A to obtain 4-[[3-[3-(trifluoromethyl)-6,8-dihydro-5H-[1,2,4]triazolo [4,3 -a]pyrazine-7-carbonyl]phenyl]methy1]-2H-phthalazin-1 -one 15 (100
36 = =

mg, yield 20.6%) as a white solid.

MS m/z (ESI): 455.1 [M+11 11-1 NMR (400 MHz, CDC13): 6 10.30 (hr. s, 1H), 8.49 (d, 1H), 8.02 (m, 1H), 7.78 (m, 3H), 7.43 (m, 3H), 5.31 (s, 211), 4.35 (s, 2H), 4.21 (m, 2H), 4.12 (m, 2H) Example 16 44[3 -(6,8-dihydro-5H41,2,4]triazolo [1,5-a]pyrazine-7-carbony1)-4-fluoro-phenylimethyl]-2H-phthalazin-1-one =r 0 411 F NON71.1NI

,N 0rs,J=H
_NHN 0 la 010 F OH 16a 2-Fluoro-54(4-oxo-3H-phthalazin-1-yOmethylThenzoic acid la (360 mg, 1.20 mmol) was dissolved in 10 mL of N, N-dimethylformamide, followed by addition of 0-(1-benzotriazoly1)-N, N, N', N'-tetramethyluronium hexafluorophosphate (600 mg, 1.80 mmol), 5,6,7,8-tetrahydro41,2,4]triazolo[1,5-a]pyrazine 16a (150 mg, 1.20 mmol, 15 prepared according to a known method disclosed by "patent application W02009090055") and N, N-diisopropylethylamine (0.4 mL, 2.40 mmol). After stirring for 20 hours, the reaction mixture was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A to obtain 44[3-(6,8-dihydro-5H41,2,4]triazolo [1,5-a]pyrazine-7-carbony1)-4-fluoro-20 phenyl]methy1]-2H-phthalazin-1-one 16 (100 mg, yield 21.0%) as a yellow solid.

MS m/z (ESI): 405.1 [M+1]

1H NMR (400 MHz, CDC13): 6 10.47 (hr. s, 1H), 8.51-8.49 (m, 1H), 7.99-1.77 (m, 4H), 7.42-7.30 (m, 2H), 7.30-7.12 (m, 1H), 4.76 (m, 2H), 4.37-4.28 (m, 4H), 3.77-3.73 (m, 2H) Example 17 44 [3-(6,8-dihydro-5H41,2,4]triazolo [4,3 -alpyrazine-7-carbony1)-4-fluoro-phenylimethy1]-2H-phthalazin- 1-one N%I\I=N
F LNJ
37 0 NH 0 HN.M=0"N
0 NH 0 1,1N
la * F OH Ha 17a 17 Q-1C-(/
2-Fluoro-5-[(4-oxo-3H-phthalazin- 1 -yl)methyl]benzoic acid la (170 mg, 0.57 mmol) was dissolved in 10 mL of N, N-dimethylformamide, followed by addition of 0-(1-benzotriazoly1)-N, N, N', N'-tetramethyluronium hexafluorophosphate (323 mg, 0.85 mmol), 5,6,7,8-tetrahydro-[1,2,41triazolo[4,3-a]pyrazine hydrochloride 17a (100 mg, 0.63 mmol, prepared according to a known method "Journal of Medicinal Chemistry, 2005, 48(1), 141-151") and N, N-diisopropylethylamine (302 mg, 1.70 mmol). After stirring for 12 hours, the reaction mixture was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A to obtain 44[3-(6,8-dihydro-5H-[1,2,4]triazolo[4,3-a]pyrazine-carbony1)-4-fluoro-phenyl]methyl]-2H-phthalazin- 1 -one 17 (50 mg, yield 21.7%) as a light yellow solid.
MS m/z (ESI): 405.1 [M+l]
NMR (400 MHz, CDC13): 6 10.87 (hr. s, 1H), 8.46-8.45 (m, 1H), 8.18 (s, 1H), 7.80-7.76 (m, 3H), 7.40-7.38 (m, 2H), 7.12-7.07 (m, 1H), 4.79 (m, 2H), 4.31-4.20 (m, 4H), 3.75-3.62 (m, 2H) Example 18 4- [[4-fluoro-3- [1-(pyrrolidine-1 -carbonyl)-3 -(trifluoromethyl)-6,8-dihydro-imidazo [1,5 -c]pyrazine-7-carbonyl]phenyl]methy1]-2H-phthalazin-1 -one :r 0 0 N

0 0 HNTh=-X 0 r0 io 0 NH 0 -.-F4 step 1 --F F FA-F
St 2 3b 18a la Step 1 pyrrolidin-l-y143 -(trifluoromethyl)-5,6,7,8-tetrahydroimidazo [1,5 -c]pyrazin-1 -yl] methanone Pyrrolidine (560 mg, 8 mmol), methyl 3-(trifluoromethyl)-5,6,7,8-tetrahydroimidazo[1,5-a]pyrazine- 1 -carboxylate 3b (400 mg, 1.60 mmol) and 0.4 mL of H20 were mixed in a sealed tube. After stirring at 50 C for 4 hours, the reaction
38 mixture was concentrated under reduced pressure to obtain crude pyrrolidin-1-y143-(trifluoromethyl)-5,6,7,8-tetrahydroimidazo [1,5 -a] pyrazin-l-yl]methanone 18a (460 mg) as a light yellow solid. The product was used directly in the next reaction without purification.
MS m/z (ESI): 289.1 [M+1]
Step 2 4- [[4-fluoro-3- [1-(pyrrolidine-1-carbony1)-3 -(trifluoromethyl)-6,8-dihydro-imidazo [1,5-a]pyrazine-7-c arbonyl] phenyl]methy1]-2H-phthalazin-l-one 2-Fluoro-5-[(4-oxo-3H-phthalazin-1-yl)methyl]benzoic acid la (417 mg, 1.40 mmol) was dissolved in 5 mL of N, N-dimethylformamide, followed by addition of 0-(1-benzotriazoly1)-N, N, N', N'-tetramethyluronium hexafluorophosphate (1 g, 2.80 mmol), crude pyrrolidin-l-y1-[3-(trifluoromethyl)-5,6,7,8-tetrahydroimidazo [1,5-a]
pyrazin-1 -yl]methanone 18a (400 mg, 1.40 mmol) and N, N-diisopropylethylamine (0.7 mL, 4.20 mmol). After stirring for 12 hours, the reaction mixture was concentrated under reduced pressure, added with 20 mL of H20, extracted with ethyl acetate (10 mL x3). The organic phase was combined, washed with saturated sodium chloride solution (10 mL), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A to obtain 44[4-fluoro-341-(pyrrolidine-1-carbonyl)-3-(trifluoromethyl)-6,8-dihydro-5H-imidazo [1,5-a]pyrazine-7-carbonyl]phenyl]methy1]-2H-phthalazin-1-one 18 (150 mg, yield 18.0%) as a light yellow solid.
MS m/z (ESI): 569.2 [M+1]
1H NMR (400 MHz, DMSO-d6): 6 12.57 (br. s, 1H), 8.26 (d, 1H), 7.83-7.93 (m, 3H), 7.46-7.50 (m, 2H), 7.26-7.31 (m, 1H), 5.07 (s, 1H), 4.84 (s, 1H), 4.27-4.34 (m, 2H), 4.26-4.27 (m, 1H), 4.07-4.17 (m, 2H), 3.89-3.92 (m, 2H), 3.66-3.68 (m, 1H), 3.48-3.49 (m, 1H), 3.36-3.38 (m, 1H), 1.76-1.91 (m, 4H) Example 19 44[4- fluoro-342-(trifluoromethyl)-6,8-dihydro-5H41,2,4]triazolo [1,5-a]pyrazine-7-c arb onyl] phenyl]methyl] -2H-phthalazin-1 -one op ,..,111-1 0 N 0 HNI.rN> (FF + F
0 NH 0 Sti F
(FFF 0 NH 0 19a la 441111147 40 F (FFF
2-Fluoro-5-[(4-oxo-311-phthalazin-1-yOmethyl]benzoic acid la (780 mg, 2.65 mmol) was dissolved in 15 mL of N, N-dimethylformamide, followed by addition of
39 =

0-(1-benzotriazoly1)-N, N, N', N'-tetramethyluronium hexafluorophosphate (1.80 g, 4.77 mmol), 2-(trifluoromethyl)-5,6,7,8-tetrahydro-[1,2,4]triazolo[1,5-a]pyrazine 19a (560 mg, 2.92 mmol, prepared according to a known method disclosed by "patent application W02009025784") and N, N-diisopropylethylamine (1.4 mL, 7.95 mmol).

After stirring for 12 hours, the reaction mixture was concentrated under reduced pressure, added with 30 mL of 1120, extracted with ethyl acetate (30 mL x3).
The organic phase was combined, washed with saturated sodium chloride solution (20 mL), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A to obtain 44[4-fluoro-342-(trifluoromethyl)-6,8-dihydro-[1,2,4]triazolo[1,5-a]pyrazine-7-carbonyl]phenylimethyl] -2H-phthalazin-1-one 19 (205 mg, yield 16.4%) as a light yellow solid.

MS m/z (ESI): 473.1 [M+l]

1H NMR (400 MHz, CDC13): 6 10.67 (br. s, 111), 8.48 (s, 1H), 7.77 (m, 314), 7.42 (m, 211), 7.11 (t, 1H), 5.10 (s, 1H), 4.75 (s ,1H),4.39 (s, 21-1), 4.32 (d, 311), 3.88 (s, 1H) Example 20 4- [[4-fluoro-3-[1-(morpholine-4-carbony1)-3-(trifluoromethyl)-6,8-dihydro-5H-imidazo [1,5 -a]pyrazine-7-c arbonyl] phenylimethyl] -2H-phthalazin-1 -one ,NH 0 0 r`o
40 , F F

0 0 Th_2--OH
0 o N
0 r'o 0 N \ 0 N J-L N
\N ON
INI-1111 F F step 1 F F
step 2 F4--F step 3 HCI

3a F 20a F
20b F
20c step 4 VP- NH
0 step 5 NH 0 0 r"-\ 0 io OH N--1--"="-Z-N

20d la F

Step 1 7-tert-butoxycarbony1-3-(trifluoromethyl)-6,8-dihydro-5H-imidazo[1,5-c]pyrazine-1-carboxylic acid 07-tert-butyl 01-methyl 3 -(trifluoromethyl)-6,8-dihydro-5H-imidazo [1,5-a]

pyrazine-1,7-dicarboxylate 3a (4.10 g, 12 mmol) was dissolved in a mixed solvent of 15 mL of tetrahydrofuran and methanol (V/V = 2:1), followed by addition of 20 mL
of a 2 M sodium hydroxide solution. After stirring for 12 hours, the reaction mixture was concentrated under reduced pressure, added dropwise with 1 M hydrochloric acid until = CA 02806324 2013-01-21 the pH of the reaction mixture was between 5 and 7. The reaction mixture was filtered and the filter cake was dried in vacuum to obtain 7-tert-butoxycarbony1-3-(trifluoromethyl)-6,8-dihydro-5H-imidazo[1,5-a]pyrazine- 1 -carboxylic acid 20a (2 g, yield 50.0%) as a light yellow solid.
MS miz (ESI): 334.1 [M+1]
Step 2 tert-butyl 1-(morpholine-4-carbony1)-3-(trifluoromethyl)-6,8-dihydro-5H-imidazo [1, 5-a]pyrazine -7-c arboxy late 7-Tert-butoxycarbony1-3-(trifluoromethyl)-6,8-dihydro-5H-imidazo [1,5-a]pyrazine -1-carboxylic acid 20a (330 mg, 1 mmol) was dissolved in 5 mL of N, N-dimethylformamide, followed by addition of 0-(1-benzotriazoly1)-N, N, N' , N

-tetramethyluronium hexafluorophosphate (756 mg, 2 mmol), morpholine (174 mg, mmol) and N, N-diisopropylethylamine (0.5 mL, 3 mmol). After stirring for 12 hours, the reaction mixture was concentrated under reduced pressure, added with 20 mL
of saturated ammonium chloride solution, extracted with dichloromethane (20 mL
x3). The organic phase was combined, dried over anhydrous sodium sulfate and filtered.
The filtrate was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A to obtain tert-butyl 1-(morpho line-4-carbony1)-3-(tri fluoromethy 1)-6, 8- dihy dro-5H-imi dazo [1,5-a]pyrazine-7-carboxylate 20b (400 mg, yield 100.0%) as a yellow solid.
MS m/z (ESI): 405.1 [M-1]
Step 3 morpholino- [3 -(trifluoromethyl)-5,6,7,8-tetrahydroimidazo [1,5 -a]pyrazin-1-yl]methanone hydrochloride Tert-butyl 1-(morpholine -4-carbony1)-3-(trifluoromethy 1)-6,8-dihydro-5H-imidazo [1,5-a]pyrazine-7-carboxylate 20b (470 mg, 1.27 mmol) was dissolved in mL of a 2 M solution of hydrogen chloride in 1,4-dioxane. After stirring for 12 hours, the reaction mixture was concentrated under reduced pressure to obtain crude morpholino- [3 -(trifluoromethyl)-5,6,7,8-tetrahydroimidazo [1,5-a]pyrazin-1-yl]methanone hydrochloride 20c (300 mg) as a light yellow oil. The product was used directly in the next reaction without purification.
Step 4 morpholino- [3 -(trifluoromethyl)-5,6,7,8-tetrahydroimidazo [1,5 -a]pyrazin-1-yl]methanone Crude morpholino- [3 -(trifluoromethyl)-5,6,7,8-tetrahy dro imidazo [1 ,5 -a]pyrazin-1-yl]methanone hydrochloride 20c (330 mg, 1 mmol) was dissolved in 10 mL of ethyl acetate, followed by addition of potassium carbonate (10 g, 72 mmol). After stirring for 4 hours, the reaction mixture was filtered and the filtrate was concentrated under reduced pressure to obtain crude morpholino-[3-(trifluoromethyl)-5,6,7,8-tetrahydroimidazo[1,5-a]pyrazin- 1 -yl]methanone 20d (300 mg) as a light yellow solid.
The product was used directly in the next reaction without purification.
41 == = CA 02806324 2013-01-21 Step 5 4- [[4-fluoro-3- [1 -(morpholine-4-carbonyl)-3 -(trifluoromethyl)-6,8-dihydro-imidazo [1,5 -a] pyrazine-7-carbonyl] phenyl]methyl] -2H-phthalazin-1 -one 2-Fluoro-5-[(4-oxo-3H-phthalazin-1-yOmethylibenzoic acid la (390 mg, 1.30 mmol) was dissolved in 10 mL of N, N-dimethylformamide, followed by addition of 0-(1-benzotriazoly1)-N, N, N', N'-tetramethyluronium hexafluorophosphate (983 mg, 2.60 mmol), crude morpholino- [3 -(trifluoromethyl)-5,6,7,8-tetrahydroimidazo[1,5-a]pyrazin-1-yl]methanone 20d (400 mg, 1.30 mmol) and N, N-diisopropylethylamine (0.7 mL, 3.90 mmol). After stirring for 12 hours, the reaction mixture was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A to obtain 4-[[4-fluoro-3-[1-(morpholine-4-carbony1)-3 -(trifluoromethyl)-6,8-dihydro-5H-imidazo [1,5-a]pyrazine-7-carbonyl Iphenyl]methy11-2H-phthalazin- 1 -one 20 (150 mg, yield 20.0%) as a light yellow solid.

MS m/z (ESI): 585.2 [M+1]

1H NMR (400 MHz, DMSO-d6): 6 12.58 (br. s, 1H), 8.27 (d, 1H), 7.83-7.98 (m, 3H), 7.48-7.50 (m, 2H), 7.27-7.32 (m, 1H), 5.07 (s, 1H), 4.82 (s, 1H), 4.27-4.35 (m, 2H), 4.26-4.27 (m, 1H), 4.07-4.12 (m, 3H), 3.59-3.66 (m, 6H), 3.17-3.18 (m, 2H) Example 21 N-methyl-7- [3- [(4-oxo-3H-phthalazin-l-y Omethyl]benzoyl] -3 -(trifluoromethyl)-6,8-dihydro-5H-imidazo [1 ,5 -carboxamide 40 .r /
0 ,..õ,,,TXNH

/NF

N \ so ,17 H
0 =--N

io OH

F F
13a 15a 21 F

3-[(4-0xo-3H-phthalazin- 1 -yl)methyl]benzoic acid 15a (186 mg, 0.67 mmol) was dissolved in 20 mL of N, N-dimethylformamide, followed by addition of 1-hydroxybenzotriazole (98 mg, 0.73 mmol), crude N-methy1-3-(trifluoromethyl)-5 ,6,7,8-tetrahydroimidazo [1,5-a]pyrazine-1-carboxamide 13a (150 mg, 0.61 mmol), 1-ethyl-(3-dimethylaminopropyl) carbodiimide hydrochloride (173 mg, 0.91 mmol) and triethylamine (253 tL, 1.82 mmol). After stirring for 12 hours, the reaction mixture was was concentrated under reduced pressure, added with 50 mL of H20 and extracted with ethyl acetate (50 mL x3). The organic phase was combined, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and
42 = CA

the resulting residue was purified by thin layer chromatography with elution system A to obtain N-methyl-7- [3- [(4-oxo-3H-phthalazin-1 -y methyl] benzoy1]-3-(trifluoromethyl)-6,8-dihydro-5H-imidazo [1,5-a]pyrazine- 1 -carboxamide 21(280 mg, yield 90.0%) as a light yellow solid.

MS m/z (ESI): 511.2 [M+l]

NMR (400 MHz, CDC13): 6 11.80 (br. s, 1H), 8.49 (d, 1H), 7.89 (m, 2H), 7.79 (t, 1H), 7.52 (m, 2H), 7.43 (m, 2H), 5.26 (s, 2H), 4.35 (s, 2H), 4.22 (m, 4H), 3.01 (m, 3H) Example 22 7- [2-fluoro-5-[(4-oxo-3H-phthalazin-1-yOmethyllbenzoyll-N,N-dimethyl-3-(trifluoromethyl)-6,8-dihydro-5H-imidazo[1,5-a]pyrazine-1-carboxamide 40 ,1\1 y N
F
F F

1 N \ 0 HN-rt N'1111 step 1 step 2 step 3 F F HCI F
20a 22a 22b 22c ,1\111H
NH

0 0 r.,tN
step 4 N N
io OH LN
1a 22 F
F FA"-F

Step 1 tert-butyl 1 -(dimethylcarbamoy1)-3 -(trifluoromethyl)-6,8-dihydro-5H-imidazo [1,5-c]pyrazine-7-carboxylate 7-Tert-butoxycarbony1-3 -(trifluoromethyl)-6,8- dihydro-5H-imidazo [1,5-a]pyrazine-1-carboxylic acid 20a (330 mg, 1 mmol) was dissolved in 5 mL of N, N-dimethylformamide, followed by addition of 0-(1-benzotriazoly1)-N, N, N', N' -tetramethyluronium hexafluorophosphate (756 mg, 2 mmol), dimethylamine hydrochloride (156 mg, 2 mmol) and N, N-diisopropylethylamine (387 mg, 3 mmol).

After stirring for 12 hours, the reaction mixture was concentrated under reduced pressure, added with 50 mL of ethyl acetate, and washed successively with saturated ammonium chloride solution (30 mL), saturated sodium chloride solution (20 mL
x3).

The organic phase was combined, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure to obtain crude tert-butyl 1 -(dimethylcarbamoy1)-3 -(trifluoromethyl)-6,8-dihydro-5H-imidazo [1,5-c]pyrazine-7-
43 = = CA 02806324 2013-01-21 carboxylate 22a (362 mg) as a light yellow solid. The product was used directly in the next reaction without purification.
MS m/z (ESI): 363.1 [M+1]
Step 2 N,N-dimethy1-3-(trifluoromethyl)-5,6,7,8-tetrahydroimidazo[1,5-a]pyrazine-1-carboxa mide hydrochloride Crude tert-butyl 1 -(dimethylcarbamoy1)-3 -(trifluoromethyl)-6,8-dihydro-5H-imidazo [1,5-a]pyrazine-7-carboxylate 22a (362 mg, 1 mmol) was dissolved in 3 mL of a 2 M solution of hydrogen chloride in 1,4-dioxane. After stirring for 12 hours, the reaction mixture was concentrated under reduced pressure to obtain crude N,N-dimethy1-3 -(trifluoromethyl)-5,6,7,8-tetrahydroimidazo [1,5 -a]pyrazine-1 -carboxamide hydrochloride 22b (262 mg) as a light yellow solid. The product was used directly in the next reaction without purification.
Step 3 N,N-dimethy1-3 -(trifluoromethyl)-5,6,7,8-tetrahy droimidazo [1,5 -a] pyrazine-l-carboxa mide N,N-dimethy1-3-(trifluoromethyl)-5,6,7,8-tetrahydroimidazo [1,5 -a] pyrazine-1-carboxamide hydrochloride 22b (234 mg, 0.80 mmol) was dissolved in 10 mL of ethyl acetate, followed by addition of potassium carbonate (10 g, 72 mmol). After stirring for 4 hours, the reaction mixture was filtered and the filtrate was concentrated under reduced pressure to obtain crude N,N-dimethy1-3-(trifluoromethyl)-5,6,7,8-tetrahydroimidazo[1,5-c]pyrazine- 1 -carboxamide 22c (200 mg) as a light yellow solid.
The product was used directly in the next reaction without purification.
Step 4 7- [2-fluoro-5- [(4-oxo-3H-phthalazin-1-yl)methyllbenzoyl] -N,N-dimethy1-3 -(trifluoromethy 1)-6,8- dihydro-5H-imidazo [1,5-alpyrazine-1 -carboxamide 2-Fluoro-5-[(4-oxo-3H-phthalazin- 1 -yOmethyl]benzoic acid la (300 mg, 1 mmol) was dissolved in 5 mL of N, N-dimethylformamide, followed by addition of 0-(1-benzotriazoly1)-N, N, N', N'-tetramethyluronium hexafluorophosphate (756 mg, 2 mmol), N,N-dimethy1-3-(trifluoromethyl)-5,6,7,8-tetrahydroimidazo [1,5 -a]
pyrazine-1-carboxamide 22c (200 mg, 0.80 mmol) and N, N-diisopropylethylamine (0.5 mL, 3 mmol). After stirring for 12 hours, the reaction mixture was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A to obtain 742-fluoro-5-[(4-oxo-3H-phthalazin- 1 -yl)methyl]benzoy1]-N,N-dimethy1-3 -(trifluoromethy 1)-6,8-dihydro-5H-imidazo [1,5-a]pyrazine-1-carboxamide 22 (45 mg, yield 11.0%) as a light yellow solid.
MS m/z (ESI): 543.1 [M+1]
II-I NMR (400 MHz, DMSO-d6): 6 12.58 (hr. s, 1H), 8.27 (d, 1H), 7.83-7.96 (m, 3H), 7.49-7.51 (m, 2H), 7.27-7.31 (m, 1H), 4.80 (s, 1H), 4.35 (s, 2H), 4.26-4.27 (m, 1H), 4.05-4.07 (m, 1H), 3.66-3.67 (m, 1H), 3.30-3.39 (m, 6H), 2.88-2.97 (m, 2H)
44 = = CA 02806324 2013-01-21 Example 23 44 [3 43 -(difluoromethyl)-6,8-dihydro-5H41,2,41triazolo [4,3-a]pyrazine-7-carbonyl] -4-fluoro-phenyl] methyl] -2H-phthalazin-l-one =NH

N

F

F F

HN N
N,NH2 re step 1step 3CFCOOF H step 2 2 F/¨F F Fstep 4 23a 23b 23c 23d 23 F F

Step 1 2,2-difluoro-N'-pyrazin-2-yl-acetohydrazide difluoroacetate Pyrazin-2-ylhydrazine 23a (1 g, 9 mmol) was added in an eggplant-shaped bottle (25 mL). followed by dropwise addition of difluoroacetic anhydride (4 g, 22.98 mmol) at 0 C. After stirring at room temperature for 3 hours, the reaction mixture was concentrated under reduced pressure to obtain crude 2,2-difluoro-N'-pyrazin-2-yl-acetohydrazide difluoroacetate 23b (2 g) as a brown oil.

The product was used directly in the next reaction without purification.

Step 2 3-(difluoromethy1)41,2,4]triazolo [4,3-c]pyrazine 2,2-Difluoro-N'-pyrazin-2-yl-acetohydrazide difluoroacetate 23b (2 g, 0.01 mol) was dissolved in 10 mL of polyphosphoric acid. After stirring at 140 C for 7 hours, the reaction mixture was cooled to 50 C and stirred for another 12 hours. The reaction mixture was poured into 50 mL of ice-water while hot, added dropwise with 30%

aqueous ammonia until the pH of the reaction mixture was between 7 and 8, and extracted with ethyl acetate (30 mL x3). The organic phase was combined, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure. The resulting residue was dissolved in 30 mL of ethyl acetate and added with activated carbon. After stirring for 30 minutes, the mixture was filtered and the filtrate was concentrated under reduced pressure to obtain 3-(difluoromethyl)-[1,2,4]triazolo[4,3-a]pyrazine 23c (460 mg, yield 30%) as a yellow solid.

MS m/z (EST): 171 [M+1]

Step 3 3-(difluoromethyl)-5,6,7,8-tetrahydro- [1,2,4]triazolo [4,3-a]pyrazine 3-(Difluoromethy1)41,2,4]triazolo[4,3-a]pyrazine 23c (460 mg, 2.70 mmol) was dissolved in 10 mL of methanol, followed by addition of Pd-C (10%, 46 mg), and the
45 reactor was purged with hydrogen for three times. After stirring for 3 hours, the reaction mixture was filtered and the filter cake was washed with methanol (10 mL). The filtrate was concentrated under reduced pressure to obtain crude 3-(difluoromethyl)-5,6,7,8 -tetrahydro-[1,2,41triazolo[4,3-a]pyrazine 23d (400 mg) as a light yellow oil.
The product was used directly in the next reaction without purification.
MS m/z (ESI): 175.0 [M+11 Step 4 4-[[3-[3 -(difluoromethyl)-6,8-dihydro-5H-[1,2,4]triazolo[4,3-a]pyrazine-7-carbonyl]-4-fluoro-phenylimethy1]-2H-phthalazin-1 -one 2-Fluoro-5-[(4-oxo-3H-phthalazin-1-yOmethyl]benzoic acid la (685 mg, 2.30 mmol) was dissolved in 10 mL of N, N-dimethylformamide, followed by addition of 0-(1-benzotriazoly1)-N, N, N', N'-tetramethyluronium hexafluorophosphate (1.10 g, 3.45 mmol), crude 3-(difluoromethyl)-5,6,7,8-tetrahydro-[1,2,4]triazolo[4,3-c]pyrazine 23d (400 mg, 2.30 mmol) and N, N-diisopropylethylamine (1.2 mL, 6.90 mmol).
After stirring for 12 hours, the reaction mixture was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A to obtain 44[343-(difluoromethyl)-6,8-dihydro-5H41,2,4]triazolo [4,3 -c]pyrazine-7-carbony1]-4-fluoro-phenyl]methy1]-2H-phthalazin-1-one 23 (200 mg, yield 20.0%) as a white solid.
MS tri/z (ESI): 454.6 [M+1]

Example 24 N-(cyclopropylmethyl)-7- [2-fluoro-5- [(4-oxo-3H-phthalazin-1-yl)methyl]benzoyl]-3-(trifluoromethyl)-6,8-dihydro-5H-imidazo [1,5-a]pyrazine-1-carboxamide NH 0 0 re4NH
a F F F
46 40)00 0 0 r-4 HN-M --XN o m N o 20a F F 1 step 1 24a F --F step 2 HCI F F 24b step 3 HN
24c F F

NHSNH 0 step 4 0 0 r-4 NH
* OH
NYN

la 24 r F
Step 1 tert-butyl 1 -(cyclopropylmethylc arbamoy1)-3 -(trifluoromethyl)-6,8-dihy dro-imidazo [1,5-a]pyrazine-7-carboxylate 7-Tert-butoxycarbony1-3 -(trifluoromethyl)-6,8-dihydro-5H-imidazo [1,5-a]pyrazine -1-carboxylic acid 20a (330 mg, 1 mmol) was dissolved in 5 mL of N, N-dimethylformamide, followed by addition of 0-(1-benzotriazoly1)-N, N, N', N'-tetramethyluronium hexafluorophosphate (756 mg, 2 mmol), cyclopropylmethylamine (142 mg, 2 mmol) and N, N-diisopropylethylamine (0.5 mL, 3 mmol). After stirring for 12 hours, the reaction mixture was concentrated under reduced pressure and added with 50 mL of ethyl acetate and washed successively with saturated ammonium chloride (15 mL x3) and saturated sodium chloride solution (10 mL).
The organic phase was collected, dried over anhydrous sodium sulfate and filtered.The filtrate was concentrated under reduced pressure to obtain crude tert-butyl 1 -(cyclopropylmethy Icarbamoy1)-3 -(trifluoromethyl)-6,8-dihydro-5H-imidazo [1,5-a]
pyrazine-7-carboxylate 24a (300 mg) as a brown-red oil. The product was used directly in the next reaction without purification.
MS m/z (ESI): 389.1 [M+1]
Step 2 N-(cyclopropylmethyl)-3-(trifluoromethyl)-5,6,7,8-tetrahydroimidazo[1,5-a]pyrazine-1-carboxamide hydrochloride Crude tert-butyl 1-(cyclopropylmethylcarbamoy1)-3-(trifluoromethyl)-6,8-dihydro-5H-imidazo[1,5-a]pyrazine-7-carboxylate 24a (300 mg, 0.77 mmol) was dissolved in 20 mL of a 2 M solution of hydrogen chloride in 1,4-dioxane. After stirring for 12 hours, the reaction mixture was concentrated under reduced pressure to obtain crude N-(cyclopropylmethyl)-3-(trifluoromethyl)-5,6,7,8-tetrahydroimidazo [1,5 -c]
pyrazine-1-carboxamide hydrochloride 24b (250 mg) as a light yellow oil. The product was used directly in the next reaction without purification.
MS m/z (ESI): 289.1 [M+1]
Step 3
47 = . CA 02806324 2013-01-21 N-(cyclopropylmethyl)-3-(trifluoromethyl)-5,6,7,8-tetrahydroimidazo[1,5-a]pyrazine-1-carboxamide N-(cyclopropylmethyl)-3 -(trifluoromethyl)-5,6,7,8-tetrahydroimidazo [1,5-a]

pyrazine- 1 -carboxamide hydrochloride 24b (250 mg, 0.77 mmol) was dissolved in 10 mL of dichloromethane, followed by addition of potassium carbonate (320 mg, 2.30 mmol). After stirring for 4 hours, the reaction mixture was filtered and the filtrate was concentrated under reduced pressure to obtain crude N-(cyclopropylmethyl)-3-(trifluoromethyl)-5,6,7,8-tetrahydroimidazo [1,5 -a]pyrazine-l-carboxamide 24c (250 mg) as a yellow solid. The product was used directly in the next reaction without purification.

Step 4 N-(cyclopropylmethyl)-742-fluoro-5-[(4-oxo-3H-phthalazin-1-yOmethyl]benzoyl]-3-(trifluoromethyl)-6,8-dihydro-5H-imidazo [1,5-a]pyrazine-1-carboxamide 2-Fluoro-5-[(4-oxo-3H-phthalazin-1-yl)methyl]benzoic acid la (300 mg, 1 mmol) was dissolved in 5 mL of N, N-dimethylformamide, followed by addition of 0-(1-benzotriazoly1)-N, N, N', N'-tetramethyluronium hexafluorophosphate (756 mg, 2 mmol), crude N-(cyclopropylmethyl)-3-(trifluoromethyl)-5,6,7,8-tetrahydroimidazo[1,5-a]pyrazine-1-carboxamide 24c (250 mg, 0.87 mmol) and N, N-diisopropylethylamine (0.5 mL, 3 mmol). After stirring for 12 hours, the reaction mixture was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A to obtain N-(cyclopropylmethyl)-7-[2-fluoro-5-[(4-oxo-3H-phthalazin-1-yOmethyl]benzoyl]-(trifluoromethyl)-6,8-dihydro-5H-imidazo [1,5-alpyrazine-1-carboxamide 24 (150 mg, yield 30.0%) as a light yellow solid.

MS miz (ESI): 569.2 [M+1]

Example 25 7- [2-fluoro-5- [(4-oxo-3H-phthalazin-1 -yl)methyl]benzoyl] -3-(trifluoromethyl)-6,8-dihydro-5H- imidazo [1,5-a]pyrazine-l-c arbonitrile o S , r o CN

L\l-/
F

NH
411 r, .

101 .NINH
+ io 0 40 step 1 step 2 F4¨F OH F

10a 25a la Step 1
48 = = CA 02806324 2013-01-21 3-(trifluoromethyl)-5,6,7,8-tetrahydroimidazo [1,5 -a] pyrazine-l-c arbonitrile 3 -(Trifluoromethyl)-5,6,7,8-tetrahydroimidazo [1,5 -a]pyrazine-l-carboxamide 10a (100 mg, 0.43 mmol) was dissolved in 5 mL of phosphorus oxychloride. The reaction mixture was heated to reflux for 4 hours. The reaction mixture was concentrated under reduced pressure, added with 10 mL of saturated sodium carbonate solution and extracted with ethyl acetate (25 mL x3). The organic phase was combined, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure to obtain crude 3-(trifluoromethyl)-5,6,7,8-tetrahydroimidazo[1,5-a]pyrazine-1-carbonitrile 25a (100 mg) as a brown solid. The product was used directly in the next reaction without purification.

MS m/z (ESI): 217.0 [M+11 Step 2 7- [2-fluoro -5- [(4-oxo-3H-phthalazin-1 -y 1)methyl] benzoy1]-3 -(trifluoromethyl)-6,8-dihy dro-5H-imidazo [1,5 -a] pyrazine-l-c arbonitrile 2-Fluoro-5-[(4-oxo-3H-phthalazin-1-yl)methyl]benzoic acid la (210 mg, 0.70 mmol) was dissolved in 5 mL of N, N-dimethylformamide, followed by addition of 0-(1-benzotriazoly1)-N, N, N', N'-tetramethyluronium hexafluorophosphate (350 mg, 0.92 mmol), crude 3-(trifluoromethyl)-5,6,7,8-tetrahydroimidazo[1,5-a]pyrazine-carbonitrile 25a (100 mg, 0.46 mmol), and N, N-diisopropylethylamine (250 pt, 1.18 mmol). After stirring for 12 hours, the reaction mixture was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A to obtain 742-fluoro-5-[(4-oxo-3H-phthalazin-1-yOmethyl]benzoy11-3-(trifluoromethyl)-6,8-dihydro-5H-imidazo[1,5-a]pyrazine-1-carbonitrile 25 (50 mg, yield 21.9%) as a white solid.

MS rniz (ESI): 496.6 [M+1]

Example 26 [[4-fluoro-3 43-(2,2,2-trifluoroethyl)-6,8-dihydro-5H41,2,4]triazolo [4,3 -alpyrazine-7-carbonyl]phenyl] methyl]-2H-phthalazin- 1-one = NF

NH
HN-Th---%N= N 0 =NH 0 F step 1 F io OH step 2 410 26a 26b F la F 26 F
49 = = CA 02806324 2013-01-21 Step 1 3 -(2,2,2-trifluoroethyl)-5,6,7,8-tetrahydro- [1,2,4]triazolo [4,3 -a]pyrazine 3-(2,2,2-trifluoroethy1)41,2,4]triazolo[4,3-alpyrazine 26a (464 mg, 2.29 mmol, prepared according to a known method "Journal of Medicinal Chemistry, 2005, 48(1), 141-151") was dissolved in 20 mL of methanol, followed by addition of Pd-C
(10%, 200 mg). and the reactor was purged with hydrogen for three times. After stirring for 3 hours, the reaction mixture was filtered and the filtrate was concentrated under reduced pressure to obtain crude 3-(2,2,2-trifluoroethyl)-5,6,7,8-tetrahydro-[1,2,4]triazolo[4,3-a]pyrazine 26b (480 mg) as a colorless oil. The product was used directly in the next reaction without purification.
Step 2 [[4-fluoro -343-(2,2,2-trifluoroethyl)-6,8-dihydro -5H- [1,2,4]triazolo [4,3-alpyrazine-7-carbonyllphenyl]methy1]-2H-phthalazin-1-one 2-Fluoro-5-[(4-oxo-3H-phthalazin-1-yl)methyl]benzoic acid la (801 mg, 2.69 mmol) was dissolved in 25 mL of N, N-dimethylformamide, followed by addition of 0-(1-benzotriazoly1)-N, N, N', N'-tetramethyluronium hexafluorophosphate (1.27 g, 3.36 mmol), crude 3 -(2,2,2-trifluoroethyl)-5 ,6,7,8-tetrahydro-[1,2,4]triazolo[4,3-a]pyrazine 26b (460 mg, 2.24 mmol) and N, N-diisopropylethylamine (0.8 mL, 4.48 mmol). After stirring for 12 hours, the reaction mixture was concentrated under reduced pressure, added with 30 mL of H20 and extracted with ethyl acetate (30 mL x3). The organic phase was combined, concentrated under reduced pressure, added with 30 mL of ethyl acetate, washed with saturated sodium chloride solution (20 mL), dried over anhydrous sodium sulfate and filtered.
The filtrate was concentrated under reduced pressure and the resulting residue was purified by thin layer chromatography with elution system A to obtain [[4-fluoro-343 -(2,2,2-trifluoroethyl)-6,8-dihydro-5H- [1,2,41triazolo [4,3-a]pyrazine-7-carbony 1] phenyl]methyl] -2H-phthalazin- 1 -one 26 (240 mg, yield 22.1%) as a white solid.
MS miz (ESI): 486.6 [M+1]
TEST EXAMPLES

BIOLOGICAL ASSAYS

Example 1 Assay for determining the Enzyme Activity of PAPR
The following in vitro screening assay is used to determine the activity of the compounds of the present invention for inhibiting the enzyme activity of PAPR.
The assay described below is to determine the activity of the compounds of the present invention for inhibiting the enzyme activity of PAPR by using the Trevigen HT
F homologous poly (adenosine diphosphate-ribose) polymerase inhibition assay kit (TREVIGEN HT F homogeneous PARP Inhibition Assay Kit, No. 4690-096-K). The
50 assays are based on the NAD+ amount need to be consumed during the DNA repair process, which is also used in another reaction for catalyzing the substrate without fluorescence activity into molecules with high fluorescence activity.
Therefore, the NAD+ level in the reaction system can be learned by measuring the enhancement degree of the fluorescence signal, and then the inhibition degree of the test compound on the enzyme activity of PARP was calculated.
The instructions of TREVIGEN HT F homologous poly (adenosine diphosphate-ribose) polymerase inhibition assay kit can be used as reference for the detailed operation of the assays as well as the preparation of the reagents, such as the reaction mixture (reaction mix), cycling reaction mixture (cycling mix), a buffer solution (buffer), and the like.
The procedures for the assay are summarized as follows: The tested compounds were dissolved in dimethylsulfoxide and then diluted with 1 x buffer to the concentration desired in the experiment. 25 [LL of a 200 nM NAD+ solution was added to a 96-well round bottomed plate, followed by the addition of 1 [t1_, of tested compounds solution, and the control of replicate wells were installed. Then 25 1_, of the reaction mixture containing DNA, PARP enzyme and reaction buffer was added into each well.
After incubating for 30 minutes at room temperature, 50 [IL of cycling reaction mixture was added into each well and incubated in the dark at room temperature for 15 to 40 minutes.
Then 50 [IL of stop solution was added into each well and the fluorescence values of each well were read on an ELIASA (Ex544 nm, Em590 nm). The inhibition rate of the test compound on the enzyme activity of PARP could be calculated by the standard curve equation of NAD+.
The IC50 values of the compounds could be calculated by the inhibition rate at different concentrations.
Example compounds No. IC50 (PARP-1)/ iM
1 0.015 2 0.005 3 0.052 15 0.0023 19 0.0102 Conclusion: The preferable compounds of the present invention have significantly inhibition activity on the proliferation inhibition of PARP-1 kinase.

Example 2 Cell Proliferation Inhibition Assay The following assay is to determine the activity of the compounds of the present invention for inhibiting the proliferation of triple negative phenotype of breast cancer cell line MDA-MB-436 in vitro.
The in vitro cellular assay described below is to determine the activity of the tested compounds for inhibiting the proliferation of triple negative phenotype of breast cancer cell. The inhibition activity is represented by the IC50 value.
51 The procedures for the assay are summarized as follows: The MDA-MB-436 cells were seeded to 96-well cell culture plate at a suitable cell concentration (e.g. 3000 cells/m1 medium) by using DMEM F12 with 10% FBS (both purchased from Gibco) as complete medium. Under the conditions of 37 C and 5% carbon dioxide, the cells were cultured in constant temperature incubator and grew overnight. The tested compounds were dissolved in dimethylsulfoxide and then diluted with culture medium without FBS
to the concentration desired in the asaays. After the cells adhere to the walls, the cell culture medium was replaced by fresh culture medium, in which the tested compounds at serial concentrations (general 7 to 9 concentrations) was contained. Then the cell plates were cultured for continuous 72 hours under the conditions of 37 C and 5%
carbon dioxide. 72 hours later, the activity of the tested compounds for inhibiting the cell proliferation was determined by using CCK8 (Cell Counting kit-8, No.:
CK04, purchased from Dojindo) method.
IC50 values of the tested compounds were calculated by the data of inhibition rates of the tested compounds at different concentrations.

Example compounds No. IC50 (MDA-MB-436)/ tM

1 0.0008 3 0.19 5 0.32 7 0.071 10 0.14 12 0.59 13 0.12 15 0.0009 16 0.099 17 0.061 18 0.61 19 0.049 21 0.78 22 0.65 23 0.002 24 0.072 26 0.003 Conclusion: The preferable compounds of the present invention have significantly inhibition activity on the proliferation inhibition of MDA-MB-436 cell.

PHARMACOKINETICS ASSAY
Test Example 1: The pharmacokinetics assay of the compounds of Example 7, Example 13 and Example 19 of the invention.
1. Abstract
52 The compounds of Example 7, Example 13 and Example 19 were administrated intragastrically or intravenous injection to rats to determine the drug concentration in plasma at different time points by LC/MS/MS method and using SD rats as test animals.
The pharmacokinetic behavior of the compounds of the present invention was studied and evaluated in rats.
2. Protocol 2.1 Samples Compounds of Example 7, Example 13 and Example 19 2.2 Test animals 24 Healthy adult SD rats, male and female in half, were purchased from SINO-BRITSH SIPPR/BK LAB. ANIMAL LTD., CO, Certificate No.: SCXK (Shanghai) 2003-0002.
2.3 Preparation of the tested compounds The intragastrical administration group: the right amount of tested compounds were weighted and dissolved in 0.5 mL of DMSO, diluted with physiological saline to 10mL
and prepared to 1.5 mg/mL.
The intravenous injection administration group: the right amount of tested compounds were weighted and added into 0.5% CMC-Na to prepare a 1.5 mg/mL
suspension.
2.4 Administration After an overnight fast, 24 healthy adult SD rats, male and female in half, were administered intragastrically at a dose of 15.0 mg / kg and an administration volume of 10 mL/kg.
2.5 Sample Collection The intragastrical administration group: blood samples (0.2 mL) were taken from orbital sinus at pre administration and at 0.25 hour, 0.5 hour, 1.0 hour, 1.5 hours, 2.0 hours, 3.0 hours, 4.0 hours, 6.0 hours, 7.0 hours, 9.0 hours, 12.0 hours and 24.0 hours post administration, stored in heparinized tubes and centrifuged for 20 minutes at 3,500 rpm to separate plasma. The plasma samples were stored at -20 C. The rats were fed at 2 hours after administration.
The intravenous injection administration group: blood samples (0.2 mL) were taken from orbital sinus at pre administration and at 2 minutes, 15 minutes, 0.5 hour, 1.0 hour, 2.0 hours, 3.0 hours, 4.0 hours, 6.0 hours, 8.0 hours, 12.0 hours and 24.0 hours post administration, stored in heparinized tubes and centrifuged for 20 minutes at 3,500 rpm to separate plasma. The plasma samples were stored at -20 C.
3. Operation 20 [iL of rat blank plasmas taken at various time points after administration were added with 50 !IL of internal standard solution and 140 IA, of methanol and mixed for 3 minutes by a vortexer. The mixture was centrifuged for 10 minutes at 13,500 rpm. 20 [iL
of the supernatant was analyzed by LC-MS/MS. The main pharmacokinetic parameters were calculated by software DAS 2Ø
53 =

4. Results of Pharmacokinetic Parameters Pharmacokinetic Parameters of the compounds of the present invention were shown as follows:

Pharmacokinetics Assay (15mg/kg) Mean Apparent Plasma Area Under Number oral Conc.
Curve Half-Life Residence Clearance Distributio Time n Volume bioavai lability Cmax AUC

CL/F Vz/F

(ng/mL) (ng/mL*h) t1/2(h) MRT(h) (lib/kg) (1/kg) 971+1400 4495+6671 3.87+4.03 12.7+15.4 15.4+12.4 103+134 oral gavage Example12.9 A 7 intravenous 34820+154540.94+0.26 1.25+0.53 0.52+0.29 0.64+0.19 injection 3073+719 4298+3252 6.01+2.27 1.87+0.53 4.47+3.78 49.9+52.9 Example oral gavage 13 16 8%
intravenous 29414+185435.05+1.34 0.89+0.44 0.72+0.45 4.70+2.17 injection Example 2335+1652 oral gavage 12557+123729.79+4.82 3.50+1.46 3.45+3.21 7.97+5.38 5 Conclusion: The example compounds of the present invention had better pharmacokinetic data and significantly improved pharmacokinetic properties.

ANTITUMOR EFFECT ASSAY

Test Example 2 the assay is to determine the antitumor effect of the compounds of the 10 present invention in mice 1. Purpose The therapeutic effect of the compounds of the present invention administered in combination with temozolomide (TMZ) on transplanted tumors of human colon carcinoma SW620 or human breast cancer cells MX-1 in nude mice was evaluated by 15 using BALB/cA-nude mice as test animals.

2. Test drug The compounds of Example 1 and Example 19 3. Test animals BALB/cA-nude mice, SPF, 16-20g, female(), were purchased from SINO-BRITSH

20 SIPPR/BK LAB. ANIMAL LTD., CO. Certificate No.:
SCXK(Shanghai) 2008-0016.

4. Experimental Procedures 4.1 Nude mice were adapt to the lab environment for three days.

4.2 The right rib of the nude mice was subcutaneously inoculated with colon carcinoma cells SW620. After tumors grew to 339 132 mm3, mice were randomly divided into 25 teams (d0).

Nude mice were subcutaneously inoculated with human breast cancer cells MX-1.
54 After tumors grew to 100 to 200 mm3, mice were randomly divided into teams (d0).
4.3 Dosage and dosage regimens were shown in the table below. The volume of tumors and the weight of the mice were measured and recorded for 2 to 3 times per week.
The volume of tumor (V) was calculated by the follow equation:
V = 1/2xaxb2 wherein: a, b represents length and width respectively.
The antitumor rate (%) = (C-T)/C(%) wherein: T, C represents the tumor volume of the experimental group (tested compounds) and blank control group at the end of the experiment respectively.
5. Dosage, dosage regimens and the results TMZ dosage dosage Time antitumor Compound cell (mg/kg) (mg/kg) (day) rate (%) Example 1 (oral gavage) colon 50 1 44 ++
+ TMZ (oral gavage) carcinoma Example 19 (oral gavage) colon 50 10 52 ++
+ TMZ (oral gavage) carcinoma 50 1 8 +++
Example 19 (oral gavage) breast 50 3 8 +++
+ TMZ (oral gavage) cancer 50 10 8 +++

Conlusion: the range of antitumor rate data (%) was shown as follows: "+":
50%60%;
"++": 60%-80%; "+++": 80%-100%. The tested compounds of the present invention administered in combination with temozolomide (TMZ) had significant antitumor rates on colon cancer cell SW620 and human breast carcinoma cell MX-1, which were all higher than 60%.
55

Claims (20)

1. The compounds of formula (I) or pharmaceutically acceptable salts thereof:

wherein:

A and B are taken together with the attached carbon atoms to form cycloalkyl, heterocyclyl, aryl or heteroaryl, wherein said cycloalkyl, heterocyclyl, aryl or heteroaryl are each independently and optionally substituted with one or more groups selected from the group consisting of alkyl, halogen, hydroxyl, alkoxyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -C(O)OR5, -OC(O)R5, -O(CH2)C(O)OR5, -C(O)R5, -NHC(O)R5, -NR6R7, -OC(O)NR6R7 and -C(O)NR6R7;

R1, R2, R3 or R4 is each independently selected from the group consisting of hydrogen, halogen, alkyl, cyano and alkoxyl, wherein said alkyl or alkoxyl is each independently and optionally substituted with one or more groups selected from the group consisting of halogen, hydroxyl, alkyl and alkoxyl;

D, E, or G is each independently selected from the group consisting of nitrogen atom and C(R8);

R5 is selected from the group consisting of hydrogen, alkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein said alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl is each independently and optionally substituted with one or more groups selected from the group consisting of alkyl, halogen, hydroxyl, alkoxyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, carboxyl and alkoxycarbonyl;

R6 or R7 is each independently selected from the group consisting of hydrogen, alkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein said alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl is each independently and optionally substituted with one or more groups selected from the group consisting of alkyl, halogen, hydroxyl, alkoxyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, carboxyl and alkoxycarbonyl;

or, R6 and R7 are taken togetherwith the attached N atom to form heterocyclyl, heterocyclyl is optionally substituted with one or more groups selected from the group consisting of alkyl, halogen, hydroxyl, alkoxyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, wherein said heterocyclyl contains one or more N, O or S(O)m heteratoms, and said carboxyl and alkoxycarbonyl;

R8 is selected from the group consisting of hydrogen, alkyl, halogen, hydroxyl, cyano, alkoxyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, benzyl, -C(O)OR5, -OC(O)R5, -O(CH2)n C(O)OR5, -(CH2)n NR6R7, -C(O)R5, -NHC(O)R5, -NR6R7, -OC(O)NR6R7 and -C(O)NR6R7, wherein said alkyl, alkoxyl, cycloalkyl, heterocyclyl, aryl, heteroaryl or benzyl is each independently and optionally substituted with one or more groups selected from the group consisting of alkyl, halogen, hydroxyl, alkoxyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, oxo, -C(O)OR5, -OC(O)R5, -O(CH2)n C(O)OR5, -C(O)R5, -NHC(O)R5, -NR6R7, -OC(O)NR6R7 and -C(O)NR6R7;
m is selected from the group consisting of 0, 1 and 2; and n is selected from the group consisting of 0, 1 and 2.
2. The compounds of formula (I) or pharmaceutically acceptable salts thereof according to claim 1, wherein A and B are taken together with the attached carbon atoms to form aryl, the preferable aryl is phenyl.
3. The compounds of formula (I) or pharmaceutically acceptable salts thereof according to claim 1, wherein R1 is hydrogen.
4. The compounds of formula (I) or pharmaceutically acceptable salts thereof according to claim 1, wherein R1 is halogen, preferably fluorine atom.
5. The compounds of formula (I) or pharmaceutically acceptable salts thereof according to claim 1, wherein R8 is selected from the group consisting of hydrogen, alkyl, halogen, cyano, -C(O)OR5, -(CH2)n NR6R7 and -C(O)NR6R7, wherein said alkyl is optionally substituted with one or more halogen atoms.
6. The compounds or pharmaceutically acceptable salts thereof according to claim 5, wherein R8 is trifluoromethyl.
7. The compounds of formula (I) or pharmaceutically acceptable salts thereof according to claim 1, wherein R1, R2, R3 or R4 is each independently hydrogen.
8. The compounds of formula (I) or pharmaceutically acceptable salts thereof according to claim 1, wherein R2, R3 or R4 is each independently hydrogen, R1 is halogen, preferably fluorine atom.
9. The compounds of formula (I) or pharmaceutically acceptable salts thereof according to any one of claims 1 to 8, wherein the compounds are selected from the group consisting of:

<1MG>
10. The preparation process for the compounds of formula (I) or the pharmaceutically acceptable saltss thereof according to claim 1, wherein said process comprising the steps of:

optionally hydrolyzing the compound of formula (IA) to a carboxylic acid, reacting the carboxylic acid with the compound of formula (IB) or a salt thereof to obtain the compound of formula (I);
wherein:
R a is selected from the group consisting of halogen, hydroxyl and alkoxyl;
A, B, D, E, G and R1 to R4 are defined as those in claim 1.
11. The pharmaceutical composition, comprising a therapeutically effective amount of the compounds of formula (I) or pharmaceutically acceptable salts thereof according to any on of claims 1 to 9, and a pharmaceutically acceptable carrier or excipient.
12. The use of the compounds of formula (I) or pharmaceutically acceptable salts thereof according to any one of claims 1 to 9, or the pharmaceutical composition according to claim 11 in the preparation of a PARP inhibitor.
13. The compounds of formula (I) or pharmaceutically acceptable salts thereof according to any one of claims 1 to 9, or the pharmaceutical composition according to claim 11 for use as a PARP inhibitor.
14. The methods for inhibiting PARP, comprising administrating the subject in need thereof a therapeutically effective amount of the compounds of formula (I) or pharmaceutically acceptable salts thereof according to any one of claims 1 to 9, or the pharmaceutical composition according to claim 11.
15. The use of the compounds of formula (I) or pharmaceutically acceptable salts thereof according to any one of claims I to 9, or the pharmaceutical composition according to claim 11 in the preparation of an adjuvant in the treatment of cancer or a medicament causing tumor cells sensitive to ionizing radiation or chemotherapy.
16. The compounds of formula (I) or pharmaceutically acceptable salts thereof according to any one of claims 1 to 9, or the pharmaceutical composition according to claim 11, for use as an adjuvant in the treatment of cancer or a medicament causing tumor cells sensitive to ionizing radiation or chemotherapy.
17. The use of the compounds of formula (I) or pharmaceutically acceptable salts thereof according to any one of claims 1 to 9, or the use of the pharmaceutical composition according to claim 11 in the preparation of a medicament for the treatment of cancer, wherein said cancer is selected from the group consisting of breast cancer, ovarian cancer, pancreatic cancer, prostate cancer, rectal cancer, liver cancer and colon cancer.
18. The compounds of formula (I) or pharmaceutically acceptable salts thereof according to any one of claims 1 to 9, or the pharmaceutical composition according to claim 11, for use as a medicament for the treatment of cancer, wherein said cancer is selected from the group consisting of breast cancer, ovarian cancer, pancreatic cancer, prostate cancer, rectal cancer, liver cancer and colon cancer.
19. The method for treating cancer, comprising administrating the subject in need thereof a therapeutically effective amount of the compounds of formula (I) or pharmaceutically acceptable salts thereof according to any one of claims 1 to 9, or the pharmaceutical composition according to claim 11.
20. The use according to any one of claims 12, 15 or 17, wherein the medicament is further co-administered with a therapeutically effective amount of drug selected from the group consisting of Temozolomide, Adriamycin, Taxol, Cisplatin, Carboplatin, Dacarbazine, Topotecan, Irinotecan, Gemcitabine and Bevacizumab.
CA2806324A 2010-08-09 2011-07-26 Phthalazinone ketone derivative, preparation method thereof, and pharmaceutical use thereof Active CA2806324C (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
CN201010248307.5 2010-08-09
CN2010102483075A CN102372716A (en) 2010-08-09 2010-08-09 Phthalazone derivative, its preparation method and application in medicine thereof
PCT/CN2011/001223 WO2012019427A1 (en) 2010-08-09 2011-07-26 Phthalazinone ketone derivative, preparation method thereof, and pharmaceutical use thereof

Publications (2)

Publication Number Publication Date
CA2806324A1 true CA2806324A1 (en) 2012-02-16
CA2806324C CA2806324C (en) 2019-02-19

Family

ID=45567315

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2806324A Active CA2806324C (en) 2010-08-09 2011-07-26 Phthalazinone ketone derivative, preparation method thereof, and pharmaceutical use thereof

Country Status (16)

Country Link
US (2) US9273052B2 (en)
EP (1) EP2604610B1 (en)
JP (1) JP5808408B2 (en)
KR (1) KR101829940B1 (en)
CN (2) CN102372716A (en)
AU (1) AU2011288876B2 (en)
BR (1) BR112013002220B1 (en)
CA (1) CA2806324C (en)
ES (1) ES2582315T3 (en)
HK (1) HK1174030A1 (en)
HU (1) HUE029275T2 (en)
PL (1) PL2604610T3 (en)
PT (1) PT2604610T (en)
RU (1) RU2564527C2 (en)
UA (1) UA111161C2 (en)
WO (1) WO2012019427A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2881395A4 (en) * 2012-08-01 2016-02-24 Shanghai Inst Materia Medica Piperazinotrizole compound, preparation method therefor, and use thereof in drug preparation

Families Citing this family (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102898377B (en) * 2012-02-14 2016-01-20 南京圣和药业股份有限公司 One class phthalazinone derivatives and uses thereof
CN102702108A (en) * 2012-06-27 2012-10-03 上海大学 1,2-dihydrophthalazin compound and synthetic method thereof
CN102863393A (en) * 2012-09-26 2013-01-09 上海大学 1,2-dihydro phthalazine compound and synthetic method thereof
WO2014102817A1 (en) * 2012-12-31 2014-07-03 Cadila Healthcare Limited Substituted phthalazin-1 (2h)-one derivatives as selective inhibitors of poly (adp-ribose) polymerase-1
KR101670126B1 (en) 2013-09-13 2016-10-27 일동제약(주) A novel phtalazinone derivatives and manufacturing process thereof
AR098414A1 (en) 2013-11-14 2016-05-26 Bristol Myers Squibb Co PIPERAZINAS DE PIRAZOLO REPLACED AS INHIBITORS OF CASEÍNA QUINASA 1 d / e
EP3325623B3 (en) 2015-07-23 2021-01-20 Institut Curie Use of a combination of dbait molecule and parp inhibitors to treat cancer
CN106749261A (en) * 2015-11-23 2017-05-31 中国科学院上海药物研究所 One class substituted triazole and piperazines PARP inhibitor and its production and use
WO2017101796A1 (en) * 2015-12-16 2017-06-22 四川科伦博泰生物医药股份有限公司 Phthalazinone derivative, and preparation method and use thereof
WO2017156350A1 (en) 2016-03-09 2017-09-14 K-Gen, Inc. Methods of cancer treatment
TWI771344B (en) * 2016-12-01 2022-07-21 大陸商江蘇恆瑞醫藥股份有限公司 Combination use of vegfr inhibitor and parp inhibitor in the preparation of a medicament for the treatment of gastric cancer
WO2018162439A1 (en) 2017-03-08 2018-09-13 Onxeo New predictive biomarker for the sensitivity to a treatment of cancer with a dbait molecule
KR102296703B1 (en) 2017-03-20 2021-09-01 포르마 세라퓨틱스 인크. Pyrrolopyrrole compositions as pyruvate kinase (pkr) activators
EP3610040A1 (en) 2017-04-12 2020-02-19 H. Hoffnabb-La Roche Ag A method for sequencing reaction with tagged nucleoside obtained via pictet spengler reaction
CN110914272A (en) * 2017-08-24 2020-03-24 江苏恒瑞医药股份有限公司 Crystal form of PARP-1 inhibitor and preparation method thereof
MX2020005659A (en) * 2017-12-06 2020-08-20 Jiangsu Hengrui Medicine Co Use of parp inhibitor in treating chemotherapy-resistant ovarian cancer or breast cancer.
CA3085660A1 (en) 2018-01-09 2019-07-18 Jiangsu Hengrui Medicine Co., Ltd. Method for preparing parp inhibitor and intermediate thereof
KR20200120915A (en) * 2018-01-26 2020-10-22 레코다티인더스트리아치미카이파마슈티카에스.피.에이. Triazole, imidazole and pyrrole condensed piperazine derivatives as modulators of mGlu5 receptors and their use
JP2021516229A (en) 2018-02-28 2021-07-01 ザ トラスティーズ オブ ザ ユニバーシティ オブ ペンシルバニア Low-affinity poly (AD-ribose) polymerase 1-dependent cytotoxic agent
WO2019175132A1 (en) 2018-03-13 2019-09-19 Onxeo A dbait molecule against acquired resistance in the treatment of cancer
US11058688B2 (en) * 2018-03-29 2021-07-13 Board Of Regents, The University Of Texas System Imidazopiperazine inhibitors of transcription activating proteins
BR112021005188A2 (en) 2018-09-19 2021-06-08 Forma Therapeutics, Inc. treating sickle cell anemia with a pyruvate kinase r activating compound
US20230055923A1 (en) 2018-09-19 2023-02-23 Forma Therapeutics, Inc. Activating pyruvate kinase r
TW202029961A (en) * 2018-10-22 2020-08-16 大陸商江蘇恒瑞醫藥股份有限公司 Use of ar antagonist combined with parp inhibitor in preparation of medicament for treating prostate cancer
JP7469304B2 (en) * 2018-11-16 2024-04-16 江▲蘇▼恒瑞医▲薬▼股▲フン▼有限公司 Pharmaceutical Compositions Comprising PARP Inhibitors
TW202110448A (en) * 2019-05-28 2021-03-16 大陸商江蘇恆瑞醫藥股份有限公司 Use of parp inhibitor in combination with vegfr inhibitor for treating ovarian cancer or breast cancer
CN112870365A (en) * 2019-11-29 2021-06-01 江苏恒瑞医药股份有限公司 Use of EZH2 inhibitor and/or PARP inhibitor in combination with chemotherapeutic drug for preparing medicine for treating tumor
WO2021148581A1 (en) 2020-01-22 2021-07-29 Onxeo Novel dbait molecule and its use
MX2022013304A (en) 2020-04-21 2022-12-15 Idience Co Ltd Crystalline forms of phthacaz1nonf compound.
CN111732594B (en) * 2020-08-18 2022-03-04 苏州富士莱医药股份有限公司 Preparation method of fluxaparide
KR20240012437A (en) * 2021-05-24 2024-01-29 지앙수 헨그루이 파마슈티컬스 컴퍼니 리미티드 Nitrogen-containing heterocyclic compounds, methods for their preparation and their applications in medicine
EP4370125A1 (en) * 2021-07-16 2024-05-22 Oregon Health and Science University Phthalazinone-based parp-1 inhibitors
EP4378938A1 (en) * 2021-07-29 2024-06-05 Shanghai Qilu Pharmaceutical Research and Development Centre Ltd. Novel parp7 inhibitor and use thereof
CN115650988A (en) * 2022-10-27 2023-01-31 江苏恒瑞医药股份有限公司 Preparation method of PARP inhibitor
WO2024109871A1 (en) * 2022-11-23 2024-05-30 江苏恒瑞医药股份有限公司 Pharmaceutically acceptable salt of nitrogen-containing heterocyclic compound, crystal form thereof, and preparation method therefor

Family Cites Families (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EA006300B1 (en) 2000-10-30 2005-10-27 Кудос Фармасеутикалс Лимитед Phthalazinone derivatives
DE60336890D1 (en) * 2002-02-19 2011-06-09 Ono Pharmaceutical Co CONDENSED PYRIDAZINE DERIVATIVE COMPOUNDS AND THE COMPOUNDS CONTAINING ACTIVE SUBSTANCES
AR043443A1 (en) 2003-03-07 2005-07-27 Merck & Co Inc PROCEDURE FOR THE PREPARATION OF TETRAHYDROTRIAZOLOPIRAZINS AND INTERMEDIATE PRODUCTS
GB0305681D0 (en) 2003-03-12 2003-04-16 Kudos Pharm Ltd Phthalazinone derivatives
GB2415430B (en) 2003-03-12 2006-07-12 Kudos Pharm Ltd Phthalazinone derivatives
MX2007002318A (en) 2004-08-26 2007-04-17 Kudos Pharm Ltd 4-heteroarylmethyl substituted phthalazinone derivatives.
GB0610680D0 (en) 2006-05-31 2006-07-12 Istituto Di Ricerche D Biolog Therapeutic compounds
KR101491998B1 (en) * 2006-12-28 2015-02-10 애브비 인코포레이티드 Inhibitors of poly(ADP-ribose)polymerase
US20090023727A1 (en) * 2007-07-05 2009-01-22 Muhammad Hashim Javaid Phthalazinone derivatives
AU2008289573B2 (en) 2007-08-21 2013-05-16 Merck Sharp & Dohme Corp. Heterocyclic compounds as dipeptidyl peptidase-IV inhibitors for the treatment or prevention of diabetes
CN101468988A (en) 2007-12-26 2009-07-01 上海恒瑞医药有限公司 Piperazine derivative, preparation thereof and use thereof in medicine
JP2011509962A (en) 2008-01-17 2011-03-31 グリュネンタール・ゲゼルシャフト・ミト・ベシュレンクテル・ハフツング Substituted sulfonamide derivatives

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2881395A4 (en) * 2012-08-01 2016-02-24 Shanghai Inst Materia Medica Piperazinotrizole compound, preparation method therefor, and use thereof in drug preparation

Also Published As

Publication number Publication date
WO2012019427A8 (en) 2012-05-10
HK1174030A1 (en) 2013-05-31
JP2013535491A (en) 2013-09-12
US20160151367A1 (en) 2016-06-02
RU2564527C2 (en) 2015-10-10
CN102686591B (en) 2014-03-19
CN102686591A (en) 2012-09-19
ES2582315T3 (en) 2016-09-12
WO2012019427A1 (en) 2012-02-16
US9273052B2 (en) 2016-03-01
KR101829940B1 (en) 2018-02-19
US20130131068A1 (en) 2013-05-23
RU2013106754A (en) 2014-09-20
JP5808408B2 (en) 2015-11-10
PL2604610T3 (en) 2016-11-30
HUE029275T2 (en) 2017-02-28
AU2011288876B2 (en) 2014-08-21
EP2604610A4 (en) 2013-12-25
PT2604610T (en) 2016-07-13
CA2806324C (en) 2019-02-19
CN102372716A (en) 2012-03-14
EP2604610A1 (en) 2013-06-19
EP2604610B1 (en) 2016-05-11
KR20130110149A (en) 2013-10-08
BR112013002220A2 (en) 2016-05-24
BR112013002220B1 (en) 2021-09-21
AU2011288876A1 (en) 2013-01-31
AU2011288876A2 (en) 2013-02-21
US9566277B2 (en) 2017-02-14
UA111161C2 (en) 2016-04-11

Similar Documents

Publication Publication Date Title
EP2604610B1 (en) Phthalazinone ketone derivative, preparation method thereof, and pharmaceutical use thereof
TWI527800B (en) 1-(arylmethyl)quinazoline-2,4(1h,3h)-diones as parp inhibitors and the use thereof
EP2968316B1 (en) 2-hydroxy-1-{4-[(4-phenylphenyl)carbonyl]piperazin-1-yl}ethan-1-one derivatives and related compounds as fatty acid synthase (fasn) inhibitors for the treatment of cancer
ES2770693T3 (en) Imidazopyridazine derivatives as casein kinase 1 delta / epsilon inhibitors
CN103703004B (en) As the Fourth Ring condensed of PARP inhibitor or the dihydro diaza * at five rings carbazole ketone
EA009875B1 (en) 6-alkenyl and 6-phenylalkyl substituted 2-quinolinones and 2-quinoxalinones as poly(adp-ribose) polymerase inhibitors
CN102952118B (en) Poly-(ADP-ribose) AG14361, preparation method and its usage
EA028175B1 (en) Pyrazolo-pyrrolidin-4-one derivatives as bet inhibitors and their use in the treatment of disease
JP7247092B2 (en) Substituted Fused Heteroaryl Compounds and Uses Thereof as Kinase Inhibitors
WO2013064083A1 (en) 1-(arylmethyl)-5,6,7,8-tetrahydroquinazoline-2,4-diones and analogs and the use thereof
TW201319067A (en) Triazolopyridine compounds
WO2019011228A1 (en) Imidazo[1,2-b]pyrimido[4,5-d]pyridazin-5(6h)-one compound and use thereof
EP3699179A1 (en) Pyrazolyl-containing tricyclic derivative, preparation method therefor and use thereof
KR20200078610A (en) Amino-substituted nitrogen-containing condensed-ring compounds and preparation methods and uses thereof
KR20130097178A (en) Quinoxaline compound
CN111153891B (en) Substituted benzimidazole PI3K alpha/mTOR double-target inhibitor and pharmaceutical composition and application thereof
WO2021078227A1 (en) Fused heteroaryl derivative, preparation method therefor, and application thereof in medicine
CN116162099A (en) Heterocyclic derivative and preparation method and application thereof
TWI548637B (en) Phthalazinone derivatives, preparation process and pharmaceutical use thereof
WO2014106763A1 (en) Pyridopyrazines as anticancer agents
WO2022237844A1 (en) Pyrrolopyrimidine derivative containing pyrazine structure
WO2021143821A1 (en) Fused heteroaryl derivative, preparation method therefor, and application thereof in medicine
CN117886813A (en) SHP2 phosphatase allosteric inhibitors
CN116157400A (en) Heterocyclic derivative and preparation method and application thereof
CN102382128A (en) Three-ring fused PI3K and mTOR dual inhibitor

Legal Events

Date Code Title Description
EEER Examination request

Effective date: 20160601