GB2589912A - Method of suppressing cancer by RNA m6A methyltransferase mettl16 inhibitors - Google Patents

Method of suppressing cancer by RNA m6A methyltransferase mettl16 inhibitors Download PDF

Info

Publication number
GB2589912A
GB2589912A GB1918338.3A GB201918338A GB2589912A GB 2589912 A GB2589912 A GB 2589912A GB 201918338 A GB201918338 A GB 201918338A GB 2589912 A GB2589912 A GB 2589912A
Authority
GB
United Kingdom
Prior art keywords
compound
formula
rna
pharmaceutically acceptable
leukemia
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
GB1918338.3A
Other versions
GB201918338D0 (en
Inventor
Karelson Mati
Seli Neinar
Selberg Simona
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
Original Assignee
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 filed Critical Chemestmed Ltd
Priority to GB1918338.3A priority Critical patent/GB2589912A/en
Publication of GB201918338D0 publication Critical patent/GB201918338D0/en
Priority to PCT/EP2020/085872 priority patent/WO2021116477A1/en
Publication of GB2589912A publication Critical patent/GB2589912A/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
    • 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
    • A61K31/17Amides, e.g. hydroxamic acids having the group >N—C(O)—N< or >N—C(S)—N<, e.g. urea, thiourea, carmustine
    • A61K31/175Amides, e.g. hydroxamic acids having the group >N—C(O)—N< or >N—C(S)—N<, e.g. urea, thiourea, carmustine having the group, >N—C(O)—N=N— or, e.g. carbonohydrazides, carbazones, semicarbazides, semicarbazones; Thioanalogues thereof
    • 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
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4406Non condensed pyridines; Hydrogenated derivatives thereof only substituted in position 3, e.g. zimeldine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Landscapes

  • Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

A pharmaceutical composition comprises a compound of formula (I) as defined herein, or a pharmaceutically effective salt thereof, and an excipient, for use in inhibiting RNA demethylation at the 6-position of adenine (m6A). Preferably the compound has formula (II), (III), (IV) or (V): The composition may be used in the treatment of glioblastoma, astrocytoma, acute myeloid leukemia, acute monocytic leukemia, chronic myelogenous leukemia, or T-acute lymphoblastic leukemia.

Description

METHOD OF SUPPRESSING CANCER BY RNA m6A METHYLTRANSFERASE METTL16 INHIBITORS Inventors: Mati Karelson, Neinar Seli, Simona Se/berg; Assignee: Chemestmed, Ltd.
TECHNICAL FIELD
The present invention relates generally to compositions and methods for treating cancer. Provided herein are compounds are reported that specifically modulate RNA methylation by inhibition of the RNA methyltransferase METTL16. Furthermore, the subject compounds and compositions are useful for the treatment of cancer, such as glioblastoma, astrocytoma, acute myeloid leukemia, acute monocytic leukemia, chronic myelogenous leukemia, T acute lymphoblastic leukemia and the like.
BACKGROUND ART
The presently disclosed subject matter generally relates to the epitrancriptomic regulation of ribonucleic acid (RNA) methylation through small-molecule inhibitors of the RNA m6A methyltransferase METTL16.
Chemical modifications of RNA have been shown to have important effects on critical cellular functions, such as proliferation, survival and differentiation (Helm et al., 2017). The most widespread modification in messenger RNA is N6-methyladenosine (m6A) (Roundtree et al., 2017). It has been shown that m6A modifications of RNA affect its splicing, intracellular distribution, translation, and cytoplasmic degradation, playing thus a crucial role in regulating cell differentiation, neuronal signaling, carcinogenesis and immune tolerance (Maity et al., 2016). The m6A presence in RNA is regulated by specific enzymes, i.e. the RNA methyltransferases, RNA methylases and RNA reader proteins.
The N-methylation of the adenosine is a reversible process, catalysed by specific proteins. (Figure 1). Those include the RNA methyltransferase enzyme complex METTL3/METTL14/VVTAP (Scholler et al, 2018) consisting of the following three components: METTL3 (methyltransferase-like protein 3) (Bokar et.al., 1998), METTL14 (methyltransferase-like protein 14) (Liu et al, 2014), and VVTAP (Wilm's tumour-1-associated protein) (Horiuchi et al, 2013) ; RNA m6A methyltransferase METTL16 (Pendleton et al, 2017); the RNA demethylases FTO (fat mass and obesity-associated protein) (Jia, et al, 2011) and AlkBH5 (AlkB family member 5) (Zheng, et al., 2013), called "erasers". The fate of the RNA in post-transcriptomic processes is also directed by the "reader" enzymes that recognize specific m6A methylation in RNA that include YTHDF1 (YTH N6-Methyladenosine RNA Binding Protein 1), YTHDF2 (YTH N6-Methyladenosine RNA Binding Protein 2) YTHDF3 (YTH N6-Methyladenosine RNA Binding Protein 3), YTHDC1 (YTH domain-containing protein 1) and YTHDC2 (YTH domain-containing protein 2) (Park et al., 2017).
The homodimeric complex of METTL16 (methyltransferase-like protein 16) acts on a specific stem-loop structure of RNA containing the UACAGAGAA sequence. Whereas this complex acts on only some methylated mRNAs and IncRNAs, it targets importantly the mRNA of the human S-adenosylmethionine (SAM) synthetase MAT2A (Pendleton et al., 2017; Shima et al., 2017; Warda et al., 2017) that regulates cellular levels of the methylation donor S-adenosyl-methionine (SAM). Consequently, METTL16 regulate the activity of all cellular methyltransferases and thus indirectly also the level of the m6A in the cell (Doxtader et al, 2018). METTL16 can be therefore be critical in various physiological and pathological processes. It has been show that the regulation of Mat2a mRNA by METTL16 is essential for mouse embryonic development (Mendel et al, 2018). The enhanced expression levels of METTL16 have been associated with the tumorigenesis and clinical outcomes of colon adenocarcinoma patients (Liu et al, 2019a).
The m6A level in the cell has been shown to be strongly related to the pathological processes of cancer (Cui et al., 2017, Li et al., 2017, Wang et al., 2017, Deng et al.., 2018; Yanhong et al, 2018; Delaunay et al., 2019; Sun et al., 2019; Lin et al., 2019; Kandimalla et al., 2019; Fazi et al., 2019). For instance, it has been shown that the reduction of the m6A level by overexpression of the RNA m6A demethylase AlkBH5 significantly promotes cell proliferation in human cervical cancer cell line SiHa (Wang, X. et al. 2017). ALKBH5 has been also reported to promote tumorigenesis and proliferation in glioblastoma stem-like cells (GSCs) (Zhang, S. et al. 2017) and breast cancer stem cells (BCSCs) (Zhang, C. et al. 2016). Contrary to this, ALKBH5 is expressed at a low level in acute myeloid leukemia (AML) (Gao et al, 2013). The functional importance of m6A mRNA modification in GSC self-renewal and glioblastoma tumor progression has been demonstrated by functional studies through manipulating expression of METTL3 or METTL14, or pharmacologically inhibiting activity of FTO in GSCs (Cui et al, 2017). It has been also shown that during epithelialmesenchymal transition in cancer cell metastasis, the m6A modification of mRNAs increases in cancer cells. The deletion of methyltransferase complex protein METTL3 down-regulates m6A and impairs the migration, invasion and EMT of cancer cells both in vitro and in vivo (Lin et al, 2019). The down-regulating METTL3 expression by m iR600 inhibited also lung cancer (Wei et al, 2019).
Based on above, the small-molecule inhibitors of METTL16 are expected to regulate the RNA methylation level in the cell and thus be potentially anti-cancer drugs.
SUMMARY OF INVENTION
The present invention is related to a method of cancer cure by of modulating the RNA methylation at 6-position of adenine (m6A) using effective amount of a compound having binding and/or inhibition of RNA m6A methyltransferase METTL16.
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 DESCRIPTION OF DRAWINGS
The present invention is disclosed further with references to accompanying drawings where: FIG. 1 illustrates dynamic and reversible m6A methylation in RNA (SAM -5-adenosyl-L-methionine; SAH -S-adenosyl-L-homocystein) (Niu, et al., 2013), FIG 2 illustrates the binding site of the compound (II) of the present invention, FIG 3 illustrates the binding site of the compound (III) of the present invention, FIG 4 illustrates the binding site of the compound (IV) of present invention, FIG 5 illustrates the binding site of the compound (V) of present invention, FIG. 6 illustrates the inhibitory effect IF of the compound (III) on the methylation of the probe RNA by METTL16 according to present invention, FIG. 7 illustrates the inhibitory effect IF of the compound (IV) on the methylation of the probe RNA by METTL16 according to present invention, FIG. 8 illustrates the inhibitory effect INH% of the compound (III) at 1 pM concentration on the glioblastoma cell culture A-172 of the present invention, FIG. 9 illustrates the time dependence of the inhibitory effect INH% of the compound (II) at 1 pM concentration on the adult acute myeloid leukemia (AML) cell culture HL-60 according to present invention, FIG. 10 illustrates the time dependence of the inhibitory effect INH% of the compound (III) at 1 pM concentration on the adult acute myeloid leukemia (AML) cell culture HL-according to present invention.
DETAILED DESCRIPTION OF INVENTION
Disclosed herein are compounds and methods of modulating the RNA methylation through inhibition of the RNA m6A methyltransferase METTL16. 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, RNA m6A methyltransferase METTL16 inhibitor has a structure of Formula (I), wherein: R1 is independently selected from the group consisting aryl or heteroary1;, R2 and R3 are independently selected from the group consisting of H, alkyl, aryl, aralkyl, acyl, and R4 is either a oxygen or a sulfur atom; or a pharmaceutically acceptable salt thereof.
In some embodiments, the RNA m6A methyltransferase METTL16 inhibitor compound has a structure of Formula (II) In some embodiments, the METTL16 complex activator compound has a structure of Formula (111) In some embodiments, the METTL16 complex activator compound has a structure of Formula (IV) NI-12 (IV) In some embodiments, the METTL16 complex activator compound has a structure of Formula (V) (V) 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, norbornyl, adamantyl, bicyclo[2.2.2]octyl, bicyclo[2.2.1]heptyl, bicyclo[3.2.1]octyl, or decahydronaphthyl. Alkyl groups optionally can be substituted, for example, with hydroxy (OH), halo, amino, and sulfonyl. An "alkoxy" group is an alkyl group having an oxygen substituent, e.g., -0-alkyl.
The term "alkenyl" refers to straight chained and branched hydrocarbon groups containing carbon atoms having 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 having a further defined substituent. For example, the term "alkylenearyl" 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. As used herein, the term "heteroaryl" refers to a monocyclic or polycyclic aromatic group containing 0, N, or S atoms, e.g., pyridinyl or pyrimidyl. 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. 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, pharmaceutically acceptable salts are described in detail in (Berge, S.M. et al, 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, cam phorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hem isulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palm itate, 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 1. Computational Modeling, Pharmacophore Generation, Virtual and Functional Screening.
In order to generate a productive pharmacophore, computational docking of the prospective RNA m6A methyltransferase METTL16 binding compounds was carried out using the complex crystal structures. The structure of the RNA m6A methyltransferase METTL16 was chosen as describing the potential target binding site for a small-molecule inhibitor. The crystal structure of this complex (PDB: 6GFN) had been measured by X-ray diffraction with resolution 1.9 A (Chen et al., 2018). 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).Glide 7.8 (Friesner et al., 2006) was used for the docking studies to find out binding modes and binding energies of ligands to the receptor. The structure of ligand molecules was optimized using the density functional theory B3LYP method (Stephens et al., 1994) with 6-31G basis set.
A virtual screening on ZINC compound library (Irwin et al, 2005) was carried out using in vivo subset. The docking free energies AG and ligand efficiencies LE of the best binding compounds are given in Table 1. The ligand efficiency was calculated as the ratio of the docking free energy AG and the number of the non-hydrogen atoms in the compound, N. LE = IAGI
I N I
of the best binding compounds are given in Table 1. The binding sites of the compounds (II), (Ill), (IV) and (V) are shown in Figures 2, 3, 4, and 5, respectively.
Table 1. The compounds with the highest ligand efficiencies to RNA m6A methyltransferase METTL16.
No. Compound structure AG (kcal/mol) LE al N NMt, 0.481 -6.255 mt, N H \ On F HN NH2 0.463 -6.486 1 S Li -, H a NH2 0.326 -4.238 \ fyi / H N 2 0.315 -4.095
N
Example 2. Screening of computationally predicted RNA m6A methyltransferase METTL16 ligands in enzyme inhibition assay.
The inhibitory effect on RNA m6A methylation by METTL16 was studied using biotin-marked RNA oligonucleotide with the sequence 5'-uacacucgaucuggacuaaagcugcucbiotin-3 (biotin-RNA). The reaction mixture included 10 nM METTL16 protein, 5 pM of the co-enzyme S-adenosylmethionine (SAM), 4 pM of the biotin-RNA, 20 mM Tris buffer, 1 mM dithiothretiol, 100 nM Triton X-100 and 6 nM RNaseOUT enzyme to eliminate the possible side effect by ribonucleases. The reaction mixture was incubated 22 hours at 37 °C.
The m6A methylation level in biotin-RNA was measured using EpiQuik m6A RNA methylation Quantification Colorimetric Kit (Epigentek).
The inhibitory effect IE of compounds on RNA probe methylation by METTL16 was calculated as the decrease of the m6A amount as compared to the negative control 5 (DMSO) relative to the difference between m6A amounts of the positive control (max inhibition) and the negative control (eq. (1)):
C
IE - inh ICinh(naX)-CDMS01 (1) where Cinh, Cinh(MaX) and CDMSO are the amounts of m6A at a given concentration of the inhibitor, maximum inhibition and in the case of DMSO, respectively. The dependence of the /Eon the inhibitor concentration for the compound (II) is shown on Fig. 6 and for the compound (III) on Fig. 7. The inhibitory concentrations are IC50 = 73.912 nM for the compound (II) and IC50 = 35.347 nM for the compound (III), respectively. Therefore, both compounds are efficient inhibitors of the RNA m6A methyltransferase METTL16.
Example 3. Inhibitory effect of RNA m6A methyltransferase METTL16 inhibitor compounds on glioblastoma cell line A-172.
The A-172 cell line was obtained from the Glioma Tumor Cell Panel (ATCC® TCP-1018Tm). The HEK-293T cells (ATCCEACS-4500Tm). Both HEK-293T and A-172 cells were grown in Dulbecco's Modified Eagle's medium (DMEM) supplemented with 10% heat-inactivated FBS and Pen/Strep. The cells were grown at 37 °C in the presence of 5% CO2.
8x 103 HEK-293T and A-172 cells were seeded in 200 pL on a 16-well E-plate. Cells were incubated for 48 h with added compounds at given concentrations and 0.5% DMSO was used as a vehicle control. Cells viability were measured using real-time 25 xCELLigence machine (RTCA xCELLigence).
The inhibitory effect of the compounds on the proliferation of the malignant A-172 cells was estimated relative to the proliferation of the normal HEK-293T cells. First, the cell counts pc in the HEK-293T and A-172 cell cultures at a given concentration c of the inhibitor were normalized by the cell counts 17Dmso in the case of negative control (DMSO). The normalized cell counts N, nc (A-172) N( A-172) -npmso (A-172) (2) N nc (HEK-293T)c(HEK-293T) - (3) 71DMS0 (HEK-2931) were then used for the calculation of the inhibitory effect of a compound at given concentration as follows: Nc (A-172) INH% - 100 N(HEK-293T) The time dependence of the inhibitory effect INH% of compound (IV) at 1 pM concentration in Fig. 8. The proliferation of the glioblastoma cells A-172 is suppressed as compared to the normal HEK-293T cells.
Example 4. Inhibitory effect of RNA m6A methyltransferase METTL16 inhibitor 10 compounds on Childhood T acute lymphoblastic leukemia cell line HL-60.
The HL-60 cell line was obtained from the ATCC® TCP1010Tm Leukemia Cell Line Panel. HL-60 cells were grown in Iscove's Modified Dulbecco's Medium (IMDM) supplemented with 20% heat-inactivated FBS and Pen/Strep. The cells were grown at 37 °C in the presence of 5% CO2.
lx 105 HL-60 cells were seeded in 1 mL on a 24-well plate. Cells were incubated for 48 h with added compounds at given concentrations and 0.5% DMSO was used as a vehicle control. Cells viability were measured using Countess Automated Cell Counter by Thermo Fisher Scientific Invitrogen.
Similarly to the glioblastoma cells, the inhibitory effect of the compounds on the proliferation of the Childhood T acute lymphoblastic leukemia cell line HL-60 was estimated relative to the proliferation of the normal HEK-293T cells. Again, the cell counts n, in the, HL-60 cell cultures at a given concentration c of the inhibitor were normalized by the cell counts nomso in the case of negative control (DMSO). The normalized cell counts N, nc (HL -60) (5) Nc(H L-60) - 71DmS00-11,-60) (4) were then used for the calculation of the inhibitory effects of a compound at given concentration as follows: N(HL-6o) (6) INH%= 100 Nc(HEK-293T) The time dependence of the inhibitory effect INH% of compound (II) on the HL-60 cells at 1 pM concentration is given in Fig. 9 and the time dependence of the inhibitory effect INH% of compound (III) at 1 pM concentration in Fig. 10. In all cases, the proliferation of the leukemia cells HL-60 is suppressed as compared to the normal HEK-293T cells.
REFERENCES CITED
1. Berge, S.M. et al. (1977) Pharmaceutical salts, J. Pharm. Sci., 66, 1-19.
2. Bokar, J. A., Shambaugh, M. E., Polayes, D., Matera, A. G., Rottman, F. M. (1997) Purification and cDNA cloning of the AdoMet-binding subunit of the human mRNA (N6-adenosine)-methyltransferase. RNA, 3, 1233-1247.
3. Chen, K.M., Mendel, M., Homolk, D., McCarthy, A.A., Pillai, R.S. (2018). Protein Data Bank. https://www.rcsb.org/structure/6GFN. Accessed 18 Oct 2018 4. Cui, Q., Shi, H., Ye, P., Li, L., Qu, Q., Sun, G., Sun, G., Lu, Z., Huang. Y., Yang, C.G., Riggs, A.D., He, C., and Shi, Y. (2017). m6A RNA Methylation Regulates the Self-Renewal and Tumorigenesis of Glioblastoma Stem Cells. Cell Rep. 18, 26222634.
5. Delaunay, S. Frye, S. (2019). RNA modifications regulating cell fate in cancer. Nat Cell Biol. 21, 552-559.
6. Deng, X., Su, R., Feng, X., Wei, M., and Chen, J. (2018). Role of N6-methyladenosine modification in cancer. Curt-. Opin. Genet. Dev. 48, 1-7.
7. Doxtader, K.A.; Wang, P.; Scarborough, A.M.; Seo, D.; Conrad, N.C.; Nam; Y. (2018). Structural Basis for Regulation of METTL16, an S-Adenosylmethionine Homeostasis Factor. Mo/. Cell 7l, 1001-1011.
8. Fazi, F., Fatica, A. (2019). Interplay Between N6-Methyladenosine (m6A) and Non- coding RNAs in Cell Development and Cancer. Front Cell Dev. Biol. 7, 116. 9. Friesner, R. A.; Murphy, R. B.; Repasky, M. P.; Frye, L. L.; Greenwood, J. R.; Halgren,T. A.; Sanschagrin, P. C.; Mainz, D. T. (2006) Extra Precision Glide: Docking and Scoring Incorporating a Model of Hydrophobic Enclosure for Protein-Ligand Complexes. J Med. Chem., 49, 6177-6196.
Gao J., Aksoy, B.A., Dogrusoz, U., Dresdner, G., Gross, B., Sumer, SO., Sun, Y., Jacobsen, A., Sinha, R., Larsson, E. et al. (2013). Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal, 6, p11.
11 Helm, M.; Motorin, Y. (2017). Detecting RNA modifications in the epitranscriptome: predict and validate. Nat Rev. Genet. 18, 275-291.
12 Horiuchi, K., Kawamura, T., lwanari, H., Ohashi, R., Naito, M., Kodama, T., Hamakubo, T. (2013). Identification of Wilms' tumor 1-associating protein complex and its role in alternative splicing and the cell cycle. J. BioL Chem., 288, 3329233302.
13. Irwin, J.J.; Shoichet, B.K. (2005). ZINC -A Free Database of Commercially Available Compounds for Virtual Screening. J Chem. Mt Model. 45, 177-182.
14 Jia G, et al. (2011) N6-methyladenosine in nuclear RNA is a major substrate of the obesity-associated FTO. Nat Chem. Biol. 7, 885-887.
15. Kandimalla, R., Gao, F., Li, Y., Huang, H., Ke, J., Deng, X., Zhao, L., Zhou, S., Goel, A., Wan, X. (2019). RNAMethyPro: a biologically conserved signature of N6methyladenosine regulators for predicting survival at pancancer level, NPJ Precis. Oncol. 3, 13.
16 Li, Z., Weng, H., Su, R., Weng, X., Zuo, Z., Li, C., Huang, H., Nachtergaele, S., Dong, L., Hu, C., Qin, X., Tang, L., Wang, Y., Hong, G.M., Huang, H., Wang, X., Chen, P., Gurbuxani, S., Arnovitz, S., Li, Y., Li, S., Strong, J., NeiIly, MB., Larson, R.A., Jiang, X., Zhang, P., Jin, J., He, C., and Chen, J. (2017). FTO Plays an Oncogenic Role in Acute Myeloid Leukemia as a N6-Methyladenosine RNA Demethylase. Cancer Cell 31, 127-141.
17 Lin, X., Chai, G., Wu, Y., Li, Y., Chen, F., Liu, J., Luo, G., Tauler, J., Du, J., Lin, S., He, C., Wang, H. (2019). RNA m6A methylation regulates the epithelial mesenchymal transition of cancer cells and translation of Snail, Nat Common., 10, 2065.
18 Liu, X.; Liu, L.; Dong, Z.; Li, J.H.; Yu, Y.; Chen, X.L.; Ren, F.; Cui, G.Y.; Sun, R.R. (2019a). Expression patterns and prognostic value of m(6)A-related genes in colorectal cancer. Am. J. Transl. Res. 11, 3972.
19 Liu, J.Z., Yue, Y.N., Han, DL., Wang, X., Fu, Y., Zhang, L., Jia, G.F.,; Yu, M., Lu, Z.K. Deng, X., Dai, Q., Chen, W.Z., He, C. (2014). A METTL3-METTL14 complex mediates mammalian nuclear RNA N-6-adenosine methylation, Nat Chem. Biol., 10, 93-95.
20. Maity, A.; Das, B. (2016). N6-methyladenosine Modification in mRNA: Machinery, Function and Implications for Health and Diseases. FEBS Journal. 283, 16071630.
21. Mendel, M.; Chen, K.; HomoIke, D.; Gos, P., Pandey, R.R.; McCarthy A.A.; Pillai, R.S. (2018). Methylation of Structured RNA by the m(6)A Writer METTL16 Is Essential for Mouse Embryonic Development, Mo/. Cell 71, 986-1000.
22. Niu, Y.; Zhao X.; Wu, Y.-Z.; Li, M-M.; Wang, X.-J.; Yang, Y.-G. (2013). N6-MethylAdenosine (m6A) in RNA: An Old Modification with A Novel Epigenetic Function. GPB. 11,8-17.
23. Park, C.H.; Hong, K. (2017). Epitranscriptome: m6A and its function in stem cell biology. Genes Genom. 39, 371-378.
24. Pendleton, K.E, Chen, B., Liu, K., Hunter, 0.V., Xie, Y., Tu, B.P., Conrad, N.K. (2017). The U6 snRNA m6A Methyltransferase METTL16 Regulates SAM Synthetase Intron Retention. Cell 169, 824-835.e14.
25. Roundtree, I.A., Evans, M.E., Pan, T. & He, C. (2017). Dynamic RNA Modifications in Gene Expression Regulation. Ce// 169, 1188-1200.
26. Sastry, G. M.; Adzhigirey, M.; Day, T.; Annabhimoju, R.; Sherman, W. (2013). Protein and Ligand Preparation: Parameters, Protocols, and influence on Virtual Screening Enrichments. J. Comput. Aid. Mol. Des. 27, 221-234.
27. Scholler, E., Weichmann, F., Treiber, T., Ringle, S., Treiber, N., Flatley, A., Feederle, R., Bruckmann, A., Meister, G. (2018) Interactions, localization, and phosphorylation of the m(6)A generating METTL3-METTL14-VVTAP complex. RNA, 24, 499-512.
28. Shima, H., Matsumoto, M., Ishigami, Y., Ebina, M., Muto, A., Sato, Y., et al. (2017).
S-Adenosylmethionine synthesis is regulated by selective N6-adenosine methylation and mRNA degradation involving METTL16 and YTHDC1. Cell Rep. 21, 3354-3363.
29. Stephens, P.J.; Devlin, F.J.; Chabalowski, C.F.; Frisch, M.J. (1994). Ab lnitio Calculation of Vibrational Absorption and Circular Dichroism Spectra Using Density Functional Force Fields. J. Phys. Chem. 98, 11623-11627.
30. Sun, T., Wu, R., Ming, L. (2019). The role of m6A RNA methylation in cancer. Biomed. & Pharmacother 112, 108613.
31 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. 31, 455-461.
32 Wang, X., Li, Z., Kong, B., Song, C., Cong, J., Hou, J., and Wang, S. (2017).
Reduced m6A mRNA methylation is correlated with the progression of human cervical cancer, Oncotarget. 8, 98918-98930.
33 Warda, A. S., Kretschmer, J., Hackert, P., Lenz, C., Urlaub, H., HObartner, C. et al. (2017). Human METTL16 is a N(6)-methyladenosine (m(6)A) methyltransferase that targets pre-mRNAs and various non-coding RNAs. EMBO Rep. 18, 2004-2014.
34 Wei, W., Huo, B., Shi, X. (2019). miR-600 inhibits lung cancer via downregulating the expression of METTL3. Cancer Manag. Res. 11, 1177-1187.
Zhang, C., Zhi, W.I., Lu, H. et al. (2016) Hypoxia-inducible factors regulate pluripotency factor expression by ZNF217-and ALKBH5-mediated modulation of RNA methylation in breast cancer cells, Oncotarget, 7, 64527-64542.
36 Zhang, S., Zhao, B.S., Zhou, A., Lin, K., Zheng, S., Lu, Z., Chen, Y., Sulman, E.P., Xie, K., Begler, 0., Majumder, S., He, C., and Huang, S. (2017). m6A Demethylase ALKBH5 Maintains Tumorigenicity of Glioblastoma Stem-like Cells by Sustaining FOXM1 Expression and Cell Proliferation Program. Cancer. 31, 591-606.
37 Zheng, G., Dahl, J.A. Niu, Y. et al., ALKBH5 Is a Mammalian RNA Demethylase that Impacts RNA Metabolism and Mouse Fertility, (2013) Mot Cell, 49, 18-29. 38 Yanhong, S., Qui, Q. (2018) m6A mRNA modification in cancer treatment, Pat. Appl. W02018169994A1.

Claims (15)

  1. Claims 1. A method of inhibiting the RNA demethylation at 6-position of adenine (m6A) by effective amount of a compound having binding and inhibition for a RNA m6A methyltransferase METTL16, wherein the compound has a structure of Formula (I) R2 // R4 \ R (I) wherein: R1 is independently selected from the group consisting aryl or heteroary1;, R2 and R3 are independently selected from the group consisting of H, alkyl, aryl, aralkyl, acyl,and R4 is either a oxygen or a sulfur atom; or a pharmaceutically acceptable salt thereof.
  2. 2. The method of claim 1, wherein the compound has a structure of Formula (II) 3. The method of inhibiting the RNA demethylation at 6-position of adenine (m6A) by effective amount of a compound having binding and inhibition for a RNA m6A methyltransferase METTL16, wherein the compound has a structure of Formula (III)
  3. N
  4. 4. The method of claim 1, wherein the compound has a structure of Formula (IV) (IV)
  5. 5. The method of claim 1, wherein the compound has a structure of Formula (V) NH2 (V)
  6. 6. A pharmaceutical composition comprising a compound of Formula (I) of claim 1, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.
  7. 7. The pharmaceutical composition comprising a compound of Formula (II) of claim 2, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.
  8. 8. A pharmaceutical composition comprising a compound of Formula (III) of claim 3, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.
  9. 9. The pharmaceutical composition comprising a compound of Formula (IV) of claim 4, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.
  10. 10. The pharmaceutical composition comprising a compound of Formula (V) of claim 5, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.
  11. 11.A compound of Formula (I) of claim 1 for use in the treatment of glioblastoma, astrocytoma, acute myeloid leukemia, acute monocytic leukemia, chronic myelogenous leukemia, T acute lymphoblastic leukemia in a patient in need thereof, comprising administering to the patient a compound of Formula (I) of claim 1, or a pharmaceutically acceptable salt thereof.
  12. 12.A compound of Formula (II) of claim 2 for use in the treatment of glioblastoma, astrocytoma, acute myeloid leukemia, acute monocytic leukemia, chronic myelogenous leukemia, T acute lymphoblastic leukemia in a patient in need thereof, comprising administering to the patient a compound of Formula (II) of claim 2, or a pharmaceutically acceptable salt thereof.
  13. 13.A compound of Formula (III) of claim 3 for use in the treatment of glioblastoma, astrocytoma, acute myeloid leukemia, acute monocytic leukemia, chronic myelogenous leukemia, T acute lymphoblastic leukemia in a patient in need thereof, comprising administering to the patient a compound of Formula (III) of claim 3, or a pharmaceutically acceptable salt thereof.
  14. 14.A compound of Formula (IV) of claim 4 for use in the treatment of glioblastoma, astrocytoma, acute myeloid leukemia, acute monocytic leukemia, chronic myelogenous leukemia, T acute lymphoblastic leukemia in a patient in need thereof, comprising administering to the patient a compound of Formula (IV) of claim 4, or a pharmaceutically acceptable salt thereof.
  15. 15.A compound of Formula (V) of claim 5 for use in the treatment of glioblastoma, astrocytoma, acute myeloid leukemia, acute monocytic leukemia, chronic myelogenous leukemia, T acute lymphoblastic leukemia in a patient in need thereof, comprising administering to the patient a compound of Formula (V) of claim 5, or a pharmaceutically acceptable salt thereof.
GB1918338.3A 2019-12-12 2019-12-12 Method of suppressing cancer by RNA m6A methyltransferase mettl16 inhibitors Pending GB2589912A (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
GB1918338.3A GB2589912A (en) 2019-12-12 2019-12-12 Method of suppressing cancer by RNA m6A methyltransferase mettl16 inhibitors
PCT/EP2020/085872 WO2021116477A1 (en) 2019-12-12 2020-12-12 METHOD OF SUPPRESSING CANCER BY RNA m6A METHYLTRANSFERASE METTL16 INHIBITORS

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
GB1918338.3A GB2589912A (en) 2019-12-12 2019-12-12 Method of suppressing cancer by RNA m6A methyltransferase mettl16 inhibitors

Publications (2)

Publication Number Publication Date
GB201918338D0 GB201918338D0 (en) 2020-01-29
GB2589912A true GB2589912A (en) 2021-06-16

Family

ID=69186633

Family Applications (1)

Application Number Title Priority Date Filing Date
GB1918338.3A Pending GB2589912A (en) 2019-12-12 2019-12-12 Method of suppressing cancer by RNA m6A methyltransferase mettl16 inhibitors

Country Status (2)

Country Link
GB (1) GB2589912A (en)
WO (1) WO2021116477A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5767134A (en) * 1997-05-15 1998-06-16 Vion Pharmaceuticals, Inc. Prodrug forms of ribonucleotide reductase inhibitors 3-AP and 3-AMP
WO2015157242A1 (en) * 2014-04-07 2015-10-15 H. Lee Moffitt Cancer Center And Research Institute, Inc. Thiosemicarbazones inhibitors of lysophosphatidic acid acyltransferase and uses thereof
WO2017030983A1 (en) * 2015-08-14 2017-02-23 Mateon Therapeutics, Inc. Compositions and methods for inhibition of cathepsins
CN109796497A (en) * 2019-01-16 2019-05-24 广西师范大学 It is a kind of using 2- acetyl group -3- ethyl pyrazine thiosemicarbazones as the bismuth compound of ligand and its synthetic method
CN109897071A (en) * 2019-03-28 2019-06-18 广西师范大学 It is a kind of using 2- pyridine carboxaldehyde thiosemicarbazones as the platinum complex of ligand and its synthetic method and application

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP3512236B2 (en) * 1994-06-29 2004-03-29 杏林製薬株式会社 Pyrazolopyridine oxoalkylene acid derivatives and their production
US5942527A (en) * 1997-08-27 1999-08-24 K & K Biosciences, Inc. Hydrazones, hydrazines, semicarbazones and thiosemicarbazones derived from pyridyl ketones as anticonvulsant drugs and excitatory amino acid antagonists
AP2006003756A0 (en) * 2004-03-05 2006-10-31 He Regents Of The University O Anti-parasitic compounds and methods of their use
JP5739329B2 (en) * 2008-06-17 2015-06-24 インスティチュート・パスツール・コリアInstitut Pasteur Korea Pyridopyrimidine compounds as antituberculosis agents
US20200129596A1 (en) 2017-03-13 2020-04-30 City Of Hope m6A mRNA MODIFICATION IN CANCER TREATMENT
CN108250058B (en) * 2018-01-19 2022-02-11 上海怡立舍生物技术有限公司 PPAR agonists and their use in the treatment of Alzheimer's disease and other diseases

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5767134A (en) * 1997-05-15 1998-06-16 Vion Pharmaceuticals, Inc. Prodrug forms of ribonucleotide reductase inhibitors 3-AP and 3-AMP
WO2015157242A1 (en) * 2014-04-07 2015-10-15 H. Lee Moffitt Cancer Center And Research Institute, Inc. Thiosemicarbazones inhibitors of lysophosphatidic acid acyltransferase and uses thereof
WO2017030983A1 (en) * 2015-08-14 2017-02-23 Mateon Therapeutics, Inc. Compositions and methods for inhibition of cathepsins
CN109796497A (en) * 2019-01-16 2019-05-24 广西师范大学 It is a kind of using 2- acetyl group -3- ethyl pyrazine thiosemicarbazones as the bismuth compound of ligand and its synthetic method
CN109897071A (en) * 2019-03-28 2019-06-18 广西师范大学 It is a kind of using 2- pyridine carboxaldehyde thiosemicarbazones as the platinum complex of ligand and its synthetic method and application

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
3-aminopyridine-2-carboxaldehyde thiosemicarbazone, see URL: https://en.wikipedia.org/wiki/3-Aminopyridine-2-carboxaldehyde_thiosemicarbazone [date accessed 5 June 2020]. *

Also Published As

Publication number Publication date
WO2021116477A1 (en) 2021-06-17
GB201918338D0 (en) 2020-01-29

Similar Documents

Publication Publication Date Title
WO2019197024A1 (en) A method of modulating the rna methylation
Xu et al. Roles and drug development of METTL3 (methyltransferase-like 3) in anti-tumor therapy
Yu et al. Targeting GLS1 to cancer therapy through glutamine metabolism
Hou et al. Dual-tail approach to discovery of novel carbonic anhydrase IX inhibitors by simultaneously matching the hydrophobic and hydrophilic halves of the active site
Zhang et al. Design, synthesis and biological evaluation of sulfamoylphenyl-quinazoline derivatives as potential EGFR/CAIX dual inhibitors
EP3372594A1 (en) Pyrimidine derivative and use thereof
WO2020207550A1 (en) METHOD OF SUPPRESSING CANCER BY RNA m6A DEMETHYLASE AlkBH5 INHIBITORS
EP3271333B1 (en) Usp7 inhibitor compounds and methods of use
An et al. Discovery of novel artemisinin-sulfonamide hybrids as potential carbonic anhydrase IX inhibitors with improved antiproliferative activities
EP2986120B1 (en) Methyltransferase inhibitors for treating cancer
SA516371644B1 (en) Pyrrole-Substituted Indolone Derivative, Preparation Method Therefor, Composition Comprising the same and use thereof
Jing et al. Discovery and optimization of tetrahydropyrido [4, 3-d] pyrimidine derivatives as novel ATX and EGFR dual inhibitors
Liu et al. Phosphamide-containing diphenylpyrimidine analogues (PA-DPPYs) as potent focal adhesion kinase (FAK) inhibitors with enhanced activity against pancreatic cancer cell lines
Sun et al. Design, synthesis and pharmacological evaluation of 2-arylurea-1, 3, 5-triazine derivative (XIN-9): a novel potent dual PI3K/mTOR inhibitor for cancer therapy
Manzoor et al. Development of benzene and benzothiazole-sulfonamide analogues as selective inhibitors of the tumor-associated carbonic anhydrase IX
Rami et al. Carbonic anhydrase inhibitors: copper (II) complexes of polyamino-polycarboxylamido aromatic/heterocyclic sulfonamides are very potent inhibitors of the tumor-associated isoforms IX and XII
Lin et al. PKM2/PDK1 dual-targeted shikonin derivatives restore the sensitivity of EGFR-mutated NSCLC cells to gefitinib by remodeling glucose metabolism
Gan et al. Discovery of novel 4-arylamino-quinazoline derivatives as EGFRL858R/T790M inhibitors with the potential to inhibit the non-small cell lung cancers
Chen et al. Design, synthesis, and biological evaluation of indazole derivatives as selective and potent FGFR4 inhibitors for the treatment of FGF19-driven hepatocellular cancer
Ji et al. Discovery of potent, orally bioavailable ERK1/2 inhibitors with isoindolin-1-one structure by structure-based drug design
EP4174064A1 (en) Tetrahydroisoquinoline compounds and use thereof
GB2589912A (en) Method of suppressing cancer by RNA m6A methyltransferase mettl16 inhibitors
Dai et al. Antitumor activity and mechanisms of dual EGFR/DNA-targeting strategy for the treatment of lung cancer with EGFRL858R/T790M mutation
Li et al. The development of small molecules targeting methyltransferase-like 3
Zhou et al. Design, synthesis and evaluation of anti-proliferative activity of 2-aryl-4-aminoquinazoline derivatives as EGFR inhibitors