Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
COMPOSITIONS AND METHODS FOR REDUCING AMYLOID BETA FORMATION ON AND COMPOSITION THEREFORE
Document Type and Number:
WIPO Patent Application WO/2020/201915
Kind Code:
A2
Abstract:
The present invention provides methods for reducing amyloid beta formation and for treating diseases associated with the accumulation of amyloid beta., The present invention provides (1) A β aggregation inhibition by A β Oligomer / Fibril formation inhibition, (2) BACE-1 reduced a through β- Amyloidogenic Processing inhibition, (3) cerebral blood flow to the increase of the cell outer Αβ Monomer, Oligomer &Αβ Fibril / Plaque reduction, (4) NO / cGMP / PKG / CREB Pathway to the activation of Neuronal cell Death inhibition and Neurogenesis, Synaptogenesis, Angiogenesis promotion, (5) DKK-1 inhibition by Wnt Signaling in the activation of synaptic plasticity recovery and Αβ production Positive Feedback Loop for inhibition of APP generates reduced and Αβ accumulation suppression, (6) Autophagy activation by cells within Toxic Mirodenafil, Sildenafil, Vardenafil, Tadalafil, Udenafil, Dasantafil, and Avanafil for the treatment of inhibition of Αβ Fibril / Plaque formation through removal of Soluble Αβ Oligomer; and a Pharmaceutically Acceptable Salt, Solvate, and Hydrate in selected compounds key of ingredient containing drug compound composition, and this with the treatment method provided.

Inventors:
CHOUNG JAI JUN (KR)
CHOI YOON PYO (KR)
KANG BYUNGWOO (KR)
KIM FRED (US)
CHOUNG MI SUN (CH)
Application Number:
PCT/IB2020/052771
Publication Date:
October 08, 2020
Filing Date:
March 24, 2020
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
ARIBIO CO LTD (KR)
SK CHEMICALS CO LTD (KR)
International Classes:
A61K31/519
Download PDF:
Claims:
CLAIMS

1. A pharmaceutical composition for treating Alzheimer comprising:

a compound selected from the group consisting of mirodenafil, sildenafil, vardenafil, tadalafil, udenafil, dasantafil, and avanafil; and a pharmaceutically acceptable salt, solvate, and hydrate thereof.

2. The pharmaceutical composition of claim 1, wherein the compound increases the blood flow in the brain by vasodilation to suppress formation and accumulation of extracellular Ab Monomer, Oligomer and/or Ab Fibril/Plaque.

3. The pharmaceutical composition of claim 1, wherein the compound activate NO/cGMP/PKG/CREB Pathwayto reduce neuronal cell death and promote neurogenesis, synaptogenesis, angiogenesis.

4. The pharmaceutical composition of claim 1, wherein the compound suppressed DKK-1 (Dickkopf WNT Signaling Pathway Inhibitor 1) to activate Wnt Signaling to restore Synaptic Plasticity and suppress Positive Feedback Loop of Ab production to reduce formation of APP (Amyloid Precusor Protien) and accumulation of Ab.

5. The pharmaceutical composition of claim 1, wherein the compound activates Autophagy to remove intracellular Toxic Soluble Ab Oligomer to suppress formation and accumulation of Ab Fibril/Plaque.

6. The pharmaceutical composition of claim 1, wherein compound improves behavioral and cognitive function.

7. A method for treating Alzheimer comprising:

administering a pharmaceutical composition comprising a compound selected from the group consisting of mirodenafil, sildenafil, vardenafil, tadalafil, udenafil, dasantafil, and avanafil; and a pharmaceutically acceptable salt, solvate, and hydrate thereof.

8. The method of claim 7, wherein the compound inhibits formation of Ab aggregation inhibition by reducing formation of Ab Oligomer or Fibril.

9. The method of claim 7, wherein the compound inhibits b-Amyloidogenic Processing through BACE-1 reduction.

10. The method of claim 7, wherein the compound reduces extracellular formation and accumulation of amyloid beta monomer, oligomer, and / or amyloid beta fibril and plaque by vasolidation.

11. The method of claim 7, wherein the compound reduces neuronal cell death and enhances neurogenesis, synaptogenesis, or angiogenesis by activating NO / cGMP / PKG / CREB Pathway.

12. The method of claim 7, wherein the compound restored Synaptic plasticity by Wnt Signaling activation through DKK-1 inhibition.

13. The method of claim 7, wherein the compound suppresses DKK-1 by inhibiting Positive Feedback Loop of Ab production to inhibit formation of APP and Ab accumulation.

14. The method of claim 7, wherein the compound activates Autophagy to remove intracellular Toxic Soluble Ab Oligomer to suppress formation and accumulation of Ab Fibril/Plaque.

15. The method of claim 7, wherein compound improves behavioral and cognitive function.

14.

Toxic Soluble Ab Oligomer Ab Fibril/Plaque Alzheimer's

disease .

15.

Alzheimer's disease

Description:
COMPOSITIONS AND METHODS FOR REDUCING AMYLOID BETA

FORMATION AND COMPOSITION THEREFORE

CROSS-REFERENCE TO RELATED APPLICATION

This application claims the benefit of priority from U.S. Provisional Application Serial No. 62/822,975, filed March 24, 2019, the contents of which are incorporated herein by reference.

FIELD OF THE INVENTION

The present invention is related to methods for reducing amyloid beta formation and for treating diseases associated with the accumulation of amyloid beta.

BACKGROUND OF THE INVENTION

Dementia refers to a clinical disease that suffers from multiple cognitive impairment (or MCD, Multiple Cognitive Deficits) as an acquired brain disease that shows multifaceted pathogenesis caused by a variety of genetic and environmental risk factors. A representative of diseases causing dementia is Alzheimer's disease, mainly endemic in senior people and contributed to 60-70% of the all dementia (Ann Neurol. 1993 May; 33(5) : 494-501.). Recent rapid aging of population, more progressive dementia in older people by Alzheimer's has increased and so does the need for treatment of dementia. However, the difficulties of research and development in additional low rate of success down the process of development of new therapeutics and treatment method for dementia. Based on 2014 GBI Research data, the success rate for final approval of a therapeutics for Alzheimer's disease is less than 1%, as 72% in clinical phase II, 92% of clinical phase III, and 99.6% of NDA application failed and about 80% of the currently on-going pipelines are only for "exploratory and/or pre-clinical stage."

The current conventional list of FDA approved drugs include AChE (Acetylcholinesterase) Inhibitor and NMDA (N-Methyl-D- Aspartate) Receptor Antagonist, used also in combination with antioxidant, NSAID (non-steroidal anti inflammatory drugs), anti-inflammatory agents, statin formulations, or Hormone formulations, etc. However, these drugs are used only to relieve symptoms and delay or improvement of cognitive disorders, but there is no fundamental treatment for dementia at this moment.

Representative AChE Inhibitors includes Donepezil (Aricept™ ), Galantamine (Reminyl™ ), Rivastigmine (ENA-713, Exelon™ ), etc., and these drugs temporarily increase the concentration of neurotransmitters Acetylcholine to provide symptomatic treatment. In addition, these drugs are prescribed for patients with mild to moderate Alzheimer's disease, vascular dementia, dementia by Parkinson's disease, and stroke or cerebral subcortical ischemic vascular disease (J Korean Med Assoc 2009; 52(4) : 417-425.).

Typical NMDA Receptor Antagonist includes Memantine (Ebixa™), which causes excitotoxicity to inhibit glutamate transportation system which blocks synaptic plasticity to effectively reduce neuronal degeneration. Memantine was confirmed to be effective in the moderate or more dementia and Lewy body dementia with relatively low side effect, but showed only limited activity in more earlier state of disease (Arch Neurol 2011 Aug; 68 (8) : 991-8). Thus, it was used more with ChE inhibitors and in mild and severe dementia than monotherapy. However, so far no clear evidence was found that the combination of AChE inhibitors and Menantine is more effective than AChE alone and additional study is required (Brain Neurorehabil 2015 Mar; 8(1) : 19-23.).

However, even though these drugs have been used for treatment of dementia and Alzheirmer's disease for a long time, there is no clear standard for the usage and furthermore, even at the maximum recommended human dose (MRHD), either there was only no improvement or continued progress of the disease to lead the patients to the terminal stage in the treated patients, there has been a continuous concern with the usage of these drugs. However, until now in the absence of any therapeutics with confirmed efficacy, there is no alternatives but to continue the effort to develop new treatment for dementia (Korean J Biol Psychiatry 2016 May; 23(2) : 48-56.).

The main targets of candidates for dementia treatments that have been developed so far include: (1) inhibitors of BACE-1 (b-Secretase 1) or y-Secretase Inhibitor to inhibit production of Ab (Amyloid b), (2) Anti-Ab Monoclonal Antibody to remove Ab (Amyloid b), (3) Tau Aggregation Inhibitor and (One Amino Acid Kinase) TAOK Inhibitor to inhibit Tau Aggregation and Phosphorylation, (4) AchE Inhibitor and NMDA Receptor Antagonist to block AChE and NMDA Receptor, etc. Recently, pipelines targeting Ab Plaque and Tau proteins have increased, among which about 70-80% of the candidates target suppression of Ab production or removal Ab. In addition, about 30% of the pipelines is biopharmaceuticals with dominantly high ratio of monoclonal antibody or peptide. However, due to the recent failures of clinical trials of candidates with high expectation, including Anti-Ab Monoclonal Antibody targeting Ab for targeted such as Aducanumab (Biogen), Solanezumab (Eli Lilly), and Gantenerumab (Roche), the assumption of Ab as the target for treatment of dementia became uncertain. Nevertheless, it is accepted in general that the key target for development of therapeutics for dementia is suppression of Ab formation and removal of the same.

One of the issues of Ab targeted drug development is that it is very difficult for the monoclonal antibodies having large molecular weight to penetrate the cell membrane and thus, it can be effective only to remove extracellular amyloid plaque, and has only limited efficacy for the removal of intracellular Ab oligomers. Furthermore, it is reported that intracellular Ab Oligomers cause more cytotoxicity than the extracellular Ab Oligomers and plays more important roles for neuronal cell death (J Biol Chem. 2002 May 3; 277 (18) : 15666-70.). Therefore, development of small molecule drug which can penetrate the cell membrane than the macromolecules such as monoclonal antibody for removal of intracellular Ab Oligomers.

One of the recent opinions drawing attention in the field is that the direction reason fo the failure of the development of therapeutics for dementia could be the conventional approach, "One Durg, One Target" paradigm as the clinical disorder dementia is a disease of multifaceted pathogenesis. It is being accepted that this traditional approach cannot achieve a successful development of therapeutics for dementia. Currently, more and more interest for development of new therapeutics based on "One Drug, Multiple Targets/Mechanisms" paradigm to overcome the above-mentioned problems. Approach such as one compound with multigarget/multimechanism activity becomes a new concept of new drug discovery and development (J Neural Transm (Vienna). 2013 Jun; 120(6) : 893-902., Future Med Chem. 2016 Apr; 8 (6) : 697-711.). Therefore, to overcome the difficulties in the development of therapeutics for dementia and to increase the success rate of development, development of small molecule/polypharmacology drug based on "One Drug, Multiple Target/Mechanisms" paradigm, especially considering multifaceted pathogenensis of dementia, than the traditional "One Drug, One Target" paradigm is needed from the beginning of the discovery of the candidates and at pre-clinical stage (Expert Rev Clin Pharmacol 2013 Jan; 6 (1) : . 10.1586 /ecp.12.74., Curr Pharm Des. 2016; 22 (21) : 3171-81.).

SUMMARY OF THE INVENTION

One embodiment of the present invention provides a method to inhibit formation of Ab Fibril/Plaque by removal of intracellular, toxic soluble Ab Oligomer by administering one of the compounds selected from Mirodenafil, Sildenafil, Vardenafil, Tadalafil, Udenafil, Dasantafil, and Avanafil; and a pharmaceutically acceptable salt, solvate, or hydrate thereof.

The method of the present invention by one embodiment provides: (1) inhibition of formation of Ab Oligomer / Fibril by suppression of Ab aggregation, (2) inhibition of b-Amyloidogenic Processing by reduction of BACE-1, (3) reduction of formation of extracellular Ab monomers, oligomers, Ab Fibril/plaque by cerebral blood flow increase (vasodilation), (4) inhibition of neuronal cell death and acceleration of neurogenesis, synaptonegenesis, and/or promoton of angiogenesis by activation of NO (Nitric Oxide) / cGMP (Cyclic Guanosine Monophosphate) / PKG (Protein Kinase G), CREB (Cyclic AMP (Adenosine Monophosphate) Response Element Binding Protein) Pathway, (5) restoration of synaptic plasticity (synaptic Plasticity) by activation of Wint Signaling by inhibition of DKK-1 (Dickkopf WNT Signaling Pathway Inhibitor 1), and inhibition of production of APP (Amyloid Precusor Protien) and reduction of Ab accumulation by suppression of positive feedback loop for Ab production, and (6) inhibition of formation of Ab Fibril/plaque by removal of intracellular toxic and soluble Ab oligomers by activation of autophage.

In another embodiment, the present invention provides a pharmaceutical composition comprising one compound selected from Mirodenafil, Sildenafil, Vardenafil, Tadalafil, Udenafil, Dasantafil, and Avanafil; and a pharmaceutically acceptable salt, solvate, and hydrate thereof as an actgive ingredient, for (1) inhibition of Ab Oligomer / Fibril formation by reduction of Ab aggregation, (2) inhibition of b-Amyloidogenic Processing decreased BACE-1, (3) reduction of extracellular Ab monomers, oligomers & Ab Fibril/Plaque by increase of the cerebral blood flow, (4) suppression of neuronal cell death Inhibition and promotion of neurogenesis, synaptogenesis and/or angiogenesis by activation of NO/cGMP/PKG/CREB Pathway, (5) restoration of synaptic plasticity (synaptic Plasticity) by activation of Wint Signaling by inhibition of DKK-1, and inhibition of production of APP and reduction of Ab accumulation by suppression of positive feedback loop for Ab production, and (6) inhibition of formation of Ab Fibril/plaque by removal of intracellular toxic and soluble Ab oligomers by activation of autophage.

BRIEF DESCRIPTION OF THE DRAWINGS

I. inhibition of Ab Oligomer/Fibril formation by reduction of Ab aggregation

Figure 1 presents the result of Thioflavin T Assay showing that there is a dose- dependent inhibition of Ab Fibril Aggregation by the composition of the present invention.

Figure 2 present the result of PICUP (Photo-Induced Cross-Linking of Unmodified Proteins) / SDS-PAGE (Sodium Dodecyl Sulfate-Polyacrylamide Gel Electrophoresis) Analysis showing that the formation of Ab oligomer aggregation has been reduced by the treatment with the composition of the present invention.

II. Suppression of b-Amyloidogenic Processing through BACE-1 reduction

Figure 3 present the result of qRT-PCR (Quantitative Real-Time Polymerase

Chain Reaction) showing that BACE-1 mRNA level was increased in the Retinoic Acid Differentiated SH-SY5Y cell with the treatment of Ab1-42 oligomers but indicating that the increased BACE-1 mRNA level is concentration- dependently decreased by the treatment with the composition of the present invention.

III. Reduction of extracellular Ab monomers, Oligomers & Ab Fibril / Plaque by the increase of brain blood flow

Figure 4 presents the result showing that intracellular calcium level, which was increased by the treatment of pericyte cells with hydrogen peroxide (H 2 O 2 ), has been decreased by the treatment with the composition of the presnt invention. IV. Inhibition of Neuronal cell death by activation of NO / cGMP / PKG / CREB Pathway

Figure 5 presents Cyclic GMP Complete ELISA results showing that the amount of cGMP in retinoic acid differentiated SH-SY5Y Cell, which was decreased by the treatment of Ab1-42 Oligomer, increased in a concentraction-dependent matter by the treatment with the composition of the present invention.

Figure 6 presents result of Western Blot Analysis showing that reduction of the ratio of apoptosis-related proteins, Capase-3 and PARP [Poly (Adenosine Diphosphate- Ribose) Polymerase)] to Cleaved Capase-3 / Caspase-3 and Cleaved PARP and Cleaved PARP / PARP, which was increased in retinoic acid differentiated SH-SY5Y Cell, has been decreased by the treatment with the composition of the present invention.

Figure 7 presents the result of JC-1 Mitochondrial Membrane Potential Assay showing that the concentration of Mitochondrial Membrane Potential in retinoic acid differentiated SH-SY5Y Cell, which was decreased by the treatment of Ab1-42 Oligomer, has been increased concentration-dependently by the treatment with the composition of the present invention.

Figure 8 presents the result of IncuCyte ® S3 Live-Cell Analysis showing that number of dead cells dyed by Cytotox Red Reagen in retinoic acid differentiated SH- SY5Y Cell, which was increased by the treatment of Ab1-42 Oligomer, has been reduced by the treatment with the composition of the present invention.

V. Promotion of Neurogenesis, Synaptogenesis, and Angiogenesis by activating NO / cGMP / PKG / CREB Pathway

Figure 9 presents result of Western Blot Analysis showing that Serl33 phosphorylation of CREB protein in retinoic acid differentiated SH-SY5Y Cell, which was decreased by the treatment of Ab1-42 Oligomer, increased concentration- dependentaly by the treatment with the composition of the present invention.

Figure 10 presents result of Western Blot Analysis showing that Protein Level of NGF (Nerve Growth Factor) and BDNF (Brain-Derived Neurotrophic Factor) in retinoic acid differentiated SH-SY5Y Cell, which was decreased by the treatment of Ab1-42 Oligomer, increased concentration-dependentaly by the treatment with the composition of the present invention.

Figure 11 presents result of NGF Immunocytochemistry showing that Protein Level of NGF (Nerve Growth Factor) in retinoic acid differentiated SH-SY5Y Cell, which was decreased by the treatment of Ab1-42 Oligomer, increased by the treatment with the composition of the present invention.

VI. Recovery of synaptic plasticity by activation of Wnt Signaling through DKK- 1 inhibition

Figure 12 presents result of qRT-PCR showing that level of DKK-1 mRNA in retinoic acid differentiated SH-SY5Y Cell and Differentiated HT-22 Mouse

Hippocampal Neuronal Cell, which was increased by the treatment of Ab1-42 Oligomer, decreased by the treatment with the composition of the present invention.

Figure 13 presents result of qRT-PCR showing that level of DKK-1 protein in retinoic acid differentiated SH-SY5Y Cell and Differentiated HT-22 Mouse

Hippocampal Neuronal Cell, which was increased by the treatment of Ab1-42 Oligomer, decreased by the treatment with the composition of the present invention.

Figure 14 presents result of qRT-PCR showing that level of Wnt3a mRNA in retinoic acid differentiated SH-SY5Y Cell and Differentiated HT-22 Mouse

Hippocampal Neuronal Cell, which was first treated with Ab1-42 Oligomer, increased by the treatment with the composition of the present invention compared to the control group.

Figure 15 presents result of Western Blot Analysis showing that reduced level fo Wntl protein in the hippocampus of Alzheimer's disease animal model (5XFAD Transgenic Mice) has been increased by the treatment with the composition of the present invention.

Figure 16 presents result of Wn -Catenin TF Array showing that activity of Wn b-Catenin related factors, VAX2, c- Myc, NR5A2, Mitf, TCF / LEF, NFAT, CEBP, GLI-1, GBX2, and AP-1 in retinoic acid differentiated SH-SY5Y Cell and Differentiated HT-22 Mouse Hippocampal Neuronal Cell, which was first treated with Ab1-42 Oligomer, has been increased more than twice by the treatment with the composition of the present invention compared to the control group. Figure 17 presents result of TOPFLASH Reporter Gene Assay (Wn b-Catenin Signaling) showing that Wnt/b-Catenin Activity in retinoic acid differentiated SH-SY5Y Cell and Differentiated HT-22 Mouse Hippocampal Neuronal Cell, which was reduced by the treatment of Ab1-42 Oligomer, increased concentration-dependently by the treatment with the composition of the present invention.

Figure 18 presents result of Western Blot Analysis showing that level of Ser9 Phosphorylation of GSK3b Protein in retinoic acid differentiated SH-SY5Y Cell, which was reduced by the treatment of Ab1-42 Oligomer, has been increased by the treatment with the composition of the present invention.

Figure 19 presents result of Phospho-GSK-3b (Ser9) Sandwich ELISA showing that level of Ser9 Phosphorylation of GSK3b Protein in retinoic acid differentiated SH- SY5Y Cell, which was reduced by the treatment of Ab1-42 Oligomer, has been increased by the treatment with the composition of the present invention.

Figure 20 presents result of Western Blot Analysis showing that level of Ser9 Phosphorylation of Tau Protein increased in the hippocampus of Alzheimer's disease animal model (NSE-hAPP-C105) has been decreased by the treatment with the composition of the present invention.

VII. Reduction of APP formation and Ab accumulation by the suppression of Ab production Positive Feedback Loop through suppression of DKK-1

Figure 21 presents result of Western Blot Analysis showing that level of APPand Ab1-42 Protein in retinoic acid differentiated SH-SY5Y Cell, which was increased by the treatment of Ab1-42 Oligomer, has been decreased by the treatment with the composition of the present invention.

Figure 22 presents result of Western Blot Analysis showing that level of APPand Ab1-42 Protein in retinoic acid differentiated SH-SY5Y Cell, which was increased by the treatment of H 2 O 2 , has been decreased by the treatment with the composition of the present invention.

VIII. Inhibition of Ab Fibril / Plaque formation by removal of intracellular toxic and soluble Ab Oligomer through activation of Autophagy Figure 23 presents result of Western Blot Analysis showing that level of Ab Protein increased in the hippocampus of Alzheimer's disease animal model (NSE- hAPP-C105) has been decreased by the treatment with the composition of the present invention.

Figure 24 presents result of Thioflavin S Staining/GFAP (Glial Fibrillary Acidic

Protein) Immunohistochemistry showing that lenumber of Ab Plaque in the hippocampus of Alzheimer's disease animal model (NSE-hAPP-C105) has been decreased by the treatment with the composition of the present invention. Number of Ab Plaque was counted by counting the number of Ab Plaque dyed by Thioflvain S (Green) and number of colocalized spot (yellow) of Astrocyte (Red) dyed by GFAP.

Figure 25 presents result of Western Blot Analysis showing that level of Thrl72 Phosphorylation of AMPK Catalytic Subunit a in retinoic acid differentiated SH-SY5Y Human Neuroblastoma Cell, which was decreased by the treatment of Ab1-42 Oligomer, has been increased concentration-dependently by the treatment with the composition of the present invention.

Figure 26 presents the result of Western Blot Analysis showing that level of LC3B protein, an autophagy marker in retinoic acid differentiated SH-SY5Y Human Neuroblastoma Cell, which was decreased by the treatment of Ab1-42 Oligomer, has been increased, at the same time, induced autophagic flux (conversion from LC3B-1 to LC3B-II) to increase LC3B-II/I Ratio, and also increased expression of Ubiquitin- Binding Protein p62 (SQSTM, Sequestosome 1), an autophagosome Cargo Protein, which was reduced by the treatment of Ab1-42 Oligomer. Further more, 3-MA (3- Methyladenine), an autophagyt/PI3K inhibitor was used in combination of the composition of the present invention at the same time, level of autophagic flux (LC3B- II/I ratio) increased by the composition of the present invention was decreased and expression of p62 which was decreased by the composition of the present invention has increased. Furthermore, level of APP and Ab1-42 Protein decreased by the composition of the present invention increased when 3-MA was used with the composition of the present invention and the results by Western Blot Analysis are shown in Figure 26.

Figure 27 presents result of Western Blot Analysis showing that level of ATG7 and ATG5/12 Protein, i.e. Autophagy Markers, increased in the hippocampus of Alzheimer's disease animal model (5XFAD Transgenic Mice) and ATG5/12 Protein Level in cortex increased by the treatment with the composition of the present invention. IX. Effect of improving cognitive ability and behavioral learning ability

Figure 28 presents result of Morris Water Maze Test showing that, in the evaluation of improvement of cognitive function level in Alzheimer's dementia animal model (NSE-hAPP-C105), group treated with the composition of the present invention achieved reduced latency to target and distance to target and increased time in target quadrant and number of cross platform compared to transgenic mouse control.

Figure 29 presents result of Passive Avoidance Test showing that, in the evaluation of improvement of cognitive ability and behavioral learning ability in Alzheimer's dementia animal model (NSE-hAPP-C105), group treated with the composition of the present invention achieved reduced latency time compared to transgenic mouse control.

DETAILED DESCRIPTION OF THE INVENTIONF

One embodiment of the present invention provides a method for (1) inhibition of formation of Ab Oligomer / Fibril by suppression of Ab aggregation, (2) inhibition of b-Amyloidogenic Processing by reduction of BACE-1, (3) reduction of formation of extracellular Ab monomers, oligomers, Ab Fibril/plaque by cerebral blood flow increase (vasodilation), (4) inhibition of neuronal cell death and acceleration of neurogenesis, synaptonegenesis, and/or promoton of angiogenesis by activation of NO (Nitric Oxide) / cGMP (Cyclic Guanosine Monophosphate) / PKG (Protein Kinase G), CREB (Cyclic AMP (Adenosine Monophosphate) Response Element Binding Protein)

Pathway, (5) restoration of synaptic plasticity (synaptic Plasticity) by activation of Wint Signaling by inhibition of DKK-1 (Dickkopf WNT Signaling Pathway Inhibitor 1), and inhibition of production of APP (Amyloid Precusor Protien) and reduction of Ab accumulation by suppression of positive feedback loop for Ab production, and (6) inhibition of formation of Ab Fibril/plaque by removal of intracellular toxic and soluble Ab oligomers by activation of autophage, by administering one of the compounds selected from Mirodenafil, Sildenafil, Vardenafil, Tadalafil, Udenafil, Dasantafil, and Avanafil; and a pharmaceutically acceptable salt, solvate, or hydrate thereof.

In another embodiment, the present invention provides a pharmaceutical composition comprising one compound selected from Mirodenafil, Sildenafil, Vardenafil, Tadalafil, Udenafil, Dasantafil, and Avanafil; and a pharmaceutically acceptable salt, solvate, and hydrate thereof as an actgive ingredient, for (1) inhibition of Ab Oligomer / Fibril formation by reduction of Ab aggregation, (2) inhibition of b-Amyloidogenic Processing decreased BACE-1, (3) reduction of extracellular Ab monomers, oligomers & Ab Fibril/Plaque by increase of the cerebral blood flow, (4) suppression of neuronal cell death Inhibition and promotion of neurogenesis, synaptogenesis and/or angiogenesis by activation of NO/cGMP/PKG/CREB Pathway, (5) restoration of synaptic plasticity (synaptic Plasticity) by activation of Wint Signaling by inhibition of DKK-1, and inhibition of production of APP and reduction of Ab accumulation by suppression of positive feedback loop for Ab production, and (6) inhibition of formation of Ab Fibril/plaque by removal of intracellular toxic and soluble Ab oligomers by activation of autophage.

The present invention has multi-mechanism distinguished from the previous or presently developed other Alzheimer therapeutics. The present invention provides significantly improved efficacy for treatment of Alzheimer's disease compared fo the current drug in the market by adopting "one Drug, Multiple Targets/Mechanisms" strategy which excels the conventional discovery strategy based on "one Drug, One Target" paradigm to provide (1) inhibition of Ab Oligomer / Fibril formation by reduction of Ab aggregation, (2) inhibition of b-Amyloidogenic Processing decreased BACE-1, (3) reduction of extracellular Ab monomers, oligomers & Ab Fibril/Plaque by increase of the cerebral blood flow, (4) suppression of neuronal cell death Inhibition and promotion of neurogenesis, synaptogenesis and/or angiogenesis by activation of NO/cGMP/PKG/CREB Pathway, (5) restoration of synaptic plasticity (synaptic Plasticity) by activation of Wint Signaling by inhibition of DKK-1, and inhibition of production of APP and reduction of Ab accumulation by suppression of positive feedback loop for Ab production, and (6) inhibition of formation of Ab Fibril/plaque by removal of intracellular toxic and soluble Ab oligomers by activation of autophage. Mechanism of treatment for Alzheimer's disease

1. Inhibition of Ab Oligomer / Fibril formation by Inhibition of Ab aggregation

Common pathological feature in sporadic and familial Alzheimer's dementia is peptide known as Ab accumulate to form extracellular senile plaque (J Alzheimers Dis 2018; 64 (si) : S567-S610.). Even though Ab also produced in normal people, it degrade quickly enough not to accumulate in the body. On the other hand, in the case of Alzheimer's disease patient, Ab is not only formed massively, but also does not degrade and accumulate in the tissues (Alzheimers Res Ther 2013 Nov 29; 5(6) : 60.). These abnormal Ab accumulation induce formation of extracellular senile plaque to disturb the signal transmission between neurons and also accumulation in Hippocampus, which plays an important role in the memory and learning ability and cortex to cause cellular inflammation and neuronal damage, and eventually damage neural networks required for normal function (EBioMedicine 2016 Apr; 6: 42-49.). In addition, the accumulated Ab Monomer is aggregate to form toxic Ab Oligomer to increase production of ROS (Reactive Oxygen Species) and RNS (Reactive Nitrogen Species) to activate the signal network related to neuronal cell death to increase apoptosis (Mt Sinai J Med. 2010 Jan-Feb; 77 (1) : 43-9.).

The inventive compositions suppressed Ab Fibril Aggregation (Figure 1) to inhibit Ab Oligomer / Fibril formation was inhibited (Figure 2).

Therefore, the composition of the present invention inhibits Ab oligomer Aggregation Oligomer to inhibit Ab Oligomer / Fibril formation.

2. Suppression of b-Amyloidogenic Processing through BACE-1 reduction

Ab is produced from APP as various Secretase cut APP to generate Ab with various numbers of amino acids. Among these, in the case of Alzheimer's patients, the ratio of Ab with 42 or 43 amino acid formed by BACE-1 rapidly increase. Currently, nerve cell damage by Ab1-42 or Ab1-43 is know to be one of the important causes in the progress of Alzherimer's disease and Ab25-35 is known to be the toxic fragment of Ab1-42 or Ab1-43 cauing neuronal cell damage (J Amino Acids 2011; 2011 : 198430., PLoS One 2013; 8 (1) : e53117, J Alzheimers Dis 2018; 62 (3) : 1345-1367). In particular, it was reported recently that SILDENAFIL, a PDE-5 Inhibitor, suppressed BACE-1 and Cathepsin B expression to inhibit b-Amyloidogenic Processing of APP to reduce the production of Ab1-42 or Ab1-43, and it was expected that PDE-5 inhibitor can suppress expression of BACE-1 to reduce formation of Ab1-42 or Ab1-43, which cause neuronal cell damage, to have therapeutic effect on Alzheimer's diseas (J Gerontol a Biol Sci Med Sci 2015 Jun; 70 (6) : 675-85., J Urol. 2016 Apr; 195 (4 Pt 1) : 1171.).

The compositions of the present invention suppressed the expression of BACE- 1 which plays the key role in b -Amyloidogenic Processing (Figure 3).

Therefore, the composition of the present invention inhibit b-Amyloidogenic Processing by APP to reduce the formation of Ab1-42 or Ab1-43 which causes damages on neuronal cells.

3. Reduction of Ab Monomer, Oligomer & Ab Fibril / Plaque cerebral blood flow to increase by cells other

One of the first step in the onset of dementia is damage of cells composing blood vessels in the brain including entodhelial cells, pericytes, and vascular smooth muscle cells by various genetic and environmental factors, followed by destruction on brain-blood barrier to allow the neuro toxic proteins sucha as Ab proteins or cells invade the brain tissues to cause inflation in brain tissue or varous degenerative changes. in the brain to the organization to infiltrate the brain tissue inflammation and various degenerative changes that cause to be known there (Nat Rev Neurol 2018 Mar; 14 (3.) : 133-150.) .

In addition, it has been reported that as the blood-brain barrier collapses with progression of decrease of cognitive function caused by Alzheimer's dementia, decrease of the cerebral blood flow in the brain region responsible for memory and the region continues to expand (Alzheimers Dement 2017 May; 13 (5) : 531-540.) . Furthermore, recent studies showed that, in the Alzheimer's dementia patient's brain tissue, increased ROS by Ab increases the concentration of calcium in pericyte to cause perivascular cells to shrink and thus, to reduce the amount of cerebral blood flow (Science. 2019 Jul 19; 365 (6450)). Therefore, proper brain blood flow should be maintained not only to provide a lot of amount of oxyten and nutrition to brain, but also to effectively remove neurotoxins, including carbon dioxide and Ab brain, or protein waste which is generated naturally through metabolism (Nat Rev Neurosci 2017 Jul; 18 (7) : 419-434).

PDE5 Inhibitors are used for treaetment of pulmonary hypertension or erectile dysfunction as it can inhibit activation of PDE5 to prevent conversion of cGMP to 5'-GMP and eventually increase the concentration of cGMP in the pericytes to induce dialation of blood vessel (Int 3 Impot Res. 2004 Jun; 16 Suppl 1 : S4-7.) . In particular, in a recent study, PDE5 Inhibitors showed Alzheimer patient's (J Cereb Blood Flow Metab 2018 Feb; 38 (2) : 189-203.).

The composition of the present invention reduced intracellular calcium concentration in pericyte which was increased by H 2 O 2 treatment to show that the composition can inhibit reduction of cerebral blood flow by inhibiting the contraction of the pericyte caused by the increased concentration of calcium (Figure 4) .

Therefore, the composition of the present inventive compositions of perivascular cells to shrink by suppressing the brain blood flow by increasing the brain tissue A b and the same neural toxicity of the protein to accumulate to inhibit the effects seem to be expected there .

4. Inhibition of Neuronal Cell Death through activation of NO / cGMP / PKG / CREB Pathway and promotion of Neurogenesis, Synaptogenesis, Angiogenesis

Ab Monomer transfected into the cells by endocytosis aggregate to form toxic soluble Ab oligomer. The aggregates can not only induce disorder of cellular functions related to neurocell death, but also increase hydrophosphorylation of Tau protein, a microtubule-associated protein, to promote the formation of NFT (Neurofibrillary Tangle). As a result, ROS increased in the process cause mitochondrial damage and activate Caspase-3 to result in death of neuron cells. (Nat Rev Neurosci. 2007 Jul;8(7) :499-509., Neuron. 2008 Nov 26;60(4) :534-42., Nat Rev Neurosci. 2011 Feb;12(2) :65-72.).

PDE5 (Phosphodiesterase 5) is exressed in the cerebral cortex and hippocampus which, among brain tissues, play an important role in cognitive functionof (J Comp Neurol 2003 Dec 22; 467 (4) : 566-80.), and in particular, it has been reported that PDE5 expression is increased in the cerebral cortex among brain tissues of patients with Alzheimer's disease compared to normal people (Neuropathol Appl Neurobiol. 2015 Jun; 41 (4) : 471-82.). Moreover, in the hippocampus of mouse, synaptic plasticity, which is important for the brain cognitive function reduced by Ab, has been reported to be restored by the action of NO / cGMP / CREB Pathway (J Neurosci 2005 Jul 20; 25 (29) : 6887-97.). It is reported that PDE5 Inhibitor can also inhibit neuronal apoptosis and it is because PED5 inhibitor can inhibit PED5 activity to prevent conversion of cGMP to 5'- GMP. As a result, cGMP accumulates in the brain to activate PKG (Mol Neurobiol. 2010 Jun; 41 (2-3) : 129-37., ACS Chem Neurosci. 2012 Nov 21; 3 (11) : 832-44., Exp Neurol. 2014 Nov; 261 : 267-77.). The activated PKG suppresses Caspase-3 which is activated by toxic soluble Ab oligomer to suppress neuronal apoptosis. (Neurobiol Aging 2014 Mar; 35 (3) : 520-31, Neuroscience 2016 Jul 22; 328: 69-79, Front Pharmacol. 2017 Mar 8; 8: 106.). Also, phosphorylation of Serl33 of CREB protein, which is gene transcription factor, activates PKG that will increase expression of factors related to neurogenesis, synaptogenesis, angiogenesis (Behav Brain Res 2013 Aug 1; 250: 230-7, DNA Cell Biol. 2018 Nov; 37 (11) : 861-865.).

The compositions of the present invention inhibit activation of PDE5 to prevent conversion of cGMP to 5'-GMP to increase the amount of cGMP in the cell (Figure 5), suppressed activity of Caspase-3 (Figure 6), restore mitochondrial membrane potential (Figures 7) to reduce Ab-induced neuronal death (Figure 8).

The compositiosn of the presention invention increased expression of factors related to neurogenesis, synaptogenesis, and angiogenesis by increasing phosphorylation of Serl33 in CREB protein (Figure 9).

The composition of the present invention increased the expression of NGF and BDNF mainly involved in the regulation of growth, maintenance, proliferation and survival of neuron (Figures 10 and 11).

Therefore, the present invention provides effect to stimulate neuronal cell apoptosis, neurogenesis, synaptogenesis, and angiogenesis in the brain of dementia patients by the activiation of NO/cGMP/PKG/CREB pathway.

5. restoration of synaptic plasticity by activation of Wnt signaling via suppression of DKK-1 and reduced production of APP and inhibition of Ab accumulation by suppression of positive feedback loop

Wnt / b-Catenin Pathway is an essential signal transmitting pathway controlling many cellular processes including cell survival. Especially, Wnt / b- Catenin Pathway in brain is not only important for the neuron survivial and neuronal signal generation, but also plays an essential role in controlling synaptic plasticity, the blood-brain barrier integrity and controlling functions of the same (Biomed Res Int 2014; 2014: 301575.)· At the present time, it is known that expression of Wnt Antagonist DKK-1 is increased in the brain tissues of Alzheimer patients to suppress Wn b-Catenin Pathway and increase synaptic plasticity (J Neurosci 2004 Jun 30; 24 (26) : 6021-7., Front Cell Neurosci 2013 Nov 5; 7 : 162). It is reported that expression of DKK-1 was increased by the accumulated Ab in Hippocampal neuron to decrease synaptic plasticity (J Neurosci 2012 Mar 7;32 (10) : 3492-8.). Moreover, DKK-1 is reported to play Positive Feedback Loop for Ab production and this gives an expectation that instead of targeting Ab as conventionally, it may be possible to slow down the progress of Alzheimer disease by inhibiting DKK-1 which is a essential controller of positive feedback loop for Ab production to reduce Ab production to reduce loss of synapse (J Mol Cell Biol. 2014 Feb;6(l) :75-80., Neuron. 2014 Oct l;84(l) :63-77., Cell Death Dis. 2014 Nov 27;5:el544., Curr Biol. 2016 Oct 10;26(19) :2551-2561., Front Neurosci. 2016 Oct 19;10:459., Transl Psychiatry.

2018 Sep 20;8(1) : 179.).

On the other hand, in several degenerative brain diseases including frontotemporal dementia, progressive supranuclear palsy, and Alzheime, NFT formed by the aggregation of Tau protein by hyperphosphorylation is the main cause of the disease and these disorders are called collectively Tauopathy' (Front Mol Neurosci. 2011 Oct 5;4:24., Front Neurosci. 2019 Dec 13;13: 1274.). Current Tauopathy is known to be caused as Wn b-Catenin Pathway is suppressed by various pathogenic memchanism in the brain (Mol Brain. 2019 Dec 4;12(1) : 104.), especially suppression of Wn b-Catenin Pathway by accumulation of Ab in the brain reduces Ser9 phosphorylation in GSK3b protein and increase Tyr 216 phosphorylation to activate GSK3b and to enhance phorphorylation of Tau protein in mouse Hippocampus neuron. As a result, formation of NFT is enhanced to reduce neuron survival, generation of neuron, synaptic plasticity, blood-brain barrier integrity and functionality to enhance the progress of Tauopathy (Neurosci Res. 1998 Aug;31(4) :317-23., Annu Rev Pathol.

2019 Jan 24;14:239-261.).

In summary, increased expression of DKK-1 by the accumulation of Ab in brain act as a Wnt antagonist to suppress Wbt^-Catenin Pathway and to activate GSK3b to increase formation of NFT to reduce synaptic plasticity, and also act as a positive feedback loop for the production of Ab to enhance production and accumulation of Ab to promote progress of degenerative brain disorders. Thus, therapeutics targeting DKK-1 expression inhibition is expected to inhibit degenerative brain diseases including Alzheimer disease by restored synaptic plasticity by the activation of Wnt signaling and by reduced formation and accumulation of Ab by suppression of positive feedback loop for Ab production.

The composition of the present invention reduced the expression of DKK-1, a regulator of the positive feedback loop of Wnt Antagonist and Ab production (Figure 12, 13).

The composition of the present invention increased expression of Wnt3a (Figure 14) and Wntl (Figure 15), activity of Wn b-Catenin -related transcription factors (VAX2, c-Myc, NR5A2 , Mitf, TCF / LEF, NFAT, CEBP, GLI -1, GBX2, AP-1) (Figure 16), and increased the activity of Wnt/b -Catenin (Figure 17).

The composition of the present invention increased Ser9 Phosphorylation of

GSK3 b Protein to inhibit the activity GSK3b activity (Figure 18, 19).

The composition of the present invention reduced Serl99 / 202

Phosphorylation of Tau protein which is increased in of Alzheimer dementia animal model (NSE-hAPP-C105) (Figure 20).

The composition of the present invention inhibits Ab production Positive

Feedback Loop and, as a result, reduced APP and Ab Protein Level (Figure 21, 22).

Therefore, the composition of the present invention is expected to restore synaptic plasticity by the activation of Wnt signaling from DKK-1 inhibition and to reduce formation of APP and accumulation of Ab by suppression of positive feedback loop for Ab production.

6. Activation of autophagy to inhibit formation of Toxic Soluble Ab Oligomer and Ab Plaque

While the brain consumes 20% or more the oxygen from the total oxygen human body consumes, the defense mechanism for removing reactive oxygen species (ROS) generated inevitably from the intracellular is weak and thus, it is weak against oxidative stress. The ROS accumulated intracellularly by the oxidative stress induces oxidation of proteins in the endoplasmic reticulum to inhibit protein folding and to accumulate misfolding protein and eventually endoplasmic reticulum stress (J Neurochem. 2006 Jun;97(6) : 1634-58., Antioxid Redox Signal. 2007

Dec;9(12) : 2277-93.). As a result, intracellular Ab accumulation caused by endoplasmic reticulum stress further increase endoplasmic reticulum stress and endosomal and lysosomal leakage and mitochondrial dysfunction to enhance neuronal cell apoptosis (J Neurosci Res. 2011 Jul;89(7) : 1031-42.) . Furthermore, recent genome-wide association study with patients with dementia by Alzheimer showed mutation of genes involved with Ab relase or removal (SORL1, BIN1, CD2AP, PICALM), which confirms intracellular accumulation of Ab is a main cause for the progress of dementia by Alzheimer (Trends Neurosci. 2017 0ct;40(10) :592-602., Nat Rev Mol Cell Biol. 2018 Dec; 19(12) :755-773.).

Recently, autophagy drew attention as the main cause of neurodegenerative disease. Autophagy is one of the main cellular mechanisms and it has been known that autophagy function to decompose long-lived proteins and organelle (Nat Med. 2013 Aug; 19(8) :983-97.). Dysfunction of autophagy causes intracellular accumulation of misfolded protein aggregates to induce various diseases, and especially closely related to the onset of the fourt main neurodegenerative diseases, i.e. Alzheimer disease, Huntington's disease, Parkinson's disease, and Amyotropic Lateral Sclerosis (Mol Cells. 2015 May;38(5) :381-9., Nat Rev Drug Discov. 2018 Sep; 17(9) :660-688.). Especially, dysfunctional autophagy in the brain of Alzheimer disease patients induces accumulation of two main onset factors for Alzheimer disease, i.e. Ab and Tau protein (J Syst Integr Neurosci. 2017;3(4) : 1-6.). As a result, the accumulated Ab and Tau protein in the brain form senile plaque to cause necropsis or neuronal cells in hippocampus and cortex that are responsible for cognitive capacity and eventually to cause rediced cognitive function in Alzheimer disease patients (J Cell Biol. 2005 Oct 10; 171(l) :87-98., Front Aging Neurosci. 2018 Jan 30;10: 04.).

AMPK (Adenosine Monophosphate-Activated Protein Kinase ) is responsible for controlling several biological functions including insulin sensitivity, cell survival, proliferation and apoptosis, among which, especially, by activating autophagy by inhibiting mTOR (mammalian target of rapamycin) protein to maintain cellular homeostasis (Nat Rev Mol Cell Biol. 2018 Feb;19(2) : 121-135., Mol Cell. 2017 Jun 15;66(6) :789-800.). Currently, Thrl72 phorphorylation by AMPK catalytic subunit a is a main starting point for the signal transmitting route for activation of autophagy (Exp Mol Med. 2016 Apr l;48:e224., Nat Rev Mol Cell Biol. 2018 Feb; 19(2) : 121- 135., Nat Rev Drug Discov. 2019 Jul;18(7):527-551.), and recently, in degenerative brain disease, it was reported that neuronal degeneration was inhibited by removing misfolded protein aggregates, related to degeneratige brain disease, including Ab and Tau that are main onset factors for Alzheimer disease byAutophagy activation from activated AMPK (Front Neurosci. 2018 May 22;12:255., Nat Rev Drug Discov. 2018 Sep;17(9) :660-688. J Alzheimers Dis. 2019; 68(1) : 33-38). Considering that 30-40% of Alzheimer disease patients have mixed pathology with other pathological symptoms, removing misfolded protein aggregates, including Ab and Tar aggregates, accumulated intracellularly by activation of autophagy from activated AMPK would be new target in the future development of therapeutics for degenerative brain disease including Alzheimer disease (Front Neurosci. 2018 May 22;12:255., Nat Rev Drug Discov. 2018 Sep;17(9) :660-688. J Alzheimers Dis. 2019; 68(1) : 33-38.).

The compositions of the present invention reduced Ab protein level in hippocampus of Alzheimer's dementia animal model (NSE-hAPP-C105) (Figure 23).

The compositions of the present invention reduced Ab Plaque number in hippocampus and cordex of Alzheimer's disease animal model (5XFAD Transgenic Mice) (Figure 24).

The compositions of the present invention AMPK Catalytic Subunit a of Thrl72 phosphorylation was increased (Figure 25).

The inventive compositions induced autophagic flux (conversion from LC3B-I to LC3B-II) of autophagy marker LC3B to increase LC3B-II/I ratio and reduced an autophagosome cargo protein, ubiquitin-binding protein p62 (SQSTM, Sequestosome 1) (Figure 26).

The compositions of the present invention enhanced expression of an autophagy marker, ATG7 and ATG5/12, and expression of ATG5/12 in cortex in the hippocampus of Alzheimer's disease animal model (5XFAD Transgenic Mice) (Figure 26). Therefore, the compositions of the present invention increased an Autophagy Marker LC3BII/I Ratio, reduced an autophagosome cargo protein Ubiquitin-Binding Protein p62, and increased expression of ATG7 and ATG5/12 to activate autophagic cascade.

7. Alzheimer's dementia animal model (NSE-hAPP-C105) for cognitive and behavioral learning skills improvement

To test the effect of the compositions of the present invention on gradual memory decreases and behavioral disorder that appears in the neurodegenerative disease Alzheimer's disease caused by formation and accumulation of Ab42, the compositions of the present invention was intraperitoneally injected to 13 month old C57BL/6-Tg(NSE-hAPP-C105)Kor Transgenic Micem, at 4 mg/kg, once per day for 4 weeks before cognitive capacity and behavioral learning skills were tested by a passisve Avoidance Test, Morris Water Maze Tese, to analyze any changes in cognitive capacity and behavioral learning skills.

It was confirmed that cognitive capacity and behavioral learning skills in the group treated with the compositions of the present invention was improved compared to transgenic couse control group in Alzheimer's dementia animal model (NSE-hAPP- C105) (Figure 28, 29) .

EXAMPLES

Example 1. Test for Selectivity of the compositions of the present invention for PDE5 (IC50, Inhibitory Concentration 50)

Example 1-1. Testing method for PDE5 selectivity

In order to test the selective inhibitory activity (IC50, Inhibitory Concentration 50) of the compositions of the present invention against PDE5 (Phosphodiesterase 5), the test was conducted by MDS Pharma Services, an analysis agency CRO, and Scottish Biomedical. The result was measured using PDE SPA analysis kit (Amersham Pharmacia Biotech). MDS Pharma Services analyzed PDE1 (bovine heart) ; PDEs 2, 3, 5 (human platelete); PDE4 (human U937 cell); and PDE6 (bovine retinal rod) and Scottish Biomedical analyzed PDEs 5, 7-11 (recombinant human enzymes). Test samples (each 100 mL) were added to a mixture of PDE Family protein (10 ml), [ 3 H]- cGMP (5 Ci/mL), Bovine Serum Albumin (0.5 mg / mL ) and MgCl 2 (5 mM) in Tris-HCI Buffer (15 mM, pH 7.5). The reaction began by the addition of PDE Family proteins. Each sample was stored in a 30 °C Water Bath for 30 minutes, and then SPA Bead (PerkinElmer) (50 mL) was added to terminate the reaction. The test tube was allowed to stand for 20 minutes, followed by measurement by Liquid Scintillation Counter (Tri-carb 1500, Packard). To determine degree of inhibition of PDE Family of protein activity, the composition of the present invention and testing samples are dissolved in DMSO (Dimethyl Sulfoxide) and the solution was then diluted with distilled water to make the final DMSO concentration at least 0.2% (v/v). All inhibition test was conducted in a condition where the rate of hydrolysis of cGMP (cyclic Guanosine Monophosphate) does not exceed 15%. The amount of GMP formation depended on time and PDE5 Family protein to increase dose dependently and proportionally.

Example 1-2. Result of PDE5 Selectivity test for the compositions of the present invention

Based on the inhibitory activities of the compositions of the present invention against 11 species of PDE Family (MDS Pharma Services and Scotish Biomedical), IC50 of the compositions of the present invention against PDE5 was 0.338 nM (MDS Pharma Services) and 30-376,471 fold higher selectivity compared to the remaining 10 species of PDE Family proteins (Table 1).

aMDS Pharma Services; PDE1 (Bovine Heart); PDEs 2, 3, 5 (Human Platelete); PDE4 (Human U937 Cell); PDE6 (Bovine Retinal Rod).

bScottish Biomedical; PDEs 5, 7-11 (Recombinant Human Enzymes); NI = No Inhibition Observed. Example 2. Test for inhibition of Ab aggregation by the compositions of the present invention-

Example 2-1. Ab aggregation inhibitory (Ab Aggregation Inhibition) Test method

Example 2-1-1. Thioflavin T Assay

If Ab1-42 Peptide, one of the Alzheimer's major biomarkers and causes, is incubated at 37 °C for 3 days, Ab Fibril and Oligomers are generated . To test the activity of the composition of the present invention for inhibiting Ab Fibril and Oligomer formation, Ab1-42 (50 mM) was treated with the composition of the present invention in various concentrations (5, 50, 500 mM) before the mixture was incubated at 37 °C for 3 days followed by measuring the amount of Ab1-42 fibril and Oligomer. Using Varioskan LUX Multimode Microplate Reader (Thermo Fisher Scientific, USA) fluorescence intensity from Thioflavin T at Excitation 450 nm/Emission 485 was measured. Thioflavin T Assay measures fluororescence response from b-Sheet fromed as the peptide aggregates and showed higher valude as there are more aggregates formed. Even though Thioflavin T Assay cannot distinguish between Ab Oligomer and Fibril, it is an advantage that it can show the total amount of aggregated proteins quantitively. Example 2-1-2. PICUP / SDS-PAGE Analysis

The production of Ab Oligomer was observed by PICUP (Photo Induced Cross-linking of Unmodified Protein) analytical method . Using PICUP process, the generated Ab Oligomer and Fibril/Plaque are immobilized followed by separated by xize using SDS-PAGE (Sodium Dodecyl Sulfate -Polyacrylamide Gel Electrophoresis). Separate Ab1-42 Peptide is observed using Silver Staining .

Example 2-2. Result of Ab aggregation inhibition by the composition of the present invention

The result of Ab-42 aggregation inhibition shows that the composition of the present invention, compared to the untreated control group, inhibits Ab1-42 aggregation formation inhibitory effects discussed results dose-dependently, based on Thioflavin T Assay (Figure 1), and also by PICUP/SDS-PAGE Analysis, the present invention inhibited Ab1-42 Oligomer and Fibril/Plaque formation compared to the untreated control group (Figure 2). Considering the result, the composition of the present invention is expected to inhibit Ab Oligomer and with Fibril/Plaque formation.

Example 3. BACE-1 expression change in Retinoic Acid Differentiated SH-SY5Y Cell by the composition of the present invention

Example 3-1. Cell culture method for SH-SY5Y neurons

Example 3-1-1. Cell Subculture

A mixture of DMEM/F12 Complete Medium, DPBS, and Trypsin-EDTA was preheated in a 37 °C constant water bath for 30 minutes. The cell culture flask was removed from the incubator, to remove all medium and rinshed with DPBS once (T25:

5 ml, T75: 10 ml and T175: 20 ml). After discarding all DPBS, trypsin-EDTA (T25: 2 mL, T75: 5 mL and T175: 10 mL) was added, and the mixture was left for 4 minutes in an incubator with 5% CO 2 at 37 °C. In a 50 ml Conical Tube, the new Complete

Medium (T25 ml, T75: 10 ml and : T175:20 ml) and the cells were mixed and centrifuged at 1500 rpm for 4 minutes. Supernatant was all removed and 1 ml of

Fresh DMEM/F12 Complete Medium was added to resuspend the cells by either tapping or pipetting. Cells and the medium was added to the cell culture flask to make T75: 20 ml and T175: 40 ml before incubating the mixture in an incubator at 37

°C under 5%C0 2 .

Example 3-1-2. SH-SY5Y Neuroblastoma Differentiation

A mixture of DMEM/F12 Complete Medium (Hyclone), DPBS (Hyclone) and Trypsin-EDTA (Hyclone) was preheated in a 37 °C constant water bath for 30 minutes. The cell culture flask was removed from the incubator, to remove all medium and rinshed with DPBS once (T75: 10 ml and T175: 20 ml). After discarding all DPBS, trypsin-EDTA (T75: 5 mL and T175: 10 mL) was added, and the mixture was left for 4 minutes in an incubator with 5% CO 2 at 37 °C. In a 50 ml Conical Tube, the new

Complete Medium (T75: 10 ml and T175:20 ml) and the cells were mixed and centrifuged at 1500 rpm for 4 minutes. Supernatant was all removed and 1 ml of

Fresh DMEM/F12 Complete Medium was added to resuspend the cells by either tapping or pipetting. Certain amount of DMEM/F12 Complete Medium was added and transferred 4 x 10 4 cells/cm 2 into Collagen Type I Coated 6-Well Plate or Collagen Type I Coated 60 mm, 100 mm Cell Culture Dish (Corning). After 24 hours, the medium was changed with DMEM/F12 differentiation medium [+ 1% FBS + 1% Penicillin/Streptomycin (Hyclone) + 10 mM Retinoic Acid (Sigma-Aldrich)] . On day 5, medium was changed again with new DMEM/F12 differentiation medium. On day 8, medium was changed again with new DMEM/F12 differentiation medium. On day 10, differentiation was completed.

Example 3-2. Ab and preparation and treatment method of the present invention

DMEM / F12 (+ 1% FBS + 1% Penicillin / Streptomycin) was prepared to have 1 mM human Ab1-42 (Abeam). Ab solution was let to stand for 3 hours at 37 °C to form Ab Oligomer. The original cell culture medium was discarded and the newly prepared Ab1-42 Oligomer (1 mM) solution was treated alone or with the composition of the present invention (maximum concentration of 40 mM) before being incubated at 37 °C for 72 hours under 5% CO2 in an incubator. 72 hours later, the medium was removed and the cells were washed once with PBS and collected before continuing with the following experiments.

Example 3-3. qRT-PCR method

Total RNA was prepared using Easy-Blue™ Total RNA extraction kit (Intron Biotechnology) and using PrimeScript™ II 1st strand cDNA Synthesis Kit (TAKARA), 1 pg RNA was reverse-transcribed. cDNA was provided as a template for EmeraldAmp ® PCR Master Mix (TAKARA) and the next Primer for BACE-1 and b-Actin PCR: the Human BACE-1 (Forward 5'-CTGGTATACACCCATCCGGC-3', Reverse 5'- CTTGGGCAAACGAAGGTTGG-3'); Human b-Actin (Forward 5'- CCAGGTCATCACCATTGG-3', Reverse 5'- CAGAGTACTTGCGCTCAG-3'). The conditions for PCR Cycle were as follows: Denaturation (45 seconds at 95 °C); Annealing, Human BACE-1 (45 seconds at 60 °C), b-Actin (45 seconds at 56 °C); Extension (45 seconds at 72 °C); 40 Cycles. qRT-PCR was performed using QuantStudio™ 5 (Thermo Scientific) .

Example 3-4. Result of Changes in BACE-1 Expression by the composition of the present invention in Retinoic Acid Differentiated SH-SY5Y Cell

The present invention dose-dependently reduced expression of BACE-1 mRNA which was increased by treatingretinoic Acid Differentiated SH-SY5Y Cell with Ab 1- 42 (Figure 3). Considering the results, the present invention is expected to suppress formation of Ab1-42 or Ab1-43 that cause neuronal cell damage by suppressing expression of BACE-1, which plays major role in b-Amyloidogenic Processing of APP.

Example 4. Change of Calcium concentration within pericyte by the compositions of the present invention

Example 4-1. Method for culturing Pericytes

Example 4-1-1. Cell Subculture

Several T-75 cm 2 Cell Culture Flask (SPL, #70075) was coated using Attachment Factor™ (Cell Systems, #4Z0-210), an Extracellular Matrix (ECM), for 1 minute, followed by adding 15 mL of Complete Classic Medium with Serum and CultureBoost™ (Cell Systems, #4Z0-500) to the flask before stabilizing the mixture in an incubator at 37 °C under CO2. The incubated Primary Human Brain Pericytes (Cell Systems, #ACBRI 498P) was washed twice with PBS buffer, followed by adding 3 mL of 0.25% Trypsin (Hyclone, #SH30042.02) and reacting the mixture at 37 °C in CO2 incubator for 2 minutes. 7 mL of Complete Classic Medium with serum and CultureBoost™ was added and the clusters of cells were separated by repetitive pipetting, and the supernant was removed by centrifugal separation (1,500 rpm, 4 minutes). The cell clusters were homogenized in 1 mL of Complete Classic Medium With Serum and CultureBoost™ before incubating the cells in stabilized T-75 cm 2 Cell Culture Flask under optimum CO2. Example 4-2. Method for Preparation of and treatment with H 2 O 2 and measurement of changes of Intracellular Calcium Concentration after treatment

Flat-Bottom 96-Well Plate (SPL, # 30096) was coated with Attachment Factor ™ and Complete Classic Medium With Serum and CultureBoost™ (100 mL/well) was added to each well followed by stabilizing the plate at 37 °C in CO2 incubator for 1 hour. Primary Human Brain Pericytes treated with 0.25% Tyrypsin was added to Attachment Facotr-Coated-96-Well Plate containing Complete Classic Medium With Serum and CultureBoost™ and the mixture was incubated in 37 °C CO2 incubator for 16 hours (5 x 10 3 Cells/Well). The culture medium of the cultures cells was removed and the cells were washed with HEPES-Buffered Saline (132 mM NaCI, 5.9 mM KCI, 1.2 mM MgCI 2 , 1.5 mM CaCI 2 , 11.5 mM Glucose, 11.5 mM HEPES, 1.2 mM NaH 2 P0 4 ) once, transferred to 96-well containing Pericyte, wherein Ratiometric Intracellular Ca 2+ Dye, Fura-2 (Molecular Probes, # F1201), was diluted to 1 mM with HEPES- Buffered Saline before being incubated at room temperature for 30 minutes. The supernatant was removed from the 96-well plate, the cells were washed with HEPES- Buffered Saline three times, and treated the composition of the present invention diluted with HEPES-Buffered Saline and H 2 O 2 before measuring the fluorescence to measure the changes of intracellular calcium concentration.

Example 4-4. Result of changes of calcium concentration in pericyte of Retinoic Acid Differentiated SH-SY5Y Cell by the composition of the present invention

The composition of the present invention reduced the intracellular calcium concentration in perivascular cells (Pericyte) which was increased by the treatment of H 2 O 2 (Figure 4). Based on the result, it is expected that the composition of the present invention can inhibit the reduction of cerebral blood flow caused by the contraction of pericytes which in turn was caused by the increased calcium concentration in pericyte by the excessive production of ROS.

Example 5. Change of Intracellular cGMP in Retinoic Acid Differentiated SH-SY5Y Cell by the composition of the present invention Example 5-1. Cell culture method of SH-SY5Y neuronal cells Example 5-1-1. Cell Subculture

Using the method described in Example 3-1-1, cells were subcultured. Example 5-1-2. SH-SY5Y Neuroblastoma Differentiation

Using the method described in Example 3-1-2, SH-SY5Y neuronal differentiation was conducted.

Example 5-2. Ab and the preparation and treatment method of the present invention

Using the method described in Example 3-2, the preparation and treatment were performed.

Example 5-3. Intracellular cGMP measurement method

The experiment was conducted according to the manual of the Cyclic GMP

Complete ELISA Kit (Abeam, #abl33052, USA). Cell culture medium was removed and the cells were treated with 0.1 mL of 0.1M HCI before standing at room temperature for 10 minutes. The cells were recovered by 600g centrifugation, and only the supernatant was collected. Where 0.1M HCI is included in the sample, 50 ml of Neutralizing Reagent was added for each well. Followign the manual, 100 ml of each agent and the standard solution (0, 0.8, 4 20, 100, 500 pmol/ml) was added to each well of 96-well plate and 50 ml of Cyclic GMP Complete Alkaline Phosphatase

Conjugate was also added to each well. After 50 ml of Cyclic GMP Complete anitibody was added, reaction was conducted for 2 hours at room temperature. Once all reaction solution was removed, the well was washed with 400 ml of wash buffer three times. After the wash buffer was completely removed, 5 ml of Cyclic GMP Complete

Alkaline Phosphatase Conjugate was added. After adding 200 ml of pNpp Substrate Solution was added to each well, the reaction was conducted at room temperature for 1 hour. 50 ml of Stop Solution was added and immediately absorption value was measured using Plate Reader at 405 nm. Theh result was calculated using the manual in the Kit. Example 5-4 Result of changes of Intracellular cGMP in Retinoic Acid

Differentiated SH-SY5Y Cell by the present invention

The composition of the present invention concentration-dependently increased the concentration of cGMP which was reduced by the treatment of Retinoic Acid Differentiated SH-SY5Y Cell with Ab1-42 Oligomer (Figure 5). Considering this result, it is expected that the present invention suppress the activity of PDE5 selectively (Table 1) to inhibit the conversion of cGMP to 5'-GMP and increase Intracellular cGMP, which suppress the activity of Caspase-3 (Figure 6) and restore Mitochondrial membrane Potential to inhibit Neuronal Cell Death (Figure 8). Also, Intracellular cGMP increased by the present invention increases the activity of CREB (Figure 9) to promote Neurotrophin expression including NGF and BDNF (Figure 10, 11) to improve the cognitive function.

Example 6. Changes in Expression of apoptosis regulation related protein in Retinoic Acid Differentiated SH-SY5Y Cell by the present invention Example 6-1. Cell culture method of SH-SY5Y neuronal cells

Example 6-1-1. Cell Subculture

Using the method described in Example 3-1-1 Cell Subculture was conducted.

Example 6-1-2. SSH-SY5Y Neuroblastoma Differentiation

Using the method described in Example 3-1-2, SH-SY5Y neuronal cells were differentiated.

Example 6-2. Methods for Preparation of Ab and Treatment by the Present Invention

Human Ab1-42 (Abeam) was mixed with DMEM/F12 (+ 1% FBS + 1%

Penicillin/Streptomycin) to be 10 mM concentration. The Ab solution was allowed to stand at 37 °C for 3 hours to form Ab Oligomer. The culture medium was removed and the newly prepared Ab1-42 Oligomer solution (10 mM) was incubated with or without the composition of the present invention (maximum concentration of 40 mM) at 37 °C for 72 hours in 5% CO2 incubator. After 72 hours the culture medium was removed and the cells were washed with PBS once to collect the cells. Further experiments wwre carried out by the following procedures.

Example 6-5. Western Blot Analysis Method

To extract the proteins, the cells were treated with RIPA Lysis Buffer (Bio-Rad), homogenized, and centrifuged (14,000 rpm, 10 , 4°c) to obtain the supernatant. 5% Stacking Gel (DW, 30% Acrylamide: Bisacrylamide, 1M Tris pH 6.8, 10% SDS,

TEMED, 10% Ammonium Persulfate) and 12% Separating Gel (DW, 30% Acrylamide: Bisacrylamide, 1.5M Tris pH 8.8, 10% SDS, TEMED, 10% Ammonium Persulfate) were used for SDS-PAGE. Supernant obtained by centrifugation was mixed with 4X Laemmli Buffer (Bio-Rad) in 3: 1 ratio and boiled at 95 °C for 10 minutes to denature the proteins. The mixture was cooled in an ice bath for 10 minutes and spinned down.

Protein Size Marker (Bio-Rad) and each sample were added to Stacking Gel Well prepared in Mini-Protein P Dual-Slab Apparatus (Bio-Rad) and electrophoresed at

150 volt till all are precipitated to the bottom. Polyvinylidine Difluoride (PVDF) Membrane was moistened with methanol to activate and washed with Transfer Buffer (190 mM Glycine, 50 mM Tris-Base, 0.05% SDS, 20% Methanol). Whatman 3M Paper moistened with Transfer Buffer was stacked in Mini Trans-Bolt Cell (Bio-Rad) and run electricity at 200 mA for 60 minutes to deposit the membrane. After the dposition, the membrane was blocked with 5% Skim milk solution (in TBS-T: lOmM Tris-Base pH 8.0, 150 mM NaCI, 0.1% Tween-20) on a Platform Shaker for 60 minutes. The primary antibodies used for the current experiments are provided in Table 2. The primary antibodies were diluted in 5 % Skim Milk in an optimum concentration for each, put on platform shaker at 4 °C for 16 hours. The primary antibodies were recovered, washed with TBS-T 3 times for 10 minutes each, the secondary antibodies (Goat Anti-Rabbit IgG H&L (HRP), Abeam; Goat Anti-Mouse IgG H&L (HRP), Abeam) were diluted with 5 % Skim milk in 1 : 10,000 ratio, followed by put on a platform shaker at room temperature for 60 minutes and washted with TBS-T solution for 10 minutes, 3 times. Finally, Membrane was added to SuperSignal™ West Femto Maximum Sensitivity Substrate (Thermo Scientific ) and expressed the colro properly before the membrane was scanned using image analysis system and the amount of protein was calculated using Image J Software (NIH, USA).

Table 2. List of Primary Antibodies

Example 6-6 Change of expression of apoptosis regulation related protein in the Retinoic Acid Differentiated SH-SY5Y Cell by the present invention

The present invention suppressed formation of Cleaved Capase-3 which was increased by treating Retinoic Acid Differentiated SH-SY5Y Cell with Ab1-42 Oligomer. As a result, the amount of RARP cleaved by Cleaved Caspase-3 was reduced and thus reduced the amount of formation of Cleaved PARP (Figure 6). Considering these results, the present invention suppresses apoptosis caused by Ab1-42 Oligomerand to reduce Neuronal Cell Death to increase Neuronal Cell Survival.

Example 7. Change Of Mitochondrial Membrane Potential In Retinoic Acid Differentiated SH-SY5Y Cell By The Present Invention

Example 7-1. Method For Culturing SH-SY5Y Neurons

Example 7-1-1. Cell Subculture

Using the method described in Example 3-1-1, Cell Subculture was conducted. Example 7-1-2. SH-SY5Y Neuroblastoma Differentiation

Using the method described in Example 3-1-2, SH-SY5Y neuronal cells were differentiated. Example 7-2. Method for Preparation Of Ab And Treatment with The

Present Invention

Using the method described in Example 3-2, preparation and treatment was conducted. Example 7-3. Method for measuring Mitochondrial Membrane Potential

After the cell culture medium was removed, the cells were washed with CPBS once. JC-1 solution of JC-1 - Mitochondrial Membrane Potential Assay Kit (Abeam, USA) was mixed with medium without FBS and treated cells with the mixture. The cells were left in the 5% incubator at 37 °C for 15 minutes. The medium was removed and the cells were washed with DPBS once and clean medium was added. Using Varioskan LUX Multimode Microplate Reader (Thermo Fisher Scientific, USA), fluorescence of JC-1 Monomer at Excitation 475nm/Emission 530nm and JC-1 Dimer at Excitation 535nm/Emission 590nm. Example 7-4. Change of Mitochondrial Membrane Potential in Retinoic

Acid Differentiated SH-SY5Y Cell

The present invention dose-dependently increased Mitochondrial Membrane Potential reduced by the treatment of Retinoic Acid Differentiated SH-SY5Y Cell with Ab1-42 Oligomer (Figure 7). Considering the results, the present invention reduced Mitochondrial Damage caused by excess ROS formed by Ab1-42 Oligomer to increase Neuronal Cell Survival.

Example 8. Live -Cell Analysis by the Effect by Suppression of Neuronal Cell Death in Retinoic Acid Differentiated SH-SY5Y Cell by The Presetn Invention

Example 8-1. Cell Culture Method Of SH-SY5Y Neurons

Example 8-1-1. Cell Subculture Using the method described in Example 3-1-1, cell subculture was conducted.

Example 8-1-2. SH-SY5Y Neuroblastoma Differentiation

Using the method described in Example 3-1-2, SH-SY5Y Neuroblastoma Cells were differentiated.

Example 8-2. Preparation of Ab and Treatment with The Present Invention

Using the method described in Example 3-2, the preparation and treatment was conducted.

Example 8-3. IncuCyte S3 Live-Cell Analysis (Satorius, USA)

Cytotox Red Reagent (Essen Bioscience Cat #4632) was added in a concentration of 1 : 1,000 (v/v) and the cells were incubated in 5% CO 2 , at 37 °C for 24 hours. To analyze the cytotoxicity, the final ratio of the microscopy was adjusted to x200 and pictures were taken every 1.5 hours. Using the woftware provided by IncuCyte, the image was produced to be 1.5 fps and for the analysis of the cycotoxicity, Total Red Obfect Area read in the image was compared. The experiments were repeated three times and Student's T-Test was used for statistical analysis. All significance tests were performed at the P <0.05 level .

Example 8-4. Live-Cell Analysis Results By The Effect Of Suppression Of Neuronal Cell Death In Retinoic Acid Differentiated SH-SY5Y Cell By The Present Invention

The present invention reduced apoptosis cells dyed by Cytotox Red Reagent increased in Retinoic Acid Differentiated SH-SY5Y Cell by the treatment with Ab1-42 Oligomer (Figure 8). Considering this result, the present invention can increase Neuronal Cell Survival by suppressing Neuronal Cell Death induced by Ab1-42 Oligomer.

Example 9. CREB Expression Change In Retinoic Acid Differentiated SH-SY5Y Cell By The Present Invention Example 9 -1. Cell Culture Method Of SH-SY5Y Neurons

Example 9-1-1. Cell Subculture

Using the method described in Example 3-1-1, cell subculture was conducted.

Example 9-1-2. SH-SY5Y Neuroblastoma Differentiation

Using the method described in Example 3-1-2, SY5Y Neuronal Cells were differentiated.

Example 9-2. Preparation And Treatment Method Of Ab And The Present Invention

Using the method described in Example 3-2, preparation and treatment was conducted.

Example 9-4. Western Blot Analysis Method

To extract the proteins, cells were treated with RIPA Lysis Buffer (Biorad), homogenized, and centrifuged (14,000 rpm, 10 minutes, 4 °C) to obtain

supernatant. 5% Stacking Gel (DW, 30% Acrylamide: Bisacrylamide, 1M Tris pH 6.8, 10% SDS, TEMED, 10% Ammonium Persulfate) and 12% Separating Gel (DW, 30% Acrylamide: Bisacrylamide, 1.5M Tris pH 8.8, 10% SDS, TEMED, 10% Ammonium Persulfate) were used for SDS-PAGE. The supernatant obtained by centrifugation and 4X Laemmli Buffer (Bio-Rad) were mixed in 3: 1 ratio and boiled at 95 °C for 10 minutes to denature the proteins. The mixture was cooled in an ice bath and spinned down. Protein Size Marker (Bio-Rad) and each sample was injected into Stacking Gel Well equipped in Mini-Protein P Dual-Slab Apparatus (Bio-Rad) and electrophoresed at 150 volt until all are settle at the bottom. Polyvinylidine Difluoride (PVDF) Membrane was moistened with methanol to activate and washed with Transfer Buffer (190 mM Glycine, 50 mM Tris-Base, 0.05% SDS, 20% Methanol). Whatman 3M Paper moistened with Transfer Buffer was stacked in Mini Trans-Bolt Cell (Bio-Rad) and run electricity at 200 mA for 60 minutes to deposit the membrane. After the dposition, the membrane was blocked with 3% BSA (Bovine Serum Albumin) solution (in TBS- T: lOmM Tris-Base pH 8.0, 150 mM NaCI, 0.1% Tween-20) on a Platform Shaker for 60 minutes. The primary antibodies used for the current experiments are provided in Table 3. The primary antibodies were each diluted in 5 % Skim Milk solution in an optimum concentration for each, put on platform shaker at 4 °C for 16 hours. The primary antibodies were recovered, washed with TBS-T 3 times for 10 minutes each, the secondary antibodies (Goat Anti-Rabbit IgG H&L (HRP), Abeam; Goat Anti-Mouse IgG H&L (HRP), Abeam) were diluted with 5 % Skim milk in 1 : 10,000 ratio, followed by put on a platform shaker at room temperature for 60 minutes and washted with TBS-T solution for 10 minutes, 3 times. Finally, Membrane was added to SuperSignal™ West Femto Maximum Sensitivity Substrate (Thermo Scientific ) and expressed the colro properly before the membrane was scanned using image analysis system and the amount of protein was calculated using Image J Software (NIH, USA).

Table 3. List of Primary Antibodies

Example 9-5. CREB Expression Changes In The Retinoic Acid Differentiated SH-SY5Y Cell By The Present Invention

The present invention concentration-dependently increased CREB Protein °.|

Serl33 Phosphorylation decreased in Retinoic Acid Differentiated SH-SY5Y Cell by the treatment with Ab1-42 Oligomer (Figure 9). Considering this result, the present invention is expected to increase Serl33 Phosphorylation in CREB Protein reduced by Ab1-42 Oligomer to restore the activities of CREB protein activitie, a Cellular Transcription Factor, to induce expression of factors related to Neurogenesis, Synaptogenesis, and Angiogenesis to improve cognitive function.

Example 10. NGF And BDNF Of Expression Change In Retinoic Acid Differentiated SH-SY5Y Cell By The Present Invention Example 10-1. Cell Culture Method Of SH-SY5Y Neurons

Example 10-1-1. Cell Subculture

Using the method described in Example 3-1-1, cell subculture was conducted. Example 10 -1-2. SH-SY5Y Neuroblastoma Differentiation

Using the method described in Example 3-1-2, SH-SY5Y Neuroblastoma was differentiated.

Example 10-2. Preparation And Treatment Method Of Ab And The Present Invention

Using the method described in Example 3-2, preparation and treatment was conducted.

Example 10-3. Western Blot Analysis Method

Using the method described in Example 6-5, Western Blot Analysis has been conducted and the primary antibodies and the conditions used for the current experiments are provided in Table 4.

Table 4. List of Primary Antibodies

Example 10-4. NGF Immunocytochemistry (ICC)

SH-SY5Y was incubated in Corning BioCoat Collagen I Culture Slides (Corning, #354630), 4.8 x 10 4 Cells/Well, for7 days, was treated with Ab1-42 Oligomer (72 hours) and the present invention (24 hours), and NGF Immunostaining experiment was conducted. Cells were fixed at 4% Paraformaldehyde (pH 7.4) for 10 minutes, and washed with Ice-Cold PBS Buffer three times. After Cell Permeabilization with 0.1% Triton X-100 in PBS, cells were washed with Ice-Cold PBS Buffer three times. Fixed cells were blocked using Blocking Solution (10% Normal Goat Serum in PBS) for 30 minutes. The primary antibodies used for the current experiment is Rabbit Anti-NGF Antibody (Abeam, #ab52918) from Table 4. Cells were incubated in Rabbit Anti-NGF Antibody (X300 Dilution) Solution at 4 °C for 16 hours, washed with PBS Buffer three times, incubated with Alexa 488 Anti-Rabbit IgG Secondary Antibody (X500 Dilution) at room temperature for 1 hour, washed with PBS once, react with DAPI Staining Solution (Abeam, #ab228549) for Nuclear Staining for 1 minute and washed with PBS Buffer 3 times. Cells were treated with Vector Shield Mounting Medium, and fluorescence image was obtained using Super Resolution Confocal Laser Microscopy (Carl Zeiss, #LSM800).

Example 10-5. NGF and BDNF of expression changes In Retinoic Acid Differentiated SH-SY5Y Cell By The Present Invention

Upon reviewing the effect of increased expression of Neurotrophin by the present invention, Western Blot Analsysis showed that the present invention concentration-dependently increased expression of NGF and BDNF decreased in Retinoic Acid Differentiated SH-SY5Y Cell by the treatment with Ab1-42 Oligomer (Figure 10), and Immunocytochemistry also showed that the present invention increased the expression of NGF in Retinoic Acid Differentiated SH-SY5Y Cell by the treatment with Ab1-42 Oligomer (Figure 11). Based on the results, the present invention is expected to increase the expression of neurotrophin including NGF and BDNF that play important role to induce neuron's survival, development and function to stimulate survival and differentiation of neuronal cells to improve the cognitive function.

Example 11. Effecty On Expression Of DKK-1 In Retinoic Acid Differentiated SH-SY5Y Cell And Differentiated HT-22 Mouse Hippocampal Neuronal Cell By The Present Invention

Example 11-1. Cell culture method of SH-SY5Y neurons

Example 11-1-1. Cell Subculture

Using the method described in Example 3-1-1, cell subculture was conducted. Example 11-1-2. SH-SY5Y Neuroblastoma Differentiation

Using the method described in Example 3-1-2, SH-SY5Y cells were differenctiated. Example 11-2. Cell culture method of HT-22 Mouse Hippocampal

Neuronal Cell Line

Example 11-2-1. Cell Subculture

DMEM/High Glucose Medium (10% FBS, 1% Penicillin-Streptomycin), DPBS and 0.25% Trypsin-EDTA were pre-heated in a 37 °C constant temperature bath for 30 minutes. Cell culture flask was taken out of the incubator, the existing medium was discarded and washed with DPBS once (T25: 5 ml, T75: 10 ml and T175: 20 ml). DPBS was all discarded, 0.25% Trypsin-EDTA (T25: 2 ml, T75: 5 ml and T175: 10 ml) was added and the mixture was left in an incubator under 5% CO2, at 37 °C for 2 minutes. New Complete Medium (T25: 4 ml, T75: 10 ml and T175: 20 ml) and cells were mixed in 50 ml Conical Tube and centrifuged at 1500 rpm for 4 minutes. After removing all supernatant, Fresh DMEM/High Glucose was added and cells were resuspended either by Tapping or Pipetting. Cells and the medium was added to a

Cell Culture Flask (T75 - 15 ml and T175 - 40 ml) and incubated under 5% CO2, at

37 °C.

Example 11-2-2. Neuronal Differentiation of HT-22 cells

DMEM/High Glucose Medium (10% FBS, 1% Penicillin-Streptomycin), DPBS and 0.25% Trypsin-EDTA were pre-heated in a 37 °C constant temperature bath for 30 minutes. Cell culture flask was taken out of the incubator, the existing medium was discarded and washed with DPBS once (T25: 5 ml, T75: 10 ml and T175: 20 ml). DPBS was all discarded, 0.25% Trypsin-EDTA (T25: 2 ml, T75: 5 ml and T175: 10 ml) was added and the mixture was left in an incubator under 5% CO2, at 37 °C for 2 minutes. New Complete Medium (T25: 4 ml, T75: 10 ml and T175: 20 ml) and cells were mixed in 50 ml Conical Tube and centrifuged at 1500 rpm for 4 minutes. After removing all supernatant, 1 ml of Fresh DMEM/High Glucose was added and cells were resuspended either by Tapping or Pipetting. Optimum amount of DMEM/High Glucose Medium was further added and 2 x 10 5 cells/cm2 were added to Collagen Type I Coated 6-Well Plate. After 24 hours, the medium was changed to differentiation medium [Neurobasal Plus medium (Gibco) + B-27 supplement (Gibco) + 1% Penicillin-streptomycin] . After 24 hours, medium was changed to new differentiatiation medium and treated with Ab1-42 Oligomer and the present invention for 6 hours.

Example 11-3. Preparation And Treatment Of Ab And The Present Invention

Using the method described in Example 3-2, preparation and treatment was conducted.

Example 11-4. qRT-PCR method

Total RNA was prepared using Easy-Blue™ Total RNA extraction kit (Intron Biotechnology) and 1 mg RNA was reverse-transcribed using PrimeScript™ II 1st strand cDNA Synthesis Kit (TAKARA). cDNA was provided as a template using EmeraldAmp® PCR Master Mix (TAKARA) and next Primer for human DKK and b- Actin PCR: Human DKK1 (Forward 5'-ATTCCAACGCTATCAAGAACC-3', Reverse 5'- CCAAGGTGCTATGATCATTACC-3'); Human b-Actin (Forward 5'- CCAGGTCATCACCATTGG-3', Reverse 5'- CAGAGTACTTGCGCTCAG-3'); Mouse DKK1 (Forward 5'-TCTGCTAGGAGCCAGTGCC-3', Reverse 5'-GATGGTGATCTTTCTGTATCC- 3'); Mouse b-Actin (Forward 5'-CTGTCCCTGTATGCCTCTG-3', Reverse 5'-

ATGTCACGCACGATTTCC-3')· The condition for PCR Cycle is as follows : Denaturation, 45 seconds at 95 °C; Annealing, 45 seconds at 56 °C; Extension, 45 seconds at 72 °C; 40 Cycles. qRT-PCR was carried out using QuantStudio™ 5 (Thermo Scientific) .

Example 11-5. Western Blot Analysis Method Using the method described in Example 6-5, Western Blot Analysis was carried out and the primary antibodies and the condition used for the current experiment is provided in Table 5. Table 5. List of Primary Antibodies

Example 11-6. Change In DKK-1 Expression In Retinoic Acid Differentiated SH-SY5Y Cell And Differentiated HT-22 Mouse Hippocampal Neuronal Cell By The Present Invention

The present invention decreased expression of DKK- 1 in mRNA (Figure 12) a nd in

Protein Level (Figure 13) in Retinoic Acid Differentiated SH-SY5Y Cell and Differentiated HT- 22 Mouse Hippoca mpa l Neuronal Cell increased by treatment with Ab1-42 Oligomer. Considering the result, the present invention is expected to decreased DKK- 1 expression increased by Ab1-42 Oligomer, restore Syna ptic Plasticity by activation of Wnt Sig na ling (Figure 14-20) and red uction of APP formation and Ab accumulation by the suppression of Positive Feedback Loop of Ab production (Fig ure 21, 22) .

Example 12. Wnt Gene Expression Changes in Differentiated HT-22 Mouse Hippocampal Neuronal Cell By The Present Invention

Example 12-1. Cell Culture Method Of HT-22 Mouse Hippocampal

Neuronal Cell Line

Example 12-1-1. Cell Subculture

Usign the method described in Example 11-2-1, Cell Subculture was carried out.

Example 12-1-2. Neuronal Differentiation of HT-22 cells

Usign the method described in Example 11-2-2, Cell Subculture was carried out. Example 12-3. Preparation And Treatment Method Of Ab And The The Present Invention

Prepared a 10 mM solution of human Ab1-42 (Abeam) in DMEM/F12 (+ 1% FBS + 1% Penicillin/Streptomycin). Solution of Ab was left at 37 °C for 3 hours to form Ab Oligomer. The existing medium was discarded and the cells were treated with newly prepared Ab1-42 Oligomer (10 mM) solution with or without the present invention (maximum concentration 5 mM), and incubated under 5% CO2 at 37 °C for 72 hours. After 72 hours, the medium was removed and the cells were washed with PBS once and recovered. Using the following procedure, further experiments were carried out.

Example 12-3. qRT-PCR method

Total RNA was prepared using Easy-Blue™ Total RNA (Intron

Biotechnology), 1 pg RNA was rever-transcribed using PrimeScript™ II 1st strand cDNA Synthesis Kit (TAKARA). cDNA was provided as a template for Wnt and b-Actin PCR using EmeraldAmp® PCR Master Mix (TAKARA) and the following Primer: Mouse Wntl (Forward 5'-CTCTTTGGCCGAGAGTTCGTGG-3', Reverse 5'- CCTCGGTTGCCGTAAAGGACGC-3'); Mouse Wnt3a (Forward 5'-

CTCGCATGGCATAGATGGGTGC-3', Reverse 5'-GCAGGTGTGCACGTCATAGAC-3'); Mouse Wnt5a (Forward 5'-CATGGAGTGTCTGGCTCCTG-3', Reverse 5'-

GTCCATCCCCTCTGAGGTCTTG-3') ; Mouse Wnt7a (Forward 5'-

CGGGAGATCAAGCAGAATGC-3', Reverse 5'-GCCTAGCTCTCGGAACTGTGGC-3'); Mouse b-Actin (Forward 5'-CTGTCCCTGTATGCCTCTG-3', Reverse 5'-

ATGTCACGCACGATTTCC-3')· The condition for PCR Cycle was as following : Denaturation (45 seconds at 95 °C); Annealing, Wnt 1, Wnt3a, Wnt 5a (45 seconds at 60 °C); Wnt7a (45 seconds at 57 °C); b-Actin (45 seconds at 56 °C); Extension (45 seconds at 72 °C); 40 Cycles. qRT-PCR was conducted using QuantStudio™ 5 (Thermo Scientific).

Example 12-4. Wnt gene expression changes In Differentiated HT-22 Mouse Hippocampal Neuronal Cell By The Present Invention The present inventionincreased the Wnt3a gene expression in Differentiated HT-22 Mouse Hippocampal Neuronal Cell on treatment with Ab1-42 Oligomer compared to control group (Figure 14). Considering the result, the present invention is expected to increase the Wnt3a expression which is known to increase synaptic activities that is important for cognitive function, and activate Canonical Wnt Signaling to restore synaptic plasticity.

Example 13. Wnt Expression Change In Alzheimer's Disease Animal Model (5XFAD Transgenic Mice) By The Present Invention

Example 13-1 Alzheimer's Disease Animal Model (5XFAD Transgenic

Mice)

Internationally widely used Alzheimer's transgenic mice model 5XFAD (C57BL / 6 x SJL) was used. This model's features is that the progress of Alzheimer's disease is significantly faster than other mouse model via 5 different genetic transformation (APP KM670/671NL (Swedish), APP I716V (Florida), APP V717I (London), PSEN1 M146L (A>C), PSEN1 L286V ). In other words, any composition therapeutically effective in this model is expected to be effective in other APP model relatively slow in progress. Analysis was conducted using brain tissues in Cortex and Hippocampus area which are related to cognitive function.

Example 13-2. Dosage Of The Present Invention

Mice at active age with progressed Alzheimer's disease pathology and cognitive function deteriorated (6 months, male) were administered daily for 4 weeks via intraperitoneal injection 5 mg/kg/day or 10 mg/kg/day.

Example 13-3. Preparation of Tissues

The present invention was administered daily for 4 weeks by Intraperitoneal Injection, animals were anesthesized in animal chambers using CO2 gas, the brain was excised to measure the amount of protein expression, and hippocampus and cerebral cortex were separated and stored in a Deep Freezer at -80 °C till analysis of protein expression. Example 13-5. Western Blot Analysis Method

Using the method described in Example 6-5, Western Blot Analysis was carried out and the primary antibodies and the condition used for the current experiment is provided in Table 6.

Table 6. List of Primary Antibodies

Antibody Dilution Ratio Catalog No Company

Wntl 1 : 500 abl5251 Abeam b-Actin (13E5) Rabbit mAb 1 :2,000 4970 Cell Signaling

Example 13-6. Wntl Expression Changes In Alzheimer's Disease Animal Model (5XFAD Transgenic Mice) By The Present Invention

The present invention increased reduced Wntl expression in hippocampus of

Alzheimer's Disease Animal Model (5XFAD Transgenic Mice) (Figure 15) . Considering the result, the present invention is expected to increase the expression of Wntl providing important function for Synpatogenesis between the neurons to restore synaptic plasticity.

Example 14. Confirmation of activation of factors related to Wnt/b- Catenin related factors in Differentiated HT-22 Mouse Hippocampal Neuronal Cell by the present invention using Wn b-Catenin TF Activation Profiling Plate Array

Example 14-1. Cell culture method of HT-22 Mouse Hippocampal

Neuronal Cell Line

Example 14-1-1. Cell Subculture

Using the method described in Example 11-2-1, Cell Subculture was carried out.

Example 14-1-2. Neuronal Differentiation of HT-22 cells

Using the method described in Example 11-2-2, Cell Subculture was carried out. Example 14-2. Method Of Preparation Of Ab And Treatment With The Present Invention

Prepared a 10 mM solution of human Ab1-42 (Abeam) in DMEM/F12 (+ 1% FBS + 1% Penicillin/Streptomycin). Solution of Ab was left at 37 °C for 3 hours to form Ab Oligomer. The existing medium was discarded and the cells were treated with newly prepared Ab1-42 Oligomer (1 mM) solution with or without the present invention (maximum concentration 5 mM), and incubated under 5% CO2 at 37 °C for 6 hours. After 6 hours, the medium was removed and the cells were washed with PBS once and recovered. Using the following procedure, further experiments were carried out.

Example 14-3. Wn b-Catenin TF Activation Profiling Plate Array Method

Nuclear Extract was extracted from the cells and Wn b-Catenin TF Activation Profiling Plate Array (Signosis, #FA-1007) was carried out following the procedure provided by the vendor. Relative Light Unit was measured using Varioskan LUX Multimode Microplate Reader (Thermo Fisher Scientific, USA).

Example 14-4. Activation of Wn b-Catenin -related transcription related factors measured by Transcription Factor Array

The present invention increased the activities of Wn b-Catenin related transcription factors including VAX2, c-Myc, NR5A2, Mitf, TCF/LEF, NFAT, CEBP, GLI- 1, GBX2, and AP-1 in Differentiated HT-22 Mouse Hippocampal Neuronal Cell treated with Ab1-42 Oligomer more than twice (Figure 16). Considering the result, the present invention is expected to increase activity of Wn b-Catenin related transcription factors to stimulate Wnt Signaling activation to recover synaptic plasticity.

Example 15. Measurement of changes in Wn b-Catenin Activity in Differentiated HT-22 Mouse Hippocampal Neuronal Cell by the present invention using TOPFLASH Reporter Gene Assay

Example 15-1. Cell culture method of HT-22 Mouse Hippocampal Neuronal Cell Line

Example 15-1-1. Cell Subculture Using the method described in Example 11-2-1, Cell subculture was carried out.

Example 15-1-2. Neuronal Differentiation of HT-22 cells

Using the method described in Example 11-2-2, Cell Subculture was carried out.

Example 15-2-3. Transfection of TOPFLASH Reporter Plasmid DNA

In a 6-Well Plate, Fugene HD Transfection Reagent (Promega, #E2311) and TOPFLASH Reporter Plasmid DNAs (pcDNA-b-Galactosidase, TOPFLASH) were added to HT-22 cells differentiated in 2 x 10 5 Cells/Well, and the cells were incubated at 37 °C under 5% CO2 for 24 hours.

Example 15-2-3. Methdo for Preparation and treatment of Ab and the present invention

Prepared 1 mM solution of Human Ab1-42 (Abeam) in DMEM/F12 (+ 1% FBS + 1% Penicillin/Streptomycin). The solution of Ab1-42 was left at 37 °C for 3 hours to form Ab Oligomer. After transfecting for 24 hours, the existing cell medium was discarded and the cells were treated with newly prepared Ab1-42 Oligomer (1 mM) solution with or without the present invention (max. concentration 5 mM) and incubated under 5% CO2 at 37 °C for 6 hours. After 6 hours, the medium was removed and the cells were washed with PBS once and recovered. Further experiments were conducted by the following procedures.

Example 15-3. TOPFLASH Reporter Gene Assay Method

HT-22 cells were washed with PBS buffertwice, Cell Lysate was prepared (100 mI/Well) with Lysis Buffer (0.1% Triton X-100, 200mM Tris-CI (pH 8.0), Complete Mini Protease Inhibitor Cocktail (Roche), and Pierce Phosphatase Inhibitor Mini-Tablet) proteins were quantified. Using part of Cell Lysate, b-Galactosidase Assay was carried out and light absorption was measured at 420 nm. Using the rest of Cell Lysate, Luciferase Assay was carried out to measure Luminescence. The obtained Luminescence value was normalized with the protein concentration using b- Galactosidase Activity (Absorbance at 420 nm) to obtain Relative Luciferase Activity value.

Example 15-4. Result of Wn b-Catenin Activity by the present invention In Differentiated HT-22 Mouse Hippocampal Neuronal Cell measured by TOPFLASH Reporter Gene Assay

The present invention concentration-dependently increased Wn b-Catenin Activity in Differentiated HT-22 Mouse Hippocampal Neuronal Cell reduced by treatment with Ab1-42 Oligomer (Figure 17) . Considering the result, the present invention is expected to increase Wnt expression (Figure 14 and 15) and activities of Wn b-Catenin related transcription factors (Figure 16), to promote activation of Wnt Signaling to recover synaptic plasticity.

Example 16. Change of GSK3 b expression in Retinoic Acid Differentiated SH-SY5Y Human Neuroblastoma Cell and Differentiated HT- 22 Mouse Hippocampal Neuronal Cell by the present invention

Example 16 -1. Cell culture method of SH-SY5Y neurons

Example 16-1-1. Cell Subculture

Using the method described in Example 3-1-1, Cell Subculture was carried out.

Example 16-1-2. SH-SY5Y Neuroblastoma Differentiation

Using the method described in Example 3-1-2, SH-SY5Y cells were differentiated.

Example 16-2. Cell culture method of HT-22 Mouse Hippocampal Neuronal Cell Line

Example 16-2-1. Cell Subculture

Using the method described in 11-2-1, Cell Subculture was carried out.

Example 16-2-2. Neuronal Differentiation of HT-22 cells

Using the method described in 11-2-2, Cell Subculture was carried out.

Example 16-3. Method For Preparation Of Ab And The Treatment With The Present Invention Usign the method described in Example 3-2. The preparation and treatment was carried out.

Example 16-4. Western Blot Analysis Method

Using the method described in Example 9-4, Western Blot Analysis was carried out and the primary antibodies and it's conditions are provided in Table 7.

Table 7. List of Primary Antibodies

Antibody Dilution Ratio Catalog No Company pSer9-GSK3b (Ser9) (D3A4) 1 ;2,000 9322 Cell Signaling

GAPDH 1 : 1,000 2118L Cell Signaling b-Actin 1 :2,000 4970 Cell Signaling Example 16-5. PathScan ® Phospho-GSK-3b (Ser9) Sandwich ELISA

HT-22 cells (1.5 x 10 5 Cells/Well) were incubated in 6-Well Plate (SPL, #30006) . The medium was changed with Neurobal Plus Meida with B-27 Supplement and, after 24 hours, cells were treated with Ab1-42 Oligomer (1 mM) and the present invention (1, 2, 5 mM) for 6 hours. Cell Culture Media was removed and cells were washed with PBS Buffer three times, and Cell Lysis Buffer (Cell Signaling, #9803) was added to each well ( 100 mI/Well) . The cells were lef ton top of ice for 5 minutes and Cell Lysate was scrapped to move to 1.5 ml and Cell Lysate was homogenized using Sonication using Bioruptor equipment. Homogenized Cell Lysate was centrifuged (14,000 rpm, 10 minutes) and the supernatant was analyzed to quantify the amount of protein using Pierce™ BCA Protein Assay Kit. GSK-3b Mouse mAb Coated Microwell included in PathScan Phospho- GSK-3b (Ser9) Sandwith ELISA Kit (Cell Signaling, #7311C) was stabilized at room temperature for 30 minutes and same amount of protein (30 mg/well) diluted with ELISA Sample Diluent was added to each Microwell and the mixture was reacted at 4 °C for 16 hours. Each Microwells was washed with ELISA Wash Buffer four times, ELISA experiment was performed using Phospho-GSK-33 (Ser9) Rabbit Detection mAb and Anti-Rabbit IgG, HRP-Linked Antibody (ELISA Formulated) . TMB Substrate Solution was used as ELISA Substrate to measure absorption at 450nm, to determine relative amount of Phospho- GSK-3b (Ser9) . Example 16-6. GSK3 b expression changes by the present invention In Retinoic Acid Differentiated SH-SY5Y Human Neuroblastoma Cell and Differentiated HT-22 Mouse Hippocampal Neuronal Cell

Reviewing the effect by the present invention on the phosphorylation of GSK33 protein, as shown in Western Blot Analysis, the present invention increased Ser9 Phosphorylation of protein in Retinoic Acid Differentiated SH-SY5Y Cell which was reduced by Ab1-42 Oligomer (Figure 18), and Phospho-GSK-3b (Ser9) Sandwich ELISA shows that the present invention increased Ser9 Phosphorylation of protein in Differentiated HT-22 Mouse Hippocampal Neuronal Cell which was reduced by Abΐ- 42 Oligomer (Figure 19). Considering the results, the present invention is expected to increase Ser9 Phosphorylation of protein to deactivate GSK3, to reduce phosphorylation of Tau protein to reduce Tau pathology.

Example 17. Change In Tau Expression By The Present Invention In Alzheimer's Dementia Animal Model (NSE-Happ-C105)

Example 17-1. Experimental Animals

The animals for experiments were C57BL/6-Tg(NSE-hAPP-C105)Kor Transgenic Mice (13 Months) in which dementia by Alzheimer was induced by overexpressing mutated APP gene expressing APP C Terminal 105 amino acids only in brain tissues. The animals were housed in the condition contolled as temperature 20±2 °C, humidity 50%, light-on for 08 :00- 20 : 00 and light-off for20 :00-08 :00. Groups were identified as Control/no treatment group [Non-tg Control, NTC (n = 6), Alzheimer disease group [tg-Control, TC (n = 6)], Alzheimer disease group treated with the present invention [tg-Presenst Invention, TM (n = 6)] and food and water were supplied without limitation during the experiment.

Example 17-2. Method Of Administration Of The Present Invention

After one time base test for Alzheimer's disease model including mouse memory test, behavioral tests by maze in water motor function test, and passive avoidance test, human's daily dose of the present invention, based on Reagan-Shaw, etc. (2008), was administered to 13 month old Alzheimer's disease model mouse by Intraperitoneal Injection at 4 mg/kg once daily for 4 weeks. The controlled group (NTC) and Alzheimer's disease group (TC) were administered with the same amount of physiological saline.

Example 17-3. Tissue preparation

The present invention was administered daily for 4 weeks by Intraperitoneal Injection, animals were anesthesized in animal chambers using CO2 gas, the brain was excised to measure the amount of protein expression, and hippocampus and cerebral cortex were separated and stored in a Deep Freezer at -80 °C till analysis of protein expression. Example 17-4. Western Blot Analysis Method

Using the method described in Example 9-4, Western Blot Analysis was carried out and the primary antibodies and it's conditions are provided in Table 8.

Table 8. List of Primary Antibodies

Example 17-5. Tau Expression Change By The Present Invention In Alzheimer's Dementia Animal Model (NSE-Happ-C105)

The present invention reduced Serl99/202 Phosphorylation in Tau Protein increased in hippocampus of the Alzheimer dementia animal model (NSE-hAPP-C105) (Figure 20). Consideering this result, the present invention is expected to reduce Serl99/202 Phosphorylation in Tau Protein to suppress aggregation of Tau Protein, to reduce the formation of NFT and to reduce Tau pathology.

Example 18. Change In APP Formation And Ab Accumulation By The Present Invention In Retinoic Acid Differentiated SH-SY5Y Human Neuroblastoma Cell

Example 18 -1. Cell culture method of SH-SY5Y neurons Example 18-1-1. Cell Subculture

Using the method described in Example 5-1-1, Cell Subculture was carried out.

Example 18-1-2. SH-SY5Y Neuroblastoma Differentiation

Using the method described in Example 5-1-2, SH-SY5Y neuronal cells were differentiated .

Example 18-2. Method For Preparation Of Ab And Treatment With The Present Invention

Example 18-2-1. Method For Preparation Of Ab And Treatment With

The Present Invention

Using the method described in Example 5-2, the preparation and treatment were performed . Example 18-2-2. Method For Preparation Of H2O2 And Treatment With

The Present Invention

DMEM/F12 (+ 1% FBS + 1% Penicillin/Streptomycin) containing 100 mM H 2 O 2 was added to the differentiated SH-SY5Y Cells every 24 hours and refreshed three times, the present invention was added in various concentration (10, 20, 40 mM) for 24 hours.

Example 18-3. Western Blot Analysis Method

Using the method described in Example 6-5, Western Blot Analysis was carried out and the primary antibodies and it's conditions are provided in Table 9.

Table 9. List of Primary Antibodies

Example 18-4. Change In APP Formation Ab Accumulation By The Present Invention In Retinoic Acid Differentiated SH-SY5Y Human Neuroblastoma Cell

The present invention reduced APP formation and Ab1-42 accumulation increased by treatment with Ab1-42 Oligomer (Figue 21)

H 2 O 2 (Figure 22) in

Retinoic Acid Differentiated SH-SY5Y Cell . Considering the result, the present invention is expected to reduce APP formation and Ab accumulation by suppressing Positive Feedback Loop of Ab production . Example 19. Change In Ab Expression By The Present Invention In

Alzheimer's Dementia Animal Model (NSE-Happ-C105)

Example 19-1. Experimental animals

The sa me type of anima l model described in Example 17- 1 was used . Example 19-2. Method Of Administration Of The Present Invention

The present invention was administered using the same method described in Example

17-2.

Example 19-3. Preparation Of Tissue

Using the method described in Example 17-3, tissue samples were prepared and stored.

Example 19-4. Western Blot Analysis Method

Using the method described in Example 6-5, Western Blot Analysis was carried out and the primary antibodies and it's conditions are provided in Table 10.

Table 10. List of Primary Antibodies

Example 19-5. Results of Ab expression change in Alzheimer's dementia animal model (NSE-hAPP-C105)

The present invention effectively reduced expression of Ab in hippocampus in Alzheimer's dementia animal model (NSE-hAPP-C105) (Figure 23). Based on this result, the present invention is expected to effectively suppress formation and accumulation of Ab.

Example 20. Ab Plaque Change By The Present Invention In Alzheimer's Disease Animal Model (5XFAD Transgenic Mice)

Example 20-1 Alzheimer's Disease Animal Model (5XFAD Transgenic

Mice)

The animal model described in Example 13-1 was used.

Example 20-2. Administration of the Present Invention Composition

Using the method described in Example 13-2, the composition of the present invention was administered.

Example 20-3. Preparation And Fixation Of Tissue Samples

The present invention was administered by Intraperitoneal Injection for 4 weeks daily, and the animal was anesthesized in an animal chamber using CO2 gas before opening the chest to administer 50 mM PBS (Phosphate Buffer Saline) to the left ventricle for 3 minutes, and perfused a fixation solution in which 4% PFA (Paraformaldehyde) was dissolved in 0.1 M phosphate buffer for 10 minutes. After perjusion and fixation, the brain was excised and added to 4% PFA fixation solution at 4 °C for 12 hours to fix, followed by precipitating the tissues in 30% Sucrose solution for 5 days, and sliced through

Continuous tubular intercept in 40 mm thickness using Freezing Microtome (Leica)for storage. Example 20-4. Ab Plaque Staining Methods Using Thioflavin S Stainin G And GFAP (Glial Fibrillary Acidic Protein) Immunohistochemistry (IHC) Method

After fixation of the mouse brain tissues in 4% Paraformaldehyde (pH 7.4) siolution for 24 hours, the tissues were dehydrated using 30% Sucrose, and cryosections were prepared . The primary antibodies and the conditions are provided in Table 11. After Antigen retrieval using 1% SDS was performed on the Cryosections of the train tissues, the sampels were reacted with Glial Fibrillary Acidic Protein (GFAP) Antibody at 4 °C for 16 hours. The sampels were treated with 500 mM Thioflavin S in 50% Ethanol for 7 minutes for Thioflavin S Staining . Alexa Fluor® 594 Goat Anti- Rabbit (IgG) Secondary Antibody was reacted with GFAP Antibody/Thioflavin S treated brain tissue cryosections, and Hoechst 33342 (Sigma-Aldrich) was used for Nuclear Staining .

Table 11. List of Primary Antibodies

Example 20-5. Change Of Ab Plaque By The Present Invention In Alzheimer's Disease Animal Model (5XFAD Transgenic Mice)7260

The present invention reduced number of Ab Plaque in hippocampus and cerebral cortex of Alzheimer's disease animal model (5XFAD Transgenic Mice) (Figure 24), which was determined by counting number of Ab Plaque (Green) dyed by Thioflavin S and Colocalized Spot (Yellow) of Astrocyte (Red) dyed with GFAP. Considering this result, the present invention is expected to suppress Ab production and Ab accumulation to reduce extracellular Ab Plaque formation . Example 21. AMPK Expression Change By The Present Invention In Retinoic Acid Differentiated SH-SY5Y Human Neuroblastoma Cell

Example 21 -1. Cell Culture Method Of SH-SY5Y Neurons

Example 21 -1-1. Cell Subculture

Using the method described in Example 3-1-1, Cell Subculture was conducted.

Example 21 -1-2. SH-SY5Y Neuroblastoma Differentiation

Using the method described in Example 3-1-2, SH-SY5Y neuronal cells were differentiated.

Example 21-2. Method For Preparation Of Ab And Treatment With The Present Invention

Prepared a 1 mM solution of human Ab1-42 (Abeam) in DMEM/F12 (+ 1% FBS + 1% Penicillin/Streptomycin). Solution of Ab was left at 37 °C for 3 hours to form Ab Oligomer. The existing medium was discarded and the cells were treated with newly prepared Ab1-42 Oligomer (1 mM) solution with or without the present invention (maximum concentration 10 mM), and incubated under 5% CO2 at 37 °C for 72 hours. After 72 hours, the medium was removed and the cells were washed with PBS once and recovered. Using the following procedure, further experiments were carried out.

Example 21-3. Western Blot Analysis Method

Using the method described in Example 9-4, Western Blot Analysis was carried out and the primary antibodies and it's conditions are provided in Table 12.

Table 12. List of Primary Antibodies

Antibody Dilution Ratio Catalog No Company pAMPKa (Thrl72) 1 : 1,000 2531 Cell Signaling

GAPDH 1 : 1,000 2118L Cell Signaling

Example 21-4. AMPK Expression Changes By The Present Invention In Retinoic Acid Differentiated SH-SY5Y Human Neuroblastoma Cell The present invention concentration-dependently increased phosphorylation of Thrl72 of AMPK Catalytic Subunit a in Retinoic Acid Differentiated SH-SY5Y Human Neuroblastoma Cell reduced by Ab1-42 Oligomer treatment (Figure 25). Considering this result, the present invention is expected to increase phosphorylation of Thrl72 in AMPK Catalytic Subunit a reduced by Ab to recover AMPK activity to activate Autophagy to remove misfolded Protein Aggregates related to degenerative brain diseases and to inhibit neuronal degeneration.

Example 22. Changes In Expression Of Autophagy Markers, Including LC3B And P62, By The Present Invention In Retinoic Acid Differentiated SH- SY5Y Human Neuroblastoma Cell

Example 22 -1. Cell culture method of SH-SY5Y neurons

Example 22 -1-1. Cell Subculture

Using the method described in Example 2-1-1, cell subculture was conducted.

Example 22 -1-2. SH-SY5Y Neuroblastoma Differentiation

Using the method described in Example 2-1-2, SH-SY5Y neuronal cells were differentiated.

Example 22-2. Method For Preparation Of Ab And Treatment With The Present Invention

Prepared a 1 mM solution of human Ab1-42 (Abeam) in DMEM/F12 (+ 1% FBS + 1% Penicillin/Streptomycin). Solution of Ab was left at 37 °C for 3 hours to form Ab Oligomer. The existing medium was discarded and the cells were treated with newly prepared Ab1-42 Oligomer (1 mM) solution with or without the present invention (maximum concentration 40 mM) and 3-MA (3-Methyladenine, Sigma- Aldrich) (5 mM), Autophagy/PI3K (Phosphoinositide 3-Kinase) Inhibitor, and incubated under 5% CO2 at 37 °C for 72 hours. After 72 hours, the medium was removed and the cells were washed with PBS once and recovered. Using the following procedure, further experiments were carried out.

Example 22-3. Western Blot Analysis Method Using the method described in Example 6-5, Western Blot Analysis was carried out and the primary antibodies and it's conditions are provided in Table 13.

Table 13. List of Primary Antibodies

Example 22-4. Changes In Expression Of Autophagy Marker Including LC3B And P62 Resulting By The Present Invention In Retinoic Acid Differentiated SH-SY5Y Human Neuroblastoma Cell

The present invention not only increase expression of LC3B, an Autophagy Marker, reduced by Ab1-42 Oligomer in Retinoic Acid Differentiated SH-SY5Y Human Neuroblastoma Cell, but also induce Autophagic Flux (Conversion of LC3B-I to LC3B- II) to increase the LC3B-II/I Ratio at the same time, and reduced expression of Ubiquitin-Binding Protein p62 (SQSTM, Sequestosome 1), an Autophagosome Cargo Protein, increased by Ab1-42 Oligomer. Also, 3-MA, an Autohpagy/PI3K Inhibitor, was used together with the present invention, Autophagic Flux (LC3B-II/I Ratio) increased by the present invention was reduced and expression of p62 reduced by the present invention increased. Furthermore, APP formation and Ab1-42 accumulation increased when the cells treated with the present invention and 3-MA together (Figure 26). Considering this result, the present invention is expected to suppress APP formation and Ab1-42 accumulation by activating Autophagy.

Example 23. Change of expression of Autophagy Markers including ATG7 and ATG5/12 by the present invention in Alzheimer's disease animal model (5XFAD Transgenic Mice) Example 23-1 Alzheimer's Disease Animal Model (5XFAD Transgenic

Mice)

The animal model described in Example 13-1 was used. Example 23-2. Administration Of The Composition Of The Present

Invention

The composition was administered using the same method described in Example 13-2. Example 23-3. Preparation Of Tissue Sample

Using the method described in Example 13-3, tissue samples were prepared and stored.

Example 23-4. Western Blot Analysis Method

Using the method described in Example 6-5, Western Blot Analysis was carried out and the primary antibodies and it's conditions are provided in Table 14.

Table 14. List of Primary Antibodies

Example 23-5. Changes in Expression of Autophagy Markers including ATG7 and ATG5/12 By the Present invention in Alzheimer's disease animal model (5XFAD Transgenic Mice)

The presen tinvention increased expression of Autophagy Markers including ATG7 and ATG5/12 in hippocampus of Alzheimer's disease animal model (5XFAD Transgenic Mice) and increased the expression of ATG5/12 in cerebral cortex (Figure 27). Considering the result, the present invention is expected to increase Autoaphgic

Flux (LC3BII/I Ratio), an Autophagy Marker, reduce p62 (Figure 26), and increase expression of ATG7 and ATG5/12 to activate Autophagic Cascade. Example 24. Effect of improving cognitive and behavioral learning abilities in the Alzheimer's dementia animal model (NSE-hAPP-C105) for the present invention

Example 23-1. Experimental animals

The animal model described in Example 17- 1 was used.

Example 23-2. Method of administration of the present invention

The composition was administered using the same method described in Example 17-

2.

Example 23-3. Test Method For Cognitive Ability And Behavioral Learning

Ability

Example 23-3-1. Morris Water Maze Test

The present invention was administered by Intraperitoneal Injection once a day for 4 weeks at 4 mg/kg before measuring the change in cognitive ability using water maze test. The test was performed in 22-25 °C water in circular water bath (diameter 1 m x height 40 cm) wherein a target (Target: diameter 12 cm) was set up about 3 cm lower than the surface of the water. Dry milk power was added to the water to make the target invisible SMART 3.0 protram (Panlab) was installed on the ceiling above the water bath to monitor the test animals' time to reach the target (Latency to Target), swimming distance (Distance to Target) and pattern of swimming (Swimming Pattern) before the experiment and 4 weeks later. Test subject was trained for the first 5 days twice a day to begin from the starting point and to end at the target. Mice that did not find the target both times were made to recognize the position of target. Each test was performed with 5 minutes internval and the target was removed on day 6 to perform the test for 1 minute to begin from the starting point and the result was used as the experimental data.

Example 23-3-2. Passive Avoidance Test

The present invention was administered by Intraperitoneal Injection at 4 mg/kg dose once a day for 4 weeks before passive Avoidance Test was performed to evaluate memory. The Passive Avoidance Test is comprised of a front room that was bright room in white color (18 x 18 x 25 cm), the back room was a dark room in black color (18 x 18 x 25 cm), strong metal stainless was installed on the floor of the dark room and the walls of the front and back rooms have holes with 4 cm diameter, which open and close in guillotine type. Each testing animal was isolated in each cage for 1 minute, moved to the front room for 10 seconds to adjust while the guillotine type doors were opened to allow the test animals to move between the rooms freely. Once all four foots of the test animals are in the back room, the door was closed quickly and recorded the time for the test animal to move from the front room to back room (Initial Latency Time) and electrified (0.5 mA) for 2 seconds. After 5 seconds, the test animal was moved to its residential cage. After 72 hours, same test was performed to measure the Latency to Enter Dark, time to move from the front to back room till maximum 300 seconds.

Example 23-3-3. Data processing method

The collected data was process using SPSS 20.0 statistical program to calculate statistical error (Mean±SD), validation of variables among species and groups was performed by one way variance analysis (One-Way ANOVA). In the event if there is a significant different between the groups, post-verification was performed using Bonferroni method. For the purpose, hypiothetical acceptance is set as a=0.05.

Example 23-4. Improvement In Cognitive And Behavioral Learning Skills By The Present Invention In Alzheimer's Dementia Animal Model (NSE-hAPP-C105)

Example 23-4-1. Result of cognitive function Improvement by The Present Invention in Alzheimer's dementia animal model (NSE-hAPP-C105): Water Maze Test (Morris Water Maze Test)

Morris Water Maze Test was performed for C57BL/6-Tg(NSE-hAPP-C105)Kor Transgenic Mice to measure the effect of the present invention on cognitive function after 4 weeks of treatment. First, analysis of time to reach the target shows that it statistically increased in TC group than NTC group (P =0.001) and statistically reduced in TM group (P =0.001) than TC group. Second, analyzing the distance of swimming to reach the target, it was statistically increased in TC group (P =0.001) than NTC group, and statistically reduced in TM group (P = 0.001) than TC group. Third, analysis of the swimming time from target in the quadrant shows that it statistically reduced in TC group (P =0.001) than NTC group and statistically increased in TM group (P =0.001) than in TC group. Fourth, analysis of the times passing through the target shows that it statistically decreased in TC group (P =0.001) than NTC group and statistically increased in TM group (P=0.037) than TC group (Figure 28). Example 23-4-2. Result Of Improvement In Action Learning And

Cognitive Abilities By The Present Invention In Alzheimer's Dementia Animal Model (NSE-hAPP-C105): Passive Avoidance Test

Passive Avoidance Test was conducted with C57BL/6-Tg(NSE-hAPP-C105)Kor Transgenic Mice to evaluate the effect of the 4 weeks of treatment with the present invention on action learning and cognitive abilities (Figures 6 A and 6B). The result shows that it statistically decreased in TC group(P=0.001) than NTC group, and statistically increased in TM group (P=0.004) than TC group (Figure 29) .