Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
GUANIDINE COMPOUNDS AND USE THEREOF
Document Type and Number:
WIPO Patent Application WO/2016/175357
Kind Code:
A1
Abstract:
The present invention relates to guanidine compounds for inhibiting mitochondrial oxidative phosphorylation (OXPHOS) and use thereof. More specifically, the present invention relates to a pharmaceutical composition for preventing or treating a OXPHOS-related disease, particularly cancer by inhibiting mitochondrial oxidative phosphorylation and reprogramming cellular metabolism.

Inventors:
KIM SUNG WUK (KR)
KIM HONG WOO (KR)
YOO SANG HEE (KR)
LEE JI SUN (KR)
HEO HYE JIN (KR)
LEE HONG BUM (KR)
KOOK JI AE (KR)
LEE YOUNG WOO (KR)
Application Number:
PCT/KR2015/004423
Publication Date:
November 03, 2016
Filing Date:
April 30, 2015
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
IMMUNOMET THERAPEUTICS INC (US)
International Classes:
C07C279/18; A61K31/155; A61P3/04; A61P3/10; A61P35/00; C07C279/26
Domestic Patent References:
WO2002074740A12002-09-26
WO1995020950A11995-08-10
Foreign References:
US20140348749A12014-11-27
US20120283299A12012-11-08
Other References:
See also references of EP 3288918A4
Attorney, Agent or Firm:
SON, Min (6th Floor,163, YangJae Cheon-ro, Gangnam-gu, Seoul, KR)
Download PDF:
Claims:
[CLAIMS ]

[Claim 1 ]

A compound selected from the group consisting of the compound having Chemical Formula 1 , the compound having Chemical Formula 6, pharmaceutically acceptable salts, pharmaceutically acceptable solvates and prodrug derivatives:

Chemical formula 1

NR3 NH

Rt. N.A..AN CH2)n

H 5

R2 R4

Where

R1 and R2 are independently H or C C6 alkyl, or C1-C haloalkyl; or are taken together with N to which they are attached for forming 3 to 8-membered saturated or unsaturated heterocycloalkyl group, where the heterocycloalkyl ring may include optionally at least a heteroatom selected from the group consisting of N, O and S, and is substituted with at least a group selected from the group consisting of H, halogen, C1-C4 alkyl, Q-C4 haloalkyl, C1-C4 alkoxy, and C1-C2 alkylamino,

R3 and R4 are independently H or C 1-C4 alkyl,

n is 0, 1 or 2, and

R5 is H, C3-C7 cycloalkyl, or aryl group represented by Chemical formula 2,

[Chemical Formula 2]

R6, R7, R8, R9 and R10 are independently H, halogen, C1-C4 alkyl, C1-C4 alkoxy, C1-C4 halolalkyl, C 1-C4 halolalkoxy, SR1 1 or OR12 where R1 1 and R12 are independently C1 -C3 alkyl, C1 -C3 haloalkyl, or C6-Cio aryl, or are linked with an adjacent substituent to form of 5-6 membered saturated ring.

[Claim 2]

The compound of claim 1 , wherein R3 and R4 are hydrogen in Chemical formula 1.

[Claim 3 ] The compound of claim 1 , wherein the compound having Chemical Formula 1 is selected from the group consisting of the guanidine compounds having Chemical Formula 4, pharmaceutically acceptable salts, pharmaceutically acceptable solvates and prodrug derivatives:

Chemical formula 4

Where

R1 and R2 are independently H or C C6 alkyl, or Ci-C6 haloalkyl; or are taken together with N to which they are attached for forming 3 to 8-membered saturated or unsaturated heterocycloalkyl group, where the heterocycloalkyl ring may include optionally at least a heteroatom selected from the group consisting of N, O and S, and is substituted with at least a group selected from the group consisting of H, halogen, C1-C4 alkyl, C1-C4 haloalkyl, CrC4 alkoxy, and Ci-C2 alkylamino,

n is 0, 1 or 2, and

R5 is H, C3-C7 cycloalkyl, or aryl group represented by Chemical formula 2,

[Chemical Formula 2]

R6, R7, R8, R9 and R10 are independently H, halogen, C1-C4 alkyl, C1 -C4 alkoxy, C C4 halolalkyl, CrC4 halolalkoxy, SR11 or OR12 where R11 and R12 are independently C1-C3 alkyl, C1-C3 haloalkyl, or C6-C10 aryl, or are linked with an adjacent substituent to form 5 to 6- membered saturated ring.

[Claim 4]

The compound of claim 1 , wherein, in Chemical formula 1 , R1 and R2 are taken together with N to which they are attached for forming 5 to 6-membered saturated or unsaturated heterocycloalkyl group and is substituted with halogen,

R3 and R4 are independently H,

n is 0, 1 or 2, and R5 is aryl group represented by Chemical formula 2 where R6, R7, R8, R9, and R10 are independently H, halogen, Ci-C4 alkyl, d-C4 alkoxy, C 1-C4 halolalkyl, Ci-C4 halolalkoxy, SRU or OR12 where R1 1 and R12 are independently C 1 -C3 alkyl, C 1-C3 haloalkyl, or C6-Cio aryl or are linked with an adjacent substituent to form of 5 to 6 membered saturated ring.

I Claim 5]

The compound of claim 1, wherein, in Chemical formula 1, R1 and R2 are taken together with N to which they are attached for forming 5 to 6-membered saturated or unsaturated heterocycloalkyl group and is substituted with halogen,

R3 and R4 are independently H,

n is 0, and

R5 is aryl group represented by Chemical formula 2 where R6, R7, R8, R9 and R10 are independently C 1-C4 halolalkoxy.

[Claim 6]

A compound selected from the group consisting of the following compounds, pharmaceutically acceptable salts, pharmaceutically acceptable solvates and prodrug derivatives: N-(N-phenylcarbamimidoyl)-4,4-difluoropiperidine-l-carboximidamide

N-(N-(5,6,7,8-teterhydronaphthalene-2-yl)carbamimidoyl)-4,4-difluoropiperidine-l- carboximidamide

N-(N-(3,4-dichlorophenyl)carbamimidoyl)-4,4-difluoropiperidine-l-carboximidamide

N-(N-(2-bromophenyl)carbamimidoyl)-4,4-difluoropiperidine-l-carboximidamide

N-(N-(3-bromophenyl)carbamimidoyl)-4,4-difluoropiperidine-l-carboximidamide

N-( -(4-bromophenyl)carbamimidoyl)-4,4-difluoropiperidine-l-carboximidamide

N-(N-(4-(trifluoromethoxy)phenyl)carbamimidoyl)-4-(dimethylamino)piperidine-l- carboximidamide

N-( -(4-(methythio)phenyl)carbamimidoyl)-4,4-difluoropiperidine-l-carboximidamide

N-(N-(4-(trifluoromethythio)phenyl)carbamimidoyl)-4,4-difluoropiperidine-l-carboximidamide

N-(N-(4-isopropylphenyl)carbamimidoyl)-4,4-difluoropiperidine-l-carboximidamide

N-( -(3-chloro-4-fluorophenyl)carbamimidoyl)-4,4-difluoropiperidine-l-carboximidamide

N-(N-(5,6,7,8-teterhydronaphthalen- 1 -yl)carbamimidoyl)-4,4-difluoropiperidine- 1 - carboximidamide

N-(N-(3-phenoxyphenyl)carbamimidoyl)-4,4-difluoropiperidine-l-carboximidamide N-(N-(4-(trifluoromethythio)phenyl)carbamimidoyl)thiazolidine-3-carboximidami

N-(N-(3,4,5-trimethoxyphenyl)carbamimidoy^

N-( -(4-(trifluoromethoxy)phenyl)carbamimidoyl)-4-chloropiperidine-l-carboximidamide

N-(N-(4-(trifluoromethoxy)phenyl)carbamimidoyl)-4,4-difluoropiperidine-l-carboxim^

N-(N-(3-chloro-4-iodophenyl)carbamimidoyl)-4,4-di^^

N-(N-(4-methoxyphenyl)carbamimidoyl)-4,4-difluoropiperidine-l -carboximidamide N-(N-(3-methoxyphenyl)carbamimidoyl)-4,4-difluoropiperidine-l -carboximidamide N-(N-(4-(trifluoromethyl)phenyl)carbamimidoyl)-4,4-difluoropiperidine- 1 -carboximidamide N-(N-(3 -bromo-4-iodophenyl)carbamimidoyl)-4,4-difluoropiperidine- 1 -carboximidamide N-(N-(4-phenoxyphenyl)carbamimidoyl)-4,4-difluoropiperidine-l -carboximidamide N-(N-(4-(trifluoromethoxy)carbamimidoyl)-4-ethoxypiperidine-l -carboximidamide

N-(N-(4-(trifluoromethoxy)phenyl)carbamimidoyl)-3,3-difluoropyrrolidine-l-carboxim N-(N-(3 ,4-dibromophenyl)carbamimidoyl)-4,4-difluoropiperidine- 1 -carboximidamide N-(N-(3-fluoro-4-(trifluoromethoxy)phenyl)carbamimidoyl)-4,4-fluoropiperidine-l- carboximidamide

N-(N-(4-phenoxyphenyl)carbamimidoyl)-3 ,3-difluoropyrrolidine- 1 -carboximidamide N-(N-(3-phenoxyphenyl)carbamimidoyl)-3,3-difluoropyrrolidine-l -carboximidamide N-(N-(4-(trifluoromethyl)phenyl)carbamimidoyl)-3,3-difluoropynOlidine-l -carboximidamide N-(N-(3-chloro-4-iodophenyl)carbamimidoyl)-3,3-difluoropyrrolidine-l -carboximidamide N-(N-(4-(trifluoromethythio)ph^

N-(N-(2-chlorophenyl)carbamimidoyl)-3,3-difluoropyrrolidine-l -carboximidamide

N-(N-(2-bromophenyl)carbamimidoyI)-3,3-difluoropyrrolidine-l -carboximidamide

N-(N-(2,4-dichlorophenyl)carbamimidoyl)-3,3-difluoropyrrolidine-l -carboximidamide N-(N-(2-propylphenyl)carbamimidoyl)-3,3-difluoropyrrolidine-l -carboximidamide

N-(N-3,4-dimethoxyphenyl)carbamimidoyl)-3,3-difluoropyrrolidine-l -carboximidamide N-( -(3-chloro-4-(trifluoromethoxy)phenyl)carbamimidoyl)-3,3-difluoropyrrolidine-l- carboximidamide

N-(N-(3 -bromo-4-(trifluoromethoxy)phenyl)carbamimidoyl)-3 ,3-difluoropyrrolidine- 1 - carboximidamide

N-(N-(2-bromobenzyl)carbamimidoyl)-4,4-difluoropiperidine-l -carboximidamide

N-(N-(4-(trifluoromethoxy)benzyl)carbam

N-(N-(3,4-dichlorobenzyl)carbamimidoyl)-4,4-difluoropiperidine-l -carboximidamide N-(^-(4-trifluoromethyl)benzyl)carbamimidoyl)-3,3-difluoropyiTolidine-l-carboximidami N-(N-(3 -trifluoromethyl)benzyl)carbamimidoyl)-3, 3 -difluoropyrrolidine-1 -carboximidamide N-(N-(3,4-dichloro)benzyl)carbamimid^

N-(N-phenethylcarbamimidoyl)-3 ,3 -difluoropyrrolidine- 1 -carboximidamide

N-(N-(4-bromophenethyl)carbamimidoyl)-3 ,3 -difluoropyrrolidine- 1 -carboximidamide

N-(N-(cyclopropylmethyl)carbamimidoyl)-4,4-difluoropiperidine-l-carboximidamide

N-(N-cyclopropylcarbamimidoyl)-4,4-difluoropiperidine-l -carboximidamide

N-(N-cyclohexylcarbamimidoyl)-4,4-difluoropiperidine-l -carboximidamide

N-( -(cyclopropylmethyl)carbamimidoyl)-3, 3 -difluoropyrrolidine- 1 -carboximidamide

N-(N-(cyclopropylcarbamimidoyl)-3,3-difluoropyrrolidine-l -carboximidamide

N-(N-(cyclohexylcarbamimidoyl)-3,3-difluoropyrrolidine-l-carboximidamide

N-(N-(3-bromobenzyl)carbamimidoyl)-4,4-difluoropiperidine-l -carboximidamide

N-(N-(4-bromobenzyl)carbamimidoyl)-4,4-difluoropiperidine-l -carboximidamide

N-(N-(4-(trifluoromethoxy)phenyl)carb

N-(N-(4-(trifluoromethoxy)phenyl)carbamimidoyl)thiazolidine-3-carboximidamide

N-(N-(4-(trifluoromethoxy)phenyl)carbamimidoyl)-N-2,2-2-trifluoroethylamino

carboximidamide

N-(N-(4-(trifluoromethoxy)phenyl)carbamimidoyl)-4-(trifluoromethyl)piperidine-l- carboximidamide

N-(N-(4-(trifluoromethoxy)phenyl)carbamimidoyl)-N-l-cycloproylmethylamino

carboximidamide

N-(N-(4-(trifluoromethoxy)phenyl)carbamimidoyl)-3 ,4,4-trifluoropiperidine- 1 -carboximidamide N-(N-(4-(trifluoromethoxy)phenyl)carbamimidoyl)-4,4-difluoro-3 -methylpiperidine- 1 - carboximidamide

N-(N-(4-(trifluoromethoxy)phenyl)carbamimidoyl)-4-fluoropiperidine-l-carboximidamide N-( -(4-(trifluoromethoxy)phenyl)carbamimidoyl)-(R)-3-fluoropyiTolidine-l-carboximidami N-(N-(4-(trifluoromethoxy)phenyl)carbamimidoyl)-6-azaspiro[2.5]octane-6-carboximidamide N-(N-(4-(trifluoromethoxy)phenyl)carbamimidoyl)-3-azabicyclo[3.1.0]hexane-3- carboximidamide

N-carbamimidoyl-3,3-difIuoropyrrolidine-l-carboximidamide

N-carbamimidoyl-4,4-difluoropiperidine- 1 -carboximidamide, and

N-((4,4-difluoropiperidin- 1 -yl)(imino)methyl)-4,4-difluoropiperidine- 1 -carboximidamide

[Claim 7]

A pharmaceutical composition for preventing or treating a disease associated with mitochondrial oxidative phosphorylation, comprising the compound or a pharmaceutically acceptable salt thereof defined in any one of claims 1 to 6 as an active ingredient.

[Claim 8]

The pharmaceutical composition according to claim 7, wherein the disease is at least one selected from the group consisting of diabetes mellitus, obesity, hyperlipemia,

hypercholesterolemia, fatty liver, coronary artery disease, osteoporosis, polycystic ovary syndrome, metabolic syndrome, cancer, muscle pain, myocyte damage and rhabdomyolysis.

[Claim 9]

The pharmaceutical composition according to claim 8, wherein the diabetes mellitus is insulin-independent diabetes mellitus.

[Claim 10]

The pharmaceutical composition according to claim 8, wherein the cancer is selected from the group consisting of uterine cancer, breast cancer, gastric cancer, brain cancer, colorectal cancer, lung cancer, skin cancer, blood cancer and liver cancer.

[Claim 1 1 ]

The pharmaceutical composition according to claim 8, wherein the treatment of the cancer includes inhibition of recurrence or metastasis of cancer.

[Claim 12]

The pharmaceutical composition according to claim 7, wherein the composition further comprises cancer is at least one active ingredient selected from the group consisting of chemotherapeutics, anti-cancer drugs, anti-cancer antibody drug, radiation, immunotherapy agents, and kinase inhibitors.

[Claim 13]

The pharmaceutical composition of Claim 7, wherein the pharmaceutical composition is formulated as a tablet, a capsule, a pill, a granule, powder, an injection or a liquid.

[Claim 14]

A method of preventing or treating a disease associated with mitochondrial oxidative phosphorylation, comprising administering to a subject in need the compound or a

pharmaceutically acceptable salt thereof defined in any one of claims 1 to 6 as an active ingredient.

[Claim 15]

A use of the compound or a pharmaceutically acceptable salt thereof defined in any one of claims 1 to 6, in in prepararion of a medicine for preventing treating a disease associated with mitochondrial oxidative phosphorylation.

Description:
.DESCRIPTION]

[Title of Invention]

GUANIDINE COMPOUNDS AND USE THEREOF [Technical Field]

The present invention relates to guanidine compounds for inhibiting mitochondrial oxidative phosphorylation (OXPHOS) and use thereof. More specifically, the present invention relates to a pharmaceutical composition for preventing or treating a disease associated with OXPHOS, particularly cancer by inhibiting mitochondrial oxidative phosphorylation and reprogramming cellular metabolism.

[Background Art]

The cellular metabolism is essential to generate resources such as ATP and biomass for their growth. The metabolic pathway to generate ATP is glycolysis and OXPHOS in mitochondria. Normal cells generate ATP via OXPHOS in mitochondria since 38 ATP molecules are generated per glucose molecule. However fast growing cells use glycolysis to generate ATP and lactate is the final metabolite in the process. For a long time, the dependency on OXPHOS is thought to be determined by availability of oxygen because oxygen is the molecule that accepts electrons during OXPHOS. Recently, the studies have shown that oxygen is not the determinant for OXPHOS, but rather cellular demands for biomass and

NADH/NADPH in fast growing cells actively choose to use glycolysis rather than OXPHOS. Cancer cells are the best example of transformed metabolism and uncontrolled proliferation. Dr. Otto Warburg in 1920s noticed the cancer cells mainly use glycolysis and produce high level of lactate. The highly glycolytic nature of cancer metabolism is currently named "Warburg effect". The glycolytic metabolic feature leads to a speculation that cancer cells might have dysfunctional mitochondria. Recent studies however showed the significance of OXPHOS in cancer cells, in particular, cancer stem cell-like population, migrating cancer cells, circulating cancer cells in metastasis.

Metformin is a biguanide used for the treatment of diabetes. It is known to be an OXPHOS inhibitor that has been clinically used for a long time. Several retrospective epidemiology studies pointed out that cancer incidence was lower in diabetic patients who were treated with metformin. The anticancer effect of metformin has been demonstrated in in vitro and in vivo models of breast, colon, prostate and lung cancer. The efficacy of metformin is limited by its weak potency and distribution due to the cationic property, therefore the dependency on Organic Cation Transporter 1 (OCT1) in order to enter cells. Many studies used a more potent biguanide and antidiabetic drug, phenformin to demonstrate the anticancer effect of OXPHOS inhibitor. Phenformin is more lipophilic than metformin and shows less dependency on OCT1 to enter cells. Several studies showed phenformin has activity of tumor growth inhibition and moreover prevent rising of cells resistant to targeted therapies (YuanP, Proc Natl Acad Sci. 2013, 1 10(45): ppl 8226-18231). Phenformin was shown to inhibit the growth of slow growing cancer cells or JARIDl B hlgh cells that might be responsible for drug resistance and relapse of disease (RoeschA, Cancer Cell, 2013, 23(6), pp81 1-825)

In the last decade, the main anticancer therapy was focused on development of inhibitors of oncogenes or signaling proteins such as kinases and growth factor receptors. The response rates were marginal in most cases. The initial responses by the best therapies apparently looked promising, but majority of patients relapsed with much more aggressive and drug resistant form of cancers. The true mechanism of relapse is still needed to be discovered, but multiple relapse mechanism have been reported such as secondary mutations on the same target or activation of different route of signaling pathway The mechanism of phenformin in overcoming drug resistance is not still clear. The OXPHOS inhibition may prevent further reprogramming after reprogramming upon co-treatment with targeted therapy, therefore it may cause energy crisis or prevent growth of slow growing population depending on OXPHOS.

Metformin has limited efficacy and tissue distribution and phenformin has been withdrawn from the market due to fatal safety issues. Thus, the conventional biguanide used for diabetic treatment have limitations as an anticancer agent.

[DISCLOSUE]

[Technical Problem]

The present invention relates to guanidine compounds or pharmaceutically-acceptable salts thereof with an improved activity of inhibiting mitochondrial oxidative phosphorylation (OXPHOS) and reprogramming cellular metabolism.

Another embodiment of the present invention is to provide a pharmaceutical composition for preventing or treating a disease-associated with mitochondrial oxidative phosphorylation (OXPHOS) or a method of preventing or treating a disease-associated with mitochondrial oxidative phosphorylation (OXPHOS) including administering the compound of the present invention to a subject in need. Further embodiment of the present invention is to provide an anti-cancer pharmaceutical composition comprising the guanidine compounds or pharmaceutically-acceptable salts thereof as active ingredient.

[Technical Solution]

To achieve the technical object, an embodiment of the present invention relates to the guanide compounds, pharmaceutically acceptable salts, pharmaceutically acceptable solvates and prodrug derivatives which have superior inhibitory effect on cancer cell growth, cancer metastasis and cancer reoccurrence to conventional drugs, even though smaller amount of the compounds are used.

In addition, an embodiment of the present invention relates to a use of the guanidine compounds for inhibiting mitochondrial oxidative phosphorylation (OXPHOS) or

reprogramming cellular metabolism.

[Detailed Description of the Invention]

Hereinafter, the present invention can be explained in more detail.

In another aspect, the present invention provides the compound selected from the group consisting of the guanidine compounds having Formula 1 , pharmaceutically acceptable salts, pharmaceutically acceptable solvates and prodrug derivatives:

[Chemical formula 1 ]

In the Chemical Formula 1 ,

R 1 and R 2 are independently H, Ci-C alkyl, or Ci-C 6 haloalkyl; or are taken together with N to which they are attached for forming 3 to 8-membered saturated or unsaturated heterocycloalkyl group, preferably 5-6 membered saturated or unsaturated heterocycloalkyl group, where the heterocycloalkyl ring may include optionally at least a heteroatom selected from the group consisting of N, O and S, and is substituted with at least a group selected from the group consisting of H, halogen, C 1 -C4 alkyl, C 1 -C4 haloalkyl, C 1 -C4 alkoxy, and C C2 alkylamino,

R 3 and R 4 are independently H or C 1 -C4 alkyl,

n is 0, 1 or 2, and

R 5 is H, C3-C7 cycloalkyl, or aryl group represented by Chemical formula 2,

[Chemical Formula 2]

R 6 , R 7 , R 8 , R 9 and R 10 are independently H, halogen, C 1 -C4 alkyl, C 1 -C4 alkoxy, C 1 -C4 halolalkyl, C C 4 halolalkoxy, SR 1 1 or OR 12 where R 1 1 and R 12 are independently C,-C 3 alkyl, C 1 -C3 haloalkyl, or C6-C 10 aryl, or

R°, R , R , R and R ,u are linked with an adjacent substituent to form of 5-6 membered saturated ring.

In Chemical formula 1 , when R 3 and R 4 are hydrogen, the compound is represented by Chemical formula 3:

[Chemical formula 3]

R 1 , R 2 , R 5 and n are the same as defined in the Chemical formula 1.

In Chemical formula 1, when R 3 and R 4 are hydrogen and R 5 is aryl group represented by Chemical formula 3, the compound is represented by Chemical formula 4:

[Chemical formula 4]

R 1 , R 2 , R 6 , R 7 , R 8 , R 9 , R 10 and n are the same as defined in the Chemical formula 1. In Chemical formulae 1 , 3 and 4, the heterocycloalkyl in definition of R 1 and R 2 can be a 3 to 8-membered saturated heterocycloalkyl which may substituted with at least a group selected from the group consisting of H, halogen, C1-C4 alkyl, C 1-C4 haloalkyl, C1-C4 alkoxy, and C]-C 2 alkylamino, R 3 and R 4 are hydrogen, and R 5 is aryl group represented by Chemical formula 2 where R 6 , R 7 , R 8 , R 9 , and R 10 are the same as defined in the Chemical formula 1.

In an embodiment of the compounds represented by Chemical Formula 1 , the

substituents of R 1 and R 2 are taken together with N to which they are attached for forming 5 to 6- membered saturated or unsaturated heterocycloalkyl group and is substituted with halogen,

R 3 and R 4 are independently H,

n is 0, 1 or 2, and

R 5 is aryl group represented by Chemical formula 2 where R 6 , R 7 , R 8 , R 9 , and R 10 are the same as defined in the Chemical formula 1 as described above.

In another embodiment of the compounds represented by Chemical Formula 1, R 1 and R 2 are taken together with N to which they are attached for forming 5 to 6 membered saturated or unsaturated heterocycloalkyl group and is substituted with halogen,

R 3 and R 4 are independently H,

n is 0, and

R 5 is aryl group represented by Chemical formula 2 where R 6 , R 7 , R 8 , R 9 and R 10 are Cj- C 4 halolalkoxy.

As used herein, the term "alkyl" refers to a saturated hydrocarbon group which is straight-chained or branched. Example groups include methyl, ethyl, propyl (n-propyl, isopropyl), tert-butyl, cyclopropylmethyl, cyclobutylmethyl, neopentyl and the like. The alkyl can be substituted with at least a halogen, such as F, CI, Br or I and the example groups include CF3, CHF 2 , CH 2 F, CH 2 C1, and the like.

As used herein, the terms "halo" or "halogen" includes fluoro, chloro, bromo, and iodo.

As used herein, the term "alkoxy" is meant to refer to a functional group containing an "alkyl" group bonded to an oxygen atom. An "alkyl" is defined above. As used herein, the term "haloalkoxy" is meant to refer to a functional group containing "haloalkyl" group bonded to an oxygen atom. An "alkyl" is defined above.

As used herein, the term "cycloalkyl" refers to non-aromatic carbocycles including cyclized alkyl, alkenyl, and alkynyl groups. Cyclolalkyl groups can include mono- or polycyclic ring systems, including spirocycles, or bridged cycles. Example cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl. C 3 -C 7 cycloalkyl refers to a cycloalkyl radical containing from 3 to 7 ring carbon atoms. Examples of cycloalkyl groups include such groups as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexl, cycloheptyl, cyclooctyl, pinenyl, and adamantanyl.

The term, "benzocycloalkyl" refers to moieties that have one or more aromatic rings fused to the cycloalkyl ring, and for examples, include benzo derivatives of cyclopentane and cyclohexane. The examples include indane, hydronaphthalene and tetrahydrohaphthalene. In

Chemical formula 2, R°, R', R°, and R 1U are linked with an adjacent substituent to form of 5-6 membered saturated ring, so that the substituted of chemical formula 2 can be 9 to 10-membered benzocycloalkyl group.

As used herein, the term "heterocycloalkyl" refers to a non-aromatic heterocycle where one or more of the ring-forming atoms are heteroatom such as O, N, or S. The heterocyclolalkyl groups can include monocyclic, bicyclic or polycyclic ring systems, including spirocycles, or bridged cycles. Examples of heterocycloalkyl groups include pyrrolidinyl, pyrrolinyl, imidazolidinyl, imidazolinyl, pirazolidinyl, pirazolinyl, pyrazalinyl, piperidyl, piperazinyl, morpholinyl, thiazolidinyl, thiomorpholinyl, tetrahydrofuranyl, dithiolyl, oxathiolyl, dioxazolyl, oxathiazolyl, pyranyl, oxazinyl, oxathiazinyl, oxadiazinyl, azospiroheptane, azospirooctane, azabicyclohexane, and azabicyclohepane.

As used herein, the term "aryl" refers to a substituted or unsubstituted, mono- or bicyclic hydrocarbon aromatic ring system having 6 to 10 ring carbon atoms. Examples include unsubstituted or substituted phenyl and naphthyl groups.

In further aspect, the the present invention provides additional various compounds as well as the compounds having Chemical Formula 1 , pharmaceutically acceptable salts, pharmaceutically acceptable solvates and prodrug derivatives. The examples of the present invention can include the following compounds:

N-(N-phenylcarbamimidoyl)-4,4-difluoropiperidine-l -carboximidamide

N-CN-(5,6,7,8-teterhydronaphthalene-2-yl)carbamimidoyI)-4 ,4-difluoropiperidine-l- carboximidamide

N-( -(3,4-dichlorophenyl)carbamimidoyl)-4,4-difluoropiperidine-l -carboximidamide

N-( -(2-bromophenyl)carbamimidoyl)-4,4-difluoropiperidine-l-carb oximidamide

N-(N-(3 -bromophenyl)carbamimidoyl)-4,4-difluoropiperidine- 1 -carboximidamide

N-(N-(4-bromophenyl)carbamimidoyl)-4,4-difluoropiperidine -l -carboximidamide

N-(N-(4-(trifluoromethoxy)phenyl)carbamimidoyl)-4-(dimeth ylamino)piperidine-l- carboximidamide

N-(N-(4-(methythio)phenyl)carbamimidoyl)-4',4-difluoropiperi dine- l -carboximidamide

N-(N-(4-(trifluoromethythio)phenyl)carbamimidoyl)-4,4-dif luoropiperidine-l -carboximidamide N-( -(4-isopropylphenyl)carbamimidoyl)^

N-(N-(3-chloro-4-fluorophenyl)carbamimidoyl)-4,4-difluoropip eridine-l -carboximidamide

N-(N-(5,6,7,8-teterhydronaphthalen-l-yl)carbamimidoyl)-4, 4-difluoropiperidine-l- carboximidamide

N-(N-(3-phenoxyphenyl)carbamimidoyl)-4,4-difluoropiperidine- l -carboximidamide

N-(N-(4-(trifluoromethythio)phenyl)carbamimidoyl)thiazoli dine-3-carboximidami

N-(N-(3,4,5-trimethoxyphenyl)carbamimidoyl)-3,3-difluorop yrrolidine-l -carboximidamide N-(TSi-(4-(trifluoromethoxy)phenyl)carbamimidoyl)-4-chloropi peridine-l -carboximidamide N-(N-(4-(trifluoromethoxy)phenyl)carba

N-(N-(3-chloro-4-iodophenyl)carbamimidoyl)-4,4-difluoropiper idine-l -carboximidamide N-(N-(4-methoxyphenyl)carbamimidoyl)-4,4-difluoropiperidine- l -carboximidamide

N-(N-(3 -methoxyphenyl)carbamimidoyl)-4,4-difluoropiperidine- 1 -carboximidamide

N-(N-(4-(trifluoromethyl)phenyl)carbamim^

N-(N-(3-bromo-4-iodophenyl)carbamimidoyl)-4,4-difluoropiperi dine-l-carboximidamide N-(N-(4-phenoxyphenyl)carbamimidoyl)-4,4-difluoropiperidine- l -carboximidamide

N-(N-(4-(trifluoromethoxy)carbamimidoyl)-4-ethoxypiperidi ne-l-carboximidamide

N-(N-(4-(trifluoromethoxy)phenyl)carbamimidoyl)-3,3-diflu oropyrrolidine-l -carboximidamide N-(N-(3 ,4-dibromophenyl)carbamimidoyl)-4,4-difluoropiperidine- 1 -carboximidamide

N-( -(3-fluoro-4-(trifluoromethoxy)phenyl)carbamimidoyl)-4,4-flu oropiperidine-l- carboximidamide

N-(N-(4-phenoxyphenyl)carbamimidoyl)-3,3-difluoropyrrolidine -l -carboximidamide

N-(N-(3-phenoxyphenyl)carbamimidoyl)-3,3-difluoropyrrolid ine-l -carboximidamide

N-(^-(4-(trifluoromethyl)phenyl)carbamimidoyl)-3,3-difluo ropyrrolidine-l-carboximidami N-(N-(3-chloro-4-iodophenyl)carbamimidoyl)-3,3-difluoropyrro lidine-l -carboximidamide N-(N-(4-(trifluoromethythio)phenyl)carbamimidoyl)-3 ,3 -difluoropyrrolidine- 1 -carboximidamide N-(N-(2-chlorophenyl)carbamimidoyl)-3 ,3 -difluoropyrrolidine- 1 -carboximidamide

N-(N-(2-bromophenyl)carbamimidoyl)-3,3-difluoropyrrolidin e-l -carboximidamide

N-(N-(2,4-dichlorophenyl)carbamimidoyl)-3,3-difluoropyrro lidine-l-carboximidamide

N-(N-(2-propylphenyl)carbamimidoyl)-3,3-difluoropyrrolidi ne-l -carboximidamide

N-(N-3,4-dimethoxyphenyl)carbamimidoyl)-3 ,3 -difluoropyrrolidine- 1 -carboximidamide N-(N-(3-chloro-4-(trifluoromethoxy)phenyl)carbamimidoyl)-3,3 -difluoropyrrolidine-l- carboximidamide

N-(N-(3-bromo-4-(trifluoromethoxy)phenyl)carbamimidoyl)-3,3- difluoropyrrolidine-l- carboximidamide N- N-(2-bromobenzyl)carbamimidoyl)-4,4-difluoropiperidine-l -carboximidamide N-(N-(4-(trifluoromethoxy)benzyl)carbamimidoyl)-4,4-difluoro piperidine-l -carboximidamide N-(N-(3 ,4-dichlorobenzyl)carbamimidoyl)-4,4-difluoropiperidine- 1 -carboximidamide

N-(N-(4-trifluoromethyl)benzyl)carbamimidoyl)-3,3-difluor opyrrolidine-l -carboximidamide N-(N-(3-trifluoromethyl)benzyl)carbamimidoyl)-3 ,3-difluoropyrrolidine- 1 -carboximidamide N-(N-(3,4-dichloro)benzyl)carbamimidoyl)-3,3-difluoropyrroli dine-l -carboximidamide N-(N-phenethylcarbamimidoyl)-3 ,3 -difluoropyrrolidine- 1 -carboximidamide

N-(N-(4-bromophenethyl)carbamimidoyl)-3,3-difluoropyrroli dine-l -carboximidamide

N-(N-(cyclopropylmethyl)carbamimidoyl)-4,4-difluoropiperi dine-l -carboximidamide

N-(N-cyclopropyIcarbamimidoyl)-4,4-difluoropiperidine-l -carboximidamide

N-( -cyclohexylcarbamimidoyl)-4,4-difluoropiperidine-l -carboximidamide

N-(N-(cyclopropylmethyl)carbamimidoyl)-3,3-difluoropyrrol idine-l -carboximidamide

N-(N-(cyclopropylcarbamimidoyl)-3 ,3-difluoropyrrolidine- 1 -carboximidamide

N-(N-(cyclohexylcarbamimidoyl)-3, 3 -difluoropyrrolidine- 1 -carboximidamide

N-(N-(3 -bromobenzyl)carbamimidoyl)-4,4-difluoropiperidine- 1 -carboximidamide

N-(TSi-(4-bromobenzyl)carbamimidoyl)-4 J 4-difluoropiperidine-l-carboximidamide

N-(N-(4-(trifluoromethoxy)phenyl)carbamimidoyl)-3,3-diflu oropiperidine-l-carboximi N-( -(4-(trifluoromethoxy)phenyl)carbamimidoyl)thiazolidine-3-ca rboximidamide

N-(N-(4-(trifluoromethoxy)phenyl)carbamimidoyl)-N-2,2-2-t rifluoroethylamino

carboximidamide

N-(N-(4-(trifluoromethoxy)phenyl)carbamimidoyl)-4-(txifluoro methyl)piperidine-l- carboximidamide

N-(N-(4-(trifluoromethoxy)phenyl)carbamimidoyl)-N-l-cyclopro ylmethylamino

carboximidamide

N-(N-(4-(trifluoromethoxy)phenyl)carbamimidoyl)-3 ,4,4-trifluoropiperidine- 1 -carboximidamide

N-( J-(4-(trifluoromethoxy)phenyl)carbamimidoyl)-4,4-difluoro-3- methylpiperidine-l- carboximidamide

N-(N-(4-(trifluoromethoxy)phenyl)carbamimidoyl)-4-fluoropipe ridine-l -carboximidamide N-( -(4-(trifluoromethoxy)phenyl)carbamimidoyl)-(R)-3-fluoropyiT olidine-l-carboximi^ N-(N-(4-(trifluoromethoxy)phenyl)carbamimidoyl)-6-azaspiro[2 .5]octane-6-carboximidamide N-(N-(4-(trifluoromethoxy)phenyl)carbamimidoyl)-3-azabicyclo [3.1.0]hexane-3 - carboximidamide

N-carbamimidoyl-3,3-difluoropyrrolidine-l -carboximidamide

N-carbamimidoyl-4,4-difluoropiperidine- 1 -carboximidamide, and N-((4,4-difluoropiperidin- 1 -yl)(imino)methyl)-4,4-difluoropiperidine- 1 -carboximidamide

The synthesis of certain compound represented by Chemical Formula 3 is illustrated in reaction scheme (1).

Reaction Scheme (1)

Cyclohexanol R" 4 Cyclohexanol R

For example, the compound of example 17 was prepared as illustrated in reaction scheme 2. The detailed synthetic procedure is as described below.

Reaction Scheme (2)

To the solution of 4,4-difluoropiperidine HCI (1.00 g, 6.34 mmol) in 30 mL cyclorohexanol was added sodium dicyanamide(0.62 g, 6.98 mmol). The reaction mixture was refluxed at 130 C for 2 h. The mixture was diluted with EtOAc and water. The separated organic layer was condensed under reduced pressure yielding yellow solid (0.89 g, 75.2 %) residue followed by triturated with n-Hexanes. To the solution of the residue (0.5 g, 2.65 mmol) in 20 mL cyclohrexanol were added cone. HCI solution (0.23 mL, 2.65 mmol) and 4- trifluoromethoxyaniline (0.35 mL, 2.65 mmol). The reaction mixture was refluxed at 130 C for 1 h. The mixture was cooled down to RT and stirred for 1 h further yielding the precipitate. The precipitate from the solution was filtered and re-dissolved in 10 mL MeOH. To the solution was added 0.5 mL 1.5 M NaOH resulting in precipitate. The solids were dissolved in 10 mL MeOH. To the solution was added acetic acid (0.23 mL, 3.98 mmol) and further stirred at RT for 1 h. The solvent was removed by a rotavapor yielding the desired white solids followed by being triturated with EtOAc (0.7 g, 56.3 %).

The guanidine compounds of present invention can function as mitochondrial oxidative phosphorylation (OXPHOS) inhibitor. As used herein, the term "OXPHOS inhibitor" refers to an agent that inhibits oxidative phosphorylation, for example, oxidative phosphorylation in the mitochondria, either by direct inhibition of proteins involved in oxidative phosphorylation, or by inhibition of expression of the proteins involved in oxidative phosphorylation. The conventional OXPHOS inhibitors are metformin, phenformin and buformin. Metformin is a mitochondrial complex 1 inhibitor that can be used to target mitochondrial OXPHOS.

Thus, the present invention relates to a use of the guanidine compounds for inhibiting mitochondrial OXPHOS or reprogramming cellular metabolism. More specifically, the present invention relates to a pharmaceutical composition for preventing or treating a disease associated with mitochondrial OXPHOS.

The disease is at least one selected from the group consisting of diabetes mellitus, obesity, hyperlipemia, hypercholesterolemia, fatty liver, coronary artery disease, osteoporosis, polycystic ovary syndrome, metabolic syndrome, cancer, muscle pain, myocyte damage and rhabdomyolysis.

The diabetes mellitus is insulin-independent diabetes mellitus.

The cancer can be uterine cancer, breast cancer, gastric cancer, brain cancer, colorectal cancer, lung cancer, skin cancer, blood cancer and liver cancer, but not limited thereto.

The guanidine compounds of present invention have superior inhibitory effect on cancer cell growth, cancer metastasis and cancer reoccurrence to conventional drugs, even though smaller amount of the compounds are used.

The compounds of the present invention are improved guanide compounds with improved potency and anticancer activity in low glucose condition. The role of OXPHOS inhibitor of the present invention is not limited in growth inhibition of cancer, but also lower cancer stem cell like population, recurrence, and enhance efficacy of other anticancer drugs in the combination.

An aspect of the invention is to provide a method of preventing or treating a disease associated with OXPHOS, particularly cancer by inhibiting mitochondrial oxidative

phosphorylation and reprogramming cellular metabolism, comprising administering the guanidine compound of present invention to a subject in need.

The compounds of invention can be used in combination with other pharmaceutical agents or treatment methods, for examples, chemotherapeutics, anti-cancer drugs, anti-cancer antibody drug, radiation, immunotherapy agents, and kinase inhibitors. The combination agent can be administered in a combined form or separate form.

Chemotherapeutic agents in combination with the compound of invention include (without limitation) alkylating agents, uracil mustards, chlormethine, cyclophosphamide

(CytoxanTM), ifosfamide, melphalan, chlorambucil, pipobroman, triethlene-melamine, rtriethylenethiophosphoramine, busulfan, carmustine, lomustine, streptozocin, dacarbazine, temozolomide, methotrexate, 5-fluorouracil, floxuridine, cytarabine, 6-mercaptopurine, 6- thioguanide, gemcitabine, doxorubicin, epirubicin, idarubicin, ara-C, paclitaxel (TaxolTM), navelbene, letrozole, anastrazole, capecitabine, cis-platin, carboplatin, and topoisomerase inhibitors. Anti-cancer antibodies include trastuzumab (Herceptin), and bevacizumab (Avastin). Immunotherapy agents include interferon, anti-PDl antibody, anti-CTLA4 antibody, IDOl inhibitors, and other immune cell therapies including adoptive T cell or NK cells. Kinase inhibitors include dasatinib, trametinib, palbociclib, or tyrosine kinase inhibitors such as erlotinib, gefatinib, but not limited thereto.

[Measurement of mitochondrial complex I inhibition]

The electron transfer complex in mitochondria is composed of 5 complexes. The complex I accept electrons from NADH produced from glycolysis and TCA cycle and the electrons move to complex II, III and IV and the electron is finally transferred to 0 2 and water molecule is generated. During the electron transfer, proton gradient is generated and the chemical gradient is a driving source to synthesize ATP at complex V. The mitochondrial inhibition of complex I indirectly assessed by measuring the oxygen consumption rate (OCR) at complex IV. When the mitochondrial ETC is inhibited, glycolysis is up-regulated and lactate production is increased. The solution outside of cells becomes acidic (lower pH) as lactate is transported to outside of cells. OCR and Extracellular acidification rate (ECAR) are determined by XF Analyzer (Seahorse Biosciences). The compounds of present invention caused lower OCR by inhibition of complex I and higher ECAR by redirecting cellular metabolism to glycolysis.

[Cytotoxicity assay in low glucose condition]

The inhibition of oxidative phosphorylation (OXPHOS) is not cytotoxic to cells in normal glucose condition, because it is postulated that normal cells have compensatory mechanism under energy stress conditions such as low glucose. However OXPHOS inhibitors show cytotoxic effect on cells in the glucose deprived condition (BirsoyK, 2014). The glucose deprived condition is observed in tumor microenvironment potentially due to poor angiogenesis. Therefore the OXPHOS inhibitors may show anti-cancer effect on cancer cells in low glucose condition that may depict tumor microenvironment.

The compounds of the present invention were evaluated their cytotoxicity in SK-MEL- 28, melanoma with 0.75 mM glucose supplement. The cytotoxic effect is compared with the cytotoxicity caused by phenformin. The cytotoxicity in low glucose condition is correlated with inhibition of oxygen consumption in mitochondria.

[In vivo xenograft study]

The compounds of the present invention were evaluated in vivo using xenograft human cancer model in mice. The compounds of invention were administrated orally or

interperitoneal injection. Tumor cell lines were cultured in vitro as monolayer culture. Using female BALB/c nude mice with immune system compromised, each mouse was inoculated subcutaneously at the right flank with tumor cells for tumor development. The treatment was started when the tumor size reaches approximately 100 mm

The pharmaceutically acceptable salt of the compound according to the present invention may be an acid addition salt formed using organic acid or inorganic acid. The organic acid may include formic acid, acetic acid, propionic acid, lactic acid, butyric acid, isobutyric acid, trifluoroacetic acid, malic acid, maleic acid, malonic acid, fumaric acid, succinic acid, succinic acid monoamide, glutamic acid, tartaric acid, oxalic acid, citric acid, glycolic acid, glucuronic acid, ascorbic acid, benzoic acid, phthalic acid, salicylic acid, anthranyl acid, dichloroacetic acid, aminooxy acetic acid, benzenesulfonic acid, 4-toluenesulfonic acid and methanesulfonic acid salts. The inorganic acid may include, for examples, hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, nitric acid, carbonic acid, and boric acid salts. The acid addition salt may be prepared by a common preparation method of salts, such as a) directly mixing the compound of the present invention and acid, b) dissolving the compound of the present invention or acid in a solvent or a an aqueous solvent and mixing them, or c) mixing the compound of the present invention and acid in a solvent or an aqueous solvent.

Thus, another embodiment of the present invention provides a pharmaceutical composition comprising the guanide compound or a pharmaceutical salt thereof as an active ingredient. The pharmaceutical composition according to the present invention has excellent cancer cell proliferation inhibition effect, and thus, it may be used as an anticancer agent for various cancers, and specific Examples of the cancer include breast, lung, melanoma, pancreas, brain, ovary, prostate, cervix, testes, renal, head and neck, liver, ymphoma, leukemia,

endometrial cancer, cholangiocarcinoma, but not limited thereto. The pharmaceutical composition of the present invention comprises at least one pharmaceutically acceptable carrier in addition to the active ingredient. As used herein,

'pharmaceutically acceptable carrier' means known pharmaceutical excipient that is useful for formulation of a pharmaceutically active compound for administration and is substantially non- toxic and non-sensitive under use conditions. The exact ratio of the excipient is determined by standard pharmaceutical practice, as well as by the solubility and the chemical properties of active compounds and the selected administration route.

The pharmaceutical composition of the present invention may be formulated into a form suitable for a desired administration method using adjuvant such as a physiologically acceptable excipient, a disintegrating agent, a sweetener, a binder, coating material, a blowing agent, a lubricant, a slip modifier, a flavoring agent and the like.

The pharmaceutical composition may be formulated in the form of tablets, capsules, pills, granule, powder, injections or liquid. The dosage form of the pharmaceutical composition and the pharmaceutically acceptable carrier may be appropriately selected according to technologies known in the art.

Meanwhile, as used herein, a 'subject' means a warm-blooded animal such as a mammal with a specific disease, disorder or condition, and for Example, it includes a human being, an orangutan, a chimpanzee, a mouse, a rat, a dog, a cow, a chicken, a pig, a goat, a sheep and the like, but is not limited thereto.

The term, 'treatment' or 'treating' includes relieving symptoms, temporarily or permanently removing the cause of symptoms, or preventing or slowing the appearance of symptoms and the progress of the disease, disorder or condition, but is not limited thereto.

The effective amount of the active ingredient of the pharmaceutical composition of the present invention means an amount required for achieving treatment of a disease. Thus, it may be controlled according to various factors including kind of disease, severity of disease, kind and content of active ingredients and other ingredients contained in the composition, kind of dosage form, and age, weight, general health state, gender and diet of a patient, administration time, administration route, secretion rate of the composition, treatment period, simultaneously used drugs, and the like. For example, in the case of an adult, the compound of the present invention may be administered once or several times a day in the total amount of 50 to 3000mg/kg.

The guanide derivatives according to the present invention may exhibit excellent cancer cell proliferation inhibition and cancer metastasis and recurrence inhibition effects even with a small amount compared to the existing drugs, and thus, may be usefully used for treatment of various cancers including breast, lung, melanoma, pancreas, brain, ovary, prostate, cervix, testes, renal, head and neck, liver, lymphoma, leukemia, endometrial cancer, cholangiocarcinoma and the like, inhibition of cancer cell proliferation, and inhibition of cancer metastasis.

[Advantageous Effect]

The guanide derivatives according to the present invention may exhibit excellent cancer cell proliferation inhibition and cancer metastasis and recurrence inhibition effects even with a small amount compared to the existing drugs, and thus, may be usefully used for treatment of various cancers including breast, lung, melanoma, pancreas, brain, ovary, prostate, cervix, testes, renal, head and neck, liver, lymphoma, leukemia, endometrial cancer, cholangiocarcinoma and the like, inhibition of cancer cell proliferation, and inhibition of cancer metastasis.

[Example]

Hereafter, the invention will be described in more detail through examples and comparative examples. However, the following examples are to merely illustrate the present invention, and the scope of the invention is not limited by them in any ways.

Example 1 : N-(N-phenylcarbamimidoyl)-4,4-difluoropiperidine-l-carboximi damide hydrochloride

4,4-difluoropiperidine hydrochloride (1.0g, 6.34mmol) was dissolved in cyclohexanol

(30ml) at a room temperature and was added by sodium dicyanamide (0.62g, 6.98mmol). The solution was refluxed at 130 ° C for 2 hours and cooled to a room temperature. By using ethylacetate and water, the aqueous layer was separated, dried under reduced pressure, and crystallized in a n-hexane to produce CN imtermediate compound in a white solid (0.89g, 75.2%). The CN imtermediate compound (0.3g, 1.59mmol) was dissolved in cyclohexanol (10ml) at a room temperature and was added by hydrochloride (0.14ml, 1.59mmol) andaniline (0.15ml, 1.59mmol). The solution was refluxed at 130 ° C for 2 hours and cooled to a room temperature. The solution was agitated and the produced solid was filterated to obtain the title compound in white sold (0.46g, 90.2%).

1H NMR (600MHz, DMSO-d6) δ 10.04 (s, 1H), 7.95 (s, 2H), 7.37 (d, J=7.8Hz, 2H), 7.31 (t, J=7.8Hz, 2H), 7.24 (s, 2H), 7.05 (t, J=7.2HZ, IH), 3.60 (m, 4H), 2.08 (m, 4H) LCMS 382.1 [M+H] +

Example 2. N-(N-(5,6,7,8-teterhydronaphthalene-2-yl)carbamimidoyl)-4,4- difluoro piperidine- 1 -carboximidamide h drochloride

The title compound (0.3g, 52.5%) in white solid was obtained according to the same method of Example 1, except that 5,6,7,8-teterhydronaphthalene-2-amine was used.

IH NMR (600MHz, DMSO-d6) δ 9.98 (s, IH), 7.95 (s, 2H), 7.31 (s, 2H), 7.08 (d, J=7.8Hz, IH), 7.01 (s, IH), 6.99 (d, J=7.8Hz, IH), 3.61 (m, 4H), 2.66 (m, 4H), 2.07 (m, 4H), 1.70 (m, 4H) LCMS 336.1 [M+H] +

Example 3 : N-(N-(3,4-dichlorophenyl)carbamimidoyl)-4,4-difluoropiperidi ne-l- carboximidamide hydrochloride ,

The title compound (0.36g, 58.1%) in white solid was obtained according to the same method of Example 1, except that 3,4-dichloroaniline was used.

IH NMR (600MHz, DMSO-d6) δ 10.43 (s, IH), 8.09 (s, 2H), 7.74 (d, J=2.4Hz, IH), 7.55 (d, J=9.0HZ, IH), 7.34 (d, J=9.0Hz, IH), 7.25 (s, 2H), 3.62 (m, 4H), 2.10 (m, 4H)

LCMS 350.0, 352.0 [M+H] +

Example 4. N-(N-(2-bromophenyl)carbamimidoyl)-4,4-difluoropiperidine- 1 - carboximidamide hydrochloride

The title compound (0.26g, 61.9%) in white solid was obtained according to the same method of Example 1, except that 2-bromoaniline was used.

IH NMR (600MHz, DMSO-d6) δ 9.02 (s, IH), 7.89 (s, IH), 7.65 (d, J=7.8Hz, IH), 7.60

(d, J=8.4Hz, IH), 7.39 (s, 2H), 7.35 (t, J=7.2Hz, 2H), 7.1 1 (t, J=7.8Hz, I H), 3.55 (m, 4H), 2.04 (m, 4H) LCMS 360.0, 362.0[M+H] +

Example 5. N-(N-(3-bromophenyl)carbamimidoyl)-4,4-difluoropiperidine- 1 carboximidamide hydrochloride

The title compound (0.33g, 78.6%) in white solid was obtained according to the same method of Example 1, except that 3-bromoaniline was used.

IH NMR (600MHz, DMSO-d6) δ 10.21 (s, IH), 8.01 (s, 2H), 7.67 (s, IH), 7.34 (d, J=7.8Hz, IH), 7.24 (t, J=8.4Hz, IH), 7.22 (s, 2H), 3.61 (m, 4H), 2.10 (m, 4H) LCMS 360.0, 362.0 [M+H] +

Example 6. N-(N-(4-bromophenyl)carbamimidoyl)-4,4-difluoropiperidine- 1 - carboximidamide hydrochloride

The title compound (0.33g, 78.6%) in white solid was obtained according to the same method of Example 1 , except that 4-bromoaniline was used.

IH NMR (600MHz, DMSO-d6) δ 10.35 (s, IH), 8.08 (s, 2H), 7.48 (d, J=8.4Hz, 2H), 7.37 (d, J=8.4HZ, 2H), 7.31 (s, 4H), 3.63 (m, 4H), 2.1 1 (m, 4H) LCMS 360.0, 362.0 [M+H] +

Example 7. N-(N-(4-(trifluoromethoxy)phenyl)carbamimidoyl)-4-(dimethyla mino) piperidine- 1 -carboximidamide hydrochloride

The title compound (0.04g, 1 1.0%) in white solid was obtained according to the same method of Example 1, except that N,N-dimethylpiperidine-4amine and 4- trifluoromethoxyaniline were used.

IH NMR (600MHz, DMSO-d6) δ 10.31 (s, IH), 7.93 (s, 2H), 7.50 (d, J=9.6Hz, 2H)(, 7.31 (d, J=8.4Hz, 2H), 7.18 (s, 2H), 4.45 (s, IH), 4.19 (d, J=12.6Hz, IH), 3.43 (m, IH), 2.97 (m, IH), 2.68 (s, 6H), 2.15 (m, 2H), 1.72 (m, 2H) LCMS 373.1 [M+H] +

Example 8. N-(N-(4-(methythio)phenyl)carbamimidoyl)-4,4-difluoropiperid ine- 1 - carboximidamide hydrochloride

The title compound (0.39g, 67.2%) in white solid was obtained according to the same method of Example 1 , except that 4-methythioaniline was used.

IH NMR (600MHz, DMSO-d6) δ 9.99 (s, IH), 7.92 (s, 2H), 7.32 (d, J=8.4Hz, 2H), 7.23 (d, J=8.4Hz, 2H), 7.20 (s, 2H), 3.60 (m, 4H), 2.44 (s, 3H), 2.08 (m, 4H) LCMS 328.3 [M+H] +

Example 9. N-(N-(4-(trifluoromethythio)phenyl)carbamimidoyl)-4,4-difluo ro piperidine- 1 -carboximidamide hydrochloride

The title compound (0.38g, 56.7%) in white solid was obtained according to the same method of Example 1 , except that 4-trifluoromethythioaniline was used.

1H NMR (600MHz, DMSO-d6) δ 10.35 (s, 1H), 8.06 (s, 2H), 7.62 (m, 2H), 7.56 (m, 2H), 7.23 (s, 2H), 3.61 (s, 4H), 2.10 (m, 4H) LCMS 382.3 [M+H] +

Example 10. N-(N-(4-isopropylphenyl)carbamimidoyl)-4,4-difluoropiperidin e-l- carboximidamide hydrochloride

The title compound (0.19g, 33.3%) in white solid was obtained according to the same method of Example 1 , except that 4-isopropylaniline was used.

1H NMR (600MHz, DMSO-d6) δ 9.91 (s, 1H), 7.90 (s, 2H), 7.26 (m, 2H), 7.25 (s, 2H), 7.18 (m, 2H), 3.60 (m, 4H), 2.84 (m, 1H), 2.07 (m, 4H) 1.18 (d, J= 7.2Hz, 6H)

LCMS 324.3 [M+H] +

Example 11. N-(N-(3-chloro-4-fluorophenyl)carbamimidoyl)-4,4-difluoropip eridine-l- carboximidamide hydrochloride

The title compound (0.37g, 62.7%) in white solid was obtained according to the same method of Example 1 , except that 3-chloro-4-fluoroaniline was used.

1H NMR (600MHz, DMSO-d6) δ 10.30 (s, 1H), 8.05 (s, 2H)), 7.66 (m, 1H), 7.36 (s, 2H), 7.32 (m, 1H), 7.30 (s, 2H), 3.61 (m, 4H), 1.99 (m, 4H) LCMS 334.1 [M+H] +

Example 12. N-(N-(5,6,7,8-teterhydronaphthalen- 1 -yl)carbamimidoyl)-4,4- difluoropiperidine- 1 -carboximidamide hydrochloride

The title compound (0.34g, 57.6%) in white solid was obtained according to the same method of Example 1, except 5,6,7, 8-teterhydronaphthalene-l-amine was used.

1H NMR (600MHz, DMSO-d6) δ 9.34 (s, IH), 7.83 (s, 2H), 7.44 (s, 2H), 7.18 (d, J=7.8HZ, IH), 7.06 (t, J=7.8Hz, IH), 6.94 (d, J=7.8Hz, IH), 3.36 (m, 4H), 2.71 (m, 4H), 2.03 (m, 4H), 1.69 (m, 4H) LCMS 336.4 [M+H] +

Example 13. N-(N-(3-phenoxyphenyl)carbamimidoyl)-4,4-difluoropiperidine- l - carboximidamide hydrochloride

The title compound (0.27g, 61.3%) in white solid was obtained according to the same method of Example 1 , except that 3- phenoxyaniline was used.

1H NMR (600MHz, DMSO-d6) δ 10.10 (s, IH), 7.91 (s, 2H), 7.42 (m, 2H), 7.27 (m, IH), 7.25 (s, IH), 7.18 (s, 2H), 7.15 (m, IH), 7.13 (m, IH), 7.07 (m, IH), 7.00 (m, 2H), 3.56 (s, 4H), 2.03 (s, 4H) LCMS 374.1 [M+H] +

Example 14. N-(N-(4-(trifluoromethythio)phenyl)carbamimidoyl)thiazolidin e-3- carboximidamide hydrochloride

The title compound (0.14g, 54.0%) in white solid was obtained according to the same method of Example 1, except that thiazolidine and 4-trifluoromethythioaniline was used. 1H NMR (600MHz, DMSO-d6) δ 10.23 (brs, 0.5H), 7.92 (brs, 1H), 7.63 (m, 2H), 7.19 (m, 1H), 4.50 (s, 1H), 3.68 (m, 1H), 3.14 (m, 1H), 3.55 (m, 2H) LCMS 350.0 [M+H] +

Example 15. N-(N-(3,4,5-trimethoxyphenyl)carbamimidoyl)-3,3-difluoropyrr olidine-l - carboximidamide hydrochloride

The title compound (0.068g, 10.0%) in white solid was obtained according to the same method of Example 1, except that 3,3-difluoropyrrolidine hydrochloride and 2,3,4- trimethoxyaniline were used.

1H NMR (600MHz, DMSO-d6) δ 6.43 (brs, 2H), 3.76 (t, J=13.2 Hz, 2H), 3.71 (s, 3H), 3.59 (s, 3H), 3.55 (t, J=7.2 Hz, 2H), 2.44 (m, 2H), 1.76 (s, 3H) LCMS 358.2 [M+H] +

Example 16. N-(N-(4-(trifluoromethoxy)phenyl)carbamimidoyl)-4-chloropipe ridine- 1 - carboximidamide hydrochloride

The title compound (610mg, 100%) in white solid was obtained according to the same method of Example 1 , except that 4-chloropiperidine and 4-trifluoromethoxyaniline was used.

1H NMR (600MHz, DMSO-d6) δ 7.79 (m, 1H), 7.46 (d, J=9.0 Hz, 1H), 7.29 (m, 1H), 7.22 (m, 1H), 4.45 (m, 1H), 3.71 (m, 2H), 3.38 (m, 2H), 2.10 (m, 2H), 1.79 (m, 2H)

LCMS 364.1 [M+H] +

Example 17. N-(TSI-(4-(trifluoromethoxy)phenyl)carbamimidoyl)-4,4-difluo ropiperidine- 1 -carboximidamide acetate

4,4-dichloropiperidine hydrochloride (l.Og, 6.34mmol) was dissolved in cyclohexanol (30ml) at a room temperature and added by sodium dicyanamide (0.62g, 6.98mmol). The solution was refluxed at 130 ° C for 2 hours and cooled to a room temperature. By using ethylacetate and water, the aqueous layer was separated, dried under reduced pressure, and crystallized in a n-hexane to produce CN imtermediate compound in a white solid (0.89g, 75.2%). The CN imtermediate compound (0.5g, 2.65mmol) was dissolved in cyclohexanol (20ml) at a room temperature and was added by hydrochloride (0.23ml, 2.65mol) and 4- trifluoromethoxyaniline (0.35ml, 2.65mmol). The solution was refluxed at 130 ° C for 1 hour and cooled to a room temperature. The produced solid was filterated, dissolved in methanol (10ml) and agitated for 1 hour with addition of 1.5M NaOH (0.5ml). The procued solid was filterated, dissolved in methanol ( 10ml) and agitated for 1 hour at a room tempearature with addition of acetic acid solution (0.23ml, 3.98mmol). The reaction solution was distilled under vacuum and crystallized in ethylacetate to produce the title compound in white solid (0.7g, 56.3%).

1H NMR (600MHz, DMSO-d6) δ 7.31 (s, 2H), 7.22 (d, J=9.0Hz, 2H), 3.58 (m, 4H), 2.00 (m, 4H), 175 (s, 3H)

LCMS 366.1 [M+H] +

Example 18. N-(N-(3-chloro-4-iodophenyl)carbamimidoyl)-4,4-difluoropiper idine-l- carboximidamide acetate

The title compound (0.10g, 19.6%) in white solid was obtained according to the same method of Example 17, except that 3-chloro-4-iodoaniline was used.

IH NMR (600MHz, DMSO-d6) δ 10.52 (s, IH), 8.08 (s, 2H), 7.81 (d, J=8.4Hz, IH), 7.71 (d, J=2.4Hz, IH), 7.29 (s, 2H), 7.10 (dd, J=8.4Hz, 2.4Hz, IH), 3.62 (s, 4H), 1.90 (s, 4H) LCMS 442.0 [M+Hf

Example 19. N-(N-(4-methoxyphenyl)carbamimidoyl)-4,4-difluoropiperidine- l- carboximidamide acetate

The title compound (0.46g, 77.9%) in white solid was obtained according to the same method of Example 17, except that 4-methoxyaniline was used.

IH NMR (600MHz, DMSO-d6) δ 7.17 (s, 2H), 6.81 (m, 2H), 3.54 (s, 4H), 1.98 (s, 4H), 1.71 (s, 3H) LCMS 312.2 [M+H] +

Example 20. N-(N-(3-methoxyphenyl)carbamimidoyl)-4,4-difluoropiperidine- l- carboximidamide acetate

The title compound (0.36g, 61.0%) in white solid was obtained according to the same method of Example 17, except that 3-methoxyaniline was used.

IH NMR (600MHz, DMSO-d6) δ 7.13 (t, J=8.4Hz, IH), 6.90 (s, IH), 6.81 (d, J=7.2Hz, IH), 6.54 (dd, J=8.4Hz, IH), 3.57 (m, 4H), 2.01 (m, 4H), 1.73 (s, 3H) LCMS 312.2 [M+H] +

Example 21. N- -(4-(trifluoromethyl)phenyl)carbamimidoyl)-4,4-difluoropiper idine-l- carboximidamide acetate.

The title compound (0.35g, 53.8%)in white solid was obtained according to the same method of Example 17, except that 4-trifluoromethylaniline was used.

IH NMR (600MHz, DMSO-d6) δ 7.57 (d, J=8.4Hz, 2H), 7.49 (d, J=7.2Hz, 2Η), 3.60 (m, 4H), 2.04 (m, 4H), 1.76 (s, 3H) LCMS 350.2 [M+H] +

Example 22. N-(N-(3-bromo-4-iodophenyl)carbamimidoyl)-4,4-difluoropiperi dine-l- carboximidamide acetate

The title compound (0.30g, 34.4%) in white solid was obtained according to the same method of Example 17, except that 3-bromo-4-iodoaniline was used.

IH NMR (600MHz, DMSO-d6) δ 7.74 (d, J=8.4Ha, IH), 7.68 (s, IH), 7.03 (d, J=7.2Hz, IH), 3.59 (m, 4H), 2.50 (m, 4H), 1.76 (s, 3H) LCMS 487.0, 488.0[M+H] +

Example 23. N-(N-(4-phenoxyphenyl)carbamimidoyl)-4,4-difluoropiperidine- l- carboximidamide acetate

The title compound (0.32g, 46.3%) in white solid was obtained according to the same method of Example 17, except that 4-phenoxyaniline was used.

1H MR (600MHz, DMSO-d6) δ 7.36 (m, IH), 7.26 (s, IH), 7.07 (m, IH), 6.93 (m, IH), 3.58 (m, 4H), 2.00 (m, 4H), 1.73 (s, 3H) LCMS 374.2 [M+H] + Example 24. N-(N-(4-(trifluoromethoxy)carbamimidoyl)-4-ethoxypiperidine- l - carboximidamide acetate

The title compound ((XlOg, 14.4%) in white solid was obtained according to the same method of Example 17, except that 4-ethoxypiperidine was used.

1H NMR (600MHz, DMSO-d6) δ 7.43 (m, 2H), 7.22 (m, 2H), 3.72 (m, 2H), 3.52 (q, 2H), 3.17 (m, 2H), 1.85 (m, 2H), 1.82 (s, 3H), 1.45 (m, 2H), 1.11 (t, 3H) LCMS 374

[M+H] +

Example 25. N-(N-(4-(trifluoromethoxy)phenyl)carbamimidoyl)-3,3- difluoropyrrolidine- 1 -carboximidamide acetate

The title compound (0.25g, 36.7%)in white solid was obtained according to the same method of Example 17, except that 3,3-difluoropyrrolidine hydrochloride was used.

1H NMR (600MHz, DMSO-d6) δ 7.36 (s, 2H), 7.23 (d, J=9.0Hz, 2H), 7.78 (t, J=12.6Hz, 2H), 2.57 (t, J=7.2Hz, 2H), 2.47 (m, 2H), 1.75 (s, 3H)

LCMS 352.1 [M+H] +

Example 26. N-(N-(3 ,4-dibromophenyl)carbamimidoyl)-4,4-difluoropiperidine- 1 - carboximidamide acetate

The title compound (0.080g, 14.0%) in white solid was obtained according to the same method of Example 17, except that 3,4-dibromoaniline was used.

1H NMR (600MHz, DMSO-d6) δ 10.42 (s, 1H), 8.06 (s, 2H), 7.86 (m, 1H), 7.65 8.4Hz, 1H), 7.31 (m, 1H), 7.23 (s, 2H), 3.62 (s, 4H), 2.10 (s, 4H)

LCMS 440.1 [M+H] +

Example 27. N-(N-(3-fluoro-4-(trifluoromethoxy)phenyl)carbamimidoyl)-4,4 - fluoropiperidine- 1 -carboximidamide acetate

The title compound (0.02g, 8.2%) in white solid was obtained according to the same method of Example 17, except that 3-fluoro-4-trifluoromethoxyaniline was used.

1H NMR (600MHz, DMSO-d6) δ 6.73 (m, 1H), 6.40 (m, 1H), 6.37 (m, 1H), 2.91 (s, 4H), 1.28 (s, 4H), 1.14 (s, 3H) LCMS 3842 [M+H] +

Example 28. N-(N-(4-phenoxyphenyl)carbamimidoyl)-3 ,3 -difluoropyrrolidine- 1 - carboximidamide acetate

The title compound (0.20g, 49.8%) in white solid was obtained according to the same method of Example 17, except that 3,3-difluoropyrrolidine hydrochloride and 4-phenoxyaniline were used.

1H NMR (600MHz, DMSO-d6) δ 7.35 (m, 2H), 7.18 (m, 2H), 7.18 (brs, 2H), 7.07 (m, 2H), 6.94 (m, 2H), 3.76 (m, 2H), 3.56 (m, 2H), 2.46 (m, 2H), 1.76 (s, 3H) LCMS 360.0 [M+H] +

Example 29. N-(N-(3-phenoxyphenyl)carbamimidoyl)-3,3-difluoropyrrolidine -l- carboximidamide acetate

The title compound (0.36g, 61.0%) in white solid was obtained according to the same method of Example 17, except that 3,3-difluoropyrrolidine hydrochloride and 3 -phenoxy aniline were used.

1H MR (600MHz, DMSO-d6) δ 7.37 (t, J=7.8Hz, 2H), 7.12 (m, 2H), 7.01 (m, 4H), 6.57 (m, 1H), 3.70 (t, J=7.2Hz, 2H), 2.44 (m, 2H), 1.74 (s, 3H)

LCMS 360.0 [M+H] +

Example 30. N-(N-(4-(trifluoromethyl)phenyl)carbamimidoyl)-3,3-difluorop yrrolidine- 1 -carboximidamide acetate

The title compound (0.2 lg, 46.3%) in white solid was obtained according to the same method of Example 17, except that 3,3-difluoropyrrolidine hydrochloride and 4- trifluoromethylaniline was used.

1H NMR (600MHz, DMSO-d6) δ 7.55 (m, 2H), 7.55 (m, 2H), 7.44 (brs, 2H), 3.79 (t, J=12.6Hz, 2H), 3.58 (t, J=7.8 Hz, 2H), 2.48 (m, 2H), 1.77 (s, 3H)

LCMS 368.0 [M+H] +

Example 31. N-(N-(3-chloro-4-iodophenyl)carbamimidoyl)-3,3-difluoropyrro lidine- 1 - carboximidamide acetate

The title compound (0.06g, 11.0%) in white solid was obtained according to the same method of Example 17, except that 3,3-difluoropiperidine hydrochloride and 3-chloro-4- iodoaniline was used.

1H NMR (600MHz, DMSO-d6) 5 7.55 (m, 2H), 7.70 (m, 1H), 7.37 (brs, 1H), 6.87 (brs, 1H), 3.77 (t, J=13.2Hz, 2H), 3.56 (t, J=7.2 Hz, 2H), 2.47 (m, 2H), 1.80 (s, 3H) LCMS 427.9 [M+H] + Example 32. N-(N-(4-(trifluoromethythio)phenyl)carbamimidoyl)-3,3- difluoropyrrolidine-l-carboximidamide acetate

The title compound (0.04g, 8.2%)in white solid was obtained according to the same method of Example 17, except that 3,3-difluoropiperidine hydrochloride and 3- trifluoromethythioaniline were used.

1H NMR (600MHz, DMSO-d6) δ 7.55 (m, 2H), 7.35 (m, 2H), 3.79 (t, J=12.6Hz, 2H), 3.58 (t, J=7.8 Hz, 2H), 2.48 (m, 2H), 1.80 (s, 3H)

LCMS 336.0 [M+H] +

Example 33. N-(N-(2-chlorophenyl)carbamimidoy l)-3 ,3 -difluoropyrrolidine- 1 - carboximidamide acetate

The title compound (0.09g, 22.0%) in white solid was obtained according to the same method of Example 17, except that 3,3-difluoropiperidine hydrochloride and 2-chloroaniline were used.

1H NMR (600MHz, DMSO-d6) δ .37 (dd, J=7.2 Hz, 1H), 7.20 (m, 1H), 7.06 (brs, 1H), 6.91 (m, 1H), 3.75 (t, J=13.2 Hz, 2H), 3.54 (t, J=7.8 Hz, 2H), 2.46 (m, 2H), 1.88 (s, 3H) LCMS 302.0 [M+H] +

Example 34. N-(N-(2-bromophenyl)carbamimidoyl)-3, 3 -difluoropyrrolidine- 1 - carboximidamide acetate

The title compound (0.1 lg, 24.6%) in white solid was obtained according to the same method of Example 17, except that 3,3-difluoro pyrrolidine hydrochloride and 2-bromoaniline were used.

IH NMR (600MHz, DMSO-d6) δ 7.53 (dd, J=18 Hz, IH), 7.22 (m, IH), 7.00 (d, J=7.2 Hz, IH), 6.88 (m, IH), 3.76 (t, J=13.2 Hz, 2H), 3.55 (t, J=7.2 Hz, 2H), 2.45 (m, 2H), 1.88 (s, 3H) LCMS 345.9, 347.0 [M+H] +

Example 35. N-(N-(2,4-dichlorophenyl)carbamimidoyl)-3 ,3 -difluoropyrrolidine- 1 - carboximidamide acetate

The title compound (0.10g, 22.0%)in white solid was obtained according to the same method of Example 17, except that 3,3-difluoropiperidine hydrochloride and 2,4-dichloroaniline were used.

IH NMR (600MHz, DMSO-d6) δ 7.42 (m, IH), 7.21 (m, 2H), 7.0 (m, IH), 3.68 (m, 2H), 3.54 (m, 2H), 2.42 (m, 2H), 1.89 (s, 3H)

LCMS 336.0, 337.9 [M+H] +

Example 36. N-(N-(2-propylphenyl)carbamimidoyl)-3,3-difluoropyrrolidine- l- carboximidamide acetate

The title compound (0.19g, 45.0%) in white solid was obtained according to the same method of Example 17, except that 3,3-difluoro pyrrolidine hydrochloride and 2-propyl aniline was used. 1H NMR (600MHz, DMSO-d6) δ 7.17 (m, 2H), 6.85 (m, 2H), 3.65 (m, 2H), 3.49 (s, 2H), 2.48 (m, 4H), 1.78 (s, 3H), 1.51 (m, 2H), 0.86 (s, 3H)

LCMS 310.1 [M+H] +

Example 37. N-(N-3 ,4-dimethoxyphenyl)carbamimidoyl)-3 ,3 -difluoropyrrolidine- 1 - carboximidamide hydrochloride

The title compound (0.16g, 24.0%) in white solid was obtained according to the same method of Example 17, except that 3, 3 -difluoropyrrolidine hydrochloride and 3,4- dimethoxyaniline were used.

IH NMR (600MHz, DMSO-d6) δ 6.41 (brs, 2H), 6.11 (d, J=18 Hz, 2H),3.77 (t, J=12.6 Hz, 2H), 3.55 (s, 3H), 3.56 (t, J=6.6 Hz, 2H), 2.46 (m, 2H), 1.76 (s, 3H) LCMS 328.1 [M+H] +

Example 38. N-(N-(3-chloro-4-(trifluoromethoxy)phenyl)carbamimidoyl)-3,3 - difluoropyrrolidine- 1 -carboximidamide acetate

The title compound (0.06g, 21.0%) in white solid was obtained according to the same method of Example 17, except that 3, 3 -difluoropyrrolidine hydrochloride and 3-chloro-4trifluoro methoxyaniline were used.

1H NMR (600MHz, DMSO-d6) δ 7.36 (m, 2H), 7.19 (brs, 1H), 3.77 (t, J=13.8 Hz, 2H), 3.56 (m, 2H), 2.46 (m, 2H), 1.81 (s, 3H) LCMS 386.1 [M+H] +

Example 39 N-(N-(3-bromo-4-(trifluoromethoxy)phenyl)carbamimidoyl)-3,3- difluoropyrrolidine- 1 -carboximidamide acetate

The title compound (0.09g, 11.0%) in white solid was obtained according to the same method of Example 17, except that 3,3-difluoropyrrolidine hydrochloride and 3-bromo-4- trifluoro methoxyaniline were used.

1H NMR (600MHz, DMSO-d6) 5 7.51 (brs, IH), 7.35 (d, J=8.4 Hz, 2Η), 7.20 (brs, 2Η), 3.78 (t, J=13.2 Hz, 2H), 3.37 (m, 2H), 2.46 (m, 2H), 1.80 (s, 3H) LCMS 432.0 [M+H] +

Example 40. N-(N-(2-bromobenzyl)carbamimidoyl)-4,4-difluoropiperidine- 1 - carboximidamide hydrochloride H CI

4,4-dichloropiperidine hydrochloride (l .Og, 6.34mmol) was dissolved in cyclohexanol (30ml) at a room temperature and was added by sodium dicyanamide (0.62g, 6.98mmol). The solution was refluxed at 130 ° C for 2 hours and cooled to a room temperature. By using ethylacetate and water, the aqueous layer was separated, dried under reduced pressure, and crystallized in a n-hexane to produce CN imtermediate compound in a white solid (0.89g, 75.2%). 2-bromobenzylamine (0.2ml, 1.06mmol) was dissolved in dichloroethane (2 mL) at a room temperature and was added by trimethylsilyltrifluoromethanesulfonate (0.23ml, 1.27mmol). The solution was agitated for 30 minutes at a room temperature, added by CN imtermediate compound (0.21 , 1.06mmol) and refuxed for 15 hours at 80°C. After the reaction was completed, the solution was cooled to room temperature, and was agitated for 1 hour after addition of 12N hydrochloride (0.18ml, 2.13mmol). The produced solid was filtered, washed with

dichloroethane(lOml), dissolved in a small amount of methanol, and agitated for 1 hour at a room temperature after addition of ethylaceate 20ml. The produced solid was filtered, washed with ethylaceate 10 ml and dried under vacuum to produce the title compound in white solid (0.25g, 56.8%).

1H NMR (600MHz, DMSO-d6) δ 7.69 (s, IH), 7.62 (d, J=8.4Hz, IH), 7.45 (d, J=6.6Hz, IH), 7.40 (t, J=7.8Hz, IH), 7.24 (t, J=7.2HZ, IH), 7.1 1 (s, 2H), 4.40 (s, 2H), 3.54 (s, 4H), 2.09 (s, 4H) LCMS 374.1, 376.1 [M+Hf

Example 41. N-(N-(4-(trifluoromethoxy)benzyl)carbamimidoyl)-4,4-difluoro piperidine- 1 -carboximidamide hydrochloride

5

The title compound (0.22g, 50.0%) in white solid was obtained according to the same method of Example 40, except that 4-trifluoromethoxybenzylamine was used.

IH NMR (600MHz, DMSO-d6) δ 7.70 (s, IH), 7.48 (s, 2H), 7.35 (d, J=7.8Hz, 2H), 7.05 (s, 2H), 4.44 (s, 2H), 3.56 (s, 4H), 1.98 (s, 4H)

LCMS 380.4 [M+H] +

Example 42. N-(N-(3,4-dichlorobenzyl)carbamimidoyl)-4,4-difluoropiperidi ne-l- carboximidamide hydrochloride

The title compound (0.20g, 46.5%) in white solid was obtained according to the same method of Example 40, except that 3,4-dichlorobenzylamine was used.

IH NMR (600MHz, DMSO-d6) δ 7.62 (d, J=7.8Hz, 2H), 7.35 (s, IH), 7.03 (s, 3H), 4.38

(s, 2H), 3.56 (s, 4H), 2.08 (s, 4H) LCMS 364.1 [M+H] +

Example 43. N-(N-(4-trifluoromethyl)benzyl)carbamimidoyl)-3 ,3-difluoropyrrolidine- 1 - carboximidamide hydrochloride

H

The title compound (0.08g, 12.0%) in white solid was obtained according to the same method of Example 40, except that 3,3-difluoropyrrolidine hydrochloride and 4- trifluoroemthylbenzylamine were used.

1H NMR (600MHz, DMSO-d6) 6 7.69 (m, 2H), 7.5 l(m, 2H), 4.42 (d, J=4.8Hz, 2H), 3.67 (brs, 2H), 3.50 (m, 2H), 2.45 (m, 2H) LCMS: 350.1 [M+H] +

Example 44. N-(N-(3-trifluoromethyl)benzyl)carbamimidoyl)-3,3-difluoropy rrolidine-l - carboximidamide hydrochloride

The title compound (0.12g, 23.0%) in white solid was obtained according to the same method of Example 40, except that 3,3-difluoropyrrolidine hydrochloride and 3- trifluoroemthylbenzylamine were used.

1H NMR (600MHz, CD30D) δ 7.64 (m, 4H), 7.54 (m, 3H), 4.50 (s, 2H), 3.71 (brs, 2H), 3.62 (m, 2H) 2.46 (m, 2H) LCMS: 350.2 [M+H] +

Example 45. N-(N-(3,4-dichloro)benzyl)carbamimidoyl)-3,3-difluoropyrroli dine-l- carboximidamide hydrochloride

The title compound (0.06g, 10.0%) in white solid was obtained according to the same method of Example 40, except that 3,3-difluoropyrrolidine hydrochloride and 3,4- dichlorobenzylamine were used.

1H NMR (600MHz, DMSO-d6) δ 8.01 (m, 1H), 7.58 (m, 2H), 7.47 (brs, 2H), 7.29 (m, 2H), 7.07 (brs, 2H), 4.33 (d, J=6.0Hz, 2H), 3.70 (brs, 2H), 3.52 (brs, 2H), 2.48 (m, 2H)

LCMS: 350.0, 352.0 [M+H] + Example 46. N-( -phenethylcarbamimidoyl)-3,3-difluoropyrrolidine- 1 - carboximidamide hydrochloride

The title compound (0.12g, 23.0%) in white solid was obtained according to the same method of Example 40, except that 3,3-difluoropyrrolidine hydrochloride and phenethylamine were used.

1H NMR (600MHz, DMSO-d6) δ 7.33 (s, 1H), 7.25 (m, 5H), 3.75 (m, 2H), 3.57 (m,

2H), 3.34 (m, 2H), 2.78 (m, 2H), 2.49 (m, 2H) LCMS 266.1 [M+H] +

Example 47. N-(N-(4-bromophenethyl)carbamimidoyl)-3,3-difluoropyrrolidin e-l- carboximidamide hydrochloride

The title compound (0.12g, 25.0%) in white solid was obtained according to the same method of Example 40, except that 3,3-difluoropyrrolidine hydrochloride and 4- bromophenethylamine were used.

1H NMR (600MHz, DMSO-d6) δ 7.48 (m, 2H), 7.24 (m, 2H), 3.78 (m, 2H), 3.57 (m, 2H), 3.35 (m, 2H), 2.76 (m, 2H), 2.49 (m, 2H) LCMS 374.0 376.0 [M+H] +

Example 48. N-(N-(cyclopropylmethyl)carbamimidoyl)-4,4-difluoropiperidin e- 1 - carboximidamide hydrochloride

The title compound (0.67g, 88.2%) in white solid was obtained according to the same method of Example 40, except that cyclopropylmethylamine was used.

1H NMR (600MHz, DMSO-d6) δ 8.19 (s, 1H), 7.59 (s, 2H), 6.89 (s 1H), 3.59 (s, 4H), 3.12 (s, 2H) 1.98 (s, 4H), 0.99 (s, 2H), 0.45 (s, 2H), 0.21 (s, 2H) LCMS 260.1 [M+H] +

Example 49. N-(N-cyclopropylcarbamimidoyl)-4,4-difluoropiperidine- 1 - carboximidamide hydrochloride

The title compound (0.58g, 88.0%) in white solid was obtained according to the same method of Example 40, except that cyclopropylamine was used.

1H NMR (600MHz, DMSO-d6) δ 8.10 (s, 1H), 7.59 (s, 2H), 6.83 (s 1H), 3.31 (s, 4H),

3.22 (s, 1H), 1.88 (s, 4H), 1.12 (s, 4H) LCMS 246.1 [M+H] +

Example 50. N-(N-cyclohexyl carbamimidoyl)-4 ,4 -difluoropiperidine- 1 - carboximidamide h drochloride

The title compound 0.25g, 53.0%) in white solid was obtained according to the same method of Example 40, except that cyclohexylamine was used.

1H NMR (600MHz, DMSO-d6) δ 8.19 (s, 1H), 7.34 (s, 2H), 7.00 (s, 2H), 3.81 (m, 4H), 3.63 (m, 1H), 3.31 (m, 2H), 2.99 (m, 2H), 2.30 (m, 4H), 2.20 (m, 4H), 1.02 (m, 2H)

LCMS 288.0 [M+H] +

Example 51. N-(N-(cyclopropylmethyl)carbamimidoyl)-3 ,3 -difluoropyrrolidine- 1 - carboximidamide hydrochloride The title compound (0.50g, 72.1%) in white solid was obtained according to the same method of Example 40, except that 3,3 difluoropyrrolidine hydrochloride and

cyclopropylmethylamine were used.

IH NMR (600MHz, DMSO-d6) δ 8.02 (s, IH), 7.48 (s, 2H), 6.88(s IH), 3.31 (m, 4H), 3.31 (s, 2H), 2.00 (s, 2H), 1.01 (s, IH) 0.45 (s, 2H), 0.21 (s, 2H) LCMS 246.0 [M+H] +

Example 52. N-(N-(cyclopropylcarbamimidoyl)-3,3-difluoropyrrolidine-l - carboximidamide hydrochloride

The title compound (0.72g, 88.0%)in white solid was obtained according to the same method of Example 40, except that 3,3-difluoropyrrolidine hydrochloride and cyclopropylamine were used.

1H NMR (600MHz, DMSO-d6) δ 8.12 (s, IH), 7.58 (s, 2H), 6.78(s IH), 3.31 (m, 4H), 3.31 (s, IH), 2.00 (s, 2H), 089 (s, 4H)

LCMS 232.0 [M+H] +

Example 53. N-(N-(cyclohexylcarbamimidoyl)-3 ,3 -difluoropyrrolidine- 1 - carboximidamide hydrochloride

The title compound (0.25g, 55.3%) in white solid was obtained according to the same method of Example 40, except that 3,3-difluoropyrrolidine hydrochloride and cyclohexylamine were used.

IH NMR (600MHz, DMSO-d6) δ 8.00 (s, IH), 7.12 (s, 2H), 6.93 (s, 2H), 3.91 (m, 4H), 3.77 (m, IH), 3.31 (m, 4H), 2.97 (m, 2H), 232 (m, 2H), 2.19 (m, 4H), 1.05 (m, 2H) Example 54. N-(N-(3-bromobenzyl)carbamimidoyl)-4,4-difluoropiperidine- 1 - carboximidamide acetate

4,4-dichloropiperidine hydrochloride (l .Og, 6.34mmol) was dissolved in cyclohexanol (30ml) at a room temperature and was added by sodium dicyanamide (0.62g, 6.98mmol). The solution was refluxed at 130 ° C for 2 hours and cooled to a room temperature. By using ethylacetate and water, the aqueous layerwas separated, dried under reduced pressure, and crystallized in a n-hexane to produce CN imtermediate compound in a white solid (0.89g, 75.2%). 3-bromobenzylamine (0.13ml, 1.06mmol) was dissolved in dichloroethane (10 mL) at a room temperature and was added by trimethylsilyltrifluoromethanesulfonate (0.23ml, 1.28mmol). The solution was agitated for 30 minutes at a room temperature, added by CN imtermediate compound (0.20, 1.06mmol) and refuxed for 15 hours at 80°C. After the reaction was completed, the solution was cooled to room temperature, and was agitated for 1 hour after addition of 12N hydrochloride (0.18ml, 2.13mmol). The produced solid was filtered, washed with

dichloroethane(lOml), dissolved in a small amount of methanol, and agitated for 1 hour at a room temperature after addition of 1.5M NaOH (0.5ml). The produced solid. was filtered, dissolved in methanol solution (10 ml) and agitated for 1 hour at room temperature after the addition of acetic acid solution (0.23ml, 3.98mmol). The reaction solution was distilled under vacuum and crystallized in ethylacetate to produce the title compound in white solid (0.16g, 34.7%).

1H NMR (600MHz, DMSO-d6) δ 7.59 (s, 1H), 7.46 (d, J=7.8Hz, 1H), 7.30 (m, 1H), 4.33 (s, 2H), 3.52 (s, 4H), 3.34 (s, 3H), 1.97 (s, 4H)

LCMS 374.1 , 376.1 [M+H] +

Example 55. N-(N-(4-bromobenzyl)carbamimidoyl)-4,4-difluoropiperidine- 1 - carboximidamide acetate

The title compound (0.02g, 4.3%) in white solid was obtained according to the same method of Example 54, except that 4-bromobenzylamine was used.

1H MR (600MHz, DMSO-d6) δ 7.54 (d, J=8.4Hz, 2H), 7.26 (d, J=7.8Hz, 2H), 4.29 (s, 2H), 3.56 (s, 4H), 3.32 (s, 3H), 1.98 (s, 4H)

LCMS 374.1, 376.1 [M+H] +

Example 56. N-(N-(4-(trifluoromethoxy)phenyl)carbamimidoyl)-3,3-difluoro piperidine- 1 -carboximidamide hydrochloride

OCF, NaN(CN) 2 ,c-HCI

Η ' V Cyclohexanol NC^ N c-HCI, Cyclohexanol F~ ^[^J H H

4-trifluoromethoxyaniline (2.0g, 11.3mmol) was dissolved in cyclohexanol (30ml) at a room temperature and was added by sodium dicyanamide (l .lg, 1.24mmol). The solution was refluxed at 130 ° C for 2 hours and cooled to a room temperature. By using ethylacetate and water, the aqueous layer was separated, dried under reduced pressure, and crystallized in a n-hexane to produce CN imtermediate compound in a white solid (1.9g, 70.0%).

3,3-difluoropiperidine (0.05g, 0.41mmol) was dissolved in dichloroethane (2 mL) at a room temperature and was added by trimethylsilyltrifluoromethanesulfonate (0.7ml, 0.41mmol). The solution was agitated for 30 minutes at a room temperature, added by CN imtermediate compound (0.20, 1.06mmol) and refuxed for 15 hours at 80°C. After the reaction was completed, the solution was cooled to room temperature, and was agitated for 1 hour after addition of 12N hydrochloride (0.34ml, 0.41mmol). The produced solid was filtered, washed with

dichloroethane(lOml), dissolved in a small amount of methanol, and agitated for 1 hour at a room temperature after addition of ethyl acetate 20ml. The produced solid was filtered, washed with ethyl acetatelO ml and dried under vacuume to produce the title compound in white solid (0.068g, 41.0%).

1H NMR (600MHz, DMSO-d6) δ 7.91 (m, 1H), 7.44 (m, 1H), 7.28 (m, 1H), 7.15 (m, 1H), 3.84 (t, J=12 Hz, 1H), 3.50 (m, 5H), 2.08 (m, 1H), 1.70 (m, 1H) LCMS 366.1 [M+H]

Example 57. N-(N-(4-(trifluoromethoxy)phenyl)carbamimidoyl)thiazolidine- 3- carboximidamide hydrochloride

The title compound (0.15g, 33.3%) in white solid was obtained according to the same method of Example 56, except that thiazolidin was used.

IH NMR (600MHz, DMSO-d6) δ 7.82 (m, IH), 7.49 (m, IH), 7.30 (d, J=18 Hz, IH), 7.13 (m, IH), 4.76 (brs, 2H), 4.47 (m, IH), 3.64 (d, J=6.6 Hz, IH), 3.12 (m, IH), 1.70 (m, IH) LCMS 334.0 [M+H] +

Example 58. N-(N-(4-(trifluoromethoxy)phenyl)carbamimidoyl)-N-2,2-2-trif luoro ethylaminocarboximidamide hydrochloride

The title compound (0.16g, 34.6%) in white solid was obtained according to the same method of Example 56, except 2,2,2-trifluoroethaneamine was used.

IH NMR (600MHz, DMSO-d6) δ 8.27 (brs, IH), 7.51 (m, 2H), 7.44 (m, 2H), 7.28 (m, 2H), 6.00 (brs, 3H), 4.00 (m, 2H)

LCMS 344.1 [M+H] +

Example 59. N-(N-(4-(trifluoromethoxy)phenyl)carbamimidoyl)-4- (trifluoromethyl)piperidine- 1 -carboximidamide hydrochloride

The title compound (0.04g, 8.0%) in white solid was obtained according to the same method of Example 56, except 4-trifluoromethylpiperidine was used.

IH NMR (600MHz, CD30D) δ 7.46 (m, 2H), 7.25 (d, J=9.0 Hz, 2H), 7.29 (m, IH), 7.21 (d, J=12.6Hz, IH), 3.05 (t, J=12.6Hz, 2H), 2.54 (m, IH), 1.95 (m, 2H),1.60 (m, 2H)

LCMS 398.1 [M+H] +

Example 60. N-(N-(4-(trifluoromethoxy)phenyl)carbamimidoyl)-N-l- cycloproylmethylaminocarboximidamide hydrochloride

The title compound (0.07g, 29.3%) in white solid was obtained according to the same method of Example 56, except cyclopropylmethylamine was used.

IH NMR (600MHz, DMSO-d6) δ 9.86 (brs, IH), 7.84 (brs, IH), 7.47 (m, J=7.2Hz, 2H), 7.29 (m,2H), 7.15 (brs, IH), 2.99 (d, J=5.4Hz, 2H), 1.16 (m, IH), 0.44 (m, 2H) 0.22 (m, 2H) LCMS 316.i l [M+H] +

Example 61. N-(N-(4-(trifluoromethoxy)phenyl)carbamimidoyl)-3,4,4- trifluoropiperidine- 1 -carboximidamide hydrochloride

The title compound (0.06g, 25.1%) in white solid was obtained according to the same method of Example 56, except 3,4,4-trifluoropiperidine was used.

IH NMR (600MHz, CD30D) δ 7.43 (m, 2H), 7.25 (m, 2H), 4.82 (m, 2H), 4.39 (m, IH), 4.05 (m, IH), 3.56 (m, IH), 2.27 (m, IH), 2.13 (m, IH)

LCMS 384.1 [M+H] +

Example 62. N-(N-(4-(trifluoromethoxy)phenyl)carbamimidoyl)-4,4-difluoro -3- methylpiperidine- 1 -carboximidamide hydrochloride

The title compound (0.07g, 29.0%) in white solid was obtained according to the same method of Example 56, except 4,4-difluoro-3-methylpiperidine was used.

IH NMR (600MHz, CD30D) δ 7.43 (m, 2H), 7.25 (m, 2H), 3.95 (m, 2H), 3.34 (m, IH), 3.06 (m, IH), 2.15 (m, IH), 1.99 (m, I H), 1.03 (s, 3H)

LCMS 380.1 [M+H

Example 63. N-(N-(4-(trifluoromethoxy)phenyl)carbamimidoyl)-4-fluoropipe ridine- 1 - carboximidamide hydrochloride

The title compound (0.0 lg, 4.4%) in white solid was obtained according to the same method of Example 56, except 4-fluoropiperidine was used.

IH NMR (600MHz, CD30D) δ 7.44 (m, 2H), 7.23 (m, 2H), 4.93 (m, IH), 3.62 (m, 4H), 1.89 (m, 4H)

LCMS 348.1 [M+H] +

Example 64. N-(N-(4-(trifluoromethoxy)phenyl)carbamimidoyl)-(R)-3- fluoropyrrolidine-1 -carboximidamide hydrochloride

The title compound (0.12g, 41.0%) in white solid was obtained according to the same method of Example 56, except (R)-3-fluoropyrrolidine was used.

IH NMR (600MHz, CD30D) 8 7.45 (d, J=9.0Hz, 2H), 7.22 (d, J=9.0Hz, 2H), 5.24 (m, IH), 3.63 (m, 4H), 2.59 (m, 2H) LCMS 334.1 [M+H] +

Example 65. N-( -(4-(trifluoromethoxy)phenyl)carbamimidoyl)-6-azaspiro[2.5]o ctane- 6-carboximidamide h drochloride

The title compound (0.02g, 8.3%) in white solid was obtained according to the same method of Example 56, except 6-azaspiro[2.5]octane was used.

1H NMR (600MHz, CD30D) δ 7.45 (d, J=9.0Hz, 2H), 7.22 (d, J=9.0Hz, 2H), 3.60 (t, J=5.4Hz, 4H), 1.47 (t, J=6.0Hz, 4H), 1.47 (m, 4H)

LCMS 356.1 [M+H] +

Example 66. N-(N-(4-(trifluoromethoxy)phenyl)carbamimidoyl)-3- azabicyclo[3.1.0]hexane-3-carboximidamide hydrochloride

The title compound (0.16g, 34.6%) in white solid was obtained according to the same method of Example 56, except 3-azabicyclo[3.1.0]hexane was used.

1H NMR (600MHz, CD30D) δ 7.45 (m, 2H), 7.22 (m, 2H), 3.69 (m, 1H), 3.54 (m, 3H), 3.31 (m, 1H), 1.74(m, 1H), 0.84 (m, 1H), 0.21 (m, 1H)

LCMS 328.1 [M+H] +

Example 67. N-carbamimidoyl-3,3-difluoropyrrolidine-l-carboximidamide

hydrochloride

3,3-difluoropyrrolidine (0.03g, 0.21 mmol) and dicyandiamide (0.019g, 0.23mmol) were added and agitated for 2 hours at 110°C. After the reaction was completed, the solution was cooled to room temperature, and was added by methanol. The solution was added by ethylacetate to obtain the solid. The solid was filerated, washed with ethyl acetate, and dried under vacuum to produce the title compound in white solid (0.02g, 58.0 %).

1H NMR (600MHz, DMSO) δ 7.37 (s, 2H), 7.02 (s , 3H), 3.78 (m, 2H), 3.57 (m, 4H) LCMS 191.1 [M+H]+

Example 68. N-carbamimidoyl-4,4-difluoropiperidine- 1 -carboximidamide hydrochloride

The title compound (0.02g, 60.0%) in white solid was obtained according to the same method of Example 67, except that 4,4,-difluoropiperidine hydrochloride was used.

1H NMR (600MHz, CD30D) δ 3.69 (m, 4H), 2.06 (m, 4H)

LCMS: 206.1 [M+H]+

Example 69. N-((4,4-difluoropiperidin-l-yl)(imino)methyl)-4,4-difluoro piperidine-1- carboximidamide hydrochloride

The title compound (0.03g, 40.0%) in white solid was obtained according to the same method of Example 67, except that 4,4,-difluoropiperidine hydrochloride was used.

1H NMR (600MHz, CD30D) δ 3.65 (m, 8H), 1.99 (m, 8H) LCMS: 310.1 [M+H]+

Test Example 1: Test for inhibition of OCR and enhancement of ECAR

A549 cells were purchased from American Type Tissue Culture Collection ( CCL-185™) and cultured in RPMI 1640 supplied with 10% fetal bovine serum (FBS) and antibiotic- antimycotic (Lifetech, CA). A549 cells were separated with 0.5% Trypsin-EDTA and 3,000 cells were plated on 1 mg/ml poly-D-lysine (Sigma, P6407) coated XF 96 well culture media. A549 cells were allowed to adhere to the wells for 24 hours under the condition of temperature, 37 °C and 5% C0 2 . The sensor cartridge of XF Analyzer was soaked in 200 μί of Calibrant solution (Seahorse, MA) in a clear 96-well plate at 37 °C for 24 hours. The compounds of the present invention were diluted with RPMI 1640 without FBS, transferred to A549 cells on XF 96 well plate, and incubated further for 23 hours at 37 °C and 5% C0 2 . After incubation, the compound solution was exchanged with pre-warmed and pH adjusted (pH7.4) XF assay media (Seahorse) supplied with 15 mM D-glucose (Sigma), 15 mM sodium pyruvate (Lifetechnologies, CA) and 4 mM L-glutamine (Lifetechnologies, CA). The compounds of present invention were prepared in XF Assay media and added to the assay plate. The assay plate was equilibrated in XF Analyzer for 1 hour, and the reading were started by sensor cartridge. The cytotoxicity assay was followed using Cyquant (Lifetechnologies, CA) in order to calibrate the inhibition with cytotoxicity of compounds. The concentrations of the compounds were tested at 0, 0.5, 1, 5, 10 and 20 μΜ and IC50 value was obtained from the inhibited values. That is, from OCR inhibition values at each concentration of the compound, IC50 was calculated according to Prism's dose response curve fitting.

In Table 1, the levels of OCR IC50 are evaluated by the following.

A Level: IC50< 2 uM

B Level : IC50 = 2-4 uM

C Level: IC50 >4 uM

According to the test results of the compounds, the compounds were classified into A, B and C. The test results are summarized in Table 1. The test compounds had good OCR inhibitory effect, which suggested the excellent OXPHOX inhibitor. The most compounds having an OCR inhibitory activity showed the increased ECAR.

Table 1

ECAR Enhancement

Example No. OCR IC50 (A549, 24 hrs)

(A549, 24hrs)

2 G No test

3 C No test

5 C No test

6 C No test

8 C No test

9 B No test

10 C No test

11 C No test

13 B No test 14 B No test

17 B No test

18 B No test

21 B +

22 B +

23 B +

25 B +

26 B No test

27 A +

28 B No test

29 A +

30 B +

31 B No test

32 B +

38 B +

39 B No test

40 C No test

42 C No test

55 C No test

Test Example 2: Cytotoxicity in low glucose condition

SK-MEL-28 (HTB-72) is a melanoma cell line obtained from ATCC (MA) and cultured in RPMI 1640 media (Lifetechnologies, CA) with supplement of 10% FBS and antibiotic- antimycotic (Lifetechnologies, CA). RPMI 1640 (-Glucose) was used to prepare low glucose media and 0.75 mM glucose was supplied with D-(+)-glucose solution from Sigma. SK-MEL-28 cells were separated from culture plate using 0.5% trypsin-EDTA and 1 ,250 cells were plated in 96-well plate with low glucose media. After incubation at 37 °C for 24 hour, 5% C0 2> the cells were treated with the compounds of present invention in FBS-free media for 72 hours.

The cytotoxicity was measured by the MTT (AMRESCO, OH) assay. NADH-dependent cellular oxidoreductase reduces MTT to its insoluble tetrazolium with purple color. The enzyme depends on cell number or energy state of cells. 10 βΐ of 5 mg/ml MTT solution was added to each well of the assay plate and skip the well for blank. The plate was incubated at 37°C and 5% C0 2 for 2 hours. The MTT solution was removed from each well and 100 fd of DMSO was added. The plate was read by VICTOR X3 Multilabel Counter at the wavelength of 550 nm. In Table 2, the levels of IC50 are evaluated by the following criteria.

A Level: IC50< 6 uM

B Level : IC50 = 6-15 uM

C Level: IC50 > 15 uM

According to the concentration of compound for killing the half of cell populations (the levels of IC50), the compounds were classified into A, B anc C, when the compounds were treated on SK-MEL-28 cells at 0.75 mM glucose. The test result is summarized in Table 1. The test compounds have good cell death effect at a low concentration, and thus are sugguested to show excellent anti-cancer activity .

Table 2

SK-MEL-280.75mM glucose

Example No.

cell viability (IC50 μΜ)

1 C

2 B

3 A

4 C

5 B

6 B

8 B

9 A

10 B

1 1 B

13 A

14 A

21 A

22 A

22 B

23 A

25 A

26 B

27 A

28 A

29 A

30 B 3 1 B

32 A

38 B

42 B

Test Example 3: In vitro combination study

A549, H1975 (CRL5908, ATCC) or U937 cell (CRL1593.2™, ATCC) lines were cultured in RPMI 1640 media (Lifetechnologies, CA) with supplement of 10% FBS and antibiotic-antimycotic (Lifetechnologies, CA). Dasatinib (Combi-Blocks, San Diego, CA) is Bcr- Abl-tyrosine kinase inhibitor and Src family tyrosine kinase inhibitor approved for CML treatment. GDC094 (Selleckchem, Houston, TX) is a pan-PI3K inhibitor and effective in various cancer cells. H1975 or A549 cells were dissociated from culture plate using 0.5% trypsin-EDTA (Lifetechnologies, CA) and were suspended. U937 cells were spun down using centrifugation. A549 cells were seeded at a density of 3,000 cells/well and HI 975 or U937 cells were seeded at a density of 5,000 cells/well in a 96-well plate, and were allowed to adhere at 37°C and 5% C0 2 for 24 hours. IC50 of each compounds were obtained from each cell line prior to the combination therapy test. Among four concentrations of the compounds of present invention, the highest concentration was set to IC 8 o and the lowest concentration was

approximately IC30. The concentrations of other anticancer drug were ranged from ICso to IC30. The media in 96-well plate were exchanged with the prepared compound solution. To determine cell viability, except U937 cells, 10 μΐ of 5mg/ml MTT in D-PBS (Lifetechnologies, CA) were added to each well after 72-hour treatment and plates were incubated at 37 °C and 5 % C02 for

2 hours. 100 μΐ of DMSO was added after removing MTT solution from each well, and were read by VICTOR X3 Multilabel Counter at the wavelength of 550 nm. The combination index was calculated using Calcusyn (Biosoft, UK).

Test Example 4: In vivo testing OXPHOS inhibitors for anticancer activity

The SK-MEL-239 tumor cell line was maintained in vitro as monolayer in RPMI1 640 medium supplemented with 10%) heat inactivated fetal bovine serum, lOOU/ml penicillin and 100 μg ml streptomycin, and L-glutamine (2 mM) at 37°C in an atmosphere of 5%> C0 2 in air. The tumor cells were routinely subcultured twice weekly with 0.5% trypsin-EDTA treatment. The cells growing in an exponential growth phase were harvested and counted for tumor cell inoculation. Female BALB/c nude mice aged 6-8 weeks and weighing approximately 18-22g were purchased from Vital River. Each mouse was inoculated subcutaneously at the right flank with SK-MEL-239 tumor cells (lx 10 7 ) in 0.1 ml of PBS for tumor development. The treatment was started, when the tumor size reaches approximately 100 mm 3 .

100 mg/kg of Phenformin and 15 mg/kg of Vemurafenib were administered via oral gavage twice daily and 100 mg/kg HLPOl (HL 176001001) was administered once daily for 21 days. Tumor volumes were measured twice a week in two dimensions using a caliper, and the volume was expressed in mm 3 using the formula: V = 0.5xa><b 2 , where a and b were the long and short diameters of the tumor, respectively. The T/C value (in percentage) was an indication of antitumor effect. T and C were the mean volumes of the treated and control groups, respectively.