CN111171113B - Small molecular compound for specifically degrading tau protein and application thereof - Google Patents

Small molecular compound for specifically degrading tau protein and application thereof Download PDF

Info

Publication number
CN111171113B
CN111171113B CN201811329209.7A CN201811329209A CN111171113B CN 111171113 B CN111171113 B CN 111171113B CN 201811329209 A CN201811329209 A CN 201811329209A CN 111171113 B CN111171113 B CN 111171113B
Authority
CN
China
Prior art keywords
tau protein
compound
disease
protein
tau
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN201811329209.7A
Other languages
Chinese (zh)
Other versions
CN111171113A (en
Inventor
汪义朋
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Wang Yipeng
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Priority to CN201811329209.7A priority Critical patent/CN111171113B/en
Publication of CN111171113A publication Critical patent/CN111171113A/en
Application granted granted Critical
Publication of CN111171113B publication Critical patent/CN111171113B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06008Dipeptides with the first amino acid being neutral
    • C07K5/06017Dipeptides with the first amino acid being neutral and aliphatic
    • C07K5/06034Dipeptides with the first amino acid being neutral and aliphatic the side chain containing 2 to 4 carbon atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Veterinary Medicine (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Psychology (AREA)
  • Psychiatry (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Hospice & Palliative Care (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Peptides Or Proteins (AREA)

Abstract

The invention relates to the technical field of bifunctional molecular compounds, and discloses a small molecular compound for specifically degrading tau protein and application thereof. The chemical structure of the small molecular compound for specifically degrading tau protein is TBM-L-ULM or pharmaceutically acceptable salt, enantiomer, stereoisomer, solvate, polymorph or N-oxide thereof, wherein TBM is a tau protein binding part, L is a linker group, ULM is an ubiquitin ligase binding part, and the tau protein binding part and the ubiquitin ligase binding part are connected through the linker group. The small molecular compound for specifically degrading tau protein can enhance the degradation of tau protein in cells, thereby reducing the content of tau protein.

Description

Small molecular compound for specifically degrading tau protein and application thereof
Technical Field
The invention relates to the technical field of bifunctional molecular compounds, in particular to a small molecular compound (named JT 01) for specifically degrading tau protein and application thereof.
Background
Alzheimer's Disease (AD) is the most common type of dementia, accounting for about 50-70% of dementia. It is counted that about 1000 ten thousand in 2016 and 4400 ten thousand AD patients worldwide. With aging population, the incidence of AD will rise further, with the expectation that about 4000 tens of thousands of AD patients in china in 2050. All current drugs for treating AD are characterized drugs (Symptomatic drugs) which only temporarily relieve symptoms but cannot delay the progression of the disease. There is a strong global expectation of new drugs (diseases-modifying drugs) that can actually alter the progression of AD.
Two characteristic pathological changes in AD are Senile Plaques (SPs) and neurofibrillary tangles (Neurofibrillary tangles, NFTs), which are polymers formed from β -amyloid (aβ) and hyperphosphorylated tau protein, respectively, where tau lesions, but not aβ lesions, are positively correlated with the degree of dementia in AD. Recent studies have shown tau-mediated aβ -induced neurotoxicity, necessary for aβ neurotoxicity; and tau is a Prion-like protein (Prion-like protein) that can spread between neurons, resulting in the spread of tau lesions, suggesting that it may be a necessary drug target for AD.
In addition to AD, tau aggregation is also seen in a range of neurodegenerative diseases linked to frontotemporal dementia with parkinsonism (frontotemporal dementia linked to chromosome-17parkinsonism, ftdp-17), pick's Disease (PiD), progressive supranuclear palsy (progressive supranuclear palsy, PSP), corticobasal degeneration (corticobasal degeneration, CBD), primary age-related tauopathies (primary age-related tauopathy, PART), silverphilic granulosis (argyrophilic grain Disease, AGD), age-related tau astrocytopathy (sting-related tau astrogliopathy, ARTAG), chronic traumatic encephalopathy (chronic traumatic encephalopathy, CTE), globoid tauopathy (Globular glial tauopathy, GGT), parkinson's Disease (PD), huntington's Disease (HD), and the like. Such diseases include AD, which is collectively known as tauopathies (tauopathies). Tau protein is an important cause of this disease and is an important therapeutic target for this disease.
At present, although there have been various therapeutic proposals based on tau protein, one of the most attractive is to reduce the intracellular tau protein content. This solution is favored mainly for the following reasons: (1) A great deal of evidence suggests that decreasing intracellular tau protein levels causes little side effects in animal models; (2) Reducing the amount of tau protein inhibits tau protein aggregation, which is a significant cause of neuronal degeneration; (3) Reducing tau protein levels reduces neuronal excitatory neurotoxic effects caused by a variety of factors, such as aβ. Thus, reduction of tau protein is also considered a new potential treatment for epilepsy and stroke.
There are two common technical approaches to reduce intracellular target proteins. (1) The expression of the target protein is reduced with siRNA, miRNA or antisense oligonucleotides. Due to the poor distribution of these oligonucleotides in tissues, poor pharmacokinetics, combined with the possibility of off-target, their clinical use is currently limited and still requires further improvement. (2) enhancing degradation of the target protein. A common approach is to enhance the activity of protein degradation systems, including protease systems and autophagy systems. However, the activity of the protein degradation system is not specifically enhanced, and other non-target proteins are easily degraded to cause serious side effects, so that no medicine for activating the protein degradation system is clinically applied at present. It is desirable to selectively enhance only the degradation of the target protein while avoiding degradation of non-target proteins due to enhanced activity of the protein degradation system.
Disclosure of Invention
The aim of the invention is to construct a small molecule compound specific for tau proteins. To achieve this object, the inventors of the present invention have found through intensive studies that a bifunctional molecular compound capable of specifically binding a target protein at one end and specifically binding a specific ubiquitin ligase at the other end, which are linked via a linker group (linker), can be constructed using a protein degradation targeting chimera (PROteolysis TArgeting Chimeras, PROTAC) technology. The compound thus constructed can bind to the target protein and ubiquitin ligase simultaneously, and the target protein is adjacent to ubiquitin ligase, so that ubiquitination of the target protein can be enhanced, and finally degradation is carried out through a proteasome. Moreover, the protoc technique has the following advantages in addition to selectivity for target proteins: (1) can act on a plurality of targets which are difficult to prepare in traditional medicines. Many traditional small molecule drugs must act on specific binding pockets (binding pockets) of the target protein to exert their inhibitory effect. The PROTAC technique is not limited in that it can interact with any segment of the target protein, and does not require a high affinity, and can cause rapid degradation of the target protein, thereby inhibiting the function of the target protein, and thus can act on many targets that are conventionally difficult to prepare. (2) The PROTAC technology can be repeatedly used in cells, and can play a similar role in catalysis, so that the treatment effect can be achieved without high concentration.
To this end, the present invention provides a small molecule compound (i.e. JT 01) for specifically degrading tau protein, wherein the chemical structure of the compound is TBM-L-ULM or a pharmaceutically acceptable salt, enantiomer, stereoisomer, solvate, polymorph or N-oxide thereof, wherein TBM is a tau protein binding moiety, L is a linker group, ULM is an ubiquitin ligase binding moiety, and the tau protein binding moiety is linked to the ubiquitin ligase binding moiety via a linker group.
In a preferred embodiment, the ULM is capable of binding to E3 ubiquitin ligase.
Further preferably, the E3 ubiquitin ligase is VHL E3 ubiquitin ligase or CRBN E3 ubiquitin ligase.
In a preferred embodiment, ULM is a group having the structure shown in formula (1),
Figure GDA0004124020990000031
wherein R is 1 Is a hydroxyl group or a group that is metabolized to a hydroxyl group in a patient or subject;
R 2 is-NH-R 4 Ar-HET wherein R 4 Is a C1-C3 alkylene group, ar is a C6-C12 arylene group, HET is an optionally substituted thiazole or isothiazole;
R 3 is-CHR 5 -M-, ULM is linked to the linker group L through M, wherein R 5 Is C1-C4 alkyl, M is a bond, C1-C4 alkylene, -NH-or-NH-C (O) -R 6 -, wherein R is 6 Is a C1-C4 alkylene group.
In a preferred embodiment, L is a group-X-Y-Z-, X is attached to TBM, Z is attached to ULM, wherein X is a bond, C1-C4 alkylene, -NH-or-NH-C (O) -R 7 -, wherein R is 7 Is a bond or a C1-C4 alkylene group;
y is-R 8 -(R 10 -E-R 11 ) n -R 9 -, wherein R is 8 And R is 9 Each is a bond or C1-C8 alkylene, R 10 And R is 11 Each is a C1-C4 alkylene group, n is an integer from 0 to 10, E is O, S, amido, piperazinyl, NR 12 、S(O)、S(O) 2 、-S(O) 2 O、-OS(O) 2 、OS(O) 2 O、
Figure GDA0004124020990000041
Figure GDA0004124020990000042
Wherein E is 1 O, S, CHR of a shape of O, S, CHR 12 Or NR (NR) 12 ,R 12 Is H or C1-C3 alkyl optionally substituted with one or two hydroxy groups;
z is-A-B-, wherein A is a bond, O or S, B is a bond, C1-C4 alkylene or-R 13 -C (O) -, wherein R 13 Is a C1-C4 alkylene group.
In a preferred embodiment, the TBM is a group having a structure represented by formula (2), or a group in which the (1), (2), (3), (4), (5), (6), (7), (8), (9) or the position(s) in the group represented by formula (2) is further modified by a substituent, wherein the TBM is linked to the linker group L via the (1), (2), (3), (4), (5), (6), (7), (8), (9) or the position(s) in formula (2),
Figure GDA0004124020990000051
wherein R is 14 Is C1-C4 alkylene, R 15 And R is 16 Each is C1-C4 alkyl, R 17 Is a bond, H, C1-C4 alkyl or-R 18 -C (O) -, wherein R 18 Is a C1-C4 alkylene group.
In a preferred embodiment, the small molecule compound that specifically degrades tau protein has the structural formula:
Figure GDA0004124020990000052
the invention also provides a method of degrading tau protein in a patient in need thereof comprising administering to said patient an effective amount of a small molecule compound as described above.
In a preferred embodiment, the small molecule compound is administered to the patient by at least one means selected from the group consisting of: nasal, inhalation, topical, oral, intramuscular, subcutaneous, transdermal, intraperitoneal, epidural, intrathecal and intravenous routes.
The invention also provides application of the small molecule compound in preparing a medicament for treating or preventing diseases related to tau protein.
Alternatively, the Disease is at least one of Alzheimer's Disease (AD), frontotemporal dementia (Frontotemporal dementia linked to chromosome-17park insonism, FTDP-17), pick's Disease (PiD), progressive supranuclear palsy (progressive supranuclear palsy, PSP), corticobasal degeneration (corticobasal degeneration, CBD), primary age-related tauopathy (primary age-related tauopathy, PART), silver-philic granulomatosis (argyrophilic grain Disease, AGD), aging-related tau astrocytopathy (imaging-related tau astrogliopathy, ARTAG), chronic traumatic encephalopathy (chronic traumatic encephalopathy, CTE), spherical glial tauopathy (Globular glial tauopathy, GGT), parkinson's Disease (PD), huntington's Disease (HD), cerebral stroke, and epilepsy.
Immunoblotting experiments prove that the small molecular compound for specifically degrading tau protein can enhance the degradation of tau protein in cells, thereby reducing the content of tau protein. Thus, it is shown that the small molecule compounds of the invention that specifically degrade tau protein can play a role in the prevention and treatment of a range of tau diseases including Alzheimer's disease.
Drawings
FIG. 1 is a nuclear magnetic resonance spectrum of a small molecular compound prepared in example 1 of the present invention;
FIG. 2 is a comparison of tau protein degradation using different concentrations of small molecule compounds of the present invention;
FIG. 3 is a graph of quantitative analysis of tau protein degradation using different concentrations of small molecule compounds of the present invention;
FIG. 4 shows a fluorescence photograph of small molecule compounds of the invention that reduce total tau protein content in HEK293 tau cells.
Detailed Description
The endpoints and any values of the ranges disclosed herein are not limited to the precise range or value, and are understood to encompass values approaching those ranges or values. For numerical ranges, one or more new numerical ranges may be found between the endpoints of each range, between the endpoint of each range and the individual point value, and between the individual point value, in combination with each other, and are to be considered as specifically disclosed herein.
The chemical structure of the small molecular compound for specifically degrading tau protein is TBM-L-ULM or pharmaceutically acceptable salt, enantiomer, stereoisomer, solvate, polymorph or N-oxide thereof, wherein TBM is a tau protein binding part, L is a connector group, ULM is an ubiquitin ligase binding part, and the tau protein binding part and the ubiquitin ligase binding part are connected through the connector group.
Preferably, the ULM is capable of binding to E3 ubiquitin ligase. Further preferably, the E3 ubiquitin ligase is VHL E3 ubiquitin ligase or CRBN E3 ubiquitin ligase.
In a more preferred embodiment, ULM is a group having the structure shown in formula (1),
Figure GDA0004124020990000071
wherein R is 1 Is a hydroxyl group or a group that is metabolized to a hydroxyl group in a patient or subject;
R 2 is-NH-R 4 Ar-HET wherein R 4 Is a C1-C3 alkylene (e.g., methylene, ethylene, or propylene), ar is a C6-C12 arylene (e.g., phenyl or naphthyl), HET is an optionally substituted thiazole or isothiazole;
R 3 is-CHR 5 -M-, ULM is linked to the linker group L through M, wherein R 5 Is a C1-C4 alkyl group (e.g., methyl, ethyl, propyl or butyl), M is a bond, a C1-C4 alkylene group (e.g., methylene, ethylene, propylene or butylene), -NH-or-NH-C (O) -R 6 -, wherein R is 6 Is a C1-C4 alkylene group (e.g., methylene, ethylene, propylene, or butylene). Wherein ULM is linked to the linker group L by "-M-". When "-M-" is-NH-C (O) -R 6 -at the time, "-R 6 - "is directly linked to the linker group L.
Further preferably, in the structure shown in the formula (1), R 1 Is hydroxyl; r is R 2 is-NH-R 4 Ar-HET wherein R 4 Is methylene, ar is phenyl; r is R 3 is-CHR 5 -NH-or-CHR 5 -NH-C(O)-R 6 -, where R is 5 is-C (CH) 3 ) 3 ,R 6 Is methylene.
In a more preferred embodiment, L is a group-X-Y-Z-, X is attached to TBM, Z is attached to ULM,
wherein X is a bond, C1-C4 alkylene (e.g., methylene, ethylene, propylene or butylene), -NH-or-NH-C (O) -R 7 -, wherein R is 7 Is a bond or a C1-C4 alkylene group (e.g., methylene, ethylene, propylene, or butylene);
y is-R 8 -(R 10 -E-R 11 ) n -R 9 -, wherein R is 8 And R is 9 Each is a bond or a C1-C8 alkylene group (e.g., methylene, ethylene, propylene, butylene, pentylene, hexylene, heptylene or octylene), R 10 And R is 11 Alkylene (e.g. methylene, ethylene, propylene or butylene) each having 1 to 4 carbon atoms, n is an integer from 0 to 10 (e.g. 0, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10), E is O, S, amido, piperazinyl, NR 12 、S(O)、S(O) 2 、-S(O) 2 O、-OS(O) 2 、OS(O) 2 O、
Figure GDA0004124020990000081
Figure GDA0004124020990000082
Wherein E is 1 O, S, CHR of a shape of O, S, CHR 12 Or NR (NR) 12 ,R 12 Is H or C1-C3 alkyl optionally substituted with one or two hydroxy groups;
z is-A-B-, wherein A is a bond, O or S, B is a bond, C1-C4 alkylene (e.g., methylene, ethylene, propylene or butylene), or-R 13 -C (O) -, wherein R 13 Is a C1-C4 alkylene group (e.g., methylene, ethylene, propylene, or butylene).
In a more preferred embodiment, the TBM is a group having a structure represented by formula (2), or a group in which the (1), (2), (3), (4), (5), (6), (7), (8), (9) or the position(s) in the group represented by formula (2) is further modified by a substituent, wherein the TBM is linked to the linker group L through the (1), (2), (3), (4), (5), (6), (7), (8), (9) or the position(s) in formula (2),
Figure GDA0004124020990000091
wherein R is 14 Is C1-C4 alkylene (e.g. methylene, ethylene, propylene or butylene), R 15 And R is 16 Each C1-C4 alkyl (e.g., methyl, ethyl, propyl or butyl), R 17 Is a bond, an alkyl group of H, C1-C4 (e.g. methyl, ethyl, propyl or butyl) or-R 18 -C (O) -, wherein R 18 Is a C1-C4 alkylene group (e.g., methylene, ethylene, propylene, or butylene).
Wherein, the substituent group at the (1), (2), (3), (4), (5), (6), (7), (8), (9) or the position in the group of the modified structure shown in the formula (2) can be halogen (such as fluorine or chlorine), C1-C4 alkyl (such as methyl, ethyl, propyl or butyl), C1-C4 alkoxy (such as methoxy, ethoxy, propoxy or butoxy), carboxyl, amino, aryl (such as phenyl) of C6-C18 or benzyl.
Further preferably, in the structure represented by formula (2), TBM is bonded to the linker group L via the (5) position in formula (2), R 14 Is ethylene, R 15 And R is 16 Each is methyl, R 17 Is a bond, methylene or-CH 2 -C(O)-。
In a most preferred embodiment, the small molecule compound that specifically degrades tau protein has the structural formula:
Figure GDA0004124020990000101
the small molecule compound for specifically degrading tau protein can be prepared according to the following process route:
Figure GDA0004124020990000102
/>
Figure GDA0004124020990000111
the preparation process comprises the following steps:
(1) Preparation of Compound B
Dissolving the above compound A in N, N-Dimethylacetamide (DMA) and then in N 2 Potassium acetate (KOAc), 4-methylthiazole and lead acetate (Pd (AcO)) were added under protection 2 ). The suspension was degassed under vacuum and replaced with hydrogen. The reaction suspension was then stirred and stirred under N 2 Reflux under heating overnight. After the reaction was completed, the reaction solution was filtered, and the filtrate was concentrated to dryness, followed by purification through a silica gel column to give compound B (yellow oil).
(2) Preparation of Compound C
Dissolving Compound B in methanol (MeOH) and NH 4 In OH, then in N 2 Raney nickel (ReneyNi) was added under protection. The suspension was degassed under vacuum and replaced with hydrogen. The reaction suspension was then taken up in H 2 Stirred overnight at room temperature. After the completion of the reaction, the reaction solution was filtered, and the filtrate was concentrated to dryness, followed by purification through a silica gel column to give compound C (pale yellow oil).
(3) Preparation of Compound D
Dissolving compound C in N, N-Dimethylformamide (DMF), in N 2 N, N-Diisopropylethylamine (DIPEA) and Boc- (2S, 4R) -4-hydroxy-2-pyrrolidinecarboxylic acid are added under protection. Then, after the reaction solution was cooled to 0 to 5 ℃,2- (7-benzotriazol-oxide) -N, N' -tetramethyluronium Hexafluorophosphate (HATU) was added. Then the reaction solution is put in N 2 Stir overnight at room temperature under protection. After completion of the reaction, the reaction mixture was poured into ice water, extracted with Ethyl Acetate (EA), and the organic phase was washed with brine. The organic phases were combined, dried over anhydrous sodium sulfate and concentratedContracted and then purified by a silica gel column to obtain a compound D.
(4) Preparation of Compound E
Compound D was dissolved in Dichloromethane (DCM) and trifluoroacetic acid (TFA) was added. The reaction solution was stirred at room temperature for reaction. After the completion of the reaction, concentration was carried out to obtain compound E.
(5) Preparation of Compounds 4-7
Compound E was dissolved in DMF at N 2 DIPEA, N-Boc-L-tert-leucine was added under protection. Then, after the reaction solution was cooled to 0 to 5 ℃, HATU was added. Then the reaction solution is put in N 2 Stir overnight at room temperature under protection. After completion of the reaction, the reaction mixture was poured into ice water, extracted with EA, and the organic phase was washed with brine. The organic phases were combined, dried over anhydrous sodium sulfate and concentrated, and then purified by a silica gel column to obtain compounds 4 to 7.
(6) Preparation of Compounds 4-8
Compound 4-7 was dissolved in DCM and TFA was added. The reaction solution was stirred at room temperature for reaction. Concentrating after the reaction is completed to obtain the compound 4-8.
(7) Preparation of Compounds 4-9
Compounds 4-8 were dissolved in DMF, N 2 DIPEA and 2, 2-dimethyl-4-oxo-3,8,11,14-tetraoxa-5-azahexadecane-16-oleic acid were added under protection. Then, after the reaction solution was cooled to 0 to 5 ℃, HATU was added. Then the reaction solution is put in N 2 Stir overnight at room temperature under protection. After completion of the reaction, the reaction mixture was poured into ice water, extracted with EA, and the organic phase was washed with brine. The organic phases were combined, dried over anhydrous sodium sulfate, concentrated, and purified by a silica gel column to obtain compounds 4 to 9.
(8) Preparation of Compounds 4-10
Compounds 4-9 were dissolved in DCM and EA/HCl was added. The reaction solution was stirred at room temperature for reaction. Concentrating after the reaction is completed to obtain the compound 4-10.
(9) Preparation of Compounds 4-12
Compound 3-3 and sarcosine were dissolved in DMF and Triethylamine (TEA) was added. Then the reaction solution is put in N 2 Stir overnight under protection. After the reaction was completed, the reaction mixture was poured into ice water, extracted with EA, and the organic phase was washed with brine. The organic phases were combined, dried over anhydrous sodium sulfate and concentrated. The crude product was purified by silica gel chromatography to give compounds 4-12.
(10) Preparation of Compound 4 (target Compound)
Compounds 4-12 were dissolved in DMF and taken up in N 2 DIPEA and Compounds 4-10 were added under protection. Then, after the reaction solution was cooled to 0 to 5 ℃, HATU was added. Then the reaction solution is put in N 2 Stir overnight at room temperature under protection. After completion of the reaction, the reaction mixture was poured into ice water, extracted with EA, and the organic phase was washed with brine. The organic phases were combined, dried over anhydrous sodium sulfate, concentrated, and purified by a silica gel column to give compound 4 (hygroscopic white foamy solid).
The invention also provides a method of degrading tau protein in a patient in need thereof comprising administering to said patient an effective amount of a compound as described above provided by the invention.
In the above method, the compound may be administered to the patient by at least one means selected from the group consisting of: nasal, inhalation, topical, oral, intramuscular, subcutaneous, transdermal, intraperitoneal, epidural, intrathecal and intravenous routes.
The invention also provides application of the compound in preparing a medicament for treating or preventing tau protein related diseases. The disease may be at least one of alzheimer's disease, frontotemporal dementia linked to chromosome 17 with parkinson's disease, pick's disease, progressive supranuclear palsy, corticobasal degeneration, primary age-related tauopathy, silver-philic granulomatosis, aging-related tauastrocytopathy, chronic traumatic encephalopathy, spherical glial tauopathy, parkinson's disease, huntington's disease, stroke, and epilepsy.
The present invention will be described in detail by examples.
Examples
(1) Preparation of Compound B
Compound A (1.82 g,10 mmol) was dissolved in 25mL DMA and then under N 2 KOAc (1.98 g,20 mmol), 4-methylthiazole (1.5 g,15 mmol) and Pd (AcO) were added under protection 2 (220 mg,1 mmol). The suspension was degassed under vacuum and replaced three times with hydrogen. The reaction suspension was then stirred and stirred under N 2 Reflux under heating overnight. The reaction was confirmed to be complete by thin layer chromatography (Thin Layer Chromatography, TLC). The reaction solution was filtered, and the filtrate was concentrated to dryness and then purified by a silica gel column to give 1.5g of compound B as a yellow oil.
(2) Preparation of Compound C
Compound B (300 mg,1.498 mmol) was dissolved in 10mLMeOH and 1mLNH 4 In OH, then in N 2 200mg of Raney nickel (ReneyNi) are added under protection. The suspension was degassed under vacuum and replaced three times with hydrogen. The reaction suspension was then taken up in H 2 Stirred overnight at room temperature. Reaction completion was confirmed by TLC (DCM/ea=1:1). The reaction solution was filtered, and the filtrate was concentrated to dryness and then purified by a silica gel column to give 110mg of compound C as a pale yellow oil.
(3) Preparation of Compound D
Compound C (110 mg,0.538 mmol) was dissolved in 3mL DMF at N 2 DIPEA (200 mg,1.547 mmol) and Boc- (2S, 4R) -4-hydroxy-2-pyrrolidinecarboxylic acid (131 mg,0.566 mmol) were added under protection. After the reaction solution was cooled to 0 to 5 ℃, HATU (245 mg,0.644 mmol) was added. Then the reaction solution is put in N 2 Stirred overnight at room temperature. Reaction completion was confirmed by TLC (DCM/ea=1:1). The reaction mixture was poured into ice water, extracted with EA, and the organic phase was washed with brine. The organic phases were combined, dried over anhydrous sodium sulfate and concentrated, then purified by a silica gel column to obtain 100mg of compound D.
(4) Preparation of Compound E
Compound D (100 mg,0.239 mmol) was dissolved in 3mL DCM and trifluoroacetic acid (TFA) (0.5 mL,6.73 mmol) was added. The reaction solution was stirred at room temperature for 1 hour. Reaction completion was confirmed by TLC (EA/meoh=9:1) and concentrated by high vacuum pump to give 220mg of crude compound E.
(5) Preparation of Compounds 4-7
Compound E (220 mg, about0.239 mmol) in 3mL DMF at N 2 DIPEA (300 mg,2.320 mmol) and N-Boc-L-tert-leucine (58 mg,0.251 mmol) were added under protection. Then after the reaction solution was cooled to 0 to 5℃HATU (110 mg,0.289 mmol) was added. Then the reaction solution is put in N 2 Stir overnight at room temperature under protection. Reaction completion was confirmed by TLC (PE/ea=1:1). The reaction mixture was poured into ice water, extracted with EA, and the organic phase was washed with brine. The organic phases were combined, dried over anhydrous sodium sulfate and concentrated, and then purified by a silica gel column to obtain 103mg of compound 4-7.
(6) Preparation of Compounds 4-8
Compound 4-7 (103 mg,0.194 mmol) was dissolved in 3mL DCM and TFA (0.5 mL, mmol) was added. The reaction solution was stirred at room temperature for 1 hour. Completion of the reaction was confirmed by TLC (PE/ea=1:1) and concentrated by a high vacuum pump to give 85mg of compound 4-8.
(7) Preparation of Compounds 4-9
Compound 4-8 (85 mg,0.194 mmol) was dissolved in 3mL DMF, N 2 DIPEA (300 mg,2.320 mmol) and 2, 2-dimethyl-4-oxo-3,8,11,14-tetraoxa-5-azahexadecane-16-oleic acid (63 mg,0.205 mmol) were added under protection. Then, after the reaction solution was cooled to 0 to 5 ℃, HATU (89 mg,0.234 mmol) was added. Then the reaction solution is put in N 2 Stir overnight at room temperature under protection. Reaction completion was confirmed by TLC (EA/meoh=9:1). The reaction mixture was poured into ice water, extracted with EA, and the organic phase was washed with brine. The organic phases were combined, dried over anhydrous sodium sulfate, concentrated, and purified by a silica gel column to obtain 75mg of compound 4-9.
(8) Preparation of Compounds 4-10
Compound 4-9 (75 mg,0.104 mmol) was dissolved in 3mL DCM and EA/HCl (0.5 mL, 2N) was added. The reaction solution was stirred at room temperature for 1 hour. Completion of the reaction was confirmed by TLC (EA/meoh=9:1) and concentrated by high vacuum pump to give 103mg (0.104 mmol) of compound 4-10.
(9) Preparation of Compounds 4-12
Compound 3-3 (150 mg, 0.390 mmol) and sarcosine (174 mg,1.953 mmol) were dissolved in 15mL DMF and TEA (400 mg,3.943 mmol) was added. ThenThe reaction solution was stirred under N 2 Stir overnight at 90 ℃ under protection. Reaction completion was confirmed by TLC (EA/dcm=1:1 and EA/meoh=9:1). The reaction mixture was poured into ice water, extracted with EA, and the organic phase was washed with brine. The organic phases were combined, dried over anhydrous sodium sulfate and concentrated. The crude product was purified by silica gel chromatography to give compound 4-12 (63 mg,0.145 mmol).
(10) Preparation of Compound 4 (target Compound)
Compound 4-12 (63 mg,0.145 mmol) was dissolved in 5mL DMF and taken up in N 2 DIPEA (200 mg,1.547 mmol) and compound 4-10 (103 mg,0.104 mmol) were added under protection. Then, after the reaction solution was cooled to 0 to 5 ℃, HATU (48 mg,0.236 mmol) was added. Then the reaction solution is put in N 2 Stir overnight at room temperature under protection. Reaction completion was confirmed by TLC (EA/meoh=9:1). The reaction mixture was poured into ice water, extracted with EA, and the organic phase was washed with brine. The organic phases were combined, dried over anhydrous sodium sulfate, concentrated and purified on a silica gel column to give 69mg of compound 4 as a hygroscopic white foam solid. The nuclear magnetic resonance spectrum of the compound is shown in figure 1. HPLC purity: 97.8% (254 nm); mass: m/z 1036[ M+1 ]] +
Test case
Cell culture and drug treatment HEK293 tau cells (stably expressing wild-type full-length tau protein) were incubated in DMEM medium for 24h, after which different concentrations of the small molecule compounds prepared as described in the examples above (0 μm,0.01 μm,0.1 μm,1 μm,10 μm,20 μm) were added and incubation was continued for 24h. The cells were lysed with RIPA solution and the cell extracts were collected, the proteins in the cell extracts were separated by 10% polyacrylamide gel electrophoresis (PAGE), immunoblotted with anti-total tau antibody (tau 5), and the tau content was quantified. The preparation of the sample, the measurement of the protein content of the sample and the detailed operation of the immunoblotting method are described below.
1. Preparation of samples
(1) Observing the growth condition and density of cells in the cell culture plate under a microscope;
(2) Pre-cooling the 1 XPBS and the cell culture plate on ice, adding 80 μl/6cm dish per well of the 6-well plate, and adding 150 μl to prepare a 1 XPBS+PMSF (1:100) +proteinase mixed inhibitor (1:1000);
(3) Sucking out the culture medium by using a 1ml sample gun to cling to the bottom wall of a cell culture plate, adding 1ml of 1 XPBS to cling to the wall, gently flushing for 1-3 times according to the cell density, sucking out the PBS by using 1ml and 200 mu l of sample guns to cling to the bottom wall of the cell culture plate, adding the mixed solution, then cleaning the cell scraper by using ultrapure water, slightly scraping the bottom of the pore plate by using the cell scraper, sucking out the cell suspension to a 1.5ml EP tube, and cleaning the cell scraper between different samples;
(4) Boiling for 10min (boiling at maximum temperature and then maintaining boiling at 150deg.C, opening antioxidant epitope);
(5) After centrifugation, 20kHz ultrasonic wave is carried out for 20 times (in order to open a DNA chain, an ultrasonic machine is opened and closed according to I, after each time one sample is exceeded, the ultrasonic machine is firstly wiped by toilet paper in double distilled water for three times, and an ultrasonic gun head cannot contact air bubbles and the bottom of an EP tube);
(6) Mixing under shaking, and placing in a refrigerator at-20deg.C.
2. Determination of sample protein content (BCA method)
(1) After shaking the protein sample, properly diluting (5 μl of each sample is mixed with 45 μl of double distilled water for dilution, 2 parallel samples are respectively arranged), and centrifuging and shaking;
(2) Six standard tubes were used, and 0. Mu.l, 10. Mu.l, 20. Mu.l, 30. Mu.l, 40. Mu.l, 50. Mu.l of BSA (100 mg BSA in 5ml double distilled water) was prepared by adding 1000. Mu.l, 990. Mu.l, 980. Mu.l, 970. Mu.l, 960. Mu.l, 950. Mu.l double distilled water to each of the samples, and 0. Mu.g/. Mu.l, 0.2. Mu.g/. Mu.l, 0.4. Mu.g/. Mu.l, 0.6. Mu.g/. Mu.l, 0.8. Mu.g/. Mu.l, 1.0. Mu.g/. Mu.l of standard protein was prepared;
(3) The diluted protein sample and the diluted standard protein were added to 96-well plates (5. Mu.l/well, each at the junction of the wall and bottom of the PCR gun, with each well replaced by a gun head) and 3 parallel wells were provided.
(4) The working solution is prepared from solution A (self-collar) and solution B (public) in the kit according to the proportion of 50:1. Adding working solution into a 96-well plate, quickly suspending and adding 95 μl into each well, covering a cover after adding, quickly attaching to the bottom, vibrating in the same direction, and incubating for 30min at 37deg.C without touching the bottom of the 96-well plate with hands;
(5) Removing bubbles by using a 1ml syringe needle, opening a BioTek switch, opening Gen5, clicking an icon with a left arrow, clicking OK, and deriving Excel;
(6) Standard protein OD values were replicated, framed OD values and standard protein concentrations were inserted into the scatter plots. Selecting data points, adding trend lines, displaying a formula, displaying an R square value (at least 29 after decimal points), and removing abnormal values. And copying the OD value of the sample protein, inputting the corresponding sample group number above, and removing abnormal values.
3. tau protein assay (immunoblotting)
1. Set up rack (two glass plates, three bottles, five reagents, filter paper, toilet paper, dustbin, gun head, comb teeth)
(1) Cleaning the table top and the underframe, cleaning comb teeth, a glass plate, a distilled water bottle, an upper rubber bottle and a lower rubber bottle, drying the upper rubber bottle and the lower rubber bottle, taking out a reagent for preparing the electrophoresis gel, and recovering to room temperature;
(2) The higher glass plates are folded inwards, the upper part is pressed to enable the lower part to be clung to the tabletop to be flush, the clamp is outwards turned and clamped, and the clamp is placed on the underframe to be buckled.
(3) And (5) injecting double distilled water to check whether liquid leaks, and if so, reinstalling and then detecting the leakage.
2. Preparation of electrophoresis gel (see Table 1, AP and TEMED can be added 1.5 times during the day cold)
TABLE 1
Figure GDA0004124020990000191
(1) Sequentially adding 20% arc/Bis, tris buffer solution, TEMED and 10% AP, and blowing and uniformly mixing by a liquid shifter, wherein the whole process prevents bubbles from being mixed in the mixed solution;
(2) Slowly injecting the separation glue into the glue film along two corners (the glue is sucked deep under the liquid surface and gently blown and evenly mixed, a small amount of liquid is reserved at each gun head to prevent bubbles), the dosage of each glue is 3 multiplied by 900 mu l, gaps of the glue film are filled with double distilled water along the two corners after the glue is not leaked, and the oxygen inhibition polymerization is prevented and the glue at the lower part is kept flat for a period of time;
(3) After the gel is solidified for about 30min, pouring out double distilled water, sucking the residual water by using filter paper, and marking the upper edge of the lower gel by using a marker;
(4) Slowly injecting concentrated gel into the adhesive film along two corners, obliquely inserting comb teeth (sample amount is smaller than 20 μl and larger comb teeth are larger than 20 μl) of required specification from left to right, and feeding gel between lanes to avoid gel shrinkage, and waiting for gel for 50min.
3. Sample processing
After the BCA method determines the protein concentration, freshly prepared bromophenol blue and β -mercaptoethanol (reducing agent), bromophenol blue, are added: beta-mercaptoethanol=1:3, mixed liquor: sample = 1:10 Boiling water at 100deg.C for 10min, shaking on a shaker for 20 seconds, packaging, and if the result is bad, boiling water at 100deg.C for 10min before loading.
4. Loading and electrophoretic separation of proteins (loading needle, sample, row plug, marker, electrophoresis liquid, electrophoresis tank, distilled water bottle)
Washing the conductive wire below the electrophoresis frame, transferring to the electrophoresis frame, marking lanes and numbers by using a Marker pen, slowly and vertically pulling out comb teeth, filling a gel groove with electrophoresis liquid, adding each lane by using a microsampler sample (1 μl of Marker is added to lanes 1 and 8, and adding bromophenol blue and 1 Xbuffer mixed liquid to lane 15 for balancing). After the sample is applied, transferring the electrophoresis frame to an electrophoresis tank, adding electrophoresis liquid, then covering a cover to enable red to be red and black to be black, after the sample is applied, firstly, performing electrophoresis for about 30min by using constant current 10 mA/block glue (according to two times of starting), and when the bromophenol blue indicator is electrophoresed until the junction of the concentrated glue and the separation glue is linear, changing the electrophoresis into constant voltage 100V (if the constant voltage can be adjusted to high current) electrophoresis for about 60min until bromophenol blue reaches the bottom of the gel, and completely running the Marker.
5. Transfer film (NC film, transfer film liquid, filter paper, ice box, basin, dish, transfer film groove, plastic plate, cleaning forceps)
(1) The NC film is marked by a marker pen and then immersed in the recovered film transferring liquid for 10min-20min (which is beneficial to fixing protein, balancing gel and removing SDS), the gel groove is taken down by pressing the bayonets at two sides, and the glass plate and the middle part of the right side of the white porcelain plate are pried by a small plate, so that the electrophoresis of the residual gel is kept during the period.
(2) Cutting the gel with glass plate vertically slightly inclined and slightly left and right according to the molecular weight range to be displayed, sticking three layers of filter paper immersed in the transfer membrane liquid on the gel with tweezers, carefully prying the gel on a sponge (with the filter paper facing downwards) with a small plate, sticking an inverted NC membrane on the other side, immersing the gel and the NC membrane in the transfer membrane liquid (with the gel on) to remove bubbles with a glass rod, carefully holding the gel on the hand (with the gel on) with tweezers, sticking the three layers of filter paper immersed in the transfer membrane liquid on the gel with tweezers, reversely putting the sponge, and sticking three layers of filter paper. Placing a black plastic plate, a layer of sponge, three layers of filter paper, glue, an NC film, three layers of filter paper, a layer of sponge and a transparent plastic plate from bottom to top, and fixing the transparent plastic plate by using rubber bands if the transparent plastic plate is not tight.
(3) The red, white and black are put into ice bath (gel is not required to be soaked in the film transferring liquid for a long time before electrifying, so that protein is prevented from being diffused and decomposed), the transferring current is constant current 276mA, the voltage is generally 140V (methanol can be properly supplemented to improve the voltage), the specific transferring time is determined according to the molecular weight of the protein to be transferred, the time is 1h when the molecular weight of the protein to be transferred is less than 100kDa, and the time is 1.5h when the molecular weight of the protein to be transferred is more than 100 kDa.
6. Immunoblotting (cleaning tweezers, box with double distilled water, milk, fresh-keeping bag, toilet paper, primary antibody, ice box, plate, transparent adhesive tape, TBST, black plastic bag, secondary antibody)
(1) Closing: after completion of transfer, NC membrane was carefully blocked with TBS blocking solution containing 5% skimmed milk powder at room temperature for 1h or overnight at 4℃with shaking, and filter paper not contacted with gum was recovered.
(2) Incubation resistance: taking out NC film, rinsing with 1 XTBS to remove residual milk stain on the surface of the film, standing on toilet paper with forceps to remove excessive water, and placing NC film Marker side outside into fresh-keeping bag, and draining with toilet paper. A primary antibody (0.1% Tween 20 added to reduce background) was added and the seal was attached to the plate (Marker and protein side facing up), and the scotch tape was incubated overnight at 4℃without pressing against the target strip.
(3) Secondary antibody incubation: the next day the NC film was removed from the incubation bag and the primary antibody was recovered, rinsed with TBST buffer for 3X 5min, rinsed with 1 XTBS to remove residual salt ions on the surface of the film, and the NC film was placed in a fresh-keeping bag with forceps standing on toilet paper to remove excess water, and drained with toilet paper. Luciferin-labeled goat anti-rabbit or goat anti-mouse Odyssey secondary antibody (0.1% tween 20 can be added to reduce background) is added in the dark, the seal is attached to a flat plate (the side with Marker and protein is upward), scotch tape is not pressed on a target strip, incubation is carried out at room temperature for 1h (more than 1h can increase background, sky cold can be prolonged to 2 h), NC membrane is taken out from incubation bag, secondary antibody is recovered, and rinsing is carried out for 3×5min with TBST buffer. After the rinsing, the residual salt ions on the surface of the film were removed by rinsing with 1 XTBS.
(4) Color development: the glass plate is wiped clean by using mirror wiping paper dipped with absolute ethyl alcohol. The Marker side is downward, and the Marker is placed in the scanner from top to bottom according to the molecular weight from small to large, covered with a plastic film and exhausted. Opening Odyssey software, clicking File, new, browse, inputting date, clicking Obtain image to select the scanning film length and width, scanning, imaging and storing original image, clicking Alter image to adjust image, marking sample and antibody type by text box, clicking Export image File, save, collecting film, and wiping glass plate with toilet paper.
FIG. 2 is a comparison of tau protein degradation using different concentrations of the small molecule compound in the above test examples. FIG. 3 is a graph of quantitative analysis of tau protein degradation using different concentrations of the small molecule compounds in the above test examples. The results show that 1. Mu.M, 10. Mu.M, 20. Mu.M compounds can dose-dependently reduce the amount of tau protein in cells. FIG. 4 shows fluorescence pictures of the reduction of total tau protein content in HEK293 tau cells using different concentrations of small molecule compounds according to the invention.
It follows that the small molecule compounds of the invention which specifically degrade tau protein content in cells. Since abnormal accumulation of tau protein in cells is involved in more than 20 neurodegenerative diseases, the amount of accumulation of tau protein is positively correlated with neurodegeneration and memory impairment of these degenerative diseases, degradation of tau protein can achieve prevention or/and treatment of tau-related neurodegenerative diseases such as Alzheimer's disease, frontotemporal dementia linked to chromosome 17 with Parkinson's disease, pick's disease, progressive supranuclear palsy, corticobasal degeneration, primary age-related tauopathy, silver-philic granulomatosis, age-related tau astrocytopathy, chronic traumatic encephalopathy, globoid tauopathy, parkinson's disease, huntington's disease, cerebral stroke and epilepsy.
The preferred embodiments of the present invention have been described in detail above, but the present invention is not limited thereto. Within the scope of the technical idea of the invention, a number of simple variants of the technical solution of the invention are possible, including combinations of the individual technical features in any other suitable way, which simple variants and combinations should likewise be regarded as being disclosed by the invention, all falling within the scope of protection of the invention.

Claims (3)

1. A small molecule compound for specifically degrading tau protein, characterized in that the compound has the structural formula:
Figure FDA0004151753560000011
2. use of a small molecule compound according to claim 1 for the manufacture of a medicament for the treatment or prophylaxis of a tau protein associated disease.
3. The use according to claim 2, wherein the disease is at least one of alzheimer's disease, frontotemporal dementia linked to chromosome 17 with parkinson's disease, pick's disease, progressive supranuclear palsy, corticobasal degeneration, primary age-related tauopathy, silver-philic granulomatosis, age-related tau astrocytosis, chronic traumatic encephalopathy, glioblastoma tauopathy, parkinson's disease, huntington's disease, cerebral stroke and epilepsy.
CN201811329209.7A 2018-11-09 2018-11-09 Small molecular compound for specifically degrading tau protein and application thereof Active CN111171113B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201811329209.7A CN111171113B (en) 2018-11-09 2018-11-09 Small molecular compound for specifically degrading tau protein and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201811329209.7A CN111171113B (en) 2018-11-09 2018-11-09 Small molecular compound for specifically degrading tau protein and application thereof

Publications (2)

Publication Number Publication Date
CN111171113A CN111171113A (en) 2020-05-19
CN111171113B true CN111171113B (en) 2023-05-16

Family

ID=70623809

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201811329209.7A Active CN111171113B (en) 2018-11-09 2018-11-09 Small molecular compound for specifically degrading tau protein and application thereof

Country Status (1)

Country Link
CN (1) CN111171113B (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116964038A (en) * 2021-03-03 2023-10-27 汪义朋 Small molecule compounds that bind to tau proteins

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012125652A2 (en) * 2011-03-14 2012-09-20 University Of Southern California Antibody and antibody mimetic for visualization and ablation of endogenous proteins
CN104736569A (en) * 2012-01-12 2015-06-24 耶鲁大学 Compounds & methods for the enhanced degradation of targeted proteins & other polypeptides by an e3 ubiquitin ligase
WO2018102067A2 (en) * 2016-11-01 2018-06-07 Arvinas, Inc. Tau-protein targeting protacs and associated methods of use

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060067942A1 (en) * 2004-09-24 2006-03-30 Salama Zoser B Pharmaceutical agent comprising amino acids, peptides, proteins and/or fractions and fragments thereof and the use of same in the prophylaxis and treatment of immune system deficiency in humans and animals
AU2016294450A1 (en) * 2015-07-13 2017-12-07 Arvinas Operations, Inc. Alanine-based modulators of proteolysis and associated methods of use

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012125652A2 (en) * 2011-03-14 2012-09-20 University Of Southern California Antibody and antibody mimetic for visualization and ablation of endogenous proteins
CN104736569A (en) * 2012-01-12 2015-06-24 耶鲁大学 Compounds & methods for the enhanced degradation of targeted proteins & other polypeptides by an e3 ubiquitin ligase
WO2018102067A2 (en) * 2016-11-01 2018-06-07 Arvinas, Inc. Tau-protein targeting protacs and associated methods of use

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
多肽介导的泛素―蛋白酶体系统特异性降解Tau蛋白;Ting-Ting Chu等;《科学新闻》;20170425(第04期);第171页 *

Also Published As

Publication number Publication date
CN111171113A (en) 2020-05-19

Similar Documents

Publication Publication Date Title
ES2915833T3 (en) Isoindoline compositions and methods for treating neurodegenerative disease and macular degeneration
US11661580B2 (en) Method of inhibiting tau phosphorylation
Douchamps et al. A second wind for the cholinergic system in Alzheimer’s therapy
CN112047935B (en) Autophagy targeting protein degradation technology and application thereof
US20110171171A1 (en) Bridged secondary amines and use thereof as iap bir domain binding compounds
WO2016105468A1 (en) Derivatives of 3-heteroarylisoxazol-5-carboxylic amide useful for the treatment of inter alia cystic fibrosis
JP2007530701A (en) Heterocyclic CGRP antagonist for the treatment of migraine
CN115279751B (en) Small molecular compound for specifically degrading tau protein and application thereof
CN102300858A (en) N-{[(ir,4s,6r-3-(2-pyridinylcarbonyl)-3-azabicyclo [4.1.0] Hept-4-yl] Methyl}-2-heteroarylamine Derivatives And Uses Thereof
CN111171113B (en) Small molecular compound for specifically degrading tau protein and application thereof
CN103328442A (en) Azetidine derivatives useful for the treatment of metabolic and inflammatory diseases
WO2020093370A1 (en) Micromolecular compound specifically degrading tau protein, and application thereof
CN102458115B (en) Iap inhibitors
CN111518215A (en) Chimera for specifically degrading tau protein, encoding gene thereof and application thereof
CN115385908B (en) Small molecule chimera for targeted promotion of tau protein dephosphorylation and application thereof
WO2022104636A1 (en) Autophagic targeted protein degradation technique and use thereof
JPWO2007091613A1 (en) Alzheimer's disease preventive or therapeutic agent, and food and drink
CN114272258B (en) Application of circular RNA circUbe2cbp
CN115998736B (en) Application of Y-27632 in specific inhibition of hallucination
CN114957219B (en) Degradation agent for targeted degradation of Gli1 protein, preparation method and application thereof
Shi The Role of Apolipoprotein E in Regulating Tau Pathogenesis and Neurodegeneration in a Tauopathy Mouse Model
CN110694068A (en) Application of substance for inhibiting angiopoietin-like protein8
CN104245698B (en) Nitrogen-containing heterocycle compound
Aida et al. UBIQUITINATION IS ASSOCIATED WITH THE INTERNALIZATION OF BILE SALT EXPORT PUMP (BSEP/ABCB11) FROM THE PLASMA MEMBRANE: 768
CN116549632A (en) New application of anti-Lingo-1 antibody Opiciinumab and medicine for new application

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
TA01 Transfer of patent application right

Effective date of registration: 20200512

Address after: 201413 room 771, floor 1, building 1, No. 2908, xinsiping Road, Fengxian District, Shanghai

Applicant after: Shanghai qiangrei Biotechnology Co.,Ltd.

Address before: 201210 No. 27, Jiangdong Road, Zhangjiang Town, Pudong New Area, Shanghai

Applicant before: Wang Yipeng

TA01 Transfer of patent application right
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
TA01 Transfer of patent application right

Effective date of registration: 20230412

Address after: No. 27, Jiangdong Road, Zhangjiang Town, Pudong New Area, Shanghai, 201210

Applicant after: Wang Yipeng

Address before: 201413 room 771, floor 1, building 1, No. 2908, xinsiping highway, Fengxian District, Shanghai

Applicant before: Shanghai qiangrei Biotechnology Co.,Ltd.

TA01 Transfer of patent application right
GR01 Patent grant
GR01 Patent grant