CN113248484A - 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
CN113248484A
CN113248484A CN202010090680.6A CN202010090680A CN113248484A CN 113248484 A CN113248484 A CN 113248484A CN 202010090680 A CN202010090680 A CN 202010090680A CN 113248484 A CN113248484 A CN 113248484A
Authority
CN
China
Prior art keywords
substituted
unsubstituted
group
aliphatic ring
heterocyclic group
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.)
Pending
Application number
CN202010090680.6A
Other languages
Chinese (zh)
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.)
Shanghai Qiangrei Biotechnology Co ltd
Original Assignee
Shanghai Qiangrei Biotechnology Co ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Shanghai Qiangrei Biotechnology Co ltd filed Critical Shanghai Qiangrei Biotechnology Co ltd
Priority to CN202010090680.6A priority Critical patent/CN113248484A/en
Priority to CN202180012446.4A priority patent/CN115279751B/en
Priority to PCT/CN2021/075201 priority patent/WO2021160012A1/en
Publication of CN113248484A publication Critical patent/CN113248484A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/14Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/551Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having two nitrogen atoms, e.g. dilazep
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/08Antiepileptics; Anticonvulsants
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Neurosurgery (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Psychology (AREA)
  • Hospice & Palliative Care (AREA)
  • Psychiatry (AREA)
  • Pain & Pain Management (AREA)
  • Urology & Nephrology (AREA)
  • Vascular Medicine (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (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 molecule 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 a ubiquitin ligase binding part, and the tau protein binding part and the ubiquitin ligase binding part are connected through the connector group. The small molecule 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 for specifically degrading tau protein and application thereof.
Background
Alzheimer's Disease (AD) is the most common dementia, accounting for about 50-70% of dementia. Statistically, about 1000 million people in 2016 and 4400 million AD patients in the world. With the aging population, the incidence of AD will rise further, with an expectation that about 4000 million AD patients will be present in china by the year 2050. At present, all the drugs for treating AD are characteristic drugs (Symptomatic drugs), and can only relieve symptoms temporarily but cannot delay the progress of the disease. There is a global need for new drugs (Disease-modifying drugs) that can actually change the progress of AD.
Two characteristic pathological changes in AD are Senile Plaques (SPs) and Neurofibrillary tangles (NFTs), which are polymers of beta-amyloid (a β) and hyperphosphorylated tau protein, respectively, where tauopathy, but not a β pathology, is positively correlated with the degree of dementia in AD. Recent studies have shown that tau mediates a β -induced neurotoxicity, essential for a β neurotoxicity; and tau is a Prion-like protein (Prion-like protein) which can propagate among neurons to cause the spread of tauopathy, suggesting that it may be an essential drug target for AD.
In addition to AD, tau aggregation is also seen in frontotemporal dementia (frontotemporal dementia, FTDP-17) linked to chromosome 17 with Parkinson's Disease, Pick's Disease (PiD), Progressive Supranuclear Palsy (PSP), corticobasal degeneration (CBD), primary age-related tauopathy (PART), Argyrophilic Granulosis (AGD), age-related Astrocytosis (AG), chronic traumatic encephalopathy (Huntington), glial cell Disease (CTE), and Parkinson's Disease (HD), Parkinson's Disease, etc. Such diseases including AD are collectively referred to as tau diseases (tauopathies). Tau protein is an important cause of such diseases and is therefore an important therapeutic target for such diseases.
Currently, although there are a number of tau-based therapeutic proposals, one of the most attractive approaches is to reduce the amount of intracellular tau protein. This solution is favored mainly for the following reasons: (1) there is a great deal of evidence that reducing the level of intracellular tau protein causes fewer side effects in animal models; (2) reducing the content of tau protein can inhibit the aggregation of tau protein, which is an important reason for causing neuron degeneration; (3) reducing the level of tau protein reduces the effects of neuronal excitotoxicity caused by a number of factors, such as a β. Therefore, reducing tau protein is also considered as a new potential treatment for epilepsy and stroke.
There are two common techniques for reducing intracellular target proteins. (1) The expression of the target protein is reduced with siRNA, miRNA or antisense oligonucleotide. Because of the poor distribution of these oligonucleotides in tissues, the poor pharmacokinetics, and the potential for off-target, their clinical use is currently limited and still further improvements are desired. (2) Enhancing the degradation of the target protein. A common approach is to enhance the activity of protein degradation systems, including protease systems and autophagy systems. However, since the non-specific enhancement of the activity of the protein degradation system is likely to cause the degradation of other non-target proteins and serious side effects, no drug activating the protein degradation system is approved for clinical application at present. It is desirable to selectively enhance the degradation of only the target protein while avoiding degradation of non-target proteins by enhancing the activity of the protein degradation system.
Disclosure of Invention
The aim of the invention is to construct a small molecule compound specific for tau protein. In order to achieve the purpose, the inventors of the present invention found through intensive research that a bifunctional molecular compound, one end of which is capable of specifically binding to a target protein and the other end of which is specifically binding to a specific ubiquitin ligase, can be constructed by using a protein degradation TArgeting chimera (PROTAC) technology, and the two are connected via a linker group (linker). The compound thus constructed can bind to the target protein and ubiquitin ligase simultaneously, so that the target protein is adjacent to the ubiquitin ligase, thereby enhancing ubiquitination of the target protein, and finally being degraded by proteasome. Moreover, the ProTAC technology has the following advantages in addition to selectivity for target proteins: (1) can act on a plurality of targets which are traditionally difficult to become drugs. Many conventional small molecule drugs must act on specific binding pockets of the target protein in order to exert an inhibitory effect. The ProTAC technology is not so limited, and can act on many conventionally difficult-to-use targets as it interacts with any segment of the target protein and does not require high affinity, resulting in rapid degradation of the target protein and thus inhibition of the function of the target protein. (2) The PROTAC technology can be repeatedly used in cells, and can play a catalytic effect, so that the treatment effect can be achieved without high concentration.
Therefore, the invention provides a small molecule compound for specifically degrading tau protein, wherein the chemical structure of the compound 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 a ubiquitin ligase binding part, the tau protein binding part and the ubiquitin ligase binding part are connected through the connector group,
wherein ULM is a group having a structure represented by formula (1) or formula (2),
Figure BDA0002383600050000031
R1is a hydroxyl group or a group that is metabolized to a hydroxyl group in the patient or subject;
R2is hydroxyl, substituted or unsubstituted aryl of C6-C24, substituted or unsubstituted ternary aliphatic ring group, substituted or unsubstituted quaternary aliphatic ring group, substituted or unsubstituted five-membered aliphatic ring group, substituted or unsubstituted six-membered aliphatic ring group, substituted or unsubstituted ternary heterocyclic group, substituted or unsubstituted quaternary heterocyclic group, substituted or unsubstituted five-membered heterocyclic group, substituted or unsubstituted six-membered heterocyclic group or substituted or unsubstituted amino, wherein, the substituted amino is mono-substituted or di-substituted, and the mono-substituted amino is-NH-R4,R4Is C1-C12 alkyl, C6-C18 aryl, substituted or unsubstitutedA substituted ternary aliphatic ring group, a substituted or unsubstituted quaternary aliphatic ring group, a substituted or unsubstituted five-membered aliphatic ring group, a substituted or unsubstituted six-membered aliphatic ring group, a substituted or unsubstituted ternary heterocyclic group, a substituted or unsubstituted quaternary heterocyclic group, a substituted or unsubstituted five-membered heterocyclic group, a substituted or unsubstituted six-membered heterocyclic group, or-R5-Ar-HET, wherein R5O, S, C1-C6 alkylene, alkylene in which at least one hydrogen atom is substituted, Ar is substituted or unsubstituted C6-C12 arylene, HET is optionally substituted thiazole, optionally substituted isothiazole, optionally substituted thiophene, optionally substituted pyridine, optionally substituted pyridazine, optionally substituted furan, optionally substituted pyrrole, optionally substituted pyridine, optionally substituted imidazole, optionally substituted quinoline or optionally substituted indole;
R3is-CHR6-M-, ULM is linked to a linker group L through M, wherein R is6Is C1-C6 alkyl, M is a bond, C1-C6 alkylene, -NH-, or-NH-C (O) -R7-, wherein R7Is C1-C6 alkylene;
Figure BDA0002383600050000041
R8h, C1-C12 alkyl, C6-C18 aryl, substituted or unsubstituted ternary aliphatic ring group, substituted or unsubstituted quaternary aliphatic ring group, substituted or unsubstituted five-membered aliphatic ring group, substituted or unsubstituted six-membered aliphatic ring group, substituted or unsubstituted ternary heterocyclic group, substituted or unsubstituted quaternary heterocyclic group, substituted or unsubstituted five-membered heterocyclic group, substituted or unsubstituted six-membered heterocyclic group, or a group represented by the following formula (3),
Figure BDA0002383600050000051
wherein R is11Is H, C1-C12 alkyl, C6-C18 aryl, substituted or unsubstituted ternary aliphatic ring radical, substituted or unsubstituted quaternary aliphatic ring radical, substituted or unsubstituted five-membered aliphatic ring radical, substituted or unsubstitutedA substituted six-membered aliphatic ring group, a substituted or unsubstituted ternary heterocyclic group, a substituted or unsubstituted quaternary heterocyclic group, a substituted or unsubstituted five-membered heterocyclic group, or a substituted or unsubstituted six-membered heterocyclic group;
R12is a bond, C1-C4 alkylene or C6-C18 arylene;
R9is C1-C6 alkylene, -NH-or-NH-C (O) -R13-, wherein R13Is C1-C6 alkylene;
R10h, C1-C12 alkyl, C6-C18 aryl, substituted or unsubstituted ternary aliphatic ring group, substituted or unsubstituted quaternary aliphatic ring group, substituted or unsubstituted five-membered aliphatic ring group, substituted or unsubstituted six-membered aliphatic ring group, substituted or unsubstituted ternary heterocyclic group, substituted or unsubstituted quaternary heterocyclic group, substituted or unsubstituted five-membered heterocyclic group, or substituted or unsubstituted six-membered heterocyclic group.
Alternatively, 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) -R19-, wherein R19Alkylene which is a bond or C1-C4;
y is-R20-(R22-E-R23)n-R21-, wherein R20And R21Each being a bond or alkylene of C1-C8, R22And R23Each is C1-C4 alkylene, n is an integer of 0-10, E is O, S, amido, piperazinyl, NR24、S(O)、S(O)2、-S(O)2O、-OS(O)2、OS(O)2O、
Figure BDA0002383600050000052
Figure BDA0002383600050000061
Wherein E1Is O, S, CHR25Or NR26,R24、R25And R26Each is H or C1-C3 alkyl optionally substituted with one or two hydroxy groups;
z is-A-B-wherein A is a bond, O orS, B is a bond, C1-C4 alkylene or-R27-C (O) -, wherein R27Is C1-C4 alkylene.
Optionally, the TBM is a group having a structure shown in formula (4), or a group further modified by a substituent group at position (r), (c) and c) in the group of formula (4),
Figure BDA0002383600050000062
wherein R is14Is a bond, C1-C4 alkylene or C6-C18 arylene;
R15and R16An alkyl group each of which is H, C1 to C12, an aryl group of C6 to C18, a substituted or unsubstituted ternary aliphatic ring group, a substituted or unsubstituted quaternary aliphatic ring group, a substituted or unsubstituted five-membered aliphatic ring group, a substituted or unsubstituted six-membered aliphatic ring group, a substituted or unsubstituted ternary heterocyclic group, a substituted or unsubstituted quaternary heterocyclic group, a substituted or unsubstituted five-membered heterocyclic group, or a substituted or unsubstituted six-membered heterocyclic group;
R17is a bond, H, C1-C4 alkyl or-R18-C (O) -, wherein R18Is C1-C4 alkylene.
In one embodiment, the small molecule compound has the formula:
Figure BDA0002383600050000071
Figure BDA0002383600050000081
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 according to any of claims 1-4.
Optionally, the small molecule compound is administered to the patient by at least one means selected from: 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 tau protein related diseases.
Optionally, the Disease is Alzheimer's Disease (AD), Frontotemporal dementia with Parkinson's Disease linked to chromosome 17 (FTDP-17), Pick's Disease (PiD), Progressive Supranuclear Palsy (PSP), corticobasal degeneration (CBD), primary age-related tauopathies (PART), Argyrophilic Granulosis (AGD), aging-related tauopathies (ag), Chronic Traumatic Encephalopathy (CTE), glial cell disorder (tau), Parkinson's Disease, stroke's, stroke ' and stroke's.
Immunoblotting experiments prove that the small molecule compound for specifically degrading tau protein can enhance the degradation of tau protein in cells, thereby reducing the content of tau protein. Therefore, the small molecule compound specifically degrading tau protein provided by the invention can play a role in preventing and treating a series of tau diseases including Alzheimer disease.
Drawings
FIG. 1 is a nuclear magnetic resonance spectrum of a small molecule compound prepared in example 1 of the present invention;
FIG. 2 shows the results of immunoblot hybridization (a) and semi-quantitative analysis (b) of the effect of intracerebral ventricular injection of a compound provided by the invention on tau protein content in mouse cerebral cortex.
Detailed Description
The following describes in detail specific embodiments of the present invention. It should be understood that the detailed description and specific examples, while indicating the present invention, are given by way of illustration and explanation only, not limitation.
The endpoints of the ranges and any values disclosed herein are not limited to the precise range or value, and such ranges or values should be understood to encompass values close to those ranges or values. For ranges of values, between the endpoints of each of the ranges and the individual points, and between the individual points may be combined with each other to give one or more new ranges of values, and these ranges of values should be considered as specifically disclosed herein.
The chemical structure of the small molecule 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 a ubiquitin ligase binding part, and the tau protein binding part and the ubiquitin ligase binding part are connected through the linker group.
In preferred cases, ULM is capable of binding E3 ubiquitin ligase. Further preferably, the E3 ubiquitin ligase is VHLE3 ubiquitin ligase or CRBNE3 ubiquitin ligase.
In a more preferred embodiment, ULM is a group having a structure represented by formula (1) or formula (2),
Figure BDA0002383600050000101
R1is a hydroxyl group or a group that is metabolized to a hydroxyl group in the patient or subject;
R2is hydroxyl, substituted or unsubstituted aryl of C6-C24, substituted or unsubstituted ternary aliphatic ring radical, substituted or unsubstituted quaternary aliphatic ring radical, substituted or unsubstituted five-membered aliphatic ring radical, substituted or unsubstituted six-membered aliphatic ring radical, substituted or unsubstituted ternary heterocyclic radical, substituted or unsubstituted quaternary heterocyclic radical, substituted or unsubstituted five-membered heterocyclic radical, substituted or unsubstituted aryl of C6-C24, substituted or unsubstituted quaternary heterocyclic radicalA six-membered heterocyclic group or a substituted or unsubstituted amino group, wherein the substituted amino group is mono-or di-substituted and the mono-substituted amino group is-NH-R4,R4Is C1-C12 alkyl, C6-C18 aryl, substituted or unsubstituted ternary aliphatic ring group, substituted or unsubstituted quaternary aliphatic ring group, substituted or unsubstituted five-membered aliphatic ring group, substituted or unsubstituted six-membered aliphatic ring group, substituted or unsubstituted ternary heterocyclic group, substituted or unsubstituted quaternary heterocyclic group, substituted or unsubstituted five-membered heterocyclic group, substituted or unsubstituted six-membered heterocyclic group or-R5-Ar-HET, wherein R5O, S, C1-C6 alkylene, alkylene in which at least one hydrogen atom is substituted, Ar is substituted or unsubstituted C6-C12 arylene, HET is optionally substituted thiazole, optionally substituted isothiazole, optionally substituted thiophene, optionally substituted pyridine, optionally substituted pyridazine, optionally substituted furan, optionally substituted pyrrole, optionally substituted pyridine, optionally substituted imidazole, optionally substituted quinoline or optionally substituted indole;
R3is-CHR6-M-, ULM is linked to a linker group L through M, wherein R is6Is C1-C6 alkyl, M is a bond, C1-C6 alkylene, -NH-, or-NH-C (O) -R7-, wherein R7Is C1-C6 alkylene;
Figure BDA0002383600050000111
R8h, C1-C12 alkyl, C6-C18 aryl, substituted or unsubstituted ternary aliphatic ring group, substituted or unsubstituted quaternary aliphatic ring group, substituted or unsubstituted five-membered aliphatic ring group, substituted or unsubstituted six-membered aliphatic ring group, substituted or unsubstituted ternary heterocyclic group, substituted or unsubstituted quaternary heterocyclic group, substituted or unsubstituted five-membered heterocyclic group, substituted or unsubstituted six-membered heterocyclic group, or a group represented by the following formula (3),
Figure BDA0002383600050000112
wherein R is11Is H, C1-C12 alkyl, C6-C18 aryl, substituted or unsubstituted ternary aliphatic ring group, substituted or unsubstituted quaternary aliphatic ring group, substituted or unsubstituted five-membered aliphatic ring group, substituted or unsubstituted six-membered aliphatic ring group, substituted or unsubstituted ternary heterocyclic group, substituted or unsubstituted quaternary heterocyclic group, substituted or unsubstituted five-membered heterocyclic group or substituted or unsubstituted six-membered heterocyclic group;
R12is a bond, C1-C4 alkylene or C6-C18 arylene;
R9is C1-C6 alkylene, -NH-or-NH-C (O) -R13-, wherein R13Is C1-C6 alkylene;
R10h, C1-C12 alkyl, C6-C18 aryl, substituted or unsubstituted ternary aliphatic ring group, substituted or unsubstituted quaternary aliphatic ring group, substituted or unsubstituted five-membered aliphatic ring group, substituted or unsubstituted six-membered aliphatic ring group, substituted or unsubstituted ternary heterocyclic group, substituted or unsubstituted quaternary heterocyclic group, substituted or unsubstituted five-membered heterocyclic group, or substituted or unsubstituted six-membered heterocyclic group.
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) -R19-, wherein R19An alkylene group having a bond or C1-C4 (e.g., methylene, ethylene, propylene, or butylene);
y is-R20-(R22-E-R23)n-R21-, wherein R20And R21Each independently a bond or C1-C8 alkylene (e.g., methylene, ethylene, propylene, butylene, pentylene, hexylene, heptylene, or octylene), R22And R23Each is C1-C4 alkylene (e.g., methylene, ethylene, propylene or butylene), 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 is24、S(O)、S(O)2、-S(O)2O、-OS(O)2、OS(O)2O、
Figure BDA0002383600050000121
Figure BDA0002383600050000122
Wherein E1Is O, S, CHR25Or NR26,R24、R25And R26Each 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-R27-C (O) -, wherein R27Is C1-C4 alkylene (such as methylene, ethylene, propylene or butylene).
In a more preferred embodiment, the TBM is a group having a structure shown in formula (4), or a group further modified by a substituent group at the position of (c), (c) or (c) in the structure shown in formula (4),
Figure BDA0002383600050000131
wherein R is14Is a bond, an alkylene group of C1-C4 (such as methylene, ethylene, propylene or butylene) or an arylene group of C6-C18 (such as phenylene or naphthylene);
R15and R16An alkyl group (e.g., methyl, ethyl, propyl, or butyl) each of H, C1 to C12, an aryl group (e.g., phenyl or naphthyl) of C6 to C18, a substituted or unsubstituted ternary aliphatic ring group, a substituted or unsubstituted quaternary aliphatic ring group, a substituted or unsubstituted five-membered aliphatic ring group, a substituted or unsubstituted six-membered aliphatic ring group, a substituted or unsubstituted ternary heterocyclic group, a substituted or unsubstituted quaternary heterocyclic group, a substituted or unsubstituted five-membered heterocyclic group, or a substituted or unsubstituted six-membered heterocyclic group;
R17is a bond,H. C1-C4 alkyl (such as methyl, ethyl, propyl or butyl) or-R18-C (O) -, wherein R18Is C1-C4 alkylene (such as methylene, ethylene, propylene or butylene).
Wherein, the substituent group at the position of (r), (C) or (C) in the group of the modified structure shown in formula (4) can be halogen (such as fluorine or chlorine), alkyl (such as methyl, ethyl, propyl or butyl) of C1-C4, alkoxy (such as methoxy, ethoxy, propoxy or butoxy) of C1-C4, carboxyl, amino, aryl (such as phenyl) of C6-C18 or benzyl.
Further preferably, in the structure of formula (4), TBM is attached to the linker group L through the position of the fifth in formula (4), and R is14Is ethylene, R15And R16Each is methyl, R17Is a bond, methylene or-CH2-C(O)-。
In a most preferred embodiment, the small molecule compound that specifically degrades tau protein has the formula:
Figure BDA0002383600050000141
Figure BDA0002383600050000151
the small molecule compound for specifically degrading tau protein can be prepared according to the following process route:
Figure BDA0002383600050000152
the preparation process comprises the following steps:
(1) preparation of Compound 2-1
2,4, 6-trichloro-1, 3, 5-triazine (compound 1-1) is added to dichloromethane, and triethylamine is added thereto. 1- (1, 4-diaza-1-yl) ethoxy-1-one was then diluted with dichloromethane and slowly added dropwise to the reaction mixture. The mixture was stirred at room temperature overnight. The reaction was quenched with water. After standing at room temperature, the upper organic solution was separated and the lower aqueous solution was extracted three times with dichloromethane. The organic solutions obtained above were combined together, dried over anhydrous magnesium sulfate and concentrated, and the solvent was removed under reduced pressure. The residue was solidified with toluene, filtered, and washed with n-hexane to give compound 2-1.
(2) Preparation of Compound 3-1
The compound 2-1 is added into dichloromethane, and triethylamine is added. The 2- (thien-2-yl) ethan-1-amine was then diluted with dichloromethane and slowly added dropwise to the reaction mixture. The mixture was stirred at room temperature overnight. The reaction was quenched with water. After standing at room temperature, the upper organic solution was separated and the lower aqueous solution was extracted three times with dichloromethane. The organic solutions obtained above were combined together, dried over anhydrous magnesium sulfate and concentrated, and the solvent was removed under reduced pressure. The residue was solidified with toluene, filtered, and washed with n-hexane to give compound 3-1.
(3) Preparation of Compound 4-1
The compound 3-1 was added to dichloromethane, and triethylamine was further added. Tert-butyl (2- (methylamino) ethyl) carbamate was then diluted with dichloromethane and slowly added dropwise to the reaction mixture. The mixture was stirred at room temperature overnight. The reaction was quenched with water. After standing at room temperature, the upper organic solution was separated and the lower aqueous solution was extracted three times with dichloromethane. The organic solutions obtained above were combined together, dried over anhydrous magnesium sulfate and concentrated, and the solvent was removed under reduced pressure. The residue was solidified with toluene, filtered, and washed with n-hexane to give compound 4-1.
(4) Preparation of Compound 5-1
The compound 4-1 was added to dichloromethane and trifluoroacetate was slowly added dropwise. The mixture was stirred at room temperature. The reaction mixture was concentrated under reduced pressure. Dichloromethane was added to the residue and concentrated under reduced pressure. Repeating the operation for multiple times to obtain the compound 5-1.
(5) Preparation of Compound 7-1
To DMF (N, N-dimethylformamide) was added 2,2' - (ethane-1, 2-dialkylbis (oxy)) diacetic acid. Followed by the addition of 4-amino-2- (2, 6-dioxopiperidin-3-yl) isoindoline-1, 3-dione and DIPEA (N, N-diisopropylethylamine) with stirring. The mixture was stirred at room temperature for three days. The residue was adjusted to pH 4-5 with HCl and extracted with ethyl acetate. The combined organic phases were dried, concentrated and purified by preparative HPLC to afford compound 7-1.
(6) Preparation of the target Compound
Compound 7-1 was added to DMF, to which HATU (2- (7-azabenzotriazol-1-yl) -N, N, N ', N' -tetramethyluranium hexafluorophosphate) was added. Followed by addition of compound 5-1 and DIEAP with stirring. The mixture was stirred at room temperature. The residue was adjusted to pH 4-5 with HCl and extracted with ethyl acetate. The combined organic phases were dried, concentrated and purified by preparative HPLC to give the title compound.
The present 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 provided herein above.
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 the 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 with parkinson's disease linked to chromosome 17, pick's disease, progressive supranuclear palsy, corticobasal degeneration, primary age-related tauopathies, silvery particle disease, aging-related tau astrocytosis, chronic traumatic encephalopathy, globoid tauopathy, parkinson's disease, huntington's disease, stroke, and epilepsy.
The present invention will be described in detail below by way of examples.
Examples
(1) Preparation of Compound 2-1
2,4, 6-trichloro-1, 3, 5-triazine (2.00g, 10.93mmol) was added to dichloromethane (30mL), and 2.21g (21.86mmol, 2.0eq.) of triethylamine was added. After cooling at 0 ℃ 1.55g (10.93mmol, 1.0eq.) of 1- (1, 4-diaza-1-yl) ethoxy-1-one are diluted with 4ml of dichloromethane and slowly added dropwise to the reaction mixture. The mixture was stirred at room temperature overnight. The reaction was quenched with water (20 mL). After standing at room temperature, the upper organic solution was separated and the lower aqueous solution was extracted three times with dichloromethane. The organic solutions obtained above were combined together, dried over anhydrous magnesium sulfate and concentrated, and the solvent was removed under reduced pressure. The residue was solidified with toluene, filtered, and washed with n-hexane to give 2.40g (76.0%) of compound 2-1 as a yellow solid.
(2) Preparation of Compound 3-1
Compound 2-1(2.40g, 8.30mmol) was added to dichloromethane (30mL) and 1.68g (16.60mmol, 2.0eq.) triethylamine was added. After cooling at 0 ℃, 1.05g (8.30mmol, 1.0eq.) of 2- (thien-2-yl) ethan-1-amine was diluted with 4ml of dichloromethane and slowly added dropwise to the reaction mixture. The mixture was stirred at room temperature overnight. The reaction was quenched with water (20 mL). After standing at room temperature, the upper organic solution was separated and the lower aqueous solution was extracted three times with dichloromethane. The organic solutions obtained above were combined together, dried over anhydrous magnesium sulfate and concentrated, and the solvent was removed under reduced pressure. The residue was solidified with toluene, filtered, and washed with n-hexane to give 2.70g (85.6%) of compound 3-1 as a yellow solid.
(3) Preparation of Compound 4-1
Compound 3-1(1.00g, 2.63mmol) was added to dichloromethane (10mL), followed by 0.531g (5.26mmol, 2.0eq.) of triethylamine. After cooling at 0 ℃, 0.915g of tert-butyl (2- (methylamino) ethyl) carbamate is diluted with 4ml of dichloromethane and slowly added dropwise to the reaction mixture. The mixture was stirred at room temperature overnight. The reaction was quenched with water (10 mL). After standing at room temperature, the upper organic solution was separated and the lower aqueous solution was extracted three times with dichloromethane. The organic solutions obtained above were combined together, dried over anhydrous magnesium sulfate and concentrated, and the solvent was removed under reduced pressure. The residue was solidified with toluene, filtered and washed with n-hexane to give 0.800g (58.7%) of compound 4-1 as a yellow solid.
(4) Preparation of Compound 5-1
Compound 4-1(248mg, 0.478mmol) was added to dichloromethane (3mL) and 46.3mg (0.772mmol, 2.0eq.) of trifluoroacetate was added slowly dropwise. The mixture was stirred at room temperature for 1 hour. The reaction mixture was concentrated under reduced pressure. Dichloromethane (3mL) was added to the residue and concentrated under reduced pressure. This procedure was repeated 3 times to give 200mg (crude) of compound 5-1 as a yellow oil.
(5) Preparation of Compound 7-1
To DMF (N, N-dimethylformamide, 3mL) was added 2,2' - (ethane-1, 2-dialkylbis (oxy)) diacetic acid (300mg, 1.69mmol) at 0 ℃. After 30 minutes at the same temperature, 4-amino-2- (2, 6-dioxopiperidin-3-yl) isoindoline-1, 3-dione (693mg, 2.54mmol) and DIPEA (N, N-diisopropylethylamine, 983mg, 7.62mmol) were added with stirring. The mixture was stirred at room temperature for three days. The residue was adjusted to pH 4-5 with 1M HCl and extracted with ethyl acetate. The combined organic phases were dried, concentrated and purified by preparative HPLC to give compound 7-1(100mg, 48.0% yield) as a white solid.
(6) Preparation of the target Compound
Compound 7-1(100mg, 0.231mmol) was added to DMF (3mL) and HATU (2- (7-azabenzotriazol-1-yl) -N, N, N ', N' -tetramethyluranium hexafluorophosphate, 132mg, 0.346mmol) was added thereto at 0 ℃. After 30 minutes at the same temperature, compound 5-1(200mg, 0.478mmol) and DIEAP (59.6mg, 0.462mmol) were added under stirring. The mixture was stirred at room temperature for 3 hours. The residue was adjusted to pH 4-5 with 1M HCl and extracted with ethyl acetate. The combined organic phases were dried, concentrated and purified by preparative HPLC to give the title compound (17mg, 8.85% yield) as a white solid. The nuclear magnetic resonance spectrum of the compound is shown in figure 1, and the nuclear magnetic resonance spectrum data is as follows:
1H NMR(400MHz,Acetone-d6):δ:ppm 10.32(s,1H),9.94(br,1H),8.70(d,J=8.4Hz,1H),7.70(t,J=7.8Hz,1H),7.45-7.44(m,2H),7.09(d,J=4.8Hz,1H),6.79-6.74(m,2H),5.93(br,1H),5.04-5.00(m,1H),4.03(s,2H),3.82(s,2H),3.63(br,7H),3.52-3.46(m,6H),3.30(br,4H),2.98(t,J=7.4Hz,3H),2.88-2.84(m,4H),2.62-2.63(m,2H),2.14-2.11(m,1H),1.94(s,2H),1.91(s,2H),1.16(s,1H)。
HPLC purity: 99.3% (254nm), 99.3% (214 nm); mass: m/z 834[ M +1]+
Test example
First, preparation of animal sample
1. Raising of animals
C57BL/6 mice were purchased from Wafukang (Beijing Corp.). The experimental animals were strictly managed according to the regulations on the management of laboratory animals in China, the temperature was controlled at 25 ℃, the circadian rhythm was reversed for 12 hours, and all animal experiments were approved by the ethical committee of the college of Tongji medical college of Huazhong university of science and technology.
2. Pharmaceutical treatment of animals
Mice were dosed by lateral ventricle injection. Anaesthetizing mice with isoflurane (purchased from Warred) on a mouse gas anaesthesia machine, cleaning the hair on the top of the head of the mice by a hair-shaving clipper, disinfecting the hair with iodophors, fixing the mice on a stereotaxic apparatus for continuous anaesthesia, fully coating the eyeball with erythromycin eye ointment for protection, cutting the skin on the top of the head of the mice into an incision along the midline of the skull by an ophthalmological scissors, fully exposing the front fontanel, the back fontanel and the midline of the skull, and adjusting the stereotaxic instrument to ensure that the front fontanel is at the same level and the plane of the skull is horizontal. And (3) performing three-dimensional positioning drilling at a position 0.2mm behind the bregma and 0.9mm beside the bregma. Sucking 5 μ l of C030019(200 μ M) or 0.04% DMSO with a Hamilton needle with a range of 10 μ l, inserting 2.3mm downward from the plane of the skull, injecting with a micro-injection pump at a constant speed for 10 min (0.25 μ l/min), retaining the needle for 10 min, taking out the needle head, and suturing the skin. The materials were obtained 48h after drug injection.
3. Preparation of samples
The instruments and consumables used for homogenization of mouse brain tissue are first prepared, fresh homogenate (50mM Tris-HCl, pH 7.4-7.5,100mM NaCl, 1% Triton,5mM EDTA,1mM PMSF (Sigma, P-7626), 1 Xprotease inhibitor cocktail (Protease inhibitors cocktail, Sigma, P8340)) is prepared and the homogenate is placed on ice for precooling. After anesthetizing a mouse with 6% chloral hydrate, the mouse was decapitated, the whole brain tissue was taken out, placed on a glass plate placed on ice, the cerebellum was cut off rapidly, the left and right brains were separated, half of the brain tissue was separated into the cortex and the hippocampus, and 1/3 near the frontotemporal lobe of the cortex was taken and placed in pre-cooled EP tubes of 1.5mL, respectively. Weighing brain tissue, putting the brain tissue into a homogenate tube, adding the homogenate at a ratio of 1:10 (brain tissue mass: homogenate volume: 1:10), opening the homogenate machine, homogenizing under 50 times for forward and reverse, collecting the homogenate liquid into an EP tube, standing on ice for 30 minutes, blowing and beating the liquid in the tube uniformly every 10 minutes, centrifuging at 12000rpm multiplied by 20 minutes in a centrifuge precooled to 4 ℃, and dividing the supernatant into two parts. The first portion was taken in appropriate volume, added to 4 x SDS loading buffer at 3:1, and placed in a fume hood at 10: 1, adding 250mM beta-mercaptoethanol, uniformly mixing, heating for 10 minutes at 95 ℃ in an iron bath, cooling, shaking, uniformly mixing, centrifuging, and storing in a refrigerator at-80 ℃. The other part is kept about 10 mu L, the protein concentration is measured, and the rest is put into liquid nitrogen for quick freezing and then transferred to a refrigerator at the temperature of 80 ℃ below zero for storage. Before immunoblotting hybridization loading, diluting the sample by 4-5 times with a proper amount of 1 xSDS loading buffer solution, reheating for 10 minutes, cooling, centrifuging for a short time, and mixing the samples uniformly.
Second, measurement of protein content of sample (BCA method)
1. Shaking a protein sample, then appropriately diluting (5 mu l of each sample is mixed and diluted with 45 mu l of double distilled water, and 2 parallel samples are arranged respectively), and centrifuging and shaking;
2. setting six standard tubes, respectively taking 0 μ l, 10 μ l, 20 μ l, 30 μ l, 40 μ l and 50 μ l of 20mg/ml BSA (100mg BSA dissolved in 5ml double distilled water), respectively adding 1000 μ l, 990 μ l, 980 μ l, 970 μ l, 960 μ l and 950 μ l double distilled water to prepare standard proteins of 0 μ g/μ l, 0.2 μ g/μ l, 0.4 μ g/μ l, 0.6 μ g/μ l, 0.8 μ g/μ l and 1.0 μ g/μ l;
3. the diluted protein sample and the diluted standard protein were added to a 96-well plate (5. mu.l/well, one tip was changed for each well at the junction between the wall and the bottom of the PCR gun) with 3 parallel wells each.
4. The working solution is prepared from solution A and solution B in the kit according to the proportion of 50: 1. Adding the working solution into a 96-well plate, quickly suspending and adding 95 mu l of the working solution into each well, covering a cover after adding, quickly attaching the cover to the bottom, vibrating along the same direction, keeping hands from touching the bottom of the 96-well plate, and incubating for 30 minutes at 37 ℃ by using a plastic box;
5. de-bubbling with a 1ml syringe needle, turning on a BioTek switch, turning on Gen5, pointing a left arrow icon, clicking OK, and exporting Excel;
6. replicate standard protein OD values, frame select OD values and standard protein concentrations and insert scatter plots. Data points are selected to add trend lines, formulas are displayed, R square values (at least 2 should be found after the decimal point is 9) are displayed, and outliers are removed. Copy the protein OD value of the sample, input the corresponding sample group number above, remove the abnormal value.
Determination of tau protein content (immunoblotting)
1. Building frame (two glass plates, three bottles, five reagents, filter paper, toilet paper, garbage can, gun head, comb teeth)
(1) Cleaning the tabletop and the underframe, cleaning the comb teeth, the glass plate, the distilled water bottle, the upper rubber bottle and the lower rubber bottle, drying the upper rubber bottle and the lower rubber bottle, taking out the reagent for preparing the electrophoresis gel, and returning to the room temperature;
(2) the higher glass plates are folded inwards together, the upper part is pressed to enable the lower part to be tightly attached to the desktop to enable the upper part to be flush with the desktop, the clamp is turned outwards to clamp the upper part, and the upper part is placed on the underframe and is buckled by the clamp.
(3) And injecting double distilled water to check whether leakage exists, and detecting leakage after reloading if leakage exists.
2. Preparation of an electrophoretic gel
TABLE 1
Figure BDA0002383600050000221
(1) Sequentially adding 20% Arc/Bis, Tris buffer solution, TEMED and 10% APS, and blowing and uniformly mixing by using a pipette, wherein bubbles are prevented from being mixed in the mixed solution in the whole process;
(2) respectively and slowly injecting separation glue into the glue film along two corners (the separation glue penetrates under the liquid surface during suction and is gently blown and uniformly mixed, a small amount of liquid is reserved at a gun head each time to prevent bubbles from being generated), the use amount of each piece of glue is 3 multiplied by 900 mu l, and after the glue is observed to be not leaked, double distilled water is respectively used for filling gaps of the glue film along the two corners (the oxygen is prevented from inhibiting polymerization, the glue at the lower part is kept horizontal, and the glue can be put for a period of time);
(3) waiting for about 30 minutes for the gel to separate, pouring back to remove double distilled water, completely absorbing the residual water by using filter paper, and marking the upper edge of the lower part of the gel by using a marker pen;
(4) the concentrated gel is respectively and slowly injected into the gel film along two corners, the comb teeth with the required specification (the small comb teeth are used for loading the sample amount less than 20 mu l, the large comb teeth are used for loading the sample amount more than 20 mu l) are obliquely inserted from left to right, the gel is supplemented between lanes to avoid the gel shrinkage, and the gel is waited (about 50 minutes is needed).
3. And (3) carrying out electrophoretic separation on the sample and the protein (a sample needle, a sample, a socket, a Marker, an electrophoretic fluid, an electrophoretic tank and a distilled water bottle).
The conductive wire under the electrophoresis frame is cleaned, the conductive wire is transferred to the electrophoresis frame, the lanes and the numbers are marked by a marking pen, the comb teeth are slowly and vertically pulled out, the gel tank is filled with electrophoresis liquid, and a sample is taken by a microsyringe and is added into each lane (Marker adds 1 mul to lane 1). After the sample is loaded, the electrophoresis rack is transferred to an electrophoresis tank, the electrophoresis tank is added with electrophoresis liquid, then a cover is covered to ensure that the red color is red, the black color is black, after the sample is loaded, the electrophoresis is firstly carried out for about 30 minutes by using a constant current of 10 mA/piece of gel (starting according to two times), when the bromophenol blue indicator electrophoresis is carried out until the junction of the concentrated gel and the separation gel is linear, the electrophoresis is changed into constant voltage of 100V (if the constant voltage can be adjusted to high current) electrophoresis for about 60 minutes until the bromophenol blue reaches the bottom of the gel and the Marker strips are completely separated.
4. Transferring membrane (marking NC membrane, transferring membrane liquid, filter paper, ice box, basin, dish, transferring membrane groove, plastic plate, cleaning tweezers)
(1) And (3) marking the NC membrane by using a marker pen, then soaking the NC membrane in the recovered membrane transferring liquid for 10-20 minutes (which is beneficial to fixing protein, balancing gel and removing SDS), pressing bayonets at two sides to take off the gel tank, prying the middle parts at the right sides of the glass plate and the white porcelain plate by using a small plate, and keeping electrophoresis of the residual gel in the process.
(2) Slightly inclining a glass plate vertically according to the molecular weight range to be displayed and slightly cutting glue back and forth left and right once, sticking three layers of filter paper soaked in the membrane transferring liquid on the glue by using a pair of tweezers, carefully prying up the glue by using a small plate and placing the glue on a sponge (the filter paper faces downwards), sticking an inverted NC membrane on the other side, immersing the glue and the NC membrane into the membrane transferring liquid (the glue is on the upper side), removing bubbles by using a glass rod, carefully clamping the glue by using the pair of tweezers and placing the glue on the hand (the glue is on the upper side), sticking the three layers of filter paper soaked in the membrane transferring liquid on the glue by using the pair of tweezers, inversely placing the glue on the sponge, and sticking three layers of filter paper. From bottom to top, a black plastic plate → a layer of sponge → three layers of filter paper → glue → NC film → three layers of filter paper → a layer of sponge → a transparent plastic plate, if not tight, can be fixed by a rubber band.
(3) After the electrodes are correctly arranged, the membrane transferring groove is placed in an ice bath (the glue is not required to be soaked in the membrane transferring liquid for a long time before the electrification so as to avoid the diffusion and the decomposition of the protein), the transferring current is constant current 276mA, the voltage is generally 140V (the methanol can be properly supplemented to increase the voltage), the specific transferring time is determined according to the molecular weight of the protein to be transferred, the time when the molecular weight of the transferred protein is less than 100kDa is 1h, and the time when the molecular weight is more than 100kDa is 1.5 h.
5. Immunoblotting color development (cleaning tweezers, box with double distilled water, milk, fresh-keeping bag, toilet paper, primary antibody, ice box, plate, transparent glue, TBST, black plastic bag, secondary antibody)
(1) And (3) sealing: after the completion of the membrane transfer, the NC membrane was carefully blocked with 5% skimmed milk powder in TBS blocking solution at room temperature for 1h or overnight at 4 ℃ to recover the filter paper not in contact with the gel.
(2) Primary antibody incubation: taking out the NC film, rinsing residual milk stain on the surface of the film by using 1 × TBS, clamping the NC film by using tweezers, standing on toilet paper to remove excessive water, placing the NC film Marker side outwards in a freshness protection package, and draining and exhausting by using the toilet paper. Add primary antibody (0.1% Tween 20 can be added to reduce background) and seal on plate (with Marker and protein side up), clear tape is not pressed on target strip, and incubate overnight at 4 deg.C.
(3) And (3) secondary antibody incubation: the next day the NC membrane was removed from the incubation bag and primary antibody was recovered, rinsed with TBST buffer for 3X 5 minutes, rinsed with 1X TBS for removing residual salt ions on the membrane surface, set up on toilet paper with tweezers to remove excess water, placed in a freshness-keeping bag and vented with toilet paper. Adding horseradish peroxide labeled goat anti-rabbit or goat anti-mouse Odyssey secondary antibody (0.1% Tween 20(Tween 20) can be added to reduce background) in a dark place, sealing and pasting the plate (the side with Marker and protein faces upwards), placing a transparent adhesive tape on a target strip, incubating for about 1 hour with slow shaking at room temperature, taking out the NC membrane from the incubation bag and recovering the secondary antibody, and rinsing for 3 multiplied by 5 minutes by TBST buffer solution. After rinsing, the residual salt ions on the membrane surface were removed by rinsing with 1 × TBS.
(4) Color development: the glass plate is firstly wiped clean by using a piece of lens wiping paper dipped with absolute ethyl alcohol. The NC film Marker was placed on a glass plate side up, incubated for about 1 minute with an ECL chemiluminescent substrate (BeyoECL Star, P0018AM) configured as described in the specification, and then photographed with an ECL developing apparatus (Shanghai volkong scientific instruments Co., Ltd., ChemiScope 3300 Mini).
(5) Semi-quantitative analysis: the obtained Image was quantified for gray scale using Image J software.
(6) Statistical analysis: statistical analysis was done using GraphPad Prism software.
FIG. 2 shows the results of immunoblot hybridization (a) and semi-quantitative analysis (b) of the effect of intracerebral ventricular injection of a compound provided by the invention on tau protein content in mouse cerebral cortex. The results show that: the compound can obviously reduce the content of the cerebral cortex and tau protein of mice.
Thus, the tau protein-directed compound constructed by the invention can reduce the content of tau protein in the brain of a mouse.
Since tau protein is abnormally accumulated in cells and is involved in more than 20 kinds of neurodegenerative diseases, the accumulation amount thereof is positively correlated with neurodegeneration and memory impairment of these degenerative diseases. Thus, the tau protein degradation can achieve the purpose of preventing or/and treating tau-related neurodegenerative diseases, such as Alzheimer disease, frontotemporal dementia with Parkinson's disease linked to chromosome 17, pick's disease, progressive supranuclear palsy, corticobasal degeneration, primary age-related tauopathies, silvery granulosis, aging-related tau astrocytosis, chronic traumatic encephalopathy, globulo-like gliosis, Parkinson's disease, Huntington's disease, stroke and epilepsy.
The preferred embodiments of the present invention have been described above in detail, but the present invention is not limited thereto. Within the scope of the technical idea of the invention, many simple modifications can be made to the technical solution of the invention, including combinations of various technical features in any other suitable way, and these simple modifications and combinations should also be regarded as the disclosure of the invention, and all fall within the scope of the invention.

Claims (8)

1. A small molecule compound for specifically degrading tau protein, having a chemical structure of 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 a ubiquitin ligase binding moiety, said tau protein binding moiety and said ubiquitin ligase binding moiety are linked by a linker group,
wherein ULM is a group having a structure represented by formula (1) or formula (2),
Figure FDA0002383600040000011
R1is a hydroxyl group or a group that is metabolized to a hydroxyl group in the patient or subject;
R2is hydroxyl, substituted or unsubstituted aryl of C6-C24, substituted or unsubstituted ternary aliphatic ring group, substituted or unsubstituted quaternary aliphatic ring group, substituted or unsubstituted five-membered aliphatic ring group, substituted or unsubstituted six-membered aliphatic ring group, substituted or unsubstituted ternary heterocyclic group, substituted or unsubstituted quaternary heterocyclic group, substituted or unsubstituted five-membered heterocyclic group, substituted or unsubstituted six-membered heterocyclic group or substituted or unsubstituted amino, wherein, the substituted amino is mono-substituted or di-substituted, and the mono-substituted amino is-NH-R4,R4Is C1-C12 alkyl, C6-C18 aryl, substituted or unsubstituted ternary aliphatic ring group, substituted or unsubstituted quaternary aliphatic ring group, substituted or unsubstituted five-membered aliphatic ring group, substituted or unsubstituted six-membered aliphatic ring group, substituted or unsubstituted ternary heterocyclic group, substituted or unsubstituted quaternary heterocyclic group, substituted or unsubstituted five-membered heterocyclic group, substituted or unsubstituted six-membered heterocyclic group or-R5-Ar-HET, wherein R5Alkylene having a value of O, S, C1-C6A group, an alkylene group having at least one hydrogen atom substituted, Ar is a substituted or unsubstituted C6-C12 arylene group, HET is optionally substituted thiazole, optionally substituted isothiazole, optionally substituted thiophene, optionally substituted pyridine, optionally substituted pyridazine, optionally substituted furan, optionally substituted pyrrole, optionally substituted pyridine, optionally substituted imidazole, optionally substituted quinoline, or optionally substituted indole;
R3is-CHR6-M-, ULM is linked to a linker group L through M, wherein R is6Is C1-C6 alkyl, M is a bond, C1-C6 alkylene, -NH-, or-NH-C (O) -R7-, wherein R7Is C1-C6 alkylene;
Figure FDA0002383600040000021
R8h, C1-C12 alkyl, C6-C18 aryl, substituted or unsubstituted ternary aliphatic ring group, substituted or unsubstituted quaternary aliphatic ring group, substituted or unsubstituted five-membered aliphatic ring group, substituted or unsubstituted six-membered aliphatic ring group, substituted or unsubstituted ternary heterocyclic group, substituted or unsubstituted quaternary heterocyclic group, substituted or unsubstituted five-membered heterocyclic group, substituted or unsubstituted six-membered heterocyclic group, or a group represented by the following formula (3),
Figure FDA0002383600040000022
wherein R is11Is H, C1-C12 alkyl, C6-C18 aryl, substituted or unsubstituted ternary aliphatic ring group, substituted or unsubstituted quaternary aliphatic ring group, substituted or unsubstituted five-membered aliphatic ring group, substituted or unsubstituted six-membered aliphatic ring group, substituted or unsubstituted ternary heterocyclic group, substituted or unsubstituted quaternary heterocyclic group, substituted or unsubstituted five-membered heterocyclic group or substituted or unsubstituted six-membered heterocyclic group;
R12is a bond, C1-C4 alkylene or C6-C18 arylene;
R9alkylene of C1 to C6-NH-or-NH-C (O) -R13-, wherein R13Is C1-C6 alkylene;
R10h, C1-C12 alkyl, C6-C18 aryl, substituted or unsubstituted ternary aliphatic ring group, substituted or unsubstituted quaternary aliphatic ring group, substituted or unsubstituted five-membered aliphatic ring group, substituted or unsubstituted six-membered aliphatic ring group, substituted or unsubstituted ternary heterocyclic group, substituted or unsubstituted quaternary heterocyclic group, substituted or unsubstituted five-membered heterocyclic group, or substituted or unsubstituted six-membered heterocyclic group.
2. The small molecule compound of claim 1, wherein 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) -R19-, wherein R19Alkylene which is a bond or C1-C4;
y is-R20-(R22-E-R23)n-R21-, wherein R20And R21Each being a bond or alkylene of C1-C8, R22And R23Each is C1-C4 alkylene, n is an integer of 0-10, E is O, S, amido, piperazinyl, NR24、S(O)、S(O)2、-S(O)2O、-OS(O)2、OS(O)2O、
Figure FDA0002383600040000031
Figure FDA0002383600040000032
Wherein E1Is O, S, CHR25Or NR26,R24、R25And R26Each 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-R27-C (O) -, wherein R27Is C1-C4 alkylene.
3. The small molecule compound according to claim 1, wherein the TBM is a group having a structure represented by formula (4), or a group in which the substituent group is further modified at the position of (4), wherein the TBM is linked to the linker group L at the position of (4) such as (i), (ii), (iii), (iv), (iii), (iv), (v) or (v) in the v),
Figure FDA0002383600040000041
wherein R is14Is a bond, C1-C4 alkylene or C6-C18 arylene;
R15and R16An alkyl group each of which is H, C1 to C12, an aryl group of C6 to C18, a substituted or unsubstituted ternary aliphatic ring group, a substituted or unsubstituted quaternary aliphatic ring group, a substituted or unsubstituted five-membered aliphatic ring group, a substituted or unsubstituted six-membered aliphatic ring group, a substituted or unsubstituted ternary heterocyclic group, a substituted or unsubstituted quaternary heterocyclic group, a substituted or unsubstituted five-membered heterocyclic group, or a substituted or unsubstituted six-membered heterocyclic group;
R17is a bond, H, C1-C4 alkyl or-R18-C (O) -, wherein R18Is C1-C4 alkylene.
4. The small molecule compound according to any one of claims 1 to 3, wherein the small molecule compound has the formula:
Figure FDA0002383600040000051
5. 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 of any of claims 1-4.
6. The method of claim 5, wherein 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.
7. Use of a small molecule compound according to any one of claims 1-4 in the manufacture of a medicament for the treatment or prevention of a tau protein associated disease.
8. The use according to claim 7, wherein the disease is at least one of Alzheimer's disease, frontotemporal dementia with Parkinson's disease linked to chromosome 17, pick's disease, progressive supranuclear palsy, corticobasal degeneration, primary age-related tauopathies, argentiphilic granulosis, aging-related tau astrocytosis, chronic traumatic encephalopathy, globoid tauopathy, Parkinson's disease, Huntington's disease, stroke and epilepsy.
CN202010090680.6A 2020-02-13 2020-02-13 Small molecular compound for specifically degrading tau protein and application thereof Pending CN113248484A (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
CN202010090680.6A CN113248484A (en) 2020-02-13 2020-02-13 Small molecular compound for specifically degrading tau protein and application thereof
CN202180012446.4A CN115279751B (en) 2020-02-13 2021-02-04 Small molecular compound for specifically degrading tau protein and application thereof
PCT/CN2021/075201 WO2021160012A1 (en) 2020-02-13 2021-02-04 Small molecular compound specifically degrading tau protein and use thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202010090680.6A CN113248484A (en) 2020-02-13 2020-02-13 Small molecular compound for specifically degrading tau protein and application thereof

Publications (1)

Publication Number Publication Date
CN113248484A true CN113248484A (en) 2021-08-13

Family

ID=77219818

Family Applications (2)

Application Number Title Priority Date Filing Date
CN202010090680.6A Pending CN113248484A (en) 2020-02-13 2020-02-13 Small molecular compound for specifically degrading tau protein and application thereof
CN202180012446.4A Active CN115279751B (en) 2020-02-13 2021-02-04 Small molecular compound for specifically degrading tau protein and application thereof

Family Applications After (1)

Application Number Title Priority Date Filing Date
CN202180012446.4A Active CN115279751B (en) 2020-02-13 2021-02-04 Small molecular compound for specifically degrading tau protein and application thereof

Country Status (2)

Country Link
CN (2) CN113248484A (en)
WO (1) WO2021160012A1 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022184111A1 (en) * 2021-03-03 2022-09-09 上海强睿生物科技有限公司 Small molecule compound bound to tau protein
CN115385908B (en) * 2022-07-26 2024-04-12 阿茨可利(杭州)医学科技有限公司 Small molecule chimera for targeted promotion of tau protein dephosphorylation and application thereof

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU2020139890A (en) * 2014-04-14 2022-01-18 Арвинас Оперэйшнз, Инк. IMIDE MODULATORS OF PROTEOLYSIS AND METHODS FOR THEIR APPLICATION
BR112017028394A2 (en) * 2015-07-10 2018-08-28 Arvinas Inc compound, composition, and methods for inducing degradation of a target protein in a cell and for treating a diseased condition or a diseased condition.
KR102570992B1 (en) * 2016-11-01 2023-08-28 아비나스 오퍼레이션스, 인코포레이티드 Tau-Protein Targeting PROTAC and Related Methods of Use
CA3105506A1 (en) * 2018-07-11 2020-01-16 H. Lee Moffitt Cancer Center And Research Institute, Inc. Dimeric immuno-modulatory compounds against cereblon-based mechanisms
CN110713480B (en) * 2019-10-28 2021-02-26 浙江省医学科学院 AChE protein degradation product and preparation method and application thereof

Also Published As

Publication number Publication date
WO2021160012A1 (en) 2021-08-19
CN115279751B (en) 2023-12-12
CN115279751A (en) 2022-11-01

Similar Documents

Publication Publication Date Title
US9790219B2 (en) Compounds, compositions, and methods for increasing CFTR activity
ES2915833T3 (en) Isoindoline compositions and methods for treating neurodegenerative disease and macular degeneration
US10344023B2 (en) Derivatives of 3-heteroarylisoxazol-5-carboxylic amide useful for the treatment of inter alia cystic fibrosis
JP7419486B2 (en) How to inhibit τ phosphorylation
BRPI0608910A2 (en) use of a compound of the formula or a pharmaceutically acceptable salt or hydrate thereof, compound or salt or hydrate thereof, pharmaceutical composition and packaged pharmaceutical composition
JP2008533155A (en) Alpha-helix mimetics and methods for treatment of fibrosis
CN107082770A (en) The alpha inhibitors of IRE 1
CN113248484A (en) Small molecular compound for specifically degrading tau protein and application thereof
US20160184383A1 (en) Method of treating cancer
US11345738B2 (en) Compounds for treating neurodegenerative proteinopathies
CN112047935B (en) Autophagy targeting protein degradation technology and application thereof
JP2020535164A (en) Compositions and Methods of Inhibiting ACSS2
WO2020093370A1 (en) Micromolecular compound specifically degrading tau protein, and application thereof
CN115385908B (en) Small molecule chimera for targeted promotion of tau protein dephosphorylation and application thereof
CN111171113B (en) Small molecular compound for specifically degrading tau protein and application thereof
WO2022104636A1 (en) Autophagic targeted protein degradation technique and use thereof
US20230322853A1 (en) Compounds and compositions for the treatment of proliferative diseases and disorders
US20190055553A1 (en) Methods for identifying and targeting non-coding rna scaffolds
US10947207B2 (en) GP130 modulators
WO2023083291A1 (en) Derivative of aspartic acid and use thereof in treatment of metabolic diseases such as hepatic fibrosis and non-alcoholic hepatitis
CA3228299A1 (en) Lpar1 antagonists and uses thereof
CA3231240A1 (en) Ink4 tumor suppressor proteins mediate resistance to cdk4/6 kinase inhibitors
JP2024516613A (en) Cyclic peptide-N-acetylgalactosamine (GalNAc) conjugates for drug delivery to liver cells - Patent Application 20070233633
WO2023044315A2 (en) Kca3.1 inhibitors for podocyte protection
TW202334148A (en) Compounds and methods for treating friedreich's ataxia

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
WD01 Invention patent application deemed withdrawn after publication

Application publication date: 20210813

WD01 Invention patent application deemed withdrawn after publication