GB2601520A - Method of survival and protection of neurons by inhibitors of RNA m6A demethylases FTO and ALKBH5 - Google Patents

Method of survival and protection of neurons by inhibitors of RNA m6A demethylases FTO and ALKBH5 Download PDF

Info

Publication number
GB2601520A
GB2601520A GB2019041.9A GB202019041A GB2601520A GB 2601520 A GB2601520 A GB 2601520A GB 202019041 A GB202019041 A GB 202019041A GB 2601520 A GB2601520 A GB 2601520A
Authority
GB
United Kingdom
Prior art keywords
compound
formula
rna
aryl
alkyl
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
GB2019041.9A
Other versions
GB202019041D0 (en
Inventor
Saarma Mart
Yu Li-Ying
Seli Neinar
Selberg Simona
Karelson Mati
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.)
Chemestmed Ltd
Chemestmed Ltd
Original Assignee
Chemestmed Ltd
Chemestmed 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 Chemestmed Ltd, Chemestmed Ltd filed Critical Chemestmed Ltd
Priority to GB2019041.9A priority Critical patent/GB2601520A/en
Publication of GB202019041D0 publication Critical patent/GB202019041D0/en
Publication of GB2601520A publication Critical patent/GB2601520A/en
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/15Oximes (>C=N—O—); Hydrazines (>N—N<); Hydrazones (>N—N=) ; Imines (C—N=C)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/192Carboxylic acids, e.g. valproic acid having aromatic groups, e.g. sulindac, 2-aryl-propionic acids, ethacrynic acid 
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/47064-Aminoquinolines; 8-Aminoquinolines, e.g. chloroquine, primaquine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/50Pyridazines; Hydrogenated pyridazines
    • A61K31/501Pyridazines; Hydrogenated pyridazines not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/08Drugs for disorders of the alimentary tract or the digestive system for nausea, cinetosis or vertigo; Antiemetics
    • 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
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • 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
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • A61P25/32Alcohol-abuse
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • A61P25/36Opioid-abuse
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Abstract

A method of support of dopaminergic neuron survival using an RNA methylation modulator compound by inhibiting an RNA m6A demethylase, wherein the RNA m6A demethylase is fat mass and obesity-associated protein (FTO) or Alpha-ketoglutarate-dependent dioxygenase alkB homolog 5 (AlkBH5). In one aspect, the RNA m6A demethylase is FTO and the compound is of Formula (I) or (II) or a salt thereof: wherein for a compound of formula (I), R1 is amino-group or halogen; R2 is H, alkyl, aryl, or aralkyl; and wherein for a compound of formula (II), R1 and R2 are independently, H, amino-group or halogen; R3 is H, alkyl, aryl, or aralkyl. In another aspect, the RNA m6A demethylase is AlkBH5 and the compound is of Formula (III) or (IV) or a salt thereof: wherein for a compound of formula (III), R1 and R2 are H, alkyl, aryl, aralkyl, acyl, alkoxycarbonyl, aryloxycarbonyl, aralkoxycarbonyl, carbamoyl, alkylcarbamoyl, dialkylcarbamoyl, aminoalkyl or aminoalkyl; wherein for a compound of formula (IV), R1 is H, alkyl, aryl, aralkyl, acyl, alkoxycarbonyl, aryloxycarbonyl, aralkoxycarbonyl, carbamoyl, alkylcarbamoyl, dialkylcarbamoyl, aminoalkyl or aminoalaryl. These compounds are useful in the treatment of Parkinson's disease, amyotrophic lateral sclerosis, Alzheimer’s disease, drug-addiction, traumatic brain injury, psychosis, depression, multiple sclerosis and nausea.

Description

METHOD OF SURVIVAL AND PROTECTION OF NEURONS BY INHIBITORS OF
RNA m6A DEMETHYLASES FTO AND ALKBH5 Inventors: Mart Saa1777(1, Li-Ying Yu, Neinar Self, Si177011a Se/berg, Mali Kate/son Assignee: Chemestmed, Ltd.
TECHNICAL FIELD
The presently disclosed subject matter generally relates to the epitrancriptomic regulation of ribonucleic acid (RNA) methylation through the use of small-molecule inhibitors of the RNA m6A demethylases FTO and AlkBH5, and their application to support the neuronal cell survival and neuroprotection.
BACKGROUND ART
It has been shown that the RNA m6A modification is important in brain development, neuron signalling and neuronal disorders (Angelova, et al, 2018, Jung, et al, 2018; Noack, et al, 2019; Widagdo, et al, 2018; Du, et al, 2019; Livneh, et al, 2020). For instance, it has been demonstrated that m6A-dependent mRNA decay is critical for proper transcriptional prepatterning in mammalian cortical neurogenesis (Yoon, et al, 2017) Weng et al had uncovered an epitranscriptom c mechanism wherein axonal injury elevates m6A levels and signaling to promote protein translation, including regeneration-associated genes, which is essential for functional axon regeneration of peripheral sensory neurons (Weng, et al, 2018). It has been shown on the limited patient group (Chinese Han population) that m6A genes may play a role in conferring risk of dementia (Du, et al, 2015). Different m6A regulating enzymes are involved directly in the processes of learning and memory. Through its binding protein YTHDF1, m6A promotes protein translation of target transcripts in response to neuronal stimuli in the adult mouse hippocampus, thereby facilitating learning and memory (Shi, et al, 2018). Similarly, the RNA m6A demethylase FTO is expressed in adult neural stem cells and neurons and displays dynamic expression during postnatal neurodevelopment. It has been demonstrated that the deficiency of FTO could reduce the proliferation and neuronal differentiation of adult neural stem cells in vivo, leading to impaired learning and memory (Li, et al, 2017) In addition, the inactivation of the Ito gene, encoding a nucleic acid demethylase, impairs dopamine receptor type 2 (D2R) and type 3 (D3R)-dependent control of neuronal activity and behavioural responses (Hess, et al, 2013). The m6A homeostasis in neurons are controlled by more than one regulator. Thus, it was found that global m6A modification of mRNAs is down-regulated in 6-OFDA-induced rat pheochromocytoma PC12 cells originating from adrenal medulla and the striatum of 6-0HDA treated rat brain that is the animal model of Parkinson's disease (PD). The reduction of the m6A level in PC12 cells by. The reduction of the m6A level in PC12 cells by overexpressing a nucleic acid demethylase, FTO, or by m6A inhibitor cycloleucine could induce the expression of N-methyl-D-aspartate (NMDA) receptor 1, and elevate oxidative stress and Ca2+ influx, resulting in PC12 neuron apoptosis (Chen, et al, 2019). Based on above, the dysregulation of the m6A can possibly be related to neurodegeneration in Parkinson's disease. In summary, either the compensatory upregulation or downregulation of m6A could be needed in neuronal cells, depending on their physiological or pathological state.
It has been shown (Chen, et al, 2019) that the downregulation of m6A is involved in dopamine neuronal death. Therefore, the inhibitors of the RNA m6A demethylases FTO or AlkBH5 should support the homeostasis of m6A in dopamine neurons, support their survival in stress conditions and protect them from neurotoxin-induced cell death.
SUMMARY OF INVENTION
The present invention is related to a method of supporting the survival of the neuronal cells and protecting them from cell death induced by neurotoxins or injury using the inhibitors of the RNA m6A demethylases FTO and A1kBH5. Also disclosed are the compounds, or salts or esters thereof, which can protect and support the survival of the neuronal cells. The compounds described can be used for treating Parkinson's disease, amyotrophic lateral sclerosis, Alzheimer's disease, drug addiction, traumatic brain injury, psychosis, depression, multiple sclerosis and nausea by administering to the subject one or more compounds, or a pharmaceutically acceptable salt thereof The "summary of invention" heading is not intended to be restrictive or limiting. The invention also includes all aspects described in the detailed description or figures as originally filed. The original claims appended hereto also define aspects that are contemplated as the invention and are incorporated into this summary by reference.
In addition to the foregoing, the invention includes, as an additional aspect, all embodiments of the invention narrower in scope in any way than the variations specifically mentioned above. For example, although aspects of the invention may have been described by reference to a genus or a range of values for brevity, it should be understood that each member of the genus and each value or sub-range within the range is intended as an aspect of the invention. Likewise, various aspects and features of the invention can be combined, creating additional aspects which are intended to be within the scope of the invention Although the applicant(s) invented the full scope of the claims appended hereto, the claims appended hereto are not intended to encompass within their scope the prior art work of others. Therefore, in the event that statutory prior art within the scope of a claim is brought to the attention of the applicants by a Patent Office or other entity or individual, the applicant(s) reserve the right to exercise amendment rights under applicable patent laws to redefine the subject matter of such a claim to specifically exclude such statutory prior art or obvious variations of statutory prior art from the scope of such a claim. Variations of the invention defined by such amended claims also are intended as aspects of the invention.
BRIEF DESCRIP l'ION OF DRAWINGS Figure 1 The binding site of the compound (V) (FTO inhibitor) to FTO protein Figure 2. The inhibitory effect IE of the compound (V) on the demethylation of the probe RNA by FTO.
Figure 3. The inhibitory effect LE of the compound (VI) on the demethylation of the probe RNA by FTO.
Figure 4. The effect of the FTO inhibitors (V) and (VI) on the survival of the dopaminergic neurons.
Figure 5. The effect of the A1kBH5 inhibitors (VII) and (VIII) on the survival of the dopam nergic neurons.
DETAILED DESCRIPTION OF IN VENT1ON
Disclosed herein are compounds and methods of supporting the survival of the neuronal cells and protecting them from neurotoxin-or injury-induced cell death by using. the RNA m6A methyltransferase complex NIETTL3/NIETTL14/WTAP activators. In some variations of the invention, the compound is administered in a composition that also includes one or more pharmaceutically acceptable diluents, adjuvants, or carriers The compound can be a small molecule. In some embodiments, the compound supporting the survival of neuronal cells has a structure of Formula (I), wherein: RI is amino-group or halogen; and R2 is selected from the group consisting of H, alkyl, aryl, or aralkyl,; or a pharmaceutically acceptable salt thereof In some embodiments, the compound supporting the survival of neuronal cells has a structure of Formula (II) R, (H) wherein: R1 and R2 are independently, hydrogen, amino-group or halogen;; and R3 is selected from the group consisting of H, alkyl, aryl, or aralkyl,, or a pharmaceutically acceptable salt thereof In some embodiments, the compound supporting the survival of neuronal cells has a structure of Formula (III) wherein: R1 and R2 are independently selected from the group consisting of H, alkyl, aryl, aralkyl, acyl, al k oxycarbonyl, aryl oxvcarbonyl, aralkoxycarbonyl, carb am oyl, al kyl carbamoyl, and di al kyl carb am oyl, aminoalkyl, ami noal aryl; or a pharmaceutically acceptable salt thereof In some embodiments, the compound supporting the survival of neuronal cells has a structure of Formula (IV) (IV) wherein: RI is selected from the group consisting of H, alkyl, aryl, aralkyl, acyl, alkoxycarbonyl, aryloxycarbonyl, aralkoxycarbonyl, carbamoyl, alkyl carbamoyl, and dialkylcarbamoyl, aminoalkyl, aminoalaryl; or a pharmaceutically acceptable salt thereof Ri In some embodiments, the compound supporting the survival of neuronal cells has a structure of Formula (V) (V) or a pharmaceutically acceptable salt thereof In some embodiments, the compound supporting the survival of neuronal cells has a structure of Formula (VI) (VI) or a pharmaceutically acceptable salt thereof In some embodiments, the compound supporting the survival of neuronal cells has a structure of Formula (VII) (VII) or a pharmaceutically acceptable salt thereof In some embodiments, the compound supporting the survival of neuronal cells has a structure of Formula (VIII) (VIII) or a pharmaceutically acceptable salt thereof The compounds described can be used for treating Parkinson's disease, amyotrophic lateral sclerosis, Alzheimer's disease, drug addiction, traumatic brain injury, psychosis, depression, multiple sclerosis and nausea by administering to the subject one or more compounds, or a pharmaceutically acceptable salt thereof As used herein, the term "alkyl" refers to straight chained and branched hydrocarbon groups containing carbon atoms, typically methyl, ethyl, and straight chain and branched propyl and butyl groups. Unless otherwise indicated, the hydrocarbon group can contain up to 20 carbon atoms. The term "alkyl" includes "bridged alkyl," i.e., a C.sub.6-C.sub.16 bicyclic or polycyclic hydrocarbon group, for example, norbomyl, adamantyl, bicyclo[2.2.2]octyl, bicyclo[2.2.1]heptyl, bicyclop.2. lloctyl, or decahydronaphthyl. Alkyl groups optionally can be substituted, for example, with hydroxy (OH), halo, amino, and sulfonyl. An "alkoxy" group is an alkyl group haying an oxygen substituent, e.g., -0-alkyl.
The term "alkenyl" refers to straight chained and branched hydrocarbon groups containing carbon atoms haying at least one carbon-carbon double bond. Unless otherwise indicated, the hydrocarbon group can contain up to 20 carbon atoms. Alkenyl groups can optionally be substituted, for example, with hydroxy (OH), halo, amino, and sulfonyl.
As used herein, the term "alkylene" refers to an alkyl group haying a further defined substituent. For example, the term "a1kylenearyl" refers to an alkyl group substituted with an aryl group, and "alkyleneamino" refers to an alkyl groups substituted with an amino group.
The amino group of the alkyleneamino can be further substituted with, e.g., an alkyl group, an alkylenearyl group, an aryl group, or combinations thereof The term "alkenylene" refers to an alkenyl group having a further defined substituent As used herein, the term "aryl" refers to a monocyclic or polycyclic aromatic group, preferably a monocyclic or bicyclic aromatic group, e.g., phenyl or naphthyl. Unless otherwise indicated, an aryl group can be unsubstituted or substituted with one or more, and in particular one to four groups independently selected from, for example, halo, alkyl, alkenyl, OCF.sub.3, NO.sub.2, CN, NC, OH, alkoxy, amino, CO.sub.2H, CO.sub.2alkyl, aryl, and heteroaryl. Exemplary aryl groups include, but are not limited to, phenyl, naphthyl, tetrahydronaphthyl, chlorophenyl, methylphenyl, methoxyphenyl, trifluoromethylphenyl, nitrophenyl, 2,4-methoxychlorophenyl, and the like. An "aryloxy" group is an aryl group haying an oxygen substituent, e.g., -0-aryl.
As used herein, the term "acyl" refers to a carbonyl group, e.g., C(0). The acyl group is further substituted with, for example, hydrogen, an alkyl, an alkenyl, an aryl, an alkenylaryl, an alkoxy, or an amino group. Specific examples of acyl groups include, but are not limited to, al koxycarb onyl (e.g., C(0)--Oalkyl); aryl oxycarbonyl (e.g., C(0)--Oaryl); alkylenearyloxycarbonyl (e.g., C(0)--OalkylenearyI); carbamoyl (e.g., C(0)--NH.sub.2); alkylcarbamoyl (e.g., C(0)--NH(alkyl)) or dialkylcarbamoyl (e.g., C(0)--NH(alkyl).sub.2).
As used herein, the term "amino" refers to a nitrogen containing substituent, which can have zero, one, or two alkyl, alkenyl, aryl, alkylenearyl, or acyl substituents. An amino group having zero substituents is --NH.sub.2.
As used herein, the term "halo" or "halogen" refers to fluoride, bromide, iodide, or chloride.
As used herein, the term "pharmaceutically acceptable salt" refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, S. M. Berge, et al, describes pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 66: 1-19 (1977). The salts can be prepared in situ during the final isolation and purification of the compounds of the invention, or separately by reacting the free base function with a suitable organic acid or inorganic acid. Examples of pharmaceutically acceptable nontoxic acid addition salts include, but are not limited to, salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, maleic acid, tartaric acid, citric acid, succinic acid lactobionic acid or malonic acid or by using other methods used in the art such as ion exchange. Other pharmaceutically acceptable salts include, but are not limited to, adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsul fon ate, citrate, cyclopentanepropi onate, di glucon ate, dodecyl sulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, m al ate, m al eate, m al onate, m ethanesul fon ate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, p-toluenesulfonate, undecanoate, valerate salts, and the like.
Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, alkyl having from 1 to 6 carbon atoms, sulfonate and aryl sulfonate.
EXAMPLES
The following Examples have been included to provide illustrations of the presently disclosed subject matter. In light of the present disclosure and the general level of skill in the art, those of skill will appreciate that the following Examples are intended to be exemplary only and that numerous changes, modifications and alterations can be employed without departing from the spirit and scope of the presently disclosed subject matter.
Example I. High-throughput virtual screening of compound libraries for the identification of 10 potential inhibitors of the RNA m6A demethylase FTO The virtual screening of the compound libraries requires the availability of the 3D structure of the target protein, in this case FTO. The crystal structure of the RNA demethylase FTO has been published as for the pure protein (Han, et al, 2010) and together with different ligands (Aik, et al, 2013; Huang, et al, 2014; Toh, et al, 2015). Crystal structure of FTO in complex with 5-carboxy-8-hydroxyquinoline (pdb: 41E4) was chosen for further molecular docking modeling. The structure had been determined using X-ray diffraction with the resolution 2.5 A. (Aik, et al, 2013). The choice of this FTO structure was made on the basis of the availability of the ligand compounds. The raw crystal structure was corrected and hydrogen atoms were automatically added to the protein using Schredinger's Protein Preparation Wizard of Maestro 10.7 (Sastry, et al, 2013).
AutoDock 4.2 (Morris et al., 2009) was used for the docking studies to find out binding modes and binding energies of ligands to the receptor. The number of rotatable bonds of ligand was set by default by AutoDock Tools 1.5.6 (Morris et al., 2009). However, if the number was greater than 6, then some of rotatable bonds were made as non-rotatable, otherwise calculations can be inaccurate. The active site was surrounded with a grid-box sized x 80 x 80 points with spacing of 0.375 A°. The AutoDock 4.2 force field was used in all molecular docking simulations. The docking efficiencies (DE) were calculated as follows DE= (AG dock)/N (1) where AG dock is the docking free energy and N -the number of non-hydrogen ("heavy") atoms in the ligand molecule. The structure of ligand molecules was optimized using the density functional theory B3LYP method with 6-31G basis set (Stephens, et al, 1994) The virtual screening was carried out on ZINC database (Irwin, et al, 2005). The structures and binding free energies of the compounds with the highest docking efficiencies to FTO protein are given in Table 1. The structures of the binding complex of the compound (V) in the FTO active centre and is presented on Figure 1.
Example 2. Screening of computationally predicted RNA m6A demethylase FTO lic,,ands in enzyme inhibition assay.
The enzymatic assay was modified from Huang et al. 2015 (Huang et al., 2015). The experiments were conducted in reaction buffer (50 mN1 Tris-HC1, pH 7.5, 300 gN1 20G, 280 RIM (NH4)2Fe(SO4)2 and 2 mIYI L-ascorbic acid). The reaction mixture contained 200 ng methylated N6-adenine RNA probe (SEQ ID NO: 1) (5'-CUUGUCAm6ACAGCAGA-3', Dharmacon) and I OnIVI FTO protein. Reactions were incubated on 96-well plate for 2h at RT.
After that, the amount of m6A that was measured using EpiQuik m6A RNA methylation Quantification Colorimetric Kit (Epigentek).
The inhibitory effect IF of compounds on RNA probe demethylation by FTO was calculated as the enhancement of the m6A amount as compared to the negative control (DMSO) relative to the difference between m6A amounts of the positive control (max inhibition) and the negative control (eq. (2)): IN - (2) where (7,"1" C,"h(max) and &Ivy() are the amounts of m6A at a given concentration of the inhibitor, maximum inhibition and in the case of JAMS°, respectively. The dependence of the 1E on the inhibitor concentration for the compound (V) is shown on Fig. 2 and for the compound (VI) on Fig. 3. The inhibitory concentrations are IC50 = 28.9 OM for the compound (V) and IC50 = 1.46 JM for the compound (VI), respectively. Therefore, both compounds are efficient inhibitors of the RNA m6A demethylase FTO.
Example 3 Treatment of primary cultures of midbrain dopaminergic neurons by m6A 25 inhibitors of RNA m6A demethylases FTO and A1kBH5.
The midbrain floors were dissected from the ventral mesencephalic of 13 days old NMRI strain mouse embryos. The tissues were incubated with 0.5% trypsin (103139, MP Biomedical) in HESS (Ca2+/Mg2+-free) (14170112, Invitrogen) for 20 mm at 37°C, then mechanically dissociated. Cells were plated onto the 96-well plates coated with poly-L-omithine (Sigma-Aldrich). Equal volumes of cell suspension were plated onto the center of the dish. The cells were grown for 5 days with different concentrations of FTO or AlkBH5 inhibitor compounds separately. Human recombinant GDNF (100 ng/ml) (Icosagen) or a medium without any neurotrophic compound added were used as positive and negative controls, respectively.
After growing 5 days, the neuronal cultures were fixed and stained with anti-Tyrosine Hydroxylase antibody (MAB318, Millipore Bioscience Research Reagents. Images were acquired by CellInsight high-content imaging equipment. Immunopositive neurons were counted by CellProfiler software and the data were analyzed using CellProfiler analyst software. (Yu, et al, 2008; Mahato et al, 2020). The measurements were carried out with the FTO inhibitors (compounds (V) and (VI)) and AlkBH5 inhibitors (compounds (VII) and (VIII)) (Selberg, et al, 2019). The results are expressed as % of cell survival compared to GDNF-maintained neurons (Figures 4 and 5). It can be seen that the compounds have some effect already at the 10 n1VI concentration and comparable to the neurotrophic factor GDNF effect at their 1 000 nM concentration. Therefore, these compounds are efficiently supporting the survival of neurons at growth factor deprivation-induced stress conditions.
Table I. The compounds with the highest docking efficiencies to FTO protein Compound structure PG DE (kcal/mol) t+..-..".....,...- -7.37 0.53 (VI) -7.70 0.51 1,-, (V) -7.03 0.50
---
i i -8.78 0.49 : H r.1"'""'N"."---" 1 t.....,, ....._ -....--."%.. -7.17 0.48 "-- :i 1 E, +.-....,,,",..) -9.45 0.47
REFERENCES CITED
Aik, W.S. Demetriades, M.; Hamdan, M.K.K.; Bagg, E.A.L.; Yeoh, K.K.; Lejeune, C.; Zhang, Z.; McDonough, M.A.; Schofield, C.J. (2013). Structural basis for Inhibition of the Fat Mass and Obesity Associated Protein (FTO). J. Med. Chem, 56, 3680-3688.
Aik, W.; Scotti, LS.; Choi, H.; Gong, L.; Demetriades, M.; Schofield, C.J.; McDonough, M.A. (2014)Structure of human RNA N6-methyladenine demethylase ALKBH5 provides insights into its mechanisms of nucleic acid recognition and dem ethyl ati on. Nucleic Acids Res. 42, 4741-54.
Angelova M.T, Dimitrova D.G, Dinges N, Lence T, Worpenberg L, Cane C, Roignant J.Y. (2018). The Emerging Field of Epitranscriptomics in Neurodevelopmental and Neuronal Disorders. Front. Bioeng. Biotechnol. 6, 46.
Chen X, Yu C, Guo Ni, Zheng X, Ali S, Huang H, Zhang L, Wang S, Huang Y, Qie S, Wang J. (2019) Down-Regulation of m6A mRNA Methylation Is Involved in Dopaminergic Neuronal Death. ACS Chem Neurosci. 10, 2355-2363.
Du, T.; Rao, S.; Wu, L.; Ye, N.; Liu, Z.; Hu, H.; Xiu, J.; Shen, Y.; Xu, Q. (2015) An association study of the m6A genes with major depressive disorder in Chinese Han population. J Affect Di sord. 183, 279-86.
Du, K; Zhang, L; Lee, T, Sun, T. (2019). m6A RNA Methylation Controls Neural 20 Development and Is Involved in Human Diseases. Mol, Neurobiol. 56, 1596-1606.
Feng, C.; Liu, Y.; Wang, G., Deng, Z.; Zhang, Q.; Wu, W.; Tong, Y.; Cheng, C., Chen, Z. (2014). Crystal structures of the human RNA demethylase Alkbh5 reveal basis for substrate recognition. J. Biol. Chem. 289, 11571-83.
Han, Z.; Niu, T.; Chang, J., Lei, X., Zhao, M., Wang, Q.; Cheng, W.; Wang, J., Feng, Y., Chai, J. (2010). Crystal Structure of the FTO Protein Reveals Basis for Its Substrate Specificity. Nature. 464, 1205-1209, Huang, Y.; Yan, I; Li, Q.; Li, J.; Gong, S.; Zhou, H.; Gan, J.; Jiang, H.; Ea, OF.; Luo, C.; Yang, C.-G. (2014). Meclofenamic Acid Selectively inhibits FTO Demethylation of m6A over ALKBH5. Nucleic Acids Res. 43, 373-384, Hess, M. E., Hess, S., Meyer, K. D., Verhagen, L. A. W., Koch, L., Bronneke, H. S., Dietrich, M. 0., Jordan, S. D., Saletore, Y., Elemento, 0., Belgardt, B. F., Franz, T., Horvath, T. L., Ruther, U., Jaffrey, S. R., Kloppenburg, P., and Bruning, J. C. (2013) The fat mass and obesity associated gene (Fto) regulates activity of the dopaminergic midbrain circuitry. Nat. Neurosci. 16, 1042-1048.
Huang J., Yan J., Li Q., Li J., Gong S., Zhou H., Gan, J., Jiang H., Jia C-F., Luo C., Yang C-G. (2015). Meclofenamic acid selectively inhibits FTO demethylation of m6A over ALKBH5. Nucleic Acids Research, 43, 373-384.
Irwin, H.; Shoichet, B.K. (2005). ZINC -A Free Database of Commercially Available Compounds for Virtual Screening, J. Chem. Inf. Model. 45, 177-182, Jung Y.; Goldman D. (2018). Role of RNA modifications in brain and behavior. Genes Brain Behav. 17, e12444.
Li, L., Zang, L., Zhang, F., Chen, J., Shen, H., Shu, L., Liang, F., Feng, C., Chen, D., Tao, H., Xu, T., Li, Z., Kang, Y., Wu, H., Tang, L., Zhang, P., Sin, P., Shu, Q., and Li, X. (2017) Fat mass and obesity associated (FTO) protein regulates adult neurogenesis. Hum. Mol, Genet, 25 26,2398-24]], Livneh, I.; Moshitch-Moshkovitz, S.; Amariglio, N. et al. The m6A epitranscriptome: transcriptome plasticity in brain development and function. Nat Rev Neurosci 21, 36-51 (2020).
Mahato AK, Kopra J, Renko JM, Visnapuu T, Korhonen I, Pulkkinen N, Bespalov MM, 30 Domanskyi A, Ronken E, Piepponen 'FP, Voutilainen MB, Tuominen RK, Karelson M, Sidorova YA, Saarma M. (2020) Glial cell line-derived neurotroph c factor receptor
H
Rearranged during transfection agonist supports dopamine neurons in vitro and enhances dopamine release in vivo. N4av Di sord. 35, 245-255.
Morris, G. NI.; Huey, R.; Lindstrom, W., Sanner, NI, F.; Belew, R. K., Goodsell, D. S.; Olson, A. J. (2009). Autodock4 and AutoDockTools4: Automated Docking with Selective Receptor Flexiblity. J. Comput. Chem. 16, 2785-91.
Noack F.; Calegari F. (2018). Epitranscriptomics: A New Regulatory Mechanism of Brain Development and Function. Front. Neurosci. 12, 85.
Sastry, G. Ni,; Adzhigirey, Ni.; Day, T.; Annabhimoju, R.; Sherman, W. (2013). Protein and Ligand Preparation: Parameters, Protocols, and influence on Virtual Screening Enrichments.
J. Comput. Aid. NIol. Des. 27, 221-234.
Selberg, S., Karelson, (2019). Ni. Method of suppressing cancer by RNA m6A demethylase AlkBH5 inhibitors, Patent application PCT/EP2019/058737 (07 04 2019) Shi, H., Zhang, X., Weng, Y. L., Lu, Z., Liu, Y., Li, J., Hao, P., Zhang, Y., Zhang, F., Wu, Y., Delgado, J. Y., Su, Y., Patel, M. J., Cao, X., Shen, B., Huang, X., Ming, G. L., Zhuang, X., Song, H., He, C., and Zhou, T. (2018) m(6)A facilitates hippocampus-dependent learning and memory through YTHDFI. Nature 563, 249-253, Stephens, Pt; Devlin, Ft; Chabalowski, C.F.; Frisch, M.J. (1994). Ab Initio Calculation of Vibrational Absorption and Circular Dichroism Spectra Using Density Functional Force Fields, J. Phys. Chem, 98, 11623-11627.
Toh, J.D.W., Sun, L. Lau, L.Z.NI.; Tan, J.; Low, J.J.A.; Qiang, W.; Tang, C.W.Q.; Yi, J.; Cheong, Eli; Tan, NI.J.H.; Chen, Y.; Hong, W.; Gao, Y.G.; Woon, E.C.Y. (2015). A Strategy Based on Nucleotide Specificity Leads to Subfamily-Selective and Cell-Active Inhibitor of N6-NIethyladenosine Demethylase FTO. Chem. Sci. 6, 112-122.
Trott, 0 Olson, A. J. (2010). AutoDock Vina: Improving the Speed and Accuracy of Docking with a New Scoring Function, Efficient Optimization and Multithreading. J. Comput, Chem, 2010, 31, 455-461, Weng, Y. L.; Wang, X.; An, R.; Cassin, J.; Vissers, C.; Liu, Y.; Liu, Y.; Xu, T.; Wang, X.; Wong, S.; Joseph, J.; Dore, L. C.; Dong, Q.; Zheng, W.; Jin, P.; Wu, H.; Shen, B.; Zhuang, X.; He, C.; Liu, K.; Song, H.; Ming, G. L. (2018). Epitranscriptomic m6A Regulation of 30 Axon Regeneration in the Adult Mammalian Nervous System. Neuron, 97(2), 313-325.e6.
N
Widagdo J.; Anggono V. (2018). The m6A-epitranscriptomic signature in neurobiology: from neurodevelopment to brain plasticity. J. Neurochem, 147, 137-152.
Xu, C.; Liu, K.; Tempel, W.; Demetriades, M.; Aik, W.; Schofield, CJ.; Min J. (2014). Structures of human A LKBH5 demethylase reveal a unique binding mode for specific single-stranded N6-methyladenosine RNA demethylation. J. Biol. Chem. 289, 17299-311.
Yoon, K.J. Ringeling, FR.; Vissers, C.; Jacob, F.; Pokrass, M.; Jimenez-Cyrus, D.; Su, Y.; Kim, N. S.; Zhu, Y.; Zheng, L.; Kim, S.; Wang, X.; Dore, L.C.; Jin, P.; Regot, S.; Zhuang, X.; Canzar, S.; He, C.; Ming, G.L.; Song, H. (2017). Temporal Control of Mammalian Cortical Neurogenesis by m6A Methyl ati on. Cell. 171, 877-889.e17.
Yu, L -Y, Saanna, M and Anima; U (2008) Death receptors and caspases but not mitochondria are activated in the GDNF-or BDNF-deprived dopaminergic neurons. J. Neurosci., 28 (30), 7467-7475.

Claims (3)

  1. Claims 1 A method of support of the dopaminergic neuron survival using a compounds that modulate the RNA methylation by inhibiting a RNA m6A demethylase, wherein the RNA m6A demethylase is fat mass and obesity-associated protein (FTO) or Alphaketoglutarate-dependent dioxygenase alkB homolog 5 (A1kBH5).
  2. 2 The method of claim 1, wherein the RNA m6A demethylase is fat mass and obesity-associated protein (FTO), and the compound is a small molecule having a structure of Formula (I) wherein: R1 is amino-group or halogen; and R2 is selected from the group consisting of H, alkyl, aryl, or aralkyl,; or a pharmaceutically acceptable salt thereof.
  3. 3 The method of claim 2, wherein the compound has a structure of Formula (II) wherein: R1 and R2 are independently, hydrogen, amino-group or halogen;; and R3 is selected from the group consisting of H, alkyl, aryl, or aralkyl,, or a pharmaceutically acceptable salt thereof 4 The method of claim 1, wherein the RNA m6A demethylase is Alpha-ketoglutaratedependent dioxygenase a1kB homolog 5 (A11cBH5) and the compound is a small molecule having a structure of Formula (Ill) wherein: RI and R2 are independently selected from the group consisting of H, alkyl, aryl, aralkyl, acyl, al koxycarbonyl, aryl oxycarbonyl, aral koxycarbonyl, carb am oyl, alkylcarbamoyl, and dialkylcarbamoyl, aminoalkyl, aminoalaryl; or a pharmaceutically acceptable salt thereof The method of claim 4, wherein the compound has a structure of Formula (IV) (IV) wherein: RI is selected from the group consisting of H, alkyl, aryl, aralkyl, acyl, alkoxycarbonyl, aryloxycarbonyl, aralkoxycarbonyl, carbamoyl, alkylcarbamoyl, and di alkyl carbamoyl, aminoalkyl, aminoal aryl; or a pharmaceutically acceptable salt thereof.6 The method of claim 2, wherein the compound has a structure of Formula (V) (V) 7. The method of claim 2, wherein the compound has a structure of Formula (VI) (VI) 8 The method of claim 4, wherein the compound has a structure of Formula (VII) (VII) Vi 9. The method of claim 4, wherein the compound has a structure of Formula (VIII) (VIII) A method of treating Parkinson's disease, amyotrophic lateral sclerosis, Alzheimer's disease, drug addiction, traumatic brain injury, psychosis, depression, multiple sclerosis and nausea by administering to the subject one or more compounds, or a pharmaceutically acceptable salt thereof, of claim 1.
GB2019041.9A 2020-12-02 2020-12-02 Method of survival and protection of neurons by inhibitors of RNA m6A demethylases FTO and ALKBH5 Pending GB2601520A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
GB2019041.9A GB2601520A (en) 2020-12-02 2020-12-02 Method of survival and protection of neurons by inhibitors of RNA m6A demethylases FTO and ALKBH5

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
GB2019041.9A GB2601520A (en) 2020-12-02 2020-12-02 Method of survival and protection of neurons by inhibitors of RNA m6A demethylases FTO and ALKBH5

Publications (2)

Publication Number Publication Date
GB202019041D0 GB202019041D0 (en) 2021-01-13
GB2601520A true GB2601520A (en) 2022-06-08

Family

ID=74099702

Family Applications (1)

Application Number Title Priority Date Filing Date
GB2019041.9A Pending GB2601520A (en) 2020-12-02 2020-12-02 Method of survival and protection of neurons by inhibitors of RNA m6A demethylases FTO and ALKBH5

Country Status (1)

Country Link
GB (1) GB2601520A (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003093240A1 (en) * 2002-04-30 2003-11-13 Yungjin Pharmaceutical Co., Ltd. Quinoline derivatives as caspase-3 inhibitor, preparation for producing the same and pharmaceutical composition comprising the same
WO2010007085A2 (en) * 2008-07-17 2010-01-21 Novartis Ag Use of organic compounds
WO2019084271A1 (en) * 2017-10-25 2019-05-02 Children's Medical Center Corporation Papd5 inhibitors and methods of use thereof
WO2020207550A1 (en) * 2019-04-07 2020-10-15 Chemestmed Ltd. METHOD OF SUPPRESSING CANCER BY RNA m6A DEMETHYLASE AlkBH5 INHIBITORS

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003093240A1 (en) * 2002-04-30 2003-11-13 Yungjin Pharmaceutical Co., Ltd. Quinoline derivatives as caspase-3 inhibitor, preparation for producing the same and pharmaceutical composition comprising the same
WO2010007085A2 (en) * 2008-07-17 2010-01-21 Novartis Ag Use of organic compounds
WO2019084271A1 (en) * 2017-10-25 2019-05-02 Children's Medical Center Corporation Papd5 inhibitors and methods of use thereof
WO2020207550A1 (en) * 2019-04-07 2020-10-15 Chemestmed Ltd. METHOD OF SUPPRESSING CANCER BY RNA m6A DEMETHYLASE AlkBH5 INHIBITORS

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
European Journal of Medicinal Chemistry, Vol. 27(9), 1992, (Nicolai, E. et al), pages 977-84, ISSN: 0223-5234. *
Organic Chemistry: An Indian Journal, Vol. 10(1), 2014, (Pamar, Malavath Geeta et al), pages 17-23, ISSN: 0974-7516. *
Pharmaceutical Chemistry Journal (Translation of Khimiko-Farmatsevticheskii Zhurnal), Vol. 33(5), 1999, (Kolotova, N. V. et al), pages 248-254, ISSN: 0091-150X. *

Also Published As

Publication number Publication date
GB202019041D0 (en) 2021-01-13

Similar Documents

Publication Publication Date Title
Pavitt Regulation of translation initiation factor eIF2B at the hub of the integrated stress response
Hetz et al. Fine-tuning of the unfolded protein response: assembling the IRE1α interactome
US9642831B2 (en) Substituted chromanes and method of use
Tanaka et al. Autophagy as a therapeutic target in diabetic nephropathy
CN100368411C (en) 7-azaindoles as inhibitors of c-Jun N-terminal kinases for the treatment of neurodegenerative disorders
EP3104706B1 (en) Compositions and methods using the same for treatment of neurodegenerative and mitochondrial disease
EP2298295B1 (en) Tumor necrosis factor inhibitors
Xuan et al. The cuprizone-induced changes in 1H-MRS metabolites and oxidative parameters in C57BL/6 mouse brain: Effects of quetiapine
EP3214935A1 (en) Ezh2 inhibitors and uses thereof
CA2966023A1 (en) Substituted tetrahydropyrans and method of use
AU2004247627A1 (en) Pyrrole compounds and uses thereof
US20210401802A1 (en) Pkc-delta-i inhibitor formulations and uses thereof
CA3072740A1 (en) Pentacyclic compound
GB2601520A (en) Method of survival and protection of neurons by inhibitors of RNA m6A demethylases FTO and ALKBH5
Shao et al. Ecto‑5'‑nucleotidase (CD73) inhibits dorsal root ganglion neuronal apoptosis by promoting the Ado/cAMP/PKA/CREB pathway
WO2020207550A1 (en) METHOD OF SUPPRESSING CANCER BY RNA m6A DEMETHYLASE AlkBH5 INHIBITORS
Mizojiri et al. The identification and pharmacological evaluation of potent, selective and orally available ACC1 inhibitor
WO2012078909A1 (en) Thiazolpyrimidine proteostasis regulators
CN102369192A (en) Derivatives of 6-(6-nh-substituted-triazolopyridazine-sulfanyl) benzothiazoles and benzimidazoles, preparation thereof, use thereof as drugs, and use thereof as met inhibitors
EP3186257B1 (en) Novel chromone oxime derivative and its use as allosteric modulator of metabotropic glutamate receptors
Jiaranaikulwanitch et al. Neuritogenic activity of bi-functional bis-tryptoline triazole
KR20010102463A (en) Use of tianeptin in the production of medicaments to treat neurodegenerative pathologies
EP3281940A1 (en) Heterocyclic naphthoquinones derivatives for use in the treatment of cancers including cushing disease
GB2601519A (en) Method of support of survival and protection of neurons by RNA m6A methyltransferase complex METTL3/METTL14 activators
Luo et al. Downregulation of DDIT4 ameliorates abnormal behaviors in autism by inhibiting ferroptosis via the PI3K/Akt pathway