Nucleic acid molecules and uses thereof

Information

  • Patent Grant
  • 11866704
  • Patent Number
    11,866,704
  • Date Filed
    Sunday, December 6, 2020
    3 years ago
  • Date Issued
    Tuesday, January 9, 2024
    8 months ago
Abstract
A method of treating a subject suffering from cancer comprising a step of administering an effective amount of a group of double-stranded RNA molecules to the subject, wherein the RNA molecule is isolated or derived from a bacteria of the genus Escherichia. A method of inhibiting growth or proliferation of cancer cells comprising a step of contacting said cells with said RNA molecule; and a pharmaceutical composition for treating cancer comprising said RNA molecule and a pharmaceutically tolerable excipient. Also a double-stranded RNA molecule and a recombinant vector comprising the double-stranded RNA molecule.
Description
CROSS-REFERENCE TO RELATED APPLICATION

This application claims priority to, and the benefit of, Chinese Patent Application No. 202010083971.2 filed on Feb. 10, 2020. The entire contents of the foregoing application are hereby incorporated by reference for all purposes.


REFERENCE TO SEQUENCE LISTING

This application contains a sequence listing which has been submitted electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on Jul. 25, 2022, is named “M006_092_NPRUS_Sequence_list_revised.txt” and is 83 kilobytes in size.


TECHNICAL FIELD

The present invention relates to a method of treating a subject suffering from cancer by administering a nucleic acid to the subject. Said nucleic acid is in particular but not exclusively a RNA molecule. The invention further relates to a pharmaceutical composition comprising a nucleic acid for the treatment and use thereof.


BACKGROUND OF THE INVENTION

Cancer has become the most common disease causing death worldwide.


Traditional Chinese medicines (TCMs) have been applied for treating and preventing cancer whereas lots of research efforts have been contributed to investigate the effectiveness of isolated small molecules such as alkaloids, terpenoids, flavonoids or the like in treating cancer. Some alkaloids are found to have effect in inhibiting cancer such as by enhancing the efficacy of an anti-cancer drug. However, most of them are often toxic to human. Also, macromolecules such as DNAs, RNAs, and proteins are generally considered unstable and have poor effect in living human body and therefore have not been widely considered as suitable in said treatment.


Currently, some studies show that non-coding RNAs (ncRNAs) such as microRNAs have diverse regulatory roles through targeting different aspects of RNA transcription or post-transcription process in nearly all eukaryotic organisms. Mlotshwa, S. et al. (Cell research 2015, 25 (4), 521-4) suggested that exogenous plant microRNAs in foods could be taken up by the mammalian digestive tract and trafficked via the bloodstream to a variety of tissue cells, where they are capable of regulating the expression of mammalian genes. Goodarzi, H. et al. (Cell 2015, 161 (4), 790-802) revealed that endogenous tRNA derived fragments could suppress the stability of multiple oncogenic transcripts in breast cancer cells through binding and antagonizing activities of pathogenesis-related RNA-binding proteins.



Escherichia coli (Migula) Castellani & Chalmers, a species from the genus Escherichia that belong to the family of Enterobacteriaceae. It is a famous gut microbiota mainly distributed in the colon of human beings and animals, which possess almost 0.1% of gut microorganisms. Non-pathogenic Escherichia coli as a part of normal bacteria in human gut intestinal can produce vitamin K, as well as avoiding the progression of other pathogenic strains, which is beneficial to the human beings. Nevertheless, there still remains a need to derive effective molecules from various sources such as human gut microbiota for treatments.


SUMMARY OF THE INVENTION

According to the limitations of current techniques, through lots of experiments and investigations, the inventors successfully separated and purified tRNA-derived fragments from bacteria of the genus Escherichia for cancer prevention and/or treatment, especially for the double-stranded RNA molecules containing antisense from SEQ ID NO: 1 to 3 and sense from SEQ ID NO: 4 to 6, which provides a novel and effective method for preventing and/or treating cancer.


In a first aspect, the invention provides a group of double-stranded RNA molecules comprising an antisense sequence selected from SEQ ID NO: 7 to SEQ ID NO: 100 and a sense sequence selected from SEQ ID NO: 101 to SEQ ID NO: 194, or a functional variant or homologue therefore.


Preferably, the said double-stranded RNA molecules or a functional variant or homologue therefore comprising an antisense sequence selected from SEQ ID NO: 47, 48, 89, 90, 91 and 92, and a sense sequence selected from SEQ ID NO: 141, 142, 183, 184, 185 and 186.


In a further aspect, the antisense sequence of said double-stranded RNA molecules or a functional variant or homologue therefore is shown as SEQ ID NO: 47, 48, 89, 90, 91 and 92, and the sense sequence of said double-stranded RNA molecules or a functional variant or homologue therefore is shown as SEQ ID NO: 141, 142, 183, 184, 185 and 186.


Still further, the antisense sequence of said double-stranded RNA molecules or a functional variant or homologue therefore is shown as SEQ ID NO: 89, and the sense sequence of said double-stranded RNA molecules or a functional variant or homologue therefore is shown as SEQ ID NO: 183.


Preferably, the said double-stranded RNA molecules or a functional variant or homologue therefore comprising a 3′ overhang.


Preferably, the said double-stranded RNA molecules or a functional variant or homologue therefore comprising one or more preferably modified nucleotides. The said nucleotides comprising m1A, m2A, m5A, m7A, m2G, m6A, m22G, Um, i6A, ms2i6A, t6A, m6t6A, s2C, s2U, s4U, ac4C, f5C, acp3U, mo5U, cmo5U, mcmo5U, mcm5U, mcm5Um, mcm5s2U, nm5s2U, mnm5U, mnm5s2U, ncm5U, ncm5Um, cmnm5U, cmnm5Um, cmnm5s2U, tm5U and tm5s2U. Preferably, said chemical modifications comprising one or more selected from m1A, m7G, m6A, Gm, Cm, Am, Um, m22G, s4U and cmo5U. Preferably, the said chemical modified nucleotides comprising uridine or guanosine. In a further aspect, the said chemical modified nucleotides comprising s4U and/or Gm. Still further, the said double-stranded RNA molecules or a functional variant or homologue therefore comprising an antisense sequence selected from SEQ ID NO: 1, 2 and 3, and a sense sequence selected from SEQ ID NO: 4, 5 and 6. Most preferably, the said double-stranded RNA molecules or a functional variant or homologue therefore is shown as SEQ ID NO: 1, 2 and 3, and a sense sequence is shown as SEQ ID NO: 4, 5 and 6.


In a further aspect, the invention provides to a pharmaceutical composition for cancer prevention and/or treatment. The pharmaceutical composition comprises the said double-stranded RNA molecules or a functional variant or homologue therefore and pharmaceutically tolerable carrier, diluent and/or excipient.


Preferably, the said pharmaceutical composition comprises the said nucleic acid stabilizer.


In a further aspect, the invention provides a delivery system for cancer prevention and/or treatment, which comprises the said double-stranded RNA molecules or a functional variant or homologue therefore and pharmaceutically tolerable carrier, diluent and/or excipient.


In another aspect, the invention provides the use of the said double-stranded RNA molecules or a functional variant or homologue therefore in preparation of pharmaceutical composition for cancer prevention and/or treatment.


Correspondingly, the invention provides a method for cancer prevention and treatment, said method comprising administrating an effective amount of said double-stranded RNA molecules or a functional variant or homologue therefore to objects for cancer prevention and/or treatment.


In the mentioned pharmaceutical composition, delivery system, use or method, said cancer prevention and/or treatment can be inhibiting growth, proliferation or migration of cancer cells.


According to an embodiment, in the mentioned pharmaceutical composition, delivery system, use or method, said cancer prevention and/or treatment is colorectal cancer prevention and/or treatment. Preferably, said cancer prevention and/or treatment is inhibiting colorectal cancer cells. Preferably, cancer prevention and/or treatment is fluorouracil-resistant cancer.


The inventors have found that non-coding RNA molecules isolated from a bacteria of the genus Escherichia, particularly transfer RNA molecules, and RNA molecules derived from Escherichia are particularly useful in treatment of cancer. The RNA molecules with a sequence length of about 10 to 200 nucleotides and their homologue double-stranded RNA molecules with a sequence length of about 10 to 30 nucleotides are highly effective in inhibiting growth and proliferation of cancer cells in vitro and exhibit an antitumor effect in vivo. Said RNA molecules are also effective against fluorouracil-resistant cell lines. Further, the pharmaceutical composition comprising the RNA molecule that is isolated or derived from a bacteria of the genus Escherichia and a pharmaceutically tolerant excipient can act directly on the cancer cells or tumor, and therefore can have a faster-acting therapeutic effect.


Those skilled in the art will appreciate that the invention described herein is susceptible to variations and modifications other than those specifically described.


The invention includes all such variations and modifications. The invention also includes all steps and features referred to or indicated in the specification, individually or collectively, and any and all combinations of the steps or features.


Other features and aspects of the invention will become apparent by consideration of the following detailed description and accompanying drawings.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1A shows chromatogram under UV 260 nm of mixed tRNA separated by high-performance weak-anion exchange chromatographic method in accordance with an example embodiment.



FIG. 1B shows urea denatured polyacrylamide gel electrophoresis profile of 6 mixed tRNA fractions separated by high-performance weak-anion exchange chromatographic method in accordance with an example embodiment.



FIG. 2A shows chromatogram under UV 260 nm of fraction 2 separated by high-performance weak-anion exchange chromatographic method, total ion chromatogram of tRNA-1 analyzed by ultra-high performance liquid chromatography coupled with mass spectrometry, multiple charge distribution and deconvolution chromatogram of tRNA-1 in accordance with an example embodiment.



FIG. 2B shows chromatogram under UV 260 nm of fraction 3 separated by high-performance weak-anion exchange chromatographic method, total ion chromatogram of tRNA-2 analyzed by ultra-high performance liquid chromatography coupled with mass spectrometry, multiple charge distribution and deconvolution chromatogram of tRNA-2 in accordance with an example embodiment.



FIG. 2C shows urea denatured polyacrylamide gel electrophoresis profile of tRNA-1 and tRNA-2, including microRNA marker, total tRNA fraction and low range ssRNA ladder in accordance with an example embodiment.



FIG. 3 shows the fragmentation rule of oligonucleotides for characterization of purified tRNA in accordance with an example embodiment.



FIG. 4A shows the identification of specific fragment of tRNA-1 digested by RNase T1 in total ion chromatography in accordance with an example embodiment.



FIG. 4B shows the identification of specific fragment of tRNA-2 digested by RNase T1 in total ion chromatography in accordance with an example embodiment.



FIG. 5A shows chromatograms under UV 260 nm of fragments of tRNA-2 digested by S1 nuclease, 5′-tRNA-half molecules and 3′-tRNA-half molecules by ultra-high performance liquid chromatography in accordance with an example embodiment.



FIG. 5B shows the urea denatured polyacrylamide gel electrophoresis profile of fragments of tRNA-2 digested by S1 nuclease, including low range ssRNA ladder, total tRNA fraction and tRNA-2 in accordance with an example embodiment.



FIG. 6A is a bar chart showing the cell viability of HCT-8 cell line treated by 50 nM RNA molecules 5′-tRNA half fragments and 3′-tRNA half fragments of tRNA-Val(UAC) and tRNA-Leu(CAG), 5′-tRF mimic and 3′-tRF mimic from Escherichia coli (Migula) Castellani & Chalmers compared to a control group and a liposome group, in accordance with an example embodiment (mean±SD n=3; ***, p<0.001, ****, p<0.0001 vs. vehicle control).



FIG. 6B is a heatmap showing the death rate of HCT-8 cell line treated by a total of 82 RNA molecules in Table 3, including EC1-EC24, EC26-EC33, EC35-EC47, EC49-EC54, EC56-EC58, EC61-EC68, EC70-EC71, EC73-EC74, EC77-EC92 derived from Escherichia coli (Migula) Castellani & Chalmers with a sequence length of 22 bp at a dose of 50 nM, compared to a control group and a liposome group in accordance with an example embodiment (mean±SD n=3).



FIG. 6C is a line chart showing the cell viability of HCT-8 cells after treatment with RNA molecule EC83 mimic derived from Escherichia coli (Migula) Castellani & Chalmers at different concentrations, i.e. 3.125 nM, 6.25 nM, 12.5 nM, 25 nM, 50 nM and 100 nM, compared to a control group in accordance with an example embodiment (mean±SD n=3).



FIGS. 7A and 7B are line charts showing the cell viability of HCT-8 cells, HCT-8/5-FU cells, LoVo cells and LoVo/5-FU cells after treatment with RNA molecule EC83 mimic, EC83-M1 mimic, EC83-M2 mimic and EC83-M3 mimic derived from Escherichia coli (Migula) Castellani & Chalmers at different concentrations, i.e. 3.125 nM, 6.25 nM, 12.5 nM, 25 nM, 50 nM and 100 nM, compared to a control group in accordance with an example embodiment (mean±SD n=3; *, p<0.05, ***, p<0.001, ****, p<0.0001 vs. vehicle control).



FIG. 8A shows the inhibition effects of RNA molecule EC83 mimic, EC83-M1 mimic, EC83-M2 mimic and EC83-M3 mimic derived from Escherichia coli (Migula) Castellani & Chalmers at a dose of 50 nM on the proliferation of HCT-8 cells in accordance with an example embodiment (mean±SD n=3; ****, p<0.0001 vs. vehicle control).



FIG. 8B shows the inhibition effects of RNA molecule EC83 mimic, EC83-M1 mimic, EC83-M2 mimic and EC83-M3 mimic derived from Escherichia coli (Migula) Castellani & Chalmers at a dose of 25 nM on the proliferation of LoVo cells in accordance with an example embodiment (mean±SD n=3; ****, p<0.0001 vs. vehicle control).



FIG. 9A shows the inhibition effects of RNA molecule EC83 mimic, EC83-M1 mimic, EC83-M2 mimic and EC83-M3 mimic derived from Escherichia coli (Migula) Castellani & Chalmers at a dose of 50 nM on the migration of HCT-8 cells in accordance with an example embodiment (mean±SD n=3; ****, p<0.0001 vs. vehicle control).



FIG. 9B shows the inhibition effects of RNA molecule EC83 mimic, EC83-M1 mimic, EC83-M2 mimic and EC83-M3 mimic derived from Escherichia coli (Migula) Castellani & Chalmers at a dose of 25 nM on the migration of LoVo cells in accordance with an example embodiment (mean±SD n=3; ****, p<0.0001 vs. vehicle control).





DETAILED DESCRIPTION OF THE EMBODIMENTS

Unless otherwise defined, all technical terms used herein have the same meaning as commonly understood by one skilled in the art to which the invention belongs.


As used herein and in the claims, “comprising” means including the following elements but not excluding others. “Essentially consisting of” means that the material consists of the respective element along with usually and unavoidable impurities such as side products and components usually resulting from the respective preparation or method for obtaining the material such as traces of further components or solvents. “Consisting of” means that the material solely consists of, i.e. is formed by the respective element.


As used herein and in the claims, the singular forms of “a” and “an” also include the corresponding plurals unless the context clearly dictates otherwise.


The present invention in the first aspect provides a method of treating a subject suffering from cancer. The method comprises a step of administering an effective amount of a RNA molecule to said subject. The RNA molecule administered according to the present invention may be naturally present, modified or artificially synthesized according to the sequences disclosed in the present invention, and preferably the RNA molecule is isolated or derived from a bacteria of the genus Escherichia. The RNA molecule of the present invention is not provided in the form of boiled extract obtained from the plant such as decoction, as it would be appreciated that RNA molecule is susceptible to spontaneous degradation at elevated temperature, alkaline pH, and the presence of nucleases or divalent metal ions. In an embodiment, the RNA molecule of the present invention is provided together with a gene delivery carrier which will be described in detail later. The RNA molecule of the present invention has a sequence length of from about 10 to 200 nucleotides which can be regarded as a small RNA molecule. Preferably, the RNA molecule has a sequence length of from about 50 to about 200 nucleotides, from about 60 to about 150 nucleotides, in particular from about 70 to about 100 nucleotides.


The RNA molecule of the present invention comprises a sequence selected from SEQ ID NO: 195 to SEQ ID NO: 241 or a functional variant or homologue thereof. The term “functional variant” of the RNA molecule refers to a molecule substantially similar to said RNA molecule with one or more sequence alterations that do not affect the biological activity or function of the RNA molecule. The alterations in sequence that do not affect the functional properties of the resultant RNA molecules are well known in the art. For example, nucleotide changes which result in alteration of the −5′-terminal and −3′-terminal portions of the molecules would not be expected to alter the activity of the polynucleotides. In an embodiment, the RNA molecule of the present invention comprises at least one modified nucleoside selected from inosine, 1-methyladenosine, 2-methyladenosine, N6-methyladenosine, N6-isopentenyladenosine, 2′-Omethyladenosine, N6-acetyladenosine, 1-methylinosine, pseudouridine, dihydrouridine, or 2-methylthio-N6-methyladenosine. In another embodiment, the RNA molecule comprises at least one nucleotide having one or more chemical modifications. In some embodiments, the one or more chemical modifications is selected from the group consisting of 1-methyl, 2-methyl, 5-methyl, 7-methyl, N2 methyl, N6 methyl, N2,N2 dimethyl, 2′-O-methyl, N6-isopentenyl, 2-methylthio-N6-isopentenyl, N6 threonide carbamoyl, N6-methyl-N6-threosylcarbamoyl, 2-thio, 4-thio, N4 acetyl, 5-formyl, 3-(3-amino-3-carboxypropyl), 5-methoxy, 5-oxoacetic acid, 5-oxoacetate methyl ester, 5-methoxycarbonylmethyl, 5-methoxycarbonylmethyl-2′-O-methyl, 5-methoxycarbonylmethyl-2-thio, 5-aminomethyl-2-thio, 5-methylaminomethyl, 5-methylaminomethyl-2-thio, 5-aminoformylmethyl, 5-aminoformylmethyl-2′-O-methyl, 5-carboxymethyl aminomethyl, 5-carbamoylmethyl-2′-O-methyl, 5-carboxymethylaminomethyl-2-methyl, 5-taurine, 5,2′-O-dimethyl, and 5-tauromethyl-2-thio.


In particular, the functional variant of the RNA molecule has at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% overall sequence identity to the non-variant RNA molecule according to the present invention.


The term “homologue” used herein refers to nucleotides having a sequence identity of at least 50%, at least 60%, at least 70%, at least 80%, at least 90% or at least 95% to the RNA molecules according to the present invention. In an embodiment, the homologue of the RNA molecule has at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% overall sequence identity to the RNA molecule.


Unless otherwise indicated, the term “thereof” in the term “functional variant or homologue thereof” as used herein in the claims refers to each and every member of the entire group(s) or list(s) preceding the term in the claim. For example, in a claim that claims “an antisense sequence selected from one of SEQ ID NO: 7 to SEQ ID NO: 100 and a sense sequence selected from one of SEQ ID NO: 101 to SEQ ID NO: 194; or a functional variant or homologue thereof”, the group of double-stranded RNA molecules covered by the claim includes functional variants or homologs of all the sequences from SEQ ID NO: 7 to SEQ ID NO: 100, and functional variants or homologs of all the sequences from SEQ ID NO: 101 to SEQ ID NO: 194.


In an embodiment, the RNA molecule is a non-coding molecule preferably selected from a transfer RNA molecule, a ribosomal RNA molecule, a micro RNA molecule, a siRNA molecule, or a piwi-interacting RNA molecule; and more preferably is a transfer RNA molecule. tRNA molecules are highly conserved RNAs with function in various cellular processes such as reverse transcription, porphyrin biosynthesis or the like. In a particular embodiment, the double-stranded RNA molecule of the invention comprises an antisense sequence selected from SEQ ID NO: 7 to SEQ ID NO: 100 or a functional variant or homologue thereof; or the RNA molecule comprises a sense sequence selected from SEQ ID NO: 101 to SEQ ID NO: 194 or a functional variant or homologue thereof; or the RNA molecule consists of a sequence selected from SEQ ID NO: 7 to SEQ ID NO: 100 or SEQ ID NO: 101 to SEQ ID NO: 194 or a functional variant or homologue thereof.


In an alternative embodiment where the RNA molecule is a small RNA molecule having a sequence length of from about 10 to about 30 base pairs, from about 15 to about 25 base pairs, from about 19 to about 22 base pairs, 19 base pairs or 22 base pairs.


In an alternative embodiment, the RNA molecule or a functional variant or homologue thereof comprises a sequence selected from SEQ ID NO: 195 to SEQ ID NO: 241, in particular SEQ ID NO: 218 or SEQ ID NO: 237; or consists of a sequence selected from SEQ ID NO: 195 to SEQ ID NO:241, in particular SEQ ID NO: 218 to SEQ ID NO: 237. Preferably, the RNA molecule is a double-stranded RNA molecule having an antisense sequence selected from SEQ ID NO: 7 to SEQ ID NO: 100 or a functional variant or homologue thereof, and a complementary sense sequence. The antisense sequence is complementary to the sense sequence and the antisense sequence is preferably derived from SEQ ID NO: 195 to SEQ ID NO: 241 or functional variant or homologue thereof. Preferably, said double-stranded RNA molecule comprises an antisense sequence selected from SEQ ID NO: 47, 48, 89, 90, 91 or 92, and said double-stranded RNA molecule comprises an sense sequence selected from SEQ ID NO: 141, 142, 183, 184, 185 or 186; Preferably, said double-stranded RNA molecule comprises an antisense sequence shown as SEQ ID NO: 89, and said double-stranded RNA molecule comprises a sense sequence shown as SEQ ID NO: 183. In particular, RNA molecule is chemically modified double-stranded RNA molecule or functional variant or homologue thereof, and comprises an antisense sequence selected from SEQ ID NO: 1 to SEQ ID NO: 3, and comprises a complementary sense sequence selected from SEQ ID NO: 4 to SEQ ID NO: 6 in accordance with an example embodiment. The inventors unexpectedly found that the double-stranded RNA molecules of the present invention are particularly useful in treatment of cancer such as fluorouracil-resistant cancer as described in detail below.


The RNA molecule of the present invention is preferably isolated or derived from the bacteria of the genus Escherichia. The bacteria of the genus Escherichia include and only include Escherichia coli (Migula) Castellani & Chalmers. In an example embodiment, RNA molecule is isolated or derived from Escherichia coli (Migula) Castellani & Chalmers.


In more detail, the preferred sequences of the RNA molecules and double-stranded RNA molecules of the present invention are listed in Tables 1 and 3 below. In an embodiment, RNA molecules of SEQ ID NO: 195 to 241 as shown in Table 1 are isolated from a bacteria of genus Escherichia in particular from Escherichia coli (Migula) Castellani & Chalmers. These sequences are obtained by purification of RNA from Escherichia coli (Migula) Castellani & Chalmers. One possible approach to obtain the RNA molecules from a particular bacteria Escherichia co/i (Migula) Castellani & Chalmers is illustrated in Example 1. It would be appreciated that other suitable methods for obtaining the isolated and purified RNA molecules of the present invention according to the disclosure herein can be applied, and the methods can be subject to appropriate modification to obtain an improved yield of the RNA molecules, without departing from the scope of the present invention.









TABLE 1







RNA molecules in particular tRNAs isolated from Escherichia coli


(Migula) Castellani & Chalmers according to the present invention.









SEQ ID




NO.
tRNA
Sequence (5′-3′)





195
tRNA-Ala(UGC)
GGGGGCA[s4U]AGCUCAGCDGGGAGAGCGCCUGCUU[cmo5U]GCACGCAGGAG




[m7G]UCUGCGGTΨCGAUCCCGCGCGCUCCCACCA





196
tRNA-Ala(UGC)
GGGGCUAUAGCUCAGCDGGGAGAGCGCCUGCUU[cmo5U]GCACGCAGGAG




[m7G]UCUGCGGTΨCGAUCCCGCAUAGCUCCACCA





197
tRNA-Ala(GGC)
GGGGCUANAGCUCAGCDGGGAGAGCGCUUGCAUGGCAUGCAAGAG[m7G]UCA




GCGGTΨCGAUCCCGCUUAGCUCCACCA





198
tRNA-Arg(CCG)
GCGCCCGUAGCUCAGCDGGADAGAGCGCUGCC[s2C]UCCG[m1G]AGGCAGAG




[m7G]UCUCAGGTΨCGAAUCCUGUCGGGCGCGCCA





199
tRNA-Arg(ICG)
GCAUCCG[s4U]AGCUCAGCDGGDAGAGUACUCGG[s2C]UICG[m2A]ACCGAGCG




[m7G][acp3U]CGGAGGTΨCGAAUCCUCCCGGAUGCACCA





200
tRNA-Arg(ICG)
GCAUCCG[s4U]AGCUCAGCDGGADAGAGUACUCGGCUICG[m2A]ACCGAGCG




[m7G][acp3U]CGGAGGTΨCGAAUCCUCCCGGAUGCACCA





201
tRNA-Arg(UCU)
GUCCUCUUAGUUAAAUGGADAUAACGAGCCC[s2C]U[mnm5U]CU[t6A]AGGGCU




AAUUGCAGGTΨCGAUUCCUGCAGGGGACACCA





202
tRNA-Arg(UCU)
GCGCCCUUAGCUCAGUUGGAUAGAGCAACGAC[s2C]U[mnm5U]CU[t6A]AGΨCG




UGGGCCGCAGGTΨCGAAUCCUGCAGGGCGCGCCA





203
tRNA-Asn(GUU)
UCCUCUG[s4U]AGUUCAGDCGGDAGAACGGCGGACUQUU[t6A] Ψ




CCGUAU[m7G]UCACUGGTΨCGAGUCCAGUCAGAGGAGCCA





204
tRNA-Asp(QUC)
GGAGCGG[s4U]AGUUCAGDCGGDDAGAAUACCUGCCU[gluQ]UC[m2A]CGCAGG




GG[m7G]UCGCGGGTΨCGAGUCCCGΨCCGUUCCGCCA





205
tRNA-Cys(GCA)
GGCGCGU[s4U]AACAAAGCGGDDAUGUAGCGGA Ψ UGCA[ms2i6A]A Ψ




CCGUCUAGUCCGGTΨCGACUCCGGAACGCGCCUCCA





206
tRNA-Gln(UUG)
UGGGGUA[s4U]CGCCAAGC[Gm]GDAAGGCACCGGU[Um]U[cmnm5s2U]UG[m2A]




ΨACCGGCAUUCCCUGGTΨCGAAUCCAGGUACCCCAGCCA





207
tRNA-Gln(CUG)
UGGGGUA[s4U]CGCCAAGC[Gm]GDAAGGCACCGGA[Um]UCUG[m2A] ψ ψ




CCGGCAUUCCGAGGTUψCGAAUCCUCGUACCCCAGCCA





208
tRNA-Glu(UUC)
GUCCCCUUCGUCψAGAGGCCCAGGACACCGCCCU[mnm5s2U]UC[m2A]CGGCGG




UAACAGGGGTψCGAAUCCCCUGGGGGACGCCA





209
tRNA-Glu(UUC)
GUCCCCUUCGUCψAGAGGCCCAGGACACCGCCCU[mnm5s2U]UC[m2A]CGGCGG




UAACAGGGGTψCGAAUCCCCUAGGGGACGCCA





210
tRNA-Glu(UUC)
GUCCCCUUCGUCψAGAGGCCAGGACACCGCCCU[mnm5s2U]UC[m2A]CGGCGGU




AACAGGGGTψCGAAUCCCCUAGGGGACGCCA





211
tRNA-Gly(CCC)
GCGGGCG[s4U]AGUUCAAUGGDAGAACGAGAGCUUCCCAAGCUCUAUACGAG




GGTψCGAUUCCCUUCGCCCGCUCCA





212
tRNA-Gly(UCC}
GCGGGCAUCGUAUAAUGGCUAUUACCUCAGCCU[mnm5U]CCAAGCUGAUGAU




GCGGGTψCGAUUCCCGCUGCCCGCUCCA





213
tRNA-Gly(GCC)
GCGGGAAUAGCUCAGDDGGDAGAGCACGACCUUGCCAAGGUCGGG[m7G]UCG




CGAGTψCGAGUCUCGUUUCCCGCUCCA





214
tRNA-His(GUG)
GGUGGCUA[s4U]AGCUCAGDDGGDAGAGCCCUGGAUUQUG[m2A] ψ ψ




CCAGUU[m7G]UCGUGGGTψCGAAUCCCAUUAGCCACCCCA





215
tRNA-Ile(GAU)
AGGCUUGUAGCUCAGGDGGDDAGAGCGCACCCCUGAU[t6A]AGGGUGAG[m7G]




[acp3U]CGGUGGTψCAAGUCCACψCAGGCCUACCA





216
tRNA-Ile(GAU)
AGGCUUGUAGCUCAGGUGGDDAGAGCGCACCCCUGAU[t6A]AGGGUGAG[m7G]




[acp3U]CGGUGGTψCAAGUCCACψCAGGCCUACCA





217
tRNA-Ile(CAU)
GGCCCCU[s4U]AGCUCAGU[Gm]GDDAGAGCAGGCGACU[k2C]AU[t6A]A ψ




CGCUUG[m7G][acp3U]CGCUGGTψCAAGUCCAGCAGGGGCCACCA





218
tRNA-Leu(CAG)
GCGAAGGUGGCGGAADD[Gm]GDAGACGCGCUAGCUUCAG[m1G] ψ G ψ




UAGUGUCCUUACGGACGUGGGGGTψCAAGUCCCCCCCCUCGCACCA





219
tRNA-Leu(GAG)
GCCGAGGUGGUGGAADD[Gm]GDAGACACGCUACCUUGAG[m1G] ψ




GGUAGUGCCCAAUAGGGCUUACGGGTψCAAGUCCCGUCCUCGGUACCA





220
tRNA-Leu(AAA)
GCCCGGA[s4U]GGUGGAADC[Gm]GDAGACACAAGGGA ψ




U[cmnm5Um]AA[ms2i6A]A ψ CCCUCGGCGUUCGCGCUGUGCGGGT ψ




CAAGUCCCGCUCCGGGUACCA





221
tRNA-Leu(CAA)
GCCGAAG[s4U]GGCGAAADC[Gm]GDAGACGCAGUUGA ψ U[Cm]AA[ms2i6A]A ψ




CAACCGUAGAAAUACGUGCCGGTψCGAGUCCGGCCUUCGGCACCA





222
tRNA-Lys(UUU)
GGGUCGUUAGCUCAGDDGGDAGAGCAGUUGACU[mnm5s2U]UU[t6A]A ψ




CAAUUG[m7G][acp3U]CGCAGGTψCGAAUCCUGCACGACCCACCA





223
tRNA-Met(CAU)
GGCUACG[s4U]AGCUCAGDD[Gm]GDDAGAGCACAUCACU[ac4C]AU[t6A]A ψ




GAUGGG[m7G][acp3U]CACAGGTψCGAAUCCCGUCGUAGCCACCA





224
tRNA-Phe(GAA)
GCCCGGA[s4U]AGCUCAGDCGGDAGAGCAGGGGA ψ UGAA[ms2i6A]A Ψ




CCCCGU[m7G][acp3U]CCUUGGTψCGAUUCCGAGUCCGGGCACCA





225
tRNA-Pro(CGG)
CGGUGAUUGGCGCAGCCUGGDAGCGCACUUCGUUCGG[m1G]ACGAAGGG[m7G]




UCGGAGGTΨCGAAUCCUCUAUCACCGACCA





226
tRNA-Sec(UCA)
AAGAUCG[s4U]CGUCUCCGGDGAGGCGGCUGGACUUCA[i6A]AUCCAGUUGGG




GCCGCGCGGUCCCGGGCAGGTΨCGACUCCUGUGAUCUUGCCA





227
tRNA-Ser(UGA)
GGAAGUG[s4U]GGCCGAGC[Gm]GDDGAAGGCACCGGU[Cm]U[cmo5U]GA[ms2i6A]




AACCGGCGACCCGAAAGGGUUCCAGAGTΨCGAAUCUCUGCGCUUCCGCCA





228
tRNA-Ser(CGA)
GGAGAGAUGCCGGAGC[Gm]GCDGAACGGACCGGUCUCGA[ms2i6A]AACCGGA




GUAGGGGCAACUCUACCGGGGGTΨCAAAUCCCCCUCUCUCCGCCA





229
tRNA-Ser(GCU)
GGUGAGG[s4U]GGCCGAGAGGCDGAAGGCGCUCCC[s2C]UGCU[t6A]AGGGAGU




AUGCGGUCAAAAGCUGCAUCCGGGGTΨCGAAUCCCCGCCUCACCGCCA





230
tRNA-Ser(GGA)
GGUGAGG[s4U]GUCCGAGU[Gm]GDDGAAGGAGCACGCCUGGAAAG Ψ




GUGUAUACGGCAACGUAUCGGGGGTΨCGAAUCCCCCCCUCACCGCCA





231
tRNA-Ser(GGA)
GGUGAGGUGUCCGAGU#GCDGAAGGAGCACGCCUGGAAAGΨGUGUAUACGGC




AACGUAUCGGGGGTΨCGAAUCCCCCCCUCACCGCCA





232
tRNA-Thr(GGU)
GCUGAUAUGGCUCAGDDGGDAGAGCGCACCCUUGGU[m6t6A]AGGGUGAG[m7G]




UCCCCAGTΨCGACUCUGGGUAUCAGCACCA





233
tRNA-Thr(GGU)
GCUGAUAUAGCUCAGDDGGDAGAGCGCACCCUUGGU[m6t6A]AGGGUGAG[m7G]




UCGGCAGTΨCGAAUCUGCCUAUCAGCACCA





234
tRNA-Trp(CCA)
AGGGGCG[s4U]AGUUCAADDGGDAGAGCACCGGU[Cm]UCCA[ms2i6A]AACCGG




GU[m7G]UUGGGAGTΨCGAGUCUCUCCGCCCCUGCCA





235
tRNA-Tyr(QUA)
GGUGGGG[s4U][s4U]CCCGAGC[Gm]GCCAAAGGGAGCAGACUQUA[ms2i6A]A Ψ




CUGCCGUCAUCGACUUCGAAGGTΨCGAAUCCUUCCCCCACCACCA





236
tRNA-Tyr(QUA)
GGUGGGG[s4U][s4U]CCCGAGC[Gm]GCCAAAGGGAGCAGACUQUA[ms2i6A]A Ψ




CUGCCGUCACAGACUUCGAAGGTΨCGAAUCCUUCCCCCACCACCA





237
tRNA-Val(UAC)
GGGUGAU[s4U]AGCUCAGCDGGGAGAGCACCUCCCU[cmo5U]AC[m6A]AGGAGG




GG[m7G]UCGGCGGTΨCGAUCCCGUCAUCACCCACCA





238
tRNA-Val(GAC)
GCGUCCG[s4U]AGCUCAGDDGGDDAGAGCACCACCUUGACAUGGUGGGG[m7G]




[acp3U]CGGUGGTΨCGAGUCCACUCGGACGCACCA





239
RNA-Val(GAC)
GCGUUCA[s4U]AGCUCAGDDGGDDAGAGCACCACCUUGACAUGGUGGGG[m7G]




[acp3U]CGUUGGTΨCGAGUCCAAUUGAACGCACCA





240
tRNA-Ini(CAU)
CGCGGGG[s4U]GGAGCAGCCUGGDAGCUCGUCGGG[Cm]UCAUAACCCGAAG[m7G]




UCGUCGGTΨCAAAUCCGGCCCCCGCAACCA





241
tRNA-Ini(CAU)
CGCGGGG[s4U]GGAGCAGCCUGGDAGCUCGUCGGG[Cm]UCAUAACCCGAAGAU




CGUCGGTΨCAAAUCCGGCCCCCGCAACCA
















TABLE 2







Abbreviations of chemical modifications symbols of tRNA sequences








Symbols
Common name





m1A
1-methyladenosine


m2A
2-methyladenosine


m6A
N6 methyladenosine


Am
2′-O-methyladenosine


i6A
N6-isopentenyl adenosine


ms2i6A
2-methylthio-N6-isopentenyl adenosine


t6A
N6 threonide carbamoyl adenosine


m6t6A
N6-methyl-N6-threosylcarbamoyl adenosine


I
Inosine


m5C
5-methylcytidine


Cm
2′-O-methylcytidine


s2C
2-thiocytidine


ac4C
N4 acetylcytidine


f5C
5-formylcytidine


k2C
Lysidine


m1G
1-methylguanosine


m2G
N2 methylguanosine


m7G
7-methylguanosine


Gm
2′-O-methylguanosine


m22G
N2, N2 dimethylguanosine


Q
Queuosine


galQ
Galactosyl queuosine


ψ
Pseudouridine


D
Dihydrouridine


T
5-methyluridine


Um
2′-O-methyluridine


s2U
2-thiouridine


s4U
4-thiouridine


acp3U
3-(3-amino-3-carboxypropyl) uridine


mo5U
5-methoxyuridine


cmo5U
Uridine 5-oxoacetic acid


mcmo5U
Uridine 5-oxoacetate methyl ester


mcm5U
5-methoxycarbonylmethyl uridine


mcm5Um
5-methoxycarbonylmethyl-2′-O-methyluridine


mcm5s2U
5-methoxycarbonylmethyl-2-thiouridine


nm5s2U
5-aminomethyl-2-thiouridine


mnm5U
5-methylaminomethyl uridine


mnm5s2U
5-methylaminomethyl-2-thiouridine


ncm5U
5-aminoformylmethyluridine


ncm5Um
5-carbamoylmethyl-2′-O-methyluridine


cmnm5U
5-carboxymethyl aminomethyl uridine


cmnm5Um
5-carboxymethylaminomethyl-2′-O-methyluridine


cmnm5s2U
5-carboxymethylaminomethyl-2-methyluridine


tm5U
5-taurine methyluridine


tm5s2U
5-tauromethyl-2-thiouridine









The antisense sequences of SEQ ID NO: 7 to SEQ ID NO: 100 and the sense sequences of SEQ ID NO: 101 to SEQ ID NO: 194 as shown in Table 3 are artificially synthesized in accordance with the present invention. In particular, these sequences are derived sequence fragments prepared according to the sequences in Table 1 isolated from Escherichia coli (Migula) Castellani & Chalmers. Said derived sequence fragments are classified into 2 groups, namely a 5′-tRFs, and a 3′-tRFs. The 5′-t group RNA molecules contain a 2-35 nucleotides cleaved at 5′ terminal portion, D loop, D stem loop, anticodon loop or anticodon stem loop of the corresponding full-length tRNA molecules isolated from the bacteria; and the 3′-t group RNA molecules contain a 2-35 nucleotides cleaved at 3′-CCA terminal portion, T loop, T stem loop, anticodon loop, anticodon stem loop of the corresponding full-length tRNA molecules isolated from the bacteria. In another embodiment, tRF obtained from tRNA-Cys(GCA) comprises 22 nucleotides long 5′-tRFs “GGCGCGUUAACAAAGCGGUUAU”, which corresponds to SEQ ID No: 7 and 22 nucleotides long 5′-tRFs “UCGACUCCGGAACGCGCCUCCA”, which corresponds to SEQ ID No: 8.


Each of the sense sequences together with the corresponding antisense sequence form a double-stranded RNA molecule. As shown in Table 3, the sense sequence of SEQ ID NO: 101 and the antisense sequence of SEQ ID NO: 7 form a double-stranded RNA molecule with a length of 22 base pairs, and the resultant RNA molecule is denoted as EC for easy reference.


The double-stranded RNA molecules are classified into 2 groups, namely a 5′-terminal group (5′-t), and a 3′-terminal group (3′-t). The 5′-t group RNA molecules contain a 5′ terminal portion of the corresponding full-length RNA molecules isolated from the bacteria; and the 3′-t group RNA molecules contain a 3′ terminal portion of the corresponding full-length RNA molecules isolated from the bacteria. In another embodiment, RNA molecules may contain the anticodon loop portion of the corresponding full-length RNA molecules isolated from the gut microorganisms and referred as anticodon group RNA molecules. The antisense sequences of SEQ ID NO: 7 to SEQ ID NO: 100 can be generated by cleavage at different sites on the full-length RNA molecules SEQ ID NO: 195 to 241.


Further, the RNA molecule of the present invention may comprise a 3′ overhang, preferably comprise 2 mer 3′ overhangs. The provision of the 3′ overhang improves the stability of the RNA molecules.









TABLE 3







RNA molecules derived from the sequences in Table 1 through artificial


synthesis according to the present invention.














Code
SEQ ID
Antisense sequence
SEQ ID
Sense sequence



Source
(mimic)
NO.
(5′-3′)
NO.
(5′-3′)
Group
















tRNA-Cys(GCA)
EC1
7
GGCGCGUUAACAAAGC
101
AUAACCGCUUUGUUA
5′-t





GGUUAU

ACGCGCC




EC2
8
UCGACUCCGGAACGCG
102
UGGAGGCGCGUUCCG
3′-t





CCUCCA

GAGUCGA






tRNA-His(GUG)
EC3
9
GGUGGCUAUAGCUCA
103
UACCAACUGAGCUAU
5′-t





GUUGGUA

AGCCACC




EC4
10
UCGAAUCCCAUUAGCC
104
UGGGGUGGCUAAUG
3′-t





ACCCCA

GGAUUCGA






tRNA-Lys(UUU)
EC5
11
GGGUCGUUAGCUCAG
105
CUACCAACUGAGCUA
5′-t





UUGGUAG

ACGACCC




EC6
12
UCGAAUCCUGCACGAC
106
UGGUGGGUCGUGCAG
3′-t





CCACCA

GAUUCGA






tRNA-Met(CAU)
EC7
13
GGCUACGUAGCUCAG
107
UAACCAACUGAGCUA
5′-t





UUGGUUA

CGUAGCC




EC8
14
UCGAAUCCCGUCGUA
108
UGGUGGCUACGACG
3′-t





GCCACCA

GGAUUCGA






tRNA-Asn(GUU)
EC9
15
UCCUCUGUAGUUCAG
109
CUACCGACUGAACUA
5′-t





UCGGUAG

CAGAGGA




EC10
16
UCGAGUCCAGUCAGA
110
UGGCUCCUCUGACUG
3′-t





GGAGCCA

GACUCGA






tRNA-Phe(GAA)
EC11
17
GCCCGGAUAGCUCAG
111
CUACCGACUGAGCUA
5′-t





UCGGUAG

UCCGGGC




EC12
18
UCGAUUCCGAGUCCG
112
UGGUGCCCGGACUCG
3′-t





GGCACCA

GAAUCGA






tRNA-Trp(CCA)
EC13
19
AGGGGCGUAGUUCAA
113
CUACCAAUUGAACUA
5′-t





UUGGUAG

CGCCCCU




EC14
20
UCGAGUCUCUCCGCCC
114
UGGCAGGGGCGGAG
3′-t





CUGCCA

AGACUCGA






tRNA-Asp(QUC)
EC15
21
GGAGCGGUAGUUCAG
115
UAACCGACUGAACUA
5′-t





UCGGUUA

CCGCUCC




EC16
22
UCGAGUCCCGUCCGUU
116
UGGCGGAACGGACG
3′-t





CCGCCA

GGACUCGA






RNA-Pro(CGG)
EC17
23
CGGUGAUUGGCGCAG
117
UACCAGGCUGCGCCA
5′-t





CCUGGUA

AUCACCG




EC18
24
UCGAAUCCUCUAUCAC
118
UGGUCGGUGAUAGA
3′-t





CGACCA

GGAUUCGA






tRNA-Sec(UCA)
EC19
25
AAGAUCGUCGUCUCC
119
CCUCACCGGAGACGA
5′-t





GGUGAGG

CGAUCUU




EC20
26
UUCGACUCCUGUGAU
120
UGGCAAGAUCACAG
3′-t





CUUGCCA

GAGUCGAA






tRNA-Ala(GGC)
EC21
27
GGGGCUAUAGCUCAG
121
CUCCCAGCUGAGCUA
5′-t





CUGGGAG

UAGCCCC




EC22
28
UCGAUCCCGCUUAGCU
122
UGGUGGAGCUAAGC






CCACCA

GGGAUCGA






tRNA-Ala(UGC)
EC23
29
GGGGGCAUAGCUCAG
123
CUCCCAGCUGAGCUA
5′-t





CUGGGAG

UGCCCCC




EC24
30
UCGAUCCCGCGCGCUC
124
UGGUGGGAGCGCGC
3′-t





CCACCA

GGGAUCGA






tRNA-Ala(UGC)
EC25
31
GGGGCUAUAGCUCAG
125
CUCCCAGCUGAGCUA
5′-t





CUGGGAG

UAGCCCC




EC26
32
UCGAUCCCGCAUAGCU
126
UGGUGGAGCUAUGC
3′-t





CCACCA

GGGAUCGA






tRNA-Ser(CGA)
EC27
33
GGAGAGAUGCCGGAG
127
UCAGCCGCUCCGGCA
5′-t





CGGCUGA

UCUCUCC




EC28
34
UCAAAUCCCCCUCUCU
128
UGGCGGAGAGAGGG
3′-t





CCGCCA

GGAUUUGA






tRNA-Ser(GCU)
EC29
35
GGUGAGGUGGCCGAG
129
UCAGCCUCUCGGCCA
5′-t





AGGCUGA

CCUCACC




EC30
36
UCGAAUCCCCGCCUCA
130
UGGCGGUGAGGCGG
3′-t





CCGCCA

GGAUUCGA






tRNA-Ser(GGA)
EC31
37
GGUGAGGUGUCCGAG
131
UCAGCCACUCGGACA
5′-t





UGGCUGA

CCUCACC




EC32
38
UCGAAUCCCCCCCUCA
132
UGGCGGUGAGGGGG
3′-t





CCGCCA

GGAUUCGA






tRNA-Ser(GGA)
EC33
39
GGUGAGGUGUCCGAG
133
UCAACCACUCGGACA
5′-t





UGGUUGA

CCUCACC




EC34
40
UCGAAUCCCCCCCUCA
134
UGGCGGUGAGGGGG
3′-t





CCGCCA

GGAUUCGA






tRNA-Ser(UGA)
EC35
41
GGAAGUGUGGCCGAG
135
UCAACCGCUCGGCCA
5′-t





CGGUUGA

CACUUCC




EC36
42
UCGAAUCUCUGCGCU
136
UGGCGGAAGCGCAG
3′-t





UCCGCCA

AGAUUCGA






tRNA-Val(GAC)
EC37
43
GCGUCCGUAGCUCAG
137
UAACCAACUGAGCUA
5′-t





UUGGUUA

CGGACGC




EC38
44
UCGAGUCCACUCGGAC
138
UGGUGCGUCCGAGU
3′-t





GCACCA

GGACUCGA






tRNA-Val(GAC)
EC39
45
GCGUUCAUAGCUCAG
139
UAACCAACUGAGCUA
5′-t





UUGGUUA

UGAACGC




EC40
46
UCGAGUCCAAUUGAA
140
UGGUGCGUUCAAUU
3′-t





CGCACCA

GGACUCGA






tRNA-Val(UAC)
EC41
47
GGGUGAUUAGCUCAG
141
CUCCCAGCUGAGCUA
5′-t





CUGGGAG

AUCACCC




EC42
48
UCGAUCCCGUCAUCAC
142
UGGUGGGUGAUGAC
3′-t





CCACCA

GGGAUCGA






tRNA-Arg(CCG)
EC43
49
GCGCCCGUAGCUCAGC
143
UAUCCAGCUGAGCUA
5′-t





UGGAUA

CGGGCGC




EC44
50
UCGAAUCCUGUCGGG
144
UGGCGCGCCCGACAG
3′-t





CGCGCCA

GAUUCGA






tRNA-Arg(ICG)
EC45
51
GCAUCCGUAGCUCAGC
145
CUACCAGCUGAGCUA
5′-t





UGGUAG

CGGAUGC




EC46
52
UCGAAUCCUCCCGGAU
146
UGGUGCAUCCGGGA
3′-t





GCACCA

GGAUUCGA






tRNA-Arg(CCG)
EC47
53
GCAUCCGUAGCUCAGC
147
UAUCCAGCUGAGCUA
5′-t





UGGAUA

CGGAUGC




EC48
54
UCGAAUCCUCCCGGAU
148
UGGUGCAUCCGGGA
3′-t





GCACCA

GGAUUCGA






tRNA-Arg(UCU)
EC49
55
GUCCUCUUAGUUAAA
149
AUAUCCAUUUAACU
5′-t





UGGAUAU

AAGAGGAC




EC50
56
UCGAUUCCUGCAGGG
150
UGGUGUCCCCUGCAG
3′-t





GACACCA

GAAUCGA






tRNA-Arg(UCU)
EC51
57
GCGCCCUUAGCUCAGU
151
UAUCCAACUGAGCUA
5′-t





UGGAUA

AGGGCGC




EC52
58
UCGAAUCCUGCAGGG
152
UGGCGCGCCCUGCAG
3′-t





CGCGCCA

GAUUCGA






tRNA-Gln(CUG)
EC53
59
UGGGGUAUCGCCAAG
153
CUUACCGCUUGGCGA
5′-t





CGGUAAG

UACCCCA




EC54
60
UCGAAUCCUCGUACCC
154
UGGCUGGGGUACGA
3′-t





CAGCCA

GGAUUCGA






tRNA-Gln(UUG)
EC55
61
UGGGGUAUCGCCAAG
155
CUUACCGCUUGGCGA
5′-t





CGGUAAG

UACCCCA




EC56
62
UCGAAUCCAGGUACCC
156
UGGCUGGGGUACCU
3′-t





CAGCCA

GGAUUCGA






tRNA-Ile(GAU)
EC57
63
AGGCUUGUAGCUCAG
157
UAACCACCUGAGCUA
5′-t





GUGGUUA

CAAGCCU




EC58
64
UCAAGUCCACUCAGGC
158
UGGUAGGCCUGAGU
3′-t





CUACCA

GGACUUGA






tRNA-Ile(GAU)
EC59
65
AGGCUUGUAGCUCAG
159
UAACCACCUGAGCUA
5′-t





GUGGUUA

CAAGCCU




EC60
66
UCAAGUCCACUCAGGC
160
UGGUAGGCCUGAGU
3′-t





CUACCA

GGACUUGA






tRNA-Ile(UAU)
EC61
67
GGCCCCUUAGCUCAGU
161
CUAACCACUGAGCUA
5′-t





GGUUAG

AGGGGCC




EC62
68
UCAAGUCCAGCAGGG
162
UGGUGGCCCCUGCUG
3′-t





GCCACCA

GACUUGA






tRNA-Thr(GGU)
EC63
69
GCUGAUAUAGCUCAG
163
CUACCAACUGAGCUA
5′-t





UUGGUAG

UAUCAGC




EC64
70
UCGAAUCUGCCUAUC
164
UGGUGCUGAUAGGC
3′-t





AGCACCA

AGAUUCGA






tRNA-Thr(GGU)
EC65
71
GCUGAUAUGGCUCAG
165
CUACCAACUGAGCCA
5′-t





UUGGUAG

UAUCAGC




EC66
72
UCGACUCUGGGUAUC
166
UGGUGCUGAUACCCA
3′-t





AGCACCA

GAGUCGA






tRNA-Glu(UUC)
EC67
73
GUCCCCUUCGUCUAGA
167
UGGGCCUCUAGACGA
5′-t





GGCCCA

AGGGGAC




EC68
74
UCGAAUCCCCUGGGG
168
UGGCGUCCCCCAGGG
3′-t





GACGCCA

GAUUCGA






tRNA-Glu(UUC)
EC69
75
GUCCCCUUCGUCUAGA
169
UGGGCCUCUAGACGA
5′-t





GGCCCA

AGGGGAC




EC70
76
UCGAAUCCCCUAGGG
170
UGGCGUCCCCUAGGG
3′-t





GACGCCA

GAUUCGA






tRNA-Glu(UUC)
EC71
77
GUCCCCUUCGUCUAGA
171
CUGGCCUCUAGACGA
5′-t





GGCCAG

AGGGGAC




EC72
78
UCGAAUCCCCUAGGG
172
UGGCGUCCCCUAGGG
3′-t





GACGCCA

GAUUCGA






tRNA-lni(CAU)
EC73
79
CGCGGGGUGGAGCAG
173
UACCAGGCUGCUCCA
5′-t





CCUGGUA

CCCCGCG




EC74
80
UCAAAUCCGGCCCCCG
174
UGGUUGCGGGGGCC
3′-t





CAACCA

GGAUUUGA






tRNA-lni(CAU)
EC75
81
CGCGGGGUGGAGCAG
175
UACCAGGCUGCUCCA
5′-t





CCUGGUA

CCCCGCG




EC76
82
UCAAAUCCGGCCCCCG
176
UGGUUGCGGGGGCC
3′-t





CAACCA

GGAUUUGA






tRNA-Gly(CCC)
EC77
83
GCGGGCGUAGUUCAA
177
UCUACCAUUGAACUA
5′-t





UGGUAGA

CGCCCGC




EC78
84
UCGAUUCCCUUCGCCC
178
UGGAGCGGGCGAAG
3′-t





GCUCCA

GGAAUCGA






tRNA-Gly(GCC)
EC79
85
GCGGGAAUAGCUCAG
179
CUACCAACUGAGCUA
5′-t





UUGGUAG

UUCCCGC




EC80
86
UCGAGUCUCGUUUCCC
180
UGGAGCGGGAAACG
3′-t





GCUCCA

AGACUCGA






tRNA-Gly(UCC)
EC81
87
GCGGGCAUCGUAUAA
181
AUAGCCAUUAUACG
5′-t





UGGCUAU

AUGCCCGC




EC82
88
UCGAUUCCCGCUGCCC
182
UGGAGCGGGCAGCG
3′-t





GCUCCA

GGAAUCGA






tRNA-Leu(CAA)
EC83
89
GCCGAAGUGGCGAAA
183
CUACCGAUUUCGCCA
5′-t





UCGGUAG

CUUCGGC




EC84
90
UCGAGUCCGGCCUUCG
184
UGGUGCCGAAGGCCG
3′-t





GCACCA

GACUCGA






tRNA-Leu(CAG)
EC85
91
GCGAAGGUGGCGGAA
185
CUACCAAUUCCGCCA
5′-t





UUGGUAG

CCUUCGC




EC86
92
UCAAGUCCCCCCCCUC
186
UGGUGCGAGGGGGG
3′-t





GCACCA

GGACUUGA






tRNA-Leu(GAG)
EC87
93
GCCGAGGUGGUGGAA
187
CUCCCAAUUCCACCA
5′-t





UUGGGAG

CCUCGGC




EC88
94
UCAAGUCCCGUCCUCG
188
UGGUACCGAGGACG
3′-t





GUACCA

GGACUUGA






tRNA-Leu(AAA)
EC89
95
GCCCGGAUGGUGGAA
189
CUACCGAUUCCACCA
5′-t





UCGGUAG

UCCGGGC




EC90
96
UCAAGUCCCGCUCCGG
190
UGGUACCCGGAGCGG
3′-t





GUACCA

GACUUGA






tRNA-Tyr(QUA)
EC91
97
GGUGGGGUUCCCGAG
191
UUGGCCGCUCGGGAA
5′-t





CGGCCAA

CCCCACC




EC92
98
UCGAAUCCUUCCCCCA
192
UGGUGGUGGGGGAA
3′-t





CCACCA

GGAUUCGA






tRNA-Tyr(QUA)
EC93
99
GGUGGGGUUCCCGAG
193
UUGGCCGCUCGGGAA
5′-t





CGGCCAA

CCCCACC




EC94
100
UCGAAUCCUUCCCCCA
194
UGGUGGUGGGGGAA
3′-t





CCACCA

GGAUUCGA









The inventors unexpectedly found that the natural chemical modifications of RNA sequence derived from EC83 in Table 3 can enhance its inhibition effects on proliferation of colorectal cancer cells. The said antisense sequences of SEQ ID NO: 1 to SEQ ID NO: 3 and the sense sequences of SEQ ID NO: 4 to SEQ ID NO: 6 as shown in Table 4 are artificially synthesized in accordance with the present invention.









TABLE 4







RNA molecules derived from the EC83 mimic sequences in Table 2


through artificial synthesis according to the present invention.











Code
SEQ ID

SEQ ID



(mimic)
NO.
Antisense sequence (5′-3′)
NO.
Sense sequence (5′-3′)














EC83
89
GCCGAAGUGGCGAAAUCGGUAG
183
CUACCGAUUUCGCCACUUCGGC





EC83-M1
1
GCCGAAG[s4U]GGCGAAAUCGGUAG
4
CUACCGAUUUCGCCACUUCGGC





EC83-M2
2
GCCGAAGUGGCGAAAUC[Gm]GUAG
5
CUACCGAUUUCGCCACUUCGGC





EC83-M3
3
GCCGAAG[s4U]GGCGAAAUC[Gm]GUAG
6
CUACCGAUUUCGCCACUUCGGC









The inventors unexpectedly found that the RNA molecules isolated or derived from a bacteria of genus Escherichia in particular Escherichia coli (Migula) Castellani & Chalmers are effective against cancer cells, in particular they are capable of inhibiting the growth, proliferation and/or metastasis of cancer cells.


Turning back to the method of treatment, the method comprises the step of administering an effective amount of a RNA molecule as described above to the subject suffering from a cancer. In an embodiment, the step of administering the RNA molecule to the subject comprises contacting cancer cells of the subject with the RNA molecule.


The term “cancer” describes a physiological condition in subjects in which a population of cells are characterized by unregulated malignant (cancerous) cell growth. In an embodiment, the cancer to be treated is ovarian cancer, liver cancer, breast cancer, colorectal cancer, or lung cancer. In a particular embodiment, the cancer is colorectal cancer. In an alternative embodiment, the RNA molecules of the present invention are effective in treating cancer which is resistant against currently existing drugs such as fluorouracil, i.e. can be used to treat cancer which is resistant against fluorouracil. Specifically, the RNA molecules of the present invention can be used to treat fluorouracil-resistant colorectal cancer Accordingly, the method of the present invention can be applied to treat a subject suffering from a multi-drug resistant cancer and related disorders.


The term “subject” used herein refers to a living organism and can include but is not limited to a human and an animal. The subject is preferably a mammal, preferably a human. The RNA molecules may be administered through injection to the subject, preferably a human. The term injection encompasses intravenous, intramuscular, subcutaneous and intradermal administration. In an embodiment, the RNA molecule of the present invention is administered together with suitable excipient(s) to the subject through intravenous injection. For instance, the RNA molecule may be delivered to the subject or cells via transfection, electroporation or viral-mediated delivery.


The expression “effective amount” generally denotes an amount sufficient to produce therapeutically desirable results, wherein the exact nature of the result varies depending on the specific condition which is treated. In this invention, cancer is the condition to be treated and therefore the result is usually an inhibition or suppression of the growth or proliferation of cancer cells, a reduction of cancerous cells or the amelioration of symptoms related to the cancer cells, in particular inhibition of the proliferation of the cancer cells or induction of cell death, i.e. apoptosis of the cancer cells. In an embodiment where the cancer is metastatic cancer, the result is usually an inhibition of migration of cancer cells, suppression of the invasion of cancer cells to other tissues, inhibition of formation metastasis cancer cells at a secondary site distant from the primary site, or amelioration of symptoms related to metastatic cancer.


The effective amount of the RNA molecules of the present invention may depend on the species, body weight, age and individual conditions of the subject and can be determined by standard procedures such as with cell cultures or experimental animals.


The RNA molecule of the present invention may be administered in form of a pharmaceutical composition comprising the RNA molecule and at least one pharmaceutically tolerable excipient. The pharmaceutically tolerable excipient may be one or more of a diluent, a filler, a binder, a disintegrant, a lubricant, a coloring agent, a surfactant, a gene delivery carrier and a preservative. The pharmaceutical composition can be present in solid, semisolid or liquid form, preferably in liquid form. The pharmaceutical composition may comprise further pharmaceutical effective ingredients such as therapeutic compounds which are used for treating cancer such as fluorouracil. The skilled person is able to select suitable pharmaceutically tolerable excipients depending on the form of the pharmaceutical composition and is aware of methods for manufacturing pharmaceutical compositions as well as able to select a suitable method for preparing the pharmaceutical composition depending on the kind of pharmaceutically tolerable excipients and the form of the pharmaceutical composition.


In an embodiment, the RNA molecule is provided in a pharmaceutical composition comprising a gene delivery carrier. The gene delivery carrier refers to any molecules that can act as a carrier for delivering a gene into a cell. In an embodiment where the RNA molecule is transfected into a cell, the gene delivery carrier is considered as a transfecting agent. In an embodiment where the RNA molecule is delivered through a recombinant viral vector, the gene delivery carrier is a viral vector carrying the double-stranded RNA molecule of the present invention. The gene delivery carriers include, but is not limited to, a vector such as a viral vector, a collagen such as atelocollagen, a polymer such as polyethylenimine (PEI), a polypeptide such as poly (L-lysine) and protamine, and a lipid for forming a liposome such as Lipofectamine. The gene delivery carriers may be commercially available such as Lipofectamine RNAiMAX Transfection Reagent, Lipofectamine 3000 Reagent, and Lipofectamine® 2000 Transfection Reagent from Thermo Fisher, U.S.A.; RNAi-Mate from GenePharma, China; atelocollagen from Koken Co., Ltd., Japan); and Histidine-Lysine peptide copolymer from siRNAomics, China. The gene delivery carriers may be viral vectors based on retrovirus, adeno-associated virus, adenovirus, and lentivirus. The gene delivery carriers should have a low toxicity and cannot induce significant immune response in the subject. In an embodiment, the RNA molecule is provided in a pharmaceutical composition comprising atelocollagen, wherein atelocollagen forms a complex with the RNA molecule for delivery. In another embodiment, the RNA molecule is provided in a pharmaceutical composition comprising Lipofectamine such as Lipofectamine® RNAiMAX transfection reagent for delivering the RNA molecule to the cells. In a further embodiment, the RNA molecule is inserted into a plasmid and form recombinant vector.


In an embodiment, the pharmaceutical composition may further comprise a nucleic acid stabilizer. The nucleic acid stabilizer refers to any chemicals that are capable of maintaining the stability of the RNA molecule in the composition to minimize or avoid degradation, in particular those having ability to deactivate activity of nucleases or the like degrading the RNA molecules.


Accordingly, the present invention also pertains to a pharmaceutical composition as described above, in particular comprising the RNA molecule and a pharmaceutically tolerable excipient as defined above. In an embodiment, the RNA molecule comprises at least one sequence selected from SEQ ID NO: 1 to 100 or a functional variant or homologue thereof. Preferably, the RNA molecule is isolated or derived from a bacteria of the genus Escherichia as described above, in particular from Escherichia coli (Migula) Castellani & Chalmers.


The administration step of the RNA molecule according to the method of the present invention may be performed by injecting a pharmaceutical composition containing the RNA molecule to the target site of the subject, i.e. where cancer cells exist or body tissue adjacent to cancer cells. This is advantageous in that the RNA molecule can be directly delivered to the cancer cells before any cellular degradation such as first pass metabolism.


The RNA molecules of the present invention are also suitable for inhibiting growth or proliferation of cancer cells. In another aspect of the invention, there is provided a method of inhibiting growth or proliferation of cancer cells comprising a step of contacting said cells with an effective amount of a RNA molecule as defined above. Preferably the RNA molecule is isolated or derived from a bacteria of the genus Escherichia or comprises a sequence selected from SEQ ID NO: 195 to SEQ ID NO: 241 or a functional variant or homologue thereof. The cancer cells are as defined above. Preferably, the cancer cells are colorectal cancer cells. The cancer cells may be resistant against currently existing cancer drugs such as but are not limited to fluorouracil.


In an embodiment, the RNA molecule has a sequence length of from about 50 to 200 nucleotides, more preferably has a length of from about 60 to about 150 nucleotides, in particular from about 70 to about 100 nucleotides. The RNA molecule is a noncoding molecule preferably a transfer RNA molecule. Preferably, the RNA molecule comprises a sequence selected from SEQ ID NO: 7 to SEQ ID NO: 100 or a functional variant or homologue thereof; or the RNA molecule comprises SEQ ID NO: 101 to SEQ ID NO: 194 or a functional variant or homologue thereof, or the RNA molecule consists of a sequence selected from SEQ ID NO: 7 to SEQ ID NO: 100 and/or SEQ ID NO: 101 to SEQ ID NO: 194 or a functional variant or homologue thereof.


In an alternative embodiment, the RNA molecule has a sequence length of from about 10 to about 30 base pairs, from about 15 to about 25 base pairs, from about 19 to about 22 base pairs, 19 base pairs or 22 base pairs. Preferably, the RNA molecule is a double-stranded RNA molecule comprising an antisense sequence selected from SEQ ID NO: 7 to SEQ ID NO: 100 or a functional variant or homologue thereof. Preferably, said double-stranded RNA molecule or a functional variant or homologue thereof comprises an antisense sequence selected from SEQ ID NO: 7 to SEQ ID NO: 100 and a complementary sense sequence selected from SEQ ID NO: 101 to SEQ ID NO: 194. Preferably, said double-stranded RNA molecule or a functional variant or homologue thereof comprises an antisense sequence as shown in SEQ ID NO: 47, 48, 89, 90, 91 or 92, and said double-stranded RNA molecule or a functional variant or homologue thereof comprises a sense sequence as shown in SEQ ID NO: 141, 142, 183, 184, 185 or 186. Preferably, said double-stranded RNA molecule or a functional variant or homologue thereof comprises an antisense sequence as shown in SEQ ID NO: 89, and said double-stranded RNA molecule or a functional variant or homologue thereof comprises a sense sequence as shown in SEQ ID NO: 183. In particular, in an embodiment, RNA molecule or a functional variant or homologue thereof is chemically modified double-stranded RNA molecules, comprising an antisense sequence selected from SEQ ID NO: 1 to SEQ ID NO: 3, and comprising a complementary sense sequence selected from SEQ ID NO: 4 to SEQ ID NO: 6. The RNA molecule may further comprise 2 mer 3′ overhangs.


The step of contacting the cancer cells with the RNA molecule of the present invention may be carried out by applying a composition in particular an incubation solution comprising the RNA molecule to said cancer cells which incubation solution may further comprise suitable excipients as defined above, a buffer or a suitable growth medium. In such embodiment of the present invention, the cancer cells are taken from a subject such as an animal or human, in particular a human. The RNA molecule is provided in the composition at a concentration of at least 3 nM, at least 5 nM, from about 5 nM to about 200 nM, from about 10 nM to about 100 nM, or from about 25 nM to about 50 nM. Further, the excipients may include a gene delivery carrier such as but is not limited to a collagen based carrier or a liposome forming agent. In an embodiment, the collagen based carrier is atelocollagen and the liposome forming agent is Lipofectamine.


The present invention pertains to a double-stranded RNA molecule as described above, i.e. comprising an antisense sequence selected from SEQ ID NO: 7 to SEQ ID NO: 100 or a functional variant or homologue thereof, and a complementary sense sequence. In particular, the double-stranded RNA molecule consists of an antisense sequence selected from SEQ ID NO: 7 to SEQ ID NO: 100 or a functional variant or homologue thereof, a complementary sense sequence selected from SEQ ID NO: 101 to SEQ ID NO: 194, and optionally a 3′ overhang.


Example embodiments of the double-stranded RNA molecule are presented in Table 3. The double-stranded RNA may be subject to modification and therefore may carry at least one modified nucleoside selected form inosine, 1-methyladenosine, 2-methyladenosine, N6-methyladenosine, N6-isopentenyladenosine, 2′-O-methyladenosine, N6-acetyladenosine, 1-methylinosine, pseudouridine, dihydrouridine, or 2-methylthio-N6-methyladenosine.


In addition to the above, the present invention pertains to tRNA-half cleaved from tRNA molecule in Table 1 using specific method such as S1 nuclease or other enzymes or reagents to probably cleave tRNA into tRNA-half. The tRNA-half may further being chemically modified, and therefore may carry at least one modified nucleoside selected form inosine, 1-methyladenosine, 2-methyladenosine, N6-methyladenosine, N6-isopentenyladenosine, 2′-O-methyladenosine, N6-acetyladenosine, 1-methylinosine, pseudouridine, dihydrouridine, or 2-methylthio-N6-methyladenosine.


In further aspect of the invention, there is provided a vector comprising a nucleic acid molecule, wherein the nucleic acid molecule is a RNA molecule as described above. In particular, the RNA molecule having a sequence selected from SEQ ID NO: 1 to SEQ ID NO: 6 or a functional variant or homologue thereof. In an embodiment, the vector is a recombinant vector comprising the double-stranded RNA molecule as described above. The vector may be viral-based vector derived from retrovirus, adeno-associated virus, adenovirus, or lentivirus. An ordinary skilled in the art would appreciate suitable approach to incorporate the RNA molecule of the present invention into a vector.


Still further, the present invention pertains to use of a nucleic acid molecule in the preparation of a medicament for treating cancer. The nucleic acid is a RNA molecule as described above including a functional variant or homologue thereof. It would also be appreciated that the RNA molecule of the present invention can be used as a small interfering RNA molecule to interfere the expression of certain genes in the target cancer cells, thereby to cause gene silencing, apoptosis, inhibition of cell growth and proliferation, or the like to achieve the desired therapeutic effect.


Accordingly, the present invention provides a novel and effective approach for treating cancers from various origins by administration of a RNA molecule that is isolated or derived from a bacteria of the genus Escherichia, or in particular a RNA molecule comprising a sequence selected from SEQ ID NO: 1 to 6. Administration of said RNA molecule is also suitable for inhibiting growth or proliferation of cancer cells. The RNA molecules are found to be highly effective at inhibiting growth and proliferation of cancer cells in vitro and exhibit an antitumor effect in vivo. Said RNA molecules are also effective against fluorouracil-resistant cell lines.


The invention is now described in the following non-limiting examples.


EXAMPLES
Chemicals and Materials


Escherichia coli MRE 600 total transfer ribonucleic acid was purchased from Roche (Basel, Switzerland). MicroRNA marker and low range ssRNA ladder were purchased from New England BioLabs (Massachusetts, U.S.A.). Diethylpyrocarbonate (DEPC)—treated water, S1 nuclease, RNase T1 and polyacrylamide containing a ratio of Acrylamide/Bis (19:1, w/w), tris/borate/EDTA (TBE), ammonium persulphate (APS) and tetramethylethylenediamine (TEMED), mirVana™ miRNA isolation kit, SYBR gold nucleic acid gel stain and gel loading buffer II were purchased from Thermo Fisher Scientific (U.S.A.). Guanidinium thiocyanate, triethylammonium acetate, hexafluoro-2-propanol and fluorouracil (5-FU) were purchased from Sigma (Missouri, U.S.A.). Ethanol was purchased from Anaqua Chemicals Supply Inc. Ltd. (U.S.A.). Deionized water was prepared by a Millipore Milli-Q Plus system (Millipore, U.S.A.). HCT-8 human ileocecal colorectal adenocarcinoma cell line and its 5-FU-resistant strain (HCT-8/5-FU), LoVo colorectal adenocarcinoma cell line and its 5-FU-resistant strain (LoVo/5-FU) were purchased from American Type Culture Collection (ATCC, U.S.A.). Opti-MEM medium, RPMI 1640 medium, 10% fetal bovine serum (FBS) and 1% penicillin/streptomycin (P/S) were purchased from Gibco (New Zealand). F-12K medium was purchased from Thermo (U.S.A.), MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] was purchased from Sigma (St Louis, MO, U.S.A.).


Example 1

Isolation of RNA molecules from mixed total tRNA of Escherichia coli Weighed powders of Escherichia coli MRE600 total tRNA were dissolved in DEPC-treated water. RNA concentration of the solutions was determined by NanoDrop (Thermo, U.S.A.). 100 μg of Escherichia coli total tRNA was directly injected into high-performance liquid weak-anion chromatography coupled with a diode array detector for separation of six fractions using the chromatographic conditions as follows: Column: TSKgel DNA-STAT column (4.6×100 i.d., 5 μm, Tosoh, Tokyo, Japan); The flow rate was set as 0.2 mL/min; The column was equilibrated with 20 mM Tris buffer (pH=8.5). Gradient elution with (A) 20 mM Tris buffer (pH=8.5) and (B) 20 mM Tris buffer+1 M NaCl (pH=8.5) was 0-120 min, 53%-63% B. The obtained six fractions were freeze-dried using a Speed-Vac system RVC 2-18 (Marin Christ, Germany). Powders of RNA and inorganic salts were then dissolved with DEPC-treated water and desalted by mirVana™ miRNA Isolation Kit. FIGS. 1A and 1B show that total tRNA was successfully separated into six different fractions by using this method.


Subsequently, fraction 2 and fraction 3 were separated by high-performance liquid ion-pair chromatography coupled with a diode array detector. The chromatographic conditions are as follows: Column: DNAPac RP column (3.0×100 i.d., 4 μm, Thermo); The flow rate was set as 0.2 mL/min; The column was equilibrated with 100 mM triethylammonium acetate (pH=7.0). Gradient elution with (A) 100 mM triethylammonium acetate (pH=7.0) and (B) 25% acetonitrile in A was 0-5 min, 30%-37% B; 5-25 min, 37%-45% B; 25-35 min, 45%-100% B; 35-45 min, 100% B. FIGS. 2A, 2B and 2C show that two RNA molecules with high purity were obtained from fraction 2 and fraction 3.


Example 2
Chemical Characterization of Purified RNA Molecules

Further, to characterize the tRNAs by mass spectrometry. The inventors employed ultra-high performance liquid chromatography coupled with quadruple time-of-flight mass technique (UHPLC-QTOF-MS) to quantitative analyze the accurate sequence information and chemical modifications of digested products of purified tRNAs. tRNA would be cleaved to several oligonucleotide fragments in length of 2-15 nt with terminal of guanine 3′-phosphoric acid. In negative mode of ESI source, oligonucleotides would have molecular ion peak with multiple charges. Charge numbers depend on the length of nucleotides, which means that nucleotide with longer length would carry more charges. The sequence information and chemical modifications are determined through collision-induced dissociation (CID) analysis of the rule and products information of tRNA-RNase T1 digestions. At the same impact voltage, the stronger the excimer ion peak intensity is, the greater the fragment response intensity is. In addition, the molecular ion peaks with more charges are easier to be cleaved, and the more fragment information is generated. Due to the complexity of CID profiles of oligonucleotide fragments longer than 8 nt, multiple excimer ions and the corresponding optimal collision energy were selected in the process of sequence analysis. CID cleavage of oligonucleotides is most easily cleaved at the phosphodiester bond and the junction between base and ribose, resulting in a series of characteristic fragments, mainly a-B, c, y and w type ions. FIG. 3 shows that the sequence of oligonucleotides can be analyzed by c, y and w type ions, and the type of nucleotide modification can be further determined by type a-B ions.


The purified RNA was freeze-dried and redissolved with RNase-treated water. Each 1 μg of purified RNA was mixed with 50 units of RNase T1 and mixed with ammonium acetate (220 mM). After incubation in water bath at 37° C. for 1.5 h, the mixed solutions were incubated at 70° C. for 10 minutes to terminate the reaction. After centrifugation at 10000×g for 1 min, the supernatant was collected for UHPLC-MS analysis.


UHPLC-MS/MS was performed on an Agilent UHPLC 1290 system (Agilent Technologies, Santa Clara, CA, USA), equipped with a vacuum degasser, a quaternary pump, an autosampler, a diode array detector and an Agilent ultrahigh definition 6545 Q-TOF mass spectrometer. Separation was carried out on an ACQUITY UPLC OST C18 Column (2.1×100 mm i.d., 1.7 μm, Waters, Massachusetts, U.S.A.) at 60° C. The flow rate was set at 0.2 mL/min and sample injection volume was 20 μL. Gradient elution with (A) 100 mM hexafluoro-2-propanol (HFIP)+15 mM trimethylamine (TEA) and (B) 50% MeOH in A was 0-1.5 min, 2% B, 1.5-8.3 min, 2%-28% B, 8.3-16.5 min, 28%-34% B, followed by washing with 80% B and equilibration with 2% B. ESI conditions were as follows: gas temperature 320° C., spray voltage 3.5 kV, sheath gas flow and temperature were set as 12 L/min and 350° C., respectively. FIGS. 4A and 4B show the MS2 results of specific RNase T1 digestion products of purified RNA molecules, the signals are in accordance with that of the specific fragments of Escherichia coli tRNA in database. Thus, the purified RNA molecules are characterized as tRNA-Val(UAC) (tRNA-1) and tRNA-Leu(CAG) (tRNA-2).









TABLE 5







MS/MS data of RNase T1 signature digestion products of tRNA-1


(SEQ ID NO:237) and tRNA-2 (SEQ ID NO:218).















Calculated,
Deconvoluted


Measured



Signature T1 digestion
massª
mass
Differenceb

mass


tRNA
sequence
(Da)
(Da)
(Da)
m/z
(Da)

















AU[s4U]AGp
1649.189
1649.189
0
[M − 2H]2−
823.587



CUCAGp
1608.216
1608.216
0
[M − H]
1607.206



CDGp
976.138
976.139
0.001
[M − H]
975.132



CACCUCCCU
4820.644
4820.652
0.008
[M − 3H]3−
1605.876



[cmo5U]AC







tRNA-1
[m6A]AGp








(SEQ ID NO:242)








[m7G]UCGp
1333.186
1333.185
−0.001
[M − H]
1332.177



TΨPCGp
1294.163
1294.164
0.001
[M − H]
1293.155



AUCCCGp
1913.257
1913.262
0.005
[M − 2H]2−
955.623



UCAUCACCCACCA
4001.590
4001.593
0.003
[M − 3H]3−
1333.191



(SEQ ID NO:243)







tRNA-2
AAGp
1021.161
1021.164
0.003
[M − H]−
1020.156



AADD[Gm]Gp
1996.305
1996.315
0.01
[M − 2H]2−
997.150



DAGp
1000.150
1000.154
0.004
[M − H]
999.146



ACGp
997.150
997.154
0.004
[M − H]
996.149



CUAGp
1303.175
1303.182
0.007
[M − H]
1302.171



CUUCAGp
1914.241
1914.251
0.010
[M − 2H]2−
956.118



ΨUAGp
1304.159
1304.161
0.002
[M − H]
1303.154



UCCUUACGp
2525.307
2525.320
0.013
[M − 2H]2−
1262.159



TΨCAAGp
1952.268
1952.278
0.01
[M − 2H]2−
975.131



UCCCCCCCCUCGp
3720.476
3720.492
0.016
[M − 3H]3−
1239.492



(SEQ ID NO:244)








CACCA
1511.271
1511.279
0.008
[M − 2H]2−
754.633






atheoretical monoisotopic mass




bDifference = (Deconvoluted mass) − (Calculated mass)







Example 3
Preparation of tRNA-Half Molecules

Further, the inventors employed specific S1 nuclease to prepare tRNA-half molecule. Each 500 ng of purified tRNA molecule was dissolved with DEPC-treated RNase-free water and mixed with 8 units of S1 nuclease, followed by added with 2 μL of 5× reaction buffer solutions and mixed with deionized water to 20 μL. After incubation in water bath at 25° C. for 40 min, the mixed solutions were with 0.5 μL of 0.5 M EDTA solution to terminate the reaction. After centrifugation at 10000×g for 1 min, the supernatant was collected and analyzed by urea denatured polyacrylamide gel electrophoresis. FIG. 5A shows that compared to tRNA-Leu(CAG), the digestion products clearly exhibited 2 fragments in the length of almost 30 nt long, which demonstrated the enzyme digestion succussed and the products were tRNA-Leu(CAG) half. Subsequently, the digestion products were injected to ultra-high performance liquid chromatography and separated using the chromatographic condition in EXAMPLE 2. See the prepared tRNA-half molecules in FIG. 5B, the prepared solutions were concentrated and freeze-dried, and stored at −80° C. The tRNA-Val(UAC) half was obtained using the above method.


Example 4
Synthesis of RNA Molecule

The inventors designed and synthesized RNA molecules having a length of about 22 bp based on the 147 isolated tRNA sequences in database. In particular, the tRNA sequences are considered to have at least 3 portions, namely a 5′-terminal portion (5′-t), a 3′-terminal portion (3′-t) and an anticodon portion. Each of the specifically designed RNA molecules contains any one of the portions. For instance, designed RNA molecules containing a 5′ terminal portion of the corresponding full-length tRNA sequence are referred as 5′-t group RNA molecules; designed RNA molecules containing a 3′ terminal portion of the corresponding full-length tRNA sequence are referred as 3′-t group RNA molecules; designed RNA molecules containing an anticodon portion of the corresponding full-length tRNA sequence are referred as anticodon group RNA molecules. The RNA molecules having a sense sequence selected from SEQ ID NO: 101 to SEQ ID NO: 194 and a complementary antisense sequence selected from SEQ ID NO: 7 to SEQ ID NO: 100, as shown in Table 3, were designed and synthesized by cleavage at different sites on the tRNA sequences in Table 1. The RNA molecules having an antisense sequence selected from SEQ ID NO: 1 to SEQ ID NO: 3 and a complementary sense sequence selected from SEQ ID NO: 4 to SEQ ID NO: 6, as shown in Table 4, were designed and synthesized by cleavage at different sites on the EC83 sequence in Table 3.


Example 5
Cytotoxic Effect of tRNA, tRNA-Half and tRF Mimic Molecules on Colorectal Cancer Cells

HCT-8, fluorouracil-resistant HCT-8, fluorouracil-resistant LoVo cell lines were cultured in RPMI Medium 1640 medium containing 10% FBS and 1% penicillin/streptomycin. LoVo cell line was cultured in F-12K medium containing 10% FBS and 1% penicillin/streptomycin. All cell lines above were cultured at humidified atmosphere containing 5% CO2 at 37° C.


In the cytotoxicity assay, exponentially growing cells of each cancer cell line were plated in 96-well microplate at a density of 5000 cells per well in 100 μL of culture medium and allowed to adhere for 24 h before treatment. Serial concentrations of RNA molecules obtained in Example 1 to 4 in a mixture containing a gene delivery carrier, i.e. Lipofectamine™ RNAiMAX Transfection Reagent (Thermo Fisher Scientific, U.S.A.) were then added to the cells. After treated for 48 h, MTT solution (50 μL per well, 1 mg/mL solution) was added to each well and incubated for 4 h at 37° C. Subsequently, 200 μL dimethyl sulfoxide (DMSO) were added and the optical densities of the resulting solutions were calorimetrically determined at 570 nm using a SpectraMax 190 microplate reader (Molecular Devices, Sunnyvale, CA, U.S.A). Dose-response curves were obtained, and the IC50 values were calculated by GraphPad Prism 5 (GraphPad, La Jolla, CA, USA). Each experiment was carried out for three times. IC50 results were expressed as means±standard deviation.


With reference to Figure. 6A, HCT-8 cells were treated with 50 nM RNA molecules of tRNA-Val(UAC), tRNA-Leu(CAG), 5′-tRNA-Val(UAC) half, 3′-tRNA-Val(UAC) half, 5′-tRNA-Leu(CAG) half, 3′-tRNA-Leu(CAG) half, EC41 mimic, EC42 mimic, EC85 mimic and EC86 mimic for 48 h before addition of MTT solution. The cell viability of these cells is compared to a control group and a RNAiMAX group. A comparative example using fluorouracil was conducted. FIG. 6A shows that these RNA molecules are also effective to colorectal cancer cells.



FIG. 6B shows the cytotoxic effect of synthetic RNA molecules in Example 4 on HCT-8 cells. The results demonstrated that in this example, RNA molecules designed and synthesized based on the ensured tRNA sequences in Example 4 are also effective to inhibit cancer cells, particularly are effective in inhibition on the growth and proliferation of colorectal cancer cells. Specifically, FIG. 6C shows that EC83 mimic exhibited the strongest cytotoxicity.


The inventors then specifically determined the cytotoxic effect and IC50 of RNA molecule EC83 mimic and different chemically modified RNA molecules EC83-M1, EC83-M2 and EC83-M3 mimic on HCT-8 and LoVo cells, at different concentrations, i.e. 6.25 nM, 12.5 nM, 25 nM, 50 nM and 100 nM. As shown in FIGS. 7A and 7B, the results are compared to a control group and a RNAiMAX group containing a transfecting agent. The results demonstrated that RNA molecule EC83 mimic and different chemically modified EC83-M1, EC83-M2, EC83-M3 mimic have a dose-dependent effect on inhibiting the growth and proliferation of colorectal cancer cells and their fluorouracil-resistant strains. Their IC50 values were summarized in Table 6. A comparative example was conducted using fluorouracil with results.









TABLE 6







IC50 values of EC83 mimic and different chemically modified


RNA molecules of EC83-M1, EC83-M2 and EC83-M3 mimic


on colorectal cancer cells and their fluorouracil-resistant strains.









IC50 (nM)













EC83
EC83-M1
EC83-M2
EC83-M3



Cell line
mimic
mimic
mimic
mimic
5-FU















HCT-8
70.6
77.9
58.9
74.5
 41400


HCT-8/5-FU
63.4
69.6
48.8
72.2
170600


LoVo
28.8
43.6
19.9
24.1
 63900


LoVo/5-FU
29.5
54.2
18.3
23.0
133200









In the clonogenic assay, HCT-8 or LoVo cells were plated in 6-well microplate at a density of 1000 cells per well in 2 mL of culture medium and allowed to adhere for 24 h before treatment. Single concentration of RNA molecules obtained in Example 1 to 4 in a mixture containing a gene delivery carrier, i.e. Lipofectamine™ RNAiMAX Transfection Reagent (Thermo Fisher Scientific, U.S.A.) were then added to the cells. After treated for 48 h, discarded the drug solutions and replaced with 2 mL of fresh culture medium. After culture 14 days, discarded the culture medium and the cells were fixated with 4% paraformaldehyde fix solution for 20 min, followed by stained with crystal violet for 10 min and then washed by deionized water. The 6-well microplate was photographed and the number of colonies with more than 50 individual cells were counted using ImageJ software (U.S.A.). Each experiment was carried out for three times. Results were expressed as means±standard deviation.


With reference to FIGS. 8A and 8B, HCT-8 cells and LoVo cells were treated with 50 nM or 25 nM RNA molecules of EC83 mimic and different chemically modified RNA molecules EC83-M1, EC83-M2 and EC83-M3 mimic. The colony number of these cells is compared to a control group. A comparative example using fluorouracil was conducted. The results show that these RNA molecules can significantly inhibit the proliferation of colorectal cancer cells. Specifically, EC83-M2 mimic exhibited the strongest anti-proliferation activity.


In the wound-healing assay, HCT-8 or LoVo cells were plated in 6-well microplate at a density of 500,000 cells per well in 2 mL of culture medium and allowed to adhere for 24 h before treatment. Serial concentrations of RNA molecules obtained in Example 1 to 4 in a mixture containing a gene delivery carrier, i.e. Lipofectamine™ RNAiMAX Transfection Reagent (Thermo Fisher Scientific, U.S.A.) were then added to the cells. After treated for 48 h, discarded the drug solutions and replaced with 2 mL of fresh culture medium. After culture 14 days, discarded the culture medium and the cells were fixated with 4% paraformaldehyde fix solution for 20 min, followed by stained with crystal violet for 10 min and then washed by deionized water. The 6-well microplate was photographed and the number of colonies with more than 50 individual cells were counted using ImageJ software (U.S.A.). Each experiment was carried out for three times. Results were expressed as means±standard deviation. A 1 mL bacteria-free pipette tip was used to make a cross-scratch at the bottom of 6-well microplate. The cells in the scratch were washed away using phosphor buffer solution and the 6-well microplate was photographed under a microscope. Single concentration of RNA molecules obtained in Example 1 to 4 in a mixture containing a gene delivery carrier, i.e. Lipofectamine™ RNAiMAX Transfection Reagent (Thermo Fisher Scientific, U.S.A.) were then added to the cells. The cells were then photographed under microscope at 24 and 48 h. ImageJ software was applied to quantify the area of wound created. Each experiment was carried out for three times. Results were expressed as means±standard deviation.


With reference to FIGS. 9A and 9B, HCT-8 cells and LoVo cells were treated with 50 nM or 25 nM RNA molecules of EC83 mimic and different chemically modified RNA molecules EC83-M1, EC83-M2 and EC83-M3 mimic. The wound area of these cells is compared to a control group. A comparative example using fluorouracil was conducted. The results show that these RNA molecules can significantly inhibit the migration of colorectal cancer cells. Specifically, EC83-M2 mimic exhibited the strongest inhibition effect on the wound healing of colorectal cancer cells.


NUMBERED EMBODIMENTS

The implementation is further described with reference to the following numbered embodiments.


Embodiment 1: A double-stranded RNA molecule comprising an antisense sequence selected from one of SEQ ID NO: 7 to SEQ ID NO: 100, and a sense sequence selected from one of SEQ ID NO: 101 to SEQ ID NO: 194; or a functional variant or homologue thereof.


Embodiment 2: The double-stranded RNA molecule of embodiment 1, wherein the antisense sequence is selected from one of SEQ ID NO: 47, 48, 89, 90, 91 and 92, and the sense sequence is selected from one of SEQ ID NO: 141, 142, 183, 184, 185 and 186; or a functional variant or homologue thereof.


Embodiment 3: The double-stranded RNA molecule of embodiment 1, wherein the antisense sequence is SEQ ID NO: 47, 48, 89, 90, 91 and 92, and the sense sequence is SEQ ID NO: 141, 142, 183, 184, 185 and 186; or a functional variant or homologue thereof.


Embodiment 4: The double-stranded RNA molecule of embodiment 1, wherein the antisense sequence is SEQ ID NO: 89, and the sense sequence is SEQ ID NO: 183; or a functional variant or homologue thereof.


Embodiment 5: The double-stranded RNA molecule of embodiment 1 comprising a 3′ overhang; or a functional variant or homologue thereof.


Embodiment 6: The double-stranded RNA molecule of embodiment 1, wherein the antisense sequence and/or the sense sequence comprises one or more chemically modified nucleotides; wherein the one or more chemically modified nucleotides are selected from the group consisting of m1A, m2A, m5A, m7A, m2G, m6A, m22G, Um, i6A, ms2i6A, t6A, m6t6A, s2C, s2U, s4U, ac4C, f5C, acp3U, mo5U, cmo5U, mcmo5U, mcm5U, mcm5Um, mcm5s2U, nm5s2U, mnm5U, mnm5s2U, ncm5U, ncm5Um, cmnm5U, cmnm5Um, cmnm5s2U, tm5U and tm5s2U; or a functional variant or homologue thereof.


Embodiment 7: The double-stranded RNA molecule of embodiment 1, wherein the antisense sequence and/or the sense sequence comprises one or more chemically modified nucleotides; wherein the one or more chemically modified nucleotides are selected from the group consisting of m1A, m7G, m6A, Gm, Cm, Am, Um, m22G, s4U and cmo5U; or a functional variant or homologue thereof.


Embodiment 8: The double-stranded RNA molecule of embodiment 7, wherein the one or more chemically modified nucleotides are s4U and/or Gm; or a functional variant or homologue thereof.


Embodiment 9: The double-stranded RNA molecule of embodiment 1, wherein the antisense sequence and/or the sense sequence comprises at least one nucleotide having one or more chemical modifications; wherein the nucleotide is selected from the group consisting of adenosine, guanosine, cytidine and uridine; or a functional variant or homologue thereof; and the one or more chemical modifications is selected from the group consisting of 1-methyl, 2-methyl, 5-methyl, 7-methyl, N2 methyl, N6 methyl, N2,N2 dimethyl, 2′-O-methyl, N6-isopentenyl, 2-methylthio-N6-isopentenyl, N6 threonide carbamoyl, N6-methyl-N6-threosylcarbamoyl, 2-thio, 4-thio, N4 acetyl, 5-formyl, 3-(3-amino-3-carboxypropyl), 5-methoxy, 5-oxoacetic acid, 5-oxoacetate methyl ester, 5-methoxycarbonylmethyl, 5-methoxycarbonylmethyl-2′-O-methyl, 5-methoxycarbonylmethyl-2-thio, 5-aminomethyl-2-thio, 5-methylaminomethyl, 5-methylaminomethyl-2-thio, 5-aminoformylmethyl, 5-aminoformylmethyl-2′-O-methyl, 5-carboxymethyl aminomethyl, 5-carbamoylmethyl-2′-O-methyl, 5-carboxymethylaminomethyl-2-methyl, 5-taurine, 5,2′-O-dimethyl, and 5-tauromethyl-2-thio.


Embodiment 10: The double-stranded RNA molecule of embodiment 9, wherein the one or more chemical modifications is selected from the group consisting of 1-methyl, 7-methyl, N6 methyl, 2′-O-methyl, 5,2′-O-dimethyl, 4-thio, and 5-oxoacetic acid.


Embodiment 11: The double-stranded RNA molecule of embodiment 9, wherein the nucleotide is uridine and/or guanosine; or a functional variant or homologue thereof.


Embodiment 12: A double-stranded RNA molecule comprising an antisense sequence selected from the group consisting of SEQ ID NO: 1, 2 and 3, and a sense sequence selected from the group consisting of SEQ ID NO: 4, 5 and 6; or a functional variant or homologue thereof.


Embodiment 13: The double-stranded RNA molecule embodiment 12, wherein the antisense sequence is SEQ ID NO: 1, 2 or 3, and the sense sequence is SEQ ID NO: 4, 5 or 6; or a functional variant or homologue thereof.


Embodiment 14: A pharmaceutical composition comprising the double-stranded RNA molecule of embodiment 1 or a functional variant or homolog thereof, and a pharmaceutically tolerable carrier, diluent and/or excipient.


Embodiment 15: The pharmaceutical composition of embodiment 14, wherein the pharmaceutical composition further comprises a nucleic acid stabilizer.


Embodiment 16: The pharmaceutical composition of embodiment 14, wherein the pharmaceutical composition is used for preventing and/or treating cancer, wherein the cancer is colorectal cancer or fluorouracil resistant cancer.


Embodiment 17: A method of preventing and/or treating cancer in a subject in need thereof, comprising administering an effective amount of the double-stranded RNA molecule of embodiment 1 to the subject.


Embodiment 18: The method of embodiment 17, wherein the cancer is colorectal cancer or fluorouracil resistant cancer.

Claims
  • 1. A double-stranded RNA molecule comprising an antisense sequence selected from one of SEQ ID NO: 7 to SEQ ID NO: 70 and SEQ ID NO: 72 to SEQ ID NO: 100, and a sense sequence selected from one of SEQ ID NO: 101 to SEQ ID NO: 164 and SEQ ID NO: 166 to SEQ ID NO: 194.
  • 2. The double-stranded RNA molecule of claim 1, wherein the antisense sequence is selected from one of SEQ ID NO: 47, 48, 89, 90, 91 and 92, and the sense sequence is selected from one of SEQ ID NO: 141, 142, 183, 184, 185 and 186.
  • 3. The double-stranded RNA molecule of claim 1, wherein the antisense sequence is SEQ ID NO: 89, and the sense sequence is SEQ ID NO: 183.
  • 4. The double-stranded RNA molecule of claim 1 comprising a 3′ overhang.
  • 5. The double-stranded RNA molecule of claim 1, wherein the antisense sequence and/or the sense sequence comprises one or more chemically modified nucleotides; wherein the one or more chemically modified nucleotides are selected from the group consisting of m1A, m2A, m5A, m7A, m2G, m6A, m22G, Um, i6A, ms2i6A, t6A, m6t6A, s2C, s2U, s4U, ac4C, f5C, acp3U, moSU, cmo5U, mcmo5U, mcm5U, mcm5Um, mcm5s2U, nm5s2U, mnm5U, mnm5s2U, ncm5U, ncm5Um, cmnm5U, cmnm5Um, cmnm5s2U, tm5U and tm5s2U.
  • 6. The double-stranded RNA molecule of claim 1, wherein the antisense sequence and/or the sense sequence comprises one or more chemically modified nucleotides; wherein the one or more chemically modified nucleotides are selected from the group consisting of m1A, m7G, m6A, Gm, Cm, Am, Um, m22G, s4U and cmo5U.
  • 7. The double-stranded RNA molecule of claim 6, wherein the one or more chemically modified nucleotides are s4U and/or Gm.
  • 8. The double-stranded RNA molecule of claim 1, wherein the antisense sequence and/or the sense sequence comprises at least one nucleotide having one or more chemical modifications; wherein the nucleotide is selected from the group consisting of adenosine, guanosine, cytidine and uridine; and the one or more chemical modifications is selected from the group consisting of 1-methyl, 2-methyl, 5-methyl, 7-methyl, N2 methyl, N6 methyl, N2,N2 dimethyl, 2′-O-methyl, N6-isopentenyl, 2-methylthio-N6-isopentenyl, N6 threonide carbamoyl, N6-methyl-N6-threosylcarbamoyl, 2-thio, 4-thio, N4 acetyl, 5-formyl, 3-(3-amino-3-carboxypropyl), 5-methoxy, 5-oxoacetic acid, 5-oxoacetate methyl ester, 5-methoxycarbonylmethyl, 5-methoxycarbonylmethyl-2′-O-methyl, 5-methoxycarbonylmethyl-2-thio, 5-aminomethyl-2-thio, 5-methylaminomethyl-2-thio, 5-aminoformylmethyl, 5-aminoformylmethyl-2′-O-methyl, 5-carboxymethyl aminomethyl, 5-carbamoylmethyl-2′-O-methyl, 5-carboxymethylaminomethyl-2-methyl, 5-taurine, 5,2′-O-dimethyl, and 5-tauromethyl-2-thio.
  • 9. The double-stranded RNA molecule of claim 8, wherein the one or more chemical modifications is selected from the group consisting of 1-methyl, 7-methyl, N6 methyl, 2′-O-methyl, 5,2′-O-dimethyl, 4-thio, and 5-oxoacetic acid.
  • 10. The double-stranded RNA molecule of claim 8, wherein the nucleotide is uridine and/or guanosine.
  • 11. A double-stranded RNA molecule comprising an antisense sequence selected from the group consisting of SEQ ID NO: 1, 2 and 3, and a sense sequence selected from the group consisting of SEQ ID NO: 4, 5 and 6.
  • 12. A pharmaceutical composition comprising the double-stranded RNA molecule of claim 1; and a pharmaceutically tolerable carrier, diluent and/or excipient.
  • 13. The pharmaceutical composition of claim 12, wherein the pharmaceutical composition further comprises a nucleic acid stabilizer.
  • 14. The pharmaceutical composition of claim 12, wherein the pharmaceutical composition is used for treating cancer, wherein the cancer is colorectal cancer or fluorouracil resistant cancer.
  • 15. A method of treating cancer in a subject in need thereof, comprising administering an effective amount of the double-stranded RNA molecule of claim 1 to the subject.
  • 16. The method of claim 15, wherein the cancer is colorectal cancer or fluorouracil resistant cancer.
Priority Claims (1)
Number Date Country Kind
202010083971 .2 Feb 2020 CN national
Foreign Referenced Citations (2)
Number Date Country
WO-03106631 Dec 2003 WO
WO-2004046375 Jun 2004 WO
Non-Patent Literature Citations (3)
Entry
Ahmed et al. PLoS One 6(8): e23443, pp. 1-8 (Year: 2011).
Mlotshwa, S., et al.(2015). A novel chemopreventive strategy based on therapeutic microRNAs produced in plants. Cell research, 25(4), 521-524.
Goodarzi, H., et al.(2015). Endogenous tRNA-derived fragments suppress breast cancer progression via YBX1 displacement. Cell, 161(4), 790-802.
Related Publications (1)
Number Date Country
20210254066 A1 Aug 2021 US